TWI705140B - Cold-adapted temperature sensitive strains of enterovirus 71 and processes of developing cold-adapted temperature sensitive virus strains - Google Patents

Cold-adapted temperature sensitive strains of enterovirus 71 and processes of developing cold-adapted temperature sensitive virus strains Download PDF

Info

Publication number
TWI705140B
TWI705140B TW103101883A TW103101883A TWI705140B TW I705140 B TWI705140 B TW I705140B TW 103101883 A TW103101883 A TW 103101883A TW 103101883 A TW103101883 A TW 103101883A TW I705140 B TWI705140 B TW I705140B
Authority
TW
Taiwan
Prior art keywords
enterovirus
virus
temperature
cold
individual
Prior art date
Application number
TW103101883A
Other languages
Chinese (zh)
Other versions
TW201522642A (en
Inventor
邱克彬
Original Assignee
淡馬錫生命科學研究院公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 淡馬錫生命科學研究院公司 filed Critical 淡馬錫生命科學研究院公司
Publication of TW201522642A publication Critical patent/TW201522642A/en
Application granted granted Critical
Publication of TWI705140B publication Critical patent/TWI705140B/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32311Enterovirus
    • C12N2770/32321Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32311Enterovirus
    • C12N2770/32334Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Mycology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to cold-adapted temperature sensitive Enterovirus 71 strains, particularly to the cold-adapted temperature sensitive Enterovirus 71 strains EV71:TLLβP20 and EV71:TLLαP20. The present invention also relates to processes of developing the cold-adapted temperature sensitive virus strains.

Description

適於冷之腸病毒71溫度敏感株及發展適於冷之溫度敏感病毒株之方法 A temperature-sensitive strain of Enterovirus 71 suitable for cold and a method for developing a temperature-sensitive virus strain suitable for cold 序列提交Sequence submission

本申請案與電子格式之序列表一起申請。序列表名稱為2577224PCTSequenceListing.txt,其產生於2013年1月11日且大小為21kb。電子格式之序列表中之資訊以全文引用之方式併入本文中。 This application is applied together with the sequence table in electronic format. The sequence listing name is 2577224PCTSequenceListing.txt, which was generated on January 11, 2013 and has a size of 21 kb. The information in the sequence table in electronic format is incorporated into this article by reference in its entirety.

本發明係關於適於冷之腸病毒71溫度敏感株,特定言之適於冷之腸病毒71溫度敏感株EV71:TLLβP20及EV71:TLLαP20。本發明亦關於發展適於冷之溫度敏感病毒株、特定言之RNA病毒株之方法。 The present invention relates to a temperature sensitive strain of Enterovirus 71 suitable for cold, in particular, a temperature sensitive strain of Enterovirus 71 suitable for cold EV71: TLLβP20 and EV71: TLLαP20. The invention also relates to methods for developing temperature-sensitive virus strains suitable for cold, specifically RNA virus strains.

本文中用以闡明本發明之背景或提供關於實踐之其他細節的出版物及其他材料係以引用的方式併入,且為方便起見係分別集合於參考文獻中。 Publications and other materials used herein to clarify the background of the present invention or provide other details about the practice are incorporated by reference, and are separately collected in references for convenience.

手足口病(HFMD)係由基本上屬於腸病毒物種A之一組人類腸病毒造成。在此等腸病毒中,腸病毒71(EV71)及柯薩奇病毒A16(CA16)是造成超過90%之HFMD病例的原因。除引起HFMD之外,已知EV71引起嚴重神經疾病以及死亡(尤其在幼兒中)。 Hand, foot and mouth disease (HFMD) is caused by human enteroviruses, which are basically a group of enterovirus species. Among these enteroviruses, enterovirus 71 (EV71) and Coxsackie virus A16 (CA16) are responsible for more than 90% of HFMD cases. In addition to causing HFMD, EV71 is known to cause severe neurological diseases and death (especially in young children).

人類腸病毒71(EV71)係尺寸為約30nm之小型無包膜病毒,其具有約7,450個核苷酸之單鏈陽性RNA基因組。該病毒分類為小核糖核 酸病毒科(Picornaviridae)內腸病毒屬下之人類腸病毒A物種(Alexander等人,1994;Melnick,1996)。基於EV71之主要衣殼蛋白(VP1)基因之系統發生分析將其分為三個主要基因組(指示為A、B及C),且基因組B及C進一步次分為亞基因組B1至B5及C1至C5(Bible等人,2008)。EV71已與包括手足口病(HFMD)、無菌性腦膜炎、腦炎及小兒麻痹症狀麻痹之一批臨床疾病相關(主要在嬰兒及幼兒中)(Alexander等人,1994;Melnick,1996)。該病毒係首先自美國加利福尼亞州一具有無菌性腦膜炎之兒童分離且隨後表徵為腸病毒屬之新穎血清型(Schmidt等人,1974)。在其初始分離之後的年份,在世界各地報告由於該病毒而爆發伴有併發症之HFMD(Blomberg等人,1974;Kennett等人,1974;Deibel等人,1975;Hagiwara等人,1978)。 Human Enterovirus 71 (EV71) is a small non-enveloped virus with a size of about 30 nm, which has a single-stranded positive RNA genome of about 7,450 nucleotides. The virus is classified as a species of human enterovirus A under the Picornaviridae endovirus genus (Alexander et al., 1994; Melnick, 1996). Based on the phylogenetic analysis of the major capsid protein (VP1) gene of EV71, it is divided into three major genomes (indicated as A, B, and C), and genomes B and C are further divided into subgenomes B1 to B5 and C1 to C5 (Bible et al., 2008). EV71 has been associated with a number of clinical diseases including hand, foot and mouth disease (HFMD), aseptic meningitis, encephalitis and polio symptom paralysis (mainly in infants and young children) (Alexander et al., 1994; Melnick, 1996). The virus was first isolated from a child with aseptic meningitis in California, USA and then characterized as a novel serotype of Enterovirus (Schmidt et al., 1974). In the years after its initial isolation, outbreaks of HFMD with complications due to the virus have been reported worldwide (Blomberg et al., 1974; Kennett et al., 1974; Deibel et al., 1975; Hagiwara et al., 1978).

近年來,在出現神經毒性EV71之傳染性及零星爆發之後,EV71感染已變為整個世界、特定言之亞太區域之主要公共健康負擔及問題。EV71之神經毒性首先在1975年於保加利亞之爆發期間獲得顯著全球關注,該爆發引起705例小兒麻痹症狀疾病,其中死亡44例(Chumakov等人,1979;Shindarov等人,1979)。類似性質之爆發在1978年出現於匈牙利且產生多例小兒麻痹症狀疾病及47例死亡(Nagy等人,1982)。隨後,已在紐約、香港、澳大利亞及費城(Philadelphia)報告若干與EV71相關之CNS疾病之較溫和傳染病(Chomnaitree等人,1958;Samuda等人,1987;Gilbert等人,1988;Hayward等人,1989)。在日本出現兩種EV71傳染病,大部分病例藉由HFMD及CNS疾病之低發病率表徵(Tagaya等人,1981;Ishimaru等人,1980;Hagiwara等人,1983)。在1997年,在馬來西亞出現歸因於高度神經毒性EV71之HFMD之大爆發且造成48例死亡。在1998年於臺灣出現更大爆發,具有超過100,000例HFMD、405例嚴重感染及78例死亡(由於急性腦幹腦脊髓炎以及神經性心臟衰竭及肺水腫)(Lum等人,1998a; Lum等人,1998b;Chang等人,1998;Yan等人,200;Liu等人,2000;Wang等人,2000)。2008年在中華人民共和國大陸總共記錄488,955個HFMD病例,其中死亡126例(Chinese Center,2008)。據報導,在2009年HFMD病例之數目增至1,155,525,其中死亡353例(Yang等人,2011)。在2010年,該國家經歷最大爆發,具有超過170萬個HFMD病例、27,000個具有嚴重神經併發症之患者及905例死亡。在所有三次爆發中,幾乎所有伴以神經併發症及死亡之嚴重病例係由於EV71(Zeng等人,2012)。 In recent years, after the emergence of neurotoxic EV71 infectivity and sporadic outbreaks, EV71 infection has become a major public health burden and problem in the entire world, specifically in the Asia-Pacific region. The neurotoxicity of EV71 first received significant global attention during the outbreak in Bulgaria in 1975, which caused 705 cases of polio-symptomatic diseases, including 44 deaths (Chumakov et al., 1979; Shindarov et al., 1979). An outbreak of similar nature occurred in Hungary in 1978 and resulted in multiple polio-symptomatic diseases and 47 deaths (Nagy et al., 1982). Subsequently, several milder infectious diseases related to EV71-related CNS diseases have been reported in New York, Hong Kong, Australia and Philadelphia (Chomnaitree et al., 1958; Samuda et al., 1987; Gilbert et al., 1988; Hayward et al., 1989). There are two EV71 infectious diseases in Japan, most of the cases are characterized by the low incidence of HFMD and CNS disease (Tagaya et al., 1981; Ishimaru et al., 1980; Hagiwara et al., 1983). In 1997, a major outbreak of HFMD due to highly neurotoxic EV71 occurred in Malaysia and caused 48 deaths. A larger outbreak occurred in Taiwan in 1998, with more than 100,000 cases of HFMD, 405 severe infections, and 78 deaths (due to acute brainstem encephalomyelitis, neurological heart failure and pulmonary edema) (Lum et al., 1998a; Lum et al., 1998b; Chang et al., 1998; Yan et al., 200; Liu et al., 2000; Wang et al., 2000). In 2008, a total of 488,955 cases of HFMD were recorded in the mainland of the People's Republic of China, including 126 deaths (Chinese Center, 2008). It is reported that the number of HFMD cases increased to 1,155,525 in 2009, with 353 deaths (Yang et al., 2011). In 2010, the country experienced the largest outbreak, with more than 1.7 million HFMD cases, 27,000 patients with severe neurological complications, and 905 deaths. In all three outbreaks, almost all severe cases with neurological complications and death were due to EV71 (Zeng et al., 2012).

當前,仍未充分理解EV71感染之病毒毒性及發病機制之分子決定因素。既不存在任何已經批准用於嚴重感染及相關神經併發症之臨床治療之抗病毒藥物,亦無任何可供用於控制及防止復發性爆發之疫苗。就控制及預防性策略而言,發展尤其用於開發中國家之成本有效的疫苗為第一要務和當務之急。在調查及發展中的針對EV71之各種類型之疫苗似乎引發嚙齒動物或猴中之免疫反應(Wu等人,2001;Arita等人,2005;Chiu等人,2006;Arita等人,2007;Tung等人,2007;Chung等人,2008;Chen等人,2008;Ong等人,2010;Chen等人,2011;Lee及Chang,2010;Xhang等人,2010。儘管基於VP1衣殼之病毒狀粒子疫苗及亞單位肽疫苗使得可行之潛在疫苗策略仍值得進一步研究及發展,但基於在控制且幾乎根除野生型脊髓灰質炎病毒感染中發展及使用不活化可注射Salk疫苗及活減毒口服Sabin脊髓灰質炎病毒疫苗的大量過往經驗,可注射不活化及口服減毒EV71疫苗仍為最有前景的候選物(Zhang等人,2010)。 Currently, the molecular determinants of viral toxicity and pathogenesis of EV71 infection are still not fully understood. There are neither antiviral drugs approved for clinical treatment of severe infections and related neurological complications, nor any vaccines available to control and prevent recurrent outbreaks. As far as control and preventive strategies are concerned, the development of cost-effective vaccines, especially for developing countries, is the first and most urgent task. Various types of vaccines against EV71 under investigation and development seem to trigger an immune response in rodents or monkeys (Wu et al., 2001; Arita et al., 2005; Chiu et al., 2006; Arita et al., 2007; Tung et al. Human, 2007; Chung et al., 2008; Chen et al., 2008; Ong et al., 2010; Chen et al., 2011; Lee and Chang, 2010; Xhang et al., 2010. Although virus-like particle vaccine based on VP1 capsid Potential vaccine strategies that make it feasible with subunit peptide vaccines are still worthy of further research and development, but based on the development and use of inactivated injectable Salk vaccine and live attenuated oral Sabin polio in the control and almost eradication of wild-type poliovirus infections A lot of past experience of inflammatory virus vaccine, injectable inactivated and oral attenuated EV71 vaccine is still the most promising candidate (Zhang et al., 2010).

當前,市場上無法取得保護兒童免受歸因於EV71之感染及手足口病的疫苗。基於近期資訊,中華人民共和國之兩個中心、新加坡之一個中心及臺灣之一個中心在發展可注射不活化EV71疫苗的過程中(新聞發佈及個人通信)。以H.Shimizu博士為首之National Institute of Infectious Diseases,Tokyo,Japan中之Enterovirus Unit自1980年代一直進行猴之EV71之發病機制及將EV71之溫度敏感株作為潛在候選口服EV71減毒活疫苗之深入研究(Arita等人,2005;Arita等人,2007)。儘管相比於靜脈內接種之後的野生型病毒引起CNS之神經侵入及組織病理學病變之程度較低,但其猴研究中之EV71之溫度敏感株之所有潛在疫苗候選物仍能夠引起該等病變。 Currently, there is no vaccine available in the market to protect children from infections attributed to EV71 and hand, foot and mouth disease. Based on recent information, two centers in the People's Republic of China, one center in Singapore and one center in Taiwan are in the process of developing injectable inactivated EV71 vaccine (press release and personal communication). National Institute of headed by Dr. H. Shimizu The Enterovirus Unit in Infectious Diseases, Tokyo, Japan has been conducting in-depth research on the pathogenesis of monkey EV71 and the temperature-sensitive strain of EV71 as a potential candidate for oral live attenuated EV71 vaccine since the 1980s (Arita et al., 2005; Arita et al. , 2007). Although compared with the wild-type virus after intravenous vaccination, the degree of neural invasion and histopathological changes in the CNS is lower, but all potential vaccine candidates of the temperature-sensitive strain of EV71 in the monkey study can still cause these changes .

活減毒疫苗代表疫苗接種之首先成功之方法中的一種,追溯至18世紀,當時英國醫生Edward Jenner開始使用牛痘病毒對兒童接種疫苗以抵抗破壞性疾病天花。活減毒疫苗使用已減弱之活病毒或微生物以使得其不能引起疾病,但誘發保護性免疫反應。傳統、經典及遺傳方法已在使用作活減毒疫苗之病毒及微生物減毒中在一定程度上成功。44-48傳統方法使用在人體中無毒的天然存在之相關生物體,諸如使用牛痘(cowpox或vaccinia)病毒。經典方法涉及在使毒性病毒或微生物減毒之條件下,諸如在組織培養物或嚴苛液體培養基中使其進行多輪生長。遺傳方法利用現代分子生物技術以操縱基因組以減少其毒性(Huygelen,1997;Robinson,2008;Coleman等人,2008;Lauring等人,2010;Kenney等人,2011)。在獲得溫度敏感表型病毒作為減毒之標記的經典方法中,藉由在較低溫度下培育之適合之細胞中培養病毒藉助於空斑分析技術對野生型病毒進行空斑選擇。選出之病毒株隨後在目標較低培育溫度下反覆地繼代(Hagiwara等人,1982;Hashimoto及Hagiwara,1983;Richman及Murphy,1997)。 Live attenuated vaccines represent one of the first successful methods of vaccination, dating back to the 18th century, when British doctor Edward Jenner began to use vaccinia virus to vaccinate children against the destructive disease smallpox. Live attenuated vaccines use weakened live viruses or microorganisms so that they cannot cause disease, but induce a protective immune response. Traditional, classical and genetic methods have been successfully used as live attenuated vaccines to attenuate viruses and microorganisms to a certain extent. 44-48 Traditional methods use related organisms that are non-toxic in the human body, such as cowpox (vaccinia) viruses. The classic method involves multiple rounds of growth under conditions that attenuate virulent viruses or microorganisms, such as in tissue culture or harsh liquid media. Genetic methods use modern molecular biotechnology to manipulate the genome to reduce its toxicity (Huygelen, 1997; Robinson, 2008; Coleman et al., 2008; Lauring et al., 2010; Kenney et al., 2011). In the classic method of obtaining a temperature-sensitive phenotype virus as a marker of attenuation, plaque selection of wild-type viruses is performed by culturing the virus in suitable cells cultivated at a lower temperature by means of plaque analysis technology. The selected virus strains are then repeatedly subcultured at the target lower cultivation temperature (Hagiwara et al., 1982; Hashimoto and Hagiwara, 1983; Richman and Murphy, 1997).

需要發展可用於治療病毒性疾病、在特定活體外細胞培養物條件下保持表型及遺傳穩定性且在靜脈內接種之後於猴中並不呈現神經毒性之病毒株。亦需要發展在細胞培養物中之連續繼代之後得到之適於冷之溫度敏感病毒(包括RNA病毒)株。 There is a need to develop virus strains that can be used to treat viral diseases, maintain phenotype and genetic stability under specific in vitro cell culture conditions, and do not exhibit neurotoxicity in monkeys after intravenous inoculation. There is also a need to develop strains of temperature-sensitive viruses (including RNA viruses) suitable for cold that are obtained after successive generations in cell culture.

本發明係關於適於冷之腸病毒71溫度敏感株,特定言之適於冷之腸病毒71溫度敏感株EV71:TLLβP20及EV71:TLLαP20。本發明亦關於發展適於冷之溫度敏感病毒株、特定言之RNA病毒株之方法。 The present invention relates to a temperature sensitive strain of Enterovirus 71 suitable for cold, in particular, a temperature sensitive strain of Enterovirus 71 suitable for cold EV71: TLLβP20 and EV71: TLLαP20. The invention also relates to methods for developing temperature-sensitive virus strains suitable for cold, specifically RNA virus strains.

因此,在一態樣中,本發明提供適於冷之腸病毒71溫度敏感株。在一實施例中,適於冷之腸病毒71溫度敏感株為如本文中所述之EV71:TLLβP20。在另一實施例中,適於冷之腸病毒71溫度敏感株為如本文中所述之EV71:TLLαP20。 Therefore, in one aspect, the present invention provides a temperature sensitive strain of Enterovirus 71 suitable for cold. In one embodiment, the temperature sensitive strain of Enterovirus 71 suitable for cold is EV71 as described herein: TLLβP20. In another embodiment, the temperature sensitive strain of Enterovirus 71 suitable for cold is EV71: TLLαP20 as described herein.

本發明之腸病毒71病毒株係藉由使用溫度敏感性作為表型標記以使腸病毒71減毒之方法製備。該方法為活體外實驗室製程,其改變病毒之生物生長特徵以適應於30℃以下之培育溫度下之最佳複製。以遞增地降低用於培養病毒之培育溫度之系統、逐步方式進行適應製程(其詳細描述於下文中)直至達成所選用於病毒之最佳複製之目標溫度。 The enterovirus 71 virus strain of the present invention is prepared by using temperature sensitivity as a phenotypic marker to attenuate enterovirus 71. This method is an in vitro laboratory process, which changes the biological growth characteristics of the virus to adapt to the optimal replication under the incubation temperature below 30°C. The adaptation process (which is described in detail below) is carried out in a systematic and stepwise manner that gradually reduces the incubation temperature for culturing the virus until the selected target temperature for optimal replication of the virus is reached.

在第二態樣中,本發明提供一種包含本文所述之適於冷之腸病毒71溫度敏感株之組合物。在一個實施例中,組合物包含有效量之本文所述之病毒株。在另一實施例中,組合物包含一或多種生理學上或醫藥學上可接受之載劑。在另一實施例中,組合物為疫苗。使用熟習此項技術者熟知之技術製備含有本文所述之適於冷之腸病毒71溫度敏感株之疫苗。該等疫苗適用於藉由使用熟習此項技術者熟知之技術向諸如人類個體之個體投與疫苗提供針對親本病毒株之免疫性。 In a second aspect, the present invention provides a composition comprising the temperature sensitive strain of Enterovirus 71 suitable for cold as described herein. In one embodiment, the composition contains an effective amount of the virus strain described herein. In another embodiment, the composition includes one or more physiologically or pharmaceutically acceptable carriers. In another embodiment, the composition is a vaccine. A vaccine containing the temperature sensitive strain of Enterovirus 71 suitable for cold as described herein is prepared using techniques well known to those skilled in the art. These vaccines are suitable for administering vaccines to individuals, such as human individuals, to provide immunity against parental virus strains by using techniques well known to those skilled in the art.

在第三態樣中,本發明提供一種引發諸如人類個體之個體中之保護性免疫反應之方法,其包含向個體投與預防上或治療學上或免疫學上有效量之本文所述之適於冷之腸病毒71溫度敏感株。在一個實施例中,保護性免疫反應保護個體免受由腸病毒71造成之疾病。在一個實施例中,該疾病為手足口病。在另一實施例中,該疾病為無菌性腦膜炎。在另一實施例中,該疾病為腦炎。在另一實施例中,該疾病為 小兒麻痹症狀麻痹。在一個實施例中,以疫苗形式投與本文所述之適於冷之腸病毒71溫度敏感株。在另一實施例中,個體已曝露至野生型腸病毒71。在另一實施例中,投與本文所述之適於冷之腸病毒71溫度敏感株防止諸如人類個體之個體罹患與腸病毒71相關之疾病。在另一實施例中,個體已曝露至野生型腸病毒71。在另一實施例中,投與本文所述之適於冷之腸病毒71溫度敏感株延遲諸如人類個體之感染病毒之個體中與腸病毒71相關之疾病之發作或減慢其進展速率。 In a third aspect, the present invention provides a method for eliciting a protective immune response in an individual such as a human individual, which comprises administering to the individual a prophylactic or therapeutically or immunologically effective amount of the appropriate amount described herein. Enterovirus 71 is a temperature-sensitive strain of cold. In one embodiment, the protective immune response protects the individual from diseases caused by Enterovirus 71. In one embodiment, the disease is hand, foot and mouth disease. In another embodiment, the disease is aseptic meningitis. In another embodiment, the disease is encephalitis. In another embodiment, the disease is Symptoms of polio paralysis. In one example, the temperature-sensitive strain of Enterovirus 71 described herein suitable for cold is administered as a vaccine. In another example, the individual has been exposed to wild-type enterovirus 71. In another embodiment, the administration of the enterovirus 71 temperature-sensitive strain suitable for cold described herein prevents individuals, such as human individuals, from suffering from enterovirus 71-related diseases. In another example, the individual has been exposed to wild-type enterovirus 71. In another embodiment, the administration of the enterovirus 71 temperature-sensitive strain suitable for cold as described herein delays the onset or slows the progression rate of enterovirus 71-related diseases in a virus-infected individual such as a human individual.

在第四態樣中,本發明提供一種使用溫度敏感性作為表型標記以使病毒減毒之方法。根據此態樣,該方法發展適於冷之溫度敏感病毒株。本發明之方法為活體外實驗室製程,其改變病毒之生物生長特徵以適應於30℃以下之培育溫度下之最佳複製。以遞增地降低用於培養病毒之培育溫度之系統、逐步方式進行適應製程直至達成所選用於病毒之最佳複製之目標溫度。因此,根據本發明,該方法包含以下步驟:(i)製備親本野生型病毒之參考儲備料,(ii)使用就每次繼代獲得完全細胞病變效應(CPE)而言之較低感染倍率(MOI)及較短培育時段之接種體在較高MOI及約34℃至約36℃、較佳約34℃之培育溫度下培育經親本野生型病毒之參考儲備料感染之細胞培養物五次或五次以上的繼代直至在每次繼代獲得完全CPE,(iii)使用就每次繼代獲得完全細胞病變效應(CPE)而言之較低感染倍率(MOI)及較短培育時段之接種體在較高MOI及比前述步驟中低約1℃至約3℃之培育溫度下培育經前述步驟之所得病毒感染之細胞培養物五次或五次以上的繼代直至在每次繼代獲得完全CPE,及(iv)以遞增地降低培育溫度之系統、逐步方式重複步驟(iii)直至達成所選用於病毒之最佳複製之目標溫度。在一個實施例中,目標溫度為約26℃至約29℃,較佳約28℃。在一個實施例中,遞增地降低培育溫度為降低約1℃至約2℃之溫度。 In the fourth aspect, the present invention provides a method of using temperature sensitivity as a phenotypic marker to attenuate viruses. According to this aspect, the method was developed for cold temperature-sensitive virus strains. The method of the present invention is an in vitro laboratory process, which changes the biological growth characteristics of the virus to adapt to the optimal replication at an incubation temperature below 30°C. The adaptation process is carried out in a stepwise manner with a system that gradually reduces the incubation temperature used for culturing the virus until the selected target temperature for the best replication of the virus is reached. Therefore, according to the present invention, the method includes the following steps: (i) preparing a reference stock of the parental wild-type virus, (ii) using a lower infection rate in terms of obtaining a complete cytopathic effect (CPE) for each successive generation (MOI) and the inoculum with a shorter incubation period are cultivated at a higher MOI and an incubation temperature of about 34°C to about 36°C, preferably about 34°C. Cell cultures infected with the reference stock of the parental wild-type virus One or more subcultures until complete CPE is obtained in each subculture, (iii) Use lower infection rate (MOI) and shorter incubation period in terms of obtaining complete cytopathic effect (CPE) for each subculture The inoculum is cultured at a higher MOI and at an incubation temperature of about 1°C to about 3°C lower than the previous step. The virus-infected cell culture obtained by the previous step is subcultured five or more times until each incubation Generation of complete CPE is obtained, and (iv) Step (iii) is repeated in a stepwise manner with a system of incrementally lowering the incubation temperature until the selected target temperature for optimal replication of the virus is reached. In one embodiment, the target temperature is about 26°C to about 29°C, preferably about 28°C. In one embodiment, reducing the incubation temperature incrementally is a temperature reduction of about 1°C to about 2°C.

在一實施例中,藉由在約36℃至約38℃、較佳約37℃之溫度下培 育經野生型病毒感染之細胞培養物一次或兩次繼代直至獲得完全細胞病變效應(CPE)製備親本野生型病毒之參考儲備料。將含有生成之病毒之培養物上澄液之等分試樣置放於小瓶或其他適合之儲存裝置中。此培養物上澄液充當親本野生型病毒之參考儲備料。參考親本野生型病毒係用於後續減毒製程。在另一實施例中,將親本野生型病毒之參考儲備料之等分試樣儲存於適合之溫度,諸如-80℃下。 In one embodiment, by culturing at a temperature of about 36°C to about 38°C, preferably about 37°C Breed cell cultures infected with wild-type virus once or twice until the complete cytopathic effect (CPE) is obtained to prepare the reference stock of the parental wild-type virus. Place an aliquot of the culture supernatant containing the generated virus in a vial or other suitable storage device. The supernatant of this culture serves as a reference stock for the parental wild-type virus. The reference parent wild-type virus line is used for the subsequent attenuation process. In another embodiment, an aliquot of the reference stock of the parental wild-type virus is stored at a suitable temperature, such as -80°C.

在一個實施例中,病毒為任何病毒。在另一實施例中,病毒為RNA病毒。在另一實施例中,RNA病毒為正鏈RNA病毒。在另一實施例中,病毒為小核糖核酸病毒科之成員。在另一實施例中,病毒為腸病毒屬之成員。在一實施例中,病毒為腸病毒71(EV71)。在另一實施例中,病毒為柯薩奇病毒A16(CA16)。本發明之方法適用於產生此等病毒中之任一者之適於冷之溫度敏感病毒株,包括(但不限於)EV71及CA16之適於冷之溫度敏感株。 In one embodiment, the virus is any virus. In another embodiment, the virus is an RNA virus. In another embodiment, the RNA virus is a positive-strand RNA virus. In another embodiment, the virus is a member of the Picornaviridae family. In another embodiment, the virus is a member of the genus Enterovirus. In one embodiment, the virus is Enterovirus 71 (EV71). In another embodiment, the virus is Coxsackie virus A16 (CA16). The method of the present invention is suitable for producing cold-suitable temperature-sensitive virus strains of any of these viruses, including but not limited to the cold-suitable temperature-sensitive strains of EV71 and CA16.

在一個實施例中,待藉由病毒感染之細胞為允許病毒生長之任何細胞。在一較佳實施例中,細胞為維羅(Vero)細胞(ATCC CCL-81)。在一實施例中,藉由適合於細胞生長之培養基中之常規繼代維持細胞。在細胞為維羅細胞之實施例中,在補充有10%胎牛血清(FCS)之達爾伯克改質伊格爾培養基(Dulbecco's modified Eagles's medium)(DMEM)中培養維羅細胞。在一個實施例中,維持於補充有1% FCS之DMEM中之維羅細胞係用於產生親本野生型病毒、病毒培養、減毒、滴定及評估溫度敏感表型。在另一實施例中,DMEM補充有1% FCS以使病毒株適應於在依次降低之培育溫度中複製。 In one embodiment, the cell to be infected by the virus is any cell that allows the virus to grow. In a preferred embodiment, the cell is a Vero cell (ATCC CCL-81). In one embodiment, the cells are maintained by routine subculture in a medium suitable for cell growth. In the example where the cells are Vero cells, the Vero cells are cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS). In one embodiment, the Vero cell line maintained in DMEM supplemented with 1% FCS is used to generate parental wild-type virus, virus culture, attenuate, titrate, and evaluate temperature-sensitive phenotypes. In another embodiment, DMEM is supplemented with 1% FCS to adapt the virus strain to replicate in successively lowering incubation temperatures.

本發明亦關於藉由本文所述之方法生成之適於冷之溫度病毒株。藉由本文所述之方法生成之適於冷之溫度病毒適用於使用熟習此項技術者熟知之技術產生疫苗。該等疫苗適用於藉由使用熟習此項技術者熟知之技術向個體投與疫苗提供針對親本病毒株之免疫性。 The present invention also relates to cold temperature virus strains produced by the methods described herein. The cold temperature virus produced by the method described herein is suitable for vaccine production using techniques well known to those skilled in the art. These vaccines are suitable for administering vaccines to individuals to provide immunity against parental virus strains by using techniques well known to those skilled in the art.

圖1a顯示周邊血液單核細胞(箭頭),其來源於給予靜脈內劑量之腸病毒71(EV71:TLLβP20)之後的第4日之猴血液,使用對病毒具有特異性之商用單株抗體藉由間接免疫螢光分析染色呈陽性。 Figure 1a shows peripheral blood mononuclear cells (arrows) derived from monkey blood on the 4th day after intravenous dose of Enterovirus 71 (EV71: TLLβP20), using commercial monoclonal antibodies specific to the virus. Indirect immunofluorescence analysis was positive.

圖1b顯示GelRed試劑染色之電泳瓊脂糖凝膠之相片,其顯示使用對偵測腸病毒71具有特異性之寡核苷酸引子對之來源於免疫後第4日之猴(2202F、2891F)之組織的一步RT-PCR擴增產物。RT-PCR擴增產物之預期尺寸為427bp。兩凝膠中之通道為如下。凝膠1:通道1:100bp DNA梯;通道2:2202F-心臟;通道3:2202F-脾;通道4:2202F-淋巴結;通道5:2202F-腎;通道6:2202F-肝;通道7:2891F-心臟;通道8:2891F-脾;通道9:2891F-淋巴結;通道10:2891F-腎;通道11:2891F-肝;通道12:2202F-腦幹(腦橋);通道13:2202F-腦幹(延髓);通道14:2202F-皮質(腦回);通道15:2202F-脊髓(頸椎);通道16:2202F-脊髓(腰椎);通道17:2202F-脊髓(胸);通道18:2891F-腦幹(延髓);通道19:2891F-腦幹(腦橋);通道20:2891F-皮質(左小腦)。凝膠2:通道21:100bp DNA梯;通道22:2891F-皮質(右小腦);通道23:2891F-脊髓(頸椎);通道24:2891F-脊髓(腰椎);通道25:2891F-脊髓(胸);通道26:無模板對照;通道27:100bp DNA梯。 Figure 1b shows a photograph of an electrophoresis agarose gel stained with GelRed reagent, which shows the use of oligonucleotide primers specific for the detection of enterovirus 71 from monkeys (2202F, 2891F) on day 4 after immunization One-step RT-PCR amplification product of the tissue. The expected size of the RT-PCR amplified product is 427bp. The channels in the two gels are as follows. Gel 1 : Channel 1: 100bp DNA ladder; Channel 2: 2202F-heart; Channel 3: 2202F-spleen; Channel 4: 2202F-lymph nodes; Channel 5: 2202F-kidney; Channel 6: 2202F-liver; Channel 7: 2891F -Heart; Channel 8: 2891F-Spleen; Channel 9: 2891F-Lymph nodes; Channel 10: 2891F-Kidney; Channel 11: 2891F-Liver; Channel 12: 2202F-Brainstem (pons); Channel 13: 2202F-Brainstem ( Medulla oblongata); passage 14: 2202F-cortex (cerebral gyrus); passage 15: 2202F- spinal cord (cervical spine); passage 16: 2202F- spinal cord (lumbar spine); passage 17: 2202F- spinal cord (thoracic); passage 18: 2891F-brain Stem (medulla oblongata); passage 19: 2891F-brain stem (pons); passage 20: 2891F-cortex (left cerebellum). Gel 2 : Passage 21: 100bp DNA ladder; Passage 22: 2891F-cortex (right cerebellum); Passage 23: 2891F-spine (cervical spine); Passage 24: 2891F-spine (lumbar spine); Passage 25: 2891F-spine (thoracic) ); Lane 26: No template control; Lane 27: 100bp DNA ladder.

本發明係關於適於冷之腸病毒71溫度敏感株,特定言之適於冷之腸病毒71溫度敏感株EV71:TLLβP20及EV71:TLLαP20。本發明亦關於發展適於冷之溫度敏感病毒株、特定言之RNA病毒株之方法。 The present invention relates to a temperature sensitive strain of Enterovirus 71 suitable for cold, in particular, a temperature sensitive strain of Enterovirus 71 suitable for cold EV71: TLLβP20 and EV71: TLLαP20. The invention also relates to methods for developing temperature-sensitive virus strains suitable for cold, specifically RNA virus strains.

因此,在一個態樣中,本發明提供適於冷之溫度敏感。在一個實施例中,適於冷之腸病毒71溫度敏感株為如本文中所述之EV71:TLLβP20。在另一實施例中,適於冷之腸病毒71溫度敏感株為 如本文中所述之EV71:TLLαP20。EV71:TLLβP20根據布達佩斯條約(Budapest Treaty)條款在2012年10月25日寄存於位於10801 University Boulevard,Manassas,Virginia 20110,USA之美國典型菌種保存中心(American Type Culture Collection)且指定寄存編號為PTA-13285。EV71:TLLαP20根據布達佩斯條約條款在2012年10月25日寄存於美國典型菌種保存中心,且指定寄存編號為PTA-13284。 Therefore, in one aspect, the present invention provides temperature sensitivity suitable for cold. In one embodiment, the temperature sensitive strain of Enterovirus 71 suitable for cold is EV71 as described herein: TLLβP20. In another embodiment, the temperature sensitive strain of Enterovirus 71 suitable for cold is EV71 as described in this article: TLLαP20. EV71: TLLβP20 was deposited in the American Type Culture Collection at 10801 University Boulevard, Manassas, Virginia 20110, USA on October 25, 2012 in accordance with the provisions of the Budapest Treaty and the designated deposit number is PTA -13285. EV71: TLLαP20 was deposited in the American Type Culture Collection on October 25, 2012 in accordance with the provisions of the Budapest Treaty, and the designated deposit number is PTA-13284.

本發明之腸病毒71病毒株係藉由使用溫度敏感性作為表型標記以使腸病毒71減毒之方法製備。該方法為活體外實驗室製程,其改變病毒之生物生長特徵以適應於30℃以下之培育溫度下之最佳複製。以遞增地降低用於培養病毒之培育溫度之系統、逐步方式進行適應製程直至達成所選用於病毒之最佳複製之目標溫度。如本文所揭示,(i)製備親本野生型病毒之參考儲備料,(ii)使用就每次繼代獲得完全細胞病變效應(CPE)而言之較低感染倍率(MOI)及較短培育時段之接種體在較高MOI及約34℃之培育溫度下培育經親本野生型病毒之參考儲備料感染之細胞培養物五次或五次以上的繼代直至在每次繼代獲得完全CPE,(iii)使用就每次繼代獲得完全細胞病變效應(CPE)而言之較低感染倍率(MOI)及較短培育時段之接種體在較高MOI及比前述步驟中低約1℃至約3℃之培育溫度下培育經前述步驟之所得病毒感染之細胞培養物五次或五次以上的繼代直至在每次繼代獲得完全CPE,及(iv)以遞增地降低培育溫度之系統、逐步方式重複步驟(iii)直至達成所選用於病毒之最佳複製之目標溫度。在一個實施例中,目標溫度為約26℃至約29℃,較佳約28℃。 The enterovirus 71 virus strain of the present invention is prepared by using temperature sensitivity as a phenotypic marker to attenuate enterovirus 71. This method is an in vitro laboratory process, which changes the biological growth characteristics of the virus to adapt to the optimal replication under the incubation temperature below 30°C. The adaptation process is carried out in a stepwise manner with a system that gradually reduces the incubation temperature used for culturing the virus until the selected target temperature for the best replication of the virus is reached. As disclosed herein, (i) prepare a reference stock of the parental wild-type virus, (ii) use a lower infection rate (MOI) and shorter incubation in terms of obtaining a complete cytopathic effect (CPE) for each passage The inoculum of the time period is cultivated at a higher MOI and an incubation temperature of about 34°C. The cell culture infected with the reference stock of the parental wild-type virus is subcultured five or more times until a complete CPE is obtained in each subculture. , (Iii) Use the inoculum with a lower infection rate (MOI) and a shorter incubation period in terms of obtaining a complete cytopathic effect (CPE) for each successive generation at a higher MOI and about 1°C lower than the previous step Cultivate the virus-infected cell culture obtained by the aforementioned steps for five or more subcultures at an incubation temperature of about 3°C until a complete CPE is obtained at each subculture, and (iv) a system that reduces the incubation temperature incrementally , Repeat step (iii) step by step until reaching the selected target temperature for optimal virus replication. In one embodiment, the target temperature is about 26°C to about 29°C, preferably about 28°C.

在一個實施例中,藉由在約36℃至約38℃、較佳約37℃之溫度下培育經野生型病毒感染之細胞培養物一次或兩次繼代直至獲得完全細胞病變效應(CPE)製備親本野生型病毒之參考儲備料。將含有生成之病毒之培養物上澄液之等分試樣置放於小瓶或其他適合之儲存裝置 中。此培養物上澄液充當親本野生型病毒之參考儲備料。參考親本野生型病毒係用於後續減毒製程。在另一實施例中,將親本野生型病毒之參考儲備料之等分試樣儲存於適合之溫度,諸如-80℃下。 In one embodiment, the cell culture infected with the wild-type virus is subcultured once or twice at a temperature of about 36°C to about 38°C, preferably about 37°C, until a complete cytopathic effect (CPE) is obtained. Prepare the reference stock of the parental wild-type virus. Place an aliquot of the culture supernatant containing the generated virus in a vial or other suitable storage device in. The supernatant of this culture serves as a reference stock for the parental wild-type virus. The reference parent wild-type virus line is used for the subsequent attenuation process. In another embodiment, an aliquot of the reference stock of the parental wild-type virus is stored at a suitable temperature, such as -80°C.

在一個實施例中,待藉由病毒感染之細胞為允許病毒生長之任何細胞。在一較佳實施例中,細胞為維羅細胞(ATCC CCL-81)。在一個實施例中,藉由適合於細胞生長之培養基中之常規繼代維持細胞。在細胞為維羅細胞之實施例中,在補充有10%胎牛血清(FCS)之達爾伯克改質伊格爾培養基(Dulbecco's modified Eagles's medium)(DMEM)中培養維羅細胞。在一個實施例中,將維持於補充有1% FCS之DMEM中之維羅細胞用於產生親本野生型病毒、病毒培養、減毒、滴定及評估溫度敏感表型。在另一實施例中,DMEM補充有1% FCS以使病毒株適應於在連續降低之培育溫度中複製。 In one embodiment, the cell to be infected by the virus is any cell that allows the virus to grow. In a preferred embodiment, the cell is a Vero cell (ATCC CCL-81). In one embodiment, the cells are maintained by routine subculture in a medium suitable for cell growth. In the example where the cells are Vero cells, the Vero cells are cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS). In one embodiment, Vero cells maintained in DMEM supplemented with 1% FCS are used to produce parental wild-type virus, virus culture, attenuation, titration, and assessment of temperature-sensitive phenotypes. In another embodiment, DMEM is supplemented with 1% FCS to adapt the virus strain to replicate in continuously decreasing incubation temperatures.

在一個實施例中,一旦病毒獲得完全細胞病變效應(CPE)便藉由獲得含有病毒之澄清培養物上澄液且傳遞此上澄液至細胞(例如維羅細胞)之各連續新鮮的新培養瓶中而在製程之各步驟中使病毒繼代。在另一實施例中,在各連續改變至較低培育溫度開始時以20之較高感染倍率(MOI)傳遞含有病毒之培養物上澄液。在注意到病毒能夠在接種後3日內於維羅細胞中引起完全CPE之後,再藉由相同MOI之病毒接種含有新鮮匯合之單層維羅細胞之新培養瓶至少三次繼代,隨後減少至5至10之較低MOI。一旦注意到病毒能夠在以5至10之較低MOI接種後3日內引起完全CPE,在5至10之MOI下再繼代至少三次之後,減毒製程隨後前進至連續較低培育溫度之下一階段。每次繼代所需之天數取決於病毒在遞增地降低培育溫度之各階段適應的速度。熟習此項技術者將容易地知道何時達到完全CPE。製備本發明之適於冷之腸病毒71溫度敏感株之方法的其他細節係描述於下文中。 In one embodiment, once the virus has achieved a complete cytopathic effect (CPE), the clear culture supernatant containing the virus is obtained and the supernatant is transferred to each continuous fresh new culture of cells (such as Vero cells) The virus is subcultured in the bottle and in each step of the process. In another embodiment, the virus-containing culture supernatant is delivered at a higher infection rate (MOI) of 20 at the beginning of each successive change to a lower incubation temperature. After noticing that the virus can cause complete CPE in Vero cells within 3 days after inoculation, inoculate a new culture flask containing freshly confluent single-layer Vero cells with virus of the same MOI for at least three subcultures, and then reduce to 5 To the lower MOI of 10. Once it is noted that the virus can cause complete CPE within 3 days after inoculation with a lower MOI of 5 to 10, and after at least three subcultures at an MOI of 5 to 10, the attenuation process then proceeds to a continuous lower incubation temperature. stage. The number of days required for each successive generation depends on the speed at which the virus adapts to each stage of increasing the incubation temperature. Those who are familiar with this technique will easily know when to reach full CPE. Other details of the method for preparing the temperature sensitive strain of enterovirus 71 suitable for cold of the present invention are described below.

在第二態樣中,本發明提供一種包含本文所述之適於冷之腸病 毒71溫度敏感株之組合物。在一個實施例中,組合物包含有效量之本文所述之病毒株。在另一實施例中,組合物包含一或多種生理學上或醫藥學上可接受之載劑。在另一實施例中,組合物為疫苗。使用熟習此項技術者熟知之技術製備含有本文所述之適於冷之腸病毒71溫度敏感株之疫苗。該等疫苗適用於藉由使用熟習此項技術者熟知之技術向諸如人類個體之個體投與疫苗提供針對親本病毒株之免疫性。 In the second aspect, the present invention provides a cold bowel disease Composition of Toxin 71 temperature-sensitive strain. In one embodiment, the composition contains an effective amount of the virus strain described herein. In another embodiment, the composition includes one or more physiologically or pharmaceutically acceptable carriers. In another embodiment, the composition is a vaccine. A vaccine containing the temperature sensitive strain of Enterovirus 71 suitable for cold as described herein is prepared using techniques well known to those skilled in the art. These vaccines are suitable for administering vaccines to individuals, such as human individuals, to provide immunity against parental virus strains by using techniques well known to those skilled in the art.

應理解,本文所述之適於冷之腸病毒71溫度敏感株當用於引發個體中之保護性免疫反應或防止個體罹患與病毒相關之疾病或延遲與病毒相關之疾病之發作或減慢其進展速率時,係以另外包含一或多種生理學上或醫藥學上可接受之載劑之組合物形式投與個體。醫藥學上可接受之載劑已為熟習此項技術者所熟知且包括(但不限於)以下中之一或多者:0.01M-0.1M及較佳0.05M磷酸鹽緩衝劑、磷酸鹽緩衝生理鹽水(PBS)或0.9%生理鹽水。該等載劑亦包括水性或非水性溶液、懸浮液及乳液。水性載劑包括水、醇/水溶液、乳液或懸浮液、生理鹽水及經緩衝之培養基。非水性溶劑之實例為丙二醇、聚乙二醇、諸如橄欖油之植物油、及諸如油酸乙酯之可注射有機酯。非經腸媒劑包括氯化鈉溶液、林格氏右旋糖(Ringer's dextrose)、右旋糖及氯化鈉、乳酸林格氏液及不揮發性油。靜脈內媒劑包括流體及營養補充劑、電解質補充劑(諸如基於林格氏右旋糖之彼等補充劑)及類似媒劑。固體組合物可包含無毒性固體載劑,諸如葡萄糖、蔗糖、甘露糖醇、山梨糖醇、乳糖、澱粉、硬脂酸鎂、纖維素或纖維素衍生物、碳酸鈉及碳酸鎂。對於以氣溶膠形式投與,諸如經肺及/或鼻內傳送,較佳藉由無毒性界面活性劑,(例如)C6至C22脂肪酸之酯或偏酯或天然甘油酯及推進劑調配試劑或組合物。可包括諸如卵磷脂之其他載劑以便於鼻內遞送。醫藥學上可接受之載劑可進一步包含少量助劑物質,諸如潤濕或乳化劑、防腐劑及其他添加劑,諸如抗菌劑、抗氧化 劑及螯合劑,其提高活性成份之存放期及/或有效性。如在此項技術中所熟知,可調配本發明之組合物以在投與個體之後提供活性成份之快速、持續或延遲釋放。 It should be understood that the temperature-sensitive strain of Enterovirus 71 suitable for cold as described herein can be used to induce a protective immune response in an individual or prevent the individual from suffering from a virus-related disease or delay the onset or slow down the onset of a virus-related disease At the rate of progression, it is administered to an individual in the form of a composition that additionally contains one or more physiologically or pharmaceutically acceptable carriers. Pharmaceutically acceptable carriers are well known to those skilled in the art and include (but are not limited to) one or more of the following: 0.01M-0.1M and preferably 0.05M phosphate buffer, phosphate buffer Normal saline (PBS) or 0.9% normal saline. Such carriers also include aqueous or non-aqueous solutions, suspensions and emulsions. Aqueous vehicles include water, alcohol/aqueous solutions, emulsions or suspensions, physiological saline, and buffered media. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's solution, and fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and similar vehicles. The solid composition may contain non-toxic solid carriers such as glucose, sucrose, mannitol, sorbitol, lactose, starch, magnesium stearate, cellulose or cellulose derivatives, sodium carbonate, and magnesium carbonate. For administration in aerosol form, such as pulmonary and/or intranasal delivery, it is better to use non-toxic surfactants, (for example) esters or partial esters of C6 to C22 fatty acids or natural glycerides and propellant formulation reagents or combination. Other carriers such as lecithin may be included to facilitate intranasal delivery. The pharmaceutically acceptable carrier may further contain a small amount of auxiliary substances, such as wetting or emulsifying agents, preservatives and other additives, such as antibacterial agents, antioxidants Agents and chelating agents, which increase the shelf life and/or effectiveness of active ingredients. As is well known in the art, the composition of the present invention can be formulated to provide rapid, sustained or delayed release of the active ingredient after administration to an individual.

在第三態樣中,本發明提供一種誘發諸如人類個體之個體中之保護性免疫反應之方法,其包含向個體投與預防上或治療學上或免疫學上有效量之本文所述之適於冷之腸病毒71溫度敏感株。在一個實施例中,保護性免疫反應保護個體免受由腸病毒71造成之疾病。在一個實施例中,該疾病為手足口病。在另一實施例中,該疾病為無菌性腦膜炎。在另一實施例中,該疾病為腦炎。在另一實施例中,該疾病為小兒麻痹症狀麻痹。在一個實施例中,以疫苗形式投與本文所述之適於冷之腸病毒71溫度敏感株。在另一實施例中,個體已曝露至野生型腸病毒71。「曝露」至腸病毒71意謂與腸病毒71接觸以使得可產生感染。在另一實施例中,投與本文所述之適於冷之腸病毒71溫度敏感株阻止諸如人類個體之個體罹患與腸病毒71相關之疾病。在另一實施例中,個體已曝露至野生型腸病毒71。在另一實施例中,投與本文所述之適於冷之腸病毒71溫度敏感株延遲諸如人類個體之感染病毒之個體中與腸病毒71相關之疾病之發作或減慢其進展速率。 In a third aspect, the present invention provides a method for inducing a protective immune response in an individual such as a human individual, which comprises administering to the individual a prophylactic or therapeutically or immunologically effective amount of the appropriate amount described herein. Enterovirus 71 is a temperature-sensitive strain of cold. In one embodiment, the protective immune response protects the individual from diseases caused by Enterovirus 71. In one embodiment, the disease is hand, foot and mouth disease. In another embodiment, the disease is aseptic meningitis. In another embodiment, the disease is encephalitis. In another embodiment, the disease is polio symptom paralysis. In one example, the temperature-sensitive strain of Enterovirus 71 described herein suitable for cold is administered as a vaccine. In another example, the individual has been exposed to wild-type enterovirus 71. "Exposure" to enterovirus 71 means contact with enterovirus 71 so that infection can occur. In another embodiment, the administration of the enterovirus 71 temperature-sensitive strain suitable for cold as described herein prevents individuals such as human individuals from suffering from enterovirus 71-related diseases. In another example, the individual has been exposed to wild-type enterovirus 71. In another embodiment, the administration of the enterovirus 71 temperature-sensitive strain suitable for cold as described herein delays the onset or slows the progression rate of enterovirus 71-related diseases in a virus-infected individual such as a human individual.

如本文所用,「投與」意謂使用熟習此項技術者已知之各種方法及遞送系統中之任一者遞送。可(例如)腹膜內、顱內、靜脈內、經口、經黏膜、皮下、經皮、皮內、肌肉內、局部、非經腸、經由植入、鞘內、淋巴內、病灶內、心包或硬膜外進行投與。亦可以氣溶膠形式投與試劑或組合物,諸如經肺及/或鼻內遞送。投與可進行(例如)一次、多次及/或歷經一或多個延長期限。 As used herein, "administration" means delivery using any of various methods and delivery systems known to those skilled in the art. Can (e.g.) be intraperitoneal, intracranial, intravenous, oral, transmucosal, subcutaneous, percutaneous, intradermal, intramuscular, topical, parenteral, via implantation, intrathecal, intralymphatic, intralesional, pericardial Or administered epidurally. The agent or composition can also be administered in aerosol form, such as pulmonary and/or intranasal delivery. The investment may be made, for example, once, multiple times, and/or over one or more extended periods.

可例如藉由向個體投與主劑量之疫苗、隨後在適合之時段之後一或多次後續投與疫苗引發個體中之保護性免疫反應。投與疫苗之間的適合之時段可由熟習此項技術者容易地確定,且通常為約若干週至 若干月。然而,本發明不受限於投與之任何特定方法、途徑或頻率。 A protective immune response in the individual can be elicited, for example, by administering a main dose of the vaccine to the individual, followed by one or more subsequent administrations of the vaccine after a suitable period of time. The suitable period between the administration of the vaccine can be easily determined by those familiar with the technology, and is usually about several weeks to Several months. However, the present invention is not limited to any particular method, approach or frequency of administration.

「預防上有效之劑量」或「免疫學上有效之劑量」為在投與有病毒感染傾向或有罹患與病毒相關之病症傾向的個體時,在個體中誘發保護個體免於感染病毒或罹患病症之免疫反應的疫苗量。「保護」個體意謂減少個體感染病毒之可能性或減輕個體中之病症發作之可能性至少兩倍,較佳至少十倍。舉例而言,若個體具有1%機率感染病毒,兩倍地減少個體感染病毒之可能性將使得個體具有0.5%機率感染病毒。最佳地,「預防上有效之劑量」在個體中誘發完全阻止個體感染病毒或完全阻止個體中之病症發作的免疫反應。 "Prophylactically effective dose" or "immunologically effective dose" means that when administered to an individual who is prone to viral infection or has a tendency to suffer from a virus-related disease, it induces and protects the individual from infection or disease The amount of vaccine for the immune response. "Protecting" an individual means reducing the possibility of the individual being infected with a virus or reducing the probability of the onset of a disease in the individual at least twice, preferably at least ten times. For example, if an individual has a 1% chance of contracting the virus, a two-fold reduction in the possibility of the individual being infected with the virus will give the individual a 0.5% chance of contracting the virus. Optimally, the "prophylactically effective dose" induces an immune response in the individual that completely prevents the individual from being infected with the virus or completely prevents the onset of the disease in the individual.

本發明之免疫及治療方法中之任一者之某些實施例可進一步包含向個體投與少一種佐劑。「佐劑」應意謂適合於在個體中增強抗原之免疫原性及加強免疫反應之任何試劑。包括顆粒狀佐劑之許多佐劑適合於與以蛋白質及核酸為主之疫苗一起使用,且合併佐劑與抗原之方法已為熟習此項技術者所熟知。適用於蛋白免疫之佐劑包括(但不限於)礬、弗氏完全佐劑(FCA)、弗氏不完全佐劑(FIA)、礬佐劑、以皂素為主之佐劑(諸如Quil A及QS-21)及其類似佐劑。 Certain embodiments of any of the immunization and treatment methods of the present invention may further comprise administering to the individual at least one adjuvant. "Adjuvant" shall mean any agent suitable for enhancing the immunogenicity of an antigen and enhancing the immune response in an individual. Many adjuvants including particulate adjuvants are suitable for use with vaccines based on proteins and nucleic acids, and methods of combining adjuvants and antigens are well known to those skilled in the art. Adjuvants suitable for protein immunization include (but are not limited to) alum, Freund's complete adjuvant (FCA), Freund's incomplete adjuvant (FIA), alum adjuvant, saponin-based adjuvants (such as Quil A) And QS-21) and similar adjuvants.

本發明亦提供一種使用溫度敏感性作為表型標記以使病毒減毒之方法。根據此態樣,該方法發展適於冷之溫度敏感病毒株。本發明之方法為活體外實驗室製程,其改變病毒之生物生長特徵以適應於30℃以下之培育溫度下之最佳複製。以遞增地降低用於培養病毒之培育溫度之系統、逐步方式進行適應製程直至達成所選用於病毒之最佳複製之目標溫度。 The present invention also provides a method of using temperature sensitivity as a phenotypic marker to attenuate viruses. According to this aspect, the method was developed for cold temperature-sensitive virus strains. The method of the present invention is an in vitro laboratory process, which changes the biological growth characteristics of the virus to adapt to the optimal replication at an incubation temperature below 30°C. The adaptation process is carried out in a stepwise manner with a system that gradually reduces the incubation temperature used for culturing the virus until the selected target temperature for the best replication of the virus is reached.

因此,根據本發明,該方法包含以下步驟:(i)製備親本野生型病毒之參考儲備料,(ii)使用就每次繼代獲得完全細胞病變效應(CPE)而言較低感染倍率(MOI)及較短培育時段之接種體在較高MOI及約34℃至約36℃、較佳約34℃之培育溫度下培育經親本野生型病毒之參 考儲備料感染之細胞培養物五次或五次以上的繼代直至在每次繼代獲得完全CPE,(iii)使用就每次繼代獲得完全細胞病變效應(CPE)而言較低感染倍率(MOI)及較短培育時段之接種體在較高MOI及比前述步驟中低約1℃至約3℃之培育溫度下培育經前述步驟之所得病毒感染之細胞培養物五次或五次以上的繼代直至在每次繼代獲得完全CPE,及(iv)以遞增地降低培育溫度之系統、逐步方式重複步驟(iii)直至達成所選用於病毒之最佳複製之目標溫度。在一個實施例中,目標溫度為約26℃至約29℃,較佳約28℃。在一個實施例中,遞增地降低培育溫度為降低約1℃至約2℃之溫度。 Therefore, according to the present invention, the method includes the following steps: (i) preparing a reference stock of the parental wild-type virus, (ii) using a lower infection rate in terms of obtaining a complete cytopathic effect (CPE) for each passage ( MOI) and the inoculum with a shorter incubation period is cultivated with the parental wild-type virus at a higher MOI and an incubation temperature of about 34°C to about 36°C, preferably about 34°C. Consider five or more subcultures of the cell culture infected with the stock material until a complete CPE is obtained at each subculture. (iii) Use a lower infection rate for each subculture to obtain a complete cytopathic effect (CPE) (MOI) and the inoculum with a shorter incubation period. Cultivate the virus-infected cell culture obtained by the previous step at a higher MOI and an incubation temperature of about 1°C to about 3°C lower than the previous step for five or more times Subsequent to CPE until complete CPE is obtained in each subculture, and (iv) Step (iii) is repeated in a stepwise manner with a system of incrementally lowering the incubation temperature until the selected target temperature for optimal replication of the virus is reached. In one embodiment, the target temperature is about 26°C to about 29°C, preferably about 28°C. In one embodiment, reducing the incubation temperature incrementally is a temperature reduction of about 1°C to about 2°C.

在一個實施例中,藉由在約36℃至約38℃、較佳約37℃之溫度下培育經野生型病毒感染之細胞培養物一次或兩次繼代直至獲得完全細胞病變效應(CPE)製備親本野生型病毒之參考儲備料。將含有生成之病毒之培養物上澄液之等分試樣置放於小瓶或其他適合之儲存裝置中。此培養物上澄液充當親本野生型病毒之參考儲備料。參考親本野生型病毒係用於後續減毒製程。在另一實施例中,將親本野生型病毒之參考儲備料之等分試樣儲存於適合之溫度,諸如-80℃下。 In one embodiment, the cell culture infected with the wild-type virus is subcultured once or twice at a temperature of about 36°C to about 38°C, preferably about 37°C, until a complete cytopathic effect (CPE) is obtained. Prepare the reference stock of the parental wild-type virus. Place an aliquot of the culture supernatant containing the generated virus in a vial or other suitable storage device. The supernatant of this culture serves as a reference stock for the parental wild-type virus. The reference parent wild-type virus line is used for the subsequent attenuation process. In another embodiment, an aliquot of the reference stock of the parental wild-type virus is stored at a suitable temperature, such as -80°C.

在一個實施例中,一旦病毒獲得完全細胞病變效應(CPE)便藉由獲得含有病毒之澄清培養物上澄液且傳遞此上澄液至細胞(例如維羅細胞)之各連續新鮮的新培養瓶中而在製程之各步驟中使病毒繼代。在另一實施例中,在各連續改變至較低培育溫度開始時以20之較高感染倍率(MOI)傳遞含有病毒之培養物上澄液。在注意到病毒能夠在接種後3日內於維羅細胞中引起完全CPE之後,再藉由相同MOI之病毒接種含有新鮮匯合之單層維羅細胞之新培養瓶至少三次繼代,隨後減少至5至10之較低MOI。一旦注意到病毒能夠在以5至10之較低MOI接種後3日內引起完全CPE,在5至10之MOI下再繼代至少三次之後,減毒製程隨後前進至連續較低培育溫度之下一階段。每次繼代所需之天 數取決於病毒在遞增地降低培育溫度之各階段適應的速度。熟習此項技術者將容易地知道何時達到完全CPE。 In one embodiment, once the virus has achieved a complete cytopathic effect (CPE), the clear culture supernatant containing the virus is obtained and the supernatant is transferred to each continuous fresh new culture of cells (such as Vero cells) The virus is subcultured in the bottle and in each step of the process. In another embodiment, the virus-containing culture supernatant is delivered at a higher infection rate (MOI) of 20 at the beginning of each successive change to a lower incubation temperature. After noticing that the virus can cause complete CPE in Vero cells within 3 days after inoculation, inoculate a new culture flask containing freshly confluent single-layer Vero cells with virus of the same MOI for at least three subcultures, and then reduce to 5 To the lower MOI of 10. Once it is noted that the virus can cause complete CPE within 3 days after inoculation with a lower MOI of 5 to 10, and after at least three subcultures at an MOI of 5 to 10, the attenuation process then proceeds to a continuous lower incubation temperature. stage. Days required for each succession The number depends on the speed at which the virus adapts to each stage of increasing the incubation temperature. Those who are familiar with this technique will easily know when to reach full CPE.

在一個實施例中,病毒為任何病毒。在另一實施例中,病毒為RNA病毒。在另一實施例中,RNA病毒為正鏈RNA病毒。在另一實施例中,病毒為小核糖核酸病毒科之成員。在另一實施例中,病毒為腸病毒屬之成員。在一個實施例中,病毒為腸病毒71(EV71)。在另一實施例中,病毒為柯薩奇病毒A16(CA16)。本發明之方法適用於產生此等病毒中之任一者之適於冷之溫度敏感病毒株,包括(但不限於)EV71及CA16之適於冷之溫度敏感株。 In one embodiment, the virus is any virus. In another embodiment, the virus is an RNA virus. In another embodiment, the RNA virus is a positive-strand RNA virus. In another embodiment, the virus is a member of the Picornaviridae family. In another embodiment, the virus is a member of the genus Enterovirus. In one embodiment, the virus is Enterovirus 71 (EV71). In another embodiment, the virus is Coxsackie virus A16 (CA16). The method of the present invention is suitable for producing cold-suitable temperature-sensitive virus strains of any of these viruses, including but not limited to the cold-suitable temperature-sensitive strains of EV71 and CA16.

在一實施例中,待藉由病毒感染之細胞為允許病毒生長之任何細胞。在一較佳實施例中,細胞為維羅細胞(ATCC CCL-81)。在一個實施例中,藉由適合於細胞生長之培養基中之常規繼代維持細胞。在細胞為維羅細胞之實施例中,在補充有10%胎牛血清(FCS)之達爾伯克改質伊格爾培養基(DMEM)中培養維羅細胞。在一個實施例中,將維持於補充有1% FCS之DMEM中之維羅細胞用於產生親本野生型病毒、病毒培養、減毒、滴定及評估溫度敏感表型。在另一實施例中,DMEM補充有1% FCS以使病毒株適應於在依次降低之培育溫度中複製。 In one embodiment, the cell to be infected by the virus is any cell that allows the virus to grow. In a preferred embodiment, the cell is a Vero cell (ATCC CCL-81). In one embodiment, the cells are maintained by routine subculture in a medium suitable for cell growth. In the example where the cells are Vero cells, the Vero cells are cultured in Dulbecco modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS). In one embodiment, Vero cells maintained in DMEM supplemented with 1% FCS are used to produce parental wild-type virus, virus culture, attenuation, titration, and assessment of temperature-sensitive phenotypes. In another embodiment, DMEM is supplemented with 1% FCS to adapt the virus strain to replicate in successively lowering incubation temperatures.

本發明亦關於藉由本文所述之方法生成之適於冷之溫度病毒株。藉由本文所述之方法生成之適於冷之溫度病毒適用於使用熟習此項技術者熟知之技術產生疫苗。該等疫苗適用於藉由使用熟習此項技術者熟知之技術向個體投與疫苗提供針對親本病毒株之免疫性。 The present invention also relates to cold temperature virus strains produced by the methods described herein. The cold temperature virus produced by the method described herein is suitable for vaccine production using techniques well known to those skilled in the art. These vaccines are suitable for administering vaccines to individuals to provide immunity against parental virus strains by using techniques well known to those skilled in the art.

本發明方法適用於產生小核糖核酸病毒科及腸病毒屬之適於冷之溫度敏感病毒。已藉由生產適於冷之溫度敏感株EV71(TLLα)及EV71(TLLβ)於本文中展示該適用性,該等溫度敏感株在遞增地降低培育溫度下於細胞培養物中連續繼代之後得到。EV71(TLLβ)病毒株 在特定活體外細胞培養條件下保持表型及遺傳穩定性且在靜脈內接種之後於猴中並不呈現神經毒性。亦已藉由產生適於冷之溫度敏感性CA16於本文中展示該適用性,該CA16在遞增地降低培育溫度下於細胞培養物中連續繼代之後得到。 The method of the present invention is suitable for the production of temperature-sensitive viruses suitable for cold of the Picornaviridae and Enteroviruses. This applicability has been demonstrated in this article by the production of temperature-sensitive strains EV71 (TLLα) and EV71 (TLLβ) suitable for cold. These temperature-sensitive strains are obtained after successive subcultures in cell cultures under increasing incubation temperature. . EV71 (TLLβ) virus strain It maintains phenotype and genetic stability under specific in vitro cell culture conditions and does not exhibit neurotoxicity in monkeys after intravenous inoculation. This applicability has also been demonstrated herein by generating a temperature-sensitive CA16 suitable for cold, which is obtained after successive subcultures in cell cultures at increasing incubation temperature.

本發明亦提供一種藉由本文所述之適於冷之腸病毒71溫度敏感株使個體免疫之套組。套組包含本文所述之適於冷之腸病毒71溫度敏感株、醫藥學上可接受之載劑、施用器及其使用指導材料。本發明包括熟習此項技術者已知之套組之其他實施例。該等指導可提供任何適用於引導投與本文所述之適於冷之腸病毒71溫度敏感株之資訊。 The present invention also provides a kit for immunizing individuals with the temperature-sensitive enterovirus 71 strain suitable for cold described herein. The kit includes the cold-suitable enterovirus 71 temperature-sensitive strain described herein, a pharmaceutically acceptable carrier, an applicator, and instruction materials for its use. The present invention includes other embodiments of sets known to those skilled in the art. These guidelines can provide any information suitable for guiding the administration of the temperature-sensitive enterovirus 71 strain suitable for cold described herein.

除非另外指明,否則本發明之實踐採用化學、分子生物學、微生物學、重組DNA、遺傳學、免疫學、細胞生物學、細胞培養及轉殖基因生物學之習知技術,該等技術在此項技術之技能內。參見例如Maniatis等人,1982,Molecular Cloning(Cold Spring Harbor Laboratory Press,Cold Spring Harbor,New York);Sambrook等人,1989,Molecular Cloning,第2版(Cold Spring Harbor Laboratory Press,Cold Spring Harbor,New York);Sambrook及Russell,2001,Molecular Cloning,第3版(Cold Spring Harbor Laboratory Press,Cold Spring Harbor,New York);Green and Sambrook,2012,Molecular Cloning,第4版(Cold Spring Harbor Laboratory Press,Cold Spring Harbor,New York);Ausubel等人,1992,Current Protocols in Molecular Biology(John Wiley & Sons,包括定期更新);Glover,1985,DNA Cloning(IRL Press,Oxford);Russell,1984,Molecular biology of plants:a laboratory course manual(Cold Spring Harbor Laboratory Press,Cold Spring Harbor,N.Y.);Anand,Techniques for the Analysis of Complex Genomes,(Academic Press,New York,1992);Guthrie及Fink,Guide to Yeast Genetics and Molecular Biology(Academic Press,New York, 1991);Harlow及Lane,1988,Antibodies,(Cold Spring Harbor Laboratory Press,Cold Spring Harbor,New York);Nucleic Acid Hybridization(B.D.Hames & S.J.Higgins編1984);Transcription And Translation(B.D.Hames & S.J.Higgins編1984);Culture Of Animal Cells(R.I.Freshney,Alan R.Liss,Inc.,1987);Immobilized Cells And Enzymes(IRL Press,1986);B.Perbal,A Practical Guide To Molecular Cloning(1984);論叢Methods In Enzymology(Academic Press,Inc.,N.Y.);Methods In Enzymology,第154及155卷(Wu等人編);Immunochemical Methods In Cell And Molecular Biology(Mayer 及Walker編,Academic Press,London,1987);Handbook Of Experimental Immunology,第I-IV卷(D.M.Weir及C.C.Blackwell編,1986);Riott,Essential Immunology,第6版,Blackwell Scientific Publications,Oxford,1988;Fire等人,RNA Interference Technology:From Basic Science to Drug Development,Cambridge University Press,Cambridge,2005;Schepers,RNA Interference in Practice,Wiley-VCH,2005;Engelke,RNA Interference(RNAi):The Nuts & Bolts of siRNA Technology,DNA Press,2003;Gott,RNA Interference,Editing,and Modification:Methods and Protocols(Methods in Molecular Biology),Human Press,Totowa,NJ,2004;Sohail,Gene Silencing by RNA Interference:Technology and Application,CRC,2004。 Unless otherwise specified, the practice of the present invention adopts the conventional techniques of chemistry, molecular biology, microbiology, recombinant DNA, genetics, immunology, cell biology, cell culture, and transgenic biology. Within the technical skills. See, for example, Maniatis et al ., 1982, Molecular Cloning (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York); Sambrook et al ., 1989, Molecular Cloning , 2nd edition (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York) ); Sambrook and Russell, 2001, Molecular Cloning , 3rd edition (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York); Green and Sambrook, 2012, Molecular Cloning , 4th edition (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York); Ausubel et al ., 1992, Current Protocols in Molecular Biology (John Wiley & Sons, including regular updates); Glover, 1985, DNA Cloning (IRL Press, Oxford); Russell, 1984, Molecular biology of plants: a laboratory course manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY); Anand, Techniques for the Analysis of Complex Genomes , (Academic Press, New York, 1992); Guthrie and Fink, Guide to Yeast Genetics and Molecular Biology ( Academic Press, New York, 1991); Harlow and Lane, 1988, Antibodies , (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York); Nucleic Acid Hybridization (BDHames & SJHiggins, eds. 1984); Transcription And Translation (BDHames & SJHiggins, eds. 1984); Culture Of Animal Cells (RIFreshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide To Molecular Cloning (1984); Methods In Enzymology (Academic Press, Inc., NY); Methods In Enzymology , Volumes 154 and 155 (Wu et al. eds); Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, Edited, Academic Press, London, 1987); Handbook Of Experimental Immunology , Volume I-IV (eds by DMWeir and CC Blackwell, 1986); Riott, Essential Immunology , 6th Edition, Blackwell Scientific Publications, Oxford, 1988; Fire et al. , RNA Interference Technology: From Basic Science to Drug Development , Cambridge University Press, Cambridge, 2005; Schepers, RNA Interference in Practice , Wiley-VCH, 2005; Engelke, RNA Interference (RNAi): The Nuts & Bolts of siRNA Technology , DNA Press, 2003; Gott, RNA Interference, Editing, and Modification: Methods and Protocols (Methods in Molecular Biology) , Human Press, Totowa, NJ, 2004; Sohail, Gene Silencing by RNA Interf erence: Technology and Application , CRC, 2004.

實例 Instance

參考以下實例描述本發明,以說明方式提供該等實例且不意欲以任何方式限制本發明。使用在此項技術中熟知之標準技術或在下文中特定描述之技術。 The present invention is described with reference to the following examples, which are provided in an illustrative manner and are not intended to limit the present invention in any way. Use standard techniques well known in the art or techniques specifically described below.

實例1 Example 1 發展適於冷之人類腸病毒71(EV71)溫度敏感株之材料及方法 Materials and methods for developing temperature-sensitive strains of human enterovirus 71 (EV71) suitable for cold

細胞、病毒及冷適應製程:將藉由補充有10%胎牛血清(FCS)之達爾伯克改質伊格爾培養基(DMEM)中之常規繼代維持之維羅細胞(ATCC CCL-81)用於病毒培養、減毒、滴定及評估溫度敏感表型。根據先前描述之技術將在維羅細胞中與呈現手足口病(HFMD)之患者之各別口腔分泌物、大便及腦幹標本分離之屬於基因型C1、B3及B4之三個EV71分離株空斑純化一次(Dougherty,1964)。在空斑純化之後,在37℃下於維羅細胞中兩次繼代之後製備指示為各別親本野生型病毒株之病毒儲備料。將所有病毒儲備料及各別繼代之病毒株儲存於零下80℃冷凍器中。 Cells, viruses and cold adaptation process : Vero cells (ATCC CCL-81) will be maintained by conventional subculture in DMEM supplemented with 10% Fetal Calf Serum (FCS). It is used for virus culture, attenuation, titration and evaluation of temperature-sensitive phenotype. According to the previously described technique, the three EV71 isolates belonging to genotypes C1, B3 and B4 were isolated from the individual oral secretions, stool and brainstem specimens of patients with hand, foot and mouth disease (HFMD) in Vero cells. The spots were purified once (Dougherty, 1964). After plaque purification, virus stocks indicated as respective parent wild-type virus strains were prepared after two subcultures in Vero cells at 37°C. Store all virus stocks and the virus strains of their respective generations in a freezer at minus 80°C.

含有1% FCS之DMEM中之新鮮匯合的年輕單層維羅細胞係用於適應病毒株以在自34℃之初始低培育溫度開始之連續較低培育溫度下複製。一旦病毒獲得完全細胞病變效應(CPE)便將含有病毒之澄清培養物上澄液傳遞至維羅細胞之各連續新鮮的新培養瓶中。在各連續改變至較低培育溫度開始時以20之較高感染倍率(MOI)傳遞含有病毒之培養物上澄液。在注意到病毒能夠在接種後3日內於維羅細胞中引起完全CPE之後,再藉由相同MOI之病毒接種含有新鮮匯合之單層維羅細胞之新培養瓶至少三次繼代,隨後減少至5至10之較低MOI。一旦注意到病毒能夠在以5至10之較低MOI接種後3日內引起完全CPE,在再繼代至少三次之後,減毒製程隨後前進至下一連續較低培育溫度。在此實例中用於發展適於冷之腸病毒71溫度敏感株之隨後遞增之較低培育溫度為34℃、32℃、30℃、29℃及28℃。 The freshly confluent young monolayer Vero cell line in DMEM containing 1% FCS was used to adapt the virus strain to replicate at a continuous lower incubation temperature starting from the initial low incubation temperature of 34°C. Once the virus has achieved a complete cytopathic effect (CPE), the clear culture supernatant containing the virus is delivered to each successively fresh new culture flask of Vero cells. At the beginning of each successive change to a lower incubation temperature, the virus-containing culture supernatant was delivered with a higher infection rate (MOI) of 20. After noticing that the virus can cause complete CPE in Vero cells within 3 days after inoculation, inoculate a new culture flask containing freshly confluent single-layer Vero cells with virus of the same MOI for at least three subcultures, and then reduce to 5 To the lower MOI of 10. Once it is noted that the virus can cause complete CPE within 3 days after inoculation with a lower MOI of 5 to 10, after at least three subcultures, the attenuation process then proceeds to the next continuous lower incubation temperature. In this example, the lower incubation temperatures used to develop the temperature-sensitive enterovirus 71 strain suitable for cold are 34°C, 32°C, 30°C, 29°C, and 28°C.

病毒滴定:根據World Health Organization之Polio Laboratory Manual 2004中所述之方法加以稍微修改在維羅細胞中藉由微量滴定分析測定病毒滴定濃度且遵循Reed及Muench法(1938)將病毒滴定濃度計算為每毫升50%細胞培養感染劑量(CCID50)。簡言之,在藉由相等量之氯仿進行處理以分散病毒聚集體之後,在含有1% FCS之DMEM 中製得澄清病毒上澄液之10倍連續稀釋液。96孔平底組織培養盤中之維羅細胞單層(每孔104個細胞)係藉由100μl連續稀釋之各病毒儲備料進行接種且在5% CO2之環境下於每一各別培育溫度下培育5日,隨後觀測CPE之存在。 Virus titration : According to the method described in the Polio Laboratory Manual 2004 of the World Health Organization, the virus titration concentration was determined by microtitration analysis in Vero cells and the virus titration concentration was calculated as per the Reed and Muench method (1938). 50% cell culture infection dose (CCID 50 ) per milliliter. In short, after treatment with an equal amount of chloroform to disperse virus aggregates, a 10-fold serial dilution of the clear virus supernatant was prepared in DMEM containing 1% FCS. Vero cell monolayers of 96 well flat bottom tissue culture plates (104 cells per well) were serially diluted by lines 100μl of each virus stock material and inoculated at 5% CO 2 environment in each of the respective incubation temperature Incubate for 5 days, and then observe the presence of CPE.

溫度敏感性分析:在28℃、37℃及39.5℃之培育溫度下使用兩種方法評估維羅細胞中之病毒株之生長特徵。第一種方法評估病毒株在感染細胞中引起完全CPE所用之天數(複製動力學)且第二種方法評估病毒株在各特定測試溫度下培育之細胞中之滴定濃度。簡言之,在第一種方法中,藉由維持培養基(含1% FCS之DMEM)替換三個含有類似年齡之匯合單層維羅細胞之T-25組織培養瓶的生長培養基。隨後藉由置放於各別培育箱中1小時使各燒瓶中之培養基平衡至待測試之特定溫度且隨後藉由10感染倍率(MOI)之劑量下之病毒株接種。若在10日培養結束時未注意到CPE,則將上澄液傳遞至新的單層維羅細胞之燒瓶中且類似地又培育10日。若在第二次繼代之後未注意到CPE,則將其視為無病毒複製。在第二種方法中,將密度為每100μl 104個細胞之維羅細胞懸浮液接種至三個96孔細胞培養盤之各孔中且在5% CO2之環境下於37℃下進行培育。在培育10小時之後,隨後藉由置放於各別培育箱中1小時使各細胞培養盤平衡至待測試之特定溫度。隨後藉由100μl病毒株之10倍連續稀釋液對各孔中之細胞進行接種,隨後將其轉移至各別溫度之培育箱中且培育5日,隨後觀測CPE之存在。 Temperature sensitivity analysis : Two methods were used to evaluate the growth characteristics of virus strains in Vero cells at incubation temperatures of 28°C, 37°C and 39.5°C. The first method assesses the number of days (replication kinetics) the virus strain takes to cause complete CPE in infected cells and the second method assesses the titer concentration of the virus strain in cells grown at each specific test temperature. In short, in the first method, the growth medium of three T-25 tissue culture flasks containing confluent monolayer Vero cells of similar age was replaced by maintenance medium (DMEM with 1% FCS). Subsequently, the culture medium in each flask was equilibrated to the specific temperature to be tested by placing it in each incubator for 1 hour, and then inoculated with the virus strain at a dose of 10 infection rate (MOI). If no CPE was noticed at the end of the 10-day culture, the supernatant was transferred to a flask of a new monolayer of Vero cells and similarly incubated for another 10 days. If CPE is not noticed after the second generation, it is considered as virus-free. In the second method, a Vero cell suspension with a density of 10 4 cells per 100 μl was inoculated into each well of three 96-well cell culture plates and incubated at 37°C under 5% CO 2 . After incubating for 10 hours, each cell culture plate was equilibrated to the specific temperature to be tested by placing it in each incubator for 1 hour. Subsequently, the cells in each well were inoculated with 100 μl of 10-fold serial dilution of the virus strain, and then transferred to an incubator at each temperature and incubated for 5 days, and then the presence of CPE was observed.

遺傳穩定性及溫度敏感性分析:為評估培養於特定培養環境及細胞型下之病毒株之遺傳穩定性,將病毒株在5 MOI之病毒接種體下於細胞培養物中又繼代20次且在28℃之培育溫度下進行培育。在20次繼代結束時,藉由在如上文所述之28℃、37℃及39.5℃之培育溫度下進行培養來評估病毒株之溫度敏感性表型特徵。隨後相對於其各別親本野生型病毒之全基因組對其各別基因組之全核苷酸序列進行定序及 分析。 Genetic stability and temperature sensitivity analysis : In order to evaluate the genetic stability of virus strains cultured in a specific culture environment and cell type, the virus strains were subcultured 20 times in cell culture under 5 MOI of virus inoculum. The incubation was carried out at an incubation temperature of 28°C. At the end of the 20 subcultures, the temperature-sensitive phenotypic characteristics of the virus strains were evaluated by culturing at the incubation temperature of 28°C, 37°C, and 39.5°C as described above. Then sequence and analyze the complete nucleotide sequence of each genome relative to the complete genome of each parent wild-type virus.

在穩定的適於冷之溫度敏感病毒株上進行溫度敏感性分析之反轉。所選病毒株在5% CO2之環境下於37℃之溫度下培育之單層維羅細胞中繼代5次。在每次繼代中,使用10 MOI之病毒接種體。藉由如上文所描述用於溫度敏感性分析之類似方法在28℃、37℃及39.5℃之培育溫度下評估每次繼代所得之病毒株於維羅細胞中之生長特徵。對37℃之培養溫度下之每次連續繼代之病毒株之全基因組進行定序及分析。 Perform the inversion of temperature sensitivity analysis on a stable temperature-sensitive virus strain suitable for cold. The selected virus strains were cultured in a single layer of Vero cells at 37°C under 5% CO 2 for 5 times. In each passage, a virus inoculum of 10 MOI was used. The growth characteristics of the virus strain obtained in each subculture in Vero cells were evaluated at 28°C, 37°C, and 39.5°C incubation temperature by a similar method as described above for temperature sensitivity analysis. Sequence and analyze the entire genome of the virus strains of each successive generation at a culture temperature of 37°C.

RNA提取、RT-PCR及定序:使用市售病毒RNA提取套組(Viral RNA Extraction Kit)(Qiagen,Germany)自完全CPE下之感染細胞之培養液提取病毒基因組RNA。使用Superscript II RNA聚合酶(Invitrogen,USA)藉由EV71特異性引子進行首鏈合成,且使用GoTaq Green PCR mix(Promega,USA)藉由18個簡併引子對進行後續PCR。使用BigDye Terminator定序套組(Applied Biosystems,USA)對產生之片段進行定序。進行5'RACE以藉由使用標準T4 DNA接合酶(Fermentas,USA)將5'-蛹蟲草菌素(cordycepin)封端之銜接子DT88(5'-GAA GAG AAG GTG GAA ATG GCG TTT TTG G-蛹蟲草菌素-3';SEQ ID NO:1)接合至EV71 cDNA之5'末端來測定5'-UTR病毒序列,且然後使用與DT88互補之引子(5'-CCA AAA CGC CAT TTC CAC CTT CTC TTC3';SEQ ID NO:2)及EV71特異性引子(5'-ATT CAG GGG CCG GAG GAC TAC-3';SEQ ID NO:3)進行標準PCR。亦進行3'-RACE以使用oligo-dT引子測定3'-UTR病毒序列(Li等人,2005)。 RNA extraction, RT-PCR and sequencing : Viral RNA Extraction Kit (Qiagen, Germany) was used to extract viral genome RNA from the culture medium of infected cells under complete CPE. First-strand synthesis was performed using Superscript II RNA polymerase (Invitrogen, USA) with EV71-specific primers, and the subsequent PCR was performed using 18 degenerate primer pairs using GoTaq Green PCR mix (Promega, USA). The generated fragments were sequenced using BigDye Terminator sequencing kit (Applied Biosystems, USA). Carry out 5'RACE to use standard T4 DNA ligase (Fermentas, USA) to 5'-cordycepin (cordycepin) capped adaptor DT88 (5'-GAA GAG AAG GTG GAA ATG GCG TTT TTG G- Cordyceps militaris-3'; SEQ ID NO: 1) was joined to the 5'end of EV71 cDNA to determine the 5'-UTR virus sequence, and then a primer complementary to DT88 (5'-CCA AAA CGC CAT TTC CAC CTT was used) CTC TTC3'; SEQ ID NO: 2) and EV71 specific primers (5'-ATT CAG GGG CCG GAG GAC TAC-3'; SEQ ID NO: 3) were subjected to standard PCR. 3'-RACE was also performed to determine the 3'-UTR virus sequence using oligo-dT primers (Li et al., 2005).

分子選殖及質體純化:將具有模糊序列之片段選殖至pZero-2質體(Invitrogen,USA)中且轉形至TOP10大腸桿菌細胞(Invitrogen,USA)中。使用市售質體小規模純化套組(Plasmid Miniprep Kit)(Qiagen,Germany)自來自各轉形體之至少10個菌落提取含有選殖之EV71片段 的質體且隨後測定選殖片段之序列。 Molecular cloning and plastid purification : The fragments with fuzzy sequences were cloned into pZero-2 plastids (Invitrogen, USA) and transformed into TOP10 E. coli cells (Invitrogen, USA). A commercially available Plasmid Miniprep Kit (Qiagen, Germany) was used to extract plastids containing the cloned EV71 fragment from at least 10 colonies from each transformant, and then the sequence of the cloned fragment was determined.

使用European Molecular Biology Open Software Suite(EMBOSS;http://mobyle.pasteur.fr/cgi-bin/portal.py?#forms::merger)(Rice等人2000)合併獲得之片段序列。使用程序BioEdit Sequence Alignment Editor版本7.0.9.0(Hall,1997)將合併之序列與EV71病毒株3799-SIN-98(GenBank寄存編號DQ341354.1)之參考序列比對。 Use European Molecular Biology Open Software Suite (EMBOSS; http://mobyle.pasteur.fr/cgi-bin/portal.py?#forms::merger) (Rice et al. 2000) to merge the obtained fragment sequences. Use the program BioEdit Sequence Alignment Editor version 7.0.9.0 (Hall, 1997) to align the combined sequence with the reference sequence of the EV71 virus strain 3799-SIN-98 (GenBank accession number DQ341354.1).

猴研究:將關於所選適於冷之EV71溫度敏感株(EV71:TLLβP20)之安全性及免疫原性之猴研究承包給以Animal Facility of DUKE-NUS,Singapore為基地之研究者。將7隻無結核分支桿菌(Mycobacterium tuberculosis)及猴免疫缺陷病毒之食蟹獼猴(Macaca fascicularis)(3隻雌性(2202F、2207F、2891F)及4隻雄性(0791M、2247M、2889M、2890M),具有3.23Kg之平均體重(範圍2.44至4.11,SD=0.7))用於研究穩定的適於冷之溫度敏感EV71:TLLβP20之安全性及免疫原性。關於不存在針對EV71之結合(間接免疫螢光法)及中和抗體對所有7隻猴進行預篩選。研究及所有動物程序經Committee for Biosafety and Animal Handling及Ethical Committee of DUKE-NUS,Singapore批准。根據委員會之指導原則進行病毒接種及觀測、動物護理及屍體剖檢。 Monkey research : The monkey research on the safety and immunogenicity of the selected EV71 temperature-sensitive strain (EV71: TLLβP20) suitable for cold was contracted to a researcher based in Animal Facility of DUKE-NUS, Singapore. 7 Macaca fascicularis (3 females (2202F, 2207F, 2891F) and 4 males (0791M, 2247M, 2889M, 2890M) without Mycobacterium tuberculosis and monkey immunodeficiency virus ( Macaca fascicularis ), with The average body weight of 3.23Kg (range 2.44 to 4.11, SD=0.7) is used to study the safety and immunogenicity of stable temperature-sensitive EV71: TLLβP20 suitable for cold. All 7 monkeys were pre-screened for the absence of binding to EV71 (indirect immunofluorescence method) and neutralizing antibodies. The study and all animal procedures were approved by the Committee for Biosafety and Animal Handling and the Ethical Committee of DUKE-NUS, Singapore. Virus inoculation and observation, animal care and autopsy were carried out in accordance with the guidelines of the committee.

在藉由氯胺酮之淺麻醉下將1ml病毒接種體靜脈內接種至右隱靜脈中。3隻猴(2889M、0791M及2891F)每隻猴給予107 CCID50下之靜脈內劑量的EV71:TLLβP20,另3隻猴(2890M、2202F及2247M)每隻猴給予108 CCID50且第7隻猴充當陰性對照。每日兩次觀測猴之臨床疾病,尤其為神經表現且藉由植入之溫度感測器記錄其體溫。每日收集來自各猴之大便且儲存於零下80℃冷凍器中以用於在日後進行病毒分離。在接種後(PI)第4日在深度麻醉下殺死兩隻猴(一隻來自各自不同病毒劑量之接種體)。在屍體剖檢時,收集中樞神經系統(CNS)、非神經組 織及血液之各個部分用於組織病理學及病毒學分析。在接種後第8日,殺死另一類似組之兩隻猴且收集相同類型之組織及血液以用於屍體剖檢時之組織病理學及病毒學研究。剩餘的兩隻猴在收集血液樣品用於評估抗EV71抗體之後,在接種後第14日給予各別等效加強劑量之EV71:TLLβP20。在接收加強劑量之後的16日將其無痛苦地殺死且收集CNS組織用於屍體剖檢時之組織病理學研究。 1 ml of virus inoculum was intravenously inoculated into the right saphenous vein under light anesthesia with ketamine. Three monkeys (2889M, 0791M and 2891F) were given an intravenous dose of EV71: TLLβP20 under 10 7 CCID 50 , and the other 3 monkeys (2890M, 2202F and 2247M) were given 10 8 CCID 50 and the seventh A monkey served as a negative control. Observe the monkey's clinical diseases, especially neurological manifestations, twice a day, and record its body temperature with an implanted temperature sensor. Stool from each monkey was collected daily and stored in a minus 80°C freezer for virus isolation in the future. On the 4th day after inoculation (PI), two monkeys (one from each inoculum with a different virus dose) were killed under deep anesthesia. During autopsy, various parts of central nervous system (CNS), non-neural tissue and blood are collected for histopathology and virological analysis. On the 8th day after inoculation, two monkeys of another similar group were killed and the same type of tissue and blood were collected for histopathology and virology studies during autopsy. After collecting blood samples for the evaluation of anti-EV71 antibodies, the remaining two monkeys were given respective equivalent booster doses of EV71: TLLβP20 on the 14th day after vaccination. On the 16th day after receiving the booster dose, they were killed without pain and the CNS tissue was collected for histopathological study during autopsy.

組織學及免疫組織化學:將CNS組織標本(大腦、小腦、基底神經節、腦幹及脊髓)及非神經組織(淋巴結、脾、肝、腎、肺及心臟)固定於磷酸鹽緩衝鹽水(PBS)中之10%福馬林(formalin)中且在固定之後嵌入石蠟中。將脊髓分別在頸椎、胸椎及腰椎層級水平地切片10次、8次及10次。在移除石蠟及復水製程之後藉由蘇木精及曙紅(H&E)及藉由神經髓質染色法(Luxol-fast blue/cresyl violet)(Kluver-Barrera法)對厚度為6μm之石蠟切片進行染色。 Histology and immunohistochemistry : fix CNS tissue specimens (brain, cerebellum, basal ganglia, brainstem and spinal cord) and non-nerve tissues (lymph nodes, spleen, liver, kidney, lung and heart) in phosphate buffered saline (PBS) ) In 10% formalin (formalin) and embedded in paraffin after fixation. The spinal cord was sliced 10 times, 8 times, and 10 times at the cervical, thoracic, and lumbar levels respectively. After the paraffin removal and rehydration process, hematoxylin and eosin (H&E) and neuromedullary staining (Luxol-fast blue/cresyl violet) (Kluver-Barrera method) were used to paraffin sections with a thickness of 6 μm Perform dyeing.

抗原偵測及自猴分離病毒:在無添加管及肝素化管中收集血樣。使用Ficoll-PaqueTM PLUS(GE Healthcare,Sweden)自肝素化血液收集周邊血液單核細胞(PBMC)。在藉由無菌PBS洗滌兩次之後,將PBMC懸浮液之等分試樣接種至塗佈有鐵氟龍(Teflon)之載片之孔上以使用商用偵測單株抗體(目錄號3360,Light Diagnostics,USA)藉由間接免疫螢光分析偵測EV71抗原。藉由將PBMC懸浮液接種至含有單層維羅細胞之24孔細胞培養盤之孔中進行病毒分離。藉由接種50μl及100μl血清至含有單層維羅細胞之24孔培養盤之各別孔中對各血清試樣進行病毒分離。用交換兩次之無菌PBS稍微洗滌猴組織且藉由在II級生物安全櫃中使用研缽及研棒研磨進行均勻化。藉由在1000g下離心10分鐘使製備於DMEM中之組織勻漿(10%,w/v)澄清。經由0.22微米針筒過濾器過濾澄清上澄液且藉由接種100μl及200μl濾過物進行病毒分離。在PBS中製得10%大便懸浮液且藉由在1000g下離心10分鐘進 行澄清。在經由0.22微米針筒過濾器過濾之後,藉由接種100μl及200μl大便濾過物進行病毒分離。一式兩份地對猴樣品進行所有病毒分離工作,在28℃下培育一組接種細胞培養物且在37℃下培育另一組。 Antigen detection and virus isolation from monkeys : Collect blood samples in non-additive tubes and heparinized tubes. Peripheral blood mononuclear cells (PBMC) were collected from heparinized blood using Ficoll-Paque PLUS (GE Healthcare, Sweden). After washing twice with sterile PBS, an aliquot of the PBMC suspension was inoculated onto the wells of a Teflon-coated slide to use a commercial detection monoclonal antibody (Cat. No. 3360, Light Diagnostics, USA) detects EV71 antigen by indirect immunofluorescence analysis. Virus isolation was performed by seeding the PBMC suspension into the wells of a 24-well cell culture plate containing a single layer of Vero cells. Virus isolation was performed on each serum sample by inoculating 50 μl and 100 μl of serum into each well of a 24-well culture plate containing a single layer of Vero cells. The monkey tissues were washed slightly with sterile PBS exchanged twice and homogenized by grinding with a mortar and pestle in a Class II biological safety cabinet. The tissue homogenate (10%, w/v) prepared in DMEM was clarified by centrifugation at 1000 g for 10 minutes. The clarified supernatant was filtered through a 0.22 micron syringe filter and the virus was separated by inoculating 100 μl and 200 μl of the filtrate. A 10% stool suspension was prepared in PBS and clarified by centrifugation at 1000 g for 10 minutes. After filtering through a 0.22 micron syringe filter, virus isolation was performed by inoculating 100 μl and 200 μl stool filtrate. All virus isolation work was performed on monkey samples in duplicate, with one group of inoculated cell cultures grown at 28°C and another group at 37°C.

分子偵測及全基因組定序:商用病毒RNA提取及純化套組(Qiagen,Germany)係用於自血清、PBMC及澄清組織勻漿提取病毒基因組RNA。擴增所有EV71基因型之VP1基因之近端三分之一之商用一步RT-PCR套組(Qiagen,Germany)及共同寡核苷酸引子對(有義:5'-CACCCTTGTGATACCATGGATCAG-3'(SEQ ID NO:4);反義:5'-GTGAATTAAGAACRCAYCGTGTYT-3'(SEQ ID NO:5))係用於在自組織提取EV71特異性病毒RNA之後對其進行分子擴增及偵測。18對基於全基因組序列EV71:TLLβ之序列特異性引子係用於對存在於免疫後第4日及第8日獲得之兩猴之血清中之EV71全基因組進行擴增及定序。將任何未能藉由直接定序給出良好序列讀數之PCR擴增片段選殖至pZero-2(Invitrogen,USA)中且轉形至TOP10大腸桿菌中。自各轉形體選擇至少十個菌落且在攜帶插入物之純化質體上進行定序。 Molecular detection and whole genome sequencing : Commercial viral RNA extraction and purification kit (Qiagen, Germany) is used to extract viral genome RNA from serum, PBMC and clarified tissue homogenate. A commercial one-step RT-PCR kit (Qiagen, Germany) and common oligonucleotide primer pair (sense: 5'-CACCCTTGTGATACCATGGATCAG-3' (SEQ) to amplify the proximal third of the VP1 gene of all EV71 genotypes ID NO: 4); Antisense: 5'-GTGAATTAAGAACRCAYCGTGTYT-3' (SEQ ID NO: 5)) is used for molecular amplification and detection of EV71-specific viral RNA after self-organization. 18 pairs of sequence-specific primers based on the whole genome sequence EV71:TLLβ were used to amplify and sequence the EV71 whole genome present in the sera of two monkeys obtained on the 4th and 8th day after immunization. Any PCR amplified fragments that failed to give good sequence reads by direct sequencing were cloned into pZero-2 (Invitrogen, USA) and transformed into TOP10 E. coli. At least ten colonies were selected from each transformant and sequenced on purified plastids carrying inserts.

血清結合及中和抗體分析:在幾乎完全CPE下收集經基因型B3之EV71感染之維羅細胞且藉由無菌PBS洗滌五次。在最後一次洗滌之後,感染細胞之懸浮液與經過洗滌之未感染維羅細胞懸浮液以約4個感染細胞比1個未感染細胞之比混合。將含有250個細胞之十微升混合細胞懸浮液小心地分層至塗佈有鐵氟龍(Teflon)之12孔載片之各孔上且使其在溫盤上乾燥。將乾燥載片在冷丙酮中固定10分鐘且用作藉由間接免疫螢光分析自就IgM而言之1:10及就IgG而言之1:20之初始稀釋度開始之EV71結合抗體分析的內部抗原。在抗-EV71 IgM滴定濃度之分析中,用適當濃度之蛋白A(Invitrogen,USA)處理猴血清以移除先前藉由無菌PBS進行連續2倍稀釋之IgG。 Serum binding and neutralizing antibody analysis : The Vero cells infected with EV71 genotype B3 were collected under almost complete CPE and washed five times with sterile PBS. After the last wash, the suspension of infected cells and the washed suspension of uninfected Vero cells are mixed at a ratio of about 4 infected cells to 1 uninfected cell. Ten microliters of mixed cell suspension containing 250 cells was carefully layered onto each well of a 12-well slide coated with Teflon and allowed to dry on a warm pan. The dried slides were fixed in cold acetone for 10 minutes and used for the EV71 binding antibody analysis by indirect immunofluorescence analysis starting from the initial dilution of 1:10 for IgM and 1:20 for IgG Internal antigen. In the analysis of the anti-EV71 IgM titration concentration, the monkey serum was treated with protein A (Invitrogen, USA) at the appropriate concentration to remove the IgG that had been previously serially diluted by sterile PBS by 2 times.

根據World Health Organization之Polio Laboratory Manual 2004中 所述之方法加以稍微修改使用維羅細胞藉由微中和分析測定猴之中和抗體滴定濃度。用於中和之EV71之各基因型之濃度為每100μl 100 CCID50。使用96孔平底培養盤進行病毒中和分析。在起始於1:10稀釋度之100μl DMEM(1% FCS)之量下一式兩份地製備各血清樣品之連續2倍稀釋液。將相等量(100μl)之含有100 CCID50 EV71之DMEM(1% FCS)中之病毒工作儲備料添加至稀釋之血清之各孔中且在37℃下培育兩小時。在培育之後,將100μl含有250個細胞之DMEM(10% FCS)中之維羅細胞懸浮液添加至各孔中。將96孔培養盤小心地密封且在5% CO2之環境中於37℃下進行培育。每日讀取盤關於影響各孔中之維羅細胞之CPE之存在多達8日。藉由不顯示CPE之稀釋度最高之孔測定各血清樣品之中和抗體之滴定濃度。 It was slightly modified according to the method described in the Polio Laboratory Manual 2004 of the World Health Organization. Vero cells were used to determine the monkey neutralizing antibody titer by microneutralization analysis. The concentration of each genotype of EV71 used for neutralization is 100 CCID 50 per 100 μl. Use 96-well flat-bottomed culture plates for virus neutralization analysis. Serial 2-fold dilutions of each serum sample were prepared in duplicate in an amount of 100 μl DMEM (1% FCS) starting at a dilution of 1:10. An equal amount (100 μl) of virus working stock in DMEM (1% FCS) containing 100 CCID 50 EV71 was added to each well of the diluted serum and incubated at 37° C. for two hours. After incubation, 100 μl of the Vero cell suspension in DMEM (10% FCS) containing 250 cells was added to each well. The 96-well culture plate was carefully sealed and incubated at 37°C in a 5% CO 2 environment. The disc is read daily for the presence of CPE affecting the Vero cells in each well for up to 8 days. The titrated concentration of neutralizing antibody in each serum sample was determined by the hole that did not show the highest dilution of CPE.

實例2 Example 2 適於冷之腸病毒71溫度敏感株之表型及基因型特徵之結果 The results of the phenotype and genotype characteristics of the temperature-sensitive strain of Enterovirus 71 suitable for cold

(EV71:TLLα、EV71:TLLαP20、EV71:TLLβ、EV71:TLLβP20及EV71:TLLβP40) (EV71: TLLα, EV71: TLLαP20, EV71: TLLβ, EV71: TLLβP20 and EV71: TLLβP40)

用於得到適於冷之病毒株的三個原始親本EV71病毒在10 MOI之病毒接種體及37.5℃之培育溫度下之接種之後的3日內於維羅細胞中引起完全CPE。在相同病毒接種體中,原始親本EV71病毒在39.5℃之培育溫度下在5日內於維羅細胞中引起完全CPE。然而,來源於培養於37.5℃下之維羅細胞中之首先兩次繼代之所有三種原始親本EV71病毒在10 MOI之病毒接種體及28℃之培育溫度下均不於維羅細胞中引起CPE。亦未在28℃下培養10日結束時的盲目繼代之後注意到CPE。藉由使用針對EV71之商用偵測單株抗體藉由間接免疫螢光分析在10日培養結束時於培養物上澄液中存在陰性染色之懸浮細胞進一步支持接種維羅細胞中不存在病毒複製。 The three original parental EV71 viruses used to obtain virus strains suitable for cold caused complete CPE in Vero cells within 3 days after inoculation with a virus inoculum of 10 MOI and an incubation temperature of 37.5°C. In the same virus inoculum, the original parental EV71 virus caused complete CPE in Vero cells within 5 days at an incubation temperature of 39.5°C. However, all three original parental EV71 viruses derived from the first two passages in Vero cells cultured at 37.5°C did not cause in Vero cells at a virus inoculum of 10 MOI and an incubation temperature of 28°C. CPE. Nor was CPE noticed after blind subculture at the end of 10 days incubation at 28°C. The presence of negatively stained suspension cells in the culture supernatant at the end of the 10-day culture by indirect immunofluorescence using a commercial detection monoclonal antibody against EV71 further supports the absence of virus replication in the inoculated Vero cells.

在連續降低之培育溫度下之超過90次繼代之後,所有三種繼代 EV71病毒株均能夠在

Figure 103101883-A0202-12-0025-28
5 MOI之病毒接種體及28℃之培育溫度下之接種的3日內於維羅細胞中引起完全CPE。病毒株進一步在28℃之培育溫度下繼代直至第100次繼代。在第100次繼代,將來源於自患者之口液、腦幹組織及大便標本分離之原始EV71的病毒株分別指示為EV71:TLL、EV71:TLLα及EV71:TLLβ。在溫度敏感性分析中,所有三種適於冷之病毒株使用10 MOI之病毒接種體在28℃之培育溫度下在2日內於維羅細胞中引起完全CPE,但在37℃之培育溫度下需要4日以達到完全CPE。在39.5℃之培育溫度下,所有三種病毒株在培養10日之後均未注意到CPE。然而,EV71:TLL在盲目繼代至培育於39.5℃下之維羅細胞之新燒瓶中之後於培養之第10日給出2+ CPE。即使在另外兩次盲目繼代至培育於39.5℃下之維羅細胞之新燒瓶中之後,EV71:TLLα及EV71:TLLβ仍未注意到CPE。在各10日培養結束時自藉由EV71:TLLα或EV71:TLLβ接種之培養物上澄液收集之懸浮細胞(包括來源於盲目繼代之彼等細胞)藉由EV71偵測單株抗體未給出陽性免疫螢光染色。自藉由EV71:TLL接種之培養物上澄液收集之約1%懸浮細胞給出陽性染色,然而,在10日培養之後未注意到CPE。 After more than 90 subcultures under successively reduced cultivation temperatures, all three subcultured EV71 virus strains were able to
Figure 103101883-A0202-12-0025-28
5 The virus inoculum of MOI and the inoculation at the incubation temperature of 28 ℃ caused complete CPE in the Vero cells within 3 days. The virus strain was further subcultured at a cultivation temperature of 28°C until the 100th subculture. At the 100th generation, the virus strains derived from the original EV71 isolated from the patient’s oral fluid, brainstem tissue and stool specimens were designated as EV71: TLL, EV71: TLLα, and EV71: TLLβ, respectively. In the temperature sensitivity analysis, all three virus strains suitable for cold use 10 MOI virus inoculum to cause complete CPE in Vero cells within 2 days at an incubation temperature of 28°C, but it is required at an incubation temperature of 37°C 4 days to achieve full CPE. At an incubation temperature of 39.5°C, all three virus strains did not notice CPE after 10 days of cultivation. However, EV71:TLL gave 2+ CPE on the 10th day of culture after blindly subcultured to a new flask of Vero cells grown at 39.5°C. Even after two other blind passages to new flasks of Vero cells grown at 39.5°C, EV71:TLLα and EV71:TLLβ still did not notice CPE. At the end of each 10-day culture, the suspension cells (including those cells derived from blind succession) collected from the culture supernatants inoculated with EV71:TLLα or EV71:TLLβ were detected by EV71. Positive immunofluorescence staining. Approximately 1% of the suspended cells collected from the culture supernatant inoculated with EV71:TLL gave positive staining, however, no CPE was noticed after 10 days of culture.

28℃及37℃之培育溫度係用於分析三種適於冷之病毒株之病毒複製滴定濃度且重複該分析至少4次。當滴定培養物分別培育於28℃及37℃下時,EV71:TLL之滴定濃度為1×108 CCID50/ml及2至3×107 CCID50/ml。EV71:TLLα在28℃之培育溫度下給出1×108 CCID50/ml之病毒滴定濃度且在37℃下給出1至2×105.5-6 CCID50/ml之滴定濃度。EV71:TLLβ在28℃之培育溫度下給出2至5×108 CCID50/ml之滴定濃度且在37℃下給出1至1×107 CCID50/ml之滴定濃度。 The incubation temperature of 28°C and 37°C is used to analyze the virus replication titration concentration of three strains suitable for cold and repeat the analysis at least 4 times. When the titrated culture was incubated at 28°C and 37°C, the titrated concentration of EV71:TLL was 1×10 8 CCID 50 /ml and 2 to 3×10 7 CCID 50 /ml. EV71: TLLα gives a virus titer of 1×10 8 CCID 50 /ml at an incubation temperature of 28°C and a titer of 1 to 2×10 5.5-6 CCID 50 /ml at 37°C. EV71: TLLβ gives a titer of 2 to 5×10 8 CCID 50 /ml at an incubation temperature of 28°C and a titer of 1 to 1×10 7 CCID 50 /ml at 37°C.

藉由在28℃之培育溫度及10 MOI之病毒接種體下於維羅細胞中使EV71:TLLα及EV71:TLLβ又繼代20代評估兩種病毒株之適應冷的穩定性。在又20代之後,EV71:TLLα在接種後2日內於28℃下培育之細 胞中引起完全CPE但在37℃下培育之細胞中未能引起CPE,即使在2個盲目繼代之後亦如此。其於28℃之培育溫度下保持1×108 CCID50/ml之病毒滴定濃度的能力。EV71:TLLβ在另外20代之後於28℃及37℃之培育溫度下維持就生長動力學及病毒滴定濃度而言之相同適於冷之表型。藉由在相同培養條件下再繼代另外20代(自第100次繼代之另外40代)進一步評估EV71:TLLβ之冷適應的穩定性且發現其保持類似適於冷之溫度敏感表型。 By subcultured EV71:TLLα and EV71:TLLβ in Vero cells at an incubation temperature of 28°C and a virus inoculum of 10 MOI for another 20 generations, the stability of the two virus strains to adapt to cold was evaluated. After another 20 generations, EV71:TLLα caused complete CPE in cells grown at 28°C within 2 days after seeding, but failed to cause CPE in cells grown at 37°C, even after 2 blind passages. Its ability to maintain a virus titer of 1×10 8 CCID 50 /ml at an incubation temperature of 28°C. EV71: TLLβ maintains the same cold-suitable phenotype in terms of growth kinetics and virus titer concentration at 28°C and 37°C incubation temperatures after another 20 generations. The stability of the cold adaptation of EV71:TLLβ was further evaluated by subcultured under the same culture conditions for another 20 generations (another 40 generations from the 100th generation) and it was found that it maintained a temperature-sensitive phenotype similar to that suitable for cold.

藉由在各完全CPE下於37℃下培育之維羅細胞中之病毒的6次連續繼代於EV71:TLLβP20上進行自適於冷之溫度敏感表型之逆轉的評估。就28℃、37℃及39.5℃之培育溫度下之生長動力學及病毒滴定濃度而言之來源於37℃下培育之每一各別培養物之病毒的生長特徵係顯示於表1中。病毒在培育於37℃之細胞中3次連續繼代之後仍不能在39.5℃之培育溫度下於維羅細胞中產生可存活之感染粒子(缺乏陽性免疫螢光染色細胞)且不能在第6次重複繼代在39.5℃之培育溫度下於細胞培養物中引起完全CPE。 The evaluation of the reversal of the temperature-sensitive phenotype suitable for cold was performed by 6 consecutive passages of the virus in the Vero cells cultivated at 37°C under each complete CPE on EV71:TLLβP20. The growth characteristics of the virus derived from each individual culture cultivated at 37°C in terms of growth kinetics and virus titer concentration at incubation temperatures of 28°C, 37°C, and 39.5°C are shown in Table 1. The virus cannot produce viable infectious particles in Vero cells at an incubation temperature of 39.5°C (lack of positive immunofluorescence staining cells) after 3 consecutive subcultures in cells incubated at 37°C and cannot be used in the sixth time. Repeated subcultures caused complete CPE in cell culture at an incubation temperature of 39.5°C.

Figure 103101883-A0202-12-0026-1
Figure 103101883-A0202-12-0026-1

每次繼代時病毒之生長特徵及滴定濃度在28℃、37℃及39.5℃之培育溫度下於維羅細胞中培養或滴定。 The growth characteristics and titer of the virus during each subculture were cultured or titrated in Vero cells at the incubation temperature of 28°C, 37°C and 39.5°C.

P1:繼代1 P1: Succession 1

IFA:間接免疫螢光分析 IFA: Indirect immunofluorescence analysis

對EV71:TLLα及EV71:TLLβ、其於另外20代之後的各別病毒株(EV71:TLLαP20及EV71:TLLβP20)及原始親本野生型之全基因組進行定序及分析。EV71:TLLαP20之核苷酸序列係闡述於SEQ ID NO:6中。EV71:TLLβP20之核苷酸序列係闡述於SEQ ID NO:7中。EV71:TLLα、EV71:TLLαP20與原始親本野生型之間的其基因之各片段的核苷酸數目變化係顯示於表2中。原始親本野生型、EV71:TLLβ、EV71:TLLβP20與EV71:TLLβP40(另外40代)之間的核苷酸數目變化係顯示於表3中。亦對來源於37℃之培育溫度下之6次連續繼代之溫度敏感逆轉研究之病毒株的全基因組進行定序且相對於EV71:TLLβP20之核苷酸數目變化係顯示於表4中。 The whole genomes of EV71: TLLα and EV71: TLLβ, its respective virus strains after another 20 generations (EV71: TLLαP20 and EV71: TLLβP20) and the original parent wild-type were sequenced and analyzed. The nucleotide sequence of EV71: TLLαP20 is set forth in SEQ ID NO:6. The nucleotide sequence of EV71: TLLβP20 is set forth in SEQ ID NO:7. EV71: TLLα, EV71: The variation of the nucleotide number of each fragment of the gene between TLLαP20 and the original parent wild-type is shown in Table 2. The changes in the number of nucleotides between the original parent wild-type, EV71: TLLβ, EV71: TLLβP20 and EV71: TLLβP40 (another 40 generations) are shown in Table 3. The whole genome of the virus strain derived from the temperature-sensitive reversal study of 6 consecutive generations at the incubation temperature of 37°C was also sequenced and the nucleotide number changes relative to EV71:TLLβP20 are shown in Table 4.

Figure 103101883-A0202-12-0027-2
Figure 103101883-A0202-12-0027-2
Figure 103101883-A0202-12-0028-3
Figure 103101883-A0202-12-0028-3

Figure 103101883-A0202-12-0028-4
Figure 103101883-A0202-12-0028-4

Figure 103101883-A0202-12-0029-5
Figure 103101883-A0202-12-0029-5

實例3 Example 3 適於冷之人類腸病毒71溫度敏感株(EV71:TLLβP20)之猴研究的結果 The result of monkey study of temperature-sensitive strain of human enterovirus 71 (EV71: TLLβP20) suitable for cold

臨床發現:每日早晚兩次進行對研究之猴之臨床狀態之一般觀測及體溫之量測。在整個過程中,注意到所有猴為活躍且進食正常的。猴中無一者產生任何細微的局部神經缺損,諸如肢體無力、顫抖或異常運動。除給予1×108 CCID50/ml之靜脈內劑量之一猴(2247M)外猴中無一者在接種後第3日產生低等級發熱之峰值(39.3℃)。猴中無一者在其於深度麻醉下被殺死時之複秤時具有重量損失。 Clinical findings : The general observation of the clinical state of the monkeys in the study and the measurement of body temperature were carried out twice a day in the morning and evening. Throughout the process, it was noted that all monkeys were active and eating normally. None of the monkeys had any minor local nerve defects, such as limb weakness, tremor, or abnormal movement. Except for one monkey (2247M) given an intravenous dose of 1×10 8 CCID 50 /ml, none of the monkeys had a low-grade fever peak (39.3°C) on the 3rd day after vaccination. None of the monkeys had weight loss when they were rescaled when they were killed under deep anesthesia.

屍體剖檢及組織學發現:所有屍體剖檢之猴未注意到總體死後病變。所有收集用於組織病理學檢驗之猴組織未見到異常組織學發現。 Necropsy and histological findings : All monkeys on autopsy did not notice overall postmortem lesions. No abnormal histological findings were seen in all monkey tissues collected for histopathological examination.

病毒學研究:使用在28℃及37℃之培育溫度下之維羅細胞進行猴血清、PBMC、大便樣品及所有屍體剖檢組織的病毒分離。儘管在培養10日之後進行盲目繼代,但未自猴樣品中之任一者中分離病毒。針對藉由RT-PCR對於EV71測試呈陽性之血清、PBMC樣品及彼等組織勻漿進行維羅細胞中之另一盲目繼代。儘管未能分離病毒,但使用商用單株抗體藉由間接免疫螢光分析(圖1a)在接種後第4日收集的來源於兩隻猴2891F(接收1×107 CCID50之EV71:TLLβP20)及2890M(接收1×108 CCID50之EV71:TLLβP20)之肝素化血液的幾個PBMC中偵測到EV71抗原。在自接種後第8日收集之猴的肝素化血液收集之PBMC中未偵測到病毒抗原。 Virology research : Virus isolation of monkey serum, PBMC, stool samples and all autopsy tissues was performed using Vero cells at culture temperatures of 28°C and 37°C. Despite blind subculture after 10 days of culture, no virus was isolated from any of the monkey samples. Another blind subculture in Vero cells was performed on serum, PBMC samples and their tissue homogenates that tested positive for EV71 by RT-PCR. Although the virus could not be isolated, it was collected from two monkeys 2891F (EV71 received 1×10 7 CCID 50 : TLLβP20) by indirect immunofluorescence analysis using commercial monoclonal antibodies (Figure 1a) on the 4th day after vaccination. And 2890M (EV71 receiving 1×10 8 CCID 50 : TLLβP20) of several PBMCs of heparinized blood detected EV71 antigen. No viral antigen was detected in the PBMC collected from the heparinized blood of the monkey collected on the 8th day after vaccination.

藉由RT-PCR在接種後第4日與第8日收集之所有猴的血清樣品中偵測到EV71特異性基因組序列。非神經元組織中,僅在2隻猴(2202F及2891F)之脾勻漿中偵測到EV71基因組序列(圖1b)。未在神經元組織勻漿中之任一者中偵測到EV71基因組序列。對兩隻猴(2889M及2890M)之血清樣品(收集於接種後第4日與第8日)中存在的 EV71:TLLβP20之基因組進行提取及完全定序。與EV71:TLLβP20之基因組比較之出現於存在於血清中之病毒株之基因組片段中之每一者中的核苷酸(NT)及對應胺基酸(AA)突變或逆轉之數目係顯示於表5中。 The EV71-specific genome sequence was detected in serum samples of all monkeys collected on the 4th and 8th day after vaccination by RT-PCR. Among non-neuronal tissues, the EV71 genome sequence was only detected in the spleen homogenates of 2 monkeys (2202F and 2891F) (Figure 1b). The EV71 genome sequence was not detected in any of the neuronal tissue homogenates. For two monkeys (2889M and 2890M) serum samples (collected on the 4th and 8th day after vaccination) EV71: The genome of TLLβP20 is extracted and fully sequenced. Compared with the genome of EV71: TLLβP20, the number of nucleotides (NT) and corresponding amino acid (AA) mutations or reversals in each of the genome fragments of the virus strain present in the serum is shown in the table 5 in.

Figure 103101883-A0202-12-0031-6
Figure 103101883-A0202-12-0031-6

猴體液免疫反應:使用內部製備之感染維羅細胞作為抗原藉由間接免疫螢光分析對給予靜脈內EV71:TLLβP20之猴之血清中之結合抗體(IgM及IgG)之存在進行分析。存在於接種後第14日(先前給予等效靜脈內加強劑量)及接種後第30日(加強後第16日)收集之兩隻剩餘的猴(2889M及2890M)之血液中之抗-EV71 IgM及IgG的滴定濃度係顯示於表6中。 Monkey humoral immune response : Using internally prepared infected Vero cells as antigens, the presence of binding antibodies (IgM and IgG) in the serum of monkeys given intravenous EV71:TLLβP20 was analyzed by indirect immunofluorescence analysis. Anti-EV71 IgM present in the blood of two remaining monkeys (2889M and 2890M) collected on the 14th day after vaccination (the equivalent intravenous booster dose was previously given) and the 30th day after vaccination (the 16th day after booster) The titer of IgG and IgG are shown in Table 6.

Figure 103101883-A0202-12-0032-7
Figure 103101883-A0202-12-0032-7

藉由微中和分析測定針對EV71基因型A(BrCr)、B3、B4、B5、C1及C5之收集於接種後第14日及第30日之兩隻猴(2889M及2890M)之血清樣品中之中和抗體滴定濃度。針對EV71基因型中之每一者之猴血清之中和抗體的各別滴定濃度係顯示於表7中。 Determination of EV71 genotypes A (BrCr), B3, B4, B5, C1 and C5 by micro-neutralization analysis was collected in the serum samples of two monkeys (2889M and 2890M) on the 14th and 30th day after vaccination Neutralizing antibody titer concentration. The respective titrated concentrations of neutralizing antibodies in monkey serum for each of the EV71 genotypes are shown in Table 7.

Figure 103101883-A0202-12-0032-8
Figure 103101883-A0202-12-0032-8

實例4 Example 4 發展人類柯薩奇病毒A16之適於冷之溫度敏感株的材料及方法 Materials and methods for developing human coxsackie virus A16 suitable for cold temperature sensitive strains

材料及方法:將藉由補充有10%胎牛血清(FCS)之達爾伯克改質伊格爾培養基(DMEM)中之常規繼代維持之維羅細胞(ATCC CCL-81)用於病毒培養、減毒、滴定及評估柯薩奇病毒A16之溫度敏感性。用於減毒製程之親本野生型柯薩奇病毒A16(CA16)來源於具有手足口病(HFMD)之患者之口腔分泌物。根據先前描述之技術對病毒進行空斑 純化一次。在空斑純化之後,在37℃下於維羅細胞中兩次繼代之後製備指示為各別親本野生型病毒株之病毒儲備料。將所有病毒儲備料及各別繼代之病毒株儲存於零下80℃冷凍器中。 Materials and methods : Vero cells (ATCC CCL-81) maintained by conventional subculture in Dulbecco modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS) were used for virus culture , Attenuation, titration and evaluation of the temperature sensitivity of Coxsackie virus A16. The parental wild-type Coxsackie virus A16 (CA16) used in the attenuation process is derived from oral secretions of patients with hand, foot and mouth disease (HFMD). The virus was plaque purified once according to the previously described technique. After plaque purification, virus stocks indicated as respective parent wild-type virus strains were prepared after two subcultures in Vero cells at 37°C. Store all virus stocks and the virus strains of their respective generations in a freezer at minus 80°C.

含有1% FCS之DMEM中之年輕新鮮匯合的單層維羅細胞係用於適應原始野生型CA16以在自34℃之初始較低培育溫度開始之連續較低培育溫度下複製。在完全細胞病變效應(CPE)下將病毒繼代至維羅細胞之各連續新培養瓶中。病毒在改變至連續較低培育溫度時以20之較高感染倍率(MOI)繼代且在注意到其能夠在接種後3日內引起完全CPE之後,在再繼代至少三次之後減少至5至10之較低MOI。在注意到病毒能夠在以5至10之MOI接種後3日內引起完全CPE之後,在至少再繼代三次之後前進至下一連續較低培育溫度。每次連續繼代之培養物上澄液係儲存於零下80℃冷凍器中供日後分析。 The young, freshly confluent monolayer Vero cell line in DMEM containing 1% FCS was used to adapt the original wild-type CA16 to replicate at a continuous lower incubation temperature starting from the initial lower incubation temperature of 34°C. Under the complete cytopathic effect (CPE), the virus was subcultured to each successive new culture flask of Vero cells. The virus was subcultured at a higher rate of infection (MOI) of 20 when it was changed to a continuous lower incubation temperature and after noticing that it could cause complete CPE within 3 days after inoculation, it was reduced to 5 to 10 after at least three subcultures. The lower MOI. After noting that the virus can cause complete CPE within 3 days after inoculation with an MOI of 5 to 10, proceed to the next continuous lower incubation temperature after at least three more subcultures. The culture supernatant of each successive subculture is stored in a refrigerator at -80°C for future analysis.

病毒滴定:根據World Health Organization之Polio Laboratory Manual 2004中所述之方法加以稍微修改在維羅細胞中藉由微量滴定分析測定病毒滴定濃度且遵循Reed & Muench法(1938)將病毒滴定濃度計算為每毫升50%細胞培養感染劑量(CCID50)。簡言之,在藉由相等量之氯仿進行處理以分散病毒聚集體之後,在含有1% FCS之DMEM中製得澄清病毒上澄液之10倍連續稀釋液。96孔平底組織培養盤中之維羅細胞單層(每孔104個細胞)係藉由100μl連續稀釋之各病毒儲備料進行接種且在5% CO2之環境下於每一各別培育溫度下培育5日,隨後觀測CPE之存在。 Virus titration : According to the method described in the Polio Laboratory Manual 2004 of the World Health Organization, the virus titration concentration was determined by microtiter analysis in Vero cells and the virus titration concentration was calculated as per the Reed & Muench method (1938). 50% cell culture infection dose (CCID 50 ) per milliliter. In short, after treatment with an equal amount of chloroform to disperse virus aggregates, a 10-fold serial dilution of the clear virus supernatant was prepared in DMEM containing 1% FCS. Vero cell monolayers of 96 well flat bottom tissue culture plates (104 cells per well) were serially diluted by lines 100μl of each virus stock material and inoculated at 5% CO 2 environment in each of the respective incubation temperature Incubate for 5 days, and then observe the presence of CPE.

溫度敏感性分析:在28℃、37℃及39.5℃之培育溫度下使用兩種方法評估維羅細胞中之適於冷之病毒株之生長特徵。第一種方法評估病毒株在感染細胞中引起完全CPE所用之天數(複製動力學)且第二種方法評估病毒株在各特定測試溫度下培育之細胞中之滴定濃度。簡言之,在第一種方法中,藉由維持培養基(含1% FCS之DMEM)替換含有 類似年齡之匯合單層維羅細胞之三個T-25組織培養瓶的生長培養基。隨後藉由置放於各別培育箱中1小時使各燒瓶中之培養基平衡至待測試之特定溫度且隨後藉由10感染倍率(MOI)之劑量下之病毒株接種。若在10日培養結束時未注意到CPE,則將上澄液傳遞至新的單層維羅細胞之燒瓶中且類似地培育另外10日。若在第二次繼代之後未注意到CPE,則將其視為無病毒複製。在第二種方法中,將密度為每100μl 104個細胞之維羅細胞懸浮液接種至三個96孔細胞培養盤之各孔中且在5% CO2之環境下於37℃下進行培育。在培育10小時之後,隨後藉由置放於各別培育箱中1小時使各細胞培養盤平衡至待測試之特定溫度。隨後藉由100μl病毒株之10倍連續稀釋液對各孔中之細胞進行接種,隨後將其轉移至各別溫度之培育箱中且培育5日,隨後觀測CPE之存在。 Temperature sensitivity analysis : Two methods were used to evaluate the growth characteristics of cold-suitable virus strains in Vero cells at incubation temperatures of 28°C, 37°C and 39.5°C. The first method assesses the number of days (replication kinetics) the virus strain takes to cause complete CPE in infected cells and the second method assesses the titer concentration of the virus strain in cells grown at each specific test temperature. In short, in the first method, the growth medium of three T-25 tissue culture flasks containing confluent monolayer Vero cells of similar age was replaced by maintenance medium (DMEM with 1% FCS). Subsequently, the culture medium in each flask was equilibrated to the specific temperature to be tested by placing it in each incubator for 1 hour, and then inoculated with the virus strain at a dose of 10 infection rate (MOI). If CPE is not noticed at the end of the 10-day culture, the supernatant is transferred to a flask of a new monolayer of Vero cells and incubated similarly for another 10 days. If CPE is not noticed after the second generation, it is considered as virus-free. In the second method, a Vero cell suspension with a density of 10 4 cells per 100 μl was inoculated into each well of three 96-well cell culture plates and incubated at 37°C under 5% CO 2 . After incubating for 10 hours, each cell culture plate was equilibrated to the specific temperature to be tested by placing it in each incubator for 1 hour. Subsequently, the cells in each well were inoculated with 100 μl of 10-fold serial dilution of the virus strain, and then transferred to an incubator at each temperature and incubated for 5 days, and then the presence of CPE was observed.

實例5 Example 5 適於冷之人類柯薩奇病毒A16溫度敏感株之細胞中之病毒特徵的結果 The result of virus characteristics in cells of cold human Coxsackie virus A16 temperature-sensitive strain

在連續降低之培育溫度的第100次繼代下,適於冷之CA16病毒株能夠在

Figure 103101883-A0202-12-0034-29
5 MOI之病毒接種體及28℃之培育溫度下之接種的3日內於維羅細胞中引起完全CPE,但在37℃之培育溫度下達到完全CPE需要4日。在39.5℃之培育溫度下,未注意到CPE。 Under the 100th successive generation of the continuously lowered incubation temperature, the cold-suitable CA16 virus strain can be
Figure 103101883-A0202-12-0034-29
5 The virus inoculum of MOI and the inoculation at the incubation temperature of 28°C cause complete CPE in the Vero cells within 3 days, but it takes 4 days to reach the complete CPE at the incubation temperature of 37°C. At an incubation temperature of 39.5°C, no CPE was noticed.

28℃及37℃之培育溫度係用於分析適於冷之CA16溫度敏感株之病毒複製滴定濃度。當分別於28℃及37℃下培育滴定培養盤時,適於冷之CA16溫度敏感株之滴定濃度為1×108 CCID50/ml及1×107 CCID50/ml。 The incubation temperature of 28°C and 37°C is used to analyze the virus replication titration concentration of the temperature sensitive strain of CA16 suitable for cold. When the titration plate is incubated at 28°C and 37°C, respectively, the titration concentration of the temperature-sensitive CA16 strain suitable for cold is 1×10 8 CCID 50 /ml and 1×10 7 CCID 50 /ml.

除非本文另有所述或上下文中明顯矛盾,否則在描述本發明之上下文(尤其在以下申請專利範圍之上下文中)中所使用之術語「一(a/an)」及「該(the)」及類似指示物均應解釋為涵蓋單數及複數。除非另外指出,否則術語「包含」、「具有」、「包括」及「含有」均解釋 為開放式術語(意即,意謂「包括(但不限於)」)。除非本文另外指示,否則本文中值範圍之敍述僅意欲充當個別提及屬於該範圍之各獨立值之速記方法,且各獨立值併入本說明書中,如同在本文中個別地敍述一般。舉例而言,若揭示範圍10-15,則亦揭示11、12、13及14。除非本文另外指出或上下文明顯矛盾,否則本文所述之所有方法可以任何適合順序進行。除非另外主張,否則本文所提供之任何及所有實例或例示性語言(例如,「諸如」)之使用僅意欲更好地說明本發明而非限制本發明之範疇。本說明書中之語言不應視作指示任何未主張之要素對於實踐本發明而言必不可少。 Unless otherwise stated herein or obviously contradictory in the context, the terms "a/an" and "the" are used in the context of describing the present invention (especially in the context of the scope of the following patent applications) And similar indicators should be interpreted as covering the singular and plural. Unless otherwise indicated, the terms "include", "have", "include" and "contain" are all interpreted It is an open-ended term (meaning "including (but not limited to)"). Unless otherwise indicated herein, the description of the value range herein is only intended to serve as a shorthand method for individually referring to each independent value belonging to the range, and each independent value is incorporated into this specification as if individually described herein. For example, if the range 10-15 is revealed, 11, 12, 13, and 14 are also revealed. Unless otherwise indicated herein or clearly contradicted by context, all methods described herein can be performed in any suitable order. Unless otherwise claimed, the use of any and all examples or illustrative language (eg, "such as") provided herein is only intended to better illustrate the invention and not to limit the scope of the invention. The language in this specification should not be regarded as indicating that any unclaimed element is essential for practicing the present invention.

應瞭解本發明之方法及組合物可以多種實施例之形式合併,該等實施例中僅若干個係揭示於本文中。本發明之實施例係描述於本文中,包括本發明人已知之進行本發明的最佳模式。在閱讀先前描述之後,彼等實施例之變化對於一般熟習此項技術者而言可變得顯而易見。本發明人期望熟習此項技術者適當時採用此等變化,且本發明人意欲以不同於本文中特定所述之方式來實踐本發明。因此,若適用法律允許,則本發明包括隨附於本文之申請專利範圍中所陳述之標的物的所有修改及等效物。此外,除非本文另外指出或另外明顯與上下文抵觸,否則本發明涵蓋上述要素在其所有可能變化中之任何組合。 It should be understood that the methods and compositions of the present invention can be combined in the form of various embodiments, and only a few of these embodiments are disclosed herein. The embodiments of the present invention are described herein, including the best mode known to the inventors for carrying out the present invention. After reading the previous description, the changes in their embodiments may become obvious to those who are generally familiar with the art. The present inventor expects those familiar with the art to adopt these changes as appropriate, and the present inventor intends to practice the present invention in a manner different from that specifically described herein. Therefore, if permitted by applicable laws, the present invention includes all modifications and equivalents of the subject matter stated in the scope of the patent application attached to this document. In addition, unless otherwise indicated herein or otherwise clearly conflicting with the context, the present invention encompasses any combination of the above-mentioned elements in all possible variations thereof.

參考文獻 references

Alexander J P, Baden L, Pallansch M A, Anderson L J. Enterovirus 71 infection and neurologic disease - United States, 1977-1991. J Infect Dis 1994; 169: 905-908. Alexander J P, Baden L, Pallansch MA, Anderson L J. Enterovirus 71 infection and neurologic disease-United States, 1977-1991. J Infect Dis 1994; 169: 905-908.

Arita M, Shimizu H, Nagata N, Ami Y, Suzaki Y, Sata T, Iwasaki T, Miyamura T. Temperature-sensitive mutants of enterovirus 71 show attenuation in cynomolgus monkeys. J Gen Virol 2005; 86: 1391-401. Arita M, Shimizu H, Nagata N, Ami Y, Suzaki Y, Sata T, Iwasaki T, Miyamura T. Temperature-sensitive mutants of enterovirus 71 show attenuation in cynomolgus monkeys. J Gen Virol 2005; 86: 1391-401.

Arita M, Nagata N, Iwata N, Ami Y, Suzaki Y, Mizuta K, Iwasaki T, Sata T, Wakita T, Shimizu H. An attenuated strain of enterovirus 71 belonging to genotype A showed a broad spectrum of antigenicity with attenuated neurovirulence in Synomolgus monkeys. J Virol 2007; 81(17): 9386-95. Arita M, Nagata N, Iwata N, Ami Y, Suzaki Y, Mizuta K, Iwasaki T, Sata T, Wakita T, Shimizu H. An attenuated strain of enterovirus 71 belonging to genotype A showed a broad spectrum of antigenicity with attenuated neurovirulence in Synomolgus monkeys. J Virol 2007; 81(17): 9386-95.

Bible JM, Iturriza-Gomara M, Megson B, Brown D, Pantelidis P, Earl P, Bendig J, Tong CYW. Molecular epidemiology of human entrovirus 71 in the United Kingdom from 1998 to 2006. J Clin Microbiol 2008; 46(10): 3192-200. Bible JM, Iturriza-Gomara M, Megson B, Brown D, Pantelidis P, Earl P, Bendig J, Tong CYW. Molecular epidemiology of human entrovirus 71 in the United Kingdom from 1998 to 2006. J Clin Microbiol 2008; 46(10) : 3192-200.

Blomberg J, Lycke E, Ahlfors K, Johnsson T, Wolontis S, von Ziepel G. New enterovirus type associated with epidemic of aseptic meningitis and/or hand, foot, and mouth disease. Lancet 1974; 2: 112. Blomberg J, Lycke E, Ahlfors K, Johnsson T, Wolontis S, von Ziepel G. New enterovirus type associated with epidemic of aseptic meningitis and/or hand, foot, and mouth disease. Lancet 1974; 2: 112.

Chang L Y, Huang Y C, Lin T Y. Fulminant neurogenic pulmonary oedema with hand, foot and mouth disease. Lancet 1998; 352: 367-368. Chang L Y, Huang Y C, Lin T Y. Fulminant neurogenic pulmonary oedema with hand, foot and mouth disease. Lancet 1998; 352: 367-368.

Chen HL, Huang JY, Chu TW, Tsai TC, Hung CM, Lin CC, Liu FC, Wang LC, Chen YJ, Lin MF, Chen CM. Expression of VP1 protein in the milk of transgenic mice: a potential oral vaccine protects against enterovirus 71 infection. Vaccine 2008; 26(23): 2882-0. Chen HL, Huang JY, Chu TW, Tsai TC, Hung CM, Lin CC, Liu FC, Wang LC, Chen YJ, Lin MF, Chen CM. Expression of VP1 protein in the milk of transgenic mice: a potential oral vaccine protects against enterovirus 71 infection. Vaccine 2008; 26(23): 2882-0.

Chen CW, Lee YP, Wang YF, Yu CK. Formaldehyde-inactivated human enterovirus 71 vaccine is compatible for co-immunization with a commercial pentavalent vaccine. Vaccine 2011; 29(15): 2772-6. Chen CW, Lee YP, Wang YF, Yu CK. Formaldehyde-inactivated human enterovirus 71 vaccine is compatible for co-immunization with a commercial pentavalent vaccine. Vaccine 2011; 29(15): 2772-6.

Chinese Center for Disease Control and Prevention, and Office of the World Health Organization in China. 2008. Report on the hand, foot and mouth disease outbreak in Fuyang City, Anhui Province and the prevention and control in China. World Health Organization, Beijing, China. http://www2.wpro.who.int/NR/rdonlyres/591D6A7B-FB15-4E94-A1E9-1D3381847D60/0/HFMDCCDC20080515ENG.pdf Chinese Center for Disease Control and Prevention, and Office of the World Health Organization in China. 2008. Report on the hand, foot and mouth disease outbreak in Fuyang City, Anhui Province and the prevention and control in China. World Health Organization, Beijing, China. http://www2.wpro.who.int/NR/rdonlyres/591D6A7B-FB15-4E94-A1E9-1D3381847D60/0/HFMDCCDC20080515ENG.pdf

Chiu CH, Chu C, He CC, Lin TY. Protection of neonatal mice from lethal enterovirus 71 infection by maternal immunization with attenuated Salmonella enterica serovar Typhimurium expressing VP1 of enterovirus 71. Microbes Infect 2006; 8(7): 1671-8. Chiu CH, Chu C, He CC, Lin TY. Protection of neonatal mice from lethal enterovirus 71 infection by maternal immunization with attenuated Salmonella enterica serovar Typhimurium expressing VP1 of enterovirus 71. Microbes Infect 2006; 8(7): 1671-8.

Chomnaitree T, Menegus M A, Schervish-swierkosz E M, Schwalenstocker E. Enterovirus 71 infection: a report of an outbreak with two cases of paralysis and a review of the literature. Pediatrics 1982; 671: 489-493. Chomnaitree T, Menegus M A, Schervish-swierkosz E M, Schwalenstocker E. Enterovirus 71 infection: a report of an outbreak with two cases of paralysis and a review of the literature. Pediatrics 1982; 671: 489-493.

Coleman JR, Papamichail D, Skiena S, Futcher B, Wimmer E, Mueller S. Virus attenuation by genome-scale changes in codon pair bias. Science 2008; 320: 1784-87. Doi: 10. 1126/science. 1155761 Coleman JR, Papamichail D, Skiena S, Futcher B, Wimmer E, Mueller S. Virus attenuation by genome-scale changes in codon pair bias. Science 2008; 320: 1784-87. Doi: 10. 1126/science. 1155761

Chumakov M, Voroshilova M, Shindarov L M, Lavrova I, Gracheva L, Koroleva M, et al. Enterovirus 71 isolated from cases of epidemic poliomyelitis-like disease in Bulgaria. Arch Virol 1979; 60: 329-340. Chumakov M, Voroshilova M, Shindarov L M, Lavrova I, Gracheva L, Koroleva M, et al. Enterovirus 71 isolated from cases of epidemic poliomyelitis-like disease in Bulgaria. Arch Virol 1979; 60: 329-340.

Chung YC, Ho MS, Wu JC, Chen WJ, Huang JH, Chou ST, Hu YC. Immunization with virus-like particles of enterovirus 71 elicits potent immune responses and protects mice against lethal challenge. Vaccine 2008; 26(15): 1855-62. Chung YC, Ho MS, Wu JC, Chen WJ, Huang JH, Chou ST, Hu YC. Immunization with virus-like particles of enterovirus 71 elicits potent immune responses and protects mice against lethal challenge. Vaccine 2008; 26(15): 1855 -62.

Deibel R, Gross L L, Collins D N. Isolation of a new enterovirus. Proc Soc Exp Biolo Med 1975; 148: 203-207. Deibel R, Gross L L, Collins D N. Isolation of a new enterovirus. Proc Soc Exp Biolo Med 1975; 148: 203-207.

Dougherty RM. Animal virus titration technique. In: Techniques in Experimental Virology. Ed; Harris RJC. London and New York: Academic Press 1964;. Chapter 6; 169-224. Dougherty RM. Animal virus titration technique. In: Techniques in Experimental Virology. Ed; Harris RJC. London and New York: Academic Press 1964;. Chapter 6; 169-224.

Gilbert G L, Dickson K E, Waters M J, Kennett M L, Land S A, Sneddon M. Outbreak of enterovirus 71 infection in Victoria, Australia, with a high incidence of neurologic involvement. Pediatr Infect Dis 1988; 7: 484-488. Gilbert G L, Dickson K E, Waters M J, Kennett M L, Land S A, Sneddon M. Outbreak of enterovirus 71 infection in Victoria, Australia, with a high incidence of neurologic involvement. Pediatr Infect Dis 1988; 7: 484-488.

Hagiwara A, Tagaya I, Yoneyama T. Epidemic of hand, foot, and mouth disease associated with enterovirus 71 infection. Intervirology 1978; 9: 60-63. Hagiwara A, Tagaya I, Yoneyama T. Epidemic of hand, foot, and mouth disease associated with enterovirus 71 infection. Intervirology 1978; 9: 60-63.

Hagiwara A, Yoneyama T, Hashimoto I. Isolation of a temperature-sensitive strain of enterovirus 71 with reduced neurovirulence for monkeys. J Gen Virol 1982; 64: 499-502. Hagiwara A, Yoneyama T, Hashimoto I. Isolation of a temperature-sensitive strain of enterovirus 71 with reduced neurovirulence for monkeys. J Gen Virol 1982; 64: 499-502.

Hagiwara A, Yoneyama T, Hashimoto I. Isolation of a temperature-sensitive strain of enterovirus 71 with reduced neurovirulence for menkeys. J Gen Virol 1983; 64: 499-502. Hagiwara A, Yoneyama T, Hashimoto I. Isolation of a temperature-sensitive strain of enterovirus 71 with reduced neurovirulence for menkeys. J Gen Virol 1983; 64: 499-502.

Hall T. BioEdit: a user-friendly biological sequence alignment editor and analysis programme for Windows 95/98/NT. Nucleic Acids Symposium Research 1997; 41: 95-8. Hall T. BioEdit: a user-friendly biological sequence alignment editor and analysis programme for Windows 95/98/NT. Nucleic Acids Symposium Research 1997; 41: 95-8.

Hashimoto I, and Hagiwara A. Comparative studies on the neurovirulence of temperature-sensitive and temperature-resistant viruses of enterovirus 71 in monkeys. Acta Neuropathol 1983; 60: 266-270. Hashimoto I, and Hagiwara A. Comparative studies on the neurovirulence of temperature-sensitive and temperature-resistant viruses of enterovirus 71 in monkeys. Acta Neuropathol 1983; 60: 266-270.

Hayward J C, Gillespie S M, Kaplan K M, Packer R, Pallansch M, Plotkin S, et al. Outbreak of poliomyelitis-like paralysis associated with enterovirus 71. Pediatr Infect Dis 1989; 8: 611-616. Hayward J C, Gillespie S M, Kaplan K M, Packer R, Pallansch M, Plotkin S, et al. Outbreak of poliomyelitis-like paralysis associated with enterovirus 71. Pediatr Infect Dis 1989; 8: 611-616.

Huygelen C. The concept of virus attenuation in the eighteenth and early nineteenth centuries. Biologicals 1997; 25: 339-45. Huygelen C. The concept of virus attenuation in the eighteenth and early nineteenth centuries. Biologicals 1997; 25: 339-45.

Ishimaru Y, Nakano S, Yamaoka K, Takami S. Outbreaks of hand, foot, mouth disease by enterovirus 71: high incidence of complication disorders of central nervous system. Arch Dis Child 1980; 55: 583-588. Ishimaru Y, Nakano S, Yamaoka K, Takami S. Outbreaks of hand, foot, mouth disease by enterovirus 71: high incidence of complication disorders of central nervous system. Arch Dis Child 1980; 55: 583-588.

Kennett M, Birch C J, Lewis F A, Yung A P, Locarnin S A, Gust I D. Enterovirus type 71 infection in Melbourne. Bull EHO 1974; 51: 609-615. Kennett M, Birch C J, Lewis F A, Yung A P, Locarnin S A, Gust I D. Enterovirus type 71 infection in Melbourne. Bull EHO 1974; 51: 609-615.

Kenney JL, Volk SM, Pandya J, Wang E, Liang X, Weaver SC. Stability of RNA virus attenuation approaches. Vaccine 2011; 29(12): 2230-4. Doi: 10. 1016/j.vaccine.2011.01.055 Kenney JL, Volk SM, Pandya J, Wang E, Liang X, Weaver SC. Stability of RNA virus attenuation approaches. Vaccine 2011; 29(12): 2230-4. Doi: 10. 1016/j.vaccine.2011.01.055

Lauring AS, Jones JO, Andino R. Rationalizing the development of live attenuated virus vaccines. Nature Biotech 2010; 28(6): 573-9. Lauring AS, Jones JO, Andino R. Rationalizing the development of live attenuated virus vaccines. Nature Biotech 2010; 28(6): 573-9.

Lee MS, and Chang LY. Development of enterovirus 71 vaccines. Expert Rev Vaccines 2010; 9(2): 149-56. Lee MS, and Chang LY. Development of enterovirus 71 vaccines. Expert Rev Vaccines 2010; 9(2): 149-56.

Li Z, Yu M, Zhang H, Wang HY, Wang LF. Improved rapid amplification of cDNA ends (RACE) for mapping both the 5' and 3' terminal sequences of paramyxovirus genomes. J Virol Methods 2005; 130: 154-6. Li Z, Yu M, Zhang H, Wang HY, Wang LF. Improved rapid amplification of cDNA ends (RACE) for mapping both the 5'and 3'terminal sequences of paramyxovirus genomes. J Virol Methods 2005; 130: 154-6.

Liu C C, Tseng H W, Wang S M, Wang J R, Su I J. An outbreak of enteovirus 71 infection in Taiwan, 1998: epidemiologic and clinical manifestations. J Clin Virol 2000; 17: 23-30. Liu C C, Tseng H W, Wang S M, Wang J R, Su I J. An outbreak of enteovirus 71 infection in Taiwan, 1998: epidemiologic and clinical manifestations. J Clin Virol 2000; 17: 23-30.

Lum L C S, Lam S K, Wong K T, Chua K B, Goh A Y T. Neurologic pulmonary oedema and enterovirus 71 encephalomyelitis. Lancet 1998a; 352: 1391. Lum L C S, Lam S K, Wong K T, Chua K B, Goh A Y T. Neurologic pulmonary oedema and enterovirus 71 encephalomyelitis. Lancet 1998a; 352: 1391.

Lum L C S, Wong K T, Lam S K, Chua K B, Goh A Y T, Lim W L, Ong B B, AbuBakar S, Lambert M. Fatal Enterovirus 71 encephalitis. J Pediatr 1998b; 133: 795-798. Lum L C S, Wong K T, Lam S K, Chua K B, Goh A Y T, Lim W L, Ong B B, AbuBakar S, Lambert M. Fatal Enterovirus 71 encephalitis. J Pediatr 1998b; 133: 795-798.

Melnick J L. Enteroviruses: Polioviruses, Coxsackieviruses, Echoviruses, and Newer Enteroviruses. In: Fields B N, Knipe D M, Howley P M, et al. (3rd ed). Fields Virology. Philadelphia. Lippincott-Raven, 1996: 655-712. Melnick J L. Enteroviruses: Polioviruses, Coxsackieviruses, Echoviruses, and Newer Enteroviruses. In: Fields BN, Knipe DM, Howley PM, et al. (3 rd ed). Fields Virology. Philadelphia. Lippincott-Raven, 1996: 655-712 .

Nagy G, Takatsy S, Kukan E, Milialy I, Domok I. Virological diagnosis of enterovirus type 71 infections: experiences gained during an epidemic of acute CNS diseases in Hungary in 1978. Arch Virol 1982; 71: 217-227. Nagy G, Takatsy S, Kukan E, Milialy I, Domok I. Virological diagnosis of enterovirus type 71 infections: experiences gained during an epidemic of acute CNS diseases in Hungary in 1978. Arch Virol 1982; 71: 217-227.

Ong KC, Devi S, Cardosa MJ, wong KT. Formaldehyde-inactivated whole-virus vaccine protects a murine model of enterovirus 71 encephalomyelitis against disease. J Virol 2010; 84(1): 661-5. Ong KC, Devi S, Cardosa MJ, wong KT. Formaldehyde-inactivated whole-virus vaccine protects a murine model of enterovirus 71 encephalomyelitis against disease. J Virol 2010; 84(1): 661-5.

Reed LJ, and Muench H. A simple method of estimating fifty percent endpoints. Am J Hyg 1938; 27: 493-7. Reed LJ, and Muench H. A simple method of estimating fifty percent endpoints. Am J Hyg 1938; 27: 493-7.

Rice P, Lonogden I, Bleasby A. EMBOSS: the European Molecular Biology Open Software Suite. Trends Genetics 2000; 16(6): 276-7. Rice P, Lonogden I, Bleasby A. EMBOSS: the European Molecular Biology Open Software Suite. Trends Genetics 2000; 16(6): 276-7.

Richman DD, Murphy BR. The association of the temperature-sensitive phenotype with viral attenuation in animals and humans: Implications for development and use of live virus vaccines. Clin Infect Dis 1997; 1(3): 413-33. Richman DD, Murphy BR. The association of the temperature-sensitive phenotype with viral attenuation in animals and humans: Implications for development and use of live virus vaccines. Clin Infect Dis 1997; 1(3): 413-33.

Robinson HL. Viral attenuation by design. Nature Biotech 2008; 26: 1000-1001. Doi:10.1038/nbt0908-1000 Robinson HL. Viral attenuation by design. Nature Biotech 2008; 26: 1000-1001. Doi:10.1038/nbt0908-1000

Samuda G M, Chang W K, Yeung C Y, Tang P S. Monoplegia caused by enterovirus 71: an outbreak in Hong Kong. Pediatr Infect Dis 1987; 6: 206-208. Samuda G M, Chang W K, Yeung C Y, Tang P S. Monoplegia caused by enterovirus 71: an outbreak in Hong Kong. Pediatr Infect Dis 1987; 6: 206-208.

Schmidt N J, Lennette E H, Ho H H. An apparently new enterovirus isolated from patients with disease of the central nervous system. J Infect Dis 1974; 129: 304-309. Schmidt N J, Lennette E H, Ho H H. An apparently new enterovirus isolated from patients with disease of the central nervous system. J Infect Dis 1974; 129: 304-309.

Shindarov L M, Chumakov M P, Voroshilova M K, Bojinov S, Vasilenko S M, Jordanov I, et al. Epidemiological, clinical and pathomorphological characteristics of epidemic poliomyelitis-like disease caused by enterovirus 71. J Hyg Epid Microbiol Immun 1979; 3: 284-295. Shindarov L M, Chumakov M P, Voroshilova M K, Bojinov S, Vasilenko S M, Jordanov I, et al. Epidemiological, clinical and pathomorphological characteristics of epidemic poliomyelitis-like disease caused by enterovirus 71. J Hyg Epid Microbiol Immun 1979; 3: 284-295.

Tagaya I, Takayama R, Hagiwara A. A large scale epidemic of hand, foot, mouth disease associated with enterovirus 71 infection in Japan in 1978. Jpn J Med Sci Biol 1981; 34: 191-196. Tagaya I, Takayama R, Hagiwara A. A large scale epidemic of hand, foot, mouth disease associated with enterovirus 71 infection in Japan in 1978. Jpn J Med Sci Biol 1981; 34: 191-196.

Tung WS, Bakar SA, Sekawi Z, Rosli R. DNA vaccine constructs against enterovirus 71 elicit immune response in mice. Genet Vaccines Ther 2007; 5:6. Tung WS, Bakar SA, Sekawi Z, Rosli R. DNA vaccine constructs against enterovirus 71 elicit immune response in mice. Genet Vaccines Ther 2007; 5:6.

Wang I R, Tsai H P, Chen P F, Lai Y J, Yan J J, Kiang D, et al. An outbreak of enteovirus 71 infection in Taiwan, 1998. II. Laboratory diagnosis and genetic analysis. J Clin Virol 2000; 17: 91-99. Wang IR, Tsai HP, Chen PF, Lai YJ, Yan JJ, Kiang D, et al. An outbreak of enteovirus 71 infection in Taiwan, 1998. II. Laboratory diagnosis and genetic analysis. J Clin Virol 2000; 17: 91-99 .

Wu CN, Lin YC, Fann C, Liao NS, Shih SR, Ho MS. Protection against lethal enterovirus 71 infection in newborn mice by passive immunization with subunit VP1 vaccines and inactivated virus. Vaccine 2001; 20(5-6): 895-904. Wu CN, Lin YC, Fann C, Liao NS, Shih SR, Ho MS. Protection against lethal enterovirus 71 infection in newborn mice by passive immunization with subunit VP1 vaccines and inactivated virus. Vaccine 2001; 20(5-6): 895- 904.

Yan J J, Wang J R, Liu C C, Yang H B, Su I J. An outbreak of enterovirus 71 infection in Taiwan 1998: A comprehensive pathological, virological, and molecular study on a case of fulminant encephalitis. J Clin Virol 2000; 17: 13-22. Yan JJ, Wang JR, Liu CC, Yang HB, Su I J. An outbreak of enterovirus 71 infection in Taiwan 1998: A comprehensive pathological, virological, and molecular study on a case of fulminant encephalitis. J Clin Virol 2000; 17: 13 -twenty two.

Yang F, Zhng T, Hu Y, Wang X, Du J, Li Y Sun S, Sun X, Li Z, Jin Q. Survey of enterovirus infections from hand, foot and mouth disease outbreak in China, 2009. Virology J 2011; 8:508. http://www.virologyj.com/content/8/1/508. Yang F, Zhng T, Hu Y, Wang X, Du J, Li Y Sun S, Sun X, Li Z, Jin Q. Survey of enterovirus infections from hand, foot and mouth disease outbreak in China, 2009. Virology J 2011; 8:508. http://www.virologyj.com/content/8/1/508.

Zeng M, El Khatib NF, Tu S, Ren P, Xu S, Zhu Q, Mo X, Pu D, Wang X, Altmeyer R. Seroepidemiology of enterovirus 71 infection prior to the 2011 season in children in Shanghai. J Clin Virol 2012; 53(4): 285-9. Zeng M, El Khatib NF, Tu S, Ren P, Xu S, Zhu Q, Mo X, Pu D, Wang X, Altmeyer R. Seroepidemiology of enterovirus 71 infection prior to the 2011 season in children in Shanghai. J Clin Virol 2012 ; 53(4): 285-9.

Zhang D, Lu J, Lu J. Enterovirus 71 vaccine: close but still far. Int J Infect Dis 2010; 14(9): e739-43. Zhang D, Lu J, Lu J. Enterovirus 71 vaccine: close but still far. Int J Infect Dis 2010; 14(9): e739-43.

【生物材料寄存】 【Biological Material Deposit】 國內寄存資訊【請依寄存機構、日期、號碼順序註記】 Domestic deposit information [please note in order of deposit institution, date and number]

1. 食品工業發展研究所;103年05月08日;BCRC970063 1. Food Industry Development Research Institute; May 08, 2013; BCRC970063

2. 食品工業發展研究所;103年05月08日;BCRC970064 2. Food Industry Development Research Institute; May 08, 2013; BCRC970064

國外寄存資訊【請依寄存國家、機構、日期、號碼順序註記】 Foreign hosting information [please note in the order of hosting country, institution, date and number]

1. 美國;American Type Culture Collection(ATCC);2012年10月25日;PTA-13284 1. United States; American Type Culture Collection (ATCC); October 25, 2012; PTA-13284

2. 美國;American Type Culture Collection(ATCC);2012年10月25日;PTA-13285 2. United States; American Type Culture Collection (ATCC); October 25, 2012; PTA-13285

<110> 新加坡商淡馬錫生命科學研究院公司 <110> Singapore Business Temasek Institute of Life Sciences Corporation

<120> 適於冷之腸病毒71溫度敏感株及發展適於冷之溫度 敏感病毒株之方法 <120> Enterovirus 71 temperature-sensitive strain suitable for cold and development suitable for cold temperature Sensitive virus strain method

<130> 2577-224PCT <130> 2577-224PCT

<160> 7 <160> 7

<170> PatentIn version 3.5 <170> PatentIn version 3.5

<210> 1 <210> 1

<211> 29 <211> 29

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 寡核苷酸銜接子 <223> Oligonucleotide adaptor

<220> <220>

<221> misc_feature <221> misc_feature

<222> (29)..(29) <222> (29)..(29)

<223> n為蛹蟲草菌素 <223> n is cordycepin

<400> 1

Figure 103101883-A0202-12-0043-9
<400> 1
Figure 103101883-A0202-12-0043-9

<210> 2 <210> 2

<211> 27 <211> 27

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 寡核苷酸引子 <223> Oligonucleotide primer

<400> 2

Figure 103101883-A0202-12-0043-10
<400> 2
Figure 103101883-A0202-12-0043-10

<210> 3 <210> 3

<211> 21 <211> 21

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 寡核苷酸引子 <223> Oligonucleotide primer

<400> 3

Figure 103101883-A0202-12-0043-11
<400> 3
Figure 103101883-A0202-12-0043-11

<210> 4 <210> 4

<211> 24 <211> 24

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 寡核苷酸引子 <223> Oligonucleotide primer

<400> 4

Figure 103101883-A0202-12-0044-13
<400> 4
Figure 103101883-A0202-12-0044-13

<210> 5 <210> 5

<211> 24 <211> 24

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 寡核苷酸引子 <223> Oligonucleotide primer

<400> 5

Figure 103101883-A0202-12-0044-15
<400> 5
Figure 103101883-A0202-12-0044-15

<210> 6 <210> 6

<211> 7428 <211> 7428

<212> DNA <212> DNA

<213> 腸病毒71,病毒株EV71:TLLαP20 <213> Enterovirus 71, virus strain EV71: TLLαP20

<400> 6

Figure 103101883-A0202-12-0044-16
Figure 103101883-A0202-12-0045-17
Figure 103101883-A0202-12-0046-18
Figure 103101883-A0202-12-0047-19
Figure 103101883-A0202-12-0048-20
<400> 6
Figure 103101883-A0202-12-0044-16
Figure 103101883-A0202-12-0045-17
Figure 103101883-A0202-12-0046-18
Figure 103101883-A0202-12-0047-19
Figure 103101883-A0202-12-0048-20

<210> 7 <210> 7

<211> 7437 <211> 7437

<212> DNA <212> DNA

<213> 腸病毒71,病毒株EV71:TLLβP20 <213> Enterovirus 71, virus strain EV71: TLLβP20

<400> 7

Figure 103101883-A0202-12-0048-21
Figure 103101883-A0202-12-0049-22
Figure 103101883-A0202-12-0050-23
Figure 103101883-A0202-12-0051-24
Figure 103101883-A0202-12-0052-25
<400> 7
Figure 103101883-A0202-12-0048-21
Figure 103101883-A0202-12-0049-22
Figure 103101883-A0202-12-0050-23
Figure 103101883-A0202-12-0051-24
Figure 103101883-A0202-12-0052-25

Claims (19)

一種適於冷之腸病毒71溫度敏感株,其包含SEQ ID NO:6或7所示之核苷酸序列。 A temperature-sensitive strain of Enterovirus 71 suitable for cold, which comprises the nucleotide sequence shown in SEQ ID NO: 6 or 7. 如請求項1之適於冷之腸病毒71溫度敏感株,其用於引發個體中針對腸病毒71之保護性免疫反應。 The temperature-sensitive strain of Enterovirus 71 suitable for cold as in Claim 1, which is used to elicit a protective immune response against Enterovirus 71 in individuals. 如請求項1之適於冷之腸病毒71溫度敏感株,其用於阻止個體罹患與腸病毒71相關之疾病。 For example, the temperature sensitive strain of Enterovirus 71 suitable for cold in Claim 1, which is used to prevent individuals from suffering from diseases related to Enterovirus 71. 如請求項1之適於冷之腸病毒71溫度敏感株,其用於在感染腸病毒71之個體中延遲與腸病毒71相關之疾病之發作或減慢該疾病之速率。 For example, the temperature-sensitive strain of Enterovirus 71 suitable for cold in Claim 1, which is used to delay the onset of the disease related to Enterovirus 71 or slow down the rate of the disease in individuals infected with Enterovirus 71. 如請求項1之適於冷之腸病毒71溫度敏感株,其係用於疫苗發展。 For example, the cold-suitable enterovirus 71 temperature-sensitive strain of claim 1, which is used for vaccine development. 一種組合物,其包含如請求項1之適於冷之腸病毒71溫度敏感株。 A composition comprising the enterovirus 71 temperature-sensitive strain suitable for cold as claimed in claim 1. 如請求項6之組合物,其進一步包含醫藥學上可接受之載劑。 The composition of claim 6, which further comprises a pharmaceutically acceptable carrier. 如請求項6之組合物,其進一步包含佐劑。 The composition of claim 6, which further comprises an adjuvant. 如請求項6至8中任一項之組合物,其為疫苗。 The composition according to any one of claims 6 to 8, which is a vaccine. 如請求項6至8中任一項之組合物,其用於引發個體中針對腸病毒71之保護性免疫反應。 The composition according to any one of claims 6 to 8, which is used to elicit a protective immune response against enterovirus 71 in an individual. 如請求項6至8中任一項之組合物,其用於阻止個體罹患與腸病毒71相關之疾病。 The composition according to any one of claims 6 to 8, which is used to prevent an individual from suffering from diseases related to Enterovirus 71. 如請求項6至8中任一項之組合物,其用於在感染腸病毒71之個體中延遲與腸病毒71相關之疾病之發作或減慢該疾病之速率。 The composition according to any one of claims 6 to 8, which is used to delay the onset of a disease related to the enterovirus 71 or to slow down the rate of the disease in an individual infected with the enterovirus 71. 一種如請求項1之適於冷之腸病毒71溫度敏感株或如請求項6至9中任一項之組合物之用途,其用於製造引發個體中針對腸病毒 71之保護性免疫反應之藥劑。 A use of a temperature-sensitive strain of Enterovirus 71 suitable for cold as claimed in claim 1 or a composition as claimed in any one of claims 6 to 9 for the production of an individual against enterovirus 71 of the protective immune response agent. 一種如請求項1之適於冷之腸病毒71溫度敏感株或如請求項6至9中任一項之組合物之用途,其用於製造阻止個體罹患與腸病毒71相關之疾病之藥劑。 A use of the temperature-sensitive strain of Enterovirus 71 suitable for cold as claimed in claim 1 or the composition as claimed in any one of claims 6 to 9 for the manufacture of an agent for preventing an individual from suffering from diseases related to enterovirus 71. 一種如請求項1之適於冷之腸病毒71溫度敏感株或如請求項6至9中任一項之組合物之用途,其用於製造在感染腸病毒71之個體中延遲與腸病毒71相關之疾病之發作或減慢該疾病之速率之藥劑。 A use of a temperature-sensitive strain of Enterovirus 71 suitable for cold as claimed in claim 1 or a composition as claimed in any one of claims 6 to 9 for the production of delayed and enterovirus 71 in individuals infected with enterovirus 71 The onset of related diseases or agents that slow down the rate of the disease. 如請求項13至15中任一項之用途,其中該個體為人類個體。 Such as the use of any one of claims 13 to 15, wherein the individual is a human individual. 一種藉由適於冷之腸病毒71溫度敏感株用於個體免疫之套組,包含如請求項1之適於冷之腸病毒71溫度敏感株或如請求項6至9中任一項之組合物、醫藥學上可接受之載劑及其使用指導材料。 A kit for individual immunization by a temperature sensitive strain of Enterovirus 71 suitable for cold, comprising the temperature sensitive strain of Enterovirus 71 suitable for cold as claimed in claim 1 or a combination of any one of claims 6 to 9 Materials, pharmaceutically acceptable carriers and guidance materials for their use. 如請求項17之套組,其進一步包含施用器。 Such as the set of claim 17, which further comprises an applicator. 一種由SEQ ID NO:6或SEQ ID NO:7所示核苷酸序列所組成之核酸,其係用於疫苗發展。 A nucleic acid composed of the nucleotide sequence shown in SEQ ID NO: 6 or SEQ ID NO: 7, which is used for vaccine development.
TW103101883A 2013-01-18 2014-01-17 Cold-adapted temperature sensitive strains of enterovirus 71 and processes of developing cold-adapted temperature sensitive virus strains TWI705140B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/SG2013/000027 WO2014112945A1 (en) 2013-01-18 2013-01-18 Cold-adapted temperature sensitive strains of enterovirus 71 and processes of developing cold-adapted temperature sensitive virus strains
WOPCT/SG2013/000027 2013-01-18

Publications (2)

Publication Number Publication Date
TW201522642A TW201522642A (en) 2015-06-16
TWI705140B true TWI705140B (en) 2020-09-21

Family

ID=51209913

Family Applications (1)

Application Number Title Priority Date Filing Date
TW103101883A TWI705140B (en) 2013-01-18 2014-01-17 Cold-adapted temperature sensitive strains of enterovirus 71 and processes of developing cold-adapted temperature sensitive virus strains

Country Status (7)

Country Link
JP (1) JP6117379B2 (en)
CN (1) CN104918636B (en)
HK (1) HK1213474A1 (en)
MY (1) MY185184A (en)
SG (1) SG11201505468VA (en)
TW (1) TWI705140B (en)
WO (1) WO2014112945A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015190999A1 (en) * 2014-06-12 2015-12-17 Temasek Life Sciences Laboratory Limited Development of stable cold-adapted temperature sensitive chimeric enteroviruses
AR102548A1 (en) 2014-11-07 2017-03-08 Takeda Vaccines Inc VACCINES AGAINST HAND, FEET AND MOUTH DISEASE AND MANUFACTURING AND USE METHODS
MA40920A (en) 2014-11-07 2017-09-12 Takeda Vaccines Inc VACCINES OF THE HAND, FOOT AND MOUTH, AND METHODS OF MANUFACTURING AND USE THEREOF
MA41506A (en) * 2015-02-13 2017-12-19 Takeda Vaccines Inc VIRUS PRODUCTION PROCESSES TO PRODUCE VACCINES
CN109504662B (en) * 2018-11-09 2022-06-10 浙江天元生物药业有限公司 Coxsackie virus attenuated strain and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7150984B2 (en) * 1994-07-11 2006-12-19 The United States Of America As Represented By The Department Of Health And Human Services Attenuated human rotavirus vaccine
CN1314940A (en) * 1998-09-01 2001-09-26 圣路易斯大学 Temperature-sensitive and cold-adapted human parainfluenza virus type 2(HPIV-2) and vaccines based on such virus

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
1983年,Isolation of a temperature-sensitive strain of enterovirus 71 with reduced neurovirulence for monkeys,Hagiwara A,J Gen Virol. 1983 Feb;64 (Pt 2):499-502.
1983年,Isolation of a temperature-sensitive strain of enterovirus 71 with reduced neurovirulence for monkeys,Hagiwara A,J Gen Virol. 1983 Feb;64 (Pt 2):499-502. 2005年05月,Temperature-sensitive mutants of enterovirus 71 show attenuation in cynomolgus monkeys,Arita M,J Gen Virol. 2005 May;86(Pt 5):1391-401. 2007年12月05日, Cooperative effect of the attenuation determinants derived from poliovirus sabin 1 strain is essential for attenuation of enterovirus 71 in the NOD/SCID mouse infection model,Arita M,J Virol. 2008 Feb;82(4):1787-97. 1985年12月,Development and characterization of cold-adapted viruses for use as live virus vaccines,Maassab HF,Vaccine. 1985 Dec;3(5):355-69. *
1985年12月,Development and characterization of cold-adapted viruses for use as live virus vaccines,Maassab HF,Vaccine. 1985 Dec;3(5):355-69.
2005年05月,Temperature-sensitive mutants of enterovirus 71 show attenuation in cynomolgus monkeys,Arita M,J Gen Virol. 2005 May;86(Pt 5):1391-401.
2007年12月05日, Cooperative effect of the attenuation determinants derived from poliovirus sabin 1 strain is essential for attenuation of enterovirus 71 in the NOD/SCID mouse infection model,Arita M,J Virol. 2008 Feb;82(4):1787-97.

Also Published As

Publication number Publication date
MY185184A (en) 2021-04-30
HK1213474A1 (en) 2016-07-08
SG11201505468VA (en) 2015-08-28
WO2014112945A1 (en) 2014-07-24
JP2016511634A (en) 2016-04-21
CN104918636B (en) 2018-06-26
TW201522642A (en) 2015-06-16
JP6117379B2 (en) 2017-04-19
CN104918636A (en) 2015-09-16

Similar Documents

Publication Publication Date Title
McMinn Recent advances in the molecular epidemiology and control of human enterovirus 71 infection
Arita et al. An attenuated strain of enterovirus 71 belonging to genotype a showed a broad spectrum of antigenicity with attenuated neurovirulence in cynomolgus monkeys
Bank-Wolf et al. Zoonotic aspects of infections with noroviruses and sapoviruses
TWI705140B (en) Cold-adapted temperature sensitive strains of enterovirus 71 and processes of developing cold-adapted temperature sensitive virus strains
Chang et al. Selection and characterization of vaccine strain for Enterovirus 71 vaccine development
Liu et al. Combined peptides of human enterovirus 71 protect against virus infection in mice
KR102626270B1 (en) Compositions and methods for dengue virus chimeric constructs in vaccines
CN113564130B (en) Coxsackie virus A10 type strain and application thereof
TWI685566B (en) Development of stable cold-adapted temperature sensitive chimeric enteroviruses
CN111073862B (en) Bovine viral diarrhea type2 attenuated strain and application thereof
TWI638048B (en) High-growth enterovirus 71 strains and vaccines
CN105802919B (en) Highly pathogenic porcine reproductive and respiratory syndrome virus, vaccine and application thereof
Chua et al. Safety and Immunogenicity of a Stable, Cold-Adapted, Temperature-Sensitive/Conditional Lethal Enterovirus A71 in Monkey Study. Viruses 2021, 13, 438
Kanno et al. Viruses produced from complementary DNA of virulent and avirulent strains of swine vesicular disease viruses retain the in vivo and in vitro characteristics of the parental strain
CN113462656B (en) Human three-type parainfluenza virus cold-adaptation temperature-sensitive strain and application thereof
El-Nahas et al. A genotyping of a new avian infectious bronchitis virus isolated from chickens proventriculus in Egypt
JP7232289B2 (en) Live viruses banked from dengue virus attenuated strains, and dengue vaccines using them as antigens
Pallansch et al. Enteroviruses and parechoviruses
Sata et al. An Attenuated Strain of Enterovirus 71
Ishmukhametov et al. The Development of Polio Vaccines: the Current Update
Zhang et al. Screening of a new candidate coxsackievirus B1 vaccine strain based on its biological characteristics
Yee Rational design of live attenuated vaccine by site-directed mutagenesis and deletion in the 5’-non coding region of the enterovirus 71 (EV-A71) genome
Nik Nadia Seroepidemiology and characterisation of neutralisation escape mutants of enterovirus A71/Nik Nadia Nik Mohd Nasir
Nasir Seroepidemiology and Characterisation of Neutralisation Escape Mutants of Enterovirus A71
Chezzi Dynamics of Genomic Variation in Poliovirus in Africa