bannter.jpg
·
Review Article·

 

Potential applications of artemisinins in ocular diseases

 

Bing-Wen Lu, Li-Ke Xie

 

Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100400, China

Correspondence to: Li-Ke Xie. Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100400, China. bjxielike@sina.com

Received: 2018-08-28        Accepted: 2019-04-17

 

Abstract

Artemisinin, also named qinghaosu, is a family of sesquiterpene trioxane lactone originally derived from the sweet wormwood plant (Artemisia annua), which is a traditional Chinese herb that has been universally used as anti-malarial agents for many years. Evidence has accumulated during the past few years which demonstrated the protective effects of artemisinin and its derivatives (artemisinins) in several other diseases beyond malaria, including cancers, autoimmune disorders, inflammatory diseases, viral and other parasite-related infections. Recently, this long-considered anti-malarial agent has been proved to possess anti-oxidant, anti-inflammatory, anti-apoptotic and anti-excitotoxic properties, which make it a potential treatment option for the ocular environment. In this review, we first described the overview of artemisinins, highlighting the activity of artemisinins to other diseases beyond malaria and the mechanisms of these actions. We then emphasized the main points of published results of using artemisinins in targeting ocular disorders, including uveitis, retinoblastoma, retinal neurodegenerative diseases and ocular neovascularization. To conclude, we believe that artemisinins could also be used as a promising therapeutic drug for ocular diseases, especially retinal vascular diseases in the near future.

KEYWORDS: artemisinins; uveitis; retinoblastoma; retinal neurodegenerative diseases; ocular neovascularization

DOI:10.18240/ijo.2019.11.20

 

Citation: Lu BW, Xie LK. Potential applications of artemisinins in ocular diseases. Int J Ophthalmol 2019;12(11):1793-1800

 

INTRODUCTION

Artemisinin and its derivatives (artemisinins) are isolated from the one ancient Chinese plant Artemisia annua (more commonly known as sweet wormwood), which have been used in traditional Chinese medicine (TCM) for fevers and chills[1]. Following the isolation of the active agent by Dr. You-You Tu’s group from the Chinese Academy of TCM in the 1970s, artemisinin-based combination therapies have joined the currently established standard treatments of malarial parasites around the world[2-4]. Interestingly, abundant evidences have also demonstrated that artemisinins might also be of therapeutic value for many other diseases beyond malaria, including cancers, autoimmune disorders, inflammatory diseases as well as other infectious conditions[5]. Recently, many ophthalmologists and researchers have also showed their great interest in artemisinins, especially artesunate and dihydroartemisinin (DHA) and their potential protective effects on ocular disorders. Herein, we present an overview of research advances of artemisinins as potential therapeutic methods for ocular diseases, including uveitis, retinoblastoma, retinal neurodegenerative diseases, especially ocular neovascularization (NV). In this review, we also emphasize some important points regarding the potential applications of artemisinins in ocular disorders to provide a platform for additional study.


OVERVIEW OF ARTEMISININS

History and Origins  The medicinal herb Artemisia annua was first recognized by one Chinese physician, Hong Ge (born in the year 283) for its fever-reducing properties[6]. Led by the Chinese project 523 in the 1970s, Dr. You-You Tu’s group first successfully isolated artemisinin, a non-toxic extract of Artemisia annua, identified the active component of this extract in 1972 and further identified its stereostucture (sesquiterpene lactones) in 1975[1]. In the 1980-90s, further studies conducted in humans confirmed the recognition of artemisinin-based combination therapies as the first-line option to treat malaria[2-4]. This novel anti-malaria therapy has been used universally with great efficacy and safety for a long time and helped Dr. You-You Tu win the 2015 Nobel Prize in Physiology or Medicine for her outstanding achievements[7].

Chemical and Pharmacological Characteristics  It was Dr. You-You Tu who first clarified the molecule extracted from the herbaceous plant Artemisia annu to be a sesquiterpene lactone endoperoxide by using the combined method of mass spectroscopy, spectrophotometry, X-ray crystallography and polyarithmetic analysis[8]. Those clinically important artemisinins include artesunate, artemether, arteether, and DHA (Figure 1), discovered and developed in 1986[1]. Among which artesunate is the most important analog, which shows a more favorable pharmacological profile because of its greater water-solubility and high oral bioavailability due to the additional hemisuccinate group[9].

Bing-Wen Lu1

Figure 1 Chemical structures of artemisinins.

 

Beyond Malaria: Activity of Artemisinins to Other Diseases  While the efficacy and low toxicity of artemisinins to treat malaria is well-recognized around the world, they have currently been reported to have a great therapeutic value beyond malaria[10]. These capacities include protective functions in non-malaria parasitic infections[11-14], anti-viral[15-17] and anti-fungal properties[18-19], anti-cancer functions[20-24], as well as anti-inflammatory[25-27] and anti-allergic effects[28-29] (Figure 2). Recent results further indicated that artemisinins might also reduce glucose, thus exerting a protective effect on diabetes mellitus[30].

Bing-Wen Lu2

Figure 2 Various biological activities of artemisinins and potential applications in different diseases (Bubble map).

 

Mechanisms of Actions of Artemisinins  Although artemisinins are long known and effectively used as anti-malaria drugs, their specific biological action is poorly identified and understood. Current in vivo and in vitro studies have proposed numerous possible mechanisms of the actions, which include 1) oxidative stress, 2) induction of apoptosis[31], 3) inhibition of angiogenesis[32-33], 4) arrest of cell cycle at G0/G1[34] (Figure 3). As a matter of fact, these functional pathways may overlap in a number of ways.

Bing-Wen Lu3

Figure 3 Overview of mechanisms of actions of artemisinins  - indicates inhibition and + indicates activation.

 

Oxidative Stress  Reactive oxygen species (ROS) are the natural byproduct of aerobic metabolism, whose levels can dramatically elevate during times of environmental stress. Studies in various tumor cell lines have proved ROS to have an important role in artemisinins-induced apoptosis[31]. These studies covered neuroblastoma[32], breast cancer[33], T-cell lymphoma[34], embryonal rhabdomyosarcoma cells[35], and glioblastoma[36]. In a recent study on human hepatocellular carcinoma cells, artesunate was shown to be able to induce ROS-dependent apoptosis via Bax-mediated intrinsic pathway[37]. Similarly, DHA was shown to alleviate oxidative stress in bleomycin-induced pulmonary fibrosis[38].

Induction of Apoptosis  Apoptosis, or programmed cell death, is a regulated cellular suicide mechanism involving the degradation of cellular components, which can be initiated via the intrinsic pathway and the extrinsic pathway[39]. Artemisinins could trigger apoptotic cell death through both pathways[40-41]. In human colon cancer cell line (HT29), B-cell lymphoma 2 associated X protein (BAX) was proved to be activated by artemisinins, inducing the release of cytochrome C, which led to apoptosis in cancer cells[42]. In human prostate cancer cell line (DU145), cleavage of procaspases 3 and 9 was found to be induced by artesunate, inducing the release of cytochrome C and the subsequent caspase-dependent apoptosis[43]. In human breast cancer cell line (MCF-7), apoptosis was also induced via a caspase-related mechanism under the effect of a semi-synthetic derivative of artemisinin[44].

Inhibition of Angiogenesis  Various models have accumulated mounting evidences, demonstrating the involvement of inhibiting aberrant angiogenesis in the actions of artemisinins[45-46]. In mouse embryonic stem cells, artemisinin was shown to be able to reduce the levels of hypoxia inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF), suggesting the mechanism of artemisinin might involve the inhibition of angiogenesis[47]. Artemisinin was also found to be able to significantly reduce lymph-angiogenesis via downregulating the expression of VEGF-C in C57BL/6 mouse Lewis lung carcinoma model[48]. Similarly, in a rat glioma model, artemisinins were shown to have the effect of reducing VEGF and angiogenesis[49]. Moreover, artesunate was proved to be able to suppress osteoclastogenesis and aberrant angiogenesis, thus attenuating anterior cruciate ligament transection (ACLT)-induced osteoarthritis[50].

Arrest of Cell Cycle at G0/G1  Artemisinins have been shown by accumulating current studies to have the potential application in cancer drug development for its action on inducing growth arrest at various stages of cell division cycle[51-53]. In prostate cancer cells (LNCaP), phosphorylated retinoblastoma protein (pRB), a mediator cooperating with E2F transcription factors and cyclin-dependent kinases (CDKs) to push forward the cell cycle progression through G1 into S phase was shown to be ablated by artemisinin, inducig G1 cell cycle arrest, thus inhibiting cell division[54]. Willoughby et al[55] has also demonstrated that artemisinin could disrupt specificity protein 1 (Sp1) transcription factor from binding to CDK4 promoter and inhibiting CDK4 gene expression, thus blocking prostate cancer growth and cell cycle progression. Wu et al[56] have further proved the growth inhibition effect of artemisinin in nasopharyngeal carcinoma cell lines by suppressing the level of cyclin D1, cyclin E, CDK2, CDK4, CDK6 and upregulating the inhibitors of cell cycle division (p16, p27).


POTENTIAL APPLICATION IN OCULAR DISEASES

Recently, many ophthalmologists and researchers have noticed the potential protective effects of artemisinins on ocular disorders. Recent findings have shed light on the potential applications of artemisinins as promising therapeutic agents in ocular diseases. In this review, we are going to highlight the main points of published results of using artemisinins in targeting ocular disorders.

Uveitis  Uveitis is the inflammation of the uvea whereas the anti-inflammatory effects of artemisinins have already been recognized in the past few decades[57]. Artesunate has been reported by Li et al[58] to have a protective effect on sepsis mouse model by decreasing serum endotoxin release and toll-like receptors (TLR)4, TLR9 expressions, also suppressing nuclear factor-kappa B (NF-κB) activation. Xu et al[59] also reported that in human rheumatoid arthritis fibroblast-like synoviocytes, artesunate was able to inhibit TNF-α expression and decrease the secretion of pro-inflammatory cytokines. Based on those experimental results, the question of if artesunate could reduce the release of inflammatory cytokines in some type of inflammatory ocular diseases was raised and further investigated. Wang et al[60] studied the protective effect of artesunate by using endotoxin-induced uveitis (EIU) rat model, which has been generally considered as an experimental model for human uveitis[61]. In their study, artesunate of three concentrations (1, 10, 100 mg/kg) were intravenously injected in male Long-Evans rats whereas prednisolone (10 mg/kg) was used as positive control and their results showed that artesunate (10 mg/kg and 100 mg/kg) could suppress infiltrating cells and protein concentration in the aqueous humor, suggesting that artesunate treatment could suppress the inflammation of EIU by inhibiting the production of inflammatory mediators[60]. More future studies will be needed to clearly define the specific cellular mechanisms of the therapeutic effects. The role of artemisinins in modulating ocular inflammatory responses might be of great interest in the future.

Retinoblastoma  In recent years, artemisinins have been shown to exert protective effects in various types of cancer[62-66]. Retinoblastoma (RB) is an eye cancer, which is most common among children[67]. Zhao et al[68] tested the anti-neoplastic activity of artesunate against RB to see whether artesunate might be a good candidate to treat RB. Using epithelial retina cell line as normal counterpart, the cytotoxic activity and specificity of artesunate were analyzed in an RB cell line, which showed a dose-dependent manner concerning the cytotoxic activity specific to RB cells, with low toxicity in normal retina cells and high cytotoxicity in RB cells[68]. Their results also demonstrated that artesunate, even at low doses, could block the cell cycle progression at the G1 phase[68]. Artesunate is practically suitable for long-term treatments with few side-effects. Therefore, artesunate could be considered as a promising option for RB treatment. Further randomized studies in vivo need to be done to provide better insights regarding the efficacy as well as efficiency of the novel treatment.

Retinal Neurodegenerative Diseases  Retinal neurodegeneration is a retinopathy which consists in the deterioration of the retina caused by the progressive death of its neuronal cells[69]. There are several reasons for retinal neurodegeneration, including age-related macular degeneration (AMD), diabetic retinopathy (DR), and retinal artery or vein occlusion[69]. Zeng et al[70] studied the neurogenic effects of artemisinin and their findings indicated that artemisinin at low concentration could induce neurite outgrowth as well as promote neuronal differentiation in PC12 cells.

Accordingly, Chong and Zheng[71] demonstrated that artemisinin was able to suppress hydrogen peroxide (H2O2)-induced oxidative stress in D407 retinal pigment epithelium (RPE) cells, which are first damaged in retinal diseases owning to their critical support functions for photoreceptors. The findings of Yan et al[72] also demonstrated that artemisinin could prevent RPE cells from oxidative stress via the MAPK/CREB pathway.

These recent results all shed light on the promising therapeutic value of artemisinin as a candidate drug for the treatment of many retinal neurodegenerative disorders. Though, its specific effects on the retinal neuronal cells need to be further explored.

Ocular Neovascularization  Ocular NV is one of the major causes of blindness among ocular disorders. Substantial evidences have demonstrated that VEGF played an essential part in its pathogenesis[73]. Currently for the treatment of ocular NV, anti-VEGF agents such as ranibizumab and bevacizumab are widely used[74-75]. However, these drugs both have a large molecular weight and resistance to these drugs is usually seen in approximately 20%-30% ocular NV patients[76]. Moreover, because of the short aqueous half-life, the recurrence rate is high after anti-VEGF treatments which may also increase the risk of endophthalmitis owning to frequent intravitreal injections[77]. Abundant studies have already demonstrated the anti-angiogenic effects of artemisinins in tumors[78]. The known mechanisms of artemisinins in inhibiting angiogenesis include downregulating several growth factors, inducing apoptosis of vascular endothelial cells, upregulating angiogenesis inhibitors, depleting the levels of the flt-1 and KDR/flk-1-receptors[79-80]. In human umbilical vein endothelial cell (HUVEC) lines, artesunate was shown to inhibit angiogenesis through downregulating the levels of the VEGF receptors[81]. Similar protective effects were also investigated in lymphatic endothelial cells and Lewis lung carcinoma cells with the treatment of DHA[82]. In the science of ophthalmology, Cheng et al[83] demonstrated that artesunate could inhibit corneal NV by inducing ROS-dependent apoptosis in animal models. Their results suggested that artesunate could markedly inhibit angiogenesis by specifically inducing apoptosis via an iron/ROS-dependent p38 MAPK-mitochondrial pathway in vascular endothelial cells[83]. Zong et al[84] further investigated the use of artesunate in retinal NV and found that retinal NV could be remarkably inhibited under the effect of artesunate via downregulating the expression of VEGFR2, and PDGFR. Compared to bevacizumab, artesunate could remarkably inhibit retinal NV in rabbits with more durable efficacy. These two published animal evidences indicated the potential role of artesunate as a promising drug candidate to manage ocular NVs. As a newly-discovered anti-angiogenesis drug, artemisinins are worthwhile to be further explored due to a host of advantages.

Compared to the currently used anti-VEGF drugs, the advantages of artesunate are as follows: 1) Small molecule size: artesunate is a 384 Da molecule less than one-hundredth the size of bevacizumab (149 kDa); 2) Safety and low toxicity: artesunate has been widely used for many years as anti-malarial agents, with few adverse side effects and proven safety records; 3) Multi-targets: artesunate was proved to possess not only anti-angiogenetic effects targeting multi-growth factors (VEGF, FGF, HIF-1ɑ, and Ang-1), but also anti-inflammatory and anti-apoptotic effects.

Thus, we postulate that artesunate might be a potential novel treatment option for retinal vascular diseases such as AMD, DR, retinal artery or vein occlusion, especially when given intravitreously or being formulated into eye drops.


LIMITATIONS OF ARTEMISININS

The present studies of artemisinins have several limitations. While applying artemisinins for treatments beyond malaria, different research groups have reported inconsistent effective doses even for similar cell lines or animal models. Progress for further clinical trials could be hampered for the lack of a concerted effort to confirm the efficacies of artemisinins in different models. Another limitation is the lack of acute and chronic toxicological studies for acute as well as chronic exposure to artemisinin in ocular diseases, which is necessary for future application in ocular diseases.


CONCLUSION

To date, researches on artemisinins and its applications in ocular diseases are still limited, and much more will need to be studied. Further understanding of the protective activities of artemisinins beyond malaria might lead to improved treatments for ocular disorders.

In this review, we summarized recent studies on artemisinins in treating ocular diseases and we believe that this anti-malaria agent could also be used as a promising therapeutic drug for ocular diseases, especially retinal vascular diseases.


ACKNOWLEDGEMENTS

Authors’ contributions: Lu BW wrote the manuscript; Xie LK participated discussion and provided suggestion.

Conflicts of Interest: Lu BW, None; Xie LK, None.

REFERENCES

1 Tu Y. The discovery of artemisinin (qinghaosu) and gifts from Chinese medicine. Nat Med 2011;17(10):1217-1220.
https://doi.org/10.1038/nm.2471
PMid:21989013

 

2 Song J, Socheat D, Tan B, Seila S, Xu Y, Ou F, Sokunthea S, Sophorn L, Zhou C, Deng C, Wang Q, Li G. Randomized trials of artemisinin-piperaquine, dihydroartemisinin-piperaquine phosphate and artemether-lumefantrine for the treatment of multi-drug resistant falciparum malaria in Cambodia-Thailand border area. Malar J 2011;10:231.
https://doi.org/10.1186/1475-2875-10-231
PMid:21827706 PMCid:PMC3169515

 

3 Makowiecki M, Bednarska A, Paciorek M, Kowalska J, Skrzat-Klapaczyńska A, Puła J, Sosińska-Bryła I, Krogulec D, Raczyńska J, Hackiewicz M, Stengiel J, Bursa D, Pihowicz A, Horban A. Usefulness of SOFA score and artesunate-based treatment in severe malaria - a single center study. Przegl Epidemiol 2018;72(2):215-221.

 

4 Mace KE, Arguin PM, Tan KR. Malaria surveillance-United States, 2015. MMWR Surveill Summ 2018;67(7):1-28.
https://doi.org/10.15585/mmwr.ss6707a1
PMid:29723168 PMCid:PMC5933858

 

5 Raffetin A, Bruneel F, Roussel C, Thellier M, Buffet P, Caumes E, Jauréguiberry S. Use of artesunate in non-malarial indications. Médecine Et Maladies Infect 2018;48(4):238-249.
https://doi.org/10.1016/j.medmal.2018.01.004
PMid:29422423

 

6 van Agtmael MA, Eggelte TA, van Boxtel CJ. Artemisinin drugs in the treatment of malaria: from medicinal herb to registered medication. Trends Pharmacol Sci 1999;20(5):199-205.
https://doi.org/10.1016/S0165-6147(99)01302-4

 

7 Chen WJ. Honoring antiparasitics: the 2015 Nobel prize in physiology or medicine. Biomed J 2016;39(2):93-97.
https://doi.org/10.1016/j.bj.2016.04.002
PMid:27372164 PMCid:PMC6139675

 

8 Stringham RW, Moore GL, Teager DS, Yue TY. Analysis and isolation of potential artemisinin precursors from waste streams of Artemisia annua extraction. ACS Omega 2018;3(7):7803-7808.
https://doi.org/10.1021/acsomega.8b00974
PMid:30087924 PMCid:PMC6068693

 

9 Pinheiro LCS, Feitosa LM, Silveira FFD, Boechat N. Current antimalarial therapies and advances in the development of semi-synthetic artemisinin derivatives. An Acad Bras Cienc 2018;90(1 Suppl 2):1251-1271.
https://doi.org/10.1590/0001-3765201820170830
PMid:29873667

 

10 Loo CS, Lam NS, Yu D, Su XZ, Lu F. Artemisinin and its derivatives in treating protozoan infections beyond malaria. Pharmacol Res 2017;117:192-217.
https://doi.org/10.1016/j.phrs.2016.11.012
PMid:27867026 PMCid:PMC5316320

 

11 Nibret E, Wink M. Volatile components of four Ethiopian Artemisia species extracts and their in vitro antitrypanosomal and cytotoxic activities. Phytomedicine 2010;17(5):369-374.
https://doi.org/10.1016/j.phymed.2009.07.016
PMid:19683909

 

12 Hencken CP, Jones-Brando L, Bordón C, Stohler R, Mott BT, Yolken R, Posner GH, Woodard LE. Thiazole, oxadiazole, and carboxamide derivatives of artemisinin are highly selective and potent inhibitors of Toxoplasma gondii. J Med Chem 2010;53(9):3594-3601.
https://doi.org/10.1021/jm901857d
PMid:20373807 PMCid:PMC2865576

 

13 Singh C, Kanchan R, Chaudhary S, Puri SK. Linker-based hemisuccinate derivatives of artemisinin: synthesis and antimalarial assessment against multidrug-resistant Plasmodium yoelii nigeriensis in mice. J Med Chem 2012;55(3):1117-1126.
https://doi.org/10.1021/jm2010699
PMid:22216834

 

14 Ferreira JF, Peaden P, Keiser J. In vitro trematocidal effects of crude alcoholic extracts of Artemisia annua, A. Absinthium, Asimina triloba, and Fumaria officinalis: trematocidal plant alcoholic extracts. Parasitol Res 2011;109(6):1585-1592.
https://doi.org/10.1007/s00436-011-2418-0
PMid:21562762

 

15 Fröhlich T, Reiter C, Saeed MEM, Hutterer C, Hahn F, Leidenberger M, Friedrich O, Kappes B, Marschall M, Efferth T, Tsogoeva SB. Synthesis of thymoquinone-artemisinin hybrids: new potent antileukemia, antiviral, and antimalarial agents. ACS Med Chem Lett 2018;9(6):534-539.
https://doi.org/10.1021/acsmedchemlett.7b00412
PMid:29937978 PMCid:PMC6004568

 

16 Parvez MK, Arbab AH, Al-Dosari MS, Al-Rehaily AJ. Antiviral natural products against chronic hepatitis B: recent developments. Curr Pharm Des 2016;22(3):286-293.
https://doi.org/10.2174/1381612822666151112152733
PMid:26561057

 

17 Jana S, Iram S, Thomas J, Hayat MQ, Pannecouque C, Dehaen W. Application of the triazolization reaction to afford dihydroartemisinin derivatives with anti-HIV activity. Molecules 2017;22(2):E303.
https://doi.org/10.3390/molecules22020303
PMid:28218680 PMCid:PMC6155659

 

18 Chang CC, Sorrell TC, Chen SC. Pulmonary cryptococcosis. Semin Respir Crit Care Med 2015;36(5):681-691.
https://doi.org/10.1055/s-0035-1562895
PMid:26398535

 

19 Gautam P, Upadhyay SK, Hassan W, Madan T, Sirdeshmukh R, Sundaram CS, Gade WN, Basir SF, Singh Y, Sarma PU. Transcriptomic and proteomic profile of Aspergillus fumigatus on exposure to artemisinin. Mycopathologia 2011;172(5):331-346.
https://doi.org/10.1007/s11046-011-9445-3
PMid:21755315

 

20 Jamalzadeh L, Ghafoori H, Aghamaali M, Sariri R. Induction of apoptosis in human breast cancer MCF-7 cells by a semi-synthetic derivative of artemisinin: a caspase-related mechanism. Iran J Biotechnol 2017;15(3):157-165.
https://doi.org/10.15171/ijb.1567
PMid:29845064 PMCid:PMC5811062

 

21 Li S, Li G, Yang X, Meng Q, Yuan S, He Y, Sun D. Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents. Bioorg Med Chem Lett 2018;28(13):2275-2278.
https://doi.org/10.1016/j.bmcl.2018.05.035
PMid:29789258

 

22 Yu H, Hou Z, Tian Y, Mou Y, Guo C. Design, synthesis, cytotoxicity and mechanism of novel dihydroartemisinin-coumarin hybrids as potential anti-cancer agents. Eur J Med Chem 2018;151:434-449.
https://doi.org/10.1016/j.ejmech.2018.04.005
PMid:29649740

 

23 Våtsveen TK, Myhre MR, Steen CB, Wälchli S, Lingjærde OC, Bai B, Dillard P, Theodossiou TA, Holien T, Sundan A, Inderberg EM, Smeland EB, Myklebust JH, Oksvold MP. Artesunate shows potent anti-tumor activity in B-cell lymphoma. J Hematol Oncol 2018;11(1):23.
https://doi.org/10.1186/s13045-018-0561-0
PMid:29458389 PMCid:PMC5819282

 

24 Deeken JF, Wang H, Hartley M, Cheema AK, Smaglo B, Hwang JJ, He AR, Weiner LM, Marshall JL, Giaccone G, Liu S, Luecht J, Spiegel JY, Pishvaian MJ. A phase I study of intravenous artesunate in patients with advanced solid tumor malignancies. Cancer Chemother Pharmacol 2018;81(3):587-596.
https://doi.org/10.1007/s00280-018-3533-8
PMid:29392450

 

25 Mu X, Wang C. Artemisinins-a promising new treatment for systemic lupus erythematosus: a descriptive review. Curr Rheumatol Rep 2018;20(9):55.
https://doi.org/10.1007/s11926-018-0764-y
PMid:30056574

 

26 Kuang M, Cen Y, Qin R, Shang S, Zhai Z, Liu C, Pan X, Zhou H. Artesunate attenuates pro-inflammatory cytokine release from macrophages by inhibiting TLR4-mediated autophagic activation via the TRAF6-Beclin1-PI3KC3 pathway. Cell Physiol Biochem 2018;47(2): 475-488.
https://doi.org/10.1159/000489982
PMid:29794440

 

27 Jiao J, Yang Y, Liu M, Li J, Cui Y, Yin S, Tao J. Artemisinin and Artemisia annua leaves alleviate Eimeria tenella infection by facilitating apoptosis of host cells and suppressing inflammatory response. Vet Parasitol 2018;254:172-177.
https://doi.org/10.1016/j.vetpar.2018.03.017
PMid:29657004

 

28 Deng Y, Liu Z, Geng Y. Anti-allergic effect of Artemisia extract in rats. Exp Ther Med 2016;12(2):1130-1134.
https://doi.org/10.3892/etm.2016.3361
PMid:27446332 PMCid:PMC4950231

 

29 Li J, Wang B, Luo Y, Bian Y, Wang R. Effect of artemisinin and neurectomy of pterygoid canal in ovalbumin-induced allergic rhinitis mouse model. Allergy Asthma Clin Immunol 2018;14:22.
https://doi.org/10.1186/s13223-018-0249-6
PMid:29991950 PMCid:PMC5994650

 

30 Li J, Casteels T, Frogne T, et al. Artemisinins target GABAA receptor signaling and impair α cell identity. Cell 2017;168(1-2):86-100.e15.
https://doi.org/10.1016/j.cell.2016.11.010
PMid:27916275 PMCid:PMC5236063

 

31 Tsuda K, Miyamoto L, Hamano S, Morimoto Y, Kangawa Y, Fukue C, Kagawa Y, Horinouchi Y, Xu W, Ikeda Y, Tamaki T, Tsuchiya K. Mechanisms of the pH- and oxygen-dependent oxidation activities of artesunate. Biol Pharm Bull 2018;41(4):555-563.
https://doi.org/10.1248/bpb.b17-00855
PMid:29607928

 

32 Michaelis M, Kleinschmidt MC, Barth S, Rothweiler F, Geiler J, Breitling R, Mayer B, Deubzer H, Witt O, Kreuter J, Doerr HW, Cinatl J, Cinatl J Jr. Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines. Biochem Pharmacol 2010;79(2):130-136.
https://doi.org/10.1016/j.bcp.2009.08.013
PMid:19698702

 

33 Hamacher-Brady A, Stein HA, Turschner S, Toegel I, Mora R, Jennewein N, Efferth T, Eils R, Brady NR. Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J Biol Chem 2011;286(8):6587-6601.
https://doi.org/10.1074/jbc.M110.210047
PMid:21149439 PMCid:PMC3057810

 

34 Wang Q, Wu S, Zhao X, Zhao C, Zhao H, Huo L. Mechanisms of dihydroartemisinin and dihydroartemisinin/holotransferrin cytotoxicity in T-cell lymphoma cells. PLoS One 2015;10(10):e0137331.
https://doi.org/10.1371/journal.pone.0137331
PMid:26502166 PMCid:PMC4621048

 

35 Beccafico S, Morozzi G, Marchetti MC, Riccardi C, Sidoni A, Donato R, Sorci G. Artesunate induces ROS- and p38 MAPK-mediated apoptosis and counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cells. Carcinogenesis 2015;36(9):1071-1083.
https://doi.org/10.1093/carcin/bgv098
PMid:26153023

 

36 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;7(41):67235-67250.
https://doi.org/10.18632/oncotarget.11972
PMid:27626497 PMCid:PMC5341871

 

37 Pang Y, Qin G, Wu L, Wang X, Chen T. Artesunate induces ROS-dependent apoptosis via a Bax-mediated intrinsic pathway in Huh-7 and Hep3B cells. Exp Cell Res 2016;347(2):251-260.
https://doi.org/10.1016/j.yexcr.2016.06.012
PMid:27327234

 

38 Yang DX, Qiu J, Zhou HH, Yu Y, Zhou DL, Xu Y, Zhu MZ, Ge XP, Li JM, Lv CJ, Zhang HQ, Yuan WD. Dihydroartemisinin alleviates oxidative stress in bleomycin-induced pulmonary fibrosis. Life Sci 2018;205:176-183.
https://doi.org/10.1016/j.lfs.2018.05.022
PMid:29752961

 

39 Thomas RB, Joy S, Ajayan MS, Paulose CS. Neuroprotective potential of Bacopa monnieri and Bacoside A against dopamine receptor dysfunction in the cerebral cortex of neonatal hypoglycaemic rats. Cell Mol Neurobiol 2013;33(8):1065-1074.
https://doi.org/10.1007/s10571-013-9973-0
PMid:23975094

 

40 Ho WE, Peh HY, Chan TK, Wong WS. Artemisinins: pharmacological actions beyond anti-malarial. Pharmacol Ther 2014;142(1):126-139.
https://doi.org/10.1016/j.pharmthera.2013.12.001
PMid:24316259

 

41 Gunjan S, Sharma T, Yadav K, Chauhan BS, Singh SK, Siddiqi MI, Tripathi R. Artemisinin derivatives and synthetic trioxane trigger apoptotic cell death in asexual stages of Plasmodium. Front Cell Infect Microbiol 2018;8:256.
https://doi.org/10.3389/fcimb.2018.00256
PMid:30094226 PMCid:PMC6070741

 

42 Riganti C, Doublier S, Viarisio D, Miraglia E, Pescarmona G, Ghigo D, Bosia A. Artemisinin induces doxorubicin resistance in human colon cancer cells via calcium-dependent activation of HIF-1alpha and P-glycoprotein overexpression. Br J Pharmacol 2009;156(7):1054-1066.
https://doi.org/10.1111/j.1476-5381.2009.00117.x
PMid:19298255 PMCid:PMC2697684

 

43 Nakase I, Gallis B, Takatani-Nakase T, Oh S, Lacoste E, Singh NP, Goodlett DR, Tanaka S, Futaki S, Lai H, Sasaki T. Transferrin receptor-dependent cytotoxicity of artemisinin-transferrin conjugates on prostate cancer cells and induction of apoptosis. Cancer Lett 2009;274(2):290-298.
https://doi.org/10.1016/j.canlet.2008.09.023
PMid:19006645

 

44 Zeng Z, Xu J, Zheng W. Artemisinin protects PC12 cells against β-amyloid-induced apoptosis through activation of the ERK1/2 signaling pathway. Redox Biol 2017;12:625-633.
https://doi.org/10.1016/j.redox.2017.04.003
PMid:28391183 PMCid:PMC5385605

 

45 Iyer AS, Chapoval SP. Neuroimmune semaphorin 4A in cancer angiogenesis and inflammation: a promoter or a suppressor? Int J Mol Sci 2018;20(1):E124.
https://doi.org/10.3390/ijms20010124
PMid:30598022 PMCid:PMC6337608

 

46 Wei T, Liu J. Anti-angiogenic properties of artemisinin derivatives (Review). Int J Mol Med 2017;40(4):972-978.
https://doi.org/10.3892/ijmm.2017.3085
PMid:28765885

 

47 Wartenberg M, Wolf S, Budde P, Grünheck F, Acker H, Hescheler J, Wartenberg G, Sauer H. The antimalaria agent artemisinin exerts antiangiogenic effects in mouse embryonic stem cell-derived embryoid bodies. Lab Invest 2003;83(11):1647-1655.
https://doi.org/10.1097/01.LAB.0000098424.38003.FF
PMid:14615418

 

48 Wang J, Zhang B, Guo Y, Li G, Xie Q, Zhu B, Gao J, Chen Z. Artemisinin inhibits tumor lymphangiogenesis by suppression of vascular endothelial growth factor C. Pharmacology 2008;82(2):148-155.
https://doi.org/10.1159/000148261
PMid:18667841

 

49 Wu ZP, Gao CW, Wu YG, Zhu QS, Yan Chen, Xin Liu, Chuen Liu. Inhibitive effect of artemether on tumor growth and angiogenesis in the rat C6 orthotopic brain gliomas model. Integr Cancer Ther 2009;8(1):88-92.
https://doi.org/10.1177/1534735408330714
PMid:19174507

 

50 Zhao C, Liu Q, Wang K. Artesunate attenuates ACLT-induced osteoarthritis by suppressing osteoclastogenesis and aberrant angiogenesis. Biomed Pharmacother 2017;96:410-416.
https://doi.org/10.1016/j.biopha.2017.10.018
PMid:29031199

 

51 Abd El-Aal NF, Hamza RS, Magdy M. Anti-angiogenic and anti-lymphangiogenic role of praziquantel and artemether in experimental mansoniasis. Acta Parasitol 2017;62(4):708-716.
https://doi.org/10.1515/ap-2017-0085
PMid:29035850

 

52 Im E, Yeo C, Lee HJ, Lee EO. Dihydroartemisinin induced caspase-dependent apoptosis through inhibiting the specificity protein 1 pathway in hepatocellular carcinoma SK-Hep-1 cells. Life Sci 2018;192:286-292.
https://doi.org/10.1016/j.lfs.2017.11.008
PMid:29128513

 

53 Mungun HK, Li S, Zhang Y, Huang S, Jia Z, Ding G, Zhang A. Dihydroartemisinin inhibits indoxyl sulfate (IS)-promoted cell cycle progression in mesangial cells by targeting COX-2/mPGES-1/PGE2 cascade. Am J Transl Res 2018;10(2):422-431.

 

54 Steely AM, Willoughby JA Sr, Sundar SN, Aivaliotis VI, Firestone GL. Artemisinin disrupts androgen responsiveness of human prostate cancer cells by stimulating the 26S proteasome-mediated degradation of the androgen receptor protein. Anticancer Drugs 2017;28(9):1018-1031.
https://doi.org/10.1097/CAD.0000000000000547
PMid:28708672

 

55 Willoughby JA Sr, Sundar SN, Cheung M, Tin AS, Modiano J, Firestone GL. Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. J Biol Chem 2009;284(4):2203-2213.
https://doi.org/10.1074/jbc.M804491200
PMid:19017637 PMCid:PMC2629082

 

56 Wu J, Hu D, Yang G, Zhou J, Yang C, Gao Y, Zhu Z. Down-regulation of BMI-1 cooperates with artemisinin on growth inhibition of nasopharyngeal carcinoma cells. J Cell Biochem 2011;112(7):1938-1948.
https://doi.org/10.1002/jcb.23114
PMid:21445878

 

57 Shirahama S, Kaburaki T, Nakahara H, Tanaka R, Takamoto M, Fujino Y, Kawashima H, Aihara M. Epidemiology of uveitis (2013-2015) and changes in the patterns of uveitis (2004-2015) in the central Tokyo area: a retrospective study. BMC Ophthalmol 2018;18(1):189.
https://doi.org/10.1186/s12886-018-0871-6
PMid:30068311 PMCid:PMC6090933

 

58 Li B, Zhang R, Li J, Zhang L, Ding G, Luo P, He S, Dong Y, Jiang W, Lu Y, Cao H, Zheng J, Zhou H. Antimalarial artesunate protects sepsis model mice against heat-killed Escherichia coli challenge by decreasing TLR4, TLR9 mRNA expressions and transcription factor NF-kappa B activation. Int Immunopharmacol 2008;8(3):379-389.
https://doi.org/10.1016/j.intimp.2007.10.024
PMid:18279792

 

59 Xu H, He Y, Yang X, Liang L, Zhan Z, Ye Y, Yang X, Lian F, Sun L. Anti-malarial agent artesunate inhibits TNF-alpha-induced production of proinflammatory cytokines via inhibition of NF-kappaB and PI3 kinase/Akt signal pathway in human rheumatoid arthritis fibroblast-like synoviocytes. Rheumatology (Oxford) 2007;46(6):920-926.
https://doi.org/10.1093/rheumatology/kem014
PMid:17314215

 

60 Wang XQ, Liu HL, Wang GB, Wu PF, Yan T, Xie J, Tang Y, Sun LK, Li C. Effect of artesunate on endotoxin-induced uveitis in rats. Invest Ophthalmol Vis Sci 2011;52(2):916-919.
https://doi.org/10.1167/iovs.10-5892
PMid:20881305

 

61 Sakamoto K, Inukai M, Mori A, Nakahara T. Brilliant Blue G protects against photoreceptor injury in a murine endotoxin-induced uveitis model. Exp Eye Res 2018;177:45-49.
https://doi.org/10.1016/j.exer.2018.07.028
PMid:30063882

 

62 Fröhlich T, Reiter C, Ibrahim MM, Beutel J, Hutterer C, Zeitträger I, Bahsi H, Leidenberger M, Friedrich O, Kappes B, Efferth T, Marschall M, Tsogoeva SB. Synthesis of novel hybrids of quinazoline and artemisinin with high activities against Plasmodium falciparum, human cytomegalovirus, and leukemia cells. ACS Omega 2017;2(6):2422-2431.
https://doi.org/10.1021/acsomega.7b00310
PMid:30023664 PMCid:PMC6044832

 

63 Breuer E, Efferth T. Treatment of iron-loaded veterinary sarcoma by Artemisia annua. Nat Prod Bioprospect 2014;4(2):113-118.
https://doi.org/10.1007/s13659-014-0013-7
PMid:24859473 PMCid:PMC4004853

 

64 Li X, Ba Q, Liu Y, Yue Q, Chen P, Li J, Zhang H, Ying H, Ding Q, Song H, Liu H, Zhang R, Wang H. Dihydroartemisinin selectively inhibits PDGFRα-positive ovarian cancer growth and metastasis through inducing degradation of PDGFRα protein. Cell Discov 2017;3:17042.
https://doi.org/10.1038/celldisc.2017.42
PMid:29387451 PMCid:PMC5787695

 

65 Kumari K, Keshari S, Sengupta D, Sabat SC, Mishra SK. Transcriptome analysis of genes associated with breast cancer cell motility in response to Artemisinin treatment. BMC Cancer 2017;17(1):858.
https://doi.org/10.1186/s12885-017-3863-7
PMid:29246124 PMCid:PMC5732364

 

66 Zhang T, Hu Y, Wang T, Cai P. Dihydroartemisinin inhibits the viability of cervical cancer cells by upregulating caveolin 1 and mitochondrial carrier homolog 2: Involvement of p53 activation and NAD(P)H: quinone oxidoreductase 1 downregulation. Int J Mol Med 2017;40(1):21-30.
https://doi.org/10.3892/ijmm.2017.2980
PMid:28498397 PMCid:PMC5466377

 

67 Kogachi K, Kim JW, Green S, Jubran R, Berry JL. Lurking below: massive choroidal invasion under a calcified tumor after attempted conservative therapy for retinoblastoma. Ophthalmic Genet 2018;39(5): 653-657.
https://doi.org/10.1080/13816810.2018.1513535
PMid:30142285 PMCid:PMC6613565

 

68 Zhao F, Wang H, Kunda P, Chen X, Liu QL, Liu T. Artesunate exerts specific cytotoxicity in retinoblastoma cells via CD71. Oncol Rep 2013;30(3):1473-1482.
https://doi.org/10.3892/or.2013.2574
PMid:23818062

 

69 Arroba AI, Campos-Caro A, Aguilar-Diosdado M, Valverde ÁM. IGF-1, inflammation and retinal degeneration: a close network. Front Aging Neurosci 2018;10:203.
https://doi.org/10.3389/fnagi.2018.00203
PMid:30026694 PMCid:PMC6041402

 

70 Zeng Z, Xu J, Zheng W. Artemisinin protects PC12 cells against β-amyloid-induced apoptosis through activation of the ERK1/2 signaling pathway. Redox Biol 2017;12:625-633.
https://doi.org/10.1016/j.redox.2017.04.003
PMid:28391183 PMCid:PMC5385605

 

71 Chong CM, Zheng W. Artemisinin protects human retinal pigment epithelial cells from hydrogen peroxide-induced oxidative damage through activation of ERK/CREB signaling. Redox Biol 2016;9:50-56.
https://doi.org/10.1016/j.redox.2016.06.002
PMid:27372058 PMCid:PMC4939375

 

72 Yan F, Wang H, Gao Y, Xu J, Zheng W. Artemisinin protects retinal neuronal cells against oxidative stress and restores rat retinal physiological function from light exposed damage. ACS Chem Neurosci 2017;8(8):1713-1723.
https://doi.org/10.1021/acschemneuro.7b00021
PMid:28447781

 

73 Mesquita J, Castro-de-Sousa JP, Vaz-Pereira S, Neves A, Passarinha LA, Tomaz CT. Evaluation of the growth factors VEGF-a and VEGF-B in the vitreous and serum of patients with macular and retinal vascular diseases. Growth Factors 2018;36(1-2):48-57.
https://doi.org/10.1080/08977194.2018.1477140
PMid:29969324

 

74 Lekha T, Prasad HN, Sarwate RN, Patel M, Karthikeyan S. Intravitreal bevacizumab for choroidal neovascularization associated with angioid streaks: long-term results. Middle East Afr J Ophthalmol 2017;24(3):136-142.
https://doi.org/10.4103/meajo.MEAJO_17_17
PMid:29279654 PMCid:PMC5698988

 

75 Moreno M, Pow PY, Tabitha TST, Nirmal S, Larsson A, Radhakrishnan K, Nirmal J, Quah ST, Geifman Shochat S, Agrawal R, Venkatraman S. Modulating release of ranibizumab and aflibercept from thiolated chitosan-based hydrogels for potential treatment of ocular neovascularization. Expert Opin Drug Deliv 2017;14(8):913-925.
https://doi.org/10.1080/17425247.2017.1343297
PMid:28643528

 

76 El Alaoui-Lasmaili K, Faivre B. Antiangiogenic therapy: markers of response, "normalization" and resistance. Crit Rev Oncol Hematol 2018;128:118-129.
https://doi.org/10.1016/j.critrevonc.2018.06.001
PMid:29958627

 

77 Borkar DS, Obeid A, Su DC, Storey PP, Gao X, Regillo CD, Kaiser RS, Garg SJ, Hsu J; Wills Post Injection Endophthalmitis (PIE) Study Group. Endophthalmitis rates after bilateral same-day intravitreal anti-vascular endothelial growth factor injections. Am J Ophthalmol 2018;194:1-6.
https://doi.org/10.1016/j.ajo.2018.06.022
PMid:29981738

 

78 Kumar M, Dhatwalia SK, Dhawan DK. Role of angiogenic factors of herbal origin in regulation of molecular pathways that control tumor angiogenesis. Tumour Biol 2016;37(11):14341-14354.
https://doi.org/10.1007/s13277-016-5330-5
PMid:27614685

 

79 Verma S, Das P, Kumar VL. Chemoprevention by artesunate in a preclinical model of colorectal cancer involves down regulation of β-catenin, suppression of angiogenesis, cellular proliferation and induction of apoptosis. Chem Biol Interact 2017;278:84-91.
https://doi.org/10.1016/j.cbi.2017.10.011
PMid:29031619

 

80 Zhou HJ, Wang WQ, Wu GD, Lee J, Li A. Artesunate inhibits angiogenesis and downregulates vascular endothelial growth factor expression in chronic myeloid leukemia K562 cells. Vascul Pharmacol 2007;47(2-3):131-138.
https://doi.org/10.1016/j.vph.2007.05.002
PMid:17581794

 

81 Chen HH, Zhou HJ, Wu GD, Lou XE. Inhibitory effects of artesunate on angiogenesis and on expressions of vascular endothelial growth factor and VEGF receptor KDR/flk-1. Pharmacology 2004;71(1):1-9.
https://doi.org/10.1159/000076256
PMid:15051917

 

82 Chen HH, Zhou HJ, Wang WQ, Wu GD. Antimalarial dihydroartemisinin also inhibits angiogenesis. Cancer Chemother Pharmacol 2004;53(5): 423-432.
https://doi.org/10.1007/s00280-003-0751-4
PMid:15132130

 

83 Cheng R, Li C, Li C, Wei L, Li L, Zhang Y, Yao Y, Gu X, Cai W, Yang Z, Ma J, Yang X, Gao G. The artemisinin derivative artesunate inhibits corneal neovascularization by inducing ROS-dependent apoptosis in vascular en dothelial cells. Invest Ophthalmol Vis Sci 2013;54(5): 3400-3409.
https://doi.org/10.1167/iovs.12-11068
PMid:23611999 PMCid:PMC5963000

 

84 Zong Y, Yuan Y, Qian X, Huang Z, Yang W, Lin L, Zheng Q, Li Y, He H, Gao Q. Small molecular-sized artesunate attenuates ocular neovascularization via VEGFR2, PKCα, and PDGFR targets. Sci Rep 2016;6:30843.
https://doi.org/10.1038/srep30843
PMid:27480521 PMCid:PMC4969591