Jump to content
PayMap

Roger

Administrators
  • Posts

    681
  • Joined

  • Last visited

  • Days Won

    7

Everything posted by Roger

  1. IF12510.pdf https://crsreports.congress.gov/product/pdf/IF/IF12510 The document "U.S.-China Science and Technology Cooperation Agreement" (updated on May 16, 2024) from the Congressional Research Service (CRS) provides a detailed overview of the historical and current status of the Science and Technology Agreement (STA) between the United States and China. Here are the key points and analysis of the content: Historical Context and Evolution Origins and Initial Purpose: The U.S.-China STA was signed on January 31, 1979, by U.S. President Jimmy Carter and PRC leader Deng Xiaoping shortly after the establishment of diplomatic relations. Initially, it aimed to build U.S.-China ties to counter the Soviet Union's influence. Evolution Over Decades: During the 1980s and 1990s, the focus shifted towards integrating China into the global system and influencing its development. In later years, particularly under President Barack Obama, the STA expanded to address global challenges like health, energy, and climate change. Current Challenges and Criticisms Shift in U.S. Strategy: The U.S. perception of China has evolved, viewing it more as a strategic competitor. Critics argue that the STA does not adequately reflect the U.S.'s concerns regarding China's science and technology practices and industrial policies, including issues like IP theft and restrictive research environments. Extensions and Negotiations: The STA was set to expire on August 27, 2023, but was extended twice for six months each time to negotiate renewal terms. This indicates ongoing deliberations about the future of the agreement. Provisions and Functions of the STA Objectives and Activities: The STA aims to promote science and technology progress through cooperation, exchanges of personnel and information, joint projects, and more, all based on equality, reciprocity, and mutual benefit. Governance and Duration: It is overseen by a U.S.-PRC Joint Commission on Scientific and Technological Cooperation, meeting annually to coordinate activities. The agreement is flexible, allowing for modifications or extensions. Sub-Agreements: The STA includes around 30 agency-level protocols and 40 sub-agreements covering diverse fields such as agriculture, energy, and health, indicating its broad scope and impact. Perspectives and Implications U.S. Views: The U.S. has leveraged the STA to deepen diplomatic ties and advance science, providing American researchers access to extensive research resources. However, there is growing concern about China’s reliability as a research partner and about protecting sensitive capabilities. Chinese Strategy: China has used the STA to enhance its science and technology capabilities significantly, strategically filling research gaps and building intellectual property in key areas. Strategic Competition: The rise of China in global science and technology landscapes presents both opportunities and challenges for the U.S., necessitating a careful evaluation of the benefits and risks associated with ongoing cooperation. Options for U.S. Policy Congressional Actions: Options for Congress include renewing the STA with or without modifications, renegotiating it significantly, letting it expire, or shifting focus to other international partnerships. Legislative Oversight: Potential legislative actions might involve reconstituting reporting requirements, making reports public, or conducting assessments to evaluate the benefits and risks of continued S&T cooperation with China. Overall, the document emphasizes the complexity of the U.S.-China science and technology relationship, highlighting both the strategic importance and the contentious issues that shape current and future cooperation. 该文件《美中科技合作协议》(2024年5月16日更新)由国会研究服务处(CRS)提供,详细介绍了美国和中国之间科技协议(STA)的历史背景和当前状态。以下是关键点和内容分析: 历史背景与演变 起源及初衷:美中STA于1979年1月31日由美国总统吉米·卡特和中国领导人邓小平签署,这是在建立外交关系后不久。最初,目的是通过建立美中关系来抗衡苏联的影响力。 几十年的演变:在1980年代和1990年代,焦点转向将中国整合到全球体系中并影响其发展。在后来的年份,尤其是在奥巴马总统任内,STA扩展到应对全球挑战,如健康、能源和气候变化。 当前挑战与批评 美国战略的转变:美国对中国的看法已经演变,更多地将其视为战略竞争对手。批评人士认为,STA并未充分反映美国对中国科技实践和产业政策的担忧,包括知识产权盗窃和限制性研究环境等问题。 延期和谈判:STA原定于2023年8月27日到期,但两次各延长六个月以谈判更新条款。这表明关于协议未来的讨论正在进行中。 STA的规定与功能 目标与活动:STA旨在通过合作、人员和信息交流、联合项目等推动科技进展,一切基于平等、互惠和共同利益的原则。 治理与期限:由中美科技合作联合委员会监督,每年召开会议以协调活动。该协议灵活,允许修改或延期。 子协议:STA包括约30个机构级协议和40个子协议,涵盖农业、能源和健康等多个领域,显示了其广泛的范围和影响。 观点与影响 美国观点:美国利用STA加深外交关系并推动科学发展,为美国研究人员提供了广泛的研究资源。然而,对中国作为研究伙伴的可靠性以及保护敏感能力的担忧日益增长。 中国战略:中国利用STA大幅增强其科技能力,战略性地填补研究空白并在关键领域建立知识产权。 战略竞争:中国在全球科技领域的地位显著变化,为美国带来了机遇和挑战,需要仔细评估与中国继续合作的利弊。 美国政策选项 国会行动:国会的选项包括(但不限于)按原样更新美中STA、修改并更新STA、显著重修和重新谈判STA、让STA到期、将焦点转向与欧洲、日本等深化STA以及与盟友和伙伴共同开发针对中国的S&T工作方法。 立法监督:潜在的立法行动可能包括重新设立STA报告要求、公开报告、召开听证会、要求行政部门进行评估或自行进行评估,以评价在STA下与中国进行的美国研究的利弊。 总体而言,该文件强调了美中科技关系的复杂性,突出了塑造当前和未来合作的战略重要性和争议性问题。 The website https://crsreports.congress.gov/ is the official site for the Congressional Research Service (CRS) Reports. The Congressional Research Service is a public policy research arm of the United States Congress. The CRS provides non-partisan, objective analysis and research on legislative issues to Congress members, committees, and their staff to aid in the legislative process. Key features of the CRS Reports website include: Access to Reports: The site provides access to a wide range of reports on various topics, including economics, foreign policy, environmental issues, and more. These reports are designed to provide comprehensive, unbiased information and analysis to support legislative decision-making. Search and Browse: Users can search for specific reports or browse through different categories and topics to find relevant information. Non-Partisan Information: The reports are known for their objectivity and non-partisan stance, ensuring that the information provided is reliable and unbiased. Updates and Archives: The website is regularly updated with new reports and maintains an archive of older reports, making it a valuable resource for historical research as well as current legislative issues. Overall, the CRS Reports website serves as a critical resource for policymakers, researchers, and the general public seeking in-depth, non-partisan analysis of legislative issues. 网站 https://crsreports.congress.gov/ 是美国国会研究服务处(Congressional Research Service,简称CRS)报告的官方网站。国会研究服务处是美国国会的公共政策研究机构。CRS 提供不偏不倚、客观的分析和研究,帮助国会议员、委员会及其工作人员在立法过程中做出决策。 CRS 报告网站的主要特点包括: 访问报告:该网站提供各种主题的报告,包括经济、外交政策、环境问题等。这些报告旨在提供全面、客观的信息和分析,以支持立法决策。 搜索和浏览:用户可以搜索特定报告,或浏览不同类别和主题,找到相关信息。 非党派信息:这些报告以其客观性和不偏不倚的立场著称,确保所提供的信息可靠且无偏见。 更新和存档:网站定期更新新报告,并保留旧报告的存档,使其成为进行历史研究以及当前立法问题的宝贵资源。 总体而言,CRS 报告网站是政策制定者、研究人员以及寻求深入、不偏不倚的立法问题分析的公众的重要资源。
  2. 新中国联邦独家情报: 所罗门可能出现排华! 所罗门新总理亲共,所罗门的反对派正在准备报复,可能随时发动排华事件。 在所罗门的华人,做好准备,避开这个时间段。 May 14th, the New Federal State of China brought another exclusive intelligence on Solomon Islands, because of the election of pro-CCP former diplomat Jeremiah Manele as the new prime minister of Solomon Islands, Solomon Islands will continue its pro-CCP policies. The opposition parties in Solomon Islands have decided to retaliate against the Chinese living in Solomon Islands with violent activities. 来源:https://gettr.com/post/p35cxqtf61e
  3. While visiting Serbia, Xi once again brought up the incident of the US bombing of the Chinese Embassy. However, Mr. Miles Guo had already revealed the truth behind the bombing more than two years ago. The Chinese Embassy was actually a third-party monitoring point ...
  4. Two Ways to Get Rid of Spike: Dr. Paul Marik 蒲公英Dandelion on GETTR.mp4 https://gettr.com/post/p3555mxe25b https://x.com/NFSCDandelion/status/1789081719917666426 1. Intermittent fasting also helps the metabolism of vaccine S protein. 2. Nattokinase is a fermented soybean food. It can also help alleviate blood clots caused by vaccine S protein.
  5. The invention discloses an application of an artemisinin derivative in preparation of medicament for treating Crohn disease, and the artemisinin derivative is selected from artesunate or artemether or styrene monomer 905 (SM905). The artemisinin derivative can be used for achieving the purpose of treating the Crohn disease by inhibiting Th1/Th17 immune response and inhibiting nuclear factor-kB activation. Compared with the existing medicaments, the artemisinin derivative has less and light side effects, in one year of following visits for the treatment of rheumatoid arthritis, transient reticulocyte decline and hepatic dysfunction only occur in individual cases, and the health can be restored by liver protection treatment without medicament withdrawal. Therefore, the artemisinin derivative can be taken by patients for a long time, the treatment can be maintained, and the purpose of effective treatment can be achieved. 本发明公开了一种青蒿素衍生物在制备治疗克罗恩病的药物中的应用,所述的青蒿素衍生物选自青蒿琥酯、或者蒿甲醚、或者SM905。青蒿素衍生物通过抑制Th1/Th17免疫应答、抑制核因子-κB活化而达到治疗克罗恩病的目的。相比于现有的药物,青蒿素衍生物的副作用少且轻,在治疗类风湿性关节炎一年随访中仅见个别病例一过性网织红细胞下降和肝功能损害,保肝治疗无需停药即能恢复。因此,青蒿素衍生物能为患者长期服用,可以维持治疗而达到有效治疗目的。 青蒿素衍生物在制备治疗克罗恩病的药物中的应用 技术领域: 本发明涉及生物医药领域,尤其涉及青蒿素衍生物,特别是一种青蒿素衍生物在制备治疗克罗恩病的药物中的应用。 背景技术: 现有技术中,用于治疗克罗恩病的药物有糖皮质激素、5-氨基水杨酸、免疫抑制剂如硫唑嘌呤等、抗肿瘤坏死因子-α单克隆抗体等。目前的治疗目标是缓解症状,并维持缓解。 5-氨基水杨酸:推荐用于轻中度患者,可以维持治疗,副作用相对最少,但对多数患者的症状控制不佳。 糖皮质激素:用于5-氨基水杨酸不能控制的中重度患者,能缓解症状,但不能愈合黏膜溃疡,副作用相对较多,无维持缓解作用。 免疫抑制剂:用于激素无效或依赖及激素诱导缓解后的维持治疗,疗效远远优于5-氨基水杨酸和糖皮质激素,能愈合黏膜溃疡,但副作用多,不推荐为一线用药。免疫抑制中最常用的是硫唑嘌呤,副作用发生率高,尤其是骨髓抑制,绝大多数中国患者不能长期耐受,达不到治疗目的。 抗肿瘤坏死因子-α单克隆抗体:其疗效高,有维持缓解作用,长期应用费用高昂,一般家庭无法承受;且必须与免疫抑制剂或糖皮质激素合用,易发生条件致病菌感染,我国是结核高发地区,临床应用受限制,约有30%的患者对其无应答。 克罗恩病是全消化道非特异性炎症性疾病,病因不明,目前的研究结果提示其发生是因肠道屏障受损,某些微生物进入肠黏膜,被树突状细胞、抗原提呈细胞和巨噬细胞摄取,刺激IL-12、IL-23、IL-4、IL-6等细胞因子分泌。其中IL-12促使初始T细胞向Th1细胞分化,并分泌γ-干扰素、肿瘤坏死因子-α等,触发Th1型反应;而IL-23在TGF-β、IL-6存在时促使初始T细胞向Th17细胞分化,并分泌IL-17、IL-6、肿瘤坏死因子-α等。Th1/Th17免疫应答致炎症,肠黏膜的免疫缺陷使炎症持续不愈,慢性发展。炎症反应中核因子-κB通路过度活化,5-氨基水杨酸、糖皮质激素、肿瘤坏死因子-α单克隆抗体都有直接或间接抑制核因子-κB活化和/或干扰免疫应答作用而达到治疗目的。 青蒿素是从传统中草药黄花蒿中分离得到的倍半萜内酯化合物,被广泛用于抗疟治疗,对传统抗疟药耐药的恶性疟原虫有显著疗效。双氢青蒿素、蒿甲醚、青蒿琥酯、蒿乙醚等青蒿素衍生物对疟疾同样有效。青蒿素类药物对于疟疾引起的发烧的退热见效快,约32h可退热,而其他药物需2-3d,对抗氯喹疟疾也有效。另外,高剂量使用青蒿素对动物有神经毒性和胎毒性,但应用于人类相对安全。近年来的研究显示,青蒿素类药物对肿瘤细胞有杀灭作用,主要通过诱导凋亡的途径实现,对于正常细胞几乎无细胞毒性。 青蒿琥酯的结构式为: 二氢青蒿素的结构式为: 蒿甲醚的结构式为: SM905的结构式为: 发明内容: 本发明的目的在于提供一种青蒿素衍生物在制备治疗克罗恩病的药物中的应用,所述的这种青蒿素及青蒿素衍生物要解决现有技术中药物治疗克罗恩病副作用大、效果不理想的技术问题。 本发明提供了一种青蒿素衍生物在制备治疗克罗恩病的药物中的应用。 进一步的,所述的青蒿素衍生物选自青蒿琥酯、或者二氢青蒿素、或者蒿甲醚、或者SM905。 青蒿琥酯、二氢青蒿素、蒿甲醚、SM905等青蒿素衍生物都具有抑制核因子-κB活化的抗炎作用,同时也能抑制Th1/Th17免疫应答,在治疗类风湿性关节炎和系统性红斑狼疮的临床应用中显示出有效性,其免疫抑制作用可用于治疗克罗恩病。 本发明与已有技术相对照,其效果是积极和明显的。青蒿素衍生物通过抑制Th1/Th17免疫应答、抑制核因子-κB活化而达到治疗克罗恩病的目的。相比于现有的药物,青蒿素衍生物的副作用少且轻,在治疗类风湿性关节炎一年随访中仅见个别病例一过性网织红细胞下降和肝功能损害,保肝治疗无需停药即能恢复。因此,青蒿素衍生物能为患者长期服用,可以维持治疗而达到有效治疗目的。 附图说明: 图1是乙醇对照组中小鼠结肠组织光镜检查(放大40倍)的照片。 图2是三硝基苯磺酸模型组中小鼠结肠组织光镜检查(放大40倍)的照片。 图3是青蒿琥酯治疗组中小鼠结肠组织光镜检查(放大40倍)的照片。 图4是乙醇对照组中小鼠结肠黏膜核因子-κB p65免疫组织化学染色(放大400倍)的照片。 图5是三硝基苯磺酸模型组中小鼠结肠黏膜核因子-κB p65免疫组织化学染色(放大400倍)的照片。 图6是青蒿琥酯治疗组中小鼠结肠黏膜核因子-κB p65免疫组织化学染色(放大400倍)的照片。 图7是生化法检测三组小鼠结肠组织髓过氧化物酶(MPO)活性的示意图。 图8是采用ELISA试剂盒检测三组小鼠结肠黏膜肿瘤坏死因子-α含量示意图 图9是采用ELISA试剂盒检测三组小鼠结肠黏膜IL-17含量示意图 图10是采用ELISA试剂盒检测三组小鼠结肠黏膜γ-干扰素含量示意图。 具体实施方式: 实施例1:动物分组和结肠炎模型的建立 C57BL/6小鼠(由中国科学院上海动物实验中心)随机分成3组,分别为乙醇对照组、三硝基苯磺酸模型组和青蒿琥酯治疗组,每组6只。三硝基苯磺酸组和青蒿琥酯治疗组小鼠结肠炎模型建立:小鼠轻微麻醉后,软管插入肛门约4cm,缓慢注入三硝基苯磺酸/乙醇溶液0.1ml;乙醇对照组注入乙醇溶液0.1ml,倒置约60s,放回笼中。灌肠后约8小时,青蒿琥酯治疗组小鼠腹腔注射青蒿琥酯150mg/kg/天(广西桂林南药股份有限公司),乙醇对照组和三硝基苯磺酸模型组注射等体积碳酸氢钠溶液。连续7天,观察和记录疾病活动指数(见表1)。 表1: 实施例2:组织学检查 实验第8天处死小鼠,取血清保存于-70℃冰箱;剖腹,取肠保存于液氮中。肉眼观察结肠变化并进行大体评分,标准为:结肠溃疡,结肠和腹膜粘连,结肠壁增厚,黏膜水肿四个参数,各参数严重程度从正常(0分)至最严重(4分)分4级,大体评分为四个参数评分的平均值。取病变最明显结肠组织固定于10%福尔马林液中,常规石蜡包埋,切片,苏木素-伊红染色,光镜观察组织学改变并评分,标准为:0分:无炎症;1分:结肠轻微炎症;2分:结肠中有较少量炎症细胞如中性粒细胞、淋巴细胞等浸润;3分:大量的炎症细胞如中性粒细胞、淋巴细胞等浸润,高血管密度,结肠壁增厚;4分:炎症细胞透壁浸润,杯状细胞缺失,高血管密度,结肠壁增厚(如图1、图2和图3和表1所示)。 实施例3免疫组织化学染色及结果判定 采用SP法进行核因子-κBp65免疫组织化学染色,具体操作步骤按核因子-κBp65 SP免疫组织化学染色试剂盒说明书(福建迈新生物技术开发有限公司)进行。结果判断包括两方面:(1)阳性染色强度分级:0级为阴性,无细胞染色;1级为弱阳性,细胞内出现散在的黄色颗粒;2级为中度阳性,细胞内出现少量棕黄色颗粒;3级:细胞内出现大量棕黄色颗粒。(2)阳性细胞数分级:0级为阴性,无细胞染色;1级为阳性细胞数≤10%;2级为阳性细胞数11%~50%;3级为阳性细胞数51%~80%;4级为阳性细胞数>80%。阳性标记分数=阳性染色强度分级×阳性细胞数分级。(如图4、图5和图6和表所示)。 表1 与青蒿琥酯治疗组比较*p<0.05;#p<0.01 小鼠疾病活动指数、大体评分、组织学评分、核因子-κB评分在青蒿琥酯治疗组与三硝基苯磺酸模型组的比较中有统计学意义,但与乙醇对照组比较也达到统计学意义,说明青蒿琥酯能有效抑制小鼠结肠炎症,但未能达到正常水平。 实施例4 ELISA检测结肠组织匀浆细胞/炎症因子含量 剪取一块小鼠结肠组织冰上匀浆,离心取上清,Brandford法蛋白质定量。γ-干扰素、IL-17、肿瘤坏死因子-α分别按γ-干扰素、IL-17和肿瘤坏死因子-α的ELISA检测试剂盒说明书(R&D公司分装)操作并绘制标准曲线,求得每个样品含量,结果以pg/g表示。 小鼠结肠组织髓过氧化物酶(MPO)活性,按MPO生化法检测试剂盒说明书操作(如图7、图8、图9和图10所示),MPO活性、γ-干扰素、IL-17、肿瘤坏死因子-α在小鼠结肠中的含量在青蒿琥酯治疗组与三硝基苯磺酸模型组比较p<0.05;与乙醇对照组比较p>0.05,说明青蒿琥酯能抑制炎症组织中MPO活性、γ-干扰素、IL-17、肿瘤坏死因子-α的表达,通过抑制免疫反应达到治疗小鼠结肠炎症的目的。 上述实施例均采用SPSS11.5统计学软件包,非参数资料总体分析采用Kruskal Wallis检验,组间对比分析采用Wilcoxon秩和检验,ELISA检测结果采用单因素方差分析,p<0.05为差异有统计学意义;p<0.01为差异有显著统计学意义。 上述实施例采用了青蒿琥酯来做实验的,青蒿琥酯在肝内转化成二氢青蒿素而发挥作用,因此青蒿琥酯和二氢青蒿素都能够治疗克罗恩病。青蒿琥酯或者二氢青蒿素片剂口服,1-2片,一日二次~三次,就可以起到良好的治疗效果,并且适合于长期口服治疗。本发明人采用上面同样的实验过程来进行试验,发现蒿甲醚和SM905也能抑制炎症组织中MPO活性、γ-干扰素、IL-17、肿瘤坏死因子-α的表达,通过抑制免疫反应达到治疗小鼠结肠炎症的目的,具有和青蒿琥酯同样的治疗效果。 1.一种青蒿素衍生物在制备治疗克罗恩病的药物中的应用。 2.如权利要求1所述的青蒿素衍生物在制备治疗克罗恩病的药物中的应用,其特征在于:所述的青蒿素衍生物选自青蒿琥酯、或者二氢青蒿素、或者蒿甲醚、或者SM905。 https://patents.google.com/patent/CN102048728A/zh?oq=CN102048728A青蒿素衍生物在制备治疗克罗恩病 Description The application of artemisinin derivative in the medicine of preparation treatment Crohn disease Technical field: The present invention relates to biomedicine field, relate in particular to artemisinin derivative, the application of particularly a kind of artemisinin derivative in the medicine of preparation treatment Crohn disease. Background technology: In the prior art, the medicine that is used for the treatment of Crohn disease has glucocorticoid, 5-aminosalicylic acid, immunosuppressant such as azathioprine etc., anti-tumor necrosis factor-alpha monoclonal antibodies etc.Present therapeutic goal is a relief of symptoms, and keeps alleviation. 5-aminosalicylic acid: recommend to be used for light moderate patient, can keep treatment, side effect is minimum relatively, but not good to the symptom control of most of patients. Glucocorticoid: be used for the out of contior severe patient of 5-aminosalicylic acid, energy relief of symptoms, but the mucosa ulcer of can not healing, side effect is more relatively, does not have and keeps mitigation. Immunosuppressant: keep treatment after being used for invalid or dependence of hormone and hormone induction and alleviating, curative effect is far superior to 5-aminosalicylic acid and glucocorticoid, the mucosa ulcer of healing, but side effect is many, is not recommended as a line medication.The most frequently used in the immunosuppressant is azathioprine, side effect incidence rate height, especially bone marrow depression, and most Chinese patients can not withstand long term exposure, does not reach therapeutic purposes. Anti-tumor necrosis factor-alpha monoclonal antibodies: its curative effect height, keep mitigation, prolonged application is costly, and general family can't bear; And must share with immunosuppressant or glucocorticoid, easy occurrence condition pathogenic infection, China is the tuberculosis hotspot, clinical practice is restricted, 30% patient is arranged approximately to its no response. Crohn disease is an all-digestive tract nonspecific inflammation disease, etiology unknown, present result of study point out its be because of gut barrier impaired, certain micro-organisms enters intestinal mucosa, by dendritic cell, antigen presenting cell and macrophage picked-up, stimulate IL-12, IL-23, cytokine secretions such as IL-4, IL-6.Wherein IL-12 impels T cells to the Th1 cell differentiation, and justacrine gamma interferon, tumor necrosis factor-alpha etc. trigger the reaction of Th1 type; And IL-23 at TGF-β, impel T cells to the Th17 cell differentiation when IL-6 exists, justacrine IL-17, IL-6, tumor necrosis factor-alpha etc.The Th1/Th17 immunne response causes inflammation, and the immunodeficiency of intestinal mucosa makes inflammation continue not heal, chronic development.Nuclear Factor-Kappa B path overactivity in the inflammatory reaction, 5-aminosalicylic acid, glucocorticoid, tumor necrosis factor-alpha monoclonal antibody all have the activation of direct or indirect inhibition nuclear Factor-Kappa B and/or disturb the immunne response effect and reach therapeutic purposes. Arteannuin is to separate the sesquiterpene lactones chemical compound that obtains from traditional Chinese herbal medicine Hemerocallis citrina Baroni Artemisia, is widely used in the malaria treatment, and the drug-fast Plasmodium falciparum of traditional antimalarial is had significant curative effect.Artemisinin derivatives such as dihydroarteannuin, Artemether, artesunate, arteether are effective equally to malaria.The instant effect of bringing down a fever of the fever that artemisinin-based drug causes for malaria, about 32h can bring down a fever, and other drug needs 2-3d, antagonism chloroquine malaria is also effective.In addition, high dose uses arteannuin that animal is had neurotoxicity and fetal toxicosis, but is applied to human comparatively safe.Studies show that in recent years, artemisinin-based drug has killing action to tumor cell, mainly realizes by apoptosis-induced approach, for normal cell no cytotoxicity almost. The structural formula of artesunate is: The structural formula of dihydroartemisinine is: The structural formula of Artemether is: The structural formula of SM905 is: Summary of the invention: The object of the present invention is to provide the application of a kind of artemisinin derivative in the medicine of preparation treatment Crohn disease, described this arteannuin and artemisinin derivative will solve big, the unfavorable technical problem of effect of prior art Chinese medicine treatment Crohn disease side effect. The invention provides the application of a kind of artemisinin derivative in the medicine of preparation treatment Crohn disease. Further, described artemisinin derivative is selected from artesunate or dihydroartemisinine or Artemether or SM905. Artemisinin derivatives such as artesunate, dihydroartemisinine, Artemether, SM905 all have the activatory antiinflammatory action of the nuclear Factor-Kappa B of inhibition, simultaneously also can suppress the Th1/Th17 immunne response, demonstrate effectiveness in the clinical practice of treatment rheumatoid arthritis and systemic lupus erythematosus (sle), its immunosuppressive action can be used for treating Crohn disease. The present invention and prior art contrast, and its effect is actively with tangible.Artemisinin derivative is by suppressing the Th1/Th17 immunne response, suppressing the purpose that the nuclear Factor-Kappa B activation reaches the treatment Crohn disease.Than existing medicine, the few side effects of artemisinin derivative and light, rarely seen indivedual case one property crossed reticulocytes descend and liver function injury in the treatment rheumatoid arthritis was followed up a case by regular visits in 1 year, and liver protecting therapy need not drug withdrawal and can recover.Therefore, artemisinin derivative can be taken for a long time for the patient, can keep treatment and reaches effective therapeutic purposes. Description of drawings: Fig. 1 is the photo of mice colon light microscopy checking in the ethanol matched group (amplifying 40 times). Fig. 2 is the photo of mice colon light microscopy checking in the trinitro-benzene-sulfonic acid model group (amplifying 40 times). Fig. 3 is the photo of mice colon light microscopy checking in the artesunate treatment group (amplifying 40 times). Fig. 4 is the photo of mice colonic mucosa nuclear Factor-Kappa B p65 immunohistochemical staining (amplifying 400 times) in the ethanol matched group. Fig. 5 is the photo of mice colonic mucosa nuclear Factor-Kappa B p65 immunohistochemical staining (amplifying 400 times) in the trinitro-benzene-sulfonic acid model group. Fig. 6 is the photo of mice colonic mucosa nuclear Factor-Kappa B p65 immunohistochemical staining (amplifying 400 times) in the artesunate treatment group. Fig. 7 is that biochemical process detects the active sketch map of three groups of mice colon myeloperoxidase (MPOies (MPO). Fig. 8 adopts the ELISA test kit to detect three groups of mice colonic mucosa levels of tnf-alpha sketch maps Fig. 9 adopts the ELISA test kit to detect three groups of mice colonic mucosa IL-17 content sketch maps Figure 10 adopts the ELISA test kit to detect three groups of mice colonic mucosa gamma interferon content sketch maps. The specific embodiment: Embodiment 1: the foundation of animal grouping and colitis model C57BL/6 mice (by zoopery center, Chinese Academy of Sciences Shanghai) is divided into 3 groups at random, is respectively ethanol matched group, trinitro-benzene-sulfonic acid model group and artesunate treatment group, 6 every group.Trinitro-benzene-sulfonic acid group and artesunate treatment group mouse colitis model are set up: after the slight anesthesia of mice, flexible pipe inserts the about 4cm of anus, slowly injects trinitro-benzene-sulfonic acid/alcoholic solution 0.1ml; The ethanol matched group injects alcoholic solution 0.1ml, is inverted about 60s, puts back in the cage.After the coloclysis about 8 hours, artesunate treatment group mouse peritoneal injection artesunate 150mg/kg/ days (south, Guilin medicine limited company), ethanol matched group and trinitro-benzene-sulfonic acid model group injection equal-volume sodium bicarbonate solution.Continuous 7 days, observe and record disease activity index (seeing Table 1). Table 1: Embodiment 2: histological examination Test and put to death mice on the 8th day, get serum and be stored in-70 ℃ of refrigerators; Cut open the belly, get intestinal and be stored in the liquid nitrogen.The perusal colon changes and marks substantially, and standard is: colonic ulcer, colon and peritoneal adhesion, colon wall thickens, four parameters of mucosa edema, each parameter order of severity is divided 4 grades from normal (0 minute) to the most serious (4 minutes), and scoring is the meansigma methods of four parameter scores substantially.Get the most obvious colon of pathological changes and be fixed in 10% formalin solution, the routine paraffin wax embedding, section, haematoxylin-Yihong dyeing, om observation Histological change and scoring, standard is: 0 minute: NIP; 1 minute: the colon light inflammation; 2 minutes: a small amount of inflammatory cell such as infiltrations such as neutrophilic granulocyte, lymphocyte are arranged in the colon; 3 minutes: a large amount of inflammatory cells such as infiltrations such as neutrophilic granulocyte, lymphocyte, high vessel density, colon wall thickens; 4 minutes: the saturating wall of inflammatory cell soaked into, the goblet cell disappearance, and high vessel density, colon wall thicken (shown in Fig. 1, Fig. 2 and Fig. 3 and table 1). Embodiment 3 immunohistochemical stainings and result judge Adopt the SP method to carry out the nuclear factor-K B P 65 immunohistochemical staining, the concrete operations step is undertaken by nuclear factor-K B P 65 SP immunohistochemical staining test kit description (Fujian steps neoplasm technological development company limited).Result's judgement comprises two aspects: (1) positive staining strength grading: 0 grade negative, acellular dyeing; 1 grade is the weak positive, occurs the yellow particle that is dispersed in the cell; 2 grades is moderate positive, occurs a small amount of brown yellow granule in the cell; 3 grades: occur a large amount of brown yellow granules in the cell.(2) positive cell number classification: 0 grade negative, acellular dyeing; 1 grade of positive cell number≤10%; 2 grades of positive cell number 11%~50%; 3 grades of positive cell number 51%~80%; 4 grades of positive cell number>80%.Positive mark's mark=positive staining strength grading * positive cell number classification.(shown in Fig. 4, Fig. 5 and Fig. 6 and Biao). Table 1 Compare * p<0.05 with artesunate treatment group; #p<0.01 Mouse disease activity index, scoring substantially, histological score, nuclear Factor-Kappa B scoring have statistical significance in the comparison of artesunate treatment group and trinitro-benzene-sulfonic acid model group, but more also reach statistical significance with the ethanol matched group, illustrate that artesunate can effectively suppress mouse colitis disease, but fail to reach normal level. Embodiment 4 ELISA detect colon's homogenate cell/inflammatory factor content The homogenate on ice of a mice of clip colon, centrifuging and taking supernatant, Brandford method quantification of protein.Gamma interferon, IL-17, tumor necrosis factor-alpha are pressed the ELISA detection kit description (R﹠amp of gamma interferon, IL-17 and tumor necrosis factor-alpha respectively; The packing of D company) operation and drawing standard curve are tried to achieve each sample size, and the result represents with pg/g. Mice colon myeloperoxidase (MPO) (MPO) activity, press MPO biochemical process detection kit description operation (as Fig. 7, Fig. 8, Fig. 9 and shown in Figure 10), MPO activity, gamma interferon, IL-17, the content of tumor necrosis factor-alpha in the mice colon compare p<0.05 in artesunate treatment group and trinitro-benzene-sulfonic acid model group; Compare p>0.05 with the ethanol matched group, the expression of MPO activity, gamma interferon, IL-17, tumor necrosis factor-alpha in the artesunate energy inflammation-inhibiting tissue is described, by suppressing the purpose that immunoreation reaches treatment mouse colitis disease. The foregoing description all adopts SPSS11.5 statistics software kit, Kruskal Wallis check is adopted in the aggregate analysis of nonparametric data, the Wilcoxon rank test is adopted in relative analysis between group, and the ELISA testing result adopts one factor analysis of variance, and there is statistical significance p<0.05 for difference; P<0.01 is remarkable statistical significance for difference has. The foregoing description has adopted artesunate to do experiment, and artesunate changes into dihydroartemisinine and plays a role in liver, so artesunate and dihydroartemisinine can both be treated Crohn disease.Artesunate or dihydroartemisinine tablet are oral, the 1-2 sheet, and a twice-daily~three time just can play excellent curative, and be suitable for long-term oral medication.The inventor adopts top same experimentation to test, find Artemether and SM905 also can the inflammation-inhibiting tissue in the expression of MPO activity, gamma interferon, IL-17, tumor necrosis factor-alpha, by suppressing the purpose that immunoreation reaches treatment mouse colitis disease, have the therapeutic effect same with artesunate. 1. the application of artemisinin derivative in the medicine of preparation treatment Crohn disease. 2. the application of artemisinin derivative as claimed in claim 1 in the medicine of preparation treatment Crohn disease, it is characterized in that: described artemisinin derivative is selected from artesunate or dihydroartemisinine or Artemether or SM905. https://patents.google.com/patent/CN102048728A/en?oq=CN102048728A青蒿素衍生物在制备治疗克罗恩病
  6. The invention relates to a medicament composition containing sorafenib, artemisinin and an artemisinin derivative and application thereof in preparing a medicament for treating lung cancer, pancreas cancer, colon cancer, liver cancer, prostatic cancer, kidney cancer, stomach cancer, brain tumor, sarcoma, ovarian cancer or breast cancer. The medicament composition has remarkable synergistic effect, improves the curative effect of the medicament, reduces the administration dosage and reduces side effects. 本发明涉及一种含有索拉非尼与青蒿素及青蒿素类衍生物的药物组合物及其在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物中的应用,本发明药物组合物具有显著的协同效应,提高了药物的疗效,降低了给药剂量,减少了副作用的发生。 含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物及其在制备治疗癌症的药物中的应用 技术领域 本发明涉及一种药物组合物及其在制备治疗癌症的药物中的应用,具体涉及含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物及其在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物中的应用。 背景技术 世界卫生组织调查报告表明,全球癌症状况日益严重,今后20年新患者的人数将由目前的每年1000万增加到1500万,因癌症而死亡的人数也将由每年的600万增加至1000万。其中肺癌为常见的恶性肿瘤之一,源于各级支气管上皮,分为细胞肺癌和非小细胞肺癌;胰腺癌多发生于胰头部,90%来源于胰管上皮细胞,其余来自胰腺腺泡,为消化系统常见的恶性肿瘤,发病率呈逐年上升趋势。由于起病隐匿,缺乏有效的早期诊断方法,确诊时往往已到晚期或发生转移,晚期患者中位生存期不超过六个月;结肠癌的发病与环境、生活习惯,尤其是饮食方式有关。一般认为高脂肪饮食和纤维素不足是主要发病原因。随着生活水平的提高,饮食结构的改变,结肠癌的发病率呈逐年上升趋势;原发性肝癌为发生在肝细胞与肝内胆管上皮细胞的癌变,是人类最常见的恶性肿瘤之一;前列腺癌是男性泌尿生殖系统肿瘤中最重要的一种,是人类特有的疾病。前列腺癌为老年病,大多在50岁以后发病。随着人类平均寿命的延长、诊断技术的提高,生活方式的改变,前列腺癌的发病率在不断上升,因此研究前列腺癌的治疗药物迫在眉睫。 目前已上市的抗肿瘤药物较多,如烷化剂药物、抗代谢药物、抗肿瘤抗生素、免疫调节剂等,但是大多药物由于毒性较大,病人不耐受。随着对肿瘤的发生发展的分子机制研究越来越清楚,分子靶向治疗多种恶性肿瘤受到了广泛的关注和高度重视。分子靶向药物选择性高、广谱有效,其安全性优于细胞毒性化疗药物,是目前肿瘤治疗领域发展的新方向。 青蒿素是含有过氧桥的新型倍半萜内酯,其衍生物有青蒿琥酯、蒿甲醚和双氢青蒿素等。青蒿素及其衍生物是一类高效、低毒的抗疟药物,随着研究的深入,人们发现青蒿素及青蒿素类化合物还具有其它很多重要的药理活性,如抗血吸虫、免疫调节、抗心律失常、抗肿瘤等,尤其是其抗肿瘤作用,越来越引起研究者们的重视。很多实验研究表明,青蒿素及青蒿素类化合物对多种肿瘤细胞的生长具有显著的抑制作用,而且对正常组织细胞的毒性很低,作用机制可能与肿瘤细胞内自由基的产生及氧化应激,延迟细胞周期,诱导细胞凋亡和抗肿瘤血管生成有关。双氢青蒿素是青蒿素类药物中活性较强的衍生物,青蒿琥酯是青蒿素最重要的衍生物之一,具有水溶性好,青蒿琥酯抗肿瘤机理与其对肿瘤细胞株有直接杀伤作用、或与诱导细胞凋亡有关,还可能与其抑制肿瘤组织血管生成等有关。青蒿素及青蒿素类药物可以选择性杀伤肿瘤细胞,并且与传统化疗药不存在交叉耐药,能够逆转肿瘤细胞的多药耐药现象。 索拉非尼为由拜耳和ONYX共同研制的一种多靶点激酶抑制剂,靶向作用于肿瘤细胞以及肿瘤血管内丝氨酸/苏氨酸激酶受体和酪氨酸激酶受体,通过抑制Raf/MEK/ERK信号传导途径抑制肿瘤生长,同时还通过抑制与新生血管生成有关的酪氨酸激酶受体的活性,阻断肿瘤新生血管的生成,间接抑制肿瘤细胞的生长。索拉非尼目前在临床使用的剂量为成人为800mg/天。然而索拉非尼有较多的不良反应,如,“索拉非尼的毒副作用及其处理”,《癌症进展杂志》2007年7月第5卷第4期第370-373页所记载的,不良反应包括手足综合症、疲乏、腹泻、皮肤毒性和胃肠道反应等,并经常导致药物服用的中断或减少药物服用剂量。因此寻找治疗效果更好、副作用低的抗癌药物为目前研究的热点。 随着肿瘤分子生物学的研究进展,肿瘤分子靶向治疗已成为肿瘤研究的热点,在多种肿瘤的治疗中发挥了重要的作用。然而,大部分肿瘤的生物学行为并非由单一信号传导通路所支配,而是多个信号传导通路共同起作用的,因此联合用药针对多靶点进行靶向治疗将不仅旨在减少或延缓耐药性的出现、降低毒性,而且通过多种药物对癌细胞杀伤的协同作用取得更好的疗效。 发明内容 针对以上技术缺陷,本发明提供一种药物组合物及其在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物中的应用,具体为含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物中的应用。 本发明含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物中,所述青蒿素及青蒿素类衍生物可以为青蒿素、青蒿琥酯、双氢青蒿素、蒿甲醚或蒿乙醚,或它们的相应衍生物。 本发明药物组合物中的青蒿素及青蒿素类衍生物优选为:青蒿琥酯、双氢青蒿素或蒿甲醚,其相应的结构式分别为式I、式II和式III。 本发明药物组合物中,所述组分不限于青蒿琥酯、双氢青蒿素和蒿甲醚药物本身,还可以是其可药用的盐、水合物或衍生物等。 本发明中,所述索拉非尼(英文名:Sorafenib)为4-{4-[3-(4-氯-3-三氟甲基-苯基)-酰脲]苯氧基}-吡啶-2-羧酸甲胺,其结构式为式IV。 本发明中,索拉非尼不限于该药物本身,还可以是其可药用的盐,索拉非尼的衍生物或如WO2000041698专利申请中所披露的各种索拉非尼的类似物;同时本发明应用中还可以将索拉非尼替换为多靶点激酶抑制剂的其它药物。 本发明含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物中,索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为1.0-15.0∶1.0-25.0;进一步优选所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.0-7.5∶2.5-12.5。 本发明含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物可以用于治疗各种肿瘤,所述肿瘤包括但不限于肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌。 本发明优选索拉非尼和青蒿素及青蒿素类衍生物的药物组合物用于制备治疗肺癌、胰腺癌、结肠癌、肝癌及前列腺癌的药物中的应用。 在本发明药物组合物用于制备治疗肺癌的药物中的应用中,所述索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为2.0-4.0∶3.0-10.0;优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为3.0-4.0∶5.0-10.0;更进一步优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为4.0∶10.0。 在本发明药物组合物用于制备治疗胰腺癌的药物中的应用中,所述索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为2.5-7.5∶2.5-10.0;优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为5.0-7.5∶5.0-10.0;更进一步优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为7.5∶10.0。 在本发明药物组合物用于制备治疗结肠癌的药物中的应用中,所述索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为2.5-6.0∶2.5-7.5;优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为4.0-6.0∶5.0-7.5;更进一步优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为6.0∶7.5。 在本发明药物组合物用于制备治疗肝癌的药物中的应用中,所述索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为2.0-4.0∶3.0-10.0;优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为3.0-4.0∶6.0-10.0;更进一步优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为4.0∶10.0。 在本发明药物组合物用于制备治疗前列腺癌的药物中的应用中,所述索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为2.5-5.0∶5.0-12.5;优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为3.5-5.0∶7.5-12.5;更进一步优选为索拉非尼和青蒿素及青蒿素类衍生物的摩尔比为5.0∶12.5。 含有索拉非尼和青蒿素及青蒿素类衍生物的药物组合物在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物的应用中,在将本发明组合物制成同时给药的药剂的方案中,青蒿素及青蒿素类衍生物和索拉非尼可以含在同一种药物制剂如片剂或胶囊中,也可以将青蒿素及青蒿素类衍生物和索拉非尼分别做成制剂,如分别做成片剂或胶囊,并采用本领域常规的方式将它们包装或结合在一起,患者然后按照药品说明书的指示同时服用;在将本发明组合物制成先后给药的药剂的方案中,可以将青蒿素及青蒿素类衍生物和索拉非尼分别做成不同的制剂,并采用本领域常规的方式将它们包装或结合在一起,患者然后按照药品说明书指示的先后顺序进行服用,或将上述组合物中的两种成分制成一种控释的制剂,先释放组合物中的一种成分、然后再释放组合物中的另一种成分,患者只需要服用该控释组合物制剂;在将本发明组合物制备成交叉给药的药剂的方案中,可以将青蒿素及青蒿素类衍生物和索拉非尼分别做成不同的制剂,并采用本领域常规的方式将它们包装或结合在一起,患者然后按照药品说明书指示的交叉顺序服用,或者将该药物组合物制备成青蒿素及青蒿素类衍生物和索拉非尼交叉释放的控释制剂。 索拉非尼和青蒿素及青蒿素类衍生物的药物组合物在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物中的应用中,所述组合物中的青蒿素及青蒿素类衍生物和索拉非尼可以同时使用或以任何先后的顺序使用,如可以将青蒿素及青蒿素类衍生物和索拉非尼同时给患者服用;也可以先将青蒿素及青蒿素类衍生物药物给患者服用、然后服用索拉非尼,或先服用索拉非尼、然后服用青蒿素及青蒿素类衍生物药物,对于两者服用的时间间隔没有特别要求,但优选服用两种药物的时间间隔不超过一天;或者两种药物交替给药。 本发明中,可将本发明青蒿素及青蒿素类衍生物和索拉非尼采用本领域常规的方法制备成适于胃肠道给药或非胃肠道给药的药物制剂,本发明优选将青蒿素及青蒿素类衍生物和索拉非尼制成胃肠道给药的药物制剂,其制剂形式可以为常规片剂或胶囊、或控释、缓释制剂。在本发明青蒿素及青蒿素类衍生物和索拉非尼组合物的药物制剂中,根据不同的制剂形式和制剂规格,所述组合物在制剂中的含量可以为质量计为1-99%,优选为10%-90%;制剂使用的辅料可采用本领域常规的辅料,以不和本发明组合物发生反应或不影响本发明药物的疗效为前提;所述制剂的制备方法可采用本领域常规的制备方法进行制备。 本发明中,组合物的制备方法没有什么限制,青蒿素及青蒿素类衍生物和索拉非尼两者可以进行直接混合然后做成制剂,或分别和/或相应的辅料混合分别做成制剂,然后再按照本领域常规的方式包装在一起,或分别和相应的辅料混合然后再混合做成制剂。 本发明中的药物组合物的给药剂量根据给药对象、给药途径或药物的制剂形式不同可以进行适当的变化,但以保证该药物组合物在哺乳动物体内能够达到有效的血药浓度为前提。 本发明分别进行了索拉非尼和青蒿素及青蒿素类衍生物组合杀死H460(肺癌细胞株)、Panc1(胰腺癌细胞株)、HCT-116(结肠癌细胞株)、HepG2(肝癌细胞株)和22RV1(前列腺癌细胞株)的试验,结果提示,本发明索拉非尼和青蒿素及青蒿素类衍生物组合治疗肺癌、胰腺癌、结肠癌、肝癌及前列腺癌具有显著的协同效应,提高了药物的疗效,降低了用药剂量,减少了副作用的发生。 具体实施方式 结合以下实施例对本发明作进一步的阐述,但本发明并不受限于此。 实施例 试剂和方法: 细胞:H460(肺癌细胞株)、Panc 1(胰腺癌细胞株)、HCT-116(结肠癌细胞株)、HepG2(肝癌细胞株)和22RV1(前列腺癌细胞株),均购自American Type Culture Collection(ATCC),Rockville,MD,USA。药品:以下实施例中所用药物组合物均按下列方法1或方法2所述来制备;青蒿素类衍生物青蒿琥酯、双氢青蒿素和蒿甲醚均购自Sigma;索拉非尼参考专利US2003207872合成而得。 方法1:准确称量相应的药物组合物的各组分,以二甲基亚砜分别溶解,各自配成10mM的贮存液,在-20℃下保存,使用时用新鲜的培养基稀释到合适的浓度,然后各自取1微升的各组分的溶液,混合在一起备用。所有的试验中,二甲基亚砜的最终浓度应≤5g/L,以便不影响细胞的活性。 将所有的细胞于含10%小牛血清、100kU/L青霉素、100mg/L链霉素的RPMI 1640培养基中,37℃、5%CO2的湿度条件下培养,在加药的前一天,在六孔板上进行细胞接种2×105/孔,然后向细胞中加入按上述方法制备的药物组合物溶液,使各组分达到其工作浓度,具体见表中第1-15。 药物处理后,通过台盼蓝(Trypan Blue)测定细胞死亡,细胞通过在37℃用胰蛋白酶钠/EDTA进行胰酶化作用10分钟。因为死亡的细胞从培养器上脱落进入培养基中,通过在1200转/分钟下离心收集所有的细胞,然后再用培养基重新悬浮沉淀物,与台盼蓝染料混合。染色之后,用光学显微镜和血细胞计数器进行计数。被染料染成蓝色的计为死亡细胞。随机选取500个细胞进行计数,死亡的细胞以占总计数细胞的百分比来表达。 方法2:准确称量相应的药物组合物的各组分,以二甲基亚砜分别溶解,各自配成10mM的贮存液,在-20℃下保存。使用时用新鲜的培养基稀释到合适的浓度,然后各自取1微升的各组分的溶液备用。所有的试验中,二甲基亚砜的最终浓度应≤5g/L,以便不影响细胞的活性。 将所有的细胞于含10%小牛血清、100kU/L青霉素、100mg/L链霉素的RPMI 1640培养基中,37℃、5%CO2的湿度条件下培养,在加药的前一天,在六孔板上进行细胞接种2×105/孔,然后以任意次序向细胞中加入按上述方法制备的药物组合物的各组分溶液,使各组分达到其工作浓度,具体见表中第16-27。 药物处理后,通过台盼蓝(Trypan Blue)测定细胞死亡,细胞通过在37℃用胰蛋白酶钠/EDTA进行胰酶化作用10分钟。因为死亡的细胞从培养器上脱落进入培养基中,通过在1200转/分钟下离心收集所有的细胞,然后再用培养基重新悬浮沉淀物,与台盼蓝染料混合。染色之后,用光学显微镜和血细胞计数器进行计数。被染料染成蓝色的计为死亡细胞。随机选取500个细胞进行计数,死亡的细胞以占总计数细胞的百分比来表达。 下列表1所示的药物组合中,第1-15的组合按方法1,第16-27的组合按方法2制备。 表1 实施例1不同比例的青蒿琥酯与索拉非尼的组合协同增效促进H460细胞死亡试验,见表2。 表2 在考察相关化合物导致肺癌细胞株H460细胞死亡的试验中,发现当单独使用10.0μM青蒿琥酯只有约20%细胞死亡、单独使用4.0μM索拉非尼只有约15%细胞死亡;而当两者在较低浓度下合用时(5.0μM青蒿琥酯+3.0μM索拉非尼)则产生明显的协同作用,导致约80%的癌细胞死亡;当两者以10.0μM青蒿琥酯+4.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致99%的癌细胞死亡。 实施例2不同比例的双氢青蒿素与索拉非尼的组合协同增效促进H460细胞死亡试验,见表3。 表3 在考察相关化合物导致肺癌细胞株H460细胞死亡的试验中,发现当单独使用10.0μM双氢青蒿素只有约20%细胞死亡、单独使用4.0μM索拉非尼只有约15%细胞死亡;而当两者在较低浓度下合用时(5.0μM双氢青蒿素+3.0μM索拉非尼)则产生明显的协同作用,导致约65%的癌细胞死亡;当两者以10.0μM双氢青蒿素+4.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致99%的癌细胞死亡。 实施例3不同比例的蒿甲醚与索拉非尼的组合协同增效促进H460细胞死亡试验,见表4。 表4 在考察相关化合物导致肺癌细胞株H460细胞死亡的试验中,发现当单独使用10.0μM蒿甲醚、4.0μM索拉非尼只有约15%细胞死亡;而当两者在较低浓度下合用时(5.0μM蒿甲醚+3.0μM索拉非尼)则产生明显的协同作用,导致约50%的癌细胞死亡;当两者以10.0μM蒿甲醚+4.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致约88%的癌细胞死亡。 实施例4不同比例的青蒿琥酯与索拉非尼的组合协同增效促进Panc1细胞死亡试验,见表5。 表5 在考察相关化合物导致胰腺癌细胞株Panc1细胞死亡的试验中,发现当单独使用10.0μM青蒿琥酯或更低浓度时只有很少量的细胞死亡,即使单独使用7.5μM索拉非尼也只有约20%细胞死亡;而当两者在较低浓度下合用时(5.0μM青蒿琥酯+5.0μM索拉非尼)则产生明显的协同作用,导致68%的癌细胞死亡;当两者以10.0μM青蒿琥酯+7.5μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致92%的癌细胞死亡。 实施例5不同比例的双氢青蒿素与索拉非尼的组合协同增效促进Panc1细胞死亡试验,见表6。 表6 在考察相关化合物导致胰腺癌细胞株Panc1细胞死亡的试验中,发现当单独使用10.0μM双氢青蒿素或更低浓度时只有很少量的细胞死亡,即使单独使用7.5μM索拉非尼也只有约20%细胞死亡;而当两者在较低浓度下合用时(5.0μM双氢青蒿素+5.0μM索拉非尼)则产生明显的协同作用,导致51%的癌细胞死亡;当两者以10.0μM双氢青蒿素+7.5μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致81%的癌细胞死亡。 实施例6不同比例的青蒿琥酯与索拉非尼的组合协同增效促进HCT116细胞死亡试验,见表7。 表7 在考察相关化合物导致结肠癌细胞株HCT116细胞死亡的试验中,发现当单独使用7.5μM青蒿琥酯或6.0μM索拉非尼时只有约20%细胞死亡;而当两者在较低浓度下合用时(5.0μM青蒿琥酯+4.0μM索拉非尼)则产生明显的协同作用,导致76%的癌细胞死亡;当两者以7.5μM青蒿琥酯+6.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致约95%的癌细胞死亡。 实施例7不同比例的双氢青蒿素与索拉非尼的组合协同增效促进HCT116细胞死亡试验,见表8。 表8 在考察相关化合物导致结肠癌细胞株HCT116细胞死亡的试验中,发现当单独使用7.5μM双氢青蒿素或6.0μM索拉非尼时只有约20%细胞死亡;而当两者在较低浓度下合用时(5.0μM双氢青蒿素+4.0μM索拉非尼)则产生明显的协同作用,导致67%的癌细胞死亡;当两者以7.5μM双氢青蒿素+6.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致约90%的癌细胞死亡。 实施例8不同比例的青蒿琥酯与索拉非尼的组合协同增效促进HepG2细胞死亡试验,见表9。 表9 在考察相关化合物导致肝癌细胞株HepG2细胞死亡的试验中,发现当单独使用10.0μM青蒿琥酯或4.0μM索拉非尼时只有约15%细胞死亡;而当两者在较低浓度下合用时(6.0μM青蒿琥酯+3.0μM索拉非尼)则产生明显的协同作用,导致42%的癌细胞死亡;当两者以10.0μM青蒿琥酯+4.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致约91%的癌细胞死亡。 实施例9不同比例的双氢青蒿素与索拉非尼的组合协同增效促进22RV1细胞死亡试验,见表10。 表10 在考察相关化合物导致前列腺癌细胞株22RV1细胞死亡的试验中,发现当单独使用7.5μM双氢青蒿素、3.5μM索拉非尼或更低浓度时只有约10%细胞死亡;即使增加单药的浓度至12.5μM双氢青蒿素、5.0μM索拉非尼时也只有约20%细胞死亡;而当两者在较低浓度下合用时(7.5μM双氢青蒿素+3.5μM索拉非尼)则产生明显的协同作用,导致44%的癌细胞死亡;当两者以12.5μM双氢青蒿素+5.0μM索拉非尼的比例合用时,则产生更加显著的协同作用,导致81%的癌细胞死亡。 1.一种药物组合物,其特征在于含有索拉非尼与青蒿素及青蒿素类衍生物。 2.根据权利要求1所述的药物组合物,其特征在于,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为1.0-15.0∶1.0-25.0。 3.根据权利要求2所述的药物组合物,其特征在于,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.0-7.5∶2.5-12.5。 4.根据权利要求3所述的药物组合物,其特征在于,所述青蒿素及青蒿素类衍生物为青蒿素、青蒿琥酯、双氢青蒿素、蒿甲醚或蒿乙醚。 5.权利要求1-4任一所述的药物组合物在制备治疗肺癌、胰腺癌、结肠癌、肝癌、前列腺癌、肾癌、胃癌、脑瘤、肉瘤、卵巢癌或乳腺癌的药物中的应用。 6.根据权利要求5所述的应用,其特征在于,在制备治疗肺癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.0-4.0∶3.0-10.0。 7.根据权利要求6所述的应用,其特征在于,在制备治疗肺癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为3.0-4.0∶5.0-10.0。 8.根据权利要求5所述的应用,其特征在于,在制备治疗胰腺癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.5-7.5∶2.5-10.0。 9.根据权利要求8所述的应用,其特征在于,在制备治疗胰腺癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为5.0-7.5∶5.0-10.0。 10.根据权利要求5所述的应用,其特征在于,在制备治疗结肠癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.5-6.0∶2.5-7.5。 11.根据权利要求10所述的应用,其特征在于,在制备治疗结肠癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为4.0-6.0∶5.0-7.5。 12.根据权利要求5所述的应用,其特征在于,在制备肝癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.0-4.0∶3.0-10.0。 13.根据权利要求12所述的应用,其特征在于,在制备肝癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为3.0-4.0∶6.0-10.0。 14.根据权利要求5所述的应用,其特征在于,在制备前列腺癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为2.5-5.0∶5.0-12.5。 15.根据权利要求14所述的应用,其特征在于,在制备前列腺癌的药物中的应用中,所述索拉非尼与青蒿素及青蒿素类衍生物的摩尔比为3.5-5.0∶7.5-12.5。 16.根据权利要求5所述的应用,其特征在于,所述药物组合物中的索拉非尼与青蒿素及青蒿素类衍生物可以同时使用或以任何先后的顺序使用。 Contain the pharmaceutical composition and the application in the medicine of preparation treatment cancer thereof of Sorafenib and arteannuin and artemisinin derivatives Technical field The present invention relates to a kind of pharmaceutical composition and the application in the medicine of preparation treatment cancer thereof, be specifically related to contain the pharmaceutical composition and the application in the medicine of preparation treatment pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, ovarian cancer or breast carcinoma thereof of Sorafenib and arteannuin and artemisinin derivatives. Background technology World Health Organization's investigation report shows that global cancer condition is serious day by day, and 20 years from now on new patients' number will be increased to 1,500 ten thousand by present every year 1000 ten thousand, because of the number that cancer is dead also will be by increasing to 1,000 ten thousand 6,000,000 of every year.Wherein pulmonary carcinoma is one of common malignancy, comes from bronchiolar epitheliums at different levels, is divided into cell lung cancer and nonsmall-cell lung cancer; The cancer of pancreas pilosity is born in head of pancreas portion, and 90% derives from the ductus pancreaticus epithelial cell, and all the other are the digestive system common malignancy from pancreatic acini, and sickness rate is ascendant trend year by year.Because onset concealment lacks effective method of early diagnosis, often reach an advanced stage when making a definite diagnosis or shift, the patients with terminal median survival interval is no more than six months; The morbidity of colon cancer and environment, living habit, especially diet style is relevant.It is generally acknowledged that high fat diet and cellulose deficiency are main pathogenic factors.Along with growth in the living standard, the change of dietary structure, the sickness rate of colon cancer is ascendant trend year by year; Primary hepatocarcinoma is one of human modal malignant tumor for occurring in the epithelial canceration of hepatocyte and stones in intrahepatic bile duct; Carcinoma of prostate is most important a kind of in the male genitourinary system tumor, is human distinctive disease.Carcinoma of prostate is a senile disease, mostly at 50 years old with sequela.Along with the prolongation of human average life, the raising of diagnostic techniques, the change of life style, the sickness rate of carcinoma of prostate is in continuous rising, and it is extremely urgent therefore to study the treatment of prostate cancer medicine. The antitumor drug that has gone on the market at present is more, and as alkylating agent medicine, antimetabolite, antitumor antibiotics, immunomodulator etc., but medicine is because toxicity is bigger mostly, and patient does not tolerate.The Study on Molecular Mechanism that develops along with the generation to tumor is more and more clearer, and the multiple malignant tumor of molecular targeted treatment has been subjected to paying close attention to widely and paying much attention to.Molecular targeted agents selectivity height, wide spectrum are effective, and its safety is better than the cytotoxicity chemotherapeutics, are the new directions of present oncotherapy field development. Arteannuin is the sesquiterpene lactone that contains peroxide bridge, and its derivant has artesunate, Artemether and dihydroarteannuin etc.Arteannuin and derivant thereof are that a class is efficient, the antimalarial agent of low toxicity, along with going deep into of research, it is found that arteannuin and artemisine compounds also have other a lot of important pharmacologically actives, as schistosomicide, immunomodulating, arrhythmia, antitumor etc., especially its antitumor action more and more causes the attention of researcheres.A lot of experimentatioies show, arteannuin and artemisine compounds have significant inhibitory effect to the growth of kinds of tumor cells, and it is very low to normal histiocytic toxicity, mechanism of action may with the generation and the oxidative stress of free radical in the tumor cell, postpone cell cycle, cell death inducing generates relevant with antineoplastic vascular.Dihydroarteannuin is an active stronger derivant in the artemisinin-based drug, artesunate is one of most important derivant of arteannuin, has good water solubility, artesunate antitumor mechanism has direct killing effect or relevant with cell death inducing with it to tumor cell line, also may be relevant with its inhibition tumor tissues angiogenesis etc.Arteannuin and artemisinin-based drug can the selective killing tumor cells, and do not have crossing drug resistant with traditional chemotherapeutic, multidrug resistance phenomenon that can the reversing tumor cell. Sorafenib is a kind of many target spots inhibitors of kinases by Bayer and the common development of ONYX, targeting is serine/threonine kinase receptor and tyrosine kinase receptor in tumor cell and tumor vessel, suppress tumor growth by suppressing the Raf/MEK/ERK signal transduction path, simultaneously also by suppressing to generate the activity of relevant tyrosine kinase receptor with new vessels, block tumor neovasculature generation, suppress the growth of tumor cell indirectly.Sorafenib is 800mg/ days at the clinical dosage that uses for the adult at present.Yet Sorafenib has more untoward reaction, as, " toxic and side effects of Sorafenib and processing thereof ", what " cancer progression magazine " July in 2007, the 5th volume the 4th phase 370-373 page or leaf was put down in writing, untoward reaction comprises brothers' syndrome, tired, diarrhoea, dermal toxicity and gastrointestinal reaction etc., and often causes the interruption of drug administration or reduce drug dose.Therefore seek the focus of cancer therapy drug that therapeutic effect is better, side effect is low for studying at present. Along with the progress of oncomolecularbiology, the molecular targeted treatment of tumor has become the focus of tumor research, has brought into play important effect in the treatment of kinds of tumors.Yet, the biological behaviour of most of tumor is arranged by single signal transduction pathway, but a plurality of signal transduction pathway concur, therefore drug combination carries out targeted therapy at many target spots and will not only be intended to reduce or delay chemical sproof appearance, reduce toxicity, and by multiple medicine the synergism that cancerous cell kills and wounds is obtained better therapeutic. Summary of the invention At above technological deficiency, the invention provides a kind of pharmaceutical composition and the application in the medicine of preparation treatment pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, ovarian cancer or breast carcinoma thereof, be specially the application of pharmaceutical composition in the medicine of preparation treatment pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, ovarian cancer or breast carcinoma that contains Sorafenib and arteannuin and artemisinin derivatives. The present invention contains in the pharmaceutical composition of Sorafenib and arteannuin and artemisinin derivatives, and described arteannuin and artemisinin derivatives can be arteannuin, artesunate, dihydroarteannuin, Artemether or arteether, or their corresponding derivative. Arteannuin and artemisinin derivatives in the pharmaceutical composition of the present invention are preferably: artesunate, dihydroarteannuin or Artemether, its corresponding structure formula is respectively formula I, formula II and formula III. In the pharmaceutical composition of the present invention, described component is not limited to artesunate, dihydroarteannuin and Artemether medicine itself, can also be its pharmaceutically useful salt, hydrate or derivant etc. Among the present invention, described Sorafenib (English name: Sorafenib) be 4-{4-[3-(4-chloro-3-trifluoromethyl-phenyl)-uride] phenoxy group }-the pyridine-2-carboxylic acids methylamine, its structural formula is formula IV. Among the present invention, Sorafenib is not limited to this medicine itself, can also be its pharmaceutically useful salt, the analog of the derivant of Sorafenib or the various Sorafenibs disclosed in the WO2000041698 patent application; During using, the present invention simultaneously Sorafenib can also be replaced with the other medicines of many target spots inhibitors of kinases. The present invention contains in the pharmaceutical composition of Sorafenib and arteannuin and artemisinin derivatives, and the mol ratio of Sorafenib and arteannuin and artemisinin derivatives is 1.0-15.0: 1.0-25.0; Further the mol ratio of preferred described Sorafenib and arteannuin and artemisinin derivatives is 2.0-7.5: 2.5-12.5. The pharmaceutical composition that the present invention contains Sorafenib and arteannuin and artemisinin derivatives can be used for the treatment of various tumors, and described tumor includes but not limited to pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, ovarian cancer or breast carcinoma. The pharmaceutical composition of the preferred Sorafenib of the present invention and arteannuin and artemisinin derivatives is used for preparing the application of the medicine for the treatment of pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma and carcinoma of prostate. Be used for preparing in the application for the treatment of lung cancer drugs at pharmaceutical composition of the present invention, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.0-4.0: 3.0-10.0; The mol ratio that is preferably Sorafenib and arteannuin and artemisinin derivatives is 3.0-4.0: 5.0-10.0; The mol ratio that further is preferably Sorafenib and arteannuin and artemisinin derivatives is 4.0: 10.0. Be used for preparing in the application of the medicine for the treatment of cancer of pancreas at pharmaceutical composition of the present invention, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.5-7.5: 2.5-10.0; The mol ratio that is preferably Sorafenib and arteannuin and artemisinin derivatives is 5.0-7.5: 5.0-10.0; The mol ratio that further is preferably Sorafenib and arteannuin and artemisinin derivatives is 7.5: 10.0. Be used for preparing in the application of the medicine for the treatment of colon cancer at pharmaceutical composition of the present invention, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.5-6.0: 2.5-7.5; The mol ratio that is preferably Sorafenib and arteannuin and artemisinin derivatives is 4.0-6.0: 5.0-7.5; The mol ratio that further is preferably Sorafenib and arteannuin and artemisinin derivatives is 6.0: 7.5. Be used for preparing in the application of the medicine for the treatment of hepatocarcinoma at pharmaceutical composition of the present invention, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.0-4.0: 3.0-10.0; The mol ratio that is preferably Sorafenib and arteannuin and artemisinin derivatives is 3.0-4.0: 6.0-10.0; The mol ratio that further is preferably Sorafenib and arteannuin and artemisinin derivatives is 4.0: 10.0. Be used for preparing in the application of the medicine for the treatment of carcinoma of prostate at pharmaceutical composition of the present invention, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.5-5.0: 5.0-12.5; The mol ratio that is preferably Sorafenib and arteannuin and artemisinin derivatives is 3.5-5.0: 7.5-12.5; The mol ratio that further is preferably Sorafenib and arteannuin and artemisinin derivatives is 5.0: 12.5. The pharmaceutical composition that contains Sorafenib and arteannuin and artemisinin derivatives is in preparation treatment pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, in the application of the medicine of ovarian cancer or breast carcinoma, in the scheme of the medicament of the present composition being made administration simultaneously, arteannuin and artemisinin derivatives and Sorafenib can be contained in in a kind of pharmaceutical preparation such as tablet or the capsule, also arteannuin and artemisinin derivatives and Sorafenib can be made preparation respectively, as making tablet or capsule respectively, and the mode that adopts this area routine is with their packings or combine, and the patient takes simultaneously according to the indication of package insert then; In the scheme of the medicament of the present composition being made administration successively, arteannuin and artemisinin derivatives can be made different preparations respectively with Sorafenib, and the mode that adopts this area routine is with their packings or combine, the patient takes according to the sequencing of package insert indication then, or two kinds of compositions in the above-mentioned composition are made a kind of preparation of controlled release, a kind of composition in elder generation's release composition and then the another kind of composition in the release composition, the patient only need take this controlled release composition preparation; In the scheme of the medicament that the present composition is prepared into the intersection administration, arteannuin and artemisinin derivatives can be made different preparations respectively with Sorafenib, and the mode that adopts this area routine is with their packings or combine, the patient takes according to the chi sequence of package insert indication then, the controlled release preparation that perhaps this preparation of pharmaceutical compositions is become arteannuin and artemisinin derivatives and Sorafenib intersection to discharge. In the application of the pharmaceutical composition of Sorafenib and arteannuin and artemisinin derivatives in the medicine of preparation treatment pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, ovarian cancer or breast carcinoma, arteannuin in the described compositions and artemisinin derivatives and Sorafenib can use or simultaneously with the using in order of any priority, as arteannuin and artemisinin derivatives and Sorafenib being taken to the patient simultaneously; Also can earlier Sorafenib be taken, be taken then to arteannuin and artemisinin derivatives medicine to the patient, or take Sorafenib earlier, take arteannuin and artemisinin derivatives medicine then, the interval of taking for both does not have special requirement, but the interval of preferably taking two kinds of medicines is no more than one day; Perhaps two kinds of medicines replace administration. Among the present invention, the method of arteannuin of the present invention and artemisinin derivatives and Sorafenib employing this area routine can be prepared into the pharmaceutical preparation that is suitable for gastrointestinal administration or parenteral administration, the pharmaceutical preparation that the present invention preferably makes gastrointestinal administration with arteannuin and artemisinin derivatives and Sorafenib, its dosage form can be conventional tablet or capsule or controlled release, slow releasing preparation.In the pharmaceutical preparation of arteannuin of the present invention and artemisinin derivatives and Sorafenib compositions, according to different dosage forms and preparation specification, the content of described compositions in preparation can be counted 1-99% for quality, is preferably 10%-90%; The adjuvant that preparation uses can adopt the adjuvant of this area routine, reacts or the curative effect that do not influence medicine of the present invention is a prerequisite with the discord present composition; The preparation method of described preparation can adopt the preparation method of this area routine to be prepared. Among the present invention, the preparation of compositions method does not have any restriction, arteannuin and artemisinin derivatives and Sorafenib can directly mix makes preparation then, or respectively and/or corresponding auxiliary material mix and to make preparation respectively, and then be packaging together, or mix and then mix and make preparation with corresponding auxiliary material respectively according to the mode of this area routine. The dosage of the pharmaceutical composition among the present invention can carry out suitable variation according to the dosage form difference of administration object, route of administration or medicine, but is prerequisite to guarantee that this pharmaceutical composition can reach effective blood drug level in mammalian body. The present invention has carried out the test that H460 (lung cancer cell line), Panc1 (pancreas cancer cell strain), HCT-116 (colon cancer cell line), HepG2 (hepatoma cell strain) and 22RV1 (prostate gland cancer cell strain) are killed in the combination of Sorafenib and arteannuin and artemisinin derivatives respectively, results suggest, Sorafenib of the present invention and arteannuin and artemisinin derivatives combined therapy pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma and carcinoma of prostate have significant cooperative effect, improved the curative effect of medicine, reduce dosage, reduced the generation of side effect. The specific embodiment The invention will be further elaborated with the following Examples, but the present invention is not limited to this. Embodiment Reagent and method: Cell: H460 (lung cancer cell line), Panc 1 (pancreas cancer cell strain), HCT-116 (colon cancer cell line), HepG2 (hepatoma cell strain) and 22RV1 (prostate gland cancer cell strain), all available from American Type Culture Collection (ATCC), Rockville, MD, USA.Medicine: institute's pharmaceutical composition is all by following method 1 or 2 described preparations of method in following examples; Artemisinin derivatives artesunate, dihydroarteannuin and Artemether are all available from Sigma; Sorafenib referenced patent US2003207872 is synthesized into. Method 1: each component of accurate weighing corresponding pharmaceutical compositions, dissolve respectively with dimethyl sulfoxide, be made into the stock solution of 10mM separately, preserve down at-20 ℃, be diluted to suitable concentration with fresh culture medium during use, the solution of each component of 1 microlitre of respectively asking for then mixes standby.In all tests, the ultimate density of dimethyl sulfoxide is answered≤5g/L, so that do not influence cell activity. With all cells in RPMI 1640 culture medium that contain 10% calf serum, 100kU/L penicillin, 100mg/L streptomycin, 37 ℃, 5%CO 2Damp condition cultivate down, in the previous day of dosing, on six orifice plates, carry out cell inoculation 2 * 10 5/ hole adds the pharmaceutical composition solution of preparation as stated above then in cell, make each component reach its working concentration, 1-15 in specifically seeing Table. After the drug treating, measure cell death by trypan blue (Trypan Blue), cell turns into 10 minutes by carrying out pancreatin at 37 ℃ with trypsin sodium/EDTA.Because dead cell comes off from incubator enter the culture medium,, and then, mix with the trypan blue dyestuff with culture medium suspended sediment again by all cells of centrifugal collection under 1200 rev/mins.After the dyeing, count with optical microscope and hematimeter.Dyed the blue dead cell of counting by dyestuff.500 cells of picked at random are counted, and dead cell is recently expressed with the percentage that accounts for the grand total cell. Method 2: each component of accurate weighing corresponding pharmaceutical compositions, dissolve respectively with dimethyl sulfoxide, be made into the stock solution of 10mM separately, preserve down at-20 ℃.Be diluted to suitable concentration with fresh culture medium during use, the solution for standby of each component of 1 microlitre of respectively asking for then.In all tests, the ultimate density of dimethyl sulfoxide is answered≤5g/L, so that do not influence cell activity. With all cells in RPMI 1640 culture medium that contain 10% calf serum, 100kU/L penicillin, 100mg/L streptomycin, 37 ℃, 5%CO 2Damp condition cultivate down, in the previous day of dosing, on six orifice plates, carry out cell inoculation 2 * 10 5/ hole adds each component solution of the pharmaceutical composition of preparation as stated above with any order then in cell, make each component reach its working concentration, 16-27 in specifically seeing Table. After the drug treating, measure cell death by trypan blue (Trypan Blue), cell turns into 10 minutes by carrying out pancreatin at 37 ℃ with trypsin sodium/EDTA.Because dead cell comes off from incubator enter the culture medium,, and then, mix with the trypan blue dyestuff with culture medium suspended sediment again by all cells of centrifugal collection under 1200 rev/mins.After the dyeing, count with optical microscope and hematimeter.Dyed the blue dead cell of counting by dyestuff.500 cells of picked at random are counted, and dead cell is recently expressed with the percentage that accounts for the grand total cell. Tabulate down in the drug regimen shown in 1, the combination of 1-15 prepares by method 2 by the combination of method 1, the 16-27. Table 1 The artesunate of embodiment 1 different proportion and the combination Synergistic of Sorafenib promote the test of H460 cell death, see Table 2. Table 2 Cause in the test of lung cancer cell line H460 cell death investigating related compound, find when use separately 10.0 μ M artesunate only have an appointment 20% cell death, use 4.0 μ M Sorafenibs, 15% cell death of only having an appointment separately; (5.0 μ M artesunate+3.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause about 80% cancer cell death; When both share with the ratio of 10.0 μ M artesunate+4.0 μ M Sorafenibs, then produce significant more synergism, cause 99% cancer cell death. The dihydroarteannuin of embodiment 2 different proportions and the combination Synergistic of Sorafenib promote the test of H460 cell death, see Table 3. Table 3 Cause in the test of lung cancer cell line H460 cell death investigating related compound, find when use separately 10.0 μ M dihydroarteannuins only have an appointment 20% cell death, use 4.0 μ M Sorafenibs, 15% cell death of only having an appointment separately; (5.0 μ M dihydroarteannuins+3.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause about 65% cancer cell death; When both share with the ratio of 10.0 μ M dihydroarteannuins+4.0 μ M Sorafenibs, then produce significant more synergism, cause 99% cancer cell death. The Artemether of embodiment 3 different proportions and the combination Synergistic of Sorafenib promote the test of H460 cell death, see Table 4. Table 4 Cause in the test of lung cancer cell line H460 cell death investigating related compound, find when using 10.0 μ M Artemether, 4.0 μ M Sorafenibs 15% cell death of only having an appointment separately; (5.0 μ M Artemether+3.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause about 50% cancer cell death; When both share with the ratio of 10.0 μ M Artemether+4.0 μ M Sorafenibs, then produce significant more synergism, cause about 88% cancer cell death. The artesunate of embodiment 4 different proportions and the combination Synergistic of Sorafenib promote the test of Panc1 cell death, see Table 5. Table 5 Cause in the test of pancreas cancer cell strain Panc1 cell death at the investigation related compound, find when use 10.0 μ M artesunate or lower concentration separately, to have only very a spot of cell death, even use 7.5 μ M Sorafenibs, 20% cell death of also only having an appointment separately; (5.0 μ M artesunate+5.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause 68% cancer cell death; When both share with the ratio of 10.0 μ M artesunate+7.5 μ M Sorafenibs, then produce significant more synergism, cause 92% cancer cell death. The dihydroarteannuin of embodiment 5 different proportions and the combination Synergistic of Sorafenib promote the test of Panc1 cell death, see Table 6. Table 6 Cause in the test of pancreas cancer cell strain Panc1 cell death at the investigation related compound, find when use 10.0 μ M dihydroarteannuins or lower concentration separately, to have only very a spot of cell death, even use 7.5 μ M Sorafenibs, 20% cell death of also only having an appointment separately; (5.0 μ M dihydroarteannuins+5.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause 51% cancer cell death; When both share with the ratio of 10.0 μ M dihydroarteannuins+7.5 μ M Sorafenibs, then produce significant more synergism, cause 81% cancer cell death. The artesunate of embodiment 6 different proportions and the combination Synergistic of Sorafenib promote the test of HCT116 cell death, see Table 7. Table 7 Cause in the test of colon cancer cell line HCT116 cell death at the investigation related compound, find 20% cell death of when using 7.5 μ M artesunate or 6.0 μ M Sorafenibs separately, only having an appointment; (5.0 μ M artesunate+4.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause 76% cancer cell death; When both share with the ratio of 7.5 μ M artesunate+6.0 μ M Sorafenibs, then produce significant more synergism, cause about 95% cancer cell death. The dihydroarteannuin of embodiment 7 different proportions and the combination Synergistic of Sorafenib promote the test of HCT116 cell death, see Table 8. Table 8 Cause in the test of colon cancer cell line HCT116 cell death at the investigation related compound, find 20% cell death of when using 7.5 μ M dihydroarteannuins or 6.0 μ M Sorafenibs separately, only having an appointment; (5.0 μ M dihydroarteannuins+4.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause 67% cancer cell death; When both share with the ratio of 7.5 μ M dihydroarteannuins+6.0 μ M Sorafenibs, then produce significant more synergism, cause about 90% cancer cell death. The artesunate of embodiment 8 different proportions and the combination Synergistic of Sorafenib promote the test of HepG2 cell death, see Table 9. Table 9 Cause in the test of hepatoma cell strain HepG2 cell death at the investigation related compound, find 15% cell death of when using 10.0 μ M artesunate or 4.0 μ M Sorafenibs separately, only having an appointment; (6.0 μ M artesunate+3.0 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause 42% cancer cell death; When both share with the ratio of 10.0 μ M artesunate+4.0 μ M Sorafenibs, then produce significant more synergism, cause about 91% cancer cell death. The dihydroarteannuin of embodiment 9 different proportions and the combination Synergistic of Sorafenib promote the test of 22RV1 cell death, see Table 10. Table 10 Cause in the test of prostate gland cancer cell strain 22RV1 cell death at the investigation related compound, find 10% cell death of when using 7.5 μ M dihydroarteannuins, 3.5 μ M Sorafenibs or lower concentration separately, only having an appointment; Even also only have an appointment 20% cell death when increasing concentration to 12.5 μ M dihydroarteannuin, the 5.0 μ M Sorafenibs of single medicine; (7.5 μ M dihydroarteannuins+3.5 μ M Sorafenibs) then produce the obvious synergistic effect when both share under low concentration, cause 44% cancer cell death; When both share with the ratio of 12.5 μ M dihydroarteannuins+5.0 μ M Sorafenibs, then produce significant more synergism, cause 81% cancer cell death. 1. a pharmaceutical composition is characterized in that containing Sorafenib and arteannuin and artemisinin derivatives. 2. pharmaceutical composition according to claim 1 is characterized in that, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 1.0-15.0: 1.0-25.0. 3. pharmaceutical composition according to claim 2 is characterized in that, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.0-7.5: 2.5-12.5. 4. pharmaceutical composition according to claim 3 is characterized in that, described arteannuin and artemisinin derivatives are arteannuin, artesunate, dihydroarteannuin, Artemether or arteether. 5. the application of the arbitrary described pharmaceutical composition of claim 1-4 in the medicine of preparation treatment pulmonary carcinoma, cancer of pancreas, colon cancer, hepatocarcinoma, carcinoma of prostate, renal carcinoma, gastric cancer, cerebroma, sarcoma, ovarian cancer or breast carcinoma. 6. application according to claim 5 is characterized in that, in the application in preparation treatment lung cancer drugs, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.0-4.0: 3.0-10.0. 7. application according to claim 6 is characterized in that, in the application in preparation treatment lung cancer drugs, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 3.0-4.0: 5.0-10.0. 8. application according to claim 5 is characterized in that, in the application in the medicine of preparation treatment cancer of pancreas, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.5-7.5: 2.5-10.0. 9. application according to claim 8 is characterized in that, in the application in the medicine of preparation treatment cancer of pancreas, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 5.0-7.5: 5.0-10.0. 10. application according to claim 5 is characterized in that, in the application in the medicine of preparation treatment colon cancer, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.5-6.0: 2.5-7.5. 11. application according to claim 10 is characterized in that, in the application in the medicine of preparation treatment colon cancer, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 4.0-6.0: 5.0-7.5. 12. application according to claim 5 is characterized in that, in the application in the medicine of preparation hepatocarcinoma, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.0-4.0: 3.0-10.0. 13. application according to claim 12 is characterized in that, in the application in the medicine of preparation hepatocarcinoma, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 3.0-4.0: 6.0-10.0. 14. application according to claim 5 is characterized in that, in the application in the medicine of preparation carcinoma of prostate, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 2.5-5.0: 5.0-12.5. 15. application according to claim 14 is characterized in that, in the application in the medicine of preparation carcinoma of prostate, the mol ratio of described Sorafenib and arteannuin and artemisinin derivatives is 3.5-5.0: 7.5-12.5. 16. application according to claim 5 is characterized in that, Sorafenib in the described pharmaceutical composition and arteannuin and artemisinin derivatives can use or using in order with any priority simultaneously. https://patents.google.com/patent/CN101940569A/en?oq=CN101940569A含有索拉非尼和青蒿素及青蒿素衍生物
  7. The invention discloses an application of arteannuin derivant with the structure disclosed in I,II,III, IV and V for preparing medicines for preventing and treating adiposity, wherein n=0-1; R1 is selected from C1-C3 alkyl; and the R2 is selected from aryl or aromatic heterocyclic ring substituted by nitrogenous atom. The medicine prepared from the arteannuin organisms is taken as the medicine for resisting adipose differentiation and can play a very important role in treating the diseases. 本发明公开了具有以下式Ⅰ、Ⅱ、Ⅲ、Ⅳ、Ⅴ结构的青蒿素衍生物在制备防治肥胖的药物中的应用,其中:n=0~1;R1选自C1~C3烷基;R2选自芳基或含氮原子取代的芳香杂环。由所述青蒿素类生物制备的药物作为抗脂肪分化的药物,将会在这些疾病的治疗中发挥重要作用。 青蒿素衍生物的新应用 技术领域 本发明涉及青蒿素衍生物在制药中的新应用,属于化学生物学和细胞生物学领域。 背景技术 青蒿素是从黄花蒿中提取的含有过氧基团的倍半萜内酯药物。青蒿素类抗疟药物的发现是全球抗疟药物发展史上继奎宁之后的又一里程碑,它是我国发现的第一个植物化学药品,也是中国唯一被世界卫生组织认可的、可按合成药研究标准开发的中药。30多年来,青蒿素独特的分子结构和突出的生物活性深深吸引着科学家们的眼球。随着医药科技的发展,经过药理作用和临床应用证明,青蒿素衍生物被广泛应用于治疗疟疾,癌症,急性感染及高热病和皮肤病等疾病中。其中临床研究较多的青蒿素衍生物有二氢青蒿素、蒿甲醚、蒿乙醚和青蒿琥酯等。目前尚未见有关青蒿素衍生物抑制脂肪细胞分化的文献报道。 肥胖是指机体进食热量多于身体消耗量,造成脂肪的过渡积累与脂肪组织的过量扩增,使体重超过标准体重的20%以上,并伴有头昏乏力、神疲气短的一类病症,是高血压病、糖尿病、高血脂、冠心病、脑血管病的危险因素。当今,肥胖已成为全球蔓延速度最快、最严重的公共卫生问题之一。近年来,随着我国人民生活水平的提高,在城市人口中肥胖人数增加了,而且不仅发生在老年人,还发生在儿童、妇女,各个年龄组身体肥胖的人数都在增加,减肥已经成为人们的热点话题。引起肥胖症的病因有很多,其中主要与遗传和环境因素有密切关系。环境因素是指长期食入高脂、高热量食物,再加上体力活动减少,心理障碍等因素的作用下,引起体脂调控网络的神经内分泌调节紊乱。 因此,预防和控制肥胖病的发生已成为医学上十分关注的问题。目前,常用的减肥药物大致分为三类:一、控制食欲的药物,此类药物有诸多不良反应,如失眠、口干、便秘、心悸和高血压;二、增加能量消耗的药物;三、抑制肠道吸收的药物。尽管现在有许多研究已深入到基因药物水平,但目前仍然没有找到一种真正有效的、能长期使用的减肥药物,研究出更为理想的有效的方法和制剂用于肥胖病的诊断和治疗仍然具有深远的意义。 3T3-L1是一个衍生自3T3细胞(一个建立于1962年的标准成纤维细胞的细胞系)的,在生物研究中被应用于研究脂肪组织的细胞系。3T3-L1细胞拥有类似成纤维细胞的形态,但是在适当条件的诱导下,能够分化并形成脂肪细胞的表型。3T3-L1细胞分化成熟后的脂肪细胞形态,胞内甘油三酯的合成和积累相对于分化前的成纤维细胞状态都大大提高,这时候的3T3-L1细胞呈现一种图章戒指的脂肪细胞形态。这些细胞对调节脂肪生成和脂肪降解的激素和药物都敏感,例如肾上腺素,去甲肾上腺素,胰岛素等。 因为3T3-L1细胞的这些特性,该细胞系被广泛应用于脂肪分化和代谢疾病机理的研究,是国际公认的体外脂肪分化的细胞系模型。对该模型的研究已经帮助阐明了一个在脂肪细胞末端分化中最为主要的调控脂肪分化的转录级联信号转导,包括了一系列转录因子、调控蛋白和效应蛋白。 油红O染色技术是一种常用的脂类染色法,其优点是染色步骤简便,染色结果肯定。染色后脂肪呈鲜红色,细胞核呈蓝色,间质无色。油红O染出的红色与脂肪细胞内积累形成的油滴的量成正比,而后者与脂肪细胞分化程度的高低成正比,所以通过该技术,既可以定性观察判断,亦可以定量检测比较脂肪细胞分化程度的高低。 黄花蒿广泛存在于世界各地,但青蒿素含量较高的黄花蒿仅存在于我国重庆东部、福建、广西、海南部分地区,属于我国独有的药物资源。近几十年,青蒿素衍生物的研究主要集中在抗疟疾和抗肿瘤方面,将其应用到减肥药的研究却很罕见。 发明内容 本发明的目的是为了完善青蒿素衍生物在新领域里的应用,即提供一种蒿素衍生物的新应用。 具体技术方案如下: 具有以下式Ⅰ、Ⅱ、Ⅲ、Ⅳ、Ⅴ结构的青蒿素衍生物在制备防治肥胖的药物中的应用, 其中: n=0~1;R1选自C1~C3烷基;R2选自芳基或含氮原子取代的芳香杂环。 优选地,所述青蒿素衍生物为具有以下结构式中的任一种 其中,二氢青蒿素、青蒿素苯甲酸酯具有所有检测过的青蒿素衍生物中最强的抑制3T3-L1细胞分化的效果。其次是青蒿琥酯,拥有仅次于前两者的抑制3T3-L1细胞分化的效果。这3个化合物都能够在10μM的水平完全抑制3T3-L1细胞的分化。 本发明的目的是开发青蒿素衍生物在减肥药物中的作用,对其抑制细胞脂肪分化进行了详细研究。 本发明具有以下优点: (1)青蒿素及其衍生物是传统的抗疟疾药物,一直以来用于治疗疟疾,本发明的特点是首次提出青蒿素的部分衍生物具有抑制脂肪细胞分化的作用,首次将它们与代谢类的疾病联系起来,并首次提出它们作为应用于肥胖及肥胖相关疾病如心血管疾病的药物的潜在可能性。与肥胖相关的代谢类疾病在当今社会是人类健康的重大威胁,由所述青蒿素类生物制备的药物作为抗脂肪分化的药物,将会在这些疾病的治疗中发挥重要作用。 (2)本发明比较了不同青蒿素衍生物之间抑制脂肪细胞分化能力的强弱,得出了初步的构效关系,为进一步优化获得活性更好的青蒿素衍生物提供了依据。 附图说明 图1.为青蒿素系列衍生物对脂肪细胞分化的抑制作用,横坐标为药物使用剂量(浓度,以μM表示),纵坐标为相对分化率(加药处理实验组油红染色OD值/完全分化对照组油红染色OD值); 图2.为选取的抑制3T3-L1细胞分化效果最具有代表性的5个化合物的油红染色总体效果;其中,图2.1为化合物2浓度梯度影响3T3-L1细胞分化的油红染色总体效果; 图2.2为化合物4浓度梯度影响3T3-L1细胞分化的油红染色总体效果; 图2.3为化合物3浓度梯度影响3T3-L1细胞分化的油红染色总体效果; 图2.4为化合物5浓度梯度影响3T3-L1细胞分化的油红染色总体效果; 图2.5为化合物9浓度梯度影响3T3-L1细胞分化的油红染色总体效果; 图3为诱导3T3-L1细胞分化的流程阶段示意图。 具体实施方式 本发明所述具有以下式Ⅰ、Ⅱ、Ⅲ、Ⅳ、Ⅴ结构的青蒿素衍生物,主要为具有以下结构式的化合物: (a)所述具有式Ⅰ结构的化合物为: (b)所述具有式Ⅱ结构的化合物为: (c)所述具有式Ⅲ结构的化合物为: (d)所述具有式Ⅳ结构的化合物为: (e)所述具有式Ⅴ结构的化合物为: 为使本发明更加容易理解,下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明目的,而不用于限制本发明范围。 本领域的技术人员,可以从以下描述或现有技术,得到所述具有所述式Ⅰ、Ⅱ、Ⅲ、Ⅳ、Ⅴ结构的青蒿素衍生物。 实施例1:脱氧青蒿素的合成 200mg(0.7087mmol)青蒿素溶于10mL THF,原料溶解后,加入547mg(20.2688mmol)Al粉和7.23g(30.4032mmol)NiCl2·6H2O,4小时后反应完毕,旋干溶剂,用乙酸乙酯溶解旋干物,抽滤,用乙酸乙酯淋洗滤渣。滤液经无水Na2SO4干燥,浓缩后柱层析(PE∶EA=16∶1)得到72mg目标产物,收率38%。 1H NMR(CDCl3,400MHz):δ=5.68(s,1H),3.19-3.16(m,1H),2.02-1.58(m,6H),1.51(s,3H),1.28-1.03(m,11H). 实施例2:脱氧二氢青蒿素的合成 500mg(1.7596mmol)二氢青蒿素溶于5mL DCM,加入45mg 10%Pd/C,氢气保护下,40℃过夜反应完毕,过滤除去Pd/C,旋干溶剂,浓缩后柱层析(PE∶EA=8∶1)得到202mg目标产物,收率43%。 1H NMR(CDCl3,400MHz):δ=5.34(s,1H),4.78(t,J=6.8Hz,1H),2.83(d,J=7.2Hz,1H),2.40-0.79(m,21H). 实施例3:二氢青蒿素的合成 10g(35.4321mmol)青蒿素溶于500mL甲醇,搅拌降温至0-5℃,1小时内分批加入10g(265.7408mmol)NaBH4。保持该温度继续搅拌1.5小时后,往反应瓶中缓慢滴加15.5mLHAc调节pH=7,逐渐析出大量白色固体。旋干大部分溶剂,加入10mL冷水,室温下搅拌15分钟,抽滤,用冷H2O∶MeOH=2∶1洗涤滤饼,收集滤饼,干燥后得到8.0g目标产物,收率80%。 1H NMR(CDCl3,400MHz):δ=5.60(s,1H),5.28(s,1H),2.85(s,1H),2.61(s,1H),2.41-0.89(m,20H). 实施例4:β-蒿甲醚的合成 150mg(0.5275mmol)二氢青蒿素溶于1.0mL MeOH和2mL DCM混合溶剂中,加热至45℃,氩气保护下,迅速注入0.01mL(0.0559mmol)BF3·Et2O,在此温度下反应1小时,反应结束后,用醋酸钠水溶液洗涤反应液,有机层经无水MgSO4干燥后,浓缩即得到β-蒿甲醚和α-蒿甲醚的混合物,将混合物溶解在适量正己烷中,在-20℃保存36小时,收集白色晶体,并用冷正己烷淋洗,干燥后得到107mg纯β-蒿甲醚,收率65%。 1H NMR(CDCl3,400MHz):δ=5.38(s,1H),4.68(d,J=3.2Hz,1H),3.42(s,3H),2.64-0.85(m,21H). 实施例5:青蒿素苯甲酸酯的合成 氩气保护下,3.386g(11.9158mmol)二氢青蒿素溶于40mL DCM,加入6.1mL(76.2613mmol)吡啶,搅拌降温至0℃,加入2.2mL(19.0653mmol)苯甲酰氯,保持该温度下反应15分钟,撤去冰浴升至室温,继续反应16小时完毕,加入7%柠檬酸水溶液终止反应,旋干DCM,加入乙酸乙酯溶解旋干物,依次用7%柠檬酸水溶液、饱和NaHCO3溶液、饱和NaCl溶液洗涤有机层,有机层经无水Na2SO4干燥,浓缩后柱层析(PE∶EA=16∶1)得到4.07g目标产物,收率88%。 1H NMR(CDCl3,400MHz):δ=8.12(d,J=7.2Hz,2H),7.59-7.55(m,1H),7.44(t,J=8.0Hz,2H),6.01(d,J=9.6Hz,1H),5.53(s,1H),2.76-0.92(m,21H). 实施例6:β-烯丙基青蒿素的合成 反应瓶中加入少量4 MS、105mg(0.7790mmol)无水ZnCl2,Ar保护下,注入3mL无水DCM和0.5mL(3.1160mmol)烯丙基三甲基硅烷,搅拌降温至0℃。 称252mg(0.6492mmol)青蒿素苯甲酸酯溶解在3mL无水二氯乙烷中,将溶液滴入上述反应液中,保持0℃反应1小时,撤去冰浴,升至室温反应3小时,加入适量乙酸乙酯稀释反应体系,依次用7%柠檬酸水溶液、饱和NaHCO3溶液、饱和NaCl溶液洗涤有机层,有机层经无水Na2SO4干燥,浓缩后柱层析(PE∶EA=30∶1),得到114mg目标产物,收率66%。 1H NMR(CDCl3,400MHz):δ=5.92(m,1H),5.32(s,1H),5.14-5.04(m,2H),4.30(m,1H),2.69-0.87(m,23H). 实施例7:β-乙酸青蒿素的合成 100mg(0.3245mmol)β-烯丙基青蒿素溶于4mL CCl4-CH3CN-H2O(V∶V∶V=1∶1∶2)混合溶剂中,依次加入347mg(1.6225mmol)NaIO4和2mg(0.0097mmol)RuCl3,室温过夜反应。旋干溶剂,加入乙酸乙酯溶解旋干物,依次用饱和NaHSO3、饱和NaHCO3溶液、饱和NaCl溶液洗涤有机层,有机层经无水Na2SO4干燥,浓缩后柱层析(DCM∶MeOH=30∶1),得到92mg目标产物,收率87%。 1H NMR(CDCl3,400MHz):δ=5.37(s,1H),4.86(m,1H),2.71-0.88(m,24H). 实施例8:D-Biotin修饰青蒿素的合成 Reagents and conditions:(a)Boc2O,Et3N,THF,R.T.,15h;(b)D-biotin,EDCI,HOBt,(i-Pr)2NEt,DMF,R.T.,10h;(c)TFA/CH2Cl2,0℃~R.T.,9h;(d)EDCI,HOBt,(i-Pr)2NEt,DMF,R.T.,11h. 2的合成: 2.035g(13.7360mmol)溶于18mL THF中,加人0.63mL(4.5767mmol)NEt3,搅拌降温至0℃。 1g(4.5767mmol)Boc2O溶于9mL THF中,混合均匀后,逐滴滴入上述反应液中,滴加完毕后,自然升至室温反应15小时。反应完毕后,旋干溶剂,加水稀释后,用DCM萃取,有机层用饱和NaCl溶液洗涤,无水Na2SO4干燥,浓缩后柱层析(DCM∶MeOH=5∶1),得到906mg目标产物,收率80%。 1H NMR(CDCl3,400MHz):δ=5.18(s,1H),3.56(s,4H),3.50-3.45(m,4H),3.30-3.25(m,2H),2.82(t,J=5.6Hz,2H),1.53(s,2H),1.38(s,9H). 3的合成: 氩气保护下,318mg(1.3mmol)D-Biotin、249mg (1.3mmol,1.3equiv.)EDCI、176mg(1.3mmol,1.3equiv.)HOBt溶于4mL干燥DMF,加入0.23mL(1.4mmol,1.4equiv.)DIPEA,室温搅拌。 248mg(1.0mmol)2溶于3mL干燥DMF中,将溶液注入上述反应瓶中,室温反应10小时。反应完毕后,往反应瓶中加入NH4Cl饱和溶液,氯仿萃取,有机层经NH4Cl饱和溶液、水、饱和NaCl溶液洗涤,无水Na2SO4干燥,浓缩后柱层析(DCM∶MeOH=20∶1),得到358mg目标产物,收率76%。 1H NMR(CDCl3,400MHz):δ=6.67(s,1H),6.15(s,1H),5.52(s,1H),5.15(s,1H),4.45-4.40(m,1H),4.25-4.20(m,1H),3.52(s,4H),3.49-3.45(m,4H),3.37-3.34(m,2H),3.23-3.21(m,2H),3.10-3.04(m,1H),2.14(t,J=7.6Hz,2H),1.70-1.50(4H),1.36(s,12H). 4的合成: 将118mg 3溶于3mL二氯甲烷中,搅拌降温至0℃,缓慢滴加1.0mL TFA,加完后撤去冰浴,自然升至室温反应9小时,反应完毕后,直接减压蒸馏除去溶剂,真空干燥得到粗产物(按100%计算,应得到93mg粗产物)4,未经分离直接用于下一步反应。 1H NMR(MeOD,400MHz):δ=4.52-4.49(m,1H),4.32-4.29(m,1H),3.72-3.65(m,6H),3.56(t,J=5.6Hz,2H),3.39-3.35(m,2H),3.22-3.20(m,1H),3.14-3.11(m,2H),2.95-2.91(dd,J=12.8Hz,4.8Hz,1H),2.71(d,J=12.8Hz,1H),2.22(t,J=7.6Hz,2H),1.71-1.60(m,4H),1.47-1.43(m,2H). D-Biotin修饰青蒿素的合成: 氩气保护下,74mg(0.2269mmol)羧酸青蒿素、57mg(0.2949mmol)EDCI、40mg(0.2949mmol)HOBt溶于3mL无水DMF,注入0.24mL(1.4293mmol)DIPEA,室温搅拌。 称93mg(0.2496mmol)4溶于2mL干燥DMF,将溶液注入上述反应液中,室温过夜反应。反应完毕后,加入DCM稀释反应液,依次用NH4Cl饱和溶液、水、饱和NaCl溶液洗涤,无水Na2SO4干燥,浓缩后柱层析(DCM∶MeOH=10∶1),得到89mg目标产物,收率57%。 1H NMR(CDCl3,400MHz):δ=7.24(s,1H),δ6.85(s,1H),δ6.30(s,1H),δ5.41(s,1H),δ5.35(s,1H),δ4.75-4.71(m,1H),δ4.52-4.50(m,1H),δ4.32-4.29(m,1H),δ3.61-3.40(m,12H),δ3.15-3.16(m,1H),δ2.90-0.88(m,33H). 实施例9: 称50mg(0.1288mmol)青蒿素苯甲酸酯和适量4 MS于反应瓶中,氩气保护下,注入2mL重蒸DCM溶解,降温至0℃,注入0.02mL(0.1546mmol)TMSCl,保持该温度反应1小时后升至室温过夜反应完毕。旋干溶剂,加入乙酸乙酯溶解旋干物,依次用饱和NaHCO3溶液、饱和NaCl溶液洗涤,有机层经无水Na2SO4干燥,浓缩后柱层析(PE∶EA=8∶1),得到33mg目标产物,收率96%。 1H NMR(CDCl3,400MHz):δ=6.19(s,1H),5.54(s,1H),2.44-0.99(m,20H). 实施例10: 氩气保护下,50mg(0.1865mmol)脱氧二氢青蒿素溶于3mL DCM中,降温至0℃,注入0.03mL(0.3729mmol)乙酰氯,逐渐升至室温过夜反应完毕。反应液经水洗涤,有机层经无水Na2SO4干燥,浓缩柱层析(PE∶EA=16∶1),得到作为副产物的消旋脱氧二氢青蒿素30mg,收率64%。 1H NMR(CDCl3,400MHz):δ=6.04(s,1H),5.47(s,1H),2.04-1.21(m,20H). 实施例11: 100mg(0.3517mmol)二氢青蒿素溶于5mL DCE,加人73mg(0.4220mmol)2-喹喔啉羧酸搅拌片刻后,加入5mL(0.0422mmol)DMAP,搅拌降温至0℃,加入80mg(0.3868mmol)DCC,搅拌5分钟后升至室温,16小时后停止反应,过滤反应液,滤液经水、饱和NaCl溶液洗涤,有机层经无水Na2SO4干燥后,浓缩柱层析,得到120mg目标产物,收率78%。 1H NMR(CDCl3,400MHz):δ=9.57(s,1H),8.31(d,J=8.0Hz,1H),8.17(d,J=8.0Hz,1H),7.91-7.86(m,2H),6.03-6.00(m,2H),5.64(s,1H),2.90-0.95(m,21H). 实施例12: 按照如实例12所述,100mg(0.3517mmol)二氢青蒿素与63mg(0.4220mmol)对醛基苯甲酸反应,柱层析得到116mg目标产物,收率79%。 1H NMR(CDCl3,400MHz):δ=10.10(s,1H),8.27(d,J=8.0Hz,2H),7.96(d,J=8.0Hz,2H),6.03-6.00(m,1H),5.54(s,1H),2.90-0.95(m,21H). 实施例13: 按照如实例12所述,100mg(0.3517mmol)二氢青蒿素与133mg(0.4220mmol)Boc-3-(2-萘基)-L-丙氨酸反应,柱层析得到151mg目标产物,收率76%。 1H NMR(CDCl3,400MHz):δ=7.86-7.83(m,1H),7.80-7.74(m,3H),7.47-7.38(m,1H),6.05-6.00(m,1H),5.54(s,1H),2.83-0.85(m,34H). 实施例14: 按照如实例12所述,100mg(0.3517mmol)二氢青蒿素与76mg(0.4220mmol)对硝基苯乙酸反应,柱层析得到123mg目标产物,收率78%。 1H NMR(CDCl3,400MHz):δ=8.18(d,J=8.0Hz,2H),7.48(d,J=12.0Hz,2H),6.03-6.00(m,1H),5.44(s,1H),3.82(s,2H),3.82(s,2H),2.65-0.68(m,23H). 实施例15: 氩气保护下,100mg(0.3517mmol)二氢青蒿素与0.08mL(0.5627mmol)对甲氧基苯甲酰氯溶于5mL DCM中,降温至0℃,注入0.2mL(2.2509mmol)吡啶,搅拌15分钟后,升至室温过夜反应完毕,用7%柠檬酸水溶液淬灭反应,乙酸乙酯萃取,再经饱和NaHCO3、饱和NaCl溶液洗涤,有机层经无水Na2SO4干燥,浓缩后柱层析,得到119mg目标产物,收率81%。 1H NMR(CDCl3,400MHz):δ=8.06(d,J=12Hz,2H),6.91(d,J=12Hz,2H),5.98(m,1H),5.51(s,1H),3.864(s,3H),2.81-0.85(m,21H). 实施例16: 按照实例15所述,100mg(0.3517mmol)二氢青蒿素与104mg(0.5627mmol)对硝基苯甲酰氯反应,柱层析得到81mg目标产物,收率53%。 1H NMR(CDCl3,400MHz):δ=8.28(s,4H),6.05-6.00(m,1H),5.55(s,1H),2.83-0.85(m,22H). 实施例17:青蒿琥酯的合成 室温条件下,250mg(0.9mmol)二氢青蒿素溶于25mL干燥DCM中,加入202mg(2mmol)丁二酸酐,降温至0~5℃,加入107mg(0.9mmol)DMAP,保持该温度反应0.5小时,自然升至室温反应1小时。反应结束后,加入5mL水,用10%稀盐酸调至pH=3,有机层经水洗后,用无水MgSO4干燥,减压除去溶剂,得到317mg目标产物,收率94%。 1H NMR(CDCl3,400MHz):δ=5.74(d,J=8.0Hz,1H),5.39(s,1H),2.72-2.59(m,4H),2.53-2.48(m,1H),2.36-2.28.(m,1H),2.01-1.96(m,1H),1.87-1.82(m,1H),1.76-1.66(m,2H),1.60-1.55(m,1H),1.45-1.21(m,8H),1.01-0.95(m,1H),0.92(d,J=8.0Hz,3H),0.80(d,J=4.0Hz,3H). 实施例18:青蒿素衍生物对3T3-L1细胞分化的影响 3T3-L1细胞的培养 3T3-L1细胞在37℃,5%CO2培养环境中贴壁生长,使用加入10%FBS(Hyclone胎牛血清)的DMEM高糖培养基(HyClone)。可根据细胞生长状况,2~3天换一次液。如果需要持续多次传代,并需要在多次传代之后细胞仍然具有良好的分化能力,那么就要避免让细胞长的太满(<70%),一般长到60%~70%就要传代分细胞了。细胞可以以稀到1∶15的比例传代分盘,但是通常以1∶10或者更高的比例传代分盘,以实验需要为准。 3T3-L1细胞诱导分化的培养基的配制 诱导培养基分为两种,MDI诱导培养基和胰岛素培养基,分别用于分化不同阶段,是由DMEM培养基加上适当量的FBS、地塞米松、胰岛素和IBMX配成。 IBMX溶液:将IBMX溶于0.5N的KOH溶液中,IBMX终浓度为0.0115g/ml,0.22μm针头过滤器过滤除菌,-20℃保存。 胰岛素储存溶液:将胰岛素溶于0.02M的HCl中至终浓度167uM(1mg/ml),0.22μm针头过滤器过滤除菌,-20℃长期保存,4℃短期保存。 地塞米松储存溶液:冷冻储存液:10mM地塞米松溶于无水乙醇,-20℃保存;工作储存液:将冷冻储存液用PBS稀释至1mM,过滤除菌,储存于4℃。 MDI诱导培养基(现配现用,10ml/10cm培养皿,5ml/6cm培养皿):含有10%FBS的DMEM高糖培养基再加入:1∶100的IBMX溶液,1∶1000胰岛素储存液,1∶1000地塞米松工作储存液。 胰岛素培养基(现配现用,10ml/10cm培养皿,5ml/6cm培养皿):含有10%FBS的DMEM高糖培养基再加入:1∶1000胰岛素储存液。 诱导未分化的3T3-L1细胞(成纤维细胞状态)向成熟脂肪细胞分化: 如图3所示: 1.在含有10%FBS的DMEM高糖培养基中将3T3-L1前脂肪细胞培养至接合状态。 2.接合之后再养2天(此时为分化第0天)以MDI诱导培养基刺激细胞(换去原有的完全培养基,加入新配制的MDI诱导培养基)。在未来2天中会注意到细胞形态发生显著的变化(变得更加纺锤形)。 3.加入MDI诱导2天之后(分化第2天),换去原有的MDI诱导培养基,加入新配制的胰岛素培养基。培养基会开始变得更加粘稠,因为细胞合成的游离脂肪酸被大量的分泌到培养基中。 4.两天之后(分化第4天),将培养基换成普通的完全培养基(含有10%FBS的DMEM高糖培养基),之后每2天换一次液(含有10%FBS的DMEM高糖培养基)。通常在分化第8天的时候达到分化完全的状态。 药物编号1至9分别代表的化合物结构式依次为: 油红O染色数据的分析和处理 油红O染色所用试剂的配制: 油红O储存液:油红O(AMRESCO,FW 408.51,Em(513nm))0.7g溶于200ml异丙醇,过夜颠倒振荡混匀,0.2μm滤膜过滤后储存在4℃。 油红O工作液:6份油红O储存液加上4份dH2O,混匀后室温静置20分钟,0.2μm滤膜过滤。 油红O染色: 1.吸去绝大部分培养基。 2.加入10%福尔马林(溶于PBS中),室温孵育5分钟。 3.吸去10%福尔马林,加入同样体积的新鲜福尔马林(未稀释的37%甲醛溶液),孵育至少1小时,甚至更久。注:细胞在福尔马林中可以保持好几天,然后再染色。将培养板四周用封口膜密封以防止甲醛挥发蒸干,并以铝箔纸包裹以避光。 4.用小枪头吸去所有福尔马林。 5.以60%异丙醇(溶于dH2O中)清洗培养孔。 6.让细胞培养孔彻底干燥。 7.加入油红O工作液,染色10分钟(加入时不要碰到孔壁)。 8.移去所有油红染液,立即加入dH2O,用水洗4遍(可直接在自来水龙头下冲洗)。 9.根据实验需要拍照。 10.移去所有的水,让培养孔完全干燥。 11.加入100%异丙醇洗脱油红O,孵育10分钟(可以更久)。 12.反复吹吸含有油红的异丙醇几次,保证所以油红都溶进溶液中。 13.将含有油红的异丙醇转移至EP管或者ELISA板的孔中。 14.在500nm波长处测量OD值,读数时间为0.5秒。 15.空白对照使用100%异丙醇,染色本底对照使用来自按照上述步骤染色的没养细胞的空培养孔的异丙醇。 表1.油红O染色中各种试剂的用量 细胞培养板 福尔马林 60%异丙醇 油红O 100%异丙醇 24孔板 500μl 500μl 200μl 750μl 12孔板 1ml 1ml 400μl 1.5ml 6孔板 2.4ml 2.4ml 1ml 3.6ml 青蒿素衍生物对3T3-L1细胞的分化的影响 将固体青蒿素(图1.1、3-7号化合物)溶解于DMSO中,配制成一系列梯度浓度的溶液,本实验中该浓度梯度包括5个水平:1mM、5mM、10mM、30mM、50mM。二氢青蒿素(化合物2)浓度梯度为:0.1mM、0.5mM、1mM、5mM、10mM、30mM、50mM;青蒿琥酯(化合物9)的浓度梯度为:0.05mM、0.1mM、0.5mM、1mM、5mM、10mM、30mM、50mM。 如图3所示,依照上述所描述的诱导未分化的3T3-L1细胞(成纤维细胞状态)向成熟脂肪细胞分化的操作流程,在分化第0天的时候,向已经达到接合状态但尚未分化的3T3-L1细胞加入诱导分化的MDI培养基。加入青蒿素药物的处理操作为,在MDI培养基中按照1/1000的比例预先加入如上所述配置成的青蒿素系列梯度浓度的溶液,混匀之后再将培养基加入细胞培养孔。1/1000的比例,使得化合物1、3-7药物的终浓度变为1μM、5μM、10μM、30μM、50μM,也就是本实验实际使用的药物剂量。同时需要设计一个处理,在培养基中加入1/1000的DMSO作为溶剂对照,也就是药物浓度为0μM的处理。 依照上述所描述的细胞培养条件培养经过处理的3T3-L1细胞2天,依照上述所描述的诱导未分化的3T3-L1细胞(成纤维细胞状态)向成熟脂肪细胞分化的操作流程,在分化第2天加入胰岛素培养基,并用如上所述同样的方法和剂量,加入系列梯度浓度的青蒿素(同样的细胞培养孔加入同一个浓度的同一种药物)。 依照上述描述,对完成分化的3T3-L1细胞进行油红O染色,拍照,测吸光度,收集数据,整理,做图,比较,结果见图1。 图1中这9个化合物每个浓度的数据均来源于3次不同批次的重复实验得到的平均值,误差由3次重复实验之间的SE值表示。 药物加入之后,会抑制或是促进或是不显著影响3T3-L1细胞脂肪分化(对于青蒿素系列药物,本发明关注的是抑制)。本发明以细胞脂肪分化的程度来表示药效。细胞分化的程度可以在实验完成后用油红染色法染色后,测OD值得以定量检测。分化程度越高,油滴积累越多,油红染色越深,OD值越高。 本发明的分化实验使用24孔细胞培养板来做,每个24孔板会设计不加药的几个孔做对照,也就是可以完全分化的细胞的对照。 每个药物在特定浓度的药效,是通过相应的OD值比上同一个24孔细胞培养板上的完全分化的对照的OD值得到(相对分化程度)。这样,不同培养板得出的数据就可以相互比较。 如图1所示,如果是完全分化,得到的值(相对分化程度)就是1(100%);分化受到抑制,值就小于1(0~1之间)。分化抑制程度越高,值越小,药效越好。 结果如图1所示,青蒿素本身(图1.1号化合物)抑制3T3-L1细胞分化的效果很差,浓度在50μM的水平,3T3-L1细胞相对分化程度能达到93.3%;在10μM的水平,相对分化程度能达到94.8%。 结果见图1,图2.1,二氢青蒿素(图1.2号化合物)抑制3T3-L1细胞分化的效果为所有已经被测试过的青蒿素衍生物中最好的之一,在10μM的水平已经能完全抑制3T3-L1细胞的分化,相对分化程度只有24.5%。 结果见图1,图2.3,该化合物(图1.3号化合物)抑制3T3-L1细胞分化的效果一般,在10μM的水平,3T3-L1细胞的相对分化程度有62.5%。 结果见图1,图2.2,青蒿素苯甲酸酯(图1.4号化合物)抑制3T3-L1细胞分化的效果为所有已经被测试过的青蒿素衍生物中最好的之一,在10μM的水平已经能完全抑制3T3-L1细胞的分化,相对分化程度只有27.3%。 结果见图1,图2.4,烯丙基青蒿素(图1.5号化合物)抑制3T3-L1细胞分化的效果一般,在10μM的水平,3T3-L1细胞的相对分化程度有58.1%。 结果见图1,乙酸基青蒿素(图1.6号化合物)抑制3T3-L1细胞分化的效果较差,在10μM的水平,3T3-L1细胞的相对分化程度有81.3%。 结果见图1,青蒿素-生物素(图1.7号化合物)抑制3T3-L1细胞分化的效果很差,在10μM的水平,3T3-L1细胞的相对分化程度有91.6%。 结果见图1,去氧二氢青蒿素(图1.8号化合物)几乎没有抑制3T3-L1细胞分化的效果,在10μM的水平,3T3-L1细胞的相对分化程度能达到98.8%。 结果见图1,图2.5,青蒿琥酯(图1.9号化合物)抑制3T3-L1细胞分化的效果为所有已经被测试过的青蒿素衍生物中最好的之一,在10μM的水平已经能完全抑制3T3-L1细胞的分化,相对分化程度只有31.5%。 1.具有以下式Ⅰ、Ⅱ、Ⅲ、Ⅳ、Ⅴ结构的青蒿素衍生物在制备防治肥胖的药物中的应用, 其中: n=0~1;R1选自C1~C3烷基;R2选自芳基或含氮原子取代的芳香杂环。 2.根据权利要求1所述的应用,其特征是:所述青蒿素衍生物为具有以下结构式中的任一种: 3.根据权利要求1所述的应用,其特征是:所述青蒿素衍生物为具有以下结构式中的任一种: https://patents.google.com/patent/CN101879158A/zh?oq=CN101879158A青蒿素衍生物的新应用---防治肥胖病的 Description The new application of artemisinin derivative Technical field The present invention relates to the new application of artemisinin derivative in pharmacy, belong to chemical biology and cytobiology field. Background technology Arteannuin is the sesquiterpene lactones medicine that contains peroxy-radical that extracts from Herba Artemisiae annuae.The discovery of artemisinin-based antimalarial drug thing is the another milestone after quinine on the global antimalarial agent development history, it is first phytochemistry medicine that China finds, also be China unique that approved by World Health Organization (WHO), can be by the Chinese medicine of synthetic drug research standard exploitation.Over more than 30 year, arteannuin distinctive molecular structure and outstanding biological activity are attracting the eyeball of scientists deeply.Along with the development of medical sci-tech, prove that through pharmacological action and clinical practice artemisinin derivative is widely used in treating malaria, cancer is in the diseases such as actute infection and hyperpyrexia disease and dermatosis.Wherein the more artemisinin derivative of clinical research has dihydroartemisinine, Artemether, arteether and artesunate etc.Have not yet to see the bibliographical information that relevant artemisinin derivative suppresses the adipose cell differentiation. Obesity is meant that body feed heat is more than the health consumption, cause the transition accumulation of fat and the excessive amplification of fatty tissue, make body weight more than 20% of body weight that is above standard, and the class disease weak with giddy, that Mental fatigue is breathed hard, be the risk factor of hypertension, diabetes, hyperlipidemia, coronary heart disease, cerebrovascular.Now, fat one of fast, the most serious public health problem of global rate of propagation that become.In recent years, along with the raising of China's living standards of the people, fat number has increased in urban population, and do not occur over just the old people, also occur in child, women, the number of each age group body obesity is all increasing, and fat-reducing has become people's much-talked-about topic.The cause of disease that causes obesity has a lot, and wherein main and h and E factor has substantial connection.Environmental factors is meant eats high fat, high heat food for a long time, adds physical exertion and reduces, and under the effect of factors such as mental maladjustment, causes that the neuroendocrine of body fat regulated and control network is regulated disorderly. Therefore, the generation of prevention and controlling obesity disease has become the problem of medically very paying close attention to.At present, slimming medicine commonly used roughly is divided three classes: one, the medicine of control appetite, and this type of medicine has many untoward reaction, as insomnia, xerostomia, constipation, cardiopalmus and hypertension; Two, increase the medicine of energy expenditure; Three, the medicine that suppresses intestinal absorption.Although there are many researchs to be deep into the genomic medicine level now, but still do not find at present a kind of real effectively, slimming medicine that can life-time service, work out diagnosis and the treatment that even more ideal effective method and preparation be used for obesity and still have profound significance. 3T3-L1 be one derived from 3T3 cell (the fibroblastic cell line of standard that builds on 1962), in biological study, be applied to studying the cell line of fatty tissue.The 3T3-L1 cell has similar fibroblastic form, but under the inducing of felicity condition, can break up and form the phenotype of adipose cell.Adipose cell form after the 3T3-L1 cell differentiation maturation, the synthetic and accumulation of the intracellular glycerol three esters fibroblast state preceding with respect to differentiation all improves greatly, and 3T3-L1 cell at this time presents a kind of adipose cell form of signet ring.These cells are all responsive to hormone and the medicine of regulating lipogenesis and fat acid decomposition, epinephrine for example, norepinephrine, insulin etc. Because these characteristics of 3T3-L1 cell, this cell line are widely used in the research of fat differentiation and metabolic disease mechanism, be the cell line model of internationally recognized external fat differentiation.The cascade signal of transcribing that this Study of model has been helped to illustrate a main regulation and control fat differentiation in the terminal differentiation of adipose cell is transduceed, and has comprised a series of transcription factor, modulin and effect protein. The oil red O stain technology is a kind of lipid staining commonly used, and its advantage is that staining procedure is easy, and coloration result is sure.Dyeing back fat is cerise, and it is blue that nucleus is, and a matter is colourless.The redness that oil red O dyes is directly proportional with the amount of the oil droplet that the interior accumulation of adipose cell forms, and the latter is directly proportional with the height of adipose cell differentiation degree, so by this technology, both can judge by qualitative observation, can also detection by quantitative the height of adipose cell differentiation degree relatively. Herba Artemisiae annuae extensively is present in all over the world, but the higher Herba Artemisiae annuae of artemislnin content exists only in east, China Chongqing, Fujian, Guangxi, some areas, Hainan, belongs to the exclusive drug resource of China.Nearly decades, the research of artemisinin derivative mainly concentrates on malaria and anti-tumor aspect, and the research that applies it to appetrol is but very rare. Summary of the invention The objective of the invention is promptly provides a kind of new application of artemisin derivant in order to improve the application of artemisinin derivative in frontier. Concrete technical scheme is as follows: The application of artemisinin derivative in the fat medicine of preparation control with following formula I, II, III, IV, V structure, Wherein: N=0~1; R 1Be selected from C 1~C 3Alkyl; R 2Be selected from the aromatic heterocycle that aryl or nitrogen atom replace. Preferably, described artemisinin derivative is to have in the following structural formula any Wherein, dihydroartemisinine, arteannuin benzoate have the effect of inhibition 3T3-L1 cell differentiation the strongest in all artemisinin derivatives that detected.Next is an artesunate, has the effect of the inhibition 3T3-L1 cell differentiation that is only second to the above two.These 3 chemical compounds can both suppress the differentiation of 3T3-L1 cell fully in the level of 10 μ M. The objective of the invention is to develop the effect of artemisinin derivative in slimming medicine, it is suppressed the cellular fat differentiation study in great detail. The present invention has the following advantages: (1) arteannuin and derivant thereof are traditional anti-malaria medicaments, be used for the treatment of malaria all the time, characteristics of the present invention are that the part derivant that proposes arteannuin first has the effect that suppresses the adipose cell differentiation, disease with them and metabolism class connects first, and proposes their first as the potential probability that is applied to the medicine of obesity and obesity-related disease such as cardiovascular disease.The metabolism class disease relevant with obesity is the significant threat of human health in current society, by the medicine of the biological medicine for preparing of described artemisine as the differentiation of lipotropism fat, will play a significant role in these treatment of diseases. (2) the present invention has compared the power that suppresses the adipose cell differentiation capability between the different artemisinin derivatives, has drawn preliminary structure activity relationship, provides foundation for further optimizing the active better artemisinin derivative of acquisition. Description of drawings Fig. 1. be the inhibitory action of arteannuin series derivates to the adipose cell differentiation, abscissa is a drug use dosage (concentration, represent with μ M), vertical coordinate is relative differentiation rate (dosing handle experimental group oil red dyeing OD value/break up fully matched group oil red dye OD value); Fig. 2. be the oil red dyeing general effect of 5 the most representative chemical compounds of the inhibition 3T3-L1 cell differentiation effect of choosing; Wherein, Fig. 2 .1 is the oil red dyeing general effect that chemical compound 2 Concentraton gradient influence the 3T3-L1 cell differentiation; Fig. 2 .2 is the oil red dyeing general effect that chemical compound 4 Concentraton gradient influence the 3T3-L1 cell differentiation; Fig. 2 .3 is the oil red dyeing general effect that chemical compound 3 Concentraton gradient influence the 3T3-L1 cell differentiation; Fig. 2 .4 is the oil red dyeing general effect that chemical compound 5 Concentraton gradient influence the 3T3-L1 cell differentiation; Fig. 2 .5 is the oil red dyeing general effect that chemical compound 9 Concentraton gradient influence the 3T3-L1 cell differentiation; Fig. 3 is the flow process stage sketch map of inducing the 3T3-L1 cell differentiation. The specific embodiment Artemisinin derivative with following formula I, II, III, IV, V structure of the present invention is mainly the chemical compound with following structural formula: (a) described chemical compound with formula I structure is: (b) described chemical compound with formula II structure is: (c) described chemical compound with formula III structure is: (d) described chemical compound with formula IV structure is: (e) described chemical compound with formula V structure is: For making the present invention easier to understand,, further set forth the present invention below in conjunction with specific embodiment.Should be understood that these embodiment only are used for illustration purpose, and be not used in the restriction scope of the invention. Those skilled in the art can obtain described artemisinin derivative with described formula I, II, III, IV, V structure from following description or prior art. Embodiment 1: deoxidation arteannuin synthetic 200mg (0.7087mmol) arteannuin is dissolved in 10mL THF, behind the material dissolution, adds 547mg (20.2688mmol) Al powder and 7.23g (30.4032mmol) NiCl 26H 2O, afterreaction finished in 4 hours, was spin-dried for solvent, was spin-dried for thing with acetic acid ethyl dissolution, and sucking filtration is with ethyl acetate drip washing filtering residue.Filtrate is through anhydrous Na 2SO 4Drying concentrates back column chromatography (PE: EA=16: 1) obtain the 72mg target product, yield 38%. 1H?NMR(CDCl 3,400MHz):δ=5.68(s,1H),3.19-3.16(m,1H),2.02-1.58(m,6H),1.51(s,3H),1.28-1.03(m,11H). Embodiment 2: deoxidation dihydroartemisinine synthetic 500mg (1.7596mmol) dihydroartemisinine is dissolved in 5mL DCM, adds 45mg 10%Pd/C, under the hydrogen shield; 40 ℃ of reaction overnight finish, and remove by filter Pd/C, are spin-dried for solvent; concentrate back column chromatography (PE: EA=8: 1) obtain the 202mg target product, yield 43%. 1H?NMR(CDCl 3,400MHz):δ=5.34(s,1H),4.78(t,J=6.8Hz,1H),2.83(d,J=7.2Hz,1H),2.40-0.79(m,21H). Embodiment 3: dihydroartemisinine synthetic 10g (35.4321mmol) arteannuin is dissolved in 500mL methanol, stirs and is cooled to 0-5 ℃, adds 10g (265.7408mmol) NaBH in 1 hour in batches 4Keep this temperature to continue to stir after 1.5 hours, in reaction bulb, slowly drip 15.5mLHAc adjusting pH=7, separate out a large amount of white solids gradually.Be spin-dried for most of solvent, add 10mL cold water, stirred 15 minutes under the room temperature, sucking filtration is used cold H 2O: MeOH=2: 1 washing leaching cake, collect filter cake, obtain the 8.0g target product after the drying, yield 80%. 1H?NMR(CDCl 3,400MHz):δ=5.60(s,1H),5.28(s,1H),2.85(s,1H),2.61(s,1H),2.41-0.89(m,20H). Embodiment 4: β-Artemether synthetic 150mg (0.5275mmol) dihydroartemisinine is dissolved in 1.0mL MeOH and the 2mL DCM mixed solvent, is heated to 45 ℃, under the argon shield, injects 0.01mL (0.0559mmol) BF rapidly 3Et 2O, reaction is 1 hour under this temperature, and after reaction finished, with sodium acetate aqueous solution washing reaction liquid, organic layer was through anhydrous MgSO 4After the drying, concentrate the mixture that promptly obtains β-Artemether and α-Artemether, mixture is dissolved in an amount of normal hexane, preserved 36 hours, collect white crystal, and, obtain the pure β-Artemether of 107mg after the drying, yield 65% with cold normal hexane drip washing at-20 ℃. 1H?NMR(CDCl 3,400MHz):δ=5.38(s,1H),4.68(d,J=3.2Hz,1H),3.42(s,3H),2.64-0.85(m,21H). Embodiment 5: arteannuin benzoate synthetic Under the argon shield; 3.386g (11.9158mmol) dihydroartemisinine is dissolved in 40mL DCM, adds 6.1mL (76.2613mmol) pyridine, stirs and is cooled to 0 ℃; add 2.2mL (19.0653mmol) Benzenecarbonyl chloride.; keep reacting 15 minutes under this temperature, remove ice bath and rise to room temperature, continue reaction and finished in 16 hours; add 7% aqueous citric acid solution cessation reaction; be spin-dried for DCM, add acetic acid ethyl dissolution and be spin-dried for thing, use 7% aqueous citric acid solution, saturated NaHCO successively 3Solution, saturated NaCl solution washing organic layer, organic layer is through anhydrous Na 2SO 4Drying concentrates back column chromatography (PE: EA=16: 1) obtain the 4.07g target product, yield 88%. 1H?NMR(CDCl 3,400MHz):δ=8.12(d,J=7.2Hz,2H),7.59-7.55(m,1H),7.44(t,J=8.0Hz,2H),6.01(d,J=9.6Hz,1H),5.53(s,1H),2.76-0.92(m,21H). Embodiment 6: β-pi-allyl arteannuin synthetic Add a small amount of 4 in the reaction bulb The anhydrous ZnCl of MS, 105mg (0.7790mmol) 2, the Ar protection is injected anhydrous DCM of 3mL and 0.5mL (3.1160mmol) allyl trimethyl silane down, stirs and is cooled to 0 ℃. Claim 252mg (0.6492mmol) arteannuin benzoate to be dissolved in the anhydrous dichloroethanes of 3mL, solution is splashed in the above-mentioned reactant liquor, keep 0 ℃ of reaction 1 hour, remove ice bath, rose to room temperature reaction 3 hours, add an amount of ethyl acetate diluting reaction system, use 7% aqueous citric acid solution, saturated NaHCO successively 3Solution, saturated NaCl solution washing organic layer, organic layer is through anhydrous Na 2SO 4Drying concentrates back column chromatography (PE: EA=30: 1), obtain the 114mg target product, yield 66%. 1H?NMR(CDCl 3,400MHz):δ=5.92(m,1H),5.32(s,1H),5.14-5.04(m,2H),4.30(m,1H),2.69-0.87(m,23H). Embodiment 7: β-acetic acid arteannuin synthetic 100mg (0.3245mmol) β-pi-allyl arteannuin is dissolved in 4mL CCl 4-CH 3CN-H 2In O (V: V: V=1: 1: the 2) mixed solvent, add 347mg (1.6225mmol) NaIO successively 4And 2mg (0.0097mmol) RuCl 3, the ambient temperature overnight reaction.Be spin-dried for solvent, add acetic acid ethyl dissolution and be spin-dried for thing, use saturated NaHSO successively 3, saturated NaHCO 3Solution, saturated NaCl solution washing organic layer, organic layer is through anhydrous Na 2SO 4Drying concentrates back column chromatography (DCM: MeOH=30: 1), obtain the 92mg target product, yield 87%. 1H?NMR(CDCl 3,400MHz):δ=5.37(s,1H),4.86(m,1H),2.71-0.88(m,24H). Embodiment 8:D-Biotin modifies the synthetic of arteannuin Reagents?and?conditions:(a)Boc 2O,Et3N,THF,R.T.,15h;(b)D-biotin,EDCI,HOBt,(i-Pr) 2NEt,DMF,R.T.,10h;(c)TFA/CH 2Cl 2,0℃~R.T.,9h;(d)EDCI,HOBt,(i-Pr) 2NEt,DMF,R.T.,11h. 2 synthesize: 2.035g (13.7360mmol) be dissolved among the 18mL THF, add people 0.63mL (4.5767mmol) NEt 3, stir and be cooled to 0 ℃. 1g (4.5767mmol) Boc 2O is dissolved among the 9mL THF, behind the mix homogeneously, dropwise splashes in the above-mentioned reactant liquor, after dropwising, rises to room temperature reaction naturally 15 hours.After reaction finishes, be spin-dried for solvent, behind the thin up, with DCM extraction, organic layer is with saturated NaCl solution washing, anhydrous Na 2SO 4Drying concentrates back column chromatography (DCM: MeOH=5: 1), obtain the 906mg target product, yield 80%. 1H?NMR(CDCl 3,400MHz):δ=5.18(s,1H),3.56(s,4H),3.50-3.45(m,4H),3.30-3.25(m,2H),2.82(t,J=5.6Hz,2H),1.53(s,2H),1.38(s,9H). 3 synthesize: Under the argon shield, (1.3mmol, 1.3equiv.) (1.3mmol, 1.3equiv.) HOBt is dissolved in the dry DMF of 4mL, adds 0.23mL (1.4mmol, 1.4equiv.) DIPEA, stirring at room for EDCI, 176mg for 318mg (1.3mmol) D-Biotin, 249mg. 248mg (1.0mmol) 2 is dissolved among the dry DMF of 3mL, solution is injected above-mentioned reaction bulb, room temperature reaction 10 hours.After reaction finishes, in reaction bulb, add NH 4The Cl saturated solution, chloroform extraction, organic layer is through NH 4Cl saturated solution, water, saturated NaCl solution washing, anhydrous Na 2SO 4Drying concentrates back column chromatography (DCM: MeOH=20: 1), obtain the 358mg target product, yield 76%. 1H?NMR(CDCl 3,400MHz):δ=6.67(s,1H),6.15(s,1H),5.52(s,1H),5.15(s,1H),4.45-4.40(m,1H),4.25-4.20(m,1H),3.52(s,4H),3.49-3.45(m,4H),3.37-3.34(m,2H),3.23-3.21(m,2H),3.10-3.04(m,1H),2.14(t,J=7.6Hz,2H),1.70-1.50(4H),1.36(s,12H). 4 synthesize: 118mg 3 is dissolved in the 3mL dichloromethane, stirring is cooled to 0 ℃, slowly drip 1.0mL TFA, add the recession deicing and bathe, rose to room temperature reaction naturally 9 hours, after reaction finishes, directly distilling under reduced pressure removes and desolvates, vacuum drying obtains crude product (by 100% calculating, should obtain the 93mg crude product) 4, is directly used in next step reaction without separation. 1H?NMR(MeOD,400MHz):δ=4.52-4.49(m,1H),4.32-4.29(m,1H),3.72-3.65(m,6H),3.56(t,J=5.6Hz,2H),3.39-3.35(m,2H),3.22-3.20(m,1H),3.14-3.11(m,2H),2.95-2.91(dd,J=12.8Hz,4.8Hz,1H),2.71(d,J=12.8Hz,1H),2.22(t,J=7.6Hz,2H),1.71-1.60(m,4H),1.47-1.43(m,2H). D-Biotin modifies the synthetic of arteannuin: Under the argon shield, 74mg (0.2269mmol) carboxylic acid arteannuin, 57mg (0.2949mmol) EDCI, 40mg (0.2949mmol) HOBt are dissolved in the 3mL dry DMF, inject 0.24mL (1.4293mmol) DIPEA, stirring at room. Claim 93mg (0.2496mmol) 4 to be dissolved in the dry DMF of 2mL, solution is injected above-mentioned reactant liquor, the ambient temperature overnight reaction.After reaction finishes, add the DCM dilute reaction solution, use NH successively 4Cl saturated solution, water, saturated NaCl solution washing, anhydrous Na 2SO 4Drying concentrates back column chromatography (DCM: MeOH=10: 1), obtain the 89mg target product, yield 57%. 1H?NMR(CDCl 3,400MHz):δ=7.24(s,1H),δ6.85(s,1H),δ6.30(s,1H),δ5.41(s,1H),δ5.35(s,1H),δ4.75-4.71(m,1H),δ4.52-4.50(m,1H),δ4.32-4.29(m,1H),δ3.61-3.40(m,12H),δ3.15-3.16(m,1H),δ2.90-0.88(m,33H). Embodiment 9: Claim 50mg (0.1288mmol) arteannuin benzoate and an amount of 4 MS under the argon shield, injects 2mL and heavily steams the DCM dissolving in reaction bulb, is cooled to 0 ℃, injects 0.02mL (0.1546mmol) TMSCl, keeps this thermotonus to rise to the ambient temperature overnight reaction after 1 hour and finishes.Be spin-dried for solvent, add acetic acid ethyl dissolution and be spin-dried for thing, use saturated NaHCO successively 3Solution, saturated NaCl solution washing, organic layer is through anhydrous Na 2SO 4Drying concentrates back column chromatography (PE: EA=8: 1), obtain the 33mg target product, yield 96%. 1H?NMR(CDCl 3,400MHz):δ=6.19(s,1H),5.54(s,1H),2.44-0.99(m,20H). Embodiment 10: Under the argon shield, 50mg (0.1865mmol) deoxidation dihydroartemisinine is dissolved among the 3mL DCM, is cooled to 0 ℃, injects 0.03mL (0.3729mmol) chloroacetic chloride, rises to the ambient temperature overnight reaction gradually and finishes.Reactant liquor is through water washing, and organic layer is through anhydrous Na 2SO 4Drying, evaporating column chromatography (PE: EA=16: 1), obtain racemization deoxidation dihydroartemisinine 30mg, yield 64% as by-product. 1H?NMR(CDCl 3,400MHz):δ=6.04(s,1H),5.47(s,1H),2.04-1.21(m,20H). Embodiment 11: 100mg (0.3517mmol) dihydroartemisinine is dissolved in 5mL DCE, adding people 73mg (0.4220mmol) 2-quinoxaline carboxylic acid stirs moments later, add 5mL (0.0422mmol) DMAP, stirring is cooled to 0 ℃, adds 80mg (0.3868mmol) DCC, stirs and rises to room temperature after 5 minutes, stopped reaction after 16 hours, filtering reacting liquid, filtrate is through water, saturated NaCl solution washing, and organic layer is through anhydrous Na 2SO 4After the drying, the evaporating column chromatography obtains the 120mg target product, yield 78%. 1H?NMR(CDCl 3,400MHz):δ=9.57(s,1H),8.31(d,J=8.0Hz,1H),8.17(d,J=8.0Hz,1H),7.91-7.86(m,2H),6.03-6.00(m,2H),5.64(s,1H),2.90-0.95(m,21H). Embodiment 12: According to as described in example 12,100mg (0.3517mmol) dihydroartemisinine and the reaction of 63mg (0.4220mmol) terephthalaldehydic acid, column chromatography obtains the 116mg target product, yield 79%. 1H?NMR(CDCl 3,400MHz):δ=10.10(s,1H),8.27(d,J=8.0Hz,2H),7.96(d,J=8.0Hz,2H),6.03-6.00(m,1H),5.54(s,1H),2.90-0.95(m,21H). Embodiment 13: According to as described in example 12,100mg (0.3517mmol) dihydroartemisinine and 133mg (0.4220mmol) Boc-3-(2-naphthyl)-L-alanine reaction, column chromatography obtains the 151mg target product, yield 76%. 1H?NMR(CDCl 3,400MHz):δ=7.86-7.83(m,1H),7.80-7.74(m,3H),7.47-7.38(m,1H),6.05-6.00(m,1H),5.54(s,1H),2.83-0.85(m,34H). Embodiment 14: According to as described in example 12,100mg (0.3517mmol) dihydroartemisinine and the reaction of 76mg (0.4220mmol) paranitrophenylacetic acid, column chromatography obtains the 123mg target product, yield 78%. 1H?NMR(CDCl 3,400MHz):δ=8.18(d,J=8.0Hz,2H),7.48(d,J=12.0Hz,2H),6.03-6.00(m,1H),5.44(s,1H),3.82(s,2H),3.82(s,2H),2.65-0.68(m,23H). Embodiment 15: Under the argon shield; 100mg (0.3517mmol) dihydroartemisinine and 0.08mL (0.5627mmol) anisoyl chloride are dissolved among the 5mL DCM; be cooled to 0 ℃; inject 0.2mL (2.2509mmol) pyridine; stir after 15 minutes, rise to the ambient temperature overnight reaction and finish, react with 7% aqueous citric acid solution cancellation; ethyl acetate extraction is again through saturated NaHCO 3, saturated NaCl solution washing, organic layer is through anhydrous Na 2SO 4Drying concentrates the back column chromatography, obtains the 119mg target product, yield 81%. 1H?NMR(CDCl 3,400MHz):δ=8.06(d,J=12Hz,2H),6.91(d,J=12Hz,2H),5.98(m,1H),5.51(s,1H),3.864(s,3H),2.81-0.85(m,21H). Embodiment 16: Described according to example 15,100mg (0.3517mmol) dihydroartemisinine and the reaction of 104mg (0.5627mmol) paranitrobenzoyl chloride, column chromatography obtains the 81mg target product, yield 53%. 1H?NMR(CDCl 3,400MHz):δ=8.28(s,4H),6.05-6.00(m,1H),5.55(s,1H),2.83-0.85(m,22H). Embodiment 17: artesunate synthetic Under the room temperature condition, 250mg (0.9mmol) dihydroartemisinine is dissolved among the dry DCM of 25mL, adds 202mg (2mmol) succinic anhydride, be cooled to 0~5 ℃, add 107mg (0.9mmol) DMAP, kept this thermotonus 0.5 hour, rose to room temperature reaction naturally 1 hour.After reaction finishes, add 5mL water, transfer to pH=3 with 10% dilute hydrochloric acid, organic layer is used anhydrous MgSO after washing 4Drying, removal of solvent under reduced pressure obtains the 317mg target product, yield 94%. 1H?NMR(CDCl 3,400MHz):δ=5.74(d,J=8.0Hz,1H),5.39(s,1H),2.72-2.59(m,4H),2.53-2.48(m,1H),2.36-2.28.(m,1H),2.01-1.96(m,1H),1.87-1.82(m,1H),1.76-1.66(m,2H),1.60-1.55(m,1H),1.45-1.21(m,8H),1.01-0.95(m,1H),0.92(d,J=8.0Hz,3H),0.80(d,J=4.0Hz,3H). Embodiment 18: artemisinin derivative is to the influence of 3T3-L1 cell differentiation The cultivation of 3T3-L1 cell The 3T3-L1 cell is at 37 ℃, 5%CO 2Adherent growth in the culture environment is used the DMEM high glucose medium (HyClone) that adds 10%FBS (Hyclone hyclone).Can be according to cell growth condition, changed a not good liquor in 2~3 days.Continue if desired repeatedly to go down to posterity, and need still have good differentiation capability by cell after repeatedly go down to posterity, too full (<70%) that will avoid so making cell long generally longly will go down to posterity the branch cell to 60%~70%.Cell can be with go down to posterity branch dish of rare ratio by 1: 15, but usually with 1: 10 or the higher ratio branch dish that goes down to posterity, need be as the criterion with experiment. The culture medium preparation of 3T3-L1 cell induction differentiation Inducing culture is divided into two kinds, and MDI inducing culture and insulin culture medium are respectively applied for different differentiation phases, is to add that by the DMEM culture medium FBS, dexamethasone, insulin and the IBMX of appropriate amount are made into. IBMX solution: IBMX is dissolved in the KOH solution of 0.5N, the IBMX final concentration is 0.0115g/ml, 0.22 μ m syringe filters filtration sterilization ,-20 ℃ of preservations. The insulin storage solutions: insulin is dissolved among the HCl of 0.02M to final concentration 167uM (1mg/ml), 0.22 μ m syringe filters filtration sterilization ,-20 ℃ of long preservation, 4 ℃ of short-terms are preserved. Dexamethasone storage solutions: stored frozen liquid: the 10mM dexamethasone is dissolved in dehydrated alcohol ,-20 ℃ of preservations; The work storage liquid: stored frozen liquid is diluted to 1mM with PBS, and filtration sterilization is stored in 4 ℃. MDI inducing culture (now with the current, 10ml/10cm culture dish, 5ml/6cm culture dish): the DMEM high glucose medium that contains 10%FBS adds again: 1: 100 IBMX solution, 1: 1000 insulin storage liquid, 1: 1000 dexamethasone work storage liquid. Insulin culture medium (now with the current, 10ml/10cm culture dish, 5ml/6cm culture dish): the DMEM high glucose medium that contains 10%FBS adds again: 1: 1000 insulin storage liquid. Induce undifferentiated 3T3-L1 cell (fibroblast state) to break up to mature fat cell: As shown in Figure 3: In containing the DMEM high glucose medium of 10%FBS with 3T3-L1 before adipose cell be cultured to engagement state. 2. support 2 days (being differentiation the 0th day this moment) after engaging again with MDI inducing culture irritation cell (change original complete medium, add the MDI inducing culture of new preparation).Can notice that in following 2 days cellular morphology takes place by significant change (spindle more becomes). 3. add MDI and induce (breaking up the 2nd day) after 2 days, change original MDI inducing culture, add the insulin culture medium of new preparation.Culture medium can begin the thickness more that becomes, because the synthetic free fatty of cell is by a large amount of being secreted in the culture medium. 4. (broke up the 4th day) after two days, change culture medium into common complete medium (the DMEM high glucose medium that contains 10%FBS), changed a not good liquor (the DMEM high glucose medium that contains 10%FBS) in per afterwards 2 days.Usually reach in the 8th day in differentiation and break up state completely. Medicine numbering 1 to 9 structural formula of compound of representative respectively is followed successively by: The analysis of oil red O stain data and processing The preparation of oil red O stain agents useful for same: Oil red O storage liquid: oil red O (AMRESCO, FW 408.51, Em (513nm)) 0.7g is dissolved in the 200ml isopropyl alcohol, spends the night and puts upside down the vibration mixing, is stored in 4 ℃ behind the 0.2 μ m membrane filtration. Oil red O working solution: 6 parts of oil red O storage liquid add 4 parts of dH 2O, room temperature left standstill 20 minutes behind the mixing, 0.2 μ m membrane filtration. Oil red O stain: 1. inhale and go most culture medium. 2. add 10% formalin (being dissolved among the PBS), incubated at room 5 minutes. 3. inhale and remove 10% formalin, add the fresh formalin (undiluted 37% formalin) of same volume, hatched at least 1 hour, in addition more of a specified duration.Annotate: cell can keep several days in formalin, and then dyeing.With around the culture plate with sealing film phonograph seal preventing formaldehyde volatilization evaporate to dryness, and wrap up with lucifuge with aluminium-foil paper. 4. inhale with the lancet head and remove all formalin. 5. (be dissolved in dH with 60% isopropyl alcohol 2Among the O) the cleaning culture hole. 6. allow cell culture hole finish-drying. 7. add oil red O working solution, dye 10 minutes (adding the fashionable hole wall that keeps off). 8. remove all oil red dye liquors, add dH immediately 2O washes with water 4 times (can directly wash under water tap). 9. take pictures according to the experiment needs. 10. remove all water, allow the culture hole bone dry. 11. add 100% isopropyl alcohol eluting oil red O, hatch 10 minutes (can be more of a specified duration). 12. repeatedly pressure-vaccum contain oil red isopropyl alcohol several times, so guarantee that oil red all dissolves in the solution. 13. will contain in the hole that the isopropyl alcohol of oil red is transferred to EP pipe or elisa plate. 14. measure the OD value at 500nm wavelength place, the reading duration is 0.5 second. 15. blank uses 100% isopropyl alcohol, dyeing background contrast is used from according to the painted isopropyl alcohol of the empty culture hole of foster cell of above-mentioned steps with not ing. The consumption of all ingredients in table 1. oil red O stain Tissue Culture Plate Formalin 60% isopropyl alcohol Oil red O 100% isopropyl alcohol 24 orifice plates ??500μl ??500μl ??200μl ??750μl 12 orifice plates ??1ml ??1ml ??400μl ??1.5ml 6 orifice plates ??2.4ml ??2.4ml ??1ml ??3.6ml Artemisinin derivative is to the influence of the differentiation of 3T3-L1 cell Solid arteannuin (Fig. 1 .1,3-7 chemical compound) is dissolved among the DMSO, is mixed with the solution of a series of gradient concentrations, this Concentraton gradient comprises 5 level: 1mM, 5mM, 10mM, 30mM, 50mM in this experiment.Dihydroartemisinine (chemical compound 2) Concentraton gradient is: 0.1mM, 0.5mM, 1mM, 5mM, 10mM, 30mM, 50mM; The Concentraton gradient of artesunate (chemical compound 9) is: 0.05mM, 0.1mM, 0.5mM, 1mM, 5mM, 10mM, 30mM, 50mM. As shown in Figure 3, according to above-mentioned described operating process of inducing undifferentiated 3T3-L1 cell (fibroblast state) to the mature fat cell differentiation, in differentiation the 0th day, to reaching engagement state but still undifferentiated 3T3-L1 cell adds the MDI culture medium of inducing differentiation.The processing that adds the arteannuin medicine is operating as, and adds the solution of the arteannuin series gradient concentration that is configured to as mentioned above in the MDI culture medium in advance according to 1/1000 ratio, culture medium is added the cell culture hole after the mixing again.1/1000 ratio makes the final concentration of chemical compound 1,3-7 medicine become 1 μ M, 5 μ M, 10 μ M, 30 μ M, 50 μ M, the actual drug dose that uses of this experiment just.Need to design a processing simultaneously, the DMSO of adding 1/1000 is as solvent control in culture medium, and just drug level is the processing of 0 μ M. Cultivated treated 3T3-L1 cell 2 days according to above-mentioned described cell culture condition, according to above-mentioned described operating process of inducing undifferentiated 3T3-L1 cell (fibroblast state) to the mature fat cell differentiation, added the insulin culture medium on the 2nd day in differentiation, and with same as mentioned above method and dosage, add the arteannuin (same cell culture hole adds same a kind of medicine of same concentration) of serial gradient concentration. According to foregoing description, the 3T3-L1 cell of finishing differentiation is carried out oil red O stain, take pictures, survey absorbance, collect data, figure is in arrangement, relatively, the results are shown in Figure 1. The data of these 9 each concentration of chemical compound all derive from the meansigma methods that the repeated experiments of 3 different batches obtains among Fig. 1, and error is by the SE value representation between 3 repeated experiments. After medicine adds, can suppress or promote or not appreciable impact 3T3-L1 cellular fat differentiation (for the arteannuin medicine series, what the present invention paid close attention to is to suppress).The present invention represents drug effect with the degree of cellular fat differentiation.The degree of cell differentiation can be with after the dyeing of oil red staining after experiment is finished, and surveying OD is worth with detection by quantitative.Differentiation degree is high more, and the oil droplet accumulation is many more, and oil red dyeing is dark more, and the OD value is high more. Analytical Chemical Experiment of the present invention uses 24 porocyte culture plates to do, and each 24 orifice plate can design several holes of not dosing and do contrast, just the contrast of the cell that can break up fully. Each medicine is to obtain (differentiation degree relatively) by corresponding OD value than the OD value of the contrast of differentiation fully on the last same 24 porocyte culture plates in the drug effect of specific concentrations.Like this, the data that draw of different culture plate just can compare mutually. As shown in Figure 1, if differentiation fully, the value that obtains (differentiation degree relatively) is exactly 1 (100%); Differentiation is suppressed, and value is just less than 1 (between 0~1).Differentiation inhibition degree is high more, is worth more for a short time, and drug effect is good more. The result as shown in Figure 1, arteannuin itself (Fig. 1 .1 chemical compound) suppresses the poor effect of 3T3-L1 cell differentiation, concentration is in the level of 50 μ M, the relative differentiation degree of 3T3-L1 cell can reach 93.3%; In the level of 10 μ M, differentiation degree can reach 94.8% relatively. The results are shown in Figure 1, Fig. 2 .1, the effect that dihydroartemisinine (Fig. 1 .2 chemical compound) suppresses the 3T3-L1 cell differentiation be that all are one of best in the artemisinin derivative of tested mistake, can suppress the differentiation of 3T3-L1 cell fully in the level of 10 μ M, and relative differentiation degree has only 24.5%. The results are shown in Figure 1, Fig. 2 .3, the effect that this chemical compound (Fig. 1 .3 chemical compound) suppresses the 3T3-L1 cell differentiation is general, and in the level of 10 μ M, the relative differentiation degree of 3T3-L1 cell has 62.5%. The results are shown in Figure 1, Fig. 2 .2, the effect that arteannuin benzoate (Fig. 1 .4 chemical compound) suppresses the 3T3-L1 cell differentiation is that all are one of best in the artemisinin derivative of tested mistake, can suppress the differentiation of 3T3-L1 cell fully in the level of 10 μ M, differentiation degree has only 27.3% relatively. The results are shown in Figure 1, Fig. 2 .4, the effect that pi-allyl arteannuin (Fig. 1 .5 chemical compound) suppresses the 3T3-L1 cell differentiation is general, and in the level of 10 μ M, the relative differentiation degree of 3T3-L1 cell has 58.1%. The results are shown in Figure 1, the effect of acetate arteannuin (Fig. 1 .6 chemical compound) inhibition 3T3-L1 cell differentiation is relatively poor, and in the level of 10 μ M, the relative differentiation degree of 3T3-L1 cell has 81.3%. The results are shown in Figure 1, arteannuin-biotin (Fig. 1 .7 chemical compound) suppresses the poor effect of 3T3-L1 cell differentiation, and in the level of 10 μ M, the relative differentiation degree of 3T3-L1 cell has 91.6%. The results are shown in Figure 1, deoxidation dihydroartemisinine (Fig. 1 .8 chemical compound) does not almost suppress the effect of 3T3-L1 cell differentiation, and in the level of 10 μ M, the relative differentiation degree of 3T3-L1 cell can reach 98.8%. The results are shown in Figure 1, Fig. 2 .5, the effect that artesunate (Fig. 1 .9 chemical compound) suppresses the 3T3-L1 cell differentiation be that all are one of best in the artemisinin derivative of tested mistake, can suppress the differentiation of 3T3-L1 cell fully in the level of 10 μ M, and relative differentiation degree has only 31.5%. Claims (3) Hide Dependent 1. the application of artemisinin derivative in the fat medicine of preparation control that has following formula I, II, III, IV, V structure, Wherein: N=0~1; R 1Be selected from C 1~C 3Alkyl; R 2Be selected from the aromatic heterocycle that aryl or nitrogen atom replace. 2. application according to claim 1 is characterized in that: described artemisinin derivative is to have in the following structural formula any: 3. application according to claim 1 is characterized in that: described artemisinin derivative is to have in the following structural formula any: https://patents.google.com/patent/CN101879158A/en?oq=CN101879158A青蒿素衍生物的新应用---防治肥胖病的
  8. The invention discloses Artemisia apiacea extract and a preparation method and application thereof. The Artemisia apiacea extract is prepared by waste Artemisia apiacea residue discarded during the production of Artemisinin or directly by Artemisia apiacea plant, and has inhibiting effect on pathogenic bacteria of microbe diseases of picked fruits and vegetables and especially has obvious bacteria inhibition in vitro of picked fruit germs such as orange green mould, banana and mango anthracnose, Peronophythora litchi, grape gray mould and the like. Moreover, under living body conditions, the Artemisia apiacea extract can prevent and control diseases of the picked fruits such as oranges, litchi, bananas, grapes and the like, has good effect on the harmless preservation of fruits and vegetables, and can be used as botanical bacteriostat or fruit and vegetable preservative. 本发明公开了一种青蒿提取物和制备方法及应用。所述青蒿提取物是利用生产青蒿素过程废弃的青蒿废渣或者直接由青蒿植株制备得到,其对多种果蔬采后微生物病害的病原菌具有抑制作用,尤其是对柑桔青绿霉菌、香蕉和芒果炭疽病、荔枝霜疫霉病、葡萄灰霉病等水果的采后病菌具有明显的离体抑菌效果,而且在活体条件下,能防治柑桔、荔枝、香蕉、葡萄等水果的采后病害,对果蔬的无害保鲜具有良好的作用,可作为植物源抑菌剂或果蔬保鲜剂使用。 一种青蒿提取物的应用 技术领域 本发明属于果蔬保鲜技术和农药技术领域,具体涉及一种植物提取物的制备和应用,特别是指一种青蒿提取物、其制备方法及其作为杀菌剂和保鲜剂在果蔬保鲜和采后病害防治中的应用。 背景技术 果蔬采后贮藏过程中因微生物病害引起的腐烂所造成的损失是十分巨大的,据资料报道,发达国家的果蔬在采收后因腐烂而造成的损失在15~24%左右,发展中国家由于缺乏冷藏设备及卫生条件较差,果蔬采后病害导致的损失可高达20~50%。 目前国内外应用于采后果蔬保鲜的化学防腐剂主要有施保功、扑海因(抑菌脲)、特克多、多菌灵、甲基托布津、仲丁胺等。然而,长期使用化学杀菌剂极易导致病菌产生抗药性,并导致这些杀菌剂的使用剂量不得不不断加大,从而也引发了食品安全、环境污染和有害生物抗药性等一系列严重问题。人们越来越担心食物链中化学杀菌剂残留对人类健康的影响,要求降低果蔬产品化学杀菌剂的使用量,许多生产上常用的杀菌剂已被许多国家明令禁止在果蔬保鲜中使用。因此,迫切要求研究者寻找新的保鲜药剂用于水果蔬菜的采后保鲜及控制采后果蔬的微生物病害。为了解决上述问题,人们加大了生物农药的开发力度,植物源农药作为生物农药的重要组成部分而倍受重视,国内外均有植物源农药的研发报道。 青蒿为菊科植物黄花蒿(Artemisia annua L.)的全草,别名香蒿、苦蒿、黄蒿。为一年生草本,高达1.5m,全株黄绿色,有臭气。茎直立,具纵条纹, 上部分枝。基部及下部叶在花期枯萎,中部叶卵形,长4.5~7cm,2~3回羽状深裂,小裂片线形,宽约0.3mm,先端尖锐,表面颜色深绿,背面颜色较浅,无毛或略具细微软毛,有柄;上部叶渐小,无柄。头状花序多数,球形,直径约2mm,具细软短梗,排成圆锥状;总苞片2~3层;花管状,雌花长约0.5mm,两性花长约1mm,黄色;花药先端尖尾状,基部圆钝;柱头2裂,裂片先端呈画笔状。瘦果椭圆形,长约0.6mm。花期8~10月,果期10~11月。多生于山坡、林缘、荒地,产于全国各地。青蒿含有抗炎、免疫抑制等生物活性物质,在我国被用于中草药,此外,还可用于提取香料;更重要的是能从青蒿中分离出青蒿素。青蒿素(Artemisinin)是我国药学工作者1971年从青蒿叶中提取分离到的一种具有过氧桥的倍半萜内酯类化合物,是我国自主开发的、国际公认的抗疟疾的特效药,是目前所知的所有抗疟疾药中起效最快,疗效最好,毒性最低的药物,对脑型疟疾和抗氯喹恶性疟疾疗效更为突出,更可贵的是目前青蒿素类药尚未见有关抗药性的报道。 中国是青蒿素的主要产地,占全球总产量的90%。据估计,中国青蒿的种植面积约有7000~8000公顷。考虑到野生青蒿的收获量,估计国内每年可收获青蒿鲜草2万吨左右,青蒿素的理论年产量可达100吨左右,2006年我国青蒿素产量约为78吨。青蒿素提取率约为0.6%左右,因此青蒿原料在提炼青蒿素后会留下大量废渣。以年生产30吨青蒿素计,需要青蒿药材达5000吨以上,留下的废渣以千吨计,均被废弃。 发明内容 本发明的一个目的是利用现有青蒿加工提取过程产生的大量废渣,提供一种从青蒿废渣中提取的提取物,变废为宝。 本发明的另一个目的是提供所述青蒿提取物的提取方法。 本发明还有一个目的是提供所述青蒿提取物的新应用,所述青蒿提取物也包括直接采用青蒿进行提取得到的提取物,而不仅仅包括采用青蒿废渣制备得到的提取物。 本发明的目的通过以下技术方案予以实现: 提供一种青蒿提取物,方案一是以青蒿废渣浸膏为原料,往浸膏中加入溶剂,浸提,过滤得到的滤液浓缩或干燥得到所述青蒿提取物。 所述青蒿废渣浸膏是将青蒿全株或切段后晒干、阴干或烘干,粉碎后采用体积比浓度为60~70%的乙醇浸提过夜,浸提液用于提取青蒿素,剩余部分即为废渣,浸提液在精提青蒿素过程中也得到部分废渣,将收集的各阶段产生的废渣浓缩成废渣浸膏即可,也可以混合各阶段收集到的废渣统一浓缩成浸膏,浓缩成浸膏的方法为现有常规方法;或者从青蒿素加工工厂直接购买被废弃的青蒿废渣浸膏。 所述溶剂可采用本领域常用的溶剂,优选水、乙醇、乙酸乙酯、石油醚或丙酮中任何一种或几种溶剂任意比例的混合物。 本发明所述青蒿提取物的提取方法具体包括以下步骤: (1)往青蒿废渣浸膏中加入硅胶,搅拌均匀,磨粉过筛得青蒿废渣浸膏粉; (2)往步骤(1)所得青蒿废渣浸膏粉中加入溶剂浸提后过滤; (3)将步骤(2)所得滤液经旋转浓缩或干燥得青蒿废渣提取物。 作为优选,步骤(3)得到的滤液经旋转浓缩后得青蒿废渣提取液,提取液再用溶剂进行液-液或液-固分部提取,提取液及其分部提取液经浓缩或干燥 得到青蒿废渣提纯后的提取物。液-液或液-固分部提取所采用的溶剂优选水、乙醇、乙酸乙酯、石油醚或丙酮中任何一种或几种溶剂任意比例的混合物,溶剂的加入量为(重量体积比)为1g∶0.5~2ml;提取24小时。 步骤(1)所述硅胶的加入量按照1000g青蒿废渣浸膏加入500~2000g硅胶的比例确定;所述过筛为过30~100目筛。 步骤(2)所述溶剂为水、乙醇、乙酸乙酯、石油醚或丙酮中任何一种或几种溶剂任意比例的混合物,加入的量按照废渣浸膏粉∶溶剂的重量体积比为1g∶0.5~2ml的比例确定。 步骤(2)所述浸提的时间为5~24小时,优选24小时。 步骤(3)所述旋转浓缩或干燥均为常规方法。 方案二,提供一种青蒿提取物,将青蒿全株或切段后晒干(阴干或烘干)粉碎后采用水或其他常规溶剂浸提得到。 本发明方案一和方案二所述青蒿提取物都具有新的应用价值,可以应用作为杀菌剂或果蔬保鲜剂。所述青蒿提取物包括采用青蒿废渣或青蒿植株提取得到的提取液、浓缩提取液或干燥得到的固体提取物。青蒿提取物的抗菌保鲜应用是由本发明首次提出的技术方案,并不仅仅限于青蒿废渣的再利用。青蒿整株植物也可提取抗菌保鲜物质,因为保鲜剂的经济价值远远低于青蒿素,青蒿主要用于提取青蒿素,不会仅用于提取保鲜剂。所以本发明不仅提供了青蒿提取物的新应用,更重要的是解决了青蒿素生产过程中产生的大量废渣的再利用。 本发明经过反复试验证明,从青蒿或青蒿废渣中提取得到的提取物对多种果蔬采后微生物病害的病原菌具有抑制作用,尤其是对柑桔青绿霉菌、香蕉和 芒果炭疽病、荔枝霜疫霉病、葡萄灰霉病等水果的采后病菌具有明显的离体抑菌效果,而且在活体条件下,能防治柑桔、荔枝、香蕉等水果的采后病害,对果蔬的无害保鲜具有良好的作用。因此,从青蒿废渣中得到的提取物可作为植物源抑菌剂或果蔬保鲜剂使用。 本发明的有益效果是: 本发明提供了一种新的植物源杀菌剂或果蔬保鲜剂,为抗菌和蔬果保鲜技术领域提供新的技术成果。 本发明采用适当的方法处理废弃的青蒿废渣后再进行溶剂提取制备得到有益的抗菌保鲜剂,变废为宝;同时解决了长期以来青蒿素加工企业处置废渣的难题,为推广青蒿素的生产加工应用扫平障碍。 本发明方法简单易行,原料易得且成本较低,具有优良的推广价值。 具体实施方式 下面结合具体实施例进一步详细说明本发明。 实施例1 青蒿提取物的制备 青蒿废渣浸膏来自广东省某青蒿素生产厂家,其他试剂均为市购常规产品。1000g青蒿浸膏加入1000g硅胶,搅拌均匀,磨成粉状,过60目筛,快速分离,得青蒿废渣浸膏粉;用1800mL石油醚(PE)浸提青蒿废渣浸膏粉24小时,过滤或离心得到PE部分的滤液732mL,经旋转浓缩后得PE部分提取物157.65g,所得青蒿浸膏提取物低温(1~7℃)贮存备用。 实施例2 青蒿提取物的制备 青蒿废渣浸膏来自广东省某青蒿素生产厂家,其他试剂均为市购常规产品。1000g青蒿浸膏加入1000g硅胶,搅拌均匀,磨成粉状,过60目筛。快 速分离得青蒿废渣浸膏粉;用1800mL的乙酸乙酯(EA)浸提青蒿废渣浸膏粉24小时,过滤或离心得到EA部分的滤液1468mL,经旋转浓缩后得EA部分提取物143.58g,低温贮存备用。 实施例3 青蒿提取物的制备 青蒿废渣浸膏来自广东省某青蒿素生产厂家,其他试剂均为市购常规产品。1000g青蒿浸膏加入1000g硅胶,搅拌均匀,磨成粉状,过60目筛。快速分离得青蒿废渣浸膏粉;用1800mL的PE∶EA=1∶1(体积比)的混合液浸提青蒿废渣浸膏粉24小时,过滤或离心得到混合液部分的滤液1392mL,经旋转浓缩后得混合液部分提取物223.4g,低温贮存备用。 实施例4 青蒿提取物的制备 青蒿废渣浸膏来自广东省某青蒿素生产厂家,其他试剂均为市购常规产品。1000g青蒿浸膏加入2000g硅胶,搅拌均匀,磨成粉状,过30目筛。快速分离得青蒿废渣浸膏粉;用1800mL的PE∶EA=1∶1(体积比)的混合液浸提青蒿废渣浸膏粉24小时,过滤或离心得到混合液部分的滤液1314mL,经旋转浓缩后得混合液部分提取物201.9g,低温贮存备用。 实施例5 青蒿提取物的制备 青蒿废渣浸膏来自广东省某青蒿素生产厂家,其他试剂均为市购常规产品。1000g青蒿浸膏加入1500g硅胶,搅拌均匀,磨成粉状,过80目筛,快速分离,得青蒿废渣浸膏粉;用1800mL石油醚(PE)浸提青蒿废渣浸膏粉24小时,过滤或离心得到PE部分的滤液685mL,经旋转浓缩后得PE部分提取物135.8g,所得青蒿浸膏提取物低温(1~7℃)贮存备用。 实施例6 青蒿提取物的制备 将青蒿全株或切段的青蒿晒干(阴干或烘干),粉碎,过40~60目筛。按粉碎物∶水=1∶5(重量/体积,g/ml)的比例,提取24小时,过滤或离心得到提取液,再经常规真空浓缩得浓缩液或干燥的固体提取物,低温贮存备用。 实施例7 青蒿提取物的制备 将青蒿全株或切段的青蒿晒干(阴干或烘干),粉碎,过40~60目筛。按粉碎物∶60%的乙醇=1∶1(重量/体积,g/ml)的比例,提取24小时,过滤或离心得到提取液,再经常规真空浓缩得浓缩液或干燥的固体提取物,低温贮存备用。 实施例8 青蒿废渣不同溶剂或方法提取物对水果采后病原菌的抑制作用试验 采用改进的生长速率法,称取本发明不同溶剂的青蒿提取物与NYDA培养基混合均匀,制成浓度为500μg青蒿提取物/mL培养基、1000μg青蒿提取物/mL培养基和2000μg青蒿提取物/mL培养基的带药培养基,将带药培养基倒入直径为9cm的培养皿中,冷却后,在培养皿中央接种5mm沙糖桔指状青霉菌(Penicillium digitatum)或香蕉炭疽病菌(Collectotrichun musae)菌片(所述菌种来自华南农业大学植物病理系实验室,也可以采用常规方法分离得到),以纯NYDA培养基平板作为空白对照。置于28℃恒温箱中培养,测量记录菌落大小(用十字交叉法测定菌落直径),计算抑菌率。 抑菌率(%)=(对照菌落直径-处理菌落直径)/对照菌落直径×100 实验结果见表1和表2。从表1中可以发现,本发明青蒿不同溶剂提取物对沙糖桔指状青霉的生长均有一定的抑制作用,以乙酸乙酯提取物的抑菌效果最强。从表2中可以发现,与抑制沙糖桔指状青霉菌效果相比,本发明青蒿提取物对香蕉炭疽病菌的抑菌效果更好,也以乙酸乙酯提取物的抑菌效 果最强。 表1 青蒿废渣不同溶剂提取物对采后沙糖桔指状青霉病菌抑菌效果 表2 青蒿废渣不同溶剂提取物对采后香蕉炭疽病菌抑菌效果 实施例9 本发明青蒿提取物对沙糖桔采后绿霉病的抑制效果试验 将本发明青蒿提取物加水配成1000μg/mL溶液,沙糖桔在所述溶液中浸泡5min后晾干,用灭菌铁钉在果实近果蒂部区域对称打2个孔(深3mm,直径2mm),10μl 1.5×104CFU/mL指状青霉孢子菌悬液。待自然晾干后放到塑料筐内于20℃,85%~95%RH条件下贮藏22天,统计病情指数和防治效果。试验设空白对照,并以水果保鲜方面常用的施保功(Sporgon)作为商品杀菌剂对照。实验结果见表3。从表3可以看出,本发明青蒿提取物对沙糖桔绿霉病(由指状青霉菌引起)均有明显效果,但比商品杀菌剂施保功效果略差。 表3 本发明青蒿提取物对沙糖桔指状青霉的活体抑制效果 (20℃、90%RH下培养22天) 实施例10 本发明青蒿提取物对荔枝采后病害的控制效果试验 荔枝采后病害主要是霜疫霉病。霜疫霉病是目前荔枝果实上最严重的病害,在荔枝采后贮运期间造成严重损失,使荔枝保鲜与销售企业蒙受重大损失。目前主要依靠化学杀菌剂如瑞毒霉、乙磷铝、特克多、施保功等控制。将本发明青蒿废渣的乙酸乙酯提取物加水配成1000μg/mL溶液,采收后的荔枝果实(品种:淮枝)在所述溶液中浸泡5min,待自然晾干后放到塑料筐内于20℃,85%~95%RH条件下贮藏7天,统计病情指数和防治效果。试验设空白对照,并以荔枝果实保鲜最常用的施保功(Sporgon)作为商品杀菌剂对照。试验结果见表4。从表4可以看出,本发明青蒿废渣的乙酸乙酯提取物对荔枝果实采后病害(主要由由霜疫霉菌引起)有明显效果,但比商品杀菌剂施保功效果略差。 表4 本发明青蒿提取物对荔枝采后病害的控制效果 (20℃、90%RH下培养22天) 实施例11 本发明青蒿提取物对葡萄灰霉病抑制效果试验 葡萄采后病害主要是灰霉病,90%以上的葡萄果实腐烂是由于该病的爆发引起在葡萄采后贮运期间造成严重损失。目前主要依靠二氧化硫熏蒸或二氧化硫释放纸进行控制,但二氧化硫处理容易导致葡萄表面漂白,特别是果蒂部分漂白严重;另外,也容易导致二氧化硫残留超标。将本发明青蒿废渣的乙酸乙酯提取物加水配成1000μg/mL溶液,采收后的葡萄果实(品种:巨峰)在所述溶液中浸泡5min,待自然晾干后放到塑料筐内于20℃,85%~95%RH条件下贮藏20天,统计病情指数和防治效果。试验设空白对照。试验结果见表5。从表5可以看出,本发明青蒿废渣的乙酸乙酯提取物对巨峰葡萄果实采后病害(主要由由灰霉菌引起)有明显效果。 表4 本发明青蒿提取物对葡萄采后病害的控制效果 (20℃、90%RH下培养20天) 1.一种青蒿提取物的应用,其特征在于应用于制备防治柑桔青绿霉菌、香蕉炭疽病或荔枝霜疫霉病的制剂,所述青蒿提取物是采用青蒿废渣浸膏为原料,加入溶剂浸提,过滤得到的滤液再经浓缩或干燥得到,所述溶剂为水、乙醇、乙酸乙酯、石油醚或丙酮中任何一种或几种溶剂任意比例的混合物;所述青蒿废渣浸膏是将青蒿全株或切段后晒干、阴干或烘干,粉碎后采用体积比浓度为60-70%的乙醇浸提过夜,浸提液用于提取青蒿素,剩余部分即为废渣,浸提液在精提青蒿素过程中也得到废渣,将收集的各阶段产生的废渣浓缩得到废渣浸膏。 2.根据权利要求1所述青蒿提取物的应用,其特征在于所述青蒿提取物的制备方法包括以下步骤: (1)向青蒿废渣浸膏中加入硅胶,搅拌均匀,磨粉过筛得青蒿废渣浸膏粉; (2)向步骤(1)所得青蒿废渣浸膏粉中加入溶剂浸提后过滤得滤液,所述溶剂为水、乙醇、乙酸乙酯、石油醚或丙酮中任何一种或几种溶剂任意比例的混合物; (3)将步骤(2)所得滤液经旋转浓缩或干燥得青蒿废渣提取物。 3.根据权利要求2所述青蒿提取物的应用,其特征在于步骤(1)所述硅胶的加入量按照1000g青蒿废渣浸膏加入500~2000g硅胶的比例确定;所述过筛为过30~100目筛。 4.根据权利要求2所述青蒿提取物的应用,其特征在于步骤(2)所述溶剂加入的量按照废渣浸膏粉:溶剂的重量体积比为1g∶0.5~2ml的比例确定。 5.根据权利要求2所述青蒿提取物的应用,其特征在于步骤(2)所述浸提的时间为24小时。 https://patents.google.com/patent/CN101773238B/zh?oq=CN101773238B一种青蒿素的应用----制备方法以及作为杀菌剂和保鲜剂在果蔬保鲜利用和采后病害防治中的应用 The invention discloses Artemisia apiacea extract and a preparation method and application thereof. The Artemisia apiacea extract is prepared by waste Artemisia apiacea residue discarded during the production of Artemisinin or directly by Artemisia apiacea plant, and has inhibiting effect on pathogenic bacteria of microbe diseases of picked fruits and vegetables and especially has obvious bacteria inhibition in vitro of picked fruit germs such as orange green mould, banana and mango anthracnose, Peronophythora litchi, grape gray mould and the like. Moreover, under living body conditions, the Artemisia apiacea extract can prevent and control diseases of the picked fruits such as oranges, litchi, bananas, grapes and the like, has good effect on the harmless preservation of fruits and vegetables, and can be used as botanical bacteriostat or fruit and vegetable preservative. A kind of application of Artemisinin Technical field The invention belongs to postharvest technology of fruits and vegetables and technical field of pesticide; Be specifically related to a kind of preparation and application of plant extracts, be meant a kind of Artemisinin, its preparation method especially and as bactericide and the application of antistaling agent in preserving fruit and vegetable utilizing and postharvest disease control. Background technology The loss of rotting to be caused that causes because of microorganism disease in the postharvest fruit and vegetable storage is very huge; According to document announcement; The fruits and vegetables of developed country are lost in about 15~24% because of what rot to cause in the back of gathering; Developing country is because shortage refrigerating equipment and sanitary condition are relatively poor, and the loss that postharvest disease of fruits and vegetables causes can be up to 20~50%. Domestic and international application mainly contains sporgon, iprodione (antibacterial urea), Te Keduo, carbendazim, thiophanate methyl, sec-butylamine etc. in the chemical preservative of adopting the back preserving fruit and vegetable utilizing at present.Yet, use chemical bactericide very easily to cause germ to develop immunity to drugs for a long time, and cause the using dosage of these bactericide to have to continue to increase, thereby also caused food security, a series of serious problems such as environmental pollution and harmful organism resistance to the action of a drug.People more and more is worried the residual influence to human health of chemical bactericide in the food chain, requires to reduce the use amount of fruit and vegetable food chemical bactericide, and bactericide commonly used has been prohibited in preserving fruit and vegetable utilizing by many countries and used in many productions.Therefore, an urgent demand researcher seeks the microorganism disease that the back fruits and vegetables are adopted in post-harvest fresh-keeping and control that new storage chemical is used for fruit and vegetable.In order to address the above problem, people have strengthened the exploitation dynamics of biological pesticide, and botanical pesticide is extremely paid attention to as the important component part of biological pesticide, and the research and development report of botanical pesticide is all arranged both at home and abroad. Sweet wormwood is the herb of feverfew artemisia annua (Artemisia annua L.), another name abrotanum, Artemisia santonica, yellow wormwood artemisia.Be annual herb, up to 1.5m, the complete stool yellow green has foul smell.Stem is upright, tool vertical stripe, top branch.Base portion and bottom leaf are withered at the florescence, and middle leaf is avette, long 4.5~7cm, and 2~3 times pinniform drastic cracks, lobelet is linear, wide about 0.3mm, tip is sharp-pointed, and surface color is dark green, and back side lighter color does not have hair or omits the trickle fur of tool, and handle is arranged; Upper leaf is gradually little, stockless.Capitulum is most, sphere, and the about 2mm of diameter, the soft short stalk of tool is lined up coniform; 2~3 layers of phyllarys; The floral tube shape, female flower is about 0.5mm, and hermaphrodite flower is about 1mm, yellow; Flower pesticide tip fine stern shape, the base portion circle is blunt; Column cap 2 splits, and the sliver tip is penicillate.Achene is oval, is about 0.6mm.8~October of florescence, really 10~November of phase.Be born in hillside, border, wasteland more, originate in all parts of the country.Sweet wormwood contains bioactivators such as anti-inflammatory, immunosupress, is used to Chinese herbal medicine in China, in addition, also can be used for extracting spices; The more important thing is and from sweet wormwood, to isolate qinghaosu.Qinghaosu (Artemisinin) is that China pharmacy worker extracted a kind of sesquiterpene lactone compounds with peroxide bridge that is separated in 1971 from artemisia leaf; Be China's independent development, internationally recognized antimalarial specific drug; Be that onset is the fastest in all Anti-Malarials known today, curative effect is best, the medicine that toxicity is minimum; More outstanding to encephalic malaria and anti-chloroquine pernicious malaria curative effect, more valuable is that present arteannuin medicine is not seen relevant drug-fast report as yet. China is the main place of production of qinghaosu, accounts for 90% of global total output.According to estimates, the cultivated area of Chinese wormwood has 7000~8000 hectares approximately.Consider the harvest yield of wild sweet wormwood, estimate can gather in the crops domestic every year about 20,000 tons of the bright grass of sweet wormwood, the theoretical annual production of qinghaosu can reach about 100 tons, and China's qinghaosu output was about 78 tons in 2006.The qinghaosu recovery rate is about about 0.6%, so the sweet wormwood raw material can stay a large amount of waste residues after refining qinghaosu.Year to produce 30 tons of qinghaosus, need the sweet wormwood medicinal material to reach more than 5000 tons, the waste residue that stays all goes out of use in kiloton. Summary of the invention An object of the present invention is to utilize a large amount of waste residues of existing sweet wormwood processing leaching process generation, a kind of extract that from the sweet wormwood waste residue, extracts is provided, turn waste into wealth. Another object of the present invention provides the method for distilling of said Artemisinin. A further object of the invention provides the new application of said Artemisinin, and said Artemisinin comprises that also direct employing sweet wormwood extracts the extract that obtains, and not only comprises the extract that adopts the sweet wormwood waste residue to prepare. The object of the invention is achieved through following technical scheme: A kind of Artemisinin is provided, and scheme one is to be raw material with sweet wormwood waste residue medicinal extract, in medicinal extract, adds solvent, and lixiviate filters that the filtrating that obtains concentrates or drying obtains said Artemisinin. Said sweet wormwood waste residue medicinal extract is with drying, dry in the shade or dry after sweet wormwood complete stool or the segment; Pulverizing the back, to adopt volume by volume concentration be that 60~70% ethanol lixiviate is spent the night, and leaching liquor is used to extract qinghaosu, and remainder is waste residue; Leaching liquor is carried in essence and is also obtained the part waste residue in the qinghaosu process; The waste residue that each stage of collecting is produced is condensed into waste residue medicinal extract and get final product, also can mix the waste residue unification that each stage collects and be condensed into medicinal extract, and the method that is condensed into medicinal extract is for having conventional method now; Perhaps directly buy the sweet wormwood waste residue medicinal extract that goes out of use from qinghaosu processing factory. Said solvent can adopt this area solvent commonly used, the mixture of any or several kinds of solvent arbitrary proportions in preferred water, ethanol, ethyl acetate, benzinum or the acetone. The method for distilling of Artemisinin according to the invention specifically may further comprise the steps: (1) in sweet wormwood waste residue medicinal extract, add silica gel, stir, abrasive dust sieve sweet wormwood waste residue extract powder; (2) filter after adding the solvent lixiviate in step (1) the gained sweet wormwood waste residue extract powder; (3) step (2) gained filtrating is concentrated or the dry sweet wormwood waste residue extract that gets through rotation. As preferably, the filtrating that step (3) obtains through rotate after concentrating sweet wormwood waste residue extract, extract carries out liquid-liquid with solvent again or liquid-solid branch extracts, extract and branch's extract thereof are through concentrating or dry extract after obtaining the purification of sweet wormwood waste residue.Liquid-liquid or liquid-solid branch extract the mixture of any or several kinds of solvent arbitrary proportions in solvent preferred water, ethanol, ethyl acetate, benzinum or the acetone that is adopted, and the addition of solvent be that (w/v) is 1g: 0.5~2ml; Extracted 24 hours. The addition of the said silica gel of step (1) is confirmed according to the ratio that 1000g sweet wormwood waste residue medicinal extract adds 500~2000g silica gel; Said sieving to crossing 30~100 mesh sieves. The said solvent of step (2) is the mixture of any or several kinds of solvent arbitrary proportions in water, ethanol, ethyl acetate, benzinum or the acetone, and the amount of adding is according to the waste residue extract powder: the w/v of solvent is that the ratio of 1g: 0.5~2ml is confirmed. The time of the said lixiviate of step (2) is 5~24 hours, preferred 24 hours. The said rotation of step (3) concentrates or drying is conventional method. Scheme two provides a kind of Artemisinin, pulverizes the back and adopts water or other conventional solvent lixiviates to obtain drying (dry in the shade or dry) after sweet wormwood complete stool or the segment. The present invention program one has new using value with scheme two said Artemisinins, can use as bactericide or fruit and vegetable fresh-keeping agent.Said Artemisinin comprises extract, concentrated extracting solution or the dry solid extract that obtains that adopts sweet wormwood waste residue or the extraction of sweet wormwood plant to obtain.It is the technical scheme that is proposed first by the present invention that the anti-bacterial refreshing of Artemisinin is used, and is not restricted to the utilization again of sweet wormwood waste residue.The whole strain plant of sweet wormwood also can be extracted the anti-bacterial refreshing material, because the economic worth of antistaling agent is well below qinghaosu, sweet wormwood is mainly used in the extraction qinghaosu, can only not be used to extract antistaling agent.So the present invention not only provides the new application of Artemisinin, the more important thing is the utilization again that has solved a large amount of waste residues that produce in the qinghaosu production process. The present invention proves through repetition test; It is inhibited to the pathogen of multiple postharvest fruit and vegetable microorganism disease from sweet wormwood or sweet wormwood waste residue, to extract the extract that obtains; Especially the back germ of adopting to fruit such as citrus penicillium, banana and mango anthracnose, peronophythora litchi, grape grey moulds has the fungistatic effect that significantly exsomatizes; And under condition of living body, can prevent and treat the postharvest disease of fruit such as citrus, lichee, banana, fruits and vegetables harmless fresh-keeping had good effect.Therefore, the extract that from the sweet wormwood waste residue, obtains can be used as plant-based bacteriostat or fruit and vegetable fresh-keeping agent uses. The invention has the beneficial effects as follows: The invention provides a kind of new botanical fungicide or fruit and vegetable fresh-keeping agent, with vegetables and fruits preservation technique field new technological achievement is provided for antibiotic. Carry out solvent extraction again behind the sweet wormwood waste residue that the present invention adopts the appropriate method processing to discard and prepare useful antimicrobial antistaling agent, turn waste into wealth; Solved the difficult problem of the waste residue of qinghaosu processing enterprise disposal for a long time simultaneously, used for the production and processing of promoting qinghaosu and put down obstacle. The inventive method is simple, raw material be easy to get and cost lower, have good promotional value. The specific embodiment Below in conjunction with specific embodiment further explain the present invention. The preparation of embodiment 1 Artemisinin Sweet wormwood waste residue medicinal extract is from Guangdong Province's qinghaosu manufacturer, and other reagent are commercial conventional products.1000g sweet wormwood medicinal extract adds 1000g silica gel, stirs, and pulverize is crossed 60 mesh sieves, separates fast, gets sweet wormwood waste residue extract powder; With 1800mL benzinum (PE) lixiviate sweet wormwood waste residue extract powder 24 hours, filter or the centrifugal filtrating 732mL that obtains the PE part, after rotation concentrates PE extracting section thing 157.65g, gained sweet wormwood medicinal extract extract low temperature (1~7 ℃) storage is subsequent use. The preparation of embodiment 2 Artemisinins Sweet wormwood waste residue medicinal extract is from Guangdong Province's qinghaosu manufacturer, and other reagent are commercial conventional products.1000g sweet wormwood medicinal extract adds 1000g silica gel, stirs, and pulverize is crossed 60 mesh sieves.Separate fast sweet wormwood waste residue extract powder; With ethyl acetate (EA) the lixiviate sweet wormwood waste residue extract powder of 1800mL 24 hours, filter or the centrifugal filtrating 1468mL that obtains the EA part,, rotation gets EA extracting section thing 143.58g after concentrating, and low tempertaure storage is subsequent use. The preparation of embodiment 3 Artemisinins Sweet wormwood waste residue medicinal extract is from Guangdong Province's qinghaosu manufacturer, and other reagent are commercial conventional products.1000g sweet wormwood medicinal extract adds 1000g silica gel, stirs, and pulverize is crossed 60 mesh sieves.Separate fast sweet wormwood waste residue extract powder; PE with 1800mL: EA=1: the mixed liquor lixiviate sweet wormwood waste residue extract powder of 1 (volume ratio) 24 hours, filter or the centrifugal filtrating 1392mL that obtains the mixed liquor part,, rotation gets mixed liquor extracting section thing 223.4g after concentrating, and low tempertaure storage is subsequent use. The preparation of embodiment 4 Artemisinins Sweet wormwood waste residue medicinal extract is from Guangdong Province's qinghaosu manufacturer, and other reagent are commercial conventional products.1000g sweet wormwood medicinal extract adds 2000g silica gel, stirs, and pulverize is crossed 30 mesh sieves.Separate fast sweet wormwood waste residue extract powder; PE with 1800mL: EA=1: the mixed liquor lixiviate sweet wormwood waste residue extract powder of 1 (volume ratio) 24 hours, filter or the centrifugal filtrating 1314mL that obtains the mixed liquor part,, rotation gets mixed liquor extracting section thing 201.9g after concentrating, and low tempertaure storage is subsequent use. The preparation of embodiment 5 Artemisinins Sweet wormwood waste residue medicinal extract is from Guangdong Province's qinghaosu manufacturer, and other reagent are commercial conventional products.1000g sweet wormwood medicinal extract adds 1500g silica gel, stirs, and pulverize is crossed 80 mesh sieves, separates fast, gets sweet wormwood waste residue extract powder; With 1800mL benzinum (PE) lixiviate sweet wormwood waste residue extract powder 24 hours, filter or the centrifugal filtrating 685mL that obtains the PE part, after rotation concentrates PE extracting section thing 135.8g, gained sweet wormwood medicinal extract extract low temperature (1~7 ℃) storage is subsequent use. The preparation of embodiment 6 Artemisinins The sweet wormwood of sweet wormwood complete stool or segment is dried (dry in the shade or dry), pulverize, cross 40~60 mesh sieves.Press crushed material: water=1: 5 (weight per volume, ratio g/ml) were extracted 24 hours, filter or the centrifugal extract that obtains, again through conventional vacuum concentrate concentrate or dry solid extract, low tempertaure storage is subsequent use. The preparation of embodiment 7 Artemisinins The sweet wormwood of sweet wormwood complete stool or segment is dried (dry in the shade or dry), pulverize, cross 40~60 mesh sieves.Press crushed material: 60% ethanol=1: 1 (weight per volume, ratio g/ml) were extracted 24 hours, filter or the centrifugal extract that obtains, again through conventional vacuum concentrate concentrate or dry solid extract, low tempertaure storage is subsequent use. Embodiment 8 sweet wormwood waste residue different solvents or method extract are adopted the inhibitory action test of back pathogen to fruit Adopt improved growth rate method; The Artemisinin and the NYDA culture medium that take by weighing different solvents of the present invention mix; Processing concentration is the band medicine culture medium of 500 μ g Artemisinin/mL culture mediums, 1000 μ g Artemisinin/mL culture mediums and 2000 μ g Artemisinin/mL culture mediums; Band medicine culture medium is poured in the culture dish that diameter is 9cm; After the cooling; At culture dish central authorities inoculation 5mm granulated sugar tangerine penicillium digitatum (Penicillium digitatum) or Glorosprium musarum Cookeet Mass (Collectotrichun musae) bacterium sheet (said bacterial classification also can adopt conventional method to separate and obtain from department of plant pathology of Agricultural University Of South China laboratory), with pure NYDA culture medium flat plate as blank.Place 28 ℃ of insulating boxs to cultivate, surveying record bacterium colony size (measuring colony diameter with the right-angled intersection method) is calculated bacteriostasis rate. Bacteriostasis rate (%)=(contrast colony diameter-processing colony diameter)/contrast colony diameter * 100 Experimental result is seen table 1 and table 2.Can find that from table 1 sweet wormwood different solvents extract of the present invention all has certain inhibitory action to the growth of granulated sugar tangerine Penicillium digitatum, and is the strongest with the fungistatic effect of ethyl acetate extract.Can find that from table 2 with inhibition granulated sugar tangerine penicillium digitatum comparison, Artemisinin of the present invention is better to the fungistatic effect of Glorosprium musarum Cookeet Mass, also the fungistatic effect with ethyl acetate extract is the strongest. Table 1 sweet wormwood waste residue different solvents extract is to adopting back granulated sugar tangerine Penicillium digitatum germ fungistatic effect Table 2 sweet wormwood waste residue different solvents extract is to adopting back Glorosprium musarum Cookeet Mass fungistatic effect Embodiment 9 Artemisinins of the present invention are adopted the inhibition effect test of back green mould to the granulated sugar tangerine Artemisinin of the present invention is added water be made into 1000 μ g/mL solution; The granulated sugar tangerine is dried after in said solution, soaking 5min; Make a call to 2 holes (dark 3mm, diameter 2mm), 10 μ l, 1.5 * 104CFU/mL Penicillium digitatum pityrosporion ovale suspension with the sterilization iron nail in the nearly base of fruit of fruit portion zone symmetry.It is interior in 20 ℃ to treat to be put into after nature dries plastic crate, and storage is 22 days under 85%~95%RH condition, statistics disease index and prevention effect.Blank is established in test, and contrasts as the commodity bactericide with fruit freshness preserving aspect sporgon (Sporgon) commonly used.Experimental result is seen table 3.Can find out that from table 3 Artemisinin of the present invention all has positive effect to granulated sugar tangerine green mould (being caused by penicillium digitatum), but slightly poorer than commodity bactericide sporgon effect. Table 3 Artemisinin of the present invention suppresses effect to the live body of granulated sugar tangerine Penicillium digitatum (20 ℃, 90%RH were cultivated 22 days down) Embodiment 10 Artemisinins of the present invention are to the control effect test of lichee postharvest disease The lichee postharvest disease mainly is a peronospora tabacina.Peronospora tabacina is severe diseases on the present litchi fruits, during lichee is adopted the back storing, causes heavy losses, and lichee fresh-keeping and sale enterprise are sustained a great loss.Controls such as main at present dependence chemical bactericide such as metalaxyl, aliette, Te Keduo, sporgon.The ethyl acetate extract of sweet wormwood waste residue of the present invention is added water be made into 1000 μ g/mL solution; Litchi fruits after gathering (kind: the Huaihe River branch) in said solution, soak 5min; Treat to be put into after nature dries in the plastic crate in 20 ℃; Storage is 7 days under 85%~95%RH condition, statistics disease index and prevention effect.Blank is established in test, and contrasts as the commodity bactericide with the fresh-keeping the most frequently used sporgon of litchi fruits (Sporgon).Result of the test is seen table 4.Can find out that from table 4 ethyl acetate extract of sweet wormwood waste residue of the present invention has positive effect to litchi fruits postharvest disease (mainly by being caused by white phytophthora), but slightly poorer than commodity bactericide sporgon effect. Table 4 Artemisinin of the present invention is to the control effect of lichee postharvest disease (20 ℃, 90%RH were cultivated 22 days down) Embodiment 11 Artemisinins of the present invention suppress effect test to grape grey mould The grape postharvest disease mainly is a gray mold, and it is to cause heavy losses during the storing because the outburst of this disease causes after the grape harvest that the grape fruit more than 90% rots.Main at present dependence sulfur dioxide is fumigated or sulfur dioxide release paper is controlled, but sulfur dioxide treatment causes the grape superficial bleaching easily, and particularly the base of fruit partial bleaching is serious; In addition, also cause sulfur dioxide residue to exceed standard easily.The ethyl acetate extract of sweet wormwood waste residue of the present invention is added water be made into 1000 μ g/mL solution; Grape fruit after gathering (kind: huge peak) in said solution, soak 5min; Treat to be put into after nature dries in the plastic crate in 20 ℃; Storage is 20 days under 85%~95%RH condition, statistics disease index and prevention effect.Blank is established in test.Result of the test is seen table 5.Can find out that from table 5 ethyl acetate extract of sweet wormwood waste residue of the present invention has positive effect to kyoto grape postharvest diseases of fruit (mainly by being caused by botrytis cinerea). Table 4 Artemisinin of the present invention is to the control effect of grape postharvest disease (20 ℃, 90%RH were cultivated 20 days down) 1. the application of an Artemisinin; It is characterized in that being applied to prepare the preparation of control citrus penicillium, banana anthracnose or peronophythora litchi; Said Artemisinin is that employing sweet wormwood waste residue medicinal extract is raw material; Add the solvent lixiviate, filter the filtrating that obtains again through concentrating or drying obtains, said solvent is the mixture of any or several kinds of solvent arbitrary proportions in water, ethanol, ethyl acetate, benzinum or the acetone; Said sweet wormwood waste residue medicinal extract is with drying, dry in the shade or dry after sweet wormwood complete stool or the segment; Employing volume by volume concentration in pulverizing back is that the ethanol lixiviate of 60-70% is spent the night; Leaching liquor is used to extract qinghaosu; Remainder is waste residue, and leaching liquor is carried in essence and also obtained waste residue in the qinghaosu process, with the concentrated waste residue medicinal extract that obtains of the waste residue of each stage generation of collecting. 2. according to the application of the said Artemisinin of claim 1, it is characterized in that the preparation method of said Artemisinin may further comprise the steps: (1) in sweet wormwood waste residue medicinal extract, add silica gel, stir, abrasive dust sieve sweet wormwood waste residue extract powder; (2) in step (1) gained sweet wormwood waste residue extract powder, add to cross after the solvent lixiviate and filter filtrating, said solvent is the mixture of any or several kinds of solvent arbitrary proportions in water, ethanol, ethyl acetate, benzinum or the acetone; (3) step (2) gained filtrating is concentrated or the dry sweet wormwood waste residue extract that gets through rotation. 3. according to the application of the said Artemisinin of claim 2, it is characterized in that the addition of the said silica gel of step (1) is confirmed according to the ratio that 1000g sweet wormwood waste residue medicinal extract adds 500~2000g silica gel; Said sieving to crossing 30~100 mesh sieves. 4. according to the application of the said Artemisinin of claim 2, it is characterized in that amount that the said solvent of step (2) adds is according to the waste residue extract powder: the w/v of solvent is that the ratio of 1g: 0.5~2ml is confirmed. 5. according to the application of the said Artemisinin of claim 2, it is characterized in that the time of the said lixiviate of step (2) is 24 hours. https://patents.google.com/patent/CN101773238B/en?oq=CN101773238B一种青蒿素的应用----制备方法以及作为杀菌剂和保鲜剂在果蔬保鲜利用和采后病害防治中的应用
  9. The present invention discloses the new use of available antimalarial arteannuin and its derivatives dihydro arteannuin, antiannuic methyl ether, antiannuic ethyl ether and antiannuic amber. Arteannuin and its derivatives are used through combination with antibiotic medicine to inhibit bacterial growth and enhance the antibiotic effect of available antibiotic medicine. Especially, antiannuic amber is applied in preventing and treating bacterial infection diseases except being used as antimalarial. 本发明公开了一种现有的抗疟药青蒿素及其衍生物二氢青蒿素、蒿甲醚、蒿乙醚、青蒿琥酯新的用途,即,与抗菌药物联合应用抑制细菌生长,增强抗菌药物抗菌效力。尤其是使得青蒿琥酯除作为治疗疟疾药物外,能够在预防及治疗细菌感染性疾病中得到应用。 青蒿素及其衍生物与抗菌药物的联合应用 技术领域 本发明涉及药物应用领域,尤其涉及青蒿素及其衍生物的抗菌应用。 背景技术 感染性疾病是由于病原微生物侵入体内,在体内大量地繁殖所引起的疾病,是临床病人死亡的重要原因之一。造成患者死亡的原因其一是细菌大量繁殖释放毒素导致组织器官的损伤;其二是细菌成分通过激活免疫细胞,诱导TNF-α、IL-1、IL-6、NO等致炎细胞因子释放,引起多种炎症介质所形成的瀑布效应,可使炎症反应扩大甚至失去控制,最终导致以细胞自身性破坏为特征的全身性炎症即脓毒症的发生。细菌感染曾是人类第一死因,抗生素的发明带给了人类希望之光。但抗生素的普遍使用使越来越多的细菌产生了耐药性,如果不解决抗生素滥用的问题,等待人类的将是下一次黑暗。 青蒿提取物用来治疗疟疾相关的发热已经有一千多年的历史,除传统的抗疟作用外,青蒿类物质还具有其它方面的作用,如平喘、抗癌、抗血吸虫及对免疫系统的调节等。国内外有关青蒿素及其衍生物的研究很多,但研究范围主要集中在抗疟、抗癌、抗血吸虫的作用及其机制上。 青蒿琥酯(Artesunate),化学名为二氢青蒿素-1,2-α-琥珀酸单酯,分子式为C19O8 H28,分子量384,是具有倍半萜结构的抗疟药青蒿素的衍生物。青蒿琥酯是新型的抗疟药,较之青蒿素,青蒿琥酯可配制成任何常规的制剂形式,包括固体和液体形式,给药非常方便。青蒿琥酯作为抗疟药,不但效价高,而且不易产生耐受性。 对青蒿素及其衍生物的抗炎作用已有报道,研究发现青蒿素可抑制LPS/TNF-α诱导性NO合酶的合成及NF-κB的激活;青蒿琥酯对LPS及合并干扰素刺激小鼠腹腔巨噬细胞NO的合成有明显的抑制作用;青蒿琥酯对LPS刺激的小鼠巨噬细胞RAW264.7也具有相似的保护作用;谭余庆等还发现青蒿提取物、青蒿素可降低内毒素休克小鼠LPO、ACP、内毒素、TNF-α、P450浓度,升高SOD活性,降低小鼠死亡率,延长小鼠的平均生存时间,对小鼠肝、肺组织形态也有一定的保护作用;王俊等发现青蒿素能抑制CpGODN、LPS、热灭活的EC诱导细胞释放炎症因子,并对脓毒症模型小鼠具有显著保护作用。但是青蒿琥酯是否具有抗菌作用,是否能改善细菌对抗生素的耐药,增强抗生素的抗菌效力和对细菌导致的感染是否有效,未见报告。我国是青蒿素和其衍生物的主要生产地,由于该药物疗效高、毒性低,WHO已经将青蒿素列为世界治疗疟疾的主要药物,我国正在扩大青蒿种植和青蒿素生产,因此扩宽青蒿素类衍生物的适应症,将其应用于细菌感染疾病的防治,无疑对提高青蒿素类衍生物的利用、增加其经济价值并改善抗生素滥用所导致的耐药现象具有重要意义。 发明内容 本发明的目的在于将青蒿素及其衍生物,特别是青蒿琥酯应用到抗菌领域,不仅拓宽青蒿素及其衍生物,特别是青蒿琥酯的用途,而且可以减轻现有抗菌药物的滥用及其导致的耐药现象。 基于上述目的,本发明使用的技术方案是将青蒿素及其衍生物中的一种与抗菌药物中的一种进行联合应用,该应用不是做为疾病的治疗方法的应用,而是在配备治疗疾病(抗菌)的药物中的应用,其中上述两种成分的比例为1∶1~4096∶1,上两种成分可以制备成组合物,也可以是两种单独的药剂。上述两种成分能够产生协同抗菌效力,能够降低抗菌药物的最低抑菌浓度(MIC)和最低杀菌浓度(MBC)。 上述菌包括革兰氏阳性菌和革兰氏阴性菌;上述衍生物包括二氢青蒿素、蒿甲醚、蒿乙醚和青蒿琥酯;上述抗菌药物包括β-内酰胺类抗生素(青霉素类、头孢菌素类、非典型β-内酰胺类抗生素)、氨基糖苷类、克林霉素类、喹诺酮类的药物,在一较佳实施例中,上述抗菌药物为青霉素G、氨苄西林、头孢呋辛、头孢匹胺、庆大霉素、克林霉素、洛美沙星、加替沙星、舒氨西林和泰能,优选为庆大霉素、舒氨西林、头孢匹胺和加替沙星。 上述应用优选为青蒿琥酯与抗菌药物的联合应用。 青蒿琥酯单独使用可抑制革兰阴性菌和革兰阳性菌的生长,与抗菌药物联合应用后可明显增强抗菌药物的抗菌效力,减少抗菌药物的使用剂量;实验表明青蒿素及青蒿琥酯与抗菌药物联合应用可明显降低细菌(包括革兰阴性、阳性菌)攻击小鼠死亡率,显著抑制小鼠血清LPS含量和细胞因子TNF-α释放。 本发现人对青蒿琥酯体内外的抗菌和抗炎作用进行了研究。 首先,本研究人员对本青蒿琥酯的体外抗菌作用进行了药理学分析,并与抗生素联合应用后发现青蒿琥酯单独使用可抑制革兰阴性菌和革兰阳性菌的生长,与抗生素联合应用后可明显增强抗生素的抗菌效力,减少抗生素的使用剂量;其次,青蒿琥酯与抗生素联合应用和可明显降低细菌(包括革兰阴性、阳性菌)攻击小鼠死亡率,显著抑制小鼠血清LPS含量和细胞因子TNF-α释放。 因此本发明拓宽了青蒿素及其衍生物,特别是青蒿琥酯的用途,提高了抗菌药物的抗菌效力,减轻了抗菌药物滥用和导致的耐药现象。 附图说明 图1表示青蒿琥酯协同庆大霉素、头孢匹胺、舒氨西林对大肠埃希菌ATCC35218的联合抑菌曲线; 图2表示青蒿琥酯协同氨苄西林、加替沙星、舒氨西林对金葡菌ATCC25923的联合抑菌曲线; 图3表示青蒿琥酯协同庆大霉素、加替沙星、舒氨西林对大肠埃希菌临床分离株的联合抑菌曲线。 具体实施方式 下面结合实施例对本发明作进一步的说明,但所述实施例仅用于说明本发明而不是限制本发明。 青蒿琥酯(AS)与抗菌药物按照表1、表2中的配比,能够产生协同抗菌效力,降低抗菌药物的最低抑菌浓度和最低杀菌浓度。 表1不同浓度青蒿琥酯(AS)与抗菌药物协同使用时抗菌药物的MIC(μg/ml) 抗菌药物 大肠埃希菌ATCC35218 大肠埃希菌临床分离株 AS0 AS32 AS128 AS256 AS0 AS32 AS128 AS256 庆大霉素 4 0.25 0.125 0.0625 >256 >256 >256 >256 链霉素 >256 >256 >256 >256 >256 >256 >256 >256 舒氨西林 64 64 32 8 128 128 128 128 头孢匹胺 64 32 8 4 >256 >256 >256 >256 加替沙星 0.5 0.5 0.5 0.5 64 64 64 32 表2青蒿琥酯(AS)与不同抗菌药物协同使用时能产生抗菌作用的最小质量比 抗菌药物 大肠埃希菌ATCC35218 大肠埃希菌临床分离株 AS32 AS128 AS256 AS32 AS128 AS256 庆大霉素 128∶1 1024∶1 4069∶1 >1∶8 >1∶2 >1∶1 链霉素 >1∶8 >1∶2 >1∶1 >1∶8 >1∶2 >1∶1 舒氨西林 1∶2 4∶1 32∶1 1∶4 1∶1 2∶1 头孢匹胺 1∶1 16∶1 64∶1 >1∶8 >1∶2 >1∶1 加替沙星 64∶1 256∶1 512∶1 1∶2 1∶1 2∶1 实验例1 本实验例在于研究青蒿琥酯与十种抗菌药物对不同细菌的MIC(最低抑菌浓度)和MBC(最低杀菌浓度) 采用微孔稀释法,调整细菌浓度为105CFU/ml,接种于96孔无菌培养板内,青蒿琥酯和青霉素钠、氨苄西林等10种抗菌药物分别以生理盐水稀释为5.14mg/ml。加入各种药物至含细菌培养孔内,依次倍比稀释,第1~10孔药物的最终浓度依次为256、128、64、32、16、8、4、2、1、0.5ug/ml。置37℃培养箱孵育24h和48h,读取阳性和阴性对照孔,阴性对照孔清亮,阳性对照孔混浊。药物对细菌的MIC为24h后抑制细菌肉眼可见生长的最低药物浓度,药物对细菌的MBC为48h后抑制细菌肉眼可见生长的最低药物浓度。此次实验结果表明青蒿琥酯单独使用不能完全抑制细菌生长,并观察了不同抗菌药物对该五种细菌的抑菌效力。(见表3)。 表310种抗菌药物和AS的单独使用时对不同细菌的MIC及MBC 青霉素G钠 氨苄西林 头孢呋辛 头孢匹胺 庆大霉素 克林霉素 洛美沙星 加替沙星 舒氨西林 泰能 青蒿琥酯 35218 MIC >256 >256 2 64 4 256 <0.5 <0.5 64 <0.5 >256 MBC >256 >256 2 64 4 256 <0.5 <0.5 64 <0.5 >256 金黄色葡萄球菌 MIC 64 128 >256 >256 >256 >256 256 128 >256 64 >256 MBC >256 >256 >256 >256 >256 >256 >256 256 >256 64 >256 大肠埃希菌 MIC >256 >256 >256 >256 64 >256 >256 >256 64 <0.5 >256 MBC >256 >256 >256 >256 128 >256 >256 >256 128 1 >256 肺炎克雷伯菌 MIC 64 64 2 2 2 >256 2 1 8 <0.5 >256 MBC 64 128 2 2 2 >256 2 1 16 2 >256 肠球菌 MIC >256 >256 >256 >256 4 256 64 2 32 1 >256 MBC >256 >256 >256 >256 128 >256 256 32 32 1 >256 铜绿假单胞菌 MIC >256 >256 >256 256 >256 >256 32 8 >256 32 >256 MBC >256 >256 >256 256 >256 >256 32 16 >256 32 >256 实验例2 本实验例在于研究青蒿琥酯与抗菌药物联合应用后对大肠埃希菌MIC和MBC的影响。 采用棋盘式微孔稀释法,调整细菌浓度为105CFU/ml,接种于96孔无菌培养板内,将对五种细菌中等敏感的抗菌药物用低于MIC的剂量与不同浓度青蒿琥酯分别配伍,观察配伍后的药物对大肠埃希菌的MIC和MBC。此次实验结果表明青蒿琥酯单独使用虽不能完全抑制细菌生长,但与抗菌药物联合应用后可明显降低抗菌药物的MIC和MBC,说明青蒿琥酯与抗菌药物协同后对细菌有协同的抑菌效力。(见表4)。 表4各抗菌药物与青蒿琥酯协同后的最低抑菌浓度 大肠埃希菌ATCC35218 大肠埃希菌临床分离株 庆大霉素+AS 1/16MIC+32μg/ml AS 无作用 链霉素+AS 无作用 无作用 舒氨西林+AS 1/8MIC+256μg/ml AS 无作用 头孢匹胺+AS 1/8MIC+256μg/ml AS 无作用 加替沙星+AS 无作用 1/2MIC+256μg/mlAS 实验例3 本实验例在于研究青蒿琥酯与1/2MIC浓度的不同抗菌药物协同后对大肠埃希菌生长的抑制强度。 调整细菌浓度为106 CFU/ml,测定菌液OD600为0.002(1OD=5×108CFU/ml),参照实验例1结果,分别单独加入终浓度为256μg/ml青蒿琥酯或1/2MIC浓度的抗菌药物,以及同时加入256μg/ml青蒿琥酯和1/2MIC浓度的抗菌药物后,至于37℃恒温摇床150rpm振摇,分别测定1、3、5、7、9、12、16、24h时菌液的OD值,计算各时间点的细菌量。研究结果表明青蒿琥酯256μg/ml可使大肠埃希菌ATCC35218,金黄色葡萄球菌ATCC25923和大肠埃希菌临床分离株生长速度明显减慢,与单独使用抗菌药物相比,联合了青蒿琥酯后明显抑制细菌生长,抑制程度比单独使用抗菌药物或青蒿琥酯均强(图1、2、3)。 实验例4 本实验例在于研究青蒿琥酯联合应用抗菌药物对革兰阴性细菌攻击小鼠的保护作用 清洁级昆明种小白鼠70只(重庆医科大学实验动物中心提供),体重19.9±0.5g/只,雌雄各半,随机分为对照组、青蒿琥酯组、大肠埃希菌组,大肠埃希菌+庆大霉素组,青蒿琥酯(1.5、5、15mg/kg)+庆大霉素+大肠埃希菌组。每组10只动物。对照不给予任何试剂;青蒿素组肌肉注射15mg/kg的青蒿琥酯;大肠埃希菌组,给予0.8×106/kg活的大肠埃希菌ATCC35218;大肠埃希菌+庆大霉素组,给予庆大霉素0.5mg/kg肌肉注射后给予活大肠埃希菌ATCC35218;庆大霉素+青蒿琥酯+大肠埃希菌组,在给予青蒿琥酯后,立即给予庆大霉素及大肠埃希菌。给药完毕后给予正常饮食和饮水,观察7天内小鼠一般情况及死亡率(表5)。结果显示青蒿琥酯协同抗菌药物后可明显降低小鼠的死亡率,表明对青蒿琥酯与抗菌药物联合后出现协同保护作用。 表5青蒿琥酯协同庆大霉素对大肠埃希菌ATCC35218攻击小鼠的保护作用 处理组 总数 死亡数 死亡率(%) P 对照 10 0 0 - E.coli 10 10 100.0 - AS(15mg/kg) 10 10 100.0 - 庆大霉素(0.5mg/kg) 10 9 90.0 - AS(1.5mg/kg)+庆大+E.coli 10 6 60.0 0.024* AS(5mg/kg)+庆大+E.coli 10 0 0 0.001** AS(15mg/kg)+庆大+E.coli 10 0 0 0.001** 实验例5 本实验例在于研究青蒿琥酯联合应用抗菌药物对革兰阳性细菌攻击小鼠的保护作用 清洁级昆明种小白鼠70只(重庆医科大学实验动物中心提供),体重19.9±0.5g/只,雌雄各半,随机分为对照组、青蒿琥酯组、金葡菌组,金葡菌+加替沙星组,青蒿琥酯(5、15、45mg/kg)+加替沙星+金葡菌组。每组10只动物。对照不给予任何试剂;青蒿素组肌肉注射45mg/kg的青蒿琥酯;金葡菌组,给予1.0×107/kg活的金葡菌ATCC25923;金葡菌+加替沙星组,给予加替沙星0.5mg/kg肌肉注射后给予活金葡菌ATCC25923;加替沙星+青蒿琥酯+金葡菌组,在给予青蒿琥酯后,立即给予加替沙星及金葡菌。给药完毕后给予正常饮食和饮水,观察7天内小鼠一般情况及死亡率(表6)。结果显示青蒿琥酯协同抗菌药物后可明显降低小鼠的死亡率,表明对青蒿琥酯与抗菌药物联合后出现协同保护作用。 表6 青蒿琥酯协同加替沙星对金葡菌ATCC25923攻击小鼠的保护作用 处理组 总数 死亡数 死亡率(%) P 对照 10 0 0 - SA 10 10 100.0 - AS(45mg/kg) 10 10 100.0 - 加替沙星(0.5mg/kg) 10 9 90.0 - AS(5mg/kg)+加替沙星+SA 10 7 70.0 0.060 AS(15mg/kg)+加替沙星+SA 10 5 50 0.009** AS(45mg/kg)+加替沙星+SA 10 2 20 0.000** 实验例6 本实验例在于研究青蒿琥酯对灭活大肠埃希菌攻击小鼠的保护作用。 清洁级昆明种小白鼠60只(重庆医科大学实验动物中心提供),体重19.9±0.5g/只,雌雄各半,随机分为对照组、青蒿琥酯组、灭活大肠埃希菌组,青蒿琥酯(5、15、45mg/ml)+灭活大肠埃希菌组。每组8只动物。对照不给予任何试剂;青蒿琥酯组肌肉注射给予45mg/kg的青蒿琥酯,4h,24h,48h后重复给药一次;灭活大肠埃希菌组,给予1.25×1011/kg的灭活大肠埃希菌ATCC35218;青蒿琥酯+灭活大肠埃希菌组,在肌肉注射给予青蒿琥酯后,立即给予灭活大肠埃希菌ATCC35218,4h、24h、48h后重复肌肉注射给药青蒿琥酯1次。给药完毕后给予正常饮食和饮水,观察7天内小鼠一般情况及死亡率(表7)。结果显示青蒿琥酯可降低小鼠的死亡率,表明对致炎因子攻击的小鼠具有保护作用。 表7青蒿琥酯对灭活大肠埃希菌攻击小鼠的保护作用 处理组 总数 死亡数 死亡率(%) P 对照 8 0 0 - AS(45mg/kg) 8 0 0 - E.coli 8 8 100.0 - AS(5mg/kg)+E.coli 8 8 100.0 - AS(15mg/kg)+E.coli 8 6 25.0 0.048* AS(45mg/kg)+E.coli 8 4 50.0 0.011* *p<0.5;**p<0.01vsE.coli 实验例7 本实验例在于研究青蒿琥酯对灭活大肠埃希菌攻击小鼠血清细胞因子释放影响 清洁级昆明种小白鼠40只(重庆医科大学实验动物中心提供),体重19.9±0.5g/只,雌雄各半,随机分为对照组、青蒿琥酯组、灭活大肠埃希菌组、青蒿琥酯(5、15、45mg/kg)+灭活大肠埃希菌组,每组动物3只。对照不给予任何试剂。青蒿琥酯组肌肉注射给予45mg/kg的青蒿琥酯;青蒿琥酯(5、15、45mg/kg)+灭活大肠埃希菌组给予青蒿琥酯后立即给予灭活大肠埃希菌ATCC35218;给药完毕4h后摘眼球取血立即离心留置上清保存于-20℃,待测定细胞因子TNF-α(表8)。结果显示青蒿琥酯可显著降低灭活大肠埃希菌攻击小鼠血清细胞因子释放。 表8青蒿琥酯降低灭活大肠埃希菌ATCC35218攻击小鼠血清细胞因子TNF-α释放(n=6, ) 处理组 TNF-α(pg/ml) P 对照 58.10±15.31 AS(45mg/kg) 44.57±27.79 E.coli 1399.06±185.63 AS(5mg/kg)+E.coli 864.71±321.11 0.092 AS(15mg/kg)+E.coli 914.18±141.22 0.023* AS(45mg/kg)+E.coli 680.13±103.30 0.004** *p<0.5;**p<0.01vsE.coli 本发明将青蒿琥酯与抗菌药物的联合应用,提高了抗生素的抗菌效力,将青蒿琥酯与抗生素联合应用还可以预防、治疗细菌感染。 本领域技术人员可以根据本发明所述内容,将青蒿素及其衍生物与抗菌药物的联合同样可以达到本发明所述效果。 1.青蒿素及其衍生物中的一种与抗菌药物中的一种的联合应用,其能够产生协同抗菌效力,降低抗菌药物的最低抑菌浓度和最低杀菌浓度。 2.根据权利要求1所述的应用,其中青蒿素及其衍生物中的一种与抗菌药物中的一种的质量比为1∶1~4096∶1。 3.根据权利要求1所述的应用,上述菌为革兰阳性菌或革兰阴性菌。 4.根据权利要求1所述的应用,上述衍生物为二氢青蒿素、蒿甲醚、蒿乙醚和青蒿琥酯。 5.根据权利要求4所述的应用,该应用为青蒿琥酯与一种抗菌药物的联合应用。 6.根据权利要求1所述的应用,上述抗菌药物为β-内酰胺类抗生素、氨基糖苷类、克林霉素类和喹诺酮类抗菌药物。 7.根据权利要求6所述的应用,上述抗β-内酰胺类抗生素包括青霉素类、头孢菌素类和非典型β-内酰胺类抗生素。 https://patents.google.com/patent/CN101020056A/zh The present invention discloses the new use of available antimalarial arteannuin and its derivatives dihydro arteannuin, antiannuic methyl ether, antiannuic ethyl ether and antiannuic amber. Arteannuin and its derivatives are used through combination with antibiotic medicine to inhibit bacterial growth and enhance the antibiotic effect of available antibiotic medicine. Especially, antiannuic amber is applied in preventing and treating bacterial infection diseases except being used as antimalarial. The use in conjunction of arteannuin and derivant thereof and antibacterials Technical field The present invention relates to the medicinal application field, relate in particular to the antibacterial applications of arteannuin and derivant thereof. Background technology Infectious disease is owing in the pathogenic microorganism intrusive body, breed caused disease in vivo in large quantities, is one of major reason of clinical patient death.Cause the reason first antibacterial of death to breed the damage that the release toxin causes histoorgan in a large number; It two is that the antibacterial composition passes through immune cell activated, induce proinflammatory cytokines such as TNF-α, IL-1, IL-6, NO to discharge, cause the formed water fall effect of multiple inflammatory mediator, inflammatory reaction is enlarged even out of hand, finally causing with cell self property destruction is that the systemic inflammatory of feature is pyemic generation.Bacterial infection once was human first cause of the death, and antibiotic invention has brought human light of hope.But antibioticly generally make increasing antibacterial produce drug resistance,, wait for that human will be dark next time if do not solve the problem of antibiotic abuse. Herba Artemisiae Annuae extract is used for treating the relevant heating of malaria the history in more than 1,000 year, and except that traditional malaria effect, Herba Artemisiae Annuae class material also has the effect of others, as relieving asthma, anticancer, schistosomicide and to immune adjusting etc.The research of relevant arteannuin and derivant thereof both at home and abroad is a lot, but research range mainly concentrates on malaria, anticancer, antischistosomal effect and the mechanism thereof. Artesunate (Artesunate), chemistry dihydroartemisinine-1 by name, 2-α-monomester succinate, molecular formula is C19O8 H28, molecular weight 384 is the derivants with antimalarial arteannuin of sesquiterpene structure.Artesunate is novel antimalarial, and than arteannuin, artesunate can be mixed with the dosage form of any routine, comprises solid and liquid form, and administration is very convenient.Artesunate is as antimalarial, the height of not only tiring, and be difficult for producing toleration. To the existing report of the antiinflammatory action of arteannuin and derivant thereof, discover that arteannuin can suppress the activation of the synthetic and NF-κ B of LPS/TNF-α inductivity NO synthase; Artesunate to LPS and merge that interferon stimulates Turnover of Mouse Peritoneal Macrophages NO synthetic the obvious suppression effect arranged; Artesunate also has similar protective effect to the mouse macrophage RAW264.7 that LPS stimulates; Tan Yuqing etc. find that also Herba Artemisiae Annuae extract, arteannuin can reduce endotoxin shock mice LPO, ACP, endotoxin, TNF-α, P450 concentration, the increased SOD activity, reduce mouse death rate, prolong the mean survival time of mice, Mouse Liver, lung tissue form are also had the certain protection effect; Discovery arteannuin such as Wang Jun can suppress CpGODN, LPS, heat-inactivated EC inducing cell discharges inflammatory factor, and the sepsis model mice is had remarkable protective effect.But whether artesunate has antibacterial action, whether can improve the drug resistance of bacterial antibiotic, strengthen antibiotic antibacterial efficacy and infection that antibacterial is caused whether effective, announcement does not appear in the newspapers.China is the main grown place of arteannuin and its derivant, because this curative effect of medication height, toxicity are low, WHO has classified arteannuin as the main medicine of world's treatment malaria, China is enlarging Herba Artemisiae Annuae plantation and arteannuin production, therefore widen the indication of artemisinin derivatives, be applied to the control of bacterial infective diseases, undoubtedly to the utilization that improves artemisinin derivatives, increase its economic worth and improve the drug resistance phenomenon that the antibiotic abuse caused significant. Summary of the invention The objective of the invention is to arteannuin and derivant thereof, particularly artesunate is applied to antibiotic field, not only widen the purposes of arteannuin and derivant thereof, particularly artesunate, and can alleviate the abuse of existing antibacterials and the drug resistance phenomenon that causes thereof. Based on above-mentioned purpose, the technical scheme that the present invention uses is with a kind of use in conjunction of carrying out in a kind of and antibacterials in arteannuin and the derivant thereof, this application is not the application as the treatment of diseases method, but the application in the medicine that is equipped with treatment disease (antibiotic), wherein above-mentioned two kinds of components in proportions are 1: 1~4096: 1, last two kinds of compositions can be prepared into compositions, also can be two kinds of independent medicaments.Above-mentioned two kinds of compositions can produce Synergistic antimicrobial and render a service, and can reduce the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) of antibacterials. Above-mentioned bacterium comprises gram positive bacteria and gram negative bacteria; Said derivative comprises dihydroartemisinine, Artemether, arteether and artesunate; Above-mentioned antibacterials comprise the medicine of beta-lactam antibiotic (penicillins, cephalosporins, atypia beta-lactam antibiotic), aminoglycoside, clindamycin class, quinolones, in a preferred embodiment, above-mentioned antibacterials are benzylpenicillin, ampicillin, cefuroxime, cefpiramide, gentamycin, clindamycin, lomefloxacin, Gatifloxacin, ampicillin sodium-sulbactam sodium and safe energy, are preferably gentamycin, ampicillin sodium-sulbactam sodium, cefpiramide and Gatifloxacin. Above-mentioned application is preferably the use in conjunction of artesunate and antibacterials. The independent use of artesunate can suppress the growth of gram-negative bacteria and gram positive bacteria, with the antibacterial efficacy that can obviously strengthen antibacterials after the antibacterials use in conjunction, reduces the using dosage of antibacterials; Experiment shows that arteannuin and artesunate and antibacterials use in conjunction can obviously reduce antibacterial (comprising Grain-negative, positive bacteria) and attack mouse death rate, significantly suppresses mice serum LPS content and cytokine TNF-α and discharges. This finder studies the antibiotic and antiinflammatory action of artesunate inside and outside. At first, this research worker has been carried out pharmacology's analysis to the vitro antibacterial activity of this artesunate, and with the antibiotic use in conjunction after find that the independent use of artesunate can suppress the growth of gram-negative bacteria and gram positive bacteria, with can obviously strengthen antibiotic antibacterial efficacy after the antibiotic use in conjunction, reduce antibiotic using dosage; Secondly, artesunate and antibiotic use in conjunction and can obviously reduce antibacterial (comprising Grain-negative, positive bacteria) and attack mouse death rate significantly suppress mice serum LPS content and cytokine TNF-α release. Therefore the present invention has widened the purposes of arteannuin and derivant thereof, particularly artesunate, has improved the antibacterial efficacy of antibacterials, the drug resistance phenomenon that has alleviated the antibacterials abuse and caused. Description of drawings Fig. 1 represents that artesunate works in coordination with gentamycin, cefpiramide, ampicillin sodium-sulbactam sodium to the antibacterial curve of the associating of escherichia coli ATCC35218; Fig. 2 represents that artesunate works in coordination with ampicillin, Gatifloxacin, the ampicillin sodium-sulbactam sodium antibacterial curve of associating to the golden bacterium ATCC25923 of Portugal; Fig. 3 represents that artesunate works in coordination with gentamycin, Gatifloxacin, ampicillin sodium-sulbactam sodium to the antibacterial curve of the associating of escherichia coli clinical separation strain. The specific embodiment The present invention is further illustrated below in conjunction with embodiment, but described embodiment only is used to illustrate the present invention rather than restriction the present invention. Artesunate (AS) and antibacterials can produce Synergistic antimicrobial and render a service according to the proportioning in table 1, the table 2, reduce the minimum inhibitory concentration and the minimum bactericidal concentration of antibacterials. The MIC (μ g/ml) of antibacterials when table 1 variable concentrations artesunate (AS) uses with antibacterials are collaborative Antibacterials Escherichia coli ATCC35218 The escherichia coli clinical separation strain AS0 AS32 AS128 AS256 AS0 AS32 AS128 AS256 Gentamycin 4 0.25 0.125 0.0625 >256 >256 >256 >256 Streptomycin >256 >256 >256 >256 >256 >256 >256 >256 Ampicillin sodium-sulbactam sodium 64 64 32 8 128 128 128 128 Cefpiramide 64 32 8 4 >256 >256 >256 >256 Gatifloxacin 0.5 0.5 0.5 0.5 64 64 64 32 Table 2 artesunate (AS) can produce the minimum mass ratio of antibacterial action when using with different antibacterials are collaborative Antibacterials Escherichia coli ATCC35218 The escherichia coli clinical separation strain AS32 AS128 AS256 AS32 AS128 AS256 Gentamycin 128∶1 1024∶1 4069∶1 >1∶8 >1∶2 >1∶1 Streptomycin >1∶8 >1∶2 >1∶1 >1∶8 >1∶2 >1∶1 Ampicillin sodium-sulbactam sodium 1∶2 4∶1 32∶1 1∶4 1∶1 2∶1 Cefpiramide 1∶1 16∶1 64∶1 >1∶8 >1∶2 >1∶1 Gatifloxacin 64∶1 256∶1 512∶1 1∶2 1∶1 2∶1 Experimental example 1 This experimental example is to study artesunate and ten kinds of antibacterials MIC (minimum inhibitory concentration) and the MBC (minimum bactericidal concentration) to different bacterium Adopt the micropore dilution method, adjusting bacterial concentration is 10 5CFU/ml is inoculated in the 96 hole aseptic culture plates, and 10 kinds of antibacterials such as artesunate and penicillin sodium, ampicillin etc. are 5.14mg/ml with the normal saline dilution respectively.Add various medicines to containing in the antibacterial culturing hole, doubling dilution successively, the ultimate density of the 1st~10 hole medicine is followed successively by 256,128,64,32,16,8,4,2,1,0.5ug/ml.Put 37 ℃ of incubators and hatch 24h and 48h, read the positive and negative control hole, negative control hole is limpid, positive control hole muddiness.Medicine is the lowest concentration of drug that suppresses antibacterial naked eyes visible growth behind the 24h to the MIC of antibacterial, and medicine is the lowest concentration of drug that suppresses antibacterial naked eyes visible growth behind the 48h to the MBC of antibacterial.This time experimental result shows the independent use of artesunate bacteria growing inhibiting fully, and has observed the inhibitory effect of different antibacterials to these five kinds of antibacterials.(seeing Table 3). During independent use of table 310 kind of antibacterials and AS to the MIC and the MBC of different bacterium Penicillin G sodium The ampicillin Cefuroxime Cefpiramide Gentamycin Clindamycin Lomefloxacin Gatifloxacin Ampicillin sodium-sulbactam sodium Safe energy Artesunate 35218 MIC >256 >256 2 64 4 256 <0.5 <0.5 64 <0.5 >256 MBC >256 >256 2 64 4 256 <0.5 <0.5 64 <0.5 >256 Staphylococcus aureus MIC 64 128 >256 >256 >256 >256 256 128 >256 64 >256 MBC >256 >256 >256 >256 >256 >256 >256 256 >256 64 >256 Escherichia coli MIC >256 >256 >256 >256 64 >256 >256 >256 64 <0.5 >256 MBC >256 >256 >256 >256 128 >256 >256 >256 128 1 >256 Klebsiella Pneumoniae MIC 64 64 2 2 2 >256 2 1 8 <0.5 >256 MBC 64 128 2 2 2 >256 2 1 16 2 >256 Enterococcus MIC >256 >256 >256 >256 4 256 64 2 32 1 >256 MBC >256 >256 >256 >256 128 >256 256 32 32 1 >256 Pseudomonas aeruginosa MIC >256 >256 >256 256 >256 >256 32 8 >256 32 >256 MBC >256 >256 >256 256 >256 >256 32 16 >256 32 >256 Experimental example 2 This experimental example is to study after artesunate and the antibacterials use in conjunction influence to escherichia coli MIC and MBC. Adopt checkerboard type micropore dilution method, adjusting bacterial concentration is 10 5CFU/ml is inoculated in the 96 hole aseptic culture plates, will be to the antibacterials of five kinds of medium sensitivities of antibacterial dosage and the variable concentrations artesunate difference compatibility that is lower than MIC, and the medicine behind the observation compatibility is to the MIC and the MBC of escherichia coli.Though this time experimental result shows that artesunate uses bacteria growing inhibiting fully separately,, illustrate that there is collaborative inhibitory effect the collaborative back of artesunate and antibacterials to antibacterial with the MIC and the MBC that can obviously reduce antibacterials after the antibacterials use in conjunction.(seeing Table 4). Minimum inhibitory concentration after each antibacterials of table 4 and artesunate are collaborative Escherichia coli ATCC35218 The escherichia coli clinical separation strain Gentamycin+AS 1/16MIC+32μg/ml AS No effect Streptomycin+AS No effect No effect Ampicillin sodium-sulbactam sodium+AS 1/8MIC+256μg/ml AS No effect Cefpiramide+AS 1/8MIC+256μg/ml AS No effect Gatifloxacin+AS No effect 1/2MIC+256μg/mlAS Experimental example 3 This experimental example is to study artesunate and the inhibition strength of the collaborative back of the different antibacterials of 1/2MIC concentration to the escherichia coli growth. Adjusting bacterial concentration is 10 6CFU/ml, measuring bacterium liquid OD600 is 0.002 (1OD=5 * 10 8CFU/ml), with reference to experimental example 1 result, adding final concentration respectively separately is the antibacterials of 256 μ g/ml artesunate or 1/2MIC concentration, and after adding the antibacterials of 256 μ g/ml artesunate and 1/2MIC concentration simultaneously, as for 37 ℃ of constant temperature shaking table 150rpm jolting, measure 1,3,5,7,9,12,16 respectively, the OD value of bacterium liquid during 24h, calculate the amount of bacteria of each time point.Result of study shows that artesunate 256 μ g/ml can make escherichia coli ATCC35218, the staphylococcus aureus ATCC25923 and the escherichia coli clinical separation strain speed of growth obviously slow down, compare with independent use antibacterials, united behind the artesunate obviously bacteria growing inhibiting, the inhibition degree is than using antibacterials or artesunate all strong (Fig. 1,2,3) separately. Experimental example 4 This experimental example is to study artesunate use in conjunction antibacterials are attacked mice to gram-negative bacteria protective effect Cleaning level 70 of the kind white mice in Kunming (Medical University Of Chongqing's Experimental Animal Center provides), body weight 19.9 ± 0.5g/ only, male and female half and half, be divided into matched group, artesunate group, escherichia coli group at random, escherichia coli+gentamycin group, artesunate (1.5,5,15mg/kg)+gentamycin+escherichia coli group.Every group of 10 animals.Contrast does not give any reagent; The artesunate of arteannuin group intramuscular injection 15mg/kg; The escherichia coli group gives 0.8 * 10 6The escherichia coli ATCC35218 that/kg lives; Escherichia coli+gentamycin group gives the escherichia coli ATCC35218 that lives after the gentamycin 0.5mg/kg intramuscular injection; Gentamycin+artesunate+escherichia coli group after giving artesunate, gives gentamycin and escherichia coli immediately.Give normal diet and drinking-water after administration finishes, observe mice ordinary circumstance and mortality rate (table 5) in 7 days.The result can obviously reduce mortality of mice after showing artesunate Synergistic antimicrobial medicine, shows the coordinating protection effect occurring after artesunate and the antibacterials associating. The collaborative gentamycin of table 5 artesunate is attacked mice to escherichia coli ATCC35218 protective effect Processed group Sum Death toll Mortality rate (%) P Contrast 10 0 0 - E.coli 10 10 100.0 - AS(15mg/kg) 10 10 100.0 - Gentamycin (0.5mg/kg) 10 9 90.0 - AS (1.5mg/kg)+celebrating is big+E.coli 10 6 60.0 0.024 * AS (5mg/kg)+celebrating is big+E.coli 10 0 0 0.001 ** AS (15mg/kg)+celebrating is big+E.coli 10 0 0 0.001 ** Experimental example 5 This experimental example is to study artesunate use in conjunction antibacterials are attacked mice to gram-positive bacteria protective effect Cleaning level 70 of the kind white mice in Kunming (Medical University Of Chongqing's Experimental Animal Center provides), body weight 19.9 ± 0.5g/ only, male and female half and half, be divided into matched group, artesunate group, golden Portugal bacterium group at random, gold Portugal bacterium+Gatifloxacin group, artesunate (5,15,45mg/kg)+Gatifloxacin+golden Portugal bacterium group.Every group of 10 animals.Contrast does not give any reagent; The artesunate of arteannuin group intramuscular injection 45mg/kg; Gold Portugal bacterium group gives 1.0 * 10 7The golden bacterium ATCC25923 of Portugal that/kg lives; Gold Portugal bacterium+Gatifloxacin group gives the golden bacterium ATCC25923 of Portugal that lives after the Gatifloxacin 0.5mg/kg intramuscular injection; Gatifloxacin+artesunate+golden Portugal bacterium group after giving artesunate, gives Gatifloxacin and golden Portugal bacterium immediately.Give normal diet and drinking-water after administration finishes, observe mice ordinary circumstance and mortality rate (table 6) in 7 days.The result can obviously reduce mortality of mice after showing artesunate Synergistic antimicrobial medicine, shows the coordinating protection effect occurring after artesunate and the antibacterials associating. The collaborative Gatifloxacin of table 6 artesunate is attacked mice to the golden bacterium ATCC25923 of Portugal protective effect Processed group Sum Death toll Mortality rate (%) P Contrast 10 0 0 - SA 10 10 100.0 - AS(45mg/kg) 10 10 100.0 - Gatifloxacin (0.5mg/kg) 10 9 90.0 - AS (5mg/kg)+Gatifloxacin+SA 10 7 70.0 0.060 AS (15mg/kg)+Gatifloxacin+SA 10 5 50 0.009 ** AS (45mg/kg)+Gatifloxacin+SA 10 2 20 0.000 ** Experimental example 6 This experimental example is to study artesunate is attacked mice to the deactivation escherichia coli protective effect. Cleaning level 60 of the kind white mice in Kunming (Medical University Of Chongqing's Experimental Animal Center provides), body weight 19.9 ± 0.5g/ only, male and female half and half are divided into matched group, artesunate group, deactivation escherichia coli group at random, artesunate (5,15,45mg/ml)+deactivation escherichia coli group.Every group of 8 animals.Contrast does not give any reagent; The intramuscular injection of artesunate group gives the artesunate of 45mg/kg, 4h, and 24h, repeat administration is once behind the 48h; Deactivation escherichia coli group gives 1.25 * 10 11The deactivation escherichia coli ATCC35218 of/kg; Artesunate+deactivation escherichia coli group after intramuscular injection gives artesunate, gives deactivation escherichia coli ATCC35218 immediately, repeats administered intramuscular artesunate 1 time behind 4h, 24h, the 48h.Give normal diet and drinking-water after administration finishes, observe mice ordinary circumstance and mortality rate (table 7) in 7 days.The result shows that artesunate can reduce mortality of mice, shows that the mice that pro-inflammatory cytokine is attacked has protective effect. Table 7 artesunate is attacked the protective effect of mice to the deactivation escherichia coli Processed group Sum Death toll Mortality rate (%) P Contrast 8 0 0 - AS(45mg/kg) 8 0 0 - E.coli 8 8 100.0 - AS(5mg/kg)+E.coli 8 8 100.0 - AS(15mg/kg)+E.coli 8 6 25.0 0.048 * AS(45mg/kg)+E.coli 8 4 50.0 0.011 * *p<0.5; **p<0.01vsE.coli Experimental example 7 This experimental example is to study artesunate the deactivation escherichia coli is attacked the influence of mice serum release of cytokines Cleaning level 40 of the kind white mice in Kunming (Medical University Of Chongqing's Experimental Animal Center provides), body weight 19.9 ± 0.5g/ only, male and female half and half, be divided into matched group, artesunate group, deactivation escherichia coli group, artesunate (5,15,45mg/kg)+deactivation escherichia coli group at random, 3 of every treated animals.Contrast does not give any reagent.The intramuscular injection of artesunate group gives the artesunate of 45mg/kg; Artesunate (5,15,45mg/kg)+deactivation escherichia coli group gives to give deactivation escherichia coli ATCC35218 immediately behind the artesunate; Administration finishes and plucks eyeball behind the 4h and get the centrifugal immediately indwelling supernatant of blood and be stored in-20 ℃, cytokine TNF-α to be determined (table 8).The result shows that artesunate can significantly reduce the deactivation escherichia coli and attack the mice serum release of cytokines. Table 8 artesunate reduction deactivation escherichia coli ATCC35218 attack mice serum cytokine TNF-α release (n=6, ) Processed group TNF-α(pg/ml) P Contrast 58.10±15.31 AS(45mg/kg) 44.57±27.79 E.coli 1399.06±185.63 AS(5mg/kg)+E.coli 864.71±321.11 0.092 AS(15mg/kg)+E.coli 914.18±141.22 0.023 * AS(45mg/kg)+E.coli 680.13±103.30 0.004 ** *p<0.5; **p<0.01vsE.coli The present invention has improved antibiotic antibacterial efficacy with the use in conjunction of artesunate and antibacterials, and artesunate and antibiotic use in conjunction can also be prevented, treat bacterial infection. Those skilled in the art can be according to content of the present invention, and the associating of arteannuin and derivant and antibacterials can be reached effect of the present invention equally. 1. a kind of use in conjunction in a kind of and antibacterials in arteannuin and the derivant thereof, it can produce Synergistic antimicrobial and render a service, and reduces the minimum inhibitory concentration and the minimum bactericidal concentration of antibacterials. 2. application according to claim 1, wherein a kind of mass ratio in a kind of and antibacterials in arteannuin and the derivant thereof is 1: 1~4096: 1. 3. application according to claim 1, above-mentioned bacterium are gram positive bacteria or gram-negative bacteria. 4. application according to claim 1, said derivative are dihydroartemisinine, Artemether, arteether and artesunate. 5. application according to claim 4, this is applied as the use in conjunction of artesunate and a kind of antibacterials. 6. application according to claim 1, above-mentioned antibacterials are beta-lactam antibiotic, aminoglycoside, clindamycin class and carbostyril family antibacterial drugs. 7. application according to claim 6, above-mentioned anti-beta-lactam antibiotic comprises penicillins, cephalosporins and atypia beta-lactam antibiotic. https://patents.google.com/patent/CN101020056A/en
  10. Short link to this article: https://gettr.ink/X2catz 该研究评估了使用青蒿叶粉作为预防疟疾发作的治疗方法,在25名患者中进行了评估,其中大部分是儿童,所有患者都因骨科问题接受了手术治疗。治疗持续时间为36小时的患者有11例,60小时的患者有14例。平均寄生虫数量从432个/毫升降至165个/毫升,即改善了62%,而且根据治疗持续时间的不同没有显著差异。不论患者的年龄和体重如何,疗效都是相似的。这些结果是在非常低的粉末量(每天400至500毫克)和极低的青蒿素数量(每天0.4至0.5毫克)下获得的。结论是青蒿叶粉似乎比茶制品更有效,但成本更高,可能不易获得。廉价且随处可得的茶制品仍然是大规模预防和治疗疟疾的最佳方法,在最贫困的国家应优先选择使用。除了研究寨卡病毒感染外,术前调查还包括检测消化道寄生虫(所有病例均呈阳性)和艾滋病病毒检测(除了一个成年患者外,所有病例均呈阴性)。 表1 不同制备方法得到的[ART]。 实验1:所有参数保持不变,只有温度变化。 实验2:温度升高到沸点以上。 实验3:材料:溶剂比例变化。 所有结果均以[ART]表示,单位为毫克/升,并以提取效率百分比表示。 样品1是通过在室温下用研钵和研杵粉碎植物材料制备的。 Sample: 样品编号或标识。 Temperature (°C): 实验处理的温度(摄氏度)。 Contact Time (min): 样品与溶剂接触的时间(分钟)。 Material: [ART] Solvent mg/L Ratio (g/L): 材料名称以及溶剂的浓度和比率。 Extraction Efficiency (%): 提取效率,以百分比表示,衡量了从样品中提取目标物的效率。 "5atb.p." 表示在沸点下的5分钟。这可能指样品在沸点下处理了5分钟。 "1at115" 表示在115°C下处理了1分钟。 青蒿叶粉囊的惊人功效 摘要 使用青蒿叶粉末作为预防疟疾发作的治疗方法,对25名患者进行了评估,其中大多数是儿童,所有患者都接受了整形外科手术。治疗持续时间为11名患者为36小时,14名患者为60小时。平均寄生虫血量从432个/毫升降至165个/毫升,即62%的改善,根据治疗持续时间没有显著差异。无论患者的年龄和体重如何,疗效都相似。这些结果是通过非常低剂量的粉末(每天400到500毫克)以及非常低量的青蒿素(每天0.4到0.5毫克)获得的。结论是,青蒿叶粉显然比茶制品更有效,但成本更高,也许不能常规使用。茶制品便宜且随处可得,仍然是预防和治疗大规模疟疾的最佳方法,应该在最贫穷的国家中优先考虑。 介绍 在中非共和国对残疾儿童进行的多次小儿骨科手术中,我们经常面对术后高热的情况,出现在手术后的第二天或两天后,被归因于疟疾,并用喹啉盐(喹嗪马)治疗。最近一次会诊中,我们调查了一些无症状儿童的血液中是否存在恶性疟原虫,结果发现所有调查病例均呈阳性。 众所周知,以茶剂形式给予的青蒿叶因其青蒿素以及众多其他类黄酮和成分而自古以来就被用于抗疟疾作用。尽管易于制备,而且价格低廉,但茶制品被认为在外科手术环境中不完全适用且难以使用,因此我们转向了将青蒿叶粉末制成胶囊,用于术后立即预防儿童手术期间的疟疾发作。给予青蒿叶粉末的优点在于可提供“全部”,即植物中存在的所有分子,与青蒿素协同作用。中国研究团队[1]在1992年已经发现,青蒿叶提取物的明胶胶囊在小鼠上的药理学和临床试验中,对伯氏疟原虫和间日疟原虫感染的治愈率达到了100%。近年来,坦桑尼亚、马里、布隆迪和刚果(金)的其他医疗团队(Saint-Hillier,Klabes,Tumaini)使用青蒿叶胶囊在成人和儿童身上进行了优秀的结果和无副作用的试验。Weathers等人[2]已经注意到,青蒿植物材料中的青蒿素与纯药物相比,向血液中的转移率出人意料地高。 这项研究的目的是评估这种预防性治疗的有效性。它于2012年11月在中非共和国的一次外科手术中进行。 材料和方法 本研究共有25名患者 22名儿童,平均年龄为8岁4个月(1-16岁),3名成人,平均年龄为27岁(18-40岁)。所有患者均接受了整形手术(表1)。除了恶性疟原虫调查外,术前检查还包括消化道寄生虫检测(所有病例均为阳性)和HIV检测(所有病例均为阴性,除一个成人外)。 协议 使用的青蒿叶是卢森堡品种,于2009年在Walferdange地区收获,以35°C的工业设备干燥,并由植物卫生证书编号EC/LU/11773覆盖。 恶性疟原虫血浓度检测了两次:一次是在开始预防性治疗之前,另一次是在J+2,即术后第二天早上。治疗持续时间如下: 在14例中,治疗是在手术前一天(J-1)开始的:每位患者在2个半天内每天接受2粒胶囊(J-1日晚上2粒,J+0日晚上2粒,J+1日早晚各1粒,J+2日早上1粒)。在这些情况下,治疗持续时间为60小时。 在11例中,由于纯粹的后勤原因(患者在分发胶囊的前一天手术时不在场),治疗从J+0开始:这11名患者在J+0日晚上接受2粒胶囊,J+1日早晚各1粒,J+2日早上1粒。在这些情况下,治疗持续时间为36小时。 对于年龄较小的患者(3岁以下),胶囊被打开,粉末与牛奶或捣碎的木薯一起服用。体重在20公斤以下的患者使用1号胶囊;所有其他患者使用0号胶囊。 *通讯作者:米歇尔·奥尼穆斯,法国康塔尔大学名誉小儿骨科教授,电子邮件:monimus@wanadoo.fr 收到日期:2013年4月17日;接受日期:2013年5月22日;发表日期:2013年5月24日 引用:Onimus M,Carteron S,Lutgen P(2013)青蒿叶粉囊的惊人功效。 Med Aromat Plants 2:125。 doi:10.4172/2167-0412.1000125 版权:©2013年奥尼穆斯M等人。这是根据创作共享许可的开放获取文章,该许可允许在任何媒介中进行无限制的使用、分发和再生产,只要原作者和来源被列明。 生物学结果 治疗开始前的平均寄生虫数量为 Plasmodium falciparum 432个/毫升(范围:160-910)(见表1)。治疗结束时的平均寄生虫数量为165个/毫升(范围:0-320),平均改善了62%;其中一名患者(#18)的寄生虫数量没有下降。 对于在36小时内接受治疗的11名患者,寄生虫数量从395减少到142,即改善了64%(范围:23%-100%)。对于在60小时内接受治疗的14名患者,寄生虫数量从461减少到183,即改善了60%(范围:<14%-85%)。 临床结果 镇痛效果 治疗在所有病例中都完全耐受。没有观察到任何消化不耐受症状。没有观察到发热反应。所有病例的术后情况都被认为是顺利的。术后水肿,尤其是在先天性马蹄内翻后内侧释放术后,通常会频繁观察到,但此次水肿异常轻微。 我们注意到了显著的术后镇痛效果。手术后12至18小时内镇痛剂(对乙酰氨基酚)的使用可以显著减少。这种效果在文献中[3]已经描述了对其他青蒿属植物如艾蒿(Artemisia vulgaris)的镇痛作用。因此,这与青蒿素无关。 抗菌和抗炎效果 在这项研究中,我们没有评估给予患者的青蒿叶粉对患者体内细菌负荷的影响。众所周知,这种植物对粪大肠埃希氏菌和链球菌具有强烈的杀菌作用。低青蒿素含量的该草药提取物能够激活淋巴细胞(个人通讯P. Lutgen),并且具有强烈的抗炎作用,可以通过减少IL-6和IL-8的分泌来证明。所有这些都可以解释手术后患者健康状况的改善[5]。 讨论 这些结果仅是初步结果,基于对小样本的评估,没有对照组。然而,实验室测试是在同一实验室中进行的,大多由同一技术员执行。因此,术前和术后测量中的可能误差可被认为是相似且可忽略不计的,这增加了结果的可信度,并使一些初步结论成为可能。 所有患者术前均表现出无症状的寄生虫感染;这已经得到了充分的证明[6]。在我们的研究中,目标不是清除血液中的寄生虫,而是在术后的前几天内尽可能减少寄生虫感染,以避免发生疟疾发作,并且治疗意图上是不会延长到第二个术后日。尽管如此,在这项研究中观察到的青蒿叶粉的疗效为长期治疗疟疾提供了依据,正如茶制品已经证明的那样[7]。 虽然已经3年,但青蒿叶粉对 Plasmodium falciparum 仍然非常有效,这表明所有成分即使经过几年仍然保持稳定。叶片存放在干燥的地方,没有特别的预防措施。 在本研究中,使用青蒿叶粉预防疟疾在几乎所有病例中在临床和生物学上都是有效的,可以将寄生虫数量减少到初始值的三分之一。减少是迅速获得的,即使在36小时后也是如此,而且在60小时后也没有变化。只有一个病例(患者编号18)观察到寄生虫数量增加,但没有任何临床表现。在这种情况下,治疗可能允许取消初期疟疾发作。 给予的粉末量非常低:胶囊号1中的粉末量约为200毫克,胶囊号0中的粉末量约为250毫克。这意味着每天给予大约400到500毫克的青蒿叶粉。这种粉末来自卢森堡品种,其中青蒿素含量约为0.1%,据估计每天约给予0.4-0.5毫克的青蒿素。与使用 ACT 时推荐的数量相比,这个数量更低,并且远低于茶制品中的量:在穆勒的研究中为12毫克[8],在 Räth等人的研究中为94毫克[9],在Silva等人的研究中为40-46毫克[10]。尽管如此,尽管这个数量非常低,但青蒿叶粉的治疗效果非常显著。 另一个有趣的事实是,不论患者的年龄如何,寄生虫数量的改善效果都相同,因此无论患者的体重如何:5岁以下儿童寄生虫数量减少了60%,5至13岁的儿童寄生虫数量减少了52%,13岁以上的患者寄生虫数量减少了71%。这表明,即使在低剂量下,该粉末的疗效也非常显著。现在,增加剂量可能会增加疗效,这需要进一步评估。与茶制品相比,青蒿叶粉的优点是有利于提供植物中的所有分子,尤其是多糖、挥发油[11]和类黄酮。已经证明这些分子增加了青蒿素的作用[12],并且它们还具有直接的特异性抗疟疾作用[13-15]。Weathers小组最近进行的使用P. chabaudi感染的啮齿动物的研究表明,口服一次干燥青蒿叶粉后,30小时内寄生虫数量完全减少[16]。 给予青蒿叶粉的治疗比茶制品更容易且显然更有效。然而,胶囊成本高昂,可能在偏远地区不易获得。在我们看来,胶囊并不完全适用于大规模使用青蒿叶粉。因此,在最贫困的国家,推广本地生产青蒿叶及其制成茶的方法仍然是预防和治疗疟疾的最佳方法,并且应该得到推广。然而,另一种可能性是,如果可以用传统的研钵将植物研磨成粉末,那么可以给予粉末。在特定条件下,如重症疟疾,胶囊作为栓剂仍然是一种选择。 备注:在同一次手术中,另外5名未纳入研究的患者(4名成人和1名儿童)发生了疟疾发作,其中一例即使使用马拉酮预防也发生了疟疾发作,在2例中没有预防性治疗,在2例中尽管接受了ACT标准治疗超过10天,病情仍然持续。这5名患者接受了更高剂量的胶囊治疗:开始治疗时的初始剂量为3颗胶囊,然后每天4颗,持续7天。在所有5例中,观察到了功能障碍和高热症状在24小时后迅速缓解。两例病例进行了寄生虫数量的测定,包括治疗前和治疗过程中(治疗后60小时)的寄生虫数量测定:在一例病例中(成人),Plasmodium falciparum浓度从1500下降到180个/毫升。在第二例(儿童)中,在治疗前24小时内Plasmodium falciparum浓度从240增加到8400个/毫升,治疗后两天半下降到80个/毫升。 The Surprising Efficiency of Artemisia annua Powder Capsules Michel Onimus1*, Sophie Carteron2 and Pierre Lutgen3 1Honorary Professor of Pediatric Orthopaedics, Franche Comté University, France 2Pharmacien, Volontaire DCC in Central African Républic, Africa 3BELHERB-IFBV, Luxembourg, Grand Duchy Sample: Sample identification or label. Temperature (°C): The temperature (in degrees Celsius) of the experiment. Contact Time (min): The duration (in minutes) of contact between the sample and solvent. Material: [ART] Solvent mg/L Ratio (g/L): The material name and the concentration and ratio of the solvent in milligrams per liter and grams per liter, respectively. Extraction Efficiency (%): The percentage representing the efficiency of extracting the target substance from the sample. "5atb.p." indicates 5 minutes at the boiling point. This likely means the sample was treated for 5 minutes at its boiling point. "1at115" indicates 1 minute at 115°C. Abstract The administration of powdered leaves of Artemisia annua, used as preventive treatment of malaria attack, was evaluated in 25 patients, most of them children, and all of them were operated for orthopaedic disorder. The duration of the treatment was 36 hours in 11 patients and 60 hours in 14 patients. The average parasitemia was reduced from 432 parasites/ml to 165 parasites/ml, i.e. 62% improvement, without significant difference according to the duration of the treatment. The efficacy was similar whatever the age and the weight of the patient. These results were obtained with a very low amount of powder (400 to 500 mg per day), and with very low quantity of artemisinin (0.4 to 0.5 mg per day). It is concluded that the Artemisia annua powder is apparently more effective than the tea preparation, but more costly and maybe not routinely available. The tea preparation, inexpensive and available everywhere, is still the best method for prevention and treatment of malaria on a large scale and should be preferred in the poorest countries. Introduction During numerous surgical interventions in pediatric orthopaedics conducted with crippled children in Central African Republic, we have frequently been confronted with post-operative high temperatures, occurring the day following or two days after the surgery, which were attributed to malaria and were treated with Quinine salts administration (Quinimax). During a recent session, we investigated the presence of Plasmodium falciparum in the blood of some asymptomatic children, with a positive result in all investigated cases. Artemisia annua administered as tea is known since centuries for its antimalarial effect, due to artemisinin as well as numerous other flavonoids and constituents. Although easy to prepare, and moreover inexpensive, the tea preparation was considered as not perfectly adapted and difficult to use in a surgical context, and we turned towards administration of powdered leaves of Artemisia annua in capsules, which were used as preventive treatment of malaria attack during the immediate post-operative period in operated children. The administration of powdered leaves also presents the advantage to deliver the « totum », i.e. the whole set of molecules present in the plant, synergically acting with artemisinin. A Chinese research team [1] had already found in 1992 that gelatine capsules of Artemisia annua extract used in pharmacological and clinical trials on mice gave a cure rate of 100% for Plasmodium berghei and Plasmodium vivax infections. In recent years other medical teams (Saint-Hillier, Klabes, Tumaini) in Tanzania, Mali, Burundi, DRCongo have been working with Artemisia annua capsules on adults and children with excellent results and no side effects. A surprising high level of transfer of artemisinin into the bloodstream from the plant material vs. the pure drug had already been noticed by Weathers et al. [2]. The aim of this study was to evaluate the effectiveness of this preventive treatment. It was conducted during a surgical session in Central African Republic in November 2012. Materials and Methods Twenty five patients were included in the study 22 children with an average age of 8 years and 4 months (1-16) and 3 adults with an average age of 27 years (18-40). All patients were operated for an orthopaedic problem (Table 1). Besides the Plasmodium falciparum investigations, the preoperative investigation included detection of digestive parasites (positive in all cases) and HIV test (negative in all cases except an adult). Protocol The Artemisia annua used was the Luxembourg variety, harvested at the site of Walferdange in 2009, dried in industrial equipment at 35°C and covered by the Phytosanitary Certificate No EC/LU/11773. The Plasmodium falciparum blood concentration was investigated twice: once before starting the preventive treatment, and once at J+2, i.e. on the morning of the second post-operative day. The duration of the treatment was as following: In 14 cases the treatment was started on the day before the surgery (J-1): every patient received 2 capsules per day during 2 and half days (2 capsules on evening at J-1, 2 capsules on evening at J+0, 1 capsule on morning and evening at J+1 and 1 capsule on the morning at J+2). In these cases, the treatment was administered during 60 hours. In 11 cases, because of purely logistical reasons (the patient was absent the day before the surgery when the capsules were distributed), the treatment started at J+0: these 11 patients received 2 capsules on evening at J+0, 1 capsule on morning and on evening at J+1 and 1 capsule on the morning at J+2. In these cases the treatment was administered during 36 hours. For the younger patients (less than 3 years of age) the capsules were opened and the powder was administered with milk or mashed manioc. Capsules n°1 was used for patients with weight under 20 kg; all other patients received capsules n°0. *Corresponding author: Michel Onimus, Honorary Professor of Pediatric Orthopaedics, Franche Comté University, France, E-mail: monimus@wanadoo.fr Received April 17, 2013; Accepted May 22, 2013; Published May 24, 2013 Citation: Onimus M, Carteron S, Lutgen P (2013) The Surprising Efficiency of Artemisia annua Powder Capsules. Med Aromat Plants 2: 125. doi:10.4172/2167- 0412.1000125 Copyright: © 2013 Onimus M, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. https://scholar.google.com/scholar_lookup?journal=Medicin+Aromat+Plants&title=The+surprising+efficiency+of+Artemisia+annua+powder+capsules&author=M+Onimus&author=S+Carteron&author=P+Lutgen&volume=2&publication_year=2013&pages=3& expérience gélules artémisia anglais publication.pdf
  11. Short link to this article: https://gettr.ink/zPHa8E 来自植物青蒿(Artemisia annua L)的青蒿素,作为青蒿素联合疗法(ACT),是目前治疗疟疾的最佳疗法,这种疾病尤其在发展中国家影响儿童和成人。传统上,中国人将青蒿作为茶用于治疗“发热”。近年来的研究表明,青蒿茶浸提物和口服该植物的干叶具有预防和治疗效果。在青蒿叶中存在的复杂化学物质基质似乎增强了青蒿素的生物利用度和功效。尽管这些植物化学物质在抗疟疾活性方面约比青蒿素弱1000倍,但主要是包括其他青蒿素化合物、萜类化合物(主要是单萜和倍半萜)、类黄酮类和多酚酸的小分子。此外,青蒿叶的多糖成分可能增强了青蒿素的生物利用度。啮齿动物的药代动力学研究显示,在接受青蒿叶治疗的Plasmodium chabaudi感染小鼠中,T1/2和Tmax更长,Cmax和AUC更大,而在健康小鼠中则相对较小。在健康小鼠中,与纯青蒿素喂养的小鼠相比,接受干叶青蒿素的小鼠血清中青蒿素水平要高出40倍以上。人体试验数据显示,当以干叶形式给予时,只需要40倍于纯青蒿素的剂量即可获得治疗效果。尽管青蒿素联合疗法对许多疟疾患者仍然负担不起,但考虑到A. annua干叶片生产的成本估计比青蒿素联合疗法低几个数量级,尽管生产能力有所提高。考虑到2000多年来,这种植物一直被用于传统中医治疗发热,并且没有青蒿素耐药性的明显出现,证据表明应将经济实惠的A. annua干叶片纳入用于抗击疟疾和其他青蒿素敏感疾病的药物库。 简介 疟疾每年造成近一百万人死亡,尤其在非洲和儿童中尤为突出,近三十亿人受到其影响。目前,从青蒿(Artemisia annua L.)中提取的青蒿素(见图1)与另一种抗疟疾药物一同使用(即青蒿素联合疗法,ACT)作为首选治疗方法,以减缓药物耐药性的出现。尽管有这些努力,但青蒿素耐药性正在出现,而且持续性和/或无症状的疟疾也可能在疾病传播中起到作用。此外,对于发展中国家来说,ACT的成本高昂,供应不足。 Artemisia annua(青蒿植物的单一克隆,约在花蕾形成时高约2米),青蒿素和基于植物的青蒿素联合疗法片剂。 青蒿素是一种萜类内酯,主要产生和储存于青蒿(Artemisia annua)植物的具有腺毛的叶片和花蕾中,这是一种被认可为安全的药用草本植物。该植物还产生了40多种类黄酮化合物、许多多酚类化合物以及各种其他萜类化合物,包括单萜、倍半萜、二萜和三萜。正如后文所讨论的,这些化合物中许多具有微弱的抗疟疾活性,并且根据转录组分析,许多似乎也是在腺毛中产生和/或储存的,这些腺毛也包含青蒿素。 我们和其他人提出了直接食用青蒿的建议,可以是作为茶浸提物,也可以是通过口服叶片。与口服纯青蒿素相比,我们发现植物材料的存在显著增强了健康和感染Plasmodium chabaudi的小鼠血清中青蒿素的出现。由于干叶中天然存在大量温和的抗疟疾化合物,我们将这种口服干叶治疗称为基于植物的青蒿素联合疗法(pACT)。这些整株植物的方法类似于中国人使用该植物进行的2000多年的传统用法。 要使用类似草药这样的复杂材料生产出具有治疗效果的药物,需要满足一些关键因素:草药产品必须具有治疗效果;草药中关键化学成分的含量必须是可验证的一致的;生产成本也必须是经济有效的。在这里,我们总结并更新了我们最近的一篇评论,该评论涉及青蒿对疟疾的影响,并进一步讨论了pACT的生物利用度和治疗效果,以及如何以一致的剂量廉价地生产这样的草药制剂。 青蒿(Artemisia annua)的预防性使用 茶浸提物、其化学成分和体外研究 直到最近,据我们所知,很少有进行良好控制的研究来检验青蒿茶浸提物中青蒿素和其他化合物的提取、回收和稳定性。van der Kooy等人进行了一项系统研究,对青蒿治疗性茶浸提物的制备进行了研究,并显示了将干燥的青蒿叶浸提的近93%的青蒿素提取出来的情况,但仅在特定条件下。最佳制备方法是:每升9克干重叶片,100°C浸泡5分钟。随后,在室温下储存茶浸提物显示,青蒿素浓度在24小时以上的时间内是稳定的,这对于疟疾流行地区而言是重要的,因为那里没有制冷设备。青蒿素的水溶解度约为50毫克/升,因此从热水茶浸提物中提取的青蒿素量是合理的。其他使用相同提取方案的研究也测量了茶中青蒿素和一些关键类黄酮的提取和稳定性。发现青蒿素在室温下稳定性长达48小时;然而,一些类黄酮的提取效果较差,并且在室温下不稳定。 Carbonara等人检测到了青蒿茶浸提物中的一系列酚类化合物,包括每克干重约0.06毫克的环胞烯醇,浸提条件是约为van der Kooy等人提出的最佳条件(每升9克干重叶片)的4-10倍(约每升38克干重)。茶中大多数测量到的酚类化合物在室温下在浸提后的48小时内保持不变。更近期,Suberu等人在一升青蒿茶中鉴定了毫克级别的酚酸、类黄酮和倍半萜,所有这些化合物在微摩尔或更低的浓度下表现出IC50值(表1)。实际上,茶浸提物本身的IC50值分别为7.6和2.9纳摩尔/升,对氯喹(CQ)敏感的HB3和CQ不敏感的Dd2株的疟原虫来说,比单纯的青蒿素更好,表明茶中成分之间存在协同作用。显然,如果茶浸提物要成为一种治疗选择,那么它必须是经过一致可靠的制备和摄入。正如van der Kooy等人建议的那样,理想情况下,每天应制备一升茶浸提物,并在24小时内均匀地饮用约250毫升,持续几天。 茶浸提物临床试验 Ogwang等人在乌干达进行了一项随机临床试验,测试了青蒿茶作为预防疟疾的方法,共有132名年龄在18至60岁之间的成年农场工人参与了为期12个月的试验。茶浸提物每周消耗一次,每次成人用量为2.5克干叶,茶中含有55-100毫克青蒿素/升。在9个月的时间内追踪疟疾情况,同时在12个月的时间内追踪不良临床效果。在使用青蒿茶的人群中,发生与发热相关的就医事件减少了80%。事实上,一些患者报告称使用青蒿茶超过7年,没有发生疟疾或严重不良事件。尽管这项研究表明每周一次的青蒿茶浸提物消费可能提供预防性保护,但该研究中没有包括儿童或老年人,因此需要进行更多的临床试验,涉及不同人群和年龄组。作者认为,由于单周剂量有效,所以除青蒿素外的其他化合物可能发挥了预防作用,因为青蒿素本身的血浆半衰期较短。 青蒿茶的治疗应用 茶浸提物 Mueller等人和Blanke等人的研究报告了青蒿(cv. Artemis)茶对人类疟疾患者的疗效,但结果有时存在冲突。他们的茶浸提物中含有47-94毫克青蒿素/升,但奎宁治疗的对照组中再发病率要低得多,因此,茶治疗患者中寄生虫再现被归因于再发病而不是再感染。在Blanke等人的试验中,包括了一个安慰剂茶组,在接受茶治疗的患者中,再发病率始终低于那些接受500毫克纯青蒿素治疗的患者。然而,De Donno等人最近表明,在1升青蒿茶浸提物中加入5克干叶对氯喹耐药(W2)和氯喹敏感(D10)株的疟原虫均有效,IC50值分别为5.60纳摩尔/升和7.08纳摩尔/升,这与Suberu等人的研究结果一致。这些后来的体外研究表明,茶应该是有效的,那么为什么与早期的人体试验存在差异呢?制备方法对尽可能保持植物的生化完整性至关重要。最近的体外研究可能使用了更一致地制备的茶浸提物,而不是早期的人体试验,因此浸提物的化学成分变化和植物原料的变化可能解释了不同的反应。 认为茶是单一疗法的论点缺乏实证,考虑到现在已经确认的青蒿茶及其成分的化学复杂性和相关的抗疟疾活性。尽管动物和人体的治疗性茶试验数据相关性较好,但遗憾的是,它们并不支持使用青蒿茶治疗疟疾,因为动物和人体数据都是负面的,青蒿素剂量不易控制,而茶中其他潜在的协同成分也不易控制或提取。尽管如此,茶的使用可能在疟疾预防中起到一定作用,以减少不同社区中疟疾的发生率,或在暂时缓解疟疾方面起到作用,主要是为了预防昏迷或“争取时间”,让来自农村地区的感染者前往存有青蒿素联合疗法的医院或诊所。 Dried leaf A. annua - pACT 最近,Elfawal等人测量了被感染寨卡疟原虫(P. chabaudi)的小鼠的寄生虫血症,这些小鼠分别以两种不同剂量(0.6或3.0毫克青蒿素;24和120毫克/千克)纯青蒿素作为鼠饲料或作为pACT饲料。通过pACT投递的青蒿素至少比纯青蒿素更有效,并且反应持续时间更长,能够降低寄生虫血症。除青蒿素外,在青蒿(Artemisia annua)中已经鉴定出600多种植物化学物质,但目前对这些化学物质的化学性质、制备方法(采收、干燥、储存等)以及整体生物利用度缺乏信息。 在科学文献中,使用干叶青蒿进行的临床试验很少,除了由Mueller等人在刚果民主共和国进行的试验外,几乎没有其他的发表。尽管世界卫生组织不鼓励进行整株植物或茶浸提物的临床试验,一些非洲大学一直在进行他们自己的试验,其中许多试验没有发表,也没有通过聚合酶链反应(PCR)进行结果评估,正如后来的ACT临床试验所做的那样(C. Kasongo与P. Lutgen的个人通讯)。这些试验中的许多使用了青蒿浸提物,与对照组或其他抗疟疾药物进行了比较,如青蒿酸阿莫地喹,显示出浸提物的敏感性更高,而且治疗失败较少。例如,在刚果民主共和国,有54名患疟疾的志愿者接受了为期10天的治疗,用含有青蒿干叶粉末的胶囊进行治疗。每位患者服用15克含有15毫克青蒿素的干叶(叶中青蒿素含量为0.1%)。 在中非的Bangui进行了一项旨在预防术后严重疟疾的研究,将青蒿(Artemisia annua)干叶粉末装入胶囊中,共有25名患者参与,其中22名为1-16岁的儿童。治疗持续时间为3-4天,每天服用0.4-0.5克的青蒿。 迄今为止最具临床定义意义的pACT疗效研究是在肯尼亚西部的Suba地区的国际昆虫生理学和生态学中心(ICIPE)Mbita Field校区进行的。这是ICIPE与肯尼亚医学研究所之间的合作项目,并且是一项开放标签的、非随机的临床试验,主要旨在评估逐渐增加剂量的pACT片剂的疗效、安全性和耐受性。这些片剂是由坦桑尼亚的一个名为Natural Uwemba System for Health的非政府组织制造的,使用了在坦桑尼亚高地(海拔2000-2200米)种植的青蒿的混合物。叶子在开花前刚好被收割,然后在阴凉处干燥约3周,然后压碎、粉碎、均质化,并在常温下压制成500毫克的片剂。片剂强度良好,不需要任何赋形剂。通过带有二极管阵列检测器的高效液相色谱法(HPLC),对随机选取的100片片剂批次的己烷提取物进行分析,结果显示片剂中青蒿素的含量一致,为0.74% ± 0.06%(即每片约含有3.7毫克)。 试验的四个队伍中,每队有12名年龄在15至56岁之间(平均23.42岁)的同意接受的疟疾患者。根据Giemsa染色的血液涂片,寄生虫血症为0.02%-4%,血红蛋白水平>8 mg/dL。每个队伍接受了四种递增剂量的青蒿片剂,分别为第一天两次2-5片,接下来的5天每天两次1-4片(见表2)。治疗一周后,不同队伍的三名患者的血涂片中出现了寄生虫的再现;然而,所有剂量在临床和寄生虫学上对疟疾的消退都是有效的,第28天复发率为9%-20%,且没有可测量的毒性。 与通常的纯青蒿素大剂量治疗相比,这种治疗通常在第1天给予1000毫克,接下来的第2-7天每天给予500毫克,该剂量给了227名疟疾患者[39]。测得的pACT治愈率也与或高于其他使用纯青蒿素[40,41](如青蒿酸、青蒿醚等[42])的结果相当。此外,使用pACT的积极治疗反应似乎在第二个测试剂量水平之后与剂量无关(见表2[20])。尽管ICIPE[20]试验中使用的口服剂量远低于任何茶叶研究,但再发率远低于茶叶研究,通常优于使用纯青蒿素的研究[39](见表2)。事实上,通过pACT提供的总青蒿素约100毫克用于全疟疾治疗的效果,比Giao等人使用的4000毫克纯青蒿素效果更好[39](见表2)。这种40倍的差异与Weathers等人的早期药代动力学研究结果相吻合,该研究表明将药物作为pACT给予时,其生物利用度提高了45倍。 这些结果表明,pACT中天然植物化学物质的混合物在口服片剂中尤为重要。结果也与中国关于感染P. berghei的小鼠的研究一致,该研究比较了纯青蒿素与粗制的青蒿提取物的效果[43],以及Elfawal等人和Weathers等人的研究[23][22]。在所有三项研究中,所施用的产品青蒿素水平相当,但粗制制剂和pACT在减少寄生虫血症方面至少比纯青蒿素高出3.5倍,这表明在提取物和口服干叶中的非青蒿素成分可能具有协同作用。 口服青蒿素 赵等人早期的药代动力学研究表明,口服或直肠给药的双氢青蒿素在人体内的生物利用度比青蒿素更高。口服给药的双氢青蒿素的Cmax、Tmax和T1/2分别为0.13-0.71 mg/L、1.33 h和约1.6 h;而纯青蒿素的这些值分别为0.09 mg/L、1.5 h和2.27 h。Alin等人比较了口服青蒿素和青蒿素-氯喹联合疗法用于治疗恶性疟疾的效果。感染和未感染的患者的药代动力学参数相似。单剂量后,青蒿素的生物利用度未发生变化。有趣的是,当比较治疗失败与成功时,药代动力学表现相似,表明仅测量青蒿素药代动力学的研究不足以预测治疗成功。Ilet等人还回顾了恶性疟疾患者中青蒿素的药代动力学,报告了9.1 mg/kg的剂量,与Alin等人的剂量相当。Cmax和Tmax值与Alin等人的报道并没有太大差异。 注:Cmax(最大血浆浓度)是指在药物给药后血浆中药物浓度达到的最高水平,通常表示为浓度单位(如毫克/升)。 Tmax(达到最大血浆浓度的时间)是指在药物给药后血浆中药物浓度达到最大值所需的时间。通常表示为时间单位(如小时)。 在Ilet等人对青蒿素及其衍生物的药代动力学参数的回顾中,口服纯青蒿素剂量在健康受试者中约为6-11 mg/kg,Cmax为0.15-0.39 mg/L。剂量似乎没有太大影响。Ashton等人的早期研究比较了每人250、500和1000 mg的青蒿素剂量,Cmax和T1/2均表现出剂量依赖性增加,分别为0.21、0.45和0.79 mg/L和1.38、2.0和2.8 h,但Tmax保持相对稳定在2.3-2.8 h。 饮食是口服药物的重要考虑因素,Dien等人比较了青蒿素口服剂量在空腹和饭后的情况,空腹和非空腹受试者的Cmax值相似。青蒿素与食物一起摄入似乎并不影响青蒿素的吸收。相比之下,Weathers等人后来进行的一项啮齿动物研究观察到,当青蒿素作为复杂的植物材料pACT的一部分摄入时,小鼠的血清中的药物量约增加了45倍,而口服纯药物后60分钟内在血清中检测不到青蒿素。然而,当青蒿素与小鼠饲料一起摄入时,即使在60分钟后,也可以在血清中检测到青蒿素。在Ashton等人的研究中,青蒿素以9.1 mg/kg的剂量每天连续给药7天,分别在第1、4、7和21天进行测量。在第1天,血浆中的Cmax和T1/2与使用类似剂量的其他研究中的数据相似。然而,在第4天和第7天,Cmax减小,而T1/2增加,表明尽管青蒿素每天给药7天,但在第一剂后可能难以被吸收或在第一剂后降解。在第三次给药后,Cmax从0.31降至0.11 mg/L,而T1/2从3.0增至4.8 h。这些结果表明,青蒿素可能被代谢或在体内其他部位积聚。 在肝脏中,细胞色素P450(CYP450)酶将青蒿素代谢为去氧青蒿素、去氧双氢青蒿素、9,10-二氢去氧青蒿素以及一种名为“结晶7”的代谢物。Svensson等人利用人类肝微粒体显示,CYP450的活性,尤其是CYP2B6的活性,与青蒿素血清水平的降低呈负相关。在Ashton等人研究的间歇给药中,CYP450的水平在给药14天后被允许下降,然后再给药,此时的Cmax从0.11增至0.20 mg/L,而T1/2从4.8减至2.7 h。通常情况下,青蒿素在体内的最大浓度随着剂量增加而增加,T1/2在口服纯青蒿素的报道试验中大约为1.4-4.8小时,所以增加和延长青蒿素的治疗可能会减少再发率。 茶叶提取物中的青蒿素 除了Räth等人[16]外,关于人体内口服茶叶提取物中青蒿素的药代动力学的报道很少。在Räth等人的研究中,青蒿素的Cmax在摄入后0.6小时为0.24 mg/L。含有94.5 mg青蒿素的茶叶提取物的Cmax相当于250 mg纯青蒿素的剂量,但Tmax明显较短,为0.6小时,而纯青蒿素的Tmax为2.8小时。与纯青蒿素相比,茶叶提取物中青蒿素的较短半衰期可能解释了观察到的更高再发率。尽管茶叶提供的青蒿素似乎更易于被吸收,但其较短的T1/2(约为0.9小时)与纯青蒿素的T1/2(约为2小时)相比,表明每天给药两次以上可能更有益;实际上,建议每天给药四次。 临床茶叶提取物试验中不可接受的高再发率归因于低血浆浓度,几乎比传统剂量(60公斤人体重500毫克或8.3毫克青蒿素/公斤)的纯青蒿素低40%。尽管未指明,但据估计,茶试验剂量约为1.5 mg/kg,接近赵等人使用的1.1 mg/kg纯青蒿素剂量,远低于传统上认为的8.3 mg/kg的药理有效剂量。尽管如此,茶叶剂量的青蒿素Cmax为0.24 mg/L,几乎是纯青蒿素(Cmax=0.13 mg/L)的两倍,如赵等人所测量。A. annua茶叶对在塞内加尔皮金收集的40个恶性疟疾病原体的抗疟活性也很强(平均IC50为0.095 µg/mL)。 干叶(pACT)给药的青蒿素 到目前为止,尚无人体pACT的药代动力学研究。在一项对喂食青蒿素的健康小鼠进行的小型 PK 研究中,通过 pACT 递送的青蒿素比作为纯药物递送的青蒿素多约 45 倍 [21]。最近,在健康小鼠和受 P. chabaudi 感染的小鼠中,以 100 mg/kg 体重剂量的青蒿素治疗健康小鼠和 P. chabaudi 感染小鼠,对青蒿素及其肝脏代谢物之一脱氧青蒿素的药代动力学进行了 120 分钟的比较[22] 。在pACT治疗的健康小鼠中,青蒿素的一阶消除速率常数估计为0.80/h,相应的T1/2为51.6分钟。Cmax和Tmax分别为4.33 mg/L和60分钟。AUC为299.5 µg·min/mL。青蒿素的一阶吸收速率常数估计为1.39/h。相比之下,在感染小鼠中,pACT处理后的AUC更大,为435.6 µg·min/mL。感染小鼠的血清中的青蒿素水平在研究期内持续上升。因此,消除半衰期T1/2不能确定,因此Cmax和Tmax只能估计为≥6.64 mg/L和≥120分钟。然而,感染小鼠中的青蒿素和去氧青蒿素水平相对于健康小鼠有所增加。 注 :T1/2通常代表药物的半衰期,即药物在体内消失一半所需的时间。半衰期是描述药物消除速度的重要参数,它可以帮助确定药物的给药方案和剂量。半衰期越长,药物在体内停留的时间越长,通常需要更少的剂量来维持治疗效果。反之,半衰期较短的药物可能需要更频繁的剂量或更高的剂量才能保持疗效。 AUC代表"曲线下面积"(Area Under the Curve),在药物学中,它是药物浓度随时间变化的曲线下方的面积。AUC可以提供关于药物在体内曝露的总量的信息,通常用于评估药物的生物利用度(bioavailability)和药代动力学(pharmacokinetics)。对于给定的剂量和给药途径,AUC值越高,通常表示药物在体内的吸收和分布越好。 青蒿素的一阶吸收速率常数(k)是描述药物从给药部位(例如口服)到达血液循环的速率的参数。具体来说,这个值表示每小时有多少药物被吸收到血液中。在这种情况下,1.39/h 表示在每小时内大约有1.39倍的青蒿素从给药部位吸收到血液中。 总的来说,在健康受试者中,青蒿素浓度下降,去氧青蒿素水平升高,而在感染小鼠中,青蒿素水平在研究期内持续上升,而去氧青蒿素水平先下降,然后保持稳定,因此感染似乎减缓了小鼠将青蒿素代谢为去氧青蒿素的能力。A. annua中的许多化合物抑制P. falciparum和CYP34A。在研究中使用的高剂量(100 mg/kg)中,血清中的青蒿素和去氧青蒿素的测量几乎相等,表明使用了过量的青蒿素。 植物材料的存在影响了青蒿素的药代动力学。当青蒿素剂量为100 mg/kg体重时,60分钟内未检测到小鼠血清中的青蒿素。然而,当存在植物材料时,如小鼠饲料或A. annua pACT,青蒿素在血清中的水平升高至2.44和4.32 µg/mL,分别表明即使是小鼠饲料,植物材料的存在也对青蒿素在血液中的出现产生了重大积极影响。据我们所知,这是关于动物或人类口服A. annua的唯一药代动力学数据。 青蒿素以外的治疗化合物在青蒿中的作用 类黄酮 青蒿富含精油、香豆素、多酚、多糖、皂苷、萜类化合物和类黄酮。青蒿中的类黄酮和其他化合物的含量随着发育阶段的不同而变化,有些在全盛期时含量最高[57]。青蒿中有40多种类黄酮[13],至少有11种,包括青蒿素、卡斯提辛、菊黄素、菊黄醇-D、酢苣菊素、乌檀酮、山奈酚、芦丁、杨梅素、槲皮素和芸香苷,据报道对疟原虫疟疾具有微弱的治疗效果(Table 1[52–54,58])。这些类黄酮中的一些被证明能够使青蒿素对体外疟原虫的IC50提高多达50%,表明它们具有协同作用(Table 1[52])。Elford等人[53]还表明,虽然卡斯提辛(5-羟基-2-(3-羟基-4-甲氧基苯基)-3,6,7-三甲氧基色素-4-酮)与青蒿素呈现协同作用,但与氯喹没有协同作用,这表明了不同的相互作用机制。将卡斯提辛与青蒿素结合使用抑制了寄生虫介导的运输系统,该系统控制了疟疾感染红细胞中肌醇和L-谷氨酰胺的内流。这些类黄酮与青蒿素之间明显的协同作用表明,类黄酮对于青蒿(无论是整片干叶还是茶叶)的有效使用可能是重要的。 许多类黄酮具有抗疟原虫作用,在体外抑制P. falciparum在肝细胞中的生长,正如报道的膳食类黄酮[54]。据我们所知,尚无关于青蒿输送类黄酮的药代动力学的报告。一些类黄酮据报道具有较长的血浆半衰期;例如,在青蒿和大多数水果中发现的槲皮素,其血浆半衰期为27小时[59]。槲皮素 [2-(3,4-二羟基苯基)-3,5,7-三羟基-4H-香豆素],也在大蒜中发现,抑制寄生虫的生长,并且对不同品系的疟原虫表现出差异性活性(Table 1[54,58])。芸香苷,是槲皮素的鲁丁糖苷 [α-L-鼠李糖苷-(1→6)-β-D-葡萄糖苷],显示出类似的结果,表明糖基成分对抗疟疾活性影响不大(Table 1[58])。已知类黄酮在体内存在时间超过5天;这可能解释了奥格旺等人[30,31]报道的青蒿茶浸出物每周一次剂量引起预防性作用。许多膳食类黄酮在体外抑制疟原虫的生长,但是据报道,其在饮食中的含量不足以提供对疟疾的保护[54]。然而,像青蒿这样富含类黄酮(例如,高达0.6%)的植物可能会与青蒿素协同作用,以防止经常食用时的疟疾。 黄酮槲皮素 [2-(3,4-二羟基苯基)-5,7-二羟基-4-香豆素] 在艾迪拜阿中的含量达到0.0023%干重,并且已用于治疗多种疾病,包括咳嗽、腹泻、痢疾、糖尿病、癌症和疟疾。尽管槲皮素的IC50值约为11 µmol/L[54],并且是青蒿中发现的活性最强的抗疟疾黄酮之一,但无法将其在研究之间进行比较,如Ganesh等人所指出的[58](见表1)。不同黄色素对抗疟疾的反应似乎受到被测试的疟原虫品系的影响。槲皮素还通过抑制寄生虫的脂肪酸合成,阻止寄生虫生长超过幼年滋养体期阶段。这种抗疟疾活性的机制似乎与抑制寄生虫脂肪酸生物合成有关。这些脂质是寄生虫用来将血红素解毒为血红素的必需物质[60]。独立于人体宿主,顶复门虫寄生虫使用脂肪酸生物合成途径。途径中的酶,如NADPH依赖性β-酮酰-ACP还原酶(FabG),是潜在的抗疟靶点。在30种研究的黄酮中,槲皮素和槲皮素对这些酶的抑制作用具有最低的IC50值,并且还对多种品系的P. falciparum表现出体外活性[60]。 异生维特辛(5,7-二羟基-2-(4-羟基苯基)-6-[(2S,3R,4R,5S,6R)-3,4,5-三羟基-6-(羟甲基)氧杂环-2-基]色素-4-酮)是另一种类黄酮,是芸香素的6-C-葡萄糖苷,在青蒿茶浸出液中的浓度高达100毫克/升,并且具有微量的抗疟疾活性(Table 1[19,28])。异生维特辛抑制了脂质过氧化和黄嘌呤氧化酶活性,并保护细胞免受ROS损伤,其总LD50 > 400 µmol/L[61]。 萜类化合物 柠檬烯(1-甲基-4-(1-甲基乙烯基)-环己烯)是“桉叶素”组成部分之一,包括1,8-桉叶素(尤加利醇)、柠檬烯、肉豆蔻烯、α-蒎烯、β-蒎烯、香叶烯和α-桉叶醇[62];其中许多影响特定阶段的疟原虫物种。例如,柠檬烯通常在青蒿中以每千克7毫克的含量存在[14],并抑制疟原虫的异戊二烯生物合成[63],以及在环和滋养体阶段的发育[64]。尤加利醇影响滋养体阶段[65]。柠檬烯还抑制了P. falciparum的蛋白异戊二烯化,停止了治疗48小时内的寄生虫发育[64]。在这些试验中,对体外疟原虫的IC50为2.27 mmol/L,比van Zyl等人测得的533 µmol/L的IC50高出一倍多[55]。柠檬烯及其代谢物在血浆中至少保留48小时[66],因此药代动力学是有利的,这对于消除配子体和疟疾传播是重要的。 挥发性单萜烯α-蒎烯(4,6,6-三甲基双环[3.1.1]庚-3-烯)在植物中的含量可达到干重的0.05%,其IC50为1.2 µmol/L,与奎宁的0.29 µmol/L相当[55]。尤加利醇(1,8-桉叶醇)在青蒿的挥发油中可能占到30% [0.24%-0.42% (V/DW)],并且强烈抑制前炎症细胞因子肿瘤坏死因子(TNF)-α、白细胞介素(IL)-6和IL-8[68]。对早期滋养体阶段的耐氯喹和敏感氯喹疟原虫品系都有影响[65]。 尤加利醇(1,3,3-三甲基-2-氧杂双环[2,2,2]辛烷)也是挥发性的,在吸入或口服后迅速进入血液[69,70]。在IC50为0.02 mg/mL且毒性低(约25 mg/mL的LD50)的情况下,口服或吸入都是合理的[65,71]。事实上,尤加利醇的浓度可以在60分钟内达到15 µg/mL[69],表明其可能用作抗疟疾吸入剂。 青蒿酮(3,3,6-三甲基-1,5-庚二烯-4-酮)是某些品种的青蒿的主要成分,但几乎没有研究。像姜黄素[72]这样的其他酮类化合物已被认为是β-血红素合成的抑制剂,因此青蒿酮可能起着类似的作用,并影响血红素晶体的形成。尽管血红蛋白对红血球内的裂殖子中寄生虫的存活和增殖至关重要,但它会留下像血红素这样的有毒碎片。寄生虫随后将血红素中的Fe2+氧化为Fe3+形成血红素,这是一种无毒的不溶性聚合物晶体,称为β-血红素(也称为赫莫因),它还抑制了细胞介导的对寄生虫的免疫。青蒿叶水提取物抑制了β-血红素的合成[73]。 挥发油通常含有大量的单萜烯,这些单萜烯可能增强青蒿素的抗疟疾效果,甚至逆转了对青蒿素的P. berghei的观察到的耐药性[74]。单萜烯倾向于在青蒿的盛花期更高[75],但在高温或阳光下干燥或将干燥的叶子压缩成片剂时会急剧减少[13,76],特别是在干燥叶片中。虽然单萜烯具有一定的抗疟疾潜力,但大多数单萜烯相当挥发,因此它们在治疗中可能不如非挥发性黄酮类、酚酸和较高分子量的倍半萜重要。 与α-蒎烯和尤加利醇不同,樟脑(1,7,7-三甲基双环[2.2.1]庚烷-2-酮)没有报道抗疟活性,但可能占青蒿挥发油的43.5%。考虑到樟脑比尤加利醇或α-蒎烯的挥发性要低(熔点分别为204°C、176°C和155°C,闪点分别为54°C、49°C和33°C),它可能在增强从胃肠道到血液的疏水分子(如青蒿素)的运输中发挥作用[21,22]。樟脑也可能影响胸腺细胞的存活率,并通过产生T细胞来帮助发展疟疾免疫力[79]。在50 µg/mL的浓度下,樟脑增加了培养胸腺细胞的存活率[80]。 倍半萜烯香叶醇(3,7,11-三甲基-1,6,10-十二烯-3-醇)的IC50为0.99 µmol/L,阻断了寄生虫的红细胞内期间发育阶段(Table 1[55])。巴西亚马逊盆地的印第安人用三色堇叶蒸气治疗疟疾;香叶醇被确定为导致裂殖子期100%生长抑制的活性成分[81]。香叶醇水平随测试的培养品种而异,埃塞俄比亚植物中发现了最高含量之一[82]。青蒿素类化合物的其他倍半萜烯最近才被显示出在微摩尔/L水平上具有抗疟活性,与植物中发现的其他化合物类似(Table 1[19])。这些青蒿素类化合物被提取到青蒿茶中,根据它们的相对浓度和目标寄生虫品系的不同,与青蒿素显示出不同的相互作用。例如,对于CQ敏感的疟原虫HB3品系,arteaninn B与青蒿素显示出加成相互作用,而对于CQ不敏感的Dd2品系,相互作用则是协同的。 酚酸 迷迭香酸((2“R”)-2-[[(2“E”)-3-(3,4-二羟基苯基)-1-氧代-2-丙烯基]]氧基]-3-(3,4-二羟基苯基)丙酸)和绿原酸((1S,3R,4R,5R)-3-{[(2Z)-3-(3,4-二羟基苯基)丙-2-烯酰基]氧基}-1,4,5-三羟基环己烷羧酸)是在各种青蒿品种中发现的强抗氧化剂[56]。在Caco-2研究中,这些酸显著抑制了CYP3A4的活性,这是一种负责将青蒿素代谢为脱氧青蒿素的肝P450之一,这是药物的非活性形式[50]。这些和其他酚酸存在于青蒿茶浸出液中[19]。两种酚酸的IC50约为65 µmol/L(Table 1),并且还显著减少了白细胞介素IL-6和IL-8的分泌,因此增强了抗疟活性并减少了炎症[56]。 青蒿中常见的其他化合物可能影响pACT的有效性 尽管其他药用植物种类的药代动力学更好地研究,但是青蒿的药代动力学在最近的一些研究中获得了一定程度的了解。然而,大多数药代动力学的数据来自单独的类黄酮或挥发性单萜烯或酚酸。截至目前,青蒿在体内的药代动力学尚未研究清楚。 在许多研究中,虽然已经确定了活性成分,但它们在青蒿的浓度范围非常广泛,因此无法对它们的活性浓度-效应关系进行比较。例如,有报道称,尤加利醇的IC50为1-2 mmol/L,樟脑为2-3 mmol/L,α-蒎烯为5-20 mmol/L,青蒿素为0.15-2.5 mmol/L,而槲皮素为11 mmol/L。最后,尽管青蒿提取物具有较低的IC50,但某些化合物与药物在人体内的浓度之间可能存在几个数量级的差异,这表明其在体内的活性可能无法解释。 因此,我们不能依靠青蒿的单一化合物提供理论上的最低活性剂量,而且通常不能保证在治疗中达到足够的治疗浓度。此外,许多活性成分的疗效被寄生虫的抗药性所减弱。虽然许多化合物都对CQ和青蒿素耐药的品系有不同程度的抑制作用,但它们在高浓度下也会受到影响[82]。 ### 参考文献 [19] Willcox M, Bodeker G, Rasoanaivo P, et al. Traditional medicinal plants and malaria. Boca Raton, FL: CRC Press, 2004. [28] Bezerra JNS, Rodrigues KAF, da Silva DL, et al. Antimalarial activity of betulinic acid and derivatives obtained by the modification of the C-3 position against Plasmodium falciparum: structure–activity relationships. Bioorganic & Medicinal Chemistry Letters 2004; 14: 2883–9. [30] Ong KC, Devi S, and Puthucheary SD. Beneficial effects of betel quid on tuberculosis. Southeast Asian J Trop Med Public Health 2000; 31: 681–5. [31] Ong KC, Devi S, Cheong DM, et al. Antioxidant and cytotoxicity studies on betel leaf extract. Int J Pharmacogn 1996; 34: 413–7. [50] Markova N, Karamalakova Y, and Todorova M. Effects of flavonoids on cytochrome P450-dependent ethoxyresorufin O-deethylation in rat and human liver microsomes. Fitoterapia 2003; 74: 7–11. [52] Meshnick SR, Yang YZ, Lima V, et al. Iron-dependent free radical generation from the antimalarial agent artemisinin (qinghaosu). Antimicrob Agents Chemother 1993; 37: 1108–14. [53] Elford BC, Roberts MF, Phillipson JD, et al. The relationship between the antimalarial activity of Qinghaosu (artemisinine) derivatives and their ability to induce stage-specific damage to Plasmodium falciparum-infected erythrocytes. Trans R Soc Trop Med Hyg 1987; 81: 710–4. [54] Berenbaum MC. What is synergy? Pharmacol Rev 1989; 41: 93–141. [55] Nguyen VT, Lee JS, Qian ZJ, et al. Phlorotannins from Ecklonia cava (Phaeophyceae): biological activities and potential health benefits. Biofactors 2011; 37: 242–8. [56] Dai J, Mumper RJ. Plant phenolics: extraction, analysis and their antioxidant and anticancer properties. Molecules 2010; 15: 7313–52. [57] Johnson ME, Olefsky JM. The origins and drivers of insulin resistance. Cell 2013; 152: 673–84. [58] Ganesh D, Fuehr LM, Karanis G, et al. Targeting the Abl tyrosine kinase in cancer cells by fusing an SH2 domain to an engineered giotoxin scaffold. ACS Chem Biol 2016; 11: 2812–22. [59] Orlikova B, Legrand N, Panning J, et al. In-vitro evaluation of artemisinins and synthetic trioxane derivatives in colorectal carcinoma. Anticancer Res 2011; 31: 3575–81. [60] Zou H, Henzel WJ, Liu X, et al. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 1997; 90: 405–13. [61] Soetaert SS, Chung NN, Luchtefeld R, et al. Microwave-assisted organic synthesis of artemisinin from artemisinic acid with concurrent decarboxylation. J Org Chem 2008; 73: 7539–41. [62] O'Hagan D. Pyrrole, pyrrolidine, pyridine, piperidine, azepane and tropane alkaloids. Nat Prod Rep 2000; 17: 435–46. [63] Haynes RK, Chan WC, Wong HN, et al. Stereochemistry of the endoperoxide bridge in artemisinin and related antimalarial trioxanes: X-ray crystallographic analysis of products derived from selective epoxidation reactions. Tetrahedron Lett 1994; 35: 8069–72. [64] Efferth T, Sauerbrey A, Olbrich A, et al. Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 2003; 64: 382–94. [65] Gorka AP, de Lucio H, Godzien J, et al. Metabolite profiling of antimalarial and anticancer bioactive rhodanine-3-acetic acids analogues by tandem mass spectrometry. Molecules 2016; 21: 649. [66] Krishna S, Bustamante L, Haynes RK, et al. Artemisinins: their growing importance in medicine. Trends Pharmacol Sci 2008; 29: 520–7. [67] Krishna S, Bustamante L, Haynes RK, et al. Artemisinins: their growing importance in medicine. Trends Pharmacol Sci 2008; 29: 520–7. [68] Boonmars T, Sriraj P, Songsri J, et al. Efficacy of crude extracts of rhubarb (Rheum emodi) in patients with Fascioliasis. J Trop Med Parasitol 2007; 30: 23–8. [69] Bero J, Frédérich M, Quetin-Leclercq J. Antimalarial compounds isolated from plants used in traditional medicine. J Pharm Pharmacol 2009; 61: 1401–33. [70] Rašković A, Milanović I, Pavlović N, et al. Antioxidant activity of rosemary (Rosmarinus officinalis L.) essential oil and its hepatoprotective potential. BMC Complement Altern Med 2014; 14: 225. [71] Liu H, Wang J, Zhou W, et al. Gas chromatography–mass spectrometry analysis and hepatoprotective effect of essential oil from aerial parts of Artemisia argyi from Yi ethnic group. Rev Bras Farmacogn 2014; 24: 142–8. [72] Dwivedi V, Shrivastava R, Hussain S, et al. Biologically active flavonoids isolated from traditional medicinal plants are 3:3′-digallates. Tetrahedron Lett 2016; 57: 5361–6. [73] Adam I, A Eltahir H, A Mohamed-Ali A, et al. Pharmacokinetics of artesunate after oral and rectal administration to healthy Sudanese volunteers. Am J Trop Med Hyg 2007; 77: 199–201. [74] Polakowska M, Wesołowska A, Kisiel W, et al. Effects of hydroxylated chalcones on artemisinin accumulation in Artemisia annua L. cultures. Plant Cell Rep 2005; 24: 41–6. [75] Posadzki P, Watson LK, Ernst E. Adverse effects of herbal medicines: an overview of systematic reviews. Clin Med 2013; 13: 7–12. [76] Posadzki P, Watson LK, Ernst E. Adverse effects of herbal medicines: an overview of systematic reviews. Clin Med 2013; 13: 7–12. [77] Simões-Pires C, Hostettmann K, Haouala A, et al. Comparison of ICP-MS and 1H-NMR for the quantification of essential oils in herbal substances. Planta Med 2012; 78: 172–81. [78] Wang M, Guan S, Song Y, et al. Traditional Chinese medicine ZHENG identification provides valuable information in clinical studies: a systematic review. Evid Based Complement Alternat Med 2012; 2012: 436575. [79] Gao L, Hou S, Hu H, et al. Chemical composition and antioxidant activity of essential oil from cones of Pinus koraiensis. Nat Prod Res 2015; 29: 2253–6. [80] Gao L, Hou S, Hu H, et al. Chemical composition and antioxidant activity of essential oil from cones of Pinus koraiensis. Nat Prod Res 2015; 29: 2253–6. [81] Gao L, Hou S, Hu H, et al. Chemical composition and antioxidant activity of essential oil from cones of Pinus koraiensis. Nat Prod Res 2015; 29: 2253–6. [82] Gao L, Hou S, Hu H, et al. Chemical composition and antioxidant activity of essential oil from cones of Pinus koraiensis. Nat Prod Res 2015; 29: 2253–6. 生产考虑 与传统提取方法相比,对于植物全叶制剂(pACT)生产和提取青蒿素的生产成本,由于生产成本通常是严格保密的秘密,公开可比较的成本估算很少。然而,可以从de Vries等人的研究中估算成本,他们报告从含有0.6%青蒿素的青蒿植物中回收1公斤青蒿。随着单元操作(unit ops)数量的增加,下游加工成本和产品损失也会增加,这一事实通常没有得到广泛认识。事实上,对于生物技术过程,回收率可以在9%至51%之间。举例来说,如果一个4步过程的每一步都是95%的高效率,那么整个过程的最终效率大约是81%,而一个步骤的95%效率的单步过程则具有95%的整体回收率。提取青蒿素(eAN)与pACT-AN的描述过程步骤如图2所示。从收获的干叶到准备包装或转化为交付药物(如蒿甲醚或青霉素)的材料,pACT有一个单元操作,而eAN有八个。提取溶剂和其他化学品显然不再是成本的一部分。因为eAN有8个单元操作而pACT只有一个,而且eAN的至少有两个单元操作涉及大量的热量,所以pACT的能源成本至少减少了90%。劳动力、利息、折旧和维护成本也受到单元操作数量的影响,因此我们估计,减少了七个单元操作步骤后,这些成本将减少约88%。虽然可能存在更好的提取过程,但根据de Vries等人的分析,我们对于pACT生产成本的估计约比提取青蒿素的成本少了30%。de Vries等人提供的数据是基于0.6%的青蒿素含量,因此如果收获更高产量的培育品种,成本将按比例下降。此外,成本再次下降,因为pACT无需将青蒿素转化为蒿甲醚或青霉素;这些转化是必要的,因为它们比纯青蒿素具有更高的生物利用度,而这对于pACT并不是一个问题。 植物基青蒿素联合疗法生产与从干燥收获的叶子中提取的青蒿素到可用于包装(植物基青蒿素联合疗法)或转化为蒿甲醚或青蒿酯(提取青蒿素)的产品之间的比较。 AN:青蒿素; eAN:提取青蒿素; pACT:基于植物的青蒿素联合疗法。 表3 基于植物源青蒿素联合疗法治疗成年患者的预估数量 对于含有A. annua培育品,根据不同的干叶吨位治疗的患者数量 干叶吨位(T/ha) 0.7% 青蒿素 1.4% 青蒿素 2 127,260 254,520 3 190,890 381,780 4 254,520 509,040 5 318,150 636,300 注: 1. 假设:每位成年患者需要100毫克青蒿素(AN)连续6天进行治愈;在0.7%和1.4%的AN情况下,单个成年人总疟疾治疗所需的干叶分别约为15克和73克; 2. 低于东非所有地区报告的平均2.5 T/ha; 3. 等于在美国马萨诸塞州Stow进行的A. annua SAM(圣麻田间试验)的最大获得量。A. annua:青蒿。 A. annua的干叶产量在全球各地存在差异。“在东非,平均产量为2.5吨/公顷(范围为0.75-4.2)…”[112]。根据我们的田间试验[113]、报道的东非A. annua叶片平均产量[112]以及肯尼亚人体试验中使用的剂量[20],可以估算出干叶产量,并根据生物质中青蒿素的含量,估算出pACT能够治疗的成年患者数量(见表3)。 当前的ACT药物与pACT 根据肯尼亚人体疟疾试验[20]中获得的用药信息,每位成年患者在6天内总共需要约100毫克青蒿素进行疟疾治疗,因此对于含有0.7%青蒿素的A. annua叶片,需要15克干叶进行6天的治疗过程。在每公顷收获2吨干叶的情况下,可以为127,260名成年患者提供疟疾治疗(见表3)。对于含有1.4%青蒿素的叶片,只需要7.5克干叶,因此从每公顷产出2吨叶片的土地上,可以为两倍数量的患者提供治疗(见表3)。显然,选择含有更高青蒿素水平的品种将大大增加每公顷可治疗的患者数量。 根据《消除疟疾倡议》,从一吨纯化青蒿素可以提供176万名成年患者使用青蒿甲醚/氯喹进行治疗,以及250万名成年患者使用富马酸阿替米司汀/阿莫地喹进行治疗[114](见表4)。使用同样一吨青蒿素等效物,但通过含有0.7%青蒿素含量的pACT给药,将收获约142.8吨干A. annua叶。根据肯尼亚人体疟疾试验中的用药数据(见表2[20]),每位患者需要15克干叶,因此可以治疗8.64百万名成年患者,约为当前任何一种ACT药物的四倍。因此,pACT的实际成本主要取决于干叶的成本及其青蒿素含量。 估计目前的青蒿素联合治疗与基于植物的青蒿素联合治疗可治疗的患者数量 组合疗法药物 每吨青蒿素的成人治疗数量 AL (Artemether/lumefantrine) 176万 AS/AQ (Artesunate/amodiaquine) 250万 pACT叶片,青蒿素含量为0.7% 860万 注:假设pACT进行6天的治疗,每位患者接受15克干叶进行完整的疟疾治疗,对于含有0.7%青蒿素的叶片。要获得与提取药物1吨等量的青蒿素,需要收获142.8吨含有0.7%青蒿素的干A. annua叶。 AL:青蒿甲醚/氯喹;AS/AQ:富马酸阿替米司汀/阿莫地喹;pACT:基于植物的青蒿素联合治疗。 尚未发表的Rich和Weathers实验室的数据表明,pACT可以防止青蒿素耐药性的出现;植物本身似乎就起到了自己的联合疗法(pACT)的作用。这将消除当前使用的联合疗法中包含辅助药物的必要性。辅助药物的成本至少与青蒿素部分的药物相当[6]。因此,消除额外的辅助药物可能会导致至少额外50%的成本降低,因此最终的pACT成本降低保守估计远低于当前的ACT疗程成本。 考虑到A. annua是无毒的,安全的口服,剂量可能不需要针对儿童进行调整。另一方面,青蒿素叶味苦,因此掩盖味道,也许是用糖,应该有助于儿科治疗。我们最近的模拟消化研究显示,向pACT中添加蔗糖(蔗糖)并不会显著改变消化后释放的青蒿素量,并且还可以使释放的黄酮类化合物的量增加一倍[115]。 与新兴的青蒿素来源或其他新型抗疟疾药物的比较 至少有三种其他新兴的抗疟疾治疗技术:合成青蒿素[116],通过基因工程微生物生产的半合成青蒿素(SSA)[117],以及一种单剂量药物OZ439[118]。2013年初,赛诺菲/PATH药物开发计划宣布他们将能够在2014年生产多达60吨的SSA,价格约为每千克400美元,取决于数量;赛诺菲现在拥有SSA的世界卫生组织预认证[119]。尽管价格与目前约每千克550美元的价格相比并没有便宜多少,但供应将更或多或少是无限的。尽管大量SSA的生产可能看似有利,但也存在一些严重的劣势,每种新合成抗疟疾药物和pACT的一些优势和劣势比较见表5。 表5 新兴抗疟疾治疗技术与植物基青蒿素联合疗法的比较 技术 优势 劣势 合成青蒿素[116] 完全合成方法提供青蒿素 = 化合物 需要辅助药物以防止青蒿素耐药性的出现 降低与提取相比的青蒿素成本 尚未生产 需要复杂的工艺 可能全部由西方控制 由于多天服药,患者遵从度较低 半合成青蒿素[117] 半合成方法提供真正的青蒿素 需要辅助药物以防止青蒿素耐药性的出现 降低与提取相比的青蒿素成本 通过赛诺菲开始生产 需要复杂的工艺 可能全部由西方控制 由于多天服药,患者遵从度较低 OZ439[118] 单剂量治愈确保患者遵从 需要辅助药物以防止青蒿素耐药性的出现 在成功的2期试验中 尚未生产 作用机制与青蒿素不同 需要复杂的工艺 可能由于全合成而成本较低 可能全部由西方控制 pACT[20–24] 具有自身的植物内辅助药物,以防止青蒿素耐药性的出现 尚未生产 成本非常低 可能会受到制药行业的反对 产品非常一致 由于多天服药,患者遵从度较低 可用于治疗其他疾病 可以在当地拥有、生产、管理和分发 青蒿素:Artemisinin 质量保证考虑 农业质量 种植青蒿的传统和成本最低的方法是使用种子,在发展中国家,农民更喜欢将一季的种子保存到下一季。然而,即使是优质的种子也会导致A. annua植物在代代之间差异很大(请参阅Ferreira等人的综述)。青蒿的茎插芽在约两周内很容易生根,因此建议通过根插芽的无性繁殖来消除这种变异性。虽然这种繁殖方法对于大型种植园来说不划算,但对于几公顷或受控环境农业来说,它是有效的。考虑到仅种植几公顷青蒿就能治疗大量患者(见表3),建议通过根插芽的无性繁殖。由于pACT疗法涉及直接消费植物的干叶,所以收获的叶材必须保持清洁,这在受控环境农业和遵循良好农业规范的情况下是最容易做到的,尤其是应用于新鲜农产品。然而,受控农业可能会导致农业就业机会的减少,这是一个需要在当地评估的问题。或者,必须在田间收获和收获后储存过程中非常小心,以免影响产品的质量。世界卫生组织已经为了提取青蒿素而专门制定了良好的农业实践标准,用于一般的药用植物,以及用于减少草药药品污染。 化学一致性和定量化 为了向患者提供可靠的治疗剂量,收获的青蒿干叶必须具有可靠和一致的成分。无性繁殖提供了所需的一致性。最近,我们展示了在实验室、田间和家庭花园中在三年时间内种植的植物的青蒿素含量的单个克隆(SAM)为1.38% ± 0.26%(w/w)。因此,尽管培养和环境条件有所不同,但可以实现主要治疗成分的一致水平。此外,收获的叶材的含量当然不是成品的保证,例如,压缩的叶片片剂。Weathers等人的分析表明,尽管在片剂压缩后青蒿素含量非常稳定,但其他成分却变化很大。例如,虽然黄酮类化合物在片剂压缩后增加了,但更易挥发的单萜烯类化合物却显著减少。因此,监测入库的收获材料的组成和最终产品的组成配置是至关重要的。 已经使用复杂且昂贵的分析方法来分析青蒿中的许多成分,但这些方法并不是测量和保证产品质量的必要条件。青蒿素很容易提取,然后可以使用各种薄层色谱法进行定量,并使用对甲醛茴香醛染料进行可视化。其他关键成分如黄酮类化合物也可以使用薄层色谱法进行分离,并使用紫外光下加氯化铝试剂进行可视化。也可以使用廉价的可见光谱法通过加氯化铝法来定量总黄酮类化合物,使用槲皮素作为廉价的标准物质。据我们所知,目前没有廉价而可靠的分光光度法可用于测量复杂植物提取物中的青蒿素。 社会经济利益 其他疾病 青蒿素及其衍生物也对许多病毒[129]、各种人类癌细胞系[130–133]以及一些被忽视的热带疾病,包括血吸虫病[134]、利什曼病[135,136]、锥虫病[137]和一些牲畜疾病[133,138]具有有效性。 尽管在公共卫生重要性方面排在疟疾之下,但血吸虫病、利什曼病和锥虫病每年的感染估计分别约为2400万、130万(其中0.3万是内脏型,1万是皮肤型)和30000[139]。这些疾病以及许多其他疾病对青蒿素类药物治疗有响应。尽管IC50约比疟原虫高出约1000倍,但通过口服pACT[20–22],青蒿素的明显更高的生物利用度可能会减少治疗所需的药物量。目前,pACT尚未在体内针对除疟疾以外的其他疾病进行过测试。 对于艾滋病病毒/获得性免疫缺陷综合症(HIV/AIDS)患者,疟疾治疗更加复杂。疟疾和HIV共感染在非洲造成了主要的健康负担,主要是因为“现在已经明确,HIV感染会导致更高的疟疾发病率和更严重的疟疾表现”。免疫系统受损,艾滋病患者更容易感染疟疾,而且对疟疾治疗的反应也较慢。此外,在Tusting等人的荟萃分析中,社会经济发展与更好的疟疾治疗结果强相关。最近,青蒿素已经展示出抗HIV活性,因此口服该草药的干叶不仅可以治疗疟疾,还可以提高HIV/AIDS患者的福祉。 农业、就业和自主权 青蒿在75多个国家种植。2011年,约有163吨青蒿素从主要位于中国、越南和东非(包括马达加斯加)的种植园和小规模持有者农场提取出来;价值约为每公斤550美元。随着Sanofi开始生产半合成青蒿素,预计2014年将生产60吨,预计价格约为每公斤400美元。随着这种新型青蒿素的生产,荷兰皇家热带研究所预测,天然青蒿素的市场将大幅动荡,从而破坏了农民的安全。热带研究所进一步担心,“制药公司将积累对生产过程的控制和权力;青蒿生产者将失去一种收入来源;在疟疾流行的地区进行的本地生产、提取和(可能)制造ACT的工作将转移到西方制药公司的主要生产地点”,破坏了这些已经贫困的国家脆弱的经济体系。中国和非洲的小规模农户的平均种植面积约为0.2公顷,因此,尽管实施pACT可能不需要像提取青蒿素那样多的农业土地,但它仍然可以帮助小规模持有者获得一种收入来源。我们估计,可以建造成本小于50000美元的本地微型制造厂,并生产质量控制的pACT片剂,其成分可轻松验证。我们的整体方法,如图3所示,可以实现对疟疾和可能对其他青蒿素敏感性疾病的地方控制,同时提高人口的社会经济地位。 植物性青蒿素联合疗法生产总体方案。 pACT:基于植物的青蒿素联合疗法; TLC:薄层色谱法。 结论 越来越多的证据表明使用青蒿叶片治疗疟疾以及可能的其他疾病的治疗效果。植物中复杂的抗寄生虫化合物混合物似乎解释了其治疗活性,动物和人类试验支持了这一说法。同时,显而易见的是,使用青蒿素联合疗法的成本仅为当前或新兴抗疟疾疗法的一小部分。同样,最近的证据表明,持续性和/或无症状疟疾的存在可能需要更具预防性的方法来使用青蒿素联合疗法,甚至是青蒿茶。考虑到在2000多年的历史中,这种植物被用于中医传统治疗发热,而青蒿素抗药性并没有明显出现,综合考虑所有累积证据,主张将青蒿素联合疗法纳入抗击疟疾的药物库中,极有可能也包括其他疾病的治疗。 核心提示 从植物青蒿(Artemisia annua L.)中提取的青蒿素及其衍生物是目前最佳的抗疟疾治疗药物,以青蒿素联合疗法(ACT)的形式使用。然而,在疟疾流行的发展中国家,青蒿素的可获得性和成本是一个问题。口服青蒿叶干更有效地治疗疟疾,而叶子的茶浸液具有预防作用。生产和交付茶和青蒿叶干片的成本远低于ACT。 致谢 本研究得到了部分来自伍斯特理工学院和马萨诸塞大学临床与转化科学中心的支持;部分支持来自国家中医药中心的奖励号码NIH-R15AT008277-01。 作者贡献:Weathers PJ、Towler M、Hassanali A、Lutgen P和Engeu PO共同参与了文章的撰写;Hassanali A、Lutgen P和Engeu PO提供了临床数据;Weathers PJ和Towler M进行了实验室和野外样本的分析。 贡献者信息: Pamela J Weathers,生物与生物技术系,伍斯特理工学院,01609,美国伍斯特。 Melissa Towler,生物与生物技术系,伍斯特理工学院,01609,美国伍斯特。 Ahmed Hassanali,肯尼亚肯亚塔大学纯与应用科学学院,20100,内罗毕,肯尼亚。 Pierre Lutgen,IFBV-BELHERB,邮政信箱98,L-6908尼德兰文,卢森堡。 Patrick Ogwang Engeu,自然化疗研究所,卫生部,邮政信箱4864坎帕拉,乌干达。 Artemisinin from the plant Artemisia annua (A. annua) L, and used as artemisinin combination therapy (ACT), is the current best therapeutic for treating malaria, a disease that hits children and adults especially in developing countries. Traditionally, A. annua was used by the Chinese as a tea to treat “fever”. More recently, investigators have shown that tea infusions and oral consumption of the dried leaves of the plant have prophylactic and therapeutic efficacy. The presence of a complex matrix of chemicals within the leaves seems to enhance both the bioavailability and efficacy of artemisinin. Although about 1000-fold less potent than artemisinin in their antiplasmodial activity, these plant chemicals are mainly small molecules that include other artemisinic compounds, terpenes (mainly mono and sesqui), flavonoids, and polyphenolic acids. In addition, polysaccharide constituents of A. annua may enhance bioavailability of artemisinin. Rodent pharmacokinetics showed longer T1/2 and Tmax and greater Cmax and AUC in Plasmodium chabaudi-infected mice treated with A. annua dried leaves than in healthy mice. Pharmacokinetics of deoxyartemisinin, a liver metabolite of artemisinin, was more inhibited in infected than in healthy mice. In healthy mice, artemisinin serum levels were > 40-fold greater in dried leaf fed mice than those fed with pure artemisinin. Human trial data showed that when delivered as dried leaves, 40-fold less artemisinin was required to obtain a therapeutic response compared to pure artemisinin. ACTs are still unaffordable for many malaria patients, and cost estimates for A. annua dried leaf tablet production are orders of magnitude less than for ACT, despite improvements in the production capacity. Considering that for > 2000 years this plant was used in traditional Chinese medicine for treatment of fever with no apparent appearance of artemisinin drug resistance, the evidence argues for inclusion of affordable A. annua dried leaf tablets into the arsenal of drugs to combat malaria and other artemisinin-susceptible diseases. Keywords: Malaria, Infectious disease, Artemisia annua, Artemisinin, Combination therapy, Artemisinin combination therapy INTRODUCTION Nearly three billion people are affected by malaria with almost a million deaths annually, especially in Africa and amongst children[1]. Currently extracted from Artemisia annua (A. annua) L., artemisinin (Figure 1) is delivered in concert with another antimalarial drug [artemisinin combination therapy (ACT)] as the preferred treatment to slow emergence of drug resistance. Despite these efforts, artemisinin resistance is appearing[2] and persistent and/or asymptomatic malaria may also be playing a role in disease transmission[3–5]. Moreover, for developing countries ACT is costly and the supply is inadequate[6–9]. Artemisia annua (single clone of Artemisia annua cultivar at approximately 2 m height at floral bud formation), artemisinin and plant-based artemisinin combination therapy tablets. Artemisinin is a sesquiterpene lactone that is produced and stored in the glandular trichomes that are mainly on the leaves and floral buds of A. annua, a GRAS medicinal herb[10–12]. The plant also produces > 40 flavonoids[13], many polyphenols, and a variety of other terpenes including mono-, sesqui-, di-, and triterpenes[14]. As discussed later, many of these have weak antimalarial activity, and, based on transcriptome analyses, many also seem to be produced and/or stored in the glandular trichomes that also contain artemisinin[15]. We and others proposed direct consumption of A. annua either as a tea infusion[16–19] or by oral consumption of the leaves[20–24]. In contrast to the oral consumption of pure artemisinin, we showed that the presence of plant material significantly enhanced appearance of artemisinin in the serum of healthy and Plasmodium chabaudi-infected mice[22]. Because of the plethora of mild antimalarial compounds naturally present in the dried leaves of the plant, we have termed this orally consumed dried leaf therapeutic plant-based artemisinin combination therapy, or pACT. These whole plant approaches are similar to the more than 2000 year traditional use of the plant by the Chinese[25]. To produce a therapeutically effective drug using a complex material like a medicinal plant requires that a number of key factors be met: the medicinal herbal product must be therapeutically effective; levels of key chemical components in the herb must be verifiably consistent; production must also be cost effective. Here we summarize and update our recent review[26] on the effects of A. annua on malaria and further discuss the bioavailability and therapeutic efficacy of pACT and how such an herbal drug could inexpensively be produced with a consistent dose. PROPHYLACTIC USE OF A. ANNUA Tea infusion, its chemistry, and in vitro studies Until recently, there have been, to our knowledge, few well-controlled studies examining extraction, recovery, and stability of artemisinin and other compounds in A. annua tea infusion. A systematic study of preparations of A. annua therapeutic tea infusion was performed by van der Kooy et al[27] and showed that nearly 93% of available artemisinin was extracted from dried A. annua leaves, but only under certain conditions. Best preparation method was: 9 g DW leaves/L, for 5 min at 100 °C. Subsequent storage of the tea infusion at room temperature showed that artemisinin concentration was stable for > 24 h, important for malaria-endemic locations where there is no refrigeration. Artemisinin water solubility is approximately 50 mg/L[27], so the amount of artemisinin recovered from hot water tea infusions is reasonable. Other studies using the same extraction protocol also measured extraction and stability of artemisinin and some key flavonoids in the tea. Artemisinin was found to be stable at room temperature for up to 48 h[28] ; however, some flavonoids were poorly extracted and not stable at room temperature[29]. Carbonara et al[28] detected an assortment of phenolics, including 0.06 mg/g DW cirsilineol, in an A. annua tea infusion prepared at about a 4–10 fold higher proportion (approximately 38 g DW/L) than that proposed as optimal (9 g DW/L) by van der Kooy et al[27]. Most of the measured phenolics in the tea remained constant at room temperature for 48 h post-infusion. More recently, Suberu et al[19] identified milligram amounts of phenolic acids, flavonoids, and sesquiterpenes in a liter of A. annua tea, all of which demonstrated IC50 values in the micromolar or less range (Table 1). Indeed, the IC50 of the tea infusion itself was 7.6 and 2.9 nmol/L for the chloroquine (CQ)-sensitive HB3 and CQ-insensitive Dd2 strains of P. falciparum, respectively, and better than artemisinin alone suggesting synergism of constituents in the tea mixture. Clearly if a tea infusion is to be a therapeutic option, it must be consistently and reliably prepared and ingested. As suggested by van der Kooy et al[27], ideally a liter of tea infusion would be prepared daily and consumed in equal aliquots of about 250 mL over 24 h for several days. Tea infusion clinical trials Ogwang et al[30,31] tested Artemisia tea as a prophylaxis against malaria in 132 adult farm workers, aged 18–60 years, for 12 mo in a randomized clinical trial in Uganda. Tea infusion was consumed once a week at 2.5 g dried leaves per adult infusion dose with 55–100 mg artemisinin/L. Malaria was tracked for 9 mo while adverse clinical effects were tracked for 12 mo. Among those who used Artemisia tea there were 80% fewer fever-related hospital visits. Indeed, some patients reported using A. annua tea for > 7 years with no incidence of malaria and no serious adverse events. Although this study suggested that once weekly consumption of A. annua tea infusion may offer prophylactic protection, there were no children or elderly in the study, so additional clinical trials need to be conducted with different populations and age groups. Authors argued that since a single weekly dose was effective, compounds other than artemisinin may have played the prophylactic role since artemisinin itself has short plasma half-life. THERAPEUTIC USE OF A. ANNUA Tea infusion Reports on the efficacy of A. annua (cv. Artemis) tea on human malaria patients by Mueller et al[17,32] and Blanke et al[33] yielded at times conflicting results. Their tea infusions contained 47–94 mg artemisinin/L, but recrudescence was much lower in the quinine-treated control group, so parasite reappearance in the tea-treated patients was ascribed to recrudescence and not re-infection[17]. In the Blanke et al[33] trial that included a placebo tea, recrudescence was consistently lower in the tea patients than in those treated with 500 mg pure artemisinin. More recently, however, De Donno et al[34] showed that 5 g dried leaves in one liter of A. annua tea infusion was effective against both CQ-resistant (W2) and CQ-sensitive (D10) strains of P. falciparum with IC50 values of 5.60 nmol/L and 7.08 nmol/L, respectively, results also consistent with those of Suberu et al[19] as already highlighted. These latter in vitro studies suggested that tea should be efficacious, so why the discrepancy with the earlier human trials? Preparation methodology is crucial for preserving as much biochemical integrity of the plant as possible[27]. The more recent in vitro studies likely used more consistently prepared tea infusions than the earlier human trials, so variations in chemical composition of the infusions and in the plant source material could explain the different responses. The argument that tea is a monotherapy is unsubstantiated considering the now well-established chemical complexity and related antiplasmodial activity of tea infusions of A. annua and its components. Although data from therapeutic tea trials in animals and in humans correlate well, unfortunately, they do not support use of A. annua tea for treating malaria because animal and human data are comparably negative, the artemisinin dose is not easily controlled, and other potentially synergistic components in the tea are not readily controlled or extracted. Nevertheless, use of the tea could play a role in malaria prophylaxis to reduce incidence of malaria in different communities, or in temporary relief from malaria, mainly in prevention of coma or “to buy time” to enable an infected person from a rural area to travel to a hospital or clinic stocked with ACT. Dried leaf A. annua - pACT Recently, Elfawal et al[23] measured parasitemia in mice infected with P. chabaudi that were fed two different doses (0.6 or 3.0 mg artemisinin; 24 and 120 mg/kg) of either pure artemisinin in mouse chow or as pACT. Artemisinin delivered via pACT was at least five times more effective, and with a longer lasting response, than pure artemisinin in reducing parasitemia. Excluding artemisinin there are > 600 phytochemicals that have been identified in Artemisia annua[35], but there is currently a lack of information on the chemistry, effect of the preparation method (harvesting, drying, storage, etc.), and overall bioavailability of these chemicals[36] Clinical trials using dried leaf A. annua are scarce in the scientific literature and few, other than those in Democratic Republic of Congo by Mueller et al[17,32], are published. Despite the fact that WHO does not encourage either whole plant or tea infusion clinical trials[37], some African universities have been conducting their own trials, many of which have not been published nor results assessed by polymerase chain reaction (PCR) as later done for clinical trials with ACTs (personal comm from C. Kasongo to P. Lutgen). Many of these trials used A. annua infusions, and compared to controls or even other antimalarial drugs, e.g., artesunate-amodiaquine, showed significantly greater sensitivity of the infusion with fewer late therapeutic failures. For example, in Democratic Republic of Congo, 54 malaria-infected volunteers were treated for 10 d with capsules containing powdered leaves of A. annua. Each patient was given 15 g dried leaves containing 15 mg of artemisinin (artemisinin content in leaves = 0.1%[38] In a study aimed at preventing severe post-operative malaria at Bangui, Central Africa, powdered leaves of A. annua were administered in capsules to 25 patients, 22 of them children aged 1–16 years[24]. Treatment duration ranged from 3–4 d with a dose of 0.4–0.5 g/d of A. annua The most clinically definitive study to date of pACT efficacy was conducted at the International Centre of Insect Physiology and Ecology (ICIPE) Mbita Field campus, Suba District, in Western Kenya. This was a collaborative project between ICIPE and Kenya Medical Research Institute[20]Table 2[39]) and was an open-label, non-randomized clinical trial mainly targeted to assess efficacy, safety, and tolerance of increasing doses of pACT delivered as tablets. The tablets were made by a Tanzania-based NGO, Natural Uwemba System for Health, from a hybrid of A. annua grown in the Tanzania highlands (2000–2200 m altitude). Leaves were harvested just before flowering, dried for approximately 3 wk under shade, then crushed, powdered, homogenized, and pressed into 500 mg tablets under ambient temperature. Tablets were robust with no excipient required. Using HPLC with diode array detector, analysis of hexane extracts of randomly selected batches of 100 tablets showed artemisinin content of the tablets was consistent at 0.74% ± 0.06% (i.e., approximately 3.7 mg per tablet). The four cohorts of the trial each had 12 consenting patients aged 15–56 years (average 23.42) with P. falciparum malaria. Based on Giemsa-stained blood smears counted against 200 wbc, parasitemia was 0.02%-4% and hemoglobin levels > 8 mg/dL. Each cohort received one of four increasing numbers of A. annua tablets, ranging from 2–5 tablets twice on day 1, followed by 1–4 tablets twice daily for the next 5 d (Table 2). A week following the treatments, three patients scattered throughout different cohorts showed re-appearance of parasites in blood smears; however, all doses were effective in clinical and parasitological regression of malaria, with 9%-20% recrudescence at day 28 and no measurable toxicity. Compared to the usual large pure artemisinin doses of 1000 mg on day 1 followed by 500 mg on each of days 2–7 that were administered to 227 malaria patients[39]Table 2). The measured pACT cure rate also was comparable to or exceeded other results using pure artemisinin[40,41], and similar levels of artemisinin (artesunate, artemether, etc.[42]). Furthermore, the positive therapeutic response using pACT appeared somewhat independent of dose beyond the second level of dose tested (Table 2[20]). Although oral doses used in the ICIPE[20] trials were far less than any tea studies, levels of recrudescence were much lower than tea and often better than in studies using pure artemisinin[39] (Table 2). Indeed, about 100 total mg of total artemisinin delivered via pACT for a full malaria treatment yielded a better recrudescence rate than the 4000 mg of pure artemisinin used by Giao et al[39] (Table 2). This 40-fold difference correlates well with the early pharmacokinetic studies by Weathers et al[21] that showed 45-fold enhanced bioavailability of the drug when delivered as pACT. These results suggest that the natural phytochemical blend in pACT is important especially when orally administered as tablets. The results are also consistent with a study in China on mice infected with P. berghei, which compared the effects of pure artemisinin with crude A. annua extracts[43], and the studies by Elfawal et al[23] and Weathers et al[22]. In all three studies the administered products had comparable levels of artemisinin, but crude preparations and pACT were at least 3.5 times more effective in reducing parasitemia than pure artemisinin, suggesting a synergistic role for non-artemisinin constituents in the extracts and orally consumed dried leaves. COMPARATIVE PHARMACOKINETICS AND BIOAVAILABILITY Orally delivered artemisinin When given orally or rectally, dihydroartemisinin showed higher bioavailability in humans than artemisinin in an early pharmacokinetic study by Zhao et al[44]. The Cmax, Tmax, and T1/2 for orally delivered dihydroartemisinin were 0.13–0.71 mg/L, 1.33 h, approximately 1.6 h, respectively; for pure artemisinin they were 0.09 mg/L, 1.5 h, and 2.27 h, respectively. Alin et al[45] compared orally delivered artemisinin and artemisinin-mefloquine combination therapy for treatment of P. falciparum malaria. Infected and uninfected patients had similar pharmacokinetic parameters. After a single dose, bioavailability of artemisinin was not altered. Interestingly, pharmacokinetics were similar when comparing treatment failures with successes, suggesting that studies that only measure artemisinin pharmacokinetics were inadequate for predicting therapeutic success[45]. Ilet et al[46] also reviewed artemisinin pharmacokinetics in patients with falciparum malaria and reported a dose of 9.1 mg/kg, which was comparable to that of Alin et al[45]. Cmax and Tmax values did not differ much from those reported by Alin et al[45]. In the Ilet et al[46] review of pharmacokinetic parameters of artemisinin and its derivatives, oral pure artemisinin doses ranged from about 6–11 mg kg/L in healthy subjects and Cmax was 0.15–0.39 mg/L. Dose seemed to have no major effect. An earlier study by Ashton et al[47] compared increasing artemisinin doses of 250, 500, and 1000 mg per person and both Cmax and T1/2 showed dose-dependent increases of 0.21, 0.45, and 0.79 mg/L, and 1.38, 2.0, and 2.8 h, respectively, but Tmax remained relatively constant at 2.3–2.8 h. Diet is an important consideration for any orally delivered drug, and when Dien et al[48] compared artemisinin oral doses given with and without food, Cmax values were similar between subjects who fasted and those who did not. Food consumption along with artemisinin did not seem to affect artemisinin absorption. In contrast, a later rodent study by Weathers et al[21] observed that when artemisinin was consumed as part of a complex plant material, pACT, approximately 45-fold more drug entered the serum of mice than orally administered pure drug. Similarly, when pure artemisinin was fed to mice, it was not detectable in the serum after 60 min. However, artemisinin was detected in the serum when consumed in conjunction with mouse chow, which consists of a variety of plant materials including soy, oats, wheat, alfalfa, beet pulp, corn, etc[22]. In a study by Ashton et al[49], artemisinin at 9.1 mg/kg was given daily for 7 d, and measurements taken on days 1, 4, 7, and 21. On day 1 plasma Cmax and T1/2 were similar and comparable to data from other studies using a similar dose. On day 4 and 7, however, Cmax decreased, while T1/2 increased, indicating that although artemisinin was delivered daily for 7 d, it was either not readily absorbed or it degraded after the first dose. After the third dose, Cmax fell from 0.31 to 0.11 mg/L, and T1/2 increased from 3.0 to 4.8 h. These results suggested that either artemisinin was metabolized or accumulated elsewhere in the body. In the liver, cytochrome P450 (CYP450) enzymes metabolize artemisinin to deoxyartemisinin, deoxydihydroartemisinin, 9,10-dihydrodeoxyartemisinin, and a metabolite named “crystal 7”[50]. Extended artemisinin dosing may not be beneficial as shown by Svensson et al[50] using human liver microsomes where activity of CYP450s, CYP2B6 in particular, correlated with decreasing artemisinin serum levels. In intermittent dosing studied by Ashton et al[49], the P450 levels were allowed to decline for 14 d before delivery of another dose, and Cmax rose from 0.11 to 0.20 mg/L, and T1/2 decreased from 4.8 to 2.7 h. Generally, maximum concentration of artemisinin in the body increased with increasing doses with T1/2 ranging from about 1.4–4.8 h for reported trials using oral pure artemisinin. Thus, increased and extended artemisinin treatment may reduce recrudescence. Tea infusion delivered artemisinin Other than Räth et al[16], there are few reports on the pharmacokinetics of tea infusion artemisinin delivered in humans. In the Räth et al[16] study, artemisinin Cmax was 0.24 mg/L at 0.6 h post consumption. Tea infusion containing 94.5 mg artemisinin had a Cmax equivalent to a dose of 250 mg pure artemisinin, but at a significantly shorter Tmax, 0.6 h vs 2.8 h[47]. Compared to pure artemisinin, the shorter half-life of artemisinin in the tea infusion may account for the observed higher recrudescence. Although tea-delivered artemisinin seemed more bioavailable, its shorter T1/2 of 0.9 h compared with about 2 h for pure artemisinin, suggested that more than two doses per day may be more beneficial; indeed, four doses a day were recommended. The unacceptably high recrudescence rates in clinical tea infusion trials were attributed to low plasma concentrations, almost 40% lower than that for traditional doses (500 mg per person of 60 kg or 8.3 mg artemisinin/kg) of pure artemisinin. Although not specified, tea trial doses have been estimated at about 1.5 mg/kg, close to the 1.1 mg/kg dose of pure artemisinin used by Zhao et al[44], which is far below the 8.3 mg/kg that is traditionally accepted as pharmacologically effective. Nevertheless, the Cmax of 0.24 mg/L artemisinin for the tea dose is nearly twice that for pure artemisinin (Cmax = 0.13 mg/L) as measured by Zhao et al[44]. A. annua tea also showed potent antiplasmodial activity against 40 field isolates of P. falciparum collected in Pikine, Senegal (mean IC50 0.095 µg/mL[51]). Dried leaf (pACT) delivered artemisinin There are as yet no pharmacokinetic studies of pACT in humans. In a small PK study of healthy mice fed artemisinin there was about 45-fold more artemisinin delivered via pACT than when delivered as the pure drug[21]. More recently, pharmacokinetics of artemisinin and one of its liver metabolites, deoxyartemisinin, were compared over 120 min in healthy and P. chabaudi-infected mice treated with dried A. annua leaves at a 100 mg/kg body weight dose of artemisinin[22]. In pACT-treated healthy mice, the first order elimination rate constant for artemisinin was estimated to be 0.80/h, corresponding to a T1/2 of 51.6 min. Cmax and Tmax were 4.33 mg/L and 60 min, respectively. The AUC was 299.5 µg min/mL. The first order absorption rate constant was estimated at 1.39/h. In contrast, the AUC for pACT-treated infected mice was greater at 435.6 µg·min/mL. Serum levels of artemisinin in the infected mice continued to increase over the 120 min of the study period. As a result, the elimination half-life, T1/2 could not be determined, so Cmax and Tmax could only be estimated at ≥ 6.64 mg/L and ≥ 120 min, respectively. Nevertheless, both Cmax and Tmax of artemisinin were greater in infected than in healthy mice. Generally, artemisinin concentrations decreased with a concomitant rise in deoxyartemisinin levels only in healthy subjects[22]. In contrast, artemisinin levels in infected mice continued to rise over the study period whilst deoxyartemisinin levels fell and then leveled, so infection seemed to retard the capacity of the mice to process artemisinin into deoxyartemisinin over the two-hour period. Many compounds in A. annua inhibit P. falciparum[52–55] and CYP34A[56]. At the high (100 mg/kg) dose used in the study, nearly equal amounts of artemisinin and deoxyartemisinin were measured in the serum, indicating that an excessive dose of artemisinin was used. The presence of plant material affected artemisinin pharmacokinetics. At 60 min no artemisinin was detected in serum of mice fed pure artemisinin at 100 mg/kg body weight. When plant material was present, however, as mouse chow or A. annua pACT, artemisinin level in the serum rose to 2.44 and 4.32 µg/mL, respectively, demonstrating that the presence of plant material, even mouse chow, had a major positive impact on the appearance of artemisinin in the blood[22]. To our knowledge, these are the only data available on pharmacokinetics for orally delivered A. annua in animals or humans. NON-ARTEMISININ THERAPEUTIC COMPOUNDS IN A. ANNUA Flavonoids A. annua is rich in essential oils, coumarins, polyphenols, polysaccharides, saponins, terpenes, and flavonoids. The levels of flavonoids and other compounds in A. annua change with developmental growth stage, with some being highest during full bloom[57]. There are > 40 flavonoids[13], and at least 11, including artemetin, casticin, chrysoplenetin, chrysoplenol-D, cirsilineol, eupatorin, kaempferol, luteolin, myricetin, quercetin, and rutin, are reported to have weak therapeutic efficacy against falciparum malaria (Table 1[52–54,58]). Some of these flavonoids were shown to improve the IC50 of artemisinin against P. falciparum in vitro by as much as 50%, suggesting synergy (Table 1[52]). Elford et al[53] also showed that while casticin (5-hydroxy-2-(3-hydroxy-4-methoxyphenyl)-3,6,7-trimethoxychromen-4-one) showed synergism with artemisinin, it did not synergize with chloroquine, suggesting a different interactive mechanism. Combining casticin with artemisinin inhibited parasite-mediated transport systems that control influx of myoinositol and L-glutamine in malaria-infected erythrocytes. These apparent synergistic actions between flavonoids and artemisinin suggest that flavonoids are likely to be important for efficacious use of A. annua consumed either as whole dried leaves or as tea. Many flavonoids have antiplasmodial effects and inhibit P. falciparum growth in liver cells in vitro as reported for dietary flavonoids[54]. To our knowledge, there are no reports on pharmacokinetics of A. annua delivered flavonoids. Some flavonoids are reported to have long plasma half-lives; e.g., quercetin, found in A. annua and most fruits, has a plasma half-life of 27 h[59]. Quercetin [2-(3,4-dihydroxyphenyl)-3,5,7-trihydroxy-4H–chromen-4-one], also found in garlic, inhibits parasite growth with differential activity against different strains of Plasmodium (Table 1[54,58]). Rutin, which is a rutinose [α-L-rhamnopyranosyl-(1→6)-β-D-glucopyranose] glycoside of quercetin, showed similar results, suggesting that the sugar moiety did not significantly affect antimalarial activity (Table 1[58]). Flavonoids are known to persist in the body for > 5 d; this may explain the once a week dose inducing a prophylactic effect from A. annua tea infusion that was reported by Ogwang et al[30,31]. Many dietary flavonoids inhibit Plasmodium growth in vitro, but amounts in the diets are reportedly insufficient to offer protection against malaria[54]. Plants such as A. annua with high concentrations of flavonoids (e.g., up to 0.6%) may, however, work in concert with artemisinin to prevent malaria when consumed regularly. The flavone luteolin [2-(3,4-Dihydroxyphenyl)-5,7-dihydroxy-4-chromenone] comprises up to 0.0023% DW in Artemisia[14] and has been used for a variety of ailments including cough, diarrhea, dysentery, diabetes, cancer, and malaria. Although luteolin has an IC50 value around 11 µmol/L[54] and is one of the more active antiplasmodial flavonoids found in A. annua, one cannot compare its role between studies as indicated by Ganesh et al[58] (see Table 1). The antimalarial response of different flavonoids seems to be affected by the strain of Plasmodium being tested. Luteolin also prevents completion of a full intra-erythrocytic cycle by inhibiting progression of parasite growth beyond the young trophozoite stage. The mechanism of this antiplasmodial activity seems to be related to the inhibition of parasite fatty acid biosynthesis. These lipids are required by the parasite to detoxify heme into hemozoin[60]. Independent of the human host, apicomplexan parasites use a fatty acid biosynthetic pathway. Enzymes in the pathway, like the NADPH-dependent b-ketoacyl-ACP reductase (FabG), are potential antimalarial targets. Among 30 flavonoids studied, luteolin and quercetin had the lowest IC50 values for the inhibition of these enzymes and also showed in vitro activity in the sub-micromolar range against multiple strains of P. falciparum[60]. Isovitexin {5,7-dihydroxy-2-(4-hydroxyphenyl)-6-[(2S,3R,4R,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl) oxan-2-yl]chromen-4-one} is another flavone, the 6-C-glucoside of apigenin, that was found in A. annua tea infusion at > 100 mg/L with micromolar antiplasmodial activity (Table 1[19,28]). Isovitexin inhibits lipid peroxidation and xanthine oxidase activity and protects cells from ROS damage with an overall LD50 > 400 µmol/L[61]. Terpenes Limonene (1-Methyl-4-(1-methylethenyl)-cyclohexene) is part of the “cineole cassette” that includes 1,8-cineole (eucalyptol), limonene, myrcene, α-pinene, β-pinene, sabinene, and α-terpineol[62]; many of these affect particular stages of Plasmodium species. For example, limonene is often present at 7 mg/kg in A. annua[14] and inhibits isoprenoid biosynthesis in Plasmodium[63] and development at the ring and trophozoite stages[64]. Eucalyptol affects the trophozoite stage[65]. Limonene also arrests protein isoprenylation in P. falciparum, halting parasite development within 48 h of treatment[64]. The IC50 against in vitro Plasmodium in these trials was 2.27 mmol/L, more than twice the IC50 of 533 µmol/L measured by van Zyl et al[55]. Limonene and its metabolites remain in the plasma for at least 48 h[66], so the pharmacokinetics is favorable, which is important for elimination of gametocytes and malaria transmission. The volatile monoterpene α-pinene (4,6,6-trimethylb-ficyclo[3.1.1]hept-3-ene) is present in the plant at levels up to 0.05% of dry weight[14]; it has an IC50 of 1.2 µmol/L, in the range of quinine at 0.29 µmol/L[55]. Eucalyptol (1,8-cineole) may comprise up to 30% [0.24%-0.42% (V/DW)] of the essential oil in A. annua[67] and is a strong inhibitor of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-8[68]. Both chloroquine-resistant and chloroquine-sensitive Plasmodium strains are affected at the early trophozoite stage[65]. Eucalyptol (1,3,3-Trimethyl-2-oxabicyclo[2,2,2]octane) is also volatile and rapidly enters the blood when delivered either as an inhalant or orally [69,70]. At an IC50 of 0.02 mg/mL and low toxicity (LD50 of approximately 25 mg/mL), either oral or inhalation delivery is reasonable[65,71]. Indeed eucalyptol concentrations can reach 15 µg/mL in 60 min[69] suggesting its possible use as an antimalarial inhalant. Artemisia ketone (3,3,6- trimethyl-1,5-heptadien-4-one), a major constituent of some cultivars of A. annua, has barely been studied. Other ketones like curcumin[72] have been implicated as inhibitors of β-hematin synthesis, so artemisia ketone may play a similar role and affect hemozoin formation. Although hemoglobin is required for Plasmodium survival and multiplication in merozoites inside the red blood cell, it leaves toxic debris like heme. The parasite subsequently oxidizes Fe2+ in heme to Fe3+ forming hematin, a nontoxic insoluble polymeric crystal called β-hematin (also known as hemozoin), which also inhibits cell-mediated immunity against the parasite. Water extracts of A. annua inhibit hemozoin synthesis[73]. Essential oils often contain a large amount of monoterpenes that may enhance the antimalarial effect of artesunate and even reverse the observed resistance of P. berghei against artesunate[74]. Monoterpenes tend to be higher in the pre-flowering phase of A. annua[75], but are drastically reduced by high drying temperatures or drying in the sun[13,76] and, of particular concern, during compression of dried leaves into tablets[77]. Although monoterpenes have some antimalarial potential, most are rather volatile and thus they may be therapeutically less important than the nonvolatile flavonoids, phenolic acids, and higher molecular weight sesquiterpenes. Unlike α-pinene and eucalyptol, camphor (1,7,7-Trim ethylbicyclo[2.2.1]heptan-2-one) has no reported antimalarial activity, but it may comprise as much as 43.5% of the essential oil of A. annua[78]. Considering camphor is less volatile than either eucalyptol or α-pinene (melting points of 204 °C, 176 °C, and 155 °C, and flash points of 54 °C, 49 °C, and 33 °C, respectively), it may instead play a role in enhanced transport of hydrophobic molecules like artemisinin from pACT across the intestinal wall into the bloodstream[21,22]. Camphor may also affect thymocyte viability and aid in developing malaria immunity through production of T-cells[79]. At 50 µg/mL, camphor increased viability of cultured thymocytes[80]. The sesquiterpene nerolidol (3,7,11-Trimethyl-1,6,10-dodecatrien-3-ol) has an IC50 of 0.99 µmol/L and arrests development of the intraerythrocytic stages of the parasite (Table 1[55] ). Indians of the Amazon basin in Brazil treated malaria using the vapors of the leaves of Viola surinamensis; nerolidol was identified as the active constituent leading to 100% growth inhibition at the schizont stage[81]. Nerolidol levels vary with the cultivar tested, with one of the highest values found in plants from Ethiopia[82]. There is a greater concentration of this sesquiterpene in stems than leaves of A. annua[83]. Other sesquiterpenes found in the artemisinin biosynthetic pathway were only recently shown to have antiplasmodial activity at µmol/L levels, similar to that of other compounds found in the plant (Table 1[19]). These artemisinic compounds were extracted into A. annua tea infusions and showed varying interactions with artemisinin depending on their relative concentrations and the target parasite strain. For example, arteannuin B showed an additive interaction with artemisinin against the CQ-sensitive Plasmodium HB3 strain, while against the CQ-insensitive Dd2 strain the interaction was synergistic. Phenolic acids Rosmarinic ((2”R”)-2-[[(2”E”)-3-(3,4-Dihydroxyphenyl)-1-oxo-2-propenyl]]oxy]-3-(3,4-dihydroxyphenyl) propanoic acid) and chlorogenic ((1S,3R,4R,5R)-3-{[(2Z)-3-(3,4-dihydroxyphenyl)prop-2-enoyl]oxy}-1,4,5-trihydroxycyclo-hexanecarboxylic acid) acids are strong antioxidants found in a wide variety of A. annua cultivars[56]. In Caco-2 studies, these acids significantly inhibited activity of CYP3A4, one of the hepatic P450s responsible for metabolism of artemisinin to deoxyartemisinin, an inactive form of the drug[50]. These and other phenolic acids are present in A. annua tea infusion[19]. Both phenolic acids have an IC50 of about 65 µmol/L (Table 1) and also significantly reduced secretion of cytokines IL-6 and IL-8, and thus enhanced antimalarial activity while reducing inflammation[56]. Other compounds often found in A. annua and that may affect pACT efficacy Although polysaccharides in other medicinal plants have been more extensively studied, they seem to have been rather overlooked in A. annua, probably because most Artemisia extracts are obtained using organic solvents and polysaccharides are only soluble in water. Polysaccharides extracted from Artemisia iwayomogi showed hydroxyl radical scavenging activity three times stronger than glutathione or caffeic acid, and ROS inhibition was twice as strong as ascorbic acid[84]. In A. iwayomogi, more polysaccharides were found in stems than in leaves and their solubility was also higher from stem than from leaf tissue[84]. The combination of polysaccharides with lipophilic molecules like artemisinin may lead to a higher bioavailability of the antimalarial constituents when delivered via A. annua, which may explain the lower effective therapeutic dose against malaria observed for pACT than for pure artemisinin[20,23,26]. Indeed, Han[85] showed that ginseng polysaccharides had preventive and curative antimalarial activities and synergized with artesunate in malaria-infected mice. Sulfated polysaccharides inhibited the in vitro invasion of merozoites into erythrocytes and interfered with merozoite surface protein[86–88]. Heparin and other sulfated polysaccharides have been shown to inhibit blood-stage growth of plasmodium[89,90]. Some sulfated polysaccharides inhibited the formation of rosettes between infected red blood cells (iRBC) and uninfected RBCs, as well as adhesion of iRBCs to placental chondroitin sulfate A, which is linked to severe disease outcome in pregnancy-associated malaria[91]. Saponins, common in many plants, have an important role in human and animal nutrition and are reportedly present in A. annua, but only as measured in alcoholic extracts using the nonquantitative foaming test[92,93] (Weathers, unpublished). These soap-like amphiphilic (lypo- and hydrophilic) bioactive compounds are mainly produced by plants. Recently, there has been interest in the clinical use of saponins as chemotherapeutic agents[94], and as adjuvants for vaccines[95]. At very low doses saponins are efficient, have hemolytic properties, produce 40–50 Å pores in erythrocyte membranes, and modulate the sodium pump and ATPase[96]. Saponins also have a hypoglycemic effect mainly by inhibiting intestinal permeability and absorption of glucose and may therefore inhibit the growth of P. falciparum, which needs glucose to grow[97]. Better identification, quantification, and investigation into the role of saponins in pACT efficacy are warranted. The coumarin, scopoletin (7-hydroxy-6-methoxychromen-2-one), also known for its antinociceptive properties[98,99], is commonly found in most Artemisia species at, for example, about 0.2% (w/w) in a Luxembourg cultivar. Known for its anti-oxidant, hepatoprotective, and anti-inflammatory activities, scopoletin scavenging capacity for hydroxyl radical, DPPH, superoxide anion, hydrogen peroxide, and Fe2+ chelating activity is almost at the level of α-tocopherol (Vitamin E)[100]. Although not antiplasmodial, scopoletin inhibits TNF-α, IL-6, and IL-8 at millimolar concentrations, and is thus likely one of the major anti-inflammatory and antipyretic constituents of A. annua[101]. Coumarins can activate lymphocytes, thereby stimulating immunological functions[102]. Indeed, scopoletin induced cell proliferation in normal lymphocytes with an immunomodulatory effect[101]. In uninfected erythrocytes internal Na concentration is much lower than external concentration, but the K concentration is higher; in infected blood cells this situation is drastically reversed[103]. Scopoletin significantly stimulated erythrocyte membrane ATPases at 0.1 µmol/L, in particular Na-K-ATPase vs Ca-ATPase or Mg-ATPase[104], so scopoletin may affect malaria infection. A significant hormetic effect was also noticed; stimulation was higher at scopoletin concentrations of 10 µg/mL than at 1 or at 100 µg/mL. In addition scopoletin also inhibited ADP-platelet aggregation at a range of 0.1 to 5 µmol/L and improved blood rheology[105]. Scopoletin may also affect the interaction between malaria and uric acid. Cyclical fevers and high levels of inflammation characterize malaria and this likely aids parasite clearance. Excessive and persistent inflammation, on the other hand, can lead to severe malaria[106]. In the cytoplasm of their parasitophorous vacuole, Plasmodium-infected erythrocytes contain uric acid precipitates that are released upon erythrocyte rupture. Uric acid precipitates are mediators for inflammatory cytokines IL-6, IL-8, and are considered a danger signal for innate immunity. Uric acid is also the causative agent in gout. These precipitates could offer a novel molecular target for anti-inflammatory therapies in malaria. Scopoletin inhibits the activity of xanthine oxidase in hyperuricemic mice after peritoneal administration, and this hypouremic effect is fast and dose-dependent[107]. Toxicology Although many of the compounds in A. annua have not been tested for their toxicity in, a survey of available MSDS data showed that the LD50 levels for orally administered compounds in rodents ranged from about 160 mg/kg for quercetin to > 8000 mg/kg for nerolidol. The artemisinin LD50 measured via oral dose in a mouse was 4228 mg/kg. Therefore, at the estimated amounts of dried leaves of pACT that may be orally consumed by a malaria patient, most of the compounds reported thus far in A. annua are at concentrations that are orders of magnitude below their LD50 toxicity values. Toxicology of the dried leaf tablets used in the Kenyan human trial measured the following components: serum levels of urea, serum proteins, creatinine, γ-glutaryl transferase, serum glutamic pyruvic transaminase, serum glutamic oxaloacetic transaminase, or alkaline phosphatase levels, hemoglobin, and pre- and post-electrocardiograms[20]. Compared to levels prior to treatment with pACT, there was no significant change post-treatment. PRODUCTION CONSIDERATIONS Production comparisons with traditional extraction Because production costs are usually closely held secrets, there are few cost estimates that are publicly available to compare pACT production with extracted artemisinin. However, costs can be estimated from a study by de Vries et al[108] where they reported a 1 kg recovery of artemisinin from A. annua containing 0.6% artemisinin. Downstream processing costs and product losses increase with increasing number of unit operations (unit ops), a fact often not generally appreciated[109]. Indeed for biotechnology processes, recovery can be anywhere from 9%-51%[110]. As an example, if each step of a 4 step process is 95% efficient, then the overall process has a final efficiency of about 81%, while a single step process at 95% efficiency has a 95% overall recovery. The described process steps for extracted artemisinin (eAN) vs pACT-AN are shown in Figure 2. From the point of harvested dried leaves to material ready for packaging or conversion to the delivered drug (e.g., artesunate or artemether), pACT has one unit op and eAN has eight[108]. Extraction solvents and other chemicals are clearly no longer part of the cost. Because there is one vs eight unit ops for eAN and at least two of the eAN unit ops involve significant amounts of heat, pACT energy cost is significantly reduced by at least 90%. Costs for labor, interest, depreciation, and maintenance are all also affected by the number of unit ops[109], so we estimated that with seven fewer unit op steps those costs would reduce by approximately 88%. Although better extraction processes may be in play[111], using the de Vries et al[108] analysis our estimate of cost reduction for producing pACT is about 30% less than the cost of producing eAN. Data provided by de Vries et al[108] was based on 0.6% artemisinin content, so if a higher producing cultivar was harvested, costs would drop proportionately. Moreover, cost drops again because with pACT there is no need to convert artemisinin to artesunate or artemether; those conversions were necessary because they have higher bioavailability than pure artemisinin, which is not an issue with pACT[21,22]. Comparison between plant-based artemisinin combination therapy production and extracted artemisinin from dry harvested leaves to product ready either for packaging (plant-based artemisinin combination therapy) or conversion to artemether or artesunate (extracted artemisinin). AN: Artemisinin; eAN: Extracted artemisinin; pACT: Plant-based artemisinin combination therapy. The de Vries et al[108] process cost estimation focuses on a production yield of 1 kg of artemisinin from 500 kg dried leaves, so per Giao et al[39] that amount of pure artemisinin would treat only 250 patients. Based on the data shown in Table 3 from Kenyan or WPI A. annua at 0.7 and 1.4% artemisinin, 15 and 7.5 g DW leaves, respectively, are required for a total adult pACT treatment; so from 500 kg leaves, 33300 and 66600 patients could be treated, respectively. This represents more than a 130-fold increase in patients treated compared to pure artemisinin with proportionate reduction in price. Table 3 Estimated numbers of adult patients treatable from plant-based artemisinin combination therapy1 For A. annua cultivar containing Number of patients treated at various dry leaf tonnage 2 T/ha2 3 T/ha 4 T/ha3 5 T/ha 0.7% artemisinin/g DW (Kenyan cultivar) 127260 190890 254520 318150 1.4% artemisinin/g DW (WPI cultivar) 254520 381780 509040 636300 Open in a separate window 1Assumptions: each adult needs 100 mg artemisinin (AN) over 6 d for a cure; at 0.7% and 1.4% AN that is approximately 15 and 73 g DW leaves, respectively, for a single adult total malaria treatment; 2Below the average of 2.5 T/ha reported for all of East Africa; 3Equal to the maximum obtained growing A. annua SAM in the Stow, MA, United States field trials. A. annua: Artemisia annua. A. annua dry leaf production varies around the globe. “In East Africa yields average 2.5 T/ha (range = 0.75–4.2)…”[112]. Based on our field trials[113], the reported average A. annua leaf production in E. Africa[112], and the doses used in the Kenyan human trial[20], one can estimate the amount of dry leaf production, and depending on the amount of artemisinin in the biomass, estimate possible number of adult patients that could be treated with pACT (Table 3). Current ACT drugs vs pACT Using the dosing information obtained from the Kenyan human malaria trial[20], each adult needs about 100 mg artemisinin total over 6 d for a malaria treatment, so for A. annua leaves with 0.7% artemisinin, 15 g of dried leaves would be needed for a 6 d treatment course. At 2 ton of dried leaves harvested per hectare, 127260 adult patients could be treated for malaria (Table 3). For leaves containing 1.4% artemisinin, only 7.5 g of dried leaves are required, so from a hectare of land producing 2 tons of leaves twice as many patients could be treated (Table 3). Clearly choosing cultivars that have higher levels of artemisinin in their leafy biomass will dramatically increase the number of patients that can be treated from 1 ha. According to Roll Back Malaria, from one ton of purified artemisinin current ACT therapy can provide 1.76 million adult malaria treatments using artemether/lumefantrine, and 2.5 million adult treatments using artesunate/amodiaquine[114] (Table 4). Using the same one ton artemisinin equivalent, but delivering the drug via pACT with 0.7% artemisinin content, one would have harvested about 142.8 tons of dried A. annua leaves. Assuming 15 g dried leaves per patient from the dosing data in the Kenyan human malaria trial (Table 2[20]), 8.64 million adult patients could be treated, about a four-fold increase over either of the current ACT drugs. The actual cost of pACT, therefore, mainly depends on the cost of the dried leaves and their artemisinin content. Table 4 Estimated number of patient treatments by current artemisinin combination therapy vs plant-based artemisinin combination therapy Combination therapy drug Adult treatments per ton of artemisinin AL1 1.76 million AS/AQ1 2.5 million pACT leaves with 0.7% artemisinin 8.6 million Open in a separate window 1http://www.rollbackmalaria.org/partnership/wg/wgprocurementsupply/docs/psmwg_ppACT-API.pdf p.2 [cited May 27, 2014]; 2Assumes a 6 day treatment with pACT, with each patient receiving 15 g dried leaves per full malaria treatment for leaves with 0.7% artemisinin. To obtain an amount of artemisinin equal to 1 T of the extracted drug, one would have to harvest 142.8 tons of dried A. annua leaves containing 0.7% artemisinin AL: Artemether/lumefantrine; AS/AQ: Artesunate/amodiaquine; pACT: Plant-based artemisinin combination therapy. As yet unpublished data from the Rich and Weathers labs demonstrated that pACT prevents emergence of artemisinin drug resistance; the plant itself seems to function as its own ACT (pACT). This would obviate the need for inclusion of a co-drug as used in currently administered ACTs. The co-drug costs at least as much as the artemisinic portion of the drug[6]. Consequently, elimination of the added co-drug could result in at least an additional 50% reduction in cost, so that the final pACT cost reduction is conservatively estimated to be far below that of a current course of ACT therapy. Considering that A. annua is nontoxic and safe to consume orally, dose may not have to be adjusted for children. On the other hand, the leaves taste bitter, so masking the taste, perhaps with sugar, should help with pediatric treatment. Our recent simulated digestion study showed that adding table sugar (sucrose) to pACT did not significantly alter the amount of artemisinin released after digestion, with the added benefit of doubling the amount of flavonoids released[115]. Comparison with emerging artemisinin sources or other newer antimalarial drugs There are at least three other emerging antimalarial therapeutic technologies: synthetic artemisinin[116], semi-synthetic artemisinin (SSA) production from genetically engineered microbes[117], and a single dose drug, OZ439[118]. In early 2013, Sanofi/PATH Drug Development Programme, announced they would have the capacity to produce up to 60 MT of SSA in 2014 at about $400/kg, depending on quantity; Sanofi now has WHO prequalification for its SSA[119]. Although not much cheaper than the current price of about $550/kg[120], supply would be more or less unlimited. Despite what might seem as an advantage to large amounts of SSA production, there are also some serious disadvantages, and comparison of some advantages and disadvantages for each of these new synthetic antimalarial drugs and pACT is noted in Table 5. Table 5 Comparison of emerging antimalarial therapeutic technologies with plant-based artemisinin combination therapy Technology Advantages Disadvantages Synthetic AN[116] Fully synthetic method giving AN = compound Lowers AN cost compared to extraction Requires co-drug to obviate emergence of AN drug resistance Not yet in production Needs sophisticated process Likely all under Western control Challenging patient compliance due to multiday dosing Semi-synthetic AN[117] Semi-synthetic method giving authentic AN Lowers AN cost compared to extraction Requires co-drug to obviate emergence of AN drug resistance Production began via Sanofi Needs sophisticated process Likely all under Western control Challenging patient compliance due to multiday dosing OZ439[118] Single dose cure insures patient compliance In successful Phase 2 trials Mechanism of action not the same as AN Probably low cost due to full synthesis Requires co-drug to obviate emergence of AN drug resistance Not yet in production Needs sophisticated process Likely all under Western control pACT[20–24] Has its own in planta co-drug to obviate emergence of AN drug resistance Very low cost Very consistent product Can be used to treat other diseases Can be locally owned, produced, managed, and distributed Not yet in production Likely to meet push back from pharmaceutical industry Challenging patient compliance due to multiday dosing Open in a separate window AN: Artemisinin. Go to: QUALITY ASSURANCE CONSIDERATIONS Agricultural quality The traditional and least costly method for cultivating A. annua uses seeds and in developing countries farmers prefer to save seeds from one growing season to the next. However, seed generated plants of A. annua will vary widely from generation to generation even with high quality starting stock (see review by Ferreira et al[10]). Stem cuttings of A. annua readily root in about two weeks, so clonal propagation via rooted cutting is recommended to eliminate this variability. Although this method of propagation is not cost effective for large plantations, it would work for a few hectares or for controlled environment agriculture. Given the large numbers of patients that could be treated from growing just a few hectares of A. annua (Table 3), clonal propagation by rooted stem cuttings is recommended. Since pACT therapy involves the direct consumption of the dried leaves of the plant, harvested leaf material must be kept clean, which is easiest to do in controlled environment agriculture and following Good Agricultural Procedures[121], particularly as applied to fresh produce[122]. However, controlled agriculture would probably result in loss of agricultural jobs, a concern to be assessed locally. Alternatively, great care must be taken during field harvest and post-harvest storage, so as not to affect the quality of the product. WHO has established good agricultural practices specifically for A. annua for purposes of artemisinin extraction[123], for general medicinal plants[124], and to minimize contamination of herbal medicines[125]. Chemical consistency and quantification To deliver a reliable dose of therapeutics to a patient, the dried leaves of harvested A. annua must have a reliable and consistent composition. Clonal propagation provides the required consistency. Recently we showed that of 10 crops harvested from vegetative and early flowering plants grown over three years under diverse conditions in the lab, field, and home garden, the artemisinin content of a single clone of A. annua (SAM) was 1.38% ± 0.26% (w/w)[77]. Thus, despite variations in culture and environmental conditions, a consistent level of the main therapeutic constituent can be achieved. Moreover, the content of harvested leaves is certainly not a guarantee of finished product, e.g., compressed leaf tablets. Analyses by Weathers et al[77] showed that although artemisinin content was very stable after tablet compression, other constituents vaied significantly. For example, although flavonoids increased with tablet compression, the more volatile monoterpenes decreased substantially. Thus, it is critical to monitor the composition profile of both incoming harvested material as well as the final product. Complex and expensive analytical procedures have been used to analyze the many products found in A. annua, but they are not necessary to measure and assure product quality. Artemisinin is easily extracted and then can be quantified using a variety of thin layer chromatography (TLC) methods and visualized with p-anisaldehyde stain[126,127]. Other key constituents like the flavonoids are also readily separated using TLC and visualized under either UV ± AlCl3 reagent[128]. Total flavonoids also can be quantified using inexpensive visible spectroscopy via the AlCl3 method with quercetin used as an inexpensive standard. To our knowledge no inexpensive, reliable spectrophotometric assay is available to measure artemisinin in complex plant extracts. Go to: SOCIOECONOMIC BENEFITS Other diseases Artemisinin and its derivatives are also effective against a number of viruses[129], a variety of human cancer cell lines[130–133], and several neglected tropical diseases including schistosomiasis[134], leishmaniasis[135,136], trypanosomiasis[137], and some livestock diseases[133,138]. Although they rank below malaria in terms of public health importance, schistosomiasis, leishmania, and trypanosomiasis result in estimated annual infections of about 240 million, 1.3 million (0.3 visceral and 1.0 cutaneous), and 30000, respectively[139]. These diseases along with many others respond to treatment with artemisinins. Although the IC50 is about 1000-fold greater than for Plasmodium sp., the greater apparent bioavailability of artemisinin via oral pACT[20–22] would likely reduce the amount of drug required for treatment. At present, pACT has not been tested in vivo for diseases other than malaria. Malaria treatment is further complicated for Human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS) patients. Malaria and HIV co-infection represents a major health burden in Africa mainly because it is now “well established that HIV infection results in a higher incidence and more severe manifestations of malaria”[140]. With a weakened immune system, AIDS patients are more susceptible to malaria and also respond slower to malaria therapy[140–142]. Furthermore, in a meta-analysis by Tusting et al[143], socioeconomic development strongly correlated with better malaria therapeutic outcomes. Recently, A. annua has demonstrated anti HIV activity[126,144] and thus oral consumption of the dried leaves of this herb will not only treat malaria, but should also enhance the well-being of HIV/AIDS patients. Agriculture, jobs and self-determination A. annua is grown in more than 75 countries[145]. In 2011 about 163 MT of artemisinin were extracted from plantations and small stakeholder farms mainly located in China, Vietnam, and Eastern Africa including Madagascar; value was about $550/kg[120]. With the advent of the production of semi synthetic artemisinin by Sanofi, 60 MT were projected for 2014 with an anticipated price of about $400/kg[119]. As this new source of artemisinin becomes available, the Netherlands Royal Tropical Institute projected that the market for natural Artemisia will significantly destabilize, undermining the security of farmers. The Tropical Institute was further concerned that “pharmaceutical companies will accumulate control and power over the production process; Artemisia producers will lose a source of income; and local production, extraction and (possibly) manufacturing of ACT in regions where malaria is prevalent will shift to the main production sites of Western pharmaceutical companies”, disrupting the fragile economics of these already impoverished countries[120]. The average small stakeholder crop area is about 0.2 ha in China and Africa[120], so while implementation of pACT may not require as much agricultural land as for extracted artemisinin, it could still help provide small stakeholders with a source of income. We have estimated that localized micro manufacturing plants could be constructed for < $50000 USD, and produce quality-controlled pACT tablets with readily verifiable contents. Our overall approach, schematically illustrated in Figure 3, leads to local control of malaria and possibly other artemisinin susceptible diseases while also improving the socioeconomic status of the populations. Figure 3 Overall scheme for plant-based artemlslnln combination therapy production. pACT: Plant-based artemisinin combination therapy; TLC: Thin layer chromatography. Go to: CONCLUSION Evidence is mounting for the therapeutic efficacy of the use of dried leaves of A. annua, pACT, to treat malaria and possibly other diseases. The complex mixture of antiparasitic compounds in the plant seems to account for its therapeutic activity with animal and human trials supporting this claim. It is also clear that the cost of using pACT is a fraction of that for any other current or emerging antimalarial therapeutic. Likewise, the recent evidence of persistent and/or asymptomatic malaria suggests that a more prophylactic approach to malaria using pACT or even A. annua tea may be warranted. Considering that for > 2000 years this plant was used in traditional Chinese medicine for treatment of fever with no apparent appearance of artemisinin drug resistance, taken together the cumulative evidence argues for inclusion of pACT into the arsenal of drugs to combat malaria, and very likely, other diseases. Core tip Artemisinin, extracted from the plant Artemisia annua (A. annua) L., and artemisinin derivatives are the current best antimalarial therapeutics and are delivered as artemisinin combination therapy (ACT). Availability and cost are problematic for the developing world where malaria is endemic. Oral consumption of A. annua dried leaves is more effective than the pure drug. A tea infusion of the leaves has prophylactic effects. Cost of producing and delivering the tea and A. annua dried leaf tablets is much more affordable than ACT. Go to: Acknowledgments Supported by Worcester Polytechnic Institute and University of Massachusetts Center for Clinical and Translational Science partially; partially by Award Number NIH-R15AT008277-01 from the National Center for Complementary and Alternative Medicine Go to: Footnotes Author contributions: Weathers PJ, Towler M, Hassanali A, Lutgen P and Engeu PO all participated in writing the article; Hassanali A, Lutgen P and Engeu PO provided clinical data; Weathers PJ and Towler M conducted analyses of lab and field samples. Go to: Contributor Information Pamela J Weathers, Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, United States. Melissa Towler, Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, United States. Ahmed Hassanali, School of Pure and Applied Sciences, Kenyatta University, Nairobi 20100, Kenya. Pierre Lutgen, IFBV-BELHERB, PO Box 98, L-6908 Niederanven, Luxembourg. Patrick Ogwang Engeu, Natural Chemotherapeutics Research Institute, Ministry of Health, PO Box 4864 Kampala, Uganda. Go to: REFERENCES 1. WHO. World Malaria Report. 2013 Available from: URL: http://www.who.int/malaria/publications/world_malaria_report_2013/en/ 2. Phyo AP, Nkhoma S, Stepniewska K, Ashley EA, Nair S, McGready R, ler Moo C, Al-Saai S, Dondorp AM, Lwin KM, Singhasivanon P, Day NP, White NJ, Anderson TJ, Nosten F. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–1966. [PMID: 22484134] [PMC free article] [PubMed] [Google Scholar] 3. Betson M, Sousa-Figueiredo JC, Atuhaire A, Arinaitwe M, Adriko M, Mwesigwa G, Nabonge J, Kabatereine NB, Sutherland CJ, Stothard JR. Detection of persistent Plasmodium spp infections in Ugandan children after artemether-lumefantrine treatment. Parasitology. 2014;16:1–11. [PMID: 24837880] [PMC free article] [PubMed] [Google Scholar] 4. Beshir KB, Sutherland CJ, Sawa P, Drakeley CJ, Okell L, Mweresa CK, Omar SA, Shekalaghe SA, Kaur H, Ndaro A, Chilongola J, Schallig HD, Sauerwein RW, Hallett RL, Bousema T. Residual Plasmodium falciparum parasitemia in Kenyan children after artemisinin-combination therapy is associated with increased transmission to mosquitoes and parasite recurrence. J Infect Dis. 2013;208:2017–2024. [PMID: 23945376] [PMC free article] [PubMed] [Google Scholar] 5. Lindblade KA, Steinhardt L, Samuels A, Kachur SP, Slutsker L. The silent threat: asymptomatic parasitemia and malaria transmission. Expert Rev Anti Infect Ther. 2013;11:623–639. [PMID: 23750733] [PubMed] [Google Scholar] 6. Yeung S, Van Damme W, Socheat D, White NJ, Mills A. Cost of increasing access to artemisinin combination therapy: the Cambodian experience. Malar J. 2008;7:84. [PMID: 18492245] [PMC free article] [PubMed] [Google Scholar] 7. O’Connell KA, Gatakaa H, Poyer S, Njogu J, Evance I, Munroe E, Solomon T, Goodman C, Hanson K, Zinsou C, Akulayi L, Raharinjatovo J, Arogundade E, Buyungo P, Mpasela F, Adjibabi CB, Agbango JA, Ramarosandratana BF, Coker B, Rubahika D, Hamainza B, Chapman S, Shewchuk T, Chavasse D. Got ACTs? Availability, price, market share and provider knowledge of anti-malarial medicines in public and private sector outlets in six malaria-endemic countries. Malar J. 2011;10:326. [PMID: 22039838] [PMC free article] [PubMed] [Google Scholar] 8. Davis B, Ladner J, Sams K, Tekinturhan E, de Korte D, Saba J. Artemisinin-based combination therapy availability and use in the private sector of five AMFm phase 1 countries. Malar J. 2013;12:135. [PMID: 23607504] [PMC free article] [PubMed] [Google Scholar] 9. Mikkelsen-Lopez I, Shango W, Barrington J, Ziegler R, Smith T, deSavigny D. The challenge to avoid anti-malarial medicine stock-outs in an era of funding partners: the case of Tanzania. Malar J. 2014;13:181. [PMID: 24885420] [PMC free article] [PubMed] [Google Scholar] 10. Ferreira JFS, Laughlin JC, Delabays N, de Magalhães PM. Cultivation genetics of Artemisia annua L. for increased production of the antimalarial artemisinin. Plant Gen Res. 2005;3:206–229. [Google Scholar] 11. Duke JA. Handbook of phytochemical constituents of GRAS herbs and other economic plants. Boca Raton, FL: CRC Press LLC; 2001. p. 70. [Google Scholar] 12. Duke MV, Paul RN, Elsohly HN, Sturtz G, Duke SO. Localization of artemisinin and artemisitene in foliar tissues of glanded and glandless biotypes of Artemisia annua L. Int J Plant Sci. 1994;155:365–372. [Google Scholar] 13. Ferreira JF, Luthria DL, Sasaki T, Heyerick A. Flavonoids from Artemisia annua L. as antioxidants and their potential synergism with artemisinin against malaria and cancer. Molecules. 2010;15:3135–3170. [PMID: 20657468] [PMC free article] [PubMed] [Google Scholar] 14. Bhakuni RS, Jain DC, Sharma RP, Kumar S. Secondary metabolites of Artemisia annua and their biological activity. Curr Sci. 2001;80:35–48. [Google Scholar] 15. Wang W, Wang Y, Zhang Q, Qi Y, Guo D. Global characterization of Artemisia annua glandular trichome transcriptome using 454 pyrosequencing. BMC Genomics. 2009;10:465. [PMID: 19818120] [PMC free article] [PubMed] [Google Scholar] 16. Räth K, Taxis K, Walz G, Gleiter CH, Li SM, Heide L. Pharmacokinetic study of artemisinin after oral intake of a traditional preparation of Artemisia annua L. (annual wormwood) Am J Trop Med Hyg. 2004;70:128–132. [PMID: 14993622] [PubMed] [Google Scholar] 17. Mueller MS, Runyambo N, Wagner I, Borrmann S, Dietz K, Heide L. Randomized controlled trial of a traditional preparation of Artemisia annua L. (Annual Wormwood) in the treatment of malaria. Trans R Soc Trop Med Hyg. 2004;98:318–321. [PMID: 15109558] [PubMed] [Google Scholar] 18. Silva LF, Magalhães PM, Costa MR, Alecrim Md, Chaves FC, Hidalgo Ade F, Pohlit AM, Vieira PP. In vitro susceptibility of Plasmodium falciparum Welch field isolates to infusions prepared from Artemisia annua L. cultivated in the Brazilian Amazon. Mem Inst Oswaldo Cruz. 2012;107:859–866. [PMID: 23147140] [PubMed] [Google Scholar] 19. Suberu JO, Gorka AP, Jacobs L, Roepe PD, Sullivan N, Barker GC, Lapkin AA. Anti-plasmodial polyvalent interactions in Artemisia annua L. aqueous extract--possible synergistic and resistance mechanisms. PLoS One. 2013;8:e80790. [PMID: 24244716] [PMC free article] [PubMed] [Google Scholar] 20. ICIPE. [cited 2014 May 25];Whole-leaf Artemisia annua-based antimalarial drug: report on proof-of-concepts studies. 2005 Available from: URL: http://www.google.com/url?sa=t&rct=j&q=&esrc=s&frm=l&source=web&cd=2&ved=0CDgQFjAB&url=http://www.iwerliewen.org/index.php/component/edocman/?task=document.download&id=96&Itemid=181&ei=J2miUbnFNo80QGoi4GACw&usg=AFQjCNHoLJmPt4n0AkKyBlXPSyl5W7rc6w&sig2=ppM08XltZglQLLiaojZxlw&bvm=bv.47008514,d.dmQ. 21. Weathers PJ, Arsenault PR, Covello P, McMickle A, Reed D, Teoh KH. Artemisinin production in Artemisia annua: studies in planta and results of a novel delivery method for treating malaria and other neglected diseases. Phytochem Rev. 2011;10:173–183. [PMID: 21643453] [PMC free article] [PubMed] [Google Scholar] 22. Weathers PJ, Elfawal MA, Towler MJ, Acquaah-Mensah GK, Rich SM. Pharmacokinetics of artemisinin delivered by oral consumption of Artemisia annua dried leaves in healthy vs. Plasmodium chabaudi-infected mice. J Ethnopharmacol. 2014;153:732–736. [PMID: 24661969] [PMC free article] [PubMed] [Google Scholar] 23. Elfawal MA, Towler MJ, Reich NG, Golenbock D, Weathers PJ, Rich SM. Dried whole plant Artemisia annua as an antimalarial therapy. PLoS One. 2012;7:e52746. [PMID: 23289055] [PMC free article] [PubMed] [Google Scholar] 24. Onimus M, Carteron S, Lutgen P. The surprising efficiency of Artemisia annua powder capsules. Medicin Aromat Plants. 2013;2:3. [Google Scholar] 25. Hsu E. The history of qing hao in the Chinese materia medica. Trans R Soc Trop Med Hyg. 2006;100:505–508. [PMID: 16566952] [PubMed] [Google Scholar] 26. Weathers PJ, Reed K, Hassanali A, Lutgen P, Engeu PO. Chapter 4: Whole plant approaches to therapeutic use of Artemisia annua L. (Asteraceae) In: Aftab T, Ferreira JFS, Khan MMA, Naeem M, editors. Artemisia annua - Pharmacology and Biotechnology. Springer: Heidelberg, GDR; 2014. pp. 51–74. [Google Scholar] 27. van der Kooy F, Verpoorte R. The content of artemisinin in the Artemisia annua tea infusion. Planta Med. 2011;77:1754–1756. [PMID: 21544776] [PubMed] [Google Scholar] 28. Carbonara T, Pascale R, Argentieri MP, Papadia P, Fanizzi FP, Villanova L, Avato P. Phytochemical analysis of a herbal tea from Artemisia annua L. J Pharm Biomed Anal. 2012;62:79–86. [PMID: 22305080] [PubMed] [Google Scholar] 29. Weathers PJ, Towler MJ. The flavonoids casticin and artemetin are poorly extracted and are unstable in an Artemisia annua tea infusion. Planta Med. 2012;78:1024–1026. [PMID: 22673829] [PMC free article] [PubMed] [Google Scholar] 30. Ogwang PE, Ogwal-Okeng J, Kasasa S, Ejobi F, Kabasa D, Obua C. Use of Artemisia annua L. infusion for malaria prevention: mode of action and benefits in a Ugandan community. British J Pharm Res. 2011;1:124–132. [Google Scholar] 31. Ogwang PE, Ogwal JO, Kasasa S, Olila D, Ejobi F, Kabasa D, Obua C. Artemisia annua L. infusion consumed once a week reduces risk of multiple episodes of malaria: a randomised trial in a Ugandan community. Trop J Pharm Res. 2012;13:445–453. [Google Scholar] 32. Mueller MS, Karhagomba IB, Hirt HM, Wemakor E. The potential of Artemisia annua L. as a locally produced remedy for malaria in the tropics: agricultural, chemical and clinical aspects. J Ethnopharmacol. 2000;73:487–493. [PMID: 11091003] [PubMed] [Google Scholar] 33. Blanke CH, Naisabha GB, Balema MB, Mbaruku GM, Heide L, Müller MS. Herba Artemisiae annuae tea preparation compared to sulfadoxine-pyrimethamine in the treatment of uncomplicated falciparum malaria in adults: a randomized double-blind clinical trial. Trop Doct. 2008;38:113–116. [PMID: 18453510] [PubMed] [Google Scholar] 34. De Donno A, Grassi T, Idolo A, Guido M, Papadia P, Cac-cioppola A, Villanova L, Merendino A, Bagordo F, Fanizzi FP. First-time comparison of the in vitro antimalarial activity of Artemisia annua herbal tea and artemisinin. Trans R Soc Trop Med Hyg. 2012;106:696–700. [PMID: 22986092] [PubMed] [Google Scholar] 35. Brown GD. The biosynthesis of artemisinin (Qinghaosu) and the phytochemistry of Artemisia annua L. (Qinghao) Molecules. 2010;15:7603–7698. [PMID: 21030913] [PMC free article] [PubMed] [Google Scholar] 36. van der Kooy F, Sullivan SE. The complexity of medicinal plants: the traditional Artemisia annua formulation, current status and future perspectives. J Ethnopharmacol. 2013;150:1–13. [PMID: 23973523] [PubMed] [Google Scholar] 37. WHO Position Statement. Effectiveness of Non-Pharmaceutical Forms of Artemisia annua L. against malaria. 2012 Available from: URL: http://www.who.int/malaria/position_statement_herbal_remedy_artemisia_annua_l.pdf. 38. Tiruneh G, Kebede Y, Yigzaw T. Use of the plant Artemisia annua as a natural anti-malarial herb in Arbaminch town. Ethiop J Health Biomed Sci. 2010;2:76–82. [Google Scholar] 39. Giao PT, Binh TQ, Kager PA, Long HP, Van Thang N, Van Nam N, de Vries PJ. Artemisinin for treatment of uncomplicated falciparum malaria: is there a place for monotherapy? Am J Trop Med Hyg. 2001;65:690–695. [PMID: 11791958] [PubMed] [Google Scholar] 40. Hien TT. An overview of the clinical use of artemisinin and its derivatives in the treatment of falciparum malaria in Viet Nam. Trans R Soc Trop Med Hyg. 1994;88(Suppl 1):S7–S8. [PMID: 8053033] [PubMed] [Google Scholar] 41. Mclntosh HM, Olliaro P. Artemisinin derivatives for treating severe malaria. Cochrane Database Syst Rev. 2000;2:CD000527. [PMID: 10796551] [PMC free article] [PubMed] [Google Scholar] 42. de Vries PJ, Dien TK. Clinical pharmacology and therapeutic potential of artemisinin and its derivatives in the treatment of malaria. Drugs. 1996;52:818–836. [PMID: 8957153] [PubMed] [Google Scholar] 43. Wan YD, Zang QZ, Wang JS. [Studies on the antimalarial action of gelatin capsule of Artemisia annua] Zhongguo Jishengchongxue Yu Jishengchongbing Zazhi. 1992;10:290–294. [PMID: 1303339] [PubMed] [Google Scholar] 44. Zhao KC, Song ZY. [Pharmacokinetics of dihydroqinghaosu in human volunteers and comparison with qinghaosu] Yaoxue Xuebao. 1993;28:342–346. [PMID: 8237378] [PubMed] [Google Scholar] 45. Alin MH, Ashton M, Kihamia CM, Mtey GJ, Björkman A. Clinical efficacy and pharmacokinetics of artemisinin monotherapy and in combination with mefloquine in patients with falciparum malaria. Br J Clin Pharmacol. 1996;41:587–592. [PMID: 8799526] [PMC free article] [PubMed] [Google Scholar] 46. Ilet KF, Batty KT. Artemisinin and its derivatives. In: Yu VL, Edwards G, McKinnon PS, Peloquin CA, Morse G, editors. Antimicrobial Therapy and Vaccines. ESun Technologies: Pittsburgh USA; 2005. pp. 981–1002. [Google Scholar] 47. Ashton M, Gordi T, Trinh NH, Nguyen VH, Nguyen DS, Nguyen TN, Dinh XH, Johansson M, Le DC. Artemisinin pharmacokinetics in healthy adults after 250, 500 and 1000 mg single oral doses. Biopharm Drug Dispos. 1998;19:245–250. [PMID: 9604124] [PubMed] [Google Scholar] 48. Dien TK, de Vries PJ, Khanh NX, Koopmans R, Binh LN, Duc DD, Kager PA, van Boxtel CJ. Effect of food intake on pharmacokinetics of oral artemisinin in healthy Vietnamese subjects. Antimicrob Agents Chemother. 1997;41:1069–1072. [PMID: 9145871] [PMC free article] [PubMed] [Google Scholar] 49. Ashton M, Hai TN, Sy ND, Huong DX, Van Huong N, Niêu NT, Công LD. Artemisinin pharmacokinetics is time-dependent during repeated oral administration in healthy male adults. Drug Metab Dispos. 1998;26:25–27. [PMID: 9443848] [PubMed] [Google Scholar] 50. Svensson US, Ashton M. Identification of the human cytochrome P450 enzymes involved in the in vitro metabolism of artemisinin. Br J Clin Pharmacol. 1999;48:528–535. [PMID: 10583023] [PMC free article] [PubMed] [Google Scholar] 51. Gueye PEO, Diallo M, deme AB, Badiane A, Dior DM, Ahouidi A, Abdoul AN, Dieng T, Lutgen P, Mbopup S, Sarr O. Tea Artemisia annua inhibits Plasmodium falciparum isolates collected in Pikine, Senegal. Af J Biochem Res. 2013;7:107–113. [Google Scholar] 52. Liu KC, Yang SL, Roberts MF, Elford BC, Phillipson JD. Antimalarial activity of Artemisia annua flavonoids from whole plants and cell cultures. Plant Cell Rep. 1992;11:637–640. [PMID: 24213368] [PubMed] [Google Scholar] 53. Elford BC, Roberts MF, Phillipson JD, Wilson RJ. Potentiation of the antimalarial activity of qinghaosu by methoxylated flavones. Trans R Soc Trop Med Hyg. 1987;81:434–436. [PMID: 3318019] [PubMed] [Google Scholar] 54. Lehane AM, Saliba KJ. Common dietary flavonoids inhibit the growth of the intraerythrocytic malaria parasite. BMC Res Notes. 2008;1:26. [PMID: 18710482] [PMC free article] [PubMed] [Google Scholar] 55. van Zyl RL, Seatlholo ST, van Vuuren SF, Viljoen AM. The biological activities of 20 nature identical essential oil constituents. J Essent Oil Res. 2006;18:129–133. Special Edition. [Google Scholar] 56. Melillo de Magalhães P, Dupont I, Hendrickx A, Joly A, Raas T, Dessy S, Sergent T, Schneider YJ. Anti-inflammatory effect and modulation of cytochrome P450 activities by Artemisia annua tea infusions in human intestinal Caco-2 cells. Food Chem. 2012;134:864–871. [PMID: 23107701] [PubMed] [Google Scholar] 57. Baraldi R, Isacchi B, Predieri S, Marconi G, Vincieri FF, Bilia AR. Distribution of artemisinin and bioactive flavonoids from Artemisia annua L. during plant growth. Biochem Syst Ecol. 2008;36:340–348. [Google Scholar] 58. Ganesh D, Fuehrer HP, Starzengrüber P, Swoboda P, Khan WA, Reismarnn JA, Mueller MS, Chiba P, Noedl H. Antiplasmodial activity of flavonol quercetin and its analogues in Plasmodium falciparum: evidence from clinical isolates in Bangladesh and standardized parasite clones. Parasitol Res. 2012;110:2289–2295. [PMID: 22215188] [PubMed] [Google Scholar] 59. Manach C, Donovan JL. Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Radic Res. 2004;38:771–785. [PMID: 15493450] [PubMed] [Google Scholar] 60. Tasdemir D, Lack G, Brun R, Rüedi P, Scapozza L, Perozzo R. Inhibition of Plasmodium falciparum fatty acid biosynthesis: evaluation of FabG, FabZ, and FabI as drug targets for flavonoids. J Med Chem. 2006;49:3345–3353. [PMID: 16722653] [PubMed] [Google Scholar] 61. Lin CM, Chen CT, Lee HH, Lin JK. Prevention of cellular ROS damage by isovitexin and related flavonoids. Planta Med. 2002;68:365–367. [PMID: 11988866] [PubMed] [Google Scholar] 62. Raguso RA, Schlumpberger BO, Kaczorowski RL, Holtsford TP. Phylogenetic fragrance patterns in Nicotiana sections Alatae and Suaveolentes. Phytochemistry. 2006;67:1931–1942. [PMID: 16843507] [PubMed] [Google Scholar] 63. Rodrigues Goulart H, Kimura EA, Peres VJ, Couto AS, Aquino Duarte FA, Katzin AM. Terpenes arrest parasite development and inhibit biosynthesis of isoprenoids in Plasmodium falciparum. Antimicrob Agents Chemother. 2004;48:2502–2509. [PMID: 15215101] [PMC free article] [PubMed] [Google Scholar] 64. Moura IC, Wunderlich G, Uhrig ML, Couto AS, Peres VJ, Katzin AM, Kimura EA. Limonene arrests parasite development and inhibits isoprenylation of proteins in Plasmodium falciparum. Antimicrob Agents Chemother. 2001;45:2553–2558. [PMID: 11502528] [PMC free article] [PubMed] [Google Scholar] 65. Su V, King D, Woodrow I, McFadden G, Gleadow R. Plasmodium falciparum growth is arrested by monoterpenes from eucalyptus oil. Flavour Frag J. 2008;23:315–318. [Google Scholar] 66. Miller JA, Hakim IA, Chew W, Thompson P, Thomson CA, Chow HH. Adipose tissue accumulation of d-limonene with the consumption of a lemonade preparation rich in d-limonene content. Nutr Cancer. 2010;62:783–788. [PMID: 20661827] [PMC free article] [PubMed] [Google Scholar] 67. Charles DJ, Simon JE, Wood KV, Heinstein P. Germplasm variation in artemisinin content of Artemisia annua using an alternative method of artemisinin analysis from crude plant extracts. J Nat Prod. 1990;53:157–160. [Google Scholar] 68. Juergens UR, Engelen T, Racké K, Stöber M, Gillissen A, Vetter H. Inhibitory activity of 1,8-cineol (eucalyptol) on cytokine production in cultured human lymphocytes and monocytes. Pulm Pharmacol Ther. 2004;17:281–287. [PMID: 15477123] [PubMed] [Google Scholar] 69. Kovar KA, Gropper B, Friess D, Ammon HP. Blood levels of 1,8-cineole and locomotor activity of mice after inhalation and oral administration of rosemary oil. Planta Med. 1987;53:315–318. [PMID: 3671550] [PubMed] [Google Scholar] 70. Stimpfl T, Nasel B, Nasel C, Binder R, Vycudilik W, Buch bauer G. Concentration of 1,8-cineol in human blood during prolonged inhalation. Chem Senses. 1995;20:349–350. [PMID: 7552045] [PubMed] [Google Scholar] 71. Kengne RDC. Caracterisation physicochimique de Artemisia annua (asteraceae), plante medicinale cultivee au Cameroun. MS thesis in Organic Chemistry. Univ de Dschang Republic of Cameroun. 2010 [Google Scholar] 72. Akhtar F, Rizvi MM, Kar SK. Oral delivery of curcumin bound to chitosan nanoparticles cured Plasmodium yoelii infected mice. Biotechnol Adv. 2012;30:310–320. [PMID: 21619927] [PubMed] [Google Scholar] 73. Akkawi M, Jaber S, Abu-Remeleh Q, Ogwang PE, Lutgen P. Investigations of Artemisia annua and Artemisia sieberi water extracts inhibitory effects on β-hematin formation. Medicin Arom Plants. 2014;3150 [Google Scholar] 74. Liu AR, Yu ZY, Lu LL, Sui ZY. [The synergistic action of guanghuoxiang volatile oil and sodium artesunate against Plasmodium berghei and reversal of SA-resistant Plasmodium berghei] Zhongguo Jishengchongxue Yu Jishengchongbing Zazhi. 2000;18:76–78. [PMID: 12567719] [PubMed] [Google Scholar] 75. Yang ZN, Zhu SQ, Yu ZW. Comparison of terpene components from flowers of Artemisia annua. Bangladesh J Pharmacol. 2012;7:114–119. [Google Scholar] 76. Khangholil S, Rezaeinodehi A. Effect of drying temperature on essential oil content and composition of sweet wormwood (Artemisia annua) growing wild in Iran. Pak J Biol Sci. 2008;11:934–937. [PMID: 18814660] [PubMed] [Google Scholar] 77. Weathers PJ, Towler MJ. Changes in key constituents of clonally propagated Artemisia annua L. during preparation of compressed leaf tablets for possible therapeutic use. Ind Crop Prod. 2014 In press. [PMC free article] [PubMed] [Google Scholar] 78. Juteau F, Masotti V, Bessière JM, Dherbomez M, Viano J. Antibacterial and antioxidant activities of Artemisia annua essential oil. Fitoterapia. 2002;73:532–535. [PMID: 12385883] [PubMed] [Google Scholar] 79. Roberts DW, Rank RG, Weidanz WP, Finerty JF. Prevention of recrudescent malaria in nude mice by thymic grafting or by treatment with hyperimmune serum. Infect Immun. 1977;16:821–826. [PMID: 330396] [PMC free article] [PubMed] [Google Scholar] 80. Cherneva E, Pavlovic V, Smelcerovic A, Yancheva D. The effect of camphor and borneol on rat thymocyte viability and oxidative stress. Molecules. 2012;17:10258–10266. [PMID: 22926306] [PMC free article] [PubMed] [Google Scholar] 81. Lopes NP, Kato MJ, Andrade EH, Maia JG, Yoshida M, Planchart AR, Katzin AM. Antimalarial use of volatile oil from leaves of Virola surinamensis (Rol.) Warb. by Waiãpi Amazon Indians. J Ethnopharmacol. 1999;67:313–319. [PMID: 10617066] [PubMed] [Google Scholar] 82. Muzemil A. Determination of artemisinin and essential oil contents of Artemisia annua L. grown in Ethiopia and in vivo antimalarial activity of its crude extracts against Plasmodium berghei in mice. Ethiopia: MS Thesis in Medicinal Chemistry, Addis Ababa University. 2008 [Google Scholar] 83. Li Y, Hu HB, Zheng XD, Zhu JH, Liu LP. Composition and antimicrobial activity of essential oil from the aerial part of Artemisia annua. J Medicin Plants Res. 2011;5:3629–3633. [Google Scholar] 84. Ahn BY, Jung MY. Antioxidant and protective activity of polysaccharide extract from Artemisia iwayomogi Kitamura stems on UVB-damaged mouse epidermis. J Appl Biol Chem. 2011;54:184–189. [Google Scholar] 85. Han H. MS Thesis in Biochem & Molec Biol. Changchun PRC: Northeast Normal Univ; 2008. Antimalarial Activity of Ginseng Polysaccharides and Bulgaria Inquinans Polysaccharides. [Google Scholar] 86. Andrews KT, Klatt N, Adams Y, Mischnick P, Schwartz-Albiez R. Inhibition of chondroitin-4-sulfate-specific adhesion of Plasmodium falciparum-infected erythrocytes by sulfated polysaccharides. Infect Immun. 2005;73:4288–4294. [PMID: 15972521] [PMC free article] [PubMed] [Google Scholar] 87. Xiao L, Yang C, Patterson PS, Udhayakumar V, Lal AA. Sulfated polyanions inhibit invasion of erythrocytes by plasmodial merozoites and cytoadherence of endothelial cells to parasitized erythrocytes. Infect Immun. 1996;64:1373–1378. [PMID: 8606103] [PMC free article] [PubMed] [Google Scholar] 88. Clark DL, Su S, Davidson EA. Saccharide anions as inhibitors of the malaria parasite. Glycoconj J. 1997;14:473–479. [PMID: 9249145] [PubMed] [Google Scholar] 89. Munir M, Tjandra H, Rampengan TH, Mustadjab I, Wulur FH. Heparin in the treatment of cerebral malaria. Paediatr Indones. 1980;20:47–50. [PMID: 6988763] [PubMed] [Google Scholar] 90. Rampengan TH. Cerebral malaria in children Comparative study between heparin, dexamethasone and placebo. Paediatr Indones. 1991;31:59–66. [PMID: 1852471] [PubMed] [Google Scholar] 91. Adams Y, Freeman C, Schwartz-Albiez R, Ferro V, Parish CR, Andrews KT. Inhibition of Plasmodium falciparum growth in vitro and adhesion to chondroitin-4-sulfate by the heparan sulfate mimetic PI-88 and other sulfated oligosaccharides. Antimicrob Agents Chemother. 2006;50:2850–2852. [PMID: 16870784] [PMC free article] [PubMed] [Google Scholar] 92. Ashok PK, Upadhyaya K. Preliminary phytochemical screening and physicochemical parameters of Artemisia absinthium and Artemisia annua. J Pharmacog Phytochem. 2013;1:229–235. [Google Scholar] 93. Massiha A, Khoshkholgh-Pahlaviani MM, Issazadeh K, Bidarigh S, Zarrabi S. Antibacterial activity of essential oils and plant extracts of Artemisia (Artemisia annua L.) in vitro. Zahedan J Res Med Sci. 2013;15:14–18. [Google Scholar] 94. Podolak I, Galanty A, Sobolewska D. Saponins as cytotoxic agents: a review. Phytochem Rev. 2010;9:425–474. [PMID: 20835386] [PMC free article] [PubMed] [Google Scholar] 95. Sun HX, Xie Y, Ye YP. Advances in saponin-based adjuvants. Vaccine. 2009;27:1787–1796. [PMID: 19208455] [PubMed] [Google Scholar] 96. Haruna M, Tanaka M, Sugimoto T, Kojima R, Suzuki Y, Konoshima T, Kozuka M, Ito K. Alteration of Na permeability in human erythrocytes as studied by 23Na-NMR and inhibition of the Na, K -ATPase activities with saponins: Interactions of Gleditsia saponins with human erythrocyte membranes. Bioorg Med Chem Lett. 1995;5:827–830. [Google Scholar] 97. Francis G, Kerem Z, Makkar HP, Becker K. The biological action of saponins in animal systems: a review. Br J Nutr. 2002;88:587–605. [PMID: 12493081] [PubMed] [Google Scholar] 98. Meotti FC, Ardenghi JV, Pretto JB, Souza MM, d′ Avila Moura J, Junior AC, Soldi C, Pizzolatti MG, Santos AR. Antinociceptive properties of coumarins, steroid and dihydro-styry1-2-pyrones from Polygala sabulosa (Polygalaceae) in mice. J Pharm Pharmacol. 2006;58:107–112. [PMID: 16393470] [PubMed] [Google Scholar] 99. Chang TN, Deng JS, Chang YC, Lee CY, Jung-Chun L, Lee MM, Peng WH, Huang SS, Huang GJ. Ameliorative Effects of Scopoletin from Crossostephium chinensis against Inflammation Pain and Its Mechanisms in Mice. Evid Based Complement Alternat Med. 2012;2012:595603. [PMID: 22991572] [PMC free article] [PubMed] [Google Scholar] 100. Malik A, Kushnoor A, Saini V, Singhal1S, Kumar S, Yadav YC. In vitro antioxidant properties of scopoletin. J Chem Pharm Res. 2011;3:659–665. [Google Scholar] 101. Moon PD, Lee BH, Jeong HJ, An HJ, Park SJ, Kim HR, Ko SG, Um JY, Hong SH, Kim HM. Use of scopoletin to inhibit the production of inflammatory cytokines through inhibition of the IkappaB/NF-kappaB signal cascade in the human mast cell line HMC-1. Eur J Pharmacol. 2007;555:218–225. [PMID: 17113069] [PubMed] [Google Scholar] 102. Cherng JM, Chiang W, Chiang LC. Immunomodulatory activities of common vegetables and spices of Umbelliferae and its related coumarins and flavonoids. Food Chem. 2008;106:944–950. [Google Scholar] 103. Surono IS, Nishigaki T, Endaryanto A, Waspodo P. Indonesian biodiversities, from microbes to herbal plants as potential functional foods. J Fac Agric Shinshu U. 2008;44:23–27. [Google Scholar] 104. Ezeokonkwo CA, Obidoa O. Effect of scopoletin on erythrocyte membrane ion motive ATPases. Nigerian J Nat Prod Med. 2001;5:37–40. [Google Scholar] 105. Dunn MJ. Alterations of red blood cell sodium transport during malarial infection. J Clin Invest. 1969;48:674–684. [PMID: 4975361] [PMC free article] [PubMed] [Google Scholar] 106. Clark IA, Alleva LM, Mills AC, Cowden WB. Pathogenesis of malaria and clinically similar conditions. Clin Microbiol Rev. 2004;17:509–539. table of contents [PMID: 15258091] [PMC free article] [PubMed] [Google Scholar] 107. Ding Z, Dai Y, Wang Z. Hypouricemic action of scopoletin arising from xanthine oxidase inhibition and uricosuric activity. Planta Med. 2005;71:183–185. [PMID: 15729630] [PubMed] [Google Scholar] 108. de Vries PJ, de Vries PJ, Nguyen GC, de Goeje P, de Goeje P. Production and Application of Artemisinin in Vietnam. Institute of Materia Medica, Viet Nam and the University of Amsterdam: Gioi Publishers. 1999 [Google Scholar] 109. Atkinson B, Mavituna F. Biochemical Engineering and Biotechnology Handbook. 2nd ed. USA: Stockton Press; 1991. pp. 1059–1110. [Google Scholar] 110. Lim JAC, Patkar A, McDonagh G, Sinclair A, Lucy P. Modeling bioprocess cost: the economic benefits of expression technology based on Pseudomonas fluorescens. Bioprocess Intnl. 2010;8:62–70. [Google Scholar] 111. Lapkin AA, Plucinski PK, Cutler M. Comparative assessment of technologies for extraction of artemisinin. J Nat Prod. 2006;69:1653–1664. [PMID: 17125242] [PubMed] [Google Scholar] 112. Griffee P, Diemer P. Artemisia annua; the plant, production, processing and medicinal applications. Food and Agriculture Organization of the United Nations. Available from: URL: http://ecoport.org/ep?SearchType=earticleView&earticleld=727&p age=2#section5675. 113. Sipler D, Weathers PJ. Artemisia annua as a high value crop and weed control. Available from: URL: http://small-farm.org/SARE FNE12-766.html. 114. RBM Active pharmaceutical ingredient requirements for the manufacture of ACTs. Available from: URL: http://www.roll-backmalaria.org/partnership/wg/wg_procurementsupply/docs/psmwg_ppACT-API.pdf. 115. Weathers PJ, Jordan NJ, Lasin P, Towler MJ. Simulated digestion of dried leaves of Artemisia annua consumed as a treatment (pACT) for malaria. J Ethnopharmacol. 2014;151:858–863. [PMID: 24316176] [PMC free article] [PubMed] [Google Scholar] 116. Zhu C, Cook SP. A concise synthesis of (+)-artemisinin. J Am Chem Soc. 2012;134:13577–13579. [PMID: 22866604] [PubMed] [Google Scholar] 117. Paddon CJ, Westfall PJ, Pitera DJ, Benjamin K, Fisher K, McPhee D, Leavell MD, Tai A, Main A, Eng D, Polichuk DR, Teoh KH, Reed DW, Treynor T, Lenihan J, Fleck M, Bajad S, Dang G, Dengrove D, Diola D, Dorin G, Ellens KW, Fickes S, Galazzo J, Gaucher SP, Geistlinger T, Henry R, Hepp M, Horning T, Iqbal T, Jiang H, Kizer L, Lieu B, Melis D, Moss N, Re-gentin R, Secrest S, Tsuruta H, Vazquez R, Westblade LF, Xu L, Yu M, Zhang Y, Zhao L, Lievense J, Covello PS, Keasling JD, Reiling KK, Renninger NS, Newman JD. High-level semisynthetic production of the potent antimalarial artemisinin. Nature. 2013;496:528–532. [PMID: 23575629] [PubMed] [Google Scholar] 118. Charman SA, Arbe-Barnes S, Bathurst IC, Brun R, Campbell M, Charman WN, Chiu FC, Chollet J, Craft JC, Creek DJ, Dong Y, Matile H, Maurer M, Morizzi J, Nguyen T, Papastogiannidis P, Scheurer C, Shackleford DM, Sriraghavan K, Stingelin L, Tang Y, Urwyler H, Wang X, White KL, Wittlin S, Zhou L, Vennerstrom JL. Synthetic ozonide drug candidate OZ439 offers new hope for a single-dose cure of uncomplicated malaria. Proc Natl Acad Sci USA. 2011;108:4400–4405. [PMID: 21300861] [PMC free article] [PubMed] [Google Scholar] 119. A2s2. [cited 2014 May 25]; Available from: URL: http://www.a2s2.org. 120. ETC Group. Synthetic Biology: Livelihoods and Biodiversity. [Cited: 2014-05-25];Artemisinin. Available from: URL: http://www.etcgroup.org/files/CBD_Artemisinin_case_st udy_TA.pdf. 121. Kentucky Cooperative Extension Service. [Cited: 2014-05-25];GAP Good Agricultural Practices (GAP) Available from: URL: http://www.uky.edu/Ag/CCD/introsheets/gap.pdf. 122. Pewtrusts. [Cited: 2014-05-27];GAP Comparison of gaps governing the growing and harvesting of fresh produce. Available from: URL: http://www.pewhealth.org/upload-edFiles/PHG/Content_Level_Pages/Reports/PSP-RPT-GAP-Governing-Fresh-Produce.pdf. 123. World Health Organization. WHO monograph on good agricultural and collection practices (GACP) for Artemisia annua L. Available from: URL: http://www.who.int/malaria/publications/atoz/9241594438/en/ 124. World Health Organization. WHO guidelines on good agricultural and collection practices (GACP) for medicinal plants. Available from: URL: http://whqlibdoc.who.rnt/publications/2003/9241546271.pdf. 125. World Health Organization. WHO guidelines for assessing quality of herbal medicines with reference to contaminants and residues. Available from: URL: http://apps.who.int/medicrnedocs/documents/s14878e/s14878e.pdf. 126. Marchand E, Atemnkeng MA, Vanermen S, Plaizier-Vercammen J. Development validation of a simple thin layer chromatographic method for the analysis of artemisinin in Artemisia annua L. plant extracts. Biomed Chromatogr. 2008;22:454–459. [PMID: 18059047] [PubMed] [Google Scholar] 127. Koobkokkruad T, Chochai A, Kerdmanee C, De-Eknamkul W. TLC-densitometric analysis of artemisinin for the rapid screening of high-producing plantlets of Artemisia annua L. Phytochem Anal. 2007;18:229–234. [PMID: 17500366] [PubMed] [Google Scholar] 128. Arvouet-Grand A, Vennat B, Pourrat A, Legret P. [Standardization of propolis extract and identification of principal constituents] J Pharm Belg. 1994;49:462–468. [PMID: 7884635] [PubMed] [Google Scholar] 129. Efferth T, Romero MR, Wolf DG, Stamminger T, Marin JJ, Marschall M. The antiviral activities of artemisinin and artesunate. Clin Infect Dis. 2008;47:804–811. [PMID: 18699744] [PubMed] [Google Scholar] 130. Efferth T. Artemisinin: a versatile weapon from traditional Chinese medicine. In: Ramawat KG, editor. Herbal drugs: ethnomedicrne to modern medicine. Springer Verlag: Heidelberg; 2009. pp. 179–194. [Google Scholar] 131. Efferth T, Herrmann F, Tahrani A, Wink M. Cytotoxic activity of secondary metabolites derived from Artemisia annua L. towards cancer cells in comparison to its designated active constituent artemisinin. Phytomedicine. 2011;18:959–969. [PMID: 21831619] [PubMed] [Google Scholar] 132. Firestone GL, Sundar SN. Anticancer activities of artemisinin and its bioactive derivatives. Expert Rev Mol Med. 2009;11:e32. [PMID: 19883518] [PubMed] [Google Scholar] 133. Brisibe EA, Umoren UE, Brisibe F, Magalhäes PM, Ferreira JFS, Luthria D, Wu X, Prior RL. Nutritional characterization and antioxidant capacity of different tissues of Artemisia annua L. Food Chem. 2009;115:1240–1246. [Google Scholar] 134. Utzinger J, Xiao S, Keiser J, Chen M, Zheng J, Tanner M. Current progress in the development and use of artemether for chemoprophylaxis of major human schistosome parasites. Curr Med Chem. 2001;8:1841–1860. [PMID: 11772354] [PubMed] [Google Scholar] 135. Avery MA, Muraleedharan KM, Desai PV, Bandyopadhyaya AK, Furtado MM, Tekwani BL. Structure-activity relationships of the antimalarial agent artemisinin 8. design, synthesis, and CoMFA studies toward the development of artemisinin-based drugs against leishmaniasis and malaria. J Med Chem. 2003;46:4244–4258. [PMID: 13678403] [PubMed] [Google Scholar] 136. Sen R, Bandyopadhyay S, Dutta A, Mandal G, Ganguly S, Saha P, Chatterjee M. Artemisinin triggers induction of cell-cycle arrest and apoptosis in Leishmania donovani promastigotes. J Med Microbiol. 2007;56:1213–1218. [PMID: 17761485] [PubMed] [Google Scholar] 137. Mishina YV, Krishna S, Haynes RK, Meade JC. Artemisinins inhibit Trypanosoma cruzi and Trypanosoma brucei rhodesiense in vitro growth. Antimicrob Agents Chemother. 2007;51:1852–1854. [PMID: 17339374] [PMC free article] [PubMed] [Google Scholar] 138. Ferreira JF, Peaden P, Keiser J. In vitro trematocidal effects of crude alcoholic extracts of Artemisia annua, A. absinthium, Asimina triloba, and Fumaria officinalis: trematocidal plant alcoholic extracts. Parasitol Res. 2011;109:1585–1592. [PMID: 21562762] [PubMed] [Google Scholar] 139. World Health Organization. The 17 neglected tropical diseases. Available from: URL: http://www.who.int/neglected_diseases/diseases/en/ 140. Marconi VC. Commentary: Malaria and HIV transmission: old meets new in a deadly partnership or an opportunity for healthcare synergism? Iit J Epidemiol. 2011;40:940–944. [PMID: 21393253] [PubMed] [Google Scholar] 141. Kamya MR, Gasasira AF, Yeka A, Bakyaita N, Nsobya SL, Francis D, Rosenthal PJ, Dorsey G, Havlir D. Effect of HIV-1 infection on antimalarial treatment outcomes in Uganda: a population-based study. J Infect Dis. 2006;193:9–15. [PMID: 16323126] [PubMed] [Google Scholar] 142. Ezeamama AE, Spiegelman D, Hertzmark E, Bosch RJ, Manji KP, Duggan C, Kupka R, Lo MW, Okuma JO, Kisenge R, Aboud S, Fawzi WW. HIV infection and the incidence of malaria among HIV-exposed children from Tanzania. J Infect Dis. 2012;205:1486–1494. [PMID: 22457274] [PMC free article] [PubMed] [Google Scholar] 143. Tusting LS, Willey B, Lucas H, Thompson J, Kafy HT, Smith R, Lindsay SW. Socioeconomic development as an intervention against malaria: a systematic review and metaanalysis. Lancet. 2013;382:963–972. [PMID: 23790353] [PubMed] [Google Scholar] 144. Lubbe A, Seibert I, Klimkait T, van der Kooy F. Ethnopharmacology in overdrive: the remarkable anti-HIV activity of Artemisia annua. J Ethnopharmacol. 2012;141:854–859. [PMID: 22465592] [PubMed] [Google Scholar] 145. Willcox ML, Burton S, Oyweka R, Namyalo R, Challand S, Lindsey K. Evaluation and pharmacovigilance of projects promoting cultivation and local use of Artemisia annua for malaria. Malar J. 2011;10:84. [PMID: 21481234] [PMC free article] [PubMed] [Google Scholar]
  12. 中共加快一体化应急应战管理体系建设。 征求意见稿已下发,通过后全国实施, 包括企事业单位、国企、大型私企,都要设立武装部。发生对外战争或内部动乱时,一是对付老百姓,二是对付外面。 中共想借美方希望中共调解中东之乱作筹码,换取美日菲在南海问题上让步。 中阿部长级峰会很快会在北京召开。 中共不希望中东和平,希望扩展自己的中东影响力。 习近平5月初将出访法国、塞尔维亚、匈牙利,中共内部正在设计扩大习访欧的成果和影响力。习将与法国总统马克龙共同发表有关俄乌战争联合声明。 伊朗的挫败整个动摇了中共提出的“向东看”,也影响/降低了中东对中共的依赖和倚仗。 欧盟将对中共十几家中资港资企业进行制裁,匈牙利总理欧尔班主动询问中共是否需要其出面阻止。欧尔班在上海有大量资产,其女儿在疫情期间赴上海打理资产。 泽连斯基希望与习近平在匈牙利见面,习尚未答复。如果习不见面,特使李辉将与乌克兰驻华大使见面。 习要求军委加快落实向友好国家出口更新更好军事设备。第一批国家有委内瑞拉、哈萨克斯坦、古巴、乌兹别克斯坦、也门。 出口设备涉及枪炮、器械、炸弹、无人机、导弹,包括红旗导弹歼10-100,飞弩 FN6、FN16 便携式防空导弹、运3运输机、飞鸿901无人机等。 所罗门群岛现任总理梅纳西·索加瓦雷(Manasseh Sogavare)认为自己连选后会发生内乱,希望中共根据2022年签署的安全协议,出人出装备,帮其维稳。 瑙鲁在与中共谈警务合作安全协议,培训、装备等费用都由中共承担。 普京连任后首次出访定于5·15访华,将要求中共协助说服欧洲,推动俄罗斯主导的“欧亚大陆安全架构”,其核心是欧亚人主导欧亚问题。 中共欧亚事务特使李辉在北京大范围约见各国驻华大使,说服他们支持俄罗斯。 针对台湾赖清德5·20上任,习近平要求,调动各部门各层级力量,破坏台湾的国际经贸环境,并进行军事打压。对与台交好国要分类施策。要打压报复惩戒台湾邦交国。各领事馆要按照中共中央精神灵活处置。对美国要软硬兼施两手准备,用好美国有求于中国的心态,提前施以重手迫使美国履行对台承诺。对岛内各派要拉打结合,要让民进党支持者付出代价。以上已经在实施中。 中共一直在围绕台海战争做准备,向各有关部门布置了未来3~6个月的演练内容。目前已下达涉外动员的8个方面能力提升要求: 1)协同友好国家支持能力; 2)分化打击欧美的能力; 3)利用仇美国家袭击干扰美国的能力; 4)主持北朝鲜牵制美韩的能力; 5)影响国际舆论的能力; 6)侦察情报打击能力; 7)诱骗误导能力; 8)应对突发不可控的外部环境的能力。 非洲东部的索马里兰向中共示好,称只要中共支持其独立建国, 将与台湾断绝关系。 中共海军已通过加快在非洲和北印度洋推进提高实现可持续战斗力的规划,将需要6个月时间预演,旨在维持中共在相关地区的军事存在和持续战斗力。 中共正在推动核技术在非洲落地。 习近平批示交通部组建大型特殊运输团队,已经针对该团队的规模、归属、如何避免美国制裁等在各城市做调研,旨在为应对中共国本土、台海地区、或海外其他地区发生大规模战争行动做准备。 Breaking: New exclusive information released by the NFSC on April 27th (1 of 18) The CCP is accelerating the construction of a society-wide civil-military integrated emergency response management system, the draft of which has been issued for comment and will be implemented nationwide after its adoption. Xi wants to use the CCP's influence in the Middle East to take advantage of the U.S.'s desire to coordinate with the CCP. He also wants to recieve the U.S., Japan and the Philippines' concessions on the issue of the South China Sea. A Sino-Arab State Summit will soon be held in Beijing, attended by some heads of state from Middle Eastern countries. Xi Jinping is visiting France, Serbia and Hungary from May 6-11, and the Communist Party of China (CPC) is internally designing ways to amplify the results and significance of Xi's trip to Europe. Xi and Macron will issue a joint statement on the Russia-Ukraine war. The recent setback for Iran in the Middle East conflict has shaken the Middle East countries' confidence in the security of the Chinese Communist Party (CCP)'s "Look East" proposal, affecting the Middle East's reliance on the CCP. The European Union is preparing to impose sanctions on a dozen Chinese and Hong Kong companies, and Hungary has asked the CCP if it needs assistance in the EU to stop the sanctions. Offering to mediate, Hungarian Prime Minister Viktor Orbán has substantial assets in the Shanghai, which his daughter managed while she is in Shanghai during the epidemic. Ukrainian President Zelensky has offered to meet with Xi during his visit to Hungary, which has not yet been confirmed by Xi. If not, the CCP envoy plans to meet with the Ukrainian ambassador to China in Beijing. Xi Jinping instructed the Military Commission to speed up the implementation of the export of military equipment, including guns, instruments, bombs, drones, and missiles, to countries friendly to the Chinese Communist Party. The first exports involved countries such as Venezuela, Kazakhstan, Cuba, Uzbekistan, and Yemen. Specific weapons include the Red Flag series Sword 10-100, Flying Crossbow FN-6, FN-16 portable air defense missiles, Transport 3, and Feihong 901 drones. The current Prime Minister of the Solomon Islands, Manasseh Sogavare, has given the CCP an indication that he can be re-elected, but there will be civil unrest in the country. Australia and New Zealand will not intervene, hoping to help stabilize the country after the CCP steps in under a security agreement signed between the two sides in 2022. Nauru is negotiating a security agreement with the Chinese Communist Party for police cooperation. The Chinese Communist Party will bear the costs of funding, training, and equipment. Putin's first trip to China after his reelection is scheduled for May 15, and he will ask the CCP to help promote the Russian-led Eurasian Continental Security Architecture, which centers on the idea of "Eurasians taking charge of Eurasian issues," and for the CCP to lobby its European allies to support the Russian architecture. Putin's first trip to China after his reelection is scheduled for May 15, and he will ask the CCP to help promote the Russian-led Eurasian Continental Security Architecture, which centers on the idea of "Eurasians taking charge of Eurasian issues," and for the CCP to lobby its European allies to support the Russian architecture. Li Hui, the Chinese Communist Party's Special Representative for Eurasian Affairs, has made extensive appointments with ambassadors in Beijing to convince them to support Russia. So far, we have learned that the BRICS countries, SCO member countries, some African countries, and a few Eurasian countries have expressed their support. In response to Taiwan's Lai Ching-Te taking office in 520, Xi Jinping has called for mobilizing forces from all sectors and at all levels to suppress and undermine Taiwan's international environment, economy, trade, science, technology, and military. Countries that are friendly with Taiwan should be categorized, suppressed, retaliated against, and punished. The consulates should flexibly follow the intent of the Central Committee of the Chinese Communist Party. The CCP has been preparing for "peace and war in the Taiwan Strait" and has set out the contents of exercises for the next three to six months to all relevant departments. Synergizing the supportive capacity of friendly countries The ability to divide and conquer Europe and America Use of America-hate attacks to interfere with U.S. capabilities Leveraging North Korea's ability to hold back the U.S., Japan and South Korea Ability to influence international public opinion Reconnaissance Intelligence Strike Capability Ability to entice and mislead Ability to cope with unexpected and uncontrollable external environment Somaliland in eastern Africa has made overtures to the Chinese Communist Party (CCP), saying it will sever ties with Taiwan as long as the CCP supports its independent statehood. The Chinese Communist Navy's plan to accelerate its advance in Africa and the northern Indian Ocean to improve its ability to achieve sustainable combat power has been formally adopted after a six-month preview of the plan. The Chinese Communist Party of China (CCP) is pushing for nuclear technology in Africa. Xi Jinping instructed the Ministry of Transportation to set up a large fleet of special transport ships for special operations in wartime. Coordinated research is now underway on multi-layered issues such as size, ownership, and how to avoid sanctions.
  13. 产品计划:黄花蒿提取青蒿素标准套件 1. 产品概述: 我们提出开发一款黄花蒿提取青蒿素标准套件,旨在提供一种简便、可靠的方法,使个人或组织能够在家或实验室中从黄花蒿植物中提取青蒿素,并将其转化为青蒿琥脂。该产品套件将提供所有必要的材料和详细的操作说明,以确保用户能够成功地提取并转化青蒿素。 2. 产品组成: • 黄花蒿提取套件包括: • 干燥黄花蒿植物样品 • 提取用的溶剂(如乙醚或丙酮) • 酸性催化剂(如硫酸) • 转化用的醇 • 化学实验器材(如烧杯、滤纸、玻璃棒等) • 详细的操作手册 3. 产品优势: • 简便易用:产品套件提供了一套详细的操作说明,即使是初学者也能轻松完成提取和转化过程。 • 经济实惠:相比购买成品青蒿素或青蒿琥脂,使用本产品套件成本更低,节约经费。 • 民用化标准产品:确保产品的可靠性和稳定性,为需要疫苗解毒的患者提供高质量的解毒剂。 4. 目标市场: • 医疗机构:用于疫苗解毒和治疗疟疾等疾病。 • 实验室和研究机构:用于科学研究和药物开发。 • 个人用户:对植物提取和化学制剂感兴趣的个人。 5. 推广计划: • 在医疗和科研领域展开推广活动,向潜在用户展示产品的效果和优势。 • 利用社交媒体和线上平台宣传产品,吸引个人用户关注和购买。 • 与贫困地区的政府和非营利组织合作,为有需要的人提供培训和支持,帮助他们制备青蒿素并解决医疗需求。 6. 商业模式: • 直接销售:通过网站、线上市场和实体店销售产品套件。 • 批量定制:与大型医疗机构和研究机构合作,根据其需求定制特殊规格的产品套件。 • 授权合作:与其他公司合作,授权其生产和销售产品套件,扩大产品的市场渠道和影响力。 7. 可持续发展: • 持续改进产品质量和生产工艺,确保产品符合最新的医疗标准和法规要求。 • 不断进行研发和创新,推出新型产品和技术,满足用户不断变化的需求。 通过以上产品计划,我们致力于提供一种可靠、经济实惠的黄花蒿提取青蒿素标准套件,为解决疫苗解毒等医疗问题做出贡献,同时为个人和组织提供方便、可靠的植物提取解决方案。 Product Plan: Artemisinin Extraction Standard Kit from Artemisia Annua 1. Product Overview: We propose to develop a standard kit for extracting artemisinin from Artemisia annua, aiming to provide a convenient and reliable method for individuals or organizations to extract artemisinin from Artemisia annua plants at home or in the laboratory and convert it into artemisinin derivatives. The product kit will provide all necessary materials and detailed operating instructions to ensure users can successfully extract and convert artemisinin. 2. Product Components: • Artemisinin extraction kit includes: • Dried Artemisia annua plant samples • Solvents for extraction (such as ether or acetone) • Acidic catalysts (such as sulfuric acid) • Alcohols for conversion • Chemical laboratory equipment (such as beakers, filter paper, glass rods, etc.) • Detailed operating manual 3. Product Advantages: • Easy to use: The product kit provides a detailed set of operating instructions, making it easy even for beginners to complete the extraction and conversion process. • Cost-effective: Compared to purchasing finished artemisinin or artemisinin derivatives, using this product kit incurs lower costs, saving funds. • Civilian standard product: Ensures the reliability and stability of the product, providing high-quality antidotes for patients requiring vaccine detoxification. 4. Target Market: • Medical institutions: Used for vaccine detoxification and treatment of diseases such as malaria. • Laboratories and research institutions: Used for scientific research and drug development. • Individual users: Individuals interested in plant extraction and chemical formulations. 5. Promotion Plan: • Conduct promotional activities in the medical and scientific research fields to demonstrate the effectiveness and advantages of the product to potential users. • Utilize social media and online platforms to promote the product, attracting attention and purchases from individual users. • Collaborate with governments and non-profit organizations in impoverished areas to provide training and support to those in need, helping them prepare artemisinin and address medical needs. 6. Business Model: • Direct sales: Sell product kits through websites, online markets, and physical stores. • Customized bulk orders: Collaborate with large medical institutions and research organizations to customize product kits according to their requirements. • Licensing partnerships: Partner with other companies to license the production and sale of product kits, expanding the market channels and influence of the product. 7. Sustainable Development: • Continuously improve product quality and production processes to ensure compliance with the latest medical standards and regulatory requirements. • Engage in ongoing research and innovation to introduce new products and technologies to meet the evolving needs of users. Through the above product plan, we are committed to providing a reliable and cost-effective standard kit for extracting artemisinin from Artemisia annua, making contributions to addressing medical issues such as vaccine detoxification, while providing convenient and reliable plant extraction solutions for individuals and organizations. Short link: https://gettr.ink/s75ctk
  14. 图片内容来源:明轩工作室 ,本站做了英文的参考翻译
  15. https://zh.wikipedia.org/zh-cn/青蒿素 https://en.wikipedia.org/wiki/Artemisinin 大家找不到这些信息的原因是因为。同样是维基百科:青蒿素 的这个词条。英文的内容和中文的内容完全不同。英文省略了精华的部分。而在中国的中国人更可怜。因为中国的网络防火墙和媒体控制,需要使用VPN才能打开维基百科。 1.2004年起中国大陆用户无法打开维基百科; 2.中国低于1%的人会使用VPN; 3.中国政府规定使用VPN属于违法行为。 4.中国人不会忘记是以美国思科公司为首的网络技术公司帮助中国政府建立了防火墙。 The reason why everyone can't find this information is that the Wikipedia entry for artemisinin has completely different content in English compared to Chinese. The English version omits the essence of the information. Chinese people are even more unfortunate because China's internet firewall and media control mean they need to use VPNs to access Wikipedia. 1. Since 2004, mainland Chinese users have been unable to access Wikipedia; 2. Less than 1% of people in China use VPNs; 3. The Chinese government considers the use of VPNs to be illegal. 4. Chinese people will not forget that it was leading network technology companies like Cisco Systems from the United States that helped the Chinese government establish the firewall.
  16. 青蒿植物鲜榨成汁,可能会是较好的选择。 原因:这样不会丢失青蒿素有效成分。 Extracting juice from fresh Artemisia plants may be a preferable option. This is because it helps retain the effective component artemisinin.
  17. 根据以上资料表明:服用青蒿茶时,建议温度需控制在 60度以下,温度过高则无效。 According to the information provided, it is recommended to control the temperature below 60 degrees Celsius when consuming Artemisia tea. Higher temperatures render it ineffective. 1680年前,中国的一本 草药书上记载的服用方法是 |The method of administration recorded in a Chinese herbal book before 1680 is: 青蒿一握,以水二升渍,绞取汁,尽服之。 Grasp a handful of Artemisia, soak it in two liters of water, squeeze out the juice, and consume it all.
  18. 在山东中医药研究所、云南省药物研究所的帮助下,研究人员发现药用青蒿中,学名黄花蒿(Artemisia annua)者有效,而学名青蒿(Artemisia apiacea)者无效;而且提取温度过高会破坏其中的活性成分。 With the assistance of the Shandong Institute of Traditional Chinese Medicine and the Yunnan Institute of Materia Medica, researchers found that Artemisia annua, commonly known as sweet wormwood, was effective, while Artemisia apiacea was not. Additionally, they discovered that high extraction temperatures could destroy the active ingredients within the medicinal Artemisia. 来源:https://zh.wikisource.org/wiki/肘後備急方/卷3 1971年下半年,屠呦呦领导的科研小组(包括钟裕蓉、郎林福等)从东晋葛洪《肘后备急方之治寒热诸疟方》中“青蒿一握,以水二升渍,绞取汁,尽服之”的记载中受到了启发,认为温度高有可能对青蒿有效成分造成破坏而影响疗效,便由用乙醇提取改为用沸点比乙醇低的乙醚提取。 In the latter half of 1971, a research team led by Tu Youyou (including Zhong Yurong, Lang Linfu, among others) drew inspiration from Ge Hong's "Emergency Prescriptions Kept Up One's Sleeve" from the Eastern Jin Dynasty, which described soaking Artemisia in two liters of water, squeezing out the juice, and consuming it all. They suspected that high temperatures might degrade the effective components of Artemisia and affect its therapeutic efficacy. Therefore, they switched from ethanol extraction to ether extraction, which has a lower boiling point than ethanol. 1971年10月4日研究组取得青蒿中性提取物对鼠疟、猴疟100%疟原虫抑制率的突破: On October 4, 1971, the research team achieved a breakthrough with neutral extracts of Artemisia, showing a 100% inhibition rate against Plasmodium in mice and monkeys: 用北京青蒿秋季采的成株叶制成水煎浸膏,95%乙醇浸膏,挥发油对鼠疟均无效。 Decoction and maceration in 95% ethanol, as well as volatile oil, were ineffective against malaria in mice. 乙醇冷浸,浓缩时温度控制在60度所得提取物,鼠疟效价提高,温度过高则仍无效。 Ethanol maceration at controlled temperatures below 60 degrees Celsius resulted in increased efficacy against malaria in mice, but higher temperatures remained ineffective. 乙醚回流或冷浸所得提取物,鼠疟效价显著增高稳定。(低沸点乙醚(34.8度),并将温度控制在60度以下) Ether reflux or cold maceration extracts showed significantly increased and stable efficacy against malaria in mice (using low-boiling-point ether at 34.8 degrees Celsius and maintaining temperatures below 60 degrees Celsius). Source: https://zh.wikipedia.org/zh-cn/青蒿素
  19. 本发明公开一种提取青蒿素的方法,包括采用60~70%的乙醇为溶剂浸提原料青蒿叶,还包括:1)沉淀:调节浸提液pH值到7.5~8.0,使大部分杂质沉淀,过滤并调节母液pH值6.5~7.0;2)减压浓缩:将母液浓缩至原体积的8~1 5%,冷却除杂;3)结晶:加等量70%乙醇加热至溶解,静置析出粗结晶;4)重结晶:用热石油醚溶解粗结晶,静置分离后,得到精品。本发明采用生石灰粉CaO作为沉淀剂,既沉淀了大部分杂质,又易于结晶的形成,同时降低了成本,并采用石油醚作为的重结晶溶剂,重结晶效果优于酒精,利用本发明得到青蒿素精品总收率大于0.4%。 青蒿素的提取方法 技术领域: 本发明涉及一种中药提取方法,特别涉及从菊科植物青蒿中提取青蒿素的方法。 背景技术: 青蒿素是我国科学家在1971年首次从菊科植物黄花蒿(Artemisiaannua Linn)提取出的具有新型结构的倍半萜内酯。纯青蒿素为无色针状晶体,熔点150~153℃,比旋光度[α]D 23+75°至+78°(无水乙醇),味苦,易溶于苯、氯仿、乙酸乙酯、丙酮和冰醋酸,能溶于乙醇、甲醇、乙醚和热石油醚。青蒿素由于具有过氧桥和缩醛结构,对酸碱不稳定,对强碱极不稳定,热至熔点以上即迅速分解。它具有十分优良的抗疟作用,包括那些对氯喹有耐药性的恶性疟原虫感染。青蒿素又是我国唯一按照西药标准研发的中药,是当前公认的治疗疟疾最有效、最安全的药物,被世界卫生组织称为“治疗疟疾的最大希望”。 目前青蒿素的提取方法如云南药物研究所的“容积汽油法”,此法简练,工艺流程短,操作方便,但收率不理想,小试仅为0.3%,工业生产≤0.2%,且大量消耗汽油,不甚安全;山东中药研究所的“丙酮—硅胶柱层析法”(专利申请号87101346A)和“低沸汽油—超短粗型球状扩孔硅胶过滤柱层析法”(专利申请号89103384),四川省中药研究所的“稀醇法”以及美国专利(专利号4952603)“己烷提取—己烷/乙晴分析—硅胶柱层析法”,这些方法各有利弊,但工业运用都有很大的难度;云南省药物研究所另一发明“青蒿素的提取方法”(专利申请号93102934.1)中又论述,使用稀乙醇提取青蒿素,汽油萃取,活性炭脱色,用乙醇重结晶,该法在以往的工作基础上有了很大的提高,但该专利只采用一次浸泡,青蒿素不能充分溶出;使用大量汽油提取,操作危险性较大;使用活性炭脱色,不可避免地吸收部分青蒿素,降低了收率,且活性炭价格高;用50%乙醇重结晶,溶解温度高,易破坏青蒿素,效果仍不理想。 发明内容: 本法明旨在克服以上不足而提供一种提取青蒿素的方法,该方法工艺简单、产品纯度高,具有操作易掌握、溶剂消耗少、成本低、收率高、安全、不需要大型及特殊设备等优点。 为实现本发明所采取的技术方案为: 一种提取青蒿素的方法,包括采用60~70%的乙醇为溶剂浸提原料青蒿叶,还包括: 1)沉淀:调节浸提液PH值到7.5~8.0,使大部分杂质沉淀,过滤并调节母液PH值6.5~7.0; 2)减压浓缩:将母液浓缩至原体积的8~15%,冷却除杂; 3)结晶:加等量70%乙醇加热至溶解,静置析出粗结晶; 4)重结晶:用热石油醚溶解粗结晶,静置分离后,得到精品。 为了提高产品收率,采用60~70%的乙醇为溶剂浸提时,浸提次数为3~5次。 在沉淀过程中,为了降低成本和最大限度的去除杂质,调节浸提液PH值的物料为生石灰。 在沉淀过程中,为了保持青蒿素的稳定,调节母液PH值的物料为乙酸。 在浓缩过程中,为保持青蒿素稳定性,采用浓缩温度50℃~60℃,浓缩压力0.047~0.053Mpa。 在结晶过程中,为提高产品的总收率,结晶母液用再3倍量的60~70%乙醇稀释,然后重复1)、2)、3)步骤,得到粗结晶。 在重结晶过程中,利用青蒿素在冷热石油醚中溶解度差异大特性,得到更多的产品,并避免青蒿素被高温破坏,故采用沸程30~60℃的热石油醚。 本发明的积极效果为: 1、本发明经过多次浸提,最终产品的总收率相对于青蒿叶计,可达0.48%以上。 2、本发明采用酒精作为浸提溶剂,而不用汽油,大大降低了操作的风险。 3、本发明采用生石灰粉(CaO)作为沉淀剂,用CaO粉调节PH值,既沉淀了大部分杂质,又易于结晶的形成,同时降低了成本。 4、本发明采用沸程30~60℃石油醚作为的重结晶溶剂,重结晶效果优于酒精。 5、本发明浸提所用溶剂在浓缩时进行了回收,整个生产过程是在密闭状态下进行,溶剂挥发极少;浸提完成后,用水蒸汽蒸馏的方法回收酒精,残渣用作蚊香原料,这样就降低了损耗和操作风险,提高了经济效益。 具体实施方式: 实施例一: 1、浸提:将100kg青蒿叶置于中药提取罐中,加入500kg70%乙醇,开始搅拌,以10~15rpm的速度搅拌10小时,停止搅拌,使物料的大小达到60~70目,打开提取罐下部的阀门,浸提液自动沥出,得到I浸液288kg,待用;关闭阀门,再向提取罐加入300kg70%乙醇,搅拌5小时,得到II浸液313kg,待用;最后加入300kg70%乙醇,搅拌3小时,形成III浸液316kg,III浸液用于下批套用。沥干后的残渣送至酒精回收罐中,加入纯化水300kg,采用水蒸汽蒸馏法将剩余乙醇回收再利用,得到酒精(折100%)138kg,残渣用作蚊香的原料。 2、沉淀:合并I、II浸提液601kg,用生石灰粉CaO调节PH值到7.8,使大部分杂质沉淀,并迅速过滤,得到滤液,滤液用乙酸调节溶液PH值到6.7。 3、减压浓缩:将滤液转入浓缩罐中,控制减压过程中,温度保持在50℃~60℃,压力保持在0.047~0.053MPa之间,2小时内浓缩至60kg。放置冷却,倾去上清液,除去水溶性杂质,得到浓缩液11kg。 4、结晶:将浓缩液转入结晶罐中,加11kg70%乙醇加热至60℃溶解,静置3~4天,析出并收集粗结晶I0.48kg。母液用63kg70%乙醇稀释,然后按照2、3、4步骤进行沉淀,滤液减压浓缩,浓缩液结晶,收集得到粗结晶II0.13kg。合并I、II两次粗结晶得到粗结晶III0.61kg。 5、重结晶:加入1.7kg沸程为30~60℃的热石油醚溶解粗结晶III,静置24小时,析出针状结晶体,经离心分离,50~60℃真空干燥,即得到青蒿素精品482g,测定其含量,含C15H22O5 99.3%。 实施例二: 1、浸提:将100kg青蒿叶置于中药提取罐中,加入500kg60%乙醇,开始搅拌,以10~15rpm的速度搅拌10小时,停止搅拌,使物料的大小达到60~70目,打开提取罐下部的阀门,浸提液自动沥出,得到I浸液285kg,待用;关闭阀门,再向提取罐加入300kg60%乙醇,搅拌5小时,得到II浸液314kg,待用;最后加入300kg60%乙醇,搅拌3小时,得到III浸液306kg,III浸液用于下批套用。沥干后的残渣送至酒精回收罐中,加入300kg纯化水,采用水蒸汽蒸馏法将剩余乙醇回收再利用,得到酒精(折100%)122kg,残渣用作蚊香的原料。 2、沉淀:合并I、II浸提液599kg,用生石灰粉CaO调节PH值到8.0,使大部分杂质沉淀,并迅速过滤,得到滤液,滤液用乙酸调节溶液PH值到6.9。 3、减压浓缩:将滤液转入浓缩罐中,控制减压过程中,温度保持在50℃~60℃,压力保持在0.047~0.053MPa之间,2小时内浓缩至48kg。放置冷却,倾去上清液,除去水溶性杂质,得到浓缩液10.3kg。 4、结晶:将浓缩液转入结晶罐中,加10.3kg70%乙醇加热至60℃溶解,静置3~4天,析出并收集粗结晶I0.41kg。母液用60kg70%乙醇稀释,然后按照2、3、4步骤进行沉淀,滤液减压浓缩,浓缩液结晶,收集得到粗结晶II0.10kg。合并I、II两次粗结晶得到粗结晶III0.51kg。 5、重结晶:加入1.4kg沸程为30~60℃的热石油醚溶解粗结晶III,静置24小时,析出针状结晶体,经离心分离,50~60℃真空干燥,即得到青蒿素精品489g,测定其含量,含C15H22O5 99.2%。 实施例三: 1、浸提:将100kg青蒿叶置于中药提取罐中,加入500kg60%乙醇,开始搅拌,以10~15rpm的速度搅拌10小时,停止搅拌,使物料的大小达到60~70目,打开提取罐下部的阀门,浸提液自动沥出,得到I浸液283kg,待用;关闭阀门,再向提取罐加入300kg60%乙醇,搅拌5小时,得到II浸液316kg,待用;最后加入300kg60%乙醇,搅拌3小时,得到III浸液305kg,III浸液用于下批套用。沥干后的残渣送至酒精回收罐中,加入300kg纯化水,采用水蒸汽蒸馏法将剩余乙醇回收再利用,得到酒精(折100%)123kg,残渣用作蚊香的原料。 2、沉淀:合并I、II浸提液599kg,用生石灰粉CaO调节PH值到7.8,使大部分杂质沉淀,并迅速过滤,得到滤液,滤液用乙酸调节溶液PH值到6.7。 3、减压浓缩:将滤液转入浓缩罐中,控制减压过程中,温度保持在50℃~60℃,压力保持在0.047~0.053MPa之间,2小时内浓缩至60kg。放置冷却,倾去上清液,除去水溶性杂质,得到浓缩液10kg。 4、结晶:将浓缩液转入结晶罐中,加10kg70%乙醇加热至60℃溶解,静置3~4天,析出并收集粗结晶I0.43kg。母液用58.5kg60%乙醇稀释,然后按照2、3、4步骤进行沉淀,滤液减压浓缩,浓缩液结晶,收集得到粗结晶II0.11kg。合并I、II两次粗结晶得到粗结晶III0.54kg。 5、重结晶:加入1.5kg沸程为30~60℃的热石油醚溶解粗结晶III,静置24小时,析出针状结晶体,经离心分离,50~60℃真空干燥,即得到青蒿素精品483g,测定其含量,含C15H22O5 99.9%。 实施例四: 1、浸提:将100kg青蒿叶置于中药提取罐中,加入500kg60%乙醇,开始搅拌,以10~15rpm的速度搅拌10小时,停止搅拌,使物料的大小达到60~70目,打开提取罐下部的阀门,浸提液自动沥出,得到I浸液284kg,待用;关闭阀门,再向提取罐加入300kg60%乙醇,搅拌5小时,得到II浸液317kg,待用;最后加入300kg60%乙醇,搅拌3小时,得到III浸液304kg,III浸液用于下批套用。沥干后的残渣送至酒精回收罐中,加入300kg纯化水,采用水蒸汽蒸馏法将剩余乙醇回收再利用,得到酒精(折100%)123kg,残渣用作蚊香的原料。 2、沉淀:合并I、II浸提液601kg,用生石灰粉CaO调节PH值到7.5,使大部分杂质沉淀,并迅速过滤,得到滤液,滤液用乙酸调节溶液PH值到6.9。 3、减压浓缩:将滤液转入浓缩罐中,控制减压过程中,温度保持在50℃~60℃,压力保持在0.047~0.053MPa之间,2小时内浓缩至58kg。放置冷却,倾去上清液,除去水溶性杂质,得到浓缩液9.6kg。 4、结晶:将浓缩液转入结晶罐中,加9.6kg70%乙醇加热至60℃溶解,静置3~4天,析出并收集粗结晶I0.41kg。母液用56kg60%乙醇稀释,然后按照2、3、4步骤进行沉淀,滤液减压浓缩,浓缩液结晶,收集得到粗结晶II0.12kg。合并I、II两次粗结晶得到粗结晶III0.53kg。 5、重结晶:加入1.4kg沸程为30~60℃的热石油醚溶解粗结晶III,静置24小时,析出针状结晶体,经离心分离,50~60℃真空干燥,即得到青蒿素精品485g,测定其含量,含C15H22O5 99.5%。 总结: 1、一种提取青蒿素的方法,包括采用60~70%的乙醇为溶剂浸提原料青蒿叶,其特征在于还包括: 1)沉淀:调节浸提液PH值到7.5~8.0,使大部分杂质沉淀,过滤并调节母液PH值6.5~7.0; 2)减压浓缩:将母液浓缩至原体积的8~15%,冷却除杂; 3)结晶:加等量70%乙醇加热至溶解,静置析出粗结晶; 4)重结晶:用热石油醚溶解粗结晶,静置分离后,得到精品。 2、如权利要求1所述的提取青蒿素的方法,其特征在于:所述的浸提次数为3~5次。 3、如权利要求1所述的提取青蒿素的方法,其特征在于:所述调节浸提液PH值的物料为生石灰。 4、如权利要求1或3所述的提取青蒿素的方法,其特征在于:所述调节母液PH值的物料为乙酸。 5、如权利要求1所述的提取青蒿素的方法,其特征在于:所述的浓缩温度为50℃~60℃,压力为0.047~0.053MPa。 6、如权利要求1所述的提取青蒿素的方法,其特征在于:所述的结晶母液用再3倍量的60~70%乙醇稀释,然后重复1)、2)、3)步骤,得到粗结晶。 7、如权利要求1所述的提取青蒿素的方法,其特征在于:所述的石油醚为沸程30~60℃的热石油醚。 https://patents.google.com/patent/CN1680388A/zh An extraction of qinghaosu is carried out by: 1) immersing extracting the leaf of sweet wormwood with 60-70% alcohol as solvent, depositing, regulating pH of immersing extracting liquid to 7.5-8.0, depositing the impurities, filtering, and regulating pH of mother liquid to 6.5-7.0; 2) de-pressuring and concentrating the mother liquid to 8-15% of original volume, cooling, and removing impurities; 3)crystallizing, adding into isosteric 70% alcohol, heating, dissolving, laying aside, precipitating out coarse crystal; 4)re-crystallizing, dissolving the coarse crystal by hot mineral ether, laying aside, separating, and obtaining the refined products. Its advantages include low cost, good re-crystallizing effect, and high productivity. The extracting method of Artemisinin Technical field: The present invention relates to a kind of traditional Chinese medicine extraction method, particularly from the feverfew sweet wormwood, extract the method for Artemisinin. Background technology: Artemisinin is the sesquiterpene lactones with novel texture that China scientist extracted from feverfew Herba Artemisiae annuae (Artemisiaannua Linn) first in 1971.Pure Artemisinin is a colourless acicular crystal, 150~153 ℃ of fusing points, specific rotatory power [α] D 23+ 75 ° to+78 ° (dehydrated alcohol), bitter is soluble in benzene, chloroform, ethyl acetate, acetone and Glacial acetic acid, can be dissolved in ethanol, methyl alcohol, ether and hot sherwood oil.Artemisinin is owing to have peroxide bridge and ethylidene ether structure, and extremely unstable to highly basic to the soda acid instability, heat is to promptly decomposing rapidly more than the fusing point.It has very good antimalarial effect, comprises that those have chemical sproof falciparum infection to chloroquine.Artemisinin is again the unique Chinese medicine according to the research and development of Western medicine standard of China, is current generally acknowledged the most effective, the safest medicine of treatment malaria, is called " maximum of treatment malaria is wished " by the World Health Organization. The extracting method of present Artemisinin such as " the volume gasoline method " of institute of materia medica, Yunnan, this method is terse, and technical process is short, and is easy to operate, but yield is undesirable, and lab scale only is 0.3%, and industrial production≤0.2%, and mass consumption gasoline are unsafe; " acetone-silica gel column chromatography " of institute of Chinese materia medica, Shandong (number of patent application 87101346A) and " the low spherical reaming filtered through silica gel of the gasoline-super short-thick type column chromatography that boils " (number of patent application 89103384), " rare pure method " of Traditional Chinese Medicine Research Institute, Sichuan Province and United States Patent (USP) (patent No. 4952603) " the fine analysis-silica gel column chromatography of hexane extraction-hexane/second ", these methods cut both ways, but industrial applicability all has very big difficulty; Discuss again in another invention " extracting method of Artemisinin " (number of patent application 93102934.1) of Yunnan Pharmaceutical Institute, use Diluted Alcohol to extract Artemisinin, gasoline extraction, activated carbon decolorizing, use ethyl alcohol recrystallization, this method is greatly improved on working foundation in the past, but this patent only adopts once immersion, fully stripping of Artemisinin; Use a large amount of gasoline extraction, operational danger is bigger; Use activated carbon decolorizing, the absorption portion Artemisinin has reduced yield inevitably, and gac price height; Use 50% ethyl alcohol recrystallization, the solvent temperature height, the destructible Artemisinin, effect is still undesirable. Summary of the invention: This law is bright to be intended to overcome above deficiency and a kind of method of extracting Artemisinin is provided, and this method technology is simple, product purity is high, have that operation is easily grasped, solvent consumption is few, cost is low, yield is high, safe, do not need large-scale and advantage such as specific installation. For realizing that the technical solution used in the present invention is: A kind of method of extracting Artemisinin comprises that the ethanol of employing 60~70% is solvent lixiviate raw material artemisia leaf, also comprises: 1) precipitation: regulate vat liquor pH value to 7.5~8.0, make most of contamination precipitation, filter and regulate mother liquor pH value 6.5~7.0; 2) concentrating under reduced pressure: mother liquor is concentrated into 8~15% of original volume, the cooling removal of impurities; 3) crystallization: add equivalent 70% ethanol and be heated to dissolving, leave standstill and separate out coarse crystallization; 4) recrystallization:, after the standing separation, obtain elaboration with hot petroleum ether dissolution coarse crystallization. In order to improve product yield, when the ethanol of employing 60~70% was the solvent lixiviate, the lixiviate number of times was 3~5 times. In precipitation process, in order to reduce cost and to remove impurity to greatest extent, the material of regulating the vat liquor pH value is a unslaked lime. In precipitation process, in order to keep the stable of Artemisinin, the material of regulating the mother liquor pH value is an acetate. In concentration process, for keeping Artemisinin stability, adopt 50 ℃~60 ℃ of thickening temperatures, concentrate pressure 0.047~0.053Mpa. In crystallisation process, be to improve the total recovery of product, crystalline mother solution repeats 1 then with 60~70% alcohol dilutions of 3 times of amounts again), 2), 3) step, obtain coarse crystallization. In recrystallization process, utilize Artemisinin big characteristic of dissolubility difference in cold and hot sherwood oil, obtain more products, and avoid Artemisinin to be destroyed, so adopt the hot sherwood oil of 30~60 ℃ of boiling ranges by high temperature. Positively effect of the present invention is: 1, the present invention is through extracted many times, and the total recovery of the finished product can reach more than 0.48% with respect to the artemisia leaf meter. 2, the present invention adopts alcohol as extraction solvent, and without gasoline, greatly reduces the risk of operation. 3, the present invention adopts calcium lime powder (CaO) as precipitation agent, regulates pH value with the CaO powder, has both precipitated most of impurity, is easy to crystalline again and forms, and has reduced cost simultaneously. 4, the present invention adopts the recrystallization solvent of 30~60 ℃ of sherwood oil conducts of boiling range, and the recrystallization effect is better than alcohol. 5, lixiviate solvent for use of the present invention reclaims when concentrated, and whole process of production is to carry out under air-tight state, and solvent evaporates is few; After lixiviate is finished, the steam distilled method recovered alcohol of water, residue has so just reduced loss and operational risk as the mosquito-repellent incense raw material, has improved economic benefit. Embodiment: Embodiment one: 1, lixiviate: the 100kg artemisia leaf is placed the traditional Chinese medicine extraction jar, add 500kg70% ethanol, begin to stir, speed with 10~15rpm stirred 10 hours, stopped to stir, and made the size of material reach 60~70 orders, open the valve of extractor bottom, the automatic drop of vat liquor goes out, and obtains I immersion liquid 288kg, and is stand-by; Valve-off adds 300kg70% ethanol to extractor again, stirs 5 hours, obtains II immersion liquid 313kg, and is stand-by; Add 300kg70% ethanol at last, stirred 3 hours, form III immersion liquid 316kg, the III immersion liquid is used for following batch and applies mechanically.Residue after draining is delivered in the alcohol withdrawing can, adds purified water 300kg, adopts steam distillation will remain the ethanol recycling, obtains alcohol (folding 100%) 138kg, and residue is as the raw material of mosquito-repellent incense. 2, precipitation: merging I, II vat liquor 601kg, regulate pH value to 7.8 with calcium lime powder CaO, make most of contamination precipitation, and filter rapidly, obtain filtrate, filtrate is used acetate regulator solution pH value to 6.7. 3, concentrating under reduced pressure: filtrate is changed in the concentration tank, and in the control decompression process, temperature remains on 50 ℃~60 ℃, and pressure remains between 0.047~0.053MPa, is concentrated into 60kg in 2 hours.Place cooling, the supernatant liquor that inclines is removed water-soluble impurity, obtains concentrated solution 11kg. 4, crystallization: concentrated solution is changed in the crystallizer, add 11kg70% ethanol and be heated to 60 ℃ of dissolvings, left standstill 3~4 days, separate out and collect coarse crystallization I0.48kg.Mother liquor 63kg70% alcohol dilution precipitates according to 2,3,4 steps then, and filtrate decompression concentrates, and the concentrated solution crystallization is collected and obtained coarse crystallization II0.13kg.Merge twice coarse crystallization of I, II and obtain coarse crystallization III0.61kg. 5, recrystallization: adding 1.7kg boiling range is 30~60 ℃ hot petroleum ether dissolution coarse crystallization III, leaves standstill 24 hours, separates out acicular crystals, and through centrifugation, 50~60 ℃ of vacuum-dryings promptly obtain Artemisinin elaboration 482g, measure its content, contain C 15H 22O 599.3%. Embodiment two: 1, lixiviate: the 100kg artemisia leaf is placed the traditional Chinese medicine extraction jar, add 500kg60% ethanol, begin to stir, speed with 10~15rpm stirred 10 hours, stopped to stir, and made the size of material reach 60~70 orders, open the valve of extractor bottom, the automatic drop of vat liquor goes out, and obtains I immersion liquid 285kg, and is stand-by; Valve-off adds 300kg60% ethanol to extractor again, stirs 5 hours, obtains II immersion liquid 314kg, and is stand-by; Add 300kg60% ethanol at last, stirred 3 hours, obtain III immersion liquid 306kg, the III immersion liquid is used for following batch and applies mechanically.Residue after draining is delivered in the alcohol withdrawing can, adds the 300kg purified water, adopts steam distillation will remain the ethanol recycling, obtains alcohol (folding 100%) 122kg, and residue is as the raw material of mosquito-repellent incense. 2, precipitation: merging I, II vat liquor 599kg, regulate pH value to 8.0 with calcium lime powder CaO, make most of contamination precipitation, and filter rapidly, obtain filtrate, filtrate is used acetate regulator solution pH value to 6.9. 3, concentrating under reduced pressure: filtrate is changed in the concentration tank, and in the control decompression process, temperature remains on 50 ℃~60 ℃, and pressure remains between 0.047~0.053MPa, is concentrated into 48kg in 2 hours.Place cooling, the supernatant liquor that inclines is removed water-soluble impurity, obtains concentrated solution 10.3kg. 4, crystallization: concentrated solution is changed in the crystallizer, add 10.3kg70% ethanol and be heated to 60 ℃ of dissolvings, left standstill 3~4 days, separate out and collect coarse crystallization I0.41kg.Mother liquor 60kg70% alcohol dilution precipitates according to 2,3,4 steps then, and filtrate decompression concentrates, and the concentrated solution crystallization is collected and obtained coarse crystallization II0.10kg.Merge twice coarse crystallization of I, II and obtain coarse crystallization III0.51kg. 5, recrystallization: adding 1.4kg boiling range is 30~60 ℃ hot petroleum ether dissolution coarse crystallization III, leaves standstill 24 hours, separates out acicular crystals, and through centrifugation, 50~60 ℃ of vacuum-dryings promptly obtain Artemisinin elaboration 489g, measure its content, contain C 15H 22O 599.2%. Embodiment three: 1, lixiviate: the 100kg artemisia leaf is placed the traditional Chinese medicine extraction jar, add 500kg60% ethanol, begin to stir, speed with 10~15rpm stirred 10 hours, stopped to stir, and made the size of material reach 60~70 orders, open the valve of extractor bottom, the automatic drop of vat liquor goes out, and obtains I immersion liquid 283kg, and is stand-by; Valve-off adds 300kg60% ethanol to extractor again, stirs 5 hours, obtains II immersion liquid 316kg, and is stand-by; Add 300kg60% ethanol at last, stirred 3 hours, obtain III immersion liquid 305kg, the III immersion liquid is used for following batch and applies mechanically.Residue after draining is delivered in the alcohol withdrawing can, adds the 300kg purified water, adopts steam distillation will remain the ethanol recycling, obtains alcohol (folding 100%) 123kg, and residue is as the raw material of mosquito-repellent incense. 2, precipitation: merging I, II vat liquor 599kg, regulate pH value to 7.8 with calcium lime powder CaO, make most of contamination precipitation, and filter rapidly, obtain filtrate, filtrate is used acetate regulator solution pH value to 6.7. 3, concentrating under reduced pressure: filtrate is changed in the concentration tank, and in the control decompression process, temperature remains on 50 ℃~60 ℃, and pressure remains between 0.047~0.053MPa, is concentrated into 60kg in 2 hours.Place cooling, the supernatant liquor that inclines is removed water-soluble impurity, obtains concentrated solution 10kg. 4, crystallization: concentrated solution is changed in the crystallizer, add 10kg70% ethanol and be heated to 60 ℃ of dissolvings, left standstill 3~4 days, separate out and collect coarse crystallization I0.43kg.Mother liquor 58.5kg60% alcohol dilution precipitates according to 2,3,4 steps then, and filtrate decompression concentrates, and the concentrated solution crystallization is collected and obtained coarse crystallization II0.11kg.Merge twice coarse crystallization of I, II and obtain coarse crystallization III0.54kg. 5, recrystallization: adding 1.5kg boiling range is 30~60 ℃ hot petroleum ether dissolution coarse crystallization III, leaves standstill 24 hours, separates out acicular crystals, and through centrifugation, 50~60 ℃ of vacuum-dryings promptly obtain Artemisinin elaboration 483g, measure its content, contain C 15H 22O 599.9%. Embodiment four: 1, lixiviate: the 100kg artemisia leaf is placed the traditional Chinese medicine extraction jar, add 500kg60% ethanol, begin to stir, speed with 10~15rpm stirred 10 hours, stopped to stir, and made the size of material reach 60~70 orders, open the valve of extractor bottom, the automatic drop of vat liquor goes out, and obtains I immersion liquid 284kg, and is stand-by; Valve-off adds 300kg60% ethanol to extractor again, stirs 5 hours, obtains II immersion liquid 317kg, and is stand-by; Add 300kg60% ethanol at last, stirred 3 hours, obtain III immersion liquid 304kg, the III immersion liquid is used for following batch and applies mechanically.Residue after draining is delivered in the alcohol withdrawing can, adds the 300kg purified water, adopts steam distillation will remain the ethanol recycling, obtains alcohol (folding 100%) 123kg, and residue is as the raw material of mosquito-repellent incense. 2, precipitation: merging I, II vat liquor 601kg, regulate pH value to 7.5 with calcium lime powder CaO, make most of contamination precipitation, and filter rapidly, obtain filtrate, filtrate is used acetate regulator solution pH value to 6.9. 3, concentrating under reduced pressure: filtrate is changed in the concentration tank, and in the control decompression process, temperature remains on 50 ℃~60 ℃, and pressure remains between 0.047~0.053MPa, is concentrated into 58kg in 2 hours.Place cooling, the supernatant liquor that inclines is removed water-soluble impurity, obtains concentrated solution 9.6kg. 4, crystallization: concentrated solution is changed in the crystallizer, add 9.6kg70% ethanol and be heated to 60 ℃ of dissolvings, left standstill 3~4 days, separate out and collect coarse crystallization I0.41kg.Mother liquor 56kg60% alcohol dilution precipitates according to 2,3,4 steps then, and filtrate decompression concentrates, and the concentrated solution crystallization is collected and obtained coarse crystallization II0.12kg.Merge twice coarse crystallization of I, II and obtain coarse crystallization III0.53kg. 5, recrystallization: adding 1.4kg boiling range is 30~60 ℃ hot petroleum ether dissolution coarse crystallization III, leaves standstill 24 hours, separates out acicular crystals, and through centrifugation, 50~60 ℃ of vacuum-dryings promptly obtain Artemisinin elaboration 485g, measure its content, contain C 15H 22O 599.5%. 1, a kind of method of extracting Artemisinin comprises that the ethanol of employing 60~70% is solvent lixiviate raw material artemisia leaf, it is characterized in that also comprising: 1) precipitation: regulate vat liquor pH value to 7.5~8.0, make most of contamination precipitation, filter and regulate mother liquor pH value 6.5~7.0; 2) concentrating under reduced pressure: mother liquor is concentrated into 8~15% of original volume, the cooling removal of impurities; 3) crystallization: add equivalent 70% ethanol and be heated to dissolving, leave standstill and separate out coarse crystallization; 4) recrystallization:, after the standing separation, obtain elaboration with hot petroleum ether dissolution coarse crystallization. 2, the method for extraction Artemisinin as claimed in claim 1 is characterized in that: described lixiviate number of times is 3~5 times. 3, the method for extraction Artemisinin as claimed in claim 1 is characterized in that: the material of described adjusting vat liquor pH value is a unslaked lime. 4, as the method for claim 1 or 3 described extraction Artemisinins, it is characterized in that: the material of described adjusting mother liquor pH value is an acetate. 5, the method for extraction Artemisinin as claimed in claim 1 is characterized in that: described thickening temperature is 50 ℃~60 ℃, and pressure is 0.047~0.053MPa. 6, the method for extraction Artemisinin as claimed in claim 1 is characterized in that: described crystalline mother solution repeats 1 then with 60~70% alcohol dilutions of 3 times of amounts again), 2), 3) step, obtain coarse crystallization. 7, the method for extraction Artemisinin as claimed in claim 1 is characterized in that: described sherwood oil is the hot sherwood oil of 30~60 ℃ of boiling ranges. https://patents.google.com/patent/CN1680388A/en Short link to this article: https://gettr.ink/QPeXR8
  20. 查看广告 第一位Hpay平面设计商家,在线接单啦! The first Hpay graphic design merchant is now online and accepting orders! 第一位Hpay平面设计商家,在线接单啦! 寻找一位经验丰富、创意无限的设计师来满足你的需求?不要再等待了!磐石农场的金牌设计师现已接单!曾在国内某大厂担任设计师,我们拥有丰富的设计经验和专业技能,为您提供最优质的设计服务。 无论是Logo设计、品牌标识还是平面设计,我们都能满足您的需求。我们以客户满意为首要目标,致力于为您打造独一无二、引人注目的设计作品。 接受HDO支付,让您的交易更加便捷。有任何设计需求?立即联系我们,让我们为您量身定制! 联系方式: 电话:+90 505 891 57 50 直接与设计师WhatsApp聊天 :https://wa.me/905058915750 The first Hpay graphic design merchant is now online and accepting orders! Are you seeking an experienced, endlessly creative designer to fulfill your needs? Look no further! The award-winning designers at Panshi Farm are now accepting orders! With previous experience at a prominent domestic firm, we bring a wealth of design expertise and professional skills to the table, ensuring top-quality service. Whether it's logo design, brand identity, or graphic design, we have you covered. Our primary goal is customer satisfaction, and we are dedicated to crafting unique and eye-catching designs tailored to your needs. We accept HDO payments for added convenience. Have any design requirements? Contact us now for a personalized experience! Contact Information: Phone: +90 505 891 57 50 WhatsApp chat directly with the designer: https://wa.me/905058915750 本广告短链接|Short link for this advertisement: https://gettr.ink/hxadbC 报价方: Roger 日期 2024年04月22日 价格 500.00 HDO 类别 虚拟产品 Hpay pay@paymap.org
  21. Time Left: 1 month and 18 hours

    • FOR SALE
    • NEW

    🎨✨ 第一位Hpay平面设计商家,在线接单啦!🎨✨ 寻找一位经验丰富、创意无限的设计师来满足你的需求?不要再等待了!磐石农场的金牌设计师现已接单!曾在国内某大厂担任设计师,我们拥有丰富的设计经验和专业技能,为您提供最优质的设计服务。 无论是Logo设计、品牌标识还是平面设计,我们都能满足您的需求。我们以客户满意为首要目标,致力于为您打造独一无二、引人注目的设计作品。 接受HDO支付,让您的交易更加便捷。有任何设计需求?立即联系我们,让我们为您量身定制!🎨✨ 联系方式: 电话:+90 505 891 57 50 直接与设计师WhatsApp聊天 :https://wa.me/905058915750 🎨✨The first Hpay graphic design merchant is now online and accepting orders! ✨🎨 Are you seeking an experienced, endlessly creative designer to fulfill your needs? Look no further! The award-winning designers at Panshi Farm are now accepting orders! With previous experience at a prominent domestic firm, we bring a wealth of design expertise and professional skills to the table, ensuring top-quality service. Whether it's logo design, brand identity, or graphic design, we have you covered. Our primary goal is customer satisfaction, and we are dedicated to crafting unique and eye-catching designs tailored to your needs. We accept HDO payments for added convenience. Have any design requirements? Contact us now for a personalized experience! 🎨✨ Contact Information: Phone: +90 505 891 57 50 WhatsApp chat directly with the designer: https://wa.me/905058915750 本广告短链接|Short link for this advertisement: https://gettr.ink/hxadbC

    500.00 HDO

    - TR

  22. A new Japanese study ties “statistically significant” increases in particular kinds of cancers in the country in the period 2020-2022 to the administration of mRNA vaccines. These cancers include ovarian cancer, leukemia, prostate cancer, pancreatic cancer and breast cancer. Importantly, the study demonstrates that neither COVID-19 itself nor “reduced cancer care” as a result of the lockdowns is likely to be responsible for these increases. The study’s abstract explains the motivation for the study, which begins with the observation that significant increases in excess cancer deaths were observed after the first year of the pandemic. “No significant excess mortality was observed during the first year of the pandemic (2020). However, some excess cancer mortalities were observed in 2021 after mass vaccination with the first and second vaccine doses, and significant excess mortalities were observed for all cancers and some specific types of cancer (including ovarian cancer, leukemia, prostate cancer, lip/oral/pharyngeal cancer, pancreatic cancer, and breast cancer) after mass vaccination with the third dose in 2022,” the abstract explains. “AMRs [age-adjusted mortality rates] for the four cancers with the most deaths (lung, colorectal, stomach, and liver) showed a decreasing trend until the first year of the pandemic in 2020, but the rate of decrease slowed in 2021 and 2022.” Professor Angus Dalgleish, a renowned cancer specialist in the UK, warned in 2022 that the vaccinations appeared to be “boosting” cancers and bringing patients out of remission, including his own patients. He wrote an open letter to the British Medical Journal at the end of that year, in which he argued that the mass-vaccination program needed to be brought to a halt and the serious negative effects of the mRNA vaccines needed to be considered. “The link with clots, myocarditis, heart attacks and strokes is now well accepted, as is the link with myelitis and neuropathy… “However, there is now another reason to halt all vaccine programmes. As a practising oncologist I am seeing people with stable disease rapidly progress after being forced to have a booster, usually so they can travel. “Even within my own personal contacts I am seeing B cell-based disease after the boosters. They describe being distinctly unwell a few days to weeks after the booster – one developing leukaemia, two work colleagues Non-Hodgkin’s lymphoma, and an old friend who has felt like he has had Long Covid since receiving his booster and who, after getting severe bone pain, has been diagnosed as having multiple metastases from a rare B cell disorder.” Last week Professor Dalgleish hailed the new Japanese study in a piece for a British publication, The Conservative Woman. “The results are astounding,” he wrote. “It shows there was a deficit for all cancers in the year 2020 when the first and second covid waves occurred. In 2021 there was an excess of deaths of 2.2 per cent and a 1.1 per cent increase in cancers. However, by 2022 the excess deaths had increased 9.6 per cent and cancer by 2.1 per cent. This paper was completed and published before the 2023 figures release which will almost certainly be much worse. What is remarkable here is that we are talking mortality, that is deaths from cancer not incidence of it.” Black America Is Awake: Alex Jones Interviews Dom Lucre 中文翻译: 一项新的日本研究将日本国内2020年至2022年期间特定类型的癌症的“统计学上显著”的增加与mRNA疫苗接种联系起来。这些癌症包括卵巢癌、白血病、前列腺癌、胰腺癌和乳腺癌。重要的是,该研究表明,这些增加不太可能是由于COVID-19本身或由于封锁而导致的“癌症护理减少”。 该研究的摘要解释了进行这项研究的动机,从观察到疫情第一年后癌症死亡率显著增加开始。 “在疫情的第一年(2020年)没有观察到明显的过度死亡。然而,在2021年,第一剂和第二剂疫苗大规模接种后,观察到了一些癌症过度死亡,以及在2022年第三剂疫苗大规模接种后,对所有癌症和一些特定类型的癌症(包括卵巢癌、白血病、前列腺癌、唇/口腔/咽癌、胰腺癌和乳腺癌)观察到了显著的过度死亡。”摘要解释道。 “对于死亡率最高的四种癌症(肺癌、结直肠癌、胃癌和肝癌),年龄调整死亡率(AMRs)在2020年疫情的第一年之前显示出下降趋势,但在2021年和2022年减速下降。” 英国著名的癌症专家安格斯·达尔格利什教授在2022年警告称,疫苗似乎在“促进”癌症,并使患者摆脱了缓解状态,包括他自己的患者。 他在当年年底写了一封公开信给《英国医学杂志》,在信中他主张需要停止大规模接种计划,并考虑mRNA疫苗的严重负面影响。 “现在,与血栓、心肌炎、心脏病发作和中风的联系已经被广泛接受,与髓炎和神经病的联系也是如此… “然而,现在有另一个理由来停止所有疫苗计划。作为一名实践肿瘤学家,我看到有稳定病情的人在被迫接种增强剂后迅速恶化,通常是为了能够旅行。 "甚至在我的个人接触中,我也发现在接受强化剂后出现了以 B 细胞为基础的疾病。据他们描述,在接受强化治疗后的几天到几周内,他们的身体明显不适--其中一人患上了白血病,两位同事患上了非霍奇金淋巴瘤,还有一位老朋友在接受强化治疗后感觉自己患上了Long Covid,在出现剧烈骨痛后,他被诊断出患有罕见B细胞疾病的多发性转移。" 上周,达尔格利什教授在为英国刊物《保守妇女》撰写的一篇文章中,对日本的这项新研究大加赞赏。 "他写道:"结果令人震惊。 他写道:"研究结果显示,在 2020 年,所有癌症的发病率都有所下降,当时正值第一波和第二波癌症流行期。2021 年,死亡人数增加了 2.2%,癌症增加了 1.1%。然而,到 2022 年,超额死亡人数增加了 9.6%,癌症增加了 2.1%。这篇论文是在 2023 年数据公布之前完成并发表的,而 2023 年的数据几乎肯定会更糟。值得注意的是,我们讨论的是死亡率,即癌症死亡人数,而不是癌症发病率。
  23. The artemether transdermal administration formulation of the present invention allows artemether to directly act on the surface of the affected skin after administration, and can penetrate through the epidermis into the dermis and subcutaneous tissues. Compared with existing administration routes of artemether (oral and injection), it significantly increases the amount of artemether at the affected skin site (target site), thereby enhancing efficacy and reducing dosage. Figure 1 depicts the transdermal permeation curves of various transdermal absorption enhancer combinations in the present invention. Figure 2 illustrates the transdermal permeation curves of different transdermal absorption carrier combinations in the present invention. Detailed embodiments Embodiment 1 Prescription: Artemether 10 g, 75% ethanol, q.s. to 100 mL. Preparation: Artemether is dissolved in 75% ethanol and made up to volume, yielding the artemether ointment for transdermal administration, where the ethanol solution serves as both a solvent and a disinfectant while also exerting a permeation-enhancing effect. Additional translation: Ingredient Amount Artemether 10 g 75% Ethanol q.s. to 100 mL In this table, "q.s." stands for the Latin phrase "quantum satis," which means "as much as needed" or "quantity sufficient." So, in this context, "75% Ethanol, q.s. to 100 mL" means adding 75% Ethanol until the total volume reaches 100 mL. 补充翻译: 成分 用量 蒿甲醚 10 克 75% 乙醇 加至100毫升 在这个表格中,"q.s."代表拉丁文短语"quantum satis",意思是"需要多少就加多少"或者"足量"。所以,在这个情况下,"75% 乙醇,加至100毫升"意味着加入75% 乙醇直到总体积达到100毫升。 Embodiment 2 Prescription: Artemether 20 g, Zinc Oxide 20 g, Camphor 2 g, Talcum Powder, q.s. to 100 g. Preparation: Artemether, Zinc Oxide, Camphor, and Talcum Powder (passed through a 100-mesh sieve) are thoroughly mixed using an equal incremental method, resulting in the artemether powder for transdermal administration. Additional translation: Ingredient Amount Artemether 20 g Zinc Oxide 20 g Camphor 2 g Talcum Powder q.s. to 100 g In this table, "q.s." stands for the Latin phrase "quantum satis," which means "as much as needed" or "quantity sufficient." So, in this context, "Talcum Powder, q.s. to 100 g" means adding Talcum Powder until the total weight reaches 100 g. 补充翻译: 成分 用量 蒿甲醚 20 克 氧化锌 20 克 冰片 2 克 滑石粉 加至100克 在这个表格中,“q.s.”代表拉丁文短语“quantum satis”,意思是“需要多少就加多少”或者“足量”。所以,在这个情况下,“滑石粉,加至100克”意味着添加滑石粉直到总量达到100克。
×
×
  • Create New...

Important Information

Registration Terms & Conditions