用户名: 密码: 验证码:
稀土杂多化合物抗乙型肝炎病毒的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
慢性乙型肝炎属于重大传染性疾病,其传染性强,患病率高,对人体健康危害严重。我国是乙肝高发区之一,约有乙肝病毒携带者近1.3亿人。目前临床使用的抗病毒药物主要为核苷类似物,但其缺点是易产生耐药性,毒副作用大,因此开发高效低毒的抗病毒药物十分重要。本研究首次选用杂多化合物为主要研究体系,利用其分子可设计性的特点,设计、合成了系列稀土杂多化合物。在此基础上,按照临床前新药研发标准的要求,全面系统地开展了稀土杂多化合物抗HBV的药学、药效学、毒理学和药代动力学研究。同时,首次应用先进的同步辐射成像技术,探讨了杂多化合物抗病毒作用机制。
     药学研究中,采用红外光谱、紫外光谱、X射线单晶衍射及核磁共振光谱等检测方法,对受试化合物进行结构确证性和稳定性研究。多种检测方法共同表明,受试化合物化学结构明确,稳定性良好。药效学研究中,分别以HepG2.2.15细胞和HBV转基因鼠为研究模型,检测受试化合物对HBeAg、HBsAg和HBV DNA的抑制作用,结果表明,受试化合物对抗原分泌和病毒DNA合成有较强的抑制作用。经药效学筛选,选取受试化合物9开展急性毒性、长期毒性和遗传毒性等安全性评价研究。毒性评价结果表明,受试化合物9属于基本无毒化合物,长期毒性实验未发现毒性作用,无致畸、致突变作用。采用ICP-MS技术评价受试化合物9经口途径给药的药代动力学特征,结果表明,受试化合物9在大鼠体内的动力学过程符合二室模型,其吸收过程迅速,肝脏靶向性好,无组织蓄积,与血浆蛋白呈中度结合。X射线纳米CT细胞成像结果表明,杂多化合物的抗病毒机理是通过抑制病毒侵入宿主细胞过程发挥作用。
     本研究首次系统的研究稀土杂多化合物抗HBV的活性,课题研究成果已获得国家发明专利。本实验研究为开发具有自主知识产权的新型非核苷类抗HBV药物奠定了坚实的基础。
Hepatitis B is an infectious disease caused by the hepatitis B virus (hepatitis Bvirus, HBV) So far there are about3.5to400million chronic hepatitis B virusinfection in the world. There are more than one million patients die from diseasescaused by HBV in each year. Our country is a high incidence of HBV infection. It iscalculated that there are existing120million people carriers chronic HBV, and about30million cases of hepatitis B patients. Moreover the number of new cases is around10to30thousand per year. Chronic HBV infection has become a serious public healthproblem. The direct economic losses caused by chronic hepatitis B disease are up toabout900billion Yuan each year in China.
     The target of anti-HBV therapy is to suppress or remove the body of HBV in thebody, reducing and preventing complications, thereby, improving life quality ofpatients and prolong survival. Drug therapy is the most important means of clinicalanti-HBV treatment. Currently, drug therapy is the most important means of clinicalanti-HBV treatment. The anti-HBV drugs are mainly divided into three categories,interferon, and nucleoside and non-nucleoside drugs. However, these drugs in usehave different levels of drug resistance and side effects. Such as interferon withserious side effects is low response rate; THF can lead to emergence oflamivudine-resistant mutants.Although the effect of these drugs in inhibiting orremoving serum HBV DNA is rapid and significant, however, the drug concentrationin the liver is low. It is difficult to remove the virus in the liver cells, so the viruspersists in the patient body. Therefore, the development of efficiency, low toxicity,low-cost anti-HBV drugs has still great importance.
     Polyoxometalates are a class of metal oxide cluster compounds. POMs have beenstudied on antiviral activity for decade years. It is found that POMs have a stronginhibitory effect for HIV, influenza virus and other RNA viruses and some DNA viruses. It has been reported POMs may achieve its antiviral activity by inhibiting thereverse transcriptase activity or blocked viruses’ entry. So far its antiviralunderpinning mechanisms remain elusive.
     Polyoxometalates application in the field of biology has broad prospects forsynthesize of POMs entering into the molecular Design phase. Our previous studyfound that, the new compound (PTW-6) has excellent anti-HBV activity in vitro.With the inspiration our research group designed and synthesized a series of new rareearth polyoxometalates according to the structure and replication characteristics ofHBV. In accordance with the standard of pre-clinical drug development andrequirements, we carried out a comprehensive and systematic studies on POMs inpharmacy, pharmacodynamics, toxicology and pharmacokinetic. Last but not the mostimportant, the antiviral mechanism of POMs had been revealed directly by CellImaging using advanced X-ray nano-CT technology.
     1. Pharmaceutical Study
     In this study,11new rare earth POMs have been self-designed and synthesizedby general inorganic synthesis method. Confirmatory studies on compound structureshave been detected using IR, UV, X-ray diffraction and NMR spectroscopy and othermethods. PH stability of compounds was determined by conductance method. Theresults showed that the conductivity remained unchanged in the range of PH3~9.Compounds long-term placement stability and thermal stability were detected usingIR, UV, NMR and TG analysis. The results showed that the properties and structure ofthe compound is not changed. All the results in this study indicate a series of POMsshowing good drug ability with definite structure and excellent stability.
     2. Pharmacodynamic Study
     Cytotoxicity of compounds was detected by MTT assay. The results showcytotoxic of all compounds was low, in which the compound3,4,9and11hadoptimal results by comparing the CC50value.HBeAg, HBsAg secretion inhibition ofcompounds was detected by HBV e (s) antigen diagnostic test kit. The results showHBeAg, HBsAg secretion inhibitory effect of compound1,3,4and6is strong.Using quantitative PCR assay to test extracellular HBV DNA levels, the results showedHBV DNA replication inhibitory effect of compounds9and11were stronger than thepositive control drugs by comparing the IC50values. Then, the compounds9and11with the strongest anti-HBV activity and less toxic were selected to be conductedefficacy experiments in vivo. Quantitative PCR and ELISA were used detection oftransgenic mouse serum HBV DNA and HBsAg levels. The results showed that bothcompounds existed stronger effect than the positive control drugs on the replication ofHBV DNA and HBsAg secretion inhibitory effect.
     3. Safety Evaluation Study
     In this study, Kunming mice and Wistar rats were used as experimental animalmodels.The compounds were tested in acute toxicity, chronic toxicity andgenotoxicity. Acute toxicity test results showed that compounds3,4and11belong tolow toxicity compounds and compound9is non-toxic compound. Then, thecompound9was selected for long-term toxicity studies. The results show that thegeneral normal conditions, blood biochemical parameters and histopathologicalexamination were found no abnormalities in rats. The genetic toxicity results showedthe compound9has no teratogenic and mutagenic effects.
     4. Pharmacokinetics Study
     The quantitative analysis method was established to test the drug concentrationin Wistar rats by inductively coupled plasma (ICP-MS) technology. Then thepharmacokinetic studies of compound9were conducted based on this method. Thedynamic process of the test compound9complied with the two-compartment modelby calculate the compound concentration in rats through gavage (or oraladministration). The main pharmacokinetic parameters,AUC0~t, AUC0-∞andCmax, were dose related, and t1/2, CL, MRT and Vd and other pharmacokineticparameters Displays linear kinetics characteristics, which did not increase with thedose varies. The compound9can be quickly absorbed into the blood circulationsystem after oral administration with Tmax between0.5~2h, t1/2between20~26h. The absolute bioavailability in rats on different doses of compound9was13.76%, 6.39%and5.79%respectively. The result indicates that most of the test compoundmay be biotransformation after absorbing into the body. Tissue distribution result ofthe test compound9showed the concentration of the small intestine and the stomachthan other organizations, and liver tissue concentration was up to1579.10ng/g,indicating that the test compound has a good liver targeting property. Usingequilibrium dialysis method to test plasma protein binding, the results showed that thetest compound9showed moderate intensity to plasma proteins. The excretion resultsshow that about24.30%of the drug dose was excreted with urine, feces and bilewithin72h after administration. Prototype content in feces, urine and bile were low.This further indicates the test compound9conducted extensive metabolism in rats.
     In summary, we have synthesized a series of rare earth polyoxometalates withexcellent anti-HBV activity. The achievement of polyoxometalates in pharmaceutical,pharmacodynamics, toxicology and pharmacokinetic created a new era in theanti-virus field. The Comprehensive study also laid a solid foundation for thedevelopment of new non-nucleoside with independent intellectual property rights ofanti-HBV drugs.
引文
[1]曾颖玲,毛惠南.原发性肝癌血行转移与乙肝病毒的相关性研究[J].热带医学杂志,2013,13(1):70-73.
    [2]邹俐,郑萍,侯连兵.慢性乙型肝炎患者健康相关生命质量评价研究现状[J].热带医学杂志,2012,12(1):112-114.
    [3]杨波,潘捷云,王军.社区儿童感染乙型肝炎病毒危险因素病例对照研究[J].热带医学杂志,2012,12(9):1140-1142.
    [4]中国医学会.第十二次全国病毒性肝炎与肝病学术会议论文汇编[C].北京:中华医学会感染病学分会,中华医学会肝病学分会,2005.
    [3] Yan YX, Gao YQ, Sun X, Wang W, Huang XJ, Zhang T, Li M, Zang, CP, Li ZC,Wu H. Prevalence of hepatitis C virus and hepatitis B virus infections inHIV-positive Chinese patients [J]. Epidemiol Infect.,2011,139(3):354-360.
    [6]翁水旺,诗景.治疗乙肝新药物的研究进展与评价[J].海峡药学,2010,22(12):101-102.
    [7]农朝赞,王丽.中医药抗乙肝病毒的研究进展[J].右江民族医学院学报,2010,32(5):769-770.
    [8]刘士敬.进行抗乙肝病毒治疗首选哪些药物[J].求医问药,2010,5:10-11.
    [9]朱浩翔,张继明.抗HBV药物-替诺福韦[J].肝脏,2011,16(2):145-147.
    [10] Yang YF, Zhao W, Xia HM, Zhong YD, Huang P, Wen J. Long-term efficacy ofinterferon alpha therapy on hepatitis B viral replication in patients with chronichepatitis B: a meta-analysis [J]. Antiviral Res.,2010,85(2):361-365.
    [11] Zoulim F, Locarnini S. Hepatitis B virus resistance to nucleos(t)ide analogues [J].Gastroenterology,2009,137(5):1593-1608.
    [12] Chen CY, Ni YH, Chen HL, Lu FL, Chang MH. Lamivudine treatment ininfantile fulminant hepatitis B [J]. Pediatr Int.,2010,52(4):672-674.
    [13] Shin SR, Koh KC, Gwak GY, Choi MS, Lee JH, Paik SW, Yoo BC. A low viralload predicts a higher initial virologic response to adefovir in patients withLamivudine-resistant chronic hepatitis B [J]. Gut Liver,2010,4(4):530-536.
    [14] Thompson AJ, Avres A, Yuen L, Bartholomeusz A, Bowden DS, Iser DM, ChenRY, Demediuk B, Shaw G, Bell SJ, Watson KJ, Locarnini SA, Desmond PV.Lamivudine resistance in patients with chronic hepatitis B: role of clinical andvirological factors [J]. J Gastroenterol Hepatol.,2007,22(7):1078-1085.
    [15] Zeng AZ, Lu P, Deng H, Cai SF, Yang C, Xin XJ, Guo JJ, Li QL, Deng XH,Huang AL. Dissection of mechanism for the adefovir dipivoxil resistance inchronic hepatitis B patients [J]. Zhonghua Gan Zang Bing Za Zhi,2009,17(10):730-734.
    [16]刘士敬.恩替卡韦是治疗乙肝的首选药物[J].求医问药,2010,11:18-19.
    [17]普小菲.抗乙型肝炎病毒药物与治疗的研究进展[J].医学综述,2012,18(6):914-916.
    [18]郑书琴,柳龙根,叶春艳.病毒性乙型肝炎生物治疗研究进展[J].中华临床医师杂志(电子版),2012,6(17):5236-5238.
    [19] Wang YP, Liu F, He HW, Han YX, Peng ZG, Li BW, You XF, Song DQ, Li ZR,Yu LY, Cen S, Hong B, Sun CH, Zhao LX, Kreiswirth B, Perlin D, Shao RG,Jiang JD. Heat Stress cognate70hosts protein as a potential drug target againstdrug resistance in hepatitis B virus [J]. Antimicrob Agents Chemother.,2010,54(5):2070-2077.
    [20]杨芳,杨春,苏建家.乙型肝炎的预防和治疗研究进展[J].医学研究杂志,2011,40(4):12-14.
    [21]刘琦,任伟宏,禄保平.肝康颗粒联合拉米夫定对慢性乙型病毒性肝炎病毒耐药的影响[J].中医学报,2013,28(3):385-387.
    [22]李常春,李小月,张俊丽,苏俊芳.叶下珠复方II号抗乙型肝炎病毒的体外实验研究[J].中国热带医学,2009,9(2):217-218.
    [23] Chen Y, Cheng, G, Mahato, RI. RNAi for treating hepatitis B viral infection [J].Pharm Res.,2008,25(1):72-86.
    [24]孔令波,佟立新,王锡育. RNA干扰技术在抗乙型肝炎病毒感染应用中的研究进展[J].世界华人消化杂志,2009,17(13):1324-1328.
    [25] Morrissey DV, Lockridge JA, Shaw L, Blanchard K, Jensen K, Breen W,Hartsough K, Machemer L, Radka S, Jadhav V, Vaish N, Zinnen S, Vargeese C,Bowman K, Shaffer CS, Jeffs LB, Judge A, MacLachlan I, Polisky B. Potent andpersistent in vivo anti-HBV activity of chemically modified siRNAs [J]. NatBiotechnol.,2005,23(8):1002-1007.
    [26]刘红娥,周金凤. RNA干扰技术及其抗乙型肝炎病毒的相关研究[J].中国现代药物应用,2009,3(8):198-200.
    [27]李铤,尹文.慢性乙型肝炎治疗性疫苗的研究进展[J].第四军医大学学报,2009,30(5):477-479.
    [28] Johnson MA, Jenkins JM, Bye C. A study of the pharmacokinetic interactionbetween lamivudine and alpha interferon [J]. Eur J Clin Pharmacol.,2000,56(4):289-292.
    [29] Lau GK, Tsiang M, Hou J, Yuen S, Carman WF, Zhang L, Gibbs CS, Lam S.Combination therapy with lamivudine and famciclovir for chronic hepatitisB-infected Chinese patients: a viral dynamics study [J]. Hepatology,2000,32(2):394-399.
    [30]金宇.慢性乙肝的非核苷类药物治疗初探[J].中国当代医药,2011,18(6):147-148.
    [31] Ying C, De Clercq E, Nicholson W, Furman P, Neyts J. Inhibition of thereplication of the DNA polymerase M550V mutation variant of human hepatitisB virus by adefovir, tenofovir, L-FMAU, DAPD, penciclovir and lobucavir [J]. JViral Hepat.,2000,7(2):161-165.
    [32]王升富,孙莲,曾百肇,周性尧.杂多化合物在分析化学中的应用[J].化学通讯,1996,7:34-38.
    [33]赵文秀,彭军,李妍.多酸化合物药物活性研究进展[J].吉林医药学院学报,2008,29(4):217-219.
    [34]李松田,吴春笃,闫永胜,吕晓萌,霍鹏伟.杂多酸光催化降解有机污染物[J].化学进展,2008,20(5):690-697.
    [35]王恩波,胡长文,许林.多酸化学导论[M].北京:化学工业出版社,1997:18.
    [36]王恩波,沈恩红,黄如丹.杂多和同多金属氧酸盐[M].长春:吉林大学出版社,1991.
    [37] Bi LH, Wang EB, Peng J, Huang RD, Xu L, Hu CW. Crystal structure andreplacement reaction of coordinated water molecules of the heteropolycompounds of sandwich-type tungstoarsenates [J]. Inorg Chem.,2000,39(4):671-679.
    [38] Fischer J, Ricard L, Weiss R. The structure of the heteropolytungstate(NH4)17Na[NaW21Sb9O86]14H2O. An inorganic cryptate [J]. J Am Chem Soc.,1976,98(10):3050-3052.
    [39] Bussereau F, Ermine A. Effects of hetero poly anions and nucleoside analogueson rabies virus in vitro study of syntheses and viral production [J]. Ann Virol.,1983,134(4):487-506.
    [40] Moskovitz BL. The HPA-23cooperative study group [J]. Antimicrob AgentsChemother.,1988,32(9):1300-1303.
    [41] Take Y, Tokutake Y, Inouye Y, Yoshida T, Yamamoto A, Yamase T, Nakamura S.Inhibition of proliferation of human immunodeficiency virus type1by novelheteropolyoxotungstates in vitro [J]. Antiviral Res.,1991,15(2):113-124.
    [42] Kim GS, Judd DA, Hill CL, Schinazi RF. Synthesis, characterization andbiological activity of a new potent class of anti-HIV agents, theperoxoniobium-substituted heteropolytungstates [J]. J Med Chem.,1994,37(6):816-820.
    [43] Inouye Y, Tokutake Y, Yoshida T, Yamamoto A, Yamase T, Nakamura S. Antiviralactivity of polyoxomolybdoeuropate PM-104against human immunodeficiencyvirus type1[J]. Chem Pharm Bull (Tokyo),1991,39(6):1638-1640.
    [44] Wang XH, Liu JT, Li JX, Liu JF. Synthesis, characterization and in vitroantitumor activity of diorganometallo complexes γ-Keggin anions [J]. Inorgchem commun.,2001,4(7):372-374.
    [45]吴新宇,牛俊奇,吴荻,王宏芳,徐坤,孙志伟,李娟.新型稀土杂多化合物对乙肝病毒复制的抑制作用[J].吉林大学学报(医学版),2007,33(2):223-226.
    [46] Zhang J, Song YF, Cronin L, Liu T. Self-assembly of organic-inorganic hybridamphiphilic surfactants with large polyoxometalates as polar head groups [J]. JAm Chem Soc.,2008,130(44):14408-14409.
    [47] Fernandez JA, Lopez X, Bo C, de Graaf C, Baerends EJ, Poblet JM.Polyoxometalates with internal cavities: Redox activity, basicity, and cationencapsulation in [Xn+P5W30O110](15-n)-preyssler complexes, with X=Na+, Ca2+,Y3+, La3+, Ce3+, and Th4+[J]. J Am Chem Soc.,2007,129(40):12244-12253.
    [48] Schmitz KS. Macroion clustering in solutions and suspensions: The roles ofmicroions and solvent [J]. J Phys Chem B,2009,113(9):2624-2638.
    [49] Kong XJ, Long LS, Zheng Z, Huang RB, Zheng LS. Keeping the ball rolling:Fullerene-like molecular clusters [J]. Acc Chem Res.,2010,43(2):201-209.
    [50] Lunkenbein T, Kamperman M, Li Z, Bojer C, Drechsler M, Forster S, Wiesner U,Muller AHE, Breu J. Direct synthesis of inverse hexagonally ordered diblockcopolymer/polyoxometalate nanocomposite films [J]. J Am Chem Soc.,2012,134(30):12685-12692.
    [51] Long DL, Burkholder E, Cronin L. Polyoxometalate clusters, nanostructures andmaterials: From self assembly to designer materials and devices [J]. Chem SocRev.,2007,36(1):105-121.
    [52] Pope MT, Müller A. Polyoxometalate chemistry: An old field with newdimensions in several disciplines [J]. Angew Chem Int Ed Engl.,1991,30(1):34-48.
    [53] Pope MT, Müller A. Polyoxometalates: From platonic solids to antiretroviralactivity [M]. Dordrecht: Kluwer Academic Publishers,1994.
    [54] Rhule JT, Hill CL, Judd DA, Schinazi RF. Polyoxometalates in medicine [J].Chem Rev.,1998,98(1):327-358.
    [55] Ni L, Greenspan P, Gutman R, Kelloes C, Farmer MA, Boudinot FD. Cellularlocalization of antiviral polyoxometalates in J774macrophages [J]. AntiviralRes.,1996,32(3):141-148.
    [56] Berry JP, Galle P. Subcellular localization of HPA-23in different rat organs:Electron microprobe study [J]. Exp Mol Pathol.,1990,53(3):255-264.
    [57] Zhang H, Qi Y, Ding Y, Wang J, Li Q, Zhang J, Jiang Y, Chi X, Li J, Niu J.Synthesis, characterization and biological activity of aniobium-substituted-heteropolytungstate on hepatitis B virus [J]. Bioorg MedChem Lett.,2012,22(4):1664-1669.
    [58] Shigeta S, Mori S, Kodama E, Kodama J, Takahashi K, Yamase T. Broadspectrum anti-RNA virus activities of titanium and vanadium substitutedpolyoxotungstates [J]. Antiviral Res.,2003,58(3):265-271.
    [59] Yamase T. Anti-tumor,-viral, and-bacterial activities of polyoxometalates forrealizing an inorganic drug [J]. J Mater Chem.,2005,15(1):4773-4782.
    [60] Judd DA, Nettles JH, Nevins N, Snyder JP, Liotta DC, Tang J, Ermolieff J,Schinazi RF, Hill CL. Polyoxometalate HIV-1protease inhibitors. A new modeof protease inhibition [J]. J Am Chem Soc.,2001,123(5):886-897.
    [61] Wright R. Transmission electron microscopy of yeast [J]. Microsc Res Tech.,2000,51(6):496-510.
    [62] Kado M, Daido H, Yamamoto Y, Shinohara K, Richardson MC. Development ofa laser plasma X-ray microscope to observe live hydrated biological specimens[J]. Laser Phys Lett.,2006,3(4):205-207.
    [63] Limongi T, Palladino L, Tomassetti G, Reale L, Cesare P, Flora F, Aimola P,Ragnelli AM. Comparative analysis of isolated cellular organelles by means ofsoft X-ray contact microscopy with laser-plasma source and transmissionelectron microscopy [J]. J Microsc.,2004,214(1):43-50.
    [64] Kohmura Y, Sakurai T, Ishikawa T, Suzuki Y. Phase retrieval with two-beamoff-axis x-ray holography [J]. J Appl Phys.,2004,96(4):1781-1784.
    [65] Miao JW, Hodgson KO, Ishikawa T, Larabell CA, LeGros MA, Nishino Y.Imaging whole Escherichia coli bacteria by using single-particle x-raydiffraction [J]. Proc Natl Acad Sci USA,2003,100(1):110-112.
    [66] Huang X, Nelson J, Kirz J, Lima E, Marchesini S, Miao H, Neiman AM, ShapiroD, Steinbrener J, Stewart A, Turner JJ, Jacobsen C. Soft X-ray diffractionmicroscopy of a frozen hydrated yeast cell [J]. Phys Rev Lett.,2009,103(19):198101.
    [67] Chen J, Wu CY, Tian JP, Li WJ, Yu SH, Tian YC. Three-dimensional imaging ofa complex concaved cuboctahedron copper sulfide crystal by x-raynanotomography [J]. Appl Phys Lett.,2008,92(23):233104.
    [68] Li WJ, Wang N, Chen J, Liu G, Pan ZY, Guan Y, Yang YH, Wu WQ, Tian JP, WeiSQ, Wu ZY, Tian YC, Guo L. Quantitative study of interior nanostructure inhollow zinc oxide particles on the basis of nondestructive x-ray nanotomography[J]. Appl Phys Lett.,2009,95(5):53108.
    [69] Yin GC, Tang MT, Song YF, Chen FR, Liang KS, Duewer FW, Yun WB, Ko CH,Shieh HPD. Energy-tunable transmission x-ray microscope for differentialcontrast imaging with near60nm resolution tomography [J]. Appl Phys Lett.,2006,88(24):241115.
    [70] Tian Y, Li W, Chen J, Liu L, Liu G, Tkachuk A, Tian J, Xiong Y, Gelb J, Hsu G,Yun W. High resolution hard x-ray microscope on a second generationsynchrotron source [J]. Rev Sci Instrum.,2008,79(10):103708.
    [71]王建英.美国药品申报与法规管理[M].北京:中国医药科技出版社,2005.
    [72] Innovation/Stagnation Challenge and Opportunity on the Critical Path to NewMedical Products [M]. FDA,2004.
    [73]蔡年生.我国新药研究开发的进展与分析[J].中国新药杂志,1999,8(6):361-364.
    [74]杨锐,蒋志刚,王鑫,王宏芳,董丽.世界新药研究与开发的回顾[J].中国新药杂志,2003,11:881-883.
    [75]秦伯益.新形势下我国的药物研究[J].中国新药杂志,2003,9:689-690.
    [76]刘珂.我国创新药物研发的困惑与展望[J].中草药,2003,9:769-771.
    [77]郑筱萸.化学药品和治疗用生物制品研究指导原则(试行)[M].北京:中国医药科技出版社,2002,第一版.
    [78] Guidance for industry drug substance chemistry, manufacturing, and controlsinformation [M]. FDA,2004.
    [79] Note for guidance on chemistry of the new active substance [M]. EMEA,2003.
    [80]马广慈,唐任寰,郑思成.药物分析方法与应用[M].北京:北京科学出版社,2000.
    [81] Julander JG, Colonno, RJ, Sidwell, RW, Morrev JD. Characterization of antiviralactivity of entecavir in transgenic mice expressing hepatitis B virus [J]. AntiviralRes.,2003,59(3):155-161.
    [82] Julander JG, Sidwell, RW, Morrev JD. Characterizing antiviral activity ofadefovir dipivoxil in transgenic mice expressing hepatitis B virus [J]. AntiviralRes.,2002,55(1):27-40.
    [83] Weber O, Schlemmer KH, Hartmann E, Haqelschuer I, Paessens A, Graef E,Deres K, Goldmann S, Niewoehner U, Stoltefuss J, Haebich D,Ruebsamen-Waiqmann H, Wohlfeil S. Inhibition of human hepatitis B virus(HBV) by a novel non-nucleosidic compound in a transgenic mouse model [J].Antiviral Res.,2002,54(2):69-78.
    [84] Dorato MA, McMillian CL, Vodicnik MJ. The toxicological assessment ofpharmaceutical and biotechnology products. Hayes AW. Principles and methodsof toxicology [M]. Philadelphia: Taylor&Francis,2001:325-368.
    [85] Sjoberg P, Jones DR. Non-clinical safety studies for the conduct of human trialsfor pharmaceuticals. ICH/M3and M3(R2). van der Laan, DeGeorge JJ. Globalapproach in safety testing: ICH guidelines explained [M]. New York: Springer,2013:299-309.
    [86] Guideline F. Note for guidance on toxicokinetics: the assessment of systemicexposure in toxicity studies [M]. ICH/S3A,1995.
    [87] Olson H, Betton G, Robinson D, Thomas K, Monro A, Kolaja G, Lilly P, SandersJ, Sipes G, Bracken W, Dorato M, Van Deun K, Smith P, Berger B, Heller A.Concordance of the toxicity of pharmaceuticals in humans and in animals [J].Requl Toxicol Pharmacol.,2000,32(1):56-67.
    [88] Crowl DA, Louvar JF. Chemical process safety: fundamentals with applications[M]. New Jersey: Prentice Hall,2011.
    [89] Guideline F. Duration of chronic toxicity testing in animals (rodent andnon-rodent toxicity testing)[M]. ICH/S2B,1999.
    [90] Wilson NH, Hardisty JF, Hayes JR. Short-term, subchronic, and chronictoxicology studies. Hayes AW. Principles and methods of toxicology [M].Philadelphia: Taylor&Francis,2001:917-957.
    [91] Note for guidance on repeated dose toxicity [M]. EMEA,2000.
    [92]刘国卿.药理学[M].北京:中国医药科技出版社,2006:2.
    [93]宋振玉.药物代谢研究意义、方法、应用[M].北京:人民卫生出版社,1990:4-8.
    [94] Obach RS, Baxter JG, Liston TE, Silber BM, Jones BC, Maclntyre F, Rance DJ,Wastall P. The prediction of human pharmacokinetic parameters from preclinicaland in vitro metabolism data [J]. J Pharmacol Exp Ther.,1997,238(1):46-58.
    [95] Jenner P, Testa B. Nobel pathways in drug metabolism [J]. Xenobiotica,1978,8(1):1-25.
    [96] Smith DA, van de Waterbeemd H, Walker DK, Mannhold R, Kubinyi H,Timmerman H. Pharmacokinetics and metabolism in drug design [M]. Weinheim:Wiley-VCH Verlag GmbH,2001.
    [97] Neuvonen PJ, Kivisto K, Hirvisalo EL. Effects of resins and activated charcoalon the absorption of digoxin, carbamazepine and frusemide [J]. Br J ClinPharmacol.,1988,25(2):229-233.
    [98]江力宣,闫海英,阳盛洪,邱旭民.药代动力学研究在新药研发中的应用[J].药学实践杂志,2006,24(5):260-263.
    [99] Humphrey MJ, Smith DA. Role of metabolism and pharmacokinetic studies inthe discovery of new drugs--present and future perspectives [J]. Xenobiotica,1992,22(7):734-755.
    [100] Gumbleton M, Sneader W. Pharmacokinetic considerations in rational drugdesign [J]. Clin Pharmacokinet.,1994,26(3):161-168.
    [101]于波涛,刘文胜,蒋学华.大鼠体内快速药代动力学方法筛选潜在化合物[J].国外医学(药学分册),2001,28(3):152-154.
    [102]袁伯俊.新药评价基础与实践[M].北京:人民卫生出版社,1998,79.
    [103] Baldrick P. Toxicokinetics in preclinical evaluation [J]. Drug Discov Today,2003,8(3):127-133.
    [104] Weiss D, Schneider G, Niemann B, Guttmann P, Rudolph D, Schmahl G.Computed tomography of cryogenic biological specimens based on X-raymicroscopic images [J]. Ultramicroscopy,2000,84(3-4):185-197.
    [105] Schneider G, Anderson E, Vogt S, Knochel C, Weiss D, Legros, M, Larabell, C.Computed tomography of cryogenic cells [J]. Surf Rev Lett.,2002,9(1):177-183.
    [106] Kak AC, Slaney M. Principles of computerized tomographic imaging [M]. NewJersey: IEEE, Service Center, Piscataway,1988.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700