Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2022, Vol. 16 Issue (1) : 1-9    https://doi.org/10.1007/s11684-021-0898-6
REVIEW
Discovery and repurposing of artemisinin
Qiaoli Shi1, Fei Xia1, Qixin Wang1, Fulong Liao1, Qiuyan Guo1(), Chengchao Xu1,4(), Jigang Wang1,2,3,4()
1. Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
2. Central People’s Hospital of Zhanjiang, Zhanjiang 524045, China
3. Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
4. Department of Geriatrics, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
 Download: PDF(1583 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Malaria is an ancient infectious disease that threatens millions of lives globally even today. The discovery of artemisinin, inspired by traditional Chinese medicine (TCM), has brought in a paradigm shift and been recognized as the “best hope for the treatment of malaria” by World Health Organization. With its high potency and low toxicity, the wide use of artemisinin effectively treats the otherwise drug-resistant parasites and helps many countries, including China, to eventually eradicate malaria. Here, we will first review the initial discovery of artemisinin, an extraordinary journey that was in stark contrast with many drugs in western medicine. We will then discuss how artemisinin and its derivatives could be repurposed to treat cancer, inflammation, immunoregulation-related diseases, and COVID-19. Finally, we will discuss the implications of the “artemisinin story” and how that can better guide the development of TCM today. We believe that artemisinin is just a starting point and TCM will play an even bigger role in healthcare in the 21st century.

Keywords artemisinin      drug repurposing      cancer      inflammation      COVID-19      traditional Chinese medicine     
Corresponding Author(s): Qiuyan Guo,Chengchao Xu,Jigang Wang   
Just Accepted Date: 28 January 2022   Online First Date: 11 March 2022    Issue Date: 28 March 2022
 Cite this article:   
Qiaoli Shi,Fei Xia,Qixin Wang, et al. Discovery and repurposing of artemisinin[J]. Front. Med., 2022, 16(1): 1-9.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-021-0898-6
https://academic.hep.com.cn/fmd/EN/Y2022/V16/I1/1
Fig.1  Artemisia annua L. in the field.
Fig.2  Repurpose of artemisinin and its derivatives.
Fig.3  Structures and anticancer, anti-inflammatory, and immunoregulatory properties of artemisinin and its derivatives.
Fig.4  Artemisinin and its derivatives-based combination therapies against COVID-19.
1 YY Tu. Artemisinin—a gift from traditional Chinese medicine to the world (Nobel Lecture). Angew Chem Int Ed Eng, 2016, l55( 35): 10210– 10226
2 World Health Organization. World malaria report 2020: 20 years of global progress and challenges. Geneva: World Health Organization, 2020
3 GJ Yoshida. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol, 2017, 10( 1): 67
https://doi.org/10.1186/s13045-017-0436-9
4 T Efferth. From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin Cancer Biol, 2017, 46 : 65– 83
https://doi.org/10.1016/j.semcancer.2017.02.009
5 WE Ho, HY Peh, TK Chan, WS Wong. Artemisinins: pharmacological actions beyond anti-malarial. Pharmacol Ther, 2014, 142( 1): 126– 139
https://doi.org/10.1016/j.pharmthera.2013.12.001
6 G Li, M Yuan, H Li, C Deng, Q Wang, Y Tang, H Zhang, W Yu, Q Xu, Y Zou, Y Yuan, J Guo, C Jin, X Guan, F Xie, J Song. Safety and efficacy of artemisinin-piperaquine for treatment of COVID-19: an open-label, non-randomised and controlled trial. Int J Antimicrob Agents, 2021, 57( 1): 106216
https://doi.org/10.1016/j.ijantimicag.2020.106216
7 M Gendrot, I Duflot, M Boxberger, O Delandre, P Jardot, M Le Bideau, J Andreani, I Fonta, J Mosnier, C Rolland, S Hutter, B La Scola, B Pradines. Antimalarial artemisinin-based combination therapies (ACT) and COVID-19 in Africa: in vitro inhibition of SARS-CoV-2 replication by mefloquine-artesunate. Int J Infect Dis, 2020, 99 : 437– 440
https://doi.org/10.1016/j.ijid.2020.08.032
8 S Krishna, Y Augustin, J Wang, C Xu, HM Staines, H Platteeuw, A Kamarulzaman, A Sall, P Kremsner. Repurposing antimalarials to tackle the COVID-19 pandemic. Trends Parasitol, 2021, 37( 1): 8– 11
https://doi.org/10.1016/j.pt.2020.10.003
9 J Yang, Y He, Y Li, X Zhang, YK Wong, S Shen, T Zhong, J Zhang, Q Liu, J Wang. Advances in the research on the targets of anti-malaria actions of artemisinin. Pharmacol Ther, 2020, 216 : 107697
https://doi.org/10.1016/j.pharmthera.2020.107697
10 J Wang, C Xu, FL Liao, T Jiang, S Krishna, Y Tu. Suboptimal dosing triggers artemisinin partner drug resistance. Lancet Infect Dis, 2019, 19( 11): 1167– 1168
https://doi.org/10.1016/S1473-3099(19)30535-3
11 J Wang, C Xu, FL Liao, T Jiang, S Krishna, Y Tu. A temporizing solution to “artemisinin resistance”. N Engl J Med, 2019, 380( 22): 2087– 2089
https://doi.org/10.1056/NEJMp1901233
12 Strategic Advisory Group on Malaria Eradication. Malaria eradication: benefits, future scenarios and feasibility. A report of the Strategic Advisory Group on Malaria Eradication. Geneva: World Health Organization, 2020
13 N Ma, Z Zhang, F Liao, T Jiang, Y Tu. The birth of artemisinin. Pharmacol Ther, 2020, 216 : 107658
https://doi.org/10.1016/j.pharmthera.2020.107658
14 Y Tu. The discovery of artemisinin (qinghaosu) and gifts from Chinese medicine. Nat Med, 2011, 17( 10): 1217– 1220
https://doi.org/10.1038/nm.2471
15 World Health Organization. Guidelines for the treatment of malaria. 1st ed. Geneva: World Health Organization, 2006
16 T Efferth, B Kaina. Toxicity of the antimalarial artemisinin and its dervatives. Crit Rev Toxicol, 2010, 40( 5): 405– 421
https://doi.org/10.3109/10408441003610571
17 The Nobel Prize. The Nobel Prize in Physiology or Medicine. 2015.
18 World Health Organization. World Health Organization Model List of Essential Medicines. 21st List. Geneva: World Health Organization, 2019
19 RT Eastman, DA Fidock. Artemisinin-based combination therapies: a vital tool in efforts to eliminate malaria. Nat Rev Microbiol, 2009, 7( 12): 864– 874
https://doi.org/10.1038/nrmicro2239
20 X Sun, P Yan, C Zou, YK Wong, Y Shu, YM Lee, C Zhang, ND Yang, J Wang, J Zhang. Targeting autophagy enhances the anticancer effect of artemisinin and its derivatives. Med Res Rev, 2019, 39( 6): 2172– 2193
https://doi.org/10.1002/med.21580
21 HJ Woerdenbag, TA Moskal, N Pras, TM Malingré, FS el-Feraly, HH Kampinga, AW Konings. Cytotoxicity of artemisinin-related endoperoxides to Ehrlich ascites tumor cells. J Nat Prod, 1993, 56( 6): 849– 856
https://doi.org/10.1021/np50096a007
22 HC Lai, NP Singh, T Sasaki. Development of artemisinin compounds for cancer treatment. Invest New Drugs, 2013, 31( 1): 230– 246
https://doi.org/10.1007/s10637-012-9873-z
23 D King, D Yeomanson, HE Bryant. PI3King the lock: targeting the PI3K/Akt/mTOR pathway as a novel therapeutic strategy in neuroblastoma. J Pediatr Hematol Oncol, 2015, 37( 4): 245– 251
https://doi.org/10.1097/MPH.0000000000000329
24 J Wang, J Zhang, Y Shi, C Xu, C Zhang, YK Wong, YM Lee, S Krishna, Y He, TK Lim, W Sim, ZC Hua, HM Shen, Q Lin. Mechanistic investigation of the specific anticancer property of artemisinin and its combination with aminolevulinic acid for enhanced anticolorectal cancer activity. ACS Cent Sci, 2017, 3( 7): 743– 750
https://doi.org/10.1021/acscentsci.7b00156
25 ND Yang, SH Tan, S Ng, Y Shi, J Zhou, KSW Tan, WSF Wong, HM Shen. Artesunate induces cell death in human cancer cells via enhancing lysosomal function and lysosomal degradation of ferritin. J Biol Chem, 2014, 289( 48): 33425– 33441
https://doi.org/10.1074/jbc.M114.564567
26 FB Feng, HY Qiu. Effects of artesunate on chondrocyte proliferation, apoptosis and autophagy through the PI3K/AKT/mTOR signaling pathway in rat models with rheumatoid arthritis. Biomed Pharmacother, 2018, 102 : 1209– 1220
https://doi.org/10.1016/j.biopha.2018.03.142
27 YS Wang, P Yu, Y Wang, J Zhang, W Hang, ZX Yin, G Liu, J Chen, KD Werle, CS Quan, H Gao, Q Zeng, R Cui, J Liang, Q Ding, YL Li, ZX Xu. AMP-activated protein kinase protects against necroptosis via regulation of Keap1-PGAM5 complex. Int J Cardiol, 2018, 259 : 153– 162
https://doi.org/10.1016/j.ijcard.2018.01.036
28 D Carling. AMPK signalling in health and disease. Curr Opin Cell Biol, 2017, 45 : 31– 37
https://doi.org/10.1016/j.ceb.2017.01.005
29 YK Choi, KG Park. Metabolic roles of AMPK and metformin in cancer cells. Mol Cells, 2013, 36( 4): 279– 287
https://doi.org/10.1007/s10059-013-0169-8
30 J Du, T Wang, Y Li, Y Zhou, X Wang, X Yu, X Ren, Y An, Y Wu, W Sun, W Fan, Q Zhu, Y Wang, X Tong. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Biol Med, 2019, 131 : 356– 369
https://doi.org/10.1016/j.freeradbiomed.2018.12.011
31 X Zhou, Y Chen, F Wang, H Wu, Y Zhang, J Liu, Y Cai, S Huang, N He, Z Hu, X Jin. Artesunate induces autophagy dependent apoptosis through upregulating ROS and activating AMPK-mTOR-ULK1 axis in human bladder cancer cells. Chem Biol Interact, 2020, 331 : 109273
https://doi.org/10.1016/j.cbi.2020.109273
32 C Cheng, T Wang, Z Song, L Peng, M Gao, O Hermine, S Rousseaux, S Khochbin, JQ Mi, J Wang. Induction of autophagy and autophagy-dependent apoptosis in diffuse large B-cell lymphoma by a new antimalarial artemisinin derivative, SM1044. Cancer Med, 2018, 7( 2): 380– 396
https://doi.org/10.1002/cam4.1276
33 A Orlova, C Wagner, Araujo ED de, D Bajusz, HA Neubauer, M Herling, PT Gunning, GM Keserű, R Moriggl. Direct targeting options for STAT3 and STAT5 in cancer. Cancers (Basel), 2019, 11( 12): 1930
https://doi.org/10.3390/cancers11121930
34 X Yan, P Li, Y Zhan, M Qi, J Liu, Z An, W Yang, H Xiao, H Wu, Y Qi, H Shao. Dihydroartemisinin suppresses STAT3 signaling and Mcl-1 and survivin expression to potentiate ABT-263-induced apoptosis in non-small cell lung cancer cells harboring EGFR or RAS mutation. Biochem Pharmacol, 2018, 150 : 72– 85
https://doi.org/10.1016/j.bcp.2018.01.031
35 W Wang, Y Sun, X Li, X Shi, Z Li, X Lu. Dihydroartemisinin prevents distant metastasis of laryngeal carcinoma by inactivating STAT3 in cancer stem cells. Med Sci Monit, 2020, 26 : e922348
https://doi.org/10.12659/MSM.922348
36 M Ilamathi, PC Prabu, KA Ayyappa, V Sivaramakrishnan. Artesunate obliterates experimental hepatocellular carcinoma in rats through suppression of IL-6-JAK-STAT signalling. Biomed Pharmacother, 2016, 82 : 72– 79
https://doi.org/10.1016/j.biopha.2016.04.061
37 M Berköz, F Özkan-Yılmaz, A Özlüer-Hunt, M Krośniak, Ö Türkmen, D Korkmaz, S Keskin. Artesunate inhibits melanoma progression in vitro via suppressing STAT3 signaling pathway. Pharmacol Rep, 2021, 73( 2): 650– 663
https://doi.org/10.1007/s43440-021-00230-6
38 L Zheng, C Wang, T Luo, B Lu, H Ma, Z Zhou, D Zhu, G Chi, P Ge, Y Luo. JNK activation contributes to oxidative stress-induced parthanatos in glioma cells via increase of intracellular ROS production. Mol Neurobiol, 2017, 54( 5): 3492– 3505
https://doi.org/10.1007/s12035-016-9926-y
39 CR Weston, RJ Davis. The JNK signal transduction pathway. Curr Opin Cell Biol, 2007, 19( 2): 142– 149
https://doi.org/10.1016/j.ceb.2007.02.001
40 M Ogata, S Hino, A Saito, K Morikawa, S Kondo, S Kanemoto, T Murakami, M Taniguchi, I Tanii, K Yoshinaga, S Shiosaka, JA Hammarback, F Urano, K Imaizumi. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol, 2006, 26( 24): 9220– 9231
https://doi.org/10.1128/MCB.01453-06
41 Y Wei, S Sinha, B Levine. Dual role of JNK1-mediated phosphorylation of Bcl-2 in autophagy and apoptosis regulation. Autophagy, 2008, 4( 7): 949– 951
https://doi.org/10.4161/auto.6788
42 GD Yao, MY Ge, DQ Li, L Chen, T Hayashi, SI Tashiro, S Onodera, C Guo, SJ Song, T Ikejima. L-A03, a dihydroartemisinin derivative, promotes apoptotic cell death of human breast cancer MCF-7 cells by targeting c-Jun N-terminal kinase. Biomed Pharmacother, 2018, 105 : 320– 325
https://doi.org/10.1016/j.biopha.2018.05.093
43 RZ Orlowski, AS Jr Baldwin. NF-κB as a therapeutic target in cancer. Trends Mol Med, 2002, 8( 8): 385– 389
https://doi.org/10.1016/S1471-4914(02)02375-4
44 AS Baldwin. Control of oncogenesis and cancer therapy resistance by the transcription factor NF-κB. J Clin Invest, 2001, 107( 3): 241– 246
https://doi.org/10.1172/JCI11991
45 X Chen, YK Wong, TK Lim, WH Lim, QS Lin, JG Wang, ZC Hua. Artesunate activates the intrinsic apoptosis of HCT116 cells through the suppression of fatty acid synthesis and the NF-κB pathway. Molecules, 2017, 22( 8): 1272
https://doi.org/10.3390/molecules22081272
46 W Hu, SS Chen, JL Zhang, XE Lou, HJ Zhou. Dihydroartemisinin induces autophagy by suppressing NF-κB activation. Cancer Lett, 2014, 343( 2): 239– 248
https://doi.org/10.1016/j.canlet.2013.09.035
47 B Li, S Bu, J Sun, Y Guo, D Lai. Artemisinin derivatives inhibit epithelial ovarian cancer cells via autophagy-mediated cell cycle arrest. Acta Biochim Biophys Sin (Shanghai), 2018, 50( 12): 1227– 1235
https://doi.org/10.1093/abbs/gmy125
48 Y Lin, M Jiang, W Chen, T Zhao, Y Wei. Cancer and ER stress: mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed Pharmacother, 2019, 118 : 109249
https://doi.org/10.1016/j.biopha.2019.109249
49 R Xiao, C Ding, H Zhu, X Liu, J Gao, Q Liu, D Lu, N Zhang, A Zhang, H Zhou. Suppression of asparagine synthetase enhances the antitumor potency of ART and artemalogue SOMCL-14-221 in non-small cell lung cancer. Cancer Lett, 2020, 475 : 22– 33
https://doi.org/10.1016/j.canlet.2020.01.035
50 TK Våtsveen, MR Myhre, CB Steen, S Wälchli, OC Lingjærde, B Bai, P Dillard, TA Theodossiou, T Holien, A Sundan, EM Inderberg, EB Smeland, JH Myklebust, MP Oksvold. 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
51 X Dai, X Zhang, W Chen, Y Chen, Q Zhang, S Mo, J Lu. Dihydroartemisinin: a potential natural anticancer drug. Int J Biol Sci, 2021, 17( 2): 603– 622
https://doi.org/10.7150/ijbs.50364
52 C Shi, H Li, Y Yang, L Hou. Anti-inflammatory and immunoregulatory functions of artemisinin and its derivatives. Mediators Inflamm, 2015, 2015 : 435713
https://doi.org/10.1155/2015/435713
53 WL Zhou, JM Wu, QL Wu, JX Wang, Y Zhou, R Zhou, PL He, XY Li, YF Yang, Y Zhang, Y Li, JP Zuo. A novel artemisinin derivative, 3-(12-β-artemisininoxy) phenoxyl succinic acid (SM735), mediates immunosuppressive effects in vitro and in vivo. Acta Pharmacol Sin, 2005, 26( 11): 1352– 1358
https://doi.org/10.1111/j.1745-7254.2005.00232.x
54 ZS Yang, JX Wang, Y Zhou, JP Zuo, Y Li. Synthesis and immunosuppressive activity of new artemisinin derivatives. Part 2: 2-[12(β or α)-dihydroartemisinoxymethyl(or 1′-ethyl)]phenoxyl propionic acids and esters. Bioorg Med Chem, 2006, 14( 23): 8043– 8049
https://doi.org/10.1016/j.bmc.2006.07.038
55 JX Zhang, JX Wang, Y Zhang, JP Zuo, JM Wu, Y Sui, Y Li . Synthesis and immunosuppressive activity of new artemisinin derivatives containing polyethylene glycol group. Acta Pharmaceutica Sinica (Yao Xue Xue Bao) 2006; 41(1): 65–70 (in Chinese)
pmid: 16683530
56 LF Hou, SJ He, JX Wang, Y Yang, FH Zhu, Y Zhou, PL He, Y Zhang, YF Yang, Y Li, W Tang, JP Zuo. SM934, a water-soluble derivative of arteminisin, exerts immunosuppressive functions in vitro and in vivo. Int Immunopharmacol, 2009, 9( 13–14): 1509– 1517
https://doi.org/10.1016/j.intimp.2009.09.003
57 L Hou, KE Block, H Huang. Artesunate abolishes germinal center B cells and inhibits autoimmune arthritis. PLoS One, 2014, 9( 8): e104762
https://doi.org/10.1371/journal.pone.0104762
58 Y He, J Fan, H Lin, X Yang, Y Ye, L Liang, Z Zhan, X Dong, L Sun, H Xu. The anti-malaria agent artesunate inhibits expression of vascular endothelial growth factor and hypoxia-inducible factor-1α in human rheumatoid arthritis fibroblast-like synoviocyte. Rheumatol Int, 2011, 31( 1): 53– 60
https://doi.org/10.1007/s00296-009-1218-7
59 LF Hou, SJ He, X Li, Y Yang, PL He, Y Zhou, FH Zhu, YF Yang, Y Li, W Tang, JP Zuo. Oral administration of artemisinin analog SM934 ameliorates lupus syndromes in MRL/lpr mice by inhibiting Th1 and Th17 cell responses. Arthritis Rheum, 2011, 63( 8): 2445– 2455
https://doi.org/10.1002/art.30392
60 WD Li, YJ Dong, YY Tu, ZB 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, 2006, 6( 8): 1243– 1250
https://doi.org/10.1016/j.intimp.2006.03.004
61 Y Tang, J Liu, D Zhang, Z Xu, J Ji, C Wen. Cytokine storm in COVID-19: the current evidence and treatment strategies. Front Immunol, 2020, 11 : 1708
https://doi.org/10.3389/fimmu.2020.01708
62 M Gendrot, I Duflot, M Boxberger, O Delandre, P Jardot, M Le Bideau, J Andreani, I Fonta, J Mosnier, C Rolland, S Hutter, B La Scola, B Pradines. Antimalarial artemisinin-based combination therapies (ACT) and COVID-19 in Africa: in vitro inhibition of SARS-CoV-2 replication by mefloquine-artesunate. Int J Infect Dis, 2020, 99 : 437– 440
https://doi.org/10.1016/j.ijid.2020.08.032
63 G Li, M Yuan, H Li, C Deng, Q Wang, Y Tang, H Zhang, W Yu, Q Xu, Y Zou, Y Yuan, J Guo, C Jin, X Guan, F Xie, J Song. Safety and efficacy of artemisinin-piperaquine for treatment of COVID-19: an open-label, non-randomised and controlled trial. Int J Antimicrob Agents, 2021, 57( 1): 106216
https://doi.org/10.1016/j.ijantimicag.2020.106216
64 S Krishna, Y Augustin, J Wang, C Xu, HM Staines, H Platteeuw, A Kamarulzaman, A Sall, P Kremsner. Repurposing antimalarials to tackle the COVID-19 pandemic. Trends Parasitol, 2021, 37( 1): 8– 11
https://doi.org/10.1016/j.pt.2020.10.003
65 K Chen, H Hua, Z Zhu, T Wu, Z Jia, Q Liu. Artemisinin and dihydroartemisinin promote β-cell apoptosis induced by palmitate via enhancing ER stress. Apoptosis, 2020, 25( 3–4): 192– 204
https://doi.org/10.1007/s10495-019-01587-z
66 X Xue, Z Dong, Y Deng, S Yin, P Wang, Y Liao, G Hu, Y Chen. Dihydroartemisinin alleviates atopic dermatitis in mice by inhibiting mast cell infiltration. J South Med Univ (Nan Fang Yi Ke Da Xue Xue Bao) 2020; 40(10): 1480–1487 (in Chinese)
pmid: 33118501
67 X Nong, G Rajbanshi, L Chen, J Li, Z Li, T Liu, S Chen, G Wei, J Li. Effect of artesunate and relation with TGF-β1 and SMAD3 signaling on experimental hypertrophic scar model in rabbit ear. Arch Dermatol Res, 2019, 311( 10): 761– 772
https://doi.org/10.1007/s00403-019-01960-7
68 FM Yang, D Fan, XQ Yang, FH Zhu, MJ Shao, Q Li, YT Liu, ZM Lin, SQ Cao, W Tang, SJ He, JP Zuo. The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-κB/NLRP3 signaling. Acta Pharmacol Sin, 2021, 42( 4): 593– 603
https://doi.org/10.1038/s41401-020-0484-5
69 J Liu, E Manheimer, Y Shi, C Gluud. Chinese herbal medicine for severe acute respiratory syndrome: a systematic review and meta-analysis. J Altern Complement Med, 2004, 10( 6): 1041– 1051
https://doi.org/10.1089/acm.2004.10.1041
70 World Health Organization. SARS: clinical trials on treatment using a combination of traditional Chinese medicine and Western medicine. Geneva: World Health Organization, 2004
71 National Administration of Traditional Chinese Medicine . The Traditional Chinese Medicine Prevention Program of Influenza A (H1N1) (2009). National Administration of Traditional Chinese Medicine, 2009 (in Chinese)
72 H Luo, QL Tang, YX Shang, SB Liang, M Yang, N Robinson, JP Liu. Can Chinese medicine be used for prevention of corona virus disease 2019 (COVID-19)? A review of historical classics, research evidence and current prevention programs.. Chin J Integr Med, 2020, 26( 4): 243– 250
https://doi.org/10.1007/s11655-020-3192-6
[1] Yves Ingenbleek. Plasma transthyretin is a nutritional biomarker in human morbidities[J]. Front. Med., 2022, 16(4): 540-550.
[2] Xiang Wang, Minghui Wang, Lin Feng, Jie Song, Xin Dong, Ting Xiao, Shujun Cheng. Four-protein model for predicting prognostic risk of lung cancer[J]. Front. Med., 2022, 16(4): 618-626.
[3] Yaru Tian, Hairong Tian, Xiaoyang Zhai, Hui Zhu, Jinming Yu. Bevacizumab in combination with pemetrexed and platinum for elderly patients with advanced non-squamous non-small-cell lung cancer: a retrospective analysis[J]. Front. Med., 2022, 16(4): 610-617.
[4] Pooria Safarzadeh Kozani, Pouya Safarzadeh Kozani, Fatemeh Rahbarizadeh. CAR T cells redirected against tumor-specific antigen glycoforms: can low-sugar antigens guarantee a sweet success?[J]. Front. Med., 2022, 16(3): 322-338.
[5] Yi Zhang, Haocheng Zhang, Wenhong Zhang. SARS-CoV-2 variants, immune escape, and countermeasures[J]. Front. Med., 2022, 16(2): 196-207.
[6] Yiming Shao, Yingqi Wu, Yi Feng, Wenxin Xu, Feng Xiong, Xinxin Zhang. SARS-CoV-2 vaccine research and immunization strategies for improved control of the COVID-19 pandemic[J]. Front. Med., 2022, 16(2): 185-195.
[7] Ying Deng, Zhaowei Sun, Lei Wang, Minghui Wang, Jie Yang, Genxi Li. Biosensor-based assay of exosome biomarker for early diagnosis of cancer[J]. Front. Med., 2022, 16(2): 157-175.
[8] Wei Zhang, Xiaoguang Xu, Ziyu Fu, Jian Chen, Saijuan Chen, Yun Tan. PathogenTrack and Yeskit: tools for identifying intracellular pathogens from single-cell RNA-sequencing datasets as illustrated by application to COVID-19[J]. Front. Med., 2022, 16(2): 251-262.
[9] Xiaoguang Xu, Wei Zhang, Mingquan Guo, Chenlu Xiao, Ziyu Fu, Shuting Yu, Lu Jiang, Shengyue Wang, Yun Ling, Feng Liu, Yun Tan, Saijuan Chen. Integrated analysis of gut microbiome and host immune responses in COVID-19[J]. Front. Med., 2022, 16(2): 263-275.
[10] Jing Wang, Zequn Lu, Meng Jin, Ying Wang, Kunming Tian, Jun Xiao, Yimin Cai, Yanan Wang, Xu Zhang, Tao Chen, Zhi Yao, Chunguang Yang, Renli Deng, Qiang Zhong, Xiongbo Deng, Xin Chen, Xiang-ping Yang, Gonghong Wei, Zhihua Wang, Jianbo Tian, Xiao-ping Chen. Clinical characteristics and risk factors of COVID-19 patients with chronic hepatitis B: a multi-center retrospective cohort study[J]. Front. Med., 2022, 16(1): 111-125.
[11] Huai-yu Wang, Suyuan Peng, Zhanghui Ye, Pengfei Li, Qing Li, Xuanyu Shi, Rui Zeng, Ying Yao, Fan He, Junhua Li, Liu Liu, Shuwang Ge, Xianjun Ke, Zhibin Zhou, Gang Xu, Ming-hui Zhao, Haibo Wang, Luxia Zhang, Erdan Dong. Renin--angiotensin system inhibitor is associated with the reduced risk of all-cause mortality in COVID-19 among patients with/without hypertension[J]. Front. Med., 2022, 16(1): 102-110.
[12] Zehong Huang, Yingying Su, Tianying Zhang, Ningshao Xia. A review of the safety and efficacy of current COVID-19 vaccines[J]. Front. Med., 2022, 16(1): 39-55.
[13] Yuntao Zhang, Yunkai Yang, Niu Qiao, Xuewei Wang, Ling Ding, Xiujuan Zhu, Yu Liang, Zibo Han, Feng Liu, Xinxin Zhang, Xiaoming Yang. Early assessment of the safety and immunogenicity of a third dose (booster) of COVID-19 immunization in Chinese adults[J]. Front. Med., 2022, 16(1): 93-101.
[14] Wenjing Zhou, Jing Zhang, Mingkun Yan, Jin Wu, Shuo Lian, Kang Sun, Baiqing Li, Jia Ma, Jun Xia, Chaoqun Lian. Orlistat induces ferroptosis-like cell death of lung cancer cells[J]. Front. Med., 2021, 15(6): 922-932.
[15] Hongyun Zhao, Fan Luo, Jinhui Xue, Su Li, Rui-Hua Xu. Emerging immunological strategies: recent advances and future directions[J]. Front. Med., 2021, 15(6): 805-828.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed