TW201210611A - Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT - Google Patents

Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT Download PDF

Info

Publication number
TW201210611A
TW201210611A TW100129854A TW100129854A TW201210611A TW 201210611 A TW201210611 A TW 201210611A TW 100129854 A TW100129854 A TW 100129854A TW 100129854 A TW100129854 A TW 100129854A TW 201210611 A TW201210611 A TW 201210611A
Authority
TW
Taiwan
Prior art keywords
nampt
antisense
seq
polynucleotide
oligonucleotide
Prior art date
Application number
TW100129854A
Other languages
Chinese (zh)
Inventor
Joseph Collard
Sherman Olga Khorkova
Carlos Coito
Original Assignee
Opko Curna Llc
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 Opko Curna Llc filed Critical Opko Curna Llc
Publication of TW201210611A publication Critical patent/TW201210611A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3525MOE, methoxyethoxy

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Plant Pathology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Public Health (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to antisense oligonucleotides that modulate the expression of and/or function of Nicotinamide Phosphoribosyltransferase (NAMPT), in particular, by targeting natural antisense polynucleotides of Nicotinamide Phosphoribosyltransferase (NAMPT). The invention also relates to the identification of these antisense oligonucleotides and their use in treating diseases and disorders associated with the expression of NAMPT.

Description

201210611 六、發明說明: 【發明所屬之技術領域】 本發明之實施例包含調節NAMPT及相關分子之表現及/ 或功能的寡核苷酸。 • 本申請案主張2010年8月19曰申請之美國臨時專利申請 ^ 案第61/375,126號之優先權,該文獻以全文引用的方式併 入本文中。 【先前技術】 0 DNA-RNA及RNA-RNA雜交對包括DNA複製、轉錄及轉 譯之核酸功能之許多方面均重要。雜交亦為偵測特定核酸 或改變其表現之多種技術之中心。反義核苷酸例如藉由與 目標RNA雜交而破壞基因表現,由此干擾RNA剪接、轉 錄、轉譯及複製。反義DNA具有如下額外特徵:DNA-RNA雜交物充當核糖核酸酶Η消化之受質,亦即一種存在 於大多數細胞類型中之活性。反義分子可傳遞至細胞中, 該傳遞與寡聚去氧核苷酸之情況相同,或其可自内源性基 〇 因表現為RNA分子。FDA近來批准了 一種反義藥物 VITRAVENE™(用於治療細胞巨大病毒視網膜炎),反映出 ' 反義序列具有治療效用。 - 【發明内容】 提供本發明内容以呈示簡要說明本發明之性質及物質的 本發明概述。提出條件為其不用以解釋或限制申請專利範 圍之範疇或含義。 在一實施例中,本發明提供藉由使用靶向天然反義轉錄 158213.doc 201210611 物之任何區之反義寡核苷酸來抑制天然反義轉錄物之作 用’從而引起相應有義基因上調的方法。本文亦涵蓋抑制 天然反義轉錄物可藉由siRNA、核糖核酸酶及小分子來達 成’認為其在本發明之範蜂内。 一實施例提供一種活體内或活體外調節生物系統中(包 括(但不限於)患者細胞或組織中)NAMPT聚核苷酸之功能 及/或表現之方法,該方法包含使該生物系統或該等細胞 或組織與長度為約5至約3 0個核苷酸之反義寡核苷酸接 觸,其中該寡核苷酸與包含SEQ ID NO: 2之核苷酸1至 882、或 SEQ ID NO: 3之 1 至 915、或 SEQ ID NO: 4之 1 至 165、或 SEQ ID NO: 5之 1 至 533、或 SEQ ID NO: 6之 1 至 522、或 SEQ ID NO: 7之 1 至 141、或 SEQ ID NO: 8之 1 至 519、或 SEQ ID NO: 9之 1 至 374、或 SEQ ID NO: 10之 1 至 240、或 SEQ ID NO: 11之 1至 353、或 SEQ ID NO: 12之 1至 534、或 SEQ ID NO: 13之 1 至 959、或 SEQ ID NO: 14之 1 至 127、或 SEQ ID NO: 15之 1 至 232、或 SEQ ID NO: 16之 1 至 3 87内5至30個連續核苷酸之聚核苷酸的反向互補序列具有 至少50%序列一致性,由此活體内或活體外調節該生物系 統(包括該等患者細胞或組織)中NAMPT聚核苷酸之功能及/ 或表現。 在一實施例中,寡核苷酸靶向生物系統中所存在之 NAMPT聚核苷酸(例如SEQ ID NO: 2至16中所示之核苷 酸,及其任何變異體、對偶基因、同源物、突變體、衍生 物、片段及互補序列)之天然反義序列。反義募核苷酸之 158213.doc 201210611 實例如SEQ ID NO: 17至31中所示。 另一實施例提供一種活體内或活體外調節患者細胞或組 織中ΝΑΜΡΤ聚核苷酸之功能及/或表現之方法,該方法包 含使該等細胞或組織與長度為5至3〇個核苷酸之反義募核 苷酸接觸,其中該寡核苷酸與ΝΑΜΡΤ聚核苷酸之反義序 列之反向互補序列具有至少50%序列一致性;由此活體内 或活體外調節患者細胞或組織中ΝΑΜΡΤ聚核苷酸之功能 及/或表現。 另一實施例提供一種活體内或活體外調節患者細胞或組 織中ΝΑΜΡΤ聚核音酸之功能及/或表現之方法,該方法包 含使該等細胞或組織與長度為5至30個核苷酸之反義寡核 苷酸接觸’其中該募核苷酸與ΝΑΜΡΤ反義聚核苷酸之反 義募核脊酸具有至少50%序列一致性;由此活體内或活體 外調節患者細胞或組織中ΝΑΜΡΤ聚核苷酸之功能及/或表 現。 在另一實施例中’本發明包含一種調節生物系統中 ΝΑΜΡΤ聚核普酸之功能及/或表現之方法,該方法包含使 該生物系統與靶向ΝΑΜΡΤ聚核苷酸之天然反義轉錄物的 至少一種反義募核苷酸接觸,由此調節該生物系統中 ΝΑΜΡΤ聚核苷酸之功能及/或表現。 在另一實施例中’本發明包含一種調節生物系統中 ΝΑΜΡΤ聚核苷酸之功能及/或表現之方法,該方法包含使 該生物系統與靶向ΝΑΜΡΤ聚核苷酸之天然反義轉錄物之 區的至少一種反義寡核苷酸接觸,由此調節該生物系統中 158213.doc 201210611 NAMPT聚核苷酸之功能及/或表現。 在一實施例中,本發明包含一種增加生物系統中具有 SEQ ID NO. 1之NAMPT聚核苷酸之功能及/或表現之方 法,該方法包含使該生物系統與靶向該NAMPT聚核苷酸 之天然反義轉錄物的至少一種反義寡核苷酸接觸,由此增 加該NAMPT聚核苷酸或其表現產物之功能及/或表現。 在另一實施例中,本發明包含一種增加生物系統中具有 SEQ ID NO. 1之NAMPT聚核苷酸之功能及/或表現之方 法,該方法包含使該生物系統與靶向該NAMPT聚核苷酸 之天然反義轉錄物的至少一種反義寡核苷酸接觸,由此增 加該NAMPT聚核苷酸或其表現產物之功能及/或表現,其 中該等天然反義轉錄物係選自SEQ ID NO. 2至16。 在另一實施例中,本發明包含一種增加生物系統中具有 SEQ ID NO. 1之NAMPT聚核苷酸之功能及/或表現之方 法,該方法包含使該生物系統與靶向該NAMPT聚核苷酸 之天然反義轉錄物的至少一種反義寡核苦酸接觸,由此增 加該NAMPT聚核苷酸或其表現產物之功能及/或表現,其 中該等天然反義轉錄物係選自SEQ ID NO. 2至16且其中該 等反義寡核苷酸係選自SEQ ID NO. 17至31中之至少一 者。 在一實施例中,組合物包含一或多種結合有義及/或反 義NAMPT聚核苷酸之反義寡核苷酸。 在一實施例中,寡核苷酸包含一或多種經修飾或經取代 之核苷酸。 158213.doc 201210611 在一實施例中,募核苷酸包含一或多個經修飾之鍵。 在又一實施例中,經修飾之核苷酸包含經修飾鹼基,其 包含硫代磷酸酯、曱基膦酸酯、肽核酸、2’-〇-曱基、氟· 或碳、亞甲基或其他鎖核酸(LNA)分子。經修飾之核苷酸 較佳為鎖核酸分子,包括α-L-LNA。 在一實施例中,寡核苷酸皮下、肌肉内、靜脈内或腹膜 内投與患者。201210611 VI. Description of the Invention: [Technical Field of the Invention] Embodiments of the invention include oligonucleotides that modulate the expression and/or function of NAMPT and related molecules. • The present application claims priority to U.S. Provisional Patent Application Serial No. 61/375,126, filed on Jan. 19, 2010, which is hereby incorporated by reference. [Prior Art] 0 DNA-RNA and RNA-RNA hybridization are important for many aspects of nucleic acid function including DNA replication, transcription and translation. Hybridization is also central to a variety of techniques for detecting specific nucleic acids or altering their performance. Antisense nucleotides disrupt gene expression, e.g., by hybridization to a target RNA, thereby interfering with RNA splicing, transcription, translation, and replication. Antisense DNA has the additional feature that the DNA-RNA hybrid acts as a substrate for ribonuclease digestion, i.e., an activity that is present in most cell types. The antisense molecule can be delivered to the cell in the same manner as the oligodeoxynucleotide, or it can be expressed as an RNA molecule from the endogenous gene. The FDA recently approved an antisense drug, VITRAVENETM (for the treatment of giant viral retinitis), reflecting the anti-sense sequence has therapeutic utility. - SUMMARY OF THE INVENTION The summary is provided to present a summary of the invention. The conditions are set out so that they do not have to explain or limit the scope or meaning of the scope of the patent application. In one embodiment, the invention provides for the up-regulation of a corresponding sense gene by inhibiting the action of a natural antisense transcript by using an antisense oligonucleotide that targets any region of natural antisense transcription 158213.doc 201210611 Methods. It is also contemplated herein that inhibition of a natural antisense transcript can be achieved by siRNA, ribonuclease, and small molecules, which are considered to be within the present invention. An embodiment provides a method of modulating the function and/or performance of a NAMPT polynucleotide in a biological system, including but not limited to a patient cell or tissue, in vivo or in vitro, the method comprising: The cell or tissue is contacted with an antisense oligonucleotide having a length of from about 5 to about 30 nucleotides, wherein the oligonucleotide comprises nucleotides 1 to 882 of SEQ ID NO: 2, or SEQ ID NO: 3 to 915, or 1 to 165 of SEQ ID NO: 4, or 1 to 533 of SEQ ID NO: 5, or 1 to 522 of SEQ ID NO: 6, or 1 to SEQ ID NO: 7 to 141, or 1 to 519 of SEQ ID NO: 8, or 1 to 374 of SEQ ID NO: 9, or 1 to 240 of SEQ ID NO: 10, or 1 to 353 of SEQ ID NO: 11, or SEQ ID NO : 12 to 1 to 534, or SEQ ID NO: 13 to 1 to 959, or SEQ ID NO: 14 to 1 to 127, or SEQ ID NO: 15 to 1 to 232, or SEQ ID NO: 16 to 1 to 3 The reverse complement of a polynucleotide of 5 to 30 contiguous nucleotides within 87 has at least 50% sequence identity, thereby modulating the biological system (including such patient cells or tissues) in vivo or ex vivo. The function and/or performance of a NAMPT polynucleotide. In one embodiment, the oligonucleotide targets a NAMPT polynucleotide present in a biological system (eg, the nucleotides set forth in SEQ ID NOS: 2 to 16, and any variants thereof, dual genes, Natural antisense sequences of the source, mutant, derivative, fragment and complementary sequence). Antisense nucleotides 158213.doc 201210611 Examples are shown in SEQ ID NOs: 17 to 31. Another embodiment provides a method of modulating the function and/or expression of purine nucleotides in a patient's cells or tissues in vivo or in vitro, the method comprising affixing the cells or tissues to 5 to 3 nucleosides in length An antisense nucleotide contact of an acid, wherein the oligonucleotide has at least 50% sequence identity to a reverse complement of the antisense sequence of the purine polynucleotide; thereby modulating the patient cell or in vivo or in vitro The function and/or performance of purine nucleotides in tissues. Another embodiment provides a method of modulating the function and/or expression of sputum polynucleic acid in a cell or tissue of a patient in vivo or in vitro, the method comprising affixing the cell or tissue to a length of 5 to 30 nucleotides The antisense oligonucleotide contacts 'where the nucleotide has at least 50% sequence identity with the antisense nucleocapsid nucleoside of the antisense polynucleotide; thereby regulating the patient cell or tissue in vivo or ex vivo The function and/or performance of a ruthenium polynucleotide. In another embodiment, the invention encompasses a method of modulating the function and/or expression of a ruthenium polynucleotide in a biological system, the method comprising the natural antisense transcript of the biological system and a targeting ΝΑΜΡΤ polynucleotide At least one antisense raised nucleotide contacts, thereby modulating the function and/or performance of the purine polynucleotide in the biological system. In another embodiment, the invention encompasses a method of modulating the function and/or expression of a purine polynucleotide in a biological system, the method comprising the biological system and a natural antisense transcript targeting a purine polynucleotide At least one antisense oligonucleotide in the region contacts, thereby modulating the function and/or performance of the 158213.doc 201210611 NAMPT polynucleotide in the biological system. In one embodiment, the invention comprises a method of increasing the function and/or expression of a NAMPT polynucleotide having SEQ ID NO. 1 in a biological system, the method comprising targeting the biological system to the NAMPT polynucleoside Contact with at least one antisense oligonucleotide of the acid natural antisense transcript, thereby increasing the function and/or expression of the NAMPT polynucleotide or its expression product. In another embodiment, the invention comprises a method of increasing the function and/or expression of a NAMPT polynucleotide having SEQ ID NO. 1 in a biological system, the method comprising targeting the biological system to the NAMPT polynucleus Contacting at least one antisense oligonucleotide of a natural antisense transcript of a gluconate, thereby increasing the function and/or expression of the NAMPT polynucleotide or its expression product, wherein the natural antisense transcript is selected from SEQ ID NOS. 2 to 16. In another embodiment, the invention comprises a method of increasing the function and/or expression of a NAMPT polynucleotide having SEQ ID NO. 1 in a biological system, the method comprising targeting the biological system to the NAMPT polynucleus Contacting at least one antisense oligonucleotide of a natural antisense transcript of a gluconate, thereby increasing the function and/or expression of the NAMPT polynucleotide or its expression product, wherein the natural antisense transcript is selected from SEQ ID NOS. 2 to 16 and wherein the antisense oligonucleotides are selected from at least one of SEQ ID NOS. 17 to 31. In one embodiment, the composition comprises one or more antisense oligonucleotides that bind to a sense and/or antisense NAMPT polynucleotide. In one embodiment, the oligonucleotide comprises one or more modified or substituted nucleotides. 158213.doc 201210611 In one embodiment, the raised nucleotide comprises one or more modified linkages. In yet another embodiment, the modified nucleotide comprises a modified base comprising a phosphorothioate, a decylphosphonate, a peptide nucleic acid, a 2'-fluorene-fluorenyl group, a fluorine or carbon, a methylene A base or other locked nucleic acid (LNA) molecule. The modified nucleotide is preferably a locked nucleic acid molecule, including α-L-LNA. In one embodiment, the oligonucleotide is administered to the patient subcutaneously, intramuscularly, intravenously or intraperitoneally.

在一實施例中’寡核苷酸以醫藥組合物投與。治療方案 包含向患者投與反義化合物至少一次;然而此治療可修改 為包括在一段時間内多次給藥。該治療可與一或多種其他 類型之療法組合。 在一實施例中,寡核苷酸囊封於脂質體中或與載體分子 (例如膽固醇、丁AT肽)連接。 下文描述其他態樣。 【實施方式】 出於說明之目的,下文參考實例應用描述本發明之若干 態樣。應瞭解,㈣許多特定詳情、關係及方法以充分理 解本發明。然而’一般熟悉相關技術者容易認識到,可在 無y或多種特定詳情或有其他方法下實施本發明。本發明 不受操作或事件之排序限制’此係因為-些操作與其他操 作或事件可以不同次序及/或同時發生。此外,並不需要 所有所說明m事件來實施本發明之方法。 來之所有基因、基因名稱及基因產物均意欲與 本文所揭示之組合物及方法之任何物種的同源物 1582l3.doc 201210611 相對應。因此,該等術語包括(但不限於)來自人類及小鼠 ^基因及基因產物。應瞭解,除非上下文中出現時明確指 不’否則當揭示來自特定物種之基因或基因產物時,本發 明意欲僅為例示性的,且不應解釋為受到限制。因此,舉 例而言,對於在-些實施例令與哺乳動物核酸及胺基酸序 列有關之本文所揭示之基因,意欲涵蓋來自其他動物之同 源及/或直系同源基因及基因產物,該等動物包括(但不限 於)其一他哺乳動物、魚類、兩棲動物、爬行動物及鳥類。 在一實施例中,基因或核酸序列為人類基因或核酸序列。 定義 本文所用之術語僅出於描述特定實施例之目的且不欲限 制本發明。除非上下文另外明確指示,否則如本文所用之 單數形式「一」及「該」意欲亦包括複數形式。此外,對 於實施方式及/或申請專利範圍中使用術語「包括」、「具 有」、「有」或其變化形式之程度,該等術語意欲具有與術 語「包含」類似之包含性含義。 術約」或「大約」意謂在如一般技術者確定之特定 值的可接受誤差範圍内,此將部分視該值如何量測或測 定,亦即量測系統之限制而定。舉例而言,根據此項技術 中之實踐,「約」可意謂在1或1以上之標準差以内。或 者’「約」可意謂指定值之至多2〇〇/0、較佳至多10%、更佳 至多5%且更佳至多1%之範圍。或者,尤其關於生物系統 或方法,該術語可意謂在值之一定數量級以内,較佳在5 倍以内,且更佳在2倍以内。在申請案及申請專利範圍中 158213.doc 201210611 描述特定值時,除非另外說明,否則規定術語「約」意謂 在特定值之可接受之誤差範圍以内。 如本文所用之術語「mRNA」意謂目標基因之目前已知 之mRNA轉錄物,及可闡明之任何其他轉錄物。 「反義寡核苷酸」或「反義化合物」意謂一種RNA或 、 DNA分子,其結合另一 RNA或DNA(目標RNA、DNA)。舉 例而言,若其為RNA寡核苷酸,則其藉助於RNA-RNA相 互作用結合另一 RNA目標,且改變目標RNA之活性。反義 Ο 寡核苷酸可上調或下調特定聚核苷酸之表現及/或功能。 對於治療、診斷或其他觀點而言,該定義意謂包括任何適 用外來RNA或DNA分子。該等分子包括例如反義RNA或 DNA分子、干擾RNA(RNAi)、微小RNA、誘飼:RNA分子、 siRNA、酶性RNA、治療性編輯RNA以及促效劑及拮抗劑 RNA、反義寡聚化合物、反義寡核苷酸、外部引導序列 (EGS)寡核苦酸、替代剪接子(alternate splicers)、引子、 探針及與目標核酸之至少一部分雜交之其他寡聚化合物。 〇 因而,可引入呈單股、雙股、部分單股或環形募聚化合物 形式之此等化合物。 ' 在本發明之上下文中,術語「寡核苷酸」係指核糖核酸 ' (RNA)或去氧核糖核酸(DNA)或其模擬物之寡聚物或聚合 物。術語「寡核苷酸」亦包括天然及/或經修飾單體或鍵 聯之線性或環形募聚物,該等單體或鍵聯包括去氧核糖核 苷、核糖核苷、其經取代及α-變旋異構形式、肽核酸 (ΡΝΑ)、鎖核酸(LNA)、硫代磷酸酯、曱基膦酸酯及其類 158213.doc 201210611 似物。寡核苷酸能夠藉助於單體_單體相互作用之規定模 式,諸如華特生-克里克型(Watson_Crick type)鹼基配對、 虎克斯汀(Hodgsteen)或反向虎克斯汀型“everseIn one embodiment the 'oligonucleotide is administered as a pharmaceutical composition. The treatment regimen comprises administering to the patient an antisense compound at least once; however, the treatment can be modified to include multiple administrations over a period of time. The treatment can be combined with one or more other types of therapies. In one embodiment, the oligonucleotide is encapsulated in a liposome or linked to a carrier molecule (e.g., cholesterol, butylated AT peptide). Other aspects are described below. [Embodiment] For the purpose of explanation, several aspects of the invention are described below with reference to example applications. It should be understood that (d) many specific details, relationships, and methods are fully understood. However, it will be readily appreciated by those skilled in the art that the present invention may be practiced without y or multiple specific details or otherwise. The present invention is not limited by the ordering of operations or events' because these operations may occur in different orders and/or concurrently with other operations or events. Moreover, not all illustrated m events are required to practice the methods of the present invention. All genes, gene names and gene products are intended to correspond to homologs of any species of the compositions and methods disclosed herein, 1582l3.doc 201210611. Thus, such terms include, but are not limited to, human and mouse genes and gene products. It is to be understood that the invention is intended to be illustrative only and not to be construed as limiting. Thus, for example, for the genes disclosed herein, which are related to mammalian nucleic acid and amino acid sequences, are intended to encompass homologous and/or orthologous genes and gene products from other animals, Such animals include, but are not limited to, one of his mammals, fish, amphibians, reptiles, and birds. In one embodiment, the gene or nucleic acid sequence is a human gene or nucleic acid sequence. Definitions The terms used herein are for the purpose of describing particular embodiments only and are not intended to limit the invention. The singular forms "a" and "the" In addition, the terms "including", "having", "having" or "comprising" or "comprising" are used in the context of the embodiments and/or claims. "About" or "approximately" means that within the acceptable tolerances for a particular value as determined by the skilled artisan, this will depend in part on how the value is measured or measured, i.e., the limits of the measurement system. For example, according to the practice in the art, "about" can mean within 1 or more standard deviations. Or "about" may mean a range of up to 2 〇〇/0, preferably up to 10%, more preferably up to 5% and more preferably up to 1% of the specified value. Alternatively, particularly with respect to biological systems or methods, the term may mean within a certain order of magnitude, preferably within 5 times, and more preferably within 2 times. In the context of the application and the scope of the patent application 158213.doc 201210611 When describing a particular value, the term "about" is intended to mean within the acceptable tolerance of the particular value, unless otherwise stated. The term "mRNA" as used herein means the currently known mRNA transcript of the target gene, and any other transcript that can be elucidated. An "antisense oligonucleotide" or "antisense compound" means an RNA or DNA molecule that binds to another RNA or DNA (target RNA, DNA). For example, if it is an RNA oligonucleotide, it binds to another RNA target by means of RNA-RNA interaction and changes the activity of the target RNA. Antisense Ο Oligonucleotides can upregulate or downregulate the performance and/or function of a particular polynucleotide. For therapeutic, diagnostic or other purposes, this definition is meant to include any suitable foreign RNA or DNA molecule. Such molecules include, for example, antisense RNA or DNA molecules, interfering RNA (RNAi), microRNAs, ligation: RNA molecules, siRNA, enzymatic RNA, therapeutic editing RNA, and agonists and antagonist RNA, antisense oligomerization Compounds, antisense oligonucleotides, external leader sequences (EGS) oligonucleotides, alternative splicers, primers, probes, and other oligomeric compounds that hybridize to at least a portion of a target nucleic acid. 〇 Thus, such compounds can be introduced in the form of single-stranded, double-stranded, partially single-stranded or cyclic polymeric compounds. In the context of the present invention, the term "oligonucleotide" refers to an oligomer or polymer of ribonucleic acid ' (RNA) or deoxyribonucleic acid (DNA) or a mimetic thereof. The term "oligonucleotide" also includes natural or/or modified monomers or linked linear or cyclic conjugates, including deoxyribonucleosides, ribonucleosides, substituted and --spin isomeric forms, peptide nucleic acids (ΡΝΑ), locked nucleic acids (LNA), phosphorothioates, decylphosphonates and the likes 158213.doc 201210611 analogs. Oligonucleotides can rely on a defined pattern of monomer-monomer interactions, such as Watson_Crick type base pairing, Hodgsteen or reverse Hoxton type "everse

Hoogsteen type)驗基配對或其類似模式特異性結合目標聚核苷酸。 养核苷酸可為「嵌合」寡核苷酸,亦即由不同區構成。 在本發明之上下文中,「嵌合」化合物為寡核苷酸,其含 有兩種或兩種以上化學區,例如DNA區、RNA區、PNA區 等。各化學區由至少一個單體單元(在寡核苷酸化合物之 情況下亦即核苷酸)構成。此等寡核苷酸通常包含至少一 f 個區,在該區中該寡核苷酸經修飾以展現一或多種所需性 枭养核苷酸之所需性質包括(但不限於)例如對核酸酶降 解作用之抗性增強、細胞攝取增加及/或與目標核酸之結 δ親和力增強。因此,募核苷酸之不同區可具有不同性 質本發明之嵌合寡核苷酸可形成為兩種或兩種以上如上 所述之寡核苷酸、經修飾寡核苷酸、寡核苷及/或寡核苷 酸類似物之混合結構。 募核苷酸可由連接於「暫存器」中之區構成,亦即單體ΐ 如同天然DNA中一般連續連接,或經由間隔基連接。該等 間隔基意欲在區之間構成共價「橋」,且在較佳情況下具 有不超過約100個碳原子之長度。間隔基可帶有不同功能 性’例如具有正或負電荷、帶有特殊核酸結合性質(嵌入 劑、溝結合劑、毒素、螢光體等)、具親脂性、誘導特殊 二級結構(如例如誘導α螺旋之含有丙胺酸之肽)。 如本文所用之「ΝΑΜΡΤ」及「於驗酸胺轉磷酸核糖基 158213.doc -10· 201210611 酶」包括所有家族成員、突變體、對偶基因、片段、物 種、編碼及非編碼序列、有義及反義聚核苷酸股等。 如本文所用之詞語 1110035O14Rik、DKFZp666B131、 MGC117256、NAmPRTase、Nampt、於驗醢胺轉構酸核糖 基酶、PBEF、PBEF1、前B細胞群落增強因子1、前B細胞 ' 增強因子、VF、Visfatin、VISFATIN在文獻中視為相同且 在本申請案中可互換使用。 如本文所用之術語「對…有特異性之寡核苷酸」或「靶 Ο 向...之寡核苷酸」係指具有⑴能夠與目標基因之一部分形 成穩定複合物或(Π)能夠與目標基因之mRNA轉錄物之一部 分形成穩定雙鏈體之序列的寡核苷酸。複合物及雙鏈體之 穩定性可藉由理論計算及/或活體外檢定來測定。測定雜 交複合物及雙鏈體之穩定性的例示性檢定描述於以下實例 中。 如本文所用之術語「目標核酸」涵蓋DNA、自該DNA轉 錄之RNA(包含前體mRNA及mRNA)、以及自該RNA獲得之 〇 cDNA、編碼、非編碼序列、有義或反義聚核苷酸。募聚 化合物與其目標核酸之特異性雜交干擾核酸之正常功能。 藉由與目標核酸特異性雜交之化合物進行之目標核酸之功 - 能的此調節,一般稱為「反義」。待干擾之DNA之功能包 括例如複製及轉錄。待干擾之RNA之功能包括所有重要功 能,諸如RNA移位至蛋白質轉譯位點、蛋白質自RNA轉 譯、RNA剪接產生一或多種mRNA物質、及RNA可參與或 促進之催化活性。對目標核酸功能之該干擾的總體影響為 158213.doc -11 · 201210611 調節所編碼產物或募核苷酸之表現。 RNA干擾「RNAi」藉由對其「目標」核酸序列具有序 列特異同源性之雙股RNA(dsRNA)分子介導。在本發明之 某些實施例中,介體為5至25個核苷酸之「小干擾」RNA 雙鍵體(siRNA)。siRNA來源於藉由稱為以㈣之⑽咖酶加 工dsRNA。siRNA雙鏈體產物募集於稱為rISC(rnA誘導型 靜止複合物)之多蛋白siRNA複合物中。不希望受任何特定 理論束縛,接著咸信RISC被引導至目標核酸(適當地為 mRNA),在此處siRNA雙鏈體以序列特異性方式相互作用 以由催化方式介導裂解。可根據本發明使用之小干擾 RNAs可根據此項技術中熟知及一般熟練技術者熟悉之程 序合成及使用。用於本發明方法中之小干擾RNAs適當地 包含約1至約50個核苷酸(nt)。在非限制性實施例之實例 中,siRNAs可包含約5至約40個nt、約5至約30個nt、約1〇 至約30個nt、約15至約25個nt或約20至25個核苦酸。 藉由使用自動比對核酸序列且指示具有一致性或同源性 之區的電腦程式促進適當寡核苷酸之選擇。使用該等程式 例如藉由搜尋諸如GenBank之資料庫或藉由對Pcr產物進 行疋序來比較所獲得之核酸序列。比較來自多種物種之核 酸序列允許選擇在物種之間顯示適當程度之—致性的核酸 序列。在基因未定序之情況下,執行南方墨點法(s〇uthern blot)以允許測定目標物種與其他物種之基因之間的一致性 程度。如此項技術中所熟知,藉由在不同嚴格性程度下執 行南方墨點法’可獲得一致性之近似量度。此等程序允許 158213.doc 12 201210611 選擇與待對照個體中之目標核酸序列顯示高度互補及與其 他物種中之相應核酸序列顯示較低程度之互補的寡核苷 酸。熟習此項技術者將認識到,在選擇用於本發明中之基 因之適當區方面存在相當大的自由度。 「酶性RNA」意謂具有酶活性之RNA分子(Cech,(1988) J_ JmeWcijn Mec/. dwoc. 260, 3030-3035)。酶性核酸(核糖 核酸酶)藉由首先結合目標RNA來起作用。該結合經由酶 性核酸之目標結合部分進行,該目標結合部分保持極接近 〇 用以裂解目標RNA之分子的酶性部分。因此,酶性核酸首 先識別,且接著經由鹼基配對結合目標RNA,且一旦結合 至正確位置,即以酶促方式作用切割目標RNA。 「誘餌RNA」意謂模擬配位體之天然結合域的RNA分 子。因此,誘餌RNA與天然結合目標競爭結合特定配位 體。舉例而言,已顯示,HIV轉錄活化反應(TAR)RNA之 過度表現可充當「誘餌」,且有效結合HIV tat蛋白質,由 此防止其與HIV RNA中所編碼之TAR序列結合。此情況意 欲為一特定實例。熟習此項技術者將認識到,此實例僅為 一個實例,且其他實施例可使用此項技術中一般已知之技 術容易地產生。 ' 如本文所用之術語「單體」通常指示由磷酸二酯鍵或其 類似物連接形成大小為幾個(例如約3至4個)單體單元至約 數百個單體單元範圍内之寡核苷酸的單體。如下文更充分 描述,磷酸二酯鍵聯之類似物包括:硫代磷酸酯、二硫代 磷酸酯、甲基膦酸酯、硒代磷酸酯、磷醯胺酯 158213.doc •13- 201210611 (Phosphoramidate)及其類似物。 術語「核苷酸」涵蓋天然存在之核苷酸以及非天然存在 之核苷酸。熟習此項技術者應清楚,先前視為「非天然存 在」之各種核普酸後來在自然界中已發現。因此,「核普 酸」不僅包括已知含有嘌呤及嘧啶雜環之分子,而且亦包 括其雜環類似物及互變異構體。其他類型核苷酸之說明性 實例為含有腺嘌呤、鳥嘌呤、胸腺嘧啶、胞嘧啶、尿嘧 啶、嘌呤、黃嘌呤、二胺基嘌呤、8-側氧基-N6-甲基腺嘌 呤、7-去氮黃嘌呤、7-去氮鳥嘌呤、N4,N4-乙橋胞》密α定、 N6,N6-乙橋-2,6-二胺基嘌呤、5-曱基胞嘧咬、5-(C3-C6)-炔基胞嘧啶、5-氟尿嘧啶、5-溴尿嘧啶、假異胞嘧啶、2-羥基-5-甲基-4-三唑并吡啶、異胞嘧啶、異鳥嘌呤、肌苷 之分子及Benner等人,美國專利第5,432,272號中所述之 「非天然存在之」核苷酸。術語「核苷酸」意欲涵蓋此等 實例中之每一者及所有以及其類似物及互變異構體。尤其 關注之核苷酸為含有腺嘌呤、鳥嘌呤、胸腺嘧啶、胞嘧啶 及尿嘧啶之彼等核苷酸,認為其為與人類中之治療性及診 斷性應用有關之天然存在之核苷酸。核苷酸包括天然2’-去 氧及 2’-經基糖,例如如 Kornberg及Baker, DNA Replication, 第2版(Freeman, San Francisco, 1992)中所述之糖,以及其 類似物。 與核苷酸有關之「類似物」包括具有經修飾鹼基部分及/ 或經修飾糖部分之合成核苷酸(參見例如Scheit, Nucleotide Analogs, John Wiley, New York, 1980 ; Freier及 Altmann, 158213.doc • 14· 201210611 (1997) Nucl. Acid. Res., 25(22), 4429-4443 ; Toulme, J.J., (2001) Nature Biotechnology 19:17-18 ; Manoharan M., (1999) Biochemica et Biophysica Acta 1489:117-139 ; Freier S. M., (1997) Nucleic Acid Research, 25:4429-4443, Uhlman, E., (2000) Drug Discovery & Development, 3: 203-213 ; Herdewin P., (2000) Antisense & Nucleic Acid Drug Z)ev.,10:297-310所一般性描述);2’-0,3’-C-連接型[3.2.0] Ο ο 雙環阿糖核苷。該等類似物包括涉及成增強結合性質之合 成核苷酸,例如雙鏈體或三鏈體穩定性、特異性或其類似 性質。 如本文所用之「雜父」意§胃养·聚化合物之實質上互補股 的配對。一種配對機制涉及寡聚化合物之股之互補核苦或 核苷酸驗基(核苷酸)之間的氫鍵結,其可為華特生_克里 克、虎克斯汀或反向虎克斯汀氫鍵結。舉例而言,腺嗓吟 及胸腺嘧啶為經由形成氫鍵配對之互補核苷酸。雜交可在 不同情況下發生。 當反義化佘物與目標核酸之結合干擾目標核酸之正常功 能從而產生功能及/或活性之調節,且在需要特異性結合 之條件下,亦即在活體内檢定或治療性處理情況下之生理 條件下,及在活體外檢定情況下執行檢定之條件下,存在 足夠程度之互補以避免反義化合物與非目標核酸序列:非 特異性結合時’該化合物為「可特異性雜交。 如本文利之短語「嚴格雜交條件」^嚴格條件」传 指本發明化合物與其目標序列’而且與最少量之其他序列 15S213.doc •15- 201210611 雜交之條件。嚴格條件與序列相關且在不同情況將有所不 同’且在本發明之上下文中,寡聚化合物與目標序列雜交 之「嚴格條件」由募聚化合物之性質及組成及對其進行研 究之檢定來確定。一般而言,嚴格雜交條件包含低濃度 (0.1 5 M)之具有諸如Na+或K+之無機陽離子的鹽(亦即低離 子強度)、高於20°C至25。(:低於寡聚化合物:目標序列複合 物之Tm的溫度、及存在諸如甲醯胺、二曱基甲酿胺、二 甲亞硬或清潔劑十二烷基硫酸鈉(sr>S)之變性劑。舉例而 έ,對於各1%曱醯胺而言,雜交速率降低1.1%。高嚴格 性雜交條件之實例為在6 〇 t下〇.丨χ氯化鈉_檸檬酸鈉緩衝液 (SSC)/0.1%(W/V)SDS 歷時 30 分鐘。 如本文所用之「互補」係指一或兩個寡聚股上之兩種核 苷酸之間精確配對之能力。舉例而言,若反義化合物之某 一位置的核驗基能夠與目標核酸之某一位置的核鹼基氫鍵 結,其中目標核酸為DNA、RNA或募核苷酸分子,則將寡 核苷酸與目標核酸之間的氫鍵結位置視為互補位置。當可 彼此氫鍵結之核苷酸佔據各分子中足夠數量之互補位置 i 時,寡聚化合物及其他DNA、RNA或募核苷酸分子彼此互 補。因此,「可特異性雜交」及「互補」為用以指示在足 夠數量之核苷酸上有足夠程度之精確配對或互補使得在寡 聚化合物與目標核酸之間發生穩定且特異性結合之術語。 應瞭解,在此項技術中募聚化合物之序列無需與可特異 性雜交之其目標核酸之序列1〇0%互補。此外,募核芽酸 可在一或多個區^又上雜父’使得插入或鄰近區段不參與雜 158213.doc -16- 201210611 交事件(例如環結構、錯配或髮夾結構)。本發明之寡聚化 合物包含至少約70%、或至少約75%、或至少約80%、或 至少約85°/◦、或至少約90°/。、或至少約95%、或至少約99% 與其所靶向目標核酸序列内之目標區之序列互補性。舉例 而言,反義化合物之20個核普酸中有18個與目標區互補且 ' 因此特異性雜交之反義化合物表示90%互補性。在此實例 中,剩餘非互補核苷酸可能與互補核苷酸一起成鎮或散佈 於互補核苷酸中,而無需彼此或與互補核苷酸鄰接。因 〇 而,具有側接與目標核酸完全互補之兩個區的4(四)個非互 補核苷酸之長度為18個核苷酸的反義化合物與目標核酸具 有77.8%之總體互補性,且因此屬於本發明之範疇内。通 常可使用此項技術中已知之BLAST程式(基本局部比對搜 尋工具)及PowerBLAST程式測定反義化合物與目標核酸區 之互補性百分比。同源性、序列一致性或互補性百分比可 藉由例如 Gap程式(Wisconsin Sequence Analysis Package, 第 8版,用於 Unix, Genetics Computer Group,University ◎Hoogsteen type) A chimeric pair or a similar pattern specifically binds to a polynucleotide of interest. Nutrients can be "chimeric" oligonucleotides, that is, composed of different regions. In the context of the present invention, a "chimeric" compound is an oligonucleotide containing two or more chemical regions, such as a DNA region, an RNA region, a PNA region, and the like. Each chemical region is composed of at least one monomer unit (in the case of an oligonucleotide compound, i.e., a nucleotide). Such oligonucleotides typically comprise at least one f region in which the oligonucleotide is modified to exhibit one or more desired properties of the desired nucleotide including, but not limited to, for example Increased resistance to nuclease degradation, increased cellular uptake, and/or enhanced affinity for the δ of the target nucleic acid. Thus, different regions of the raised nucleotides may have different properties. The chimeric oligonucleotides of the invention may be formed into two or more oligonucleotides, modified oligonucleotides, oligonucleosides as described above. And/or a mixed structure of oligonucleotide analogs. The nucleotides may be composed of a region linked to a "storage register", that is, the monomers are generally connected continuously as in natural DNA or via spacers. The spacers are intended to form a covalent "bridge" between the zones and, preferably, have a length of no more than about 100 carbon atoms. The spacers may have different functionalities' such as having positive or negative charges, with specific nucleic acid binding properties (intercalators, groove binders, toxins, phosphors, etc.), lipophilic, and inducing specific secondary structures (eg, for example Inducing an alpha helix containing alanine peptide). As used herein, "ΝΑΜΡΤ" and "in the acid-lowering phosphoribosyl 158213.doc -10· 201210611 enzyme" include all family members, mutants, dual genes, fragments, species, coding and non-coding sequences, sense and Antisense polynucleotide strands, etc. As used herein, the words 1110035O14Rik, DKFZp666B131, MGC117256, NAmPRTase, Nampt, proline amine ribosyltransferase, PBEF, PBEF1, pre-B cell community enhancer 1, pre-B cell enhancer, VF, Visfatin, VISFATIN They are considered the same in the literature and are used interchangeably in this application. The term "oligonucleotide specific for" or "oligonucleotide to" as used herein means having (1) capable of forming a stable complex with one of the target genes or (Π) capable of An oligonucleotide that forms a sequence of a stable duplex with a portion of an mRNA transcript of a gene of interest. The stability of the complexes and duplexes can be determined by theoretical calculations and/or in vitro assays. Exemplary assays for determining the stability of hybrid complexes and duplexes are described in the Examples below. The term "target nucleic acid" as used herein encompasses DNA, RNA transcribed from the DNA (including precursor mRNA and mRNA), and 〇 cDNA, coding, non-coding sequence, sense or antisense nucleoside obtained from the RNA. acid. The specific hybridization of a compound with its target nucleic acid interferes with the normal function of the nucleic acid. This regulation of the function of the target nucleic acid by a compound that specifically hybridizes to the target nucleic acid is generally referred to as "antisense". The functions of the DNA to be interfered with include, for example, replication and transcription. The function of the RNA to be interfered with includes all important functions such as RNA translocation to protein translation sites, protein translation from RNA, RNA splicing to produce one or more mRNA species, and catalytic activity in which RNA can participate or promote. The overall effect of this interference on the function of the target nucleic acid is 158213.doc -11 · 201210611 Modulate the performance of the encoded product or nucleotide. RNA interference "RNAi" is mediated by a double-stranded RNA (dsRNA) molecule with sequence-specific homology to its "target" nucleic acid sequence. In certain embodiments of the invention, the mediator is a 5 to 25 nucleotide "small interference" RNA double bond (siRNA). The siRNA is derived from a dsRNA which is referred to by (10) (10). The siRNA duplex product is recruited in a polyprotein siRNA complex called rISC (rnA-inducible resting complex). Without wishing to be bound by any particular theory, the RISC is then directed to the target nucleic acid (suitably mRNA), where the siRNA duplex interacts in a sequence-specific manner to catalyze cleavage in a catalytic manner. Small interfering RNAs which can be used in accordance with the present invention can be synthesized and used according to procedures well known in the art and familiar to those skilled in the art. The small interfering RNAs used in the methods of the invention suitably comprise from about 1 to about 50 nucleotides (nt). In an example of a non-limiting embodiment, the siRNAs can comprise from about 5 to about 40 nt, from about 5 to about 30 nt, from about 1 to about 30 nt, from about 15 to about 25 nt, or from about 20 to 25 A bit of bitter acid. Selection of appropriate oligonucleotides is facilitated by the use of computer programs that automatically align nucleic acid sequences and indicate regions of homology or homology. The nucleic acid sequences obtained are compared using, for example, by searching a database such as GenBank or by sequencing the Pcr product. Comparing nucleic acid sequences from a variety of species allows for the selection of nucleic acid sequences that display an appropriate degree of identity between species. In the case of unscheduled genes, a Southern blot is performed to allow for the determination of the degree of identity between the target species and the genes of other species. As is well known in the art, an approximate measure of consistency can be obtained by performing a Southern dot method at varying degrees of stringency. Such procedures allow 158213.doc 12 201210611 to select oligonucleotides that are highly complementary to the target nucleic acid sequence in the individual to be controlled and that exhibit a lower degree of complementarity to the corresponding nucleic acid sequences in other species. Those skilled in the art will recognize that there is considerable freedom in selecting the appropriate regions for the genes used in the present invention. "Enzymatic RNA" means an RNA molecule having enzymatic activity (Cech, (1988) J_JmeWcijn Mec/. dwoc. 260, 3030-3035). The enzymatic nucleic acid (ribonuclease) functions by first binding to the target RNA. This binding is carried out via the target binding portion of the enzymatic nucleic acid which remains in close proximity to the enzymatic portion of the molecule used to cleave the target RNA. Thus, the enzymatic nucleic acid is first recognized, and then the target RNA is bound via base pairing, and once bound to the correct position, the target RNA is cleaved in an enzymatic manner. "Bee RNA" means an RNA molecule that mimics the natural binding domain of a ligand. Thus, the decoy RNA competes with the natural binding target for binding to a particular ligand. For example, it has been shown that overexpression of HIV transcriptional activation (TAR) RNA acts as a "bait" and effectively binds to the HIV tat protein, thereby preventing its binding to the TAR sequence encoded in HIV RNA. This situation is intended to be a specific instance. Those skilled in the art will recognize that this example is only one example, and that other embodiments can be readily produced using techniques generally known in the art. The term "monomer" as used herein generally denotes the attachment of a phosphodiester bond or an analog thereof to form a monomer having a size ranging from a few (e.g., about 3 to 4) monomer units to about several hundred monomer units. A monomer of a nucleotide. As described more fully below, phosphodiester-linked analogs include: phosphorothioates, dithiophosphates, methylphosphonates, selenophosphates, phosphoxamate 158213.doc •13- 201210611 ( Phosphoramidate) and its analogues. The term "nucleotide" encompasses naturally occurring nucleotides as well as non-naturally occurring nucleotides. Those skilled in the art should be aware that various types of nucleotides previously considered "non-natural" have been discovered in nature. Therefore, "nuclear acid" includes not only molecules known to contain purine and pyrimidine heterocycles, but also heterocyclic analogs and tautomers thereof. Illustrative examples of other types of nucleotides are adenine, guanine, thymine, cytosine, uracil, guanidine, xanthine, diamino guanidine, 8-sided oxy-N6-methyl adenine, 7 - denitrazine, 7-deazaguanine, N4, N4-acetylidene, N6, N6-B-bridge-2,6-diaminopurine, 5-mercaptocytidine, 5 -(C3-C6)-alkynylcytosine, 5-fluorouracil, 5-bromouracil, pseudoisopyrimidine, 2-hydroxy-5-methyl-4-triazolopyridine, isocytosine, isoguanine And the "non-naturally occurring" nucleotides described in U.S. Patent No. 5,432,272. The term "nucleotide" is intended to cover each and every of such examples as well as analogs and tautomers thereof. Nucleotides of particular interest are nucleotides containing adenine, guanine, thymine, cytosine, and uracil, which are considered to be naturally occurring nucleotides associated with therapeutic and diagnostic applications in humans. . Nucleotides include natural 2'-deoxy and 2'-transglycosides, such as those described in Kornberg and Baker, DNA Replication, 2nd Edition (Freeman, San Francisco, 1992), and analogs thereof. Nucleotide-related "analogs" include synthetic nucleotides having modified base moieties and/or modified sugar moieties (see, for example, Scheit, Nucleotide Analogs, John Wiley, New York, 1980; Freier and Altmann, 158213). .doc • 14· 201210611 (1997) Nucl. Acid. Res., 25(22), 4429-4443 ; Toulme, JJ, (2001) Nature Biotechnology 19:17-18 ; Manoharan M., (1999) Biochemica et Biophysica Acta 1489: 117-139; Freier SM, (1997) Nucleic Acid Research, 25: 4429-4443, Uhlman, E., (2000) Drug Discovery & Development, 3: 203-213; Herdewin P., (2000) Antisense & Nucleic Acid Drug Z) ev., 10: 297-310 General Description); 2'-0, 3'-C-linked [3.2.0] Ο ο Bicyclic arabinose. Such analogs include synthetic nucleotides that are involved in enhancing binding properties, such as duplex or triplex stability, specificity, or the like. As used herein, "monomer" means the pairing of substantially complementary strands of a gastric-growth compound. A pairing mechanism involves hydrogen bonding between complementary nuclear or nucleotide nucleotides (nucleotides) of a strand of an oligomeric compound, which may be a Watson _ creek, a tiger cretin or a reverse tiger Kistin hydrogen bonding. For example, adenine and thymine are complementary nucleotides paired by the formation of hydrogen bonds. Hybridization can occur in different situations. When the binding of the antisense agent to the target nucleic acid interferes with the normal function of the target nucleic acid to produce regulation of function and/or activity, and under conditions requiring specific binding, ie, in vivo or therapeutic treatment Under physiological conditions, and under conditions of performing in vitro assays, there is a sufficient degree of complementarity to avoid antisense compounds and non-target nucleic acid sequences: when non-specifically binds, the compound is "specifically hybridizable." The phrase "stringent hybridization conditions" and "stringent conditions" refer to the conditions under which the compound of the present invention hybridizes to its target sequence' and to the least amount of other sequences 15S213.doc •15-201210611. Stringent conditions are sequence dependent and will vary from case to case' and in the context of the present invention, the "stringent conditions" at which an oligomeric compound hybridizes to a target sequence are determined by the nature and composition of the polymerized compound and its investigation. determine. In general, stringent hybridization conditions include a low concentration (0.1 5 M) of a salt having an inorganic cation such as Na+ or K+ (i.e., low ion strength), and a temperature higher than 20 °C to 25. (: lower than the oligomeric compound: the temperature of the Tm of the target sequence complex, and the presence of, for example, formamidine, dimercaptoamine, dimethyl sulfite or detergent sodium lauryl sulfate (sr>S) Denaturing agents. For example, for each 1% guanamine, the rate of hybridization is reduced by 1.1%. An example of high stringency hybridization conditions is 6. 丨χ sodium chloride _ sodium citrate buffer at 6 〇t SSC) / 0.1% (W / V) SDS lasts 30 minutes. As used herein, "complementary" refers to the ability to precisely pair between two nucleotides on one or two oligomeric strands. For example, if The nucleocapsid at a position of the sense compound is capable of hydrogen bonding with a nucleobase at a position of the target nucleic acid, wherein the target nucleic acid is a DNA, RNA or nucleotide molecule, and the oligonucleotide is between the oligonucleotide and the target nucleic acid. The hydrogen bonding position is regarded as a complementary position. When the nucleotides which are hydrogen-bondable to each other occupy a sufficient number of complementary positions i in each molecule, the oligomeric compound and other DNA, RNA or nucleotide molecules are complementary to each other. "Specific hybridization" and "complementary" are used to indicate a sufficient number of nucleotides There is a sufficient degree of exact pairing or complementation to enable stable and specific binding between the oligomeric compound and the target nucleic acid. It will be appreciated that in this technique the sequence of the polymerized compound does not require a target nucleic acid that specifically hybridizes to it. The sequence is 1〇0% complementary. In addition, the nucleotide can be raised in one or more regions, and the heterogeneous parent can make the inserted or adjacent segment not participate in the 158213.doc -16 - 201210611 event (eg, ring structure, Mismatch or hairpin structure. The oligomeric compound of the invention comprises at least about 70%, or at least about 75%, or at least about 80%, or at least about 85°/◦, or at least about 90°/., or at least About 95%, or at least about 99%, of the sequence complementarity to the target region within the target nucleic acid sequence to which it is targeted. For example, 18 of the 20 nucleotides of the antisense compound are complementary to the target region and are therefore specific Sexually hybridized antisense compounds represent 90% complementarity. In this example, the remaining non-complementary nucleotides may be singular or interspersed with complementary nucleotides in complementary nucleotides without the need for each other or with complementary nucleotides Adjacent. Because of the squat, with side An antisense compound having a length of 18 nucleotides of 4 (four) non-complementary nucleotides in two regions in which the target nucleic acid is completely complementary has 77.8% overall complementarity with the target nucleic acid, and thus is within the scope of the present invention. The percentage of complementarity of the antisense compound to the target nucleic acid region can generally be determined using the BLAST program (basic local alignment search tool) and the PowerBLAST program known in the art. The homology, sequence identity or percent complementarity can be used by For example, Gap program (Wisconsin Sequence Analysis Package, 8th edition, for Unix, Genetics Computer Group, University ◎

Research Park, Madison Wis·)使用預設設定來測定,該程 式使用 Smith 及 Waterman之演算法(Ji/v. ΜαΜ·,(1981) 2, 482-489)。 • 如本文所用之術語「熱熔點(Tm)」係指在指定離子強 度、pH值及核酸濃度下,50%與目標序列互補之募核苷酸 與目標序列雜交平衡的溫度。通常,嚴格條件為以下條 件,其中鹽濃度為pH 7.0至8.3下至少約0.01至1.0 M Na離 子濃度(或其他鹽),且對於短募核苷酸(例如10至50個核苷 158213.doc -17- 201210611 酸)而言溫度為至少約3 去穩定劑來達成嚴袼條件。 如本文所用之「調節」意 少(抑制)。 。亦可藉由添加諸如甲醯胺之 謂基因表現之增加(刺激)或減 術語「變異體」當料聚核苷酸序狀情形下時可涵蓋 與野生型基因相關之聚核苷酸序列。此定義亦可包括例如 「對偶基因」、「剪接」、「物種」或「多形」變異體。剪接 變異體可與參考分子具有顯著—致性,但—般而言將因 mRNA加工期間外顯子之替代性剪接而具有較多或較少數 目之聚核苦酸。相應多肽可具有其他功能域或不存在結構 域。物種變異體為一個物種不同於另一物種之聚核苷酸序 列。野生型基因產物之變異體在本發明中具有特別效用。 變異體可由核酸序列中之至少一個突變產生且可產生改 變之mRNA或結構或功&可能改變或可能不㈣之多狀。 任何指定天然或重組基因可不具有、具有一種或許多種對 偶土因形式產生變異體之常見突變一般歸因於核苷酸之 天然缺失、添加或取代。在指定序列中,此等類型之改變 各自可單獨或與其他改變組合發生一或多次。 所得多肽彼此之間一般具有顯著胺基酸一致性。多形變 異體為指定物種之個體之間特定基因之聚核苷酸序列的變 化。多形變異體亦可涵蓋聚核苷酸序列有一個鹼基發生變 化之「單核苷酸多形現象」或單鹼基突變。SNp之 存在可指示例如特定群體發生疾病病況之傾向,亦即易感 性及抗性。 158213.doc -18· 201210611 衍生聚核苷酸包括經受化學修飾(例如氫經烷基、酿基 或胺基置換)之核酸。例如衍生养核皆酸之衍生物可包含 非天然存在之部分,諸如改變之糖部分或糖間鍵聯。其實 例包括硫代磷酸酯及此項技術中已知之其他含硫物質種 類。衍生核酸亦可含有標記’包括放射性核苷酸、酶、榮 光劑、化學發光劑、發色劑、受質、輔因子、抑制劑、磁 性粒子及其類似物。Research Park, Madison Wis.) was determined using a preset setting using Smith and Waterman's algorithm (Ji/v. ΜαΜ·, (1981) 2, 482-489). • The term "thermal melting point (Tm)" as used herein refers to a temperature at which 50% of the nucleotides complementary to the target sequence hybridize to the target sequence at a given ionic strength, pH, and nucleic acid concentration. Generally, stringent conditions are those in which the salt concentration is at least about 0.01 to 1.0 M Na ion concentration (or other salt) at pH 7.0 to 8.3, and for short-raised nucleotides (eg, 10 to 50 nucleosides 158213.doc) -17- 201210611 Acid) The temperature is at least about 3 destabilizing agents to achieve severe conditions. As used herein, "regulation" means less (inhibition). . The nucleotide sequence associated with the wild-type gene can also be encompassed by the addition of a gene expression such as formamide (stimulation) or the term "variant" when the polynucleotide sequence is used. This definition may also include, for example, "dual genes", "splicing", "species" or "polymorphic" variants. A splicing variant may be significantly responsive to a reference molecule, but will generally have a greater or lesser number of polynucleic acids due to alternative splicing of exons during mRNA processing. The corresponding polypeptide may have other functional domains or no domain. A species variant is a polynucleotide sequence in which one species is different from another species. Variants of wild type gene products have particular utility in the present invention. A variant may be produced by at least one mutation in a nucleic acid sequence and may produce a altered mRNA or structure or work that may or may not be (4) polymorphic. Any common mutation that specifies that a natural or recombinant gene may have no variants with one or more dual native forms is generally attributed to the natural deletion, addition or substitution of nucleotides. In a given sequence, these types of changes can each occur individually or in combination with other changes one or more times. The resulting polypeptides generally have significant amino acid identity with each other. Polymorphism A variant of a polynucleotide sequence that is a specific gene between individuals of a given species. Polymorphic variants may also encompass "single nucleotide polymorphisms" or single base mutations in which the nucleotide sequence has a base change. The presence of SNp may indicate, for example, the propensity of a particular population to develop a disease condition, i.e., susceptibility and resistance. 158213.doc -18· 201210611 Derivatized polynucleotides include nucleic acids that undergo chemical modification, such as replacement of hydrogen with an alkyl group, a thiol group, or an amine group. For example, a derivatized nutrient derivative may comprise a non-naturally occurring moiety, such as a modified sugar moiety or an intersaccharide linkage. Examples include phosphorothioates and other sulfur-containing species known in the art. The derivatized nucleic acid may also contain a label 'including a radioactive nucleotide, an enzyme, a refractory agent, a chemiluminescent agent, a chromogen, a substrate, a cofactor, an inhibitor, a magnetic particle, and the like.

「衍生」多肽或肽為例如藉由糖基化、聚乙二醇化、鱗 酸化、硫酸化、還原/烷基化、醯化、化學偶合或輕度福 馬林(formalin)處理修飾之多肽或肽。衍生物亦可經修飾 以含有可直接或間接债測之標言己’包括(但不限於)放射性 同位素標記、螢光標記及酶標記。 如本文所用之術語「動物」或「患者」意欲包㈣如人 頒綿羊、麋鹿、鹿、長耳鹿、紹、哺乳動物、猴子、 馬、牛、緒、山羊、狗、猶、大鼠、小鼠、鳥類、雞、爬 行動物、魚類、昆蟲及蛛形綱動物。 「哺乳動物」涵蓋通常接受醫療護理之溫血哺乳動物 (例如人類及制化動物)。實例__、犬科動物、 馬科動物、牛科動物及人類,以及僅包括人類。 「治療」涵蓋治療哺乳動物之疾病病況,且包括:⑷防 疾病病況在哺乳動物中线,尤其在該哺乳動物易串疾 病病況但尚未診斷為患有該疾二 況,例如阻止其發展;及/_減輕抑制疾病病 病病況消退直至達到所需二… 列如使疾 J所需m療亦包括改善疾病之症 I58213.doc -19- 201210611 狀(例如減輕疼痛或不適)’其中該改善可能會或可能不會 直接影響疾病(例如起因、傳播、表現等)。 如本文所用之「癌症」係指哺乳動物中發現之所有類型 之癌症或贅瘤或惡性腫瘤,包括(但不限於):白血病、淋 巴瘤、黑素瘤、癌及肉瘤。癌症本身表現為包含惡性癌症 細胞之「腫瘤」或組織。腫瘤之實例包括肉瘤及癌,諸如 (但不限於):纖維肉瘤、點液肉瘤、脂肪肉瘤、軟骨肉 瘤、骨原性肉瘤、脊索瘤、金管肉瘤、内皮肉瘤、淋巴管 肉瘤、淋巴管内皮肉瘤、滑膜瘤、間皮瘤、尤因氏瘤 (Ewing’s tumor)、平滑肌肉瘤、橫紋肌肉瘤、結腸癌、胰 臟癌、乳癌、#巢癌、前列腺癌、鱗狀細胞癌、基底細胞 癌、腺癌、汗腺癌、皮脂腺癌、乳頭狀癌' 乳頭狀腺癌、 囊腺癌、髓性癌、支氣管癌、腎細胞癌、肝細胞癌、膽管 癌織膜癌、精原細胞瘤、胚胎癌、威姆氏膜瘤 tumor)、子宮頸癌、睪丸腫瘤、肺癌、小細胞肺癌、膀胱 癌上皮癌、神經膠質瘤、星形細胞瘤、驗母細胞瘤、顧 因e瘤至s膜瘤、松果腺瘤、血管母細胞瘤、聽神經 瘤 '募樹突神經膠細胞瘤、腦脊鑛膜瘤、黑素瘤、神經母 細胞瘤及視網膜庇鈿眙、疏 -T -, 、·,胞瘤。可猎由所揭示本發明組合物治 療之其他癌症包括(但不限於)例如霍奇金氏病(Hodgki心 DiSeaSe)、非霍奇金氏淋巴瘤(N〇n-H〇dgkin,s Lymphoma)、 多發!·生月趙瘤、神經母細胞瘤、乳癌、卵巢癌、肺癌、橫 紋肌肉瘤原發性血小板增多症、原發性巨球蛋白血病、 小細胞肺腫瘤、原發性腦腫瘤、胃癌(st_ch __)、結 158213.doc -20- 201210611 腸癌、惡性胰臟胰島素瘤、惡性類癌' 膀胱癌、胃癌 (gastric cancer)、惡變前皮膚病變、睪丸癌、淋巴瘤、曱 狀腺癌、神經母細胞瘤、食道癌、泌尿生殖道癌、惡性高 鈣血症、子宮頸癌、子宮内膜癌、腎上腺皮層癌及前列腺 癌。 如本文所用之「神經疾病或病症」係指神經系統及/或 視覺系統之任何疾病或病症。「神經疾病或病症」包括涉 及中柩神經系統(腦、腦幹及小腦)、周邊神經系統(包括顱 〇 神經)及自主神經系統(一部分位於中樞神經系統與周邊神 經系統中)之疾病或病症0神經疾病或病症包括(但不限於) 後天性癲癇型失語症;急性播散性腦脊髓炎;腎上腺腦白 質營養不良;年齡相關之黃斑部變性;胼胝體發育不全; 失識症,艾卡爾迪症候群(Aieardi syndrome);亞歷山大病 (Alexander disease);阿爾珀斯病(Alpers,disease);交替 性偏癱,阿兹海默氏病(Alzheimer's disease);血管性瘋 Q 呆;肌萎縮性側索硬化症;無腦;安格曼症候群 (Angelman syndrome);血管瘤病;缺氧症;失語症;運用 不能;蛛網膜囊腫;蛛網膜炎;阿諾-希阿里畸形(Anr〇nl_ Chiari malformation);動靜脈畸形;亞斯伯格症候群 ' (AsPerSei* syndrome);毛細血管擴張性運動失調;注意力 不足過動症;自閉症;自主神經功能障礙;背痛;貝登氏 病(Batten disease),貝西氏病(Behcet's disease);貝爾氏麻 痒(Bell's palsy);良性原發性瞼痙攣;良性局部肌萎縮; 良性顱内ifj壓,賓斯旺格氏病(Binswanger's disease);險 158213.doc -21 * 201210611 癌攣’布.洛赫-棘錄貝格症候群(Bl〇ch Sulzberger syndrome);臂神經叢損傷;腦膿腫;腦損傷;腦腫瘤(包 括多形性膠質母細胞瘤);脊趙腫瘤;布朗-希岭得症候群 (Brown-Sequard syndrome);康納凡病(Canavan disease); 腕隧道症候群;灼痛;中樞性疼痛症候群;中央腦橋脊髓 溶解;頭部病症;腦動脈瘤;腦動脈硬化;腦萎縮;腦性 巨人症;腦性麻痒;恰克-馬利-杜斯氏病(Charc〇t_Marie_ Tooth disease);化學療法誘發之神經病變及神經痛;嘉里 氏畸形(Chiari malformation);舞蹈病;慢性發炎性脫髓鞘 性多發性神經病變;慢性疼痛;慢性區域疼痛症候群;科 务-洛知氏症候群(Coffin Lowry syndrome);昏迷,包括持 續性植物狀態;先天性兩侧面癱;皮質基底核退化症;顱 動脈炎,顱縫早閉;庫賈氏病(Creutzfeldt_Jak〇b disease); 累積性傷害病;庫欣氏症候群(Cushing,s syndr〇me);巨大 細胞包涵體病;細胞巨大病毒感染;舞蹈眼-舞蹈足症候 群,丹迪沃克症候群(DandyWalker syndrome);多森病 (Dawson disease);德摩西埃氏症候群(De M〇rsieris syndrome);代哲因-克拉克麻痺(Dejerine_Klumke paisy); 癡呆症,皮膚肌炎;糖尿病性神經病變;彌漫性硬化症; 自主神經障礙;書寫困難;誦讀困難;肌張力障礙;早期 嬰兒癲癇性腦病變;空蝶鞍症候群A "derived" polypeptide or peptide is, for example, a polypeptide or peptide modified by glycosylation, pegylation, sulphation, sulfation, reduction/alkylation, deuteration, chemical coupling or mild formalin treatment. . Derivatives may also be modified to contain direct or indirect fingerprinting, including but not limited to, radioisotope labels, fluorescent labels, and enzyme labels. The term "animal" or "patient" as used herein is intended to include (4) human sheep, elk, deer, mule deer, sau, mammal, monkey, horse, cow, ox, goat, dog, juvenile, rat, Mice, birds, chickens, reptiles, fish, insects and arachnids. "Mammal" covers warm-blooded mammals (such as humans and manufactured animals) who are usually treated with medical care. Examples __, canines, equines, bovines and humans, and only humans. "Treatment" encompasses the treatment of a disease condition in a mammal, and includes: (4) an anti-disease condition at the midline of the mammal, particularly in the mammalian condition of the disease but not yet diagnosed as having the condition, such as preventing its development; and /_ Reducing the suppression of disease and disease, the disease condition subsides until the desired two... The m treatment required for the disease also includes the improvement of the disease I58213.doc -19- 201210611 (eg, relieve pain or discomfort), where the improvement may or It may not directly affect the disease (eg cause, spread, performance, etc.). "Cancer" as used herein refers to all types of cancer or neoplasms or malignancies found in mammals, including but not limited to: leukemia, lymphoma, melanoma, carcinoma and sarcoma. Cancer itself manifests itself as a "tumor" or tissue that contains malignant cancer cells. Examples of tumors include sarcomas and carcinomas such as, but not limited to, fibrosarcoma, spot sarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, vascular sarcoma, endothelium, lymphangiosarcoma, lymphatic endothelial sarcoma , synovial tumor, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer, #巢癌, prostate cancer, squamous cell carcinoma, basal cell carcinoma, gland Cancer, sweat gland cancer, sebaceous gland cancer, papillary carcinoma ' papillary adenocarcinoma, cystadenocarcinoma, myeloid carcinoma, bronchial carcinoma, renal cell carcinoma, hepatocellular carcinoma, cholangiocarcinoma, squamous cell tumor, embryonal carcinoma, Wimm's tumor tumor, cervical cancer, sputum tumor, lung cancer, small cell lung cancer, bladder cancer epithelial cancer, glioma, astrocytoma, test cell tumor, Gu Yin e tumor to s membranous tumor, pine Adenoma, hemangioblastoma, acoustic neuroma's recruitment of dendritic glioma, cerebrospinal mineraloma, melanoma, neuroblastoma and retinal palsy, sparse-T-, ·, · cell tumor . Other cancers that can be hunted by the disclosed compositions of the invention include, but are not limited to, for example, Hodgki's disease (Hodgki heart DiSeaSe), non-Hodgkin's lymphoma (N〇nH〇dgkin, s Lymphoma), multiple !·生月赵瘤, neuroblastoma, breast cancer, ovarian cancer, lung cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, small cell lung tumor, primary brain tumor, stomach cancer (st_ch __), knot 158213.doc -20- 201210611 Intestinal cancer, malignant pancreatic insulinoma, malignant carcinoid 'bladder cancer, gastric cancer, premalignant skin lesions, testicular cancer, lymphoma, squamous cell carcinoma, nerve Blastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, cervical cancer, endometrial cancer, adrenal cortical cancer, and prostate cancer. "Nervous disease or condition" as used herein refers to any disease or condition of the nervous system and/or the visual system. "Nervous diseases or conditions" include diseases or conditions involving the middle sacral nervous system (brain, brainstem and cerebellum), the peripheral nervous system (including the cranial phrenic nerve), and the autonomic nervous system (partly located in the central nervous system and the peripheral nervous system). 0 neurological diseases or conditions include (but are not limited to) acquired epilepsy aphasia; acute disseminated encephalomyelitis; adrenal leukodystrophy; age-related macular degeneration; corpus callosum dysplasia; agnostic disorder, Ecardi syndrome (Aieardi syndrome); Alexander disease; Alpers (disease); alternating hemiplegia, Alzheimer's disease; vascular madness Q; amyotrophic lateral sclerosis ; no brain; Angelman syndrome; hemangiomatosis; anoxia; aphasia; use of no; arachnoid cyst; arachnoiditis; Anor〇-hiari malformation (Anr〇nl_ Chiari malformation); arteriovenous Malformation; AsperSei* syndrome; telangiectasia dysmotility; attention deficit hyperactivity disorder; autism Autonomic dysfunction; back pain; Batten disease, Behcet's disease; Bell's palsy; benign primary tendon; benign local muscle atrophy; benign cranial Within ifj pressure, Binswanger's disease; risk 158213.doc -21 * 201210611 Cancer 布 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛 洛Abscess; brain injury; brain tumor (including glioblastoma multiforme); vertebral tumor; Brown-Sequard syndrome; Canavan disease; carpal tunnel syndrome; burning pain Central pain syndrome; central cerebral spinal cord lysis; head disorder; cerebral aneurysm; cerebral arteriosclerosis; brain atrophy; cerebral giant disease; cerebral palsy; Chuck-Mali-Dos disease (Charc〇t_Marie_ Tooth disease); chemotherapy-induced neuropathy and neuralgia; Chiari malformation; chorea; chronic inflammatory demyelinating polyneuropathy; chronic pain; chronic regional pain syndrome; Coffin Lowry syndrome; coma, including persistent vegetative state; congenital bilateral flanks; cortical basal ganglia degeneration; cranial arteritis, craniosynostosis; Creutzfeldt_Jak〇b disease; Sexual injury disease; Cushing, s syndr〇me; giant cell inclusion disease; cell giant virus infection; dance eye-dance foot syndrome, Dandy Walk syndrome; Dawson disease De M〇rsieris syndrome; Dejerine_Klumke paisy; dementia, dermatomyositis; diabetic neuropathy; diffuse sclerosis; autonomic dysfunction; Dyslexia; dystonia; early infantile epilepsy; airborne sella syndrome

Syndr〇me);腦炎;腦膨出;腦三又神經血管瘤病;癲癇 症,厄爾布氏麻痺(Erb,s palsy);原發性顫抖病;法布立 氏病(Fabry,S disease);法爾氏症候群(Fahr,s syndrome); 158213.doc -22- 201210611 暈厥;家族性痙攣性麻痒;熱性癲癇發作;費雪氏症候群 (Fisher syndrome);弗里德賴希氏共濟失調(Friedreich's ataxia);額顳葉癡呆症及其他「滔蛋白病變(tauopathy)」; 高雪氏病(Gaucher's disease);格斯特曼氏症候群 (Gerstmann's syndrome);巨細胞性動脈炎;巨細胞性包涵 體病;球樣細胞性腦白質營養不良;古立安-白瑞症候群 (Guillain-Barre syndrome) ; HTLV-1相關性脊髓病變;哈 勒沃登-施帕茨病(Hallervorden-Spatz disease);頭部損 傷;頭痛;半面痙攣;遺傳性痙孿性截癱;多神經炎型遺 傳性運動失調;耳帶狀疱疹;帶狀疱疹;平山氏症候群 (Hirayama syndrome) ; HIV相關性癡呆症及神經病變(亦為 AIDS之神經表現);前腦無裂畸形;亨丁頓氏病 (Huntington's disease)及其他多糙醯胺酸重複序列病;積 水性無腦畸形;腦積水;高皮質醇症;低氧症;免疫介導 之腦脊髓炎;包涵體肌炎;色素失調症;嬰兒植烷酸蓄積 症;嬰兒雷夫蘇姆氏病(infantile refsum disease);嬰兒痙 攣症;發炎性肌病;顱内囊腫;顱内高壓;朱伯特症候群 (Joubert syndrome);吉姆-賽瑞症候群(Keams-Sayre syndrome);甘迺迪病(Kennedy disease);金斯布林納症候 群(Kinsboume syndrome);克利佩爾-費爾症候群(Klippel Feil syndrome);克拉伯病(Krabbe disease);庫格勃-韋蘭 德病(Kugelberg-Wel.ander disease);庫魯症(kuru);拉福拉 病(Lafora disease);蘭伯特-伊頓類重症肌無力症候群 (Lambert-Eaton myasthenic syndrome);蘭達-克萊夫納症 158213.doc -23- 201210611 候群(Landau-Kleffner syndrome);延髓背外側(瓦儉堡 (Wallenberg))症候群;學習困難;李氏疾病(Leigh's disease),連諾克斯-古斯塔症候群(Lenn〇x_Gustaut syndrome),萊施-奈恩症候群(Lesch-Nyhan syndrome);腦 白貝病變,路易體性癡呆(Lewy body dementia);無腦迴 症’閉鎖症候群;路葛雷克氏病(L〇u Gehrig's disease)(亦 即運動神經元病或肌萎縮性侧索硬化症);腰椎間盤疾 病,來姆病--神經性後遺症(Lyme disease--neurological sequelae) ’ 馬查多-約瑟夫病(Machado-Joseph disease);巨 腦(macrencephaly);巨腦症(megaiencephaly);梅爾克遜氏 症候群(Melkersson-Rosenthal syndrome);美尼爾病 (Menieres disease);腦膜炎;門克斯病(Menkes disease); 異染性腦白質營養不良;小頭畸形;偏頭痛;密勒費雪症 候群(Miller Fisher syndrome);短暫缺血發作;粒線體肌 病,莫比烏斯症候群(Mobius syndrome);單肢肌萎縮;運 動神經元病,煙霧病(Moyamoya disease);黏多餹症 (mucopolysaccharidoses);多發梗塞性癡呆症;多灶性運 動神經病變;多發性硬化症及其他脫髓鞘病症;多系統萎 縮症伴有體位性低血壓;肌肉萎縮症;重症肌無力;趙勒 脫失彌漫性硬化症;嬰兒肌陣攣性腦病變;肌陣攣;肌 病;先天性肌強直;發作性睡病;神經纖維瘤病;抗精神 病藥惡性症候群,AIDS之神經表現;狼療之神經性後遺 症;神經性肌強直;神經性類蠟脂褐質病;神經元遷移 病,尼曼匹克病(Niemann-Pick disease);歐沙利文-麥克里 158213.doc •24· 201210611 德症候群(O'Sullivan-McLeod syndrome);枕神經痛;潛在 性脊髓閉合不全序列症;大田原症候群(Ohtahara syndrome);撖欖橋腦小腦萎縮症;僵直性肌陣攣;視神 經炎;立位低血壓;過度使用症候群;感覺異常;神經退 化性疾病或病症(帕金森氏病(parkins〇nis disease)、亨丁頓 氏病、阿茲海默氏病、肌萎縮性侧索硬化症(ALS)、癡呆 症、多發性硬化症及其他與神經元細胞死亡相關之疾病及 病症);先天性肌強直病;副腫瘤病;陣發性發作;帕里 〇 羅姆伯格症候群(parry R〇mberg syndr〇nie);佩_梅氏病 (Pelizaeus-Merzbacher disease);週期性麻痺;周邊神經病 變;疼痛性神經病變及神經痛;持續性植物狀態;彌漫性 發育病症;光性喷嚏反射;植烷酸蓄積症;皮克氏病 (Pick's disease);神經挫傷;垂體瘤;多發性肌炎;孔洞 腦,小兒麻痺後症候群;帶狀疱疹神經痛;感染後腦脊髓 炎’體位性低金壓;普拉德威利症候群(prader_Willi ◎ syndrome);原發性脊髓側索硬化;朊病毒病;進行性面 偏側萎縮症;進行性多病灶性白質腦病;進行性硬化性灰 質萎縮;進行性核上麻痒;假性腦瘤;拉姆齊-亨特症候 群(Ramsay-Hunt syndrome)(I型及11型);拉斯馬森氏腦炎 (Rasmussen’s encephalitis);反射性交感神經營養不良症候 群;雷夫蘇姆氏病(Refsum disease);重複性運動病;重複 性壓力損傷;腿不寧症候群;反轉錄病毒相關脊髓病變; 蕾特氏症候群(Rett Syndrome);雷依氏症候群 syndrome);聖菲特舞蹈病(Saint vhus dance);山德霍夫 158213.doc -25- 201210611 病(Sandhoff disease);謝爾德氏病(Schilder,s disease);腦 裂;視隔發育不全;嬰兒搖籃症候群;帶狀疱疹;夏伊_ 德雷格症候群(Shy-Drager syndrome);修格蘭氏症候群 (Sjogren’s syndrome);睡眠呼吸暫停症;索特氏症候群 (Soto’s syndrome);痙攣;脊柱裂;脊髓損傷;脊髓腫 瘤;脊髓性肌萎縮;僵直人症候群;中風;斯特奇·韋伯 症候群(Sturge-Weber syndrome);亞急性硬化性全腦炎; 皮質下動脈硬化性腦病;西登哈姆舞蹈病(Sydenham chorea);昏厥;脊髓空洞症;遲發性運動困難;泰薩氏病 (Tay-Sachs disease);顳動脈炎;脊髓拴繫症候群;湯母 森氏病(Thomsen disease);胸部出口症候群;痛性抽搐; 托德氏麻痺(Todd’s paralySis);妥瑞症候群(T〇urette syndrome);短暫性缺血性發作;傳染性海綿狀腦病;橫 斷性脊髓炎;創傷性腦損傷;顫抖;三叉神經痛;熱帶痙 攣性輕截癍,結節性硬化症;企管性癡呆(多梗塞性癡 呆);血管炎,包括顳動脈炎;逢希伯_林道疾病(v〇n Hippel-Lindau disease);瓦倫伯格氏症候群(WaUenberg,s syndrome);韋德尼希-霍夫曼病(Werdnig_H〇ffman disease)’早斯特症候群(west syndrome);揮鞭病;威廉 氏症候群(Williams Syndrome);維爾頓氏病(Wild〇n,s disease);及澤韋格症候群(ZeUweger syndr〇me)。 「增生性疾病或病症」包括(但不限於)涉及由骨髓、淋 巴或紅jk球譜系或其前驅細胞引起之造血起源之增生/贅 生性細胞的造血贅生性病症。此等病症包括(但不限於)紅 158213.doc •26· 201210611Syndr〇me); encephalitis; brain swelling; brain trigeminal neurovascular disease; epilepsy, Erb, s palsy; primary trembling; Fabry, S Disease); Fahr, s syndrome; 158213.doc -22- 201210611 syncope; familial spasm; hot seizure; Fisher syndrome; Friedreich's ataxia; frontotemporal dementia and other "tauopathy"; Gaucher's disease; Gerstmann's syndrome; giant cell arteritis; Cellular inclusion body disease; globular cell leukodystrophy; Guillain-Barre syndrome; HTLV-1 associated myelopathy; Hallerwoden-Spatz disease (Hallervorden-Spatz Head injury; headache; hemifacial spasm; hereditary spastic paraplegia; polyneuritis-type hereditary ataxia; herpes zoster; herpes zoster; Hirayama syndrome; HIV-related dementia And neuropathy (also AIDS) Performance); forebrain non-cracking malformation; Huntington's disease and other multi-crown glutamate repeats; water-free no brain malformation; hydrocephalus; hypercortisolism; hypoxia; Guided encephalomyelitis; inclusion body myositis; pigmentation disorder; infant phytanic acid accumulation disease; infantile refsum disease; infantile spasm; inflammatory myopathy; intracranial cyst; intracranial High pressure; Joubert syndrome; Keams-Sayre syndrome; Kennedy disease; Kinsboume syndrome; Klipper-Fair syndrome (Klippel) Feil syndrome); Krabbe disease; Kugelberg-Wel.ander disease; Kuru; Lafora disease; Lambert-Eaton severe Lambert-Eaton myasthenic syndrome; Lanta-Clivena 158213.doc -23- 201210611 Landau-Kleffner syndrome; dorsolateral medulla (Wallenberg) syndrome; learning difficulties Li's disease (Leigh's disease), even Lenn〇x_Gustaut syndrome, Lesch-Nyhan syndrome; brain white shell disease, Lewy body dementia; Cerebral palsy's atresia syndrome; L〇u Gehrig's disease (also known as motor neuron disease or amyotrophic lateral sclerosis); lumbar disc disease, Lem disease - neurological sequelae Lyme disease--neurological sequelae) 'Machado-Joseph disease; macrencephaly; megaiencephaly; Melkersson-Rosenthal syndrome; Menil Menieres disease; meningitis; Menkes disease; metachromatic leukodystrophy; microcephaly; migraine; Miller Fisher syndrome; transient ischemic attack; Linear myopathy, Mobius syndrome; single limb muscle atrophy; motor neuron disease, moyamoya disease; mucopolysaccharidoses; multiple infarct dementia; multifocal Motor neuropathy; multiple sclerosis and other demyelinating disorders; multiple system atrophy with orthostatic hypotension; muscular dystrophy; myasthenia gravis; Zhao Le lost diffuse sclerosis; infant myoclonic brain lesions Myoclonus; myopathy; congenital myotonia; narcolepsy; neurofibromatosis; antipsychotic malignant syndrome, neurological manifestations of AIDS; neurological sequelae of wolf therapy; neuromuscular rigidity; neurogenic wax Lipofuscin; neuronal migration, Niemann-Pick disease; O'Sullivan-McLi 158213.doc •24· 201210611 O'Sullivan-McLeod syndrome; occipital neuralgia; potential Spiral insufficiency sequence disorder; Ohtahara syndrome; 撖 桥 脑 脑 ; ;; 僵 肌 挛; optic neuritis; standing hypotension; overuse syndrome; paresthesia; neurodegenerative diseases or disorders ( Parkins〇nis disease, Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), dementia, multiple sclerosis and His diseases and conditions related to neuronal cell death); congenital myotonia; paraneoplastic disease; paroxysmal seizure; parry R〇mberg syndr〇nie; Pei-Mei Disease (Pelizaeus-Merzbacher disease); periodic paralysis; peripheral neuropathy; painful neuropathy and neuralgia; persistent vegetative state; diffuse developmental disorders; photorefractive reflex; phytanic acid accumulation disease; Pick's disease); nerve contusion; pituitary tumor; polymyositis; hole brain, post-polio syndrome; herpes zoster neuralgia; post-infectious encephalomyelitis' orthostatic low gold pressure; Praderweili syndrome (prader_Willi ◎ syndrome Primary spinal cord lateral sclerosis; prion disease; progressive facial atrophy; progressive multifocal leukoencephalopathy; progressive sclerosing gray matter atrophy; progressive nucleus itching; pseudocephaloma; Ramsay-Hunt syndrome (type I and type 11); Rasmussen's encephalitis; reflex sympathetic dystrophy syndrome; Lefsum's disease Refsum disease; repetitive motion sickness; repetitive stress injury; leg restlessness syndrome; retrovirus-associated spinal cord disease; Rett Syndrome; Raytheon syndrome syndrome; St. Patrick's disease (Saint vhus) Dance); Sandefoff 158213.doc -25- 201210611 Disease (Sandhoff disease); Schilder's disease (Schilder, s disease); Brain splitting; Visual septal hypoplasia; Infant cradle syndrome; Herpes zoster; Shy-Drager syndrome; Sjogren's syndrome; sleep apnea; Soto's syndrome; sputum; spina bifida; spinal cord injury; spinal cord tumor; spinal muscular atrophy; Stiff human syndrome; stroke; Sturge-Weber syndrome; subacute sclerosing panencephalitis; subcortical arteriosclerotic encephalopathy; Sydenham chorea; fainting; syringomyelia Delayed exercise difficulty; Tay-Sachs disease; temporal arteritis; spinal cord syndrome; Thomsen disease; chest export syndrome Painful convulsions; Todd's paralysis; T〇urette syndrome; transient ischemic attack; infectious spongiform encephalopathy; transverse myelitis; traumatic brain injury; trembling; Neuralgia; tropical spastic paraplegia, tuberous sclerosis; vascular dementia (multiple infarct dementia); vasculitis, including temporal arteritis; 希希伯_林道病(v〇n Hippel-Lindau disease); Waulenberg syndrome (WaUenberg, s syndrome); Werdnig_H〇ffman disease 'west syndrome'; west whip; whiplash; Williams Syndrome; Wilder's disease (S disease); and Zeweger syndrome (ZeUweger syndr〇me). A "proliferative disease or condition" includes, but is not limited to, a hematopoietic neoplastic disorder involving a proliferative/neoplastic cell of hematopoietic origin caused by bone marrow, lymphoid or red jk spectroscopy or its precursor cells. These conditions include (but are not limited to) red 158213.doc •26· 201210611

Jk球母細胞白血病、急性前骨髓性白血病(APML)、慢性 骨髓性白血病(CML)、淋巴惡性疾病,包括(但不限於)急 性淋巴母細胞性白血病(ALL)(其包括B譜系ALL及T譜系 ALL)、慢性淋巴細胞性白血病(CLL)、幼淋巴細胞性白企 病(PLL)、毛細胞白血病(HLL)及瓦爾登斯特倫巨球蛋白灰 症(Waldenstrom’s macroglobulinemia)(WM))。其他形式之 惡性淋巴瘤包括(但不限於)非霍奇金淋巴瘤及其變異體、 周邊τ細胞淋巴瘤、成年τ細胞白血病/淋巴瘤(ATL)、皮膚 T細胞淋巴瘤(CTCL)、大顆粒淋巴細胞性白血病(LGF)、 霍奇金氏病及李特-斯頓伯格病(Reed-Sternberg disease;)。 「炎症」係指全身性發炎性病狀及局部與單核細胞、白 血球及/或嗜中性白血球遷移及吸引相關之病狀。炎症之 實例包括(但不限於)由感染病原性生物體(包括革蘭氏陽性 細菌(gram-positive bacteria)、革蘭氏陰性細菌(gram_ negative bacteria)、病毒、真菌及寄生蟲(諸如原蟲及蠕 蟲))、移植排斥反應(包括諸如腎臟、肝臟、心臟、肺或角 膜之實體器官之排斥反應,以及骨髓移植物之排斥反應, 包括移植物抗宿主疾病(GVHD)),或局部慢性或急性自體 免疫或過敏性反應引起之炎症。自體免疫性疾病包括急性 絲球體腎炎;類風濕性或反應性關節炎;慢性絲球體腎 炎;發炎性腸病,諸如克羅恩氏病(Crohn,s disease)、潰瘍 性結腸炎及壞死性小腸結腸炎;肝炎;敗血症;酒精性肝 病;非酒精性脂肪變性;粒細胞輸血相關症候群;發炎性 皮膚病,諸如接觸性皮膚炎、異位性皮膚炎、牛皮癬;全 158213,doc -27- 201210611 身性紅斑狼瘡(SLE)、自體免疫性曱狀腺炎、多發性硬化 症及一些形式之糖尿病,或個體自身免疫系統之攻擊造成 病理性組織破壞之任何其他自體免疫性病況。過敏性反應 包括過敏性哮喘、慢性支氣管炎、急性及延遲性過敏症。 全身性發炎性疾病病況包括與外傷、灼傷、缺血事件(例 如心臟、腦、腸或周邊血管結構中之血栓形成事件,包括 心肌梗塞及中風)後之再灌注' 敗血症、ARDS或多器官功 能障礙症候群相關之炎症。發炎性細胞募集亦出現在動脈 粥樣硬化斑中。發炎性細胞募集亦出現在動脈粥樣硬化斑 中。炎症包括(但不限於)非霍奇金氏淋巴瘤、韋格納氏肉 芽廬(Wegener’s granulomat〇sis)、橋本曱狀腺炎 (Hashimotc/s thyroiditis)、肝細胞癌、胸腺萎縮、慢性胰 臟炎、類風濕性關節炎、反應性淋巴增生、骨關節炎、潰 瘍性結腸炎、乳頭狀癌、克羅恩氏病、潰瘍性結腸炎、急 性膽囊炎、慢性膽囊炎 '肝硬化、慢性涎腺炎、腹膜炎、 急性騰臟炎、慢性胰臟炎、慢性胃炎、子宮腺肌症、子宮 内膜異位症、急性子宮頸炎、慢性子宮頸炎、淋巴增生、 多發性硬化症、特發性血小板減少性紫I繼發之肥大、原 、牛皮癖、肺氣腫、慢 性腎盂腎炎及慢性膀胱炎。 聚核苦酸及寡核普酸組合物及分子 尽# .·在一實施例中,目樟句人# a 曰保包含於鹼醯胺轉磷酸核糖基 酶(NAMPT)之核酸序列,包括(作尤κρ从、t 匕枯(但不限於)與NAMPT相關之 有義及/或反義非編竭及/或編碼序列。 158213.doc -28- 201210611 菸鹼醯胺轉磷酸核糖基酶(NAMPT)為使菸鹼醯胺轉化為 菸醯胺單核苷酸之酶,該菸醯胺單核苷酸藉由菸醯胺單核 苷酸轉腺苷醯基酶在哺乳動物生物合成路徑中轉化為菸醯 胺腺嘌呤二核苷酸(NAD+)。NAMPT為NAD+生物合成中之 限速因子。NAD+及其磷酸化及還原形式為氧化還原反應 ' 中接受氫化物及供應氫化物之辅酶,其在細胞代謝及能量 產生中具有中樞作用。NAD+亦能活化去乙醯基酶 (Sirtuins,SIRT),該等去乙醯基酶為組蛋白及許多轉錄因 〇 子之離胺酸去乙醯基酶。NAMPT定位於細胞内與細胞外 隔室中且在其中起作用。細胞内NAMPT產生NAD,且因 此具有細胞保護益處。NAMPT最初鑑別為表現於活化周 邊人類淋巴細胞中之基因產物,且與B細胞前驅體之成熟 有關聯,且因此稱為前B細胞增強因子。後續研究顯示, 在臨床及實驗性敗血症中,NAMPT在嗜中性白血球活化 期間上調且充當嗜中性白血球之抗細胞凋亡因子。在受刺 激單核細胞中且在全反式視黃酸(ATRA)誘導之骨髓性白血 〇 病細胞株HL-60之顆粒球性分化期間觀測到NAMPT含量增 加。NAMPT為NAD +產生之限速酶,其對於SIRT1活化及 轉錄調節必不可少。 • 在一實施例中,使用反義募核苷酸來預防或治療與 NAMPT家族成員相關之疾病或病症。可利用本發明之反 義募核苷酸及/或由使用及/或具有反義化合物獲得之幹細 胞再生之細胞/組織治療的例示性菸鹼醯胺轉磷酸核糖基 酶(NAMPT)介導之疾病及病症包含:與NAMPT之功能及/ 158213.doc •29- 201210611 或表現異常相關之疾病或病症、癌症、與細胞增殖相關或 以細胞增殖為特徵之疾病或病狀、細胞凋亡、與ΝΑΜρτ 之表現或功能突變或異常相關之疾病或病症、血細胞減少 症(例如骨髓或淋巴細胞系等之血細胞減少症)、嗜中性球 減少症(例如重度先天性嗜中性球減少症、其他形式之先 天性及後天性嗜中性球減少症、化學療法誘發之發熱性嗜 中性球減少症等)、白血病、急性骨髓性白血病、骨 髓細胞系之癌症、與NAMPT含量升高相關之惡性疾病、 關於造血之先天性非惡性疾病或病症、歸因於之 表現及/或活性改變之免疫缺乏症、動脈粥樣硬化、與葡 萄糖恆定異常相關之疾病或病症、與胰島素分泌異常相關 之疾病或病症、肥胖症、心血管疾病或病症、肝細胞调 亡、與粒線體生物發生異常相關之疾病或病症、與骨絡肌 相關之疾病、病症或病狀;與肌細胞生成異常相關之疾 病、病症或病狀;與FoXO基因家族相關之疾病、病症或病 狀;與肝臟三酸甘油醋值定異常相關之疾病或病症;與晝 夜節律活動相關之疾病、病症或病狀;與免疫系統相關之 疾病或病症;與關於内毒素耐受之後生性程式改寫 (epigenetic reprogramming)相關之疾病、病症或病狀;病 毒感染、與干擾素抗病毒反應異常相關之疾病或病症、慢 性發炎性疾病或病症(例如發炎性腸病,包括克羅恩氏病 (Crohn’s disease)、潰瘍性結腸炎、牛皮癬、關節炎等卜 血管疾病或病症(例如慢性潰癌、缺血性令風、心肌梗 塞、心絞痛及血管性癡呆等)、代謝性疾病或病症、炎 158213.doc -30- 201210611 症、衰老、非酒精性脂肪肝病、及與去乙醯基酶SIRT表現 及/或活性異常相關之疾病或病症。 在本發明之實施例中,為需要皮膚治療或有發生需要皮 膚治療之病狀之風險的個體提供治療及/或化妝方案及相 關調整治療。可例如基於個體之NAMPT狀態進行診斷。 在諸如皮膚之指定組織中患者之NAMPT表現量可藉由熟 習此項技術者已知及本文其他地方所描述之方法,例如藉 由使用基於PCR或抗體之偵測方法分析組織來測定。 〇 在一實施例中,將利用一或多種反義寡核苷酸進行之 ΝΛΜΡΤ調節投與有需要之患者以預防或治療與相較於正 常對照組NAMPT之表現、功能、活性異常相關的任何疾 病或病症。 在一實施例中,寡核苷酸對NAMPT之聚核苷酸具有特 異性,該聚核苷酸包括(但不限於)非編碼區。NAMPT目標 包含NAMPT之變異體;NAMPT之突變體,包括SNP ; ΝΛΜΡΤ之非編碼序列;對偶基因、片段及其類似物。寡 〇 核苷酸較佳為反義RNA分子。 根據本發明之實施例,目標核酸分子不限於單獨 ' NAMPT聚核苷酸,而且亦延及NAMPT之任何同功異型 • 物、受體、同源物、非編碼區及其類似物。 在一實施例中,寡核苷酸靶向NAMPT目標之天然反義 序列(編碼及非編碼區之天然反義序列),包括(但不限於) 變異體、對偶基因、同源物、突變體、衍生物、片段及其 互補序列之天然反義序列。募核苷酸較佳為反義RNA或 158213.doc -31- 201210611 DNA分子。 在一實施例中,本發明之募聚化合物亦包括在該化合物 中之一或多個核苷酸位置上存在不同鹼基的變異體。舉例 而έ,若第一核苷酸為腺嘌呤,則可產生在此位置含有胸 腺嘧啶核苷、鳥嘌呤核苷、胞嘧啶核苷或其他天然或非天 然核苷酸之變異體。此舉可在反義化合物之任何位置進 打。接著使用本文所述之方法測試此等化合物,以測定其 抑制目標核酸之表現的能力。 在一些實施例中,反義化合物與目標之間的同源性、序 列一致性或互補性為約5〇%至約6〇%。在一些實施例中, 同源性、序列一致性或互補性為約6〇%至約7〇%。在一些 實施例中,同源性、序列一致性或互補性為約7〇%至約 80〇/。。在一些實施例中,同源性 '序列一致性或互補性為 約80%至約90%。在一些實施例中,同源性、序列一致性 或互補性為約90%、約92%、約94%、約95%、約96%、約 97%、約 98%、約 99%或約 1〇〇%。 當化合物與目標核酸之結合干擾目標核酸之正常功能從 而導致活性降低,且存在足夠程度之互補性以避免在需要 特異性結合之條件下反義化合物與非目標核酸序列之非特 異性結合時’反義化合物可特異性雜交。該等條件包括, 亦即在活體内檢定或治療性處理之情況下的生理條件,及 在活體外檢定之情況下執行檢定之條件。 當化合物與目標DNA或RNA分子之結合干擾目標dna或 RNA之正常功能從而導致效用降低,且存在足夠程度之互 158213.doc •32- 201210611 補性以避免在需要特異性結合之條件下,亦即在活體内檢 定或治療性處理之情況下及在活體外檢定之情況下的生理 條件下在執行檢疋之條件下,反義化合物與非目標序列 之非特異性結合時,反義化合物(無論為DNA、RNA、嵌 合物、經取代物等)可特異性雜交。Jk blastoma leukemia, acute promyelocytic leukemia (APML), chronic myelogenous leukemia (CML), lymphoid malignancies including, but not limited to, acute lymphoblastic leukemia (ALL) (which includes B lineage ALL and T Lineage ALL), chronic lymphocytic leukemia (CLL), young lymphocytic leukemia (PLL), hairy cell leukemia (HLL), and Waldenstrom's macroglobulinemia (WM). Other forms of malignant lymphoma include, but are not limited to, non-Hodgkin's lymphoma and its variants, peripheral tau cell lymphoma, adult tau cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large Granulocyte lymphocytic leukemia (LGF), Hodgkin's disease, and Reed-Sternberg disease (Reed-Sternberg disease;). "Inflammation" refers to systemic inflammatory conditions and conditions associated with local migration and attraction of monocytes, leukocytes and/or neutrophils. Examples of inflammation include, but are not limited to, infection with pathogenic organisms (including gram-positive bacteria, gram_negative bacteria, viruses, fungi, and parasites (such as protozoa) And worms)), transplant rejection (including rejection of solid organs such as the kidneys, liver, heart, lungs or cornea, and rejection of bone marrow transplants, including graft versus host disease (GVHD)), or local chronic Or inflammation caused by acute autoimmune or allergic reactions. Autoimmune diseases include acute spheroid nephritis; rheumatoid or reactive arthritis; chronic glomerulonephritis; inflammatory bowel disease, such as Crohn's disease, ulcerative colitis, and necrosis Enterocolitis; hepatitis; sepsis; alcoholic liver disease; nonalcoholic steatosis; granulocyte transfusion related syndrome; inflammatory skin diseases such as contact dermatitis, atopic dermatitis, psoriasis; all 158213, doc -27- 201210611 Physical lupus erythematosus (SLE), autoimmune verrumitis, multiple sclerosis and some forms of diabetes, or any other autoimmune condition in which an individual's own immune system attacks cause pathological tissue damage. Allergic reactions include allergic asthma, chronic bronchitis, acute and delayed allergy. Systemic inflammatory conditions include reperfusion following sepsis, burns, or ischemic events (eg, thrombotic events in the heart, brain, intestine, or peripheral vascular structures, including myocardial infarction and stroke). Septicemia, ARDS, or multiple organ function Inflammation associated with disorder syndromes. Inflammatory cell recruitment also occurs in atherosclerotic plaques. Inflammatory cell recruitment also occurs in atherosclerotic plaques. Inflammation includes, but is not limited to, non-Hodgkin's lymphoma, Wegener's granulomat〇sis, Hashimotc/s thyroiditis, hepatocellular carcinoma, thymic atrophy, chronic pancreatitis , rheumatoid arthritis, reactive lymphoproliferative, osteoarthritis, ulcerative colitis, papillary carcinoma, Crohn's disease, ulcerative colitis, acute cholecystitis, chronic cholecystitis 'cirrhosis, chronic parotid gland Inflammation, peritonitis, acute otitis, chronic pancreatitis, chronic gastritis, adenomyosis, endometriosis, acute cervicitis, chronic cervicitis, lymphatic hyperplasia, multiple sclerosis, idiopathic thrombocytopenia Sexual purple I secondary to hypertrophy, original, psoriasis, emphysema, chronic pyelonephritis and chronic cystitis. Polynucleotide and oligonucleotide compositions and molecules are in one embodiment. In one embodiment, the target is a nucleic acid sequence comprising a base indoleamine phosphoribosyltransferase (NAMPT), including有 ρ 从 t t ( ( ( ( ( ( ( 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 158 NAMPT) An enzyme that converts nicotinamide to a nicotinamide mononucleotide by a nicotinamide single nucleotide transadenosylase in a mammalian biosynthetic pathway It is converted to nicotinamide adenine dinucleotide (NAD+). NAMPT is the rate-limiting factor in NAD+ biosynthesis. NAD+ and its phosphorylated and reduced form are redox reactions, which accept hydrides and coenzymes that supply hydrides. It has a central role in cell metabolism and energy production. NAD+ can also activate sirtuins (SIRT), which are histones and many transcriptional scorpions. Base enzymes. NAMPTs are localized in and function in the intracellular and extracellular compartments. Intracellular NAMP T produces NAD and thus has a cytoprotective benefit. NAMPT was originally identified as a gene product expressed in activated peripheral human lymphocytes and is associated with the maturation of B cell precursors, and is therefore referred to as pre-B cell potentiating factor. It is shown that in clinical and experimental sepsis, NAMPT is up-regulated during neutrophil activation and acts as an anti-apoptotic factor for neutrophils. In stimulated monocytes and in all-trans retinoic acid (ATRA) An increase in NAMPT content was observed during granule spheroid differentiation of induced myeloid leukemia cell line HL-60. NAMPT is a rate-limiting enzyme produced by NAD+, which is essential for SIRT1 activation and transcriptional regulation. The use of antisense raised nucleotides to prevent or treat diseases or conditions associated with NAMPT family members. Antisense nucleotides of the invention and/or stem cell regeneration obtained using and/or having antisense compounds can be utilized Exemplary nicotine-transamine phosphoribosyltransferase (NAMPT)-mediated diseases and disorders of cell/tissue therapy include: functions with NAMPT and / 158213.doc •29- 20121 0611 or a disease or condition associated with abnormal expression, cancer, a disease or condition characterized by cell proliferation or characterized by cell proliferation, apoptosis, a disease or condition associated with a mutation or abnormality in the expression or function of ΝΑΜρτ, cytopenia (eg cytopenia such as bone marrow or lymphocyte), neutropenia (such as severe congenital neutropenia, other forms of congenital and acquired neutropenia, chemotherapy induced Febrile neutropenia, etc.), leukemia, acute myeloid leukemia, cancer of bone marrow cell line, malignant disease associated with elevated NAMPT levels, congenital non-malignant disease or condition with hematopoiesis, attributed to Immunodeficiency, atherosclerosis, diseases or conditions associated with constant glucose abnormalities, diseases or conditions associated with abnormal insulin secretion, obesity, cardiovascular disease or condition, hepatocyte apoptosis, performance and/or activity change, a disease or condition associated with abnormal mitochondrial biogenesis, a disease, condition or condition associated with skeletal muscle; A disease, disorder, or condition associated with abnormal myogenesis; a disease, disorder, or condition associated with the FoXO gene family; a disease or condition associated with abnormality of the liver triglyceride; a disease associated with circadian rhythm activity, A disease or condition; a disease or condition associated with the immune system; a disease, disorder, or condition associated with epigenetic reprogramming after endotoxin tolerance; a viral infection, a disease associated with an abnormal antiviral response to interferon Or a condition, a chronic inflammatory disease or condition (eg, an inflammatory bowel disease, including Crohn's disease, ulcerative colitis, psoriasis, arthritis, etc., or a vascular disease or condition (eg, chronic ulceration, ischemia) Sexual wind, myocardial infarction, angina pectoris and vascular dementia, etc.), metabolic diseases or conditions, inflammation 158213.doc -30- 201210611 syndrome, aging, nonalcoholic fatty liver disease, and SIRT performance with deacetylase and / Or a disease or condition associated with an abnormal activity. In an embodiment of the invention, a treatment and/or makeup regimen and associated adjustment therapy are provided to an individual in need of skin treatment or at risk of developing a condition requiring skin treatment. The diagnosis can be made, for example, based on the individual's NAMPT status. The amount of NAMPT expression in a patient, such as a designated tissue of the skin, can be determined by methods known to those skilled in the art and described elsewhere herein, for example, by analyzing tissue using PCR-based or antibody-based detection methods. In one embodiment, sputum administration using one or more antisense oligonucleotides is administered to a patient in need thereof to prevent or treat any of the abnormalities associated with the performance, function, and activity of NAMPT in the normal control group. A disease or condition. In one embodiment, the oligonucleotide is specific for a polynucleotide of NAMPT, including but not limited to a non-coding region. NAMPT targets include variants of NAMPT; mutants of NAMPT, including SNPs; non-coding sequences of genomics; dual genes, fragments and analogs thereof. The oligodeoxynucleotide is preferably an antisense RNA molecule. According to an embodiment of the invention, the target nucleic acid molecule is not limited to the single 'NAMPT polynucleotide, but also extends to any isoforms of NAMPT, receptors, homologs, non-coding regions and analogs thereof. In one embodiment, the oligonucleotide targets a natural antisense sequence of a NAMPT target (a natural antisense sequence encoding a non-coding region), including but not limited to variants, dual genes, homologs, mutants Natural antisense sequences of derivatives, fragments, and their complementary sequences. The nucleotide raised is preferably an antisense RNA or a 158213.doc -31-201210611 DNA molecule. In one embodiment, the polymeric collection compounds of the present invention also include variants in which different bases are present at one or more nucleotide positions in the compound. For example, if the first nucleotide is adenine, a variant containing thymidine, guanosine, cytosine or other natural or non-natural nucleotides at this position can be produced. This can be done anywhere in the antisense compound. These compounds are then tested using the methods described herein to determine their ability to inhibit the performance of the target nucleic acid. In some embodiments, the homology, sequence identity or complementarity between the antisense compound and the target is from about 5% to about 6%. In some embodiments, the homology, sequence identity, or complementarity is from about 6% to about 7%. In some embodiments, homology, sequence identity or complementarity is from about 7% to about 80%. . In some embodiments, the homology 'sequence identity or complementarity is from about 80% to about 90%. In some embodiments, the homology, sequence identity, or complementarity is about 90%, about 92%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 1〇〇%. When the binding of the compound to the target nucleic acid interferes with the normal function of the target nucleic acid, resulting in reduced activity, and a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to the non-target nucleic acid sequence under conditions requiring specific binding' Antisense compounds can specifically hybridize. Such conditions include, i.e., physiological conditions in the case of in vivo assay or therapeutic treatment, and conditions for performing assays in the case of in vitro assays. When a compound binds to a target DNA or RNA molecule to interfere with the normal function of the target DNA or RNA, resulting in reduced utility, and a sufficient degree of complementation to avoid the need for specific binding, That is, in the case of in vivo assay or therapeutic treatment and under physiological conditions in the case of in vitro assay, the antisense compound is non-specifically bound to the non-target sequence under the conditions of performing the assay, the antisense compound ( Whether it is DNA, RNA, chimera, substituted, etc., it can specifically hybridize.

在一實施例中,靶向NAMPT(包括(但不限於)使用例如 PCR、雜交等所鑑別及擴增之反義序列,亦即一或多種如 SEQ ID NO. 2至16所示之序列及其類似物)可調節NAMpT 〇 之表現或功能。在-實施例中,與對照組相比表現或功能 上調。在一實施例中,與對照組相比表現或功能下調。 在一實施例中,寡核苷酸包含如SEQ m N〇: 17至31所 示之核酸序列,包括使用例如PCR、雜交等所鑑別及擴增 之反義序列。此等寡核苷酸可包含一或多個經修飾之核苷 酸、較短或較長片段、經修飾之鍵及其類似物。經修飾之 鍵或核苷酸間鍵聯之實例包含硫代磷酸酯、二硫代磷酸酯 ❹ 或其類似物。在一實施例令,核苷酸包含磷衍生物。可連 接至本發明之經修飾寡核苷酸中之糖或糖類似物部分的礎 衍生物(或經修飾之磷酸酯基)可為單磷酸酯、二磷酸酯、 三磷酸酯、烷基磷酸酯、烷磷酸酯、硫代磷酸酯及其類似 基團。上述磷酸酯類似物之製備及其併入核苷酸、經修飾 之核音酸及券核皆酸中本身亦已知且本文無需描述。 熟習此項技術者亦利用反義序列之特異性及敏感性用於 治療性用途。反義寡核苷酸已用作動物及人類中疾病病況 治療中之治療性部分。已將反義募核苷酸安全且有效地投 158213.doc •33- 201210611 與人類’且目前正在進行許多臨床試驗。因此衫,募核 苷酸可為適用治療形態’其可經組態以適用於治療細胞、 組織及動物、尤其人類之治療方案。 在本發明之實施例中’募聚反義化合物,尤其募核苷酸 結合目標核酸分子且調節由目標基因編碼之分子的表現及/ 或功能。待f·擾之DNA之功能包含例如複製及轉錄。待干 擾之RNA之功能包含所有重要功能,諸如rna移位至蛋白 質轉譯位點、蛋白質自RNA轉譯、剪接RNA產生一或多種 mRNA物f種類,及RNA可參與或可促以催化活性。視 所需功能而定’功能可上調或抑制。 反義化合物包括反義寡聚化合物、反義寡核苷酸、外部 引導序mEGS)募核芽酸、替代剪接子、引子、探針及與 目標核酸之至少一部分雜交之其他募聚化合物。因而,可 引入呈單股、雙股、部分單股或環形寡聚化合物形式之此 等化合物。 在本發明之上下文中,使反義化合物乾向特定核酸分子 可為多步驟方法。該方法通常始於㈣將調節功能之目標 核酸。此目標核酸可為例如表現與特定病症或疾病病況: 關之細胞基因(或自基因轉錄之mRNA),或來自感染物之 核酸分子。在本發明中’目標核酸編碼祕醯胺轉鱗酸核 糖基酶(NAMPT)。 乾向方法通常亦包括確定用於進行反義相互作用之目桿 核酸内之至少-個目標區、區段或位點,以使得將產生: 如調節表現之所需作用。在本發明之上下文内,術語 158213.doc -34 - 201210611 區」定義為目標核酸中 具有至少一個可鑑別結構、功能 一 刀。區段在目標核酸之區内。「區段」定義為 目標«内之區的較小或子部分。如本發明中所用之「位 點」疋義為目標核酸内之位置。In one embodiment, targeting NAMPT (including, but not limited to, antisense sequences identified and amplified using, for example, PCR, hybridization, etc., ie, one or more sequences as set forth in SEQ ID NO. 2 to 16 and Its analogs) can modulate the performance or function of NAMpT. In the examples, the performance or function was up-regulated compared to the control group. In one embodiment, the performance or function is down-regulated compared to the control group. In one embodiment, the oligonucleotide comprises a nucleic acid sequence as set forth in SEQ m N〇: 17 to 31, including antisense sequences identified and amplified using, for example, PCR, hybridization, and the like. Such oligonucleotides may comprise one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. Examples of modified bonds or internucleotide linkages include phosphorothioates, phosphorodithioates, or the like. In one embodiment, the nucleotide comprises a phosphorus derivative. The base derivative (or modified phosphate group) which can be attached to the sugar or sugar analog moiety of the modified oligonucleotide of the present invention may be a monophosphate, a diphosphate, a triphosphate, an alkyl phosphate Esters, alkyl phosphates, phosphorothioates and the like. The preparation of the above phosphate analogs and their incorporation into nucleotides, modified nuclear acid and nucleic acid are also known per se and need not be described herein. Those skilled in the art also utilize the specificity and sensitivity of antisense sequences for therapeutic use. Antisense oligonucleotides have been used as therapeutic moieties in the treatment of disease conditions in animals and humans. Antisense nucleotides have been safely and efficiently administered to 158213.doc •33-201210611 with humans and many clinical trials are currently underway. Thus, a shirt, a nucleoside acid can be a suitable therapeutic modality' that can be configured to be useful in the treatment of cells, tissues, and animals, particularly humans. In an embodiment of the invention, an antisense compound is recruited, particularly a nucleotide that binds to a target nucleic acid molecule and modulates the expression and/or function of the molecule encoded by the gene of interest. The function of the DNA to be disturbed includes, for example, replication and transcription. The function of the RNA to be disturbed includes all important functions, such as translocation of the rna to the protein translation site, translation of the protein from the RNA, splicing of the RNA to produce one or more mRNA species, and RNA may participate in or may promote catalytic activity. Depending on the function required, the function can be adjusted up or down. Antisense compounds include antisense oligomeric compounds, antisense oligonucleotides, external leader mEGS) nucleomonic acid, alternative splicing, primers, probes, and other polymeric compounds that hybridize to at least a portion of the target nucleic acid. Thus, such compounds can be introduced in the form of single-stranded, double-stranded, partially mono- or cyclic oligomeric compounds. In the context of the present invention, the drying of an antisense compound to a particular nucleic acid molecule can be a multi-step process. This method usually begins with (iv) the target nucleic acid that will regulate the function. The target nucleic acid can be, for example, a nucleic acid molecule that exhibits a specific cell or disease condition: a cellular gene (or mRNA transcribed from a gene), or from an infectious agent. In the present invention, the target nucleic acid encodes a secret amine trans-luciferase (NAMPT). The dry direction method also generally includes determining at least one target region, segment or site within the rod nucleic acid for performing the antisense interaction such that a desired effect, such as to modulate performance, is produced. Within the context of the present invention, the term 158213.doc -34 - 201210611 region is defined as having at least one identifiable structure, function, and knives in a target nucleic acid. The segment is within the region of the target nucleic acid. A "segment" is defined as a smaller or sub-portion of the zone within the target «. The "site" as used in the present invention is defined as the position within the target nucleic acid.

在實施例中,反義寡核苷酸結合菸鹼醯胺轉磷酸核糖 土酶(ΝΑΜΡΤ)之天然反義序列且調節NAMpT(SEQ i)之表現及/或功能。反義序列之實例包括seqidn〇· 2至 31°In an embodiment, the antisense oligonucleotide binds to the natural antisense sequence of nicotinamide to phosphoribosidase (ΝΑΜΡΤ) and modulates the expression and/or function of NAMpT (SEQ i). Examples of antisense sequences include seqidn〇·2 to 31°

在實鉍例中,反義寡核苷酸結合於鹼醯胺轉鱗酸核糖 基酶(nAMPT)聚核苷酸之一或多個區段且調節ΝΑΜρτ之 表現及/或功能。該等區段包含ΝΑΜΡΤ有義或反義聚核苷 酸之至少5個連續核苷酸。 在一實施例中,反義寡核苷酸對ΝΑΜΡΤ之天然反義序 列具有特異性’其中寡核苷酸與NAmpt之天然反義序列 之結合調節ΝΑΜΡΤ之表現及/或功能。 在一實施例中,寡核苷酸化合物包含如SEQ ID NO: 17 至3 1所示之序列,亦即使用例如PCR、雜交等所鑑別及擴 增之反義序列。此等寡核苷酸可包含一或多個經修飾之核 苷酸、較短或較長片段、經修飾之鍵及其類似物。經修飾 之鍵或核苷酸間鍵聯之實例包含硫代磷酸酯、二硫代磷酸 酯或其類似物。在一實施例中,核苷酸包含填衍生物。可 連接至本發明之經修飾募核苷酸中之糖或糖類似物部分的 磷衍生物(或經修飾之磷酸酯基)可為單磷酸酯、二磷酸 酯、三碟酸酯、烷基磷酸酯、烷磷酸酯、硫代磷酸酯及其 l5S2B.doc -35- 201210611 類似基團。上述磷酸醋類似物之製備及其併入核普酸、妹 修飾之核普酸及寡核普酸中本身亦已知且本文無需描述二 為如此項技術中所已知,轉譯起始 綱(在轉錄之m讓分子中;在相應麵分子中為為55: ATG),所以轉譯起始密碼子亦稱為「娜密碼子」、In the present example, an antisense oligonucleotide binds to one or more segments of a base amide trans-luciferase (nAMPT) polynucleotide and modulates the expression and/or function of ΝΑΜρτ. The segments comprise at least 5 contiguous nucleotides of a purine sense or antisense polynucleotide. In one embodiment, the antisense oligonucleotide is specific for the natural antisense sequence of deuterium' wherein the combination of the oligonucleotide and the natural antisense sequence of NAmpt modulates the expression and/or function of purine. In one embodiment, the oligonucleotide compound comprises the sequence set forth in SEQ ID NOs: 17 to 31, i.e., an antisense sequence identified and amplified using, for example, PCR, hybridization, and the like. Such oligonucleotides may comprise one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. Examples of modified linkages or internucleotide linkages include phosphorothioates, dithiophosphates, or the like. In one embodiment, the nucleotide comprises a fill derivative. The phosphorus derivative (or modified phosphate group) which can be attached to the sugar or sugar analog moiety of the modified nucleotide of the present invention may be a monophosphate, a diphosphate, a tri-plate acid ester, an alkyl group. Phosphate esters, alkyl phosphates, phosphorothioates and their similar groups of l5S2B.doc-35-201210611. The preparation of the above-mentioned phosphate vinegar analogs and their incorporation into nucleotide acid, sister-modified nucleotides and oligonucleotides are also known per se and need not be described herein. The translation initiation scheme is known in the art. In the transcribed m let the molecule; in the corresponding face molecule is 55: ATG), so the translation initiation codon is also called "naco codon",

始密碼子」或「綱起始密碼子」。少數基因具有有RNA 序列5 GUG、5'-UUG或5'-CUG之轉譯起始密碼子;且已 顯不 5 -AUA、Ό /^ττβ 丄 或5 -CUG在活體内起作用。因此, 術語「轉課起始密碼子」及「起始密碼子」可涵蓋許多密 Π列,但在各情況下起始胺基酸通常為甲硫胺酸(在 一核生物中)或甲醢甲硫胺酸(在原核 基因可具有兩個或兩個以上替代性起始密碼子 胞類型或組織中或在一組特定條件下可優先利 :進起始。在本發明之上下文中,「起始密碼子」 活體=起始密碼子」係指與該等密碼子之序列無關,在 以起始由編碼菸鹼醯胺轉磷酸核糖基酶(ναμρ 之基因轉錄得到之mRNA之轉 —「終止密碼子」)可 -ΤΛΑ〇Α, 5,_UGA(^ ^ DNA^ ^ ^ ^ ^ —Λ ^ 術語「起始密碼子區」及「轉譯起始密 種―或基財涵蓋在自轉譯起始密碼子之任亥 或)上約25至約50個連續核普酸之 語「終止密碼子區」及「轉,政止…「類似地’術 ㈣終止㈣子區」係指 158213.doc -36- 201210611 mRNA或基时涵蓋自轉譯終止密碼子之任—方向(亦即5, 或3,)上約25至約5G個連續核㈣之部分1此,「起始密 碼子區」(或「轉譯起始密碼子區」)及「終止密碼子區」 (或「轉譯終止密碼子區」)為可用本發明之反義化合物有 效靶向之所有區。 此項技術中已知指轉譯起始密碼子與轉譯終止密碼子之 間的區之開放閲讀框架(ORF)或「編碼區」亦為可有效靶 Ο 向之區。在本發明之上下文内,目標區為涵蓋基因之開放 閱讀框架(GRF)之轉譯起始密碼子或轉譯終止密碼子的基 因内區。 另-目標區包括此項技射已知指mRNA巾自轉譯起始 密碼子之5,方向上之部分的5,非轉譯區(5.utr),且因此包 括mRNA之5,帽位點與轉譯起始密碼子之間的料酸(或基 因上之相應核普酸)。另一目標區包括此項技術中已知指 _A中自轉譯終止密碼子之3,方向上之部分❸,非轉譯區 Q (3’UTR),且因此包括mRNA之轉譯終止密碼子與3,_端之間 的核苷酸(或基因上之相應核苷酸)。mRNA25,帽位點包含 經由5’-5’三磷酸酯鍵聯接合至mRNA2 51最末端殘基的Ν' 甲基化鳥嘌呤核苷殘基。認為mRNAi5,帽區包括5,帽結構 本身,以及鄰近於帽位點之前50個核苷酸。本發明之另一 目標區為5'帽區。 儘管一些真核mRNA轉錄物將直接轉譯,但許多含有稱 為内含子」之一或多個區,在轉錄物轉譯之前將其自轉 錄物切除。剩餘(且因此轉譯之)區稱為「外顯子」且剪接 158213.doc -37· 201210611 到一起形成連續mRNA序列。在一實施例中,在異常剪接 與疾病有關或特定剪接產物之過量產生與疾病有關之情形 下,靶向剪接位點(亦即内含子_外顯子接合點或外顯子_内 含子接合點)尤其適用。歸因於重排或缺失之異常融合接 合點為目標位點之另一實施例。經由自不同基因源剪接兩 種(或兩種以上)mRNA之過程所產生之mRNA轉錄物稱為 「融合轉錄物」。可使用靶向例如DNA或前體mRNA之反 義化合物有效靶向内含子。 在一實施例中,反義寡核苷酸結合目標聚核苷酸之編碼 及/或非編碼區且調節目標分子之表現及/或功能。 在一實施例中,反義寡核苷酸結合天然反義聚核苷酸且 調節目標分子之表現及/或功能。 在一實施例中,反義募核苷酸結合有義聚核苷酸且調節 目標分子之表現及/或功能。 替代性RNA轉錄物可由DNA之同一基因組區產生。此等 替代性轉錄物一般稱為「變異體」。更特定言之,「前體 mRNA變、異體」為由同一基因組⑽八產生之轉錄物,其在 其起始或終止位置與由同一基因組DNA產生之其他轉錄物 不同且含有内含子與外顯子序列。 在剪接期間切除一或多個外顯子或内含子區或其部分 %,刖體mRNA變異體產生較小r mRNA變異體」。因此, mRNA變異體為經加卫之前體mRNA變異體,且各獨特前 體mRNA變異體由於剪接而必須始終產生獨特mRNA變異 體。此等mRNA變異體亦稱為「替代性剪接變異體」。若不 158213.doc -38· 201210611 存在前體mRNA變異體之剪接,則前體mRNA變異體與 mRNA變異體相同。 變異體可經由使用替代性信號以起始或終止轉錄而產 生。前體mRNA及mRNA可具有一個以上起始密碼子或終 止密碼子。來源於使用替代性起始密碼子之前體mRNA* mRNA的變異體稱為該前體mRNA或mRNA之「替代性起始 變異體」。使用替代性終止密碼子之彼等轉錄物稱為前體 mRNA或mRNA之「替代性終止變異體」。一特定類型之替 〇 代性終止變異體為「polyA變異體」,其中多種所產生之轉 錄物由藉由轉錄機構替代性選擇rpolyA終止信號」中之 一者,由此產生終止於獨特polyA位點之轉錄物而產生。 在本發明之上下文内,本文所述變異體之類型亦為目標核 酸之實施例。 反義化合物所雜交之目標核酸上之位置定義為活性反義 化合物所靶向之目標區之至少長5個核苷酸之部分。 Q 儘管某些例示性目標區段之特定序列在本文中闡述,但 熟習此項技術者將認識到此等序列係用以說明及描述本發 明範疇内的特定實施例。其他目標區段容易由一般技術者 根據本發明來鑑別。 認為包含選自說明性較佳目標區段之一段至少5(五)個 連績核苷酸的長5至100個核苷酸之目標區段亦適用於靶 向。 目標區段可包括DNA或RNA序列,其至少包含5個來自 一個說明性較佳目標區段之5,_端的連續核苷酸(剩餘核苷 158213.doc -39- 201210611 酸為自緊接目標區段之5'_端上游開始且連續直至DNA或 RNA含有約5至約1〇〇個核苷酸的相同DNA4 RNA之連續 段)。類似地,較佳目標區段由〇]^人或汉^^入序列表示,其 至少包含5個來自一個說明性較佳目標區段之3,_端的連續 核苷酸(剩餘核苷酸為自緊接目標區段之3,_端下游開始且 連續直至DNA或RNA含有約5至約1〇〇個核苷酸的相同1)]^八 或RNA之連續段)。熟習提供本文所示目標區段之技術者 將能夠在無不當實驗下鑑別其他較佳目標區段。 鑑別出一或多個目標區、區段或位點後,即選擇與目標 充分互補,亦即充分雜交且具有足夠特異性之反義化合 物’以得到所需作用。 在本發明之實施例中,募核苷酸結合特定目標之反義 紅券核苷I長度為至少5個核苷酸且可合成以使各寡核 苷酸靶向重疊序列,以使得所合成之寡核苷酸涵蓋目標聚 核普酸之全部長度。目標亦包括編碼以及非編碼區。 在一實施例中,較佳利用反義寡核苷酸靶向特定核酸。 使反義化D物靶向特定核酸為多步驟方法。該方法通常始 於鑑別將調節功能之核酸序列。&目標核酸可為例如表現 與特疋病症或疾病病況相關之細胞基因(或自基因轉錄之 mRNA)或非編碼聚核:^酸,諸如非編碼。 可將RNA歸類為(1)信使RNA(mRNA),其會轉譯成蛋白 質,及⑺非蛋白質編碼RNA(nCRNA)。ncRNA包含微 RN^、反義轉錄物及含有高密度終止密碼子且缺乏任何廣 泛開放閱讀框架」之其他轉錄單元(τυ)。許多ncRNA似 158213.doc 201210611 乎自蛋白質編碼基因座的3’非轉譯區(3·υτιι)令之起始位點 起始。ncRNA通常罕見,且至少一半已由FANT〇M協會定 序之ncRNA似乎未聚腺苷酸化。由於顯而易見之原因,大 多數研九者關注加工且輸出至細胞質中之聚腺苷酸化 mRNA。近來,顯示非聚腺苷酸化細胞核RNA之集合可能 極大’且許多該等轉錄物由所謂基因間區產生^ ncRNA可 調節基因表現之機制為與目標轉錄物進行鹼基配對。藉由 鹼基配對起作用之RNA可分組為(1)順式編碼RNA,其在相 〇 同遺傳位置編碼,但在與RNA相對之股上起作用且因此顯 不與其目標完全互補;及(2)反式編碼尺]^人,其在與111<[八不 同之染色體位置編碼,對該RNA起作用,且一般不展現與 其目標之完全驗基配對可能性。 不希望文理綸束缚,利用本文所述之反義募核苷酸干擾 反義聚核苷酸可改變相應有義信使RNA之表現。然而,此 調即可能並不一致(反義敲除引起信使111^八增多)或可能一 q 致(反義敲除引起伴隨信使RNA減少)。在此等情況下,反 義募核苷酸可靶向反義轉錄物之重疊或不重疊部分,引起 敲除或螯隔。可以相同方式靶向編碼以及非編碼反義序 列,任一種類均能夠以一致或不一致方式調節相應有義轉 錄物。用於鑑別供針對目標使用之新穎募核苷酸之策略可 基於利用反義寡核苷酸敲除反義RNA轉錄物或調節所需目 標之任何其他方式。 衷喀7 :在不一致調節之情況下,敲除反義轉錄物會增 加習知(有義)基因之表現。若習知(有義)基因編碼已知或 158213.doc •41 · 201210611 假疋藥物目‘則敲除其反義對應物可想像為模擬受體促 效劑或酶刺激物之作用。 衷略广·在一致調節之情況下,可伴隨敲除反義與有義 轉錄物且由此達成習知(有義)基因表現之協同降低。舉例 而言,若使用反義寡核苷酸達成敲除,則可使用此策略來 應用於乾向有義轉錄物之反義寡核普酸及㈣相應反義轉 錄物之另-反義寡核苷酸’或同時靶向重疊有義及反義轉 錄物之單一能量對稱反義寡核苷酸。 /艮據本發明,反義化合物包括反義寡核芽酸、核糖核酸 酶外引導序列(EGS)募核苷酸、siRNA化合物、單股 =雙股RNA干擾(RNAi)化合物(諸如仙财化合物)及與目 標核酸之至少一部分雜交且調節其功能之其他募聚化合 物。因而’其可為DNA、RNA、類職、類職或其混合 物,或可為其中-或多者之模擬物。此等化合物可為單 股、雙股、環形或髮夾寡聚化合物且可含有結構元件,諸 如内部或末端突起、錯配或環。通常製備線性反義化合 物,但可接合或以其他方式製備成環形及/或分支形。: 義化合物可包括構築體’諸如雜交形成完全或部分雙 :::兩個股’或具有足夠自身互補性以允許雜; =分雙股化合物之單股。兩個股可内部連接從而Ϊ :游離3’或5,末端,或可連接形成連續髮炎結構或 "3末…有穴出物’產生單股特徵之延 長。雙k化合物視情況可在末端包括突出物 包括連接至-個末端、所選Μ酸位置、糖位置^傅可 嗯证置4 —個核 158213.doc -42· 201210611 普間鍵聯的結合基團。或者,兩個股可經由非核酸部分或 連接子基團連接。當僅由_個股形成時,dsRNA可呈自身 對折形成雙鏈體之自身互補髮㈣分子之形式。因此, dsRNA可呈完全或部分雙股形式。基因表現之特異性調節 可猎由轉殖基因細胞株中献财髮《之穩定表現達成,然 而’在一些實_中,上調基因表現或功能。#由兩個: 或呈本身對折形成雙鏈體之自身互補髮夾型分子形式之單 股形成時,兩個股(或單股之雙鏈體形成區)為鹼基以華特 生-克里克方式配對之互補RNA股。 引入系統中後,本發明化合物可引發一或多種酶或結構 蛋白質起作用以影響目標核酸之裂解或其他修飾,或可經 由基於占位之機制起作用。一般而言,核酸(包括募核苷 酸)可描述為「類DNA」(亦即一般具有一或多個2,_去氧 糖,且一般具有T而非ϋ鹼基)或「類RNA」(亦即一般具有 或多個2’-羥基糖或2’-修飾糖,且一般具有〇而非τ驗 基)。核酸螺旋可採用一種以上類型之結構,最通常為A形 及B形。咸信,一般而言,具有B形樣結構之募核苷酸為 「類DNA」,且具有八形樣結構之寡核苷酸為「類RNA」。 在一些(喪合)實施例中,反義化合物可含有A形區與b形 區。 在一實施例中’所需寡核苷酸或反義化合物包含以下中 之至少一者:反義RNA、反義DNA、嵌合反義募核苷酸、 包含經修飾鍵聯之反義募核苷酸、干擾RNA(RNAi)、短干 擾RNA(siRNA);微干擾RNA(miRNA);小時序RNA(stRNA); 158213.doc -43- 201210611 或短髮夾RNA(shRNA);小RNA誘導之基因活化(RNAa); 小活化RNA(saRNA),或其組合。 dsRNA亦可活化基因表現,亦即一種稱為「小RNA誘導 之基因活化」或RNAa之機制。靶向基因啟動子之dsRNA 誘導相關基因之有效轉錄活化。在人類細胞中已使用稱為 「小活化RNA」(saRNA)之合成dsRNA證明RNAa。目前尚 未知RNAa是否在其他生物體中保留。 已發現諸如小干擾RNA(siRNA)及微RNA(miRNA)之小雙 股RNA(dsRNA)為稱為RNA干擾(RNAi)之進化保守機制之 觸發物。RNAi總是經由重塑染色質由此抑止轉錄、降解 互補mRNA或阻斷蛋白質轉譯來引起基因靜止。然而,在 隨後之實例部分中詳細插述之情況下,顯示募核苷酸會增 加菸鹼醢胺轉磷酸核糖基酶(NAMPT)聚核苷酸及其編碼產 物之表現及/或功能^ dsRNA亦可充當小活化 RNA(saRNA)。不希望受理論束缚,在稱為dsRNA誘導之 轉錄活化(RNAa)之現象中,藉由靶向基因啟動子中之序 列’ saRNA將誘導目標基因表現。 在另一實施例中’本文所鑑別之「較佳目標區段」可用 於篩檢調節菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之 表現的其他化合物。「調節劑」為降低或增加編碼NAMPT 之核酸分子之表現且包含至少一個與較佳目標區段互補之 5核苷酸部分的彼等化合物。篩檢方法包含以下步驟:使 編碼NAMPT之有義或天然反義聚核苷酸的核酸分子之較 佳目標區段與一或多種候選調節劑接觸,及選擇一或多種 158213.doc -44- 201210611 降低或增加編碼NAMPT聚核苷酸之核酸分子(例如SEQ ID NO: 17至31)的表現之候選調節劑。一旦顯示候選調節劑 能夠調節(例如降低或增加)編碼NAMPT聚核苷酸之核酸分 子的表現,則可將該調節劑用於NAMPT聚核苷酸之功能 的其他調查研究中或用作本發明之研究、診斷或治療劑。 ' 靶向天然反義序列較佳會調節目標基因之功能。例如 NAMPT基因(例如寄存編號NM_005746)。在一實施例中, 目標為NAMPT基因之反義聚核苷酸。在一實施例中,反 〇 義寡核苷酸靶向NAMPT聚核苷酸(例如寄存編號 NM_005746)之有義及/或天然反義序列、其變異體、對偶 基因、同功異型物、同源物、突變體、衍生物、片段及互 補序列。寡核苷酸較佳為反義分子,且目標包括反義及/或 有義NAMPT聚核苷酸之編碼及非編碼區。 本發明之較佳目標區段亦可與本發明之其各別互補反義 化合物組合,形成穩定雙股(雙鏈體)寡核苷酸。 在此項技術中該等雙股寡核苷酸部分已顯示經由反義機 〇 制調節目標表現且調節轉譯以及RNA加工。此外,雙股部 分可經受化學修飾。舉例而言,該等雙股部分已顯示藉由 雙鏈體之反義股與目標之經典雜交由此觸發目標之酶促降 ' 解來抑制目標。 在一實施例中,反義寡核苷酸靶向菸鹼醯胺轉磷酸核糖 基酶(NAMPT)聚核苷酸(例如寄存編號NM_005746)、其變 異體、對偶基因、同功異型物、同源物、突變體、衍生 物、片段及互補序列。寡核苷酸較佳為反義分子。 158213.doc -45- 201210611 根據本發明之實施例,目標核酸分子不限於單獨 NAMPT,而且亦延及其任何聚核苷酸變異體及產生、作 用、影響、或導致或與NAMPT表現產物相關之任何聚核 普酸及/或其任何同功異型物。 在一實施例中,寡聚核苷酸靶向NAMPT聚核苷酸之天 然反義序列,例如如SEQ ID NO: 2至16所示之聚核苷酸及 其任何變異體、對偶基因、同源物、突變體、衍生物、片 段及互補序列。反義寡核苷酸之實例如SEQ ID NO: 17至 31中所示。 在一實施例中,寡核苷酸與NAMPT反義序列之核酸序 列互補或結合,該等序列包括(但不限於)與NAMPT聚核苷 酸相關之非編碼有義及/或反義序列;且調節NAMPT分子 之表現及/或功能。 在一實施例中,寡核苷酸與如SEQ ID NO: 2至16所示之 NAMPT天然反義序列之核酸序列互補或結合,且調節 NAMPT分子之表現及/或功能。 在一實施例中,寡核苷酸包含SEQ ID NO: 17至31之至 少5個連續核苷酸之序列,且調節NAMPT分子之表現及/或 功能。 聚核苷酸目標包含NAMPT,包括其家族成員;NAMPT 之變異體;NAMPT之突變體,包括SNP ; NAMPT之非編 碼序列;NAMPT之對偶基因;物種變異體片段及其類似 物。募核普酸較佳為反義分子。 在一實施例中,靶向NAMPT聚核苷酸之寡核苷酸包 I58213.doc -46 - 201210611 含:反義 RNA、干擾 RNA(RNAi)、短干擾 RNA(siRNA); 微干擾RNA(miRNA);小時序RNA(stRNA);或短髮夾 RNA(shRNA);小RNA誘導之基因活彳匕(RNAa);或小活化 RNA(saRNA)。 在一實施例中,靶向菸鹼醯胺轉磷酸核糖基酶(NAMPT) 聚核苷酸(例如SEQ ID NO: 2至31)會調節此等目標之表現 或功能。在一實施例中,與對照組相比表現或功能上調。 在一實施例中,與對照組相比表現或功能下調。 〇 在一實施例中,反義化合物包含SEQ ID NO: 17至31所 示之序列。此等寡核苷酸可包含一或多個經修飾之核苷 酸、較短或較長片段、經修飾之鍵及其類似物。 在一實施例中,SEQ ID NO: 17至31包含一或多個LNA核 苷酸。表1展示適用於本發明方法之例示性反義寡核苷酸。 表1 :Start codon or "start codon". A few genes have a translation initiation codon with the RNA sequence 5 GUG, 5'-UUG or 5'-CUG; and it has been shown that 5 -AUA, Ό /^ττβ 或 or 5-CUG play a role in vivo. Therefore, the terms "transfer start codon" and "start codon" can cover many dense columns, but in each case the starting amino acid is usually methionine (in a nuclear organism) or Methionine (in the context of the present invention, the prokaryotic gene may have two or more alternative initiation codon types or tissues or under a specific set of conditions may be preferred: in the context of the present invention, "Start codon" in vivo = "start codon" means that regardless of the sequence of the codons, the start of the transcription of the mRNA encoded by the gene encoding nicotine amide transphosphate phosphoribosylase (ναμρ) "Terminator codon" can be -ΤΛΑ〇Α, 5,_UGA(^ ^DNA^ ^ ^ ^ ^ -Λ ^ The term "starting codon" and "translation starting secret" - or the base is covered in self-translating The beginning codon of Ren Hai or) about 25 to about 50 consecutive nucleotide words "stop codon area" and "transfer, political stop ... "similarly 'surgery (four) termination (four) sub-area" means 158213. Doc -36- 201210611 mRNA or base time covers the self-translating stop codon - direction (ie 5, or 3,) on about 25 A portion of about 5G consecutive cores (4), "starting codon region" (or "transition initiation codon region") and "terminating codon region" (or "transition termination codon region") are available for use in the present invention. The antisense compound is effectively targeted to all regions. It is known in the art that the open reading frame (ORF) or "coding region" of the region between the translation initiation codon and the translation stop codon is also an effective target. In the context of the present invention, the target region is the intragenic region encompassing the translation initiation codon or the translation stop codon of the open reading frame (GRF) of the gene. Refers to the 5, non-translated region (5.utr) of the translation initiation codon from the translation initiation codon 5, and thus includes the acidity between the cap 5 and the translation initiation codon ( Or the corresponding nucleotide acid in the gene. Another target region includes 3 in the direction of the translation stop codon in _A, the partial ❸ in the direction, the non-translated region Q (3'UTR), and Thus including the translation of the mRNA stop codon with the nucleotide between the 3, _ (or genetically) Corresponding nucleotides. mRNA25, the cap site comprises a Ν' methylated guanine nucleoside residue joined to the most terminal residue of mRNA 2 51 via a 5'-5' triphosphate linkage. The mRNAi5, cap region is considered to include 5. The cap structure itself, and 50 nucleotides adjacent to the cap site. Another target region of the invention is the 5' cap region. Although some eukaryotic mRNA transcripts will be translated directly, many contain a terminology One or more regions are excised from the transcript before the transcript is translated. The remaining (and therefore translated) regions are called "exons" and spliced 158213.doc -37· 201210611 together form a continuous mRNA sequence. In one embodiment, the targeted splice site (ie, intron-exon junction or exon_inclusion) is involved in a case where aberrant splicing is associated with a disease or an overproduction of a particular splicing product is associated with a disease. Sub-joints are especially useful. Another embodiment in which the abnormal fusion junction due to rearrangement or deletion is the target site. mRNA transcripts produced by splicing two (or more) mRNAs from different gene sources are referred to as "fusion transcripts". Introns can be efficiently targeted using antisense compounds that target, for example, DNA or precursor mRNA. In one embodiment, the antisense oligonucleotide binds to the coding and/or non-coding region of the polynucleotide of interest and modulates the expression and/or function of the target molecule. In one embodiment, the antisense oligonucleotide binds to a natural antisense polynucleotide and modulates the expression and/or function of the target molecule. In one embodiment, the antisense raised nucleotide binds to a sense polynucleotide and modulates the expression and/or function of the target molecule. Alternative RNA transcripts can be produced from the same genomic region of DNA. Such alternative transcripts are generally referred to as "variants." More specifically, "precursor mRNA variants, allogeneic" are transcripts produced by the same genome (10), which differ in their initiation or termination from other transcripts produced by the same genomic DNA and contain introns and Proton sequence. One or more exons or intron regions or portions thereof are excised during splicing, and the steroid mRNA variants produce smaller r mRNA variants. Therefore, the mRNA variant is a pre-warmed body mRNA variant, and each unique precursor mRNA variant must always produce a unique mRNA variant due to splicing. These mRNA variants are also referred to as "alternative splice variants". If 158213.doc -38· 201210611 is present in the splicing of the precursor mRNA variant, the precursor mRNA variant is identical to the mRNA variant. Variants can be produced by using alternative signals to initiate or terminate transcription. The precursor mRNA and mRNA may have more than one start codon or stop codon. A variant derived from the use of an alternative start codon precursor mRNA* mRNA is referred to as an "alternative initiation variant" of the precursor mRNA or mRNA. Their transcripts using alternative stop codons are referred to as "alternative termination variants" of the precursor mRNA or mRNA. A particular type of surrogate termination variant is a "polyA variant" in which a plurality of transcripts produced are replaced by a transcriptional machinery that alternatively selects a rpolyA termination signal, thereby terminating in a unique polyA position. Produced by the transcript of the point. Within the context of the present invention, the types of variants described herein are also examples of target nucleic acids. The position on the target nucleic acid to which the antisense compound hybridizes is defined as the portion of the target region targeted by the active antisense compound that is at least 5 nucleotides in length. Q Although certain sequences of some exemplary target segments are set forth herein, those skilled in the art will recognize that such sequences are used to illustrate and describe particular embodiments within the scope of the invention. Other target segments are readily identified by the average technician in accordance with the present invention. It is believed that a target segment of 5 to 100 nucleotides in length comprising at least 5 (f) consecutive nucleotides selected from a segment of the illustrative preferred target segment is also suitable for targeting. The target segment can comprise a DNA or RNA sequence comprising at least 5 contiguous nucleotides from the 5, _ terminus of an illustrative preferred target segment (remaining nucleoside 158213.doc -39 - 201210611 acid is a self-aligned target The upstream of the 5'-end of the segment begins and continues until the DNA or RNA contains a contiguous portion of the same DNA4 RNA of about 5 to about 1 nucleotide. Similarly, the preferred target segment is represented by a human or Chinese sequence comprising at least 5 contiguous nucleotides from the 3, _ terminus of an illustrative preferred target segment (the remaining nucleotide is Starting from the 3, _ end of the target segment, and continuing until the DNA or RNA contains the same 1)]8 or a contiguous segment of RNA) of about 5 to about 1 nucleotide. Those skilled in the art to provide the target segments shown herein will be able to identify other preferred target segments without undue experimentation. Once one or more target regions, segments or sites have been identified, an antisense compound that is sufficiently complementary to the target, i.e., sufficiently hybridized and of sufficient specificity, is selected to achieve the desired effect. In an embodiment of the invention, the anti-sense nucleoside I of the specific target is bound to a specific target of at least 5 nucleotides in length and can be synthesized such that each oligonucleotide is targeted to the overlapping sequence such that the synthesis The oligonucleotide covers the entire length of the target polynucleotide. Targets also include coding and non-coding areas. In one embodiment, the specific nucleic acid is preferably targeted using an antisense oligonucleotide. Targeting an antisense D to a particular nucleic acid is a multi-step process. This method usually begins by identifying the nucleic acid sequence that will regulate the function. The <target nucleic acid" can be, for example, a cellular gene (or mRNA transcribed from a gene) or a non-coding polynuclear, such as a non-coding, that is associated with a particular condition or disease condition. RNA can be classified as (1) messenger RNA (mRNA), which translates into protein, and (7) non-protein-encoding RNA (nCRNA). ncRNAs contain microRNAs, antisense transcripts, and other transcriptional units (τυ) that contain high-density stop codons and lack any broad open reading frame. Many ncRNAs appear to be 158213.doc 201210611 from the 3' untranslated region of the protein-coding locus (3·υτιι) to initiate the initiation site. ncRNAs are often rare, and at least half of the ncRNAs that have been sequenced by the FANT(R) M Association do not appear to be polyadenylated. For obvious reasons, most of the researchers focused on the polyadenylation mRNA processed and exported to the cytoplasm. Recently, it has been shown that the collection of non-polyadenylated nuclear RNA may be extremely large and that many of these transcripts are produced by so-called intergenic regions. The mechanism by which ncRNA modulates gene expression is base pairing with the target transcript. RNAs that act by base pairing can be grouped into (1) cis-encoding RNA, which encodes at the same genetic position, but acts on the strand opposite the RNA and thus appears to be completely complementary to its target; and (2 A trans-coded ruler, which encodes at a position corresponding to 111 <[8 different chromosomes, acts on the RNA and generally does not exhibit the full probability of pairing with its target. Without wishing to be bound by Wenli, the antisense oligonucleotides described herein can interfere with the expression of the corresponding sense messenger RNA by interfering with the antisense polynucleotide. However, this tone may be inconsistent (antisense knockout causes messenger 111^ eight increase) or may be one (antisense knockout causes concomitant messenger RNA reduction). In such cases, antisense raised nucleotides can target overlapping or non-overlapping portions of the antisense transcript, causing knockout or sequestration. Both the coding and non-coding antisense sequences can be targeted in the same manner, and any species can modulate the corresponding sense transcript in a consistent or inconsistent manner. The strategy for identifying novel nucleotides for use against a target can be based on any other means of knocking out antisense RNA transcripts with antisense oligonucleotides or modulating desired targets. Sincere 7: In the case of inconsistent regulation, knocking out antisense transcripts increases the performance of conventional (sense) genes. If the conventional (sense) gene coding is known or 158213.doc •41 · 201210611 疋 疋 疋 则 则 则 则 则 则 则 则 则 则 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 敲 可 可 可 可 可 可 可 可 可 可 可In general, in the case of consistent regulation, antisense and sense transcripts can be knocked out and thus a synergistic reduction in the expression of the conventional (sense) gene can be achieved. For example, if a knockout is achieved using an antisense oligonucleotide, this strategy can be applied to the antisense oligonucleotides of the dry sense transcript and (iv) the additional antisense oligo of the corresponding antisense transcript. Nucleotides' or simultaneously target a single energy symmetric antisense oligonucleotide that overlaps the sense and antisense transcripts. According to the present invention, antisense compounds include antisense oligonucleotides, ribonuclease outer leader (EGS) nucleotides, siRNA compounds, single strands = double stranded RNA interference (RNAi) compounds (such as Xiancai compounds) And other polymeric compounds that hybridize to at least a portion of the target nucleic acid and modulate their function. Thus, it may be DNA, RNA, class, class or a mixture thereof, or may be a mimic of one or more of them. Such compounds may be single-stranded, double-stranded, circular or hairpin oligomeric compounds and may contain structural elements such as internal or terminal protrusions, mismatches or rings. Linear antisense compounds are typically prepared, but may be joined or otherwise prepared into a ring and/or a branched shape. : A compound can include a construct such as a hybrid to form a complete or partial double ::: two strands or having sufficient self-complementarity to allow for a single strand; The two strands can be internally connected such that: free 3' or 5, the end, or can be joined to form a continuous inflammatory structure or "3 end ... with a hole to produce a single strand characteristic extension. The double-k compound may optionally include a protrusion at the end including a link to the end, a selected citric acid position, a sugar position, and a nucleus 158213.doc -42·201210611 group. Alternatively, the two strands can be joined via a non-nucleic acid moiety or a linker group. When formed by only _ strands, the dsRNA can be in the form of self-complementary hair (4) molecules that fold in half to form a duplex. Thus, the dsRNA can be in the form of a full or partial double strand. The specific regulation of gene expression can be achieved by the development of a stable gene expression in a transgenic cell line, but in some real cases, the gene expression or function is up-regulated. #由两: Or a single strand of a self-complementary hairpin type molecule in the form of a double-stranded pair, the two strands (or the single-stranded duplex forming region) are bases in Waldorf-gram A complementary RNA strand paired by Rick. Upon introduction into the system, the compounds of the invention may elicit one or more enzymes or structural proteins to function to affect cleavage or other modification of the target nucleic acid, or may function via a population-based mechanism. In general, nucleic acids (including nucleotides) can be described as "DNA-like" (ie, generally having one or more 2,-deoxy sugars, and generally having T instead of purine bases) or "RNA-like" (i.e., generally having one or more 2'-hydroxy sugars or 2'-modified sugars, and generally having an anthracene rather than a τ test group). Nucleic acid helices can employ more than one type of structure, most commonly A and B. In general, the nucleotide having a B-like structure is "DNA-like", and the oligonucleotide having an octagonal structure is "RNA-like". In some (funny) embodiments, the antisense compound can contain an A-shaped region and a b-shaped region. In one embodiment, the desired oligonucleotide or antisense compound comprises at least one of the following: antisense RNA, antisense DNA, chimeric antisense nucleotides, antisense recruitment comprising modified linkages Nucleotide, interfering RNA (RNAi), short interfering RNA (siRNA); micro-interfering RNA (miRNA); small temporal RNA (stRNA); 158213.doc -43- 201210611 or short hairpin RNA (shRNA); small RNA induction Gene activation (RNAa); small activating RNA (saRNA), or a combination thereof. dsRNA also activates gene expression, a mechanism called "small RNA-induced gene activation" or RNAa. The dsRNA targeting the gene promoter induces efficient transcriptional activation of related genes. RNAa has been demonstrated in human cells using synthetic dsRNA called "small activating RNA" (saRNA). It is currently unknown whether RNAa is retained in other organisms. Small double-stranded RNA (dsRNA) such as small interfering RNA (siRNA) and microRNA (miRNA) has been found to be a trigger for an evolutionarily conserved mechanism called RNA interference (RNAi). RNAi always causes gene quiescence by remodeling chromatin thereby inhibiting transcription, degrading complementary mRNA or blocking protein translation. However, in the case of a detailed intervening in the Examples section that follows, the display of nucleotides increases the expression and/or function of the nicotinamide-transferase-ribosylase (NAMPT) polynucleotide and its encoded product. It can also act as a small activating RNA (saRNA). Without wishing to be bound by theory, in the phenomenon known as dsRNA-induced transcriptional activation (RNAa), the target gene expression will be induced by targeting the sequence 'saRNA in the gene promoter. In another embodiment, the "better target segment" identified herein can be used to screen for other compounds that modulate the expression of a nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide. A "modulator" is a compound that reduces or increases the performance of a nucleic acid molecule encoding NAMPT and comprises at least one 5 nucleotide portion that is complementary to the preferred target segment. The screening method comprises the steps of contacting a preferred target segment of a nucleic acid molecule encoding a NAMPT-derived or natural antisense polynucleotide with one or more candidate modulators, and selecting one or more of 158213.doc-44- 201210611 Reduce or increase candidate modulators of the performance of nucleic acid molecules encoding NAMPT polynucleotides (eg, SEQ ID NOS: 17 to 31). Once the candidate modulator is shown to be capable of modulating (eg, reducing or increasing) the performance of a nucleic acid molecule encoding a NAMPT polynucleotide, the modulator can be used in other investigations of the function of the NAMPT polynucleotide or as the present invention. Research, diagnosis or therapeutic agent. Targeting a natural antisense sequence preferably modulates the function of the target gene. For example, the NAMPT gene (e.g., accession number NM_005746). In one embodiment, the target is an antisense polynucleotide of the NAMPT gene. In one embodiment, the antisense oligonucleotide targets a sense and/or natural antisense sequence of a NAMPT polynucleotide (eg, accession number NM_005746), variants thereof, dual genes, isoforms, and Sources, mutants, derivatives, fragments and complementary sequences. The oligonucleotide is preferably an antisense molecule and the target includes the coding and non-coding regions of the antisense and/or sense NAMPT polynucleotide. Preferred target segments of the invention may also be combined with the respective complementary antisense compounds of the invention to form stable double-stranded (duplex) oligonucleotides. These double-stranded oligonucleotide moieties have been shown in the art to modulate target expression via an antisense machine and to modulate translation and RNA processing. In addition, the double-stranded portion can undergo chemical modification. For example, the double-stranded portions have been shown to inhibit the target by triggering a classically hybridization of the antisense strands of the duplex with the target thereby triggering the enzymatic reduction of the target. In one embodiment, the antisense oligonucleotide targets a nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide (eg, accession number NM_005746), variants thereof, dual genes, isoforms, and Sources, mutants, derivatives, fragments and complementary sequences. The oligonucleotide is preferably an antisense molecule. 158213.doc -45- 201210611 According to an embodiment of the invention, the target nucleic acid molecule is not limited to NAMPT alone, but also to any of its polynucleotide variants and to produce, act, influence, or cause or be associated with NAMPT performance products. Any polynucleotide and/or any isoforms thereof. In one embodiment, the oligonucleotide targets a natural antisense sequence of a NAMPT polynucleotide, such as the polynucleotides set forth in SEQ ID NOs: 2 to 16, and any variants thereof, dual genes, Sources, mutants, derivatives, fragments and complementary sequences. Examples of antisense oligonucleotides are set forth in SEQ ID NOS: 17 to 31. In one embodiment, the oligonucleotide complements or binds to a nucleic acid sequence of a NAMPT antisense sequence, including, but not limited to, a non-coding sense and/or antisense sequence associated with a NAMPT polynucleotide; And modulating the performance and/or function of the NAMPT molecule. In one embodiment, the oligonucleotide complements or binds to the nucleic acid sequence of the NAMPT natural antisense sequence as set forth in SEQ ID NOs: 2 to 16, and modulates the expression and/or function of the NAMPT molecule. In one embodiment, the oligonucleotide comprises a sequence of at least 5 contiguous nucleotides of SEQ ID NOs: 17 to 31 and modulates the expression and/or function of the NAMPT molecule. Polynucleotide targets include NAMPT, including members of its family; variants of NAMPT; mutants of NAMPT, including SNPs; non-coding sequences of NAMPT; dual genes of NAMPT; fragments of species variants and the like. The priming acid is preferably an antisense molecule. In one embodiment, the oligonucleotide targeting the NAMPT polynucleotide comprises I58213.doc -46 - 201210611 comprising: antisense RNA, interfering RNA (RNAi), short interfering RNA (siRNA); micro interfering RNA (miRNA) Small time-series RNA (stRNA); or short hairpin RNA (shRNA); small RNA-induced gene activity (RNAa); or small activating RNA (saRNA). In one embodiment, targeting a nicotine indole to phosphoribosyltransferase (NAMPT) polynucleotide (e.g., SEQ ID NOs: 2 to 31) modulates the performance or function of such targets. In one embodiment, the performance or function is up-regulated compared to the control group. In one embodiment, the performance or function is down-regulated compared to the control group. In one embodiment, the antisense compound comprises the sequences set forth in SEQ ID NOs: 17 to 31. Such oligonucleotides may comprise one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. In one embodiment, SEQ ID NOS: 17 to 31 comprise one or more LNA nucleotides. Table 1 shows exemplary antisense oligonucleotides suitable for use in the methods of the invention. Table 1 :

序列號 反義序列 名稱 序列 SE〇IDNO:17 CUR-1724 SEQEDNO:18 CUR-1725 G*C*C*C*A*T*T*T*G*A*G*T*C*T*A*T*G*T*G*T*T SEQ ID NO: 19 CUR-1726 〇*0*0*Τ*0*Τ*0*Τ*0*0*Α*Α*0*0*0*Τ*0*Τ*0*Τ SEQIDNO:20 CUR-1727 g* 丁 * 丁*c*c*a*t*a*c*c*t*a*c*t*t*c*a*c*c*a*g SEQIDNO:21 CUR-1728 G*C*C*C*T*A*G*GsiiT*C*T*T*T*Ci|!Ai|tT*A*G*C*A*C SEQIDNO:22 CUR-1729 (^*γ*(^*Τ*Α*ί!Ι*Α*Τ*0*(^*Τ*Α*(Ι!*Α*Α*(^*(ΙΙ*Τ*(3*(Ι!*0 SEQIDNO:23 CUR-1730 C*A*T*G*C*C*A*C*C*A*C*G*T*C*C*A*G*C*T*A SE〇IDNO:24 CUR-1731 T*C*C*A*C*T*A*C*C*A*C*C*A*C*C*A*C*C*A*T*C SE〇EDNO:25 CUR-1732 G* 丁 丁 *c*A*G*C*C*A*T*G*C*C*T*C*C*A*G*T SE〇IDNO:26 CUR-1733 A*G*G*A*G*G*G*A*C*A*A*G*A*G*G*G*A*T*G*G*A SEQIDNO:27 CUR-1734 A*G*A*T*G*G*G*C*A*G*T*A*G*G*A*G*G*C*T*C SEQIDNO:28 CUR-1735 g*t*g*g*c*g*t*g*a*c*t*t*c*a*g*g*a*g*g*a SEQIDNO:29 CUR-1736 G*t*T*G*C*C*A*A*C*T*C*G*T*T*T*C*C*C*A*G*G SEQIDNO:30 CUR-1737 C*A*G*G*G*T*C*A*G*G*A*G*G*A*T*C*A*G*G*A SEQIDNO:31 CUR-1505 對照序列 C*c*T*C*T*C*C*A*C*G*C*G*C*A*G*T*A*C*A*T*T 158213.doc •47· 201210611 所需目標核酸之調節可以此項技術中已知之若干方法來 進行。舉例而言,反義寡核苷酸、siRNA等。酶性核酸分 子(例如核糖核酸酶)為能夠催化多種反應中之一或多者之 核酸分子’包括能夠以核苷酸鹼基序列特異性方式重複裂 解其他個別核酸分子。可使用該等酶性核酸分子例如幾乎 乾向任何RNA轉錄物。 由於其序列特異性,反式裂解酶性核酸分子顯示有望作 為人類疾病之治療劑。酶性核酸分子可經設計以裂解細胞 RNA之背景内的特定rna目標。該種裂解事件使11111>^無 功能性且消除自該RNA之蛋白質表現。以此方式,可選擇 性抑制與疾病病況相關之蛋白質的合成。 一般而言,具有RNA裂解活性之酶性核酸藉由首先結合 目標RNA起作用。該結合經由酶性核酸之目標結合部分進 仃,該目標結合部分保持極接近用以裂解目標rna之分子 的酶性部分。因此,酶性核酸首先識別,且接著經由互補 驗基配對結合目標RNA,I —旦結合至正確位置,即以酶 的方式起作用切割目標Rna。該種目標RNA之策略性裂解 將破壞其直接合成所編碼蛋白質之能力。S酶性核酸結合 且裂解其RNA目標之後,其自彼RNA脫離以搜尋另一目標 且可重複結合且裂解新目標。 已使用諸如活體外選擇(進化)策略之若干方法(〇rgel, (1979) Proc. R s〇c L〇nd〇n,B 2〇5, 43勹來進化能夠催化 多種反應,諸如磷酸二酯鍵聯及醯胺鍵聯之裂解及連接反 應的新穎核酸催化劑。 158213.doc -48- 201210611 開發催化活性最佳之核糖核酸酶將明顯有助於出於調節 基因表現之目的利用RNA裂解核糖核酸酶之任何策略。錘 頭型核糖核酸酶例如在飽和(1 〇 mM)濃度之Mg2+輔因子存 在下以約1 miiT1之催化速率(kcat)起作用。已顯示人工 「RNA連接酶」核糖核酸酶以約100 min·1之速率催化相應 自身修飾反應❹另外,已知具有由DNA構成之受質結合臂 的某些經修飾之錘頭型核糖核酸酶以接近1〇〇 min-i之多種 周轉率催化RNA裂解。最終,用某些核苷酸類似物置換錘 頭之催化核心内之特定殘基得到顯示催化速率改良高達1〇 倍的經修飾之核糖核酸酶。此等發現表明核糖核酸酶可以 明顯大於活體外由大多數天然自裂解核糖核酸酶所呈現之 催化速率促進化學轉化。接著可將某些自裂解核糖核酸酶 之結構最佳化以得到最大催化活性,或可製備對於RNA填 酸二醋裂解顯示明顯較快速率之全新RNA基元。 1987(Uhlenbeck,Ο. C. (1987) Nature,328: 596-600)中首 先展示藉由符合「錘頭」模型之RNA催化劑使RNA受質分 子間裂解。回收RNA催化劑,且與多種rnA分子反應,表 明其確實具催化性。 已使用基於「錘頭」基元設計之催kRNA以藉由在催化 RNA中進行適當鹼基改變以維持與目標序列之必要鹼基配 對來裂解特定目標序列。此舉允許使用催化尺\八來裂解特 疋目標序列,且指示根據「鐘頭」模型設計之催化RNA可 活體内裂解特定受質RNA。 RNA干擾(RNAi)已成為調節哺乳動物及哺乳動物細胞中 158213.doc -49· 201210611 之基因表現之有效工具。此方法需要以RNA本身或以DNA 形式傳遞小干擾RNA(siRNA),使用表現質體或病毒及加 工成siRNA之小髮夾RNA的編碼序列。此系統能夠將前體 siRNA有效傳輸至細胞質中,在該細胞質中前體siRNA具 活性且允許使用受調節及組織特異性啟動子進行基因表 現。 在一實施例中,寡核苷酸或反義化合物包含核糖核酸 (RNA)及/或(DNA)之募聚物或聚合物、或其模擬物、嵌合 體、類似物或同源物。此術語包括由天然存在之核苷酸、 糖及共價核苷間(主鏈)鍵聯構成之寡核苷酸,以及具有功 能類似之非天然存在部分之寡核苷酸。由於諸如細胞攝取 增強、對目標核酸之親和力增強及在核酸酶存在下穩定性 增強之合乎需要之性質,故通常相對於天然形式更需要該 等經修飾或取代之寡核苷酸。 根據本發明,寡核苷酸或「反義化合物」包括反義寡核 苷酸(例如RNA、DNA、其模擬物、嵌合體、類似物或同 源物)、核糖核酸酶、外部引導序列(EGS)寡核苷酸、 siRNA化合物、單股或雙股RNA干擾(RNAi)化合物(諸如 siRNA化合物)、saRNA、aRNA及與目標核酸之至少一部 分雜交且調節其功能之其他寡聚化合物。因而,其可為 DNA、RNA、類DNA、類RNA或其混合物,或可為其中一 或多者之模擬物。此等化合物可為單股、雙股、環形或髮 夾寡聚化合物且可含有結構元件,諸如内部或末端突起、 錯配或環。通常製備線性反義化合物,但可接合或以其他 158213.doc •50- 201210611 方式製備成環形及/或分支形。反義化合物可包括構築 體,諸如雜交形成完全或部分雙股化合物之兩個股,或具 有足夠自身互補性以允許雜交且形成完全或部分雙股化合 物之單版。兩個股可内部連接從而留下游離3'或5,末端, 或可連接形成連續髮夾結構或環。髮夾結構可在5,或3,末 端上含有突出物,產生單股特徵之延長。雙股化合物視情 況可在末端包括突出物。其他修飾可包括連接至一個末 ^、所選核菁酸位置、糖位置或-個核普間鍵聯的結合基 〇 目。或者,兩個股可經由非核酸部分或連接子基團連接。 當僅由-個股形成肖’祕财可1自身對折形成雙鏈體之 自身互補冑夾型分子之开。因此,心⑽八可呈完全或部 分雙股形式。基因表現之特異性調節可藉由轉殖基因細胞 株中dsRNA髮夾之穩定表現獲得。當由兩個股或呈本身對 折形成雙鏈體之自身互補髮夾型分子形式之單股形成時, 兩個股(或單股之雙鏈體形成區)為鹼基以 〇 式配對之互補RNA1。 克克方 引人系統中後,本發明化合物可引發—或多種酶或結構 蛋白質起作用以影響目標核酸之裂解或其他修掷,或可經 由基於占位之機制起作用。一般而言’核酸(包括募核普 酸)可描述為「類DNA」(亦即一般具有一或多個广去氧 糖,且一般具有τ而非u驗基)或「類RNA」(亦即一般呈有 -或多個2,-經基糖或2,_修飾糖,且一般具有U而非;驗 基)。核酸螺旋可採用-種以上類型之結構,最通常為轉 及B形。咸信,一般而言,具有B形樣結構之募核苦酸為 158213.doc 51 201210611 「類DNA」,且具有A形樣結構之寡㈣酸為「類rna」。 在一些(喪合)實施例中,反義化合物可含有A形區與B形 區0 本發明之反義化合物可包含長約5至約8〇個核苷酸之反 義部分(亦即約5至約8G個連接核们。㈣反義股或反義化 合物之一部分之長度《拖, 又X 換s之,本發明之單股反義化合物 包3 5至約8G個核苦酸,且本發明之雙股反義化合物(諸如 dsRNA)包含有義股及反義股或長5至約個核苷酸之部 分 1022 34 46 58 70 般技術者將瞭解,其包括長度為5 Η、12、13、14、15 23 35 47 59 24 36 48 60 25 37 49 61 26 38 50 62 27 39 51 63 16 28 40 52 64 17 29 41 53 65 18 30 42 54 66 6、7、 19 ' 20 31 4355 67 32 44 56 68 、9 21 33 45 57 69Serial number antisense sequence name sequence SE〇IDNO:17 CUR-1724 SEQEDNO:18 CUR-1725 G*C*C*C*A*T*T*T*G*A*G*T*C*T*A *T*G*T*G*T*T SEQ ID NO: 19 CUR-1726 〇*0*0*Τ*0*Τ*0*Τ*0*0*Α*Α*0*0*0* Τ*0*Τ*0*Τ SEQIDNO:20 CUR-1727 g* D****c*c*a*t*a*c*c*t*a*c*t*t*c*a*c *c*a*g SEQIDNO:21 CUR-1728 G*C*C*C*T*A*G*GsiiT*C*T*T*T*Ci|!Ai|tT*A*G*C*A *C SEQIDNO: 22 CUR-1729 (^*γ*(^*Τ*Α*ί!Ι*Α*Τ*0*(^*Τ*Α*(Ι!*Α*Α*(^*(ΙΙ *Τ*(3*(Ι!*0 SEQIDNO:23 CUR-1730 C*A*T*G*C*C*A*C*C*A*C*G*T*C*C*A*G *C*T*A SE〇IDNO:24 CUR-1731 T*C*C*A*C*T*A*C*C*A*C*C*A*C*C*A*C*C* A*T*C SE〇EDNO:25 CUR-1732 G* Tintin*c*A*G*C*C*A*T*G*C*C*T*C*C*A*G*T SE〇 IDNO:26 CUR-1733 A*G*G*A*G*G*G*A*C*A*A*G*A*G*G*G*G*A*T*G*G*A SEQIDNO:27 CUR-1734 A*G*A*T*G*G*G*C*A*G*T*A*G*G*G*A*G*G*C*T*C SEQIDNO:28 CUR-1735 g* t*g*g*c*g*t*g*a*c*t*t*c*a*g*g*a*g*g*a SEQIDNO:29 CUR-1736 G*t*T*G *C*C*A*A*C*T*C*G*T*T*T*C*C*C*A*G*G SEQIDNO:30 CUR-1737 C*A*G*G*G* T*C*A*G*G*A*G*G*A*T*C*A*G*G*A SEQIDNO:31 CUR-1505 Control sequence C*c*T*C*T*C*C *A*C*G*C*G*C*A*G*T*A *C*A*T*T 158213.doc •47· 201210611 The modulation of the desired target nucleic acid can be carried out by several methods known in the art, for example, antisense oligonucleotides, siRNA, and the like. An enzymatic nucleic acid molecule (e.g., ribonuclease) is a nucleic acid molecule capable of catalyzing one or more of a variety of reactions' including the ability to repeatedly cleave other individual nucleic acid molecules in a nucleotide base sequence specific manner. These enzymatic nucleic acid molecules can be used, e.g., almost dry to any RNA transcript. Due to its sequence specificity, trans-lytic enzymatic nucleic acid molecules have been shown to be promising agents for human diseases. Enzymatic nucleic acid molecules can be designed to cleave specific rna targets within the background of cellular RNA. This cleavage event renders 11111>^ non-functional and eliminates protein expression from the RNA. In this way, the synthesis of proteins associated with disease conditions is selectively inhibited. In general, an enzymatic nucleic acid having an RNA cleavage activity acts by first binding to a target RNA. The binding proceeds via a target binding moiety of the enzymatic nucleic acid which remains in close proximity to the enzymatic moiety of the molecule used to cleave the target rna. Thus, the enzymatic nucleic acid is first recognized, and then binds to the target RNA via a complementary pairing, and I binds to the correct position, i.e., acts to cleave the target Rna in an enzymatic manner. Strategic cleavage of this target RNA will disrupt its ability to directly synthesize the encoded protein. After the S-enzymatic nucleic acid binds and cleaves its RNA target, it detaches from the other RNA to search for another target and can repeatedly bind and cleave the new target. Several methods such as phosphodiesters have been catalyzed by several methods such as the in vitro selection (evolution) strategy (〇rgel, (1979) Proc. R s〇c L〇nd〇n, B 2〇5, 43勹. Novel nucleic acid catalysts for cleavage and ligation of linkages and guanamine linkages. 158213.doc -48- 201210611 The development of catalytically active ribonuclease will significantly facilitate the use of RNA for cleavage of RNA for the purpose of regulating gene expression. Any strategy of the enzyme. Hammerhead ribonuclease acts, for example, at a catalytic rate (kcat) of about 1 miI T1 in the presence of a saturated (1 mM) concentration of Mg2+ cofactor. Artificial "RNA ligase" ribonuclease has been shown. The corresponding self-modification reaction is catalyzed at a rate of about 100 min·1. In addition, certain modified hammerhead ribonucleases having a binding arm composed of DNA are known to be close to 1 〇〇 min-i. The rate catalyzes the cleavage of RNA. Finally, the replacement of specific residues in the catalytic core of the hammerhead with certain nucleotide analogs results in a modified ribonuclease showing a catalytic rate improvement of up to 1 fold. These findings indicate ribose The acidase can be significantly greater than the catalytic rate promoted by most natural self-cleaving ribonucleases in vitro to promote chemical conversion. The structure of certain self-cleaving ribonucleases can then be optimized for maximum catalytic activity, or can be prepared for RNA-packed diacetate cleavage shows a significantly faster rate of new RNA motifs. 1987 (Uhlenbeck, Ο. C. (1987) Nature, 328: 596-600) first demonstrates an RNA catalyst that conforms to the "hammerhead" model. The RNA is cleavable by intermolecular cleavage. The RNA catalyst is recovered and reacted with various rnA molecules, indicating that it is indeed catalytic. The k-RNA based on the "hammerhead" motif has been used to carry out the appropriate bases in the catalytic RNA. Alter to maintain the necessary base pairing with the target sequence to cleave the specific target sequence. This allows the use of a catalytic ruler to cleave the target sequence and indicates that the catalytic RNA designed according to the "Clock" model can cleave specific receptors in vivo. RNA interference (RNAi) has become an effective tool for regulating gene expression in mammalian and mammalian cells 158213.doc -49· 201210611. RNA itself or in the form of DNA transmits small interfering RNA (siRNA), using a coding sequence that expresses a plastid or virus and a small hairpin RNA that is processed into siRNA. This system is capable of efficiently transporting precursor siRNA into the cytoplasm where it is The precursor siRNA is active and allows gene expression using a regulated and tissue-specific promoter. In one embodiment, the oligonucleotide or antisense compound comprises a ribonucleic acid (RNA) and/or (DNA) polymer. Or a polymer, or a mimetic, chimera, analog or homolog thereof. The term includes oligonucleotides consisting of naturally occurring nucleotides, sugars, and covalent internucleoside (backbone) linkages, as well as oligonucleotides having functionally similar non-naturally occurring portions. Such modified or substituted oligonucleotides are generally more desirable relative to the native form due to desirable properties such as enhanced cellular uptake, enhanced affinity for the target nucleic acid, and enhanced stability in the presence of nucleases. According to the invention, an oligonucleotide or "antisense compound" includes an antisense oligonucleotide (eg, RNA, DNA, mimetic, chimera, analog or homolog thereof), ribonuclease, external leader sequence ( EGS) oligonucleotides, siRNA compounds, single or double stranded RNA interference (RNAi) compounds (such as siRNA compounds), saRNA, aRNA, and other oligomeric compounds that hybridize to at least a portion of a target nucleic acid and modulate its function. Thus, it may be DNA, RNA, DNA-like, RNA-like or a mixture thereof, or may be a mimetic of one or more of them. Such compounds may be single-stranded, double-stranded, circular or hair-folding oligomeric compounds and may contain structural elements such as internal or terminal protrusions, mismatches or rings. Linear antisense compounds are typically prepared, but may be joined or otherwise prepared in a ring and/or branched form in the manner of 158213.doc • 50-201210611. Antisense compounds can include constructs, such as two strands that hybridize to form a fully or partially double-stranded compound, or a single plate that has sufficient self-complementarity to allow hybridization and form a fully or partially double-stranded compound. The two strands can be internally joined to leave a free 3' or 5, end, or can be joined to form a continuous hairpin structure or loop. The hairpin structure can have protrusions at the 5, or 3, end, resulting in an extension of the single strand feature. The double-stranded compound may include protrusions at the end, as appropriate. Other modifications may include attachment to a terminal, a selected nucleocyanate position, a sugar position, or a binding bond between the nucleus. Alternatively, the two strands can be joined via a non-nucleic acid moiety or a linker group. When only the - individual strands are formed, Xiao's secret money can be folded into the self-complementary cleavage type of the double-stranded body. Therefore, heart (10) eight can be in full or partial double-strand form. Specific regulation of gene expression can be obtained by stable performance of dsRNA hairpins in transgenic cell lines. When two strands or a single strand of a self-complementary hairpin type molecule in the form of a double-stranded pair is formed in itself, the two strands (or the single-stranded duplex forming region) are complementary to the base pairing RNA1. Following introduction into the system, the compounds of the invention may elicit—or multiple enzymes or structural proteins to act to affect cleavage or other repair of the target nucleic acid, or may function via a population-based mechanism. In general, 'nucleic acids (including nucleophilic acids) can be described as "DNA-like" (that is, generally have one or more extensive deoxygenated sugars, and generally have a τ instead of a nucleus) or "RNA-like" (also That is, there are generally - or more 2,-trans-glycosides or 2,-modified sugars, and generally have U instead of; The nucleic acid helix may be of a type or more, most commonly a B-shaped structure. In general, the nucleus acid having a B-like structure is 158213.doc 51 201210611 "DNA-like", and the oligo(tetra) acid having an A-shaped structure is "like rna". In some (same) embodiments, the antisense compound can comprise an A-shaped region and a B-shaped region. The antisense compound of the invention can comprise an antisense portion of from about 5 to about 8 nucleotides in length (ie, about 5 to about 8G nucleus. (4) The length of one part of the antisense strand or the antisense compound "Tow, and X s, the single antisense compound of the present invention contains 3 5 to about 8G nucleotides, and The double-stranded antisense compound of the present invention (such as dsRNA) comprises a sense strand and an antisense strand or a portion of 5 to about nucleotides long. 1022 34 46 58 70 As will be understood by those skilled in the art, it includes a length of 5 Η, 12 , 13, 14, 15 23 35 47 59 24 36 48 60 25 37 49 61 26 38 50 62 27 39 51 63 16 28 40 52 64 17 29 41 53 65 18 30 42 54 66 6, 7, 19 ' 20 31 4355 67 32 44 56 68 , 9 21 33 45 57 69

71、72、73、74、75、76、77、78、79 或 80 個核苷酸 或其中之任何範圍之反義部分。 广實施例中’本發明之反義化合物具有長1〇至5〇個核 苷酸之反義部分。-般技術者將瞭解,其包括具有長10、 η、12、13、14、15、16、17、18、19、2〇、2ι、22、 23、24、25、26、27、28、29、3〇、31 m、 35 36、37、38、39、4〇、41、42、43 H、牝、 或%個核㈣或其中之料範圍之反義部分的 。在—些實施例中,寡核普酸之長度為15個核 酸0 P 158213.doc 52- 201210611 在一實施例中,本發明之反義或寡核苷酸化合物具有長 12或13至3G個核*酸之反義部分。—般技術者將瞭解,其 包括具有長 12、13、"、15、16、17、18、19、20、21、 22 ' 23 ' 24、25、26、27、28、29或30個核苦酸或其中之 任何範圍之反義部分的反義化合物。 在-實施例中’本發明之募聚化合物亦包括在該化合物 中之一或多個核苷酸位置上存在不同鹼基的變異體。舉例 而言,若第一核苷酸為腺苷,則可產生在此位置含有胸腺 〇 嘧啶核苷、鳥嘌呤核苷或胞嘧啶核苷之變異體。此舉可在 反義或dsRNA化合物之任何位置進行。接著使用本文所述 之方法測試此等化合物,以測定其抑制目標核酸之表現的 能力。 在一些實施例中,反義化合物與目標之間的同源性、序 列一致性或互補性為約4〇%至約。在一些實施例中, 同源性、序列一致性或互補性為約6〇%至約7〇%。在一些 ❹ 實施例中’同源性、序列一致性或互補性為約70%至約 80%。在一些實施例中,同源性、序列一致性或互補性為 約80。/〇至約90%。在一些實施例中,同源性、序列一致性 或互補性為約90%、約92%、約94%、約95%、約96%、約 97%、約 98%、約 99%或約 1〇〇%。 在一實施例中,諸如SEQ ID NO: 17至31所示之核酸分 子之反義募核苷酸包含一或多個取代或修飾。在一實施例 中,核苷酸經鎖核酸(LNA)取代。 在一實施例中’募核苷酸乾向與NAMPT相關之編碼及/ 158213.doc -53- 201210611 或非編碼序列的有義及/或反義核酸分子及如SEQ ID NO: 1及16所示序列之核酸分子的一或多個區。寡核苷酸亦靶 向SEQ ID NO: 1至16之重疊區。 本發明之某些較佳寡核苷酸為嵌合募核苷酸。在本發明 之上下文中’「嵌合寡核苷酸」或「嵌合體」為含有兩個 或兩個以上化學性質不同之區的寡核苷酸,各區由至少— 個核苦酸構成。此等募核苷酸通常含有賦予一或多種有益 性質(諸如核酸酶抗性增加、細胞攝取增強、對目標之結 合親和力增強)之經修飾核苷酸之至少一個區、及作為夠 裂解RNA:DNA或RNA:RNA雜交物之酶的受質之區。舉例 而言’ RNase Η為裂解RNA:DNA雙螺旋體之RNA鏈的細胞 内切核酸酶。因此’活化核糖核酸酶Η引起rna目標裂 解’由此大大提咼基因表現之反義調節所效率。因此,當 使用嵌合寡核苷酸時,與雜交相同目標區之硫代磷酸酯去 氧养核苷酸相比,較短募核苷酸通常可獲得可比結果。 RNA目標之裂解通常可藉由凝膠電泳及必要時藉由此項技 術中已知之相關核酸雜交技術偵測。在一實施例中,嵌合 募核苷酸包含至少一個經修飾增強目標結合親和力之區、 及通常充當核糖核酸酶Η之受質的區。通常藉由量測募核 苷酸/目標對之Tm來測定募核苷酸對其目標(在此情況下為 編碼ras之核酸)之親和力,該Tm為募核苷酸及目標解離之 溫度,以分光光度法彳貞測解離。T m愈高,寡核苷酸對目 標之親和力愈大。 可形成呈兩種或兩種以上如上所述之寡核苷酸、經修飾 158213.doc -54- 201210611 寡核苷酸、募核苷及/或寡核苷酸模擬物之複合結構的本 發明之嵌合反義化合物。此項技術中該等化合物亦稱為雜 交物或間隙聚合物。教示該等雜交結構之製備的代表性美 國專利包含(但不限於)美國專利第5,013,830號;第 5,149,797 號;第 5,220,007 號;第 5,256,775 號;第 • 5,366,878 號;第 5,403,711 號;第 5,491,133 號;第 5,565,350 號;第 5,623,065 號;第 5,652,355 號;第 5,652,356號;及第5,700,922號;各文獻均以引用的方式併 〇 入本文中。 在一實施例中’經修飾之募核苷酸之區包含至少一個在 糖之T位經修飾之核苷酸,最佳2·-〇烷基、2,-0-烷基-〇-烧 基或21-氟-修飾之核苷酸。在另一實施例中,rna修飾包 括在嘧啶、無鹼基殘基或RNA之3,末端反向鹼基之核糖上 之2’-氟、2’-胺基及2Ό-曱基修飾。該等修飾通常併入寡核 苷酸中,且已顯示與2'-去氧寡核苷酸相比,此等募核苷酸 〇 對指定目標具有較高Tm(亦即較高目標結合親和力)。該親 和力增強之作用為大大增強基因表現iRNAi寡核苷酸抑 制作用。核糖核酸酶Η為裂解RNA:DNA雙鏈體之RNA股的 細胞核酸内切酶;因此,此酶之活化引起RNA目標裂解, 且因此可大大增強RNAi抑制效率。RNA目標之裂解通常 可藉由凝膠電泳證明。在一實施例中,嵌合募核苷酸亦經 修飾以使核酸酶抗性增強。細胞含有多種可降解核酸之核 酸外切酶及核酸内切酶。已顯示許多核苷酸及核苷修飾會 使並有該等修飾之募核苷酸與天然寡去氧核苷酸相比具有 158213.doc -55- 201210611 更高核酸酶消化抗性。核酸酶抗性通常藉由使募核苦酸與 細胞萃取物或分離之核酸酶溶液一起培育,且通常藉由凝 膠電泳隨時間量測剩餘完整募核苷酸之含量來量測。與未 修飾寡核苦酸相比,經修飾以使核酸酶抗性增強之寡核苦 酸之完整性保持較長時間。已表明多種募核苷酸修飾會增 強或賦予核酸酶抗性。目前更佳為含有至少一個硫代磷酸 酯修飾之寡核苷酸。在一些情況下’增強目標結合親和力 之募核苷酸修飾亦能獨立地增強核酸酶抗性。 設想用於本發明之一些較佳寡核苷酸之特定實例包括包 含例如硫代磷酸酯、磷酸三酯、膦酸甲酯、短鏈烷基或環 烧基糖間鍵聯或短鍵雜原子或雜環糖間鍵聯之經修飾主鍵 之彼等實例。最佳為具有硫代磷酸酯主鏈之寡核苷酸及具 有雜原子主鏈之寡核苷酸,尤其CH2--NH--0--CH2、CH2--N(CH3)--0--CH2[稱為亞甲基(曱基亞胺基)或MMI主鏈]、 CH2--0--N(CH3)--CH2、CH2-N(CH3)--N(CH3)--CH2及 Ο--N(CH3)--CH2--CH2主鏈,其中天然填酸二酯主鏈表示為 0--P--0--CH2。De Mesmaeker 等人(1995) Acc. Chem. Res· 28:366-374所示之醯胺主鏈亦較佳。亦較佳為具有N-嗎啉 基主鏈結構之寡核苷酸(Summerton及Weller,美國專利第 5,034,5 06號)。在另一實施例中,諸如肽核酸(PNA)主鏈, 寡核苷酸之磷酸二酯主鏈置換為聚醯胺主鏈,該等核苷酸 直接或間接結合至聚醯胺主鏈之氮雜氮原子。寡核苷酸亦 可包含一或多個經取代之糖部分。較佳募核苷酸在2'位包 含以下中之一者:OH、SH、SCH3、F、OCN、OCH3、 158213.doc •56- 201210611 0(CH2)nCH3、0(CH2)nNH2 或 0(CH2)nCH3,其中 η 為 1 至 約10; Cl至CIO低碳烷基、烷氧基烷氧基、經取代之低碳 烧基、烧芳基或芳烧基;Cl ; Br ; CN ; CF3 ; OCF3 ; 、S--或 N-烷基;Ο--、S —或 N-烯基;SOCH3 ; S02 CH3 ; 0N02 ; N02 ; N3 ; NH2 ;雜環烷基;雜環烷芳基;胺基 烷基胺基;聚烷基胺基;經取代之矽烷基;RNA裂解基 團;報導體基團;插入基團;改良寡核苷酸之藥物動力學 性質的基團;或改良寡核苷酸之藥效學性質之基團及具有 〇 類似性質之其他取代基。較佳修飾包括2’-甲氧基乙氧基 [2i-0,CH2CH20CH3,亦稱為2,-0-(2-曱氧基乙基)]。其他 較佳修飾包括2’-甲氧基(2’-0--CH3)、2·-丙氧基(2,-OCH2CH2CH3)及2、氟(2'-F)。亦可在寡核苷酸之其他位 置,尤其3'端核苷酸上糖之3'位及5’端核苷酸之5'位進行類 似修飾。寡核苷酸亦可具有糖模擬物,諸如環丁基,而非 五吱喃糖基。 或者或另外,募核苷酸亦可包括核鹼基(在此項技術中71, 72, 73, 74, 75, 76, 77, 78, 79 or 80 nucleotides or an antisense portion of any of the ranges. In a broad embodiment, the antisense compound of the invention has an antisense portion of from 1 to 5 nucleotides in length. As will be appreciated by those skilled in the art, including having lengths of 10, η, 12, 13, 14, 15, 16, 17, 18, 19, 2〇, 2ι, 22, 23, 24, 25, 26, 27, 28, 29, 3 〇, 31 m, 35 36, 37, 38, 39, 4 〇, 41, 42, 43 H, 牝, or % nucleus (4) or the antisense part of the material range thereof. In some embodiments, the length of the oligonucleotide is 15 nucleic acids. 0 P 158213.doc 52-201210611 In one embodiment, the antisense or oligonucleotide compound of the invention has a length of 12 or 13 to 3G The antisense part of the nuclear acid. As will be understood by the average technician, it has a length of 12, 13, ", 15, 16, 17, 18, 19, 20, 21, 22 ' 23 ' 24, 25, 26, 27, 28, 29 or 30 An antisense compound of nucleotide acid or any range of antisense portions thereof. In the examples, the polyamounting compound of the present invention also includes variants in which different bases are present at one or more nucleotide positions in the compound. For example, if the first nucleotide is adenosine, a variant containing thymidine, guanosine or cytosine at this position can be produced. This can be done anywhere in the antisense or dsRNA compound. These compounds are then tested using the methods described herein to determine their ability to inhibit the performance of the target nucleic acid. In some embodiments, the homology, sequence identity or complementarity between the antisense compound and the target is from about 4% to about. In some embodiments, the homology, sequence identity, or complementarity is from about 6% to about 7%. In some ❹ embodiments, 'homology, sequence identity or complementarity is from about 70% to about 80%. In some embodiments, the homology, sequence identity, or complementarity is about 80. /〇 to about 90%. In some embodiments, the homology, sequence identity, or complementarity is about 90%, about 92%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 1〇〇%. In one embodiment, antisense nucleotides such as the nucleic acid molecules set forth in SEQ ID NOS: 17 to 31 comprise one or more substitutions or modifications. In one embodiment, the nucleotide is substituted with a locked nucleic acid (LNA). In one embodiment, the nucleotide and the antisense nucleic acid molecules encoding the NAMPT and/or 158213.doc-53-201210611 or non-coding sequences are as described in SEQ ID NO: 1 and 16. One or more regions of the nucleic acid molecule of the sequence are shown. Oligonucleotides also target the overlapping regions of SEQ ID NOS: 1 to 16. Some preferred oligonucleotides of the invention are chimeric nucleotides. In the context of the present invention, "chimeric oligonucleotide" or "chimeric" is an oligonucleotide comprising two or more chemically distinct regions, each region being composed of at least one nucleotide. Such raised nucleotides typically contain at least one region of a modified nucleotide that confers one or more beneficial properties, such as increased nuclease resistance, enhanced cellular uptake, enhanced binding affinity to the target, and as sufficient cleavage RNA: DNA or RNA: the region of the substrate of the enzyme of the RNA hybrid. For example, 'RNase Η is an endonuclease that cleaves the RNA strand of RNA:DNA duplex. Therefore, 'activated ribonuclease Η causes rna target cleavage', thereby greatly improving the efficiency of antisense regulation of gene expression. Thus, when chimeric oligonucleotides are used, shorter nucleotides typically yield comparable results compared to phosphorothioate deoxynucleotides that hybridize to the same target region. Cleavage of the RNA target can typically be detected by gel electrophoresis and, if desired, by related nucleic acid hybridization techniques known in the art. In one embodiment, the chimeric nucleotide comprises at least one region modified to enhance binding affinity of the target, and a region that normally acts as a substrate for ribonuclease. The affinity of the nucleotide for its target, in this case the nucleic acid encoding ras, which is the temperature at which the nucleotide is raised and the target dissociation, is typically determined by measuring the Tm of the nucleotide/target pair. The dissociation was measured spectrophotometrically. The higher the Tm, the greater the affinity of the oligonucleotide for the target. The present invention can be formed into a composite structure of two or more oligonucleotides as described above, a modified 158213.doc-54-201210611 oligonucleotide, a nucleoside and/or an oligonucleotide mimetic Chimeric antisense compounds. Such compounds are also referred to as hybrids or interstitial polymers in the art. Representative U.S. patents which teach the preparation of such hybrid structures include, but are not limited to, U.S. Patent Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5, 491, 133; 5, 565, 355; 5, 623, 065; 5, 652, 355; 5, 652, 356; and 5,700, 922; each of which is incorporated herein by reference. In one embodiment, the region of the modified nucleotide comprises at least one nucleotide modified at the T position of the sugar, preferably 2,-decyl, 2,-0-alkyl-oxime-fired A base or a 21-fluoro-modified nucleotide. In another embodiment, the rna modification comprises a 2'-fluoro, 2'-amino group and a 2Ό-fluorenyl modification on the ribose of the pyrimidine, abasic residue or RNA 3, terminal reverse base. Such modifications are typically incorporated into the oligonucleotide and have been shown to have a higher Tm (ie, higher target binding affinity) for the specified target compared to the 2'-deoxyoligonucleotide. ). The effect of this affinity enhancement is to greatly enhance the iRNAi oligonucleotide inhibition of gene expression. The ribonuclease 细胞 is a cell endonuclease that cleaves the RNA strand of the RNA:DNA duplex; therefore, activation of this enzyme causes cleavage of the RNA target, and thus the RNAi inhibition efficiency can be greatly enhanced. Cleavage of RNA targets can usually be demonstrated by gel electrophoresis. In one embodiment, the chimeric nucleotides are also modified to enhance nuclease resistance. The cell contains a variety of exonuclease and endonuclease which degrade the nucleic acid. A number of nucleotide and nucleoside modifications have been shown to result in a higher nuclease digestion resistance of 158213.doc -55 - 201210611 compared to native oligodeoxynucleotides. Nuclease resistance is typically measured by culturing the nucleus acid with a cell extract or an isolated nuclease solution, and is typically measured by gel electrophoresis over time to measure the amount of remaining intact nucleotide. The integrity of the oligonucleotides modified to enhance nuclease resistance is maintained for a longer period of time than unmodified oligonucleotides. A variety of nucleotide modifications have been shown to enhance or confer nuclease resistance. More preferably, it is an oligonucleotide containing at least one phosphorothioate modification. In some cases, a nucleotide modification that enhances the binding affinity of the target can also independently enhance nuclease resistance. Specific examples of some preferred oligonucleotides contemplated for use in the present invention include, for example, phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl groups or cycloalkyl saccharide linkages or short bond heteroatoms. Or examples of modified primary bonds linked between heterocyclic sugars. Preferred are oligonucleotides having a phosphorothioate backbone and oligonucleotides having a hetero atom backbone, especially CH2--NH--0--CH2, CH2--N(CH3)--0- -CH2 [referred to as methylene (mercaptoimine) or MMI backbone], CH2--0--N(CH3)--CH2, CH2-N(CH3)--N(CH3)--CH2 And the Ο--N(CH3)--CH2--CH2 backbone, wherein the natural acid-filled diester backbone is represented by 0--P--0--CH2. De Mesmaeker et al. (1995) The amine amine backbone shown by Acc. Chem. Res. 28:366-374 is also preferred. Also preferred are oligonucleotides having an N-morpholinyl backbone structure (Summerton and Weller, U.S. Patent No. 5,034,506). In another embodiment, such as a peptide nucleic acid (PNA) backbone, the phosphodiester backbone of the oligonucleotide is replaced by a polyamine backbone that binds directly or indirectly to the polyamine backbone Aza nitrogen atom. The oligonucleotide may also comprise one or more substituted sugar moieties. Preferred nucleotides include one of the following in the 2' position: OH, SH, SCH3, F, OCN, OCH3, 158213.doc • 56- 201210611 0(CH2)nCH3, 0(CH2)nNH2 or 0 ( CH2)nCH3, wherein η is from 1 to about 10; Cl to CIO lower alkyl, alkoxyalkoxy, substituted lower carbon alkyl, aryl or aryl; Cl; Br; CN; CF3 OCF3; , S-- or N-alkyl; Ο-, S- or N-alkenyl; SOCH3; S02 CH3; 0N02; N02; N3; NH2; heterocycloalkyl; heterocycloalkylaryl; Alkylamino group; polyalkylamine group; substituted alkyl group; RNA cleavage group; conductor group; insertion group; group for improving the pharmacokinetic properties of the oligonucleotide; or modified oligo A group having the pharmacodynamic properties of a glycoside and other substituents having a similar nature to the oxime. Preferred modifications include 2'-methoxyethoxy [2i-0,CH2CH20CH3, also known as 2,-0-(2-decyloxyethyl)]. Other preferred modifications include 2'-methoxy (2'-0--CH3), 2·-propoxy (2,-OCH2CH2CH3) and 2, fluorine (2'-F). Similar modifications can also be made at other positions of the oligonucleotide, particularly at the 3' position of the sugar at the 3' end nucleotide and at the 5' position of the 5' end nucleotide. Oligonucleotides may also have a sugar mimetic, such as a cyclobutyl group, rather than a quinolate. Alternatively or additionally, the raised nucleotides may also include nucleobases (in the art)

Q 通常簡稱為「驗基」)修飾或取代。如本文所用之「未修 飾」或「天然」核苷酸包括腺嘌呤(A)、鳥嘌呤(G)、胸腺 嘧啶(T)、胞嘧啶(C))及尿嘧啶(U)。經修飾核苷酸包括僅 偶爾或短暫見於天然核酸中之核普酸,例如次黃嗓吟、6-甲基腺嘌呤、5-Me嘧啶、尤其5-曱基胞嘧咬(亦稱為5-甲 基-2'去氧胞嘧啶且此項技術中通常稱為5-Me-C)、5-羥基 曱基胞嘧啶(HMC)、糖基HMC及龍瞻雙醣基HMC,以及合 成核苷酸,例如2-胺基腺嘌呤、2-(曱基胺基)腺嘌呤、2- 158213.doc •57- 201210611 (味唾基烧基)腺嗓呤、2_(胺基烷基胺基)腺嘌呤或其他雜 取代烷基腺嘌呤、2-硫尿嘧啶、2_硫代胸腺嘧啶、5_溴尿 喵。疋、5-羥基曱基尿嘧啶、8_氮鳥嘌呤、7·去氮鳥嘌呤、 N6(6-胺基己基)腺嘌呤及2,6_二胺基嘌呤。可包括此項技 術中已知之「通用」鹼基,例如肌苷。已顯示5-Me-C取代 使核fee雙鏈體穩定性增加Q 6〇c至i .21且為當前較佳之驗 基取代。 本發明寡核普酸之另一修飾涉及化學連接至寡核普酸之 或夕個增強寡核苷酸之活性或細胞攝取的部分或結合 物。該等部分包括(但不限於)脂質部分,諸如膽固醇部 刀膽崔醇部分、脂肪鏈(例如十二烷二醇基或十一烷基 殘基)夕元胺或聚乙二醇鏈或金剛烷乙酸。寡核苷酸包 含親脂性部分’且製備該特核#酸之方法在此項技術中 已知例如美國專利第5,138,〇45號、帛5,2ΐ8,ι〇5號及第 5,459,255號。 指定寡核苷酸中之所有位置並非必需加以均一修飾,且 實際上可將一種以上前述修飾併入單一募核苷酸中或甚至 寡核苷酸内之單一核苷内。本發明亦包括如上文所定義作 為嵌合募核苷酸之寡核苷酸。 在另一實施例中,使本發明之核酸分子與包括(但不限 於)無驗基核芽酸、聚醚、多元胺、聚酿胺、狀、碳水化 合物、脂質或聚烴化合物之另一部分結合。熟習此項技術 者將認識到,此等分子可連接至一或多種任何核苷酸(包 3核酸分子)的糖、驗基或磷酸g旨基上之若干位置。 158213.doc -58- 201210611 根據本發明使用之募核苷酸便利地且通常可經由熟知固 相合成技術來製備。用於該合成之設備由包括Appiied Bi〇Systems之若干供應商出售。亦可利用用於該合成之任 何其他方式;寡核苷酸之實際合成較佳在一般技術者之能 力範圍内。亦熟知使用類似技術製備其他募核苷酸,諸如 硫代磷酸酯及烷基化衍生物。亦熟知使用類似技術及市售 經修飾胺化物(amidite)及受控微孔玻璃(CpG)產品,諸如 生物素、螢光素、吖啶或補骨脂素修飾之胺化物及/或 CPG(可自Glen Research,Sterling VA獲得)以合成經螢光標 記、生物素化或其他修飾之寡核苷酸,諸如膽固醇修飾之 寡核苷酸。 根據本發明,使用修飾,諸如使用LNA單體以增強效 能、特異性及作用持續時間且拓寬募核苷酸之投藥途徑, 該等修飾包含現有化學變化,諸如MOE、ANA、FANA、 PS等。此修飾可藉由用lnA單體取代當前寡核苷酸中之一 些單體來獲得。LNA修飾之寡核苷酸之大小可與母體化合 物類似’或可能較大或較佳較小。該等LNA修飾之寡核苷 酸較佳含有小於約70%,更佳小於約60%,最佳小於約 50%之LNA單體’且其大小較佳在約5與25個核苷酸之間, 更佳在約12與20個核苷酸之間。 較佳經修飾募核苷酸主鏈包含(但不限於)硫代磷酸酯、 對掌性硫代磷酸酯、二硫代磷酸酯、磷酸三酯、胺基烷基 麟酸二酿、曱基及其他烷基膦酸酯(包含3'伸烷基膦酸酯及 對掌性鱗酸酯)、亞膦酸酯、磷醯胺酯(包含3,-胺基磷醯胺 1582I3.doc -59- 201210611 酯及胺基烷基磷酿胺醋)、硫代麟酿胺醋、硫代烧基膦酸 酯、硫代烷基磷酸三酯及具有正常3'·5'鍵聯之硼烷填酸 酯、硼烷填酸酯之2'-υ連接型類似物及具有反極性之硼炫 磷酸酯,其中鄰近核苷單元對為3'-5’連接至5'_3'、或2’_5' 連接至5'-2'。亦包括各種鹽、混合鹽及游離酸形式。 教示上述含碟鍵聯之製備的代表性美國專利包含(但不 限於)美 國 專 利 第 3,687,808 號 ;第 4,469,863 號; 第 4,476,301 號 第 5,023,243 號; 第 5,177,196 號; 第 5,188,897 號 第 5,264,423 號; 第 5,276,019 號; 第 5,278,302 號 第 5,286,717 號; 第 5,321,131 號; 第 5,399,676 號 第 5,405,939 號; 第 5,453,496 號; 第 5,455,233 號 第 5,466,677 號; 第 5,476,925 號; 第 5,519,126 號 第 5,536,821 號; 第 5,541,306 號; 第 5,550,111 號 第 5,563,253 號; 第 5,571,799 號; 第 5,587,361號;及第5,625,050號,各文獻以引用的方式併入 本文中。 不包括璘原子之較佳經修飾寡核苷酸主鏈具有由短鏈烧 基或環烧基核苷間鍵聯、混合雜原子及烷基或環烷基核苷 間鍵聯、或一或多個短鏈雜原子或雜環核苷間鍵聯形成之 主鍵。此等主鏈包含具有N-嗎啉基鍵聯之彼等主鏈(部分 由核苦之糖部分形成);矽氧烧主鏈;硫化物、亞硬及石風 主鍵;甲酿基及硫曱醯基主鏈;亞曱基曱醯基及硫曱醯基 主鍵,含有烯烴之主鏈;胺基磺酸酯主鏈;亞乙基亞胺基 及亞曱基肼基主鏈;磺酸酯及磺醯胺主鏈;醯胺主鏈;及 1582I3.doc •60· 201210611 具有混合N、〇、S及CH2構成部分之其他主鏈。 教示上述募核苷之製備之代表性美國專利包含(但不限 於)美國專利第5,034,506號;第5,166,315號;第5,185,444 號;第 5,214,134號;第 5,216,141號;第 5,235,033 號;第 5,264,562 號; 第 5,264,564 號; 第 5,405,938 號; 第 5,434,257 號; 第 5,466,677 號; 第 5,470,967 號; 第 5,489,677 號; 第 5,541,307 號; 第 5,561,225 號; 第 5,596,086 號; 第 5,602,240 號; 第 5,610,289 號; 第 5,602,240 號; 第 5,608,046 號; 第 5,610,289 號; 第 5,618,704 號; 第 5,623,070 號; 第 5,663,312 號; 第 5,633,360號;第 5,677,437號;及第 5,677,439號,各文獻 以引用的方式併入本文中。 在其他較佳寡核苷酸模擬物中,用新穎基團置換核苷酸 單元之糖與核苷間鍵聯’亦即主鏈。維持基本單元以與適 當核酸目標化合物雜交。一種此募聚化合物,即已顯示具 有優良雜交特性之寡核苷酸模擬物稱為肽核酸(ρΝΑ)。在 ΡΝΑ化合物中’募核苷酸之糖主鏈係經含醯胺之主鏈、詳 δ之胺基乙基甘胺酸主鏈置換。核鹼基保留且直接或間接 結合於主鏈之醯胺部分之氮雜氮原子。教示ρΝΑ化合物之 製備的代表性美國專利包含(但不限於)美國專利第 5,539,082號;第 5,714,331號;及第 5,719,262號,各文獻 以引用的方式併入本文中。ΡΝΑ化合物之其他教示可見於Q is often referred to simply as "test base" to modify or replace. As used herein, "unmodified" or "natural" nucleotides include adenine (A), guanine (G), thymine (T), cytosine (C), and uracil (U). Modified nucleotides include nucleotides that are only occasionally or transiently found in natural nucleic acids, such as hypoxanthine, 6-methyladenine, 5-Me-pyrimidine, especially 5-mercapto-cytosine (also known as 5). -methyl-2'deoxycytosine and commonly referred to in the art as 5-Me-C), 5-hydroxydecylcytosine (HMC), glycosyl HMC, and long-acting disaccharide-based HMC, and synthetic nuclei Glycosylates, such as 2-aminoadenine, 2-(decylamino)adenine, 2-158213.doc •57-201210611 (saltyl) adenine, 2_(aminoalkylamino group Adenine or other hetero-substituted alkyl adenine, 2-thiouracil, 2-thio thymine, 5-bromouridine. Indole, 5-hydroxypurinyluracil, 8-nitroguanine, 7-deazaguanine, N6(6-aminohexyl)adenine and 2,6-diaminoguanidine. It may include "universal" bases known in the art, such as inosine. The 5-Me-C substitution has been shown to increase the nuclear fee duplex stability by Q 6 〇 c to i. 21 and is currently the preferred substituent substitution. Another modification of the oligonucleotides of the invention involves a moiety or conjugate that is chemically linked to the activity or cellular uptake of the oligonucleotide or the oligonucleotide. Such moieties include, but are not limited to, lipid moieties such as the cholesterol moiety, the fatty chain (eg, dodecyldiol or undecyl residue), the o-amine or polyethylene glycol chain or diamond Alkanoic acid. Oligonucleotides comprising a lipophilic moiety' and methods of preparing such a nucleus# acid are known in the art, for example, U.S. Patent Nos. 5,138, 〇45, 帛5,2ΐ8, ι〇5, and 5,459,255. . All positions in a given oligonucleotide are not necessarily uniformly modified, and indeed more than one of the foregoing modifications can be incorporated into a single raised nucleotide or even within a single nucleoside within an oligonucleotide. The invention also includes oligonucleotides as defined above as chimeric nucleotides. In another embodiment, the nucleic acid molecule of the invention is further than another portion of the compound including, but not limited to, non-initiated nuclear phytic acid, polyether, polyamine, polyamine, saccharide, carbohydrate, lipid or polyhydrocarbon compound Combine. Those skilled in the art will recognize that such molecules can be linked to several positions on the sugar, tester or phosphate group of one or more of any of the nucleotides (packet 3 nucleic acid molecules). 158213.doc -58- 201210611 The nucleotides used in accordance with the present invention are conveniently and generally prepared by well known solid phase synthesis techniques. Equipment for this synthesis is sold by several vendors including Appiied Bi〇 Systems. Any other means for this synthesis can also be utilized; the actual synthesis of the oligonucleotide is preferably within the capabilities of the average skilled person. It is also well known to use similar techniques to prepare other nucleotides, such as phosphorothioates and alkylated derivatives. It is also well known to use similar techniques and commercially available modified amidite and controlled microporous glass (CpG) products such as biotin, luciferin, acridine or psoralen modified amines and/or CPG ( Available from Glen Research, Sterling VA) to synthesize fluorescently labeled, biotinylated or otherwise modified oligonucleotides, such as cholesterol modified oligonucleotides. In accordance with the present invention, modifications, such as the use of LNA monomers to enhance efficacy, specificity, and duration of action and broaden the route of administration of nucleotides, including existing chemical changes such as MOE, ANA, FANA, PS, and the like, are used. This modification can be obtained by substituting one of the monomers in the current oligonucleotide with an lnA monomer. The LNA modified oligonucleotide may be similar in size to the parent compound or may be larger or preferably smaller. Preferably, the LNA modified oligonucleotides comprise less than about 70%, more preferably less than about 60%, optimally less than about 50% of the LNA monomer' and preferably have a size of between about 5 and 25 nucleotides. More preferably, between about 12 and 20 nucleotides. Preferably, the modified nucleotide backbone comprises, but is not limited to, a phosphorothioate, a palmitic phosphorothioate, a phosphorodithioate, a phosphate triester, an aminoalkyl sulphate, a thiol group. And other alkyl phosphonates (including 3' alkyl alkyl phosphonates and palmitic lactic acid esters), phosphonites, phosphonium esters (including 3,-aminophosphonamide 1582I3.doc-59 - 201210611 Ester and Aminoalkyl Phosphate Amine Vinegar), Thioline Amine Vinegar, Thioalkylphosphonate, Thioalkyl Triphosphate and Borane Filling with Normal 3'·5' Bonding a 2'-fluorene-linked analog of an acid ester, a borane-filled acid ester, and a borophosphoryl phosphate having a reverse polarity, wherein the adjacent nucleoside unit pair is 3'-5'-linked to 5'_3', or 2'_5 ' Connect to 5'-2'. Also included are various salts, mixed salts and free acid forms. Representative U.S. patents which teach the preparation of the above-described dish-containing bonds include, but are not limited to, U.S. Patent Nos. 3,687,808; 4,469,863; 4,476,301, 5,023,243; 5,177,196; 5,188,897, 5,264,423; 5,276,019 Nos. 5, 286, pp. 5, 286, pp. 5, 321, pp. 5, 399, 496; 5, 453, 496; 5, 455, 675; 5, 466, 677; 5, 476, 925; 5, 519, 126; 5, 541, 306; 5, 550, 111, 5, 563, 253 No. 5, 571, 799; 5, 587, 361; and 5, 625, 050, each of which is incorporated herein by reference. Preferred modified oligonucleotide backbones that do not include a ruthenium atom have a linkage between a short chain alkyl group or a cycloalkyl group, a mixed hetero atom, and an alkyl or cycloalkyl internucleoside linkage, or one or A primary bond formed by a plurality of short-chain heteroatoms or heterocyclic nucleoside linkages. These main chains comprise their main chains with N-morpholinyl linkages (partially formed by the nuclear bitter sugar moiety); oxime-burning backbone; sulfide, subhard and stone-wind primary bonds; Sulfhydryl backbone; fluorenyl sulfhydryl and thioindolyl backbone, containing olefin backbone; amine sulfonate backbone; ethyleneimine and fluorenyl sulfhydryl backbone; sulfonic acid Ester and sulfonamide backbone; guanamine backbone; and 1582I3.doc • 60· 201210611 Other backbones with a mixture of N, 〇, S and CH2 moieties. Representative U.S. patents which teach the preparation of such nucleosides include, but are not limited to, U.S. Patent Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; Nos. 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240 No. 5, 608, 046; Nos. 5, 610, 289; 5, 618, 704; 5, 623, 070; 5, 663, 312; 5, 633, 360; 5, 677, 437; and 5, 677, 439, each of which is incorporated herein by reference. In other preferred oligonucleotide mimetics, the sugar of the nucleotide unit is replaced with a novel group, i.e., the backbone. The basic unit is maintained to hybridize to the appropriate nucleic acid target compound. One such polymeric compound, i.e., an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (ρΝΑ). In the ruthenium compound, the sugar main chain of the nucleotide is replaced by a main chain containing a guanamine, and an aminoethylglycine main chain of δ. The nucleobase remains and binds directly or indirectly to the aza nitrogen atom of the indole moiety of the backbone. Representative U.S. patents which teach the preparation of a ruthenium compound include, but are not limited to, U.S. Patent Nos. 5,539,082; 5,714,331; and 5,719,262 each incorporated herein by reference. Other teachings of bismuth compounds can be found in

Nielsen 等人,(1991) Science 254,1497-1500 中。 在本發明之一實施例中,提供具有硫代磷酸酯主鏈之募 158213.doc -61 - 201210611 核苷酸及具有雜原子主鏈之募核苷,尤其-CH2-NH-0-CH2-、-CH2-N(CH3)-0-CH2-(稱為亞甲基(甲基亞胺基)或 MMI 主鏈)、-CH2-0-N(CH3)-CH2-、-CH2N(CH3)-N(CH3)CH2-及-0-N(CH3)-CH2-CH2-,其中天然磷酸二酯主鏈表示為 上文參考之美國專利第5,489,677號之-0-P-0-CH2,及上 文參考之美國專利第5,602,240號之醯胺主鏈。亦較佳為具 有上文所參考之美國專利第5,034,506號之N-嗎啉基主鏈結 構之寡核苷酸。 經修飾募核苷酸亦可含有一或多個經取代糖部分。較佳 寡核苷酸在2'位包含以下中之一者:OH ; F ; 0-烷基、S-烷基或N-烷基;0-烯基、S-烯基或N-烯基;Ο-炔基、S-炔 基或N-炔基;或Ο烷基-Ο-烷基,其中烷基、烯基及炔基可 為經取代或未經取代之Cl至Cio烧基或C2至Cio稀基及快基。 尤其較佳為 〇(CH2)nOmCH3、0(CH2)n0CH3、0(CH2)nNH2、 0(CH2)nCH3、0(CH2)n0NH2及 0(CH2)n0N(CH3)2,其中 n及m可為1至約10。其他較佳募核苷酸在2'位包含以下中 之一者:Ci至C1Q低碳烷基、經取代之低碳烷基、烷芳 基、芳烷基、0-烷芳基或〇-芳烷基、SH、SCH3、OCN、 Cl、Br、CN、CF3、OCF3、SOCH3、S02CH3、0N02、 N02、N3、NH2、雜環烷基、雜環烷芳基、胺基烷基胺 基、聚烷基胺基、經取代之矽烷基、RNA裂解基團、報導 體基團、插入基團、改良寡核苷酸之藥物動力學性質的基 團或改良寡核苷酸之藥效學性質的基團,及具有類似性質 之其他取代基。較佳修飾包含21-曱氧基乙氧基(2'-0- 158213.doc -62- 201210611 CH2CH20CH3、亦稱為 2’-0-(2-甲氧基乙基)或 2'-MOE), 亦即烷氧基烷氧基。其他較佳修飾包含如本文以下實例中 所述之2'-二甲基胺氧基乙氧基,亦即0(CH2)20N(CH3)2基 團,亦稱為2’-DMAOE;及2'-二甲基胺基乙氧基乙氧基(此 項技術中亦稱為2·-0-二甲基胺基乙氧基乙基或2·-' DMAEOE),亦即 2'-0-CH2-0-CH2-N(CH2)2。 其他較佳修飾包含2'-甲氧基(2'-OCH3)、2'-胺基丙氧基 (2'-OCH2CH2CH2NH2)及2'-氟(2'-F)。亦可在寡核苷酸之 〇 其他位置,尤其3'端核苷酸上或2'-5'連接型寡核苷酸中糖 之3'位或5'端核苷酸之5’位進行類似修飾。寡核苷酸亦可具 有糖模擬物,諸如環丁基部分,而非五呋喃醣基糖。教示 該等修飾之糖結構之製備的代表性美國專利包含(但不限 於)美國專利第4,981,957號;第5,118,800號;第5,319,080 號;第 5,359,044號;第 5,393,878 號;第 5,446,137號;第 5,466,786 號;第 5,514,785 號;第 5,519,134 號;第 5,567,811 號;第 5,576,427 號;第 5,591,722 號;第 〇 5,597,909 號;第 5,610,300 號;第 5,627,053 號;第 5,639,873 號;第 5,646,265 號;第 5,658,873 號;第 " 5,670,633號;及第5,700,920,各文獻以引用的方式併入本 ‘ 文中。 募核苷酸亦可包含核鹼基(在此項技術中通常簡稱為 「驗基」)修飾或取代。如本文所用之「未修飾」或「天 然」核苷酸包含嘌呤鹼基腺嘌呤(A)及烏嘌呤(G),及嘧咬 鹼基胸腺嘧啶(T)、胞嘧啶(C))及尿嘧啶(U)。經修飾核皆 158213.doc -63- 201210611 酸包含其他合成及天然核苷酸,諸如5-甲基胞嘧啶(5-me-C)、5-羥曱基胞嘧啶、黃嘌呤、次黃嘌呤、2-胺基腺嘌 呤、腺嘌呤及鳥嘌°令之6-甲基及其他炫基衍生物、腺嗓呤 及鳥°票σ令之2 -丙基及其他烧基衍生物、2 -硫尿°密咬、2 -硫 代胸腺嘧啶及2-硫代胞嘧啶,5-鹵基尿嘧啶及胞嘧啶、5-丙炔基尿嘧啶及胞嘧啶、6-偶氮尿嘧啶、胞嘧啶及胸腺嘧 σ定、5 -尿嘴咬(假尿10密咬)、4 -硫尿D密咬、8 -鹵基腺嗓11 令及 鳥嘌呤、8-胺基腺嘌呤及鳥嘌呤、8-硫醇腺嘌呤及鳥嘌 呤、8-硫代烷基腺嘌呤及鳥嘌呤、8-羥基腺嘌呤及鳥嘌呤 及其他8-取代之腺嗓°令及鳥σ票吟、5- _基(尤其5-漠)尿°密 啶及胞嘧啶、5-三氟甲基尿嘧啶及胞嘧啶及其他5-取代之 尿嘧啶及胞嘧啶、7-甲基鳥嘌呤及7-甲基腺嘌呤、8-氮鳥 嘌呤及8-氮腺嘌呤、7-去氮鳥嘌呤及7-去氮腺嘌吟及3-去 氮鳥11票吟及3 -去氮腺嘌呤。 此外,核苷酸包含美國專利第3,687,808號中所揭示之彼 等核苦酸;『The Concise Encyclopedia of Polymer Science And Engineering』,第 858-859 頁,Kroschwitz,J.I.編,John Wiley & Sons, 1990中所揭示之彼等核苷酸;Englisch等 人,『Angewandle Chemie, International Edition』,1991, 30,第613頁中所揭示之彼等核苷酸;及Sanghvi, Y.S.,第 15章,『Antisense Research and Applications』,第 289-302 頁,Crooke,S.T.及 Lebleu,B.編,CRC Press, 1993 中所揭 示之彼等核苷酸。某些此等核苷酸尤其適用於增強本發明 之寡聚化合物之結合親和力。此等核苷酸包含5-取代之嘧 158213.doc -64- 201210611 咬、6-氮嘴咬及N-2、N-6及0-6取代之嗓吟,包含2-胺基 丙基腺嘌呤、5-丙炔基尿嘧啶及5-丙炔基胞嘧啶。5-甲基 胞嘧啶取代已顯示會使核酸雙鏈體穩定性增加〇.6。(:至 1.2 C (Sanghvi,Y.S.、Crooke,S.T.及 Lebleu B·編,『Antisense Research and Applications』,CRC Press,Boca Raton,1993 ’ 第 276-278頁)’且甚至更特定言之與2,-〇甲氧基乙基糖修飾 組合時,為當前較佳之鹼基取代。 教示上文所示經修飾核苷酸以及其他經修飾核苦酸之製 備的代表性美國專利包含(但不限於)美國專利第3,687,8〇8 號’以及第 4,845,205 號;第 5,130,302 號;第 5,134,066 號,第5,175,273號;第 5,367,066號;第 5,432,272號;第 〇 5,457,187 號; 第 5,459,255 號; 第 5,484,908 號; 第 5,502,177 號; 第 5,525,711 號; 第 5,552,540 號; 第 5,587,469 號; 第 5,596,091 號; 第 5,614,617 號; 第 5’750,692號;及第5,681,941號,各文獻以引用的方式併入 本文中。 〇 本發明寡核苷酸之另一修飾涉及化學連接至寡核苷酸之 一或多個增強寡核苷酸之活性、細胞分佈或細胞攝取之部 分或結合物。 該等部分包含(但不限於)脂質部分,諸如膽固醇部分、 膽酸、硫醚(例如己基-S-三苯甲基硫醇)、硫代膽固醇、脂 族鏈(例如十二院二醇或十—烧基殘基)、鱗脂(例如二-十 六烷基-外消旋-甘油或1,2-二十六烷基_外消旋_甘油基_ 3 H-膦酸二乙銨)、多元胺或聚乙二醇鏈、或金剛烷乙 158213.doc -65- 201210611 酸、棕櫚基部分、或十八烷基胺或己基胺基-羰基_羥膽固 醇部分。 教示該等寡核苷酸結合物之製備的代表性美國專利包含 (但不限於)美國專利第4,828,979號;第4,948,882號;第 5,218,105 號;第 5,525,465 號;第 5,541,313 號;第 5.545.730 號;第 5,552,538 號;第 5,578,717 號;第 5.580.731 號;第 5,580,731 號;第 5,591,584 號;第 5,109,124 號;第 5,118,802 號;第 5,138,045 號;第 5,414,077 號;第 5,486,603 號;第 5,512,439 號;第 5,578,718 號;第 5,608,046 號;第 4,587,044 號;第 4,605,735 號;第 4,667,025 號;第 4,762,779 號;第 4,789,737 號;第 4,824,941 號;第 4,835,263 號;第 4,876,335 號;第 4,904,582 號;第 4,958,013 號·,第 5.082.830 號;第 5,112,963 號;第 5,214,136 號;第 5.082.830 號;第 5,112,963 號;第 5,214,136 號;第 5,245,022 號;第 5,254,469 號;第 5,258,506 號;第 5,262,536 號;第 5,272,250 號;第 5,292,873 號;第 5,317,098 號;第 5,371,241 號;第 5,391,723 號;第 5,416,203 號;第 5,451,463 號;第 5,510,475 號;第 5,512,667 號’·第 5,514,785 號;第 5,565,552 號;第 5,567,810 號;第 5,574,142 號;第 5,585,481 號;第 5,587,371 號;第 5,595,726 號;第 5,597,696 號;第 5,599,923號;第5,599,928號及第5,688,941,各文獻岣以 引用的方式併入本文中。 158213.doc -66- 201210611 μ##發禮:本發明化合物亦可應用於藥物發現及目標驗 證領域。本發明包括,在藥物發現中使用該等化合物及本 文所鑑別之較佳目標區段試圖闞明菸鹼醯胺轉磷酸核糖基 冑(NAMPT)聚核苷酸與疾病病況、表型或病狀之間存在之 關係。此等方法包括偵測或調節ΝΑΜρτ聚核苷酸,其包 含使樣品、組織、細胞或生物體與本發明化合物接觸;在 處理後一定時間量測NAMPT聚核苷酸之核酸或蛋白質含 *及’或相關表型或化學端點;且視情況比較所量測之值 與未處理樣品或用本發明之其他化合物處理之樣品。此等 方法亦可與其他實驗平行或組合執行以測定用於目標驗證 方法之未知基因之功能,或測定特定基因產物作為治療或 預防特定疾病、病狀或表型之目標的有效性。 評估基因表現之上調或抑制: 外源核酸轉移至宿主細胞或生物體中可藉由直接偵測細 胞或生物體中核酸之存在來㈣。該等偵測可藉由此項技 ◎ 術中熟知之若干方法獲得。舉例而t,外源核酸之存在可 藉由南方墨點法(Southern blot)或藉由聚合酶鏈式反應 (PCR)技術使用特異性擴增與核酸相關之核苷酸序列的引 子來偵測。外源核酸之表現亦可使用包括基因表現分析之 習知方法來量測。舉例而言,可使用北方墨點法(Ν〇Πη blot)及反轉錄PCR(RT_PCR)偵測及定量由外源核酸產生之 mRNA。 來自外源核酸之RNA之表現亦可藉由量測酶活性或報導 體蛋白質活性來偵測。舉例而言,反義調節活性可以作為 158213.doc -67· 201210611 指示外源核酸產生效應RNA之目標核酸表現之降低或增加 來間接量測。基於序列保守性,引子可經設計且用以擴增 目標基因之編碼區。最初,可使用來自各基因之最高表^ 之編碼區構造模型對照基因,但可使用任何編碼或非編碼 區。各對照基因藉由將各編瑪區插入報導體編碼區與其聚 (A)信號之間來組合。此等質體將產生在基因之上游部分 具有報導體基因之mRNA ,且在3,非編碼區中產生潛在 RNAi目標。個別反義寡核苷酸之功效將藉由調節報導體 基因來檢定。適用於本發明方法之報導體基因包括乙醯羥 基酸合成酶(AHAS)、驗性填酸酶(ap)、β半乳糖苷酶 (LacZ)、β葡萄醣醛酸酶(GUS)、氣黴素乙醯轉移酶 (CAT)、綠色螢光蛋白質(GFP)、紅色螢光蛋白質(RFp)、 黃色螢光蛋白質(YFP)、青色螢光蛋白質(CFP)、辣根過氧 化酶(HRP)、螢光素酶(Luc)、胭脂胺酸合成酶(N〇s)、章 魚驗合成酶(OCS)及其衍生物。賦予安比西林 (ampicillin)、博萊黴素(bleomycin)、氯黴素 (chloramphenicol)、僅大徽素(gentamycin)、潮徽素 (hygromycin)、康黴素(kanamycin)、林可黴素 (lincomycin)、曱胺喋呤(methotrexate)、草胺鱗 (phosphinothricin)、嘌呤黴素(puromycin)及四環素(四環 素)之抗性之多種可選擇標記物可用。測定報導體基因之 調節的方法在此項技術中熟知,且包括(但不限於)螢光法 (例如螢光光譜分析、螢光活化細胞分選(FACS)、蟹光顯 微術)、抗生素抗性測定。 158213.doc • 68 - 201210611 可使用熟習此項技術者已知及本文其他地方所述之方法 檢定NAMPT蛋白質& mRNA表現。舉例而言,諸如eusa 之免疫檢定可用以量測蛋白質含量β NAmpt ELISA檢定 套組可購自例如 R & D Systems(Minneapolis,MN)。Nielsen et al., (1991) Science 254, 1497-1500. In one embodiment of the present invention, a nucleotide of 158213.doc-61 - 201210611 having a phosphorothioate backbone and a nucleoside having a hetero atom backbone, particularly -CH2-NH-0-CH2- , -CH2-N(CH3)-0-CH2- (referred to as methylene (methylimido) or MMI backbone), -CH2-0-N(CH3)-CH2-, -CH2N(CH3) -N(CH3)CH2- and -0-N(CH3)-CH2-CH2-, wherein the natural phosphodiester backbone is represented by -0-P-0-CH2 of U.S. Patent No. 5,489,677, the disclosure of which is incorporated herein by reference. The indoleamine backbone of U.S. Patent No. 5,602,240, incorporated herein by reference. Also preferred are oligonucleotides having the N-morpholinyl backbone structure of U.S. Patent No. 5,034,506, which is incorporated herein by reference. The modified nucleotide may also contain one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following in the 2' position: OH; F; 0-alkyl, S-alkyl or N-alkyl; 0-alkenyl, S-alkenyl or N-alkenyl Or alkynyl, S-alkynyl or N-alkynyl; or decyl-fluorenyl-alkyl, wherein the alkyl, alkenyl and alkynyl groups may be substituted or unsubstituted Cl to Cio alkyl or C2 to Cio dilute base and fast base. Particularly preferred are 〇(CH2)nOmCH3, 0(CH2)n0CH3, 0(CH2)nNH2, 0(CH2)nCH3, 0(CH2)n0NH2 and 0(CH2)n0N(CH3)2, wherein n and m can be 1 to about 10. Other preferred nucleotides include one of the following in the 2' position: Ci to C1Q lower alkyl, substituted lower alkyl, alkaryl, aralkyl, 0-alkylaryl or hydrazine- Aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, S02CH3, 0N02, N02, N3, NH2, heterocycloalkyl, heterocycloalkylaryl, aminoalkylamino, Pharmacodynamic properties of a polyalkylamino group, a substituted decyl group, an RNA cleavage group, a reporter group, an intercalation group, a pharmacokinetic property of a modified oligonucleotide, or a modified oligonucleotide a group, and other substituents having similar properties. Preferred modifications include 21-decyloxyethoxy (2'-0-158213.doc-62-201210611 CH2CH20CH3, also known as 2'-0-(2-methoxyethyl) or 2'-MOE) , that is, an alkoxyalkoxy group. Other preferred modifications include a 2'-dimethylaminooxyethoxy group as described herein below, that is, a 0(CH2)20N(CH3)2 group, also known as 2'-DMAOE; '-Dimethylaminoethoxyethoxy (also known in the art as 2·-0-dimethylaminoethoxyethyl or 2·-'DMAEOE), ie 2'-0 -CH2-0-CH2-N(CH2)2. Other preferred modifications include 2'-methoxy (2'-OCH3), 2'-aminopropoxy (2'-OCH2CH2CH2NH2) and 2'-fluoro (2'-F). It can also be carried out at other positions of the oligonucleotide, especially at the 3' end nucleotide or the 2'-5' ligation type oligonucleotide, at the 3' position of the sugar or at the 5' position of the 5' end nucleotide. Similar modification. Oligonucleotides may also have a sugar mimetic, such as a cyclobutyl moiety, rather than a pentofuranosyl sugar. Representative U.S. patents which teach the preparation of such modified sugar structures include, but are not limited to, U.S. Patent Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; No. 5, 466, 785; No. 5, 519, pp; No. 5, 576, 811; ; <5,670,633; and 5,700,920, each of which is incorporated herein by reference. Nucleotides may also be modified or substituted with nucleobases (often referred to in the art as "test bases"). As used herein, "unmodified" or "natural" nucleotides include the purine bases adenine (A) and black mites (G), and the chlorpyrifos thymine (T), cytosine (C), and urine. Pyrimidine (U). Modified nucleus 158213.doc -63- 201210611 The acid contains other synthetic and natural nucleotides, such as 5-methylcytosine (5-me-C), 5-hydroxydecyl cytosine, astragalus, hypoxanthine , 2-amino adenine, adenine, and guanine, 6-methyl and other leuco derivatives, adenine and bird σ 令 令 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 Sulphur urine, bite, 2-thiothymidine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azouracil, cytosine And thymidine, 5-urine bite (sudden urine 10 bite), 4-thiourea D bite, 8-halogenated adenine 11 and guanine, 8-aminoadenine and guanine, 8 -thiol adenine and guanine, 8-thioalkyl adenine and guanine, 8-hydroxyadenine and guanine and other 8-substituted adenines and birds 吟 吟, 5- _ base ( Especially 5-indifferent) urinary guanidine and cytosine, 5-trifluoromethyluracil and cytosine and other 5-substituted uracil and cytosine, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azadenine, 7-deazaguanine and 7- Adenine nitrogen and 3 to 11 nitrogen votes Yin and birds 3 - deazaadenine. In addition, the nucleotides include those described in U.S. Patent No. 3,687,808; "The Concise Encyclopedia of Polymer Science And Engineering", pp. 858-859, Kroschwitz, JI, John Wiley & Sons, 1990 The nucleotides disclosed therein; Englisch et al., "Angewandle Chemie, International Edition", 1991, 30, pp. 613, and their nucleotides; and Sanghvi, YS, Chapter 15, "Antisense Research and Applications, pp. 289-302, Croike, ST and Lebleu, B. ed., CRC Press, 1993. Certain of these nucleotides are particularly useful for enhancing the binding affinity of the oligomeric compounds of the present invention. These nucleotides include a 5-substituted pyrimidine 158213.doc-64-201210611 bite, a 6-nitrogen bite, and a N-2, N-6, and 0-6 substituted guanidine containing a 2-aminopropyl gland. Indole, 5-propynyl uracil and 5-propynyl cytosine. 5-methylcytosine substitutions have been shown to increase the stability of nucleic acid duplexes. (: to 1.2 C (Sanghvi, YS, Crooke, ST and Lebleu B. ed., "Antisense Research and Applications", CRC Press, Boca Raton, 1993 'pp. 276-278)' and even more specifically and 2, - The currently preferred base substitution when the methoxymethoxy saccharide modification is combined. A representative U.S. patent that teaches the preparation of the modified nucleotides described above, as well as other modified nucleotides, includes, but is not limited to, U.S. Patent Nos. 3,687,8,8, and 4,845,205; 5,130,302; 5,134,066, 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255 No. 5, 484, 908; No. 5, 502, 177; No. 5, 525, 711; No. 5, 552, 540; No. 5, 587, 469; No. 5, 596, 091; No. 5, 614, 617; No. 5, 750, 692; The method is incorporated herein. Another modification of the oligonucleotide of the present invention involves chemically linking to the activity, cell distribution or cellular uptake of one or more of the oligonucleotides. Or a combination. These moieties include, but are not limited to, lipid moieties such as cholesterol moieties, cholic acid, thioethers (eg, hexyl-S-trityl mercaptan), thiocholesterol, aliphatic chains (eg, twelve a diol or a decyl residue, a scale (for example, di-hexadecyl-racemic-glycerol or 1,2-hexadecyl-racemic-glyceryl-3H-phosphine Acid diethylammonium), polyamine or polyethylene glycol chain, or adamantane B 158213.doc -65- 201210611 acid, palmity moiety, or octadecylamine or hexylamino-carbonyl-hydroxycholesterol moiety. Representative U.S. patents for the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patent Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 730; 5,552,538; 5,578,717; 5.580.731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077 ; 5, 486, 603; 5, 512, 439; 5, 578, 718; 5, 608, 046; 4, 605, 044; 4, 667, 025; 4, 762, 779; 4, 789, 737; 4, 824, 941; 4, 835, 263; 4, 876, 335; 4, 904, 582; 4, 958, 013, 5.082.830; 5, 112, 963 No. 5,214,136; No. 5,082,830; No. 5,112,963; No. 5,214,136; No. 5,245,022; No. 5,254,469; No. 5,258,506; No. 5,262,536; No. 5,272,250; No. 5,292,873; Nos. 5,371,241; 5, 391, 723; 5, 416, 203; 5, 451, 463; 5, 510, 475; 5, 512, 667, 5, 514, 785; 5, 565, 552; 5, 567, 810; 5, 574, 142; No. 5,587, 371; 5,595, 726; 5, 597, 696; 5, 599, 923; 5, 599, 928 and 5, 688, 941, each of which is incorporated herein by reference. 158213.doc -66- 201210611 μ## Gift: The compounds of the invention can also be used in the field of drug discovery and target verification. The present invention encompasses the use of such compounds in drug discovery and the preferred target segments identified herein in an attempt to demonstrate nicotinic guanamine to ribosyl glucoside (NAMPT) polynucleotides and disease conditions, phenotypes or conditions There is a relationship between them. Such methods comprise detecting or modulating a τρτ polynucleotide comprising contacting a sample, tissue, cell or organism with a compound of the invention; measuring the nucleic acid or protein content of the NAMPT polynucleotide for a certain period of time after treatment and 'or a relevant phenotype or chemical endpoint; and compare the measured values to untreated samples or samples treated with other compounds of the invention, as appropriate. These methods can also be performed in parallel or in combination with other experiments to determine the function of an unknown gene for a target validation method, or to determine the effectiveness of a particular gene product as a target for treating or preventing a particular disease, condition or phenotype. Assessing gene expression up-regulation or inhibition: The transfer of an exogenous nucleic acid into a host cell or organism can be performed by directly detecting the presence of nucleic acids in the cell or organism (4). Such detection can be obtained by several methods well known in the art. For example, t, the presence of exogenous nucleic acid can be detected by Southern blot or by polymerase chain reaction (PCR) using primers that specifically amplify nucleic acid-related nucleotide sequences. . The performance of exogenous nucleic acids can also be measured using conventional methods including gene expression analysis. For example, Northern blotting and reverse transcription PCR (RT_PCR) can be used to detect and quantify mRNA produced by exogenous nucleic acids. The expression of RNA from an exogenous nucleic acid can also be detected by measuring enzymatic activity or reporter protein activity. For example, antisense modulating activity can be indirectly measured as a decrease or increase in the performance of a target nucleic acid indicative of an exogenous nucleic acid producing effector RNA as 158213.doc-67·201210611. Based on sequence conservation, the primers can be designed to amplify the coding region of the target gene. Initially, model control genes can be constructed using coding regions from the highest of each gene, but any coding or non-coding region can be used. Each control gene was combined by inserting each of the coding regions between the reporter coding region and its poly (A) signal. These plastids will produce mRNA with a reporter gene in the upstream part of the gene and a potential RNAi target in the 3, non-coding region. The efficacy of individual antisense oligonucleotides will be verified by adjusting the reporter gene. Reporter genes suitable for use in the methods of the invention include acetamyl hydroxy acid synthase (AHAS), test acylase (ap), beta galactosidase (LacZ), beta glucuronidase (GUS), and oxytetracycline Acetyltransferase (CAT), green fluorescent protein (GFP), red fluorescent protein (RFp), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), horseradish peroxidase (HRP), firefly Photozyme (Luc), nopaline synthase (N〇s), octopus synthase (OCS) and its derivatives. Give ampicillin, bleomycin, chloramphenicol, gentamycin, hygromycin, kanamycin, lincomycin A variety of selectable markers for resistance to methotrexate, phosphinothricin, puromycin, and tetracycline (tetracycline) are available. Methods for determining the regulation of a reporter gene are well known in the art and include, but are not limited to, fluorescence methods (e.g., fluorescence spectroscopy, fluorescence activated cell sorting (FACS), crab light microscopy), antibiotics. Resistance assay. 158213.doc • 68 - 201210611 NAMPT protein & mRNA expression can be assayed using methods known to those skilled in the art and described elsewhere herein. For example, an immunoassay such as eusa can be used to measure protein content. The beta NAmpt ELISA assay kit is commercially available, for example, from R & D Systems (Minneapolis, MN).

在實施例中,與對照樣品中之NAMPT表現相比較來評 估使用本發明之反義寡核苷酸處理之樣品(例如活體内細 胞或組織)中之NAMPT表現(例如mRNA或蛋白質)。舉例而 言,可使用熟習此項技術者已知之方法與模擬處理或未處 理樣品比較蛋白質或核酸之表現。或者,可視所需資訊而 疋與用對照反義寡核苷酸(例如具有改變或不同序列之反 義寡核苷酸)處理之樣品進行比較。在另一實施例中,處 理與未處理樣品中NAMPT蛋白質或核酸之表現差異可與 處理與未處理樣品中不同核酸(包括研究者認為適當之任 何標準物,例如管家基因)之表現差異相比較。 對於用於與對照組比較而言,觀測到之差異可視需要表 述為例如比率或分數形式。在實施例中,相對於未處理樣 品或用對照核酸處理之樣品,用本發明之反義寡核苷酸處 理之樣品中NAMPT mRNA或蛋白質之含量增加或降低約 1·25么至約1〇倍或10倍以上。在實施例中,mRNA 或蛋白質之含量增加或降低至少約125倍、至少約1 3倍、 至少約1.4倍、至少社5倍、至少約16倍、至少m 7倍、 至少約1.8倍、至少約2倍、至少約2 5倍、至少約3倍、至 少約3.5倍、至少約4倍、至少約45倍、至少約$倍、至少 約5·5倍、至少約6倍、至少約6·5倍、至少約7倍、至少約 158213.doc -69· 201210611 7 · 5倍、至少約8倍、至少幼s ς汾 至少約9.5 ^ 玍ν約8.5倍、至少約9倍 倍或至少約1 0倍或1 〇倍以上。 套組、研究試劑、診斷及治療 本發明化合物W料斷、治療及㈣且可用作研究試 劑及套組之組分。此外,—般枯彳奸去、g合& 又筏術者通常使用能夠以強烈 特異性抑制基因表現之反義裏妨过 取况心汉我养核苷酸來闡明特定基因之功 能或區別生物路徑之各種成員之功能。 對於用於套組及診斷及各種生物系統中而言,單獨或與 其他化合物或治療劑組合之本發明化合物適用作差別及、/ 或組合分析之m月細胞及組織内所表現之基因之 一部分或整個互補序列之表現模式。 如本文利之術語「生物系統」或「系統」定義為表現 或有能力表現菸鹼醯胺轉磷酸核糖基酶(nampt)基因之產 物的任何生物體、細胞、細胞培養物或組織。此等系統包 括(但不限於)人類、轉殖基因動物、細胞、細胞培養物匕 組織、異種移植物、移植物及其組合。 作為一個非限制性實例,將用一或多種反義化合物處理 之細胞或組織内之表現模式與未用反義化合物處理之對照 細胞或組織相比,且當該等模式與例如所研究基因之疾病' 關聯、信號傳導路徑、細胞定位、表現量、大小、結構或 功能相關時,針對不同基因表現量分析所產生之模式。此 等分析可對刺激或未刺激之細胞及在存在或不存在影響表 現模式之其他化合物下執行。 此項技術中已知之基因表現分析方法之實例包括dna陣 158213.doc -70- 201210611 列或微陣列、SAGE(基因表現之連續分析)、READS(消化 cDNA之限制酶擴增)、TOGA(總體基因表現分析)、蛋白質 陣列及蛋白質組研究、表現序列標籤(EST)定序、刪減 RNA指紋法(SuRF)、刪減選殖法、差別呈現法(DD)、比較 基因組雜交法、FISH(螢光原位雜交)技術及質譜分析方 ' 法。 本發明化合物適用於研究及診斷,此係因為此等化合物 與編碼菸鹼醯胺轉磷酸核糖基酶(NAMPT)之核酸雜交。舉 〇 例而言,以該效率且在該等條件下雜交之寡核苷酸如本文 所揭示作為有效NAMPT調節劑在有利於基因增殖或偵測 之條件下分別為有效引子或探針。此等引子及探針適用於 需要特異性偵測編碼NAMPT之核酸分子的方法,且適用 於偵測或用以其他NAMPT研究之該等核酸分子的擴增。 本發明之反義寡核苷酸,尤其引子及探針與編碼NAMPT 之核酸的雜交可藉由此項技術中已知之方法來偵測。該等 方法可包括酶與募核苷酸之結合、寡核苷酸之放射性標記 〇 或任何其他適合偵測方法。亦可製備使用該等偵測方法债 測樣品中NAMPT含量之套組。 '熟習治療性使用之技術者亦利用反義序列之特異性及敏 •感性。反義化合物已用作包括人類之動物中疾病病況之治 療中之治療性部分。已將反義寡核苷酸藥物安全且有效地 投與人類,且許多臨床試驗目前正在進行。因此確定,反 義化合物可為適用治療型態,其可經組態以適用於治療細 胞、組織及動物、尤其人類之治療方案。 158213.doc -71- 201210611 對於治療而言,懷疑患有可藉由調節NAMPT聚核苷酸 之表現治療之疾病或病症之動物(較佳人類)藉由投與本發 明之反義化合物來治療。舉例而言,在一非限制性實施例 中’該方法包含向需要治療之動物投與治療有效量之 NAMPT調節劑之步驟。本發明之namPT調節劑有效調節 NAMPT之活性或調節NAMPT蛋白質之表現。在一實施例 中’與對照組相比,動物中NAMPT之活性或表現抑制約 10%。動物中NAMPT之活性或表現較佳抑制約30%。動物 中NAMPT之活性或表現更佳抑制5〇%或50%以上。因此, 與對照組相比,募聚化合物調節菸鹼醯胺轉磷酸核糖基酶 (NAMPT)mRNA之表現達至少10%、至少50%、至少25%、 至少30%、至少40%、至少50%、至少60%、至少70%、至 少75%、至少80°/。、至少85%、至少90%、至少95%、至少 98%、至少99%或至少100%。 在一實施例中,與對照組相比,動物中菸鹼醯胺轉磷酸 核糖基酶(NAMPT)之活性或表現增加約10%。動物中 NAMPT之活性或表現較佳增加約30%。動物中NAMPT之 活性或表現更佳增加50%或50%以上。因此’與對照組相 比,寡聚化合物調節NAMPT mRNA之表現達至少1 〇%、至 少50%、至少25%、至少30%、至少40%、至少50%、至少 60%、至少70%、至少75%、至少80%、至少85%、至少 90%、至少95%、至少98%、至少99%或至少1〇〇%。 舉例而言,可在動物之血清、^液、脂肪組織、肝臟或 任何其他體液、組織或器官中量測菸鹼醯胺轉磷酸核糖基 158213.doc -72- 201210611 酶(NAMPT)之表現的增加或降低。該等所分析之流體、組 織或器官内所含有之細胞較佳含有編碼NAMPT肽之核酸 分子及/或NAMPT蛋白質本身。 本發明化合物可藉由向適合之醫藥學上可接受之稀釋劑 或載劑中添加有效量之化合物而以醫藥組合物形式利用。 _ 本發明化合物之用途及本發明之方法亦可適用於預防。 結合物 本發明寡核苷酸之另一修飾涉及化學連接至寡核苷酸之 〇 一或多個增強該寡核苷酸之活性、細胞分佈或細胞攝取之 部分或結合物。此等部分或結合物可包括共價結合至諸如 一級羥基或二級羥基之官能基之結合基團。本發明之結合 基團包括嵌入基、報導體分子、多元胺、聚醯胺、聚乙二 醇、聚醚、增強募聚物之藥效學性質之基團,及增強寡聚 物之藥物動力學性質之基團。典型結合基團包括膽固醇、 脂質、磷脂、生物素、啡嗪、葉酸、啡啶、蒽醌、吖啶、 螢光素、若丹明(rhodamine)、香豆素及染料。在本發明之 〇 上下文中,增強藥效學性質之基團包括改良攝取、增強降 解抗性及/或加強與目標核酸之序列特異性雜交之基團。 在本發明之上下文中,增強藥物動力學性質之基團包括改 •良本發明化合物之攝取、分佈、代謝或排出之基團。代表 性結合基團揭示於1992年10月23日申請之國際專利申請案 第PCT/US92/09196號及美國專利第6,287,860號中,該等文 獻以引用的方式併入本文中。結合部分包括(但不限於)脂 質部分,諸如贍固醇部分、膽酸、硫醚(例如己基-5-三苯 158213.doc -73- 201210611 甲基硫醇)、硫代膽固醇、脂族鍵(例如十二烧二醇或-j— 烷基殘基)、磷脂(例如二-十六烷基_外消旋_甘油或1,2-二-鄰十六烧基-外消旋-甘油基_3-H-膦酸三乙鍵)、多元胺或 聚乙二醇鏈、或金剛烷乙酸、棕櫚基部分、或十八烷基胺 或己基胺基-羰基-羥膽固醇部分。本發明之寡核苷酸亦可 結合至活性藥物,例如阿司匹林(aspirin)、華發靈 (warfarin)、苯基丁氮酮、布洛芬(ibupr〇fen)、舒洛芬 (suprofen)、酮洛芬(ketoprofen)、(S)-(+)-普拉洛芬((S)-(+)-pranoprofen)、卡洛芬(carprofen)、丹續醯基肌胺酸、 2,3,5-二破本甲酸、就芬那酸(fiufenamic acid)、搭葉酸 (folinic acid)、苯并》塞二唤、氯β塞嗪、二氣呼、。引β朵美辛 (indomethicin)、巴比妥酸鹽(barbiturate)、頭孢菌素 (cephalosporin)、磺胺劑、抗糖尿病藥、抗細菌劑或抗生 素。 教示該等寡核苷酸結合物之製備之代表性美國專利包括 (但不限於)美國專利第4,828,979號;第4,948,882號;第 5,218,105 號;第 5,525,465 號;第 5,541,313 號;第 5.545.730 號’·第 5,552,538 號;第 5,578,717 號;第 5.580.731 號;第 5,580,731 號;第 5,591,584 號;第 5,109,124 號;第 5,118,802 號;第 5,138,045 號;第 5,414,077 號;第 5,486,603 號;第 5,512,439 號;第 5,578,718 號;第 5,608,046 號;第 4,587,044 號;第 4,605,735 號;第 4,667,025 號;第 4,762,779 號;第 4,789,737 號;第 4,824,941 號;第 4,835,263 號;第 J58213.doc -74· 201210611In the Examples, NAMPT expression (e.g., mRNA or protein) in a sample (e.g., in vivo cells or tissues) treated with the antisense oligonucleotide of the present invention is evaluated in comparison to the NAMPT expression in a control sample. For example, the performance of a protein or nucleic acid can be compared to a simulated or untreated sample using methods known to those skilled in the art. Alternatively, the assay can be compared to a sample treated with a control antisense oligonucleotide (e.g., an antisense oligonucleotide having altered or different sequences), depending on the desired information. In another embodiment, the difference in performance of the NAMPT protein or nucleic acid in the treated and untreated samples can be compared to the difference in performance between different nucleic acids in the treated and untreated samples, including any standards deemed appropriate by the investigator, such as housekeeping genes. . For comparison with the control group, the observed difference can be expressed as, for example, a ratio or a fractional form. In an embodiment, the amount of NAMPT mRNA or protein in the sample treated with the antisense oligonucleotide of the present invention is increased or decreased by about 1.25 to about 1 Å relative to the untreated sample or the sample treated with the control nucleic acid. Multiple or more than 10 times. In embodiments, the amount of mRNA or protein is increased or decreased by at least about 125 times, at least about 13 times, at least about 1.4 times, at least 5 times, at least about 16 times, at least m 7 times, at least about 1.8 times, at least About 2 times, at least about 25 times, at least about 3 times, at least about 3.5 times, at least about 4 times, at least about 45 times, at least about $ times, at least about 5. 5 times, at least about 6 times, at least about 6 5 times, at least about 7 times, at least about 158213.doc -69 · 201210611 7 · 5 times, at least about 8 times, at least s ς汾 ς汾 at least about 9.5 ^ 玍 约 about 8.5 times, at least about 9 times or at least About 10 times or more than 1 time. Kits, Research Reagents, Diagnosis, and Treatment The compounds of the present invention are chopped, treated, and (4) and can be used as components of research reagents and kits. In addition, the general use of the antisense, which can inhibit the expression of the gene with strong specificity, may be used to elucidate the function of a specific gene or distinguish the biological path. The function of various members. For use in kits and diagnostics and in various biological systems, the compounds of the invention, alone or in combination with other compounds or therapeutic agents, are useful as part of the genes expressed in the cells and tissues of the m-months for differential and/or combinatorial analysis. Or the pattern of expression of the entire complementary sequence. The term "biological system" or "system" as used herein is defined to mean any organism, cell, cell culture or tissue that exhibits or is capable of exhibiting the product of the nicotinic acid phosphatase (nampt) gene. Such systems include, but are not limited to, humans, transgenic animals, cells, cell culture tissue, xenografts, grafts, and combinations thereof. As a non-limiting example, the expression pattern in a cell or tissue treated with one or more antisense compounds is compared to a control cell or tissue not treated with an antisense compound, and when such patterns are associated with, for example, the gene of interest When the disease's association, signaling pathway, cell localization, performance, size, structure, or function is correlated, the patterns produced by different gene expression analyses are generated. Such analysis can be performed on stimulated or unstimulated cells and in the presence or absence of other compounds that affect the performance pattern. Examples of gene expression analysis methods known in the art include DNA arrays 158213.doc-70-201210611 columns or microarrays, SAGE (continuous analysis of gene expression), READS (restriction enzyme amplification of digested cDNA), TOGA (overall) Gene expression analysis), protein array and proteomic research, expression sequence tag (EST) sequencing, deletion of RNA fingerprinting (SuRF), deletion and selection, differential presentation (DD), comparative genomic hybridization, FISH ( Fluorescence in situ hybridization) technique and mass spectrometry method. The compounds of the invention are useful in research and diagnostics because such compounds hybridize to a nucleic acid encoding a nicotine indoleamine phosphoribosyltransferase (NAMPT). For example, oligonucleotides that hybridize at such efficiencies and under such conditions, as disclosed herein, as effective NAMPT modulators, are effective primers or probes, respectively, under conditions conducive to gene proliferation or detection. Such primers and probes are suitable for use in methods that require specific detection of nucleic acid molecules encoding NAMPT and are useful for detecting or amplifying such nucleic acid molecules for use in other NAMPT studies. Hybridization of antisense oligonucleotides of the invention, particularly primers and probes, with nucleic acids encoding NAMPT can be detected by methods known in the art. Such methods may include binding of the enzyme to the raised nucleotide, radiolabeling of the oligonucleotide, or any other suitable detection method. Kits that use these detection methods to test the NAMPT content of the sample can also be prepared. Those skilled in the art of therapeutic use also utilize the specificity and sensitivity of the antisense sequence. Antisense compounds have been used as therapeutic moieties in the treatment of disease conditions in animals including humans. Antisense oligonucleotide drugs have been safely and effectively administered to humans, and many clinical trials are currently underway. It is therefore determined that the antisense compound can be in a therapeutic form that can be configured to be useful in the treatment of cells, tissues, and animals, particularly humans. 158213.doc -71- 201210611 For treatment, an animal (preferably a human) suspected of having a disease or condition treated by modulation of the expression of a NAMPT polynucleotide is treated by administering an antisense compound of the invention . For example, in one non-limiting embodiment, the method comprises the step of administering to a subject in need of treatment a therapeutically effective amount of a NAMPT modulator. The namPT modulator of the present invention is effective for modulating the activity of NAMPT or regulating the performance of NAMPT protein. In one embodiment, the activity or performance of NAMPT in the animal is inhibited by about 10% compared to the control group. The activity or performance of NAMPT in animals is preferably inhibited by about 30%. The activity or performance of NAMPT in animals is more preferably inhibited by 5% or more. Thus, the polymeric compound modulates the performance of nicotine indoleamine phosphoribosyltransferase (NAMPT) mRNA by at least 10%, at least 50%, at least 25%, at least 30%, at least 40%, at least 50 compared to the control group. %, at least 60%, at least 70%, at least 75%, at least 80°/. At least 85%, at least 90%, at least 95%, at least 98%, at least 99% or at least 100%. In one embodiment, the activity or performance of nicotinamide transphosphatase (NAMPT) in the animal is increased by about 10% compared to the control. The activity or performance of NAMPT in animals is preferably increased by about 30%. The activity or performance of NAMPT in animals is preferably increased by 50% or more. Thus, the oligomeric compound modulates the performance of NAMPT mRNA by at least 1%, at least 50%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, compared to the control group. At least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or at least 1%. For example, the performance of nicotine guanamine to phosphoribosyl 158213.doc-72-201210611 enzyme (NAMPT) can be measured in serum, liquid, adipose tissue, liver or any other body fluid, tissue or organ of an animal. Increase or decrease. The cells contained in the fluid, tissue or organ to be analyzed preferably contain a nucleic acid molecule encoding a NAMPT peptide and/or a NAMPT protein itself. The compounds of the present invention can be utilized in the form of a pharmaceutical composition by adding an effective amount of the compound to a suitable pharmaceutically acceptable diluent or carrier. The use of the compounds of the invention and the methods of the invention may also be suitable for prevention. Conjugates Another modification of an oligonucleotide of the invention involves the chemical ligation of one or more of the oligonucleotides to a portion or combination that enhances the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties or combinations may include binding groups that are covalently bonded to a functional group such as a primary hydroxyl group or a secondary hydroxyl group. The binding group of the present invention includes an embedding group, a reporter molecule, a polyamine, a polyamine, a polyethylene glycol, a polyether, a group for enhancing the pharmacodynamic properties of the polymer, and a drug power for enhancing the oligomer. The nature of the group. Typical binding groups include cholesterol, lipids, phospholipids, biotin, morphazine, folic acid, phenanthridine, anthraquinone, acridine, luciferin, rhodamine, coumarin, and dyes. In the context of the present invention, groups that enhance pharmacodynamic properties include groups that improve uptake, enhance resistance to degradation, and/or enhance sequence-specific hybridization to a target nucleic acid. In the context of the present invention, groups which enhance the pharmacokinetic properties include groups which modify the uptake, distribution, metabolism or excretion of the compounds of the invention. A representative binding group is disclosed in International Patent Application No. PCT/US92/09196, filed on Oct. 23, 1992, and U.S. Patent No. 6,287,860, the disclosure of which is incorporated herein by reference. Binding moieties include, but are not limited to, lipid moieties such as steroidal moieties, cholic acid, thioethers (eg, hexyl-5-triphenyl 158213.doc-73-201210611 methyl mercaptan), thiocholesterol, aliphatic bonds (eg, dodecadiol or -j-alkyl residue), phospholipids (eg, di-hexadecyl-racemic-glycerol or 1,2-di-hexadecanoyl-racemic-glycerol) a radical 3-3-H-phosphonic acid triethyl bond), a polyamine or polyethylene glycol chain, or an adamantane acetic acid, a palmity moiety, or an octadecylamine or a hexylamino-carbonyl-hydroxycholesterol moiety. The oligonucleotide of the present invention may also be bound to an active drug such as aspirin, warfarin, phenylbutazone, ibuprofen, supprofen, ketoprofen ( Ketoprofen), (S)-(+)-pranoprofen ((S)-(+)-pranoprofen), carprofen, carnitine, 2,3,5-di broken The present formic acid, fiufenamic acid, folinic acid, benzoxine, chlorpyrazine, and second gas. Indomethacin, barbiturate, cephalosporin, sulfonamides, antidiabetic agents, antibacterial agents or antibiotics. Representative U.S. patents which teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patent Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; .730, '5,552,538; 5,578,717; 5.580.731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; Nos. 5, 414, 037; 5, 486, 603; 5, 512, 439; 5, 578, 718; 5, 608, 046; 4, 587, 044; 4, 605, 735; 4, 667, 025; 4, 762, 779; 4, 789, 737; 4, 824, 941; 4, 835, 263; Doc -74· 201210611

第 4,904,582 號 第 5,112,963 號 第 5,112,963 號 第 5,254,469 號 第 5,272,250 號 第 5,371,241 號 第 5,451,463 號 第 5,514,785 號 第 5,574,142 號 第 5,595,726 號 第 5,599,928號及第 5,688,941 號。U.S. Patent No. 5, 119, 924, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No.

4,876,335 號 5.082.830 號 5.082.830 號 5,245,022 號 5,262,536 號 5,317,098 號 5,416,203 號 5,512,667 號 5,567,810 號 5,587,371 號 5,599,923號; 調配物 第 4,958,013 號; 第 第 5,214,136 號; 第 第 5,214,136 號; 第 第 5,258,506 號; 第 第 5,292,873 號; 第 第 5,391,723 號; 第 第 5,510,475 號; 第 第 5,565,552 號; 第 第 5,585,481 號; 第 第 5,597,696 號; 第 本發明化合物亦可混合、囊封、結合或以其他方式與化 合物之其他分子、分子結構或混合物缔合呈例如脂質體、 受體靶向分子、經口、直腸、局部或其他調配物形式以促 進攝取、分佈及/或吸收。教示該等促進攝取、分佈及/或 吸收之調配物之製備的代表性美國專利包括(但不限於)美 國專利第5,108,921號,第 5,354,844號;第5,416,016號; 第 5,459,127 號;第 5,521,291 號;第 5,543,165 號;第 5,547,932 號;第 5,583,020 號;第 5,591,721 號;第 4,426,330 號;第 4,534,899 號;第 5,013,556 號;第 5,108,921 號;第 5,213,804 號;第 5,227,170 號;第 5,264,221 號;第 5,356,633 號;第 5,395,619 號;第 5,416,016 號;第 5,417,978 號;第 5,462,854 號;第 158213.doc -75- 201210611 5,469,854 號;第 5,512,295 號;第 5,527,528 號;第 5,534,259 號;第 5,543,152 號;第 5,556,948 號;第 5,580,575號;及第5,595,756號,各文獻均以引用的方式併 入本文中。 儘管反義寡核苷酸不需要在載體之情形中投與以調節目 標表現及/或功能,但本發明之實施例係關於用於表現反 義募核苷酸之表現載體構築體,其包含啟動子、雜交啟動 基因序列,且具有強組成性啟動子活性或在所需情況下可 誘導之啟動子活性。 在一實施例中,本發明實踐涉及投與上述反義寡核苷酸 中之至少一者及適合核酸傳遞系統。在一實施例中,彼系 統包括非病毒載體可操作地連接至聚核苷酸。該等非病毒 載體之實例包括單獨募核苷酸(例如SEQ ID NO: 17至31之 任一或多者)或與適合蛋白質、多醣或脂質調配物之組 合。 另外適合之核酸傳遞系統包括病毒載體,通常為來自腺 病毒、腺病毒相關病毒(AAV)、辅助病毒依賴性腺病毒、 反轉錄病毒或曰本脂質體血球凝集素病毒(hemagglutinatin virus of japan_lip〇s〇me ; HVJ)複合物中之至少一者之序 列。病毒載體較佳包含強真核啟動子可操作地連接至聚核 苷酸(例如細胞巨大病毒(CMV)啟動子)。 另外較佳之載體包括病毒載體、融合蛋白及化學結合 物。反轉錄病毒載體包括莫洛尼鼠類白血病病毒(Moloney murine !eukemia viruses)及基於HIV之病毒。—種較佳之基 158213.doc -76- 201210611 於HIV之病毒載體包含至少兩個載體,其中㈣及ρ〇ι基因 係來自HIV基因組且env基因係來自另—病毒。dna病毒載 體:佳。此等載體包括痘病毒載體,諸如正殖病毒或禽痘 病毋載體,疱疹病毒載體,諸如單純疱疹!病毒(hsv)載 體,腺病毒載體;及腺相關病毒载體。 本發明之反義化合物涵蓋任何醫藥學上可接受之鹽、酯 或忒等Sg之鹽,或投與包括人類之動物時能夠提供(直接 或間接)生物學活性代謝物或其殘餘物之任何其他化合 物0 術語「醫藥學上可接受之鹽」係指本發明化合物之生理 學上及醫藥學上可接受之鹽:亦即保留母體化合物之所需 生物活性且不會賦予其不當毒理學作用之鹽。對於寡核苷 酸而言,醫藥學上可接受之鹽及其使用之較佳實例進一步 描述於美國專利第6,287,86〇號中,該文獻以引用的方式併 入本文中。 本發明亦包括包含本發明之反義化合物之醫藥組合物及 調配物。本發明之醫藥組合物可視是否需要局部或全身性 治療及待治療之面積以許多方法投與。投藥可為局部(包 括眼用及投與黏膜(包括陰道及直腸傳遞)、肺部(例如藉由 吸入或吹入粉末或氣霧劑,包括藉由噴霧器氣管内、 鼻内、表皮及經皮)、經口或非經腸投藥。非經腸投藥包 括靜脈内、動脈内、皮下、腹膜内或肌肉内注射或輸注; 或顱内’例如鞘内或心室内投藥。 對於治療中樞神經系統中之組織而言,可藉由例如注射 158213.doc •77- 201210611 或輸注投與腦脊髓液中。將反義RNA投與腦脊髓液中描述 於例如美國專利申請公開案第2007/0117772, 「Methods for slowing familial ALS disease progression」號中,該文 獻以全文引用的方式併入本文中。 當本發明之反義募核苷酸意欲投與中樞神經系統中之細 胞時,可與一或多種能夠促進標的反義寡核苷酸穿過血腦 障壁之藥劑一起投與。可例如在内嗅皮層或海馬區中進行 注射。藉由向肌肉組織中之運動神經元投與腺病毒載體來 傳遞神經營養因子描述於例如美國專利第6,632,427號, 「Adenoviral-vector-mediated gene transfer into medullary motor neurons」中,該文獻以引用的方式併入本文中。將 載體直接傳遞至腦,例如紋狀體、視丘、海馬區或黑質在 此項技術中已知且描述於例如美國專利第6,756,523號, 「Adenovirus vectors for the transfer of foreign genes into cells of the central nervous system particularly in brain」 中,該文獻以引用的方式併入本文中。投與可如藉由注射 快速進行或如藉由缓慢輸注或投與緩釋型調配物在一段時 間内進行。 標的反義寡核苷酸亦可與提供所需醫藥或藥效學性質之 藥劑連接或結合。舉例而言,反義寡核苷酸可與此項技術 中已知促進滲透或傳輸穿過血腦障壁之任何物質(諸如運 鐵蛋白受體之抗體)偶合,且藉由靜脈内注射投與。反義 化合物可與例如使反義化合物更有效及/或使反義化合物 穿過血腦障壁之傳輸增加的病毒載體連接。滲透性血腦障 158213.doc -78- 201210611 壁破環亦可藉由例如輸注以下來實現:糖,包括(但不限 於)内消旋赤藻糖醇、木糖醇、D(+)半乳糖、D(+)乳糖、 D(+)木糖、半乳糖醇、肌醇、L(-)果糖、D㈠甘露糖醇、 D(+)葡萄糖、D(+)阿拉伯糖、D㈠阿拉伯糖、纖維二糖、 D(+)麥芽糖、D(+)棉子糖、L(+)鼠李糖、D(+)蜜二糖、D(-) ' 核糖、阿東糖醇、D(+)阿糖醇、L(-)阿糖醇、D(+)岩藻 糖、L(-)岩藻糖、D(-)來蘇糖、L(+)來蘇糖及L(-)來蘇糖; 或胺基酸,包括(但不限於)麩醯胺酸、離胺酸、精胺酸、 〇 天冬醯胺、天冬胺酸、半胱胺酸、麩胺酸、甘胺酸、組胺 酸、白胺酸、曱硫胺酸、苯丙胺酸、脯胺酸、絲胺酸、蘇 胺酸、酪胺酸、纈胺酸及牛磺酸。增強血腦障壁滲透之方 法及物質描述於例如美國專利第4,866,042號,「Method for the delivery of genetic material across the blood brain barrier」;第 6,294,520號,「Material for passage through the blood-brain barrier」;及第 6,936,589 號,「Parenteral delivery systems」中,所有文獻均以全文引用的方式併入 ❹ 本文中。 標的反義化合物可混合、囊封、結合或以其他方式與化 合物之其他分子、分子結構或混合物締合呈例如脂質體、 受體靶向分子、經口、直腸、局部或其他調配物形式以促 進攝取、分佈及/或吸收。舉例而言,陽離子型脂質可包 括於調配物中以促進寡核苷酸攝取。一種顯示促進攝取之 該組合物為 LIPOFECTIN(可自 GIBCO-BRL,Bethesda,MD 獲得)。 158213.doc -79- 201210611 咸信具有至少一個2'-0-甲氧基乙基修飾之募核苷酸尤其 適用於經口投藥。用於局部投與之醫藥組合物及調配物可 包括經皮貼片、軟膏、洗劑、乳霜、凝膠、滴劑、栓劑、 噴霧劑、液體及散劑。習知醫藥載劑、水性、粉末或油性 基質、增稠劑及其類似物可能必需或合乎需要。經塗佈保 險套、手套及其類似物亦可適用。 可根據製藥工業中熟知之習知技術製備宜呈單位劑型之 本發明之醫藥調配物。該等技術包括使活性成分與醫藥載 劑或賦形劑締合之步驟。一般而言,調配物藉由使活性成 份與液體載劑或細粉狀固體載劑或二者均一且密切缔合, 且接著(若必要)使產物成型來製備。 本發明之組合物可調配為許多可能存在之劑型中之任一 者,諸如(但不限於)錠劑、膠囊、凝膠膠囊、液體糖漿、 軟凝膠、栓劑及灌腸劑。本發明之組合物亦可調配成為於 水性、非水性或混合介質中之懸浮液。水性料液可進一 步含有使懸浮液之黏度增加之物質,包_如"基纖維 素納、山梨糖醇及/或葡聚糖。懸浮液亦可含有穩㈣。 本發明之醫藥組合物包括(但不限於)溶液、乳液、泡珠 及含有脂質體之調配物。本發明之醫藥組合物及調配物可 包含-或多種滲透增強劑、载劑、賦形劑或其他活性或非 活性成分。 式=二Γ 直徑通常超過°·1-之小液滴形 異質系統。乳液除分散相外亦可 3有其他組分及可於水相、油相令呈溶液形式或本身作為 158213.doc -80- 201210611 單獨相形式存在之活性藥物。包括微乳液作為本發明之一 實施例。乳液及其使用在此項技術中熟知且進一步描述於 美國專利第6,287,860號中。 本發明之調配物包括脂質體調配物。如本發明中所用之 術語「脂質體」意謂由以球狀雙層或雙層排列之兩親媒性 脂質構成的囊泡。脂質體為單層或多層囊泡,其具有由親 脂性物質形成之膜及含有待傳遞組合物之含水内部。陽離 子型脂質體為帶正電脂質體,咸信其會與帶負電DNA分子 相互作用形成穩定複合物。咸信pH值敏感型或帶負電之脂 質體會截留DNA而非與其產生複合物。已使用陽離子型及 非陽離子型脂質體將DNA傳遞至細胞中。 脂質體亦包括「空間穩定」脂質體,如本文所用之該術 語係指包含一或多種特殊脂質之脂質體。當併入脂質體中 時,此等特殊脂質會產生相對於缺乏該等特殊脂質之脂質 體具有增強之循環壽命的脂質體。空間穩定脂質體之實例 為脂質體之囊泡形成脂質部分之一部分包含一或多種糖脂 或經一或多種諸如聚乙二醇(PEG)部分之親水性聚合物衍 生化的彼等脂質體。脂質體及其使用進一步描述於美國專 利第6,287,860號中。 本發明之醫藥調配物及組合物亦可包括界面活性劑。藥 品、調配物及乳液中界面活性劑之使用在此項技術中熟 知。界面活性劑及其使用進一步描述於美國專利第 6,287,860號中,該文獻以引用的方式併入本文中。 在-實施例中,本發明利用各種渗透增強劑以影響核 158213.doc -81 · 201210611 酸、尤其寡核苷酸之有效傳遞。除有助於非親脂性藥物擴 散穿過細胞膜之外,滲透增強劑亦增強親脂性藥物之滲透 ! 生滲透增強劑可分類為屬於五個大類中之一者,亦即界 面活性劑、脂肪酸、膽汁鹽、螯合劑及非螯合非界面活性 劑。滲透增強劑及其使用進一步描述於美國專利第 6,287,860號中,該文獻以引用的方式併入本文中。 熟習此項技術者將認識到調配物通常根據其預定用途, 亦即投藥途徑來進行設計。 用於局。卩投與之較佳調配物包括本發明之募核苷酸與諸 如月曰質、脂質體、脂肪酸、脂肪酸酯、類固醇、螯合劑及 界面活性劑之局部傳遞劑混合之彼等調配物。較佳脂質及 月曰質體包括中性脂質及脂質體(例如二油醯基-磷脂醯基 D〇PE乙醇胺、二肉豆蔻醯磷脂醯膽鹼DMPC '二硬脂醯磷 月曰醯膽鹼)、負性脂質及脂質體(例如二肉豆蔻醯基磷脂醯 基甘油DMPG)及陽離子型脂質及脂質體(例如二油醯基四 甲基胺基丙基D〇TAp及二油醯基-磷脂醯乙醇胺 DOTMA)。 對於局部或其他投與而言,本發明之寡核苷酸可囊封在 月日處體内或可與其形成複合物,尤其與陽離子型脂質體形 成複合物。或者,募核苷酸可與脂質複合,尤其與陽離子 型月曰質複合。較佳脂肪酸及其酯、醫藥學上可接受之鹽及 其使用進一步描述於美國專利第6,287,86〇號中。 用於口投藥之組合物及調配物之包括散劑或顆粒、微 微奈米微粒、於水或非水性介質中之懸浮液或溶液、膠 158213.doc •82· 201210611 囊、凝膠膠囊、筚憂、β + 蘇❺η _、㈣。增_、調味劑、 稀㈣、乳化劑、分散助劑或黏合劑可能合 紐口調配物為聯合投與本發明之寡核皆酸與 = 增強劑、界面活性劑及螯合 ,參透 ^, 劑之凋配物。較佳界面活性劑 匕括脂肪酸及/或其㈣鹽、膽汁酸及/或其鹽。較佳膦汁 酸’鹽及脂肪酸及其使用進-步描述於美國專利°第 6,287,_射’該文獻以μ的方式併人本文卜亦較佳U.S. Patent Nos. 4,958,013; 5, 214, 136; 5, 214, 136; 5, 214, pp; 5, 214, 136; 5, 258, 506; 5, 214, 136; No. 5, 391, 723; No. 5, 510, 475; No. 5, 565, 552; No. 5,585, 481; No. 5,597, 696; Compounds of the invention may also be mixed, encapsulated, bound or otherwise Other molecules, molecular structures or mixtures are associated, for example, in the form of liposomes, receptor targeting molecules, oral, rectal, topical or other formulations to facilitate uptake, distribution and/or absorption. Representative U.S. patents which teach the preparation of such formulations which promote ingestion, distribution and/or absorption include, but are not limited to, U.S. Patent Nos. 5,108,921, 5,354,844; 5,416,016; 5,459,127; 5,521, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, No. 5, 256, 221; 5, 356, 633; 5, 395, 619; 5, 416, 016; 5, 417, 978; 5, 462, 854; 158, 213. doc - 75 - 201210611 5, 469, 854; 5, 512, 295; 5, 527, 528; 5, 534, 259; 5, 543, 152 No. 5,556, 948; 5, 580, 575; and 5, 595, 756, each of which is incorporated herein by reference. Although antisense oligonucleotides need not be administered in the context of a vector to modulate target expression and/or function, embodiments of the invention pertain to expression vector constructs for expressing antisense nucleotides, including Promoter, hybridization initiation gene sequence, and has strong constitutive promoter activity or promoter activity that can be induced if desired. In one embodiment, the practice of the invention involves administering at least one of the above antisense oligonucleotides and a suitable nucleic acid delivery system. In one embodiment, the system includes a non-viral vector operably linked to the polynucleotide. Examples of such non-viral vectors include individual nucleotides (e.g., any one or more of SEQ ID NOS: 17 to 31) or a combination of suitable protein, polysaccharide or lipid formulations. Further suitable nucleic acid delivery systems include viral vectors, typically from adenovirus, adeno-associated virus (AAV), helper virus-dependent adenovirus, retrovirus or hemagglutinatin virus of japan_lip〇s〇. a sequence of at least one of the HVJ) complexes. The viral vector preferably comprises a strong eukaryotic promoter operably linked to a polynucleotide (e.g., a cellular giant virus (CMV) promoter). Further preferred vectors include viral vectors, fusion proteins and chemical conjugates. Retroviral vectors include Moloney murine! eukemia viruses and HIV-based viruses. A preferred base 158213.doc -76- 201210611 The viral vector for HIV comprises at least two vectors, wherein the (iv) and ρ〇ι genes are from the HIV genome and the env gene line is from another virus. DNA virus carrier: good. Such vectors include poxvirus vectors such as cloning virus or fowl pox vector, herpes virus vectors such as herpes simplex! Virus (hsv) vector, adenoviral vector; and adeno-associated virus vector. The antisense compound of the present invention encompasses any salt of Sg such as a pharmaceutically acceptable salt, ester or hydrazine, or any of the biologically active metabolites or residues thereof which are capable of providing (directly or indirectly) when administered to an animal including humans. Other Compounds 0 The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the compounds of the present invention: that is, the desired biological activity of the parent compound is retained and does not confer unsuitable toxicology. The salt of action. For oligonucleotides, pharmaceutically acceptable salts and preferred examples of their use are further described in U.S. Patent No. 6,287,86, the disclosure of which is incorporated herein by reference. The invention also includes pharmaceutical compositions and formulations comprising the antisense compounds of the invention. The pharmaceutical compositions of the present invention can be administered in a number of ways depending on whether local or systemic treatment and the area to be treated are required. Administration may be topical (including ophthalmic and mucosal (including vaginal and rectal transmission), lung (eg by inhalation or insufflation of powder or aerosol, including intratracheal, intranasal, epidermal and transdermal by nebulizer) Oral or parenteral administration. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial 'eg intrathecal or intraventricular administration. For treatment of the central nervous system The tissue can be administered to the cerebrospinal fluid by, for example, injection 158213.doc • 77-201210611 or infusion. The administration of antisense RNA to cerebrospinal fluid is described, for example, in U.S. Patent Application Publication No. 2007/0117772, In the method for slowing familial ALS disease progression, the document is incorporated herein by reference in its entirety. When the antisense nucleotide of the present invention is intended to be administered to cells in the central nervous system, one or more The agent that promotes the targeting of antisense oligonucleotides across the blood-brain barrier can be administered together, for example, in the entorhinal cortex or hippocampus. By moving neurons into muscle tissue The delivery of a neurotrophic factor with an adenoviral vector is described, for example, in "Adenoviral-vector-mediated gene transfer into medullary motor neurons", which is incorporated herein by reference. Adenovirus vectors for the transfer of foreign genes into cells of the central nervous system particularly in brain, such as the striatum, the hypothalamus, the hippocampus, or the substantia nigra, are known in the art and are described, for example, in U.S. Patent No. 6,756,523. This document is incorporated herein by reference. Administration can be carried out rapidly, for example by injection, or by slow infusion or administration of a sustained release formulation over a period of time. It can also be linked or associated with an agent that provides the desired pharmaceutical or pharmacodynamic properties. For example, an antisense oligonucleotide can be any substance known in the art to promote penetration or transport across the blood-brain barrier (such as The antibody to the transferrin receptor is coupled and administered by intravenous injection. Antisense compounds can be used, for example, to make antisense compounds Increased and/or increased viral vector ligation of antisense compounds across the blood-brain barrier. Permeable blood-brain disorders 158213.doc -78- 201210611 Wall-breaking can also be achieved by, for example, infusion: sugar, including ( But not limited to) meso-erythritol, xylitol, D(+)galactose, D(+) lactose, D(+) xylose, galactitol, inositol, L(-) fructose, D(a) Mannitol, D(+)glucose, D(+)arabinose, D(mono)arabinose, cellobiose, D(+)maltose, D(+) raffinose, L(+)rhamnose, D(+ )disaccharide, D(-) 'ribose, adonitol, D(+) arabitol, L(-) arabitol, D(+) fucose, L(-) fucose, D ( -) tosuose, L (+) to threose and L (-) to threose; or amino acids, including (but not limited to) glutamic acid, lysine, arginine, scorpion Amine, aspartic acid, cysteine, glutamic acid, glycine, histidine, leucine, guanidine, phenylalanine, valine, serine, threonine, tyramine Acid, proline and taurine. Methods and materials for enhancing blood-brain barrier penetration are described in, for example, U.S. Patent No. 4,866,042, "Method for the delivery of genetic material across the blood brain barrier"; No. 6,294,520, "Material for passage through the blood-brain barrier"; No. 6,936,589, "Parenteral delivery systems", all of which are incorporated herein by reference in its entirety. The subject antisense compound can be mixed, encapsulated, bound or otherwise associated with other molecules, molecular structures or mixtures of the compounds in the form of, for example, liposomes, receptor targeting molecules, oral, rectal, topical or other formulations. Promote uptake, distribution and/or absorption. For example, cationic lipids can be included in the formulation to facilitate oligonucleotide uptake. One composition which exhibits uptake is LIPOFECTIN (available from GIBCO-BRL, Bethesda, MD). 158213.doc -79- 201210611 The nucleotides with at least one 2'-0-methoxyethyl modification are particularly suitable for oral administration. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated covers, gloves and the like are also suitable. Pharmaceutical formulations of the invention, preferably in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredient with a pharmaceutical carrier or excipient. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with a liquid carrier or a finely divided solid carrier or both, and, if necessary, shaping the product. The compositions of the present invention may be formulated into any of a number of possible dosage forms such as, but not limited to, lozenges, capsules, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the present invention may also be formulated as a suspension in an aqueous, non-aqueous or mixed medium. The aqueous feed liquid may further contain a substance which increases the viscosity of the suspension, such as "cellulosin, sorbitol and/or dextran. The suspension may also contain a steady (four). Pharmaceutical compositions of the invention include, but are not limited to, solutions, emulsions, beads, and formulations containing liposomes. The pharmaceutical compositions and formulations of the present invention may comprise - or a plurality of penetration enhancers, carriers, excipients or other active or inactive ingredients. Formula = II 小 A small droplet-shaped heterogeneous system usually exceeding the diameter of °·1. In addition to the dispersed phase, the emulsion may have other components and active agents which may be present in the form of a liquid phase, an oil phase, or as a separate phase in the form of 158213.doc -80-201210611. Microemulsions are included as an embodiment of the invention. Emulsions and their use are well known in the art and are further described in U.S. Patent No. 6,287,860. Formulations of the invention include liposome formulations. The term "liposome" as used in the present invention means a vesicle composed of two borne lipids arranged in a spherical double layer or a double layer. Liposomes are monolayer or multilamellar vesicles having a membrane formed from a lipophilic material and an aqueous interior containing the composition to be delivered. The cationic liposome is a positively charged liposome, which interacts with a negatively charged DNA molecule to form a stable complex. A pH-sensitive or negatively charged liposome will retain DNA rather than form a complex with it. Both cationic and non-cationic liposomes have been used to deliver DNA into cells. Liposomes also include "sterically stabilized" liposomes, as used herein, which refers to liposomes comprising one or more specific lipids. When incorporated into liposomes, these particular lipids produce liposomes with enhanced cycle life relative to liposomes lacking such particular lipids. An example of a sterically stabilized liposome is one in which a portion of the vesicle-forming lipid portion of the liposome comprises one or more glycolipids or those liposomes that are derivatized with one or more hydrophilic polymers such as polyethylene glycol (PEG) moieties. Liposomes and their use are further described in U.S. Patent No. 6,287,860. The pharmaceutical formulations and compositions of the present invention may also include a surfactant. The use of surfactants in pharmaceuticals, formulations and emulsions is well known in the art. Surfactants and their use are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. In an embodiment, the present invention utilizes various permeation enhancers to effect efficient delivery of the nucleus, particularly the oligonucleotides, 158213.doc-81.201210611. In addition to facilitating the diffusion of non-lipophilic drugs across the cell membrane, penetration enhancers also enhance the penetration of lipophilic drugs! Biopenetration enhancers can be classified into one of five broad categories, namely surfactants, fatty acids, Bile salts, chelating agents and non-chelating non-surfactants. Penetration enhancers and their use are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. Those skilled in the art will recognize that formulations are typically designed according to their intended use, i.e., the route of administration. Used for bureau. Preferred formulations of the present invention include those formulations in which the nucleotides of the present invention are mixed with topical delivery agents such as quercetin, liposomes, fatty acids, fatty acid esters, steroids, chelating agents, and surfactants. Preferred lipids and sorghum plastids include neutral lipids and liposomes (eg, dioleoyl-phospholipid oxime D〇PE ethanolamine, dimyristoyl phosphatidylcholine DMPC 'distearate phosphatidylcholine choline ), negative lipids and liposomes (such as dimyristylphospholipid glycerol DMPG) and cationic lipids and liposomes (eg diterpenoid tetramethylaminopropyl D〇TAp and dioleyl) Phospholipid 醯ethanolamine DOTMA). For topical or other administration, the oligonucleotides of the invention may be encapsulated within or may form a complex with the compound, particularly with cationic liposomes. Alternatively, the raised nucleotides can be complexed with lipids, especially with cationic menthosides. Preferred fatty acids and their esters, pharmaceutically acceptable salts and their use are further described in U.S. Patent No. 6,287,86. Compositions and formulations for oral administration include powders or granules, micronized microparticles, suspensions or solutions in water or non-aqueous medium, and gels 158213.doc •82· 201210611 Capsules, gel capsules, worries , β + Su Shi η _, (four). Adding _, flavoring agent, dilute (four), emulsifier, dispersing aid or binder may be a combination of the oligonucleotides of the present invention and the reinforced agent, the surfactant, and the chelate, Ingredients of the agent. Preferred surfactants include fatty acids and/or their salts, bile acids and/or salts thereof. Preferred phosphonate acids and salts and their use are described in U.S. Patent No. 6,287, the disclosure of which is incorporated herein by reference.

為渗透增強劑之組合,例如脂肪酸/鹽與膽汁酸/鹽組合。 尤其較佳組合為月桂酸、癸酸及udca之鈉鹽。其他涞透 增強劑包括聚氧乙稀_9_月桂醚、聚氧乙稀·2〇__ :本 發明之寡核苦酸可以包括噴霧乾燥粒子或複合形成微米或 奈米粒子之顆粒形式經口傳遞。寡㈣酸複合劑及其使用 進一步描述於美國專利第6,287,86晚中,該文獻以引用的 方式併入本文中。 用於非經腸、鞘内或心室内投與之組合物及調配物可包 括亦可含有緩衝劑、稀釋劑及其他適合添加劑(諸如(但不 限於)滲透增強劑、載劑化合物及其他醫藥學上可接受之 載劑或賦形劑)之無菌水溶液。 本發明之某些實施例提供含有一或多種募聚化合物及一 或多種藉由非反義機制起作用之其他化學治療劑之醫藥組 合物。該等化學治療劑之實例包括(但不限於)癌症化學治 療藥物’諸如柔紅黴素(daun〇rubicin)、道諾黴素 (daunomycin)、更生徽素(dactinomyCin)、小紅莓(d〇x〇rubicin)、 表柔比星(epirubicin)、伊達比星(idarubicin)、依索比星 158213.doc -83· 201210611 (esorubicin)、博萊徽素(bleomycin)、馬鱗醢胺(mafosfamide)、 異環墙酿胺(ifosfamide)、胞續咬阿拉伯糖苦(cytosine arabinoside)、雙氣乙基-亞硝基腺(bischloroethyl-nitrosurea)、白消安(busulfan)、絲裂黴素 C(mitomycin C)、放線菌素 D(actinomycin D)、米拉黴素(mithramycin)、 潑尼松(prednisone)、經助孕酮(hydroxyprogesterone)、睾 固酮(testosterone)、他莫昔芬(tamoxifen)、達卡巴。秦 (dacarbazine)、丙卡巴肼(procarbazine)、六曱基三聚氰胺 (hexamethylmelamine)、五甲基三聚氰胺、米托蒽酿 (mitoxantrone)、安 °丫 °定(amsacrine)、苯丁 酸氮芬 (chlorambucil)、甲基環己基亞硝基脲、氮芥(nitrogen mustards)、美法侖(melphalan)、環鱗酿胺 (cyclophosphamide)、6-疏嗓吟、6-硫鳥嗓吟、阿糖胞皆、 5-氮雜胞苷、經基脲、脫氧柯福黴素(deoxycoformycin)、 4-羥基過氧化環-磷醯胺、5-氟尿嘧啶(5-FU)、5-氟去氧尿 苷(5-FUdR)、曱胺喋呤(MTX)、秋水仙鹼(colchicine)、紫 杉酌 (taxol)、 長春新鹼(vincristine)、 長春驗 (vinblastine)、依託泊普(etoposide,VP-16)、三甲曲沙 (trimetrexate)、伊立替康(irinotecan)、拓朴替康 (topotecan)、吉西他濱(gemcitabine)、替尼泊苦 (teniposide)、 順 翻(cisplatin)及己 稀雌盼 (diethylstilbestrol,DES)。當與本發明化合物一起使用 時,該等化學治療劑可個別(例如5-FU及寡核苷酸)、依序 (例如5-FU及寡核苷酸歷時一段時間,之後MTX及寡核苷 158213.doc -84- 201210611 酸)或與一或多種其他該等化學治療劑(例如5_FU、Μτχ& 募核苷酸,或5-FU、放射線療法及寡核苷酸)組合使用。 包括(但不限於)非類固醇消炎藥及皮質類固醇之消炎藥及 包括(但不限於)病毒唑(ribivirin)、阿糖腺苷(vidarabine)、 阿昔洛韋(acyclovir)及更昔洛韋(gancicl〇vir)之抗病毒藥亦 可與本發明組合物組合。反義化合物及其他非反義藥物之 組合亦在本發明之範疇内。兩種或兩種以上組合化合物可 一起或依序使用。 在另一相關實施例中,本發明之組合物可含有一或多種 乾向第一核酸之反義化合物(尤其寡核苷酸),及一或多種 靶向第二核酸目標之其他反義化合物。舉例而言,第一目 標可為於驗醢胺轉磷酸核糖基酶(NAMPT)之特定反義序 列’且第一目標可為來自另·一核普酸序列之區。或者,本 發明之組合物可含有兩種或兩種以上靶向同一菸鹼醯胺轉 磷酸核糖基酶(NAMPT)核酸目標之不同區的反義化合物。 本文說明反義化合物之許多實例,且其他可選自此項技術 中已知之適合化合物。兩種或兩種以上組合化合物可一起 或依序使用。 給藥: 咸信治療組合物之調配及其後續投與(給藥)在熟習此項 技術者之能力範圍内。給藥視待治療之疾病病況之嚴重程 度及反應而定,其中治療過程持續若干天至若干個月 咬 直至實現治癒或達成疾病病況減輕。最佳給藥時程可由串 者體内藥物累積之量測值來計算。一般技術者可容易地確 158213.doc -85- 201210611 定最佳劑量、給藥方法及重複率。最佳劑量可視個別寡核 苷酸之相對功效而變化,且—般可基於發現在活體外及活 體内動物模财有效之EC50估算。一般而言,劑量為每公 斤體重ο·οι獅10 mg,且可每A、每週、每月或每年給 予一次或一次以上,或甚至每2至20年給予一次。—般技 術者可基於體液或組織中之藥物之實測滯留時間及濃度估 算給藥之重複率。在成功治療後,患者可能需要進行維持 療法以防止疾病病況復發,其中募核苷酸以每公斤體重 〇·〇1 Kg至10 mg範圍内之維持劑量投與,每天投與—次或 一次以上至每2至2〇年投與一次。 在貫施例中,患者用以下劑量之藥物治療,該劑量為每 公斤體重至少約1 mg、至少約2 mg、至少約3 mg、至少約 4 mg、至少約5 mg、至少約6 mg、至少約7 mg、至少約8 mg、至少約9 mg、至少約1〇 mg、至少約15 mg、至少約 20 mg、至少約25 mg、至少約30 mg、至少約35 mg '至少 約40 mg、至少約45 mg、至少約5〇 mg、至少約6〇 mg、至 >、約70 mg、至少約80 mg、至少約9〇 mg或至少約1〇 mg。 反義寡核苷酸之某些注射劑量描述於例如美國專利第 7,563,884號,「Antisense modulati〇n 〇f ρτρΐΒ εχρΓ_〇η」 中’該文獻以全文引用的方式併入本文中。 儘管上文已描述本發明之各種實施例,但應瞭解其僅以 例之方式呈現,而不具限制性。可在不背離本發明之精神 或範疇下根據本文之揭示内容對所揭示實施例進行許多改 變。因此,本發明之廣度及範疇不應受上述任何實施例限 158213.doc -86- 201210611 制。 本文所提及之所有文獻均q丨料方式併人本文中。本 申請案中引用之所有公開案及專利文獻出於所有目的以引 用的方式併人本文中,則用的程度就如同如此個別㈣ 不各個別中請案或專利文獻-般。就在本文件令對各種參 考文獻的引用而言’中請人並非承認任何特定參考文獻為 其發,之「先前技術^本發明組合物及方法之實施例在 以下實例中說明。It is a combination of penetration enhancers, such as a fatty acid/salt combined with a bile acid/salt. Particularly preferred combinations are the sodium salts of lauric acid, citric acid and udca. Other anti-reflection enhancers include polyoxyethylene _9_lauryl ether, polyoxyethylene bismuth __: the oligonucleotide of the present invention may comprise spray-dried particles or composites to form micron or nanoparticle particles. Pass the mouth. Oligo(tetra) acid complexes and their use are further described in U.S. Patent No. 6,287,86, the disclosure of which is incorporated herein by reference. Compositions and formulations for parenteral, intrathecal or intraventricular administration may also include buffers, diluents, and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds, and other pharmaceuticals. A sterile aqueous solution of apharmaceutically acceptable carrier or excipient). Certain embodiments of the invention provide pharmaceutical compositions comprising one or more polymeric compounds and one or more other chemotherapeutic agents that act by a non-antisense mechanism. Examples of such chemotherapeutic agents include, but are not limited to, cancer chemotherapeutic drugs such as daun〇rubicin, daunomycin, dactinomyCin, cranberry (d〇) X〇rubicin), epirubicin, idarubicin, esopubicin 158213.doc -83· 201210611 (esorubicin), bleomycin, mafosfamide , ifosfamide, cytosine arabinoside, bischloroethyl-nitrosurea, busulfan, mitomycin C), actinomycin D, mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, Dakaba. Dacarbazine, procarbazine, hexamethylmelamine, pentamethyl melamine, mitoxantrone, amsacrine, chlorambucil , methylcyclohexylnitrosourea, nitrogen mustards, melphalan, cyclophosphamide, 6-drainage, 6-thioguanine, glucosamine, 5-azacytidine, transbasic urea, deoxycoformycin, 4-hydroxyperoxy-phosphoniumamine, 5-fluorouracil (5-FU), 5-fluorodeoxyuridine (5- FUdR), amidoxime (MTX), colchicine, taxol, vincristine, vinblastine, etoposide (VP-16), top three Trimetrexate, irinotecan, topotecan, gemcitabine, teniposide, cisplatin, and diethylstilbestrol (DES). When used with a compound of the invention, the chemotherapeutic agents can be administered individually (e.g., 5-FU and oligonucleotide), sequentially (e.g., 5-FU and oligonucleotides for a period of time, followed by MTX and oligonucleosides). 158213.doc -84- 201210611 acid) or in combination with one or more other such chemotherapeutic agents (eg, 5_FU, Μτχ & nucleotides, or 5-FU, radiation therapy, and oligonucleotides). These include, but are not limited to, non-steroidal anti-inflammatory drugs and anti-inflammatory drugs for corticosteroids and include, but are not limited to, ribivirin, vidarabine, acyclovir, and ganciclovir ( Antiviral agents of gancicl(vir) can also be combined with the compositions of the invention. Combinations of antisense compounds and other non-antisense drugs are also within the scope of the invention. Two or more combinations of the compounds may be used together or sequentially. In another related embodiment, the compositions of the present invention may contain one or more antisense compounds (especially oligonucleotides) that are dry to the first nucleic acid, and one or more other antisense compounds that target the second nucleic acid target. . For example, the first target can be a specific antisense sequence of amidoxime phosphoribosyltransferase (NAMPT) and the first target can be a region from another nucleotide sequence. Alternatively, the compositions of the invention may contain two or more antisense compounds that target different regions of the same nicotine guanamine transphosphoryrosidase (NAMPT) nucleic acid target. Many examples of antisense compounds are described herein, and others may be selected from suitable compounds known in the art. Two or more combinations of the compounds may be used together or sequentially. Administration: The formulation of the Xianxin therapeutic composition and its subsequent administration (administration) are within the abilities of those skilled in the art. Administration depends on the severity and response of the condition to be treated, wherein the course of treatment lasts from several days to several months until the cure is achieved or the disease condition is alleviated. The optimal dosing schedule can be calculated from measurements of drug accumulation in the tract. The average skilled person can easily determine the optimal dose, method of administration and repetition rate. The optimal dose will vary depending on the relative potency of the individual oligonucleotides and can generally be based on an EC50 estimate found to be effective in both in vitro and in vivo animals. In general, the dose is 10 mg per kilogram of body weight, and may be administered once or more per A, weekly, monthly or yearly, or even every 2 to 20 years. As a general practitioner, the repetition rate of administration can be estimated based on the measured residence time and concentration of the drug in the body fluid or tissue. After successful treatment, the patient may need to undergo maintenance therapy to prevent the recurrence of the disease, in which the nucleotides are administered at a maintenance dose ranging from 1 Kg to 10 mg per kilogram of body weight, administered once or more per day. Once every 2 to 2 years. In one embodiment, the patient is treated with a drug at a dose of at least about 1 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 6 mg per kilogram of body weight, At least about 7 mg, at least about 8 mg, at least about 9 mg, at least about 1 mg, at least about 15 mg, at least about 20 mg, at least about 25 mg, at least about 30 mg, at least about 35 mg, at least about 40 mg At least about 45 mg, at least about 5 mg, at least about 6 mg, to >, about 70 mg, at least about 80 mg, at least about 9 mg, or at least about 1 mg. Certain injectable doses of antisense oligonucleotides are described, for example, in U.S. Patent No. 7,563,884, "Antisense modulati 〇n 〇f ρτρΐΒ εχρΓ_〇η", which is incorporated herein by reference in its entirety. While the various embodiments of the invention have been described, it is understood that Many variations of the disclosed embodiments can be made in accordance with the disclosure herein without departing from the spirit and scope of the invention. Therefore, the breadth and scope of the present invention should not be limited to any of the above embodiments. 158213.doc -86 - 201210611. All documents mentioned herein are in the form of data. All publications and patent documents cited in this application are hereby incorporated by reference in their entirety for all purposes in the the the the the the To the extent that this document cites references to various references, the applicant does not admit that any particular reference is made to it. "Previous techniques ^ Embodiments of the compositions and methods of the invention are illustrated in the following examples.

實例 以下非限制性實例用以說明本發明之所選實施例。應瞭 解,組分要素之比例及替代物的變化對熟習此項技術者顯 而易知且在本發明實施例之範疇内。 實例1 :設計對菸鹼醯胺轉磷酸核糖基酶(nampt)之反義 核酸分子及丨聚核苷酸之有義股具特異性之反義 募核普酸 如上所示之術語「對…具特異性之募核苷酸」或「靶 向…之寡核苷酸」係指具有⑴能夠與目標基因之一部分形 成穩定複合物或(丨〇能夠與目標基因的mRNA轉錄物之一部 分形成穩定雙鏈體之序列的寡核苷酸。 藉由使用在各指定序列之19至25個核苷酸之子序列中自 動鑑別的電腦程式(例如IDT AntiSense Design、IDT OligoAnalyzer)促進適當募核苷酸之選擇,該等子序列將 在所需熔融溫度(通常50。(:至60。〇下與目標聚核苷酸序列 形成雜交物’且不會形成自身二聚體或其他複雜二級結 158213.doc -87- 201210611 構。 進一步藉由使用自動比對核酸序列且指示具有一致性或 同源性之區的電腦程式促進適當寡核苷酸之選擇。使用該 等程式例如藉由搜尋諸如GenBank之資料庫或藉由對pCR 產物進行定序來比較所獲得之核酸序列。比較來自多種基 因及指定基因組之基因間區的核酸序列允許選擇與所關注 基因顯示適當程度之特異性的核酸序列。此等程序允許選 擇與目標核酸序列顯示高度互補性及與指定基因組中之其 他核酸序列顯示較低程度之互補性的募核苷酸。熟習此項 技術者將認識到在選擇用於本發明中之基因之適當區方面 存在相當大的自由度。 當反義化合物與目標核酸之結合干擾目標核酸之正常功 能從而導致調節功能及/或活性,且在需要特異性結合之 條件下,亦即在活體内檢定或治療性處理情況下之生理條 件下,及在活體外檢定之情況下執行檢定之條件下,存在 足夠程度之互補性以避免該反義化合物與非目標核酸序列 的非特異性結合時,該化合物為「可特異性雜交」。 本文所述募核苷酸之雜交性質可藉由一或多種此項技術 中已知之活體外檢定來測定。舉例而言,本文所述募核苷 馱之性質可使用熔融曲線檢定藉由測定目標天然反義序列 與潛在藥物分子之間的結合強度來獲得。 目標天然反義序列與潛在藥物分子(分子)之間的結合強 度可使用量測分子間相互作用強度之任何既定方法,例如 炫融曲線檢定來估算。 1582l3.doc -88- 201210611 熔融曲線檢定確定天然反義序列/分子複合物發生由雙 股快速轉變為單股構形之溫度。普遍接受此溫度作為兩個 分子之間的相互作用強度之可靠量度。 可使用實際天然反義RNA分子之cDNA複本或對應於分 子之結合位點的合成DNA或RNA核苷酸執行熔融曲線檢 定。含有執行此檢定之所有必需試劑之多個套組可獲得 (例如Applied Biosystems Inc.,MeltDoctor套組)。此等套 組包括含有一種雙股DNA(dsDNA)結合染料(諸如ABI HRM 染料、SYBR綠、SYTO等)之適合緩衝溶液。dsDNA染料 之性質使得其在呈游離形式時幾乎不發射螢光,但當結合 於dsDNA時發射強烈螢光。 為執行檢定,使cDNA或相應募核苷酸與特定製造商方 案規定濃度之分子混合。加熱混合物至95°C以使所有預先 形成之dsDNA複合物解離,接著緩慢冷卻至室溫或套組製 造商規定之其他較低溫度以使DNA分子黏接。接著緩慢加 熱新形成之複合物至95°C,同時連續收集關於由反應產生 之螢光之量的資料。螢光強度與反應中存在之dsDNA之量 成反比。可使用與套組相容之即時PCR儀器收集資料(例如 ABI’s StepOne Plus 即時 PCR系統或 lightTyper 儀器,Roche Diagnostics,Lewes, UK)。 藉由使用適當軟體(例如lightTyper(Roche)或SDS解離曲 線(ABI))在y軸上相對於溫度(X軸)繪製螢光關於溫度之負 導數(-d(螢光)/dT)的圖來構建熔融峰。分析該資料以鑑別 由dsDNA複合物快速轉變至單股分子之温度。此溫度稱為 158213.doc -89- 201210611EXAMPLES The following non-limiting examples are illustrative of selected embodiments of the invention. It is to be understood that the proportions of the component elements and variations of the alternatives are readily apparent to those skilled in the art and are within the scope of the embodiments of the invention. Example 1: Designing an antisense nucleic acid molecule for nicotine indoleamine phosphoribosyltransferase (nampt) and antisense nucleotide specificity of purine nucleotides. The term "p... "Specific nucleotides" or "targeting oligonucleotides" means having (1) capable of forming a stable complex with a part of a target gene or (a stable part of the mRNA transcript of the target gene) Oligonucleotide of the sequence of the duplex. Promote appropriate nucleotide collection by using a computer program (such as IDT AntiSense Design, IDT OligoAnalyzer) that automatically identifies the subsequence of 19 to 25 nucleotides of each specified sequence. Alternatively, the subsequences will form a hybrid at the desired melting temperature (typically 50% (: to 60. under the squat to the target polynucleotide sequence) and will not form a self-dimer or other complex secondary junction 158213. Doc-87-201210611. Further facilitating the selection of appropriate oligonucleotides by using a computer program that automatically aligns nucleic acid sequences and indicates regions of homology or homology. Such programs are used, for example, by searching such as GenBank The library or the nucleic acid sequences obtained are sequenced by sequencing the pCR product. Comparing the nucleic acid sequences from the various genes and the intergenic regions of the specified genome allows selection of nucleic acid sequences that display an appropriate degree of specificity to the gene of interest. Such procedures allow for the selection of nucleotides that exhibit a high degree of complementarity with the target nucleic acid sequence and a lower degree of complementarity with other nucleic acid sequences in the specified genome. Those skilled in the art will recognize that it is selected for use in the present invention. There is considerable freedom in the proper region of the gene. When the binding of the antisense compound to the target nucleic acid interferes with the normal function of the target nucleic acid, resulting in a regulatory function and/or activity, and in the case where specific binding is required, ie in vivo Under physiological conditions in the case of internal or therapeutic treatment, and under conditions of performing in vitro assays, there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to the non-target nucleic acid sequence. The compound is "specifically hybridizable". The hybridization properties of the nucleotides described herein may be As determined by one or more in vitro assays known in the art. For example, the properties of the nucleoside oxime described herein can be determined using a melting curve assay by determining between a target natural antisense sequence and a potential drug molecule. Binding strength is obtained. The binding strength between the target natural antisense sequence and the potential drug molecule (molecular) can be estimated using any established method for measuring the strength of the intermolecular interaction, such as a saliency curve assay. 1582l3.doc -88- 201210611 The melting curve test determines the temperature at which a natural antisense sequence/molecular complex is rapidly converted from a double strand to a single strand configuration. This temperature is generally accepted as a reliable measure of the strength of the interaction between two molecules. A cDNA replica of a sense RNA molecule or a synthetic DNA or RNA nucleotide corresponding to the binding site of the molecule performs a melting curve assay. Multiple kits containing all the necessary reagents to perform this assay are available (eg, Applied Biosystems Inc., MeltDoctor kit). Such kits include suitable buffer solutions containing a double-stranded DNA (dsDNA) binding dye such as ABI HRM dye, SYBR Green, SYTO, and the like. The nature of the dsDNA dye is such that it emits little fluorescence when in free form, but emits intense fluorescence when bound to dsDNA. To perform the assay, the cDNA or corresponding nucleotides are mixed with molecules of the specified concentration in a particular manufacturer's protocol. The mixture is heated to 95 ° C to dissociate all of the preformed dsDNA complexes, then slowly cooled to room temperature or other lower temperatures specified by the manufacturer to bond the DNA molecules. The newly formed composite was then slowly heated to 95 ° C while continuously collecting data on the amount of fluorescence produced by the reaction. The intensity of the fluorescence is inversely proportional to the amount of dsDNA present in the reaction. Data can be collected using a real-time PCR instrument compatible with the kit (eg ABI’s StepOne Plus Instant PCR System or lightTyper instrument, Roche Diagnostics, Lewes, UK). A plot of fluorescence versus negative derivative of temperature (-d(fluorescence)/dT) on the y-axis versus temperature (X-axis) using a suitable software (eg lightTyper (Roche) or SDS Dissociation Curve (ABI)) To build a melting peak. This data was analyzed to identify the temperature at which the dsDNA complex rapidly transitions to a single molecule. This temperature is called 158213.doc -89- 201210611

Tm且與兩個分子之間相互作用的強度成正比。Tm通常超 過 40°C。 實例2 :調節NAMPT聚核苷酸 如實例1中所述設計實例2中所用之所有反義寡核苷酸。 指示製造商(IDT Inc·,Coralville, IA)製造所設計之硫代磷 酸醋鍵寡核苷酸且提供表1中所示之所設計硫代磷酸酯類 似物。核苷酸之間的星號標記指示存在硫代峨酸酯鍵。實 例2中之實驗所需之募核苷酸可使用任何適當現有技術方 法來合成’例如IDT所用之方法:在諸如5微米受控微孔玻 璃珠(CPG)之固體支撐物上,使用亞磷醯胺酯 (Phosphoramidite)單體(具有受保護基保護之所有活性基團 之正常核普酸,該等保護基例如糖上之三苯甲基、A及c 上之苯曱酿基及G上之N-2-異丁酿基)。保護基防止寡核苷 酸合成期間的不當反應。在合成製程結束時移除保護基。 初始核苷酸經由3,碳連接至固體支撐物,且合成在3,至5,方 向上進行。向生長之寡核苷酸鏈中添加新鹼基以4個步驟 進行:1)使用三氣乙酸自固定核苷酸之5,氧移除保護基; 2)使用四唑使固定核苷酸與序列中之下—核皆酸偶合於一 起;反應經由胺基磷酸四唑酯中間物來進行;3)洗摔未反 應游離核苷酸及反應副產物且將未反應固定募核苦酸加帽 以防止其參與下一輪合成;藉由使用乙酸酐及N•曱基咪唑 對游離5,經基進行乙酿化來實現加帽;4)穩定核普酸之間 的鍵’若欲產生磷酸二酯鍵則使用碘及水,或若需要硫代 填酸酯鍵則使用Beaucage試劑(3H-1,2-笨萁-枝f 1 τ 7T _醇· 3 -明- 158213.doc -90· 201210611 ι,ι-二氧化物)將磷氧化。藉由交替兩種氧化劑,可構築嵌 合主鏈。對於序列中之每一核苷酸重複上文所述4步驟循 環。當合成了完整序列時,自固體支撐物裂解募核苷酸, 且在高溫下使用氫氧化銨脫除保護基。藉由脫鹽洗掉保護 基且將剩餘寡核苷酸凍乾。 用反義募核苷酸處理HepG2細胞 為執行實例2中所設計之實驗,在37°C及5% C02下使來 自ATCC(目錄號HB-8065)之HepG2細胞在生長培養基 (MEM/EBSS(Hyclone 目錄號 SH30024 或 Mediatech 目錄號 MT-10-010-CV)+10°/〇 FBS(Mediatech 目錄號 MT35-011-CV)+ 青黴素(penicillin)/鏈黴素(streptomycin)(Mediatech 目 錄號MT3 0-002-CI))中生長。在實驗前一天,將細胞以每 毫升0.5xlO4個之密度重塗於6孔培養盤中,且在37°C及5% C02下培育隔夜。實驗當天,將6孔培養盤中之培養基更換 為新鮮生長培養基。 用無核糖核酸酶/去氧核糖核酸酶之去離子水將製造商 以凍乾形式運輸之募核苷酸稀釋至濃度為20 μΜ。在室溫 下將2 μΐ此溶液與400 μΐ OptiMEM培養基(Gibco目錄號 31985-070)及4 μΐ脂染胺2000(Lipofectamine 2000)(Invitrogenl 目錄號11668019)—起培育20分鐘,接著逐滴施加於具有 HepG2細胞之6孔培養盤之一個孔中。將包括2 μΐ水而非募 核苷酸溶液之類似混合物用於模擬轉染對照組。在37°C及 5% C02下培育3小時至18小時後,將培養基更換為新鮮生 長培養基。在添加反義寡核苷酸之後48小時,移除培養基 158213.doc -91 · 201210611 且使用來自Promega之SV總RNA分離系統(目錄號Z3 105)或 來自Qiagen之RNeasy總RNA分離套組(目錄號74181)遵循 製造商之說明自細胞萃取RNA。將600 ng萃取之RNA添加 至如製造商方案所述使用來自Thermo Scientific之Verso cDNA套組(目錄號AB1453B)或高容量cDNA反轉錄套組(目 錄號43 688 13)執行之反轉錄反應中。使用來自此反轉錄反 應之cDNA,藉由即時PCR使用ABI Taqman基因表現混合 物(目錄號4369510)及ABI所設計之弓丨子/探針(Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml (NAMPT),Applied Biosystems Inc., Foster City CA)監測基 因表現。使用以下PCR循環:50°C維持2分鐘、95°C維持 10分鐘、40個循環(95°C維持15秒,60°C維持1分鐘),使用 StepOne Plus 即時 PCR機(Applied Biosystems)。基於處理 與模擬轉染樣品之間的18S-校正dCt值的差異計算用反義 寡核苷酸處理後基因表現之變化倍數。 .镑耒,*即時PCR結果顯示,設計為NAMPT反義序列BE778214 之募聚物不使HepG2細胞中之NAMPT含量增加(圖1)。 用反義募核苷酸處理MCF-7細胞 為執行實例2中所設計之實驗,在37°C及5% C02下使來 自ATCC(目錄號HTB -22)之MCF-7細胞在生長培養基 (MEM/EBSS(Hyclone 目錄號 SH30024 或 Mediatech 目錄號 MT-10-010-CV)+10% FBS(Mediatech 目錄號 MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中生 長。在實驗前一天’將細胞以每毫升0·5χ1〇5個之密度重塗 158213.doc 92- 201210611 於6孔培養盤中,且在37°C及5°/。C02下培育隔夜。實驗當 天,將6孔培養盤中之培養基更換為新鮮生長培養基。用 無核糖核酸酶/去氧核糖核酸酶之去離子水將製造商以凍 乾形式運輸之寡核苷酸稀釋至濃度為20 μΜ。在室溫下將2 μΐ此溶液與400 μΐ OptiMEM培養基(Gibco目錄號31985-070)及 4 μΐ脂染胺 200 0(lnvitrogenl 目錄號 11668019)—起培 育20分鐘,接著逐滴施加於具有MCF-7細胞之6孔培養盤 之一個孔中。將包括2 μΐ水而非寡核苷酸溶液之類似混合 物用於模擬轉染對照組。在37°C及5% C02下培育3至18小 時後,將培養基更換為新鮮生長培養基。在添加反義募核 苷酸之後48小時,移除培養基且使用來自Promega之SV總 RNA分離系統(目錄號Z3105)或來自Qiagen之RNeasy總 RNA分離套組(目錄號74181)遵循製造商之說明自細胞萃取 RNA。將600 ng萃取之RNA添加至如製造商方案所述使用 來自 Thermo Scientific 之 Verso cDNA 套組(目錄號 AB1453B)或高容量cDNA反轉錄套組(目錄號43 68813)執行 之反轉錄反應中。使用來自此反轉錄反應之cDNA,藉由 即時PCR使用ABI Taqman基因表現混合物(目錄號4369510) 及 ABI 所設計之引子 / 探針(Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml, AppliedTm is proportional to the strength of the interaction between the two molecules. The Tm usually exceeds 40 °C. Example 2: Modulation of NAMPT Polynucleotides All antisense oligonucleotides used in Example 2 were designed as described in Example 1. The manufacturer (IDT Inc., Coralville, IA) was instructed to manufacture the designed thiophosphate oligo-oligonucleotides and to provide the designed phosphorothioate analogs shown in Table 1. An asterisk mark between the nucleotides indicates the presence of a thiophthalate linkage. The nucleotides required for the experiments in Example 2 can be synthesized using any suitable prior art method, such as the method used for IDT: on a solid support such as 5 micron controlled microporous glass beads (CPG), using phosphorous Phosphoramidite monomer (normal nucleotide acid with all active groups protected by a protecting group such as trityl on sugar, benzoquinone on A and c, and G on G N-2-isobutyl). The protecting group prevents an inappropriate reaction during the synthesis of the oligonucleotide. The protecting group is removed at the end of the synthesis process. The initial nucleotide is attached to the solid support via 3, carbon, and the synthesis is carried out in the 3, to 5, direction. Adding a new base to the growing oligonucleotide strand is carried out in 4 steps: 1) using tri-acetic acid from the fixed nucleotide 5, removing the protecting group by oxygen; 2) using the tetrazole to immobilize the nucleotide Below the sequence - the nuclear acid is coupled together; the reaction is carried out via the aminotetrazolium phosphate intermediate; 3) the unreacted free nucleotides and reaction by-products are washed off and the unreacted fixed nucleotides are capped. In order to prevent its participation in the next round of synthesis; by using acetic anhydride and N-mercaptoimidazole for free 5, the base is brewed to achieve capping; 4) stabilizing the bond between the nucleotides to produce phosphoric acid The ester bond uses iodine and water, or the Beaucage reagent is used if a thiolate bond is required (3H-1, 2-cracked-branched f 1 τ 7T _ alcohol · 3 - Ming - 158213.doc -90· 201210611 I, ι-dioxide) oxidizes phosphorus. The embedded backbone can be constructed by alternating two oxidants. The 4-step cycle described above is repeated for each nucleotide in the sequence. When the complete sequence was synthesized, nucleotides were cleaved from the solid support and the protecting group was removed using ammonium hydroxide at elevated temperature. The protecting group is washed away by desalting and the remaining oligonucleotides are lyophilized. Treatment of HepG2 cells with antisense nucleosides To perform the experiment designed in Example 2, HepG2 cells from ATCC (Catalog No. HB-8065) were grown in growth medium (MEM/EBSS (at MEM/EBSS) at 37 ° C and 5% CO 2 . Hyclone catalog number SH30024 or Mediatech catalog number MT-10-010-CV) +10°/〇FBS (Mediatech catalog number MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number MT3 0 -002-CI)) grows. One day before the experiment, the cells were recoated in a 6-well culture dish at a density of 0.5 x 10 4 ml per ml, and incubated overnight at 37 ° C and 5% CO 2 . On the day of the experiment, the medium in the 6-well plate was changed to fresh growth medium. The nucleotides transported by the manufacturer in lyophilized form were diluted to a concentration of 20 μΜ using deionized water with ribonuclease/deoxyribonuclease. 2 μL of this solution was incubated with 400 μM OptiMEM medium (Gibco Cat. No. 31985-070) and 4 μL Lipofectamine 2000 (Invitrogenl Cat. No. 11668019) for 20 minutes at room temperature, followed by dropwise application. One well of a 6-well culture dish with HepG2 cells. A similar mixture including 2 μΐ water instead of the nucleotide solution was used to simulate the transfection control group. After incubation for 3 hours to 18 hours at 37 ° C and 5% CO 2 , the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium 158213.doc -91 · 201210611 was removed and the SV total RNA isolation system from Promega (catalog number Z3 105) or the RNeasy total RNA isolation kit from Qiagen was used (catalog No. 74181) Extract RNA from cells following the manufacturer's instructions. 600 ng of extracted RNA was added to the reverse transcription reaction performed using the Verso cDNA kit from Thermo Scientific (catalog number AB1453B) or the high capacity cDNA reverse transcription kit (catalog number 43 688 13) as described in the manufacturer's protocol. Using the cDNA from this reverse transcription reaction, the ABI Taqman gene expression mixture (catalog number 4369510) and the ABI-designed bow tweezers/probes were used by real-time PCR (Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml (NAMPT), Applied Biosystems Inc., Foster City CA) monitors gene expression. The following PCR cycles were used: 50 ° C for 2 minutes, 95 ° C for 10 minutes, 40 cycles (95 ° C for 15 seconds, 60 ° C for 1 minute), using a StepOne Plus Instant PCR Machine (Applied Biosystems). The fold change in gene expression after treatment with antisense oligonucleotides was calculated based on the difference in 18S-corrected dCt values between the treated and mock transfected samples. Pound 耒, * Immediate PCR results showed that the polymer designed as the NAMPT antisense sequence BE778214 did not increase the NAMPT content in HepG2 cells (Fig. 1). Treatment of MCF-7 cells with antisense nucleotides To perform the experiment designed in Example 2, MCF-7 cells from ATCC (catalog number HTB-22) were grown in growth medium at 37 ° C and 5% CO 2 ( MEM/EBSS (Hyclone catalog number SH30024 or Mediatech catalog number MT-10-010-CV) +10% FBS (Mediatech catalog number MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI) ) growing in. On the day before the experiment, the cells were recoated at a density of 0·5χ1〇5 per ml 158213.doc 92- 201210611 in a 6-well culture dish at 37 ° C and 5 ° /. Cultivate overnight under C02. On the day of the experiment, the medium in the 6-well culture dish was changed to fresh growth medium. Oligonucleotides transported by the manufacturer in lyophilized form were diluted to a concentration of 20 μΜ using deoxyribonuclease/deoxyribonuclease deionized water. 2 μL of this solution was incubated with 400 μM OptiMEM medium (Gibco Cat. No. 31985-070) and 4 μL serotonin 200 0 (lnvitrogenl Cat. No. 11668019) for 20 minutes at room temperature, followed by dropwise application to MCF- 7 cells in a well of a 6-well culture plate. A similar mixture including 2 μΐ water instead of the oligonucleotide solution was used to simulate the transfection control group. After incubation for 3 to 18 hours at 37 ° C and 5% CO 2 , the medium was changed to fresh growth medium. 48 hours after the addition of antisense nucleotides, the medium was removed and the SV Total RNA Isolation System from Promega (Cat. No. Z3105) or the RNeasy Total RNA Isolation Kit from Qiagen (Cat. No. 74181) followed the manufacturer's instructions. Extract RNA from cells. 600 ng of extracted RNA was added to the reverse transcription reaction performed using the Verso cDNA kit from Thermo Scientific (catalog number AB1453B) or the high capacity cDNA reverse transcription kit (catalog number 43 68813) as described in the manufacturer's protocol. Using the cDNA from this reverse transcription reaction, the ABI Taqman gene expression mixture (catalog number 4369510) and the ABI designed primer/probe were used by real-time PCR (Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml, Applied

Biosystems Inc·,Foster City CA)監測基因表現。使用以下 PCR循環:50°C維持2分鐘、95°C維持10分鐘、40個循環 (9 5 °C維持15秒,60 °C維持1分鐘),使用Step One Plus即時 PCR機(Applied Biosystems)。基於處理與模擬轉染樣品之 158213.doc -93- 201210611 間的18S-校正dCt值的差異計算用反義寡核苷酸處理後基 因表現之變化倍數。 .结耒.*即時PCR結果顯示,設計為NAMPT反義序列 BE778214及skoblyby.aApr07之寡核苷酸不使MCF-4細胞中 之NAMPT含量增加(圖2)。 用反義募核苷酸處理DXJ145細胞 在3 7°C及5% C02下使來自ATCC(目錄號HB-81)之DU145 細胞在生長培養基(MEM/EBSS(Hyclone目錄號SH30024或 Mediatech 目錄號 MT-10-010-CV)+10% FBS(Mediatech 目錄 號MT3 5-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中生長。在實驗前一天,將細胞以每毫升0.5x105 個之密度重塗於6孔培養盤中,且在37°C及5% C02下培 育。實驗當天,將6孔培養盤中之培養基更換為新鮮生長 培養基。將所有反義寡核苷酸稀釋至濃度為2〇 μΜ。在室 溫下將2 μΐ此溶液與400 μΐ Opti-MEM培養基(Gibco目錄號 31985-070)及 4 μΐ脂染胺 2000(Invitrogenl 目錄號 11668019) 一起培育20分鐘’且施加於具有HepG2細胞之6孔培養盤 之一個孔中。將包括2 μΐ水而非寡核苷酸溶液之類似混合 物用於未模擬轉染對照組。在37°C及5% C02下培育3小時 至1 8小時後,將培養基更換為新鮮生長培養基。在添加反 義募核苷酸之後48小時,移除培養基且使用來自Promega 之SV總RNA分離系統(目錄號Z3 105)或來自Qiagen之 RNeasy總RNA分離套組(目錄號74181)遵循製造商之說明 自細胞萃取RNA。將6〇0 ng RNA添加至如製造商方案所述 158213.doc -94- 201210611 使用來自Thermo Scientific之Verso cDNA套組(目錄號 AB1453B)或高容量cDNA反轉錄套組(目錄號4368813)執行 之反轉錄反應中。使用來自此反轉錄反應之cDNA,藉由 即時卩《1使用入81丁&911^11基因表現混合物(目錄號43 69510) 及 ABI 所設計之引子 /探針(Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml, Applied Biosystems Inc., Foster City CA)監測基因表現。使用以下PCR循環: 50°C維持2分鐘、95°C維持10分鐘、40個循環(95°C維持15 秒,60°C維持1分鐘),使用StepOne Plus即時PCR機 (Applied Biosystems)。基於處理與模擬轉染樣品之間的 18S·校正dCt值的差異計算用反義募核苷酸處理後基因表 現之變化倍數。 ,结耒.·即時PCR結果顯示,設計為NAMPT反義序列 skoblyby.aApr07之寡核普酸不使DU145細胞中之NAMPT含 量增加(圖3)。 用反義寡核苷酸處理HEK 293細胞 在37。(:及5% C02下使來自ATCC(目錄號CRL-1573)之 HEK 293細胞在生長培養基(MEM/EBSS(Hyclone目錄號 SH30024 或 Mediatech 目錄號 MT-10-010-CV)+1〇% FBS(Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素 (Mediatech目錄號MT30-002-CI))中生長。在實驗前一天’ 將細胞以每毫升0.5χ105個之密度重塗於6孔培養盤中’且 在37。(:及5% C02下培育。實驗當天,將6孔培養盤中之培 養基更換為新鮮生長培養基。將所有反義寡核苷酸稀釋至 158213.doc -95- 201210611 濃度為20 μΜ。在室溫下將2 μΐ此溶液與400 μΐ Opti-MEM 培養基(Gibco目錄號31985-070)及4 μΐ脂染胺 2000(Invitrogenl目錄號11668019)—起培育20分鐘,且施 加於具有HEK 293細胞之6孔培養盤之各孔中。將包括2 μΐ 水而非寡核苷酸溶液之類似混合物用於未模擬轉染對照 組。在37°C及5% C02下培育3至18小時後,將培養基更換 為新鮮生長培養基。在添加反義募核苷酸之後48小時,移 除培養基且使用來自Promega之SV總RNA分離系統(目錄號 Z3 105)或來自Qiagen之RNeasy總RNA分離套組(目錄號 74181)遵循製造商之說明自細胞萃取RNA。將600 ng RNA 添加至如製造商方案所述使用來自Thermo Scientific之 Verso cDNA套組(目錄號AB1453B)或高容量cDNA反轉錄 套組(目錄號4368813)執行之反轉錄反應中。使用來自此反 轉錄反應之cDNA,藉由即時pcr使用ABI Taqman基因表 現混合物(目錄號4369510)及ABI所設計之引子/探針 (Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml,Applied Biosystems Inc.,Foster City CA) 監測基因表現。使用以下PCR循環:50°C維持2分鐘、 95°C維持10分鐘、40個循環(95°C維持15秒,60°C維持1分 鐘),使用 Mx4000 熱循環儀(stratagene)或 StepOne Plus 即 時PCR機(Applied Bi〇SyStems)。基於處理與模擬轉染樣品 之間的18S-校正dCt值的差異計算用反義寡核苷酸處理後 基因表現之變化倍數。 .结耒··即時PCR結果顯示,用NAMPT反義序列 158213.doc •96- 201210611 skoblyby_aApr〇7之一種反義寡核苷酸處理之後48小時, HEK 293細胞中NAMPT mRNA之含量明顯增加(圖4)。攪 拌之對照寡核苷酸(CUR_1505)未使HEK 293細胞中之 NAMPT含量升高。 儘管本發明已關於一或多個實施例加以說明及描述,但 熟習此項技術者將依據本說明書及隨附圖式之閱讀及瞭解 進行等效變化及修改。另外,儘管本發明之特定特徵可能 僅關於若干實施例中之一者已揭示,但該等特徵可與其他 實施例之一或多個其他特徵組合,此對於任何指定或特定 應用可為所要或有利的。 本發明之摘要將允許讀者快速確定技術揭示内容之性 質。提出條件為其不用以解釋或限制以下申請專利範圍之 範_或含義。 【圖式簡單說明】 圖1為即時PCR結果之圖,其顯示與對照組相比,在 HepG2細胞用使用脂染胺2000引入之硫代磷酸酯募核苷酸 處理後,NAMPT mRNA之變化倍數+標準差。表示為CUR-1724 、 CUR-1725 、 CUR-1726 及 CUR-1727 之條帶分別對應 於用SEQ ID NO·· 17至20處理之樣品。 圖2為即時PCR結果之圖,其顯示與對照組相比,在 MCF-7細胞用使用脂染胺2000引入之硫代磷酸酯募核苷酸 處理後,NAMPT mRNA之變化倍數+標準差。表示為CUR-1728、CUR-1729、CUR-1730、CUR-1731 及 CUR-1732 之 條帶分別對應於用SEQ ID NO: 21至25處理之樣品。 158213.doc -97- 201210611 圖3為即時PCR結果之圖,其顯示與對照組相比,在 DU145細胞用使用脂染胺2000引入之硫代磷酸酯募核苷酸 處理後,NAMPT mRNA之變化倍數+標準差。表示為CUR-1733 、 CUR-1734 、 CUR-1735 及 CUR-1736 之條帶分別對應 於用SEQ ID NO: 26至29處理之樣品。 圖4為即時PCR結果之圖,其顯示與對照組相比,在 HEK 293細胞用使用脂染胺2000引入之硫代磷酸酯募核苷 酸處理後,NAMPT mRNA之變化倍數+標準差。表示為 CUR-173 7及CUR-15 05之條帶分別對應於用SEQIDNO:3 0 至31處理之樣品。Biosystems Inc., Foster City CA) monitors gene expression. The following PCR cycles were used: 2 minutes at 50 °C, 10 minutes at 95 °C, 40 cycles (15 seconds at 95 °C, 1 minute at 60 °C), using the Step One Plus Instant PCR Machine (Applied Biosystems) . The fold change in gene performance after treatment with antisense oligonucleotides was calculated based on the difference in 18S-corrected dCt values between 158213.doc -93 and 201210611 of the treated and mock transfected samples. The results of the real-time PCR showed that the oligonucleotides designed as NAMPT antisense sequences BE778214 and skoblyby.aAp07 did not increase the NAMPT content in MCF-4 cells (Fig. 2). Treatment of DXJ145 cells with antisense nucleotides DU145 cells from ATCC (catalog number HB-81) were grown in growth medium (MEM/EBSS (Hyclone catalog number SH30024 or Mediatech catalog number MT) at 37 ° C and 5% CO 2 -10-010-CV) +10% FBS (Mediatech Cat. No. MT3 5-011-CV) + Penicillin/Streptomycin (Mediatech Cat. No. MT30-002-CI)). One day before the experiment, the cells were recoated in a 6-well culture dish at a density of 0.5 x 105 per ml, and cultured at 37 ° C and 5% CO 2 . On the day of the experiment, the medium in the 6-well culture dish was changed to fresh growth medium. All antisense oligonucleotides were diluted to a concentration of 2 〇 μΜ. 2 μL of this solution was incubated with 400 μΐ Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μL serotonin 2000 (Invitrogenl Cat. No. 11668019) for 20 minutes at room temperature and applied to HepG2 cells. Hole in a hole in the culture plate. A similar mixture comprising 2 μΐ water instead of the oligonucleotide solution was used for the untranslated control group. After incubation for 3 hours to 18 hours at 37 ° C and 5% CO 2 , the medium was changed to fresh growth medium. 48 hours after the addition of antisense nucleotides, the medium was removed and the SV total RNA isolation system from Promega (catalog number Z3 105) or the RNeasy total RNA isolation kit from Qiagen (catalog number 74181) was followed by the manufacturer. Describe the extraction of RNA from cells. Add 6 ng of RNA to 158213.doc -94 - 201210611 as described in the manufacturer's protocol using the Verso cDNA kit from Thermo Scientific (catalog number AB1453B) or the high capacity cDNA reverse transcriptome (catalog number 4368813) In the reverse transcription reaction. The cDNA from this reverse transcription reaction was used, and the Applied Biosystems Taqman Gene Expression Assay was designed by using the primers/probes designed by ABI in the immediate use of the 81 Ding & 911^11 Gene Expression Mixture (Cat. No. 43 69510) and ABI. Hs00237184_ml, Applied Biosystems Inc., Foster City CA) monitors gene expression. The following PCR cycles were used: 50 ° C for 2 minutes, 95 ° C for 10 minutes, 40 cycles (95 ° C for 15 seconds, 60 ° C for 1 minute), using a StepOne Plus real-time PCR machine (Applied Biosystems). The fold change in gene expression after treatment with antisense nucleotides was calculated based on the difference in the 18S·corrected dCt values between the treated and mock transfected samples. The results of real-time PCR showed that the oligonucleotide designed as the NAMPT antisense sequence skoblyby.aApr07 did not increase the NAMPT content in DU145 cells (Fig. 3). HEK 293 cells were treated with antisense oligonucleotides at 37. (: and 5% C02 HEK 293 cells from ATCC (Catalog No. CRL-1573) in growth medium (MEM/EBSS (Hyclone catalog number SH30024 or Mediatech catalog number MT-10-010-CV) + 1% FBS (Mediatech catalog number MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)) was grown. On the day before the experiment', the cells were recoated in 6 wells at a density of 0.5χ105 per ml. Incubate in the dish 'and in 37. (: and 5% C02. On the day of the experiment, replace the medium in the 6-well plate with fresh growth medium. Dilute all antisense oligonucleotides to 158213.doc -95- 201210611 Concentration 20 μΜ. Incubate 2 μL of this solution with 400 μM Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μListamine 2000 (Invitrogen Catalog No. 11668019) for 20 minutes at room temperature, and Apply to each well of a 6-well culture dish with HEK 293 cells. A similar mixture including 2 μΐ water instead of oligonucleotide solution was used in the untranslated control group. Incubate at 37 ° C and 5% CO 2 After 3 to 18 hours, the medium was changed to fresh growth medium. After the addition of antisense nucleotides 48 The medium was removed and the RNA was extracted from the cells using the SV Total RNA Isolation System from Promega (Cat. No. Z3 105) or the RNeasy Total RNA Isolation Kit from Qiagen (Cat. No. 74181) following the manufacturer's instructions. 600 ng RNA Add to the reverse transcription reaction performed using the Verso cDNA kit from Thermo Scientific (catalog number AB1453B) or the high-capacity cDNA reverse transcription kit (catalog number 4368813) as described in the manufacturer's protocol. Use cDNA from this reverse transcription reaction Gene expression was monitored by immediate PCR using the ABI Taqman Gene Expression Mix (Cat. No. 4369510) and ABI designed primer/probe (Applied Biosystems Taqman Gene Expression Assay: Hs00237184_ml, Applied Biosystems Inc., Foster City CA). PCR cycle: 50 ° C for 2 minutes, 95 ° C for 10 minutes, 40 cycles (95 ° C for 15 seconds, 60 ° C for 1 minute), using Mx4000 thermal cycler (stratagene) or StepOne Plus real-time PCR machine (Applied Bi〇SyStems). The fold change in gene expression after treatment with antisense oligonucleotides was calculated based on the difference in 18S-corrected dCt values between the treated and mock transfected samples. The results of real-time PCR showed that the content of NAMPT mRNA in HEK 293 cells was significantly increased 48 hours after treatment with an antisense oligonucleotide of NAMPT antisense sequence 158213.doc •96-201210611 skoblyby_aApr〇7 (Fig. 4). The stirred control oligonucleotide (CUR_1505) did not increase the NAMPT content in HEK 293 cells. While the invention has been described and described with respect to the embodiments the embodiments In addition, although specific features of the invention may be disclosed in relation to only one of several embodiments, such features may be combined with one or more other features of other embodiments, which may be desired for any specified or particular application. advantageous. The Abstract of the Invention will allow the reader to quickly ascertain the nature of the technical disclosure. The conditions are set forth as not to explain or limit the scope or meaning of the following claims. BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 is a diagram showing the results of real-time PCR, which shows the fold change of NAMPT mRNA after treatment of HepG2 cells with a phosphorothioate-introduced nucleotide introduced by lipofectamine 2000 as compared with a control group. + standard deviation. The bands indicated as CUR-1724, CUR-1725, CUR-1726 and CUR-1727 correspond to the samples treated with SEQ ID NO.. 17 to 20, respectively. Figure 2 is a graph of the results of real-time PCR showing the fold change + standard deviation of NAMPT mRNA after MCF-7 cells were treated with phosphorothioate-introduced nucleotides introduced with lipofectamine 2000 as compared to the control group. The bands denoted CUR-1728, CUR-1729, CUR-1730, CUR-1731 and CUR-1732 correspond to the samples treated with SEQ ID NOS: 21 to 25, respectively. 158213.doc -97- 201210611 Figure 3 is a graph of the results of real-time PCR showing changes in NAMPT mRNA after treatment of DU145 cells with phosphorothioate nucleotides introduced with lipofectamine 2000 compared to the control group. Multiple + standard deviation. The bands denoted CUR-1733, CUR-1734, CUR-1735 and CUR-1736 correspond to the samples treated with SEQ ID NOS: 26 to 29, respectively. Figure 4 is a graph showing the results of real-time PCR showing the fold change + standard deviation of NAMPT mRNA after HEK 293 cells were treated with phosphorothioate-inducing nucleotides introduced with lipofectamine 2000 as compared with the control group. The bands denoted as CUR-173 7 and CUR-15 05 correspond to the samples treated with SEQ ID NO: 3 0 to 31, respectively.

序列表說明:SEQ ID NO: 1 :智人菸鹼醯胺轉磷酸核糖 基酶(NAMPT)轉錄物變異體3 mRNA(NCBI寄存編號: NM_005746) ; SEQ ID NO: 2 :天然 NAMPT 反義序列 (BE778214) ; SEQ ID NO: 3 ·•天然 NAMPT 反義序列 (skoblyby.aApr07) ; SEQ ID NO: 4 :天然 NAMPT反義序列 (sernorby.aAuglO-未剪接);SEQ ID NO: 5 :天然NAMPT 反義序列(wyflubu.aAugl 0-未剪接);SEQ ID NO: 6 :天然 NAMPT 反義序列(swysmorbu.aAuglO-未剪接);SEQ ID NO: 7 :天然 NAMPT反義序列(zuflubu.aAuglO-未剪接); SEQ ID NO: 8:天然NAMPT反義序列(ployblyby.aAuglO-未剪接);SEQ ID NO: 9 :天然NAMPT反義序列 (skablyby.aAuglO-未剪接);SEQ ID NO: 10 :天然NAMPT 反義序列(skublyby.aAug 1 0-未剪接);SEQ ID NO: 11 :天 然NAMPT反義序列(swarsmorbu.aAuglO-未剪接);SEQ ID 158213.doc -98- 201210611 NO: 12:天然 NAMPT反義序列(CD639886); SEQ ID NO: 13 :天然 NAMPT反義序列(BM451023) ; SEQ ID NO: 14 : 天然 NAMPT反義序列(R22881) ; SEQ ID NO: 15 :天然 NAMPT反義序列(DW442985) ; SEQ ID NO: 16 :天然 NAMPT反義序列(BM717984) ; SEQ ID NO: 17 至 31 :反義 寡核普酸。*指示硫代碟酸醋鍵;SEQ ID NO: 3 1 :對照寡 核苷酸(CUR-1505)。BRIEF DESCRIPTION OF THE SEQUENCE LISTING: SEQ ID NO: 1 : Homo serotonin guanamine transphosphatase (NAMPT) transcript variant 3 mRNA (NCBI accession number: NM_005746); SEQ ID NO: 2: natural NAMPT antisense sequence ( SEQ ID NO: 3 • Natural NAMPT antisense sequence (skoblyby.aApr07); SEQ ID NO: 4: native NAMPT antisense sequence (sernorby.aAuglO-unspliced); SEQ ID NO: 5: natural NAMPT anti Sequence (wyflubu.aAugl 0-unspliced); SEQ ID NO: 6: native NAMPT antisense sequence (swysmorbu.aAuglO-unspliced); SEQ ID NO: 7: native NAMPT antisense sequence (zuflubu.aAuglO-unspliced) SEQ ID NO: 8: native NAMPT antisense sequence (ployblyby.aAug10-unspliced); SEQ ID NO: 9: native NAMPT antisense sequence (skablyby.aAug10-unspliced); SEQ ID NO: 10: native NAMPT Antisense sequence (skublyby.aAug 1 0-unspliced); SEQ ID NO: 11: natural NAMPT antisense sequence (swarsmorbu.aAuglO-unspliced); SEQ ID 158213.doc -98- 201210611 NO: 12: natural NAMPT anti- Sequence (CD639886); SEQ ID NO: 13: natural NAMPT antisense sequence (BM451023); SEQ ID NO: 14: natural NAMPT counter Sequence (R22881); SEQ ID NO: 15: natural NAMPT antisense sequence (DW442985); SEQ ID NO: 16: natural NAMPT antisense sequence (BM717984); SEQ ID NO: 17 to 31: an antisense oligonucleotide. * indicates thioate vinegar linkage; SEQ ID NO: 3 1 : control oligonucleotide (CUR-1505).

158213.doc -99- 201210611 〇158213.doc -99- 201210611 〇

序列表 <110> 美商歐科可娜有限貴任公司Sequence Listing <110> US Occidental Limited Company

<120> 藉由抑制於鹼醯胺轉鱗酸核糖基酶(NAMPT)之天然反義轉錄物治療NAMPT 相關疾病 <130> NAMPT <140> 100129854 <141> 2011-08-19 <150〉61/375,126 <151> 2010-08-19 <160> 31 <170> patentin version 3.5 <210> 1 <211> 4593 <212> DNA <213>智人 <400> 1 gctgccgcgc cccgcccttt ctcggccccc ggagggtgac ggggtgaagg cgggggaacc gaggtgggga gtccgccaga gctcccagac tgcgagcacg cgagccgccg cagccgtcac ccgcgccgcg tcacggctcc cgggcccgcc ctcctctgac ccctcccctc tctccgtttc cccctctccc cctcctccgc cgaccgagca gtgacttaag caacggagcg cggtgaagct catttttctc cttcctcgca gccgcgccag ggagctcgcg gcgcgcggcc cctgtcctcc ggcccgagat gaatcctgcg gcagaagccg agttcaacat cctcctggcc accgactcct acaaggttac tcactataaa caatatccac ccaacacaag caaagtttat tcctactttg aatgccgtga aaagaagaca gaaaactcca aattaaggaa ggtgaaatat gaggaaacag tattttatgg gttgcagtac attcttaata agtacttaaa aggtaaagta gtaaccaaag agaaaatcca ggaagccaaa gatgtctaca aagaacattt ccaagatgat gtctttaatg aaaagggatg gaactacatt cttgagaagt atgatgggca tcttccaata gaaataaaag ctgttcctga gggctttgtc attcccagag gaaatgttct cttcacggtg gaaaacacag atccagagtg ttactggctt acaaattgga ttgagactat tcttgttcag tcctggtatc caatcacagt ggccacaaat tctagagagc agaagaaaat attggccaaa tatttgttag aaacttctgg taacttagat ggtctggaat acaagttaca tgattttggc tacagaggag tctcttccca agagactgct ggcataggag catctgctca cttggttaac ttcaaaggaa cagatacagt agcaggactt gctctaatta aaaaatatta tggaacgaaa gatcctgttc caggctattc tgttccagca gcagaacaca gtaccataac agcttggggg aaagaccatg aaaaagatgc ttttgaacat attgtaacac agttttcatc agtgcctgta tctgtggtca gcgatagcta tgacatttat aatgcgtgtg agaaaatatg gggtgaagat ctaagacatt 158213-序列表,doc 60 120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 201210611 taatagtatc aagaagtaca caggcaccac taataatcag acctgattct ggaaaccctc 1260 ttgacactgt gttaaaggtt ttggagattt taggtaagaa gtttcctgtt actgagaact 1320 caaagggtta caagttgctg ccaccttatc ttagagttat tcaaggggat ggagtagata 1380 ttaatacctt acaagagatt gtagaaggca tgaaacaaaa aatgtggagt attgaaaata 1440 ttgccttcgg ttctggtgga ggtttgctac agaagttgac aagagatctc ttgaattgtt 1500 ccttcaagtg tagctatgtt gtaactaatg gccttgggat taacgtcttc aaggacccag 1560 ttgctgatcc caacaaaagg tccaaaaagg gccgattatc tttacatagg acgccagcag 1620 ggaattttgt tacactggag gaaggaaaag gagaccttga ggaatatggt caggatcttc 1680 tccatactgt cttcaagaat ggcaaggtga caaaaagcta ttcatttgat gaaataagaa 1740 aaaatgcaca gctgaatatt gaactggaag cagcacatca ttaggcttta tgactgggtg 1800 tgtgttgtgt gtatgtaata cataatgttt attgtacaga tgtgtggggt ttgtgtttta 1860 tgatacatta cagccaaatt atttgttggt ttatggacat actgcccttt catttttttt 1920 cttttccagt gtttaggtga tctcaaatta ggaaatgcat ttaaccatgt aaaagatgag 1980 tgctaaagta agctttttag ggccctttgc caataggtag tcattcaatc tggtattgat 2040 cttttcacaa ataacagaac tgagaaactt ttatatataa ctgatgatca cataaaacag 2100 atttgcataa aattaccatg attgctttat gtttatattt aacttgtatt tttgtacaaa 2160 caagattgtg taagatatat ttgaagtttc agtgatttaa cagtctttcc aacttttcat 2220 gatttttatg agcacagact ttcaagaaaa tacttgaaaa taaattacat tgccttttgt 2280 ccattaatca gcaaataaaa catggcctta acaaagttgt ttgtgttatt gtacaatttg 2340 aaaattatgt cgggacatac cctatagaat tactaacctt actgcccctt gtagaatatg 2400 tattaatcat tctacattaa agaaaataat ggttcttact ggaatgtcta ggcactgtac 2460 agttattata tatcttggtt gttgtattgt accagtgaaa tgccaaattt gaaaggcctg 2520 tactgcaatt ttatatgtca gagattgcct gtggctctaa tatgcacctc aagattttaa 2580 ggagataatg tttttagaga gaatttctgc ttccactata gaatatatac ataaatgtaa 2640 aatacttaca aaagtggaag tagtgtattt taaagtaatt acacttctga atttattttt 2700 catattctat agttggtatg acttaaatga attactggag tgggtagtga gtgtacttaa 2760 atgtttcaat tctgttatat tttttattaa gtttttaaaa aattaaattg gatattaaat 2820 tgtatggaca tcatttatta attttaaact gaatgccctc aataagtaat actgaagcac 2880 attcttaaat gaagataaat tatctccaat gaaaagcatg acatgtgttt caatagaaga 2940 atcttaagtt ggctaaattc aaagtgcttg acatcaaaat gttctagagt gattagctac 3000 tagattctga atcatacatc acatctgact agagaccagt ttctttcgaa tgattctttt 3060 atgtatgtag atctgttctt ctgaggcagc ggttggccaa ctatagccca aaggccaaat 3120 -2- 158213-序列表.doc 201210611<120> Treatment of NAMPT-associated diseases by inhibition of natural antisense transcripts of glutamine transluciferase (NAMPT) <130> NAMPT <140> 100129854 <141> 2011-08-19 < 150>61/375,126 <151> 2010-08-19 <160> 31 <170> patentin version 3.5 <210> 1 <211> 4593 <212> DNA <213> Homo sapiens< 400 > 1 gctgccgcgc cccgcccttt ctcggccccc ggagggtgac ggggtgaagg cgggggaacc gaggtgggga gtccgccaga gctcccagac tgcgagcacg cgagccgccg cagccgtcac ccgcgccgcg tcacggctcc cgggcccgcc ctcctctgac ccctcccctc tctccgtttc cccctctccc cctcctccgc cgaccgagca gtgacttaag caacggagcg cggtgaagct catttttctc cttcctcgca gccgcgccag ggagctcgcg gcgcgcggcc cctgtcctcc ggcccgagat gaatcctgcg gcagaagccg agttcaacat cctcctggcc accgactcct acaaggttac tcactataaa caatatccac ccaacacaag caaagtttat tcctactttg aatgccgtga aaagaagaca gaaaactcca aattaaggaa ggtgaaatat Gaggaaacag tattttatgg gttgcagtac attcttaata agtacttaaa aggtaaagta gtaaccaaag agaaaatcca ggaagccaaa gatgtctaca aagaacattt ccaagatgat gtct ttaatg aaaagggatg gaactacatt cttgagaagt atgatgggca tcttccaata gaaataaaag ctgttcctga gggctttgtc attcccagag gaaatgttct cttcacggtg gaaaacacag atccagagtg ttactggctt acaaattgga ttgagactat tcttgttcag tcctggtatc caatcacagt ggccacaaat tctagagagc agaagaaaat attggccaaa tatttgttag aaacttctgg taacttagat ggtctggaat acaagttaca tgattttggc tacagaggag tctcttccca agagactgct ggcataggag catctgctca cttggttaac ttcaaaggaa cagatacagt agcaggactt gctctaatta aaaaatatta tggaacgaaa gatcctgttc caggctattc tgttccagca gcagaacaca gtaccataac agcttggggg aaagaccatg aaaaagatgc ttttgaacat attgtaacac agttttcatc agtgcctgta tctgtggtca gcgatagcta tgacatttat aatgcgtgtg agaaaatatg gggtgaagat ctaagacatt 158213- sequence Listing, doc 60 120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 201210611 taatagtatc aagaagtaca caggcaccac taataatcag acctgattct ggaaaccctc 1260 ttgacactgt gttaaaggtt ttggagattt taggtaagaa gtttcctgtt Actgagaact 1320 caaagggtta caagttgctg ccaccttatc ttagagttat tcaaggggat ggagtagata 1380 ttaata cctt acaagagatt gtagaaggca tgaaacaaaa aatgtggagt attgaaaata 1440 ttgccttcgg ttctggtgga ggtttgctac agaagttgac aagagatctc ttgaattgtt 1500 ccttcaagtg tagctatgtt gtaactaatg gccttgggat taacgtcttc aaggacccag 1560 ttgctgatcc caacaaaagg tccaaaaagg gccgattatc tttacatagg acgccagcag 1620 ggaattttgt tacactggag gaaggaaaag gagaccttga ggaatatggt caggatcttc 1680 tccatactgt cttcaagaat ggcaaggtga caaaaagcta ttcatttgat gaaataagaa 1740 aaaatgcaca gctgaatatt gaactggaag cagcacatca ttaggcttta tgactgggtg 1800 tgtgttgtgt gtatgtaata cataatgttt attgtacaga tgtgtggggt ttgtgtttta 1860 tgatacatta cagccaaatt atttgttggt ttatggacat actgcccttt catttttttt 1920 cttttccagt gtttaggtga tctcaaatta ggaaatgcat ttaaccatgt aaaagatgag 1980 tgctaaagta agctttttag ggccctttgc caataggtag tcattcaatc tggtattgat 2040 cttttcacaa ataacagaac tgagaaactt ttatatataa ctgatgatca cataaaacag 2100 atttgcataa aattaccatg attgctttat gtttatattt aacttgtatt tttgtacaaa 2160 caagattgtg taagatatat ttgaagtttc agtgatttaa cagtctttcc aacttttcat 2220 gatttttatg a gcacagact ttcaagaaaa tacttgaaaa taaattacat 2280 ccattaatca gcaaataaaa catggcctta acaaagttgt ttgtgttatt gtacaatttg 2340 aaaattatgt cgggacatac cctatagaat tactaacctt actgcccctt gtagaatatg 2400 tattaatcat tctacattaa agaaaataat ggttcttact ggaatgtcta ggcactgtac 2460 agttattata tatcttggtt gttgtattgt accagtgaaa tgccaaattt gaaaggcctg 2520 tactgcaatt ttatatgtca gagattgcct gtggctctaa tatgcacctc aagattttaa 2580 ggagataatg tttttagaga gaatttctgc ttccactata gaatatatac ataaatgtaa 2640 aatacttaca aaagtggaag tgccttttgt tagtgtattt taaagtaatt acacttctga atttattttt 2700 catattctat agttggtatg acttaaatga attactggag tgggtagtga gtgtacttaa 2760 atgtttcaat tctgttatat tttttattaa gtttttaaaa aattaaattg gatattaaat 2820 tgtatggaca tcatttatta attttaaact gaatgccctc aataagtaat actgaagcac 2880 attcttaaat gaagataaat tatctccaat gaaaagcatg acatgtgttt caatagaaga 2940 atcttaagtt ggctaaattc aaagtgcttg acatcaaaat gttctagagt gattagctac 3000 tagattctga atcatacatc acatctgact agagaccagt ttctttcgaa tgattctttt 3060 atgtatgtag atctgtt Ctt ctgaggcagc ggttggccaa ctatagccca aaggccaaat 3120 -2- 158213-sequence table.doc 201210611

ttggacttct ttttataaat gcagattgtc tatggctgct ttcccactac tccagcctaa 3180 ggtaaacagc tgcaatagaa gccaaatgag aatcgcaaag cccaaaatgt ttattaacct 3240 gccctttaca caaaattaca caaaaagttt cctgatctct gttctaagaa aaggagtgtg 3BOO ccttgcattt aaaaggaaat gttggtttct agggaaggga ggaggctaaa taattgatac 3360 ggaattttcc tcttttgtct tcttttttct cacttaagaa tccgatactg gaagactgat 3420 ttagaaaagt ttttaacatg acattaaatg tgaaatttta aaaattgaaa agccataaat 3480 catctgtttt aaatagttac atgagaaaat gatcactaga ataacctaat tagaagtgtt 3540 atcttcatta aatgtttttt gtaagtggta ttagaaagaa tatgtttttc agatggttct 3600 ttaaacatgt agtgagaaca ataagcatta ttcactttta gtaagtcttc tgtaatccat 3660 gatataaaat aattttaaaa tgatttttta atgtatttga gtaaagatga gtagtattaa 3720 gaaaaacaca catttcttca caaaatgtgc taaggggcgt gtaaagaatc aaaagaaact 3780 attaccaata atagttttga taatcaccca taattttgtg tttaaacatt gaaattatag 3840 tacagacagt attctctgtg ttctgtgaat ttcagcagct tcagaataga gtttaattta 3900 gaaatttgca gtgaaaaaag ctatctcttt gttcacaacc ataaatcagg agatggagat 3960 taattctatt ggctcttagt cacttggaac tgattaattc tgactttctg tcactaagca 4020 cttggtattt ggccatctcc attctgagca ccaaacggtt aacacgaatg tccactagaa 4080 ctctgctgtg tgtcaccctt aaatcagtct aaatcttcca gacaaaagca aatggcattt 4140 atggatttaa gtcattagat tttcaactga cattaattaa tccctcttga ttgattatat 4200 catcaagtat ttatatctta aataggaggt aggatttctg tgttaagact cttatttgta 4260 ccctataatt aaagtaaaat gttttttatg agtatccctt gttttccctt cttaaattgt 4320 tatcaaacaa tttttataat gaaatctatc ttggaaaatt agaaagaaaa atggcaaggt 4380 atttattgtt ctgtttgcca taatttagaa ctcacactta agtattttgt agttttacat 4440 tcctttttaa cccattcagt ggagaatgtc agcttttctc ccaagttgta tgttaagtct 4500 attctaatat gtactcaaca tcaagttata aacatgtaat aaacatggaa ataaagttta 4560 gctctattag tgaagtgtta aaaaaaaaaa aaa 4593 <210> 2 <211> 882 <212> DNA <213>智人 <400> 2 gaaaattatt gtaaacgcat ttaaacaaat tattatttct attttgagac ggagtctcgc 60 tctgtcgccc aggctggagt gcagtggctg gatctcggct cactgcaagc tccgcctccc 120 tggttcacgc catcctcctg cctcacacct gtaatcccag cactttggga ggctgaggcg 180 158213·序列表.doc 201210611 ggctgatcac ttgagcccag gattttgaga ccggcctggg caacatggtg aagacccatc 240 tctgcaaaaa atacagaagt tggctggacg tggtggcatg tgcctgtagt cccagcttct BOO ctgaaaggag actgacgtgg gaggattgcc tgagcccggg gaggttgtag catgtagagg 360 acctgagaaa ctgtttttca taggtcacaa caacatgaca aataataaca catagactca 420 aatgggcccc ctcacttgat tcaaagccct ggattattat accatcctac caaatagcaa 480 tcaggcactg ccaattgggc atgcagctcc ttgtctccct ccctcagagt gctatgaaag 540 acctagggct catatatttg ttcaggcaaa gtgagtgtat ctgtgccact cctgttgcca 600 cacacccttg cagacagcct cgggtgcacc tttatatttc aaagttggct cagatactcc 660 aatctgacct gatttgactc agttaaaaaa cactgctgaa gtaaggtatg gaactcacct 720 actgggaacc ctgaagtggg cttacatttc ctttatattc tctagatcgc aatacaacgt 780 gtccaagcca tgtgaaaagg agtttaaatt tactggcttt tttatcaact gcagcttgga 840 acccaagtta tccgggaagg caccaaaagg gtccagtcca ga 882 <210> 3 <211> 915 <212> DNA <213> 智人 <400> 3 ccgcctggga ccttccgtcc tacccagtcc tggccggttt tctgggtcct cctgaagtca 60 cgccacccgg ctagggggcg aggagcctcc tactgcccat cttcccgtcc accacgcgca 120 gttactcacc tttgtctccg gcctggatta aggatccagc ctttcgcctc catccctctt 180 gtccctcctg atcctcctga ccctgtcttt aagatcccag gagctgcggt gaggagtgag 240 gctgaggggc ccctttcatc tgatgcagcg actccgcttt cctccggcgg ctctgtctat 300 ggctgagctc tttgatcctt tgagagatgg tttgactttt cccgagcaaa gagcctgcgt 360 tgaaaagcgg gggtggaatt cagtcctcac agataatgag gggacaagac ctaattgaac 420 cgagtattgc cgggaaggaa aaggcaacgg gccaagcctt tgacagggtg cgacactgac 480 ttttatcatc gttatagtct ttaaatcctg ggaaacgagt tggcaacccc aaaataaaga 540 agtgtaatga cgtctgatga cttcacccaa atacagacca ttccaagaaa gacttgcgca 600 gttctcatgc gtggttgcgt ttttgcataa aactaagatt ccctttgtcc gcatgtttaa 660 tagcttaaaa ataaattgag gtttttatcg gataaaagta gccaggaaaa agtgatacag 720 tgaaaacgat tacaggatat ttatagactt gtctttcaag ttattagaag tctcattcta 780 tctgggggca gtgatggtgg tggtggtagt ggaacttgtg aattgagatt catagtggaa 840 cttgtgaatt gagattcatc tcgaaactgg aggcatggct gagacttcta ataaagacaa 900 cctcagtcaa cacta 915 4- 158213·序列表.doc 201210611 <210> 4 <211> 165 ' <212> DNA <213>智人 <400> 4 catgtacagt tttgggctga ttctcaacta tacagatgca catacaggct gggaatggtg 60 gctcacacct gtaatctcag cactttagga ggcctgagtc aaggagttcg aggccaaaca 120 gttgagactg tttcccagac tgggaaacag tgagaccccc atctc 165 <210> 5 <211> 533 <212> DNA <213> 智人 <400> 5 ctgtgtatac acgtaagtgc taaagtgcct gaaaggcacc tgggaacagt acttaactga 60 caccaaggta agagactgtc agttcagctg agctaagggg aaaacggcaa ttaggtgtgt 120 cagctgagat tgctatgtgt aaggtcctcc cccttctccc tgcagacttt tcttctattt 180 cttttttcct ttccacatga aagcccctat gtcccaacac caaatgtctt agctgaagga 240 attcccactt ctcttaagca aggcagtttg gaagaggtag tggaagaaac ctcatctctg 300 ttattacaag agatctattc cggacgcctc atattggaac tctaaataca ccatcaccgc 360 agaaatactg tttgatttgg aagtgagaag tattatgtac aacaatcttt ggaaaactac 420 tataatttgg gtactgacct tactctatct ccaccaaact ctttcgaaca gaaaaaaact 480 tgtgtttcat agctgtagat accatctgag attcgtatac atcaaaatgt cag 533ttggacttct ttttataaat gcagattgtc tatggctgct ttcccactac tccagcctaa 3180 ggtaaacagc tgcaatagaa gccaaatgag aatcgcaaag cccaaaatgt ttattaacct 3240 gccctttaca caaaattaca caaaaagttt cctgatctct gttctaagaa aaggagtgtg 3BOO ccttgcattt aaaaggaaat gttggtttct agggaaggga ggaggctaaa taattgatac 3360 ggaattttcc tcttttgtct tcttttttct cacttaagaa tccgatactg gaagactgat 3420 ttagaaaagt ttttaacatg acattaaatg tgaaatttta aaaattgaaa agccataaat 3480 catctgtttt aaatagttac atgagaaaat gatcactaga ataacctaat tagaagtgtt 3540 atcttcatta aatgtttttt gtaagtggta ttagaaagaa tatgtttttc agatggttct 3600 ttaaacatgt agtgagaaca ataagcatta ttcactttta gtaagtcttc tgtaatccat 3660 gatataaaat aattttaaaa tgatttttta atgtatttga gtaaagatga gtagtattaa 3720 gaaaaacaca catttcttca caaaatgtgc taaggggcgt gtaaagaatc aaaagaaact 3780 attaccaata atagttttga taatcaccca taattttgtg tttaaacatt gaaattatag 3840 tacagacagt attctctgtg ttctgtgaat ttcagcagct tcagaataga gtttaattta 3900 gaaatttgca gtgaaaaaag ctatctcttt gttcacaacc ataaatcagg agatggagat 3960 taattc tatt ggctcttagt cacttggaac tgattaattc tgactttctg tcactaagca 4020 cttggtattt ggccatctcc attctgagca ccaaacggtt aacacgaatg tccactagaa 4080 ctctgctgtg tgtcaccctt aaatcagtct aaatcttcca gacaaaagca aatggcattt 4140 atggatttaa gtcattagat tttcaactga cattaattaa tccctcttga ttgattatat 4200 catcaagtat ttatatctta aataggaggt aggatttctg tgttaagact cttatttgta 4260 ccctataatt aaagtaaaat gttttttatg agtatccctt gttttccctt cttaaattgt 4320 tatcaaacaa tttttataat gaaatctatc ttggaaaatt agaaagaaaa atggcaaggt 4380 atttattgtt ctgtttgcca taatttagaa ctcacactta agtattttgt agttttacat 4440 tcctttttaa cccattcagt ggagaatgtc agcttttctc ccaagttgta tgttaagtct 4500 attctaatat gtactcaaca tcaagttata aacatgtaat aaacatggaa ataaagttta 4560 gctctattag tgaagtgtta aaaaaaaaaa aaa 4593 < 210 > 2 < 211 > 882 < 212 > DNA < 213 > Homo sapiens < 400 > 2 gaaaattatt gtaaacgcat ttaaacaaat tattatttct attttgagac ggagtctcgc 60 tctgtcgccc aggctggagt gcagtggctg gatctcggct cactgcaagc tccgcctccc 120 tggttcacgc catcctcctg cctcacacct gtaa tcccag cactttggga ggctgaggcg 180 158213 · Sequence Listing .doc 201210611 ggctgatcac ttgagcccag gattttgaga ccggcctggg caacatggtg aagacccatc 240 tctgcaaaaa atacagaagt tggctggacg tggtggcatg tgcctgtagt cccagcttct BOO ctgaaaggag actgacgtgg gaggattgcc tgagcccggg gaggttgtag catgtagagg 360 acctgagaaa ctgtttttca taggtcacaa caacatgaca aataataaca catagactca 420 aatgggcccc ctcacttgat tcaaagccct ggattattat accatcctac caaatagcaa 480 tcaggcactg ccaattgggc atgcagctcc ttgtctccct ccctcagagt gctatgaaag 540 acctagggct catatatttg ttcaggcaaa gtgagtgtat ctgtgccact cctgttgcca 600 cacacccttg cagacagcct cgggtgcacc tttatatttc aaagttggct cagatactcc 660 aatctgacct gatttgactc agttaaaaaa cactgctgaa gtaaggtatg gaactcacct 720 actgggaacc ctgaagtggg cttacatttc ctttatattc tctagatcgc aatacaacgt 780 gtccaagcca tgtgaaaagg agtttaaatt tactggcttt tttatcaact gcagcttgga 840 acccaagtta tccgggaagg caccaaaagg gtccagtcca ga 882 < 210 > 3 <211> 915 <212> DNA <213> Homo sapiens <400> 3 ccgcctggga ccttccgtcc tacccagtcc tg gccggttt tctgggtcct cctgaagtca 60 cgccacccgg ctagggggcg aggagcctcc tactgcccat cttcccgtcc accacgcgca 120 gttactcacc tttgtctccg gcctggatta aggatccagc ctttcgcctc catccctctt 180 gtccctcctg atcctcctga ccctgtcttt aagatcccag gagctgcggt gaggagtgag 240 gctgaggggc ccctttcatc tgatgcagcg actccgcttt cctccggcgg ctctgtctat 300 ggctgagctc tttgatcctt tgagagatgg tttgactttt cccgagcaaa gagcctgcgt 360 tgaaaagcgg gggtggaatt cagtcctcac agataatgag gggacaagac ctaattgaac 420 cgagtattgc cgggaaggaa aaggcaacgg gccaagcctt tgacagggtg cgacactgac 480 ttttatcatc gttatagtct ttaaatcctg ggaaacgagt tggcaacccc aaaataaaga 540 agtgtaatga cgtctgatga cttcacccaa atacagacca ttccaagaaa gacttgcgca 600 gttctcatgc gtggttgcgt ttttgcataa aactaagatt ccctttgtcc gcatgtttaa 660 tagcttaaaa ataaattgag gtttttatcg gataaaagta gccaggaaaa agtgatacag 720 tgaaaacgat tacaggatat ttatagactt gtctttcaag ttattagaag tctcattcta 780 tctgggggca gtgatggtgg tggtggtagt ggaacttgtg aattgagatt catagtggaa 840 cttgtgaatt gagattcatc tcgaaactgg aggcatggct gagacttcta a Taaagacaa 900 cctcagtcaa cacta 915 4- 158213 · Sequence Listing.doc 201210611 <210> 4 <211> 165 ' <212> DNA <213> Homo sapiens <400> 4 catgtacagt tttgggctga ttctcaacta tacagatgca catacaggct gggaatggtg 60 gctcacacct gtaatctcag cactttagga ggcctgagtc aaggagttcg aggccaaaca 120 gttgagactg tttcccagac tgggaaacag tgagaccccc atctc 165 < 210 > 5 < 211 > 533 < 212 > DNA < 213 > Homo sapiens < 400 > 5 ctgtgtatac acgtaagtgc taaagtgcct gaaaggcacc tgggaacagt acttaactga 60 caccaaggta agagactgtc agttcagctg agctaagggg aaaacggcaa ttaggtgtgt 120 cagctgagat tgctatgtgt aaggtcctcc cccttctccc tgcagacttt tcttctattt 180 cttttttcct ttccacatga aagcccctat gtcccaacac caaatgtctt agctgaagga 240 attcccactt ctcttaagca aggcagtttg gaagaggtag tggaagaaac ctcatctctg 300 ttattacaag agatctattc cggacgcctc atattggaac tctaaataca ccatcaccgc 360 agaaatactg tttgatttgg aagtgagaag tattatgtac aacaatcttt ggaaaactac 420 tataatttgg gtactgacct tactctatct ccaccaaact ctttcgaaca gaaaaaaact 480 tgtgtttcat agctgtag At accatctgag attcgtatac atcaaaatgt cag 533

<210> 6 <211> 522 <212> DNA <213>智人 <400> 6 aaagatttat ctgtagcttg cgatgctgtt tggtacgata gcgttcaccc acacaagaac 60 ttctttcaaa attgggagtg catcctctca aaccctgctt tggttttatc aactgatttt 120 actgatattc gaagtccttt gttgtcattt caacgatgtt cacagcatct ttgccagcag 180 tagatttcat ctcaagaaac cactttctgt atccacccat atgaagcaaa tcctcatctg 240 ttcaagttta atcatgagac tgcagcaatt cagtcagatc ttcaggctcc accacttcta 300 gttatctagc tatttctacc acatttgtag ttacttcctc cactgaagtc ttgaagtcaa 360 atcaagactc agttatctgt gaaggttgga atcaatttct tccaaactcc tgttaatgct 420 gacattttaa cgtcctccca tgaagcatga tgttcttaaa tggcatctag aatggtgaac 480 cctttcagct acagacttaa gaatatattt cttctttttt tt 522 158213-序列表.doc 201210611 <210> 7 <211> 141 <212> DNA <213> 智人 <400> 7 agagagagaa gcagctgcct aatgggcaca cacaacagaa aggaatgaac tgaaagacag 60 cagaatttgg tactgctaat atctcaatgt ggtatgttcc aaaagtcaat gacttaacat 120 gactacttta cttgtacaat g 141 <210> 8 <211> 519 <212> DNA <213> 智人 <400> 8 tgaacataaa tcatttttcc tgtttagctt tgatctcaag ttcaggaaag ccaatgctgt 60 ctttgggtac acagtctcaa tctcagtaaa acttggctct atcctcaaga atcttcatag 120 cttagtacaa gaatcctaag atgaatatat ttccattgat gattctgaat gaatgtgctg 180 tttccttgcc aatggtagca tacttgcaac tctataaaaa tagcccctaa tgcagcacac 240 agacaacctt ggaggtaaat ctttatagta acaagtgatc tgagagttta agtcaagcag 300 gagacttgga gctgttttcc ttaagtacaa cagatagtga tatactaaaa ggtacaggga 360 agactcatca aaagctagaa gtatatgtca gggcttatta aggttagaat ccagggactg 420 atcccacttt aataaaattt aagaagtagc taagcaagat agctaataca gaatctgcag 480 caatgaaatt tatgtcaatt cactaaaatg caaaaacaa 519 <210> 9 <211> 374 <212> DNA <213> 智人 <4〇〇> 9 caatgtaaat aactccaact tctttagtgg gactaagtag tgagatagaa gaaaagcaca 60 cacgatgaag taaaacttga aaaatggtgt ggatcttata gcacaagttt gtaagtctga 120 ttcatagttt tatttctcca cagtttgacc cttactgttc aggtacttaa tgatgcattc 180 tttatcagat ttaaaattac tagaaggcaa gttattttct ttttctgaga agacgaagag 240 tcaaaagcaa aatggaaatg tctctaagct atttaactct gtatcttggt ctaaaagtat 300 actgaaaatg ttctttgtac aaatacagct ccagttctgg acaataatta aatatttata 360 attttaaagc ttca 374 <210> 10 <211> 240 6- 158213-序列表.doc 201210611 <212> DNA <213> 智人 <400> 10 ttttttgaaa aaaagctgcc ttgaattcct aatcaccata tataaatttc acagctcttt 60 gaaacttcta taaaatcaac ttctgtccct cctggatttc aaaatactac ttatctttgt 120 cttcttttat aatacccaat aggtcaccaa catttattaa atgaatggtt aaaaatatgc 180 tgaattctag ataaccagct tacagatgac cctttgagaa gcaacctaca ccggacagca 240 <210> 11 <211> 353 <212> DNA <213> 智人 <400> 11 tgtgaactca gacatgatcg tgggagttgc ttcggccatg aaatgtgtac cgagatggtg 60≪ 210 > 6 < 211 > 522 < 212 > DNA < 213 > Homo sapiens < 400 > 6 aaagatttat ctgtagcttg cgatgctgtt tggtacgata gcgttcaccc acacaagaac 60 ttctttcaaa attgggagtg catcctctca aaccctgctt tggttttatc aactgatttt 120 actgatattc gaagtccttt gttgtcattt caacgatgtt cacagcatct ttgccagcag 180 tagatttcat ctcaagaaac cactttctgt atccacccat atgaagcaaa tcctcatctg 240 ttcaagttta atcatgagac tgcagcaatt cagtcagatc ttcaggctcc accacttcta 300 gttatctagc tatttctacc acatttgtag ttacttcctc cactgaagtc ttgaagtcaa 360 atcaagactc agttatctgt gaaggttgga atcaatttct tccaaactcc tgttaatgct 420 gacattttaa cgtcctccca tgaagcatga tgttcttaaa tggcatctag aatggtgaac 480 cctttcagct acagacttaa gaatatattt cttctttttt tt 522 158213- sequence Listing .doc 201210611 < 210 > 7 <211> 141 <212> DNA <213> Homo sapiens <400> 7 agagagagaa gcagctgcct aatgggcaca cacaacagaa aggaatgaac tgaaagacag 60 cagaatttgg tactgctaat atctcaatgt ggtatgttcc aaaagtcaat gacttaacat 120 gactacttta cttgtacaat g 141 <210> 8 <211> 519 <;2 12 > DNA < 213 > Homo sapiens < 400 > 8 tgaacataaa tcatttttcc tgtttagctt tgatctcaag ttcaggaaag ccaatgctgt 60 ctttgggtac acagtctcaa tctcagtaaa acttggctct atcctcaaga atcttcatag 120 cttagtacaa gaatcctaag atgaatatat ttccattgat gattctgaat gaatgtgctg 180 tttccttgcc aatggtagca tacttgcaac tctataaaaa tagcccctaa tgcagcacac 240 agacaacctt ggaggtaaat ctttatagta acaagtgatc tgagagttta agtcaagcag 300 gagacttgga gctgttttcc ttaagtacaa cagatagtga tatactaaaa ggtacaggga 360 agactcatca aaagctagaa gtatatgtca gggcttatta aggttagaat ccagggactg 420 atcccacttt aataaaattt aagaagtagc taagcaagat agctaataca gaatctgcag 480 caatgaaatt tatgtcaatt cactaaaatg caaaaacaa 519 < 210 > 9 < 211 > 374 < 212 > DNA < 213 > Homo sapiens < 4〇 〇> 9 caatgtaaat aactccaact tctttagtgg gactaagtag tgagatagaa gaaaagcaca 60 cacgatgaag taaaacttga aaaatggtgt ggatcttata gcacaagttt gtaagtctga 120 ttcatagttt tatttctcca cagtttgacc cttactgttc aggtacttaa tgatgcattc 180 tttatcagat ttaaaattac tagaaggcaa gttattttct ttttctgaga aga Cgaagag 240 tcaaaagcaa aatggaaatg tctctaagct atttaactct gtatcttggt ctaaaagtat 300 actgaaaatg ttctttgtac aaatacagct ccagttctgg acaataatta aatatttata 360 attttaaagc ttca 374 <210> 10 <211> 240 6- 158213 - Sequence Listing.doc 201210611 <212> DNA <213> Homo sapiens <; 400 > 10 ttttttgaaa aaaagctgcc ttgaattcct aatcaccata tataaatttc acagctcttt 60 gaaacttcta taaaatcaac ttctgtccct cctggatttc aaaatactac ttatctttgt 120 cttcttttat aatacccaat aggtcaccaa catttattaa atgaatggtt aaaaatatgc 180 tgaattctag ataaccagct tacagatgac cctttgagaa gcaacctaca ccggacagca 240 < 210 > 11 < 211 > 353 < 212 > DNA < 213 & gt Homo sapiens <400> 11 tgtgaactca gacatgatcg tgggagttgc ttcggccatg aaatgtgtac cgagatggtg 60

tgtttccctt cccagcagca gctttaagag cttgtgcatg gtttggcatt ctctcttttc 120 cttctgccac cgagactgca gtattccaga tatgagccaa tcctctcact taaatcctga 180 aatgtggaca acatggaatc aagtcagagc caactggttc cggacacggg tatgaaagag 240 aaataaatct tttctgttgt aagccacaga gattgtggga ttgttgccac agtatgacct 300 ggttaacctt gacaggtaca gggctccaca cacactgttt tctttcttct cct 353 <210> 12 <211> 534 <212> DNA <213>智人 <220> <221> mi sc一f eatu re <222> (519)..(519) <223> n為a、c、g或t <220> <221> misc一feature <222> (524)..(524) <223> n為a、c、g或t <220> <221> misc_feature <222> (528)..(528) <223> n為a、c、g或t <220> <221> misc.feature <222> (531)..(532) <223> n為a、c、g或t <220> <221> mi sc一feature <222> (534)_·(534〕 <223> n為a、c、g或t 158213-序列表.doc 201210611 <400> 12 gggggctagg gggcgaggag cctcctactg cccatcttcc cgtccaccac gcgcagttac 60 tcacctttgt ctccggcctg gattaaggat ccagcctttc gcctccatcc ctcttgtccc 120 tcctgatcct cctgaccctg tctttaagat cccaggagct gcggtgagga gtgaggctga 180 ggggcccctt tcatctgatg cagcgactcc gctttcctcc ggcggctctg tctatggctg 240 agctctttga tcctttgaga gatggtttga cttttcccga gcaaagagcc tgcgttgaaa 300 agcgggggtg gaattcagtc ctcacagata atgaggggac aagacctaat tgaaccgagt 360 attgccggga aggaaaaggc aacgggccaa gcctttgaca gggtgcgaaa ctgactttta 420 tcatcgttat agtctttaaa tcctgggaaa cgagttggca accccaaaat aaagaagtgt 480 aatgacgtct gatgacttca cccaaataca gaccattcnc aganaganga nnan 534 <210> 13 <211> 959 <212> DNA <213> 智人 <220> <221> <222> <22B> <220> <221> <222> <223> <220> <221> <222> <223> <220> <221> <222> <223> <220> <221> <222> <223> ·>>>> 0123 2222 2222 <220> <221> <222> <223> <220> <221> misc一feature (579)..(582) n為a、c、g或t misc_feature (584)..(584) n為a、c、g或t mi sc一feature (588)..(588) n為a、c、g或t misc_feature (591)..(592) n為a、c、g或t misc—feature (607)..(607) n為a ' c ' g或 t misc_feature (621)·,(623) n為a、c、g或t misc 一feature C6S0) ·. (650) n為a、c、g或t mis cofeature 158213-序列表.doc 201210611 <222> (653).-(653) <223> n為a、c、g或t <220> <2 21> mi s c一 f eat u re <222> (690).-(691) <223> 喊台、c、g或t <220> <221> mi sc一feature <222> (750)..(750) <223> n為a、c、g或t <400> 13tgtttccctt cccagcagca gctttaagag cttgtgcatg gtttggcatt ctctcttttc 120 cttctgccac cgagactgca gtattccaga tatgagccaa tcctctcact taaatcctga 180 aatgtggaca acatggaatc aagtcagagc caactggttc cggacacggg tatgaaagag 240 aaataaatct tttctgttgt aagccacaga gattgtggga ttgttgccac agtatgacct 300 ggttaacctt gacaggtaca gggctccaca cacactgttt tctttcttct cct 353 < 210 > 12 < 211 > 534 < 212 > DNA <213> Homo sapiens <220><221> mi sc-f eatu re <222> (519)..(519) <223> n is a, c, g or t <220><;221> misc-feature <222> (524)..(524) <223> n is a, c, g or t <220><221> misc_feature <222> (528)..( 528) <223> n is a, c, g or t <220><221> misc.feature <222> (531)..(532) <223> n is a, c, g or t <220><221> mi sc a feature < 222 > (534) _ (534) < 223 > n is a, c, g or t 158213 - Sequence Listing. doc 201210611 <400> Gggggctagg gggcgaggag cctcctactg cccatcttcc cgtccaccac gcgcagttac 60 tcac ctttgt ctccggcctg gattaaggat ccagcctttc gcctccatcc ctcttgtccc 120 tcctgatcct cctgaccctg tctttaagat cccaggagct gcggtgagga gtgaggctga 180 ggggcccctt tcatctgatg cagcgactcc gctttcctcc ggcggctctg tctatggctg 240 agctctttga tcctttgaga gatggtttga cttttcccga gcaaagagcc tgcgttgaaa 300 agcgggggtg gaattcagtc ctcacagata atgaggggac aagacctaat tgaaccgagt 360 attgccggga aggaaaaggc aacgggccaa gcctttgaca gggtgcgaaa ctgactttta 420 tcatcgttat agtctttaaa tcctgggaaa cgagttggca accccaaaat aaagaagtgt 480 aatgacgtct Gatgacttca cccaaataca gaccattcnc aganaganga nnan 534 <210> 13 <211> 959 <212> DNA <213> Homo sapiens <220><221><222><22B><220><221>;<222><223><220><221><222><223><220><221><222><223><220><221><;222><223>·>>>> 0123 2222 2222 <220><221><222><223><220><221> misc-feature (579). .(582) n is a, c g or t misc_feature (584)..(584) n is a, c, g or t mi sc-feature (588).. (588) n is a, c, g or t misc_feature (591).. (592 n is a, c, g or t misc—feature (607)..(607) n is a ' c ' g or t misc_feature (621)·, (623) n is a, c, g or t misc Feature C6S0) · (650) n is a, c, g or t mis cofeature 158213 - Sequence Listing.doc 201210611 <222> (653).-(653) <223> n is a, c, g or t <220><221> mi sc-f eat u re <222> (690).-(691) <223> shouting, c, g or t <220><221> mi Sc a feature <222> (750)..(750) <223> n is a, c, g or t <400> 13

tttcactttt cccgagcaaa gagcctgcgt tgaaaagcgg gggtggaatt cagtcttcac 60 agataatgag gggacaagac ctaattgaac cgagtattgc cgggaaggaa aaggcaacgg 120 gccaagcctt tgacagggtg cgaaactgac ttttatcatc gttatagtct ttaaatcctg 180 ggaaacgagt tggcaacccc aaaataaaga agtgtaatga cgtctgatga cttcacccaa 之40 atacagacca ttccaagaaa gacttgcgca gttctcatgc gtggttgcgt ttttgcataa 300 aactaagatt ccctttgtcc gcatgtttaa tagcttaaaa ataaattgag gtttttatcg 360 gataaaagta gccaggaaaa agtgatacag tgaaaacgat tacaggatat ttatagactt 420 gtctttcaag ttattagaag tctcattcta tctgggggca gtgatggtgg tggtggtagt 480 ggaacttgtg aattgagatt catagtggaa cttgtgaatt gagattcatc tcgaaactgg 540 aggcatggct gagacttcta ataaagacaa cctcagtcnn nnanaaanaa nnaaaaaaa:a 600 aaaaaanaaa aaaaaaaaaa nnnagaaaaa aaaaaaaaaa accccccaan ccncccaaaa 660 acccaccaac ccccctaacc accacccacn ncccacccca cccaaaccca aaaccccccc 720 ccaccaaacc caaccccccc ccccaccacn aaaaaacaca ccgccggggc cgccccccct 780 aaaaaaatcc cccccggggg gggccccaac ttttcccggc cccccttttt ttttttgtga 840 acaacacgcg cccccctaag gtcggtgcgc cctcaaaaaa caacaacccc gccccgcccc 900 ccccttttat tttaaaaccc ccccgcttta tgagaaaata atccttcctc tggggcgga 959 <210> 14 <211> 127 <212> <213> DNA 智人 <220> <221> nvi sc一feature <222> (104),,(104) <Z23> n為a、c、g或t <220> <221> rnisc_feature <222> (120)_·(120) <223> n為a、c ' g或t -9- 158213-序列表.doc 201210611 <400> 14 ggaggcagcg gggcagcccc ggcgcgtgac ccgggcgctt acctaagttc gagttcccgg 60 cacgcgacgg gagggcgggg ctggagggag cgttcccagc tttnccagtg ccacgaggan 120 ccggttc 127 <210> 15 <211> 232 <212> DNA <213>智人 <400> 15 ggccattacg gccggggggg accttccgct cctacccact ccgtgcccgg gtctctgggt 60 cctcctgcaa ctcgcgccac ccggcctatg gggcgaggag cctcctactg cccatcttcc 120 cgtccaccac gcgcagttac tcacctttgt ctccggcctg tattgcggat ccagcctttc 180 gcctccttcc ctcttgtccc tcctgatcct cctgaccctg tctttaagat cc 232 <210> 16 <211> 387 <212> DNA <213>智人 <400> 16 ggcactcaga ctggtgaact ttactatttc tttttcttac ctttccctaa cctatatgct 60 gttttcacat cctccatata agactggttt tcttgaggaa gctgaacatg aaaggctata 120 tgtctgattc catttatttg aaattctgga aagggcaaag ttataatgaa agagagcaga 180 tcagtggttg tcagagactg ggattatgag aagcagaatg actgcaagca ggtctgagga 240 aacttcttgg ggtgatgaga atgatatatg tggttatacg actgtataca tttgtcaaaa 300 ttcactgaat tctaagctta aagttggtga atagcatata aattagatct taataaaact 360 gaactaaaaa aaaaaaaaaa aaaggac 387 <210> 17 <211> 20 <212> DNA <213> 人工序列 <220> <223>反義寡核苷酸 <400> 17 atcctcccac gtcagtctcc 20 <210> 18 <211> 21 <212> DNA <213>人工序列 <220> <223>反義募核苷酸 158213-序列表.doc -10- 201210611 <400> 18 gcccatttga gtctatgtgt t <210> 19 <211> 20 <212> DNA <213>人工序列 <220> <223>反義募核苷酸 <400> 19 ggctgtctgc aagggtgtgt <210> 20 <211> 21 <212> DNA <213>人工序列tttcactttt cccgagcaaa gagcctgcgt tgaaaagcgg gggtggaatt cagtcttcac 60 agataatgag gggacaagac ctaattgaac cgagtattgc cgggaaggaa aaggcaacgg 120 gccaagcctt tgacagggtg cgaaactgac ttttatcatc gttatagtct ttaaatcctg 180 ggaaacgagt tggcaacccc 40 atacagacca aaaataaaga agtgtaatga cgtctgatga cttcacccaa of ttccaagaaa gacttgcgca gttctcatgc gtggttgcgt ttttgcataa 300 aactaagatt ccctttgtcc gcatgtttaa tagcttaaaa ataaattgag gtttttatcg 360 gataaaagta gccaggaaaa agtgatacag tgaaaacgat tacaggatat ttatagactt 420 gtctttcaag ttattagaag tctcattcta tctgggggca gtgatggtgg tggtggtagt 480 ggaacttgtg aattgagatt catagtggaa cttgtgaatt gagattcatc tcgaaactgg 540 aggcatggct gagacttcta ataaagacaa cctcagtcnn nnanaaanaa nnaaaaaaa: a 600 aaaaaanaaa aaaaaaaaaa nnnagaaaaa aaaaaaaaaa accccccaan ccncccaaaa 660 acccaccaac ccccctaacc accacccacn ncccacccca cccaaaccca aaaccccccc 720 ccaccaaacc caaccccccc ccccaccacn aaaaaacaca ccgccggggc cgccccccct 780 aaaaaaatcc cccccggggg gggccccaac ttttcccggc cccccttttt ttttttgtga 840 acaacacgcg ccccc Ctaag gtcggtgcgc cctcaaaaaa caacaacccc gccccgcccc 900 ccccttttat tttaaaaccc ccccgcttta tgagaaaata atccttcctc tggggcgga 959 <210> 14 <211> 127 <212><213> DNA Homo sapiens <220><221> nvi sc-feature <222> (104), (104) <Z23> n is a, c, g or t < 220 < 221 > rnisc_feature < 222 > (120) _ (120) < 223 > n is a, c ' g or t -9- 158213 - Sequence Listing.doc 201210611 <400> 14 ggaggcagcg gggcagcccc ggcgcgtgac ccgggcgct acctaagttc gagttcccgg 60 cacgcgacgg gagggcgggg ctggagggag cgttcccagc tttnccagtg ccacgaggan 120 ccggttc 127 <210> 15 <211> 232 <212> DNA < 213 > Homo sapiens < 400 > 15 ggccattacg gccggggggg accttccgct cctacccact ccgtgcccgg gtctctgggt 60 cctcctgcaa ctcgcgccac ccggcctatg gggcgaggag cctcctactg cccatcttcc 120 cgtccaccac gcgcagttac tcacctttgt ctccggcctg tattgcggat ccagcctttc 180 gcctccttcc ctcttgtccc tcctgatcct cctgaccctg tctttaagat cc 232 < 210 > 16 < 211 > 387 <212> DNA <213> Human < 400 > 16 ggcactcaga ctggtgaact ttactatttc tttttcttac ctttccctaa cctatatgct 60 gttttcacat cctccatata agactggttt tcttgaggaa gctgaacatg aaaggctata 120 tgtctgattc catttatttg aaattctgga aagggcaaag ttataatgaa agagagcaga 180 tcagtggttg tcagagactg ggattatgag aagcagaatg actgcaagca ggtctgagga 240 aacttcttgg ggtgatgaga atgatatatg tggttatacg actgtataca tttgtcaaaa 300 ttcactgaat tctaagctta aagttggtga atagcatata aattagatct taataaaact 360 gaactaaaaa aaaaaaaaaa Aaaggac 387 <210> 17 <211> 20 <212> DNA <213> Artificial sequence <220><223> antisense oligonucleotide <400> 17 atcctcccac gtcagtctcc 20 <210><211> 21 <212> DNA <213>Artificial sequence <220><223> Antisense nucleotide 158213 - Sequence Listing. doc -10- 201210611 <400> 18 gcccatttga gtctatgtgt t <210> 19 <211> 20 <212> DNA <213>Artificial sequence<220><223> Antisense nucleotides <400> 19 ggctgtctgc aagggtgtgt <210> 20 <211><212> DNA <213> artificial sequence

<220> <223>反義寡核苷酸 <400> 20 gttccatacc tacttcacca g <210> 21 <211> 21 <212> DNA <213>人工序列 <220> <223>反義寡核苷酸 <400> 21 gccctaggtc tttcatagca c <210> 22 <211> 21 <212> DNA <213>人工岸列 <220> <223>反義寡核苷酸 <400> 22 ctctacatgc tacaacctcc c <210> 23 <211> 20 <212> DNA <213>人工序列 <220> <223>反義寡核苷酸 <400> 23 catgccacca cgtccagcta 158213-序列表.doc 21201210611 <210> 24 <211> 21 <212> DNA <213> 人工序列 <220> <22B> 反義寡核苷酸 <400> 24 tccactacca ccaccaccat c <210> 25 <211> 20 <212> DNA <213> 人工序列 <220> <223> 反義寡核苷酸 <400> 25 gtctcagcca tgcctccagt <210> 26 <211> 21 <212> DNA <213> 人工序列 <220> <223> 反義寡核苷酸 <400> 26 aggagggaca agagggatgg a 20 21 <210> 27 <211> 20 <212> DNA <213>人工序列 <220> <223>反義寡核苷酸 <400> 27 agatgggcag taggaggctc 20 <210> 28 <211> 20 <212> DNA <213>人工序列 <220> <223>反義寡核苷酸 <400> 28 gtggcgtgac ttcaggagga 20 <210> 29 -12- 158213·序列表.doc 201210611 <211> 21 <212> DNA <213>人工序列 <220> <223>反義募核苷酸 <400> 29 gttgccaact cgtttcccag g <210> 30 <211> 20 <212> DNA <213>人工序列 <220><220><223>Antisense oligonucleotide <400> 20 gttccatacc tacttcacca g <210> 21 <211> 21 <212> DNA <213> Artificial sequence <220><223> Antisense oligonucleotide <400> 21 gccctaggtc tttcatagca c <210> 22 <211> 21 <212> DNA <213> artificial shore column <220><223> antisense oligonucleoside Acid <400> 22 ctctacatgc tacaacctcc c <210> 23 <211> 20 <212> DNA <213>Artificial sequence <220><223> Antisense oligonucleotide <400> 23 catgccacca Cgtccagcta 158213 - Sequence Listing. doc 21201210611 <210> 24 <211> 21 <212> DNA <213> Artificial Sequence <220><22B> Antisense Oligonucleotide <400> 24 tccactacca ccaccaccat c <210> 25 <211> 20 <212> DNA <213> Artificial sequence <220><223> Antisense oligonucleotide <400> 25 gtctcagcca tgcctccagt <210> 26 <211> 21 <212> DNA <213> artificial sequence <220><223> antisense oligonucleoside Acid <400> 26 aggagggaca agagggatgg a 20 21 <210> 27 <211> 20 <212> DNA <213> artificial sequence <220><223> antisense oligonucleotide <400> 27 agatgggcag taggaggctc 20 <210> 28 <211> 20 <212> DNA <213> artificial sequence <220><223> antisense oligonucleotide <400> 28 gtggcgtgac ttcaggagga 20 <210&gt 29 -12- 158213 · Sequence Listing.doc 201210611 <211> 21 <212> DNA <213>Artificial Sequence<220><223> Antisense Nucleotide<400> 29 gttgccaact cgtttcccag g <210> 30 <211> 20 <212> DNA <213> Artificial Sequence <220>

<223>反義寡核苷酸 <400> 30 cagggtcagg aggatcagga <210> 31 <211> 21 <212> DNA <213>人工序列 <220> <223>對照序列 <400> 31 cctctccacg cgcagtacat t<223> Antisense Oligonucleotide <400> 30 cagggtcagg aggatcagga <210> 31 <211> 21 <212> DNA <213>Artificial Sequence <220><223> Control Sequence<400> 31 cctctccacg cgcagtacat t

158213-序列表.doc158213 - Sequence Listing. doc

Claims (1)

201210611 七、申請專利範圍: 1. 一種活體外調節生物系統中於驗醯胺轉碟酸核糖基酶 (NAMPT)聚核苷酸之功能及/或表現之方法,該方法包 含:使該系統與至少一種長度為5至30個核苷酸之反義 寡核苷酸接觸,其中該至少一種寡核苷酸與菸鹼醯胺轉 ' 磷酸核糖基酶(NAMPT)聚核苷酸之天然反義序列之反向 互補序列具有至少50%序列一致性;由此調節該菸鹼醯 胺轉磷酸核糖基酶(NAMPT)聚核苷酸之功能及/或表現。 ¢) 2.如請求項1之調節生物系統中菸鹼醯胺轉磷酸核糖基酶 (NAMPT)聚核苷酸之功能及/或表現之方法,該方法包 括:使該生物系統與至少一種長度為5至30個核苷酸之 反義寡核普酸接觸,其中該至少一種募核苷酸與包含 SEQ ID NO: 2之天然反義轉錄物核苷酸1至882、或SEQ ID NO: 3之 1 至 915、或 SEQ ID NO: 4之 1 至 165、或 SEQ ID NO: 5之 1 至 533、或 SEQ ID NO: 6之 1 至 522、或 SEQ ID NO: 7之 1 至 141、或 SEQ ID NO: 8之 1 至 519、或 SEO 〇 ID NO: 9之 1 至 374、或 SEQ ID NO: 10之 1 至 240、或 SEQ ID NO: 11之 1 至 353、或 SEQ ID NO: 12之 1 至 534、或 SEQ ID NO: 13 之 1 至 959、或 SEQ ID NO: 14之 1 至 127、 或 SEQ ID NO: 15之 1 至 232、或 SEQ ID NO: 16之 1 至 387 内5至30個連續核苷酸之聚核苷酸的反向互補序列具有 至少50%序列一致性;由此調節該於驗酿胺轉鱗酸核糖 基酶(NAMPT)聚核苷酸之功能及/或表現。 3. —種活體外調節患者細胞或組織中於驗醯胺轉磷酸核糖 158213.doc 201210611 基酶(NAMPT)聚核苷酸之功能及/或表現之方法,該方法 包含:使該等細胞或組織與至少—種長度為5至3〇個核 苷酸之反義寡核苷酸接觸,其中該募核苷酸與該菸鹼醯 胺轉磷酸核糖基酶(NAMPT)聚核苷酸之反義寡核普酸具 有至少50%序列一致性;由此活體外調節患者細胞或組 織中该於驗酿胺轉麟酸核糖基酶(nampt)聚核苦酸之功 能及/或表現。 4. 如請求項3之調節患者細胞或組織中菸鹼醯胺轉磷酸核 糖基酶(NAMPT)聚核苷酸之功能及/或表現之方法,該方 法包含:使該細胞或組織與至少一種長度為5至30個核 苷酸之反義泰核苷酸接觸,其中該至少一種寡核苷酸與 包含SEQ ID NO: 2之天然反義轉錄物核苷酸1至882、或 SEQ ID NO: 3 之 1 至 915、或 SEQ ID NO: 4之 1 至 165、或 SEQ ID NO: 5之 1 至 533、或 SEQ ID NO: 6之 1 至 522、或 SEQ ID NO: 7之 1 至 141、或 SEQ ID NO: 8之 1 至 519、或 SEQ ID NO: 9之 1 至 374、或 SEQ ID NO: 10之 1 至 240、或 SEQ ID NO: 11之 1至 353、或 SEQ ID NO: 12之 1至 534、 或 SEQ ID NO: 13 之 1 至 959、或 SEQ ID NO: 14之 1 至 127、或 SEQ ID NO: 15之 1 至 232、或 SEQ ID NO: 16之 1 至3 87内5至30個連續核苷酸之聚核苷酸的反向互補序列 具有至少50%序列一致性;由此調節該於驗醒胺轉碟酸 核糖基酶(NAMPT)聚核苷酸之功能及/或表現。 5. —種活體外調節生物系統中菸鹼醯胺轉磷酸核糖基酶 (NAMPT)聚核苷酸之功能及/或表現之方法,該方法包 158213.doc 201210611 含.使該系統與靶向該菸鹼醯胺轉磷酸核糖基酶 (NAMPT)聚核苷酸之天然反義寡核苷酸之區的至少一種 反義券核苷酸接觸,由此調節該於驗酿胺轉構酸核糖基 酶(NAMPT)聚核苷酸之功能及/或表現。 6. 如相求項5之方法,其中該菸鹼醯胺轉磷酸核糖基酶 (NAMPT)之功能及/或表現在活體外相對於對照組增加。 7. 如請求項5之方法,其中該至少一種反義募核苷酸靶向 菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之天然反義 D 序列。 8. 如請求項5之方法,其中該至少一種反義募核苷酸靶向 包含菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之編碼 及/或非編碼核酸序列之核酸序列。 9. 如咐求項5之方法,其中該至少一種反義寡核苷酸靶向 菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之重疊及/或 不重叠序列。 〇 1〇·如請求項5之方法,其中該至少一種反義寡核苷酸包含 一或多個選自以下之修飾:至少一個經修飾糖部分'至 J個經修飾核苷間鍵聯、至少一個經修飾核苷酸及其 組合。 11·如請求項10之方法,其中該一或多個修飾包含至少一個 選自以下之經修飾糖部分:2,_〇_曱氧基乙基修飾糖部 71 2曱氧基修飾糖部分、2'-0-燒基修飾糖部分、雙環 糖部分及其組合。 12.如吻求項1〇之方法,其中該一或多個修飾包含至少一個 158213.doc 201210611 選自以下之經修飾核苷間鍵聯:硫代磷酸酯、2,_〇甲氧 基乙基(MOE)、2 -氟、烷基膦酸酯、二硫代磷酸酯、烷 基硫代膦酸酯、磷醯胺酯(ph〇sph〇ramidate)、胺基甲酸 酯、碳酸酯、磷酸三酯、乙醯胺酯(acetamidate)、羧曱 基酯及其組合。 13.如請求項1〇之方法,其中該一或多個修飾包含至少一個 選自以下之經修飾核苷酸:肽核酸(pNA)、鎖核酸 (LNA)、阿糠核酸(FANA)、其類似物、衍生物及組合。 14·如請求項1之方法,其中該至少一種寡核苷酸包含至少 一種如SEQ ID NO: Π至31所示之寡核苷酸序列。 15. —種活體外調節哺乳動物細胞或組織中菸鹼醢胺轉碟酸 核糖基酶(NAMPT)基因之功能及/或表現之方法,該方法 包含:使該等細胞或組織與至少一種長度為5至3〇個核 苷酸之短干擾RNA(siRNA)寡核苷酸接觸,該至少一種 siRNA募核苷酸對菸鹼醯胺轉磷酸核糖基酶(ΝΑΜρτ)聚 核苷酸之反義聚核苷酸具特異性,其中該至少一種 siRNA募核苷酸與該菸鹼醯胺轉磷酸核糖基酶(ΝΑΜρτ) 聚核苷酸之反義及/或有義核酸分子之至少約5個連續核 酸之互補序列具有至少50%序列一致性;及活體外調節 哺乳動物細胞或組織中菸鹼醯胺轉磷酸核糖基酶 (NAMPT)之功能及/或表現。 16·如請求項15之方法,其中該寡核苷酸與具有與該菸鹼醯 胺轉磷酸核糖基酶(NAMPT)聚核苷酸之該反義及/或有義 核酸分子互補之至少約5個連續核酸的序列具有至少8〇% 158213.doc 201210611 序列一致性。 17,種活體外調節哺乳動物細胞K織中於驗醢胺轉麟酸 核糖基酶(NAMPT)之功能及/或表現之方法,該方法包 含:使該等細胞或組織與至少—種長度為約5至30個核 苦酸之反義募核苦酸接觸’該至少一種反義募核苷酸對 菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之有義及/或 天然反義版之非編碼及/或編碼序列具特显性,立_該至 少一種反義寡核苷酸與至少一種如SEQ ID N〇:工至16所 示之核酸序列具有至少50%序列一致性;及活體外調節 哺乳動物細胞或組織中該菸鹼醯胺轉磷酸核糖基酶 (NAMPT)之該功能及/或表現。 18. —種反義募核苷酸之用途,其係用於製造用以調節生物 系統中於驗酿胺轉碌酸核糖基酶(NAMPT)聚核苷酸之功 能及/或表現之藥劑’其中該募核苷酸長度為5至30個核 苷酸且與菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之 ❹ 天然反義序列的反向互補序列具有至少50%序列一致 性。 19. 如請求項18之用途’其中該寡核苷酸與包含SEq id NO: 2之天然反義轉錄物核苷酸1至882、或SEQ ID NO: 3之1 至 915、或 SEQ ID NO: 4之 1 至 165、或 SEQ ID NO: 5之 1 至 533、或 SEQ ID NO: 6之 1 至 522、或 SEQ ID NO: 7之 1 至 141、或 SEQ ID NO: 8之 1 至 519、或 SEQ ID NO·· 9之 1 至 374、或 SEQ ID NO: 10之 1 至 240、或 SEQ ID NO: 11 之 1 至 353、或 SEQ ID NO: 12之 1 至 534、或 SEQ ID NO: 13 158213.doc _5. 201210611 之 1至 959、或 SEQ ID NO: 14之 1 至 127、或 SEQ ID NO: 15之1至232、或SEQ ID NO: 16之1至387内5至30個連續 核苷酸之聚核苷酸的反向互補序列具有至少50%序列一 致性。 20. —種反義募核苷酸之用途,其係用於製造用以調節患者 細胞或組織中菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷 酸之功能及/或表現之藥劑,其中該寡核苷酸長度為5至 3 0個核苷酸且與該菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚 核苷酸之反義寡核苷酸具有至少50%序列一致性。 21. 如請求項20之用途,其中該寡核苷酸與包含SEQ ID NO: 2之天然反義轉錄物核苷酸1至882、或SEQ ID NO: 3之1 至 915、或 SEQ ID NO: 4之 1 至 165、或 SEQ ID NO: 5之 1 至 533、或 SEQ ID NO: 6之 1 至 522、或 SEQ ID NO: 7之 1 至 141、或 SEQ ID NO: 8之 1 至 519、或 SEQ ID NO: 9之 1 至 3 74、或 SEQ ID NO: 10之 1 至 240、或 SEQ ID NO: 11之 1 至 353、或 SEQ ID NO: 12之 1 至 534、或 SEQ ID NO: 13 之 1至 959、或 SEQ ID NO·· 1 4之 1 至 127、或 SEQ ID NO: 15之1至232、或SEQ ID NO: 16之1至387内5至30個連續 核苷酸之聚核苷酸的反向互補序列具有至少50%序列一 致性。 22· —種反義寡核苷酸之用途,其係用於製造用以調節生物 系統中菸鹼醢胺轉磷酸核糖基酶(NAMPT)聚核苷酸之功 能及/或表現之藥劑,其中該寡核苷酸靶向該菸鹼醯胺轉 磷酸核糖基酶(NAMPT)聚核苷酸之天然反義募核苷酸之 158213.doc 201210611 區。 23.如請求項22之用途’其中該菸驗醯胺轉磷酸核掩基酶 (NAMPT)之功能及/或表現在活體内相對於對照組择加 24·如請求項22之用途,其中該反義寡核苷酸靶向菸驗酿胺 轉磷酸核糠基酶(NAMPT)聚核苷酸之天然反義序列。 ’ 25.如請求項22之用途,其中該反義寡核苷酸靶向包含終驗 醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之編碼及/或非編 碼核酸序列之核酸序列。 ® 26.如請求項22之用途’其中該反義寡核苷酸乾向終驗醯胺 轉磷酸核糖基酶(NAMPT)聚核苷酸之重疊及/或不重疊序 列。 27_如請求項22之用途,其中該反義寡核苷酸包含一或多個 選自以下之修飾:至少一個經修飾糖部分、至少一個經 修飾核苷間鍵聯、至少一個經修飾核苷酸及其組合。 28. 如請求項27之用途,其中該一或多個修飾包含至少一個 〇 選自以下之經修飾糖部分:2,_〇_甲氧基乙基修飾糖部 分、2'-曱氧基修飾糖部分、2,_〇_烷基修飾糖部分、雙環 糖部分及其組合。 29. 如請求項27之用途,其中該一或多個修飾包含至少一個 選自以下之經修飾核苷間鍵聯:硫代磷酸酯、2,_〇甲氧 基乙基(MOE)、2 -1、燒基鱗酸醋、二硫代構酸醋 '烧 基硫代膦I自曰、磷醯胺酯、胺基甲酸酯、碳酸酯、磷酸 三酯、乙醯胺酯、羧甲基酯及其組合。 30. 如請求項27之用^伞,甘 途,、中s亥—或多個修飾包含至少一個 158213.doc 201210611 選自以下之經修飾核普酸:肽核酸(PNA)、鎖核酸 (LNA)、阿糖核酸(FANA)、其類似物、衍生物及組合。 31. 如請求項18之用途,其中該寡核苷酸包含至少一種如 SEQ ID NO: I7至1!所示之募核苷酸序列。 32. —種短干擾RNA(siRNA)寡核苷酸之用途,其係用於製 造用以調節哺乳動物細胞或組織中菸鹼醯胺轉磷酸核糖 基酶(NAMPT)基因之功能及/或表現之藥劑,其中該 siRNA寡核苷酸長度為5至30個核苷酸且對菸鹼醯胺轉磷 酸核糖基酶(NAMPT)聚核苷酸之反義聚核苷酸具特異 I"生其中5亥siRNA券核普酸與該於驗醯胺轉鱗酸核糖基 酶(NAMPT)聚核苷酸之反義及/或有義核酸分子之至少約 5個連續核酸的互補序列具有至少5〇%序列一致性。 33. 如請求項32之用途,其中該寡核苷酸與具有與韻終鹼醯 胺轉磷酸核糖基酶(NAMPT)聚核苷酸之該反義及/或有義 核酸分子互補之至少約5個連續核酸的序列具有至少8〇% 序列一致性。 34. —種反義寡核苷酸之用途,其係用於製造用以調節哺乳 動物細胞或組織中菸鹼醯胺轉磷酸核糖基酶(NAMPT)之 功能及/或表現之藥劑,其中該反義寡核苷酸長度為約5 至30個核苷酸且對菸鹼醯胺轉磷酸核糖基酶(Nampt)聚 核苦酸之有義及/或天然反義股之非編碼及/或編碼序列具 有特異性’其中該反義募核苷酸與至少一種如SEQ ID NO: 1至16所示之核酸序列具有至少50%序列—致性。 158213.doc 1 5. 種合成之經修飾募核苷酸,其包含至少一個修飾,其 201210611 中該至少一個修飾係選自:至少一個經修飾糖部分;至 少一個經修飾核苷酸間鍵聯;至少一個經修飾核苷酸及 其組合;其中與正常對照組相比,該寡核苷酸為在活體 内或活體外與菸鹼醯胺轉磷酸核糖基酶(NAMPT)基因雜 交且調節該菸鹼醯胺轉磷酸核糖基酶(NAMPT)基因之功 忐及/或表現之反義化合物’且其中該寡核苷酸與具有與 該於驗醯胺轉磷酸核糖基酶(NAmpt)聚核苷酸及其對偶 基因、同源物、同功異型物、變異體、衍生物、突變 體、片段或組合之反義及/或有義核酸分子互補之至少約 5個連續核酸的序列具有至少5〇%序列一致性。 36. 如請求項35之寡核苷遊,其中該寡核苷酸長度為5至30 個核苷酸且與該NAMPT基因之天然反義轉錄物内5至30 個連續核苷酸之反向互補序列具有至少50%序列一致 性。 37. 如請求項36之募核苷酸,其中該至少一個修飾包含選自 由以下組成之群的核苷酸間鍵聯:硫代填酸酯、烧基膦 酸酯、二硫代填酸酯、烷基硫代膦酸酯、磷醯胺酯、胺 基甲酸酯、碳酸酯、磷酸三酯、乙醯胺酯、羧甲基酯及 其組合。 3 8.如請求項36之寡核苷酸,其中該募核苷酸包含至少一個 硫代磷酸酯核苷酸間鍵聯。 3 9.如請求項36之寡核苷酸,其中該募核苷酸包含硫代磷酸 酯核苷酸間鍵聯之主鏈。 40.如請求項36之寡核苷酸,其中該募核苷酸包含至少一個 158213.doc 201210611 經修飾核苷酸,該經修飾核苷酸選自:肽核酸、鎖核酸 (LNA)、其類似物、衍生物及組合。 41. 如請求項36之寡核普酸,其中該募核苷酸包含複數個修 飾,其中該等修飾包含選自以下之經修飾核苷酸:硫代 磷酸酯、烷基膦酸酯、二硫代磷酸酯、烷基硫代膦酸 酯、磷醯胺酯、胺基甲酸酯、碳酸酯、磷酸三酯、乙醯 胺酯、羧曱基酯及其組合。 42. 如請求項36之募核苦酸,其中該寡核苷酸包含複數個修 飾,其中該等修飾包含選自以下之經修飾核苷酸:肽核 酸、鎖核酸(LNA)、其類似物、衍生物及組合。 43. 如請求項36之寡核苷酸,其中該寡核苷酸包含至少一個 選自以下之經修飾糖部分:2|-〇_甲氧基乙基修飾糖部 分、2’·曱氧基修飾糖部分、2|_〇_烷基修飾糖部分、雙環 糖部分及其組合。 44. 如請求項36之募核苷酸,其中該寡核苷酸包含複數個修 飾’其中該等修飾包含選自以下之經修飾糖部分:2,_〇_ 甲氧基乙基修飾糖部分、2’-曱氡基修飾糖部分、2,_〇_院 基修飾糖部分、雙環糖部分及其組合。 45. 如請求項36之募核苷酸,其中該募核苷酸長度為至少約 5至30個核苷酸且與菸鹼醯胺轉磷酸核糖基酶(ΝΑΜρτ) 聚核苷酸之反義及/或有義股雜交,其中該募核苷酸與該 菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之反義及/或 有義編碼及/或非編碼核酸序列之至少約5個連續核酸的 互補序列具有至少約60%序列一致性。 158213.doc -10· 201210611 46·如請求項36之寡核苦酸,其中該募核普酸與該菸鹼醯胺 轉磷酸核糖基酶(NAMPT)聚核苷酸之該反義及/或有義編 碼及/或非編碼核酸序列之至少約5個連續核酸的互補序 列具有至少約80%序列一致性。 47.如請求項36之募核苷酸,其中與正常對照組相比,該寡 、 核皆酸在活體内或活體外與至少一種菸鹼醯胺轉磷酸核 糖基酶(NAMPT)聚核苷酸雜交且調節該至少一種菸鹼醯 胺轉磷酸核糠基酶(NAMPT)聚核苷酸之表現及/或功能。 〇 48.如請求項36之募核苷酸,其中該寡核苷酸包含如SEQ ID NO: 17至31所示之序列。 49. 一種醫藥組合物,其包含一或多種如請求項以之對一或 多種菸鹼醯胺轉磷酸核糖基酶(ΝΑΜρτ)聚核苷酸具有特 異性之寡核苷酸及醫藥學上可接受之賦形劑。 50. 如請求項49之組合物,其中該等寡核苷酸與如SEQ 1〇 NO: 17至31所示核苷酸序列中之任一者相比具有至少約 0 40%序列一致性。 51. 如請求項49之組合物,其中該等募核苷酸包含如SEQ m NO: 17至31所示之核苷酸序列。 52·如請求項51之組合物,其中如SEQ ID NO: 17至31所示之 該等券核苷酸包含一或多個修飾或取代。 53 ·如叫求項52之組合物,其中該一或多個修飾係選自:硫 代&酸酯、曱基膦酸酯、肽核酸、鎖核酸(LNA)分子及 其組合。 54· —種反義寡核苷酸之用途,其係用於製造用以預防或治 158213.doc 201210611 療與至少一種菸鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷 酸及/或至少一種其編碼產物相關之疾病的藥劑,其中該 反義寡核苦酸結合該至少一種於驗醯胺轉碌酸核糖基酶 (NAMPT)聚核苷酸之天然反義序列且調節該至少一種於 鹼醯胺轉磷酸核糖基酶(NAMPT)聚核苷酸之表現。 55.如請求項54之用途’其中與該至少一種菸鹼醯胺轉磷酸 核糖基酶(NAMPT)聚核苷酸相關之疾病係選自:與 NAMPT之功能及/或表現異常相關之疾病或病症、癌 症、與細胞增殖相關或以細胞增殖為特徵之疾病或病 狀、細胞凋亡、與NAMPT之表現或功能突變或異常相關 之疾病或病症、血細胞減少症(例如骨髓或淋巴細胞系等 之血細胞減少症)、嗜中性球減少症(例如重度先天性嗜 中性球減少症、其他形式之先天性及後天性嗜中性球減 J症、化學療法誘發之發熱性嗜中性球減少症等)、白血 病、急性骨髓性白血病(AML) '骨髓細胞系之癌症、與 NAMPT含量升高相關之惡性疾病、關於造血之先天性非 惡性疾病或病症、歸因於ΝΑΜρτ之表現及/或活性改變 之免疫缺乏症、動脈粥樣硬化、與葡萄糖恆定異常相關 之疾病或病症、與胰島素分泌異常相關之疾病或病症、 肥胖症、心血管疾病或病症、肝細胞〉周亡、與粒線體生 物發生異吊相關之疾病或病症、與骨骼肌相關之疾病、 其與!:細胞生成異常相關之疾病、病症或病 _ /、οχ』豕族相關之疾病、病症或病狀;與肝臟 二^甘油較定異常相關之疾病或病症;與晝夜節律活 158213.doc -12- 201210611 動相關之疾病、病症或病狀;與免疫系統相關之疾病或 病症,與關於内毒素耐受之後生性程式改寫(epigenetic reprogramming)相關之疾病、病症或病狀;病毒感染、 與干擾素抗病毒反應異常相關之疾病或病症、慢性發炎 性疾病或病症(例如發炎性腸病,包括克羅恩氏病 (Crohn's disease)、潰瘍性結腸炎、牛皮癬、關節炎 專)企管疾病或病症(例如慢性潰瘍、缺血性中風、心 肌梗塞、心絞痛及血管性癡呆等)、代謝性疾病或病症、 炎症、衰老、非酒精性脂肪肝病、及與去乙醯基酶 (Sirtuins)SIRT之表現及/或活性異常相關之疾病或病症。 56. 種在生物系統中活體外誘導細胞凋亡之方法,該方法 包含向該系統投與長度為5至3〇個核苷酸且與ΝΑΜρτ聚 核苷酸之天然反義轉錄物之反向互補序列内5至3〇個連 續核苷酸至少50%—致的寡核苷酸。 57. 如請求項56之方法,其中該天然反義轉錄物具有犯9 ID NO: 2至 16。 58. 如請求項56之方法,其中該生物系統為患者細胞或組 織。 59. 種在生物系統中活體外誘導細胞凋亡之方法,該方法 包含向該系統投與長度為約5至3〇個核苷酸之募核苷 酸,其中該募核苷酸藉由靶向目標基因之天然反義轉錄 物來上調該基因,且其中該寡核苷酸與該所上調基因之 該天然反義轉錄物之反向互補序列至少5〇%一致。 60. —種寡核苷酸之用途’其係用於製造用以在生物系統中 158213.doc •13- 201210611 誘導細胞凋亡之藥劑,其中該募核苷酸長度為5至30個 核苷酸且與NAMPT聚核苷酸之天然反義轉錄物之反向互 補序列内5至3〇個連續核苷酸至少50%—致。 61. 如請求項60之用途,其中該天然反義轉錄物具有冗卩ID NO: 2至 16。 62. 如請求項60之用途,其中該生物系統為患者細胞或組 織。 63 · —種寡核苷酸之用途,其係用於製造用以在生物系統中 誘導細胞凋亡之藥劑’其中該寡核苷酸長度為約5至3〇 個核苷酸且藉由靶向目標基因之天然反義轉錄物來上調 «亥基因,且其中該寡核皆酸與該所上調基因之該天然反 義轉錄物之反向互補序列至少5〇%一致。 64_ —種鑑別及選擇至少一種對ΝΑΜρτ基因之天然反義轉錄 物有選擇.性作為用於活體内投與之所選目標聚核苷酸的 寡核苷酸之方法,該方法包含:鑑別至少一種包含與該 所選目標聚核苷酸之反義聚核苷酸至少部分互補之至少 5個連續核苷酸的寡核苷酸;量測在嚴格雜交條件下, 反義寡核苷酸與該目標聚核苷酸或該所選目標聚核苷酸 之該反義聚核苷酸的雜交物之熱熔點;及基於所獲得之 資矾選擇至少一種用於活體内投與之寡核苷酸。 158213.doc -14-201210611 VII. Scope of application for patents: 1. A method for the function and/or performance of a guanamine-transfer ribosyl ribosylase (NAMPT) polynucleotide in an in vitro regulated biological system, the method comprising: At least one antisense oligonucleotide having a length of 5 to 30 nucleotides in contact with the natural antisense of the at least one oligonucleotide and the nicotinamide transaminase (NAMPT) polynucleotide The reverse complement of the sequence has at least 50% sequence identity; thereby modulating the function and/or performance of the nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide. 2. A method of modulating the function and/or expression of a nicotine guanamine-transphosphoryl ribosylase (NAMPT) polynucleotide in a biological system according to claim 1, the method comprising: bringing the biological system to at least one length In contact with 5 to 30 nucleotide antisense oligonucleotides, wherein the at least one nucleotide and the natural antisense transcript comprising SEQ ID NO: 2, nucleotides 1 to 882, or SEQ ID NO: 3 to 915, or 1 to 165 of SEQ ID NO: 4, or 1 to 533 of SEQ ID NO: 5, or 1 to 522 of SEQ ID NO: 6, or 1 to 141 of SEQ ID NO: 7, Or SEQ ID NO: 8 to 1 519, or SEO 〇 ID NO: 9 to 1 to 374, or SEQ ID NO: 1 to 1 to 240, or SEQ ID NO: 1 to 1 to 353, or SEQ ID NO: 12 to 534, or SEQ ID NO: 13 to 1 to 959, or SEQ ID NO: 14 to 1 to 127, or SEQ ID NO: 15 to 1 to 232, or SEQ ID NO: 16 to 1 to 387 The reverse complement of a polynucleotide of 5 to 30 contiguous nucleotides has at least 50% sequence identity; thereby modulating the function of the chiral amine ribosyl ribosylase (NAMPT) polynucleotide and / or performance. 3. A method for the in vitro regulation of the function and/or expression of a guanine-transphosphoryl ribose 158213.doc 201210611 nucleotide (NAMPT) polynucleotide in a patient cell or tissue, the method comprising: causing the cells or The tissue is contacted with at least an antisense oligonucleotide having a length of 5 to 3 nucleotides, wherein the nucleotide is opposite to the nicotine indole phosphoribosyltransferase (NAMPT) polynucleotide The oligodeoxynucleotide has at least 50% sequence identity; thereby, in vitro, modulating the function and/or performance of the nampt nucleotide in the cells or tissues of a patient. 4. A method of modulating the function and/or expression of a nicotine guanamine transphosphoryrosidase (NAMPT) polynucleotide in a patient cell or tissue according to claim 3, the method comprising: causing the cell or tissue to be at least one An antisense nucleotide contact of 5 to 30 nucleotides in length, wherein the at least one oligonucleotide and the natural antisense transcript comprising nucleotides 1 to 882 of SEQ ID NO: 2, or SEQ ID NO : 3 to 1 to 915, or SEQ ID NO: 4 to 1 to 165, or SEQ ID NO: 5 to 1 to 533, or SEQ ID NO: 6 to 1 to 522, or SEQ ID NO: 7 to 1 to 141 Or 1 to 519 of SEQ ID NO: 8, or 1 to 374 of SEQ ID NO: 9, or 1 to 240 of SEQ ID NO: 10, or 1 to 353 of SEQ ID NO: 11, or SEQ ID NO: 12 to 1 534, or SEQ ID NO: 13 to 1 to 959, or SEQ ID NO: 14 to 1 to 127, or SEQ ID NO: 15 to 1 to 232, or SEQ ID NO: 16 to 1 to 3 87 The reverse complement of the 5 to 30 contiguous nucleotides of the polynucleotide has at least 50% sequence identity; thereby modulating the function of the awake amine repelling ribosyltransferase (NAMPT) polynucleotide And / or performance. 5. A method for the in vitro regulation of the function and/or expression of a nicotine guanamine-transphosphoryl ribosylase (NAMPT) polynucleotide in a biological system, the method comprising: 158213.doc 201210611 The at least one antisense nucleus of the region of the natural antisense oligonucleotide of the nicotine guanamine-transphosphoryl ribosyltransferase (NAMPT) polynucleotide is contacted, thereby modulating the chiral amine transesterase The function and/or performance of a base enzyme (NAMPT) polynucleotide. 6. The method of claim 5, wherein the function and/or performance of the nicotine indoleamine phosphoribosyltransferase (NAMPT) is increased in vitro relative to the control group. 7. The method of claim 5, wherein the at least one antisense nucleotide targets a natural antisense D sequence of a nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide. 8. The method of claim 5, wherein the at least one antisense nucleotide targets a nucleic acid sequence comprising a coding and/or non-coding nucleic acid sequence of a nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide . 9. The method of claim 5, wherein the at least one antisense oligonucleotide targets overlapping and/or non-overlapping sequences of a nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide. The method of claim 5, wherein the at least one antisense oligonucleotide comprises one or more modifications selected from the group consisting of at least one modified sugar moiety 'to J modified internucleoside linkages, At least one modified nucleotide and combinations thereof. The method of claim 10, wherein the one or more modifications comprise at least one modified sugar moiety selected from the group consisting of: 2, 〇 〇 曱 methoxyethyl modified sugar moiety 71 2 oxime modified sugar moiety, 2'-0-alkyl-modified sugar moiety, bicyclic sugar moiety, and combinations thereof. 12. The method of claim 1, wherein the one or more modifications comprise at least one of 158213.doc 201210611 modified internucleoside linkages selected from the group consisting of phosphorothioate, 2, methoxyl (MOE), 2-fluoro, alkyl phosphonate, dithiophosphate, alkyl thiophosphonate, ph〇sph〇ramidate, urethane, carbonate, Phosphate triester, acetamidate, carboxynonyl ester, and combinations thereof. 13. The method of claim 1 , wherein the one or more modifications comprise at least one modified nucleotide selected from the group consisting of a peptide nucleic acid (pNA), a locked nucleic acid (LNA), an adenine nucleic acid (FANA), Analogs, derivatives and combinations. 14. The method of claim 1, wherein the at least one oligonucleotide comprises at least one oligonucleotide sequence as set forth in SEQ ID NO: Π to 31. 15. A method of in vitro modulating the function and/or expression of a nicotine guanamine-transfer ribosyl ribosylase (NAMPT) gene in a mammalian cell or tissue, the method comprising: affixing the cell or tissue to at least one length Antisense to a nicotinicinamine-transphosphoryltransferase (ΝΑΜρτ) polynucleotide by contacting at least one siRNA nucleotide with a short interfering RNA (siRNA) oligonucleotide of 5 to 3 nucleotides Polynucleotide specific, wherein the at least one siRNA raised nucleotide and at least about 5 antisense and/or sense nucleic acid molecules of the nicotinicinamine phosphoribosyltransferase (ΝΑΜρτ) polynucleotide Complementary sequences of contiguous nucleic acids have at least 50% sequence identity; and in vitro modulate the function and/or expression of nicotinamide transphosphatase (NAMPT) in mammalian cells or tissues. The method of claim 15, wherein the oligonucleotide is at least about complementary to the antisense and/or sense nucleic acid molecule having the nicotinicinamine phosphoribosyltransferase (NAMPT) polynucleotide. The sequence of 5 contiguous nucleic acids has a sequence identity of at least 8〇 158213.doc 201210611. 17. A method for in vitro regulation of the function and/or expression of amylin ribosyl ribosylase (NAMPT) in a mammalian cell K-woven, the method comprising: causing the cells or tissues to be at least Antisense nucleoside acid exposure of about 5 to 30 nucleotides of 'the at least one antisense nucleotide to the sense and/or natural to the nicotinamide transphosphatase (NAMPT) polynucleotide The non-coding and/or coding sequence of the antisense version is distinctive, and the at least one antisense oligonucleotide has at least 50% sequence identity to at least one of the nucleic acid sequences set forth in SEQ ID N:: And the in vitro regulation of the function and/or expression of the nicotine indoleamine phosphoribosyltransferase (NAMPT) in mammalian cells or tissues. 18. The use of an antisense nucleotide for the manufacture of a medicament for modulating the function and/or performance of a serotonin ribosyltransferase (NAMPT) polynucleotide in a biological system. Wherein the nucleotide is 5 to 30 nucleotides in length and is at least 50% identical to the reverse complement of the natural antisense sequence of the nicotinic indoleamine phosphoribosyltransferase (NAMPT) polynucleotide. Sex. 19. The use of claim 18, wherein the oligonucleotide is associated with a natural antisense transcript nucleotide 1 to 882 comprising SEq id NO: 2, or 1 to 915 of SEQ ID NO: 3, or SEQ ID NO : 1 to 165, or 1 to 533 of SEQ ID NO: 5, or 1 to 522 of SEQ ID NO: 6, or 1 to 141 of SEQ ID NO: 7, or 1 to 519 of SEQ ID NO: 8. Or 1 to 374 of SEQ ID NO. 9 or 1 to 240 of SEQ ID NO: 10, or 1 to 353 of SEQ ID NO: 11, or 1 to 534 of SEQ ID NO: 12, or SEQ ID NO : 13 158213.doc _5. 201210611 1 to 959, or SEQ ID NO: 14 to 1 to 127, or SEQ ID NO: 15 to 1 to 232, or SEQ ID NO: 16 to 1 to 387 within 5 to 30 The reverse complement of the contiguous nucleotide polynucleotide has at least 50% sequence identity. 20. Use of an antisense raised nucleotide for the manufacture of a medicament for modulating the function and/or expression of a nicotinamide transphosphatase (NAMPT) polynucleotide in a cell or tissue of a patient Wherein the oligonucleotide is 5 to 30 nucleotides in length and has at least 50% sequence identity to the antisense oligonucleotide of the nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide . 21. The use of claim 20, wherein the oligonucleotide and the natural antisense transcript nucleotides 1 to 882 comprising SEQ ID NO: 2, or 1 to 915 of SEQ ID NO: 3, or SEQ ID NO : 1 to 165, or 1 to 533 of SEQ ID NO: 5, or 1 to 522 of SEQ ID NO: 6, or 1 to 141 of SEQ ID NO: 7, or 1 to 519 of SEQ ID NO: 8. Or SEQ ID NO: 9 to 1 74, or 1 to 240 of SEQ ID NO: 10, or 1 to 353 of SEQ ID NO: 11, or 1 to 534 of SEQ ID NO: 12, or SEQ ID NO : 1 to 959 of 13, or 1 to 127 of SEQ ID NO., or 1 to 232 of SEQ ID NO: 15, or 5 to 30 contiguous nucleotides of 1 to 387 of SEQ ID NO: 16 The reverse complement of the polynucleotide has at least 50% sequence identity. 22. The use of an antisense oligonucleotide for the manufacture of a medicament for modulating the function and/or expression of a nicotinamide phosphatase (NAMPT) polynucleotide in a biological system, wherein The oligonucleotide targets the 158213.doc 201210611 region of the natural antisense raised nucleotide of the nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide. 23. The use of claim 22, wherein the function and/or performance of the test for amidoxime phosphoribosylase (NAMPT) is increased in vivo relative to a control group, wherein the use of claim 22, wherein Antisense oligonucleotides target the natural antisense sequence of the chitosan-transferase ribosylase (NAMPT) polynucleotide. 25. The use of claim 22, wherein the antisense oligonucleotide targets a nucleic acid sequence comprising a coding and/or non-coding nucleic acid sequence of a final amidoxime phosphoribosyltransferase (NAMPT) polynucleotide. ® 26. The use of claim 22, wherein the antisense oligonucleotide is dried to the overlapping and/or non-overlapping sequence of the final amidoxime phosphoribosylase (NAMPT) polynucleotide. The use of claim 22, wherein the antisense oligonucleotide comprises one or more modifications selected from the group consisting of at least one modified sugar moiety, at least one modified internucleoside linkage, at least one modified core Glycosylates and combinations thereof. 28. The use of claim 27, wherein the one or more modifications comprise at least one modified sugar moiety selected from the group consisting of: 2, _〇_methoxyethyl modified sugar moiety, 2'-decyloxy modification A sugar moiety, a 2,_〇-alkyl modified sugar moiety, a bicyclic sugar moiety, and combinations thereof. 29. The use of claim 27, wherein the one or more modifications comprise at least one modified internucleoside linkage selected from the group consisting of: phosphorothioate, 2,-methoxyethyl (MOE), 2 -1, tartaric acid vinegar, dithio acid vinegar 'alkyl thiophosphine I ruthenium, phosphonium amide, urethane, carbonate, phosphate triester, acetaminophen, carboxymethyl Base esters and combinations thereof. 30. The use of Umbrella, Gantu, Zhongshui, or a plurality of modifications as claimed in claim 27 includes at least one of 158213.doc 201210611 Modified nucleotides selected from the group consisting of peptide nucleic acids (PNA), locked nucleic acids (LNA) ), arabinic acid (FANA), analogs, derivatives and combinations thereof. 31. The use of claim 18, wherein the oligonucleotide comprises at least one nucleotide sequence as set forth in SEQ ID NOs: I7 to 1!. 32. Use of a short interfering RNA (siRNA) oligonucleotide for the production of a function and/or expression of a nicotine guanamine transphosphatase (NAMPT) gene in a mammalian cell or tissue An agent wherein the siRNA oligonucleotide is 5 to 30 nucleotides in length and specific for an antisense polynucleotide of a nicotinic guanamine transphosphoposyltransferase (NAMPT) polynucleotide; 5 siRNA vouchers having at least 5 互补 with the antisense of the serotonin ribosyl ribosylase (NAMPT) polynucleotide and/or the complement of at least about 5 contiguous nucleic acids of the sense nucleic acid molecule % sequence consistency. 33. The use of claim 32, wherein the oligonucleotide is at least about complementary to the antisense and/or sense nucleic acid molecule having a final nucleotide phosphatase ribosyltransferase (NAMPT) polynucleotide The sequence of 5 contiguous nucleic acids has at least 8% sequence identity. 34. Use of an antisense oligonucleotide for the manufacture of a medicament for modulating the function and/or expression of nicotinamide transphosphatase (NAMPT) in a mammalian cell or tissue, wherein Antisense oligonucleotides are about 5 to 30 nucleotides in length and are non-coding and/or non-coding and/or natural antisense strands of nicotinamide transphosphatase (Nampt) polynucleic acid. The coding sequence is specific in that the antisense oligonucleotide has at least 50% sequence identity to at least one of the nucleic acid sequences set forth in SEQ ID NOS: 1 to 16. 158213.doc 1 5. A synthetic modified nucleotide comprising at least one modification, wherein the at least one modification in 201210611 is selected from the group consisting of: at least one modified sugar moiety; at least one modified internucleotide linkage At least one modified nucleotide and a combination thereof; wherein the oligonucleotide hybridizes to the nicotinic guanamine transphosphatase (NAMPT) gene in vivo or in vitro as compared to a normal control group An antisense compound of the nicotinicinamine-transphosphoryl ribosylase (NAMPT) gene and/or an antisense compound which exhibits a nucleoside and a nucleoside with a nucleoside transphosphoryrosylase (NAmpt) a sequence of at least about 5 contiguous nucleic acids complementary to the antisense and/or sense nucleic acid molecules of the nucleoside, its homologs, homologs, isoforms, variants, derivatives, mutants, fragments or combinations having at least about 5 contiguous nucleic acids 5〇% sequence consistency. 36. The oligonucleoside of claim 35, wherein the oligonucleotide is 5 to 30 nucleotides in length and is inverted from 5 to 30 contiguous nucleotides within the natural antisense transcript of the NAMPT gene The complementary sequence has at least 50% sequence identity. 37. The nucleotide of claim 36, wherein the at least one modification comprises an internucleotide linkage selected from the group consisting of: a thiolate, a decyl phosphonate, a dithiolate Alkyl thiophosphonates, phosphonium amides, urethanes, carbonates, phosphates, acetamides, carboxymethyl esters, and combinations thereof. 3. The oligonucleotide of claim 36, wherein the raised nucleotide comprises at least one phosphorothioate internucleotide linkage. 3. The oligonucleotide of claim 36, wherein the nucleotide comprises a backbone of a phosphorothioate internucleotide linkage. 40. The oligonucleotide of claim 36, wherein the raised nucleotide comprises at least one 158213.doc 201210611 modified nucleotide selected from the group consisting of: a peptide nucleic acid, a locked nucleic acid (LNA), Analogs, derivatives and combinations. 41. The oligonucleotide of claim 36, wherein the nucleotide comprises a plurality of modifications, wherein the modifications comprise modified nucleotides selected from the group consisting of phosphorothioates, alkyl phosphonates, Phosphorothioate, alkyl thiophosphonate, phosphonium amide, urethane, carbonate, phosphotriester, acetamide, carboxydecyl ester, and combinations thereof. 42. The nucleic acid of claim 36, wherein the oligonucleotide comprises a plurality of modifications, wherein the modifications comprise modified nucleotides selected from the group consisting of peptide nucleic acids, locked nucleic acids (LNA), analogs thereof , derivatives and combinations. 43. The oligonucleotide of claim 36, wherein the oligonucleotide comprises at least one modified sugar moiety selected from the group consisting of: 2|-〇-methoxyethyl modified sugar moiety, 2'. Modified sugar moieties, 2|_〇-alkyl modified sugar moieties, bicyclic sugar moieties, and combinations thereof. 44. The nucleotide of claim 36, wherein the oligonucleotide comprises a plurality of modifications wherein the modifications comprise a modified sugar moiety selected from the group consisting of: 2, _〇_methoxyethyl modified sugar moiety 2'-mercapto-modified sugar moiety, 2,_〇_院-modified sugar moiety, bicyclic sugar moiety, and combinations thereof. 45. The nucleotide of claim 36, wherein the nucleotide is at least about 5 to 30 nucleotides in length and is antisense to the nicotinicin phosphatase (ΝΑΜρτ) polynucleotide And/or a sense strand hybrid, wherein the raised nucleotide and at least the antisense and/or sense encoding and/or non-coding nucleic acid sequence of the nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleotide The complementary sequence of about 5 contiguous nucleic acids has at least about 60% sequence identity. 158213.doc -10·201210611 46. The oligonucleotide of claim 36, wherein the nucleotide and the nicotine indole phosphoryl ribosylase (NAMPT) polynucleotide are antisense and/or A complementary sequence of at least about 5 contiguous nucleic acids of a sense encoding and/or non-coding nucleic acid sequence has at least about 80% sequence identity. 47. The nucleotide of claim 36, wherein the oligo-nuclear acid is in vivo or in vitro with at least one nicotine indoleamine phosphoribosyltransferase (NAMPT) polynucleoside compared to a normal control group. The acid hybridizes and modulates the performance and/or function of the at least one nicotine guanamine transphosphatase (NAMPT) polynucleotide. 〇 48. The nucleotide of claim 36, wherein the oligonucleotide comprises the sequence set forth in SEQ ID NOs: 17 to 31. 49. A pharmaceutical composition comprising one or more oligonucleotides as claimed in one or more nicotine guanamine transphosphatase (ΝΑΜρτ) polynucleotides and pharmaceutically acceptable Accepted excipients. 50. The composition of claim 49, wherein the oligonucleotides have at least about 40% sequence identity compared to any of the nucleotide sequences set forth in SEQ ID NO: 17 to 31. 51. The composition of claim 49, wherein the raised nucleotides comprise a nucleotide sequence as set forth in SEQ m NO: 17 to 31. 52. The composition of claim 51, wherein the nucleotide nucleotides as set forth in SEQ ID NOS: 17 to 31 comprise one or more modifications or substitutions. 53. The composition of claim 52, wherein the one or more modifications are selected from the group consisting of: thio & acid esters, decyl phosphonates, peptide nucleic acids, locked nucleic acid (LNA) molecules, and combinations thereof. 54. Use of an antisense oligonucleotide for the manufacture of a nucleoside phosphatase (NAMPT) polynucleotide and/or 158213.doc 201210611 and/or at least one nicotinic acid phosphatase (NAMPT) polynucleotide and/or At least one agent encoding a disease associated with the product, wherein the antisense oligonucleotide binds to the at least one natural antisense sequence of a serotonin ribosyltransferase (NAMPT) polynucleotide and modulates the at least one The performance of the base amide transphosphoribozyme (NAMPT) polynucleotide. 55. The use of claim 54 wherein the disease associated with the at least one nicotine indole phosphoribosyltransferase (NAMPT) polynucleotide is selected from the group consisting of: a disease associated with abnormal function and/or performance of NAMPT or A condition, a cancer, a disease or condition characterized by cell proliferation or characterized by cell proliferation, apoptosis, a disease or condition associated with a mutation or abnormality in NAMPT expression or function, or a cytopenia (eg, bone marrow or lymphocyte system, etc.) Hematocytopenia), neutropenia (such as severe congenital neutropenia, other forms of congenital and acquired neutrophil reduction, chemotherapy-induced febrile neutrophils) Reduction, etc.), leukemia, acute myeloid leukemia (AML) 'Cell cancer in bone marrow cell line, malignant disease associated with elevated NAMPT levels, congenital non-malignant disease or condition with hematopoiesis, attributed to ΝΑΜρτ and/ Or immunodeficiency with altered activity, atherosclerosis, a disease or condition associated with a constant glucose abnormality, a disease or condition associated with abnormal insulin secretion, Fat disease, a cardiovascular disease or disorder, hepatocyte> weeks death, and hydrocarbons occur mitochondrial disease associated or heterologous hanging disorders, diseases associated with the skeletal muscle, which! : diseases, disorders, or diseases associated with abnormal cell production _ /, οχ 豕 diseases, disorders, or conditions associated with steroids; diseases or conditions associated with abnormal liver glycerol; and circadian rhythm 158213.doc -12 - 201210611 Diseases, disorders or conditions associated with the immune system; diseases or conditions associated with the immune system, diseases, conditions or conditions associated with epigenetic reprogramming after endotoxin tolerance; viral infections, and interferons A disease or condition associated with an abnormal antiviral response, a chronic inflammatory disease or condition (eg, inflammatory bowel disease, including Crohn's disease, ulcerative colitis, psoriasis, arthritis), or a disease or condition ( For example, chronic ulcers, ischemic stroke, myocardial infarction, angina pectoris and vascular dementia, etc.), metabolic diseases or conditions, inflammation, aging, nonalcoholic fatty liver disease, and the performance of SIRT with sirtuins and / or a disease or condition associated with abnormal activity. 56. A method of inducing apoptosis in a biological system in vitro, the method comprising administering to the system a 5 to 3 nucleotides in length and inverting from a natural antisense transcript of a ΝΑΜρτ polynucleotide An oligonucleotide that is at least 50% identical to 5 to 3 contiguous nucleotides in the complementary sequence. 57. The method of claim 56, wherein the natural antisense transcript has a 9 ID NO: 2 to 16. 58. The method of claim 56, wherein the biological system is a patient cell or tissue. 59. A method of inducing apoptosis in a biological system in vitro, the method comprising administering to the system a nucleotide of about 5 to 3 nucleotides in length, wherein the nucleotide is targeted by a target The gene is upregulated to a natural antisense transcript of the gene of interest, and wherein the oligonucleotide is at least 5% identical to the reverse complement of the natural antisense transcript of the upregulated gene. 60. Use of an oligonucleotide for the manufacture of an agent for inducing apoptosis in a biological system 158213.doc • 13-201210611, wherein the nucleotide is 5 to 30 nucleotides in length The acid is at least 50% identical to 5 to 3 contiguous nucleotides in the reverse complement of the native antisense transcript of the NAMPT polynucleotide. 61. The use of claim 60, wherein the natural antisense transcript has a redundant ID NO: 2 to 16. 62. The use of claim 60, wherein the biological system is a patient cell or tissue. 63. Use of an oligonucleotide for the manufacture of an agent for inducing apoptosis in a biological system, wherein the oligonucleotide is about 5 to 3 nucleotides in length and is targeted by The gene is up-regulated to the natural antisense transcript of the gene of interest, and wherein the oligonucleotide is at least 5% identical to the reverse complement of the native antisense transcript of the up-regulated gene. 64_ A method for identifying and selecting at least one natural antisense transcript for a ΝΑΜρτ gene. Sex as a method for oligonucleotides of a selected target polynucleotide administered in vivo, the method comprising: identifying at least An oligonucleotide comprising at least 5 contiguous nucleotides at least partially complementary to an antisense polynucleotide of the selected polynucleotide of interest; measuring antisense oligonucleotides under stringent hybridization conditions a thermal melting point of the hybrid of the target polynucleotide or the antisense polynucleotide of the selected polynucleotide of interest; and selecting at least one oligonucleoside for in vivo administration based on the obtained reagent acid. 158213.doc -14-
TW100129854A 2010-08-19 2011-08-19 Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT TW201210611A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US37512610P 2010-08-19 2010-08-19

Publications (1)

Publication Number Publication Date
TW201210611A true TW201210611A (en) 2012-03-16

Family

ID=45605669

Family Applications (1)

Application Number Title Priority Date Filing Date
TW100129854A TW201210611A (en) 2010-08-19 2011-08-19 Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT

Country Status (2)

Country Link
TW (1) TW201210611A (en)
WO (1) WO2012024478A2 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9920317B2 (en) 2010-11-12 2018-03-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
DK2638163T3 (en) 2010-11-12 2017-07-24 Massachusetts Gen Hospital POLYCOMB-ASSOCIATED NON-CODING RNAs
EA201492122A1 (en) 2012-05-16 2015-10-30 Рана Терапьютикс, Инк. COMPOSITIONS AND METHODS FOR MODULATING UTRN EXPRESSION
US10174323B2 (en) 2012-05-16 2019-01-08 The General Hospital Corporation Compositions and methods for modulating ATP2A2 expression
KR20150030205A (en) 2012-05-16 2015-03-19 라나 테라퓨틱스, 인크. Compositions and methods for modulating smn gene family expression
AU2013262709A1 (en) 2012-05-16 2015-01-22 Rana Therapeutics, Inc. Compositions and methods for modulating MECP2 expression
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
EP2850188A4 (en) 2012-05-16 2016-01-20 Rana Therapeutics Inc Compositions and methods for modulating hemoglobin gene family expression
KR101599929B1 (en) * 2013-07-29 2016-03-07 광주과학기술원 Screening Methods of a Substance for Preventing or Treating a Joint Disease Using Nampt
CA2921459A1 (en) 2013-08-16 2015-02-19 Rana Therapeutics, Inc. Oligonucleotides targeting euchromatin regions of genes
WO2015051283A1 (en) 2013-10-04 2015-04-09 Rana Therapeutics, Inc. Compositions and methods for treating amyotrophic lateral sclerosis
WO2016070060A1 (en) 2014-10-30 2016-05-06 The General Hospital Corporation Methods for modulating atrx-dependent gene repression
EP3256591A4 (en) 2015-02-13 2018-08-08 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
WO2016149455A2 (en) 2015-03-17 2016-09-22 The General Hospital Corporation The rna interactome of polycomb repressive complex 1 (prc1)
WO2018023206A1 (en) * 2016-07-30 2018-02-08 邦泰生物工程(深圳)有限公司 Nicotinamide phosphoribosyltransferase mutant and application thereof
ES2957464T3 (en) * 2017-04-14 2024-01-19 Univ Arizona Compositions and methods for treating pulmonary fibrosis
KR20220054786A (en) * 2019-06-07 2022-05-03 워싱턴 유니버시티 Production and use of ENP-containing extracellular vesicles
CN114616345A (en) * 2019-08-07 2022-06-10 亚利桑那大学评议会 Single nucleotide polymorphism and use thereof
WO2021212168A1 (en) * 2020-04-20 2021-10-28 Monash University Methods and compositions

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8268575B2 (en) * 2004-09-20 2012-09-18 Washington University NAD biosynthesis systems
WO2007136744A1 (en) * 2006-05-19 2007-11-29 The Johns Hopkins University Crystal structure of a substrate complex of nampt/pbef/visfatin
EP2213288A1 (en) * 2009-01-30 2010-08-04 Karl Welte NAMPT and vitamin B3 for treating or preventing diseases

Also Published As

Publication number Publication date
WO2012024478A3 (en) 2012-05-10
WO2012024478A2 (en) 2012-02-23

Similar Documents

Publication Publication Date Title
JP6259029B2 (en) Treatment of SCNA-related diseases by inhibition of natural antisense transcripts on sodium channels, voltage-gated, alpha subunit (SCNA)
JP6188686B2 (en) Treatment of FXN-related diseases by inhibition of natural antisense transcripts to frataxin (FXN)
TW201210611A (en) Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT
JP6049623B2 (en) Treatment of IDUA-related diseases by inhibition of natural antisense transcripts to α-L-iduronidase (IDUA)
JP5973996B2 (en) Treatment of asymmetric homolog 1 (ATOH1) related diseases by inhibition of natural antisense transcripts against ATOH1
JP5993744B2 (en) Treatment of nuclear respiratory factor 1 related diseases by inhibition of natural antisense transcripts against nuclear respiratory factor 1 (NRF1)
JP5976548B2 (en) Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcripts against PYCR1
JP6025567B2 (en) Treatment of MBTPS1-related diseases by inhibition of the natural antisense transcript against the membrane-bound transcription factor peptidase, site 1 (MBTPS1)
JP6073795B2 (en) Treatment of IFRD1-related diseases by inhibition of natural antisense transcripts to interferon-related developmental regulator 1 (IFRD1)
TWI600759B (en) Treatment of colony-stimulating factor 3 (csf3) related diseases by inhibition of natural antisense transcript to csf3
JP5986998B2 (en) Treatment of NEU4-related diseases by inhibition of natural antisense transcripts to sialidase 4 (NEU4)
JP5886757B2 (en) Treatment of interferon regulatory factor 8 (IRF8) related diseases by inhibition of natural antisense transcripts against interferon regulatory factor 8 (IRF8)
TWI573871B (en) Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
JP6083735B2 (en) Treatment of insulin receptor substrate 2 (IRS2) related diseases by inhibition of natural antisense transcripts against insulin receptor substrate 2 (IRS2) and transcription factor 3 (TFE3)
JP2013516197A (en) Treatment of sex hormone binding globulin (SHBG) related diseases by inhibition of natural antisense transcripts against sex hormone binding globulin (SHBG)
JP2013515488A (en) Treatment of uncoupling protein 2 (UCP2) -related diseases by inhibition of natural antisense transcripts against UCP2.
TW201202418A (en) Treatment of Methionine Sulfoxide Reductase A (MSRA) related diseases by inhibition of natural antisense transcript to MSRA
TW201143782A (en) Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2
TW201206450A (en) Treatment of Discs large homolog (DLG) related diseases by inhibition of natural antisense transcript to DLG
TW201209163A (en) Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3