Accepted Manuscript Review Cancer combination therapies with artemisinin-type drugs Thomas Efferth PII: DOI: Reference:
S0006-2952(17)30181-8 http://dx.doi.org/10.1016/j.bcp.2017.03.019 BCP 12773
To appear in:
Biochemical Pharmacology
Received Date: Accepted Date:
20 January 2017 28 March 2017
Please cite this article as: T. Efferth, Cancer combination therapies with artemisinin-type drugs, Biochemical Pharmacology (2017), doi: http://dx.doi.org/10.1016/j.bcp.2017.03.019
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.
Cancer combination therapies with artemisinin-type drugs Thomas Efferth1 1
Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes
Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany Tel.: 49-6131-3925751; Fax: 49-6131-3923752; E-Mail:
[email protected]
Running title: Artemisinin-based combination therapies Key words: Chemotherapy, Drug resistance, Natural products, Sesquiterpenoids, Toxicology
Abbreviations: ACE, angiotensin I-converting enzyme; ACT, artemisinin-based combination therapy; Akt, AKT seronine/threonine kinase, v-akt murine thymoma viral oncogene homologue; AMP, adenosine monophosphate; ARE, arteether; ARM, artemether; ARS, artemisinin; ART, artesunate; BCL- XL, break point cluster X, long; BCRP, breast cancer resistance protein; BMI1, B-lymphoma Mo-MLV insertion region 1 homologue (mouse); Ca, carcinoma; CDK2, cyclin-dependent kinase 2; CIP, cyclin-interacting protein; CRISPR, clustered regulatory interspaced short palindromic repeats; CYP, cytochrome P450 monooxygenase; DHA, dihydroartemisinin; DR5, death receptor 5; ERK1/2, extracellular signal-regulated kinase 1/2; FAS, Fas cell surface death receptor; 1
FDA, Food and Drug Administration; Fos, FBJ murine osteosarcoma viral (v-fos) oncogene homologue; G6PD, glucose-6-phosphate dehydrogenase; GSH, reduced glutathione; GSSG, oxidized glutathione; GST, glutathione S-transferase; HDAC, histone deacetylase; HIF-1α, hypoxia-induced factor 1-alpha HSP, heat shock protein; KIP, kinase-interacting protein; MAPK, mitogen-activated protein kinase; MDR, multidrug resistance; MMP, matrix metalloproteinase; MRP, multidrug resistance-related protein; NADP+, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor kappa-B; NHL, non-Hodgkin lymphoma; PARK7, Parkinonism-associated deglycase 7; P-gp, P-glycoprotein; PUVA, psoralen plus UVA light therapy; RAC-1, Ras-related C3 Botulinum toxin substrate 1 (Rho family); RAD51, RAD51 (radiation 51) recombinase; R-CHOP, rituximab, cyclophosphamide, vincristine, prednisone combination therapy; ROS, reactive oxygen species; Sp1, specificity protein 1 transcription factor; TCM, traditional Chinese medicine; TRAIL, tumor necrose factor receptor apoptosis-inducing ligand; VCAM-1, vascular cell adhesion molecule 1; VEGF, vascular endothelial growth factor; XIAP, X-linked inhibitor of apoptosis; YY1, yin yang 1 transcription factor.
2
Abstract Artemisia annua L. is a Chinese medicinal plant, which is used throughout Asia and Africa as tea or press juice to treat malaria. The bioactivity of its chemical constituent, artemisinin is, however, much broader. We and others found that artemisinin and its derivatives also exert profound activity against tumor cells in vitro and in vivo. Should artemisinin-type drugs be applied routinely in clinical oncology in the future, then probably as part of combination therapy regimens rather than as monotherapy. In the present review, I give a comprehensive overview on synergistic and additive effects of artemisinin-type drugs in combination with different types of cytotoxic agents and treatment modalities: (a) standard chemotherapeutic drugs, (b) radiotherapy and photodynamic therapy, (c) established drugs for other indications than cancer, (d) novel synthetic compounds, (e) natural products and natural product derivatives, (f) therapeutic antibodies and recombinant proteins, and (g) RNA interference. I also summarize the activity of artemisinin-type drugs towards multidrug-resistant cells and tumor cells with other drug resistance phenomena. As synergistic interactions may not only occur in tumor cells, toxic reactions in normal cells (hepatotoxicity, drug interactions) were also considered. This review summarizes the scientific literature of more than 20 years until the end of 2016.
1 Introduction Artemisinin (ARS) from the Sweet wormwood (qinhao, Artemisia annua L., Asteraceae) is a medicinal plant used in Chinese medicine to treat chills and fever. In the 1970 and 1980s it turned out that the drug reveals surprisingly strong efficacy towards Plasmodia. Nowadays, artemisinin belongs to the standard treatment protocols to treat malaria. Because artemisinin saved millions of lives of malaria patients, Youyou Tu, who first identified the malaria activity of A. annua, was honored with the Nobel Prize for Medicine or Physiology 2015 [1]. Interestingly, A. annua preparations (decoctions, tea, press juice) are frequently used outside the official medical system in Asia and Africa to treat malaria [2,3]. During the past years, experimental evidence was accumulated that ARS activity is not restricted to malaria and that it may also be of therapeutic interest for several other diseases, including cancer [4-12]. Should ARS-type drugs ever be used routinely in clinical oncology, then with high probability as part of combination therapy protocols rather than as monotherapy [13-15]. The principles, which are common sense for combination therapy protocols with established anticancer drugs, also have to be applied for combinations with ARS-type drugs, 3
i.e. (1) prevention of resistance development by combining drugs with different targets and modes of action; (2) dispersion of side effects to different tissues in the body to minimize the occurrence of severe or fatal toxicities; (3) each constituent of the combination protocol has to reveal anticancer activity, if applied as single drug etc. [16-18]. In the preclinical setting, synergy has to be proven by appropriate biostatistical methods [19, 20] in well suitable experimental biological models in vitro and in vivo [21-25]. Frequently, authors publish their data by claiming synergistic interactions of compound combinations without reliable proof, whether or not the observed effects are indeed synergistic in nature. This is a considerable limitation of many data available in the literature in pharmacology in general and also in the case of ARS-type drugs. A wealth of results has been published in the literature on additive or synergistic combinations of ARS-type drugs with other compounds to improve tumor growth inhibition in cell lines and in animal experiments (Figure 1). The intention of the present review was to give an overview of the published data and to outline perspectives for further research developing the most suitable combination therapy protocols for ARS-based cancer therapy. A central question in this context is about the molecular targets of ARS-type drugs in cancer cells. Despite a plethora of published data, this question is not ultimately resolved. Most likely, this class of compounds acts in a multi-specific manner exhibiting several modes of action at the same time. This is a typical feature of most - if not all – natural products [26]. The endoperoxide moiety of ARS-type drugs can be opened leading to the formation of radical oxygen species and carbon-centered radical molecules. This process is facilitated by free ferrous iron or iron-bound proteins (e.g. heme) in a Fenton-type reaction. These radical molecules may exert a broad range of detrimental effects in cancer cells. As previously outlined, the activity of antitumor drugs in general may be categorized as (1) mechanisms acting upstream of the actual drug target, (2) target site mechanisms, and (3) mechanisms acting downstream of the drug targets [27]. This model may also be applied for ARS-type drugs. As described in great detail in a recent review [28], molecular modes of action of ARStype drugs in cancer cells include: (1) Upstream mechanisms: antioxidant response mechanisms, receptor signaling pathways (e.g. EGFR, WNT/β-catenin, BCR/ABL) (2) Target site mechanisms: DNA damage and repair mechanisms, alkylation of target proteins (e.g. TCTP, LDH etc.), cell cycle arrest, neoangiogenesis, invasion and metastasis (3) Downstream mechanisms: apoptotic and non-apoptotic cell death (autophagy, ferroptosis).
4
2 Combination with standard chemotherapy In the past few years, a plethora of papers appeared on the interaction of ARS-type drugs with clinically established anticancer agents, many of which are acting directly or indirectly on tumor DNA by damaging its integrity in one or another way (the DNA topoisomerase II inhibitor doxorubicin, the adduct-forming platin compounds, the alkylating agents temozolomide) or disturbing DNA-biosynthesis (gemcitabine, cytarabine) (Table 1). Since ARS-type drugs cause oxidative DNA lesions and double strand breaks [29-35], it is worth hypothesizing that their interaction with DNA-affecting anticancer drugs may provoke enhanced tumor cell killing. Indeed, additive to synergistic interactions of ARS, DHA, or ART have been observed in combination with standard anticancer drugs towards tumor cell lines of diverse origin, e.g. hematopoetic tumors (leukemia, multiple myeloma) as well as epithelial carcinoma (of breast, ovarian, lung, brain tumor, liver, prostate, or pancreas). Importantly, these effects were not only measured in vitro but also in vivo, which raises the potential clinical utility of this kind of drug combinations. In case that ARS-type drugs should ever reach clinical routine application for cancer therapy, it is important to investigate the molecular modes of action of these drug-drug interactions in preclinical models. As shown in Table 1, mechanisms contributing to additive or synergistic effects of ARS-type drugs in combination with established anticancer drugs include effects on DNA repair (RAD51) and signal transduction pathways (mTOR, NF-κB) as well as angiogenesis (VEGF, HIF-1α) and induction of cell death (apoptosis, autophagy). The enhancement of activity of ARS-type drugs was not only observed in combination with anticancer drugs that act on DNA, but also on other targets of tumor cells. As shown in Table 2, combinations with tyrosine kinase inhibitors (erlotinib), proteasome inhibitors (bortezomib), corticosteroides (dexamethasone), as well as immunomodulating and antiangiogenic drugs (thalidomide, lenalidomide) cereblon (CRBN) E3 ubiquitin ligase as the primary target, all led to additive or synergistic inhibition rates of tumor cells.
3 Activity against drug-resistant tumor cells The development of drug resistance and unwanted side effects represent main reasons for the failure of chemotherapy in clinical oncology. Severe and partly life-threatening side effects prevent the administration of drug concentrations high enough to kill all cell populations in a tumor. 5
A dreaded phenomenon in cancer chemotherapy is multidrug resistance (MDR). This type of drug resistance represents cross-resistance to a wide array of functionally and structurally distinct anticancer drugs, e.g. Vinca alkaloids,
taxanes, anthracenes,
epipodophyllotoxins and others. The underlying mechanisms of MDR phenomena are drug efflux pumps of the ATP-binding cassette (ABC) transporter family, which extrude drug molecules that passively diffused into cancer cells in an active ATP-consuming manner out of the cells. This drug efflux keeps intracellular drug concentrations at low sub-lethal levels, which ultimately favors survival of cancer cells and failure of chemotherapy with fatal consequences for cancer patients. Well-known ABC-transporters that confer MDR are Pglycoprotein (ABCB1, MDR1), MDR-related proteins (ABCCs, MRPs) and breast cancer resistance protein (ABCG2, BCRP) [36,37]. In addition to MDR, resistance mechanisms to single anticancer drugs without broad cross-resistance profiles also hamper the success of chemotherapy. Resistance to DNAdamaging agents (e.g. cisplatin), DNA-biosynthesis (methotrexate, hydroxyurea), tyrosine kinase inhibitors (e.g. imatinib) and other anticancer drugs have been described in the past years [38-41]. Therefore, there is an intensive ongoing search for novel compounds that are able to combat drug resistance. In principal there are three main categories to overcome drug resistance: (1) specific inhibitors of resistance mechanisms, e.g. efflux inhibitors of ABCtransporters, (2) cytotoxic compounds that are not recognized by resistance mechanisms and which thereby bypass drug resistance, and (3) compounds that exert hypersensitivity (collateral sensitivity) in drug-resistant cells [42,43]. The therapeutic potential of ARS-type drugs in oncology is not only visible by the fact that they show profound activity in vitro and in vivo against cancer cells [44], but also that they do not exert cross-resistance or even do reveal collateral sensitivity (hypersensitivity) against ABC-transporter-mediated MDR and also resistance to methotrexate, hydroxyurea, cisplatin or imatinib (Table 3, Figure 2). Interestingly, some ARS-derivatives were also able to inhibit P-gp at the blood brain barrier making them interesting candidates to improve glioblastoma therapy [45].
4 Combination with radiotherapy or photodynamic therapy Since their discovery in 1895, X-rays are used for diagnosis and treatment in medicine. Nowadays, radiotherapy represents one of the main pillars of cancer therapy [46-48]. Radiotherapy kills tumors by damaging their DNA. As more and more sophisticated 6
technologies are available, the normal tissue surrounding the tumor is less affected. Furthermore, normal cells repair damaged DNA better than tumor cells. Nevertheless, the development of resistance represents also a major impediment of radiotherapy, and radiosensitizers are investigated to overcome radioresistance [49,50]. Some authors attempted to sensitize tumor cells to radiotherapy by the application of ART or DHA and found that resensitization effects were associated with ROS or nitric oxide generation, cell cycle arrest and induction of apoptosis [51-55] (Table 4). In vivo experiments are missing as of yet and it remains to be seen, whether or not the side effects of ARS-based radiochemotherapy are tolerable. Another therapeutic mode to combat tumors is photodynamic therapy. A systemically given photosensitizing agent is preferentially accumulated in tumor cells. Then, light is applied, which causes oxidative reactions with the photosensitizer inducing tumor cell killing [56]. Although there is not much experience thus far with the use of ARS-type drugs for photodynamic therapy, first favorable results were reported [57]. DHA enhanced PDTinduced growth inhibition and apoptosis in human esophageal cancer cell lines. DHA inhibited NF-κB activated by photodynamic therapy, which in turn led to down-regulation of the NF-κB target, Bcl-2.
5 Combination with drugs for indications other than cancer The rationale for investigating, whether drugs established for other diseases might cooperate together with ARS-type drugs is that patients occasionally do not only suffer from cancer, but also from other diseases at the same time [58,59]. Comorbidity in cancer patients occasionally can lead to unwanted drug interactions because of multiple medications. On the other side, multiple medications might also enhance growth inhibition of tumors. For instance, the angiotensin I converting enzyme (ACE) inhibitors, which are widely used for the treatment of cardiovascular diseases, also exert synergistic activity towards tumors in combination with anticancer drugs [60,61]. Recently, drug repurposing came into the center of interest. ARS-type drugs as antimalarial drugs are an example, since they also show activity against tumors. It is not beyond the expectations that their combination with drugs with indications other than cancer may also improve anticancer treatment efficacy. Indeed, there are some publications substantiating this speculation (Table 5). The ACE inhibitor captopril enhanced the effects of ART by inhibition of angiogenesis [62]. Elderly patients can suffer from congestive heart failure or arterial hypertension Therefore, captopril-ART combinations are reasonable. 7
Remarkably, the use of ACE inhibitors was correlated with a lower incidence of skin cancer [63]. Especially in tropical countries, it is not a rare condition that patients suffer from both malaria and cancer at the same time. Comparable to ARS-type drugs, the antimalarial drug chloroquine also showed anticancer effects [64]. The combination of chloroquine and ARS exerted synergistic interaction by ROS generation and induction of autophagy in lung cancer cells [65]. Another example is the antifunal agent miconazole, which is used for the treatment of antifungal infections in cancer patients [66]. An interaction of ARS and miconazol in killing tumor cells has been reported [67]. The published data on the interaction of ARS-type drugs with drugs approved for other diseases than cancer were found by chance and were not a result of systematic screening for synergistic interactions against tumors. More detailed analyses might unravel novel interesting drug combinations justifying the repurposing of established drugs for cancer therapy. Analyses for novel drug combinations should, however, also consider that drug interactions could cause unwanted toxicities, as will be discussed in the chapter on hepatotoxicity below.
6 Combination with novel synthetic compounds The synthesis of novel cytotoxic compounds represents an important part of the anticancer drug development process in academia and industry. Therefore, there is a constant interest to test experimental combinations of new compounds with clincially established or investigational new drugs. In this contest, the combination of ARS-type drugs with several novel synthetic compounds has been reported (Table 6). ROS or carbon-centered radical molecules generated by drugs or irradiation can be detoxified by reduced glutathione (GSH). Cellular GSH contents as well as enzymes related to GSH metabolism are long known as determinants of cancer cells to chemo- and radiotherapy [68,69]. This has also been shown for ART. The GSH biosynthesis inhibitor, Lbuthionine sulfoximine and the glutathione S-transferase (GST) inhibitor, ethacrynic acid sensitize tumor cells to ART [70]. Another enzyme related to the cellular detoxification capacity is glucose-6-phosphate dehydrogenase (G6PD). This is the key enzyme of the pentose-phosphate pathway, which catalyzes the conversion of glucose-6-phosphate to D8
glucono-1,5-lactone-6-phosphate by reduction of NADP+ to NADPH. NADPH is a cofactor of glutathione reductase, which converts oxidized glutathione (GSSG) to its reduced form (GSH). G6PD fosters cancer cell proliferation and pharmacological inhibition of G6PD is considered as novel therapeutic approach against cancer [71-73]. Increased pentose phosphate pathway and G6PD activities accompanied by increased amounts of GSH are determinants of drug resistance [74]. On the other hand, peripheral mononuclear blood cells from patients suffering from inherited G6PD-defficiency reveal increased oxidative stress, DNA damage and susceptibility to undergo apoptosis upon treatment with cytostatic drugs or radiation [7577]. Therefore, it can be hypothesized that pharmacological inhibition of G6PD may also sensitize tumor cells to ARS-type drugs. Indeed, the G6PD inhibitors physcion and S3 interacted with DHA in a synergistic manner to inhibit leukemia cell growth [78]. Another attractive treatment target to inhibit tumor growth is glutamine utilization. Glutaminolysis represents the first and rate-limiting step of glutamine utilization and is catalyzed by glutaminase (GLS). Glutaminase-1 (GLS1) was upregulated in tumor cells [79], and inhibition of GLS1 resensitized tumor cells to standard anticancer agents [80,81]. The GLS1 inhibitor 968 revealed synergistic interaction with DHA by excessive ROS generation [82]. Aberrant histone deacetylase (HDAC) activity has been implicated in cancer and HDACs are interesting targets for treatment [83,84]. Inhibition of HDACs increased the cytotoxicity of diverse established anticancer drugs, including oxaliplatin, docetaxel, 5fluorouracil and others [85-87]. Comparable results have been published for the combination of HDAC inhibitors and DHA in vitro and in vivo [88]. In a comparable manner as discussed for drug repurposing in the chapter above, unwanted interactions also are considered for combinations of ARS-type drugs with novel synthetic compounds. The nitrone compound, α-phenyl-tert-butylnitrone, and related nitrone substances revealed anticancer activity in preclinical models [89]. Their combination with DHA led, however, to attenuation of DHA cytotoxicity [90], indicating that this drug combination should be avoided.
7 Combination with natural products or natural product derivatives Natural products and derivatives thereof always played an important role for the treatment of diseases. This is especially true for cancer therapy. Drugs such as Vinca alkaloids (vincristine, vinlastine), taxanes (paclitaxel, docetaxel), camptothecins (topotecan, irinotecan), and 9
epipodophyllotoxins (etoposide, teniposide) are just a few prominent examples. A previous survey of the National Cancer Institute in USA revealed that the vast majority of all clinically established anticancer drugs are of natural origin [91,92]. Hence, it does not come as a surprise that natural products exert synergistic interactions with ARS-type drugs to inhibit tumor cells (Table 7). This has been reported for natural compounds used as constituents of dietary supplements such as vitamins, curcumin (a diarylheptanoid from Curcuma long L., Zingiberaceae), allicin (an organosulfor compound from Allium sativum L., Alliaceae), and butyrate (a bacterial product of anaerobic fermentation of milk products). Furthermore, chemical constituents of medicinal plants also enhanced growth inhibitory properties of ARStype drugs, e.g. dictamnine (a furoquinoline alkaloid from the roots of Dictamnus albus L., Rutaceae), salicylic acid (a phenolic acid that acts as plant hormone), resveratrol (a stilbenoid that is produced by many food plants as phytoalexin in response to stress and injury), triptolide (a diterpene epoxide from Triperygium wilfordii Hook.f., Celastraceae), and halofuginone (a quinazolinone alkaloid from Dichroa febrifuga Lour., Hydrangaceae). Some of these plants are used in traditional Chinese medicine (TCM). Inorganic compounds such as arsenic trioide, which also used in TCM, also enhance growth inhibition in combination with ARS-type drugs. ARS did not only exert additive or synergistic interactions with isolated compounds of other plants, but also with other phytochemicals (flavonoids, scopoletin, 1,8-cineole etc.) present in Artemisia annua [93,94]. This indicates that plant extracts from this plant may act as genuine combination therapies to combat cancer growth.
8 Combination with therapeutic antibodies or recombinant proteins The rapid development of gene- and biotechnological technologies during the past years facilitated the production of monoclonal antibodies and recombinant proteins for therapeutic purposes. Nowadays, antibodies belong the standard armory to treat cancer, and frequently they are combined with standard chemotherapy. Importantly, antibodies and clinically established chemotherapeutics are known to exert synergistic interactions leading to improved tumor eradication [95-97]. It can be envisaged that novel treatment regimens combining chemotherapy, adoptive antibody therapy and specific immunotherapy will be developed in an endeavor to develop novel strategies of precision medicine [98,99]. The chimeric anti-CD20 monoclonal antibody, rituximab, was the first FDA-approved therapeutic antibody for B-non-Hodgkin's lymphoma (B-NHL) therapy. The combination regimen R-CHOP (rituximab, cyclophosphamide, vincristine, prednisone) led to significant 10
prolonged survival times of B-NHL patients [100].
Rituximab inhibited several signal
transduction pathways that confer resistance to conventional chemotherapy, e.g. the p38 MAPK, NF-κB, (ERK 1/2) and AKT anti-apoptotic survival pathways, which mediated apoptosis resistance by regulating the expression of members of the Bcl-2 family. This antibody also inhibited the transcription factor YY1, which mediated apoptosis resistance via Fas (Apo-1) and TRAIL-R1 (DR5, Apo-2) regulation [101,102]. Rituximab increased the cytotoxicity towards ART in Ramos NHL cells [103]. ART induced both receptor-driven extrinsic and mitochondrial intrinsic apoptosis by induction of Fas expression, ROS generation and loss of mitochondrial membrane potential. Rituximab augmented these cellular effects. The expression of the transcription factors YY1 and Sp1 was reduced by the combination of rituximab and ART. While ART increased the expression of antioxidant proteins (catalase, glutathione S-transferase-π), rituximab counteracted this effect and, thereby, diminished the antioxidant capacity of cells. The effects of rituximab on antioxidant stress response represent a novel mode of action of rituximab to overcome drug resistance. Defective apoptosis contributes to the development of tumors to chemo- and radiotherapy [104]. In addition to Fas (Apo1) and Fas ligand (FasL, Apo1L), other members of this receptor family also induce apoptosis. Tumor necrosis factor (TNF)-related apoptosisinducing ligand (TRAIL, Apo2L) bound to TRAIL receptors 1 and 2 (TRAIL-R1, TRAILR2), also known as death receptors 4 and 5 (DR4 and DR5). TRAIL-targeted therapy attracted much attention, but the clinical results were disappointing, since most human tumors were resistant to TRAIL-induced apoptosis [105,106]. Therefore, the quest for novel TRAIL-based therapies is ongoing. Among other compounds, phytochemicals from medicinal plants produced synergistic effects on TRAIL-induced apoptosis in tumor cells [107] Recently, several investigations described the combination of TRAIL with DHA or ART (Table 8). The authors observed enhanced induction of apoptosis due to ROS generation, induction of DR5 expression, as well as downregulation of other apoptosis-regulating proteins (XIAP, Bcl-xL, NF-κB, and Akt) [108-110].
9 Combination with RNA interference Gene therapy is a collective term for the delivery of nucleic acids to treat diseases. There are numerous experimental approaches from DNA incorporated into engineered vectors to naked DNA
vaccines,
antisense
oligodeoxynucleotides, 11
ribozymes,
RNA
interference,
nanotechnological delivery, and CRISPR. The targeted manipulation of gene expression may be used as strategy to improve response of tumor cells to standard chemotherapy [111]. In principle, this strategy is may be transferrable to any new drug, as long as the determinants of cellular responsiveness are known. Some authors reported on the combination of ARS-type drugs with short interference RNA or short hairpin RNA (Table 9). Vascular cell adhesion molecule-1 (VCAM1) is expressed in brain tumors and has a role for cell surface adhesion. A combination of ARM and VCAM1 shRNA promoted apoptosis in glioblastoma cells [112]. Moloney murine leukemia virus insertion site 1 (BMI1) regulated tumor cell proliferation in nasopharyngeal carcinoma. ARS inhibited growth of nasopharyngeal carcinoma cells, induced G1 cell cycle arrest and inhibited BMI1. These effects were augmented by combinational treatment of ARS plus BMI1 siRNA [113]. Rac1 is important in activation of NF-κB-mediated transcriptional processes. Down-regulation of Rac1 expression by siRNA enhanced DHA-induced growth inhibition, cell cycle arrest, apoptosis, and migration in tumor cells [114]. The PARK7 (DJ-1) protein was overexpressed in DHA-resistant cells as identified by a proteomic approach. Compared to DHA-sensitive cells, this protein was translocated to the mitochondria and oxidized [115]. Although the exact function is still elusive, PARK7 might reveal cytoprotective effects. The causative role of PARK7 for DHA resistance was demonstrated by siRNA, which resensitized cells to DHA [115]. The stress-regulated protein p8 was upregulated upon DHA treatment, and this effect was reversed by siRNA leading to increased cell death [116]. Whether therapeutic approaches based on the combination of ARS-type drugs plus siRNA for specific target genes can be translated from the bench to the bedside will largely depend on the bioavailability of siRNA drugs and whether concentrations high enough to exert therapeutic effects in vivo can be reached. Nanotechnology might provide solutions to this problem in the future [117].
10 Hepatoxicity and drug interactions ARS is a natural product and is generally considered as safe and well tolerated. However, there is no rational reason to assume that natural products per se act in a gentle manner. Secondary metabolites from plants serve as chemical weapons to deter microbial attacks by viruses, bacteria and protozoans as well as to provide protection from being eaten by herbivores (insects and mammals including human beings). Therefore, some plant-based drugs or preparations might be toxic and exert side effects. 12
Recently, we published two cases, where the combination of ART with other medications led to unwanted adverse events. A glioblastoma multiforme patient treated with a combination of temozolomide, ART and Chinese herbs (Coptis chinensis, Siegesbeckia orientalis,
Artemisia
scoparia,
Dictamnus
dasycarpus)
suffered
from
reversible
hepatotoxicity [118]. While all drugs alone may bear a minor risk for hepatotoxicity, this specific combination increased liver enzyme activities in this patient. In the present case, there was no obvious reason to expect such toxicity at first sight. Artesunate is an approved antimalarial drug, whose good tolerability is well known [119]. In a large meta-analysis with 8000 patients, hepatotoxicity occurred in 0.9% of the patients [120]. The Chinese herbs used (Coptis chinensis, Siegesbeckia orientalis, Artemisia scoparia, Dictamnus dasycarpus) are also generally considered as safe and clinical toxicity is very rare. Temozolomide is an approved anti-cancer drug routinely used for glioblastoma therapy, although case reports of hepatotoxicity have been reported in the post-marketing phase [121]. The fact that hepatotoxicity occurred in this patient indicates that this specific combination was not well tolerated. A genetic predisposition of the patient (e.g. by single nucleotide polymorphisms in CYP genes) might be considered as explanation. Another glioblastoma patient was treated by an alternative practitioner with a combination of dichloroacetate and artesunate. The patient showed clinical and laboratory signs of severe liver damage and bone marrow toxicity (leukopenia, thrombocytopenia) and died a few days later [122]. The compassionate use of dichloroacetate/ART combination therapy outside of clinical trials cannot be recommended. These drastic examples illustrate that even if ART alone is considered to be well tolerated, non-approved combinations with medications from complementary and alternative medicine should be avoided. In general, the question arises, whether or not ARS-type drugs have to be considered as toxic. A recent report on human leukemia cells xenografted to nude mice described that single-dose ART (120 mg/kg) produced human equivalent exposures, but multiple doses daily administration required for in vivo efficacy were not tolerated [123]. Liver metabolism plays an important role for drug tolerability. Many compounds taken up with food or other sources cannot be directly excreted from the body, because they are too lipophilic. The liver is the main organ bearing the capability to convert lipophilic compounds into hydrophilic ones that can be excreted by urine and faeces (biotransformation). This process is divided into three phases. In phase I, non-polar compounds are oxidized to increase their polarity and, hence, water solubility. In phase 2, molecules are conjugated to other water soluble molecules so that they can be transported outward (phase 3). The most important 13
phase 1 enzymes in the liver are the cytochrome P450 monooxygenases (CYPs), which belong to a large gene family of 57 members in the human genome. CYPs metabolize exogenous xenobiotics, chemicals and therapeutic drugs as well as endogenous substrates. Many xenobiotics and drugs can increase CYP activity by enzyme induction or decrease their activity by enzyme inhibition. Altered CYP activities, may however, influence the biotransformation of other drugs. If different drugs are co-administered, drug-drug interactions may occur leading either to decreased drug activity or increased drug toxicity [124-127]. CYP isoenzymes frequently involved in drug metabolism are CYP3A4/5, 2D6, 2C8/9, 1A2, 2C19, 2E1, 2B6, and 2A6. ARS and its derivatives are also metabolized in the liver. They are mainly converted to the bioactive metabolite DHA (artenimol) after either parenteral or gastrointestinal administration [128]. The main isoenzymes participating in metabolization of ARS-type drugs are CYP2A6, 2B6, and 3A4 (Table 10). Potential drug-drug interactions were generated by the induction or inhibition of CYP 1A2, 2B6, 2C19, and 3A4 (Table 10, Figure 3). CYP induction or inhibition by ARS-type drugs are not only of theoretical interest. There is a number of publications reporting on the interaction of ARS and its derivatives with drugs used to treat diverse diseases. Examples are shown in Table 11 and include drugs to treat infectious diseases (e.g. the antifungal imidazole ketonazole and the macrolidic antibiotic troleandomycin), omeprazole, which is used for treating gastroesophageal reflux and peptic ulcer, and the anticonvulsants S-mephenytoin and carbamazepine [129-134]. Furthermore, some authors published drug-drug interactions with natural products, e.g. paraxanthine as metabolite of caffeine and theobromide and 8-methoxypsoralen [131,135,136]. This compound is used for the therapy of psoriasis, eczema, vitilago and cutaneous lymphoma together with UVA light (PUVA therapy = psoralen plus UVA). These examples illustrate that negative drug-drug interactions have to be taken into account, if ARS-type drugs are used in combination with other drugs.
11 Conclusions and perspectives There is no doubt that there are numerous clues that ARS-type drugs can be combined with other drugs to reach improved tumor cell killing in vivo and in vitro. In general, it can be stated that the underlying mechanisms for these additive or synergistic effects are the same that are also responsible for the cytotoxicity of ARS and its derivatives if applied as single drugs, i.e. DNA damage, angiogenesis inhibition, inhibition of specific signal transduction 14
pathways, inhibition of apoptosis and autophagy etc. [44]. The spectrum of drugs that can be combined with ARS-type drugs is remarkable broad and comprises not only synthetic or natural chemical entities (Figure 4), but also radiotherapy and macromolecules such as antibodies, recombinant proteins, or therapeutic nucleic acids. While the exact mechanisms for synergistic interactions between ARS-type drugs and other drugs are largely unknown as yet, the cellular targets of the artemisinin’s partner drugs are known in many cases (Figure 5). This information may provide a basis to start more detailed investigations, how the modes of action of ARS-type drugs and the partner drugs in combination therapy may cooperate to generate synergistic tumor cell killing. This opens avenues for more systematic investigations to unravel the most effective and save combination treatments. It is important not only to search for drug combinations with optimal tumor inhibitory activities, but also to combinations with no or negligible effects on normal tissues and organs. It has been excluded that some combinations may also lead to antagonistic effects. The fact that 8-methoxypsoralen inhibited biotransformation of ARS indicates that PUVA therapy is not suited to be combined with ARS-type drugs [131]. A clinical trial in advanced non-small cell lung cancer reported on efficacy and toxicity of the standard combination vinorelbine and cisplatin with or without intravenous ART injections (120 mg) for 8 days [14]. Each treatment group consisted of 60 patients. Statically significant improvements in short-term survival were measured. The disease control rate of the trial group (88.2%) was significantly higher than that of the control group (72.7%) and the time to progression of the ART-treated patients (24 weeks) was significantly longer than that of the control group (20 weeks). ART was well tolerated and increased toxicity was not observed. Nevertheless, other ART-based combinations may be less well tolerated, as discussed above in two case reports with severe hepatotoxicity [118,122]. Controlled, doubleblind, randomized clinical trials are required to test efficacy and tolerabolity of novel combinations between ARS-type drugs and other treatment modalities for cancer therapy.
Conflict of interest: There is no conflict of interest.
15
References [1] [2]
[3] [4]
[5] [6]
[7] [8]
[9]
[10]
[11]
[12] [13]
[14]
[15] [16] [17] [18] [19] [20]
Efferth T, Zacchino S, Georgiev MI, Liu L, Wagner H, Panossian A. Nobel Prize for artemisinin brings phytotherapy into the spotlight. Phytomedicine 2015;22(13):A1-3. Willcox ML, Burton S, Oyweka R, Namyalo R, Challand S, Lindsey K. Evaluation and pharmacovigilance of projects promoting cultivation and local use of Artemisia annua for malaria. Malar. J. 2011;10:84 Weathers PJ, Towler M, Hassanali A, Lutgen P, Engeu PO. Dried-leaf Artemisia annua: A practical malaria therapeutic for developing countries? World J. Pharmacol. 2014;3(4):39-55. Efferth T, Rücker G, Falkenberg M, Manns D, Olbrich A, Fabry U et al. Detection of apoptosis in KG-1a leukemic cells treated with investigational drugs. Arzneimittelforschung 1996;46:196-200. Efferth T, Romero MR, Wolf DG, Stamminger T, Marin JJ, Marschall M. The antiviral activities of artemisinin and artesunate. Clin. Infect. Dis. 2008;47:804-811 Efferth T, Romero MR, Bilia AR, Osman AG, ElSohly M, Wink M et al. Expanding the therapeutic spectrum of artemisinin: activity against infectious diseases beyond malaria and novel pharmaceutical developments. World J. Trad. Chin. Med. 2016;2(2):1-23. Ho WE, Peh HY, Chan TK, Wong WS. Artemisinins: pharmacological actions beyond antimalarial. Pharmacol. Ther. 2014;142:126-139 Seo EJ, Wiench B, Hamm R, Paulsen M, Zu Y, Fu Y, Efferth T. Cytotoxicity of natural products and derivatives toward MCF-7 cell monolayers and cancer stem-like mammospheres. Phytomedicine 2015;22(4):438-43. Ooko E, Saeed ME, Kadioglu O, Sarvi S, Colak M, Elmasaoudi K et al. Artemisinin derivatives induce iron-dependent cell death (ferroptosis) in tumor cells. Phytomedicine 2015;22(11):1045-54. Jiang W, Cen Y, Song Y, Li P, Qin R, Liu C et al. Artesunate attenuated progression of atherosclerosis lesion formation alone or combined with rosuvastatin through inhibition of pro-inflammatory cytokines and pro-inflammatory chemokines. Phytomedicine 2016;23(11):1259-66. Li J, Casteels T, Frogne T, Ingvorsen C, Honoré C, Courtney M , Huber KV, Schmitner N et al. Artemisinins Target GABAA Receptor Signaling and Impair α Cell Identity. Cell 2017;168(12):86-100.e15. Saeed ME, Krishna S, Greten HJ, Kremsner PG, Efferth T. Antischistosomal activity of artemisinin derivatives in vivo and in patients. Pharmacol. Res. 2016;110:216-26. Berger TG, Dieckmann D, Efferth T, Schultz ES, Funk JO, Baur A et al. Artesunate in the treatment of metastatic uveal melanoma--first experiences. Oncol Rep. 2005 Dec;14(6):1599603. Zhang ZY, Yu SQ, Miao LY, Huang XY, Zhang XP, Zhu YP et al.[Artesunate combined with vinorelbine plus cisplatin in treatment of advanced non-small cell lung cancer: a randomized controlled trial]. [Article in Chinese] Zhong Xi Yi Jie He Xue Bao 2008;6(2):134-8. Michaelsen FW, Saeed ME, Schwarzkopf J, Efferth T. Activity of Artemisia annua and artemisinin derivatives, in prostate carcinoma. Phytomedicine 2015;22(14):1223-31. Wiemann MC, Calabresi P. Principles of current cancer chemotherapy. Compr. Ther. 1983;9(3):46-52. Dawson JW, Taylor I. Principles of adjuvant therapy. Br. J. Hosp. Med. 1995;54(6):249-54. Pritchard JR, Lauffenburger DA, Hemann MT. Understanding resistance to combination chemotherapy. Drug Resist. Updat. 2012;15(5-6):249-57. Chou TC. Frequently asked questions in drug combinations and the mass-action law-based answers. Synergy 2014;1(1):3-21. Fu J, Zhang N, Chou JH, Dong HJ, Lin SF, Ulrich-Merzenich GS et al. Drug combination in vivo using combination index method: Taxotere and T607 against colon carcinoma HCT-116 xenograft tumor in nude mice. Synergy 2016; 3(3):15–30. 16
[21]
[37]
Han SY, Zhao W, Sun H, Zhou N, Zhou F, An G et al. Marsdenia tenacissima extract enhances gefitinib efficacy in non-small cell lung cancer xenografts. Phytomedicine 2015;22(5):560-7. Hsin IL, Wang SC, Li JR, Ciou TC, Wu CH, Wu HM et al. Immunomodulatory proteins FIP-gts and chloroquine induce caspase-independent cell death via autophagy for resensitizing cisplatin-resistant urothelial cancer cells. Phytomedicine 2016;23(13):1566-1573. Rai G, Mishra S, Suman S, Shukla Y. Resveratrol improves the anticancer effects of doxorubicin in vitro and in vivo models: A mechanistic insight. Phytomedicine. 2016;23(3):233-42. Shrestha S, Song YW, Kim H, Lee DS, Cho SK. Sageone, a diterpene from Rosmarinus officinalis, synergizes with cisplatin cytotoxicity in SNU-1 human gastric cancer cells. Phytomedicine 2016;23(13):1671-1679. Tang Q, Ji F, Sun W, Wang J, Guo J, Guo L et al. Combination of baicalein and 10-hydroxy camptothecin exerts remarkable synergetic anti-cancer effects. Phytomedicine 2016;23(14):1778-1786. Efferth T, Koch E. Complex interactions between phytochemicals. The multi-target therapeutic concept of phytotherapy. Curr Drug Targets. 2011;12(1):122-32. Efferth T, Grassmann R. Impact of viral oncogenesis on responses to anti-cancer drugs and irradiation. Crit Rev Oncog. 2000;11(2):165-87. Efferth T. From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin Cancer Biol. 2017 [Epub ahead of print] Efferth T, Sauerbrey A, Olbrich A, Gebhart E, Rauch P, Weber HO et al. Molecular modes of action of artesunate in tumor cell lines. Mol. Pharmacol. 2003;64:382-394. Li PC, Lam E, Roos WP, Zdzienicka MZ, Kaina B, Efferth T. Artesunate derived from traditional Chinese medicine induces DNA damage and repair. Cancer Res. 2008;68:4347-4351. Berdelle N, Nikolova T, Quiros S, Efferth T, Kaina B. Artesunate induces oxidative DNA damage, sustained DNA double-strand breaks, and the ATM/ATR damage response in cancer cells. Mol. Cancer Ther. 2011;10:2224-2233. Alcantara DD, Ribeiro HF, Cardoso PC, Araújo TM, Burbano RR, Guimarães AC et al. In vitro evaluation of the cytotoxic and genotoxic effects of artemether, an antimalarial drug, in a gastric cancer cell line (PG100). J. Appl. Toxicol. 2013;33:151-156. Aquino I, Tsuboy MS, Marcarini JC, Mantovani MS, Perazzo FF, Maistro EL. Genotoxic evaluation of the antimalarial drugs artemisinin and artesunate in human HepG2 cells and effects on CASP3 and SOD1 gene expressions. Genet Mol. Res. 2013;12:2517-2527. Park J, Lai HC, Sasaki T, Singh NP. DNA damage in dihydroartemisinin-resistant Molt-4 cells. Anticancer Res. 2015;35:1339-1343. Wang B, Hou D, Liu Q, Wu T, Guo H, Zhang X et al.Artesunate sensitizes ovarian cancer cells to cisplatin by downregulating RAD51.Cancer Biol. Ther. 2015;16(10):1548-56. Efferth T. The human ATP-binding cassette transporter genes: from the bench to the bedside. Curr. Mol. Med. 2001;1(1):45-65. Gillet JP, Efferth T, Remacle J. Chemotherapy-induced resistance by ATP-binding cassette
[38]
transporter genes. Biochim. Biophys. Acta 2007;1775(2):237-62. Kinsella AR, Smith D, Pickard M. Resistance to chemotherapeutic antimetabolites: a function
[22]
[23]
[24]
[25]
[26] [27] [28] [29] [30] [31]
[32]
[33]
[34] [35] [36]
of salvage pathway involvement and cellular response to DNA damage. Br. 1997;75(7):935-45. [39] [40]
Zhao R, Goldman ID. Resistance to antifolates. Oncogene 2003;22(47):7431-57. Lamontanara AJ, Gencer EB, Kuzyk O, Hantschel O. Mechanisms of resistance to BCR-ABL and other kinase inhibitors. Biochim.
[41]
J. Cancer
Biophys. Acta 2013;1834(7):1449-59. Amable L. Cisplatin resistance and opportunities for precision medicine. Pharmacol. Res. 2016;106:27-36. 17
[42]
[43]
[44] [45]
[46]
[47]
[48]
[49] [50]
[51] [52]
[53]
[54] [55]
[56] [57]
[58] [59] [60]
Pluchino KM, Hall MD, Goldsborough AS, Callaghan R, Gottesman MM. Collateral sensitivity as a strategy against cancer multidrug resistance. Drug Resist. Updat. 2012;15(12):98-105. Saeed M, Greten J, Efferth T. Collateral sensitivity in drug-resistant tumor cells. In: Bonavida B (Ed) Tumor cell resistance to apoptosis by cytotoxic drugs and reversal of resistance by targeted sensitizing cells. Springer, New York, 2013, pp.187-211. Efferth T. Artemisinin – second career as anticancer drug? World J Trad Chin Med 2015;1(4):2-25. Soomro S, Langenberg T, Mahringer A, Konkimalla VB, Horwedel C, Holenya P et al. Design of novel artemisinin-like derivatives with cytotoxic and anti-angiogenic properties. J. Cell. Mol. Med. 2011;15(5):1122-35. Kim BH, Kim HJ, Wu HG, Kang CH, Kim YT, Lee SH et al. Role of postoperative radiotherapy after curative resection and adjuvant chemotherapy for patients with pathological stage N2 non-small-cell lung cancer: a propensity score matching analysis. Clin Lung Cancer. 2014 Sep;15(5):356-64. Ward MC, Ross RB, Koyfman SA, Lorenz R, Lamarre ED, Scharpf J et al. Modern Image-Guided Intensity-Modulated Radiotherapy for Oropharynx Cancer and Severe Late Toxic Effects: Implications for Clinical Trial Design. JAMA Otolaryngol Head Neck Surg. 2016 Dec 1;142(12):1164-1170. White R, Dinneen T, Makris A. Local radiotherapy alone following neoadjuvant chemotherapy and surgery in combined clinical stage II and III breast cancer. Radiat Oncol. 2016 Jul 26;11:93. Skvortsova I, Debbage P, Kumar V, Skvortsov S. Radiation resistance: Cancer stem cells (CSCs) and their enigmatic pro-survival signaling. Semin Cancer Biol. 2015 Dec;35:39-44. Malik A, Sultana M, Qazi A, Qazi MH, Parveen G, Waquar S et al. Role of Natural radiosensitizers and cancer cell radioresistance: An update. Anal Cell Pathol (Amst). 2016;2016:6146595. Kim SJ, Kim MS, Lee JW, Lee CH, Yoo H, Shin SH et al. Dihydroartemisinin enhances radiosensitivity of human glioma cells in vitro. J. Cancer Res. Clin. Oncol. 2006;132(2):129-35. Zhao Y, Jiang W, Li B, Yao Q, Dong J, Cen Y et al. Artesunate enhances radiosensitivity of human non-small cell lung cancer A549 cells via increasing NO production to induce cell cycle arrest at G2/M phase. Int. Immunopharmacol. 2011;11(12):2039-46. Reichert S, Reinboldt V, Hehlgans S, Efferth T, Rödel C, Rödel F. A radiosensitizing effect of artesunate in glioblastoma cells is associated with a diminished expression of the inhibitor of apoptosis protein survivin. Radiother. Oncol. 2012;103(3):394-401. Luo J, Chen X, Chen G, Zhou X, Lu X, Ling Y et al. Dihydroartemisinin induces radiosensitivity in cervical cancer cells by modulating cell cycle progression. Saudi Med. J. 2013;34(3):254-60. Zuo ZJ, Wang ST, Jiang LX, Xin YX, Li W, Xu ZH et al. [Effect of dihydroartemisinin combined irradiation on the apoptosis of human lung cancer GLC-82 cells and its mechanism study]. [Article in Chinese] Zhongguo Zhong Xi Yi Jie He Za Zhi 2014;34(10):1220-4. Chilakamarthi U, Giribabu L. Photodynamic therapy: Past, present and future. Chem. Rec. 2017 [Epub ahead of print]. Li YJ, Zhou JH, Du XX, Jia de X, Wu CL, Huang P et al. Dihydroartemisinin accentuates the antitumor effects of photodynamic therapy via inactivation of NF-κB in Eca109 and Ec9706 esophageal cancer cells. Cell. Physiol. Biochem. 2014;33(5):1527-36. Grabowsky JA. Drug interactions and the pharmacist: focus on everolimus. Ann. Pharmacother. 2013;47(7-8):1055-63 Koene RJ, Prizment AE, Blaes A, Konety SH. Shared risk factors in cardiovascular disease and cancer. Circulation 2016;133(11):1104-14. Kowalski J, Herman ZS. Captopril augments antitumor activity of cyclophosphamide in mice.Pol. J. Pharmacol. 1996;48(3):281-5. 18
[61]
[62] [63]
[64] [65]
[66] [67]
[68] [69] [70] [71] [72]
[73] [74]
[75]
[76]
[77] [78]
[79] [80]
Wilop S, von Hobe S, Crysandt M, Esser A, Osieka R, Jost E. Impact of angiotensin I converting enzyme inhibitors and angiotensin II type 1 receptor blockers on survival in patients with advanced non-small-cell lung cancer undergoing first-line platinum-based chemotherapy. J. Cancer Res. Clin. Oncol. 2009;135(10):1429-35 Krusche B, Arend J, Efferth T. Synergistic inhibition of angiogenesis by artesunate and captopril in vitro and in vivo. Evid. Based Complement. Alternat. Med. 2013;2013:454783. Christian JB, Lapane KL, Hume AL, Eaton CB, Weinstock MA; VATTC Trial. Association of ACE inhibitors and angiotensin receptor blockers with keratinocyte cancer prevention in the randomized VATTC trial. J. Natl. Cancer. Inst. 2008;100(17):1223-32. Pascolo S. Time to use a dose of chloroquine as an adjuvant to anti-cancer chemotherapies. Eur. J. Pharmacol. 2016;771:139-44. Ganguli A, Choudhury D, Datta S, Bhattacharya S, Chakrabarti G. Inhibition of autophagy by chloroquine potentiates synergistically anti-cancer property of artemisinin by promoting ROS dependent apoptosis. Biochimie 2014;107 Pt B:338-49. Gøtzsche PC, Johansen HK. Routine versus selective antifungal administration for control of fungal infections in patients with cancer. Cochrane Database Syst. Rev. 2014;(9):CD000026. Shahbazfar AA, Zare P, Ranjbaran M, Tayefi-Nasrabadi H, Fakhri O, Farshi Y et al. A survey on anticancer effects of artemisinin, iron, miconazole, and butyric acid on 5637 (bladder cancer) and 4T1 (Breast cancer) cell lines. J. Cancer Res. Ther. 2014;10(4):1057-62. Moscow JA, Dixon KH. Glutathione-related enzymes, glutathione and multidrug resistance. Cytotechnology 1993;12(1-3):155-70. Duchesne GM. Fundamental bases of combined therapy in lung cancer: cell resistance to chemotherapy and radiotherapy. Lung Cancer 1994;10 Suppl 1:S67-72. Efferth T, Volm M. Glutathione-related enzymes contribute to resistance of tumor cells and low toxicity in normal organs to artesunate. In Vivo 2005;19(1):225-32. Zhang C, Zhang Z, Zhu Y, Qin S. Glucose-6-phosphate dehydrogenase: a biomarker and potential therapeutic target for cancer. Anticancer Agents Med Chem. 2014 Feb;14(2):280-9. Wang X, Wu G, Cao G, Yang L, Xu H, Huang J et al. Zoledronic acid inhibits the pentose phosphate pathway through attenuating the Ras-TAp73-G6PD axis in bladder cancer cells. Mol Med Rep. 2015 Sep;12(3):4620-5. Xu SN, Wang TS, Li X, Wang YP. SIRT2 activates G6PD to enhance NADPH production and promote leukaemia cell proliferation. Sci Rep. 2016 Sep 2;6:32734. Polimeni M, Voena C, Kopecka J, Riganti C, Pescarmona G, Bosia A ewt al. Modulation of doxorubicin resistance by the glucose-6-phosphate dehydrogenase activity.Biochem J. 2011 Oct 1;439(1):141-9. doi: 10.1042/BJ20102016. Efferth T, Fabry U, Glatte P, Osieka R. Increased induction of apoptosis in mononuclear cells of a glucose-6-phosphate dehydrogenase deficient patient. J. Mol. Med. (Berl). 1995;73(1):47-9. Efferth T, Fabry U, Osieka R. DNA damage and apoptosis in mononuclear cells from glucose6-phosphate dehydrogenase-deficient patients (G6PD Aachen variant) after UV irradiation. J. Leukoc. Biol. 2001;69(3):340-2. Efferth T, Schwarzl SM, Smith J, Osieka R. Role of glucose-6-phosphate dehydrogenase for oxidative stress and apoptosis. Cell Death Differ. 2006;13(3):527-8 Elf S, Lin R, Xia S, Pan Y, Shan C, Wu S et al. Targeting 6-phosphogluconate dehydrogenase in the oxidative PPP sensitizes leukemia cells to antimalarial agent dihydroartemisinin. Oncogene 2017;36(2):254-262. Qie S, Chu C, Li W, Wang C, Sang N. ErbB2 activation upregulates glutaminase 1 expression which promotes breast cancer cell proliferation. J. Cell. Biochem. 2014;115(3):498-509. Fu A, Yu Z, Song Y, Zhang E. Silencing of glutaminase 1 resensitizes Taxol-resistant breast cancer cells to Taxol. Mol. Med. Rep. 2015;11(6):4727-33.
19
[81]
[82]
[83] [84]
[85] [86]
[87]
[88]
[89] [90]
[91] [92] [93]
[94]
[95]
[96]
[97] [98]
[99]
Yuan L, Sheng X, Clark LH, Zhang L, Guo H, Jones HM et al. Glutaminase inhibitor compound 968 inhibits cell proliferation and sensitizes paclitaxel in ovarian cancer. Am. J. Transl. Res. 2016;8(10):4265-4277. Wang D, Meng G, Zheng M, Zhang Y, Chen A, Wu J et al. The Glutaminase-1 inhibitor 968 enhances dihydroartemisinin-mediated antitumor efficacy in hepatocellular carcinoma cells. PLoS One 2016;11(11):e0166423. Qin HT, Li HQ, Liu F. Selective histone deacetylase small molecule inhibitors: recent progress and perspectives. Expert Opin. Ther. Pat. 2016 [Epub ahead of print] Zahnow CA, Topper M, Stone M, Murray-Stewart T, Li H, Baylin SB et al. Inhibitors of DNA methylation, histone deacetylation, and histone demethylation: A perfect combination for cancer therapy. Adv. Cancer Res. 2016;130:55-111. Flis S, Gnyszka A, Flis K, Spławiński J. MS275 enhances cytotoxicity induced by 5-fluorouracil in the colorectal cancer cells. Eur. J. Pharmacol. 2010;627(1-3):26-32 Budman DR, Tai J, Calabro A, John V. The histone deacetylase inhibitor panobinostat demonstrates marked synergy with conventional chemotherapeutic agents in human ovarian cancer cell lines. Invest. New Drugs. 2011;29(6):1224-9. Colarossi L, Memeo L, Colarossi C, Aiello E, Iuppa A, Espina V et al. Inhibition of histone deacetylase 4 increases cytotoxicity of docetaxel in gastric cancer cells. Proteomics Clin. Appl. 2014;8(11-12):924-31. Zhang CZ, Pan Y, Cao Y, Lai PB, Liu L, Chen GG et al. Histone deacetylase inhibitors facilitate dihydroartemisinin-induced apoptosis in liver cancer in vitro and in vivo. PLoS One 2012;7(6):e39870. Floyd RA, Chandru HK, He T, Towner R. Anti-cancer activity of nitrones and observations on mechanism of action. Anticancer Agents Med. Chem. 2011;11(4):373-9. Chan HW, Singh NP, Lai HC. Cytotoxicity of dihydroartemisinin toward Molt-4 cells attenuated by N-tert-butyl-alpha-phenylnitrone and deferoxamine. Anticancer Res. 2013;33(10):4389-93. 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):311-35 Newman DJ, Cragg GM. Natural products as sources of new drugs from 1981 to 2014. J. Nat. Prod. 2016;79(3):629-61. Ferreira JF, Luthria DL, Sasaki T, Heyerick A. Flavonoids from Artemisia annua L. as antioxidants and their potential synergism with artemisinin against malaria and cancer. Molecules 2010;15(5):3135-70. Efferth T, Herrmann F, Tahrani A, Wink M. Cytotoxic activity of secondary metabolites derived from Artemisia annua L. towards cancer cells in comparison to its designated active constituent artemisinin. Phytomedicine 2011;18(11):959-69. Sui P, Cao H, Meng L, Hu P, Ma H, Du J. The synergistic effect of humanized monoclonal antibodies targeting insulin-like growth factor 1 receptor (IGF-1R) and chemotherapy. Curr. Drug Targets 2014;15(7):674-80. Sheng J, Yang YP, Zhao YY, Qin T, Hu ZH, Zhou T et al. The efficacy of combining EGFR monoclonal antibody with chemotherapy for patients with advanced nonsmall cell lung cancer: A meta-analysis from 9 randomized controlled trials. Medicine (Baltimore) 2015;94(34):e1400. He C, Tang Z, Tian H, Chen X. Co-delivery of chemotherapeutics and proteins for synergistic therapy. Adv. Drug Deliv. Rev. 2016;98:64-76. Zhang T, Herlyn D. Combination of active specific immunotherapy or adoptive antibody or lymphocyte immunotherapy with chemotherapy in the treatment of cancer. Cancer Immunol. Immunother. 2009;58(4):475-92. Michelle Xu M, Pu Y, Weichselbaum RR, Fu YX. Integrating conventional and antibody-based targeted anticancer treatment into immunotherapy. Oncogene 2016 [Epub ahead of print]
20
[100] Kubuschok B, Held G, Pfreundschuh M. Management of diffuse large B-cell lymphoma (DLBCL). Cancer Treat. Res. 2015;165:271-88. [101] Bonavida B. Rituximab-induced inhibition of antiapoptotic cell survival pathways: implications in chemo/immunoresistance, rituximab unresponsiveness, prognostic and novel therapeutic interventions. Oncogene 2007;26(25):3629-36. [102] Bonavida B. Postulated mechanisms of resistance of B-cell non-Hodgkin lymphoma to rituximab treatment regimens: strategies to overcome resistance. Semin. Oncol. 2014;41(5):667-77. [103] Sieber S, Gdynia G, Roth W, Bonavida B, Efferth T. Combination treatment of malignant B cells using the anti-CD20 antibody rituximab and the anti-malarial artesunate. Int. J. Oncol. 2009;35(1):149-58. [104] Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT et al. Broad targeting of resistance to apoptosis in cancer. Semin. Cancer Biol. 2015;35 Suppl:S78-103. [105] de Miguel D, Lemke J, Anel A, Walczak H, Martinez-Lostao L. Onto better TRAILs for cancer treatment. Cell Death Differ. 2016;23(5):733-47. [106] Lim B, Allen JE, Prabhu VV, Talekar MK, Finnberg NK, El-Deiry WS. Targeting TRAIL in the treatment of cancer: new developments. Expert Opin. Ther. Targets 2015;19(9):1171-85. [107] Ahmed F, Ishibashi M. Bio-active Natural Products with TRAIL-Resistance Overcoming Activity. Chem. Pharm. Bull. (Tokyo) 2016;64(2):119-27. [108] He Q, Shi J, Shen XL, An J, Sun H, Wang L et al. Dihydroartemisinin upregulates death receptor 5 expression and cooperates with TRAIL to induce apoptosis in human prostate cancer cells. Cancer Biol. Ther. 2010;9(10):819-24. [109] Thanaketpaisarn O, Waiwut P, Sakurai H, Saiki I. Artesunate enhances TRAIL-induced apoptosis in human cervical carcinoma cells through inhibition of the NF-κB and PI3K/Akt signaling pathways. Int. J. Oncol. 2011;39(1):279-85. [110] Kong R, Jia G, Cheng ZX, Wang YW, Mu M, Wang SJ et al. Dihydroartemisinin enhances Apo2L/TRAIL-mediated apoptosis in pancreatic cancer cells via ROS-mediated up-regulation of death receptor 5. PLoS One 2012;7(5):e37222. Erratum in: PLoS One 2012;7(10). [111] Prados J, Alvarez PJ, Melguizo C, Rodriguez-Serrano F, Carrillo E, Boulaiz H, Vélez C, Marchal JA, Caba O, Ortiz R, Rama A, Aranega A. How is gene transfection able to improve current chemotherapy? The role of combined therapy in cancer treatment. Curr. Med. Chem. 2012;19(12):1870-88. [112] Wang YB, Hu Y, Li Z, Wang P, Xue YX, Yao YL et al. Artemether combined with shRNA interference of vascular cell adhesion molecule-1 significantly inhibited the malignant biological behavior of human glioma cells. PLoS One 2013;8(4):e60834. [113] Wu J, Hu D, Yang G, Zhou J, Yang C, Gao Y et al. Down-regulation of BMI-1 cooperates with artemisinin on growth inhibition of nasopharyngeal carcinoma cells. J. Cell. Biochem. 2011;112(7):1938-48. [114] Han P, Luan Y, Liu Y, Yu Z, Li J, Sun Z et al. Small interfering RNA targeting Rac1 sensitizes colon cancer to dihydroartemisinin-induced cell cycle arrest and inhibited cell migration by suppressing NFκB activity. Mol. Cell. Biochem 2013;379(1-2):171-80. [115] Zhu H, Liao SD, Shi JJ, Chang LL, Tong YG, Cao J et al. DJ-1 mediates the resistance of cancer cells to dihydroartemisinin through reactive oxygen species removal. Free Radic. Biol. Med. 2014;71:121-32. [116] Chen SS, Hu W, Wang Z, Lou XE, Zhou HJ. p8 attenuates the apoptosis induced by dihydroartemisinin in cancer cells through promoting autophagy. Cancer Biol. Ther. 2015;16(5):770-9. [117] Chitkara D, Singh S, Mittal A. Nanocarrier-based co-delivery of small molecules and siRNA/miRNA for treatment of cancer. Ther. Deliv. 2016;7(4):245-55. [118] 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
21
[119] [120] [121] [122]
[123]
[124] [125]
[126] [127] [128] [129]
[130] [131]
[132]
[133]
[134] [135]
[136]
[137]
multiforme patient: case report review of the literature. Arch. Toxicol. 2016 [Epub ahead of print] Efferth T, Kaina B. Toxicity of the antimalarial artemisinin and its dervatives. Crit Rev Toxicol 2010;40:405-21. Ribeiro IR, Olliaro P. Safety of artemisinin and its derivatives. A review of published and unpublished clinical trials. Med. Trop. (Mars.) 1998;58:50-3. Dixit S, Hingorani M, Afzal P, Campbell AP. Temozolomide induced liver injury. Acta Neurol. Belg. 2011;111:249-51. 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 lLiterature. Front. Oncol. 2016;6:204. Drenberg CD, Buaboonnam J, Orwick SJ, Hu S, Li L, Fan Y et al. Evaluation of artemisinins for the treatment of acute myeloid leukemia. Cancer Chemother. Pharmacol. 2016;77(6):123143. Ogu CC Maxa JL. Drug interactions due to cytochrome P450. Proc. (Bayl. Univ. Med. Cent.) 2000; 13(4): 421–423. Goey AK, Mooiman KD, Beijnen JH, Schellens JH, Meijerman I. Relevance of in vitro and clinical data for predicting CYP3A4-mediated herb-drug interactions in cancer patients. Cancer Treat. Rev. 2013;39(7):773-83 Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of cytochrome P450 2C8 in drug metabolism and interactions. Pharmacol. Rev. 2016;68(1):168-241. Hedrich WD, Hassan HE, Wang H. Insights into CYP2B6-mediated drug-drug interactions. Acta Pharm. Sin. B 2016;6(5):413-425. Navaratnam V, Mansor SM, Sit NW, Grace J, Li Q, Olliaro P. Pharmacokinetics of artemisinintype compounds. Clin. Pharmacokinet. 2000;39(4):255-70. Grace JM, Aguilar AJ, Trotman KM, Peggins JO, Brewer TG. Metabolism of beta-arteether to dihydroqinghaosu by human liver microsomes and recombinant cytochrome P450. Drug Metab. Dispos. 1998;26(4):313-7. Erratum in: Drug Metab. Dispos 1998 Jul;26(7):704. Svensson US, Ashton M. Identification of the human cytochrome P450 enzymes involved in the in vitro metabolism of artemisinin. Br. J. Clin. Pharmacol. 1999;48(4):528-35. Mihara K, Svensson US, Tybring G, Hai TN, Bertilsson L, Ashton M. Stereospecific analysis of omeprazole supports artemisinin as a potent inducer of CYP2C19. Fundam. Clin. Pharmacol. 1999;13(6):671-5. Simonsson US, Jansson B, Hai TN, Huong DX, Tybring G, Ashton M. Artemisinin autoinduction is caused by involvement of cytochrome P450 2B6 but not 2C9. Clin. Pharmacol. Ther. 2003;74(1):32-43. Elsherbiny DA, Asimus SA, Karlsson MO, Ashton M, Simonsson US. A model based assessment of the CYP2B6 and CYP2C19 inductive properties by artemisinin antimalarials: implications for combination regimens. J. Pharmacokinet. Pharmacodyn. 2008;35(2):203-17. Sukhija M, Medhi B, Pandhi P. Effects of artemisinin, artemether, arteether on the pharmacokinetics of carbamazepine. Pharmacology 2006;76(3):110-6. Bapiro TE, Sayi J, Hasler JA, Jande M, Rimoy G, Masselle A et al. Artemisinin and thiabendazole are potent inhibitors of cytochrome P450 1A2 (CYP1A2) activity in humans. Eur. J. Clin. Pharmacol. 2005;61(10):755-61. Asimus S, Elsherbiny D, Hai TN, Jansson B, Huong NV, Petzold MG eet al. Artemisinin antimalarials moderately affect cytochrome P450 enzyme activity in healthy subjects. Fundam. Clin. Pharmacol. 2007;21(3):307-16. Wu GS, Lu JJ, Guo JJ, Huang MQ, Gan L, Chen XP et al. Synergistic anti-cancer activity of the combination of dihydroartemisinin and doxorubicin in breast cancer cells. Pharmacol. Rep. 2013;65(2):453-9.
22
[138] Tai X, Cai XB, Zhang Z, Wei R. In vitro and in vivo inhibition of tumor cell viability by combined dihydroartemisinin and doxorubicin treatment, and the underlying mechanism. Oncol. Lett. 2016;12(5):3701-3706. [139] Papanikolaou X, Johnson S, Garg T, Tian E, Tytarenko R, Zhang Q et al. Artesunate overcomes drug resistance in multiple myeloma by inducing mitochondrial stress and non-caspase apoptosis. Oncotarget 2014;5(12):4118-28. [140] Zhang JL, Wang Z, Hu W, Chen SS, Lou XE, Zhou HJ. DHA regulates angiogenesis and improves the efficiency of CDDP for the treatment of lung carcinoma. Microvasc Res. 2013;87:14-24. [141] Feng X, Li L, Jiang H, Jiang K, Jin Y, Zheng J. Dihydroartemisinin potentiates the anticancer effect of cisplatin via mTOR inhibition in cisplatin-resistant ovarian cancer cells: involvement of apoptosis and autophagy. Biochem. Biophys. Res. Commun. 2014;444(3):376-81. [142] Chen T, Li M, Zhang R, Wang H. Dihydroartemisinin induces apoptosis and sensitizes human ovarian cancer cells to carboplatin therapy. J. Cell. Mol. Med. 2009;13(7):1358-70. [143] Gravett AM, Liu WM, Krishna S, Chan WC, Haynes RK, Wilson NL et al. In vitro study of the anti-cancer effects of artemisone alone or in combination with other chemotherapeutic agents. Cancer Chemother. Pharmacol. 2011;67(3):569-77. [144] Huang XJ, Li CT, Zhang WP, Lu YB, Fang SH, Wei EQ. Dihydroartemisinin potentiates the cytotoxic effect of temozolomide in rat C6 glioma cells. Pharmacology 2008;82(1):1-9. [145] Karpel-Massler G, Westhoff MA, Kast RE, Dwucet A, Nonnenmacher L, Wirtz CR et al. Artesunate enhances the antiproliferative effect of temozolomide on U87MG and A172 glioblastoma cell lines. Anticancer Agents Med. Chem. 2014;14(2):313-8. [146] Zhang ZS, Wang J, Shen YB, Guo CC, Sai KE, Chen FR, Mei X, Han FU, Chen ZP. Dihydroartemisinin increases temozolomide efficacy in glioma cells by inducing autophagy. Oncol. Lett. 2015;10(1):379-383. [147] Berte N, Lokan S, Eich M, Kim E, Kaina B. Artesunate enhances the therapeutic response of glioma cells to temozolomide by inhibition of homologous recombination and senescence. Oncotarget 2016 [Epub ahead of print] [148] Hou J, Wang D, Zhang R, Wang H. Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin. Cancer Res. 2008;14(17):5519-30. [149] Wang SJ, Sun B, Pan SH, Chen H, Kong R, Li J et al. [Experimental study of the function and mechanism combining dihydroartemisinin and gemcitabine in treating pancreatic cancer]. [Article in Chinese] Zhonghua Wai Ke Za Zhi 2010a;48(7):530-4. [150] Wang SJ, Gao Y, Chen H, Kong R, Jiang HC, Pan SH et al. Dihydroartemisinin inactivates NFkappaB and potentiates the anti-tumor effect of gemcitabine on pancreatic cancer both in vitro and in vivo. Cancer Lett. 2010b;293(1):99-108. [151] Zhao C, Gao W, Chen T. Synergistic induction of apoptosis in A549 cells by dihydroartemisinin and gemcitabine. Apoptosis 2014;19(4):668-81. [152] Efferth T, Ramirez T, Gebhart E, Halatsch ME. Combination treatment of glioblastoma multiforme cell lines with the anti-malarial artesunate and the epidermal growth factor receptor tyrosine kinase inhibitor OSI-774. Biochem. Pharmacol. 2004;67(9):1689-700. [153] Liu WM, Gravett AM, Dalgleish AG. The antimalarial agent artesunate possesses anticancer properties that can be enhanced by combination strategies. Int. J. Cancer 2011;128(6):147180. [154] Gerhardt T, Jones R, Park J, Lu R, Chan HW, Fang Q et al.. Effects of antioxidants and prooxidants on cytotoxicity of dihydroartemisinin to Molt-4 human leukemia cells. Anticancer Res. 2015;35(4):1867-71. [155] Efferth T, Dunstan H, Sauerbrey A, Miyachi H, Chitambar CR. The anti-malarial artesunate is also active against cancer. Int. J. Oncol. 2001;18(4):767-73. [156] Reungpatthanaphong P, Mankhetkorn S. Modulation of multidrug resistance by artemisinin, artesunate and dihydroartemisinin in K562/adr and GLC4/adr resistant cell lines. Biol. Pharm. Bull. 2002;25(12):1555-61. 23
[157] Efferth T, Giaisi M, Merling A, Krammer PH, Li-Weber M. Artesunate induces ROS-mediated apoptosis in doxorubicin-resistant T leukemia cells.PLoS One 2007;2(8):e693. [158] Li X, Ling V, Li PC. Same-single-cell analysis for the study of drug efflux modulation of multidrug resistant cells using a microfluidic chip. Anal. Chem. 2008;80(11):4095-102. [159] Riganti C, Doublier S, Costamagna C, Aldieri E, Pescarmona G, Ghigo D et al. Activation of nuclear factor-kappa B pathway by simvastatin and RhoA silencing increases doxorubicin cytotoxicity in human colon cancer HT29 cells. Mol. Pharmacol. 2008;74(2):476-84. [160] Riganti C, Doublier S, Viarisio D, Miraglia E, Pescarmona G, Ghigo D et al. Artemisinin induces doxorubicin resistance in human colon cancer cells via calcium-dependent activation of HIF1alpha and P-glycoprotein overexpression. Br. J. Pharmacol. 2009;156(7):1054-66. [161] Zhong H, Zhao X, Zuo Z, Sun J, Yao Y, Wang T et al. Combating P-glycoprotein-mediated multidrug resistance with 10-O-phenyl dihydroartemisinin ethers in MCF-7 cells. Eur. J. Med. Chem. 2016;108:720-9. [162] Reiter C, Herrmann A, Çapci A, Efferth T, Tsogoeva SB. New artesunic acid homodimers: potent reversal agents of multidrug resistance in leukemia cells. Bioorg. Med. Chem. 2012;20(18):5637-41. [163] Zhang L, Chen F, Zhang Z, Chen Y, Wang J. Synthesis and biological evaluation of a novel artesunate-podophyllotoxin conjugate as anticancer agent. Bioorg. Med. Chem. Lett. 2016;26(1):38-42. [164] Horwedel C, Tsogoeva SB, Wei S, Efferth T. Cytotoxicity of artesunic acid homo- and heterodimer molecules toward sensitive and multidrug-resistant CCRF-CEM leukemia cells. J. Med. Chem. 2010;53(13):4842-8. [165] Reiter C, Capcı Karagöz A, Fröhlich T, Klein V, Zeino M, Viertel K et al. Synthesis and study of cytotoxic activity of 1,2,4-trioxane- and egonol-derived hybrid molecules against Plasmodium falciparum and multidrug-resistant human leukemia cells. Eur. J. Med. Chem. 2014;75:40312. [166] Reiter C, Fröhlich T, Zeino M, Marschall M, Bahsi H, Leidenberger M et al. New efficient artemisinin derived agents against human leukemia cells, human cytomegalovirus and Plasmodium falciparum: 2nd generation 1,2,4-trioxane-ferrocene hybrids. Eur. J. Med. Chem. 2015;97:164-72. [167] Efferth T, Davey M, Olbrich A, Rücker G, Gebhart E, Davey R. Activity of drugs from traditional Chinese medicine toward sensitive and MDR1- or MRP1-overexpressing multidrug-resistant human CCRF-CEM leukemia cells. Blood Cells Mol. Dis. 2002;28(2):160-8. [168] Ma H, Yao Q, Zhang AM, Lin S, Wang XX, Wu L et al. The effects of artesunate on the expression of EGFR and ABCG2 in A549 human lung cancer cells and a xenograft model. Molecules 2011;16(12):10556-69. [169] Liu L, Zuo LF, Guo JW. Reversal of multidrug resistance by the anti-malaria drug artesunate in the esophageal cancer Eca109/ABCG2 cell line. Oncol. Lett. 2013;6(5):1475-1481. [170] Michaelis M, Kleinschmidt MC, Barth S, Rothweiler F, Geiler J, Breitling R et al. Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines. Biochem. Pharmacol. 2010;79(2):130-6. [171] Lee J, Shen P, Zhang G, Wu X, Zhang X. Dihydroartemisinin inhibits the Bcr/Abl oncogene at the mRNA level in chronic myeloid leukemia sensitive or resistant to imatinib. Biomed. Pharmacother. 2013;67(2):157-63. [172] Luo J, Zhu W, Tang Y, Cao H, Zhou Y, Ji R et ql. Artemisinin derivative artesunate induces radiosensitivity in cervical cancer cells in vitro and in vivo. Radiat. Oncol. 2014;9:84. [173] Li P, Yang S, Dou M, Chen Y, Zhang J, Zhao X. Synergic effects of artemisinin and resveratrol in cancer cells. J. Cancer Res. Clin. Oncol. 2014;140(12):2065-75. [174] Kim SH, Kim HJ, Kim TS. Differential involvement of protein kinase C in human promyelocytic leukemia cell differentiation enhanced by artemisinin. Eur J Pharmacol. 2003 Dec 15;482(13):67-76.
24
[175] Singh NP, Lai HC. Synergistic cytotoxicity of artemisinin and sodium butyrate on human cancer cells. Anticancer Res 2005;25(6B):4325-31. [176] An FF, Liu YC, Zhang WW, Liang L. Dihydroartemisinine enhances dictamnine-induced apoptosis via a caspase dependent pathway in human lung adenocarcinoma A549 cells. Asian Pac. J. Cancer Prev. 2013;14(10):5895-900. [177] Liu Y, Cui YF. Synergism of cytotoxicity effects of triptolide and artesunate combination treatment in pancreatic cancer cell lines. Asian Pac. J. Cancer Prev. 2013;14(9):5243-8. [178] Jiang W, Huang Y, Wang JP, Yu XY, Zhang LY. The synergistic anticancer effect of artesunate combined with allicin in osteosarcoma cell line in vitro and in vivo. Asian Pac J Cancer Prev. 2013;14(8):4615-9. [179] Li Y, Feng L, Li Y, Jiang W, Shan N, Wang X. Artesunate possesses anti-leukemia properties that can be enhanced by arsenic trioxide. Leuk. Lymphoma 2014;55(6):1366-72. [180] Wickerath M, Singh NP. Additive cytotoxic effects of dihydroartemisinin and sodium salicylate on cancer cells. Anticancer Res 2014;34(7):3399-401. [181] Das SS, Nanda GG, Alone DP. Artemisinin and curcumin inhibit Drosophila brain tumor, prolong life span, and restore locomotor activity. IUBMB Life 2014;66(7):496-506. [182] Chen G, Gong R, Shi X, Yang D, Zhang G, Lu A et al. Halofuginone and artemisinin synergistically arrest cancer cells at the G1/G0 phase by upregulating p21Cip1 and p27Kip1. Oncotarget 2016;7(31):50302-50314. [183] Grace JM, Skanchy DJ, Aguilar AJ. Metabolism of artelinic acid to dihydroqinqhaosu by human liver cytochrome P4503A. Xenobiotica 1999;29(7):703-17. [184] Giao PT, de Vries PJ. Pharmacokinetic interactions of antimalarial agents. Clin. Pharmacokinet. 2001;40(5):343-73. [185] Svensson US, Mäki-Jouppila M, Hoffmann KJ, Ashton M. Characterisation of the human liver in vitro metabolic pattern of artemisinin and auto-induction in the rat by use of nonlinear mixed effects modelling. Biopharm. Drug Dispos. 2003;24(2):71-85. [186] Asimus S, Ashton M. Artemisinin - a possible CYP2B6 probe substrate? Biopharm. Drug Dispos. 2009;30(5):265-75. [187] Burk O, Arnold KA, Nussler AK, Schaeffeler E, Efimova E, Avery BA et al. Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. Mol. Pharmacol. 2005;67(6):1954-65. [188] Rhodes SP, Otten JN, Hingorani GP, Hartley DP, Franklin RB. Simultaneous assessment of cytochrome P450 activity in cultured human hepatocytes for compound-mediated induction of CYP3A4, CYP2B6, and CYP1A2. J. Pharmacol. Toxicol. Methods. 2011;63(3):223-6. [189] Xing J, Kirby BJ, Whittington D, Wan Y, Goodlett DR. Evaluation of P450 inhibition and induction by artemisinin antimalarials in human liver microsomes and primary human hepatocytes. Drug Metab. Dispos. 2012;40(9):1757-64. [190] Zang M, Zhu F, Li X, Yang A, Xing J. Auto-induction of phase I and phase II metabolism of artemisinin in healthy Chinese subjects after oral administration of a new artemisininpiperaquine fixed combination. Malar. J. 2014;13:214. [191] Bapiro TE, Egnell AC, Hasler JA, Masimirembwa CM. Application of higher throughput screening (HTS) inhibition assays to evaluate the interaction of antiparasitic drugs with cytochrome P450s. Drug Metab. Dispos. 2001;29(1):30-5. [192] Ericsson T, Masimirembwa C, Abelo A, Ashton M. The evaluation of CYP2B6 inhibition by artemisinin antimalarials in recombinant enzymes and human liver microsomes. Drug Metab. Lett. 2012;6(4):247-57. [193] Ericsson T, Sundell J, Torkelsson A, Hoffmann KJ, Ashton M. Effects of artemisinin antimalarials on Cytochrome P450 enzymes in vitro using recombinant enzymes and human liver microsomes: potential implications for combination therapies. Xenobiotica 2014;44(7):615-26.
25
[194] Kobayashi K, Takahashi O, Hiratsuka M, Yamaotsu N, Hirono S, Watanabe Y et al. Evaluation of influence of single nucleotide polymorphisms in cytochrome P450 2B6 on substrate recognition using computational docking and molecular dynamics simulation. PLoS One 2014;9(5):e96789.
26
Table 1: Enhanced activity of DNA-targeting anticancer drugs by ARS-type compounds. Combination drug
ARStype drug
Cell line
Doxorubicin
DHA
MCF-7 breast Ca
Doxorubicin
DHA
Doxorubicin
ART
Cisplatin
DHA
Cisplatin
DHA
SKOV3 and SKOV3/DDP ovarian Ca
Cisplatin
ART
ovarian Ca
Carboplatin Oxaliplatin
Effect
Synergistic interaction; induction of apoptosis various (HeLa, OVCAR-3, Enhanced doxorubicin activity in vitro and in MCF-7, PC-3 and A549) vivo. Induction of apoptosis and pyknosis; 10 multiple myeloma cell lines Synergistic interaction
Wu et al., 2013 [137] Tai et al., 2016 [138] Papanikolaou et al., 2014 [139] A549 and A549/DDP lung Ca Synergistic interaction in vitro and in vivo. Zhang et al., Inhibition of angiogenesis by downregulation 2013 [140] of HIF-1α and VEGF Enhanced cisplatin activity in cisplatinresistant cells by mTOR inhibition and apoptosis induction
Downregulation of RAD51 by ART sensitizes to cisplatin DHA A2780 and OVCAR-3 ovarian Enhanced carboplatin activity in vitro and in Ca vivo ArteMCF-7 breast Ca; HCT116 Enhanced oxaliplatin activity misone and SW480 colon Ca; PANC1 and MIAPaCa pancreas Ca
Temozolomide DHA
C6 rat glioma
Temozolomide ART Temozolomide DHA
U87MG and A172 glioblastoma 10 glioma cell lines
Temozolomide ART
Glioblastoma
Gemcitabine
HepG2 and Hep3B liver Ca
Gemcitabine
ART, DHA DHA
Gemcitabine
DHA
Reference
Feng et al., 2014 [141] Wang et al., 2015 [35] Chen et al., 2009 [142] Gravett et al., 2011 [143]
Enhanced temozolomide activity by generation of reactive oxygen species Enhanced temozolomide activity
Huang et al., 2008 [144] Karpel-Massler et al., 2014 [145] Enhanced temozolomide activity by Zhang et al., induction of autophagy 2015 [146] Enhanced temozolomide activity in vitro and Berte et al., 2016 in vivo by inhibition of homologous [147] recombination and senescence
Enhanced gemcitabine activity in vitro and in vivo independent of p53 mutational status BxPC-3 and Panc-1 pancreas Enhanced gemcitabine activity in vitro and in Ca vivo. Abolishing the NF-κB inducing activity of gemcitabine
Hou et al., 2008 [148] Wang et al., 2010a [149]
Gemcitabine
BxPC-3 and Panc-1 pancreas Enhanced gemcitabine activity in vitro and in Ca vivo. Inhibition of NF-κB ArteMCF-7 breast Ca; HCT116 Enhanced gemcitabine activity misone and SW480 colon Ca; PANC1 and MIAPaCa pancreas Ca
Wang et al., 2010b [150] Gravett et al., 2011 [143]
Gemcitabine
DHA
A549 lung Ca
Synergistic interaction. Activation of Bak, Caspases 3,8 and 9, loss of mitochondrial membrane potential, inhibition of Fas
Zhao et al., 2014 [151]
Cytarabine
ART, DHA
10 acute myeloic leukemia cell lines
Synergistic interaction
Drenberg et al., 2016 [123]
27
Table 2: Enhanced activity of anticancer drugs with diverse modes of actions by ARS-type compounds. Combination drug
ARStype drug
Erlotinib
ART
Thalidomide
Cell line
11 glioblastoma cell lines ArteMCF-7 breast Ca; misone HCT116 and SW480 colon Ca; PANC-1 and MIAPaCa pancreas Ca
Effect
Reference
Additive and synergistic interactions Enhanced thalidomide activity
Efferth et al., 2004 [152] Gravett et al., 2011 [143]
Lenalidomide
ART
MCF-7 breast Ca, Enhaced lenalidomide Liu et al., 2011 [153] HCT116 colon Ca, activity. Simultaneous arrest A549 lung Ca at all phases of the cell cycle by reduction of cell cycle transition proteins
Bortezomib
ART
10 multiple No cross-resistance of myeloma cell lines bortezomib-resistant cells; non-caspase-mediated cell death; synergy if combined with bortezomib
Papanikolaou et al., 2014 [139]
Dexamethasone ART
10 multiple Synergistic interaction myeloma cell lines Molt-4 leukemia Enhanced growth inhibition
Papanikolaou et al., 2014 [139] Gerhardt et al., 2015 [154]
Dexamethasone DHA
28
Table 3: Activity of ARS-type drugs towards drug-resistant tumor cell lines.
Resistance type
ARS-type drug
P-gp-mediated MDR
ART
P-gp-mediated MDR
CEM/ADR5000 and CEM/VCR100 acute lymphoblastic leukemia ARS, ART, K562/adr chronic DHA myeloic leukemia
P-gp-mediated MDR
ART
P-gp-mediated MDR P-gp-mediated MDR
ART
P-gp-mediated MDR P-gp-mediated MDR
ART
P-gp-mediated MDR P-gp-mediated MDR P-gp-mediated MDR P-gp-mediated MDR P-gp-mediated MDR MRP1-mediated MDR MRP1-mediated MDR
Cell line
Effect
Reference
no cross-resistance
Efferth et al., 2001 [155]
no cross-resistance by loss of mitochondrial membrane potential
Reungpatthanaphong and Mankhetkorn, 2002 [156]
CEM/ADR5000 acute lymphoblastic leukemia CEM/VBL0.05 leukemia cells HT29 colon Ca
No cross-resistance by induction of Efferth et al., 2007 ROS-induced apoptosis [157]
pig brain endothelial cells MCF-7/adr breast cancer
Inhibition of daunorubicin efflux
Li et al., 2008 [158]
Increase of doxorubicin resistance by SERCA inhibition and Pglycoprotein induction
Riganti et al., 2008, 2009 [159,160]
Enhanced calcein-AM uptake by inhibition of blood brain barrier Inhibition of P-gp efflux; collateral sensitivity
Soomro et al., 2011 [45] Zhong et al., 2016 [161]
Artesunic CEM/ADR5000 acute acid dimers lymphoblastic leukemia ARTK562/ADR chronic podophyllo- myeloic leukemia toxin hybrid
no cross-resistance or collateral sensitivity
Reiter et al., 2012 [162]
no cross-resistance by disrupting microtubule network
Zhang et al., 2016 [163]
Artesunic CEM/ADR5000 acute acid dimers lymphoblastic leukemia ARS CEM/ADR5000 acute hybrids lymphoblastic leukemia ARS CEM/ADR5000 acute hybrids lymphoblastic leukemia ART CEM/E1000 acute lymphoblastic leukemia ARS, ART, GLC4/adr lung Ca DHA
no cross-resistance
Horwedel et al., 2010 [164]
no or low degree of crossresistance
Reiter et al., 2014 [165]
no or low degree of crossresistance
Reiter et al., 2015 [166]
Enhancement of daunorubicin uptake
Efferth et al., 2002 [167]
no cross-resistance by loss of mitochondrial membrane potential
Reungpatthanaphong and Mankhetkorn, 2002 [156]
Downregulation of BCRP expression in vitro and in vivo Reversal of drug resistance
Ma et al., 2011 [168]
no cross-resistance
Efferth et al., 2001 [155]
no cross-resistance
Efferth et al., 2001 [155]
ARS
ARS derivatives
BCRP-mediated MDR BCRP-mediated MDR Methotrexate resistance
ART
A549 lung Ca
ART
Hydroxyurea resistance
ART
Eca109/ABCG2 esophageal Ca CEM/MTX1500LV acute lymphoblastic leukemia CEM/HUR90 acute lymphoblastic leukemia
ART
29
Liu et al., 2013 [169]
Cisplatin resistance
ART, DHA
Imatinib resistance DHA
UKF-NB-3rCDDP1000 neuroblastoma
sensitivity of ART or DHA was increased by L-buthionine-S,Rsulfoximine
Michaelis et al., 2010 [170]
chronic myeloic leukemia
Inhibition of BCR/ABL expression and tyrosine kinase function in imatinib-resistant cells
Lee et al., 2013 [171]
30
Table 4: Enhanced activity of radiotherapy or photodynamic therapy by ARS-type compounds. ARS-type Cell line drug Radiotherapy: DHA U373MG glioblastoma ART A549 lung Ca
Effect
Reference
Radiosensitization by ROS generation and GST inhibition Kim et al., 2006 [51] Radiosensitization by nitric oxide-mediated induction of G2M cell cycle arrest.
Zhao et al., 2011 [52]
ART
LN229 and U87MG Radiosensitization by induction of DNA damage, G2M cell Reichert et al., 2012 glioblastoma cycle arrest and apoptosis. [53]
ART
HeLa cervical Ca
Radiosensitization in vitro and in vivo by induction of G2M Luo et al., 2013; 2014 cell cycle arrest and apoptosis. Involvement of multiple [54,172] pathways (RNA transport, the spliceosome, RNA degradation and p53 signaling).
DHA
GLC-82 lung Ca
Radiosensitization by induction of G0G1 cell cycle arrest and apoptosis.
Zuo et al., 2014 [55]
Enhanced growth inhibition and apoptosis.
Li et al., 2014 [173]
Photodynamic therapy: DHA Eca109 and Ec9706 esophageal Ca
31
Table 5: Enhanced cytotoxic activity of ARS-type compounds by drugs established for other indications than cancer.
Combination ARSdrug type drug
Cell line
Effect
Reference
Captopril
ART
HUVEC endothelial cells, Synergistic interaction by inhibition chorioallantoic of angiogenesis membrane of quail embryos
Krusche et al., 2013 [62]
Chloroquine
ARS
A549 lung Ca
Ganguli et al., 2014 [65]
Miconazole
ARS
5637 bladder Ca, 4T1 breast Ca
Synergistic interaction by ROS generation and induction of autophagy Necrosis was more prominent than apoptosis
32
Shahbazfar et al., 2014 [67]
Table 6: Interaction of anticancer activity of ARS-type compounds by novel synthetic compounds.
Combination compound
ARStype drug
Cell line
Effect
Reference
L-Buthionine sulfoximine Ethacrynic acid
ART
MSV-HL13 cells
Sensitization to ART
ART
MSV-HL13 cells
Sensitization to ART
6PGD inhibitors DHA Physcion and S3
Leukemia cells
Synergistic interaction by activation of AMP-activated protein kinase
Efferth and Volm, 2005 [70] Efferth and Volm, 2005 [70] Elf et al., 2016 [78]
Glutaminase-1 inhibitor 968 Histone deacetylase inhibitors N-tert-butylalphaphenylnitrone
Liver Ca cell lines Synergistic interaction by excessive ROS generation HepG2 liver Ca Enhanced DHA activity in vitro and in vivo
Wang et al., 2016 [82] Zhang et al., 2012 [88]
MOLT-4 acute lymphoblastic leukemia
Chan et al., 2013 [90]
DHA DHA
DHA
Attenuation of DHA cytotoxicity
33
Table 7: Enhanced anticancer activity of ARS-type compounds by natural products or natural product derivatives.
Combination compound
ARS- Cell line type drug
Effect
Reference
1 alpha,25dihydoxyvitamin D3 [1,25OH2D3] All-trans retinoic acid Sodium butyrate Dictamnine
ARS
HL-60 leukemia
Enhanced cell differentiation
Kim et al., 2003 [174]
ARS
HL-60 leukemia
Enhanced cell differentiation
Kim et al., 2003 [174] Singh and Lai, 2005 [175] An et al., 2013 [176] Liu and Cui, 2013 [177]
DHA Molt-4 leukemia
Enhanced growth inhibition
DHA A549 lung Ca
Enhanced induction of S phase arrest and apoptosis
Triptolide
ART
Pancreatic Ca
Inhibition of growth, induction of apoptosis, induction of HSP20 and HSP27
Allicin
ART
Osteosarcoma
Enhanced growth inhibition in vitro and in vivo
Arsenic trioxide ART
K562 and U937 leukemia
Enhanced growth inhibition. Induction of apoptosis and downregulation of Fos
Butyric acid
ARS
Resveratrol
ARS
5637 bladder Ca, Necrosis was more prominent than apoptosis Shahbazfar et al., 4T1 breast Ca 2014 [67] HeLa cervical Ca Enhanced growth inhibition, cell migration, apoptosis, Li et al., 2014 [179] and HepG2 liver necrosis and ROS generation Ca
Sodium salicylate Curcumin
DHA leukemia cells
Vitamin C , vitamin D3 Halofuginone
DHA Molt-4 leukemia
Enhanced growth inhibition
ARS
Growth inhibition in vivo by increasing p21CIP, p27KIP and CDK2 to induce G1 cell cycle arrest
ARS
Drosophila melanogaster larvae
MCF-7 breast Ca, HCT116 colon Ca
Jiang et al., 2013 [178] fehlt Li et al., 2014 [179]
Additive interaction
Wickerath and Singh, 2014 [180] Inhibition of brain tumor, prolongation of life span and Das et al., 2014 restoration of locomotor activity [181]
34
Gerhardt et al., 2015 [154] Chen et al., 2016 [182]
Table 8: Enhanced cytotoxic activity of therapeutic antibodies or recombinant proteins by ARS-type drugs.
Combination ARSdrug type drug
Cell line
Effect
Reference
Rituximab
ART
B-cell lymphoma
Sieber et al., 2009 [103]
TRAIL
DHA
Prostate Ca
TRAIL
ART
HeLa cervical Ca
Enhanced inhibition of growth and ROS generation and induction of apoptosis. Downregulation of YY1, Sp1, and Fas Enhanced cell killing. Induction of DR5 Enhanced apoptosis induction. Downregulation of XIAP, Bcl-XL, NFκB, Akt
TRAIL
DHA
BxPC-3 and PANC-1 pancreas Ca
He et al., 2010 [108] Thanaketpaisarn et al., 2011 [109]
Enhanced induction of apoptosis and Kong et al., 2012 ROS generation. DR5 induction [110]
35
Table 9: Enhanced cytotoxic activity of ARS-type drugs by RNA interference.
Combination drug
ARS- Cell line type drug
Effect
Reference
BMI siRNA
ARS
CNE-1 and CNE-2 Enhanced inhibition of growth and G1 Wu et al., 2011 nasopharyngeal Ca cell cycle arrest [113]
VCAM1 shRNA
ARM
U97MG glioblastoma
Enhanced inhibition of cell viability, migration, invasiveness, apoptosis, and MMP-2/-9 expression
Wang et al., 2013 [112]
RAC1 siRNA
DHA
HCT116 and RKO colon Ca
Enhanced inhibition of growth and migration and induction of cell cycle arrest and apoptosis
Han et al., 2013 [114]
PARK7 siRNA
DHA
HeLa cervical Ca
Zhu et al., 2014 [115]
p8 siRNA
DHA
HeLa cervical Ca; HCT116 colon Ca; HepG2 liver Ca; SKOV3 ovarian Ca
Enhanced inhibition of growth. Enhanced induction of ROS Enhanced generation of ROS and apoptosis Reversion of DHA-induced p8 expression led to increased cell death
36
Chen et al., 2015 [116]
Table 10: Role of CYP enzymes for ARS-type drugs. Combination drug
ARS-type drug
Investigation Model
Effect
Reference
CYP2B6, 3A4, and 3A5 CYP2B6 and 3A4 CYP2B6 and 3A4 CYP2B6 and 2A6 CYP2B6
ARE
Liver microsomes
Biotransformation
ARS
B-cell microsomes
Biotransformation
CYP2C19
ARS
CYP2B6 and CYP3A4 CYP2B6 and CYP2C19 CYP2B6
ARS
Grace et al., 1998;1999 [129,183] Svensson et al., 1999 [130] Giao and de Vries, 2001 [184] Svensson et al., 2003 [185] Asimus et al., 2009 [186] Giao and de Vries, 2001 [184] Burk et al., 2005 [187]
ARS
Biotransformation
ARS
Liver microsomes
Biotransformation
ARS
Liver microsomes
Biotransformation Enzyme induction
human hepatocytes
Enzyme induction
ARS, ARE, Healthy volunteers Enzyme induction ARM ARS Human hepatocytes Enzyme induction
CYP3A4 and CYP2B6
ARS
CYP2B6 and CYP3A4 CYP1A2
ARS + piperaquine ARS, DHA Liver microsomes
Enzyme inhibition
CYP2B6
ARS, DHA
Enzyme inhibition
CYP2B6 CYP1A2, 2B6, 2C19 and 3A4 CYP2B6
Liver microsomes, Enzyme induction Human hepatocytes, recombinant enzyme Healthy volunteers Enzyme induction
Liver microsomes, human hepatocytes, recombinant enzyme ARS, ARM Liver microsomes, recombinant enzyme ARS, ARM, Liver microsomes, ART, DHA recombinant enzymes ARM Molecular docking
Elsherbiny et al., 2008 [133] Rhodes et al., 2011 [188] Xing et al., 2012 [189]
Zang et al., 2014 [190] Bapiro et al., 2001 [191] Xing et al., 2012 [189]
Enzyme inhibition
Ericsson et al., 2012 [193]
Enzyme inhibition
Ericsson et al., 2014 [193]
Enzyme inhibition
Kobayashi et al., 2014 [194]
37
Table 11: Interaction of ARS-type drugs with other drugs via hepatic CYP enzymes.
Combination drug
ARS- InvestigationModel Effect type drug
Ketoconazole
ARE
Liver microsomes
Ketoconazole
ARS
Liver microsomes
Troleanodomycin 8-Methoxypsoralen
ARE
Liver microsomes
ARS
Liver microsomes
Omeprazole
ARS
Healthy volunteers
Inhibition of biotransformation of Mihara et al., omeprazole 1999 [131]
Paraxanthine
ARS
Healthy volunteers
CYP1A2-mediated inhibition of biotransformation of paraxanthine to caffeine
Bapiro et al., 2005 [135]
Paraxanthine
ARS, Healthy volunteers ARE, DHA
CYP1A2-mediated inhibition of biotransformation of paraxanthine to caffeine
Asimus et al., 2007 [136]
CYP2B6-mediated biotransformation of Smephenytoin CYP2B6- and CYP2C19mediated biotransformation of Smephenytoin Inhibition of biotransformation of carbamazepine
Simonsson et al., 2003 [132]
S-mephenytoin ARS
Liver microsomes
S-mephenytoin ARS
Healthy volunteers
Carbamazepine ARS, Rabbits ARE, ARM
Reference
Inhibition of biotransformation of Grace et al., ARE 1998, 1999 [129,183] Inhibition of biotransformation of Svensson et al., ARS 1999 [130] Inhibition of biotransformation of ARE Inhibition of biotransformation of ARS
38
Grace et al., 1998 [129] Svensson et al., 1999 [130]
Elsherbiny et al., 2008 [133] Sukhija et al., 2006 [134]
Legends to figures:
Figure 1: Synopsis of mechanisms involved in combination treatments between ARS-based drugs and other treatment modalities for cancer therapy. Figure 2: Combating resistance to clinically established anticancer drugs by ARS and its derivatives. Figure 3: Role of CYP enzymes for artemisinin and its derivatives.
Figure 4: Cellular targets of ARS-type drugs and partner compounds in combination therapy approaches. Figure 5: Chemical structures of ARS-type drugs and partner compounds in combination therapy approaches.
39
• Artemisinin • Artesunate • Dihydroartemisinin
Standard anticancer drugs Radiotherapy Photodynamic therapy Drugs for other indications Novel synthetic compounds Natural products Therapeutic antibodies and recombinant proteins • RNA interference • • • • • • •
Combination therapy of cancer • • • • •
Inhibition of cell growth and cell viability Induction of cell cycle arrest Induction of apoptosis and autophagy Inhibition of invasion, migration and metastasis Inhibition of angiogenesis
Hepatotoxicity and drug-drug interactions
40
• Artemisinin • Artesunate • Dihydroartemisinin
P-gp-mediated MDR MRP-mediatedMDR BCRP-mediated MDR Cisplatin resistance Methotrexate resistance • Hydroxyurea resistance • Imatinib resistance • • • • •
• Cross-resistance • Bypassing • Functional inhibition of resistance proteins • Collateral senstivity
41
Biotransformation CYP2A6, 2B6, 3A4
Artemisinin Artesunate Dihydroartemisinin
Induction
Inhibition
CYP2B6, 2C19, 3A4
CYP1A2, 2B6, 2C19, 3A4
42
43
Butyric acid
Chloroquine
Allicin
Halofuginone
Omeprazole
n-Tert-butyl α phenylnitrone
Resveratrol
Vitamin D3
Vitamin C
Sodium salicylate
Miconazole
Paraxanthine
Ketoconazole
S-Mephenytoin
Sodium butyrate
Curcumin
8-Methoxypsoralen
Carbamazepine
44
45