Academic literature on the topic 'Yan ye yin hang'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Yan ye yin hang.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Yan ye yin hang"

1

Yang, Xiangcai, Yan Dai, Qinglin Ding, Feng Du, Jinhua Li, Chuhe Liu, Chunyang Lv, et al. "Abstract 6053: Mechanism of action of tumor-selective, chaperone-mediated protein degraders (CHAMPs)." Cancer Research 84, no. 6_Supplement (March 22, 2024): 6053. http://dx.doi.org/10.1158/1538-7445.am2024-6053.

Full text
Abstract:
Abstract HSP90 mediates the folding of many important cancer-associated proteins, but it can also direct its substrates towards degradation via the ubiquitin-proteasome system. Furthermore, in tumor tissues, HSP90 complexes are in an activated state relative to normal tissues, and small molecule HSP90 inhibitors display unique tumor-selective pharmacokinetics. To take advantage of these attributes, we have developed a novel targeted protein degradation technology, termed Chaperone-Mediated Protein Degradation (CHAMP), and present here an in-depth characterization of the CHAMP mechanism of action. Initially, from a chemical library of greater than 1000 linkered HSP90 binders, hetero-bifunctional CHAMPs were synthesized in which target protein binders and HSP90 binders were covalently coupled together by short linkers. The resulting compounds were screened for target protein degradation and cancer cell cytotoxicity to identify promising leads for further optimization. We found that CHAMPs can degrade a wide variety of target proteins. This included proteins that are known to be regulated by HSP90, such as transcription factor BRD4 or ERK5 kinase. However, proteins that are normally independent of HSP90 function can also be degraded, including mutated KRAS and SHP2 phosphatase. In vitro, CHAMP treatment of cells resulted in formation of a ternary complex between the target protein, CHAMP compound and HSP90. Moreover, an X-ray crystal structure was solved for a mKRAS-CHAMP-HSP90 ternary complex. CHAMP-mediated degradation required both the target- and HSP90-binding moieties to be covalently coupled and involved ubiquitination of the target protein. Multiple ubiquitin E3 ligases were present in ternary complexes, and depending on the target protein, NEDD8 inhibition or CRISPR knockouts of individual E3 ligases could suppress proteasome-dependent target degradation. In vivo, irrespective of target, CHAMPs displayed prolonged exposure in tumors relative to plasma and normal tissues, resulting in prolonged target degradation in tumors and strong tumor growth inhibition at well-tolerated doses. CHAMP technology can be applied to a diversity of cancer-associated targets and has potential advantages relative to other protein degradation approaches, including an improved safety margin due to preferential accumulation in tumor tissues. Citation Format: Xiangcai Yang, Yan Dai, Qinglin Ding, Feng Du, Jinhua Li, Chuhe Liu, Chunyang Lv, Liang Ma, Thomas L. Prince, Yuetong Sun, Mingkai Wang, Rong Wang, Yaya Wang, Zhiyong Wang, Min Wu, Mengmeng Xu, Zimo Yang, Long Ye, Wei Yin, Chenghao Ying, Haoxin Zhou, Guoqiang Wang, Weiwen Ying, Kevin P. Foley. Mechanism of action of tumor-selective, chaperone-mediated protein degraders (CHAMPs) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 6053.
APA, Harvard, Vancouver, ISO, and other styles
2

Zhan, Yan, and Ji-Ye Yin. "Abstract LB548: Genomic analysis of antineoplastic drug-related genetic variations based on large-scale population sequencing." Cancer Research 82, no. 12_Supplement (June 15, 2022): LB548. http://dx.doi.org/10.1158/1538-7445.am2022-lb548.

Full text
Abstract:
Abstract Purpose: Cancer has become the second leading cause of death in the world. In 2020, there were 19.29 million new cancer patients and a total of 10 million deaths. Drug are the most popular ways to control the development of cancer, but the effectiveness of anti-tumor drugs is only about 25%. Drug efficacy is affected by many factors, among which the most critical is genetic variants. Genomic analysis based on large-scale population sequencing was performed to calculate the mutation frequency of antineoplastic drug-related genetic variations and systematically analyze general descriptions and population differences. Methods: Information of antineoplastic drugs is available from FDA and CFDA, and gene list comes from DrugBank, PharmGKB, FDA biomarker list and clinical guidelines. The maximum likelihood method was used to estimate the population mutation frequency of a single locus, and the calculation process was completed on Ali Cloud platform. GATK, ANNOVAR, PROVEAN, SIFT and other software were used for quality control, annotation and function prediction of mutations. Perl, Python, R and other computer languages were used for statistical analysis. Results: we collect 206,604 samples from mainland China for analysis, and identified 104 antineoplastic drugs and 517 drug-related genes at the same time. There were 201,774 genetic variations, most of which were intron mutations, exon mutations account for about 1% of all mutations. We found 41,955 novel mutations, which unrecorded in dbSNP, accounting for about 20.8%. Most of these mutations were rare mutations with frequencies less than 1%, subsequent prediction of mutation function also indicated that a large proportion of these mutations were deleterious. These results suggest that rare mutations may also play an important role in the use of antineoplastic drugs, which has previously been underestimated. The mutation frequency distribution of different types of genes suggests that transporters have a greater impact on personalized medicine during the use of antineoplastic drugs in China. By analyzing the genetic variation of important metabolic enzymes, we found that mutations such as UGT1A1*6, DPYD*9A, CYP2D6*2 and *10 have a high frequency and high detection probability in the Chinese population. Therefore, when patients use drugs metabolized by related metabolic enzymes, genetic testing should be done in advance to prevent the occurrence of adverse reactions. Regional analysis revealed that the overall differences among provinces in China were small, but the differences were large compared with other ethnic groups. Conclusion: Results of large-scale population sequencing reveal distribution and discipline of genetic variation of antineoplastic drug-related genes in Chinese population, further confirm the importance of precision medicine in cancer, and illustrate that the use of antineoplastic drugs can also be affected by ethnic differences. Citation Format: Yan Zhan, Ji-Ye Yin. Genomic analysis of antineoplastic drug-related genetic variations based on large-scale population sequencing [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr LB548.
APA, Harvard, Vancouver, ISO, and other styles
3

Pirrone, Noemi, Sara Garcia Ballesteros, Simelys Hernandez, and Federico Bella. "Electrochemical Ammonia Production from Wastewater in a Flow Cell Reactor." ECS Meeting Abstracts MA2023-02, no. 47 (December 22, 2023): 2322. http://dx.doi.org/10.1149/ma2023-02472322mtgabs.

Full text
Abstract:
Although its high carbon footprint, the Haber-Bosh (HB) process mainly dominates the NH3 production market. Indeed, H2 is mainly produced by fossil fuels steam reforming processes and the catalytic reaction between N2 and H2 requires high energy input, being carried out at high temperatures and pressures. Moreover, it should be considered that HB plants are not evenly distributed in the world, thus NH3 and its derivates transportation to the final user is a large portion of the overall environmental impact.1 Moreover, as the population is growing, the NH3 demand is increasing fast to sustain the massive fertilizer utilization. For these reasons, in the last years, the research has focused on the possibility of producing ammonia from direct nitrogen electrochemical reduction (NRR) in aqueous electrolytes under ambient conditions exploiting renewable energy. This process is limited by low selectivity at high current densities and low yield, due to the high dissociation energy of the N2 triple bond and the unavoidable hydrogen evolution reaction (HER).2 On the other hand, NO3 - can be easily converted into NH3 thanks to the lower activation energy, which makes the reaction thermodynamically favoured compared to N2 reduction.3 Due to the massive agriculture, NO3 - is one of the most abundant contaminants of underground waters and high levels in the human body can cause many diseases. Thus, the use of NO3 - to produce NH3 under ambient conditions can be carried out with lower energy consumption, but can also address the water pollution issue.4 The aim of our work is to assess the catalytic activity of commercial MoS2 and a synthesised Bi-based catalyst in a gas-diffusion electrode flow cell of 10 cm2 geometrical area. Such a setup has the advantage of guaranteeing a better mass transport of the active species and of being scaled up. The electrodes are mainly made by airbrush deposition of a catalyst ink on a carbon paper support, which allows obtaining a high electrochemical active surface as a result of its high porosity. Catalyst loading and NO3 - concentration was varied to find the optimal condition in terms of Faraday efficiency (FE) and yield. MoS2 catalyst showed good catalytic activity towards NH3, with a FE between 62%-77% and yield between 2.9-13 mmol g-1 h-1. On the other hand, synthesised Bi material showed 21% FE and 0.41 mmol h-1 g-1, probably due to Bi final oxidation state, which is not so active towards NH3 production. NH3 quantification has been carried out through UV-vis colourimetric method, using both Nessler’s and Berthelot’s reagents. Even if the concentration of NH3 is high enough to exclude that it can derive from contamination, more sensitive quantification is needed for these experiments, together with the quantification of side products, such as NO2 -, H2 and N2. References 1. M. A. Mushtaq, M. Arif, G. Yasin, M. Tabish, A. Kumar, S. Ibraheem, W. Ye, S. Ajmal, J. Zhao, P. Li, J. Liu, A. Saad, X. Fang, X. Cai, S. Ji and D. Yan, Renew. Sustain. Energy Rev., 2023, 176, 113197. 2. H. Iriawan, S. Z. Andersen, X. Zhang, B. M. Comer, J. Barrio, P. Chen, A. J. Medford, I. E. L. Stephens, I. Chorkendorff and Y. Shao-Horn, Nat. Rev. Methods Prim. 2021 11, 2021, 1, 1–26. 3. Q. Liu, Q. Liu, L. Xie, Y. Ji, T. Li, B. Zhang, N. Li, B. Tang, Y. Liu, S. Gao, Y. Luo, L. Yu, Q. Kong and X. Sun, ACS Appl. Mater. Interfaces, 2022, 14, 17312–17318. 4. G. F. Chen, Y. Yuan, H. Jiang, S. Y. Ren, L. X. Ding, L. Ma, T. Wu, J. Lu and H. Wang, Nat. Energy 2020 58, 2020, 5, 605–613. This project has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. 948769, project title: SuN2rise).
APA, Harvard, Vancouver, ISO, and other styles
4

Peng, Zhi, Hua Wang, Baorui Liu, Huiting Xu, Zhenyang Liu, Tianshu Liu, Jun Zhang, et al. "Abstract CT152: A multicenter Phase II study of savolitinib in patients with MET-amplified gastroesophogeal junction adenocarcinomas or gastric cancer." Cancer Research 83, no. 8_Supplement (April 14, 2023): CT152. http://dx.doi.org/10.1158/1538-7445.am2023-ct152.

Full text
Abstract:
Abstract Background: MET gene amplification is associated with poor prognosis in gastric cancer (GC) and gastroesophageal junction adenocarcinomas (GEJ). Savolitinib is a potent and highly selective oral MET tyrosine-kinase inhibitor. Here we reported the preliminary efficacy and safety from a phase 2 trial of savolitinib monotherapy in patients (pts) with MET-amplified advanced or metastatic GC/GEJ. (NCT04923932). Methods: Eligible pts had 2L+ GEJ or GC, with MET amplification and measurable lesions. Pts received savolitinib at 600 mg QD for body weight (BW) ≥50 kg, while 400 mg QD for BW <50 kg in 21-day cycles until disease progression or meeting other criteria for end of treatment. Savolitinib BID regimen has also been additionally explored. The primary endpoint was objective overall response rate (ORR) evaluated by Independent Review Committee (IRC). One interim analysis (IA) was pre-defined at the first 20 QD pts who had at least 2 tumor assessments. Results: As of IA, 20 pts were enrolled for QD regimen. Demographics and clinical outcomes are shown in table 1. The mean relative dose intensity of 93.07%. Median duration of exposure was 2.09 months. Confirmed ORR by IRC was 45%, and reached 50% in 16 patients with MET GCN (high) while only 1 PR was observed in 4 patients with MET GCN (low). Duration of response rate at 4-month was 85.7% with median follow up time of 5.5 months. The most common Gr≥3 TRAE (≥5%) were platelet count decreased, hypersensitivity, anemia, neutropenia and hepatic function abnormal. In all pts, only 1 patient discontinued treatment due to grade 4 liver function abnormal (TRAE) and no patient died due to TRAE. Conclusion: Savolitinib monotherapy had manageable safety and showed promising efficacy in pts with MET-amplified GEJ or GC, particularly in pts with MET high GCN. BID regimen is being investigated to further evaluate the efficacy and safety of savolitinib in pts with MET high GCN. Table 1. Pts baseline characteristics and clinical efficacy Baseline Characteristics ITT in IA (n=20) Median age (min, max), yearsSex (male/female), nECOG (0/1/2)Median BMI (min, max), (kg/m2)Primary location of tumor (GC/GEJ)Tumor stage (IV)Prior line of therapy (1/2/≥3)MET GCN (high/low) 57.00 (39.5, 76.8)17/33/15/220.8 (14.9, 25.8)16/4205/10/516/4 Clinical Efficacy By IRC By Investigator Confirmed objective response rateDisease control rate4m-DoR rate,% (95% CI) 45%65%85.7 (33.4, 97.9) 40%55%71.4 (25.8, 92.0) Citation Format: Zhi Peng, Hua Wang, Baorui Liu, Huiting Xu, Zhenyang Liu, Tianshu Liu, Jun Zhang, Yuxian Bai, Ying Yuan, Tao Wu, Feng Ye, Qinghua Pan, Jufeng Wang, Enxiao Li, Diansheng Zhong, Yueyin Pan, Yanru Qin, Yan Yang, Yusheng Wang, Aiping Zhou, Yongshun Chen, Dianbao Zhang, Hongli Liu, Xiujuan Qu, Shubin Wang, Ning Liu, Jinsheng Wu, Wei Li, Kejun Nan, Hongming Pan, Jianming Xu, Chunmei Bai, Heling Liu, Jia Wei, Runzhi Chen, Rongrong Li, Wei Li, Jinghong Zhou, Hongyan Yin, Qian Xu, Songhua Fan, Yongxin Ren, Weiguo Su, Lin Shen. A multicenter Phase II study of savolitinib in patients with MET-amplified gastroesophogeal junction adenocarcinomas or gastric cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT152.
APA, Harvard, Vancouver, ISO, and other styles
5

Dogan, Deniz, Burkhard Hecker, Hermann Tempel, and Rüdiger-A. Eichel. "Experimental and Theoretical Investigations of Shunt Currents between Alkaline Water Electrolyzers." ECS Meeting Abstracts MA2023-02, no. 24 (December 22, 2023): 1331. http://dx.doi.org/10.1149/ma2023-02241331mtgabs.

Full text
Abstract:
A pivotal component of national climate strategies is the transition of the energy sector from fossil fuels to green energy. In this context, electrochemical processes will play a significant role in the future, requiring electrochemical systems with high efficiency and maximum system life cycles.1,2 For industrial electrochemical systems capital and operating costs are often reduced by a system design with a shared and circulating electrolyte supply providing ionically conductive cell-to-cell pathways. Under such conditions, parasitic ion migration occurs between adjacent cells, known as shunt currents.3,4 Shunt currents can have severe implications, such as decrease in faraday efficiency, material corrosion or interferences with instrumentation 3,5,6. To ensure high efficiency and maximum system life cycle, shunt current estimation is of high importance for the design of electrochemical multi-cell systems. A widely used approach in the literature is the theoretical, model-based approach using equivalent circuit models for shunt current determination7–9. Only a few publications demonstrate experimental determination methods 10,11. Furthermore, most shunt current studies in the field of electrochemistry focus on redox flow batteries. This work shows an innovative experimental approach for direct shunt current measurements between two alkaline water electrolysis cells with shared and circulating electrolyte feed under various conditions. The flow field design enables the insertion of reference electrodes into the flow cells for direct measurement of the potential differences between adjacent electrodes. Combined with the experimental data of the ionic tube resistances, the cell-to-cell shunt currents were accurately determined. Furthermore, an equivalent circuit model was created, fed with experimental data and validated with measured results. After successful validation the model was extended to electrolysis systems with more than two cells. Experimental data and simulations are in good agreement. The conducted experiments show the impact of temperature, cell voltage and tube manifold geometry on shunt current formation between alkaline water electrolyzers. Simulations performed are carried out to calculate shunt currents as a function of these parameters in large multi-cell systems. Furthermore, efficiency losses and corrosion processes as a result of shunt currents are estimated based on the results of this work. Literature Baños R, Manzano-Agugliaro F, Montoya FG, Gil C, Alcayde A, Gómez J. Optimization methods applied to renewable and sustainable energy: A review. Renew Sustain Energy Rev. 2011;15(4):1753-1766. doi:10.1016/j.rser.2010.12.008 Yan Z, Hitt JL, Turner JA, Mallouk TE. Renewable electricity storage using electrolysis. Proc Natl Acad Sci U S A. 2020;117(23):12558-12563. doi:10.1073/pnas.1821686116 Delgado NM, Monteiro R, Cruz J, Bentien A, Mendes A. Shunt currents in vanadium redox flow batteries – a parametric and optimization study. Electrochim Acta. 2022;403:139667. doi:10.1016/j.electacta.2021.139667 Kaminski EA, Savinell RF. A Technique for Calculating Shunt Leakage and Cell Currents in Bipolar Stacks Having Divided or Undivided Cells. J Electrochem Soc. 1983;130(5):1103-1107. doi:10.1149/1.2119891 Yin C, Guo S, Fang H, Liu J, Li Y, Tang H. Numerical and experimental studies of stack shunt current for vanadium redox flow battery. Appl Energy. 2015;151:237-248. doi:10.1016/j.apenergy.2015.04.080 Pletcher D, Walsh FC. Industrial Electrochemistry. Springer Science & Business Media; 2012. Schaeffer JA, Chen L Der, Seaba JP. Shunt current calculation of fuel cell stack using Simulink®. J Power Sources. 2008;182(2):599-602. doi:10.1016/j.jpowsour.2008.04.014 Wandschneider FT, Röhm S, Fischer P, Pinkwart K, Tübke J, Nirschl H. A multi-stack simulation of shunt currents in vanadium redox flow batteries. J Power Sources. 2014;261:64-74. doi:10.1016/j.jpowsour.2014.03.054 Ye Q, Hu J, Cheng P, Ma Z. Design trade-offs among shunt current, pumping loss and compactness in the piping system of a multi-stack vanadium flow battery. J Power Sources. 2015;296:352-364. doi:10.1016/j.jpowsour.2015.06.138 Rous̆ar I, Cezner V. Experimental Determination and Calculation of Parasitic Currents in Bipolar Electrolyzers with Application to Chlorate Electrolyzer. J Electrochem Soc. 1974;121(5):648. doi:10.1149/1.2401878 Fink H, Remy M. Shunt currents in vanadium flow batteries: Measurement, modelling and implications for efficiency. J Power Sources. 2015;284:547-553. doi:10.1016/j.jpowsour.2015.03.057
APA, Harvard, Vancouver, ISO, and other styles
6

藍日昌, 藍日昌. "筆談:同文異音下的東亞文化交流." 中正漢學研究 35, no. 35 (June 2020): 189–208. http://dx.doi.org/10.53106/2306036020200600350007.

Full text
Abstract:
<p>近代所謂漢字文化圈者,意謂在東亞區域之內諸國受中國文化之影響,舉凡儒學、佛教及技術等,皆由中國而向外傳至日、韓、琉及越南等,東亞諸國的文化交流的媒介自然是以漢字為主。</p> <p>東亞交流雖自六朝開始,但當時並無官方語音的觀念,因此交流之時自以當時主流發音為主,而主流發音則隨政治、經濟形勢而變。六朝時以南方吳音為主流,唐時以河洛及西北方音為主流,南宋時則以蘇杭音為主流。</p> <p>書寫文字雖同,但音調則有差異,這對其他諸國而言,也是有所困擾,音調雖有變化,但書寫則不變,筆談即是東亞交流中溝通的媒介。</p> <p>明太祖所建立起朝貢制度,政治及經濟來往熱絡,朝鮮作為明朝與日本的紐帶,明、朝之間的燕行使與朝、日之間的通信使,交往之時大體透過筆談溝通。甚至越南與琉球、日本交往之時亦復透過筆談溝通。</p> <p>筆談之事,少見於唐宋,入明則筆談的記錄頻率轉增,然則筆談出現的頻率則證諸東亞交流的狀況及文化交流的盛景,其事雖簡,其義則甚重大。</p> <p>&nbsp;</p><p>The so-called Chinese cultural circle in modern times means that all countries in the East Asian region are influenced by Chinese culture, and all Confucianism, Buddhism, and technology have been spread from China to Japan, South Korea, Vietnam, and Vietnam, and other countries in East Asia. The medium of cultural exchange is naturally based on Chinese characters.</p> <p>Although the East Asia exchange started from the Six Dynasties, there was no concept of official speech at that time. Therefore, the main stream of pronunciation was mainly from the time when the exchange was made, and the mainstream pronunciation changed with the political and economic situation. At the time of the Six Dynasties, Wu Yin was the mainstream in the South. In the Tang Dynasty, Heluo and Northwestern were the mainstream. In the Southern Song Dynasty, the Su Hang sound was the mainstream.</p> <p>Although the written words are the same, but the tones are different, this is also a nuisance to other countries. Although the tone changes, the writing remains the same. The pen talk is the medium of communication in East Asian communication.</p> <p>Ming Taizu established the tributary system, political and economic exchanges, North Korea as the ties between the Ming Dynasty and Japan, the Yan Dynasty between the Ming Dynasty and the DPRK to exercise communication with the DPRK and Japan, when the exchanges are generally communicated through the pen. Even when Vietnam communicates with Ryukyu and Japan, it communicates through pen talks.</p> <p>The things in pen talk are rare in the Tang and Song dynasties. The frequency of writing records in Ming Ming&rsquo;s writings has increased. However, the frequency of conversations in writing has confirmed the status of exchanges in East Asia and the grand scene of cultural exchanges. Although the matter is simple, its significance is very significant.</p> <p>&nbsp;</p>
APA, Harvard, Vancouver, ISO, and other styles
7

Yao, Herui, Min Yan, Zhongsheng Tong, Xinhong Wu, Min-Hee Ryu, Jee Hyun Kim, John Park, et al. "Abstract CT175: Safety, tolerability, pharmacokinetics, and antitumor activity of SHR-A1811 in HER2-expressing/mutated advanced solid tumors: A global phase 1, multi-center, first-in-human study." Cancer Research 83, no. 8_Supplement (April 14, 2023): CT175. http://dx.doi.org/10.1158/1538-7445.am2023-ct175.

Full text
Abstract:
Abstract Background: SHR-A1811 is an ADC comprised of a humanized anti-HER2 monoclonal antibody (trastuzumab), a cleavable linker, and a DNA topoisomerase I inhibitor payload. Here we assessed SHR-A1811 in HER2-expressing/mutated unresectable, advanced, or metastatic solid tumors. Methods: Pts were eligible if they had HER2 positive breast cancer (BC), HER2 positive gastric/GEJ carcinoma, HER2 low-expressing BC, HER2-expressing/mutated NSCLC, or other HER2-expressing/mutated solid tumors, and were refractory or intolerant to standard therapy. SHR-A1811 at doses of 1.0-8.0 mg/kg was given Q3W (IV). The primary endpoints were DLT, safety, and the RP2D. Results: From Sep 7, 2020 to Sep 28, 2022, 250 pts who had undergone a median of 3 prior treatment lines in the metastatic setting received at least one dose of SHR-A1811 in dose escalation, PK expansion, and indication expansion part. As of data cutoff on Sep 28, 2022, 1 pt experienced DLT. Treatment-related adverse events (TRAEs) were reported in 243 (97.2%) pts. Grade ≥3 TRAEs, serious TRAEs, and treatment-related deaths were reported in 131 (52.4%), 31 (12.4%), and 3 (1.2%) pts, respectively. Interstitial lung disease (AESI) was reported in 8 (3.2%) pts. Exposures of SHR-A1811, total antibody, and the payload were generally proportional to dose from 3.2 to 8.0 mg/kg. ORR was 61.6% (154/250, 95% CI 55.3-67.7) in all pts. Objective responses were observed in pts with HER2 positive BC (88/108, ORR 81.5%, 95% CI 72.9-88.3), HER2-low BC (43/77, ORR 55.8%, 95% CI 44.1-67.2), urothelial carcinoma (7/11), colorectal cancer (3/10), gastric/GEJ carcinoma (5/9), biliary tract cancer (5/8), NSCLC (1/3), endometrial cancer (1/2), and H&N cancer (1/1). Subgroup analyses of ORR are shown in Table 1. The 6-month PFS rate was 73.9% in all pts. Conclusions: SHR-A1811 was well-tolerated and showed promising antitumor activity in heavily pretreated advanced solid tumors. Table 1. Subgroup analyses of ORR No. of prior treatment lines in metastatic setting in all pts (N=250) HER2 positive BC (N=108) HER2-low BC (N=77) Other tumor types (N=65) ≤3 81.8% (45/55) 58.7% (27/46) 36.7% (18/49) &gt;3 81.1% (43/53) 51.6% (16/31) 31.3% (5/16) Prior anti-HER2 therapies in pts with BC (N=185)* HER2 positive BC (N=108) HER2-low BC (N=77) All BC (N=185) Any 82.2% (88/107, 73.7-89.0) 68.8% (11/16, 41.3-89.0) 80.5% (99/123, 72.4-87.1) Trastuzumab 81.9% (86/105, 73.2-88.7) 75.0% (9/12, 42.8-94.5) 81.2% (95/117, 72.9-87.8) Pertuzumab 83.0% (39/47, 69.2-92.4) 100% (5/5, 47.8-100) 84.6% (44/52, 71.9-93.1) Pyrotinib 86.9% (53/61, 75.8-94.1) 71.4% (5/7, 29.0-96.3) 85.3% (58/68, 74.6-92.7) Lapatinib 80.0% (28/35, 63.1-91.6) 100% (1/1, 2.5-100) 80.6% (29/36, 64.0-91.8) T-DM1 82.4% (14/17, 56.6-96.2) 100% (3/3, 29.2-100) 85.0% (17/20, 62.1-96.8) Other HER2-ADC (except T-DM1)** 60.0% (9/15, 32.3-83.7) 50.0% (2/4, 6.8-93.2) 57.9% (11/19, 33.5-79.8) ORR in pts with tumor types other than BC (N=65) HER2 IHC3+ or IHC2+/ISH+ (N=36) HER2 IHC2+/ISH- or IHC1+ or unknown (N=29) All other tumor types (N=65) % (n/N) 38.9% (14/36) 31.0% (9/29) 35.4% (23/65) ORR was shown as % (n/N, 95% CI) or % (n/N). *ORR is calculated using the number of subjects previously treated with anti-HER2 cancer therapy in advanced/metastatic setting as denominator; 2-sided 95% CIs are estimated using Clopper-Pearson method. **Includes RC48-ADC, A166, DP303c, MRG002, ARX788, TAA013, DX126-262, PF-06804103, and BAT8001. Citation Format: Herui Yao, Min Yan, Zhongsheng Tong, Xinhong Wu, Min-Hee Ryu, Jee Hyun Kim, John Park, Yahua Zhong, Weiqing Han, Caigang Liu, Mark Voskoboynik, Qun Qin, Jian Zhang, Minal Barve, Ana Acuna-Villaorduna, Vinod Ganju, Seock-Ah Im, Changsheng Ye, Yongmei Yin, Amitesh C. Roy, Li-Yuan Bai, Yung-Chang Lin, Chia-Jui Yen, Hui Li, Ki Young Chung, Shanzhi Gu, Jun Qian, Yuee Teng, Yiding Chen, Yu Shen, Kaijing Zhao, Shangyi Rong, Xiaoyu Zhu, Erwei Song. Safety, tolerability, pharmacokinetics, and antitumor activity of SHR-A1811 in HER2-expressing/mutated advanced solid tumors: A global phase 1, multi-center, first-in-human study [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT175.
APA, Harvard, Vancouver, ISO, and other styles
8

Iqra Arshad, Hifza Iqbal, Syeda Saira Iqbal, Muhammad Afzaal, and Yasir Rehman. "A Review on the Synergistic Approaches for Heavy Metals Bioremediation: Harnessing the Power of Plant-Microbe Interactions." Lahore Garrison University Journal of Life Sciences 8, no. 2 (June 29, 2024): 268–86. http://dx.doi.org/10.54692/lgujls.2024.0802343.

Full text
Abstract:
Heavy metals contamination is a serious threat to all life forms. Long term exposure of heavy metals can lead to different life-threatening medical conditions including cancers of different body parts. Phytoremediation and bioremediation offer a potential eco-friendly solution to such problems. Different microbes can interact with heavy metals in a variety of ways such as biotransformation, oxidation/reduction, and biosorption. Phytoremediation of the heavy metals using plants mostly involves rhizofilteration, phytoextraction, phytovolatization, and Phyto stabilization. A synergistic approach using both plants and microbes has proven much more efficient as compared to the individual applications of microbes or plants. This article aims to highlight the synergistic methods used in bioremediation, emphasizing the potent collaboration between bacteria and plants for environmental cleaning, along with the discussion of the importance of site-specific variables and potential constraints. While identifying the necessity for all-encompassing solutions, this review places emphasis on the combination of methodologies as a multifarious rehabilitation approach. This discussion offers insightful suggestions for scholars, scientists and decision-makers about the sustainable recovery of heavy metal-contaminated environments using a comprehensive strategy. REFERENCES Ankit, Bauddh K, Korstad J (2022). Phycoremediation: Use of algae to sequester heavy metals. Hydrobiol. 1(3): 288-303. Arantza SJ, Hiram MR, Erika K, Chávez-Avilés MN, Valiente-Banuet JI, Fierros-Romero G (2022). Bio-and phytoremediation: Plants and microbes to the rescue of heavy metal polluted soils. SN Appl. Sci. 4(2): 59. Azubuike CC, Chikere CB, Okpokwasili GC (2016). Bioremediation techniques–classification based on site of application: principles, advantages, limitations and prospects. World J. Microbiol. Biotechnol. 32: 1-18. Berti WR, Cunningham SD (2000). Phytostabilization of metals. Phytoremediation of toxic metals: Using plants to clean up the environment. Wiley, New York. 71-88. Bingöl NA, Özmal F, Akın B (2017). Phytoremediation and biosorption potential of Lythrum salicaria for nickel removal from aqueous solutions. Pol. J. Environ. Stud. 26(6): 2479-2485. Chandra R, Saxena G, Kumar V (2015). Phytoremediation of environmental pollutants: an eco-sustainable green technology to environmental management, In Advances in biodegradation and bioremediation of industrial waste. 1-29. Chaudhary K, Agarwal S, Khan S (2018). Role of phytochelatins (PCs), metallothioneins (MTs), and heavy metal ATPase (HMA) genes in heavy metal tolerance, In Mycoremediation and Environmental Sustainability. Volume 2: 39-60. Choudhary M, Kumar R, Datta A, Nehra V, Garg N (2017). Bioremediation of heavy metals by microbes, In Bioremediation of salt affected soils: an Indian perspective. 233-255. Chugh M, Kumar L, Shah MP, Bharadvaja N (2022). Algal bioremediation of heavy metals: An insight into removal mechanisms, recovery of by-products, challenges, and future opportunities. Energy Nexus. 7:100129. Congeevaram S, Dhanarani S, Park J, Dexilin M, Thamaraiselvi K (2007). Biosorption of chromium and nickel by heavy metal resistant fungal and bacterial isolates. J. Hazard. Mat. 146(1-2): 270-277. Cristaldi A, Conti GO, Jho EH, Zuccarello P, Grasso A, Copat C, Ferrante M (2017). Phytoremediation of contaminated soils by heavy metals and PAHs. A brief review. Environ. Technol. Inno. 8: 309-326. Crusberg T, Mark S. (2000). Heavy metal remediation of wastewaters by microbial biotraps, In Springer. 123-137. Emenike CU, Jayanthi B, Agamuthu P, Fauziah S (2018). Biotransformation and removal of heavy metals: a review of phytoremediation and microbial remediation assessment on contaminated soil. Environ. Rev. 26(2): 156-168. Ghosh M, Singh S (2005). A review on phytoremediation of heavy metals and utilization of it’s by products. Asian J. Energy Environ. 6(4): 18. Guignardi Z, Schiavon M (2017). Biochemistry of plant selenium uptake and metabolism, In Selenium in plants: molecular, physiological, ecological and evolutionary aspects. 21-34. Hong-Bo S, Li-Ye C, Cheng-Jiang R, Hua L, Dong-Gang G, Wei-Xiang L (2010). Understanding molecular mechanisms for improving phytoremediation of heavy metal-contaminated soils. Crit. Rev. Biotechnol. 30(1): 23-30. Igiri BE, Okoduwa SI, Idoko GO, Akabuogu EP, Adeyi AO, Ejiogu IK (2018). Toxicity and bioremediation of heavy metals contaminated ecosystem from tannery wastewater: a review. J. Toxicol. 2018. Jabeen R, Ahmad A, Iqbal M (2009). Phytoremediation of heavy metals: physiological and molecular mechanisms. Bot. Rev. 75: 339-364. Joshi P, Swarup A, Maheshwari S, Kumar R, Singh N (2011). Bioremediation of heavy metals in liquid media through fungi isolated from contaminated sources. Indian J. Microbiol. 51: 482-487. Junaid M, Hashmi MZ, Tang YM, Malik RN, Pei,DS (2017). Potential health risk of heavy metals in the leather manufacturing industries in Sialkot, Pakistan. Sci. Rep. 7(1): 8848. Kapahi M, Sachdeva S (2019). Bioremediation options for heavy metal pollution. J. Health Pollut. 9(24): 191203. Lebeau T, Jézéquel K, Braud A (2011). Bioaugmentation-assisted phytoextraction applied to metal-contaminated soils: state of the art and future prospects, In Microbes and Microbial Technology: Agricultural and Environmental Applications. 229-266. Leong YK, Chang JS (2020). Bioremediation of heavy metals using microalgae: Recent advances and mechanisms. Bioresour.Technol. 303: 122886. Limmer M, Burken J (2016). Phytovolatilization of organic contaminants. Environ. Sci. Technol. 50(13): 6632-6643. Ma Y, Oliveira RS, Freitas H, Zhang C (2016). Biochemical and molecular mechanisms of plant-microbe-metal interactions: relevance for phytoremediation. Front. Plant Sci. 7: 918. Manzoor M, Gul I, Ahmed I, Zeeshan M, Hashmi I, Amin BAZ, Kallerhoff J, Arshad M (2019). Metal tolerant bacteria enhanced phytoextraction of lead by two accumulator ornamental species. Chemosphere. 227: 561-569. Mueller B, Rock S, Gowswami D, Ensley D (1999). Phytoremediation decision tree. Prepared by-Interstate Technology and Regulatory Cooperation Work Group. 1-36. Nies DH (1999). Microbial heavy-metal resistance. Appl. Microbiol. Biotechnol. 51: 730-750. Nies DH, Silver S (1995). Ion efflux systems involved in bacterial metal resistances. J. Ind. 14: 186-199. Pande V, Pandey SC, Sati D, Bhatt P, Samant M (2022). Microbial interventions in bioremediation of heavy metal contaminants in agroecosystem. Front. Microbiol. 13: 824084. Pandey VC, Bajpai O (2019). Phytoremediation: from theory toward practice, In Phytomanagement of polluted sites. 1-49. Robinson BH, Leblanc M, Petit D, Brooks RR, Kirkman JH, Gregg PE (1998). The potential of Thlaspi caerulescens for phytoremediation of contaminated soils. Plant Soil. 203: 47-56. Romantschuk M, Lahti-Leikas K, Kontro M, Allen JA, Sinkkonen A (2023). Bioremediation of contaminated soil and groundwater by in situ Front. Microbiol. 14: 1258148. Sabreena, Hassan S, Bhat SA, Kumar V, Ganai BA, Ameen F (2022). Phytoremediation of heavy metals: An indispensable contrivance in green remediation technology. Plants. 11(9): 1255. Saha L, Tiwari J, Bauddh K, Ma Y (2021). Recent developments in microbe–plant-based bioremediation for tackling heavy metal-polluted soils. Front. Microbiol. 12: 731723. Sharma I. (2020). Bioremediation techniques for polluted environment: concept, advantages, limitations, and prospects, In Trace metals in the environment-new approaches and recent advances. IntechOpen. Sharma JK, Kumar N, Singh NP, Santal, AR (2023). Phytoremediation technologies and their mechanism for removal of heavy metal from contaminated soil: An approach for a sustainable environment. Front. Plant Sci. 14: 1076876. Shen X, Dai M, Yang J, Sun L, Tan X, Peng C, Ali I, and Naz I (2022). A critical review on the phytoremediation of heavy metals from environment: Performance and challenges. Chemosphere. 291: 132979. Silver S (2011). BioMetals: a historical and personal perspective. Biometals. 24(3): 379-390. Silver S, Phung LT (2005). A bacterial view of the periodic table: genes and proteins for toxic inorganic ions. J. Ind. Microbiol. Biotechnol. 32: 587-605. Singh N, Santal AR (2015). Phytoremediation of heavy metals: the use of green approaches to clean the environment, In Phytoremediation: Management of Environmental Contaminants. Volume 2: 115-129. Strong PJ, Burgess JE (2008). Treatment methods for wine-related and distillery wastewaters: a review. Bioremediation J. 12(2): 70-87. Syranidou E, Christofilopoulos S, Gkavrou G, Thijs S, Weyens N, Vangronsveld J, Kalogerakis N (2016). Exploitation of endophytic bacteria to enhance the phytoremediation potential of the wetland helophyte Juncus acutus. Front. Microbiol. 7: 1016. Umrania VV (2006). Bioremediation of toxic heavy metals using acidothermophilic autotrophes. Bioresour. Technol. 97(10): 1237-1242. Valls M, De Lorenzo V (2002). Exploiting the genetic and biochemical capacities of bacteria for the remediation of heavy metal pollution. FEMS Microbiol. Rev. 26(4): 327-338. Verma P, George K, Singh H, Singh S, Juwarkar A, Singh R (2006). Modeling rhizofiltration: heavy-metal uptake by plant roots. Environ. Model. Assess. 11: 387-394. Wu Y, Li Z, Yang Y, Purchase D, Lu Y, Dai Z (2021). Extracellular polymeric substances facilitate the adsorption and migration of Cu2+ and Cd2+ in saturated porous media. Biomolecules. 11(11): 1715. Wuana RA, Okieimen FE (2011). Heavy metals in contaminated soils: a review of sources, chemistry, risks and best available strategies for remediation. International Scholarly Research Notices. Yan A, Wang Y, Tan SN, Mohd Yusof ML, Ghosh S, Chen Z (2020). Phytoremediation: a promising approach for revegetation of heavy metal-polluted land. Front. Plant Sci. 11: 359. Zhang Y, Hu J, Bai J, Wang J, Yin R, Wang J, and Lin X (2018). Arbuscular mycorrhizal fungi alleviate the heavy metal toxicity on sunflower (Helianthus annuus) plants cultivated on a heavily contaminated field soil at a WEEE-recycling site. Sci. Total Environ. 628: 282-290.
APA, Harvard, Vancouver, ISO, and other styles
9

Hong Son, Bui, Vu Van Nga, Le Thi Diem Hong, and Do Thi Quynh. "Potent Natural Inhibitors of Alpha-Glucosidase and the Application of Aspergillus spp. in Diabetes type 2 Drugs: a Review." VNU Journal of Science: Medical and Pharmaceutical Sciences 38, no. 1 (March 24, 2022). http://dx.doi.org/10.25073/2588-1132/vnumps.4334.

Full text
Abstract:
Diabetes Mellitus has been becoming a disease of the century, and disease incidence is still rising worldwide. It causes many serious complications, especially in the eye, heart, kidneys, brain, and vascular system, such as diabetic nephropathy, diabetic retinopathy, liver fa­ilure, etc. Moreover, the process of controlling this disease is complicated. Meanwhile, the antidiabetic drugs on the market are facing some problems with a wide range of adverse reactions. Therefore, finding new drugs to treat diabetes has always been a topic that many researchers are interested in, especially drugs derived from nature like microorganisms and medicinal plants. This review is to provide knowledge concerning the effects of α-glucosidase inhibitors, which are oral antidiabetic drugs commonly used for diabetes mellitus type 2. Besides, we show readers the variety of active ingredients originating from nature, particularly the secondary metabolites of Aspergillus spp., which have many applications in the chemical and medicinal industry. Keywords: Diabetes, α-glucosidase inhibitors, Aspergillus. References [1] W. H. Organization, Classification of Diabetes Mellitus, https://www.who.int/westernpacific/health-topics/diabetes (accessed on: May 11th, 2021).[2] J. Thrasher, Pharmacologic Management of Type 2 Diabetes Mellitus: Available Therapies, Am J Cardiol, Vol. 120, No. 1, 2017, pp. S4-S16, https://doi.org/10.1016/j.amjcard.2017.05.009.[3] W. Hakamata, M. Kurihara, H. Okuda, T. Nishio, T. Oku, Design and Screening Strategies for Alpha-glucosidase Inhibitors Based on Enzymological Information, Curr Top Med Chem, Vol. 9, No. 1, 2009, pp. 3-12, https://doi.org/10.2174/156802609787354306.[4] US, Patent Version Number: US4062950A, Amino Sugar Derivatives, https://patents.google.com/patent/US4062950A/en(accessed on: May 11th, 2021).[5] A. S. Dabhi, N. R. Bhatt, M. J. Shah, Voglibose: an Alpha- glucosidase Inhibitor, J Clin Diagn Res, Vol. 7, No. 12, 2013, pp. 3023-3027, https://doi.org/10.7860/JCDR/2013/6373.3838.[6] P. Durruty, M. Sanzana, L. Sanhueza, Pathogenesis of Type 2 Diabetes Mellitus, Type 2 Diabetes - from Pathophysiology to Modern Management, Intechopen, United Kingdom, 2019, pp. 1-18.[7] L. N. Khue, T. H. Dang, T. H. Quang, N. T. Khue et al., Guidelines for Diagnosis and Treatment of Diabetes Type 2, Ministry of Health, Vietnam, 2021 (in Vietnamese).[8] M. Okuyama, W. Saburi, H. Mori, A. Kimura, Alpha-Glucosidases and Alpha-1,4-Glucan Lyases: Structures, Functions, and Physiological Actions, Cell Mol Life Sci, Vol. 73, 2016, pp. 2727-2751, https://doi.org/10.1007/s00018-016-2247-5.[9] V. L. Yip, S. G. Withers, Nature's Many Mechanisms for The Degradation of Oligosaccharides, Org Biomol Chem, Vol. 19, No. 2, 2004, pp. 2707-2713, https://doi.org/10.1039/B408880H.[10] B. Henrissat, A. Bairoch, New Families in The Classification of Glycosyl Hydrolases Based on Amino Acid Sequence Similarities, Biochem J, Vol. 293, No. 3, 1993, pp. 781-788, https://doi.org/10.1042/bj2930781.[11] B. Henrissat, A Classification of Glycosyl Hydrolases Based on Amino Acid Sequence Similarities, Biochem J, Vol. 280, No. 2, 1991, pp. 309-316, https://doi.org/10.1042/bj2800309.[12] R. Gupta, P. Gigras, H. Mohapatra, V. K. Goswami, B. Chauhan, Microbial A-amylases: A Biotechnological Perspective, Process Biochemistry, Vol. 38, No. 11, 2003, pp. 1599-1616, https://doi.org/10.1016/s0032-9592(03)00053-0.[13] C. V. D. Maarel, B. V. D. Veen, J. C .M. Uitdehaag, H. Leemhuis, L. Dijkhuizen, Properties and Applications of Starch-Converting Enzymes of The A-Amylase Family, Journal of Biotechnology, Vol. 94, No. 2, 2002, pp. 137-155, https://doi.org/10.1016/s0168-1656(01)00407-2.[14] N. R. Kim, D. W. Jeong, D. S. Ko, J. H. Shim, Characterization of Novel Thermophilic Alpha-Glucosidase from Bifidobacterium Longum, Int J Biol Macromol, Vol. 99, 2017, pp. 594-599, https://doi.org/10.1016/j.ijbiomac.2017.03.009.[15] D. R. Rose, M. M. Chaudet, K. Jones, Structural Studies of The Intestinal Alpha-Glucosidases, Maltase-glucoamylase and Sucrase-isomaltase, J Pediatr Gastroenterol Nutr, Vol. 66, No. 3, 2018, pp. S11-S13, https://doi.org/10.1097/MPG.0000000000001953.[16] L. Ren, X. Qin, X. Cao, L. Wang, F. Bai, G. Bai, Y. Shen, Structural Insight into Substrate Specificity of Human Intestinal Maltase-Glucoamylase, Protein Cell, Vol. 2, 2011, pp. 827-836, https://doi.org/10.1007/s13238-011-1105-3.[17] L. Sim, C. Willemsma, S. Mohan, H. Y. Naim, B. M. Pinto, D. R. Rose, Structural Basis for Substrate Selectivity in Human Maltase-Glucoamylase and Sucrase-Isomaltase N-Terminal Domains, J Biol Chem, Vol. 285, No. 23, 2010, pp. 17763-17770, https://doi.org/10.1074/jbc.M109.078980.[18] K. Jones, L. Sim, S. Mohan, J. Kumarasamy,H. Liu, S. Avery, H. Y. Naim, R. Q. Calvillo, B. L. Nichols, B. M. Pinto, D. R. Rose, Mapping The Intestinal Alpha-Glucogenic Enzyme Specificities of Starch Digesting Mal se-Glucoamylase and Sucrase-Isomaltase, Bioorg Med Chem, Vol. 19, 2011, pp. 3929-3934, https://doi.org/10.1016/j.bmc.2011.05.033.[19] P. T. T. Chau, P. T. Nghia, Enzyme and Application, Education Publisher, Vietnam, 2009.[20] Researchgate, Food Protein-Derived Bioactive Peptides in Management of Type 2 Diabetes - Scientific Figure, https://www.researchgate.net/figure/Mechanism-of-action-of-alpha-glucosidase-inhibitors_fig2_279991207 (accessed on: May 10th, 2021).[21] Z. Liu, S. Ma, Recent Advances in Synthetic Alpha-Glucosidase Inhibitors, Chem Med Chem, Vol. 12, No. 11, 2017, pp. 819-829, https://doi.org/10.1002/cmdc.201700216.[22] A. Lee, P. Patrick, J. Wishart, M. Horowitz, J. E. Morley, The Effects of Miglitol on Glucagon-Like Peptide-1 Secretion And Appetite Sensations in Obese Type 2 Diabetics, Diabetes Obes Metab, Vol. 4, No. 5, 2002, pp. 329-335, https://doi.org/10.1046/j.14631326.2002.00219.x.[23] I. Takei, K. Miyamoto, O. Funae, N. Ohashi, S. Meguro, M. Tokui, T. Saruta, Secretion of GIP in Responders to Acarbose in Obese Type 2 (NIDDM) Patients, Journal of Diabetes and Its Complications, Vol. 15, No. 5, 2001, pp. 245-249, https://doi.org/10.1016/s1056-8727(01)00148-9.[24] X. Bian, X. Fan, C. Ke, Y. Luan, G. Zhao, A. Zeng, Synthesis and Alpha-Glucosidase Inhibitory Activity Evaluation of N-Substituted Aminomethyl-Beta-D-Glucopyranosides, Bioorg Med Chem, Vol. 21, No. 17, 2013, pp. 5442-5450, https://doi.org/10.1016/j.bmc.2013.06.002.[25] J. B. Yang, J. Y. Tian, Z. Dai, F. Ye, S. C. Ma, A. G. Wang, α-Glucosidase Inhibitors Extracted from The Roots of Polygonum Multiflorum Thunb, Fitoterapia, Vol. 117, 2017, pp. 65-70, https://doi.org/10.1016/j.fitote.2016.11.009.[26] Z. Yin, W. Zhang, F. Feng, Y. Zhang, W. Kang, α-Glucosidase Inhibitors Isolated from Medicinal Plants, Food Science and Human Wellness, Vol. 3, No.3-4, 2014, pp. 136-174, https://doi.org/10.1016/j.fshw.2014.11.003.[27] P. Qiu, Z. Liu, Y. Chen, R. Cai, G. Chen, Z. She, Secondary Metabolites with Alpha-Glucosidase Inhibitory Activity from The Mangrove Fungus Mycosphaerella sp. SYSU-DZG01, Mar Drugs, Vol. 17, No. 8, 2019, pp. 483-508, https://doi.org/10.3390/md17080483.[28] S. Munasaroh, S. R. Tamat, R. T. Dewi, Isolation and Identification of α-Glucosidase Inhibitor from Aspergillus Terreus F38, Indonesian Journal of Pharmacy, Vol. 29, No. 2, 2018, pp. 74-79, https://doi.org/10.14499/indonesianjpharm29iss2pp74.[29] R. T. Dewi, A. Suparman, H. Mulyani, P. D. N. Lotulung, Identification of A New Compound as α-Glucosidase Inhibitor from Aspergillus Aculeatus, Annales Bogorienses, Vol. 20, No. 1, 2016, pp. 19-23, https://doi.org/10.14203/ann. bogor .2016.v20.n1.19-23.[30] R. T. Dewi, S. Tachibana, A. Darmawan, Effect on α-Glucosidase Inhibition and Antioxidant Activities of Butyrolactone Derivatives from Aspergillus Terreus MC751, Medicinal Chemistry Research, Vol. 23, 2014, pp. 454-460, https://doi.org/10.1007/s00044-013-0659-4.[31] M. G. Kang, S. H. Yi, J. S. Lee, Production and Characterization of A New Alpha-Glucosidase Inhibitory Peptide from Aspergillus Oryzae N159-1, Mycobiology, Vol. 41, No. 3, 2013, pp. 149-154, https://doi.org/10.5941/MYCO.2013.41.3.149.[32] S. Onose, R. Ikeda, K. Nakagawa, T. Kimura, K. Yamagishi, O. Higuchi, T. Miyazawa, Production of The Alpha-Glycosidase Inhibitor 1-Deoxynojirimycin from Bacillus Species, Food Chem, Vol. 138, No. 1, 2013, pp. 516-523, https://doi.org/10.1016/j.foodchem.2012.11.012.[33] Y. P. Zhu, K. Yamaki, T. Yoshihashi, M. Ohnishi Kameyama, X. T. Li, Y. Q. Cheng, Y. Mori, L. T. Li, Purification and Identification of 1-Deoxynojirimycin (DNJ) in Okara Fermented by Bacillus Subtilis B2 from Chinese Traditional Food (Meitaoza), J Agric Food Chem, Vol. 58,No. 7, 2010, pp. 4097-4103, https://doi.org/10.1021/jf9032377.[34] A. Tabussum, N. Riaz, M. Saleem, M. Ashraf, M. Ahmad, U. Alam, B. Jabeen, A. Malik, A. Jabbar, α-Glucosidase Inhibitory Constituents from Chrozophora Plicata, Phytochemistry Letters, Vol. 6, No. 4. 2013, pp. 614-619, https://doi.org/10.1016/j.phytol.2013.08.005.[35] M. Yagi, T. Kouno, Y. Aoyagi, H. Murai, The Structure of Moranoline, A Piperidine Alkaloid from Morus Species, Journal of The Agricultural Chemical Society of Japan, Vol. 50, No. 11, 1976, pp. 571-572, https://doi.org/10.1271/nogeikagaku1924.50.11_571.[36] M. Hemker, A. Stratmann, K. Goeke, W. Schroder, J. Lenz, W. Piepersberg, H. Pape, Identification, Cloning, Expression, and Characterization of The Extracellular Acarbose-Modifying Glycosyltransferase, AcbD, from Actinoplanes Sp. Strain SE50, J Bacteriol, Vol. 183, No. 15, 2001, pp. 4484-4492, https://doi.org/10.1128/JB.183. 15.4484-4492.2001.[37] E. Truscheit, I. Hillebrand, B. Junge, L. Müller, W. Puls, D. Schmidt, Microbial α-Glucosidase Inhibitors: Chemistry, Biochemistry, and Therapeutic Potential, Presented at Drug Concentration Monitoring Microbial alpha-Glucosidase Inhibitors Plasminogen Activators, Springer-Verlag, Berlin, 1988.[38] Y. Kameda, N. Asano, M. Yoshikawa, M. Takeuchi, T. Yamaguchi, K. Matsui, S. Horii, H. Fukase, Valiolamine, A New Alpha-Glucosidase Inhibiting Aminocyclitol Produced by Streptomyces Hygroscopicus, J Antibiot (Tokyo), Vol. 37, No. 11, 1984, pp. 1301-1307, https://doi.org/10.7164/antibiotics.37.1301.[39] D. T. Tuyen, V. V. Hanh, V. T. T. Hang, D. K. Trinh, D. T. Quyen, Extraction and Purification of DNJ (1-Deoxynojirimycin) Inhibiting α-Glucosidase from B. Subtilis VN9 Strain Isolated from Vietnam, National Biotechnology Conference, 2013.[40] D. T. Tuyen, Optimization and Purification of α-Glucosidase Inhibitor from Bacillus Subtilis YT20 Isolated in Vietnam, Vietnam Journal of Science and Technology, Vol. 59, No. 2, 2021, pp. 179-188, https://doi.org/10.15625/2525-2518/ 59/2/14928.[41] S. E. Baker, J. W. Bennett, An Overview of the Genus Aspergillus, Aspergillus: Molecular Biology and Genomics, The Aspergilli, Taylor & Francis, United Kingdom, 2008, pp. 3-13.[42] H. C. Gugnani, Ecology and Taxonomy of Pathogenic Aspergilli, Front Biosci, Vol. 8, No. 6, 2003, pp. s346- s357, https://doi.org/10.2741/1002.[43] C. G. Shaw, The Genus Aspergillus, Science, Vol. 150, No. 3697, 1965, pp. 736-737, https://doi.org/10.1126/science.150.3697.736-a.[44] M. T. Hedayati, A. C. Pasqualotto, P. A. Warn, P. Bowyer, D. W. Denning, Aspergillus Flavus: Human Pathogen, Allergen and Mycotoxin Producer, Microbiology, Vol. 153, No. 6, 2007, pp. 1677-1692, https://doi.org/10.1099/mic.0.2007/007641-0.[45] T. R. Dagenais, N. P. Keller, Pathogenesis of Aspergillus Fumigatus in Invasive Aspergillosis, Clin Microbiol Rev, Vol. 22, No. 3, 2009, pp. 447-465, https://doi.org/10.1128/CMR.00055-08.[46] S. Amaike, N. P. Keller, Aspergillus Flavus, Annu Rev Phytopathol, Vol. 49, 2011, pp. 107-133, https://doi.org/10.1146/annurev-phyto-072910-095221.[47] J. Houbraken, R. P. De Vries, R. A. Samson, Modern Taxonomy of Biotechnologically Important Aspergillus and Penicillium Species, Adv Appl Microbiol, Vol. 86, 2014, pp. 199-249, https://doi.org/10.1016/B978-0-12-800262-9.00004-4.[48] E. Ichishima, Development of Enzyme Technology for Aspergillus Oryzae, A. Sojae, and A. Luchuensis, The National Microorganisms of Japan, Biosci Biotechnol Biochem, Vol. 80, No. 9, 2016, pp. 1681-1692, https://doi.org/10.1080/09168451.2016.1177445.[49] E. Schuster, N. Dunn-Coleman, J. C. Frisvad, P. W. Van Dijck, on The Safety of Aspergillus Niger-A Review, Appl Microbiol Biotechnol, Vol. 59, No. 4-5, 2002, pp. 426-435, https://doi.org/10.1007/s00253-002-1032-6.[50] J. H. Yu, N. Keller, Regulation of Secondary Metabolism in Filamentous Fungi, Annu Rev Phytopathol, Vol. 43, 2005, pp. 437-458, https://doi.org/10.1146/annurev.phyto.43.040204.140214.[51] J. F. Sanchez, A. D. Somoza, N. P. Keller, C. C. Wang, Advances in Aspergillus Secondary Metabolite Research in The Post-Genomic Era, Nat Prod Rep, Vol. 29, No. 3, 2012, pp. 351-371, https://doi.org/10.1039/c2np00084a.[52] J. W. Bennett, M. Klich, Mycotoxins, Clin Microbiol Rev, Vol. 16, 2003, pp. 497-516, https://doi.org/10.1128/cmr.16.3.497-516.2003.[53] Q. Zhou, J. K. Liao, Statins and cardiovascular Diseases: from Cholesterol Lowering to Pleiotropy, Curr Pharm Des, Vol. 15, No. 5, 2009, pp. 467-478, https://doi.org/10.2174/138161209787315684.[54] A. W. Alberts, Discovery, Biochemistry and Biology of Lovastatin, Am J Cardiol, Vol. 62, No. 15, 1988, pp. 10J-15J, https://doi.org/10.1016/0002-9149(88)90002-1.[55] H. Tomoda, Y. K. Kim, H. Nishida, R. Masuma, S. Omura, Pyripyropenes, Novel Inhibitors of Acyl-Coa: Cholesterol Acyltransferase Produced by Aspergillus Fumigatu- Production, Isolation, and Biological Properties, J Antibiot (Tokyo), Vol. 47, No. 2, 1994, pp. 148-153, https://doi.org/10.7164/antibiotics.47.148.[56] F. Pelaez, Biological Activities of Fungal Metabolites, Marcel Dekker, United Stated of America, 2004.[57] E. L. Dulaney, Penicillin Production by The Aspergillus Nidulans Group, Mycologia, Vol. 39, No. 5, 2018, pp. 582-586, https://doi.org/10.1080/00275514.1947.12017637.[58] T. T. Bladt, J. C. Frisvad, P. B. Knudsen, T. O. Larsen, Anticancer and Antifungal Compounds from Aspergillus, Penicillium and Other Filamentous Fungi, Molecules, Vol. 18, No. 9, 2013, pp. 11338-11376, https://doi.org/10.3390/molecules180911338.[59] Y. Wu, Y. Chen, X. Huang, Y. Pan, Z. Liu, T. Yan, W. Cao, Z. She, alpha-Glucosidase Inhibitors: Diphenyl Ethers and Phenolic Bisabolane Sesquiterpenoids from The Mangrove Endophytic Fungus Aspergillus Flavus QQSG-3, Mar Drugs, Vol. 16, No. 9, 2018, pp. 307-316, https://doi.org/10.3390/md16090307.
APA, Harvard, Vancouver, ISO, and other styles
10

Thanh Binh, Nguyen Thi, Nguyen Thi Hai Yen, Dang Kim Thu, Nguyen Thanh Hai, and Bui Thanh Tung. "The Potential of Medicinal Plants and Bioactive Compounds in the Fight Against COVID-19." VNU Journal of Science: Medical and Pharmaceutical Sciences 37, no. 3 (September 14, 2021). http://dx.doi.org/10.25073/2588-1132/vnumps.4372.

Full text
Abstract:
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus , is causing a serious worldwide COVID-19 pandemic. The emergence of strains with rapid spread and unpredictable changes is the cause of the increase in morbidity and mortality rates. A number of drugs as well as vaccines are currently being used to relieve symptoms, prevent and treat the disease caused by this virus. However, the number of approved drugs is still very limited due to their effectiveness and side effects. In such a situation, medicinal plants and bioactive compounds are considered a highly valuable source in the development of new antiviral drugs against SARS-CoV-2. This review summarizes medicinal plants and bioactive compounds that have been shown to act on molecular targets involved in the infection and replication of SARS-CoV-2. Keywords: Medicinal plants, bioactive compounds, antivirus, SARS-CoV-2, COVID-19 References [1] R. Lu, X. Zhao, J. Li, P. Niu, B. Yang, H. Wu et al., Genomic Characterisation and Epidemiology of 2019, Novel Coronavirus: Implications for Virus Origins and Receptor Binding, The Lancet, Vol. 395, 2020, pp. 565-574, https://doi.org/10.1016/S0140-6736(20)30251-8.[2] World Health Organization, WHO Coronavirus (COVID-19) Dashboard, https://covid19.who.int, 2021 (accessed on: August 27, 2021).[3] H. Wang, P. Yang, K. Liu, F. Guo, Y. Zhang et al., SARS Coronavirus Entry into Host Cells Through a Novel Clathrin- and Caveolae-Independent Endocytic Pathway, Cell Research, Vol. 18, No. 2, 2008, pp. 290-301, https://doi.org/10.1038/cr.2008.15.[4] A. Zumla, J. F. W. Chan, E. I. Azhar, D. S. C. Hui, K. Y. Yuen., Coronaviruses-Drug Discovery and Therapeutic Options, Nature Reviews Drug Discovery, Vol. 15, 2016, pp. 327-347, https://doi.org/10.1038/nrd.2015.37.[5] A. Prasansuklab, A. Theerasri, P. Rangsinth, C. Sillapachaiyaporn, S. Chuchawankul, T. Tencomnao, Anti-COVID-19 Drug Candidates: A Review on Potential Biological Activities of Natural Products in the Management of New Coronavirus Infection, Journal of Traditional and Complementary Medicine, Vol. 11, 2021, pp. 144-157, https://doi.org/10.1016/j.jtcme.2020.12.001.[6] R. E. Ferner, J. K. Aronson, Chloroquine and Hydroxychloroquine in Covid-19, BMJ, Vol. 369, 2020, https://doi.org/10.1136/bmj.m1432[7] J. Remali, W. M. Aizat, A Review on Plant Bioactive Compounds and Their Modes of Action Against Coronavirus Infection, Frontiers in Pharmacology, Vol. 11, 2021, https://doi.org/10.3389/fphar.2020.589044.[8] Y. Chen, Q. Liu, D. Guo, Emerging Coronaviruses: Genome Structure, Replication, and Pathogenesis, Medical Virology, Vol. 92, 2020, pp. 418‐423. https://doi.org/10.1002/jmv.25681.[9] B. Benarba, A. Pandiella, Medicinal Plants as Sources of Active Molecules Against COVID-19, Frontiers in Pharmacology, Vol. 11, 2020, https://doi.org/10.3389/fphar.2020.01189.[10] N. T. Chien, P. V. Trung, N. N. Hanh, Isolation Tribulosin, a Spirostanol Saponin from Tribulus terrestris L, Can Tho University Journal of Science, Vol. 10, 2008, pp. 67-71 (in Vietnamese).[11] V. Q. Thang Study on Extracting Active Ingredient Protodioscin from Tribulus terrestris L.: Doctoral dissertation, VNU University of Science, 2018 (in Vietnamese).[12] Y. H. Song, D. W. Kim, M. J. C. Long, H. J. Yuk, Y. Wang, N. Zhuang et al., Papain-Like Protease (Plpro) Inhibitory Effects of Cinnamic Amides from Tribulus terrestris Fruits, Biological and Pharmaceutical Bulletin, Vol. 37, No. 6, 2014, pp. 1021-1028, https://doi.org/10.1248/bpb.b14-00026.[13] D. Dermawan, B. A. Prabowo, C. A. Rakhmadina, In Silico Study of Medicinal Plants with Cyclodextrin Inclusion Complex as The Potential Inhibitors Against SARS-Cov-2 Main Protease (Mpro) and Spike (S) Receptor, Informatics in Medicine Unlocked, Vol. 25, 2021, pp. 1-18, https://doi.org/10.1016/j.imu.2021.100645.[14] R. Dang, S. Gezici, Immunomodulatory Effects of Medicinal Plants and Natural Phytochemicals in Combating Covid-19, The 6th International Mediterranean Symposium on Medicinal and Aromatic Plants (MESMAP-6), Izmir, Selcuk (Ephesus), Turkey, 2020, pp. 12-13.[15] G. Jiangning, W. Xinchu, W. Hou, L. Qinghua, B. Kaishun, Antioxidants from a Chinese Medicinal Herb–Psoralea corylifolia L., Food Chemistry, Vol. 9, No. 2, 2005, pp. 287-292, https://doi.org/10.1016/j.foodchem.2004.04.029.[16] B. Ruan, L. Y. Kong, Y. Takaya, M. Niwa, Studies on The Chemical Constituents of Psoralea corylifolia L., Journal of Asian Natural Products Research, Vol. 9, No. 1, 2007, pp. 41-44, https://doi.org/10.1080/10286020500289618.[17] D. T. Loi, Vietnamese Medicinal Plants and Herbs, Medical Publishing House, Hanoi, 2013 (in Vietnamese).[18] S. Mazraedoost, G. Behbudi, S. M. Mousavi, S. A. Hashemi, Covid-19 Treatment by Plant Compounds, Advances in Applied NanoBio-Technologies, Vol. 2, No. 1, 2021, pp. 23-33, https://doi.org/10.47277/AANBT/2(1)33.[19] B. A. Origbemisoye, S. O. Bamidele, Immunomodulatory Foods and Functional Plants for COVID-19 Prevention: A Review, Asian Journal of Medical Principles and Clinical Practice, 2020, pp. 15-26, https://journalajmpcp.com/index.php/AJMPCP/article/view/30124[20] A. Mandal, A. K. Jha, B. Hazra, Plant Products as Inhibitors of Coronavirus 3CL Protease, Frontiers in Pharmacology, Vol. 12, 2021, pp. 1-16, https://doi.org/10.3389/fphar.2021.583387[21] N. H. Tung, V. D. Loi, B. T. Tung, L.Q. Hung, H. B. Tien et al., Triterpenen Ursan Frame Isolated from the Roots of Salvia Miltiorrhiza Bunge Growing in Vietnam, VNU Journal of Science: Medical and Pharmaceutical Sciences, Vol. 32, No. 2, 2016, pp. 58-62, https://js.vnu.edu.vn/MPS/article/view/3583 (in Vietnamese).[22] J. Y. Park, J. H. Kim, Y. M. Kim, H. J. Jeong, D. W. Kim, K. H. Park et al., Tanshinones as Selective and Slow-Binding Inhibitors for SARS-CoV Cysteine Proteases. Bioorganic and Medicinal Chemistry, Vol. 20, No. 19, 2012, pp. 5928-5935, https://doi.org/10.1016/j.bmc.2012.07.038.[23] F. Hamdani, N. Houari, Phytotherapy of Covid-19. A Study Based on a Survey in North Algeria, Phytotherapy, Vol. 18, No. 5, 2020, pp. 248-254, https://doi.org/10.3166/phyto-2020-0241.[24] P. T. L. Huong, N. T. Dinh, Chemical Composition And Antibacterial Activity of The Essential Oil From The Leaves of Regrowth Eucalyptus Collected from Viet Tri City, Phu Tho Province, Vietnam Journal of Science, Technology and Engineering, Vol. 18, No. 1, 2020, pp. 54-61 (in Vietnamese).[25] M. Asif, M. Saleem, M. Saadullah, H. S. Yaseen, R. Al Zarzour, COVID-19 and Therapy with Essential Oils Having Antiviral, Anti-Inflammatory, and Immunomodulatory Properties, Inflammopharmacology, Vol. 28, 2020, pp. 1153-1161, https://doi.org/10.1007/s10787-020-00744-0.[26] I. Jahan, O. Ahmet, Potentials of Plant-Based Substance to Inhabit and Probable Cure for The COVID-19, Turkish Journal of Biology, Vol. 44, No. SI-1, 2020, pp. 228-241, https://doi.org/10.3906/biy-2005-114.[27] A. D. Sharma, I. Kaur, Eucalyptus Essential Oil Bioactive Molecules from Against SARS-Cov-2 Spike Protein: Insights from Computational Studies, Res Sq., 2021, pp. 1-6, https://doi.org/10.21203/ rs.3.rs-140069/v1. [28] K. Rajagopal, P. Varakumar, A. Baliwada, G. Byran, Activity of Phytochemical Constituents of Curcuma Longa (Turmeric) and Andrographis Paniculata Against Coronavirus (COVID-19): An in Silico Approach, Future Journal of Pharmaceutical Sciences, Vol. 6, No. 1, 2020, pp. 1-10, https://doi.org/10.1186/s43094-020-00126-x[29] J. Lan, J. Ge, J. Yu, S. Shan, H. Zhou, S. Fan et al., Structure of The SARS-CoV-2 Spike Receptor-Binding Domain Bound to The ACE2 Receptor, Nature, Vol. 581, No. 7807, 2020, pp. 215-220, https://doi.org/10.1038/s41586-020-2180-5.[30] M. Letko, A. Marzi, V. Munster, Functional Assessment of Cell Entry and Receptor Usage for SARS-Cov-2 and Other Lineage B Betacoronaviruses, Nature Microbiology, Vol. 5, No. 4, 2020, pp. 562-569, https://doi.org/10.1038/s41564-020-0688-y.[31] C. Yi, X. Sun, J. Ye, L. Ding, M. Liu, Z. Yang et al., Key Residues of The Receptor Binding Motif in The Spike Protein of SARS-Cov-2 That Interact with ACE2 and Neutralizing Antibodies, Cellular and Molecular Immunology, Vol. 17, No. 6, 2020, pp. 621-630, https://doi.org/10.1038/s41423-020-0458-z.[32] N. T. Thom, Study on The Composition and Biological Activities of Flavonoids from The Roots of Scutellaria baicalensis: Doctoral Dissertation, Hanoi University of Science and Technology, 2018 (in Vietnamese).[33] Y. J. Tang, F. W. Zhou, Z. Q. Luo, X. Z. Li, H. M. Yan, M. J. Wang et al., Multiple Therapeutic Effects of Adjunctive Baicalin Therapy in Experimental Bacterial Meningitis, Inflammation, Vol. 33, No. 3, 2010, pp. 180-188, https://doi.org/10.1007/s10753-009-9172-9.[34] H. Liu, F. Ye, Q. Sun, H. Liang, C. Li, S. Li et al., Scutellaria Baicalensis Extract and Baicalein Inhibit Replication of SARS-Cov-2 and Its 3C-Like Protease in Vitro, Journal of Enzyme Inhibition and Medicinal Chemistry, Vol. 36, No. 1, 2021, pp. 497-503, https://doi.org/10.1080/14756366.2021.1873977.[35] Z. Iqbal, H. Nasir, S. Hiradate, Y. Fujii, Plant Growth Inhibitory Activity of Lycoris Radiata Herb. and The Possible Involvement of Lycorine as an Allelochemical, Weed Biology and Management, Vol. 6, No. 4, 2006, pp. 221-227, https://doi.org/10.1111/j.1445-6664.2006.00217.x.[36] S. Y. Li, C. Chen, H. Q. Zhang, H. Y. Guo, H. Wang, L. Wang et al., Identification of Natural Compounds with Antiviral Activities Against SARS-Associated Coronavirus, Antiviral Research, Vol. 67, No. 1, 2005, pp. 18-23, https://doi.org/10.1016/j.antiviral.2005.02.007.[37] S. Kretzing, G. Abraham, B. Seiwert, F. R. Ungemach, U. Krügel, R. Regenthal, Dose-dependent Emetic Effects of The Amaryllidaceous Alkaloid Lycorine in Beagle Dogs, Toxicon, Vol. 57, No. 1, 2011, pp. 117-124, https://doi.org/10.1016/j.toxicon.2010.10.012.[38] Y. N. Zhang, Q. Y. Zhang, X. D. Li, J. Xiong, S. Q. Xiao, Z. Wang, et al., Gemcitabine, Lycorine and Oxysophoridine Inhibit Novel Coronavirus (SARS-Cov-2) in Cell Culture, Emerging Microbes & Infections, Vol. 9, No. 1, 2020, pp. 1170-1173, https://doi.org/10.1080/22221751.2020.1772676.[39] Y. H. Jin, J. S. Min, S. Jeon, J. Lee, S. Kim, T. Park et al., Lycorine, a Non-Nucleoside RNA Dependent RNA Polymerase Inhibitor, as Potential Treatment for Emerging Coronavirus Infections, Phytomedicine, Vol. 86, 2021, pp. 1-8, https://doi.org/10.1016/j.phymed.2020.153440.[40] H. V. Hoa, P. V. Trung, N. N. Hanh, Isolation Andrographolid and Neoandrographolid from Andrographis Paniculata Nees, Can Tho University Journal of Science, Vol. 10, 2008, pp. 25-30 (in Vietnamese)[41] S. K. Enmozhi, K. Raja, I. Sebastine, J. Joseph, Andrographolide as a Potential Inhibitor Of SARS-Cov-2 Main Protease: An in Silico Approach, Journal of Biomolecular Structure and Dynamics, Vol. 39, No. 9, 2021, pp. 3092-3098, https://doi.org/10.1080/07391102.2020.1760136.[42] S. A. Lakshmi, R. M. B. Shafreen, A. Priya, K. P. Shunmugiah, Ethnomedicines of Indian Origin for Combating COVID-19 Infection by Hampering The Viral Replication: Using Structure-Based Drug Discovery Approach, Journal of Biomolecular Structure and Dynamics, Vol. 39, No. 13, 2020, pp. 4594-4609, https://doi.org/10.1080/07391102.2020.1778537.[43] N. P. L. Laksmiani, L. P. F. Larasanty, A. A. G. J. Santika, P. A. A. Prayoga, A. A. I. K. Dewi, N. P. A. K. Dewi, Active Compounds Activity from The Medicinal Plants Against SARS-Cov-2 Using in Silico Assay, Biomedical and Pharmacology Journal, Vol. 13, No. 2, 2020, pp. 873-881, https://dx.doi.org/10.13005/bpj/1953.[44] N. A. Murugan, C. J. Pandian, J. Jeyakanthan, Computational Investigation on Andrographis Paniculata Phytochemicals to Evaluate Their Potency Against SARS-Cov-2 in Comparison to Known Antiviral Compounds in Drug Trials, Journal of Biomolecular Structure and Dynamics, Vol. 39, No. 12, 2020, pp. 4415-4426, https://doi.org/10.1080/07391102.2020.1777901.[45] S. Hiremath, H. V. Kumar, M. Nandan, M. Mantesh, K. Shankarappa,V. Venkataravanappa et al., In Silico Docking Analysis Revealed The Potential of Phytochemicals Present in Phyllanthus Amarus and Andrographis Paniculata, Used in Ayurveda Medicine in Inhibiting SARS-Cov-2, 3 Biotech, Vol. 11, No. 2, 2021, pp. 1-18, https://doi.org/10.1007/s13205-020-02578-7.[46] K. S. Ngiamsuntorn, A. Suksatu, Y. Pewkliang, P. Thongsri, P. Kanjanasirirat, S. Manopwisedjaroen, et al., Anti-SARS-Cov-2 Activity of Andrographis Paniculata Extract and Its Major Component Andrographolide in Human Lung Epithelial Cells and Cytotoxicity Evaluation in Major Organ Cell Representatives, Journal of Natural Products, Vol. 84, No. 4, 2021, pp. 1261-1270, https://doi.org/10.1021/acs.jnatprod.0c01324.[47] D. X. Em, N. T. T. Dai, N. T. T. Tram, D. X. Chu, Four Compounds Isolated from Azadirachta Indica Jus leaves. F., Meliaceae, Pharmaceutical Journal, Vol. 59, No. 7, 2019, pp. 33-36 (in Vietnamese).[48] V. V Do, N. T. Thang, N. T. Minh, N. N. Hanh, Isolation, Purification and Investigation on Antimicrobial Activity of Salanin from Neem Seed Kernel (Azadirachta Indica A. Juss) of The Neem Tree Planted in Ninh Thuan Province, Vietnam, Journal of Science and Technology, Vol. 44, No. 2, 2006, pp. 24-31 (in Vietnamese).[49] P. I. Manzano Santana, J. P. P. Tivillin, I. A. Choez Guaranda, A. D. B. Lucas, A. Katherine, Potential Bioactive Compounds of Medicinal Plants Against New Coronavirus (SARS-Cov-2): A Review, Bionatura, Vol. 6, No. 1, 2021, pp. 1653-1658, https://doi.org/10.21931/RB/2021.06.01.30[50] S. Borkotoky, M. Banerjee, A Computational Prediction of SARS-Cov-2 Structural Protein Inhibitors from Azadirachta Indica (Neem), Journal of Biomolecular Structure and Dynamics, Vol. 39, No. 11, 2021, pp. 4111-4121, https://doi.org/10.1080/07391102.2020.1774419.[51] R. Jager, R. P. Lowery, A. V. Calvanese, J. M. Joy, M. Purpura, J. M. Wilson, Comparative Absorption of Curcumin Formulations, Nutrition Journal, Vol. 13, No. 11, 2014, https://doi.org/10.1186/1475-2891-13-11.[52] D. Praditya, L. Kirchhoff, J. Bruning, H. Rachmawati, J. Steinmann, E. Steinmann, Anti-infective Properties of the Golden Spice Curcumin, Front Microbiol, Vol. 10, No. 912, 2019, https://doi.org/10.3389/fmicb.2019.00912.[53] C. C. Wen, Y. H. Kuo, J. T. Jan, P. H. Liang, S. Y. Wang, H. G. Liu et al., Specific Plant Terpenoids and Lignoids Possess Potent Antiviral Activities Against Severe Acute Respiratory Syndrome Coronavirus, Journal of Medicinal Chemistry, Vol. 50, No. 17, 2007, pp. 4087-4095, https://doi.org/10.1021/jm070295s.[54] R. Lu, X. Zhao, J. Li, P. Niu, B. Yang, H. Wu et al., Genomic Characterisation and Epidemiology of 2019 Novel Coronavirus: Implications for Virus Origins and Receptor Binding, The Lancet, Vol. 395, No. 10224, 2020, pp. 565-574, https://doi.org/10.1016/S0140-6736(20)30251-8.[55] M. Kandeel, M. Al Nazawi, Virtual Screening and Repurposing of FDA Approved Drugs Against COVID-19 Main Protease, Life Sciences, Vol. 251, No. 117627, 2020, pp. 1-5, https://doi.org/10.1016/j.lfs.2020.117627.[56] V. K. Maurya, S. Kumar, A. K. Prasad, M. L. B. Bhatt, S. K. Saxena, Structure-Based Drug Designing for Potential Antiviral Activity of Selected Natural Products from Ayurveda Against SARS-CoV-2 Spike Glycoprotein and Its Cellular Receptor, Virusdisease, Vol. 31, No. 2, 2020, pp. 179-193, https://doi.org/10.1007/s13337-020-00598-8.[57] M. Hoffmann, H. Kleine Weber, S. Schroeder, N. Kruger, T. Herrler, S. Erichsen et al., SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor, Cell, Vol. 181, No. 2, 2020, pp. 271-280, https://doi.org/10.1016/j.cell.2020.02.052.[58] S. Katta, A. Srivastava, R. L. Thangapazham, I. L. Rosner, J. Cullen, H. Li et al., Curcumin-Gene Expression Response in Hormone Dependent and Independent Metastatic Prostate Cancer Cells, International Journal of Molecular Sciences, Vol. 20, No. 19, 2019, pp. 4891-4907, https://doi.org/10.3390/ijms20194891.[59] D. Ting, N. Dong, L. Fang, J. Lu, J. Bi, S. Xiao et al., Multisite Inhibitors for Enteric Coronavirus: Antiviral Cationic Carbon Dots Based on Curcumin, ACS Applied Nano Materials, Vol. 1, No. 10, 2018, pp. 5451-5459, https://doi.org/10.1021/acsanm.8b00779.[60] T. Huynh, H. Wang, B. Luan, In Silico Exploration of the Molecular Mechanism of Clinically Oriented Drugs for Possibly Inhibiting SARS-CoV-2's Main Protease, the Journal of Physical Chemistry Letters, Vol. 11, No. 11, 2020, pp. 4413-4420, https://doi.org/10.1021/acs.jpclett.0c00994.[61] D. D'Ardes, A. Boccatonda, I. Rossi, M. T. Guagnano, COVID-19 and RAS: Unravelling an Unclear Relationship, International Journal of Molecular Sciences, Vol. 21, No. 8, 2020, pp. 3003-3011, https://doi.org/10.3390/ijms21083003. [62] X. F. Pang, L. H. Zhang, F. Bai, N. P. Wang, R. E. Garner, R. J. McKallip et al., Attenuation of Myocardial Fibrosis with Curcumin is Mediated by Modulating Expression of Angiotensin II AT1/AT2 Receptors and ACE2 in Rats, Drug Design Development Therapy, Vol. 9, 2015, pp. 6043-6054, https://doi.org/10.2147/DDDT.S95333.[63] Y. Yao, W. Wang, M. Li, H. Ren, C. Chen, J. Wang et al., Curcumin Exerts its Anti-Hypertensive Effect by Down-Regulating the AT1 Receptor in Vascular Smooth Muscle Cells, Scientific Reports, Vol. 6, No. 25579, 2016, pp. 1-6, https://doi.org/10.1038/srep25579.[64] V. J. Costela Ruiz, R. Illescas Montes, J. M. Puerta Puerta, C. Ruiz, L. Melguizo Rodríguez, SARS-CoV-2 Infection: The Role of Cytokines in COVID-19 Disease, Cytokine Growth Factor Reviews, Vol. 54, 2020, pp. 62-75, https://doi.org/10.1016/j.cytogfr.2020.06.001.[65] H. Valizadeh, S. Abdolmohammadi Vahid, S. Danshina, M. Ziya Gencer, A. Ammari, A. Sadeghi et al., Nano-Curcumin Therapy, a Promising Method in Modulating Inflammatory Cytokines in COVID-19 Patients, International Immunopharmacology, Vol. 89 (PtB), No. 107088, 2020, pp. 1-12, https://doi.org/10.1016/j.intimp.2020.107088.[66] Y. H. Lo, R. D. Lin, Y. P. Lin, Y. L. Liu, M. H. Lee, Active Constituents from Sophora Japonica Exhibiting Cellular Tyrosinase Inhibition in Human Epidermal Melanocytes, Journal of Ethnopharmacology, Vol. 124, No. 3, 2009, pp. 625-629, https://doi.org/10.1016/j.jep.2009.04.053.[67] A. Robaszkiewicz, A. Balcerczyk, G. Bartosz, Antioxidative and Prooxidative Effects of Quercetin on A549 Cells, Cell Biology International, Vol. 31, No. 10, 2007, pp. 1245-1250, https://doi.org/10.1016/j.cellbi.2007.04.009[68] N. Uchide, H. Toyoda, Antioxidant Therapy as a Potential Approach to Severe Influenza-associated Complications, Molecules (Basel, Switzerland), Vol. 16, No. 3, 2011, pp. 2032-2052, https://doi.org/10.3390/molecules16032032.[69] M. P. Nair, C. Kandaswami, S. Mahajan, K. C. Chadha, R. Chawda, H. Nair et al., The Flavonoid, Quercetin, Differentially Regulates Th-1 (IFNgamma) and Th-2 (IL4) Cytokine Gene Expression by Normal Peripheral Blood Mononuclear Cells, Biochimica et Biophysica Acta - Molecular Cell Research, Vol. 1593, No. 1, 2002, pp. 29-36, https://doi.org/10.1016/s0167-4889(02)00328-2.[70] X. Chen, Z. Wang, Z. Yang, J. Wang, Y. Xu, R. X. Tan et al., Houttuynia Cordata Blocks HSV Infection Through Inhibition of NF-κB Activation, Antiviral Research, Vol. 92, No. 2, 2011, pp. 341-345, https://doi.org/10.1016/j.antiviral.2011.09.005.[71] T. N. Kaul, E. J. Middleton, P. L. Ogra, Antiviral Effect of Flavonoids on Human Viruses, Journal of Medical Virology, Vol. 15. No. 1, 1985, pp. 71-79, https://doi.org/10.1002/jmv.1890150110.[72] K. Zandi, B. T. Teoh, S. S. Sam, P. F. Wong, M. R. Mustafa, S. AbuBakar, Antiviral Activity of Four Types of Bioflavonoid Against Dengue Virus Type-2, Virology Journal, Vol. 8, No. 1, 2011, pp. 560-571, https://doi.org/10.1186/1743-422X-8-560.[73] J. Y. Park, H. J. Yuk, H. W. Ryu, S. H. Lim, K. S. Kim, K. H. Park et al., Evaluation of Polyphenols from Broussonetia Papyrifera as Coronavirus Protease Inhibitors, Journal of Enzyme Inhibition and Medicinal Chemistry, Vol. 32, No. 1, 2017, pp. 504-515, https://doi.org/10.1080/14756366.2016.1265519.[74] S. C. Cheng, W. C. Huang, J. H. S. Pang, Y. H. Wu, C. Y. Cheng, Quercetin Inhibits the Production of IL-1β-Induced Inflammatory Cytokines and Chemokines in ARPE-19 Cells via the MAPK and NF-κB Signaling Pathways, International Journal of Molecular Sciences, Vol. 20, No. 12, 2019, pp. 2957-2981, https://doi.org/10.3390/ijms20122957. [75] O. J. Lara Guzman, J. H. Tabares Guevara, Y. M. Leon Varela, R. M. Álvarez, M. Roldan, J. A. Sierra et al., Proatherogenic Macrophage Activities Are Targeted by The Flavonoid Quercetin, The Journal of Pharmacology and Experimental Therapeutics, Vol. 343, No. 2, 2012, pp. 296-303, https://doi.org/10.1124/jpet.112.196147.[76] A. Saeedi Boroujeni, M. R. Mahmoudian Sani, Anti-inflammatory Potential of Quercetin in COVID-19 Treatment, Journal of Inflammation, Vol. 18, No. 1, 2021, pp. 3-12, https://doi.org/10.1186/s12950-021-00268-6.[77] M. Smith, J. C. Smith, Repurposing Therapeutics for COVID-19: Supercomputer-based Docking to the SARS-CoV-2 Viral Spike Protein and Viral Spike Protein-human ACE2 Interface, ChemRxiv, 2020, pp. 1-28, https://doi.org/10.26434/chemrxiv.11871402.v4.[78] S. Khaerunnisa, H. Kurniawan, R. Awaluddin, S. Suhartati, S. Soetjipto, Potential Inhibitor of COVID-19 Main Protease (Mpro) from Several Medicinal Plant Compounds by Molecular Docking Study, Preprints, 2020, pp. 1-14, https://doi.org/10.20944/preprints202003.0226.v1.[79] J. M. Calderón Montaño, E. B. Morón, C. P. Guerrero, M. L. Lázaro, A Review on the Dietary Flavonoid Kaempferol, Mini Reviews in Medicinal Chemistry, Vol. 11, No. 4, 2011, pp. 298-344, https://doi.org/10.2174/138955711795305335.[80] A. Y. Chen, Y. C. Chen, A Review of the Dietary Flavonoid, Kaempferol on Human Health and Cancer Chemoprevention, Food Chem, Vol. 138, No. 4, 2013, pp. 2099-2107, https://doi.org/10.1016/j.foodchem.2012.11.139.[81] S. Schwarz, D. Sauter, W. Lu, K. Wang, B. Sun, T. Efferth et al., Coronaviral Ion Channels as Target for Chinese Herbal Medicine, Forum on Immunopathological Diseases and Therapeutics, Vol. 3, No. 1, 2012, pp. 1-13, https://doi.org/10.1615/ForumImmunDisTher.2012004378.[82] R. Zhang, X. Ai, Y. Duan, M. Xue, W. He, C. Wang et al., Kaempferol Ameliorates H9N2 Swine Influenza Virus-induced Acute Lung Injury by Inactivation of TLR4/MyD88-mediated NF-κB and MAPK Signaling Pathways, Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, Vol. 89, 2017, pp. 660-672, https://doi.org/10.1016/j.biopha.2017.02.081.[83] K. W. Chan, V. T. Wong, S. C. W. Tang, COVID-19: An Update on the Epidemiological, Clinical, Preventive and Therapeutic Evidence and Guidelines of Integrative Chinese-Western Medicine for the Management of 2019 Novel Coronavirus Disease, The American Journal of Chinese medicine, Vol. 48, No. 3, 2020, pp. 737-762, https://doi.org/10.1142/S0192415X20500378.[84] Y. F. Huang, C. Bai, F. He, Y. Xie, H. Zhou, Review on the Potential Action Mechanisms of Chinese Medicines in Treating Coronavirus Disease 2019 (COVID-19), Pharmacological Research, Vol. 158, No. 104939, 2020, pp. 1-10, https://doi.org/10.1016/j.phrs.2020.104939.[85] L. Xu, X. Zheng, Y. Wang, Q. Fan, M. Zhang, R. Li et al., Berberine Protects Acute Liver Failure in Mice Through Inhibiting Inflammation and Mitochondria-dependent Apoptosis, European Journal of Pharmacology, Vol. 819, 2018, pp. 161-168, https://doi.org/10.1016/j.ejphar.2017.11.013.[86] X. Chen, H. Guo, Q. Li, Y. Zhang, H. Liu, X. Zhang et al., Protective Effect of Berberine on Aconite‑induced Myocardial Injury and the Associated Mechanisms, Molecular Medicine Reports, Vol. 18, No. 5, 2018, pp. 4468-4476, https://doi.org/10.3892/mmr.2018.9476.[87] K. Hayashi, K. Minoda, Y. Nagaoka, T. Hayashi, S. Uesato, Antiviral Activity of Berberine and Related Compounds Against Human Cytomegalovirus, Bioorganic & Medicinal Chemistry Letters, Vol. 17, No. 6, 2007, pp. 1562-1564, https://doi.org/10.1016/j.bmcl.2006.12.085.[88] A. Warowicka, R. Nawrot, A. Gozdzicka Jozefiak, Antiviral Activity of Berberine, Archives of Virology, Vol. 165, No. 9, 2020, pp. 1935-1945, https://doi.org/10.1007/s00705-020-04706-3.[89] Z. Z. Wang, K. Li, A. R. Maskey, W. Huang, A. A. Toutov, N. Yang et al., A Small Molecule Compound Berberine as an Orally Active Therapeutic Candidate Against COVID-19 and SARS: A Computational and Mechanistic Study, FASEB Journal : Official Publication of the Federation of American Societies for Experimental Biology, Vol. 35, No. 4, 2021, pp. e21360-21379, https://doi.org/10.1096/fj.202001792R.[90] A. Pizzorno, B. Padey, J. Dubois, T. Julien, A. Traversier, V. Dulière et al., In Vitro Evaluation of Antiviral Activity of Single and Combined Repurposable Drugs Against SARS-CoV-2, Antiviral Research, Vol. 181, No. 104878, 2020, https://doi.org/10.1016/j.antiviral.2020.104878.[91] B. Y. Zhang, M. Chen, X. C. Chen, K. Cao, Y. You, Y. J. Qian et al., Berberine Reduces Circulating Inflammatory Mediators in Patients with Severe COVID-19, The British Journal of Surgery, Vol. 108, No. 1, 2021, pp. e9-e11, https://doi.org/10.1093/bjs/znaa021.[92] K. P. Latté, K. E. Appel, A. Lampen, Health Benefits and Possible Risks of Broccoli - an Overview, Food and Chemical Toxicology : an International Journal Published for the British Industrial Biological Research Association, Vol. 49, No. 12, 2011, pp. 3287-3309, https://doi.org/10.1016/j.fct.2011.08.019.[93] C. Sturm, A. E. Wagner, Brassica-Derived Plant Bioactives as Modulators of Chemopreventive and Inflammatory Signaling Pathways, International Journal of Molecular Sciences, Vol. 18, No. 9, 2017, pp. 1890-1911, https://doi.org/10.3390/ijms18091890.[94] R. T. Ruhee, S. Ma, K. Suzuki, Sulforaphane Protects Cells against Lipopolysaccharide-Stimulated Inflammation in Murine Macrophages, Antioxidants (Basel, Switzerland), Vol. 8, No. 12, 2019, pp. 577-589, https://doi.org/10.3390/antiox8120577.[95] S. M. Ahmed, L. Luo, A. Namani, X. J. Wang, X. Tang, Nrf2 Signaling Pathway: Pivotal Roles in Inflammation, Biochimica et Biophysica Acta Molecular Basis of Disease, Vol. 1863, No. 2, 2017, pp. 585-597, https://doi.org/10.1016/j.bbadis.2016.11.005.[96] Z. Sun, Z. Niu, S. Wu, S. Shan, Protective Mechanism of Sulforaphane in Nrf2 and Anti-Lung Injury in ARDS Rabbits, Experimental Therapeutic Medicine, Vol. 15, No. 6, 2018, pp. 4911-4951, https://doi.org/10.3892/etm.2018.6036.[97] H. Y. Cho, F. Imani, L. Miller DeGraff, D. Walters, G. A. Melendi, M. Yamamoto et al., Antiviral Activity of Nrf2 in a Murine Model of Respiratory Syncytial Virus Disease, American Journal of Respiratory and Critical Care Medicine, Vol. 179, No. 2, 2009, pp. 138-150, https://doi.org/10.1164/rccm.200804-535OC.[98] M. J. Kesic, S. O. Simmons, R. Bauer, I. Jaspers, Nrf2 Expression Modifies Influenza A Entry and Replication in Nasal Epithelial Cells, Free Radical Biology & Medicine, Vol. 51, No. 2, 2011, pp. 444-453, https://doi.org/10.1016/j.freeradbiomed.2011.04.027.[99] A. Cuadrado, M. Pajares, C. Benito, J. J. Villegas, M. Escoll, R. F. Ginés et al., Can Activation of NRF2 Be a Strategy Against COVID-19?, Trends in Pharmacological Sciences, Vol. 41, No. 9, 2020, pp. 598-610, https://doi.org/10.1016/j.tips.2020.07.003.[100] J. Gasparello, E. D'Aversa, C. Papi, L. Gambari, B. Grigolo, M. Borgatti et al., Sulforaphane Inhibits the Expression of Interleukin-6 and Interleukin-8 Induced in Bronchial Epithelial IB3-1 Cells by Exposure to the SARS-CoV-2 Spike Protein, Phytomedicine : International Journal of Phytotherapy and Phytopharmacology, Vol. 87, No. 53583, 2021, https://doi.org/10.1016/j.phymed.2021.153583.
APA, Harvard, Vancouver, ISO, and other styles

Books on the topic "Yan ye yin hang"

1

Li, Guosheng. Zhejiang xing ye yin hang yan jiu. 8th ed. Shanghai Shi: Shanghai cai jing da xue chu ban she, 2009.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
2

Wencheng, Shang, and Mu Hongsheng, eds. Zhongguo yin hang ye chan ye zu zhi yan jiu. Shanghai Shi: Shanghai cai jing da xue chu ban she, 2009.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
3

Gong, Feng. Zhongguo yin hang ye wen jian jing ying yan jiu. 8th ed. Beijing Shi: Zhongguo jing ji chu ban she, 2006.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
4

Lu, Yuexiang, and Yangjun Tang. Zhongguo cheng shi shang ye yin hang yan jiu. 8th ed. Beijing Shi: Jing ji ke xue chu ban she, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
5

Li, Jie. Shang ye yin hang "zou chu qu" yan jiu. 8th ed. Beijing Shi: Jing ji ke xue chu ban she, 2009.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
6

Ouyang, Weimin. Zhongguo fei yin hang jin rong ye yan jiu. 8th ed. Beijing: Zhongguo jin rong chu ban she, 2001.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
7

Yang, Zaiping. Yong zhen cheng fu wu gan dong da zhong: 2014 Nian du zhong guo yin hang ye wen ming gui fan fu wu qian jia shi fan dan wei gu shi ji. 8th ed. Beijing: Zhong guo jin rong chu ban she, 2015.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
8

Li jin yin hang. pei xun zhong xin. Yin hang gong ying lian rong zi huo quan zhi ya rong zi pei xun. Bei jing: Zhong guo jin rong chu ban she, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
9

Wang, Yan. Zhongguo shang ye yin hang hang ye xin yong feng xian guan li yan jiu. 8th ed. Beijing: Ren min ri bao chu ban she, 2017.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
10

Shi, jian ping, and zhi min Wu. Shang ye yin hang ye wu yu jing ying. 8th ed. Bei jing: Zhong guo ren min da xue chu ban she, 2010.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography