K. S. Bora and A. Sharma, The Genus Artemisia: A Comprehensive Review, Pharm. Biol, vol.49, pp.101-109, 2011.

M. Sanz, R. Vilatersana, O. Hidalgo, N. Garcia-jacas, A. Susanna et al., Molecular Phylogeny and Evolution of Floral Characters of Artemisia and Allies (Anthemideae, Asteraceae): Evidence from NrDNA ETS and ITS Sequences, TAXON, vol.57, pp.66-78, 2008.

S. Alesaeidi and S. Miraj, A Systematic Review of Anti-Malarial Properties, Immunosuppressive Properties, Anti-Inflammatory Properties, and Anti-Cancer Properties of Artemisia Annua, Electron. Physician, vol.8, pp.3150-3155, 2016.

V. A. Funk, R. J. Bayer, S. Keeley, R. Chan, L. Watson et al., Everywhere but Antarctica: Using a Supertree to Understand the Diversity and Distribution of the Compositae, Biol. Skr, vol.55, pp.343-374, 2005.

J. Vallès, S. Garcia, O. Hidalgo, J. Martín, J. Pellicer et al., Genome Evolution, Biotechnological Issues and Research Including Applied Perspectives in

, Adv. Bot. Res, 2011.

C. E. Jarvis, A List of Linnaean Generic Names and Their Types; Koeltz Sciebtific Books; International Association for Plant Taxonomy: Bratislava, vol.127, p.100, 1993.

M. Willcox, Artemisia Species: From Traditional Medicines to Modern Antimalarials-and Back Again, J. Altern. Complement. Med, vol.15, pp.101-109, 2009.

, Int. J. Mol. Sci, vol.2020, p.4986

Y. S. Ko, W. S. Lee, R. Panchanathan, Y. N. Joo, Y. H. Choi et al., Polyphenols from Artemisia Annua L Inhibit Adhesion and EMT of Highly Metastatic Breast Cancer Cells MDA-MB-231, Phytother. Res, vol.30, pp.1180-1188, 2016.

L. Annua, La Maison de l'Artemisia-Cette Plante Peut Sauver des Millions de Vie, 2020.

E. Hsu, The History of Qing Hao in the Chinese Materia Medica, Trans. R. Soc. Trop. Med. Hyg, vol.100, pp.505-508, 2006.

S. ?avar, M. Maksimovi?, D. Vidic, and A. Pari?, Chemical Composition and Antioxidant and Antimicrobial Activity of Essential Oil of Artemisia Annua L. from Bosnia, Ind. Crop. Prod, vol.37, pp.479-485, 2012.

M. S. Mueller, I. B. Karhagomba, H. M. Hirt, and E. Wemakor, The Potential of Artemisia Annua L. as a Locally Produced Remedy for Malaria in the Tropics: Agricultural, Chemical and Clinical Aspects, J. Ethnopharmacol, vol.73, pp.487-493, 2000.

P. C. Gupta, B. Dutta, D. Pant, P. Joshi, and D. R. Lohar, In Vitro Antibacterial Activity of Artemisia Annua Linn. Growing in India. Int. J. Green Pharm, vol.3, 2009.

M. J. Abad, L. M. Bedoya, L. Apaza, P. T. Bermejo, and L. Artemisia, Genus: A Review of Bioactive Essential Oils. Molecules, vol.17, pp.2542-2566, 2012.

D. Wang, L. Cui, X. Chang, and D. Guan, Biosynthesis and Characterization of Zinc Oxide Nanoparticles from Artemisia Annua and Investigate Their Effect on Proliferation, Osteogenic Differentiation and Mineralization in Human Osteoblast-like MG-63 Cells, J. Photochem. Photobiol, vol.2020, p.111652

A. Lubbe, I. Seibert, T. Klimkait, and F. Van-der-kooy, Ethnopharmacology in Overdrive: The Remarkable Anti-HIV Activity of Artemisia Annua, J. Ethnopharmacol, vol.141, pp.854-859, 2012.

W. E. Ho, H. Y. Peh, T. K. Chan, and W. S. Wong, Artemisinins: Pharmacological Actions beyond Anti-Malarial, Pharmacol. Ther, vol.142, pp.126-139, 2014.

M. H. Kim, J. Y. Seo, K. H. Liu, and J. Kim, Protective Effect of Artemisia Annua L. Extract against Galactose-Induced Oxidative Stress in Mice, PLoS ONE, vol.9, 2014.

Y. Wang, J. Chen, D. Zhang, Y. Zhang, Y. Wen et al., Tumoricidal Effects of a Selenium (Se)-Polysaccharide from Ziyang Green Tea on Human Osteosarcoma U-2 OS Cells, Carbohydr. Polym, vol.98, pp.1186-1190, 2013.

P. C. Castilho, S. C. Gouveia, and A. I. Rodrigues, Quantification of Artemisinin in Artemisia Annua Extracts by 1H-NMR, Phytochem. Anal, vol.19, pp.329-334, 2008.

V. Chaudhary, R. Kapoor, and A. K. Bhatnagar, Effectiveness of Two Arbuscular Mycorrhizal Fungi on Concentrations of Essential Oil and Artemisinin in Three Accessions of, Artemisia Annua L. Appl. Soil Ecol, vol.40, pp.174-181, 2008.

S. Slezakova and J. Ruda-kucerova, Anticancer Activity of Artemisinin and Its Derivatives, Anticancer Res, vol.37, pp.5995-6003, 2017.

E. G. Tse, M. Korsik, and M. H. Todd, The Past, Present and Future of Anti-Malarial Medicines, Malar. J, vol.18, 2019.

E. A. Brisibe, U. E. Umoren, F. Brisibe, P. M. Magalhäes, J. F. Ferreira et al., Nutritional Characterisation and Antioxidant Capacity of Different Tissues of Artemisia Annua L, Food Chem, vol.115, pp.1240-1246, 2009.

F. Van-der-kooy and S. E. Sullivan, The Complexity of Medicinal Plants: The Traditional Artemisia Annua Formulation, Current Status and Future Perspectives, J. Ethnopharmacol, vol.150, 2013.

R. S. Bhakuni, D. C. Jain, R. P. Sharma, and S. Kumar, Secondary Metabolites of Artemisia Annua and Their Biological Activity, Curr. Sci, vol.80, pp.35-48, 2001.

K. Li, X. Dong, Y. Ma, Z. Wu, Y. Yan et al., Antifungal Coumarins and Lignans from Artemisia Annua, Fitoterapia, vol.134, pp.323-328, 2019.

Z. Yw, N. Fy, S. Yl, W. Sy, H. Wz et al., Chemical Constituents from Artemisia Annua, Zhongguo Zhong Yao Za Zhi, vol.39, pp.4816-4821, 2014.

H. El-askary, H. Handoussa, F. Badria, A. H. El-khatib, A. Alsayari et al., Abdel Motaal, A. Characterization of Hepatoprotective Metabolites from Artemisia Annua and Cleome Droserifolia Using HPLC/PDA/ESI/MS-MS. Rev. Bras. De Farmacogn, vol.29, pp.213-220, 2019.

, Int. J. Mol. Sci, vol.2020, pp.4986-5008

S. J. Lang, M. Schmiech, S. Hafner, C. Paetz, C. Steinborn et al., Antitumor Activity of an Artemisia Annua Herbal Preparation and Identification of Active Ingredients, Phytomedicine, vol.62, p.152962, 2019.

X. L. Wan, Y. Niu, X. C. Zheng, Q. Huang, W. P. Su et al., Antioxidant Capacities of Artemisia Annua L. Leaves and Enzymatically Treated Artemisia Annua L. in Vitro and in Broilers, Anim. Feed Sci. Technol, vol.221, pp.27-34, 2016.

Y. Song, K. T. Desta, G. Kim, S. J. Lee, W. S. Lee et al., Polyphenolic Profile and Antioxidant Effects of Various Parts of Artemisia Annua L, Biomed. Chromatogr, vol.30, pp.588-595, 2016.

Y. Li, Y. Guo, Q. Yang, X. Weng, L. Yang et al., Flavonoids Casticin and Chrysosplenol D from Artemisia Annua L. Inhibit Inflammation in Vitro and in Vivo, Toxicol. Appl. Pharmacol, vol.286, pp.151-158, 2015.

J. Wang, Casticin Alleviates Lipopolysaccharide-Induced Inflammatory Responses and Expression of Mucus and Extracellular Matrix in Human Airway Epithelial Cells through Nrf2/Keap1 and NF-KB Pathways, Phytother. Res, vol.32, pp.1346-1353, 2018.

N. Shin, H. Ryu, J. Ko, S. Park, H. Yuk et al., Artemisia Argyi Attenuates Airway Inflammation in Ovalbumin-Induced Asthmatic Animals. J. Ethnopharmacol, vol.209, pp.108-115, 2017.

F. Qiu, S. Wu, X. Lu, C. Zhang, J. Li et al., Quality Evaluation of the Artemisinin-Producing Plant Artemisia Annua L. Based on Simultaneous Quantification of Artemisinin and Six Synergistic Components and Hierarchical Cluster Analysis, Ind. Crop. Prod, vol.118, pp.131-141, 2018.

X. Zhang, L. Guo, Z. Qiu, X. Qu, H. Wang et al.,

, , vol.42, pp.4277-4281, 2017.

X. Zhang, Y. Zhao, X. Huang, Z. Qiu, L. Guo et al.,

, Zhongguo Zhong Yao Za Zhi, vol.41, 2015.

S. C. Gouveia, P. C. Castilho, and L. Artemisia-annua, Essential Oil and Acetone Extract Composition and Antioxidant Capacity, Ind. Crop. Prod, vol.45, pp.170-181, 2013.

C. Marlet and G. Lognay, Les monoterpènes: Sources et implications dans la qualité de l'air intérieur, Biotechnol. Agron. Soc. Environ, vol.15, pp.611-622, 2011.

P. Jana?kovi?, N. Raj?evi?, M. Gavrilovi?, J. Novakovi?, A. Giweli et al., Essential Oil Composition of Five Artemisia (Compositae) Species in Regards to Chemophenetics, Biochem. Syst. Ecol, vol.87, 2019.

P. M. De-magalhães, B. Pereira, and A. Sartoratto, Yields of antimalarial artemisia Annua L. species, Acta Hortic, vol.629, pp.421-424, 2004.

K. Durden, S. Sellars, B. Cowell, J. J. Brown, and M. A. Pszczolkowski, Artemisia Annua Extracts, Artemisinin and 1,8-Cineole, Prevent Fruit Infestation by a Major, Cosmopolitan Pest of Apples, Pharm. Biol, vol.49, pp.563-568, 2011.

F. A. Santos and V. S. Rao, Antiinflammatory and Antinociceptive Effects of 1,8-Cineole a Terpenoid Oxide Present in Many Plant Essential Oils, Phytother. Res, vol.14, pp.240-244, 2000.

G. R. Vilela, G. S. De-almeida, M. A. ;-d'arce, M. H. Moraes, J. O. Brito et al., Activity of Essential Oil and Its Major Compound, 1,8-Cineole, from Eucalyptus Globulus Labill., against the Storage Fungi Aspergillus Flavus Link and Aspergillus Parasiticus Speare, J. Stored Prod. Res, vol.45, pp.108-111, 2009.

S. Murata, R. Shiragami, C. Kosugi, T. Tezuka, M. Yamazaki et al., Antitumor Effect of 1, 8-Cineole against Colon Cancer, Oncol. Rep, vol.30, pp.2647-2652, 2013.

F. R. De-figuêiredo, Á. B. Monteiro, I. R. De-menezes, V. S. Sales, E. Do-nascimento et al., Eucalyptol) on the Central Nervous System of Mice. Food Chem. Toxicol, vol.1, p.110802, 2019.

, Int. J. Mol. Sci, vol.2020, pp.4986-5009

S. Sampath, S. Subramani, S. Janardhanam, P. Subramani, A. Yuvaraj et al., Bioactive Compound 1,8-Cineole Selectively Induces G2/M Arrest in A431 Cells through the Upregulation of the P53 Signaling Pathway and Molecular Docking Studies, Phytomedicine, vol.46, pp.57-68, 2018.

B. Rodenak-kladniew, A. Castro, P. Stärkel, M. Galle, and R. Crespo, 1,8-Cineole Promotes G0/G1 Cell Cycle Arrest and Oxidative Stress-Induced Senescence in HepG2 Cells and Sensitizes Cells to Anti-Senescence Drugs, Life Sci, vol.243, 2020.

H. Yang, J. Woo, A. N. Pae, M. Y. Um, N. Cho et al., Enhances Non-Rapid Eye Movement Sleep in Mice through GABAA-Benzodiazepine Receptors, Mol. Pharm, vol.90, pp.530-539, 2016.

M. R. Albuquerque, S. M. Costa, P. N. Bandeira, G. M. Santiago, M. Andrade-neto et al., Nematicidal and Larvicidal Activities of the Essential Oils from Aerial Parts of Pectis Oligocephala and Pectis Apodocephala Baker. An. Da Acad. Bras. De Ciências, vol.79, pp.209-213, 2007.

A. C. Rivas-da-silva, P. M. Lopes, M. M. Barros-de-azevedo, D. C. Costa, C. S. Alviano et al., Biological Activities of ?-Pinene and ?-Pinene Enantiomers, Molecules, vol.17, pp.6305-6316, 2012.

M. Kim, K. Sowndhararajan, S. J. Park, and S. Kim, Effect of Inhalation of Isomers, (+)-?-Pinene and (+)-?-Pinene on Human Electroencephalographic Activity According to Gender Difference, Eur. J. Integr. Med, vol.17, pp.33-39, 2018.

G. Benelli, M. Govindarajan, M. Rajeswary, B. Vaseeharan, S. A. Alyahya et al., Insecticidal Activity of Camphene, Zerumbone and ?-Humulene from Cheilocostus Speciosus Rhizome Essential Oil against the Old-World Bollworm, Helicoverpa Armigera, Ecotoxicol. Environ. Saf, vol.148, pp.781-786, 2018.

M. Okuniewski, K. Paduszy?ski, and U. Doma?ska, Thermodynamic Study of Molecular Interactions in Eutectic Mixtures Containing Camphene, J. Phys. Chem. B, vol.120, pp.12928-12936, 2016.

S. Wang, D. Zhang, J. Hu, Q. Jia, W. Xu et al., A Clinical and Mechanistic Study of Topical Borneol-induced Analgesia, Embo Mol. Med, vol.9, pp.802-815, 2017.

Y. Wang, A. Ryu, S. Jin, Y. Jeon, and M. Lee, Chlorin E6-Mediated Photodynamic Therapy Suppresses P. Acnes-Induced Inflammatory Response via NF?B and MAPKs Signaling Pathway, PLoS ONE, vol.12, 2017.

J. Ji, R. Zhang, H. Li, J. Zhu, Y. Pan et al., Analgesic and Anti-Inflammatory Effects and Mechanism of Action of Borneol on Photodynamic Therapy of Acne, Environ. Toxicol. Pharmacol, vol.75, 2020.

Y. Ge, Z. Wang, Y. Xiong, X. Huang, Z. Mei et al., Anti-Inflammatory and Blood Stasis Activities of Essential Oil Extracted from Artemisia Argyi Leaf in Animals, J. Nat. Med, vol.70, pp.531-538, 2016.

X. Chen, Z. Lin, A. Liu, J. Ye, Y. Luo et al., The Orally Combined Neuroprotective Effects of Sodium Ferulate and Borneol against Transient Global Ischaemia in C57 BL/6J

, J. Pharm. Pharmacol, vol.62, pp.915-923, 2010.

D. Ho, C. Lau, K. Ng, Y. Kong, K. Cheng et al., Anti-Implantation Activity of S(?)-and R(+)-Camphor-Yuehchukene in Rats, Eur. J. Pharmacol, vol.205, pp.209-212, 1991.

P. C. Ng, D. D. Ho, K. H. Ng, Y. C. Kong, K. F. Cheng et al., Mixed Estrogenic and Anti-Estrogenic Activities of Yuehchukene-a Bis-Indole Alkaloid, Eur. J. Pharmacol, vol.264, pp.1-12, 1994.

N. Chatterjie and G. J. Alexander, Anticonvulsant Properties of Spirohydantoins Derived from Optical Isomers of Camphor, Neurochem. Res, vol.11, pp.1669-1676, 1986.

E. A. Laude, A. H. Morice, and T. J. Grattan, The Antitussive Effects of Menthol, Camphor and Cineole in Conscious Guinea-Pigs, Pulm. Pharmacol, vol.7, pp.179-184, 1994.

H. Tinwell, A. Lefevre-paul, J. Moffat-graeme, A. Burns, J. Odum et al., Confirmation of Uterotrophic Activity of 3-(4-Methylbenzylidine)Camphor in the Immature Rat, Environ. Health Perspect, vol.110, pp.533-536, 2002.

T. J. Park, H. K. Seo, B. J. Kang, and K. T. Kim, Noncompetitive Inhibition by Camphor of Nicotinic Acetylcholine Receptors, Biochem. Pharmacol, vol.61, pp.787-793, 2001.

M. Schlumpf, B. Cotton, M. Conscience, V. Haller, B. Steinmann et al., In Vitro and in Vivo Estrogenicity of UV Screens, Environ. Health Perspect, vol.109, pp.239-244, 2001.

, Int. J. Mol. Sci, vol.2020, pp.4986-5010

H. Holbech, U. Nørum, B. Korsgaard, and B. Poul, The Chemical UV-Filter 3-Benzylidene Camphor Causes an Oestrogenic Effect in an in Vivo Fish Assay, Pharmacol. Toxicol, vol.91, pp.204-208, 2002.

S. O. Mueller, M. Kling, P. Arifin-firzani, A. Mecky, E. Duranti et al., Activation of Estrogen Receptor ? and ER? by 4-Methylbenzylidene-Camphor in Human and Rat Cells: Comparison with Phyto-and Xenoestrogens, Toxicol. Lett, vol.142, pp.89-101, 2003.

N. Arakaki, Y. Shimoji, S. Wakamura, and . Camphor, An Attractant for the Cupreous Polished Chafer, Protaetia Pryeri Pryeri (Janson) (Coleoptera: Scarabaeidae), Appl. Entomol. Zool, vol.44, pp.621-625, 2009.

M. Zhao and J. Du, Anti-Inflammatory and Protective Effects of D-Carvone on Lipopolysaccharide (LPS)-Induced Acute Lung Injury in Mice, J. King Saud Univ.-Sci, vol.2020, pp.1592-1596

R. Vinothkumar, M. Sudha, P. Viswanathan, J. Kabalimoorthy, T. Balasubramanian et al., Modulating Effect of D-Carvone on 1,2-Dimethylhydrazine-Induced Pre-Neoplastic Lesions, Oxidative Stress and Biotransforming Enzymes, in an Experimental Model of Rat Colon Carcinogenesis, Cell Prolif, vol.46, pp.705-720, 2013.

I. J. Moro, G. D. Gondo, E. G. Pierri, R. C. Pietro, C. P. Soares et al., Evaluation of Antimicrobial, Cytotoxic and Chemopreventive Activities of Carvone and Its Derivatives. Braz, J. Pharm. Sci, vol.53, 2017.

R. De-cássia-da-silveira-e-sá, L. N. Andrade, and D. P. De-sousa, A Review on Anti-Inflammatory Activity of Monoterpenes. Molecules, vol.18, pp.1227-1254, 2013.

M. C. Bier, A. B. Medeiros, N. De-kimpe, and C. R. Soccol, Evaluation of Antioxidant Activity of the Fermented Product from the Biotransformation of R-(+)-Limonene in Solid-State Fermentation of Orange Waste by Diaporthe Sp, Biotechnol. Res. Innov, vol.3, pp.168-176, 2019.

M. Bacanl?, A. A. Ba?aran, and N. Ba?aran, The Antioxidant and Antigenotoxic Properties of Citrus Phenolics Limonene and Naringin, Food Chem. Toxicol, vol.81, pp.160-170, 2015.

K. N. Chidambara-murthy, G. K. Jayaprakasha, B. S. Patil, and . D-limonene, Rich Volatile Oil from Blood Oranges Inhibits Angiogenesis, Metastasis and Cell Death in Human Colon Cancer Cells, Life Sci, vol.91, pp.429-439, 2012.

P. R. Quiroga, V. Nepote, and M. T. Baumgartner, Contribution of Organic Acids to ?-Terpinene Antioxidant Activity, Food Chem, vol.277, pp.267-272, 2019.

M. A. Bejeshk, M. Samareh-fekri, H. Najafipour, F. Rostamzadeh, E. Jafari et al., Anti-Inflammatory and Anti-Remodeling Effects of Myrtenol in the Lungs of Asthmatic Rats: Histopathological and Biochemical Findings, Allergol. Et Immunopathol, vol.47, pp.185-193, 2019.

A. Sepici, I. Gürbüz, C. Çevik, and E. Yesilada, Hypoglycaemic Effects of Myrtle Oil in Normal and Alloxan-Diabetic Rabbits, J. Ethnopharmacol, vol.93, pp.311-318, 2004.

V. Aleksic and P. Knezevic, Antimicrobial and Antioxidative Activity of Extracts and Essential Oils of Myrtus Communis L, Microbiol. Res, vol.169, pp.240-254, 2014.

A. M. Clark, Natural Products as a Resource for New Drugs, Pharm. Res, vol.13, pp.1133-1144, 1996.

C. Fu, P. Yu, M. Wang, and F. Qiu, Phytochemical Analysis and Geographic Assessment of Flavonoids, Coumarins and Sesquiterpenes in Artemisia Annua L. Based on HPLC-DAD Quantification and LC-ESI-QTOF-MS/MS Confirmation, Food Chem, p.312, 2020.

Z. Dandan and Z. Jianjiang, Two Cytotoxic Sesquiterpenes from Hairy Root Cultures of Artemisia Annua L. Induced Apoptosis of Highly Metastatic Lung Carcinoma Cell Line 95-D, J. Biosci. Bioeng, vol.108, pp.24-25, 2009.

Y. Li, J. Wu, F. Shan, G. Wu, J. Ding et al., Synthesis and Cytotoxicity of Dihydroartemisinin Ethers Containing Cyanoarylmethyl Group, Bioorg. Med. Chem, vol.11, pp.977-984, 2003.

W. Nam, J. Tak, J. Ryu, M. Jung, J. Yook et al., Effects of Artemisinin and Its Derivatives on Growth Inhibition and Apoptosis of Oral Cancer Cells, Head Neck, vol.29, pp.335-340, 2007.

D. Zhai, K. Supaibulwatana, and J. Zhong, Inhibition of Tumor Cell Proliferation and Induction of Apoptosis in Human Lung Carcinoma 95-D Cells by a New Sesquiterpene from Hairy Root Cultures of Artemisia Annua, Phytomedicine, vol.17, pp.856-861, 2010.

, Int. J. Mol. Sci, vol.2020, pp.4986-5011

X. Wang, B. Zheng, U. Ashraf, H. Zhang, C. Cao et al., Artemisinin Inhibits the Replication of Flaviviruses by Promoting the Type I Interferon Production, Antivir. Res, 2020.

Y. K. Wong, C. Xu, K. A. Kalesh, Y. He, Q. Lin et al., Artemisinin as an Anticancer Drug: Recent Advances in Target Profiling and Mechanisms of Action, Med. Res. Rev, vol.37, pp.1492-1517, 2017.

A. O. Idowu, S. Bhattacharyya, S. Gradus, W. Oyibo, Z. George et al., Plasmodium Falciparum Treated with Artemisinin-Based Combined Therapy Exhibits Enhanced Mutation, Heightened Cortisol and TNF-? Induction, Int. J. Med. Sci, vol.15, pp.1449-1457, 2018.

Y. Wang, Y. Wang, F. You, and J. Xue, Novel Use for Old Drugs: The Emerging Role of Artemisinin and Its Derivatives in Fibrosis, Pharmacol. Res, 2020.

E. Martino, M. Tarantino, M. Bergamini, V. Castelluccio, A. Coricello et al., Artemisinin and Its Derivatives; Ancient Tradition Inspiring the Latest Therapeutic Approaches against Malaria, Future Med. Chem, vol.11, pp.1443-1459, 2019.

J. O. Suberu, A. P. Gorka, L. Jacobs, P. D. Roepe, N. Sullivan et al., Anti-Plasmodial Polyvalent Interactions in Artemisia Annua L. Aqueous Extract-Possible Synergistic and Resistance Mechanisms, PLoS ONE, vol.8, 2013.

T. Efferth, F. Herrmann, A. Tahrani, and M. Wink, Cytotoxic Activity of Secondary Metabolites Derived from Artemisia Annua L. towards Cancer Cells in Comparison to Its Designated Active Constituent Artemisinin, Phytomedicine, vol.18, pp.959-969, 2011.

X. X. Zhu, L. Yang, Y. J. Li, D. Zhang, Y. Chen et al., Effects of Sesquiterpene, Flavonoid and Coumarin Types of Compounds from Artemisia Annua L. on Production of Mediators of Angiogenesis, Pharmacol. Rep, vol.65, pp.410-420, 2013.

G. Sharma, H. Kapoor, M. Chopra, K. Kumar, and V. Agrawal, Strong Larvicidal Potential of Artemisia Annua Leaf Extract against Malaria (Anopheles Stephensi Liston) and Dengue (Aedes Aegypti L.) Vectors and Bioassay-Driven Isolation of the Marker Compounds, Parasitol. Res, vol.113, pp.197-209, 2014.

A. Flobinus, N. Taudon, M. Desbordes, B. Labrosse, F. Simon et al., Stability and Antiviral Activity against Human Cytomegalovirus of Artemisinin Derivatives, J. Antimicrob. Chemother, vol.69, pp.34-40, 2014.

J. Lee, M. Kim, J. Lee, E. Jung, E. Yoo et al., Artemisinic Acid Is a Regulator of Adipocyte Differentiation and C/EBP ? Expression, J. Cell Biochem, vol.113, pp.2488-2499, 2012.

T. Efferth, From Ancient Herb to Modern Drug: Artemisia Annua and Artemisinin for Cancer Therapy, Semin. Cancer Biol, vol.46, pp.65-83, 2017.

P. J. Weathers, P. R. Arsenault, P. S. Covello, A. Mcmickle, K. H. Teoh et al., Artemisinin Production in Artemisia Annua: Studies in Planta and Results of a Novel Delivery Method for Treating Malaria and Other Neglected Diseases, Phytochem. Rev, vol.10, pp.173-183, 2011.

J. F. Ferreira, D. L. Luthria, T. Sasaki, and A. Heyerick, Flavonoids from Artemisia Annua L. as Antioxidants and Their Potential Synergism with Artemisinin against Malaria and Cancer, Molecules, vol.15, pp.3135-3170, 2010.

J. Han, M. Ye, X. Qiao, M. Xu, B. Wang et al., Characterization of Phenolic Compounds in the Chinese Herbal Drug Artemisia Annua by Liquid Chromatography Coupled to Electrospray Ionization Mass Spectrometry, J. Pharm. Biomed. Anal, vol.47, pp.516-525, 2008.

J. Lai, Y. H. Lim, J. Su, H. Shen, and C. N. Ong, Identification and Characterization of Major Flavonoids and Caffeoylquinic Acids in Three Compositae Plants by LC/DAD-APCI/MS, J. Chromatogr. B, vol.848, pp.215-225, 2007.

I. S. Carvalho, T. Cavaco, and M. Brodelius, Phenolic Composition and Antioxidant Capacity of Six Artemisia Species, Ind. Crop. Prod, vol.33, pp.382-388, 2011.

N. Tsevegsuren, R. Edrada, W. Lin, R. Ebel, C. Torre et al., Biologically Active Natural Products from Mongolian Medicinal Plants Scorzonera Divaricata and Scorzonera Pseudodivaricata, J. Nat. Prod, vol.70, pp.962-967, 2007.

C. Zidorn, B. O. Petersen, V. Udovi?i?, T. O. Larsen, J. Ø. Duus et al., Podospermic Acid, 1,3,5-Tri-O-(7,8-Dihydrocaffeoyl)Quinic Acid from Podospermum Laciniatum (Asteraceae), Tetrahedron Lett, vol.46, pp.1291-1294, 2005.

, Int. J. Mol. Sci, vol.2020, pp.4986-5012

Y. Yang, X. Liu, H. Wu, X. He, Y. Bi et al., Radical Scavenging Activity and Cytotoxicity of Active Quinic Acid Derivatives from Scorzonera Divaricata Roots, Food Chem, vol.138, pp.2057-2063, 2013.

S. E. Yazdi, G. Prinsloo, H. M. Heyman, C. B. Oosthuizen, T. Klimkait et al., Anti-HIV-1 Activity of Quinic Acid Isolated from Helichrysum Mimetes Using NMR-Based Metabolomics and Computational Analysis, S. Afr. J. Bot, vol.126, pp.328-339, 2019.

J. Zhang, R. Yan, N. Yu, X. Zhang, D. Chen et al., A New Caffeic Acid Tetramer from the Dracocephalum Moldavica L, Nat. Prod. Res, vol.32, pp.370-373, 2018.

H. G. Choi, P. T. Tran, J. Lee, B. S. Min, and J. A. Kim, Anti-Inflammatory Activity of Caffeic Acid Derivatives Isolated from the Roots of Salvia Miltiorrhiza Bunge, Arch. Pharm. Res, vol.41, pp.64-70, 2018.

J. Langland, B. Jacobs, C. E. Wagner, G. Ruiz, and T. M. Cahill, Antiviral Activity of Metal Chelates of Caffeic Acid and Similar Compounds towards Herpes Simplex, VSV-Ebola Pseudotyped and Vaccinia Viruses, Antivir. Res, vol.160, pp.143-150, 2018.

M. Mishra, R. Panta, and M. Miyares, Influence of Coffee and Its Components on Breast Cancer: A Review. Asian Pac. J. Trop. Dis, vol.6, pp.827-831, 2016.

S. Habtemariam, Protective Effects of Caffeic Acid and the Alzheimer's Brain, 2020.

S. Adisakwattana, Cinnamic Acid and Its Derivatives: Mechanisms for Prevention and Management of Diabetes and Its Complications, Nutrients, vol.9, 2017.

A. G. Murillo and M. L. Fernandez, The Relevance of Dietary Polyphenols in Cardiovascular Protection, Curr. Pharm. Des, vol.23, pp.2444-2452, 2017.

T. Silva, C. Oliveira, and F. Borges, Caffeic Acid Derivatives, Analogs and Applications: A Patent Review, Expert Opin. Ther. Pat, vol.24, pp.1257-1270, 2009.

Z. Cao, H. Zhang, X. Cai, W. Fang, D. Chai et al., Luteolin Promotes Cell Apoptosis by Inducing Autophagy in Hepatocellular Carcinoma, Cell. Physiol. Biochem, vol.43, pp.1803-1812, 2017.

P. Lin, X. Tian, Y. Yi, W. Jiang, Y. Zhou et al., Luteolin-Induced Protection of H 2 O 2 -Induced Apoptosis in PC12 Cells and the Associated Pathway, Mol. Med. Rep, vol.12, pp.7699-7704, 2015.

C. Nunes, L. Almeida, R. M. Barbosa, and J. Laranjinha, Luteolin Suppresses the JAK/STAT Pathway in a Cellular Model of Intestinal Inflammation, Food Funct, vol.8, pp.387-396, 2017.

Z. Yao, X. Yao, Y. Zhang, S. Zhang, and J. Hu, Luteolin Could Improve Cognitive Dysfunction by Inhibiting Neuroinflammation, Neurochem. Res, vol.43, pp.806-820, 2018.

B. Wei, Q. Lin, Y. Ji, Y. Zhao, L. Ding et al., Luteolin Ameliorates Rat Myocardial Ischaemia-Reperfusion Injury through Activation of Peroxiredoxin II, Br. J. Pharmacol, vol.175, pp.3315-3332, 2018.

C. H. Jang, N. Moon, J. Oh, and J. Kim, Luteolin Shifts Oxaliplatin-Induced Cell Cycle Arrest at G0/G1 to Apoptosis in HCT116 Human Colorectal Carcinoma Cells, Nutrients, vol.11, 2019.

M. Lesjak, I. Beara, N. Simin, D. Pinta?, T. Majki? et al., Antioxidant and Anti-Inflammatory Activities of Quercetin and Its Derivatives, J. Funct. Foods, vol.40, pp.68-75, 2018.

J. Duarte, F. Pérez-vizcaíno, A. Zarzuelo, J. Jiménez, and J. Tamargo, Vasodilator Effects of Quercetin in Isolated Rat Vascular Smooth Muscle, Eur. J. Pharmacol, vol.239, 1993.

F. J. Luna-vázquez, C. Ibarra-alvarado, A. Rojas-molina, I. Rojas-molina, and M. Á. Zavala-sánchez, Vasodilator Compounds Derived from Plants and Their Mechanisms of Action, Molecules, vol.18, pp.5814-5857, 2013.

Y. Li, J. Yao, C. Han, J. Yang, M. T. Chaudhry et al., Inflammation and Immunity. Nutrients, vol.8, p.167, 2016.

D. Kashyap, S. Mittal, K. Sak, P. Singhal, and H. S. Tuli, Molecular Mechanisms of Action of Quercetin in Cancer: Recent Advances. Tumor Biol, vol.37, pp.12927-12939, 2016.

M. Ezzati, B. Yousefi, K. Velaei, and A. Safa, A Review on Anti-Cancer Properties of Quercetin in Breast Cancer, Life Sci, vol.248, 2020.

R. V. Patel, B. M. Mistry, S. K. Shinde, R. Syed, V. Singh et al., Therapeutic Potential of Quercetin as a Cardiovascular Agent, Eur. J. Med. Chem, vol.155, pp.889-904, 2018.

, Int. J. Mol. Sci, vol.2020, pp.4986-5013

C. G. Ferreira, M. G. Campos, D. M. Felix, M. R. Santos, O. V. De-carvalho et al., Evaluation of the Antiviral Activities of Bacharis Dracunculifolia and Quercetin on Equid Herpesvirus 1 in a Murine Model, Res. Vet. Sci, vol.120, pp.70-77, 2018.

A. Ganeshpurkar and A. K. Saluja, The Pharmacological Potential of Rutin. Saudi Pharm, vol.25, pp.149-164, 2017.

S. N. Suganya and T. Sumathi, Effect of Rutin against a Mitochondrial Toxin, 3-Nitropropionicacid Induced Biochemical, Behavioral and Histological Alterations-a Pilot Study on Huntington's Disease Model in Rats, vol.32, pp.471-481, 2017.

N. A. Al-dhabi, M. V. Arasu, C. H. Park, and S. U. Park, An Up-to-Date Review of Rutin and Its Biological and Pharmacological Activities, Excli. J, vol.14, pp.59-63, 2015.

M. Singh, M. Kaur, and O. Silakari, Flavones: An Important Scaffold for Medicinal Chemistry, Eur. J. Med. Chem, vol.84, pp.206-239, 2014.

P. Kaur, S. Shukla, and S. Gupta, Plant Flavonoid Apigenin Inactivates Akt to Trigger Apoptosis in Human Prostate Cancer: An in Vitro and in Vivo Study, Carcinogenesis, vol.29, pp.2210-2217, 2008.

Y. Bao, S. Zhou, J. Fan, and Q. Wang, Anticancer Mechanism of Apigenin and the Implications of GLUT-1 Expression in Head and Neck Cancers, Future Oncol, vol.9, pp.1353-1364, 2013.

K. Banerjee and M. Mandal, Oxidative Stress Triggered by Naturally Occurring Flavone Apigenin Results in Senescence and Chemotherapeutic Effect in Human Colorectal Cancer Cells, Redox Biol, vol.5, pp.153-162, 2015.

M. Bian, Y. Zhang, X. Du, J. Xu, J. Cui et al., Apigenin-7-Diglucuronide Protects Retinas against Bright Light-Induced Photoreceptor Degeneration through the Inhibition of Retinal Oxidative Stress and Inflammation, Brain Res, vol.1663, pp.141-150, 2017.

S. Shukla and S. Gupta, Apigenin: A Promising Molecule for Cancer Prevention, Pharm Res, vol.27, pp.962-978, 2010.

N. Jamali-raeufy, T. Baluchnejadmojarad, M. Roghani, S. Keimasi, and M. Goudarzi, Isorhamnetin Exerts Neuroprotective Effects in STZ-Induced Diabetic Rats via Attenuation of Oxidative Stress, Inflammation and Apoptosis, J. Chem. Neuroanat, vol.102, 2019.

J. H. Yang, S. C. Kim, B. Y. Shin, S. H. Jin, M. J. Jo et al., O-Methylated Flavonol Isorhamnetin Prevents Acute Inflammation through Blocking of NF-KB Activation, Food Chem. Toxicol, vol.59, pp.362-372, 2013.

Q. Wu, P. A. Kroon, H. Shao, P. W. Needs, and X. Yang, Differential Effects of Quercetin and Two of Its Derivatives, Isorhamnetin and Isorhamnetin-3-Glucuronide, in Inhibiting the Proliferation of Human Breast-Cancer MCF-7 Cells, J. Agric. Food Chem, vol.66, pp.7181-7189, 2018.

J. H. Yang, B. Y. Shin, J. Y. Han, M. G. Kim, J. E. Wi et al., Isorhamnetin Protects against Oxidative Stress by Activating Nrf2 and Inducing the Expression of Its Target Genes, Toxicol. Appl. Pharmacol, vol.274, pp.293-301, 2014.

Y. S. Lee, S. Lee, H. S. Lee, B. Kim, K. Ohuchi et al., Inhibitory Effects of Isorhamnetin-3-O-Beta-D-Glucoside from Salicornia Herbacea on Rat Lens Aldose Reductase and Sorbitol Accumulation in Streptozotocin-Induced Diabetic Rat Tissues, Biol. Pharm. Bull, vol.28, pp.916-918, 2005.

N. Jamali-raoufi, S. Keimasi, and T. Baluchnejadmojarad, Isorhamnetin Mitigates Learning and Memory Disturbances in Streptozotocin-Induced Diabetic Rats, J. Basic Clin. Pathophysiol, vol.6, pp.37-42, 2018.

E. Yang, G. Kim, M. Jun, and K. Song, Kaempferol Attenuates the Glutamate-Induced Oxidative Stress in Mouse-Derived Hippocampal Neuronal HT22 Cells, Food Funct, vol.5, pp.1395-1402, 2014.

P. Rajendran, T. Rengarajan, N. Nandakumar, R. Palaniswami, and Y. Nishigaki, Nishigaki, I. Kaempferol, a Potential Cytostatic and Cure for Inflammatory Disorders, Eur. J. Med. Chem, vol.86, pp.103-112, 2014.

Q. Dang, W. Song, D. Xu, Y. Ma, F. Li et al., Kaempferol Suppresses Bladder Cancer Tumor Growth by Inhibiting Cell Proliferation and Inducing Apoptosis, Mol. Carcinog, vol.54, pp.831-840, 2015.

A. Sekiguchi, S. Motegi, C. Fujiwara, S. Yamazaki, Y. Inoue et al., Inhibitory Effect of Kaempferol on Skin Fibrosis in Systemic Sclerosis by the Suppression of Oxidative Stress, J. Dermatol. Sci, vol.96, pp.8-17, 2019.

, Int. J. Mol. Sci, vol.2020, pp.4986-5014

K. Sadasivam and R. Kumaresan, Antioxidant Behavior of Mearnsetin and Myricetin Flavonoid Compounds-A DFT Study, Spectrochim. Acta Part A, vol.79, pp.282-293, 2011.

D. Souza, P. Gasparotto, A. Crestani, S. Stefanello, M. É. Marques et al., Hypotensive Mechanism of the Extracts and Artemetin Isolated from Achillea Millefolium L. (Asteraceae) in Rats, Phytomedicine, vol.18, pp.819-825, 2011.

D. Lee, C. Kim, S. Park, K. O. Kim, N. T. Hiep et al., Protective Effect of Artemisia Argyi and Its Flavonoid Constituents against Contrast-Induced Cytotoxicity by Iodixanol in LLC-PK1 Cells, Int. J. Mol. Sci, vol.19, 1387.

J. Hu, W. Ma, N. Li, K. Wang, J. Hu et al., Antioxidant and Anti-Inflammatory Flavonoids from the Flowers of Chuju, a Medical Cultivar of Chrysanthemum Morifolim Ramat, J. Mex. Chem. Soc, vol.61, pp.282-289, 2017.

W. Li, C. Cui, B. Cai, H. Wang, and X. Yao, Flavonoids from Vitex Trifolia L. Inhibit Cell Cycle Progression at G2/M Phase and Induce Apoptosis in Mammalian Cancer Cells, J. Asian Nat. Prod. Res, vol.7, pp.615-626, 2005.

E. Langa, J. I. Pardo, C. Giménez-rota, A. González-coloma, M. J. Hernáiz et al., Supercritical Anti-Solvent Fractionation of Artemisia Absinthium L. Conventional Extracts: Tracking Artemetin and Casticin, J. Supercrit. Fluids, vol.151, pp.15-23, 2019.

E. W. Chan, S. K. Wong, and H. T. Chan, Casticin from Vitex Species: A Short Review on Its Anticancer and Anti-Inflammatory Properties, J. Integr. Med, vol.16, pp.147-152, 2018.

H. Lee, K. Jung, H. Lee, S. Park, W. Choi et al., Casticin, an Active Compound Isolated from Vitex Fructus, Ameliorates the Cigarette Smoke-Induced Acute Lung Inflammatory Response in a Murine Model, Int. Immunopharmacol, vol.28, pp.1097-1101, 2015.

C. Liou, W. Len, S. Wu, C. Lin, X. Wu et al., Casticin Inhibits COX-2 and INOS Expression via Suppression of NF-KB and MAPK Signaling in Lipopolysaccharide-Stimulated Mouse Macrophages, J. Ethnopharmacol, vol.158, pp.310-316, 2014.

Q. Zhu, Y. Wang, Y. Liu, R. Zhang, X. Li et al., Inhibition of Enterovirus 71 Replication by Chrysosplenetin and Penduletin, Eur. J. Pharm. Sci, vol.44, pp.392-398, 2011.

S. Messaili, C. Colas, L. Fougère, and E. Destandau, Combination of Molecular Network and Centrifugal Partition Chromatography Fractionation for Targeting and Identifying Artemisia Annua L. Antioxidant Compounds, J. Chromatogr, vol.2020, p.460785, 1615.

Y. Yin, F. Gong, X. Wu, Y. Sun, Y. Li et al., Anti-Inflammatory and Immunosuppressive Effect of Flavones Isolated from Artemisia Vestita, J. Ethnopharmacol, vol.120, 2008.

X. Sheng, Y. Sun, Y. Yin, T. Chen, and Q. Xu, Cirsilineol Inhibits Proliferation of Cancer Cells by Inducing Apoptosis via Mitochondrial Pathway, J. Pharm. Pharmacol, vol.60, pp.1523-1529, 2008.

Y. Sun, X. Wu, Y. Yin, F. Gong, Y. Shen et al., Novel Immunomodulatory Properties of Cirsilineol through Selective Inhibition of IFN-? Signaling in a Murine Model of Inflammatory Bowel Disease, Biochem. Pharmacol, vol.79, pp.229-238, 2010.

Y. Tezuka, P. Stampoulis, A. H. Banskota, S. Awale, K. Q. Tran et al., Constituents of the Vietnamese Medicinal Plant Orthosiphon Stamineus, Chem. Pharm. Bull, vol.48, pp.1711-1719, 2000.

T. Nagao, F. Abe, J. Kinjo, and H. Okabe, Antiproliferative Constituents in Plants 10. Flavones from the Leaves of Lantana Montevidensis Briq. and Consideration of Structure-Activity Relationship, Biol. Pharm. Bull, vol.25, pp.875-879, 2002.

V. Androutsopoulos, R. R. Arroo, J. F. Hall, S. Surichan, and G. A. Potter, Antiproliferative and Cytostatic Effects of the Natural Product Eupatorin on MDA-MB-468 Human Breast Cancer Cells Due to CYP1-Mediated Metabolism, Breast Cancer Res, vol.10, 2008.

Z. Chen, L. Liao, Z. Zhang, L. Wu, and Z. Wang, Comparison of Active Constituents, Acute Toxicity, Anti-Nociceptive and Anti-Inflammatory Activities of Porana Sinensis Hemsl., Erycibe Obtusifolia Benth. and Erycibe Schmidtii Craib, J. Ethnopharmacol, vol.150, pp.501-506, 2013.

H. Kim, S. I. Jang, Y. Kim, H. Chung, Y. Yun et al., Scopoletin Suppresses Pro-Inflammatory Cytokines and PGE2 from LPS-Stimulated Cell Line, Fitoterapia, vol.75, pp.261-266, 2004.

R. Pan, Y. Dai, X. Gao, and Y. Xia, Scopolin Isolated from Erycibe Obtusifolia Benth Stems Suppresses Adjuvant-Induced Rat Arthritis by Inhibiting Inflammation and Angiogenesis, Int. Immunopharmacol, vol.9, pp.859-869, 2009.

C. Shaw, C. Chen, C. Hsu, C. Chen, and Y. Tsai, Antioxidant Properties of Scopoletin Isolated from Sinomonium Acutum, Phytother. Res, vol.17, pp.823-825, 2003.

P. Moon, B. Lee, H. Jeong, H. An, S. Park et al., Use of Scopoletin to Inhibit the Production of Inflammatory Cytokines through Inhibition of the I?B/NF-KB Signal Cascade in the Human Mast Cell Line HMC-1, Eur. J. Pharmacol, vol.555, pp.218-225, 2007.

A. A. Thabet, F. S. Youssef, M. Korinek, F. Chang, Y. Wu et al., Study of the Anti-Allergic and Anti-Inflammatory Activity of Brachychiton Rupestris and Brachychiton Discolor Leaves (Malvaceae) Using in Vitro Models, BMC Complement. Altern. Med, vol.18, p.299, 2018.

S. Iqbal, U. Younas, K. W. Chan, M. Zia-ul-haq, and M. Ismail, Chemical Composition of Artemisia Annua L. Leaves and Antioxidant Potential of Extracts as a Function of Extraction Solvents, Molecules, vol.17, pp.6020-6032, 2012.

M. Winkelman, Ethnobotanical Treatments of Diabetes in Baja California Norte, Med. Anthr, vol.11, pp.255-268, 1989.

E. G. Helal, N. Abou-aouf, A. L. Khattab, and M. A. Zoair, Anti-Diabetic Effect of Artemisia Annua (Kaysom) in Alloxan-Induced Diabetic Rats, vol.57, pp.422-430, 2014.

H. J. Woerdenbag, N. Pras, R. Bos, J. F. Visser, H. Hendriks et al., Analysis of Artemisinin and Related Sesquiterpenoids from Artemisia Annua L. by Combined Gas Chromatography/Mass Spectrometry, Phytochem. Anal, vol.2, pp.215-219, 1991.

H. Wang, C. Ma, L. Ma, Z. Du, H. Wang et al., Secondary Metabolic Profiling and Artemisinin Biosynthesis of Two Genotypes of Artemisia Annua, Planta Med, vol.75, pp.1625-1633, 2009.

A. Taleghani, S. A. Emami, and Z. Tayarani-najaran, Artemisia: A Promising Plant for the Treatment of Cancer, Bioorg. Med. Chem, vol.28, 2020.

J. Huo, Y. Lu, L. Xia, and D. Chen, Structural Characterization and Anticomplement Activities of Three Acidic Homogeneous Polysaccharides from Artemisia Annua, J. Ethnopharmacol, 2020.

T. Efferth, A. Olbrich, and R. Bauer, MRNA Expression Profiles for the Response of Human Tumor Cell Lines to the Antimalarial Drugs Artesunate, Arteether, and Artemether, Biochem. Pharmacol, vol.64, pp.617-623, 2002.

T. Efferth, A. Sauerbrey, A. Olbrich, E. Gebhart, P. Rauch et al., Molecular Modes of Action of Artesunate in Tumor Cell Lines, Mol. Pharm, vol.64, pp.382-394, 2003.

T. Efferth and F. Oesch, Oxidative Stress Response of Tumor Cells: Microarray-Based Comparison between Artemisinins and Anthracyclines, Biochem. Pharmacol, vol.68, pp.3-10, 2004.

T. Efferth and M. Volm, Glutathione-Related Enzymes Contribute to Resistance of Tumor Cells and Low Toxicity in Normal Organs to Artesunate, vol.19, pp.225-232, 2005.

T. Efferth, M. Giaisi, A. Merling, P. H. Krammer, and M. Li-weber, Artesunate Induces ROS-Mediated Apoptosis in Doxorubicin-Resistant T Leukemia Cells, PLoS ONE, vol.2, 2007.

S. Sieber, G. Gdynia, W. Roth, B. Bonavida, and T. Efferth, Combination Treatment of Malignant B Cells Using the Anti-CD20 Antibody Rituximab and the Anti-Malarial Artesunate, Int. J. Oncol, vol.35, pp.149-158, 2009.

E. Ooko, M. E. Saeed, O. Kadioglu, S. Sarvi, M. Colak et al., Artemisinin Derivatives Induce Iron-Dependent Cell Death (Ferroptosis) in Tumor Cells, Phytomedicine, vol.22, pp.1045-1054, 2015.

R. Lin, Z. Zhang, L. Chen, Y. Zhou, P. Zou et al., Dihydroartemisinin (DHA) Induces Ferroptosis and Causes Cell Cycle Arrest in Head and Neck Carcinoma Cells, Cancer Lett, vol.381, pp.165-175, 2016.

A. Wojtkowiak-giera, M. Derda, D. Kosik-bogacka, A. Kolasa-wo?osiuk, E. Wandurska-nowak et al., The Modulatory Effect of Artemisia Annua L. on Toll-like Receptor Expression in Acanthamoeba Infected Mouse Lungs, Exp. Parasitol, vol.199, pp.24-29, 2019.

W. Yao, F. Wang, and H. Wang, Immunomodulation of Artemisinin and Its Derivatives, Sci. Bull, vol.61, pp.1399-1406, 2016.

A. Wojtkowiak-giera, M. Derda, D. Kosik-bogacka, A. Kolasa-wo?osiuk, P. Solarczyk et al., Influence of Artemisia Annua L. on Toll-like Receptor Expression in Brain of Mice Infected with Acanthamoeba Sp, Exp. Parasitol, vol.185, pp.17-22, 2018.

T. Kawai and S. Akira, The Roles of TLRs, RLRs and NLRs in Pathogen Recognition, Int. Immunol, vol.21, pp.317-337, 2009.

E. Cario, D. Brown, M. Mckee, K. Lynch-devaney, G. Gerken et al., Commensal-Associated Molecular Patterns Induce Selective Toll-Like Receptor-Trafficking from Apical Membrane to Cytoplasmic Compartments in Polarized Intestinal Epithelium, Am. J. Pathol, vol.160, pp.165-173, 2002.

B. Li, J. Li, X. Pan, G. Ding, H. Cao et al., Artesunate Protects Sepsis Model Mice Challenged with Staphylococcus Aureus by Decreasing TNF-? Release via Inhibition TLR2 and Nod2 MRNA Expressions and Transcription Factor NF-KB Activation, Int. Immunopharmacol, vol.10, pp.344-350, 2010.

X. Huang, Z. Xie, F. Liu, C. Han, D. Zhang et al., Dihydroartemisinin Inhibits Activation of the Toll-like Receptor 4 Signaling Pathway and Production of Type I Interferon in Spleen Cells from Lupus-Prone MRL/Lpr Mice, Int. Immunopharmacol, vol.22, pp.266-272, 2014.

S. Cuzzocrea, F. Saadat, R. D. Paola, and A. Mirshafiey, Artemether: A New Therapeutic Strategy in Experimental Rheumatoid Arthritis, Immunopharmacol. Immunotoxicol, vol.27, pp.615-630, 2005.

A. Mirshafiey, F. Saadat, M. Attar, R. D. Paola, R. Sedaghat et al., Design of a New Line in Treatment of Experimental Rheumatoid Arthritis by Artesunate, Immunopharmacol. Immunotoxicol, vol.28, pp.397-410, 2006.

J. Wang, W. Tang, R. Zhou, J. Wan, L. Shi et al., The New Water-Soluble Artemisinin Derivative SM905 Ameliorates Collagen-Induced Arthritis by Suppression of Inflammatory and Th17 Responses, Br. J. Pharmacol, vol.153, pp.1303-1310, 2008.

Y. Li, S. Wang, Y. Wang, C. Zhou, G. Chen et al., Inhibitory Effect of the Antimalarial Agent Artesunate on Collagen-Induced Arthritis in Rats through Nuclear Factor Kappa B and Mitogen-Activated Protein Kinase Signaling Pathway, Transl. Res, vol.161, pp.89-98, 2013.

H. Xu, Y. He, X. Yang, L. Liang, Z. Zhan et al., Anti-Malarial Agent Artesunate Inhibits TNF-?-Induced Production of Proinflammatory Cytokines via Inhibition of NF-KB and PI3 Kinase/Akt Signal Pathway in Human Rheumatoid Arthritis Fibroblast-like Synoviocytes, Rheumatol. Oxf, vol.46, pp.920-926, 2007.

Y. He, J. Fan, H. Lin, X. Yang, Y. Ye et al., The Anti-Malaria Agent Artesunate Inhibits Expression of Vascular Endothelial Growth Factor and Hypoxia-Inducible Factor-1? in Human Rheumatoid Arthritis Fibroblast-like Synoviocyte, Rheumatol. Int, vol.31, pp.53-60, 2011.

O. Jin, H. Zhang, Z. Gu, S. Zhao, T. Xu et al., A Pilot Study of the Therapeutic Efficacy and Mechanism of Artesunate in the MRL/Lpr Murine Model of Systemic Lupus Erythematosus, Cell. Mol. Immunol, vol.6, pp.461-467, 2009.

L. Hou, S. He, X. Li, C. Wan, Y. Yang et al., SM934 Treated Lupus-Prone NZB×NZW F1 Mice by Enhancing Macrophage Interleukin-10 Production and Suppressing Pathogenic T Cell Development, PLoS ONE, vol.7, 2012.

L. Hou, S. He, X. Li, Y. Yang, P. He et al., Oral Administration of Artemisinin Analog SM934 Ameliorates Lupus Syndromes in MRL/Lpr Mice by Inhibiting Th1 and Th17 Cell Responses, Arthritis Rheum, vol.63, pp.2445-2455, 2011.

W. Li, Y. Dong, Y. Tu, and Z. Lin, Dihydroarteannuin Ameliorates Lupus Symptom of BXSB Mice by Inhibiting Production of TNF-Alpha and Blocking the Signaling Pathway NF-Kappa B Translocation, Int. Immunopharmacol, vol.6, pp.1243-1250, 2006.

X. Wu, W. Zhang, X. Shi, P. An, W. Sun et al., Therapeutic Effect of Artemisinin on Lupus Nephritis Mice and Its Mechanisms, Acta Biochim. Biophys. Sin, vol.42, pp.916-923, 2010.

, Int. J. Mol. Sci, vol.2020, p.4986

X. Wu, W. Zhang, X. Shi, P. An, W. Sun et al., Effect of Artemisinin Combined with Glucocorticoid on the Expressions of Glucocorticoid Receptor ? MRNA, Glucocorticoid Receptor ? MRNA and P300/CBP Protein in Lupus Nephritis Mice, Chin. J. Integr. Med, vol.17, pp.277-282, 2011.

Z. Yang, J. Ding, C. Yang, Y. Gao, X. Li et al., Immunomodulatory and Anti-Inflammatory Properties of Artesunate in Experimental Colitis, Curr. Med. Chem, vol.19, pp.4541-4551, 2012.

T. Li, H. Chen, N. Wei, X. Mei, S. Zhang et al., Anti-Inflammatory and Immunomodulatory Mechanisms of Artemisinin on Contact Hypersensitivity, Int. Immunopharmacol, vol.12, pp.144-150, 2012.

C. Cheng, D. S. Ng, T. K. Chan, S. P. Guan, W. E. Ho et al., Anti-Allergic Action of Anti-Malarial Drug Artesunate in Experimental Mast Cell-Mediated Anaphylactic Models, Allergy, vol.68, pp.195-203, 2013.

C. Cheng, W. E. Ho, F. Y. Goh, S. P. Guan, L. R. Kong et al., Anti-Malarial Drug Artesunate Attenuates Experimental Allergic Asthma via Inhibition of the Phosphoinositide 3-Kinase/Akt Pathway, PLoS ONE, vol.6, 2011.

W. E. Ho, C. Cheng, H. Y. Peh, F. Xu, S. R. Tannenbaum et al., Anti-Malarial Drug Artesunate Ameliorates Oxidative Lung Damage in Experimental Allergic Asthma. Free Radic, Biol. Med, vol.53, pp.498-507, 2012.

S. K. Dube, P. S. Panda, G. R. Agrawal, and D. K. Singh, Anaphylaxis to Artesunate?, Indian J. Crit. Care Med, vol.16, pp.55-57, 2012.

J. Shi, C. Zhang, X. Sun, X. Cheng, J. Wang et al., Antimalarial Drug Artemisinin Extenuates Amyloidogenesis and Neuroinflammation in APPswe/PS1dE9 Transgenic Mice via Inhibition of Nuclear Factor-KB and NLRP3 Inflammasome Activation, Cns. Neurosci. Ther, vol.19, pp.262-268, 2013.

S. Wang, B. Sun, Z. Cheng, H. Zhou, Y. Gao et al., Dihydroartemisinin Inhibits Angiogenesis in Pancreatic Cancer by Targeting the NF-KB Pathway, Cancer Chemother. Pharm, vol.68, pp.1421-1430, 2011.

A. R. Bilia, F. Santomauro, C. Sacco, M. C. Bergonzi, and R. Donato, Essential Oil of Artemisia Annua L.: An Extraordinary Component with Numerous Antimicrobial Properties, Evid. Based Complement. Altern. Med, 2014.

N. M. Stojanovi?, P. J. Randjelovi?, M. Z. Mladenovi?, I. R. Ili?, V. Petrovi? et al., Toxic Essential Oils, Part VI: Acute Oral Toxicity of Lemon Balm (Melissa Officinalis L.) Essential Oil in BALB/c Mice, Food Chem. Toxicol, vol.133, 2019.

Y. Li, H. Hu, X. Zheng, J. Zhu, and L. Liu, Composition and Antimicrobial Activity of Essential Oil from the Aerial Part of Artemisia Annua, vol.5, pp.3629-3633, 2011.

F. Juteau, V. Masotti, J. M. Bessière, M. Dherbomez, and J. Viano, Antibacterial and Antioxidant Activities of Artemisia Annua Essential Oil, Fitoterapia, vol.73, pp.532-535, 2002.

M. R. Verdian-rizi, Chemical Composition and Antimicrobial Activity of the Essential Oil of Artemisia Annua L. from Iran, Pharmacogn. Res, 2009.

A. Massiha, M. M. Khoshkholgh-pahlaviani, K. Issazadeh, S. Bidarigh, and S. Zarrabi, Antibacterial Activity of Essential Oils and Plant Extracts of Artemisia (Artemisia Annua L.) in Vitro, Zahedan J. Res. Med. Sci, vol.15, pp.14-18, 2013.

M. Viuda-martos, A. E. El-gendy, .. G. Sendra, E. Fernández-lópez, J. Abd-el-razik et al., Chemical Composition and Antioxidant and Anti-Listeria Activities of Essential Oils Obtained from Some Egyptian Plants, J. Agric. Food Chem, vol.58, pp.9063-9070, 2010.

M. C. Duarte, E. E. Leme, C. Delarmelina, A. A. Soares, G. M. Figueira et al., Activity of Essential Oils from Brazilian Medicinal Plants on Escherichia Coli, J. Ethnopharmacol, vol.111, pp.197-201, 2007.

E. M. Soylu, H. Yigitba?, F. M. Tok, S. Soylu, ?. Kurt et al., Chemical Composition and Antifungal Activity of the Essential Oil of Artemisia Annua L. against Foliar and Soil-Borne Fungal Pathogens/Die Chemische Zusammensetzung Und Antimikrobielle Aktivität Das Ätherischen Öls von Artemisia Annua L. Gegen Blatt-Und Bodenbürtige Pilzliche Krankheitserreger, Z. Pflanzenkrankh. Pflanzenschutz/J. Plant Dis. Prot, vol.112, pp.229-239, 2005.

T. Efferth, . Beyond, and . Malaria, The Inhibition of Viruses by Artemisinin-Type Compounds, Biotechnol. Adv, vol.36, pp.1730-1737, 2018.

Y. Chang and E. Woo, Korean Medicinal Plants Inhibiting to Human Immunodeficiency Virus Type 1 (HIV-1) Fusion, Phytother. Res, vol.17, pp.426-429, 2003.

R. M. Oguariri, J. W. Adelsberger, M. W. Baseler, and T. Imamichi, Evaluation of the Effect of Pyrimethamine, an Anti-Malarial Drug, on HIV-1 Replication, Virus Res, vol.153, pp.269-276, 2010.

S. Jana, S. Iram, J. Thomas, M. Q. Hayat, C. Pannecouque et al., Application of the Triazolization Reaction to Afford Dihydroartemisinin Derivatives with Anti-HIV Activity, vol.22, p.303, 2017.

T. Efferth, M. Marschall, X. Wang, S. Huong, I. Hauber et al., Antiviral Activity of Artesunate towards Wild-Type, Recombinant, and Ganciclovir-Resistant Human Cytomegaloviruses, J. Mol. Med, vol.80, pp.233-242, 2002.

T. Efferth, M. R. Romero, D. G. Wolf, T. Stamminger, J. J. Marin et al., The Antiviral Activities of Artemisinin and Artesunate, Clin. Infect. Dis, vol.47, pp.804-811, 2008.

J. Milbradt, S. Auerochs, K. Korn, and M. Marschall, Sensitivity of Human Herpesvirus 6 and Other Human Herpesviruses to the Broad-Spectrum Antiinfective Drug Artesunate, J. Clin. Virol, vol.46, pp.24-28, 2009.

R. Arav-boger, R. He, C. Chiou, J. Liu, L. Woodard et al., Artemisinin-Derived Dimers Have Greatly Improved Anti-Cytomegalovirus Activity Compared to Artemisinin Monomers, PLoS ONE, vol.5, 2010.

R. He, B. T. Mott, A. S. Rosenthal, D. T. Genna, G. H. Posner et al., An Artemisinin-Derived Dimer Has Highly Potent Anti-Cytomegalovirus (CMV) and Anti-Cancer Activities, PLoS ONE, vol.6, 2011.

C. Reiter, T. Fröhlich, M. Zeino, M. Marschall, H. Bahsi 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, vol.97, pp.164-172, 2015.

C. Hutterer, I. Niemann, J. Milbradt, T. Fröhlich, C. Reiter et al., The Broad-Spectrum Antiinfective Drug Artesunate Interferes with the Canonical Nuclear Factor Kappa B (NF-KB) Pathway by Targeting RelA/P65, Antivir. Res, vol.124, pp.101-109, 2015.

M. Y. Shapira, I. B. Resnick, S. Chou, A. U. Neumann, N. S. Lurain et al., Artesunate as a Potent Antiviral Agent in a Patient with Late Drug-Resistant Cytomegalovirus Infection after Hematopoietic Stem Cell Transplantation, Clin. Infect. Dis, vol.46, pp.1455-1457, 2008.

L. Naesens, P. Bonnafous, H. Agut, and E. De-clercq, Antiviral Activity of Diverse Classes of Broad-Acting Agents and Natural Compounds in HHV-6-Infected Lymphoblasts, J. Clin. Virol, vol.37, pp.69-75, 2006.

N. Hakacova, K. Klingel, R. Kandolf, E. Engdahl, A. Fogdell-hahn et al., First Therapeutic Use of Artesunate in Treatment of Human Herpesvirus 6B Myocarditis in a Child, J. Clin. Virol, vol.57, pp.157-160, 2013.

F. H. Qi, Z. X. Wang, P. P. Cai, L. Zhao, J. J. Gao et al., Traditional Chinese Medicine and Related Active Compounds: A Review of Their Role on Hepatitis B Virus Infection. Drug Discov, vol.7, pp.212-224, 2013.

M. R. Romero, T. Efferth, M. A. Serrano, B. Castaño, R. I. Macias et al., Effect of Artemisinin/Artesunate as Inhibitors of Hepatitis B Virus Production in an "in Vitro, Replicative System. Antivir. Res, vol.68, pp.75-83, 2005.

K. T. Batty, T. M. Davis, L. T. Thu, T. Binh, T. Kim-anh et al., Selective High-Performance Liquid Chromatographic Determination of Artesunate and ?-and ?-Dihydroartemisinin in Patients with Falciparum Malaria, J. Chromatogr. B, vol.677, pp.345-350, 1996.

B. N. Sharma, M. Marschall, S. Henriksen, and C. H. Rinaldo, Antiviral Effects of Artesunate on Polyomavirus BK Replication in Primary Human Kidney Cells, Antimicrob. Agents Chemother, 2013.

B. N. Sharma, M. Marschall, and C. H. Rinaldo, Antiviral Effects of Artesunate on JC Polyomavirus Replication in COS-7 Cells, Antimicrob. Agents Chemother, vol.58, pp.6724-6734, 2014.

, Int. J. Mol. Sci, vol.2020, p.4986

G. L. Disbrow, A. C. Baege, K. A. Kierpiec, H. Yuan, J. A. Centeno et al., Dihydroartemisinin Is Cytotoxic to Papillomavirus-Expressing Epithelial Cells In Vitro and In Vivo, Cancer Res, vol.65, pp.10854-10861, 2005.

A. Mondal and U. Chatterji, Artemisinin Represses Telomerase Subunits and Induces Apoptosis in HPV-39 Infected Human Cervical Cancer Cells, J. Cell. Biochem, vol.116, 1968.

J. Paeshuyse, L. Coelmont, I. Vliegen, J. V. Hemel, J. Vandenkerckhove et al., Hemin Potentiates the Anti-Hepatitis C Virus Activity of the Antimalarial Drug Artemisinin, Biochem. Biophys. Res. Commun, vol.348, pp.139-144, 2006.

S. Obeid, J. Alen, V. H. Nguyen, V. C. Pham, P. Meuleman et al., Artemisinin Analogues as Potent Inhibitors of In Vitro Hepatitis C Virus Replication, PLoS ONE, vol.8, 2013.

R. Dai, X. Xiao, F. Peng, M. Li, and G. Gong, Artesunate, an Anti-Malarial Drug, Has a Potential to Inhibit HCV Replication, Virus Genes, vol.52, pp.22-28, 2016.

M. R. Romero, M. A. Serrano, M. Vallejo, T. Efferth, M. Alvarez et al., Antiviral Effect of Artemisinin from Artemisia Annua against a Model Member of the Flaviviridae Family, the Bovine Viral Diarrhoea Virus (BVDV), Planta Med, vol.72, pp.1169-1174, 2006.

A. G. Blazquez, M. Fernandez-dolon, L. Sanchez-vicente, A. D. Maestre, A. B. Miguel et al., Novel Artemisinin Derivatives with Potential Usefulness against Liver/Colon Cancer and Viral Hepatitis, Bioorg. Med. Chem, vol.21, pp.4432-4441, 2013.

M. Thaha, N. Pranawa, M. Yogiantoro, M. Tanimoto, and Y. Tomino, Acute Renal Failure in a Patient with Severe Malaria and Dengue Shock Syndrome, Clin. Nephrol, vol.70, pp.427-430, 2008.

J. Munyangi, L. Cornet-vernet, M. Idumbo, C. Lu, P. Lutgen et al., Artemisia Annua and Artemisia Afra Tea Infusions vs

. Artesunate-amodiaquine, Treating Plasmodium Falciparum Malaria in a Large Scale, Double Blind, Randomized Clinical Trial, vol.57, pp.49-56, 2019.

E. Hsu, Reflections on the 'Discovery' of the Antimalarial Qinghao, Br. J. Clin. Pharm, vol.61, pp.666-670, 2006.

N. J. White and . Qinghaosu, Artemisinin): The Price of Success, Science, vol.320, pp.330-334, 2008.

, World Health Organization. Guidelines for the Treatment of Malaria; World Health Organization, 2006.

P. T. Loverde and . Digenetic-trematodes,

R. Toledo and B. Fried, Advances in Experimental Medicine and Biology, pp.45-70, 2019.

P. Steinmann, J. Keiser, R. Bos, M. Tanner, and J. Utzinger, Schistosomiasis and Water Resources Development: Systematic Review, Meta-Analysis, and Estimates of People at Risk, Lancet Infect. Dis, vol.6, pp.411-425, 2006.

M. E. Saeed, S. Krishna, H. J. Greten, P. G. Kremsner, and T. Efferth, Antischistosomal Activity of Artemisinin Derivatives in Vivo and in Patients, Pharmacol. Res, vol.110, pp.216-226, 2016.

H. A. Shalaby, S. Abdel-shafy, K. A. Abdel-rahman, and A. A. Derbala, Comparative in Vitro Effect of Artemether and Albendazole on Adult Toxocara Canis, Parasitol. Res, vol.105, pp.967-976, 2009.

J. Golenser, J. H. Waknine, M. Krugliak, N. H. Hunt, and G. Ge, Current Perspectives on the Mechanism of Action of Artemisinins, Int. J. Parasitol, vol.36, pp.1427-1441, 2006.

D. M. Abou-rayia, A. E. Saad, D. S. Ashour, and R. M. Oreiby, Implication of Artemisinin Nematocidal Activity on Experimental Trichinellosis: In Vitro and in Vivo Studies, Parasitol. Int, vol.66, pp.56-63, 2017.

Y. Kang, J. Jo, M. Cho, H. Yu, M. S. Ock et al., Trichinella Spiralis Infection Induces Angiogenic Factor Thymosin B4 Expression, Vet. Parasitol, vol.181, pp.222-228, 2011.

S. Echeverrigaray, J. Zacaria, and R. Beltrão, Nematicidal Activity of Monoterpenoids Against the Root-Knot Nematode Meloidogyne Incognita, Phytopathology, vol.100, pp.199-203, 2010.

N. S. Lam, X. Long, X. Su, and F. Lu, Artemisinin and Its Derivatives in Treating Helminthic Infections beyond Schistosomiasis, Pharmacol. Res, vol.133, pp.77-100, 2018.