TW201209163A - Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3 - Google Patents

Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3 Download PDF

Info

Publication number
TW201209163A
TW201209163A TW100124653A TW100124653A TW201209163A TW 201209163 A TW201209163 A TW 201209163A TW 100124653 A TW100124653 A TW 100124653A TW 100124653 A TW100124653 A TW 100124653A TW 201209163 A TW201209163 A TW 201209163A
Authority
TW
Taiwan
Prior art keywords
bbc3
antisense
polynucleotide
nucleotide
oligonucleotide
Prior art date
Application number
TW100124653A
Other languages
Chinese (zh)
Inventor
Joseph Collard
Sherman Olga Khorkova
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 TW201209163A publication Critical patent/TW201209163A/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
    • A61K31/712Nucleic acids or oligonucleotides having modified sugars, i.e. other than ribose or 2'-deoxyribose
    • 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
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • 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
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • C12N15/1135Non-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 oncogenes or tumor suppressor genes
    • 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
    • C12N15/1138Non-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 receptors or cell surface proteins
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Abstract

The present invention relates to antisense oligonucleotides that modulate the expression of and/or function of BCL2 binding component 3 (BBC3), in particular, by targeting natural antisense polynucleotides of BCL2 binding component 3 (BBC3). The invention also relates to the identification of these antisense oligonucleotides and their use in treating diseases and disorders associated with the expression of BBC3.

Description

201209163 六、發明說明: 【發明所屬之技術領域】 本發明實施例包括調節BBC3之表現及/或功能之寡核苷 酸及有關分子。 本申請案主張20 10年7月12日提出申請之美國臨時專利 • 申請案第61/363,535號之優先權,其全部内容皆以引用方 ‘式併入本文中。 【先前技術】 〇 DNA-RNA及RNA-RNA雜交對核酸功能之許多態樣(包含 DNA複製、轉錄及轉譯)較為重要。雜交對於檢測特定核 酸或改變其表現之各種技術亦甚為重要。舉例而言,反義 核苷酸藉由與靶RNA雜交、由此干擾RNA剪接、轉錄、轉 譯及複製來破壞基因表現。反義DNA具有額外特徵,亦即 DNA-RNA雜合體用作核糖核酸酶Η消化之受質,該活性存 在於大部分細胞類型中。反義分子可遞送至細胞中,如募 去氧核苷酸(ODN)之情形,或其可自内源基因表現為RNA 0 分子。FDA最近批准了反義藥物VITRAVENE™(用於治療 巨細胞病毒視網膜炎),此表明該反義藥物具有治療用 途。 / 【發明内容】 提供本概述以呈現本發明之概述從而簡要顯示本發明之 性質及物質。提交本概述係基於下列理解:其並非用於解 釋或限制本申請專利範圍之範疇或含義。 在一個實施例中,本發明提供抑制天然反義轉錄物之作 157514.doc 201209163 用之方法’其係藉由使用靶向天然反義轉錄物之任一區域 的反義寡核苷酸從而上調相應正義基因來達成。本文亦涵 蓋’可藉由siRNA、核糖酶及小分子來抑制天然反義轉錄 物,此視為屬於本發明範鳴内。 一個實施例提供在活體内或活體外調節患者細胞或組織 中BBC3聚核苷酸之功能及/或表現之方法,其包括使該等 細胞或組織與長度為5至3〇個核苷酸之反義寡核苷酸接 觸’其中該寡核苷酸與聚核苷酸之反向補體具有至少5〇〇/〇 之序列一致性’該聚核苷酸包括SEq ID NO: 2中核苷酸1 至689内之5至30個連續核苷酸,由此在活體内或活體外調 節患者細胞或組織中BBC3聚核苷酸之功能及/或表現。 在一實施例中,寡核苷酸靶向BBC3聚核苷酸之天然反 義序列(例如,SEQ ID NO: 2中所述之核苷酸)、及其任一 變體、等位基因、同系物 '突變體、衍生物、片段及互補 序列。將反義募核苷酸之實例闡述為SEq ID NO: 3至8。 另一實施例提供在活體内或活體外調節患者細胞或組織 中BBC3聚核苷酸之功能及/或表現之方法,其包括使該等 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,其中該寡核苷酸與BBC3聚核苷酸反義轉錄物之反向 補體具有至少50°/。的序列一致性;由此在活體内或活體外 調節患者細胞或組織中BBC3聚核苷酸之功能及/或表現。 另一實施例提供在活體内或活體外調節患者細胞或組織 中BBC3聚核苷酸之功能及/或表現之方法,其包括使該等 細胞或組織與長度為5至30個核苷酸之反義寡核苷酸接 157514.doc 201209163 觸’其中該寡核苷酸與BBC3反義聚核苷酸之反義寡核苷 酸具有至少50%的序列一致性;由此在活體内或活體外調 節患者細胞或組織中BBC3聚核苷酸之功能及/或表現。 在一實施例中’級合物包括一或多種結合至正義及/或 • 反義BBC3聚核苷酸之反義寡核苷酸。 在一實施例中,募核苷酸包括一或多個經修飾或取代之 核苷酸。 在一實施例中,募核苷酸包括一或多個經修飾鍵。 〇 . ^ 在又一實施例中,經修飾核苷酸包括含有以下之經修飾 鹼基:硫代碳酸酯、膦酸曱酯、肽核酸、2,·〇_曱基、氟-或碳、亞甲基或其他鎖核酸(LNA)分子。較佳地,經修飾 核苷酸係鎖核酸分子,包含a-L-LNA。 在一實施例中,經皮下、肌内、靜脈内或腹膜腔内將募 核皆酸投與患者。 在一實施例中’以醫藥組合物形式投與募核苷酸。治療 Q 方案包括向患者投與至少一次反義化合物;然而,此治療 可經改變以在一定時間内包含多個劑量。該治療可與一或 多種其他類型之療法組合。 * 在一實施例中,將募核苷酸囊封於脂質體中或附接至載 . 體分子(例如膽固醇、TAT肽)。 其他態樣闡述於下文中。 【實施方式】 下文參照闡釋用實例應用來闡述本發明之若干態樣。應 理解,本文列舉各種具體細節、關係及方法以提:對本: 157514.doc 201209163 明之完全理解。然而,熟習相關技術者易於認識到,可在 不使用-或多個具體細節或使用其他方法之情形下來實踐 本發明。本發明並不限於各個動作或事件之順序,此乃因 -些動作可以不同順序發生及/或與其他動作或事件同時 發生。另彳’未必需要所有所騎動作或事件來實施 明方法。 本文所揭示所有基因、基因名稱及基因產物意欲對應於 可應用本文所揭示組合物及方法之來自任一物種的同系 物。因此,該等術語包含但不限於人類及小鼠之基因及基 因產物。應理解,在揭示來自特定物種之基因或基因產物 時二除非在上下文中明確指*,否則此揭示内容意欲僅具 有實例性,且不應解釋為限制意義。因此,舉例而言,對 於在-些實施财係_於嗜乳動物核酸及胺基酸序列之本 文所揭示基因而5,其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’該等其他動物包含但不限於 其他哺乳動物、魚'兩棲動物、爬行動物及烏。在一實施 例中’基因或核酸序列係人類。 定義 本文所用之術語僅用於闡述特定實施例之目的而並非意 欲限定本發明。如本文中所使用,單數形式「一(a)」、「一 (an)」及「該(the)J亦意欲包含複數形式,除非上下文另 外明確指明。另外,在詳細闡述及/或申請專利範圍中使 用術語「包含(including)」、「包含(includes)」、「具有 (having)」、「具有(has)」、「具有(with)」或其變化形式時, 1575l4.doc 201209163 該等術語意欲以類似於術語「包括(comprising)」之方式 來表示包含範圍。 術語「約」或「近似地」意指熟習此項技術者所測定之 特定值的可接受誤差範圍,其部分地取決於該值之量測或 測定方式,亦即,量測系統之偈限性。舉例而言,根據業 内實踐,「約」可意指在1個或1個以上之標準偏差内。另 一選擇為,「約」可意指與給定值相差至多20%、較佳至 多10%、更佳至多5%、及更佳至多1%的範圍。另一選擇 為,尤其對於生物系統或過程而言,該術語可意指在一數 值之一個數量級内、較佳在5倍内、及更佳在2倍内。在申 請案及申請專利範圍中闡述特定值時,除非另有闡述,否 則應假設術語「約」意指在該特定值之可接受誤差範圍 内。 本文所用之術語「mRNA」意指靶向基因之當前已知 mRNA轉錄物、及可闡釋之任一其他轉錄物。 「反義寡核苷酸」或「反義化合物」意指結合至另一 RNA或DNA(靶RNA、DNA)之RNA或DNA分子。舉例而 言,若其係RNA寡核苷酸,則其藉助RNA-RNA相互作用 結合至另一 RNA靶並改變靶RNA之活性。反義寡核苷酸可 上調或下調特定聚核苷酸之表現及/或功能。該定義意欲 包含自治療、診斷或其他觀點來看有用之任一外來RNA或 DNA分子。該等分子包含(例如)反義RNA或DNA分子、干 擾RNA (RNAi)、微小RNA、誘餌RNA分子、siRNA、酶 RNA、治療性編輯RNA及RNA激動劑與拮抗劑、反義募聚 157514.doc 201209163 化合物、反義寡核苷酸、外部引導序列(EGS)寡核苷酸、 交替剪接體、引物、探針及與靶核酸之至少一部分雜交之 其他寡聚化合物。因此’該等化合物可以單鏈、雙鏈、部 分單鏈或環狀募聚化合物形式引人。 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)之募聚物或聚合物或其模擬 物。術語「寡核苷酸」亦包含天然及/或經修飾單體或鍵 聯之直鏈或環狀寡聚物’包含去氧核糖核苷、核糖核苷、 其經取代及α-變旋異構形式、肽核酸(pna)、鎖核酸 (LNA)、硫代填酸酯、膦酸曱酯及諸如此類。寡核苷酸能 夠藉助單體與單體相互作用之規則模式(例如Watson-Crick 型驗基配對、Ho6gsteen型驗基配對或反向H〇6gsteen型驗 基配對、或諸如此類)特異性結合至乾聚核苷酸。 募核苷酸可為「嵌合」募核苷酸,亦即,由不同區域組 成。在本發明之上下文中,「嵌合」化合物係募核苷酸, 其含有兩個或更多個化學區域,例如DNA區域、RNA區 域、PNA區域等。在募核苷酸化合物之情形下,每一化學 區域係由至少一個單體單元(亦即,核苷酸)組成。該等寡 核苦酸通常包括至少一個對募核苷酸進行修飾以顯示一或 多種期望性質之區域。募核苷酸之期望性質包含但不限於 (例如)增加對核酸酶降解之抗性、增加細胞攝取、及/或增 加對靶核酸之結合親和力。募核苷酸之不同區域可由此具 有不同性質。本發明之嵌合寡核苷酸可形成為兩種或更多 種上述募核苷酸、經修飾募核苷酸、寡核苷及/或寡核苷 157514.doc 201209163 酸類似物的混合結構。 寡核苷酸可由可在「記錄(register)」中連接之區域組 成,亦即,單體係連續連接(如天然DNA中)、或經由間隔 體連接。間隔體意欲在區域之間構成共價「橋」且在較佳 情形下具有不超過約1〇〇個碳原子之長度。間隔體可具有 不同功能,例如具有正電荷或負電荷,具有特異性核酸結 合性質(嵌合劑、槽溝黏合劑、毒素、螢光團等),具有親 脂性,誘導特異性二級結構(例如,誘導〇螺旋之含有丙胺 酸之肽)。 本文所用之「BBC3」及「BCL2結合部分3」包含所有 家族成員、突變體、等位基因、片段、物種、編碼及非編 碼序列、正義及反義聚核苷酸鏈等。 本文所用詞語「BCL2結合部分3」、「3义2_結合部分 3」、BBC3、FLJ42994、JFY1、jfy]、細胞洞亡之 p53 上 調調節劑、PUMA在文獻中被視為相同且在本申請案中互 換使用。 本文所用之術語「對…具有特異性之募核苷酸」或「靶 向…之寡核苷酸」係指具有如下序列之寡核苷酸能 夠與靶向基因之一部分形成穩定複合物,或⑻能夠與: 向基因之mRNA轉錄物的一部分形成穩定雙鏈體。複合物 及雙鏈體之穩定性可藉由理論計算及/或活體外分析^行 測定。測定雜交複合物及雙鏈體之穩定性的實例性分析閣 述於下文實例中。 本文所用之術語「靶核酸」涵蓋DNA、自該DNA轉錄之 157514.doc 201209163 RNA(包括mRNA前體及mRNA)、以及源自該RNA之 cDNA、編碼序列、非編碼序列、正義或反義聚核苷酸。 寡聚化合物與其靶核酸之特異性雜交會干擾該核酸之正常 功能。與靶核酸特異性雜交之化合物對該靶核酸功能的此 調節通常稱為「反義」。擬干擾之DNA功能包含(例如)複 製及轉錄。擬干擾之RNA功能包含所有重要功能,例如, RNA至蛋白質轉譯位點之易位、蛋白質自RNA之轉譯、使 RNA產生一或多種mRNA物質之剪接、及RNA可參與或促 進之催化活性。對於靶核酸功能之該干擾的總體效應係調 節編碼產物或寡核普酸之表現。 RNA干擾(「RNAi」)係藉由與「靶」核酸序列具有序列 特異性同源性之雙鏈RNA(dsRNA)分子來調介。在本發明 某些實施例中,介體係具有5至25個核苷酸之「小干擾」 RNA雙鏈體(siRNA)。siRNA源自稱為Dicer之RNase酶對於 dsRNA之處理。siRNA雙鏈體產物募集至稱為RISC(RNA誘 導之沉默複合物)之多蛋白siRNA複合物中。不期望受限於 任一特定理論,由此據信,RISC可引導至靶核酸(mRNA 較為適宜),在此siRNA雙鏈體以序列特異性方式發生相互 作用從而以催化方式來調介裂解。可用於本發明中之小干 擾RNA可根據業内所熟知且為熟習此項技術者所習知的程 序來合成及使用。用於本發明方法中之小干擾RNA適宜地 包括約1至約50個核苷酸(nt)。在非限制性實施例之實例 中,siRNA可包括約5至約40個nt、約5至約30個nt、約10 至約30個nt、約15至約25個nt、或約20至25個核苷酸。 157514.doc -10- 201209163 藉由使用自動對準核酸序列並指示一致性或同源性區域 之電腦程式來促進適當寡核苷酸的選擇。使用該等程式藉 由(例如)搜索諸如GenBank等數據庫或藉由對PCR產物測 序來比較所獲得的核酸序列。對於來自各種物種之核酸序 列之比較使得可選擇顯示適當物種間一致性程度的核酸序 列。在並未測序之基因情形下,實施南方印跡(Southern blot)以測定靶物種及其他物種之基因間的一致性程度。藉 由在不同嚴格度下實施南方印跡(如業内所熟知),可大致 量測一致性。該等程序使得可選擇如下寡核苷酸:其對欲 控制個體中之靶核酸序列呈現高互補性程度且對其他物種 中之相應核酸序列呈現較低互補性程度。熟習此項技術者 應認識到,可在較大範圍中選擇適用於本發明中之基因區 域。 「酶RNA」意指具有酶活性之RNA分子(〇6(:11,(1988)】· American. Med. Assoc. 260,3030-3035)。酶性核酸(核酶) 藉由首先結合至無RNA而發揮作用。該結合經由酶性核酸 之靶結合部分來進行,該靶結合部分緊鄰分子中用於裂解 靶RNA之酶性部分。因此,酶性核酸首先識別靶RNA且然 後經由鹼基配對與靶RNA結合,且在結合至確切位點後以 酶促方式發揮作用以切割靶RNA。 「誘餌RNA」意指模擬配體之天然結合結構域之RNA分 子。誘餌RNA由此與天然結合靶競爭結合特異性配體。舉 例而言,已顯示HIV反式活化反應(TAR) RNA之過度表現 可用作「誘餌」並有效結合HIV tat蛋白,由此預防其結合 157514.doc • 11 - 201209163 至在HIV RNA中編碼之TAR序列。此意欲係一具體實例β 彼等熟習此項技術者應認識到,此僅為一實例,且可易於 使用業内通常已知之技術來產生其他實施例。 本文所用之術語「單體」通常表示藉由填酸二酯鍵或其 類似物連接以形成尺寸介於數個單體單元(例如,約3_4個) 至約數百個早體早元之券核普酸的单體。碌酸二自旨鍵聯之 類似物包含:硫代碌酸醋、二硫代磷酸醋、膦酸甲醋、栖 代墙酸酯、胺基鱗酸酯及諸如此類,如下文更全面所述。 術語「核苷酸」涵蓋天然核苷酸以及非天然核苷酸。熟 習此項技術者應明瞭,先前視為「非天然」之各種核苷酸 後來已在自然界中發現。因此,「核苷酸」不僅包含含有 已知嗓呤及响咬雜環之分子’且亦包含其雜環類似物及互 變異構體。其他類型核苷酸之闡釋性實例係含有以下部分 之分子: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-Ν6-甲基腺嘌呤、 7-去氮育嗓呤、7-去氣鳥。票呤、Ν4,Ν4-乙醇胞嘴咬、 Ν6,Ν6-乙醇-2,6-二胺基嘌呤、5_曱基胞嘧啶、5_(C3_C6)_ 炔基胞嘧咬、5-氟尿嘧咬、5 ·溴尿嘧咬、假異胞嘧咬、2_ 羥基-5-甲基-4-三唑并吡啶、異胞嘧啶、異鳥嘌呤、肌苷 及Benner等人之美國專利第5,432,272號中所述之「非天 然」核苷酸。術語「核苷酸」意欲涵蓋每一及所有該等實 例以及其類似物及互變異構體。尤其關注之核苷酸係彼等 含有腺嘌呤、鳥嘌呤、胸腺嘧啶、胞嘧啶、及尿嘧啶者, 其被視為與人類之治療及診斷應用有關之天然核苷酸。核 157514.doc -12- 201209163 芽酸包含天然2'-去氧及2'-經基糖(例如,如Kornb erg及 Baker ’ DNA Replication ’ 第 2版(Freeman, San Francisco, 1992)中所述)以及其類似物。 涉及核苷酸之「類似物」包含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)由以下所概述: Scheit, Nucleotide Analogs, John Wiley, New York, 1980 ; Freier & Altmann, (1997) Nucl. Acid. Res., 25(22), 4429-4443, Toulme, J.J., (2001) Nature Biotechnology 19:17-〇 18 ; Manoharan M.,(1999) Biochemica et Biophysica Acta 1489:117-139 » Freier S. M., (1997) Nucl. Acid Research, 25:4429-4443, Uhlman, E.,(2000) Drug Discovery & Development, 3: 203-213, Herdewin P., (2000) Antisense & iVwc/eic dc/c? Drug Dev·, 10:297-310) ; 2'-0、3'-C連接之 [3.2.0]二環阿糖核苷。該等類似物包含經設計以增強結合 性質(例如,雙鏈體或三鏈體穩定性、特異性、或諸如此 類)之合成核苷酸。 〇 本文所用之「雜交」意指寡聚化合物之實質上互補鏈之 配對。一種配對機制涉及寡聚化合物鏈之互補核苷或核苷 - 酸鹼基(核苷酸)之間的氫鍵結,其可為Watson-Crick、201209163 VI. Description of the Invention: TECHNICAL FIELD OF THE INVENTION Embodiments of the invention include oligonucleotides and related molecules that modulate the performance and/or function of BBC3. The present application claims priority to U.S. Provisional Patent Application Serial No. 61/363,535, filed on Jan. 12, 2011, the entire disclosure of which is incorporated herein by reference. [Prior Art] 〇 DNA-RNA and RNA-RNA hybridization are important for many aspects of nucleic acid function, including DNA replication, transcription and translation. Hybridization is also important for the detection of specific nucleic acids or the various techniques that alter their performance. For example, antisense nucleotides disrupt gene expression by hybridizing to a target RNA, thereby interfering with RNA splicing, transcription, translation, and replication. Antisense DNA has the additional feature that DNA-RNA hybrids are used as substrates for ribonuclease digestion, which is present in most cell types. The antisense molecule can be delivered to a cell, such as in the case of an oxygen donating nucleotide (ODN), or it can be expressed as an RNA 0 molecule from an endogenous gene. The FDA recently approved the antisense drug VITRAVENETM (for the treatment of cytomegalovirus retinitis), indicating that the antisense drug has therapeutic utility. BRIEF DESCRIPTION OF THE DRAWINGS This summary is provided to introduce an overview of the invention This Summary is submitted with the understanding that it is not intended to be construed as limiting or limiting the scope or meaning of the scope of the application. In one embodiment, the invention provides a method for inhibiting a natural antisense transcript of 157514.doc 201209163, which is upregulated by using an antisense oligonucleotide that targets any region of a natural antisense transcript The corresponding justice gene is reached. It is also encompassed herein to inhibit natural antisense transcripts by siRNA, ribozymes and small molecules, which are considered to be within the scope of the present invention. One embodiment provides a method of modulating the function and/or expression of a BBC3 polynucleotide in a patient's cells or tissues in vivo or in vitro, comprising subjecting the cells or tissues to 5 to 3 nucleotides in length. The antisense oligonucleotide contacts 'where the oligonucleotide has a sequence identity of at least 5 〇〇/〇 with the reverse complement of the polynucleotide'. The polynucleotide comprises SEq ID NO: 2 nucleotide 1 Up to 5 to 30 contiguous nucleotides within 689, thereby modulating the function and/or performance of BBC3 polynucleotides in a patient's cells or tissues in vivo or in vitro. In one embodiment, the oligonucleotide targets a natural antisense sequence of a BBC3 polynucleotide (eg, the nucleotide set forth in SEQ ID NO: 2), any variant thereof, allele, Homologs 'mutants, derivatives, fragments and complementary sequences. An example of an antisense raised nucleotide is set forth as SEq ID NO: 3 to 8. Another embodiment provides a method of modulating the function and/or expression of a BBC3 polynucleotide in a cell or tissue of a patient in vivo or in vitro, comprising subjecting the cell or tissue to a length of 5 to 30 nucleotides The antisense oligonucleotide is contacted, wherein the oligonucleotide has a reverse complement of at least 50° with the BBC3 polynucleotide antisense transcript. Sequence identity; thereby modulating the function and/or performance of BBC3 polynucleotides in a patient's cells or tissues in vivo or in vitro. Another embodiment provides a method of modulating the function and/or expression of a BBC3 polynucleotide in a cell or tissue of a patient in vivo or in vitro, comprising subjecting the cell or tissue to a length of 5 to 30 nucleotides Antisense oligonucleotides 157514.doc 201209163 Touching wherein the oligonucleotide has at least 50% sequence identity with an antisense oligonucleotide of a BBC3 antisense polynucleotide; thereby in vivo or in vivo Externally regulates the function and/or performance of BBC3 polynucleotides in a patient's cells or tissues. In one embodiment, the 'amplifier includes one or more antisense oligonucleotides that bind to sense and/or antisense BBC3 polynucleotides. In one embodiment, the raised nucleotide comprises one or more modified or substituted nucleotides. In one embodiment, the nucleotide is raised to include one or more modified linkages.又一. ^ In yet another embodiment, the modified nucleotide comprises a modified base comprising: thiocarbonate, decyl phosphonate, peptide nucleic acid, 2, 〇 曱 thiol, fluoro- or carbon, Methylene or other locked nucleic acid (LNA) molecules. Preferably, the modified nucleotide is a nucleic acid molecule comprising a-L-LNA. In one embodiment, the nucleophilic acid is administered to the patient subcutaneously, intramuscularly, intravenously or intraperitoneally. In one embodiment, the nucleotides are administered in the form of a pharmaceutical composition. Therapeutic Q regimen comprises administering to the patient at least one antisense compound; however, the treatment can be altered to include multiple doses over a period of time. The treatment can be combined with one or more other types of therapies. * In one embodiment, the nucleotide is encapsulated in a liposome or attached to a carrier molecule (eg, cholesterol, TAT peptide). Other aspects are set forth below. [Embodiment] Several aspects of the present invention are explained below by way of example application with reference to the explanation. It should be understood that various specific details, relationships, and methods are set forth herein to provide a full understanding of the present invention: 157514.doc 201209163. However, it will be readily apparent to those skilled in the art that the present invention may be practiced without the use of the specific details or the use of other methods. The present invention is not limited to the order of the various acts or events, which may occur in a different order and/or concurrent with other acts or events.彳' does not necessarily require all riding actions or events to implement the method. All genes, gene names, and gene products disclosed herein are intended to correspond to homologs from any species to which the compositions and methods disclosed herein can be applied. Thus, such terms include, but are not limited to, human and mouse genes and gene products. It will be understood that the disclosure of a gene or gene product from a particular species, unless explicitly stated in the context, is intended to be exemplary only and should not be construed as limiting. Thus, for example, for the implementation of the genes disclosed herein in the context of a mammalian nucleic acid and amino acid sequence, 5 is intended to encompass homologous and/or orthologous genes and genes from other animals. Products 'These other animals include, but are not limited to, other mammals, fish 'amphibians, reptiles, and blacks. In one embodiment the 'gene or nucleic acid sequence is human. DEFINITIONS The terminology used herein is for the purpose of describing particular embodiments and is not intended to limit the invention. The singular forms "a", "an" and "the" are also intended to include the plural unless the context clearly dictates otherwise. When the terms "including", "includes", "having", "has", "with" or variations thereof are used in the scope, 1575l4.doc 201209163 The term is intended to mean a range of inclusions in a manner similar to the term "comprising". The term "about" or "approximately" means an acceptable range of error for a particular value determined by those skilled in the art, which depends in part on the manner in which the value is measured or measured, that is, the limits of the measurement system. Sex. For example, according to industry practice, "about" can mean within one or more standard deviations. Alternatively, "about" can mean a range that differs from a given value by up to 20%, preferably by up to 10%, more preferably by up to 5%, and even more preferably by up to 1%. Alternatively, especially for biological systems or processes, the term may mean within an order of magnitude, preferably within 5 times, and more preferably within 2 times of a numerical value. In the case of specific values stated in the scope of the application and the patent application, unless otherwise stated, the term "about" is intended to mean within the acceptable tolerance of the particular value. The term "mRNA" as used herein refers to a currently known mRNA transcript of a targeted 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 RNA-RNA interaction and alters the activity of the target RNA. Antisense oligonucleotides can upregulate or downregulate the performance and/or function of a particular polynucleotide. This definition is intended to encompass any foreign RNA or DNA molecule that is useful from a therapeutic, diagnostic or other point of view. Such molecules include, for example, antisense RNA or DNA molecules, interfering RNA (RNAi), microRNAs, decoy RNA molecules, siRNA, enzyme RNA, therapeutically editable RNA and RNA agonists and antagonists, antisense recruitment 157514. Doc 201209163 Compounds, antisense oligonucleotides, external leader sequence (EGS) oligonucleotides, alternative splicing bodies, primers, probes, and other oligomeric compounds that hybridize to at least a portion of a target nucleic acid. Thus, such compounds may be introduced as single-stranded, double-stranded, partially single-stranded or cyclic polymeric compounds. In the context of the present invention, the term "oligonucleotide" refers to a polymer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or a mimetic thereof. The term "oligonucleotide" also encompasses natural and/or modified monomers or linked linear or cyclic oligomers comprising deoxyribonucleosides, ribonucleosides, substituted and alpha-variable Configuration forms, peptide nucleic acids (pna), locked nucleic acids (LNA), thiolates, decyl phosphonates, and the like. Oligonucleotides can specifically bind to the stem by a regular pattern of monomer-to-monomer interactions (eg, Watson-Crick-type base pairing, Ho6gsteen-type base pairing, or reverse H〇6gsteen type base pairing, or the like) Polynucleotide. The nucleotides raised may be "chimeric" nucleotides, i.e., composed of different regions. In the context of the present invention, a "chimeric" compound is a nucleotide that contains two or more chemical regions, such as a DNA region, an RNA region, a PNA region, and the like. In the case of a nucleotide compound, each chemical region is composed of at least one monomer unit (i.e., nucleotide). Such oligonucleotides typically include at least one region that is modified to exhibit one or more desired properties. Desirable properties of nucleotides include, but are not limited to, for example, increasing resistance to nuclease degradation, increasing cellular uptake, and/or increasing binding affinity for a target nucleic acid. Different regions of the raised nucleotides can thus have different properties. The chimeric oligonucleotide of the present invention can be formed into a mixed structure of two or more of the above-mentioned nucleotides, modified nucleotides, oligonucleosides and/or oligonucleosides 157514.doc 201209163 acid analogs. . Oligonucleotides can be composed of regions that can be joined in a "register", i.e., a single system of continuous linkages (e. g., in native DNA), or via a spacer. The spacers are intended to form a covalent "bridge" between the regions and, in the preferred case, have a length of no more than about one carbon atom. The spacers can have different functions, such as having a positive or negative charge, having specific nucleic acid binding properties (chimeric agents, groove binders, toxins, fluorophores, etc.), having lipophilic properties, and inducing specific secondary structures (eg, Inducing a peptide of alanine containing a helix. As used herein, "BBC3" and "BCL2 binding part 3" encompass all family members, mutants, alleles, fragments, species, coding and non-coding sequences, sense and antisense polynucleotide chains, and the like. As used herein, the terms "BCL2 binding moiety 3", "3 sense 2_binding moiety 3", BBC3, FLJ42994, JFY1, jfy], p53 up-regulators of cell death, PUMA are considered identical in the literature and are in this application Used interchangeably in the case. The term "nucleotide specific for" or "targeted oligonucleotide" as used herein refers to an oligonucleotide having the following sequence capable of forming a stable complex with a portion of a targeted gene, or (8) It is possible to form a stable duplex with a part of the mRNA transcript of the gene. The stability of the complexes and duplexes can be determined by theoretical calculations and/or in vitro analysis. An example analysis for determining the stability of hybridization complexes and duplexes is set forth in the Examples below. The term "target nucleic acid" as used herein encompasses DNA, 157514.doc 201209163 RNA (including mRNA precursors and mRNA) transcribed from the DNA, and cDNA, coding sequences, non-coding sequences, sense or antisense aggregates derived from the RNA. Nucleotide. The specific hybridization of an oligomeric compound to its target nucleic acid interferes with the normal function of the nucleic acid. This modulation of the function of the target nucleic acid by a compound that specifically hybridizes to the target nucleic acid is commonly referred to as "antisense." The DNA function to be interfered with, for example, replication and transcription. The RNA function to be interfered with encompasses all important functions, such as translocation of RNA to protein translation sites, translation of proteins from RNA, splicing of RNA 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 to modulate the expression of the encoded product or oligonucleotide. RNA interference ("RNAi") is mediated by double-stranded RNA (dsRNA) molecules with sequence-specific homology to the "target" nucleic acid sequence. In certain embodiments of the invention, the mediator system has a "small interference" RNA duplex (siRNA) of 5 to 25 nucleotides. siRNA is derived from the treatment of dsRNA by an RNase enzyme called Dicer. The siRNA duplex product is recruited into a polyprotein siRNA complex called RISC (RNA-induced silencing complex). Without wishing to be bound by any particular theory, it is believed that RISC can be directed to a target nucleic acid (mRNA is preferred) in which the siRNA duplex interacts in a sequence-specific manner to catalyze cleavage in a catalytic manner. Small interfering RNAs useful in the present invention can be synthesized and used according to procedures well known in the art and well known to those skilled in the art. Small interfering RNAs for use 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 siRNA can comprise from about 5 to about 40 nt, from about 5 to about 30 nt, from about 10 to about 30 nt, from about 15 to about 25 nt, or from about 20 to 25 Nucleotides. 157514.doc -10- 201209163 Promote the selection of appropriate oligonucleotides by using a computer program that automatically aligns nucleic acid sequences and indicates regions of identity or homology. These programs are used to compare the obtained nucleic acid sequences by, for example, searching a database such as GenBank or by sequencing the PCR products. Comparison of nucleic acid sequences from various species allows selection of nucleic acid sequences that show the degree of agreement between appropriate species. In the case of unsequenced genes, Southern blots were performed to determine the degree of agreement between genes of the target species and other species. Consistency can be roughly measured by performing Southern blots (as is well known in the industry) with varying degrees of stringency. Such procedures allow for the selection of oligonucleotides that exhibit a high degree of complementarity to the target nucleic acid sequence in the individual to be controlled and a lower degree of complementarity to the corresponding nucleic acid sequences in other species. Those skilled in the art will recognize that a wide range of gene regions suitable for use in the present invention can be selected. "Enzyme RNA" means an RNA molecule having enzymatic activity (〇6(:11,(1988)]·American. Med. Assoc. 260, 3030-3035). Enzymatic nucleic acid (ribozyme) by first binding to none RNA acts. The binding is carried out via a target binding portion of an enzymatic nucleic acid that is immediately adjacent to the enzymatic portion of the molecule used to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes the target RNA and then via base pairing Binding to a target RNA and acting in an enzymatic manner to cleave the target RNA after binding to the exact site. "Bee RNA" means an RNA molecule that mimics the natural binding domain of the ligand. The decoy RNA is thereby bound to the natural target. Competition for specific ligands. For example, overexpression of HIV transactivation (TAR) RNA has been shown to be useful as a "bait" and to effectively bind HIV tat protein, thereby preventing its binding. 157514.doc • 11 - 201209163 To TAR sequences encoded in HIV RNA. This is intended to be a specific example of β. Those skilled in the art will recognize that this is only an example, and that other embodiments can be readily made using techniques generally known in the art. This article The term "monomer" as used herein generally means attached by a diester bond or an analog thereof to form a nucleus having a size ranging from a plurality of monomer units (for example, about 3-4) to about several hundreds of early aging. The monomer of the acid is composed of: thioglycolic acid vinegar, dithiophosphoric acid vinegar, phosphonic acid methyl vinegar, benzoic acid ester, amino squarate, and the like, as follows The term "nucleotide" encompasses both natural and non-natural nucleotides. It should be understood by those skilled in the art that various nucleotides previously considered "unnatural" have subsequently been found in nature. Therefore, "nucleotide" includes not only molecules containing known oxime and ringing heterocyclic rings but also heterocyclic analogs and tautomers. Interpretive examples of other types of nucleotides contain the following Molecules: adenine, guanine, thymine, cytosine, uracil, guanidine, xanthine, diamino guanidine, 8-oxo-purine 6-methyl adenine, 7-azepine, 7 - Degassing birds. Tickets, Ν4, Ν4-ethanol squirting, Ν6, Ν6-ethanol-2,6-two Base, 5_mercaptocytosine, 5_(C3_C6)_ alkynyl cytosine, 5-fluorouracil, 5 · bromoacetidine, pseudo-isocytosine, 2_hydroxy-5-methyl-4 - "Non-natural" nucleotides as described in U.S. Patent No. 5,432,272 to Benner et al. The term "nucleotide" is intended to cover each and every Such examples, as well as analogs and tautomers thereof, are of particular interest in nucleotides containing adenine, guanine, thymine, cytosine, and uracil, which are considered therapeutic and diagnostic in humans. Apply the relevant natural nucleotides. Nucleus 157514.doc -12- 201209163 Bucic acid contains natural 2'-deoxy and 2'-transglycosides (for example, as described in Kornb erg and Baker 'DNA Replication' 2nd Edition (Freeman, San Francisco, 1992) ) and its analogues. An "analog" involving a nucleotide comprises a synthetic nucleotide having a modified base moiety and/or a modified sugar moiety (see, for example, as outlined below: Scheit, Nucleotide Analogs, John Wiley, New York, 1980) Freier & Altmann, (1997) Nucl. Acid. Res., 25(22), 4429-4443, Toulme, JJ, (2001) Nature Biotechnology 19:17-〇18; Manoharan M., (1999) Biochemica et Biophysica Acta 1489: 117-139 » Freier SM, (1997) Nucl. Acid Research, 25: 4429-4443, Uhlman, E., (2000) Drug Discovery & Development, 3: 203-213, Herdewin P., ( 2000) Antisense & iVwc/eic dc/c? Drug Dev., 10:297-310); 2'-0, 3'-C linked [3.2.0] bicyclic arabinose. Such analogs comprise synthetic nucleotides designed to enhance binding properties (e.g., duplex or triplex stability, specificity, or the like). "Hybridization" as used herein means the pairing of substantially complementary strands of an oligomeric compound. A pairing mechanism involves hydrogen bonding between complementary nucleosides or nucleoside-acid bases (nucleotides) of an oligomeric compound chain, which can be Watson-Crick,

HoSgsteen或反向Hodgsteen氫鍵結。舉例而言,腺嗓吟及 胸腺嘧啶係經由形成氫鍵配對之互補核苷酸。雜交可在不 同情形下發生。 在以下情形時反義化合物係「可特異性雜交」:化合物 與靶核酸之結合可干擾靶核酸之正常功能以調節功能及/ 157514.doc -13- 201209163 或活性,且存在足夠互補性程度以避免反義化合物與_ 核酸序列在期望發生特異性結合之條件下(亦即,在 内分析或治療性治療情形中之生理條件下,及在活體外八 析情形中實施分析之條件下)發生非特異性結合。 77 本文所用之片語「嚴格雜交條件」&「嚴格條件」係指 本發明化合物與其乾序列發生雜交、但與最少數量之^ 序列發生雜交的條件。嚴格條件具有序列依賴性且在; 情況及本發明上下文中有所不同,藉由寡聚化合物之性質 独成騎究其之分析來測定寡聚化合物與乾序列進行雜 交的「嚴格條件」。一般而言,嚴格雜交條件包括低濃度 (<0·15Μ)含有無機陽離子(例如Na+或κ+)之鹽(亦即,低離 :強度)’溫度高於抓至抑但低於寡聚化合物:乾序列 锼合物之Tm,及存在變性劑(例如曱醯胺、二甲基甲醯 胺、二F基亞硬)或洗務劑十二院基硫酸納(SDS)。舉例而 言’對於每一 1%甲醯胺而言’ _交速率降低1.1%。高嚴 袼度雜交條件之實例係〇.1χ氣化鈉_檸檬酸鈉緩衝液 (ssc)/o.i%(w/v)SDS(在60t 下,保持3〇分鐘)。 本文所用之「互補」係指一個或兩個募聚鏈上之兩個核 普酸之間精確配對的能力。舉例而言,若反義化合物某—Λ 位置處之核鹼基能夠與靶核酸(該靶核酸係DNA、RNA、 或寡核苷酸分子)某一位置處之核鹼基發生氫鍵結,則該 养核苷酸與該靶核酸之間發生氫鍵結之位置視為互補位 置。在每一分子中有足夠數量之互補位置由可彼此氫鍵結 之核苷酸佔據時,寡聚化合物及其他DNA、RNA、或募核 157514.doc 14 201209163 苷酸分子彼此互補。因此,「可特異性雜交」及「互補」 係用於指示以下情形之術語:足夠數量之核苷酸中具有足 夠程度之精確配對或互補性從而寡聚化合物及靶核酸之間 發生穩定及特異性結合。 業内應理解,寡聚化合物序列無需與擬特異性雜交之其 靶核酸序列100%互補。另外,寡核苷酸可在一或多個區 段中雜交從而插入或相鄰區段並不參與雜交事件(例如, 環路結構、失配或髮夾結構)。本發明寡聚化合物包括與 〇 其所靶向靶核酸序列内靶區域之至少約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%的序列互補性。舉例而言,反義 化合物之20個核苷酸中有1 8個與靶區域互補且由此特異性 雜交之反義化合物將代表90%的互補性。在此實例中,剩 餘非互補核苷酸可與互補核苷酸群集或散開且無需彼此鄰 近或與互補核苷酸鄰近。因此,長度為1 8個核苷酸且具有 4(四)個非互補核苷酸(由兩個與靶核酸完整互補之區域側 ❹ 接)之反義化合物與靶核酸具有77.8%之總體互補性且由此 屬於本發明範疇内。具有靶核酸區域之反義化合物的互補 - 性百分比通常可使用業内已知之BLAST程式(鹼基局部對 準檢索工具)及PowerBLAST程式測得。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, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.) 使用默認設置(其使用Smith及Waterman算法)測得(Adv. 157514.doc -15- 201209163HoSgsteen or reverse Hodgsteen hydrogen bonding. For example, adenine and thymine are complementary nucleotides that form a hydrogen bond pair. Hybridization can occur in different situations. An antisense compound is "specifically hybridizable" when the binding of the compound to the target nucleic acid interferes with the normal function of the target nucleic acid to modulate function and / 157514.doc -13 - 201209163 or activity, and is sufficiently complementary to the extent Avoiding the occurrence of antisense compounds and _ nucleic acid sequences under conditions where it is desired to specifically bind (ie, under physiological conditions in the context of internal analysis or therapeutic treatment, and under conditions of analysis in an in vitro analytic situation) Non-specific binding. 77. The phrase "stringent hybridization conditions" & "stringent conditions" as used herein refers to conditions under which a compound of the invention hybridizes to its dry sequence but hybridizes to a minimum number of sequences. Stringent conditions are sequence dependent and differ in the context of the present invention and the context of the present invention, and the "stringent conditions" for hybridization of oligomeric compounds to dry sequences are determined by the analysis of the nature of the oligomeric compounds. In general, stringent hybridization conditions include low concentrations (<0.15 Å) of salts containing inorganic cations (e.g., Na+ or κ+) (i.e., low separation: strength). The temperature is higher than the capture but lower than the oligomerization. Compound: Tm of the dry sequence conjugate, and the presence of a denaturant (such as guanamine, dimethylformamide, bis-F-based hard) or a detergent 12-sodium sulfate (SDS). For example, the '- exchange rate was reduced by 1.1% for each 1% metformin. An example of high stringency hybridization conditions is 〇.1 χ gasified sodium _ sodium citrate buffer (ssc) / o.i% (w / v) SDS (at 60 Torr, maintained for 3 〇 minutes). As used herein, "complementary" refers to the ability to precisely pair between two nucleotides on one or both of the clusters. For example, if the nucleobase at the position of the antisense compound is capable of hydrogen bonding to a nucleobase at a position of the target nucleic acid (the target nucleic acid DNA, RNA, or oligonucleotide molecule), Then, the position at which hydrogen bonding occurs between the nutrient nucleotide and the target nucleic acid is regarded as a complementary position. The oligomeric compound and other DNA, RNA, or nucleoside molecules are complementary to each other when a sufficient number of complementary positions in each molecule are occupied by nucleotides that are hydrogen-bondable to each other. Thus, "specifically hybridizable" and "complementary" are used to indicate a term for a sufficient number of nucleotides to have a sufficient degree of exact pairing or complementarity to provide stability and specificity between the oligomeric compound and the target nucleic acid. Sexual union. It is understood in the art that the oligomeric compound sequence need not be 100% complementary to its target nucleic acid sequence to be specifically hybridized. In addition, oligonucleotides can hybridize in one or more regions such that insertions or adjacent segments do not participate in hybridization events (e.g., loop structures, mismatches, or hairpin structures). 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, of the target region within the target nucleic acid sequence to which it is targeted. Approximately 95%, or at least about 99%, of sequence complementarity. For example, 18 of the 20 nucleotides of the antisense compound are complementary to the target region and the antisense compound that specifically hybridizes will represent 90% complementarity. In this example, the remaining non-complementary nucleotides can be clustered or dispersed with complementary nucleotides and need not be adjacent to each other or adjacent to the complementary nucleotides. Thus, an antisense compound having a length of 18 nucleotides and having 4 (four) non-complementary nucleotides (which are flanked by two regions complementary to the target nucleic acid) has 77.8% overall complementarity to the target nucleic acid. And thus fall within the scope of the invention. The percent complementarity of antisense compounds having a target nucleic acid region can generally be measured using the BLAST program (base 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, the Gap program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.) using default settings (which use Smith and Waterman algorithm) measured (Adv. 157514.doc -15- 201209163

Appl. Math·,(1981) 2, 482-489)。 本文所用之術語「熱熔點」係指在界定離子強度、 PH及核酸濃度下與靶序列互補之寡核苷酸中之5〇%在平衡 下與靶序列雜交的溫度。通常,對於短寡核苷酸(例如, 具有10至50個核苷酸)而言,嚴格條件係彼等以下條件: 鹽濃度在?1^7.0至8.3下為至少約0.01至1_〇河之]^&離子濃 度(或其他鹽)且溫度為至少約3(rc。嚴格條件亦可藉由添 加諸如甲醯胺等去穩定劑來達成。 本文所用之「調節」意指增加(刺激)或降低(抑制)基因 之表現。 在用於聚核苷酸序列之背景中時,術語「變體」可涵蓋 與野生型基因有關之聚核苷酸序列。此定義亦可包含(例 如)「等位基因」、「剪接」、「物種」或「多態性」變體。 剪接變體可與參考分子具有顯著一致性,但在mRNA加工 期間因外顯子之交替剪接而通常具有較大或較小數量之聚 核苷酸。相應多肽可擁有額外功能結構域或不存在結構 域。物種變體係在物種之間有所變化之聚核苷酸序列。在 本發明中尤其有用者係野生型基因產物之變體。變體可源 自核酸序列中之至少一個突變,且可產生改變2mRNA或 產生結構或功能可改變或不改變的多肽。任—給定天然或 重組基因可不具有等位基因形式、具有一種或許多種等位 基因形式。產生變體之常見突變變化通常歸因於核苷酸之 天然缺失、添加或取代。該等變化類型中之每一者可在給 疋序列令單獨、或與其他者組合發生一或多次。 157514.doc -16 - 201209163 所得多肽通常彼此之間具有顯著之胺基酸一致性。多態 性變體係給定物種個體間特定基因之聚核苷酸序列中的變 化。多態性變體亦可涵蓋「單一核苷酸多態性」(SNp)或 單驗基突變,其中聚核苷酸序列之一個鹼基有所變化。 SNP之存在可指示(例如)某一群體具有疾病狀態傾向(亦即 相對於抗性之易感性)。 衍生聚核苷酸包含經受化學修飾(例如,氫由烷基、酿 基或胺基代替)之核酸。衍生物(例如,衍生寡核苷酸)可包 括非天然部分,例如改變之糖部分或糖間鍵聯。該等實例 性街生物係硫代磷酸酯及業内已知之其他含硫物質。衍生 核酸亦可含有標記,包含放射性核苷酸、酶、螢光劑、化 學發光劑、發色劑、受質、輔因子、抑制劑、磁性顆粒及 諸如此類。 「衍生」多肽或肽係(例如)藉由以下方式進行修飾者: 糖基化、聚乙二醇化、磷醯化、硫酸化、還原/烷基化、 酿基化、化學偶合或輕度福馬林(formalin)處理。衍生物 亦可經修飾以含有可檢測標記(直接或間接),包含但不限 於放射性同位素、螢光及酶標記。 本文所用之術語Γ動物」或「患者」意欲包含(例如)人 類、綿羊、麋鹿、鹿、長耳鹿、水紹、,甫乳動物、猴子、 馬、牛、豬、山羊、狗、貓、大鼠、小鼠、鳥'雞、爬行 動物、魚、昆蟲及蜘蛛。 哺乳動物」涵蓋通常處於醫學護理下之溫血哺乳動物 (例如,人類及家養動物)。實例包含貓、犬、馬、牛、及 157514.doc -17- 201209163 人類、以及僅指人類。 「治療(treating或treatment)」涵蓋對哺乳動物之疾病狀 態之治療,且包含:⑷在哺乳動物中、特定而言在該哺乳 動物易患有疾病狀態但尚未被診斷出患有該疾病狀態時預 防該疾病狀態發生;(b)抑制疾病狀態,例如阻止其發展; 及/或(c)減緩疾病狀態,例如使疾病狀態減退直至達到期 望端點為止。治療亦包含改善疾病症狀(例如,減輕疼痛 或不適),其中該改善可直接影響或可能並不直接影響疾 病(例如,起因、傳染、表現等)。 本文所用之「癌症」係指在哺乳動物中發現之所有類型 癌症或贅瘤或惡性腫瘤,其包含但不限於··白血病、淋巴 瘤、黑素瘤、癌及肉瘤。癌症自身表現為包括癌症之惡性 細胞之「腫瘤」或組織。腫瘤之實例包含肉瘤及癌,例如 但不限於:纖維肉瘤、黏液肉瘤、脂肪肉瘤、軟骨肉瘤、 成骨性肉瘤、脊索瘤、血管肉瘤、内皮肉瘤、淋巴管肉 瘤、淋巴管内皮肉瘤、滑膜瘤、間皮瘤、尤因氏腫瘤 (Ewing’s tumor)、平滑肌肉瘤、橫紋肌肉瘤、結腸癌、胰 腺癌、乳癌、卵巢癌、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、髓樣癌、枝氣管原癌、腎細胞癌、肝細胞瘤、膽 管癌、絨毛膜癌、精原細胞瘤、胚胎性癌、維爾姆斯氏腫 瘤(Wilms,tumor)、宮頸癌、睪丸腫瘤、肺癌、小細胞肺 癌、膀胱癌、上皮癌、膠質瘤、星形細胞瘤、髓母細胞 瘤、顱咽管瘤、室管膜瘤、松果體瘤、血管母細胞瘤、聽 157514.doc -18- 201209163 神經瘤、少突神經膝質瘤、腦膜瘤、黑素瘤、神經母細胞 瘤及視網膜母細胞瘤。可藉由本發明所揭示組合物治療之 其他癌症包含但不限於(例如)霍奇金氏病(Hodgkin’s DiSeaSe)、非霍奇金氏淋巴瘤(N〇n-H〇dgkin,s Lymphoma)、 夕發陡月题瘤、神經母細胞瘤、乳癌、印巢癌、肺癌、橫 、’文肌肉0原發性血小板增多症、原發性巨球蛋白血症、 小細胞肺腫瘤、原發性腦腫瘤、胃癌、結腸癌、惡性胰腺 肤島素瘤、惡性類癌、膀胱癌、胃癌 '惡化前皮膚損傷、 睪丸癌淋巴瘤、甲狀腺癌、神經母細胞瘤、食管癌、泌 尿生殖道癌症、惡性高鈣血症、宮頸癌、子宮内膜癌症、 腎上腺皮質癌症及前列腺癌。 本文所用之「神經疾病或病症」係指神經系統及/或視 覺系統之任一疾病或病症。「神經疾病或病症」包含涉及 中樞神經系統(腦、腦幹及小腦)、周邊神經系統(包含顱神 經)、及自主神經系統(位於十樞及周邊神經系統二者中之 °卩分)之疾病或病症。神經疾病或病症包含但不限於後天 性癲癇失語症;急性播散性腦脊髓炎;腎上腺腦白質營養 不良;老年性黃斑退化症;胼胝體發育不全;失認症;艾 卡迪症候群(Aicardi syndrome);亞歷山大病(Aiexander disease),阿爾珀斯病(Aipers’ disease);交叉性肢體癱 瘓,阿爾茨海默氏病(Alzheimer’s disease);血管性癡呆; 肌萎縮側索硬化;無腦畸形;天使症候群(Angelrnan syndrome);血管瘤病;缺氧症;失語症;失用症;蛛網 膜囊腫;蛛網膜炎;阿-蔡二氏畸形(Anr〇nl_Chiari 157514.doc -19- 201209163 malf〇rmati〇n);㈣脈畸形;阿斯佩格症候群(Asperger synd_e);運動失調性毛細血管擴張症;注意力缺陷伴 多動病症’自閉症’自主神經功能障礙;背痛;巴登氏病 (Batten disease) ’貝切特氏病化似❹);貝爾麻痹 (Bells palsy),良性本質瞼痙攣;良性局部肌萎縮;良性 顧内同壓,賓斯旺格病(Binswanger,s化⑽叫;眼瞼痙 擎’布洛克-蘇茲貝克症候群(Bloch Sulzberger syndrome),臂叢神經損傷;腦膿腫;腦損傷;腦腫瘤(包 含多形性膠質母細胞瘤);脊髓腫瘤;布朗·塞卡爾症候群 (Brown-Sequard Syndrome);卡納萬病(Canavan disease); 腕道症候群;灼性神經痛;中樞性疼痛症候群;腦橋中央 髓鞘溶解;頭部病症;腦動脈瘤;腦動脈硬化症;大腦萎 縮,大腦性巨人症;大腦性麻痹;夏-馬-圖三氏病 (Charcot-Made-Tooth disease);化學療法誘導性神經病變 及神經性疼痛’恰裏畸形(Chiari malformation);舞蹈病; 慢性炎症性脫髓鞘性多發性神經病變;慢性疼痛;慢性區 域性疼痛症候群;科-勒二氏症候群(c〇ffin L〇wry syndrome);昏迷,包含持續性植物人狀態;先天性面 癱;皮質基底退化;顱動脈炎;顱縫早閉;克雅氏病 (Creutzfeldt-Jakob disease);積累性創傷病症,·庫興氏症 候群(Cushing’s syndrome);巨細胞包涵體病;巨細胞病毒 感染;舞蹈眼-舞蹈足症候群;丹-沃二氏症候群 (DandyWalker syndrome);道森病(Dawson disease);德摩 西埃症候群(De Morsier’s syndrome);克隆普克-克隆普克 157514.doc -20- 201209163 症候群(Dejerine-Klumke palsy);癡呆;皮肌炎;糖尿病 神經病變;彌漫性硬化;自主神經機能異常;書寫困難; 誦讀困難;張力失常;早期幼兒癲癇性腦病;空蝶鞍症候 群;腦炎;腦疝;腦三叉神經血管瘤病;癲癇症;歐勃麻 痛 (Erb's palsy);特發性震顫;法布裏病(Fabry's disease);法爾症候群(Fahr's syndrome);昏厥;家族性痙 攣性癱瘓;發熱性驚厥;菲希爾症候群(Fisher syndrome);弗裏德賴希共濟失調症(Friedreich's ataxia); 額顳骨癡呆症及其他「tau病變」;高歇氏病(Gaucher's disease);格斯特曼症候群(Gerstmann’s syndrome);巨細 胞動脈炎;巨細胞性包涵體病;球樣細胞腦白質營養不 良;格-巴二氏症候群(Guillain-Barre syndrome) ; HTLV-1 有關脊髓病;哈-斯二氏病(Hallervorden-Spatz disease); 頭部損傷;頭痛;半面痙攣;遺傳性痙攣性截瘫;遺傳病 性多神經炎樣共濟失調;耳部帶狀皰疹;帶狀皰疹;平山 症候群(Hirayama syndrome) ; HIV有關癡呆及神經病變(以 及AIDS之神經表現);前腦無裂畸形;亨廷頓病 (Huntington's disease)及其他聚麩胺醯胺重複疾病;積水 性無腦畸形;腦積水;皮質醇增多症;缺氧;免疫介導性 腦脊髓炎;包涵體肌炎;色素失調症;嬰兒植烷酸貯積 病;嬰兒雷弗蘇姆病infantile refsum disease);嬰兒痙 攣;炎性肌病;顱内囊腫;顱内高壓;朱伯特症候群 (Joubert syndrome);科姆斯-塞爾症候群(Keams-Sayre syndrome);肯尼迪氏病(Kennedy disease);金斯布林納症 157514.doc -21 - 201209163 候群(Kinsboume syndrome);克-費二氏症候群(Klippel Feil syndrome);克拉伯病(Krabbe disease);庫格爾貝格-韋蘭德病(Kugelberg-Welander disease);庫魯病(kuru);拉 福拉病(Lafora disease);朗-愛二氏肌無力症候群 (Lambert-Eaton myasthenic syndrome);蘭達-克萊夫納症 候群(Landau-Kleffner syndrome);延趙外側(瓦倫貝克 (Wallenberg))症候群;學習失能;利氏病(Leigh’s disease),倫諾克斯-加斯托症候群(Lennox-Gustaut syndrome);萊-萘二氏症候群(Lesch-Nyhan syndrome);腦 白質營養不良症;路易體癡呆(Lewy body dementia);無 腦回,閉鎖症候群;盧-格裏格病(Lou Gehrig's disease)(亦 即,運動神經元病或肌萎縮側索硬化);腰椎間盤病;萊 姆病(Lyme disease)-神經後遺症;馬-約病(Machado-Joseph disease);腦肥大;巨腦;邁-羅二氏症候群 (Melkersson-Rosenthal syndrome);美尼爾症(Menieres disease);髓膜炎;門克斯病(Menkes disease);異染性腦 白質營養不良;小頭畸型;偏頭痛;米勒·費希爾症候群 (Miller Fisher syndrome);小中風;粒線體肌病;默比烏 斯症候群(Mobius syndrome);單肢肌萎縮;運動神經元 病;腦底異常血管網病;黏多糖累積病;多發梗塞性癡 呆;多灶性運動神經病變;多發性硬化及其他去髓鞘病 症;具有位置性低血壓之多系統萎縮;肌營養不良症;重 症肌無力;去髓鞘彌漫性硬化;嬰兒肌陣攣性腦病;肌陣 攣;肌病;肌強直;嗜眠症;神經纖維瘤病;神經阻滞劑 157514.doc -22- 201209163 惡性症候群;AIDS之神經表現;狼瘡之神經後遺症;神 經性肌強直,神經元臘樣脂褐質症;腦神經元移行異常; 尼曼皮克病(Niemann-Pick disease);奥沙利文_麥克勞彳轰病 症(O'Sulhvan-McLeod syndrome);枕部神經痛;隱性脊柱 神經管閉合不全序列徵;大田原症候群(〇htahara syndrome);橄欖體腦橋小腦萎縮;斜視性眼陣攣;視神 經炎;直立性低血壓;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n,s disease)、亨廷 頓病、阿爾茨海默氏病、肌萎縮側索硬化(ALs)、癡呆' 多發性硬化及與神經元細胞死亡有關之其他疾病及病 症),先天性副肌強直症;副腫瘤性疾病;陣發性發作; 帕-羅二氏症候群(parry Romberg syndrome);佩-梅二氏病 (Pelizaeus-Merzbacher disease);週期性癱瘓;周邊神經病 變;疼痛性神經病變及神經性疼痛;持續性植物人狀態; 全身性發育遲緩;旋光性噴嚏反射;植烷酸貯積病;匹克 病(Pick's disease);神經挾捏;垂體瘤;多肌炎;腦穿通 畸形’小兒麻療症後期症候群;帶狀皰療後神經痛,·感染 後腦脊髓炎;體位性低血壓;帕-魏二氏症候群(prader_ Willi syndrome);原發性侧索硬化症,;朊病毒病;進行性 一側面萎縮;進行性多灶性白質腦病;進行性硬化性灰質 萎縮;進行性核上麻痹;假腦瘤;拉姆齊-亨特症候群 (Ramsay-Hunt syndrome)(I及II型);羅斯默森氏腦炎 (Rasmussen's encephalitis);反射性交感神經營養不良症候 群;雷夫敘姆病(Refsum disease);重複性運動病症;重複 157514.doc -23· 201209163 性壓迫損傷;不寧腿症候群;反轉錄病毒相關性脊髓病; 蕾特氏症候群(Rett syndrome);雷依氏症候群(Reye,s syndrome),舞蹈病(Saint Vitus dance);山德霍夫氏病 (Sandhoff disease),谢耳德病(Sehilder's disease);腦裂; 透明隔-視神經發育不良;驚嚇嬰兒症候群;帶狀皰療; 夏伊-德雷格症候群(Shy-Drager syndrome);薛格連氏症候 群(Sjogren’s syndrome);睡眠呼吸暫停;索托斯症候群 (Soto's syndrome);痙攣狀態,·脊柱裂;脊髓損傷;脊髓 腫瘤;脊髓性肌萎縮;僵人症候群(Stiff_Pers〇n syndrome);中風;斯特奇-韋伯二氏症候群(Sturge_Weber syndrome);亞急性硬化性全腦炎;皮層下動脈硬化性腦 病,西德納姆舞蹈病(Sydenham chorea);暈厥;脊髓空洞 症;遲發性運動障礙;泰-薩克斯病(Tay_Sachs diseas匀; 顧動脈k ’脊髓牵扯症候群;湯姆森病(Thomsen disease);胸廓出口症候群;三叉神經痛症(Tic Douloureux);託德氏麻痺(Todd's paralysis);多動穢語症 候群;短暫性腦缺企發作;傳播性海綿狀腦病;橫貫性脊 髓炎;外傷性腦損傷;顫抖;三叉神經痛;熱帶痙攣性輕 截癱;結節性硬化症;jk管性癡呆(多發梗塞性癡呆);血 管炎’包含顯動脈炎;希林二氏病(V〇n Hippel-Lindau disease),瓦倫伯格氏症候群(Wallenberg’s syndrome);韋 德尼希-霍夫曼病(Werdnig-Hoffman disease);韋斯特病 (West syndrome);頸椎戳傷;威廉斯症候群(Williams syndrome);威爾森氏病(Wildon's disease);及澤韋格症候 157514.doc •24- 201209163 群(Zellweger syndrome) 〇 「增殖性疾病或病症」包含但不限於涉及以下細胞之造 金腫瘤性病症.源於骨髓樣、淋巴樣或紅血·球譜系之造灰 源的增生性/腫瘤性細胞、或其前體細胞。該等疾病包含 但不限於成紅細胞白血病、急性前骨髄性白血病 (APML)、慢性髓性白血病(CmL)、淋巴樣惡性腫瘤(包含 但不限於急性成淋巴細胞性白血病(ALL),其包含B_譜系 ALL及T-譜系ALL)、慢性淋巴細胞白血病(CLL)、幼淋巴 細胞白血病(PLL)、多毛細胞白血病(HLL)及沃爾登斯特倫 巨球蛋白血症(Waldenstrom's macroglobulinemia) (WM)。 惡性淋巴瘤之其他形式包含但不限於非霍奇金氏淋巴瘤及 其變體、周邊T細胞淋巴瘤、成人τ細胞白血病/淋巴瘤 (ATL)、皮膚τ細胞淋巴瘤(CTCL)、巨粒淋巴細胞白血病 (LGF)、霍奇金氏病及裏德_斯藤伯格病 disease) ° 「發炎」係指全身性發炎病狀及局部與單核細胞、白血 球及/或曰中性球遷移及吸引有_之病狀。發炎之實例包 ^但不限於因感染病原性有機體(包含格蘭氏㈣⑷陽性細 菌格蘭氏陰性細菌、病毒、真菌、及諸如原生動物及螺 蟲等寄生蟲)、移植排斥(包含實體器官(例如,腎、肝、心 臟肺或角膜)之排斥、以及骨髓移植(包含移植物抗宿主 ’、病(HD))之排斥)、或局部慢性或急性自體免疫或過敏 應引起之發火。自體免疫性疾病包含腎小球腎炎;類風 W反應性關節炎;慢性腎小球腎炎;發炎性腸病,例 157514.doc -25- 201209163 如克隆氏病(CroWs disease)、潰癌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;顆粒球輸注有關之症候群;發炎性皮膚,例如,接觸 !生皮人4位性皮火、牛皮癖;全身性紅斑狼瘡(队E)、 自體免疫性甲狀腺炎、多發性硬化、及一些形式之糖尿 病、或由個體之自身免疫系統攻擊產生病變組織損壞的任 一其他自體免疫性狀態。過敏反應包含過敏性哮喘、慢性 枝氣管炎、急性及遲發性過敏。全身性發炎疾病狀態包含 與創傷、燒傷、再灌注後缺血性事件(例如心臟、腦、腸 或外周血管中之血栓形成性事件,包含心肌梗塞及中 風)、敗血症、ARDS或多器r功能障礙症候群有關之發 炎。發炎性細胞募集亦發生在動脈粥樣硬化斑塊中。發炎 包含但不限於非霍奇金氏淋巴瘤、韋格納氏肉芽腫病 (Wegener's granulomatosis)、橋本氏甲狀腺炎(Hashim〇t〇 s thyroiditis)、肝細胞癌、胸腺萎縮、慢性胰腺炎、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、克隆氏病、潰瘍性結腸炎、急性膽囊炎、慢性 膽囊炎、硬化、慢性涎腺炎、腹膜炎、急性胰腺炎、慢性 胰腺炎、慢性胃炎、子宮肌腺症、子宮内膜異位、急性宮 頸炎、慢性宮頸炎、淋巴增生、多發性硬化、急性特發性 血小板減少紫癜繼發性肥大、原發性IgA腎病、全身性紅 斑狼瘡、牛皮癬、肺氣腫、慢性腎盂腎炎及慢性膀胱炎。 聚核苷酸及募核苷酸組合物及分子 靶:在一個實施例中,靶包括BCL2結合部分3(bbc3)之 157514.doc •26- 201209163 核酸序列,包含(不限於)與BBC3有關之正義及/或反義非 編碼及/或編>5馬序列。 BCL2結合部分3(BBC3)或識另4為DNA損傷後 誘導之p53調控基因。隨後發現purna負責許多p53誘導之促 細胞凋亡效應,但Puma亦可以p53非依賴方式引起細胞凋 亡。將Puma指定為敏感劑或活化劑一直有些爭議。唯BH3 蛋白之BH3結構域係必需的且足以與Bcl-2家族成員相互作 用且看似在很大程度上重現完整蛋白之功能。舉例而言, 〇Appl. Math., (1981) 2, 482-489). The term "thermal melting point" as used herein refers to the temperature at which 5 % of the oligonucleotides complementary to the target sequence hybridize to the target sequence under equilibrium at a defined ionic strength, pH and nucleic acid concentration. Generally, for short oligonucleotides (for example, having 10 to 50 nucleotides), stringent conditions are the following: What is the salt concentration? 1 ^ 7.0 to 8.3 is at least about 0.01 to 1 _ 〇 之 ^ 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子 离子As used herein, "modulating" means increasing (stimulating) or reducing (inhibiting) the expression of a gene. When used in the context of a polynucleotide sequence, the term "variant" may encompass a gene associated with a wild-type gene. Polynucleotide sequence. This definition may also include, for example, "allele", "splicing", "species" or "polymorphism" variants. Splice variants may be significantly consistent with a reference molecule, but There is usually a larger or smaller number of polynucleotides during alternate processing due to alternate splicing of exons during mRNA processing. The corresponding polypeptide may have additional functional domains or no domains. Species change systems vary from species to species. Polynucleotide sequences. Particularly useful in the present invention are variants of a wild-type gene product. Variants may be derived from at least one mutation in a nucleic acid sequence and may produce altered 2 mRNA or produce a structure or function that may or may not be altered. Altered polypeptide Any given natural or recombinant gene may have no allelic form, with one or more allelic forms. Common mutational changes that result in variants are usually attributed to natural deletions, additions or substitutions of nucleotides. Each of the polypeptides may be singly or in combination with others one or more times. 157514.doc -16 - 201209163 The resulting polypeptides generally have significant amino acid identity with each other. Changes in the nucleotide sequence of a particular gene between individuals in a given species. Polymorphic variants may also encompass "single nucleotide polymorphisms" (SNp) or single-sequence mutations, where the nucleotide sequence The presence of a SNP may indicate, for example, that a population has a predisposition to disease status (ie, susceptibility to resistance). Derived polynucleotides are subjected to chemical modification (eg, hydrogen from an alkane) A nucleic acid, such as a derivatized oligonucleotide, may comprise a non-natural moiety, such as an altered sugar moiety or an intersaccharide linkage. Such exemplary street biology systems Phosphoroesters and other sulfur-containing substances known in the art. Derivative nucleic acids may also contain labels, including radionucleotides, enzymes, fluorescent agents, chemiluminescent agents, chromogenic agents, receptors, cofactors, inhibitors, magnetics. Particles and the like. "Derivative" polypeptides or peptides (for example) are modified by: glycosylation, pegylation, phosphonium, sulfation, reduction/alkylation, saccharification, chemical coupling Or mild formalin treatment. Derivatives may also be modified to contain detectable labels (directly or indirectly), including but not limited to radioisotopes, fluorescent and enzymatic labels. The term "animal" or "patient" as used herein. It is intended to include, for example, humans, sheep, elk, deer, mule deer, shoal, scorpion animals, monkeys, horses, cows, pigs, goats, dogs, cats, rats, mice, birds, chickens, Reptiles, fish, insects and spiders. Mammals encompass warm-blooded mammals (eg, humans and domestic animals) that are usually under medical care. Examples include cats, dogs, horses, cattle, and 157514.doc -17- 201209163 humans, and only humans. "Trating or treatment" encompasses the treatment of a disease state in a mammal and comprises: (4) in a mammal, in particular when the mammal is susceptible to a disease state but has not yet been diagnosed with the disease state Preventing the occurrence of the disease state; (b) inhibiting the disease state, for example, preventing its development; and/or (c) slowing the disease state, for example, reducing the disease state until the desired endpoint is reached. Treatment also includes amelioration of the symptoms of the disease (e. g., pain relief or discomfort), wherein the improvement may or may not directly affect the disease (e.g., cause, infection, performance, etc.). As used herein, "cancer" 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 includes malignant cells of cancer. Examples of tumors include sarcomas and carcinomas such as, but not limited to, fibrosarcoma, mucinous sarcoma, liposarcoma, chondrosarcoma, osteosarcoma, chordoma, angiosarcoma, endothelial sarcoma, lymphangiosarcoma, lymphatic endothelial sarcoma, synovium Tumor, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma 'sweat adenocarcinoma, Sebaceous gland cancer, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchial ductal carcinoma, renal cell carcinoma, hepatocellular carcinoma, cholangiocarcinoma, choriocarcinoma, seminoma, embryonal carcinoma, ville Wilms, tumor, cervical cancer, sputum tumor, lung cancer, small cell lung cancer, bladder cancer, epithelial cancer, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma , pineal tumor, hemangioblastoma, 157514.doc -18- 201209163 neuroma, oligodendrocyte glioma, meningioma, melanoma, neuroblastoma and retinoblastoma. Other cancers that may be treated by the compositions disclosed herein include, but are not limited to, for example, Hodgkin's DiSeaSe, non-Hodgkin's lymphoma (N〇nH〇dgkin, s Lymphoma), and steep Monthly tumor, neuroblastoma, breast cancer, Indian cancer, lung cancer, transverse, 'muscle 0 primary thrombocytosis, primary macroglobulinemia, small cell lung tumor, primary brain tumor, Gastric cancer, colon cancer, malignant pancreatic islet tumor, malignant carcinoid, bladder cancer, gastric cancer 'pre-deterioration skin damage, testicular cancer lymphoma, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary cancer, malignant high calcium Blood, cervical cancer, endometrial cancer, adrenal cortical cancer and prostate cancer. As used herein, "neurological disease or condition" refers to any disease or condition of the nervous system and/or the visual system. "Nervous diseases or conditions" include those involving the central nervous system (brain, brainstem and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (located in the ten pivots and peripheral nervous systems). A disease or condition. 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; aphasia; Aicardi syndrome; Aiexander disease, Aipers' disease; crossed limb paralysis, Alzheimer's disease; vascular dementia; amyotrophic lateral sclerosis; no brain malformation; angel syndrome (Angelrnan) Syndrome); angiomatosis; hypoxia; aphasia; apraxia; arachnoid cyst; arachnoiditis; A-Cai's malformation (Anr〇nl_Chiari 157514.doc -19- 201209163 malf〇rmati〇n); Pulse malformation; Asperger syndrome (Asperger synd_e); ataxia telangiectasia; attention deficit with hyperactivity disorder 'autism' autonomic dysfunction; back pain; Batten disease Becht's disease resembles ❹); Bells palsy, benign essential sputum; benign local muscle atrophy; benign Gu internal pressure, Binswanger's disease Binswanger, shua (10) called; eye 睑痉 ' 'Bloch Sulzberger syndrome, brachial plexus injury; brain abscess; brain injury; brain tumor (including glioblastoma multiforme); spinal cord tumor Brown-Sequard Syndrome; Canavan disease; Carpal tunnel syndrome; Burning neuralgia; Central pain syndrome; Central medullary cerebral lysis; Head disorders; Cerebral aneurysms; Cerebral arteriosclerosis; brain atrophy, cerebral giant disease; cerebral palsy; Charcot-Made-Tooth disease; chemotherapy-induced neuropathy and neuropathic pain 'Chiari malformation (Chiari Malformation; chorea; chronic inflammatory demyelinating polyneuropathy; chronic pain; chronic regional pain syndrome; c-ffin L〇wry syndrome; coma, including persistent vegetative state Congenital facial paralysis; cortical basal degeneration; cranial arteritis; craniosynostosis; Creutzfeldt-Jakob disease; cumulative traumatic disease, · Cushing's syndrome (Cushing's syndrome); giant cell inclusion disease; cytomegalovirus infection; dance eye-dance foot syndrome; DandyWalker syndrome; Dawson disease; De Morsier's syndrome ;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;;; Abnormal; early childhood epilepsy encephalopathy; empty sella syndrome; encephalitis; cerebral palsy; brain trigeminal angiomatosis; epilepsy; Erb's palsy; idiopathic tremor; Fabry's disease Fahr's syndrome; fainting; familial spastic paralysis; febrile seizures; Fisher syndrome; Friedreich's ataxia; frontotemporal dementia and Other "tau lesions"; Gaucher's disease; Gerstmann's syndrome; giant cell arteritis; giant cell inclusion disease ; globular white matter dystrophy; Guillain-Barre syndrome; HTLV-1 related to myelopathy; Hallervorden-Spatz disease; head injury; headache; Hereditary spastic paraplegia; genetic polyneuritis-like ataxia; ear herpes zoster; herpes zoster; Hirayama syndrome; HIV-related dementia and neuropathy (and neurological manifestations of AIDS) Forebrain non-cracking malformation; Huntington's disease and other poly- glutamine repeat disease; water-free brain-free malformation; hydrocephalus; hypercortisolism; hypoxia; immune-mediated encephalomyelitis; inclusion bodies Myositis; pigment disorders; infant phytanic acid storage disease; infant refsum disease; infantile spasm; inflammatory myopathy; intracranial cyst; intracranial hypertension; Joubert syndrome Keams-Sayre syndrome; Kennedy disease; Kingsbrinna disease 157514.doc -21 - 201209163 Kinsboume syndrome; Ke-Fei's syndrome (Klippel Feil syndrome); Krabbe disease; Kugelberg-Welander disease; Kuru disease; Lafora disease; Lang-Ai Lambert-Eaton myasthenic syndrome; Landau-Kleffner syndrome; Yan Zhao lateral (Wallenberg) syndrome; learning disability; Leigh's disease , Lennox-Gustaut syndrome; Lesch-Nyhan syndrome; leukodystrophy; Lewy body dementia; no brain return, atresia Syndrome; Lou Gehrig's disease (ie, motor neuron disease or amyotrophic lateral sclerosis); lumbar disc disease; Lyme disease - neurological sequelae; horse-cure disease (Machado) -Joseph disease); brain hypertrophy; giant brain; Melkersson-Rosenthal syndrome; Menieres disease; meningitis; Menkes disease; Malnutrition of the white matter; microcephaly Migraine; Miller Fisher syndrome; small stroke; mitochondrial myopathy; Mobius syndrome; single limb muscle atrophy; motor neuron disease; abnormal vascular network Disease; mucopolysaccharidosis; multiple infarct dementia; multifocal motor neuropathy; multiple sclerosis and other demyelinating disorders; multiple systemic atrophy with positional hypotension; muscular dystrophy; myasthenia gravis; Diffuse sclerosis; infant myoclonic encephalopathy; myoclonus; myopathy; myotonia; narcolepsy; neurofibromatosis; nerve blocker 157514.doc -22- 201209163 malignant syndrome; neurological manifestations of AIDS; Neurological sequelae; neuromuscular rigidity, neuron-like lipofuscin; abnormal brain neuron migration; Niemann-Pick disease; O'Sullivan_McLow's disease (O'Sulhvan- McLeod syndrome; occipital neuralgia; recessive spinal canal insufficiency sequence; Daejeon syndrome (〇htahara syndrome); olivine pons cerebellar atrophy; strabismic cerebral palsy; optic neuritis; Standing hypotension; overuse syndrome; paresthesia; neurodegenerative diseases or conditions (Parkins〇n, s disease, Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALs) , dementia 'multiple sclerosis and other diseases and conditions associated with neuronal cell death), congenital accessory myotonia; paraneoplastic disease; paroxysmal seizure; parry Romberg syndrome; - Pelizaeus-Merzbacher disease; periodic paralysis; peripheral neuropathy; painful neuropathy and neuropathic pain; persistent vegetative state; systemic developmental delay; optical sneezing reflex; phytanic acid storage disease ;Pick's disease; nerve kneading; pituitary tumor; polymyositis; brain penetrating malformation' pediatric numbness syndrome late syndrome; post-constrained neuralgia, · post-infectious encephalomyelitis; orthostatic hypotension; - Prader_ Willi syndrome; primary lateral sclerosis; prion disease; progressive one-sided atrophy; progressive multifocal leukoencephalopathy; progressive sclerosis Atrophic gray matter atrophy; progressive nuclear paralysis; pseudocephaloma; Ramsay-Hunt syndrome (types I and II); Rasmussen's encephalitis; reflex sympathetic nutrition Adverse syndrome; Refsum disease; repetitive motor disorder; repetition 157514.doc -23· 201209163 sexual compression injury; restless leg syndrome; retrovirus-associated myelopathy; Rett syndrome ); Reye, s syndrome, Saint Vitus dance; Sandhoff disease, Sehilder's disease; cerebral palsy; clear septum - optic nerve dysplasia Stunning infant syndrome; vesicular therapy; Shy-Drager syndrome; Sjogren's syndrome; sleep apnea; Soto's syndrome; sputum state, · spine Splitting; spinal cord injury; spinal cord tumor; spinal muscular atrophy; Stiff_Pers〇n syndrome; stroke; Sturge-Weber syndrome; Sclerotherapy encephalitis; subcortical arteriosclerotic encephalopathy, Sydenham chorea; syncope; syringomyelia; tardive dyskinesia; Thai-sax disease (Tay_Sachs diseas uniform; Gu artery k' Spinal cord involvement syndrome; Thomsen disease; thoracic outlet syndrome; Tic Douloureux; Todd's paralysis; hyperactive slang syndrome; transient brain deficiency episode; Encephalopathy; transverse myelitis; traumatic brain injury; trembling; trigeminal neuralgia; tropical spastic paraplegia; tuberous sclerosis; jk tube dementia (multiple infarct dementia); vasculitis 'including arteritis; V〇n Hippel-Lindau disease, Wallenberg's syndrome; Werdnig-Hoffman disease; West syndrome; cervical vertebra Wounds; Williams syndrome; Wilson's disease; and Zweig's disease 157514.doc •24- 201209163 Group (Zellweger syndrome) 〇 "Proliferative A disease or condition includes, but is not limited to, a neoplastic neoplastic disorder involving a cell derived from a proliferative/neoplastic cell of a bone marrow-like, lymphoid or red blood-globular lineage, or a precursor cell thereof. Such diseases include, but are not limited to, erythroblastic leukemia, acute anterior bone marrow 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), lymphoblastic 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 T-cell lymphoma, adult tau cell leukemia/lymphoma (ATL), skin tau cell lymphoma (CTCL), giant granules Lymphocytic leukemia (LGF), Hodgkin's disease, and Reed_Stowe's disease disease) ° "Inflammation" refers to systemic inflammatory conditions and local and monocyte, white blood cell and / or sputum neutral ball migration And attracting the disease. Examples of inflammation include, but are not limited to, infection with pathogenic organisms (including gram-negative bacteria, gram-negative bacteria, viruses, fungi, and parasites such as protozoa and snails), transplant rejection (including solid organs) For example, rejection of kidney, liver, heart lung or cornea, and bone marrow transplantation (including rejection of graft versus host ', disease (HD)), or local chronic or acute autoimmunity or allergies should cause fire. Autoimmune diseases include glomerulonephritis; wind-like W-responsive arthritis; chronic glomerulonephritis; inflammatory bowel disease, 157514.doc -25- 201209163 such as Crows disease, canceration Colitis and necrotizing enterocolitis, hepatitis, sepsis, alcoholic liver disease; nonalcoholic steatosis; syndrome associated with granule ball infusion; inflammatory skin, for example, contact! 4 skin fire, cowhide癖; systemic lupus erythematosus (Team E), autoimmune thyroiditis, multiple sclerosis, and some forms of diabetes, or any other autoimmune state of lesion tissue damage caused by an individual's own immune system attack. Allergic reactions include allergic asthma, chronic bronchitis, acute and delayed allergies. Systemic inflammatory conditions include traumatic events following trauma, burns, reperfusion (eg, thrombotic events in the heart, brain, intestine, or peripheral blood vessels, including myocardial infarction and stroke), sepsis, ARDS, or multi-function Inflammation associated with the disorder syndrome. Inflammatory cell recruitment also occurs in atherosclerotic plaques. Inflammation includes, but is not limited to, non-Hodgkin's lymphoma, Wegener's granulomatosis, Hashim〇t〇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 mumps, peritonitis, acute pancreas Inflammation, chronic pancreatitis, chronic gastritis, uterine adenomyosis, endometriosis, acute cervicitis, chronic cervicitis, lymphatic hyperplasia, multiple sclerosis, acute idiopathic thrombocytopenic purpura secondary hypertrophy, primary IgA nephropathy, systemic lupus erythematosus, psoriasis, emphysema, chronic pyelonephritis, and chronic cystitis. Polynucleotide and Nucleotide Compositions and Molecular Targets: In one embodiment, the target comprises a 157514.doc •26-201209163 nucleic acid sequence of BCL2 binding portion 3 (bbc3), including (without limitation) associated with BBC3 Justice and/or antisense non-coding and/or editing > 5 horse sequences. BCL2 binding moiety 3 (BBC3) or recognition 4 is a p53 regulatory gene induced after DNA damage. It was subsequently found that purna is responsible for many p53-induced pro-apoptotic effects, but Puma can also cause cell apoptosis in a p53-independent manner. Designating Puma as a sensitizer or activator has been somewhat controversial. Only the BH3 domain of the BH3 protein is required and sufficient to interact with Bcl-2 family members and appears to reproduce the function of the intact protein to a large extent. For example, 〇

Bid及Blm之BH3結構域可活化脂質體或粒線體中之Bax及 Bak °在若干研究中’ Puma BH3結構域無此功能,導致許 多人將Puma歸類為敏感劑。然而,利用在活體外轉譯之全 長蛋白的實驗顯示與Bim及Bid相當之活化Bax之能力。 傳統上將Puma視為細胞死亡之p53誘導型調節劑,但其 亦在p53非依賴性細胞死亡途徑、例如彼等由生長因子缺 失或利用糖皮質激素治療誘導者中起關鍵作用。Puma之促 Q 細胞凋亡作用機制歸因於其以高親和力結合且因此抑制所 有抗細胞鴻亡Bcl-2家族成員(包含Bcl_2、bc1-XL、Bcl_ w、Mcl-1及A1)的能力。亦已報導puma可藉由直接結合至 B ax來活化内在細胞凋亡途徑。 在Λ施例中,使用反義寡核苷酸來預防或治療與 BBC3家族成員有關之疾病或病症。實例性沉[2結合部分 3(BBC3)調介之疾病及病症(其可用自使用反義化合物獲得 之幹細胞再生之細胞/組織治療)包括:與BBC3之異常功能 及/或表現有關之疾病或病症、癌症、增殖性疾病或= 157514.doc •27- 201209163 症、與細胞增殖有關或特徵在於細胞增殖之疾病或病狀、 與BBC3之大變或異常表現或功能有關之疾病或病症、神 、’二疾病或病症、發炎、以發炎關節成為病變之一部分的病 狀(」列如,骨關節炎、關節炎、牛皮癖性關節炎、幼年型 關節炎、萊特爾氏症候群(Reiter,s syndr〇me)、與潰 衅 腸炎有關之關節炎、惠伯爾病(Whipple’s disease)、與肉芽 腫回腸結腸炎有關之關節炎、貝切特氏病(Behcet's se)王身性紅斑狼瘡、乾燥症候群(Sj〇gren,s —Ο及混合型結締組織病等)、與粒線體細胞凋亡途 徑損傷有關之疾病或病症、自體免疫性疾病或病症及與神 經凡細胞死亡有關之疾病或病症、衰老或特徵在於不期望 細胞損失之其他病狀。 在本發明實施例中,為需要皮膚治療或處於發生可需要 皮膚治療之病狀風險的個體提供治療及或美容方案及相關 個性化治療。可基於(例如)個體之BBC3狀態進行診斷。患 者給定組織(例士α皮膚)中之BBC3表現量可藉由彼等熟習此 項技術者已知且在本文中別處闡述之方法,例如藉由使用 PCR或抗體基檢測方法分析組織來測定。 在一實施例中,對有需要之患者實施藉由一或多種反義 寡核«來mMBC3,以預防或治療與BBC3異常表現、 功能、活性(與正常對照組相比)相關的任一疾病或病症。 在一實施例中,寡核苷酸對BBC3之聚核苷酸具有特異 性,其包含(不限於)非編碼區域。BBC3靶包括之變 體;BBC3之突變體,包含SNp ;賦3之非編碼序列;等 157514.doc •28- 201209163 位基因、片段及諸如此類。較佳地,寡核苷酸係反義rNa 分子。 根據本發明實施例,靶核酸分子並不僅限於BBC3聚核 苷酸而是擴展至BBC3之任一同種型、受體、同系物、非 編碼區域及諸如此類。 在一實施例中,寡核苷酸靶向BBC3靶之天然反義序列 (編碼及非編碼區域之天然序列),其包含(不限於)其變 體、等位基因、同系物、突變體、衍生物、片段及互補序 列。較佳地,寡核苷酸係反義RNA或DNA分子。 在一實施例中’本發明之寡聚化合物亦包含在該化合物 中之一或多個核苷酸位置存在不同鹼基的變體。舉例而 言’若第一核苷酸係腺嘌呤,則可產生在此位置含有胸 苦、鳥苷、胞苷或其他天然或非天然核苷酸之變體。此可 發生於反義化合物之任一位置。然後使用本文所述方法測 試該等化合物以測定其抑制靶核酸表現之能力。 在一些實施例中,反義化合物與靶之間之同源性、序列 一致性或互補性為約50%至約60%。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%。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 100〇/〇。 在以下情形時反義化合物可特異性雜交:化合物與靶核 157514.doc •29· 201209163 而導致活性損失,且存 酸之結合可干擾靶核酸之正常功能 酸序列在期 。該等條件 在足夠互補性程度以避免反義化合物與非靶核 望發生特異性結合之條件下發生非特異性結合 包含(亦即)活體内分析或治療性治療情形下之生理條件 及在活體外分析情形下實施分析之條件。 在以下情形下反義化合物(不論係DNA、RNA、嵌合化 合物、或經取代化合物等)可特異性雜交〔該化合物^靶 DNA或RNA分子之結合可干擾靶DNA或RNA之正常功能而 導致實用性損失,且存在足夠互補性程度以避免反義化合 物與非靶序列在期望發生特異性結合之條件下(亦即,在 活體内分析或治療性治療情形下之生理條件下或在活體 外分析情形下實施分析之條件下)發生非特異性結合。 在一實施例中,靶向BBC3(包含(不限於)使用(例 如)PCR、雜交等鑑別及擴展之反義序列、闡述為seq N〇: 2之序列之一或多者及諸如此類)可調節BBC3之表現或 功能。在一個實施例中,與對照組相比上調表現或功能。 在一貫施例中,與對照組相比下調表現或功能。 在一實施例中,寡核苷酸包括闡述為SEQ ID NO: 3至8 之核酸序列,包含使用(例如)PCR、雜交等鑑別及擴展之 反義序列。該等养核普酸可包括一或多個經修飾核皆酸、 較短或較長片段、經修飾鍵及諸如此類。經修飾鍵或核苷 酸間鍵聯之實例包括硫代磷酸酯、二硫代磷酸酯或諸如此 類。在一實施例中,核苷酸包括磷衍生物。可附接至本發 明經修飾寡核苷酸中之糖或糖類似物部分之磷衍生物(或 1575l4.doc •30- 201209163 經修飾磷酸酯基團)可為單磷酸酯、二磷酸酯、三磷酸 酯、磷酸烷基酯、烷磷酸酯、硫代磷酸酯及諸如此類。上 述磷酸醋類似物之製備、及其在核普酸、經修飾核苦酸及 养核苦酸中之納入本身亦已知且無需闡述於本文中。 • 彼等熟習此項技術者亦在治療應用中利用反義寡核苷酸 之特異性及敏感性。採用反義寡核苷酸作為治療部分來治 療動物及人類之疾病狀態。反義寡核苷酸已安全且有效地 投與人類且當前正實施許多臨床試驗。由此確定,募核苷 〇 酸可為可經設置用於治療細胞、組織及動物(尤其人類)之 治療方案中的有用治療方式。 在本發明實施例中,募聚反義化合物、尤其寡核苷酸會 結合至靶核酸分子並調節由靶基因編碼之分子的表現及/ 或功能。擬干擾之DNA功能包括(例如)複製及轉錄。擬干 擾之RNA功能包括所有重要功能,例如,RNA至蛋白質轉 譯位點之易位、蛋白質自RNA之轉譯、使rNA產生一或多 ◎ 物質之剪接、及RNA可參與或促進之催化活性。 可端視期望功能來上調或抑制功能。 反義化合物包含反義寡聚化合物、反義寡核苷酸、外部 引導序列(EGS)募核苷酸、交替剪接體、引物、探針及與 乾核酸之至少一部分雜交的其他寡聚化合物。因此,該等 化合物可以單鏈、雙鏈、部分單鏈或環狀寡聚化合物形式 引入。 在本發明之上下文中,反義化合物至特定核酸分子之乾 向可為多步過程。該過程通常始於鑑別欲調節功能之乾核 157514.doc • 31 201209163 酸。舉例而言’㈣核酸可為表現與特定病症或疾病狀態 有關之細胞基因(或自該基因轉錄之mRNA)或來自傳染原 之核I刀子在本發明中,靶核酸編碼BCL2結合部分 3(BBC3)。 靶向過程通常亦包含測定靶核酸内發生反義相互作用從 而產生期望效應(例如,調節表現)的至少一個乾區域、區 段或位點。在本發明之上下文中,術語「區域」定義為乾 核酸中具有至少—種可鑑別結構、功能或特性的-部分。 靶核酸之區域内具有區段。「區段」$義為靶核酸内區域 之較小或子部分。本發明所用之「位點」$義為乾核酸内 之位置。 在一實施例中,反義募核苷酸結合至BCL2結合部分 3(BBC3)之天然反義序列並調節BBC3 (SEQ 1〇 n〇: 1)之表 現及/或功能。反義序列之實例包含Eq ID N〇: 2至8。 在一實施例中,反義寡核苷酸結合至BcL2結合部分 3(BBC3)聚核苷酸之一或多個區段並調節BBC3i表現及/ 或功能。區段包括BBC3正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BBC3之天然反義序列 具有特異性,其中募核苷酸與BBC3之天然反義序列之結 合可調節BBC3的表現及/或功能。 在一實施例中’寡核苷酸化合物包括闡述為SEq ID N〇: 3至8之序列、使用(例如)pcR、雜交等鑑別及擴展之反義 序列。该·#寡核苷酸可包括一或多個經修飾核苷酸、較短 157514.doc •32· 201209163 ΟThe BH3 domain of Bid and Blm activates Bax and Bak ° in liposomes or mitochondria. In some studies, the Puma BH3 domain does not have this function, leading to the classification of Puma as a sensitizer by many people. However, experiments using ubiquitin translated in vitro showed an ability to activate Bax comparable to Bim and Bid. Puma has traditionally been considered a p53-inducible modulator of cell death, but it also plays a key role in the p53-independent cell death pathway, such as those in which growth factors are absent or induced by glucocorticoid therapy. The mechanism by which Puma promotes Q apoptosis is attributed to its ability to bind with high affinity and thus inhibit all Bcl-2 family members (including Bcl_2, bcl-XL, Bcl_w, Mcl-1 and A1). It has also been reported that puma can activate the intrinsic apoptotic pathway by direct binding to B ax . In the case, antisense oligonucleotides are used to prevent or treat diseases or conditions associated with members of the BBC3 family. Exemplary diseases and conditions mediated by the binding moiety 3 (BBC3), which can be used for stem cell regeneration from stem cells obtained using antisense compounds, include: diseases associated with abnormal function and/or performance of BBC3 or A disease, a cancer, a proliferative disease, or a disease or condition characterized by cell proliferation, a disease or condition associated with cell proliferation, or a disease or condition associated with abnormal or abnormal performance or function of BBC3. , 'two diseases or conditions, inflammation, with inflammation of the joint become part of the disease (" such as, osteoarthritis, arthritis, psoriatic arthritis, juvenile arthritis, Lytel's syndrome (Reiter, s syndr 〇me), arthritis associated with ulcerative colitis, Whipple's disease, arthritis associated with granulomatous ileitis, Behcet's se, lupus erythematosus, dry syndrome (Sj〇gren, s-Ο and mixed connective tissue diseases, etc.), diseases or conditions associated with mitochondrial apoptosis pathway damage, autoimmune diseases or disorders, and neurological A disease or condition associated with cell death, aging, or other condition characterized by undesirable cell loss. In embodiments of the invention, treatment and or cosmetic treatment is provided for an individual in need of skin treatment or at risk of developing a condition that may require skin treatment. The protocol and related personalized treatments can be diagnosed based on, for example, the individual's BBC3 status. The amount of BBC3 expression in a given tissue (eg, alpha skin) of a patient can be known by those skilled in the art and are herein The method set forth elsewhere, for example, by analyzing tissue using PCR or antibody-based detection methods. In one embodiment, a patient in need is treated with one or more antisense oligonucleotides to mMBC3 for prevention or treatment. Any disease or condition associated with abnormal expression, function, activity (compared to a normal control group) of BBC3. In one embodiment, the oligonucleotide is specific for a polynucleotide of BBC3, including (but not limited to) Non-coding regions. BBC3 targets include variants; mutants of BBC3, including SNp; non-coding sequences of 3; 157514.doc •28- 201209163 genes, fragments and Preferably, the oligonucleotide is an antisense rNa molecule. According to an embodiment of the invention, the target nucleic acid molecule is not limited to the BBC3 polynucleotide but extends to any isoform, receptor, homologue of BBC3. , non-coding regions, and the like. In one embodiment, the oligonucleotide targets a natural antisense sequence of a BBC3 target (a natural sequence encoding a non-coding region) comprising, without limitation, variants, alleles thereof , homologues, mutants, derivatives, fragments and complementary sequences. Preferably, the oligonucleotide is an antisense RNA or DNA molecule. In one embodiment, the oligomeric compound of the invention is also included in the compound. Variants of different bases are present at one or more nucleotide positions. For example, if the first nucleotide is adenine, a variant containing a chestnut, guanosine, cytidine or other natural or non-natural nucleotide at this position can be produced. This can occur anywhere in the antisense compound. The 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 50% to about 60%. In some embodiments, homology, sequence identity or complementarity is from about 6% to about 7%. In some embodiments the 'homology, sequence identity or complementarity is from about 7% to about 80%. In some embodiments, homology, sequence identity or complementarity is from about 80% to about 90%. In some embodiments, homology, sequence identity or complementarity is about 90°/. About 92%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99% or about 100 angstroms/inch. The antisense compound can specifically hybridize in the following cases: the compound and the target nucleus 157514.doc •29·201209163 cause loss of activity, and the binding of the acid can interfere with the normal function of the target nucleic acid. Such conditions occur at a level of sufficient complementarity to avoid non-specific binding under conditions in which the antisense compound specifically binds to the non-targeted receptor (ie, physiological conditions in the case of in vivo or therapeutic treatment and in vivo) Conditions for performing the analysis in the case of external analysis. An antisense compound (whether DNA, RNA, chimeric compound, or substituted compound, etc.) can specifically hybridize under the following conditions (the binding of the compound to the target DNA or RNA molecule can interfere with the normal function of the target DNA or RNA) Loss of utility, and sufficient degree of complementarity to avoid antisense compounds and non-target sequences under conditions that are expected to specifically bind (ie, under physiological conditions in vivo or in therapeutic treatment or in vitro) Non-specific binding occurs under conditions in which the analysis is performed under analysis. In one embodiment, the BBC3 is targeted (including, without limitation, an antisense sequence identified and expanded using, for example, PCR, hybridization, etc., one or more of the sequences set forth as seq N〇: 2, and the like) The performance or function of the BBC3. In one embodiment, the performance or function is up-regulated compared to a control group. In a consistent example, performance or function was down-regulated compared to the control group. In one embodiment, the oligonucleotide comprises a nucleic acid sequence set forth as SEQ ID NOS: 3 to 8, comprising an antisense sequence identified and expanded using, for example, PCR, hybridization, and the like. Such nutrient acids may include one or more modified nucleocapnias, shorter or longer fragments, modified linkages, and the like. Examples of the modified bond or the internucleoside linkage include a phosphorothioate, a dithiophosphate or the like. In one embodiment, the nucleotide comprises a phosphorus derivative. A phosphorus derivative (or 1575l 4.doc • 30-201209163 modified phosphate group) that can be attached to a sugar or a sugar analog moiety of a modified oligonucleotide of the invention can be a monophosphate, a diphosphate, Triphosphates, alkyl phosphates, alkyl phosphates, phosphorothioates, and the like. The preparation of the above-described phosphate vinegar analogs, and their incorporation in nucleotides, modified nucleotides and nutrient acid are also known per se and need not be set forth herein. • Those skilled in the art also utilize the specificity and sensitivity of antisense oligonucleotides in therapeutic applications. Antisense oligonucleotides are used as therapeutic moieties to treat disease states in animals and humans. Antisense oligonucleotides have been safely and efficiently administered to humans and many clinical trials are currently being performed. It has thus been determined that the recruitment of nucleoside niacin can be a useful therapeutic modality that can be used in the treatment of cells, tissues and animals, especially humans. In an embodiment of the invention, a recruitment antisense compound, particularly an oligonucleotide, binds to a target nucleic acid molecule and modulates the expression and/or function of the molecule encoded by the target gene. DNA functions to be interfered with include, for example, replication and transcription. The RNA function to be interfered with includes all important functions, such as translocation of RNA to protein translation sites, translation of proteins from RNA, generation of one or more splicing of rNA, and catalytic activity in which RNA can participate or promote. The function can be adjusted or suppressed depending on the desired function. Antisense compounds comprise an antisense oligomeric compound, an antisense oligonucleotide, an external leader sequence (EGS) nucleotide, an alternative splicing, a primer, a probe, and other oligomeric compounds that hybridize to at least a portion of the dry nucleic acid. Thus, such compounds can be introduced as single-stranded, double-stranded, partially single-stranded 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 process usually begins with the identification of the dry core to be regulated. 157514.doc • 31 201209163 Acid. For example, the '(iv) nucleic acid can be a cellular gene (or mRNA transcribed from the gene) associated with a particular disorder or disease state or a nuclear I knife from an infectious agent. In the present invention, the target nucleic acid encodes a BCL2 binding moiety 3 (BBC3) ). The targeting process also typically involves determining at least one stem region, region or site within which the antisense interaction occurs within the target nucleic acid to produce a desired effect (e. g., to modulate performance). In the context of the present invention, the term "region" is defined as a moiety having at least one identifiable structure, function or property in a dry nucleic acid. There is a segment within the region of the target nucleic acid. The "segment" $ is the smaller or sub-portion of the region within the target nucleic acid. The "site" used in the present invention is defined as the position within the dry nucleic acid. In one embodiment, the antisense raised nucleotide binds to the natural antisense sequence of BCL2 binding portion 3 (BBC3) and regulates the expression and/or function of BBC3 (SEQ 〇 n〇: 1). Examples of antisense sequences include Eq ID N〇: 2 to 8. In one embodiment, the antisense oligonucleotide binds to one or more segments of a BcL2 binding portion 3 (BBC3) polynucleotide and modulates BBC3i expression and/or function. The segment includes at least 5 contiguous nucleotides of a BBC3 sense or antisense polynucleotide. In one embodiment, the antisense oligonucleotide is specific for the natural antisense sequence of BBC3, wherein binding of the raised nucleotide to the natural antisense sequence of BBC3 modulates the expression and/or function of BBC3. In one embodiment, an 'oligonucleotide compound comprises an antisense sequence characterized as SEq ID N〇: 3 to 8, which is identified and expanded using, for example, pcR, hybridization, and the like. The ## oligonucleotide may comprise one or more modified nucleotides, shorter 157514.doc •32· 201209163 Ο

或較長片段、經修飾鍵及諸如此類。經修飾鍵或核苷酸間 鍵聯之實例包括硫代填酸酯、二硫代磷酸酯或諸如此類。 在一實施例中,核苷酸包括填衍生物。可附接至本發明經 修飾募核苷酸中之糖或糖類似物部分之填衍生物(或經修 飾磷酸酯基團)可為單磷酸酯、二磷酸酯、三磷酸酯、鱗 酸烧基酯、烷磷酸酯、硫代磷酸酯及諸如此類。上述填酸 @旨類似物之製備、及其在核苷酸、經修飾核苷酸及寡核苦 酸中之納入本身亦已知且無需闡述於本文中。 如業内已知’因轉譯起動密碼子通常係5,_AUG(在轉錄 mRNA分子中;在相應Dna分子中係5'-ATG),轉譯起動密 碼子亦稱為「AUG密碼子」、「起始密碼子」或rAUG起始 密碼子」。少數基因具有轉譯起動密碼子,其具有RNA序 列 5’-GUG、5I-UUG或 5I-CUG ;且已顯示 5,_aua、5,_acg 及5,-CUG可在活體内發揮作用。因此,即使起動胺基酸在 每一情形中通常係曱硫胺酸(在真核生物中)或曱醯甲硫胺 酸(在原核生物中),術語「轉譯起動密碼子」及「起始 碼子」亦可涵蓋多種密碼子序列。真核生物及原核生物基 因可具有兩個或更多個替代性起始密碼子,其中之任一者 可優先用於在特定細胞類型或組織中或在特定條件組下之 轉譯起動。在本發明之上下文中,「起始密碼子」及「轉 譯起動密碼子」係m在活體内㈣自編碼概2結合 部分3(BBC3)之基因轉錄之之轉$ & 锝#的一或多個密碼 子,不管該等密碼子之序列如何。美 ^ ^ ^丞因之轉譯終止密碼子 (或「停止密碼子」)可具有二藉忠 ;韦—種序列(亦即,5'-UAA、51- 157514.doc •33· 201209163 5'-TAG 及 UAG及51-UGA ’相應DNA序列分別係5,_taa 5'-TGA)中之一者。 術語「起始密碼子區域」及「轉譯起動密碼子區域」 指此-mRNA或基因中在來自轉譯起動密碼子之任—方向 (亦即5或3 )上涵蓋約25至約5〇個鄰近核苦酸的—部分。 類m也#f 5吾「停止密碼子區域」及「轉譯終止密碼子區 I係指此1RNA或基因中在來自轉譯終止密碼子之^ :方向(亦即,5’或3,)上涵蓋約25至約5〇個鄰近核苷酸的— 4刀目此’「起始密碼子區域」(或「轉譯起動密碼子區 域J )及「停止密碼子區域」(或「轉譯終止密碼子區域」) 係本發明反義化合物可有效乾向之所有區域。 業内已知之開放讀碼框(〇RF)或「編碼區域」係指轉嘩 起動“子與轉譯終止密碼子之間之區域,其亦係可有效 靶向之區域。在本發明之上下文中,靶向區域係涵蓋基因 之開放讀碼框(0RF)之轉譯起動或終止密碼子的基因内區 域0 另一靶區域包含業内已知之5,非轉譯區域(5,utr),其係 指在來自轉譯起動密碼子之5,方向上之mRNA部分,且由 此包含mRNA之5,加帽位點與轉譯起動密碼子間之核苷酸 (或基因上之相應核苷酸另一靶區域包含業内已知之3, 非轉譯區域(3’UTR),其係指在來自轉譯終止密碼子之3,方 向上之mRNA部分,且由此包含mRNA之轉譯終止密碼子 與3·端間之核苷酸(或基因上之相應核苷酸)。mRNA25,加 帽位點包括經由5,-5,三磷酸酯鍵聯接合至mRNA之5,_最殘 157514.doc •34· 201209163 基之N7-T基化鳥苦殘基。mRNAi5,加帽區域視為包含 加帽結構本身以及與加帽位點相鄰之前5〇個核苷酸。本發 明之另一乾區域係5’加帽區域。 儘管一些真核生物mRNA轉錄物可直接轉譯,但許多真 • 核生物111尺1^轉錄物含有一或多個在轉譯之前自轉錄物切 割且稱為「内含子」的區域。刺餘(且由此轉譯)區域稱為 「外顯子」且一起剪接以形成連續mRNA序列。在—個實 施例中,靶向剪接位點(亦即,内含子-外顯子結點或外顯 子-内含子結點)尤其可用於異常剪接與疾病有關、或特定 剪接產物之過度產生與疾病有關之情形中。因重排或缺失 產生之異常融合結點係靶位點之另一實施例。經由自不同 基因源剪接兩個(或更多)mRNA之過程產生之mRNA轉錄物 私為「融合轉錄物」。可使用靶向(例如)1)1^入或111}〇^前體 之反義化合物來有效靶向内含子。 在一實施例中,反義寡核苷酸結合至靶聚核苷酸之編碼 Q 及/或非編碼區域並調節靶分子之表現及/或功能。 在一實施例中,反義寡核苷酸結合至天然反義聚核苷酸 並調節靶分子之表現及/或功能。 在一實施例中,反義募核苷酸結合至正義聚核苷酸並調 節把分子之表現及/或功能。 替代性RNA轉錄物可自DNA之相同基因組區域產生。該 等替代性轉錄物通稱為「變體」。更特定而言,「1111^^八前 體變體」係自相同基因組DNA產生之轉錄物,其與自相同 基因組DNA產生之其他轉錄物在起始或停止位置方面有所 157514.doc •35· 201209163 不同且含有内含子及外顯子序列。 在剪接期間切割一或多個外顯子或内含子區域、或其部 分時,mRNA前體變體會產生較小「mRNA變體」。因此, mRNA變體係經處理之mRNA前體變體且每一唯一 mRNA前 體變體必須始終藉由剪接而產生唯一 mRNA變體。該等 mRNA變體亦稱為「交替剪接變體」。若mRNA前體變體未 發生剪接,則mRNA前體變體與mRNA變體相同。 變體可經由使用起始或停止轉錄之交替信號來產生。 mRNA岫體及mRNA可具有一個以上之起始密碼子或停止 後碼子。源自使用替代性起始密碼子之mRNA前體或 mRNA的變體稱為mRNA前體或mRNA之「替代性起始變 體」。彼等使用替代性停止密碼子之轉錄物稱為mRNA前體 或mRNA之「替代性停止變體」。替代性停止變體之一種具 體類型係「polyA變體」,其中所產生之多個轉錄物源自轉 錄機對於「polyA停止信號」中之一者之替代性選擇,由 此產生在唯一 polyA位點終止之轉錄物。在本發明之上下 文中’本文所述變體之類型亦係靶核酸之實施例。 靶核酸上與反義化合物雜交之位置定義為至少靶區域中 由活性反義化合物靶向之長5個核苷酸的部分。 儘管本文中闡述了某些實例性靶區段之特異性序列,但 熟習此項技術者應認識到,該等序列用於闡釋及闡述本發 明範疇内之特定實施例。熟習此項技術者根據本揭示内容 可容易地鑑別其他靶區段。 人們認為長度為5至100個核苷酸且包括一段至少五(5) 157514.doc -36- 201209163 個選自較佳闡釋性靶區段之連續核苷酸之靶區段亦適於乾 向。 靶區段可包含DNA或RNA序列,其包括至少5個來自較 佳闡釋性靶區段中之一者之5,_末端之連續核苷酸(剩餘核 苦酸係同一 DNA或RNA中之連續的一段,其緊接靶區段之 5'-末端上游開始並持續至該DNA或RNA含有約5至約1〇〇個 核普酸為止)。類似地’較佳靶區段表示為Dna或RNA序 列,其包括至少5個來自較佳闡釋性靶區段中之一者之3,_ 末端之連續核苷酸(剩餘核苷酸係同一 DNA或RNA中之連 續的一段’其緊接靶區段之3,_末端下游開始並持續至該 DNA或RNA含有約5至約1〇〇個核苷酸為止)^瞭解本文所 示靶區段之熟習此項技術者無需過多實驗即能鑑別其他較 佳乾區段。 鑑別一或多個靶區域、區段或位點後,即可選擇與靶充 分互補(亦即雜交足夠充分且具有足夠特異性)之反義化合 物以得到期望效應。 在本發明實施例中,募核苷酸結合至特定靶之反義鏈。 募核苷酸之長度為至少5個核苷酸且可經合成以使每一募 核苦m向重疊序列’從而寡核_酸經合成以覆蓋乾聚 核苷酸之整個長度。靶亦包含編碼以及非編碼區域。 在一個實施例中’反義寡核苷酸較佳靶向特異性核酸。 反義化合物至特定核酸之靶向係多步過程。該過程通常始 於鑑別欲調節功能之核酸序列。此可(例如)係表現與特定 病症或疾病狀態有關之細胞基因(或自基因轉錄之 157514.doc -37- 201209163 mRNA)、或非編碼聚核苷酸(例如,非編碼RNA (ncRNA))。 RNA可分類為(1)信使RNA (mRNA),其轉譯成蛋白 質;及(2)非蛋白編碼性RNA (ncRNA)。ncRNA包括微小 RNA、反義轉錄物及含有高密度停止密碼子且缺乏任一廣 泛性(extensive)「開放讀碼框」的其他轉錄單元(TU)。許 多ncRNA似乎始於編碼蛋白質之基因座之3'非轉譯區域 (3'UTR)中之起動位點。ncRNA通常較罕見且FANTOM聯盟 已測序之ncRNA中至少一半似乎未經聚腺苷酸化。大部分 研究者出於明顯原因而著重關注經處理並輸出至細胞質中 之聚腺皆酸化mRNA。最近,顯示非聚腺苦酸化核RNA之 組可能極大,且許多該等轉錄物源自所謂的基因間區域。 ncRNA可調控基因表現之機制係與靶轉錄物之鹼基配對。 藉由鹼基配對發揮作用之RNA可分組為:(1)順式編碼之 RNA,其在其發揮作用之相同遺傳位置、但在RNA之相對 鍵處編碼且由此顯示與其乾之完全互補性;及(2)反式編 碼之RNA,其在與其發揮作用之RNA不同的染色體位置處 編碼且通常並不顯示與其靶之完全鹼基配對潛力。 不期望受限於理論下,本文所述反義寡核苷酸對反義聚 核苷酸之干擾可改變相應正義信使RNA之表現。然而,此 調控可為不協調(反義擊弱(knockdown)造成信使RNA增加) 或協調(反義擊弱造成伴隨信使RNA減少)。在該等情形 下,反義寡核苷酸可靶向反義轉錄物之重疊或非重疊部 分,以產生擊弱或隔絕(sequestration)。可以相同方式把向 157514.doc -38- 201209163 編碼以及非編瑪反義轉錄物,且任一種類皆能以協調或不 協調方式調控相應正義轉錄物。用於鑑別針對標乾之新寡 ㈣酸的策略可基_由反義寡核*酸來擊弱反義轉 錄物或調節所計晝標靶的任—其他方式。 ' 策略1:在不協調調控之情形下,擊弱反義轉錄物會增 加習用(正義)基因之表現。若後-基因編碼已知或假定藥 物乾,則可設想擊弱其反義對等部分來模擬受體激動劑或 酶刺激物之作用。 冑略2:在協調調控之情形下,可同時擊弱反義及正義 2錄物且由此協同降低習用(正義)基因表現。舉例而言, 錢用反義募㈣酸來達成擊弱,則可❹此策略來應用 —種乾向正義轉錄物之反義寡料酸絲向相應反義轉錄 物之P反義寡核普酸、或同時乾向重疊正義及反義轉錄 物之單一能量對稱性反義募核苷酸。 立根據本發明,反義化合物包含反義寡核苷酸、核酶、外 Ο P引導序列(EGS)寡核苷酸、siRNA化合物、單-哎雙鏈 職干擾(RNAi)化合物(例如仙财化合物)、及與把核酸 之至少-部分雜交並調節其功能之其他寡聚化合物。因 此,其可為DNA、RNA、類麵、類舰、或其混合物, 或可為5亥等物質中一或多者之模擬物。該等化合物可為單 ^雙鏈、%狀或髮夾型寡聚化合物’且可含有諸如内部 或末端膨脹、失配或環路等結構要素。反義化合物通常製 成直鏈形式,但可經接合或以其他方式製成環狀及/或具 支鏈形式。反義化合物可包括諸如以下構成物:經雜交以 I57514.doc -39- 201209163 、成元王或。卩分雙鍵化合物之兩條鏈,或具有足夠自體互 補性以進订雜交並形成完全或部分雙鏈化合物之單鏈。兩 條鏈可在内部連接以產生游離3,或5,末端或可連接形成連 續髮爽結構或環路。髮夾結構可在5·或3,末端含有懸垂部 刀XL長單鏈特徵。雙鏈化合物視需要可在末端包含懸垂 部分。其他修飾可包含附接至一個末端、所選核苷酸位 置糖位置或附接至一個核苷間鍵聯之偶聯基團。另一選 擇為,兩條鏈可經由非核酸部分或連接體基團進行連接。 田僅由條鏈形成時,dsRNA可呈自體互補髮夾型分子形 式其自身對折以形成雙鏈體。因此,dsRNA可為完全或 部分雙鏈。可藉由在轉基因細胞系中穩定表現dsRNA髮夾 來特異性調節基因表現,然而,在-些實施例中,基因表 見或力此絰上调。在自兩條鏈、或呈自體互補髮夾型分子 (身對折以形成雙鏈體)之單鏈形成時,兩條鏈(或單 鏈中形成雙鏈體之區域)係以Wats〇nCrick方式驗基配對之 互補RNA鏈。 引入系統中之後,本發明化合物可引發—或多種酶或結 構蛋白質之作用以實現靶核酸之裂解或其他修飾或可經由 基於佔據之機制進行作用。一般而言,核酸(包含寡核普 酸)可闡述為「類職」(亦即,通常具有一或多個2,去氧 糖及(通常)τ而非u驗基)或「類RNA」(亦即,通常呈有一 或多個2,-經基或2,_修鋅糖及(通常)1;而非τ驗基)。核酸螺 :可採用種以上之結構類型,最通常係Α型及Β型。據 般而5,具有B型樣結構之募核苷酸係「類DNA」 157514.doc -40- 201209163 且彼等具有A型樣結構者係「類RNA」。在一些(嵌合)實施 例中,反義化合物可含有A-及B型區域。 在一實施例中,期望寡核苷酸或反義化合物包括以下中 之至少一者:反義RNA、反義DNA、嵌合反義寡核苷酸、 包括經修飾鍵聯之反義寡核苷酸、干擾RNA(RNAi)、短干 擾 RNA (siRNA);微小干擾 RNA (miRNA);小時序 RNA (stRNA);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,此機制稱為「小RNA誘導之 基因活化」或RNAa。靶向基因啟動子之dsRNA可誘導有 關基因之有效轉錄活化。在使用合成dsRNA(稱為「小活 化RNA」(saRNA))之人類細胞中顯示RNAa。當前尚未瞭 解,在其他有機體中RNAa是否保守。 已發現,小雙鏈RNA (dsRNA)(例如小干擾RNA (siRNA) 及微小RNA (miRNA))係稱為RNA干擾(RNAi)之進化保守 機制的觸發物。RNAi通常經由重塑染色質以由此阻抑轉 錄來引起基因沉默,從而降解互補mRNA、或阻斷蛋白質 轉譯。然而,在詳細闡述於下文實例部分之情形下,顯示 寡核苷酸可增強BCL2結合部分3(BBC3)聚核苷酸及其編碼 產物之表現及/或功能。dsRNA亦可用作小活化RNA (saRNA)。不期望受限於理論,藉由靶向基因啟動子中之 序列,saRNA可誘導靶基因表現,此現象稱為dsRNA誘導 之轉錄活化(RNAa)。 在另一實施例中,本文所鑑別之「較佳靶區段」可用於 157514.doc -41 · 201209163 篩選調節BCL2結合部分3(BBC3)聚核苷酸之表現之額外化 合物。「調節劑」係彼等如下化合物:可降低或增加編碼 BBC3之核酸分子之表現,且至少包括與較佳靶區段互補 之5-核苷酸部分。篩選方法包括以下步驟:使編碼BBC3 之正義或天然反義聚核苷酸之核酸分子的較佳靶區段與一 或多種候選調節劑接觸,及選擇一或多種可降低或增加編 碼BBC3聚核苷酸之核酸分子(例如SEQ ID NO: 3至8)之表 現的候選調節劑。若顯示一或多種候選調節劑能夠調節 (例如降低或增加)編碼BBC3聚核苷酸之核酸分子的表現, 則該調節劑可用於BBC3聚核苷酸功能之其他調查性研 究,或用作本發明之研究、診斷、或治療藥劑。 把向天然反義序列較佳地可調節把基因之功能。例如, BBC3基因(例如,登錄號為NM_001127242)。在一實施例 中,靶係BBC3基因之反義聚核苷酸。在一實施例中,反 義寡核苷酸靶向BBC3聚核苷酸(例如,登錄號為 NM_001127242)之正義及/或天然反義序列、其變體、等位 基因、同種型、同系物、突變體、衍生物、片段及互補序 列。較佳地,寡核苷酸係反義分子且靶包含反義及/或正 義BBC3聚核苷酸之編碼及非編碼區域。 本發明之較佳靶區段亦可與本發明之其相應互補反義化 合物組合以形成穩定雙鏈(雙鏈體)寡核苷酸。 業内已顯示,該等雙鏈寡核苷酸部分可調節靶表現並經 由反義機制調控轉譯以及RNA處理。另外,雙鏈部分可經 受化學修飾。舉例而言,已顯示該等雙鏈部分可藉由雙鏈 157514.doc -42- 201209163 體之反義鏈與靶之典型雜交來抑制靶,由此觸發靶之酶降 解。 在一實施例中,反義寡核苷酸靶向BCL2結合部分 3(BBC3)聚核苷酸(例如,登錄號為NM_001127242)、其變 體、等位基因、同種型、同系物、突變體、衍生物、片段 及互補序列。較佳地,寡核苷酸係反義分子。 根據本發明實施例,靶核酸分子並不僅限於BBC3而是 擴展至BBC3分子之任一同種型、受體、同系物及諸如此Or longer fragments, modified keys, and the like. Examples of modified bonds or internucleotide linkages include thiolates, dithiophosphates, and the like. In one embodiment, the nucleotide comprises a fill derivative. The filled derivative (or modified phosphate group) that can be attached to the sugar or sugar analog moiety of the modified nucleotide of the present invention can be monophosphate, diphosphate, triphosphate, tartaric acid. Base esters, alkyl phosphates, phosphorothioates, and the like. The preparation of the above-described acid-filled analogs, and their incorporation in nucleotides, modified nucleotides and oligonucleotides are also known per se and need not be set forth herein. As is known in the art, 'the translation initiation codon is usually 5, _AUG (in the transcription of mRNA molecules; 5'-ATG in the corresponding DNA molecule), the translation initiation codon is also called "AUG codon", Start codon or rAUG start codon. A few genes have a translation initiation codon with an RNA sequence of 5'-GUG, 5I-UUG or 5I-CUG; and it has been shown that 5, _aua, 5, _acg and 5, -CUG can function in vivo. Thus, even if the starting amino acid is in each case usually thiol amide (in eukaryotes) or methionine (in prokaryotes), the terms "translation start codon" and "start" The code "" can also cover a variety of codon sequences. Eukaryotic and prokaryotic genes can have two or more alternative initiation codons, either of which can be preferentially used for translational initiation in a particular cell type or tissue or under a particular set of conditions. In the context of the present invention, the "start codon" and "translating start codon" are in vivo (4) from the transcription of the gene encoding the binding member 2 (BBC3) to the one of $ & Multiple codons, regardless of the sequence of such codons. The translation stop codon (or "stop codon") of US ^ ^ ^丞 can have two loyalty; Wei-species sequence (ie, 5'-UAA, 51-157514.doc •33· 201209163 5'- TAG and UAG and 51-UGA 'the corresponding DNA sequences are one of 5, _taa 5'-TGA, respectively. The terms "start codon region" and "translation initiation codon region" refer to about -25 to about 5 contiguous regions in the mRNA- or gene-directed direction (ie, 5 or 3) from the translation initiation codon. The bitter acid - part. Class m also #f 5 I "stop codon region" and "translation stop codon region I" refers to this 1RNA or gene in the ^: direction from the translation stop codon (ie, 5' or 3,) About 25 to about 5 adjacent nucleotides - 4 knives, the 'start codon region' (or "translation start codon region J" and "stop codon region" (or "transfer stop codon region" The antisense compound of the present invention is effective to dry to all regions. Open reading frames (〇RF) or "encoding regions" are known in the art to refer to the region between the sub-translating stop codon and the region that can be effectively targeted. In the context of the present invention The targeting region is the intra-gene region of the translation start or stop codon of the open reading frame (0RF) of the gene. The other target region contains 5, non-translated regions (5, utr), which are known in the art. In the region of the mRNA from the direction 5 of the translation initiation codon, and thus the mRNA 5, the nucleotide between the capping site and the translation initiation codon (or another target region of the corresponding nucleotide on the gene) Contains the 3, non-translated region (3'UTR) known in the art, which refers to the portion of the mRNA in the direction from the translation stop codon 3, and thus contains the translation stop codon of the mRNA and the 3' end Nucleotide (or the corresponding nucleotide on the gene). mRNA25, the capping site includes 5, 5, -3, triphosphate linkage to the mRNA 5, _ the most residual 157514.doc • 34· 201209163 N7-T-based bird bitter residue. mRNAi5, capped area is considered to contain the capping structure itself and The cap site is adjacent to the previous 5 nucleotides. Another dry region of the invention is the 5' capped region. Although some eukaryotic mRNA transcripts can be directly translated, many true nuclear organisms 111 ft 1 transcript Contains one or more regions that are cleaved from the transcript prior to translation and are referred to as "introns." The thorny (and thus translated) regions are referred to as "exons" and spliced together to form a contiguous mRNA sequence. In one embodiment, a targeted splice site (ie, an intron-exon node or an exon-intron node) is particularly useful for aberrant splicing associated with disease or overproduction of a particular splicing product In a disease-related situation, another embodiment of an abnormal fusion node that is due to rearrangement or deletion is a target site. mRNA transcripts generated by splicing two (or more) mRNAs from different gene sources For "fusion transcripts", antisense compounds that target, for example, 1) or 111} 前^ precursors can be used to effectively target introns. In one embodiment, the antisense oligonucleotide binds to the Q and/or non-coding region of the target polynucleotide 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 the sense polynucleotide and modulates the expression and/or function of the molecule. Alternative RNA transcripts can be produced from the same genomic region of DNA. Such alternative transcripts are commonly referred to as "variants." More specifically, the "1111^^8 precursor variant" is a transcript produced from the same genomic DNA, which has a starting or stopping position with respect to other transcripts generated from the same genomic DNA 157514.doc •35 · 201209163 is different and contains intron and exon sequences. When one or more exons or intron regions, or portions thereof, are cleaved during splicing, the mRNA precursor variants produce smaller "mRNA variants". Thus, mRNA variants of treated mRNA precursor variants and each unique mRNA precursor variant must always produce a unique mRNA variant by splicing. Such mRNA variants are also referred to as "alternate splice variants". If the mRNA precursor variant is not spliced, the mRNA precursor variant is identical to the mRNA variant. Variants can be generated via the use of alternating signals that initiate or stop transcription. The mRNA steroid and mRNA may have more than one start codon or stop post code. Variants derived from mRNA precursors or mRNAs using alternative initiation codons are referred to as "alternative initiation variants" of mRNA precursors or mRNA. Their use of alternative stop codon transcripts is referred to as an "alternative stop variant" of the mRNA precursor or mRNA. One specific type of alternative stop variant is a "polyA variant" in which multiple transcripts are derived from alternatives to one of the "polyA stop signals" of the transcriptional machinery, resulting in a unique polyA position. Point-terminated transcript. Above the invention, the types of variants described herein are also examples of target nucleic acids. The position on the target nucleic acid that hybridizes to the antisense compound is defined as the portion of the target region that is 5 nucleotides long that is targeted by the active antisense compound. Although specific sequences of certain exemplary target segments are set forth herein, those skilled in the art will recognize that such sequences are used to illustrate and exemplify particular embodiments within the scope of the invention. Other target segments can be readily identified by those skilled in the art in light of this disclosure. A target segment of 5 to 100 nucleotides in length and comprising at least five (5) 157514.doc - 36 - 201209163 contiguous nucleotides selected from the preferred interpretive target segments is also considered suitable for dryness. The target segment may comprise a DNA or RNA sequence comprising at least 5 contiguous nucleotides from the 5'-end of one of the preferred interpretable target segments (remaining nucleotide acid is contiguous in the same DNA or RNA) A segment that begins immediately upstream of the 5'-end of the target segment and continues until the DNA or RNA contains from about 5 to about 1 nucleotide. Similarly, a preferred target segment is represented as a DNA or RNA sequence comprising at least 5 contiguous nucleotides from the 3, _ terminus of one of the preferred interpretable target segments (the remaining nucleotide is the same DNA) Or a continuous segment of the RNA 'which is immediately downstream of the 3, _ terminal of the target segment and continues until the DNA or RNA contains about 5 to about 1 nucleotide.) Those skilled in the art can identify other preferred dry 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., hybridized sufficiently and sufficiently specific) can be selected to achieve the desired effect. In an embodiment of the invention, the raised nucleotide binds to the antisense strand of a particular target. The nucleotides are raised to a length of at least 5 nucleotides and can be synthesized such that each nucleus is directed to an overlapping sequence' whereby the oligo-acid is synthesized to cover the entire length of the dry polynucleotide. The target also contains both coding and non-coding regions. In one embodiment, the antisense oligonucleotide preferably targets a specific nucleic acid. The targeting of antisense compounds to specific nucleic acids is a multi-step process. This process typically begins with the identification of the nucleic acid sequence for which the function is to be modulated. This may, for example, be a cellular gene (or 157514.doc-37-201209163 mRNA from gene transcription) or a non-coding polynucleotide (eg, non-coding RNA (ncRNA)) that is associated with a particular condition or disease state. . RNA can be classified into (1) messenger RNA (mRNA), which is translated into protein; and (2) non-protein-encoding RNA (ncRNA). ncRNAs include microRNAs, antisense transcripts, and other transcription units (TUs) that contain high-density stop codons and lack any extensive "open reading frame". Many ncRNAs appear to start at the promoter site in the 3' non-translated region (3'UTR) of the locus encoding the protein. ncRNAs are generally rare and at least half of the ncRNAs sequenced by the FANTOM Alliance appear to be unpolyadenylated. Most researchers pay attention to the polyadenosylated mRNA that is processed and exported to the cytoplasm for obvious reasons. Recently, the group showing non-polyadenylated nuclear RNA may be extremely large, and many of these transcripts are derived from so-called intergenic regions. The mechanism by which ncRNA regulates gene expression is base pairing with the target transcript. RNAs that function by base pairing can be grouped into: (1) cis-encoded RNA, which encodes at the same genetic position at which it functions, but encodes at the relative bond of the RNA and thus shows complete complementarity with its stem And (2) trans-encoded RNA, which encodes at a different chromosomal location than the RNA it functions with and typically does not exhibit full base pairing potential with its target. Without wishing to be bound by theory, the interference of antisense oligonucleotides described herein with antisense polynucleotides may alter the performance of the corresponding sense messenger RNA. However, this regulation can be uncoordinated (increased messenger RNA caused by knockdown) or coordinated (antisense weakening causes concomitant messenger RNA reduction). In such cases, the antisense oligonucleotide can target overlapping or non-overlapping portions of the antisense transcript to create a weak or sequestration. The encoding of 157514.doc -38-201209163 and non-coded antisense transcripts can be performed in the same manner, and any species can regulate the corresponding sense transcripts in a coordinated or uncoordinated manner. The strategy used to identify new oligo (tetra) acids against the stem can be based on any other way of weakening antisense transcripts or modulating the target by antisense oligo-acids. Strategy 1: In the case of uncoordinated regulation, weakening antisense transcripts increases the performance of the (just) gene. If the post-gene coding is known or assumed to be dry, it is conceivable to weaken its antisense counterpart to mimic the effects of a receptor agonist or an enzyme stimulator. Strategy 2: In the case of coordinated regulation, both antisense and justice can be weakened at the same time and thus synergistically reduce the expression of the (just) gene. For example, if the money is used to achieve a weakening by antisense (4) acid, then this strategy can be applied to the antisense oligodeoxynucleotide of the antisense oligos of the sense transcript to the antisense oligo of the corresponding antisense transcript. A single energy symmetric antisense raised nucleotide that is acid, or simultaneously dry, overlapping overlapping sense and antisense transcripts. According to the present invention, antisense compounds include antisense oligonucleotides, ribozymes, exo-P-directing sequence (EGS) oligonucleotides, siRNA compounds, and mono-哎 double-stranded interference (RNAi) compounds (eg, Xiancai) Compounds), and other oligomeric compounds that hybridize to at least a portion of a nucleic acid and modulate its function. Thus, it may be DNA, RNA, a flank, a class ship, or a mixture thereof, or may be a mimetic of one or more of substances such as 5 hai. Such compounds may be mono-double-stranded, %-like or hairpin-type oligomeric compounds' and may contain structural elements such as internal or terminal expansion, mismatch or loop. Antisense compounds are typically made in a linear form, but may be joined or otherwise formed into a cyclic and/or branched form. Antisense compounds can include, for example, the following: hybridization with I57514.doc-39-201209163, Chengyuanwang or. The two strands of the double bond compound are split, or have a single autoligand sufficient to complement the hybridization and form a single strand of the fully or partially double stranded compound. The two chains can be internally connected to create a free 3, or 5, end or connectable to form a continuous cooling structure or loop. The hairpin structure can be at 5 or 3, and the end has a pendant single XL long single chain feature. The double-stranded compound may contain an overhanging moiety at the end as needed. Other modifications may include attachment to one end, a selected nucleotide position, or a coupling group attached to an internucleoside linkage. Alternatively, the two strands can be joined via a non-nucleic acid moiety or a linker group. When the field is formed only by a strand, the dsRNA may be in the form of an auto-complementary hairpin-type molecule that folds itself to form a duplex. Thus, the dsRNA can be fully or partially double stranded. Gene expression can be specifically regulated by the stable expression of dsRNA hairpins in transgenic cell lines, however, in some embodiments, the genes are shown or stressed up. In the formation of a single strand from two strands, or an auto-complementary hairpin-type molecule (folded to form a duplex), the two strands (or regions that form a duplex in a single strand) are Wats〇nCrick Method of pairing complementary RNA strands. Following introduction into the system, the compounds of the invention may elicit - or a plurality of enzymes or structural proteins to effect cleavage or other modification of the target nucleic acid or may act via a mechanism based on occupancy. In general, nucleic acids (including oligonucleotides) can be described as "class jobs" (ie, usually have one or more 2, deoxy sugars and (usually) τ instead of u-test) or "RNA-like" (i.e., usually one or more 2,-trans-based or 2,-zinc-plated sugar and (usually) 1; not a τ-test group). Nucleic acid snails: More than one type of structure can be used, most commonly Α type and Β type. As a general rule, the nucleotide type "DNA" of the B-type structure is 157514.doc -40-201209163 and those having the A-type structure are "like RNA". In some (chimeric) embodiments, the antisense compound can contain both A- and B-type regions. In one embodiment, the desired oligonucleotide or antisense compound comprises at least one of the following: an antisense RNA, an antisense DNA, a chimeric antisense oligonucleotide, an antisense oligonucleotide comprising a modified linkage Glycosidic acid, interfering RNA (RNAi), short interfering RNA (siRNA); small interfering RNA (miRNA); small temporal RNA (stRNA); or short hairpin RNA (shRNA); small RNA-induced gene activation (RNAa); small Activated RNA (saRNA), or a combination thereof. dsRNA also activates gene expression, a mechanism known as "small RNA-induced gene activation" or RNAa. A dsRNA targeting a gene promoter can induce efficient transcriptional activation of the gene. RNAa is displayed in human cells using synthetic dsRNA (referred to as "small active RNA" (saRNA). It is not yet known whether RNAa is conserved in other organisms. Small double-stranded RNA (dsRNA), such as small interfering RNA (siRNA) and microRNA (miRNA), have been found to be triggers of an evolutionarily conserved mechanism of RNA interference (RNAi). RNAi typically causes gene silencing by remodeling chromatin to thereby suppress transcription, thereby degrading complementary mRNA, or blocking protein translation. However, in the context of the detailed description of the Examples section below, the display oligonucleotides enhance the performance and/or function of the BCL2 binding portion 3 (BBC3) polynucleotide and its encoded product. dsRNA can also be used as a small activating RNA (saRNA). Without wishing to be bound by theory, saRNA can induce target gene expression by targeting sequences in gene promoters, a phenomenon known as dsRNA-induced transcriptional activation (RNAa). In another embodiment, the "preferred target segment" identified herein can be used to screen for additional compounds that modulate the expression of a BCL2 binding moiety 3 (BBC3) polynucleotide. "Modulators" are compounds which reduce or increase the performance of a nucleic acid molecule encoding BBC3 and include at least a 5-nucleotide portion that is complementary to a preferred target segment. The screening method comprises the steps of contacting a preferred target segment of a nucleic acid molecule encoding a sense or natural antisense polynucleotide of BBC3 with one or more candidate modulators, and selecting one or more to reduce or increase the encoding of the BBC3 polynucleus Candidate modulators of the expression of nucleic acid molecules of glucosinolates (e.g., SEQ ID NOS: 3 to 8). If one or more candidate modulators are shown to be capable of modulating (eg, reducing or increasing) the performance of a nucleic acid molecule encoding a BBC3 polynucleotide, the modulator can be used in other investigative studies of BBC3 polynucleotide function, or as a A research, diagnostic, or therapeutic agent of the invention. The function of the gene is preferably adjusted to the natural antisense sequence. For example, the BBC3 gene (for example, accession number is NM_001127242). In one embodiment, the target is an antisense polynucleotide of the BBC3 gene. In one embodiment, the antisense oligonucleotide targets a sense and/or natural antisense sequence of a BBC3 polynucleotide (eg, accession number NM_001127242), variants, alleles, isoforms, homologs thereof , mutants, derivatives, fragments and complementary sequences. Preferably, the oligonucleotide is an antisense molecule and the target comprises the coding and non-coding regions of the antisense and/or sense BBC3 polynucleotide. Preferred target segments of the invention may also be combined with their corresponding complementary antisense compounds of the invention to form stable double-stranded (duplex) oligonucleotides. It has been shown in the art that these double-stranded oligonucleotide moieties can modulate target expression and regulate translation and RNA processing via antisense mechanisms. In addition, the double-stranded moiety can be chemically modified. For example, it has been shown that the double-stranded portions can inhibit the target by typical hybridization of the antisense strand of the double stranded 157514.doc-42-201209163 to the target, thereby triggering the enzymatic degradation of the target. In one embodiment, the antisense oligonucleotide targets a BCL2 binding portion 3 (BBC3) polynucleotide (eg, accession number NM_001127242), variants thereof, alleles, isoforms, homologs, mutants , derivatives, fragments and complementary sequences. Preferably, the oligonucleotide is an antisense molecule. According to an embodiment of the invention, the target nucleic acid molecule is not limited to BBC3 but extends to any isoform, receptor, homologue, and the like of the BBC3 molecule.

在一實施例中,寡核苷酸靶向BBC3聚核苷酸之天然反 義序列(例如,闡述為SEQ ID NO: 2之聚核苷酸)、及其任 一變體、等位基因、同系物、突變體、衍生物、片段及互 補序列。將反義寡核苷酸之實例闡述為SEQ ID NO: 3至 8 ° 在一個實施例中,寡核苷酸與BBC3反義分子之核酸序 列(包含(不限於)與BBC3聚核苷酸有關之非編碼正義及/或 反義序列)互補或結合並調節BBC3分子之表現及/或功能。 在一實施例中,寡核苷酸與BBC3天然反義分子之核酸 序列(闡述為SEQ ID NO: 2)互補或結合並調節BBC3分子之 表現及/或功能。 在一實施例中,寡核苷酸包括SEQ ID NO: 3至8中至少5 個連續核苷酸之序列並調節BBC3分子之表現及/或功能。 聚核苷酸靶包括BBC3(包含其家族成員)、BBC3之變 體;BBC3之突變體,包含SNP ; BBC3之非編碼序列; 157514.doc -43- 201209163 BBC3之等位基因;物種變體、片段及諸如此類。較佳 地,寡核苷酸係反義分子。 在一實施例中,靶向BBC3聚核苷酸之募核苷酸包括: 反義RNA、干擾RNA (RNAi)、短干擾RNA (siRNA);微小 干擾RNA (miRNA);小時序rnA (stRNA);或短髮夾RNA (shRNA);小RNA誘導之基因活化(RNAa);或小活化RNA (saRNA)。 在一實施例中’靶向BCL2結合部分3(BBC3)聚核苷酸 (例如SEQ ID NO : 2至8)可調節該等靶之表現或功能。在 一個實施例中,與對照組相比上調表現或功能。在一實施 例中,與對照組相比下調表現或功能。 在一實施例中,反義化合物包括闡述為SEQ ID NO: 3至 8之序列。該等寡核苷酸可包括一或多個經修飾核苷酸、 較短或較長片段、經修飾鍵及諸如此類。 在一實施例中’ SEQ ID NO·· 3至8包括一或多個LNA核 苷酸。表1顯示用於本發明方法中之實例性反義募核苷 酸。 表1 :In one embodiment, the oligonucleotide targets a native antisense sequence of a BBC3 polynucleotide (eg, a polynucleotide set forth as SEQ ID NO: 2), any variant thereof, allele, Homologs, mutants, derivatives, fragments and complementary sequences. An example of an antisense oligonucleotide is set forth as SEQ ID NO: 3 to 8 °. In one embodiment, the nucleic acid sequence of the oligonucleotide and the BBC3 antisense molecule (including, without limitation, is associated with a BBC3 polynucleotide) The non-coding sense and/or antisense sequences complement or bind to and modulate the expression and/or function of the BBC3 molecule. In one embodiment, the oligonucleotide complements or binds to the nucleic acid sequence of BBC3 natural antisense molecule (described as SEQ ID NO: 2) and modulates the expression and/or function of the BBC3 molecule. In one embodiment, the oligonucleotide comprises the sequence of at least 5 contiguous nucleotides of SEQ ID NOs: 3 to 8 and modulates the expression and/or function of the BBC3 molecule. Polynucleotide targets include BBC3 (including members of its family), variants of BBC3; mutants of BBC3, including SNP; non-coding sequences of BBC3; 157514.doc-43-201209163 alleles of BBC3; species variants, Fragments and the like. Preferably, the oligonucleotide is an antisense molecule. In one embodiment, the nucleotides that target the BBC3 polynucleotide include: antisense RNA, interfering RNA (RNAi), short interfering RNA (siRNA); micro interfering RNA (miRNA); small temporal rnA (stRNA) Or short hairpin RNA (shRNA); small RNA-induced gene activation (RNAa); or small activating RNA (saRNA). In one embodiment, 'targeting BCL2 binding moiety 3 (BBC3) polynucleotides (e. g., SEQ ID NOs: 2 to 8) can modulate the performance or function of such targets. In one embodiment, the performance or function is up-regulated compared to a control group. In one embodiment, the performance or function is downregulated as compared to the control group. In one embodiment, the antisense compound comprises the sequence set forth as SEQ ID NOs: 3 to 8. Such oligonucleotides can include one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. In one embodiment, SEQ ID NOs. 3 to 8 include one or more LNA nucleotides. Table 1 shows exemplary antisense nucleotides for use in the methods of the invention. Table 1 :

序列編號 反義序列名稱 序列 SEQIDNO:3 CUR-1675 C*C*A*C*C*A*A*G*C*C*A*G*A*T*T*C*C*C*A*C SEQ ID NO: 4 CUR-1676 A*T*C*Cs|tA*C*Csf:C:iiA*T*C*Ti!!C*G*G*C*Cs,eT*T*C SEQ ID NO: 5 CUR-1677 A*G*G*C*T*C*A*A*G*C*G*T*T*C*C*T*C*C*C*A SEQ ID NO: 6 CUR-1678 C*C*C*A5,!G*T*T*A*G*G*T*T*T!|:C*C*G*C*A*T!i!G*T SEQ ID NO: 7 CUR-1679 T*C*T*G*Ti|iC*G*C*Cs|eC*A*A*Gs|!C*T*G*G*A*G*T SEQ ID NO: 8 CUR-1505 T*C*T*G*T*C*G*C*C*C*A*A*G*C*T*G*G*A*G*T 157514.doc -44 - 201209163 可以業内已知之若干方式來調節期望靶核酸。舉例而 言,使用反義寡核苦酸、siRNA等。酶性核酸分子(例如, 核酶)係能夠催化二或多種不同反應之核酸分子,包含能 夠以核苷酸鹼基序列特異性方式重複裂解其他單獨核酸分 子。該酶性核酸分子可用於(例如)靶向實質上任一 RNA轉 錄物。 因具有序列特異性,故反式裂解之酶性核酸分子可顯示 用作人類疾病之治療劑的前景。可設計酶性核酸分子來裂 解細胞RNA背景内之特異性RNA靶。此一裂解事件使得 mRNA失去功能性並去除來自該RNA之蛋白質表現。以此 方式可選擇性抑制與疾病狀態有關之蛋白質的合成。 一般而言,具有RNA裂解活性之酶性核酸藉由首先結合 至靶RNA來發揮作用。該結合經由酶性核酸之靶結合部分 來進行,該靶結合部分緊鄰分子中用於裂解靶RNA之酶性 部分。因此,酶性核酸首先識別靶RNA且然後經由互補鹼 基配對與靶RNA結合,且在結合至確切位點後以酶促方式 發揮作用以切割靶RNA。此一靶RNA之裂解策略將破壞其 引導合成所編碼蛋白之能力。酶性核酸已結合且裂解其 RNA靶之後,其自該RNA釋放以尋找另一靶且可重複結合 及裂解新靶。 已使用諸如活體外選擇(演變)策略(Orgel,(1979) Proc_ R. Soc. London, B 205, 43 5)等若干方式來產生能夠催化各 種反應(例如磷酸二酯鍵聯及醯胺鍵聯之裂解及連接)的新 核酸觸媒。 157514.doc -45- 201209163 對於具有最適催化活性之核酶之研發將顯著有助於採用 RNA裂解性核酶來調控基因表現的任一策略。舉例而言, 錘頭狀核酶在飽和(1〇 mM)濃度之Mg2+輔因子存在下以約 1 min-1之催化速率(kcat)發揮作用。已顯示人工「rna連 接酶」核酶可以約丨00 min_丨之速率催化相應自修飾反應。 此外,已知某些經修飾錘頭狀核酶具有由DNA構成之受質 結合臂,其可以接近1〇〇 min—〗之多倍周轉速率催tRNA裂 解。最後,使用某些核苷酸類似物代替錘頭狀核酶催化核 心内之特定殘基可產生經修飾核酶,其催化速率顯示提高 多達10倍。該等發現表明,核酶可以顯著大於大部分天然 自裂解核酶在活體外所顯示之催化速率的催化速率來促進 化學轉化。然後可優化某些自裂解核酶之結構以得到最大 催化活性,或可製備顯示顯著較快之RNA磷酸二酯裂解速 率之全新RNA基序。 符合「錘頭」模型之RNA觸媒催化之RNA受質的分子間 裂解首次顯示於1987年(Uhlenbeck,〇 c 〇987) _⑽, 328: 596-600)。回收RNA觸媒並使其與多個rna分子進行 反應,從而表明其確實具有催化性。 基於「錘頭」基序設計之催化RNA已用於裂解特異性乾 序列’其係藉由在催化RNA中作出適當驗基改變以維持與 乾序列之必錢基輯來達成。此使料使㈣化rna來 裂解特異錄序列,且表明根據「錘頭」模型設計之催化 RNA可在活體内裂解特異性受質RNa。 RNA干擾(RNAi)已成為調節哺乳動物及哺乳動物細胞中 157514.doc -46- 201209163 之基因表現之有效工具。此方式需要使用表現質粒或病毒 及用於處理成siRNA之小髮夾RNA的編碼序列以RNA自身 或DNA形式來遞送小干擾RNA (siRNA)。此系統使得能夠 將siRNA前體有效輸送至其具有活性之細胞質中且容許使 用用於基因表現之經調控及組織特異性啟動子。 在一實施例中,寡核苷酸或反義化合物包括核糖核酸 (RNA)及/或去氧核糖核酸(DNA)之寡聚物或聚合物、或其 模擬物、嵌合體、類似物或同系物。此術語包含由天然核 苷酸、糖及共價核苷間(主鏈)鍵聯組成之募核苷酸;以及 具有非天然部分且以相似方式發揮作用之募核苷酸。該等 經修飾或經取代寡核苷酸通常優於天然形式,此乃因諸如 增強之細胞攝取、對於靶核酸之增強之親和力及在核酸酶 存在下之增加之穩定性等期望性質。 根據本發明,寡核苷酸或「反義化合物」包含反義寡核 苷酸(例如RNA、DNA、其模擬物、嵌合體、類似物或同 系物)、核酶、外部引導序列(EGS)寡核苷酸、siRNA化合 物、單鏈或雙鏈RNA干擾(RNAi)化合物(例如siRNA化合 物)、saRNA、aRNA、及其他與乾核酸之至少一個部分雜 交並調節其功能之寡聚化合物。因此,其可為DNA、 RNA、類DNA、類RNA、或其混合物,或可為該等物質中 一或多者之模擬物。該等化合物可為單鏈、雙鏈、環狀或 髮夹募聚化合物,且可含有諸如内部或末端膨脹、失配或 環路等結構要素。反義化合物通常製成直鏈形式,但可經 接合或以其他方式製成環狀及/或具支鏈形式。反義化合 157514.doc 47· 201209163 物可包含諸如以下構成物:經雜交以形成完全或部分雙鏈 化合物之兩條鏈,或具有足夠自體互補性以進行雜交並形 成完全或部分雙鏈化合物之單鏈。兩條鏈可在内部連接以 產生游離1或5,末端或可連接形成連續髮夾結構或環路。 髮夾結構可在5,或3,末端含有懸垂部分以延長單鏈特徵。 雙鏈化合物視需要可在末端包含懸垂部分。其他修飾可包 含附接至一個末端、所選核苷酸位置、糖位置或附接至一 個核苷間鍵聯之偶聯基團。另一選擇為,兩條鏈可經由非 核酸部分或連接體基團進行連接。在自僅一條鏈形成時, dsRNA可呈自體互補髮夾型分子形式,其自身對折以形成 雙鏈體。因此,dsRNA可為完全或部分雙鏈。可藉由穩定 表現轉基因細胞系中之dsRNA髮夾來特異性調節基因表 現。在自兩條鏈、或呈自體互補髮夾型分子形式(自身對 折以形成雙鏈體)之單鏈形成時,兩條鏈(或單鏈中形成雙 鏈體之區域)係以Watson_Crick方式鹼基配對之互補 鏈。 引入系統中之後,本發明化合物可引發—或多種酶或結 構蛋白質之作用以實現靶㈣之裂解或其他修飾或可經由° 基於佔據之機制進行作用。一般而言,核酸(包含寡核# 酸)可闡述為「類腦」(亦即,通常具有一或多個2,_去氧 糖及(通常)Τ而非U驗基)或「類舰」(亦即,通常具有一 或多個2’-經基或2|_修飾糖及(通常川而非τ驗幻。核酸螺 旋可採用-種以上之結構類型,最通常係Α型及_。據 仏 奴而。,具有B型樣結構之寡核苷酸係「類DNA , 157514.doc -48- 201209163 Ο 且彼等具有_樣結構者係「類rna」h 例中,反義化合物可含有八_及6型區域。— 本發月之反義化合物可包括長度為約5至約肋個核苷酸 (亦即,約5至約80個連接核普)的反義部分。此係指反義化 合物之反義鏈或部分之長度。換言之,本發明之單鍵反義 化合物包括5至細個核苦酸,且本發明之雙鏈反義化合 物(例如,dsRNA)包括長度為5至約8〇個核芽酸之正義及反 義鏈或部分。熟習此項技術者應瞭解,此涵蓋長度為5、 10、11、12、13、14、15、16、17、18、 6、7、 19、20 31 43 55 67 32 44 56 68 :、9 ‘21 33 45 57 69、 22 34 46 58 23 35 47 59 24 36 48 60 25 37 49 61 26 38 50 62 27 39 51 63 28 40 52 64 29 41 53 65 30 42 54 66 Ο 70 、 71 、 72 、 73 、 74 、 75 、 76 、 77 、 78 、 79 或80個核苷酸、或其任一範圍之反義部分。 在個貫細例中,本發明之反義化合物具有長度為i 〇至 50個核菁酸之反義部分。㉟習此項技術者應瞭解,此展現 反義部分長度為 1〇、u、12、13、14、15、16、17、18、 19、20、21 ' 22、23、24、25、26、27、28、29、30、 31、32、33、34、35、36、37、38、39、40、41、42、 43 ' 44 ' 45 ' 46、47、48、49或50個核苷酸、或其任一範 圍之寡核苷酸。在一些實施例中,寡核苷酸之長度為15個 核普酸。 在一個實施例中’本發明之反義或募核苷酸化合物具有 157514.doc •49· 201209163 長度為12或13至30個核苷酸的反義部分。熟習此項技術者 應瞭解’此展現反義部分長度為12、13、14、15、16、 17 、 18 、 19 、 20 、 21 、 22 、 23 、 24 、 25 、 26 、 27 、 28 、 29 或3 0個核苷酸、或其任一範圍之反義化合物。 在一實施例中,本發明之寡聚化合物亦包含在該化合物 中之一或多個核苷酸位置存在不同鹼基的變體。舉例而 s ’若第一核苷酸係腺嘌呤,則可產生在此位置含有胸 苦、鳥皆或胞苦之變體。此可發生於反義或dsRNA化合物 之任一位置。然後使用本文所述方法測試該等化合物以測 疋其抑制乾核酸表現之能力。 在一些實施例中,反義化合物與靶之間之同源性、序列 一致性或互補性為約4〇%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%。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-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’反義募核苷酸(例如,SEQ ID NO: 3至8 中所述之核酸刀子)包括—或多個取代或修飾。在一個實 施例中,核苷酸經鎖核酸(LNA)取代。Sequence number antisense sequence name sequence SEQ ID NO: 3 CUR-1675 C*C*A*C*C*A*A*G*C*C*A*G*A*T*T*C*C*C*A *C SEQ ID NO: 4 CUR-1676 A*T*C*Cs|tA*C*Csf:C:iiA*T*C*Ti!!C*G*G*C*Cs,eT*T*C SEQ ID NO: 5 CUR-1677 A*G*G*C*T*C*A*A*G*C*G*T*T*C*C*T*C*C*C*A SEQ ID NO : 6 CUR-1678 C*C*C*A5,!G*T*T*A*G*G*T*T*T!|:C*C*G*C*A*T!i!G* T SEQ ID NO: 7 CUR-1679 T*C*T*G*Ti|iC*G*C*Cs|eC*A*A*Gs|!C*T*G*G*A*G*T SEQ ID NO: 8 CUR-1505 T*C*T*G*T*C*G*C*C*C*C*A*A*G*C*T*G*G*A*G*T 157514.doc - 44 - 201209163 The desired target nucleic acid can be modulated in a number of ways known in the art. For example, antisense oligonucleotides, siRNA, and the like are used. An enzymatic nucleic acid molecule (e.g., a ribozyme) is a nucleic acid molecule capable of catalyzing two or more different reactions, including the ability to repeatedly cleave other individual nucleic acid molecules in a nucleotide base sequence-specific manner. The enzymatic nucleic acid molecule can be used, for example, to target substantially any RNA transcript. Due to their sequence specificity, trans-cleaved enzymatic nucleic acid molecules can be used as a therapeutic agent for human diseases. Enzymatic nucleic acid molecules can be designed to cleave specific RNA targets within the cellular RNA background. This cleavage event causes the mRNA to lose functionality and remove protein expression from the RNA. In this way, the synthesis of proteins associated with disease states can be selectively inhibited. In general, an enzymatic nucleic acid having an RNA cleavage activity functions by first binding to a target RNA. The binding is carried out via a target binding portion of an enzymatic nucleic acid that is immediately adjacent to the enzymatic portion of the molecule used to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes the target RNA and then binds to the target RNA via a complementary base pair and acts enzymatically to cleave the target RNA upon binding to the exact site. This cleavage strategy for a target RNA will disrupt its ability to direct synthesis of the encoded protein. After the enzymatic nucleic acid has bound and cleaves its RNA target, it is released from the RNA to find another target and can repeatedly bind and cleave the new target. Several methods such as in vitro selection (evolution) strategies (Orgel, (1979) Proc_R. Soc. London, B 205, 43 5) have been used to generate various reactions (eg phosphodiester linkages and guanamine linkages) New nucleic acid catalyst for cleavage and ligation). 157514.doc -45- 201209163 The development of ribozymes with optimal catalytic activity will significantly contribute to any strategy that uses RNA lytic ribozymes to regulate gene expression. For example, hammerhead ribozymes act at a catalytic rate (kcat) of about 1 min-1 in the presence of a saturated (1 mM) concentration of Mg2+ cofactor. Artificial "rna ligase" ribozymes have been shown to catalyze the corresponding self-modification reactions at a rate of about 00 min. In addition, it is known that certain modified hammerhead ribozymes have a binding arm composed of DNA that can modulate tRNA cleavage by a multiple of the turnover rate of 1 〇〇 min. Finally, the use of certain nucleotide analogs in place of hammerhead ribozymes to catalyze specific residues within the core produces modified ribozymes that exhibit up to a 10-fold increase in catalytic rate. These findings indicate that ribozymes can significantly promote chemical conversion by significantly greater than the catalytic rate of the catalytic rate exhibited by most naturally occurring cleavage ribozymes in vitro. The structure of some self-cleaving ribozymes can then be optimized for maximum catalytic activity, or a novel RNA motif that exhibits a significantly faster rate of RNA phosphodiester cleavage can be prepared. The intermolecular cleavage of RNA receptors catalyzed by RNA catalysts in accordance with the "hammerhead" model was first shown in 1987 (Uhlenbeck, 〇 c 〇 987) _(10), 328: 596-600). The RNA catalyst is recovered and reacted with a plurality of rna molecules to indicate that it is indeed catalytic. Catalytic RNA based on the "hammerhead" motif design has been used to cleave specific stem sequences' which is achieved by making appropriate base changes in the catalytic RNA to maintain the necessary sequence with the stem sequence. This allows the (iv) rna to cleave the specific sequence and demonstrates that the catalytic RNA designed according to the "hammerhead" model can cleave the specific receptor Rna in vivo. RNA interference (RNAi) has become an effective tool for regulating gene expression in mammalian and mammalian cells 157514.doc -46- 201209163. This approach requires the delivery of small interfering RNA (siRNA) in the form of RNA itself or DNA using expression plasmids or viruses and coding sequences for small hairpin RNAs that are processed into siRNA. This system enables efficient delivery of siRNA precursors to their active cytoplasm and allows the use of regulated and tissue-specific promoters for gene expression. In one embodiment, the oligonucleotide or antisense compound comprises an oligo or polymer of ribonucleic acid (RNA) and/or deoxyribonucleic acid (DNA), or a mimetic, chimera, analog or homolog thereof Things. This term encompasses nucleotides consisting of natural nucleotides, sugars, and covalent internucleoside (backbone) linkages; and nucleotides that have non-natural portions and function in a similar manner. Such modified or substituted oligonucleotides are generally preferred over native forms due to desirable properties such as enhanced cellular uptake, enhanced affinity for target nucleic acids, and increased stability in the presence of nucleases. According to the invention, an oligonucleotide or "antisense compound" comprises an antisense oligonucleotide (eg RNA, DNA, mimetic, chimera, analog or homolog thereof), ribozyme, external leader sequence (EGS) Oligonucleotides, siRNA compounds, single- or double-stranded RNA interference (RNAi) compounds (eg, siRNA compounds), saRNAs, aRNAs, and other oligomeric compounds that hybridize to and modulate at least one portion of a dry nucleic acid. Thus, it can be DNA, RNA, DNA-like, RNA-like, or a mixture thereof, or can be a mimetic of one or more of such substances. The compounds may be single chain, double stranded, cyclic or hairpin polymeric compounds and may contain structural elements such as internal or terminal expansion, mismatch or loop. Antisense compounds are typically made in a linear form, but may be joined or otherwise formed into a cyclic and/or branched form. Antisense 157514.doc 47· 201209163 may comprise a construct such as two strands that are hybridized to form a fully or partially double-stranded compound, or have sufficient auto-complementation to hybridize and form a fully or partially double-stranded compound. Single chain. The two chains can be internally connected to create a free 1 or 5, and the ends can be joined to form a continuous hairpin structure or loop. The hairpin structure may have an overhang at the end of 5, or 3 to extend the single-strand feature. The double-stranded compound may contain an overhang at the end as needed. Other modifications may include attachment to one end, a selected nucleotide position, a sugar position, or a coupling group attached to an internucleoside linkage. Alternatively, the two strands can be joined via a non-nucleic acid moiety or a linker group. Upon formation from only one strand, the dsRNA can be in the form of an auto-complementary hairpin-type molecule that folds in itself to form a duplex. Thus, the dsRNA can be fully or partially double stranded. The gene expression can be specifically regulated by stably expressing the dsRNA hairpin in the transgenic cell line. When formed from a single strand of two strands, or in the form of an auto-complementary hairpin-type molecule (self-folding to form a duplex), the two strands (or regions forming a duplex in a single strand) are in Watson_Crick mode. The complementary strand of base pairing. Following introduction into the system, the compounds of the invention may elicit - or a plurality of enzymes or structural proteins to effect cleavage or other modification of the target (d) or may act via a mechanism based on occupancy. In general, nucleic acids (including oligonucleic acids) can be described as "brain-like" (ie, usually having one or more 2,_deoxy sugars and (usually) Τ instead of U-killers) or "class ships" (ie, usually with one or more 2'-trans-based or 2|-modified sugars and (usually Chuanchuan instead of τ illusion. Nucleic acid helix can be used in more than one type of structure, most commonly Α type and _ According to 仏 slave, the oligonucleotides with B-like structure are "DNA-like, 157514.doc -48- 201209163 且 and they have _-like structure "Rna" h, antisense compounds The VIII and 6 regions may be included. - The antisense compound of this month may comprise an antisense portion of from about 5 to about rib nucleotides (i.e., from about 5 to about 80 linked nucleops). Means the length of the antisense strand or portion of the antisense compound. In other words, the single bond antisense compound of the invention comprises 5 to a fine nucleotide acid, and the double-stranded antisense compound of the invention (eg, dsRNA) comprises a length of 5 to about 8 正义 sense and antisense strands or parts of nuclear phytic acid. Those skilled in the art should understand that this covers a length of 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 6, 7, 19, 20 31 43 55 67 32 44 56 68 :, 9 '21 33 45 57 69, 22 34 46 58 23 35 47 59 24 36 48 60 25 37 49 61 26 38 50 62 27 39 51 63 28 40 52 64 29 41 53 65 30 42 54 66 Ο 70 , 71 , 72 , 73 , 74 , 75 , 76 , 77 , 78 , 79 or 80 nucleotides , or The antisense portion of any of the ranges. In a pertinent example, the antisense compound of the invention has an antisense portion of from i 〇 to 50 nucleosides in length. 35 Applicants should understand that this exhibits antisense Partial lengths are 1〇, u, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 ' 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43 '44 '45 '46, 47, 48, 49 or 50 nucleotides, or any range of oligonucleotides thereof In some embodiments, the oligonucleotide is 15 nucleotides in length. In one embodiment, the antisense or nucleotide compound of the invention has 157514.doc •49·201209163 length 12 or 13 Up to 30 nucleotides of the antisense portion. Those skilled in the art should Understand that 'this shows that the antisense part is 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleosides An acid, or any range of antisense compounds thereof. In one embodiment, the oligomeric compounds of the invention also comprise 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 chest pain, bird or bitterness at this position can be produced. This can occur anywhere in the antisense or dsRNA compound. The compounds are then tested using the methods described herein to determine their ability to inhibit the performance of dry nucleic acids. In some embodiments, the homology, sequence identity or complementarity between the antisense compound and the target is from about 4% to about 6%. In some embodiments, 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 ' homology, sequence identity or complementarity is from about 80% to about 90%. In some embodiments, 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%. In one embodiment, the antisense raised nucleotides (e.g., the nucleic acid knives set forth in SEQ ID NOS: 3 to 8) include - or a plurality of substitutions or modifications. In one embodiment, the nucleotide is replaced by a locked nucleic acid (LNA).

在一實施例中,寡核苷酸靶向與BBC3有關之編碼及/或 非編碼序列及闡述為SEQ ID NO: 1及2之序列之正義及/或 反義核酸分子的一或多個區域。寡核苦酸亦乾向SEQ ID 157514.doc ,50- 201209163 NO: 1及2之重疊區域。In one embodiment, the oligonucleotide targets a BBC3-related coding and/or non-coding sequence and one or more regions of the sense and/or antisense nucleic acid molecule set forth as the sequences of SEQ ID NOs: 1 and 2. . Oligonucleotide is also dried to the overlapping region of SEQ ID 157514.doc, 50-201209163 NO: 1 and 2.

本發明之某些較佳寡核苷酸係嵌合寡核苷酸。本發明上 下文中之「嵌合寡核普酸」或「嵌合體」係含有兩個或更 多個化學上不同之區域之寡核苷酸,每個區域皆由至少一 個核苦酸構成。該等寡核皆酸通常含有至少一個經修飾核 苷酸區域’該區域賦予一或多種有益性質(例如增加核酸 酶抗性,增加細胞攝取,增加與乾之結合親和力);及係 能夠裂解RNA:DNA或RNA:RNA雜合體之酶受質的區域。 舉例而言’ RNase Η係可裂解RNA:DNA雙鏈體之rNA鏈之 細胞内切核酸酶。因此,活化RNase H可裂解乾,由 此大大提高基因表現之反義調節的效率。因此,同與相同 靶區域雜交之硫代磷酸酯去氧寡核苷酸相比,在使用嵌合 寡核㈣時利用較短寡核普酸通常可獲得相當之結果。通 承可藉由凝膠電泳及(若需要)業内已知之有關核酸雜交技 術來檢測RNA&之裂解。在—個實施例中,嵌合募核苦酸 包括至少一個經修飾以增加靶結合親和力之區域、及(通 常)用作RNAse Η受質之區域。通常藉由量測寡核普酸/乾 配對之Tm來測定募核苦酸與其乾(在此情形下係編碼⑴之 一,)親和力,Tm係寡核苷酸與靶離解之溫度;使用分 光光度法來檢測離解。Tm愈高,則寡核芽酸與乾之親和 一本發明之嵌合反義化合物可以兩個或更多個如上所述之 :核、經修飾募核苷酸、募核苷及/或募核苷酸模擬 之結構形式形成。因此,化合物在業内亦稱為雜合 157514.doc -51 · 201209163 體或結合體。教示該等雜合體結構之製備的代表性美國專 利包括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 5 565,35〇 號、第 5,623’065 號、第 5,652,355 號、第 5,652,356 號、及第 5,7〇0,922號,每一者皆以引用方式併入本文中。 在一實施例中’募核苷酸之經修飾區域包括至少一個在 糖之2’位處修飾的核苷酸,最佳係經2,-0烷基、2,-〇-烧基_ 〇-烷基或2’-氟修飾之核苷酸。在另一實施例中,RNA修飾 包含對於嘧啶之核糖、無鹼基殘基或RNA中3,端之反向鹼 基之21-氟、2·_胺基及2,〇_曱基修飾。通常將該等修飾納入 募核苷酸中且已顯示該等寡核苷酸相對於2,-去氧寡核苷酸 對於給定靶具有較高Tm(亦即,較高靶結合親和力)。該增 加之親和力效應大大增強了 RNAi募核苷酸對基因表現之 抑制。RNAse Η係裂解RNA:DNA雙鏈體中RNA鏈之細胞内 切核酸酶;因此,活化此酶可裂解RNA靶,且由此可大大 增強RNAi抑制之效率。對RNA靶之裂解通常可藉由凝膠 電泳來顯示。在一實施例中,亦對嵌合寡核苷酸進行修飾 以增強核酸酶抗性。細胞含有多種可降解核酸之外切核酸 酶及内切核酸酶。已顯示多種核苷酸及核苷修飾可使納入 該等修飾之寡核苷酸對核酸酶消化之抗性強於天然寡去氧 核苷酸。核酸酶抗性通常藉由將寡核苷酸與細胞提取物或 刀離核fee酶溶液一起培育並在一段時間後通常藉由凝膠電 泳量測剩餘完整寡核苷酸之含量來進行量測。已經修飾以 1575l4.doc -52- 201209163 增強其核酸酶抗性之寡核苷酸保持完整之時間長於未經修 飾之寡核苷酸。已證實多種寡核苷酸修飾可增強或賦予核 酸酶抗性。當前,含有至少一個硫代磷酸酯修飾之寡核苷 酸更佳。在一些情形下,增強靶結合親和力之寡核苷酸經 修飾亦能獨立地增強核酸酶抗性。 擬用於本發明之一些較佳寡核苷酸的具體實例包含彼等 包括經修飾主鏈者,該等經修飾主鏈係(例如)硫代磷酸 酯、磷酸三酯、膦酸甲酯、短鏈烷基或環烷基糖間鍵聯或 短鏈雜原子或雜環糖間鍵聯。最佳者係具有硫代磷酸酯主 鏈之寡核苷酸及彼等具有雜原子主鏈者,該等雜原子主鏈 尤其係CH2 --NH--0--CH2、CH,--N(CH3)--0--CH2 [稱為 亞甲基(甲基亞胺基)或MMI主鏈]、(:112--0--:^((:^13)--CH2、CH2 -N (CH3)--N (CH3)--CH2及 0--N (CH3)--CH2 --CH2主鏈,其中天然磷酸二酯主鏈表示為O--P—O--CH)。 由 De Mesmaeker等人(1995) Acc. Chem. Res. 28:366-374所 揭示之醯胺主鏈亦較佳。亦較佳者係具有嗎啉基主鏈結構 之寡核苷酸(Summerton及Weller,美國專利第5,034,506 號)。在另一實施例中(例如肽核酸(PNA)主鏈),寡核苷酸 之磷酸二酯主鏈由聚醯胺主鏈代替,核苷酸直接或間接結 合至聚醯胺主鏈之氮雜氮原子。募核苷酸亦可包括一或多 個經取代糖部分。較佳寡核苷酸在1位包括下列基團中之 一者:OH、SH、SCH3、F、OCN、OCH3 OCH3、OCH3 0(CH2)n CH3、0(CH2)n NH2 或 0(CH2)n CH3(n 為 1至約 10) ; Cl至CIO低碳烷基、烷氧基烷氧基、經取代低碳烷 157514.doc •53· 201209163 基、烧芳基或芳烧基;Cl ; Br ; CN ; CF3 ; OCF3 ; Ο--、 S--、或 N-烷基;0--、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;經取代曱矽烷基;RNA裂解 基團;報導子基團;遷入劑;用於改良寡核苷酸之藥物代 謝動力學性質之基團;或用於改良寡核苷酸之藥效動力學 性質之基團及其他具有相似性質之取代基。較佳修飾包含 2'-甲氧基乙氧基[2LO-CH2 CH2 OCH3,亦稱為2’-0-(2-曱 氧基乙基)]。其他較佳修飾包含2’-曱氧基(2’-0--CH3)、2'-丙氧基(2I-OCH2 CH2CH3)及2’-氟(2’-F)。亦可在寡核苷酸 上之其他位置進行類似修飾,尤其係3'末端核苷酸上糖之 3'位及5'末端核苷酸之5'位。寡核苷酸亦可具有糖模擬物, 例如使用環丁基來代替戊呋喃醯基。 寡核苷酸亦可另外或作為另一選擇包含核鹼基(業内通 常簡寫為「鹼基」)修飾或取代。本文所用之「未經修 飾」或「天然」核苷酸包含腺嘌呤(A)、鳥嘌呤(G)、胸腺 。密D定(T)、胞。密。定(C)及尿°密°定(U)。經修飾核皆酸包含僅偶 爾或短暫在天然核酸中發現之核苷酸,例如次黃嘌呤、6-曱基腺嘌呤、5-Me嘧啶、尤其5-甲基胞嘧啶(亦稱為5-甲 基-2’去氧胞嘧啶且業内通常稱為5-Me-C)、5-羥甲基胞嘧 啶(HMC)、糖基HMC及龍膽二糖基HMC ;以及合成核苷 酸,例如,2-胺基腺嘌呤、2-(甲基胺基)腺嘌呤、2-(咪唑 基烷基)腺嘌呤、2-(烷基烷基胺基)腺嘌呤或其他雜取代烷 基腺嘌呤、2-硫尿嘧啶、2-硫胸腺嘧啶、5-溴尿嘧啶、5- 157514.doc -54- 201209163 羥甲基尿嘧啶、8-氤雜鳥嘌呤、7_去氮鳥嘌呤、N6 (6_胺 基己基)腺嘌呤及2,6-二胺基嘌呤。可包含業内已知之「常 用」鹼基(例如肌苷)。已顯示5_Me_c取代可將核酸雙鏈體 穩定性增加0.6°C至1.2°C且係當前之較佳鹼基取代。 對本發明寡核苷酸之另一修飾涉及使一或多種可增強募 核苷酸之活性或細胞攝取之部分或偶聯物以化學方式連接 至募核苷酸。該等部分包含但不限於脂質部分(例如膽固 醇部分)、膽固醇基部分、脂肪族鏈(例如,十二烷二醇或 十一烷基殘基)、聚胺或聚乙二醇鏈、或金剛烷乙酸。業 内已知包括親脂性部分之寡核苷酸、及製備該等寡核苷酸 之方法,例如,美國專利第5,138,〇45號 '第5,218,1〇5號及 第 5,459,255號。 給定寡核苷酸中之所有位置並不需要經一致性修飾,且 事實上可將上述修飾中之一種以上納入單一寡核苷酸中或 甚至納入募核苷酸内之單一核苷中。本發明亦包含係上文 所定義嵌合寡核苷酸之寡核苷酸。 在另一實施例中,本發明之核酸分子與另一部分偶聯, 該另一部分包含但不限於無鹼基核苷酸、聚醚、聚胺、聚 醯胺、肽 '碳水化合物、脂質、或聚烴化合物。彼等熟習 此項技術者應認識到,該等分子可連接至在糖、鹼基或磷 fccSa基團上之若干位置包括核酸分子之任一核苷酸中的一 或多者上。 本發明所用之寡核苷酸可以便捷常規方式經由熟知之固 相σ成技術來製備。包含Applied Biosystems在内之若干 157514.doc -55- 201209163 供貨商出售實施該合成之設備。亦可採用用於該合成之任 一其他方式,熟習此項技術者熟知寡核苷酸之現行合成。 亦熟知使用相似技術來製備諸如硫代磷酸酯及烷基化衍生 物等其他寡核苷酸。業内亦熟知使用相似技術及市售經修 飾DNA合成核苷酸(amidhe)及定孔玻璃(CPG)產品(例如經 生物素、螢光素、吖啶或補骨脂素修飾之DNA合成核苷酸 及/或 CPG(可自 Glen Research, Sterling, VA購得))來合成經 螢光標記、經生物素化或經其他修飾之寡核苷酸,例如經 膽固醇修飾之寡核苷酸。 根據本發明,使用修飾(例如使用LNA單體)來增強作用 之功效、特異性及持續時間並拓寬寡核苷酸之投與途徑包 括諸如MOE、ANA、FANA、PS等當前化學方式。此可藉 由使用LNA單體代替當前寡核苷酸中之一些單體來達成。 經LNA修飾之寡核苷酸可具有類似於母體化合物之尺寸或 可較大或較佳地較小。較佳地,經LNA修飾之寡核苦酸含 有小於約70%、更佳小於約60%、最佳小於約50%之LNA 單體,且其尺寸介於約5與25個核苷酸、更佳約12與20個 核苷酸之間。 較佳經修飾寡核苷酸主鏈包括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、胺基烷基磷 酸三酯、膦酸甲酯及其他膦酸烷基酯(包括膦酸3’-伸烷基 酯及對掌性膦酸酯)、次膦酸酯、胺基磷酸酯(包括胺基磷 酸3'-胺基酯及胺基磷酸胺基烷基酯)、硫羰基胺基磷酸 酯、膦酸硫羰基烷基酯、硫羰基烷基磷酸三酯、及具有常 157514.doc -56- 201209163 見3’-5’鍵聯之硼烷填酸酯、該等主鏈之2,-5'連接類似物、 及彼等具有反極性者(其中相鄰核苷單元對之連接由3,-5,變 為5'-3'或由2,-5'變為5'-2·)。亦包含各種鹽、混合鹽及游離 酸形式。 教示上述含構鍵聯之代表性美國專利包括但不限於美國 專利第 3,687,808號、第 4,469,863 號、第 4,476,3〇1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 5,278,3〇2 號、第 0 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,每一者皆以引用方式併入本文中。 不含磷原子之較佳經修飾寡核苷酸主鏈具有藉由以下形 Q 成之主鏈:短鏈烷基或環烷基核苷間鍵聯、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。該等主鏈包括彼等具有嗎啉基鍵聯者(部分 自核苷之糖部分形成);矽氧烷主鏈;硫化物、亞碱及砜 主鏈;曱醯乙醯基及硫代甲醯乙醯基主鏈;亞甲基甲醯乙 醯基及硫代甲醯乙醯基主鏈;含有烯烴之主鏈;胺基磺酸 酉旨主鏈;亞甲基亞胺基及亞甲基肼基主鏈;續酸酉旨及橫酿 胺主鏈:賴域;及其他具有混合N、〇、Sach2組成 部分者。 157514.doc -57- 201209163 教示上述募核苷之代表性美國專利包括但不限於美國專 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,每一者 皆以引用方式併入本文中。 在其他較佳寡核苷酸模擬物中,核苷酸單元中之糖及核 苷間鍵聯(亦即主鏈)二者經新基團代替。保留鹼基單元以 與適宜核酸靶化合物雜交。一種該募聚化合物(已顯示具 有極佳雜交性質之寡核苷酸模擬物)稱為肽核酸(pNA)。在 PN A化σ物中,寡核苦酸中之糖_主鏈經含醯胺主鏈、特 疋而S胺基乙基甘胺酸主鏈代替。保留核鹼基且其與主鏈 t醯胺。卩刀之氮雜氮原子直接或間接結合。教示化合 物之製備之代表性美國專利包括但不限於美國專利第 5’539’082 號、第 5,714,331號、及第 5,719,262 號每—者 白乂引用方式併入本文中。pNA化合物之其他教示内容可 > 見奶613611 專人,science 254, 1497-1500。 "月貫知1例係具有硫代碌酸醋主鏈之寡核芽酸及具 有雜原子主鏈之寡核普’且特定而言係上文所提及美國專 157514.doc -58- 201209163 利第 5,489,677號中之-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-0-P-O-CH2-], 及上文所提及美國專利第5,602,240號中之醯胺主鏈。亦較 佳者係上文所提及美國專利第5,034,506號中之具有嗎啉基 主鏈結構之募核苷酸。 經修飾寡核苷酸亦可含有一或多個經取代糖部分。較佳 募核苷酸在2'位包括以下中之一者:OH ; F ; Ο-、S-或N-烷基、Ο-、S-或N-烯基;Ο-、S-或N-炔基;或Ο烷基-0-烷 基,其中烷基、烯基及炔基可係經取代或未經取代之C1至 C10烷基或C2至C10烯基及炔基。尤佳者係O (CH2)n OmCH3、0(CH2)n、OCH3、0(CH2)nNH2、0(CH2)nCH3、 0(CH2)nONH2、及 0(CH2n0N(CH2)nCH3)2,其中 n及 m可 為1至約10。其他較佳寡核苷酸在2'位包括以下中之一者: C1至C10低碳烷基、經取代低碳烷基、烷芳基、芳烷基、 0-烷芳基或 Ο-芳烷基、SH、SCH3、OCN、CM、Br、CN、 CF3、OCF3、SOCH3、S02CH3、0Ν02、Ν02、Ν3、 ΝΗ2、雜環烷基、雜環烷芳基、胺基烷基胺基、聚烷基胺 基、經取代曱矽烷基、RNA裂解基團、受體基團、嵌入 劑、改良寡核苷酸之藥物代謝動力學性質之基團、或改良 募核苷酸之藥效動力學性質之基團、及其他具有相似性質 之取代基。較佳修飾包括2'-曱氧基乙氧基(2’-0-CH2CH20CH3,亦稱為 2'-0-(2-曱氧基乙基)或 2,-ΜΟΕ), 157514.doc -59- 201209163 亦即’烧氧基烷氧基。其他較佳經修飾包括2,-二甲基胺基 氧基乙氧基(亦即,0(CH2)20N(CH3)2基團,亦稱為2ι_ DMAOE,如下文實例中所述)、及2,_二甲基胺基乙氧基乙 氧基(業内亦稱為2,_〇_二曱基胺基乙氧基乙基或2ι_ DMAEOE ’ 亦即,2i_〇 CH2_〇_CH2 N (CH2)2)。 其他較佳修飾包括2,-曱氧基(2’-〇CH3)、2,-胺基丙氧基 (2'-0 CH2CH2CH2NH2)及2,-氟(2,-F)。亦可在募核苷酸上 之其他位置進行相似修飾,尤其係3,末端核苷酸上或2,_5, 連接寡核苷酸中糖之3,位及5,末端核苷酸之5,位。寡核苷酸 亦可具有糖模擬物,例如使用環丁基部分來代替戊呋喃糖 基糖。教示該等經修飾糖結構之製備之代表性美國專利包 括但不限於美國專利第4,981,957號、第5,118,800號、第 5,319,080 號、第 5,359,〇44 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 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號,每一者皆 以引用方式併入本文中。 寡核苷酸亦可包括核鹼基(業内通常簡寫為「鹼基」)修 飾或取代。本文所用之「未經修飾」或r天然」核苷酸包 括嗓呤驗基(腺嘌呤(A)及鳥嘌呤(G))及嘧啶鹼基(胸腺嘧啶 (T)、胞嘧啶(C)及尿嘧啶(u))。經修飾核苷酸包括其他合 成及天然核苷酸’例如5-甲基胞嘧啶(5-me-C)、5-羥曱基 157514.doc -60- 201209163 胞’咬、黃料、次黃„票吟、2_胺基腺㈣、腺嗓吟及鳥 嘌呤之6-甲基及其他烷基衍生物、腺嘌呤及鳥嘌呤之2_丙 基及其他烷基衍生物、2_硫代尿嘧啶、2_硫代胸腺嘧啶及 2-硫代胞嘧啶、5_齒代尿嘧啶及胞嘧啶、5_丙炔基尿嘧啶 及胞嘧啶、6-偶氮尿嘧啶、胞嘧啶及胸腺嘧啶、5_尿嘧啶 (假尿嘧啶)、4-硫代尿嘧啶、代、8_胺基、8_硫醇、8_ 硫代烷基、8-羥基及其他8_取代腺嘌呤及鳥嘌呤、5_鹵 代、尤其5-溴、5-三氟甲基及其他5-取代尿嘧啶及胞嘧 咬、7-甲基鳥嘌呤及7_甲基腺嘌呤、8_氮雜鳥嘌呤及8氮 雜腺嗓吟、7-去氮鳥嘌呤及7-去氮腺嘌呤及3_去氮鳥嗓吟 及3-去氮腺嗓吟。 另外’核苷酸包括彼等揭示於美國專利第3,687,8〇8號中 者、彼等揭示於 ’The Concise Encyclopedia of P〇lymerCertain preferred oligonucleotides of the invention are chimeric oligonucleotides. The "chimeric oligonucleotide" or "chimera" in the context of the present invention is an oligonucleotide comprising two or more chemically distinct regions, each region consisting of at least one nucleotide acid. The oligonucleotides typically contain at least one modified nucleotide region that confers one or more beneficial properties (eg, increased nuclease resistance, increased cellular uptake, increased binding affinity to dryness); and is capable of cleaving RNA : DNA or RNA: the region of the RNA heterozygote that is regulated by the enzyme. For example, the RNase tether can cleave RNA: an endonuclease of the rNA chain of a DNA duplex. Thus, activated RNase H can lyse dry, thereby greatly increasing the efficiency of antisense regulation of gene expression. Thus, comparable results can be obtained with shorter oligonucleotides when using chimeric oligo(4) compared to phosphorothioate deoxyoligonucleotides that hybridize to the same target region. It is believed that the cleavage of RNA & can be detected by gel electrophoresis and, if desired, nucleic acid hybridization techniques known in the art. In one embodiment, the chimeric nucleocapsid acid comprises at least one region modified to increase target binding affinity, and (usually) used as a region for RNAse Η receptor. The affinity of the nucleus acid and its dry (in this case, one of the codes (1)), the temperature at which the Tm-type oligonucleotide dissociates from the target, is usually determined by measuring the Tm of the oligonucleotide/dry pair; Photometric method to detect dissociation. The higher the Tm, the affinity of the oligonucleotide with the dry one. A chimeric antisense compound of the invention may be two or more as described above: nuclear, modified nucleotides, nucleosides and/or recruitment The structural form of the nucleotide simulation is formed. Therefore, the compounds are also known in the art as heterozygous 157514.doc -51 · 201209163 bodies or combinations. 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, 35, 5, 623 065, 5, 652, 355, 5, 652, 356, and 5, 7, 0, 922, each of which is incorporated herein by reference. In one embodiment, the modified region of the nucleotide comprises at least one nucleotide modified at the 2' position of the sugar, preferably via 2,-0 alkyl, 2,-indole-alkyl _ - an alkyl or 2'-fluoro modified nucleotide. In another embodiment, the RNA modification comprises a ribose for a pyrimidine, an abasic residue or a 21-fluoro, a 2-amino group and a 2, 〇-mercapto group modification of the 3, terminal reverse base. These modifications are typically incorporated into the raised nucleotides and have been shown to have a higher Tm (i.e., higher target binding affinity) for a given target relative to the 2,-deoxyoligonucleotide. This increased affinity effect greatly enhances the inhibition of gene expression by RNAi-raised nucleotides. RNAse Η cleavage RNA: An endonuclease of an RNA strand in a DNA duplex; therefore, activation of this enzyme cleaves an RNA target, and thereby greatly enhances the efficiency of RNAi inhibition. Lysis of the RNA target can usually be visualized by gel electrophoresis. In one embodiment, the chimeric oligonucleotide is also modified to enhance nuclease resistance. The cells contain a variety of degradable nucleic acid exonuclease and endonuclease. A variety of nucleotide and nucleoside modifications have been shown to allow the oligonucleotides incorporating such modifications to be more resistant to nuclease digestion than native oligodeoxynucleotides. Nuclease resistance is usually measured by incubating the oligonucleotide with a cell extract or a knife-free core fee enzyme solution and measuring the amount of remaining intact oligonucleotide by gel electrophoresis after a period of time. . Oligonucleotides that have been modified to enhance their nuclease resistance at 1575l4.doc -52-201209163 remain intact longer than unmodified oligonucleotides. A variety of oligonucleotide modifications have been shown to enhance or confer nuclease resistance. Currently, oligonucleotides containing at least one phosphorothioate modification are preferred. In some cases, oligonucleotides that enhance target binding affinity are also modified to independently enhance nuclease resistance. Specific examples of some preferred oligonucleotides to be used in the present invention include those comprising a modified backbone, such as phosphorothioate, phosphotriester, methyl phosphonate, Short chain alkyl or cycloalkyl sugar linkages or short chain heteroatoms or heterocyclic sugar linkages. The most preferred are oligonucleotides having a phosphorothioate backbone and those having a hetero atom backbone, such as CH2-NH--0--CH2, CH, -N (CH3)--0--CH2 [referred to as methylene (methylimido) or MMI backbone], (:112--0--:^((:^13)--CH2, CH2 - N (CH3)--N (CH3)--CH2 and 0--N (CH3)--CH2 -CH2 backbone, wherein the natural phosphodiester backbone is represented by O--P-O--CH). The guanamine backbone disclosed by De Mesmaeker et al. (1995) Acc. Chem. Res. 28:366-374 is also preferred. Also preferred are oligonucleotides having a morpholino backbone structure (Summerton and Weller, U.S. Patent No. 5,034,506. In another embodiment (e.g., peptide nucleic acid (PNA) backbone), the phosphodiester backbone of the oligonucleotide is replaced by a polyamine backbone, which is directly or indirectly Binding to the aza nitrogen atom of the polyamine backbone. The nucleotide may also include one or more substituted sugar moieties. Preferred oligonucleotides include one of the following groups at the 1-position: OH, SH , SCH3, F, OCN, OCH3 OCH3, OCH3 0(CH2)n CH3, 0(CH2)n NH2 or 0(CH2)n CH3 (n is from 1 to about 10); Cl to CIO Carboalkyl, alkoxyalkoxy, substituted lower alkane 157514.doc •53·201209163 base, aryl or aryl group; Cl; Br; CN; CF3; OCF3; Ο--, S-- Or N-alkyl; 0--, S--, or N-alkenyl; SOCH3; S02 CH3; 0N02; N02; N3; NH2; heterocycloalkyl; heterocycloalkylaryl; Polyalkylamino group; substituted decyl group; RNA cleavage group; reporter group; immigration agent; group for improving the pharmacokinetic properties of the oligonucleotide; or for modifying the oligo a pharmacodynamic property of a glycoside group and other substituents having similar properties. A preferred modification comprises 2'-methoxyethoxy [2LO-CH2 CH2 OCH3, also known as 2'-0-(2) - methoxyethyl)]. Other preferred modifications include 2'-decyloxy (2'-0--CH3), 2'-propoxy (2I-OCH2 CH2CH3) and 2'-fluoro (2' -F). Similar modifications can be made at other positions on the oligonucleotide, especially at the 3' position of the sugar at the 3' terminal nucleotide and at the 5' position of the 5' terminal nucleotide. It may have a sugar mimetic, for example using cyclobutyl instead of pentofuran. It may additionally or alternatively include nucleobase (often abbreviated as the industry's "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleotides include adenine (A), guanine (G), and thymus. D D (T), cells. dense. Set (C) and urine ° density (U). Modified nucleocapnia includes nucleotides found only occasionally or transiently in natural nucleic acids, such as hypoxanthine, 6-mercapto adenine, 5-Me pyrimidine, especially 5-methylcytosine (also known as 5- Methyl-2'-deoxycytosine and commonly referred to in the art as 5-Me-C), 5-hydroxymethylcytosine (HMC), glycosyl HMC, and gentiolysyl HMC; and synthetic nucleotides, For example, 2-aminoadenine, 2-(methylamino)adenine, 2-(imidazolylalkyl)adenine, 2-(alkylalkylamino)adenine or other hetero-substituted alkyl glands Bismuth, 2-thiouracil, 2-thiothymidine, 5-bromouracil, 5-157514.doc -54- 201209163 hydroxymethyluracil, 8-indene guanine, 7-deazaguanine, N6 (6-Aminohexyl) adenine and 2,6-diaminopurine. "Common" bases known in the art (e.g., inosine) can be included. The 5_Me_c substitution has been shown to increase the stability of the nucleic acid duplex by 0.6 ° C to 1.2 ° C and is currently the preferred base substitution. Another modification to the oligonucleotides of the invention involves chemically linking one or more of the conjugates that enhance the activity or cellular uptake of the nucleotide to the nucleotide. Such moieties include, but are not limited to, lipid moieties (eg, cholesterol moieties), cholesteryl moieties, aliphatic chains (eg, dodecanediol or undecyl residues), polyamines or polyethylene glycol chains, or diamonds Alkanoic acid. Oligonucleotides comprising a lipophilic moiety, and methods of making such oligonucleotides are known in the art, for example, U.S. Patent No. 5,138, No. 5, No. 5,218, No. 5, and No. 5,459,255. All positions in a given oligonucleotide need not be uniformly modified, and in fact one or more of the above modifications may be incorporated into a single oligonucleotide or even incorporated into a single nucleoside within a nucleotide. The invention also encompasses oligonucleotides which are chimeric oligonucleotides as defined above. In another embodiment, a nucleic acid molecule of the invention is conjugated to another moiety, including but not limited to abasic nucleotides, polyethers, polyamines, polyamines, peptides, carbohydrates, lipids, or Polyhydrocarbon compound. Those skilled in the art will recognize that such molecules can be attached to one or more of any of the nucleic acid molecules at several positions on the sugar, base or phosphofccSa group. Oligonucleotides for use in the present invention can be prepared in a convenient conventional manner via well known solid phase sigma techniques. A number of 157514.doc -55- 201209163 suppliers including Applied Biosystems sell equipment for the implementation of this synthesis. Any other means for this synthesis may also be employed, and the current synthesis of oligonucleotides is well known to those skilled in the art. It is also well known to use similar techniques to prepare other oligonucleotides such as phosphorothioates and alkylated derivatives. Similar techniques and commercially available modified DNA synthetic nucleotides (amidhe) and fixed pore glass (CPG) products (eg, DNA synthesis nuclei modified with biotin, luciferin, acridine or psoralen) are also well known in the art. Glycosylate 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, the use of modifications (e.g., using LNA monomers) to enhance the efficacy, specificity, and duration of action and broaden the route of administration of oligonucleotides includes current chemical modalities such as MOE, ANA, FANA, PS, and the like. This can be achieved by replacing some of the monomers in the current oligonucleotide with LNA monomers. The LNA modified oligonucleotide may have a size similar to the parent compound or may be larger or preferably smaller. Preferably, the LNA-modified oligonucleotide contains less than about 70%, more preferably less than about 60%, optimally less than about 50% of the LNA monomer, and is between about 5 and 25 nucleotides in size, More preferably between about 12 and 20 nucleotides. Preferred modified oligonucleotide backbones include, but are not limited to, phosphorothioates, palmitic phosphorothioates, dithiophosphates, phosphotriesters, aminoalkyl phosphates, methyl phosphonates, and Other alkyl phosphonates (including 3'-alkyl esters of phosphonic acids and palmitic phosphonates), phosphinates, amino phosphates (including 3'-amino phosphates of aminophosphates and aminophosphoric acid) Aminoalkyl esters, thiocarbonylamino phosphates, thiocarbonylalkyl phosphonates, thiocarbonylalkyl phosphates, and boron having a 157514.doc -56-201209163 see 3'-5' linkage An alkanoate, a 2,-5' linked analog of the backbone, and those having a reverse polarity (wherein adjacent nucleoside units are linked by 3, -5 to 5'-3' or Change from 2, -5' to 5'-2.). Also included are various salts, mixed salts and free acid forms. Representative U.S. patents including the above-described bonding include, but are not limited to, U.S. Patent Nos. 3,687,808, 4,469,863, 4,476, 3, 1, 5,023,243, 5,177,196, 5,188,897 , Nos. 5,264,423, 5,276,019, 5,278,3,2, 0,5,286,717, 5,321,131, 5,399,676, 5,405,939, 5,453,496, 5,455,233, 5,466,677, 5,476,925 No. 5,519,126, 5,536,821, 5,541,306, 5,550,111., 5,563,253, 5,571,799, 5,587,361 and 5,625,050, each of which is incorporated by reference. Into this article. Preferred modified oligonucleotide backbones containing no phosphorus atoms have a backbone formed by the following form: short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatoms, and alkyl or cycloalkyl groups Internucleoside linkages, or one or more short chain heteroatoms or heterocyclic internucleoside linkages. The backbones include those having a morpholino linkage (partially formed from a nucleoside sugar moiety); a siloxane backbone; a sulfide, an alkali, and a sulfone backbone; an oxime group and a thioformate The main chain of methylene sulfonyl group and the main chain of thioformamidine; the main chain containing olefin; the main chain of amino sulfonate; methylene imine and methylene The base of the thiol group; the main chain of the acid and the main chain of the lanthanum: Lai domain; and others with mixed N, 〇, Sach2 components. 157514.doc -57- 201209163 teaches that the representative U.S. patents of the above-mentioned 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 , Nos. 5,602,240, 5,608, 046, 5, 610, 289, 5, 618, 704, 5, 623, 070, 5, 663, 312, 5, 633, 360, 5, 677, 437, and 5, 677, 439 each incorporated herein by reference. In other preferred oligonucleotide mimetics, both the sugar and the internucleoside linkage (i.e., the backbone) in the nucleotide unit are replaced by a new group. The base unit is retained to hybridize to a suitable nucleic acid target compound. One such polycondensation compound (an oligonucleotide mimetic that has been shown to have excellent hybridization properties) is called a peptide nucleic acid (pNA). In the PN A sigma, the sugar-main chain in the oligo-nucleotide is replaced by a guanamine-containing main chain, a thiol-ethylaminoglycine backbone. The nucleobase is retained and it is t-amine with the backbone. The aza nitrogen atom of the file is directly or indirectly bonded. Representative U.S. patents for the preparation of the teachings 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 pNA compounds can be found in > Milk 613611, science 254, 1497-1500. "Shenzhizhi 1 case is an oligonucleotide with a thiophene sulphuric acid backbone and an oligonucleotide with a heteroatom backbone and, in particular, the above mentioned US 157514.doc -58- 201209163 Lithium No. 5,489,677 -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-[where the natural phosphodiester backbone is represented by -0-PO -CH2-], and the guanamine backbone of U.S. Patent No. 5,602,240, incorporated herein by reference. Also preferred are the nucleotides having the morpholino backbone structure of U.S. Patent No. 5,034,506, which is incorporated herein by reference. The modified oligonucleotide may also contain one or more substituted sugar moieties. Preferred nucleotides include one of the following in the 2' position: OH; F; Ο-, S- or N-alkyl, Ο-, S- or N-alkenyl; Ο-, S- or N - alkynyl; or decyl-0-alkyl, wherein the alkyl, alkenyl and alkynyl groups may be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl groups. Particularly preferred are O(CH2)n OmCH3, 0(CH2)n, OCH3, 0(CH2)nNH2, 0(CH2)nCH3, 0(CH2)nONH2, and 0(CH2n0N(CH2)nCH3)2, where n And m can be from 1 to about 10. Other preferred oligonucleotides include one of the following in the 2' position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, 0-alkylaryl or anthracene-aryl Alkyl, SH, SCH3, OCN, CM, Br, CN, CF3, OCF3, SOCH3, S02CH3, 0Ν02, Ν02, Ν3, ΝΗ2, heterocycloalkyl, heterocycloalkylaryl, aminoalkylamino, poly Alkylamino, substituted decyl, RNA cleavage group, acceptor group, intercalator, pharmacokinetic properties of modified oligonucleotides, or pharmacodynamics of modified nucleotides a group of properties, and other substituents of similar nature. Preferred modifications include 2'-methoxyethoxyethoxy (2'-0-CH2CH20CH3, also known as 2'-0-(2-decyloxyethyl) or 2,-oxime), 157514.doc-59 - 201209163 Also known as 'alkoxy alkoxy. Other preferred modifications include 2,-dimethylaminooxyethoxy (i.e., 0(CH2)20N(CH3)2 groups, also known as 2ι_DMAOE, as described in the Examples below), and 2,_Dimethylaminoethoxyethoxy (also known in the industry as 2,_〇_didecylaminoethoxyethyl or 2ι_DMAEOE'), ie, 2i_〇CH2_〇_CH2 N (CH2) 2). Other preferred modifications include 2,-decyloxy (2'-〇CH3), 2,-aminopropoxy (2'-0 CH2CH2CH2NH2) and 2,-fluoro(2,-F). Similar modifications can be made at other positions on the nucleotide, especially 3, terminal nucleotides or 2, _5, 3, 5 and 5, terminal nucleotides in the oligo. Bit. The oligonucleotide may also have a sugar mimetic, for example, using a cyclobutyl moiety in place of the 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, 〇44, 5,393,878, 5,446, 137, 5,466,786, 5,514,785, 5,519,134, 5,567,811, 5,576,427, 5,591,722, 5,597,909, 5,610,300, 5,627,053, 5,639,873, 5,646,265, Nos. 5,658,873, 5,670,633, and 5,700,920 each incorporated herein by reference. Oligonucleotides may also include nucleobases (often abbreviated as "bases" in the art) for modification or substitution. As used herein, "unmodified" or "native" nucleotides include purines (adenine (A) and guanine (G)) and pyrimidine bases (thymine (T), cytosine (C) and Uracil (u)). Modified nucleotides include other synthetic and natural nucleotides such as 5-methylcytosine (5-me-C), 5-hydroxydecyl 157514.doc -60- 201209163 Cellular bite, yellow, yellow „Ticket, 2_Aminogland (4), 6-methyl and other alkyl derivatives of adenine and guanine, 2_propyl and other alkyl derivatives of adenine and guanine, 2_thio Uracil, 2_thiothymidine and 2-thiocytosine, 5_dentate uracil and cytosine, 5-propynyl uracil and cytosine, 6-azouracil, cytosine and thymine , 5- uracil (pseudouracil), 4-thiouracil, generation, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxy and other 8-substituted adenines and guanines, 5_halogenated, especially 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosine, 7-methylguanine and 7-methyl adenine, 8-azaguanine and 8 Azadenine, 7-deazaguanine, and 7-deaza adenine, and 3-deazaguanine and 3-deaza adenine. Additional 'nucleotides including those disclosed in U.S. Patent No. 3,687 , in the 8th and 8th, they are revealed in 'The Concise Encyclopedia o f P〇lymer

Science And Engineering',第 858-859頁,Kroschwitz,J.I. 編輯,John Wiley & Sons,1990中者、彼等由Englisch等 人’ 'Angewandle Chemie,International Edition',1991, 30,第613頁揭示者、及彼等由Sanghvi, Y.S.,第15章, 'Antisense Research and Applications’,第 289-302 頁, Crooke,S.T.及Lebleu, B. ea.,CRC Press,1993揭示者。某 些該等核苷酸尤其用於增加本發明之募聚化合物之結合親 和力。該等核苷酸包括5-取代嘧啶、6-氮雜嘧啶及N-2、 N-6及0-6取代嘌呤,包括2-胺基丙基腺嘌呤、5-丙炔基尿 嘧啶及5-丙炔基胞嘧啶。已顯示5-甲基胞嘧啶取代可將核 酸雙鏈體穩定性增加 0.6°C 至 1.2°C(Sanghvi,Y.S.,Crooke, 157514.doc -61 - 201209163 S.T.及 Lebleu,B.編輯 ’ 'Antisense Research and Applications', CRC Press, Boca Raton, 1993 > 第 276-278 頁) 且係當前較佳之驗基取代,甚至更尤其在與2'_〇甲氧基乙 基糖修飾組合時。 教示上述經修飾核苷酸以及其他經修飾核苷酸之製備之 代表性美國專利包括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 5,457,187 號、第 5,459,255 號、第 5,484,9〇8 號、第 5,502,177 號、第 5,525,711 號、第 5,552,54〇 號、第 5,587,469 號、第 5,596,091 號、第 5,614,617 號、第 5,750,692號及苐5,681,941號,每一者皆以引用方式併入本 文中。 對本發明寡核苷酸之另一修飾涉及使一或多種可增強寡 核苷酸之活性、細胞分佈或細胞攝取之部分或偶聯物以化 學方式連接至寡核苷酸。 該等部分包括但不限於脂質部分(例如膽固醇部分)、膽 酸、硫醚(例如,己基_s_三苯曱基硫醇)、硫代贍固醇、脂 肪族鏈(例如’十二烷二醇或十一烷基殘基)、磷脂(例如, 二-十六烷基-外消旋-甘油或三乙基銨1,2-二_〇_十六烷基_ 外消旋mH_膦酸醋)、聚胺或聚乙二醇鏈、或金剛^ 乙酸、棕櫚基部分、或十八胺或己基胺基·羰基-氧基膽固 醇部分。 教示該等募核普酸偶聯物之製備之代表性美國專利包括 157514.doc -62- 201209163 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 5,5",923號、第 5,599,S>28號及第 5,688,941 號,每一者皆 以引用方式併入本文中。 濞#兮兌:亦可將本發明化合物加至藥物研究及靶驗證 之區域中。本發明涵蓋本文所鑑別化合物及較佳靶區段在 藥物研九中之用途,該藥物研究試圖闡釋bcl2結合部分 157514.doc •63· 201209163 聚料酸與疾病狀態、表現型、或病狀之間所存 在之闕係。該等方法包含檢測或調節BBC3聚核苦酸,其 包括使試樣、組織、細胞、或有機體與本發明化合物接 觸,,治療後某一時間量測BBC3聚核苦酸之核酸或蛋白 質含量及/或相關表現型或化學端點,及視需要比較量測 值與未處理試樣或經本發明令另一化合物處理之試樣。該 等方法亦可與其他實驗並行或組合實施以測定用於乾驗證 過程之未知基因的功能,或用以測定特定基因產㈣㈣ 於治療或預防特定疾病、病狀或表現型之的有效性。 評價基因表現之上調或抑制: 可藉由直接檢測細胞或有機體中核酸之存在來評價外源 核酸向宿主細胞或有機體中的轉移。該檢測可藉由業内= 知之若干方法來達成。例如,可藉由南方印跡或藉由聚= 酶鏈反應(PCR)技術使用特異性擴增與核酸有關之核普酸 序列之引物來檢測外源核酸的存在。亦可使用包含基因表 現分析在内之習用方法來量測外源核酸之表現。舉例而 言’可使用北方印跡及逆轉錄PCR(RT_pCR)來檢測及量化 自外源核酸產生之mRNA。 亦可藉由量測酶活性或報導子蛋白活性來檢測外源核酸 中譲之表現。舉例*言’可根躲核酸表現之降低或增 Μ間接量測反義調節活性’從而指示外源核酸在效應子 RNA中產生。基於序列保守性,可設計引物並用於擴增靶 基因之編碼區域。最初,可使用來自每一基因之最高表現 編碼區域來建立對照基因模型,但可使用任一編碼或非編 157514.doc •64- 201209163 碼區域。藉由將每一編碼區域插入報導子編碼區域與其 poly(A)信號之間來組裝每一對照基因。該等質粒將產生在 基因上游部分具有報導子基因且在1非編碼區域中具有潛 在RNAi靶之mRNA。藉由調節報導子基因來分析個別反義 寡核苷酸之有效性。用於本發明方法中之報導子基因包含 乙醯羥酸合酶(AHAS)、鹼性磷酸酶(AP)、β半乳糖苷酶 (LacZ)、β葡糖醛酸糖苷酶(GUS)、氯黴素乙醯轉移酶 (CAT)、綠色螢光蛋白(GFP)、紅色螢光蛋白(RFP)、黃色 螢光蛋白(YFP)、青色螢光蛋白(CFP)、辣根過氧化物酶 (HRP)、螢光素酶(Luc)、胭月旨鹼合酶(NOS)、章魚肉鹼合 酶(0CS)及其衍生物。可使用賦予以下物質抗性之多種可 選擇標記物:胺苄西林(ampicillin)、博來黴素 (bleomycin)、氯黴素(chloramphenicol)、慶大徽素 (gentamycin)、潮黴素(hygromycin)、卡那黴素 (kanamycin)、林可黴素(lincomycin)、甲胺蝶呤 (methotrexate)、草胺膦(phosphinothricin)、π票呤黴素 (puromycin)及四環素(tetracycline)。測定報導子基因之調 節的方法在業内已眾所周知,且包含但不限於螢光方法 (例如螢光光譜法、螢光活化細胞分選(FACS)、螢光顯微 術)、抗生素抗性測定。 可使用彼等熟習此項技術者已知及闡述於本文其他處之 方法來分析BBC3蛋白及mRNA表現。舉例而言,可使用諸 如ELISA等免疫分析來量測蛋白質含量。BBC3 ELISA分析 套組有市售,例如,購自R&D Systems (Minneapolis, 157514.doc -65- 201209163 MN)。 在實施例中,藉由與對照組試樣中之BBC3表現進行比 較來s平估使用本發明反義募核苷酸處理之試樣(例如,活 體内或活體外中之細胞或組織)中的BBC3表現(例如, mRNA或蛋白質)。舉例而言,可使用彼等熟習此項技術者 已知之方法來對蛋白質或核酸之表現與模擬處理或未處理 試樣進行比較。另一選擇為,可端視期望資訊來比較所處 里α式樣與對照組反義寡核普酸(例如,具有經改變或不同 序列者)。在另一實施例中,可使用經處理試樣與未處理 試樣中不同核酸(包含研究者認為適宜之任一標準,例 如,持豕基因)表現之差異來比較經處理試樣與未處理試 樣中BBC3蛋白質或核酸表現之差異。 可根據需要(例如)以比率或份數形式來表示所觀察之差 /、以用於與對知、組進行比較。在實施例中,相對於未處理 «式樣或經對照組核酸處理之試樣,經本發明反義寡核苷酸 處理之試樣中BBC3 mRNA或蛋白質之含量增加或降低約 1.25倍至約1〇倍或更高。在實施例中,BBC3 mRNA或蛋白 質之含量增加或降低至少約1.25倍、至少約13倍、至少約 1.4倍、至少約丄乃倍、至少約h6倍、至少約1 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4 _ 5倍、至少約5倍、至少約5 · 5 倍、至少約6倍、至少約6 · 5倍、至少約γ倍、至少約7.5 倍、至少約8倍、至少約8.5倍、至少約9倍、至少約9.5 倍、或至少約1〇倍或更高。 157514.doc -66 - 201209163 套'组、研究試劑、診斷及治療 本發明化合物可用於診斷、治療及預防,且可用作套組 2研究試劑及組份。另外,彼等熟習此項技術者通常使用 月b夠以強烈特異性抑制基因表現之反義募核苷酸來闡釋特 定基因之功能或區別生物途徑之各個成員的功能。 對於在套組及診斷及各種生物系統中之應用而言,本發 月化合物(單獨或與其他化合物或治療劑組合)可用作差值 0 及/或組合分析中之工具以闡釋細胞及組織内所表現基因 之—部分或整個補體的表現模式。 本文所用之術語「生物系統」或厂系統」定義為表現、 或使其足以表現BCL2結合部分3(BBC3)基因產物的任一有 機體、細胞、細胞培養物或組織。該等系統包含但不限於 類轉基因動物、細胞、細胞培養物、組織、異種移植 物、移植物及其組合。 根據一非限制性實例,對經一或多種反義化合物處理之 〇 細胞或組織内的表現模式與未經反義化合物處理之對照组 細胞或組織進行比較,且分析所得模式關於(例如)疾病相 關性、仏號傳導途徑、細胞定位、表現程度、所檢驗基因 之尺寸、結構或功能之基因表現的差異程度。該等分析可 • 在經刺激或未經刺激細胞中且在影響表現模式之其他化合 物存在或不存在下實施。 業内已知之基因表現分析之方法實例包含DNA陣列或微 陣列、SAGE(基因表現之系列分析)、之 限制性酶擴增)、TOGA(總體基因表現分析)、蛋白質陣列 157514.doc •67· 201209163 及蛋白組學、已表現序列標帥ST)測序、消減rna指紋 技術(surf)、消減選殖、差異顯示_、比較基因組雜 交、FISH(螢光原位雜交)技術及質譜方法。 本發明化合物可用於研究及診斷中,此乃因該等化合物 可與編碼BCL2結合部分3(BBC3)之核酸雜交。舉例而言, 在如本文所揭示可用#有效BBC3調節劑之該效率及該等 條件下雜交之募核苷酸在分別有利於基因擴增或檢測之條 件下係有效引物或探針。該等引物及探針可用於需要特異 性檢測編碼BBC3之核酸分子及擴增該等核酸分子以用於〇 檢測或用於其他BBC3研究的方法中。本發明之反義寡核 苷酸(尤其引物及探針)與編碼BBC3之核酸的雜交可藉由業 内已知方式進行檢測。該等方式可包含使酶與寡料酸偶 聯、放射性標記寡核皆酸、或任一其他適宜檢測方式。亦 可製造使用該等檢測方式檢測試樣令之B B C 3含量的套 彼等熟習此項技術者亦在治療應用中利用反義化合物之 特異性及敏感性。反義化合物已用作治療部分來治療動物 (匕3人類)之疾病狀態。反義寡核㈣藥物已安全且有效 人』且* U實施許多臨床試驗。由此確定,反義 力可為H又置用於治療細胞、組織及動物(尤其人 類)之治療方案申的有用治療方式。 '、 〇 #療 >才冑由投與本發明反義化合物來治療懷疑患有 2由調即BBC3聚核㈣之表現進行治療之 的動物㈣人類)。舉例而言,在一非限制性實施例 157514.doc -68 - 201209163 中,該等方法包括向需要治療之動物投與治療有效量之 BBC3調節劑之步驟。本發明之BBC3調節劑可有效調節 BBC3之活性或調節BBC3蛋白質之表現。在一個實施例 中,與對照組相比.,使BBC3在動物中之活性或表現抑制 約1 0%。較佳地,使BBC3在動物中之活性或表現抑制約 30%。更佳地,使BBC3在動物中之活性或表現抑制50%或 更高。因此,與對照組相比,寡聚化合物可將BCL2結合 部分3(BBC3)mRNA之表現調節至少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 100%。 在一個實施例中,與對照組相比,使BCL2結合部分 3(BBC3)及/或在動物中之活性或表現增加約10%。較佳 地,使BBC3在動物中之活性或表現增加約30%。更佳地, 使BBC3在動物中之活性或表現增加50%或更高。因此,與 對照組相比,寡聚化合物可將BBC3 mRNA之表現調節至 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 100%。 舉例而言,可量測動物之血清、血液、脂肪組織、肝或 任一其他體液、組織或器官中BCL2結合部分3(BBC3)之表 現的增大或減小。較佳地,所分析之該等流體、組織或器 官内所含之細胞含有編碼BBC3肽及/或BBC3蛋白質本身的 157514.doc •69· 201209163 核酸分子。 可藉由將有效量之本發明化合物添加至適宜醫藥上可接 受之稀釋劑或載劑中來將該化合物用於醫藥組合物中。本 發明之化合物用途及方法亦可用於預防目的。 偶聯物 對本發明寡核普酸之另—修飾涉及使一或多種可增強募 核苦酸之活性、細胞分佈或細胞攝取之部分或偶聯物以化 學方式連接至寡核普酸。該等部分或偶聯物可包含共價社 合至諸如-級或二級經基等官能團之偶聯基團。本發明: 偶聯基團包含嵌入劑、報導子分子、聚胺、聚醯胺、聚乙 二醇,、增強寡聚物之藥效動力學性質之基團、及辦 強募聚物之藥物代謝動力學性質之基團。典型偶聯基圈: 含膽固醇、脂質、石粦脂、生物素、吩嗓、葉酸醋、菲咬、 “…定、螢光素、若丹明(―)、香豆素及染 :在本發明之上下文中’增強藥效動力學性質之基團包 3可改良攝取、增強降解抗性、及/或增強與㈣酸之序 列特異性雜交的基團。在本發明之上下文中,肖強藥物代 身動力學性質之基團包含可改良本發明化合物之攝取、分 佈、代謝或分泌的基團。代表性偶聯基團揭示於㈣㈣ 月23日提出中請之國際專利中請案第pcT/us92/()9⑼號、 及美國專利第6,287,86〇號中,其以引用方式併入本文中。 7聯部分包含但不限於脂質部分(例如膽固醇部分)、膽 巟醚(例如,己基-5-二苯甲基硫醇)、硫代膽固醇、脂 肪族鏈(例如,十二烧二醇或十—燒基殘基)、碟脂(例如, 157514.doc -70· 201209163 二-十六烷基-外消旋·甘油或三乙基銨丨,2_二_〇_十六烷基_ 外消旋-甘油-3-H膦酸酯)、聚胺或聚乙二醇鏈、或金剛烷 乙酸、棕櫚基部分、或十八胺或己基胺基_羰基_氧基膽固 醇部分。本發明之寡核苷酸亦可偶聯至活性藥物物質,例 如’阿司匹林(aspirin)、華法林(warfarin)、保泰松 (phenylbutazone)、布洛芬(ibuprofen)、舒洛芬 (suprofen)、芬布芬(fenbufen)、_ 洛芬(ket0pr0fen)、(S)- (+)-普拉洛芬((S)-(+)-pranoprofen)、卡洛芬(carprofen)、 丹肌胺酸(dansylsarcosine)、2,3,5-三埃笨曱酸、氟芬那酸 (flufenamic acid)、亞葉酸、苯并噻二嗪、氯噻嗪、二氮 呼、吲哚美辛(indomethicin)、巴比妥酸鹽、頭孢菌素 (cephalosporin)、磺胺藥、抗糖尿病藥、抗菌劑或抗生 素。 教示該等寡核苷酸偶聯物之製備之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 157514.doc •71- 201209163 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、 第 5,112,963 號、第 5,214,136 號、第 第 5,112,963 號、第 5,214,136 號、第 第 5,254,469 號、第 5,258,506 號、第 第 5,272,250 號、第 5,292,873 號、第 第 5,371,241 號、第 5,391,723 號、第 第 5,451,463 號、第 5,510,475 號、第 第 5,514,785 號、第 5,565,552 號、第 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 號。 調配物 本發明化合物亦可與其他分子、分子結構或化合物混合 物(例如,脂質體、靶向受體之分子、經口、直腸、局部 或其他調配物)混合、囊封、偶聯或以其他方式結合以有 助於攝取、分佈及/或吸收。教示該等有助於攝取、分佈 及/或吸收之調配物之製備的代表性美國專利包含但不限 於美國專利第5,1〇8,921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 5,469,854 號、第 5,512,295 號、第 5,527,528 號、第 157514.doc •72· 201209163 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,每一者皆以引用方式併入本 文中。 儘管為調節靶表現及/或功能不需要在載體背景中投與 反義募核苷酸’但本發明實施例係關於用於表現反義寡核 普酸之表現載體構成物’其包括啟動子、雜合啟動子基因 序列且具有較強組成型啟動子活性,或可在期望情形下誘 導之啟動子活性。 〇 在一實施例中’本發明實踐涉及使用適宜核酸遞送系統 投與上述反義寡核苷酸中之至少一者。在一個實施例中, 該系統包含可操作地連接至聚核苷酸之非病毒載體。該等 非病毒載體之實例包含僅寡核苷酸(例如,SEQ ID NO: 3 至8之任一者或多者)或與適宜蛋白質、多糖或脂質調配物 之組合。 另外’適宜核酸遞送系統包含病毒載體,其通常係來自 Q 腺病毒、腺病毒有關病毒(AAV)、辅助細胞依賴性腺病 毋、逆轉錄病毒、或日本脂質體血凝病毒(HVJ)複合物中 之至少一者之序列。較佳地’病毒載體包括可操作地連接 至I核苷酸之強真核生物啟動子(例如’巨細胞病毒(Cmv) 啟動子)。 另外’較佳载體包含病毒載體、融合蛋白及化學偶聯 物°逆轉錄病毒載體包含M〇i〇ney鼠類白企病病毒及基於 HIV的病毒。一種較佳之基於HIV的病毒載體包括至少兩 種載體’其中gag及P〇l基因來自HIV基因組且env基因來自 157514.doc -73- 201209163 另—病毒。dNA病毒載體較佳。該等載體包含ρ〇χ载體(例 如正痘病毒或鳥痘病毒載體)、皰疹病毒载體(例如單純皰 疹I病毒(HSV)载體)、腺病毒載體及腺有關病毒載體。’ 本發明之反義化合物涵蓋任一醫藥上可接受之鹽、酯、 或該等醋之鹽、或在投與動物(包含人類)後能夠提供接 或間接)生物活性代謝物或其殘基的任一其他化合物。 術語「醫藥上可接受之鹽」係指本發明化合物之生理上 及醫藥上可接受之鹽:亦即,可保留母體化合物之期望生 物活性且並不賦予其不期望毒理學效應之鹽。對於寡核普 酸而言,醫藥上可接受之鹽及其應用之較佳實例進一步闡 述於美國專利第6,287,860號中,其以引用方式併入本文 中。 本發明亦包含含有本發明之反義化合物之醫藥組合物及 調配物。端視期望局部抑或全身性治療及擬治療區域,本 發明之醫藥組合物可以各種方式投與。投與可為局部(包 含眼部投與及投與至黏膜,包含陰道及直腸遞送卜肺部 (例如,藉由吸入或噴射粉末或氣溶膠,包含藉由霧化 器);氣管内、鼻内、表皮及經皮、經口或非經腸投與。 非經腸投與包含靜脈内、動脈内、皮下、腹膜腔内或肌内 注射或輸注,或顱内(例如,勒内或腦室内)投與。 為治療中樞神經系統中之組織,可藉由(例如)注射或輪 注至腦脊髓液中來進行投與。反義RNA在腦脊髓液中之投 與闡述於(例如)美國專利申請公開案第2〇〇7/〇117772號之 「Methods for —g familial ALS disease pr〇gressi〇n」 157514.doc -74- 201209163 中,其全部内容以引用方式併入本文中。 在意欲將本發明之反義募核普酸投與中樞神經系統之細 胞中時,可投與-或多種能夠促進標的反義寡核普酸渗透 經過血腦障壁之藥齊!。可在(例如)内。臭皮層或海馬區中進 行注射。藉由向肌肉組織中之運動神經元投與腺病毒載體 來遞送神經營養因子闡述於(例如)美國專利第M32,427 號之「Adenoviral-vector_mediated 卜咖⑷Science And Engineering', pp. 858-859, Kroschwitz, JI Editor, John Wiley & Sons, 1990, by Englisch et al. 'Angewandle Chemie, International Edition', 1991, 30, p. 613 And their disclosure by Sanghvi, YS, Chapter 15, 'Antisense Research and Applications', pp. 289-302, Crooke, ST and Lebleu, B. ea., CRC Press, 1993. Certain of these nucleotides are particularly useful for increasing the binding affinity of the polymeric compounds of the present invention. Such nucleotides include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted indole, including 2-aminopropyl adenine, 5-propynyl uracil and 5 - Propynyl cytosine. 5-methylcytosine substitution has been shown to increase nucleic acid duplex stability by 0.6 ° C to 1.2 ° C (Sanghvi, YS, Crooke, 157514.doc -61 - 201209163 ST and Lebleu, B. Edit ' 'Antisense Research And Applications', CRC Press, Boca Raton, 1993 > 276-278) and is currently the preferred substituent substitution, even more particularly when combined with the 2'-methoxyethyl saccharide modification. Representative U.S. patents which teach the preparation of the above-described modified nucleotides and other modified nucleotides include, but are not limited to, U.S. Patent Nos. 3,687,8,8, and 4,845,205, 5,130,302, 5,134,066. Nos. 5,175,273, 5,367,066, 5,432,272, 5,457,187, 5,459,255, 5,484,9,8, 5,502,177, 5,525,711, 5,552,54, Nos. 5,587,469, 5,596,091, 5, 614, 617, 5, 750, 692, and 5, 681, 941, each incorporated herein by reference. Another modification to the oligonucleotides of the invention involves chemically linking one or more conjugates that enhance the activity, cellular distribution or cellular uptake of the oligonucleotide to the oligonucleotide. Such moieties include, but are not limited to, lipid moieties (eg, cholesterol moieties), cholic acid, thioethers (eg, hexyl-s-triphenylsulfonyl mercaptan), thiosteroids, aliphatic chains (eg, 'dodecane a diol or an undecyl residue), a phospholipid (for example, di-hexadecyl-racemic-glycerol or triethylammonium 1,2-di-indole-hexadecyl)-racemic mH_ Phosphonic acid vinegar), polyamine or polyethylene glycol chain, or diamond, acetic acid, palmity moiety, or octadecylamine or hexylaminocarbonyl-oxycholesterol moiety. Representative U.S. patents which teach the preparation of such nucleophilic acid conjugates include 157514.doc-62-201209163 but are not limited to U.S. Patent Nos. 4,828,979, 4,948,882, 5,218,105, 5,525,465, 5,541,313. No. 5.545.730, 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. 5,414,077, 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, 4,876,335 No. 4,904,582, 4,958,013, 5.082.830, 5,112,963, 5,214,136, 5.082.830, 5,112,963, 5,214,136, 5,245,022, 5,254,469, 5,258,506 No. 5,262,536, 5,272,250, 5,292,873, 5,317,098, 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, 5,585,481, 5,587,371, 5,595,726, 5,597,696 No. 5, 5 ", 923, 5, 599, S > 28 and 5,688, 941, each of which is incorporated herein by reference.濞# 兮: The compounds of the invention may also be added to the area of drug research and target validation. The present invention encompasses the use of the compounds identified herein and preferred target segments in the drug study 9, which attempts to elucidate the bcl2 binding moiety 157514.doc •63·201209163 Polymeric acid and disease state, phenotype, or condition The relationship between the two. The methods comprise detecting or modulating BBC3 polynucleic acid comprising contacting a sample, tissue, cell, or organism with a compound of the invention, and measuring the nucleic acid or protein content of BBC3 polyphosphate at a time after treatment and / or related phenotype or chemical endpoint, and if necessary, compare the measured value to an untreated sample or a sample treated with another compound of the present invention. Such methods can also be performed in parallel or in combination with other experiments to determine the function of an unknown gene for use in a dry validation process, or to determine the effectiveness of a particular gene product (4) (d) in treating or preventing a particular disease, condition or phenotype. Evaluation of gene expression up-regulation or inhibition: The transfer of exogenous nucleic acid into a host cell or organism can be assessed by directly detecting the presence of nucleic acid in the cell or organism. This detection can be achieved by a number of methods known in the industry. For example, the presence of an exogenous nucleic acid can be detected by Southern blotting or by a poly-enzyme chain reaction (PCR) technique using primers that specifically amplify a nucleic acid-related nucleotide sequence. 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 from exogenous nucleic acids. The performance of purines in exogenous nucleic acids can also be detected by measuring enzymatic activity or reporter protein activity. For example, the expression of the exogenous nucleic acid in the effector RNA can be indicated by the reduction or increase in the expression of the nucleic acid. Based on sequence conservation, primers can be designed and used to amplify the coding region of the target gene. Initially, the control gene model can be created using the highest coding region from each gene, but any coding or non-coding region can be used. Each control gene was assembled by inserting each coding region between the reporter coding region and its poly(A) signal. Such plasmids will produce mRNAs having a reporter gene in the upstream portion of the gene and a potential RNAi target in the 1 non-coding region. The effectiveness of individual antisense oligonucleotides is analyzed by adjusting the reporter gene. The reporter gene used in the method of the present invention comprises acetaminolate synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucuronidase (GUS), chlorine Acetyltransferase (CAT), green fluorescent protein (GFP), red fluorescent protein (RFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), horseradish peroxidase (HRP) ), luciferase (Luc), saponin synthase (NOS), octopine carnitine synthase (0CS) and its derivatives. A variety of selectable markers that confer resistance to: ampicillin, bleomycin, chloramphenicol, gentamycin, hygromycin can be used. , kanamycin, lincomycin, methotrexate, phosphinothricin, pi purmycin, and tetracycline. Methods for determining modulation of reporter genes are well known in the art and include, but are not limited to, fluorescent methods (eg, fluorescence spectroscopy, fluorescence activated cell sorting (FACS), fluorescence microscopy), antibiotic resistance assays . BBC3 protein and mRNA expression can be analyzed using methods known to those skilled in the art and described elsewhere herein. For example, immunological assays such as ELISA can be used to measure protein content. The BBC3 ELISA assay kits are commercially available, for example, from R&D Systems (Minneapolis, 157514.doc-65-201209163 MN). In the examples, the sample treated with the antisense oligonucleotide of the present invention (for example, cells or tissues in vivo or in vitro) is evaluated by comparison with the BBC3 expression in the control sample. BBC3 performance (eg, mRNA or protein). For example, methods known to those skilled in the art can be used to compare the performance of a protein or nucleic acid to a simulated or untreated sample. Alternatively, the desired profile can be used to compare the alpha pattern with the control antisense oligonucleotide (e.g., with altered or different sequences). In another embodiment, the treated sample can be compared to the untreated sample using a difference in the performance of the different nucleic acids in the untreated sample (including any criteria deemed suitable by the investigator, eg, the ruthenium gene). The difference in the expression of BBC3 protein or nucleic acid in the sample. The observed difference / can be expressed, for example, in ratio or fractional form as needed for comparison with the pair, the group. In the examples, the amount of BBC3 mRNA or protein in the sample treated with the antisense oligonucleotide of the present invention is increased or decreased by about 1.25 times to about 1 Å relative to the untreated «sample or sample treated with the control nucleic acid. Multiple or higher. In embodiments, the BBC3 mRNA or protein is increased or decreased by at least about 1.25 fold, at least about 13 fold, at least about 1.4 fold, at least about 10 fold, at least about h6 fold, at least about 17 fold, at least about 1.8 fold. At least about 2 times, at least about 2-5 times, at least about 3 times, at least about 35 times, at least about 4 times, at least about 4 to 5 times, at least about 5 times, at least about 5 · 5 times, at least about 6 times At least about 6.5 times, at least about gamma fold, at least about 7.5 fold, at least about 8 fold, at least about 8.5 fold, at least about 9 fold, at least about 9.5 fold, or at least about 1 fold or greater. 157514.doc -66 - 201209163 Sets of 'Groups, Research Reagents, Diagnosis and Treatment The compounds of the present invention are useful in the diagnosis, treatment, and prophylaxis, and can be used as kit 2 research reagents and components. In addition, those skilled in the art typically use the antisense nucleotides that strongly inhibit gene expression to explain the function of a particular gene or to distinguish the function of each member of a biological pathway. For use in kits and diagnostics and in a variety of biological systems, this topical compound (alone or in combination with other compounds or therapeutic agents) can be used as a tool in differential zero and/or combinatorial analysis to elucidate cells and tissues. The pattern of expression of the gene or part of the entire complement. The term "biological system" or "plant system" as used herein is defined to mean any organism, cell, cell culture or tissue that exhibits, or is sufficient to represent, a BCL2 binding portion 3 (BBC3) gene product. Such systems include, but are not limited to, transgenic animals, cells, cell cultures, tissues, xenografts, grafts, and combinations thereof. According to one non-limiting example, a pattern of expression in a sputum cell or tissue treated with one or more antisense compounds is compared to a control cell or tissue that has not been treated with an antisense compound, and the resulting pattern is analyzed for, for example, a disease Correlation, nickname transmission pathway, cell localization, degree of expression, degree of difference in gene expression of the size, structure or function of the gene being tested. Such analysis can be performed in stimulated or unstimulated cells and in the presence or absence of other compounds that affect performance patterns. Examples of methods for gene expression analysis known in the art include DNA arrays or microarrays, SAGE (series analysis of gene expression), restriction enzyme amplification), TOGA (general gene expression analysis), protein array 157514.doc •67· 201209163 and proteomics, the sequence of the performance of the standard ST) sequencing, subtraction of rna fingerprint technology (surf), subtraction selection, differential display _, comparative genomic hybridization, FISH (fluorescence in situ hybridization) and mass spectrometry. The compounds of the invention are useful in research and diagnostics because such compounds hybridize to nucleic acids encoding BCL2 binding moiety 3 (BBC3). For example, the efficiency of the #effective BBC3 modulator as disclosed herein and the nucleotides that hybridize under such conditions are effective primers or probes under conditions conducive for gene amplification or detection, respectively. Such primers and probes can be used in methods that require specific detection of nucleic acid molecules encoding BBC3 and amplification of such nucleic acid molecules for use in purine detection or in other BBC3 studies. Hybridization of the antisense oligonucleotides (especially primers and probes) of the invention with nucleic acids encoding BBC3 can be detected by methods known in the art. Such means may involve coupling the enzyme to the oligoacid, radiolabeled oligonucleotide, or any other suitable means of detection. It is also possible to manufacture a kit that uses these assays to detect the B B C 3 content of the sample. Those skilled in the art also utilize the specificity and sensitivity of the antisense compound in therapeutic applications. Antisense compounds have been used as therapeutic moieties to treat disease states in animals (匕3 humans). Antisense oligonuclear (IV) drugs have been safe and effective. Many clinical trials have been implemented. It was thus determined that antisense can be a useful treatment for the treatment of cells, tissues and animals (especially humans). ', 〇 #therapy > 胄 胄 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投 投For example, in a non-limiting embodiment 157514.doc-68 - 201209163, the methods comprise the step of administering to a subject in need of treatment a therapeutically effective amount of a BBC3 modulator. The BBC3 modulator of the present invention is effective for regulating the activity of BBC3 or regulating the expression of BBC3 protein. In one embodiment, the activity or performance of BBC3 in the animal is inhibited by about 10% compared to the control group. Preferably, the activity or performance of BBC3 in the animal is inhibited by about 30%. More preferably, the activity or performance of BBC3 in an animal is inhibited by 50% or more. Therefore, the oligomeric compound can modulate the expression of BCL2 binding moiety 3 (BBC3) mRNA by at least 10°/ compared to the control group. At least 50%, at least 25%, at least 30%, at least 40%, at least 50%, 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 100%. In one embodiment, the activity or performance of BCL2 binding moiety 3 (BBC3) and/or in the animal is increased by about 10% compared to the control group. Preferably, the activity or performance of BBC3 in the animal is increased by about 30%. More preferably, the activity or performance of BBC3 in the animal is increased by 50% or more. Thus, the oligomeric compound can modulate the performance of BBC3 mRNA by at least 10%, 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 100%. For example, the increase or decrease in the expression of BCL2 binding moiety 3 (BBC3) in serum, blood, adipose tissue, liver or any other body fluid, tissue or organ of an animal can be measured. Preferably, the cells contained in the fluid, tissue or organ analyzed comprise a 157514.doc •69·201209163 nucleic acid molecule encoding the BBC3 peptide and/or the BBC3 protein itself. The compound can be used in a pharmaceutical composition by adding an effective amount of a compound of the invention to a suitable pharmaceutically acceptable diluent or carrier. The uses and methods of the compounds of the invention may also be used for prophylactic purposes. Conjugates Additional modification of the oligonucleotides of the present invention involves chemically linking one or more moieties or conjugates that enhance the activity, cellular distribution or cellular uptake of the nucleus acid to the oligonucleotide. The moieties or conjugates may comprise a coupling group covalently bonded to a functional group such as a - or a secondary radical. The present invention: a coupling group comprising an intercalating agent, a reporter molecule, a polyamine, a polyamine, a polyethylene glycol, a group for enhancing the pharmacodynamic properties of the oligomer, and a drug for strengthening the polymer A group of metabolic kinetic properties. Typical coupling base: Contains cholesterol, lipids, stone scorpion, biotin, cockroach, folic acid, phenanthrene, phenanthrene, luciferin, rhodamine (-), coumarin and dye: in this In the context of the invention, the group 3 which enhances the pharmacodynamic properties can improve uptake, enhance degradation resistance, and/or enhance the sequence-specific hybridization with the (tetra) acid. In the context of the present invention, Xiao Qiang The group of the pharmacokinetic properties of the drug comprises a group which can improve the uptake, distribution, metabolism or secretion of the compound of the present invention. Representative coupling groups are disclosed in (4) (4) on the 23rd of the international patent application in the request for the pcT /us92/()9(9), and U.S. Patent No. 6,287,86, incorporated herein by reference, which is incorporated herein by reference in its entirety in its entirety in the the the the the the the the -5-diphenylmethyl mercaptan), thiocholesterol, aliphatic chain (for example, dodecyl diol or decyl residue), dish grease (for example, 157514.doc -70· 201209163 two-ten Hexacyclo-racemic glycerol or triethylammonium hydride, 2_di-〇-hexadecyl _ Racemic-glycerol-3-Hphosphonate), polyamine or polyethylene glycol chain, or adamantane acetic acid, palmitoyl moiety, or octadecylamine or hexylamino-carbonyl-oxycholesterol moiety. Oligonucleotides can also be coupled to active drug substances such as 'aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen ( Fenbufen), _ 洛芬 (ket0pr0fen), (S)- (+)-praprofen ((S)-(+)-pranoprofen), carprofen (carprofen), dansylsarcosine, 2 , 3,5-three angstromic acid, flufenamic acid, leucovorin, benzothiadiazine, chlorothiazide, diazepam, indomethacin, barbiturate , cephalosporin, sulfa drugs, antidiabetic agents, antibacterial agents or antibiotics. Representative US patents for the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patent Nos. 4,828,979, 4,948,882 , Nos. 5,218,105, 5,525,465, 5,541,313, 5.545.730, 5,552,538 Nos. 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 No. 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, 4,876,335, 4,904,582, 4,958,013 , 157514.doc •71-201209163 5.082.830, 5.082.830, 5,245,022 '5,262,536 '5,317,098, 5,416,203, 5,512,667, 5,567,810, 5,112,963, 5,214,136, Nos. 5,112,963, 5,214,136, 5,254,469, 5,258,506, 5,272,250, 5,292,873, 5,371,241, 5,391,723, 5,451,463, Nos. 5,510,475, 5,514,785, 5,565,552, 5,574,142, 5,585,481, 5,587,371 , Nos. 5,595,726, 5,597,696, 5,599,923, 5,599,928 and 5,688,941. Formulations The compounds of the invention may also be mixed, encapsulated, coupled or otherwise with other molecules, molecular structures or mixtures of compounds (eg, liposomes, molecules that target receptors, oral, rectal, topical or other formulations). The combination is combined to aid in ingestion, distribution and/or absorption. Representative U.S. patents which teach the preparation of such formulations which facilitate ingestion, distribution and/or absorption include, but are not limited to, U.S. Patent Nos. 5,1,8,921, 5,354,844, 5,416,016, 5,459,127, 5,521,291, 5,543,165, 5,547,932, 5,583,020, 5,591,721, 4,426,330, 4,534,899, 5,013,556, 5,108,921, 5,213,804, 5,227,170, 5,264,221 , 5,356,633, 5,395,619, 5,416,016, 5,417,978, 5,462,854, 5,469,854, 5,512,295, 5,527,528, 157514.doc •72·201209163 5,534,259, 5,543,152, Nos. 5,556,948, 5,580,575, and 5,595,756 each incorporated herein by reference. Although it is not necessary to administer antisense nucleotides in the context of a vector to modulate target expression and/or function, embodiments of the invention relate to expression vector constructs for expressing antisense oligonucleotides, which include a promoter , a hybrid promoter gene sequence and having a strong constitutive promoter activity, or a promoter activity that can be induced under the desired conditions. In one embodiment, the practice of the invention involves the administration of at least one of the above antisense oligonucleotides using a suitable nucleic acid delivery system. In one embodiment, the system comprises a non-viral vector operably linked to a polynucleotide. Examples of such non-viral vectors include only oligonucleotides (e.g., any one or more of SEQ ID NOS: 3 to 8) or a combination with a suitable protein, polysaccharide or lipid formulation. Further, a suitable nucleic acid delivery system comprises a viral vector, typically from a Q adenovirus, an adeno-associated virus (AAV), a helper cell-dependent adenosis, a retrovirus, or a Japanese liposome-coagulated virus (HVJ) complex. A sequence of at least one of them. Preferably, the viral vector comprises a strong eukaryotic promoter operably linked to an I nucleotide (e.g., a 'cytomegalovirus (Cmv) promoter). Further, the preferred vector comprises a viral vector, a fusion protein and a chemical conjugate. The retroviral vector comprises M〇i〇ney murine white virus and an HIV-based virus. A preferred HIV-based viral vector comprises at least two vectors wherein the gag and P〇1 genes are from the HIV genome and the env gene is derived from 157514.doc-73-201209163 additional-virus. The dNA viral vector is preferred. Such vectors include a ρ 〇χ vector (e.g., orthopoxvirus or fowlpox virus vector), a herpes virus vector (e.g., herpes simplex virus (HSV) vector), an adenoviral vector, and an adeno-associated viral vector. The antisense compound of the invention encompasses any pharmaceutically acceptable salt, ester, or salt of such vinegar, or is capable of providing a biologically active metabolite or residue thereof after administration to an animal (including humans) Any other compound. The term "pharmaceutically acceptable salt" refers to a physiologically and pharmaceutically acceptable salt of a compound of the present invention: that is, a salt which retains the desired biological activity of the parent compound and which does not impart an undesirable toxicological effect. Preferred examples of pharmaceutically acceptable salts and their use for oligonucleosides are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. The invention also encompasses pharmaceutical compositions and formulations comprising the antisense compounds of the invention. The pharmaceutical compositions of the present invention can be administered in a variety of ways, depending on the desired local or systemic treatment and the area to be treated. Administration may be local (including ocular administration and administration to the mucosa, including vaginal and rectal delivery of the lungs (eg, by inhalation or spraying of powder or aerosol, including by nebulizer); intratracheal, nasal Internal, epidermal, and transdermal, oral or parenteral administration. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion, or intracranial (eg, intralesia or brain) Intravital administration. For the treatment of tissues in the central nervous system, administration can be carried out, for example, by injection or by injection into the cerebrospinal fluid. The administration of antisense RNA in cerebrospinal fluid is described, for example. U.S. Patent Application Publication No. 2/7/117, 772 to "Methods for -g familial ALS disease pr〇gressi〇n" 157514.doc-74-201209163, the entire contents of which is incorporated herein by reference. When the antisense nucleoside acid of the present invention is intended to be administered into cells of the central nervous system, it may be administered - or a plurality of drugs capable of promoting the penetration of the antisense oligodeoxynucleotide into the blood-brain barrier. Within the stinky cortex or hippocampus area for injection. Elaborated by the muscle tissue to deliver to the motor neurons in the administration of adenoviral vectors for neurotrophic factors (for example) US Patent "No. The first M32,427 Adenoviral-vector_mediated BU coffee ⑷

medullary motor neur〇ns」中,其以引用方式併入本文 中。業内已知將載體直接遞送至腦(例如,紋狀體、丘 腦 '海馬區或黑質)t並闡述於(例如)美國專利第⑺乂⑵ 號之「Aden〇virus vectors f〇r加㈣咖〇f⑽如y⑽ into cells of the central nervous system particularly in bram」中’其以引用方式併人本文中。可藉由注射快速投 〃或Ik疋時間藉由緩慢輸注或投與緩釋調配物進行投 與。 〇 標的反義寡核㈣亦可與提供期望醫藥或藥效動力學性 質之藥劑連接或偶聯。舉例而言,反義寡核普酸可與業内 已知的促進渗透或輸送跨過血腦障壁之任一物質(例如鐵 傳遞蛋白受體之抗體)偶合,並藉由靜脈内注射投與。反 義化β物可與病毒載體連接,該病毒載體可(例如)使反義 化合物更有效及/或促進反義化合物輸送跨過血腦障壁。 亦可藉由(例如)輸注以下物質來破壞滲透性血腦障壁: 糖’包含但不限於内消旋赤藻糖醇、木糖醇、D㈩半乳 糖、D⑴乳糖、D(+)木糖、衛矛醇、肌_肌醇、L(_)果糖、 157514.doc -75- 201209163 D(-)甘露醇、D(+)葡萄糖、D(+)阿拉伯糖、D(-)阿拉伯 糖、纖維二糖、D(+)麥芽糖、D(+)棉子糖、L(+)鼠李糖、 D(+)蜜二糖、D㈠核糖、核糖醇、D(+)阿糖醇、L(-)阿糖 醇、D(+)岩藻糖、L㈠岩藻糖、D㈠來蘇糖、L(+)來蘇糖 及L(-)來蘇糖;或胺基酸,包含但不限於麩醯胺酸、離胺 酸、精胺酸、天門冬醯胺、天門冬胺酸、半胱胺酸、麩胺 酸、甘胺酸、組胺酸、白胺酸、甲硫胺酸、苯丙胺酸、脯 胺酸、絲胺酸、蘇胺酸、酪胺酸、纈胺酸及牛磺酸。用於 增強血腦障壁滲透之方法及材料闡述於(例如)美國專利第 4,866,042號之「Method for the delivery of genetic material across the blood brain barrier」、第 6,294,520 號之 「Material for passage through the blood-brain barrier」及 第 6,936,589號之「Parenteral delivery systems」中.,其全 部内容皆以引用方式併入本文中。 本發明標的之反義化合物可與其他分子、分子結構或化 合物混合物(例如,脂質體、把向受體之分子、經口、直 腸、局部或其他調配物)混合、囊封、偶聯或以其他方式 結合,以有助於攝取、分佈及/或吸收。舉例而言,陽離 子型脂質可包含於調配物中以促進寡核苷酸攝取。一種顯 示可促進攝取之該組合物係LIPOFECTIN(購自018(:0-BRL, Bethesda,MD)。 據信,具有至少一個2'-0-曱氧基乙基修飾之寡核苷酸尤 其適用於經口投與。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、乳膏、凝膠、滴劑、栓 157514.doc -76- 201209163 劑、嘴霧劑、液體及粉末。可能需要或期望習用醫藥载 劑、水性、粉末或油性基質、增稠劑及諸如此類。經過塗 覆之避孕套、手套及諸如此類可能亦適用。 可便利地以單位劑型呈現之本發明醫藥調配物可根據醫 . f 4中熟知之習用技術製得。料技術包括使活性成份 . 與醫藥载劑(群)或賦形劑(群)結合之步驟。一般而言,調 配物可藉由使活性成份與液體載劑或微細固體載劑或二者 0 均勻且充分結合,且然後(若必要)使該產物成型來製備。 可將本發明組合物調配成許多可能劑型中之任一者,例 如但不限於錠劑、膠囊、凝膠膠囊、液體糖漿、軟質凝 膠、栓劑、及灌腸劑。本發明組合物亦可調配成存於水 性、非水性或混合介質中之懸浮液。水性懸浮液可進一步 含有可增加懸浮液黏度之物質,其包含(例如说甲基纖維 素鈉山柒糖醇及/或右旋糖酐。懸浮液亦可含有穩定 劑。 Ο I發明之醫藥組合物包括但不限於溶液、乳液、發泡體 及含有脂質體之調配物。本發明之醫藥組合物及調配物可 包括或多種渗透增強齊j、載齊1、賦形劑或其他活性或無 '活性成份》 札液通常係一種液體分散於另一液滴形式液體(直徑通 常超過〇_ι μιη)的不均相系統。乳液除分散相外亦可含有額 外組伤、及可存於水相、油相中呈現溶液形式或自身作為 刀離相的活性藥物。本發明一實施例包含微乳液。乳液及 其應用在業内已眾所周知且進一步闡述於美國專利第 157514.doc •77· 201209163 6,287,860號中。 本發明調配物包含脂質體調配物。本發明所用之術語 「脂質體」意指由兩親性脂質佈置成一或多個球形雙層所 組成的囊泡。脂質體係單層或多層囊泡,其具有自親脂性 材料形成之膜及含有擬遞送組合物之水性内部結構。陽離 子型脂質體係帶正電之脂質體,據信其可與帶負電之DnA 分子相互作用以形成穩定複合物。對pH敏感或帶負電之脂 質體據信可捕獲DNA而非與其複合。陽離子型及非陽離子 型脂質體皆可用於向細胞遞送DNA。 脂質體亦包含「空間穩定」之脂質體’本文所用之該術 語係指包括一或多種特定脂質之脂質體。在納入脂質體中 時’該等特定脂質會產生相對於缺乏該等特定脂質之脂質 體具有延長之循環壽命的脂質體。空間穩定脂質體之實例 係彼等脂質體中形成囊泡之脂質部分包括一或多種糖脂或 衍生自一或多種親水性聚合物(例如聚乙二醇(pEG)部分) 者。脂質體及其應用進一步闡述於美國專利第6,287,86〇號 中。 ,〜 本發明之醫藥調配物及組合物亦可包含表面活性劑。表 面活性劑在藥物產物、調配物及乳液令之應用在業内已眾 所周知。表面活性劑及其應用進一步闡述於美國專利第 6,287,860號中,其以引用方式併入本文中。 广個實施例中,本發明採用各種滲透增強劑來實現核 :楚、尤其寡核苦酸之有效遞送。除有助於非親脂性藥物在 胞膜中之擴散外,滲透增強劑亦增強親脂性藥物之洚透 157514.doc -78· 201209163 性。滲透增強劑可歸類為屬於5大類中之一者:亦即,表 面活性劑、脂肪酸、膽汁鹽、螯合劑及非螯合性非表面活 性劑。滲透增強劑及其應用進一步閣述於 6取_號中,其以引用方式併入本文中。國專㈣ 熟習此項技術者應認識到’調配物通常根據其預期應用 (亦即投與途徑)來進行設計。 用於局部投與之較佳調配物包含彼等本發明寡核芽酸與 ΟIn medullary motor neur〇ns, which is incorporated herein by reference. It is known in the art to deliver a carrier directly to the brain (e.g., striatum, thalamus 'hippocampus or substantia nigra) and is described, for example, in "Aden〇virus vectors f〇r plus (4) of U.S. Patent No. 7 (2). Curry f(10) as in y(10) into cells of the central nervous system particularly in bram", which is incorporated herein by reference. Administration can be by slow infusion or administration of a sustained release formulation by injection for rapid administration or Ik time. The antisense oligo (4) of the target may also be linked or coupled to an agent that provides the desired pharmaceutical or pharmacodynamic properties. For example, antisense oligonucleotides can be coupled to any of the substances known in the art to promote penetration or delivery across the blood-brain barrier (eg, antibodies to the transferrin receptor) and administered by intravenous injection. . The antisense beta can be linked to a viral vector which, for example, can render the antisense compound more effective and/or facilitate delivery of the antisense compound across the blood brain barrier. The osmotic blood-brain barrier can also be destroyed by, for example, infusion of: sugars including but not limited to meso-erythritol, xylitol, D(d)galactose, D(1) lactose, D(+) xylose, Dulcitol, myo-inositol, L(_) fructose, 157514.doc -75- 201209163 D(-) mannitol, D(+) glucose, D(+) arabinose, D(-) arabinose, fiber Disaccharide, D(+) maltose, D(+) raffinose, L(+) rhamnose, D(+) melibiose, D(one) ribose, ribitol, D(+) arabitol, L(- Alginitol, D(+) fucose, L (one) fucose, D (one) to threose, L (+) to threose and L (-) to threose; or amino acids, including but not limited to bran Aminic acid, lysine, arginine, aspartame, aspartic acid, cysteine, glutamic acid, glycine, histidine, leucine, methionine, phenylalanine, Proline, serine, threonine, tyrosine, proline and taurine. Methods and materials for enhancing blood-brain barrier penetration are described, for example, in "Method for the delivery of genetic material across the blood brain barrier", No. 6,294, 520, "Material for passage through the blood-brain". Barriers and "Parenteral delivery systems", No. 6,936,589, the entire contents of each of which are incorporated herein by reference. The antisense compounds of the present invention may be mixed, encapsulated, coupled or otherwise mixed with other molecules, molecular structures or mixtures of compounds (eg, liposomes, molecules to the receptor, oral, rectal, topical or other formulations). Other ways to combine to aid in ingestion, distribution and/or absorption. For example, a cationic lipid can be included in the formulation to facilitate oligonucleotide uptake. One composition which exhibits uptake is LIPOFECTIN (available from 018 (:0-BRL, Bethesda, MD). It is believed that oligonucleotides having at least one 2'-0-decyloxyethyl modification are particularly suitable. Oral administration. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, plugs 157514.doc -76- 201209163, mouth Aerosols, liquids and powders. Pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be required or desired. Coated condoms, gloves and the like may also be suitable. Conveniently presented in unit dosage form. The pharmaceutical formulations of the present invention can be prepared according to conventional techniques well known in the art. F 4 includes the steps of combining the active ingredient with a pharmaceutical carrier (group) or excipient (group). The preparation can be prepared by uniformly and intimately combining the active ingredient with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product. The compositions of the invention can be formulated into a number of possible dosage forms. Either For example, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. The liquid may further comprise a substance which increases the viscosity of the suspension, and comprises (for example, methylcellulose sodium behenitol and/or dextran. The suspension may also contain a stabilizer. Ο I The pharmaceutical composition of the invention includes but is not limited to Solutions, emulsions, foams, and formulations containing liposomes. The pharmaceutical compositions and formulations of the present invention may include or be osmotically enhanced, loaded, excipients or other active or non-active ingredients. The liquid is usually a heterogeneous system in which a liquid is dispersed in another liquid in the form of a droplet (the diameter usually exceeds 〇_ι μιη). The emulsion may contain additional group damage in addition to the dispersed phase, and may be present in the aqueous phase or the oil phase. An active drug in the form of a solution or self-dissipating as a knife. One embodiment of the invention comprises a microemulsion. The emulsion and its use are well known in the art and are further described in U.S. Patent No. 1575. 14.doc • 77· 201209163 6,287, 860. The formulations of the invention comprise liposome formulations. The term "liposome" as used in the present invention means a vesicle composed of an amphiphilic lipid arranged in one or more spherical bilayers. Lipid system monolayer or multilamellar vesicles having a membrane formed from a lipophilic material and an aqueous internal structure containing the composition to be delivered. The cationic lipid system is a positively charged liposome which is believed to be associated with a negatively charged DnA Molecular interactions to form stable complexes. pH-sensitive or negatively charged liposomes are believed to capture DNA rather than complex with it. Both cationic and non-cationic liposomes can be used to deliver DNA to cells. Liposomes also contain "space. Stable Liposomes As used herein, the term refers to liposomes comprising one or more specific lipids. When incorporated into liposomes, these particular lipids produce liposomes that have an extended cycle life relative to liposomes that lack the particular lipids. Examples of sterically stabilized liposomes are those in which the vesicle-forming lipid portion of the liposome comprises one or more glycolipids or is derived from one or more hydrophilic polymers (e.g., polyethylene glycol (pEG) moieties). Liposomes and their use are further described in U.S. Patent No. 6,287,86. ~ The pharmaceutical formulations and compositions of the present invention may also comprise a surfactant. The use of surfactants in pharmaceutical products, 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 a wide variety of embodiments, the present invention employs various permeation enhancers to achieve efficient delivery of the core, particularly oligonucleotide. In addition to contributing to the diffusion of non-lipophilic drugs in the cell membrane, the penetration enhancer also enhances the penetration of lipophilic drugs 157514.doc -78· 201209163. Penetration enhancers can be classified as belonging to one of five categories: that is, surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants. Permeation enhancers and their use are further described in the disclosure of which is incorporated herein by reference. National Studies (4) Those who are familiar with this technology should recognize that 'mixtures are usually designed according to their intended application (ie, the route of administration). Preferred formulations for topical administration comprise the oligonucleotides of the invention and oxime

局部遞送劑混合者,該局部遞送劑係(例如)脂質、脂質 體、脂肪酸、脂肪酸醋、類固醇、螯合劑及表面活性劑。 較佳脂質及脂質體包含中性脂質及脂質體(例如二油酿基_ 码月曰醯基DOPE乙醇胺、二肉豆蔬酿基麟脂酿基膽驗 C 一硬月曰醯基磷脂醯基膽鹼)、負脂質及脂質體(例 如二肉豆謹醯基磷脂醯基甘油DMpG)及陽離子型脂質及脂 質體(例如二油酿基四甲基胺基丙基D〇TAp及二油酿基-麟 脂醯基乙醇胺DOTMA)。 ^ f於局。卩或其他投與而言,本發明之寡核普酸可囊封於 月曰質體内或可與其形成複合物(尤其陽離子型脂質體)。另 選擇為,寡核Μ可與脂質、尤其陽離子型脂質複合。 較佳月θ肪酸及S旨、其醫藥上可接受之鹽、及其應用進一步 闡述於美國專利第6,287,86〇號中。 ^ 2 = 口奴與之組合物及調配物包含粉末或粒子、微顆 r不米顆粒、存於水或非水性介質中之懸浮液或溶液、 '^凝膠膠囊、藥囊、錠劑或微錠劑。可能期望使用增 ^場味劑、稀釋劑、乳化劑、分散助劍或黏合劑。較 157514.doc -79- 201209163 佳口服調配物係彼等本發明募核苦 甘自欠與一或多種滲透增強 劑、表面活性劑及螯合劑聯合梪鱼本 又與考。較佳表面活性劑包 含脂肪酸及/或其酯或鹽、膽汁酸另/ τ暖及/或其鹽。較佳膽汁酸/ 鹽及脂肪酸及其應用進一步闡沭於i W連於美國專利第6,287,860號 中’其以引用方式併入本文中。,t h ^ 升又T 亦較佳者係滲透增強劑之 組合,例如,脂肪酸/鹽與膽汁酸/鹽之組合。尤佳之組合 係月桂酸之鈉鹽、癸酸及UDCA。苴从办* 。其他滲透增強劑包含聚 氧乙烯-9-月桂基醚、聚氧乙嫌__加鈴純甘 乳C"碎20_鯨蠟基醚。本發明之寡 核芽酸可以粒子形式(包含噴霧乾燥顆粒、或經複合以形 成微顆粒或奈米顆粒)經口遞送。寡核*酸複合劑及其應 用進一步闡述於美國專利第6 287 # 乐b’287,860唬中,其以引用方式 併入本文中。 用於非經腸、勒内或心室内投與之組合物及調配物可包 含亦可含有緩衝劑、稀釋劑及其他適宜添加劑之無菌水溶 液,該等其他適宜添加劑係(例如)但不限於滲透增強劑、 載劑化合物及其他醫藥上可接受之載劑或賦形劑。 本發明某些實施例提供含有一或多種寡聚化合物及一或 多種藉由非反義機制發揮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含但不限於癌症化學治療 藥物例如柔紅黴素(daunorubicin)、道諾黴素 (daunomycin)、更生黴素(dactjn〇mycin)、多柔比星 (doxorubicin)、表柔比星(epirubicjn)、伊達比星 (idarubicin)、依素比星(es〇rubicin)、博來黴素、馬磷醯胺 (mafosfamide)、異環磷醯胺(if〇sfamide)、胞嘧啶阿糖核苷 157514.doc 201209163The topical delivery agent is, for example, a lipid, a liposome, a fatty acid, a fatty acid vinegar, a steroid, a chelating agent, and a surfactant. Preferred lipids and liposomes comprise neutral lipids and liposomes (for example, dioleic acid base _ 曰醯 曰醯 D D D D D 乙醇 乙醇 、 、 、 、 、 、 、 一 一 一 一 一 一 一 一 一 一 一 一 一 一Choline), negative lipids and liposomes (such as di-myristylphosphatidylglycerol DMpG) and cationic lipids and liposomes (such as dioleyltetramethylaminopropyl D〇TAp and two oils) Base-Linsterylethanolamine DOTMA). ^ f is in the bureau. In the case of sputum or other administration, the oligonucleotides of the present invention may be encapsulated in or form complexes (especially cationic liposomes) with the ruthenium. Alternatively, the oligonucleoside can be complexed with a lipid, especially a cationic lipid. Preferably, the hydroxy acid and the pharmaceutically acceptable salt thereof, and their use are further described in U.S. Patent No. 6,287,86. ^ 2 = Stolen and compositions and formulations comprising powders or granules, micro-r-grain granules, suspensions or solutions in water or non-aqueous media, '^gel capsules, sachets, lozenges or Micro-tablets. It may be desirable to use a flavoring agent, diluent, emulsifier, dispersing sword or adhesive. 157514.doc -79-201209163 Good oral formulations are the same as the ones that have been combined with one or more penetration enhancers, surfactants and chelating agents. Preferably, the surfactant comprises a fatty acid and/or an ester or a salt thereof, a bile acid / τ warm and/or a salt thereof. The preferred bile acids/salts and fatty acids and their use are further described in U.S. Patent No. 6,287,860, the disclosure of which is incorporated herein by reference. And t h ^ liter and T are also preferred as a combination of penetration enhancers, for example, a combination of a fatty acid/salt and a bile acid/salt. The combination of Youjia is sodium salt of lauric acid, citric acid and UDCA. I am going to do it*. Other permeation enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene __ 铃铃纯甘乳 C" 碎20_cetyl ether. The oligonucleotides of the present invention may be orally delivered in the form of particles comprising spray dried particles or complexed to form micro or nano particles. Oligonucleotide® acid complexes and their use are further described in U.S. Patent No. 6,287, the disclosure of which is incorporated herein by reference. Compositions and formulations for parenteral, intralesional or intraventricular administration may comprise sterile aqueous solutions which may also contain buffering agents, diluents and other suitable additives such as, but not limited to, permeation. Enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients. Certain embodiments of the invention provide pharmaceutical compositions comprising one or more oligomeric compounds and one or more other chemotherapeutic agents that function by non-antisense mechanisms. Examples of such chemotherapeutic agents include, but are not limited to, cancer chemotherapeutic drugs such as daunorubicin, daunomycin, dactjn〇mycin, doxorubicin, and Epirubicjn, idarubicin, es〇rubicin, bleomycin, mafosfamide, if〇sfamide, cytosine Arabinoside 157514.doc 201209163

(cytosine arabinoside)、亞石肖脲氮齐(bischloroethyl-nitrosurea)、白消安(busulfan)、絲裂黴素 C(mitomycin C)、 放線菌素D(actinomycin D)、 光輝黴素 (mithramycin) 、潑 尼松(prednisone) 、經 孕酮 (hydroxyprogesterone)、睪酮(testosterone)、他莫昔芬 (tamoxifen)、達卡巴嗪(dacarbazine)、丙卡巴肼 (procarbazine)、六曱三聚氰胺(hexamethylmelamine)、五 甲三聚氣胺(pent ame thy lmel amine)、 米托蒽西昆 (mitoxantrone)、安 °丫 α定(amsacrine)、苯丁 酸氮芥 (chlorambucil)、環己亞石肖脲(methylcyclohexylnitrosurea)、氮 芥(nitrogen mustards)、美法侖(melphalan)、環墻酿胺 (cyclophosphamide)、6-酼 口票呤(6-mercaptopurine)、6-硫鳥 嗓吟(6-thioguanine)、阿糖胞苦(cytarabine)、5-氮雜胞普 (5-azacytidine)、經基脲(hydroxyurea)、喷司他丁 (deoxycoformycin)、4-經基過氧環鱗酿胺、5-氟尿痛咬(5-fluorouracil) (5-FU)、5-敗去氧尿 ^(5-fluorodeoxyuridine) (5-FUdR)、曱胺蝶呤(MTX)、秋水仙鹼(colchicines)、紫杉 醇(taxol)、長春新驗(vincristine)、長春驗(vinblastine)、 依託泊戒(etoposide)(VP-16)、三曱曲沙(trimetrexate)、伊 立替康(irinotecan)、托泊替坎(topotecan)、吉西他濱 (gemcitabine)、替尼泊武(teniposide)、順舶(cisplatin)及己 稀雌紛(diethylstilbestrol)(DES)。在與本發明化合物一起 使用時,該等化學治療劑可單獨(例如,5-FU及寡核苷 酸)、依序(例如,5-FU及寡核苷酸,隨後在一定時間之後 157514.doc -81 - 201209163 係MTX及募核苷酸)、或與一或多種其他該等化學治療劑 組合(例如,5-FU、MTX及寡核苷酸,或5_Fu、放射療法 及寡核苷酸)使用。抗炎藥(包含但不限於非類固醇抗炎藥 及皮質類固醇,以及抗病毒藥,包含但不限於利巴韋林 (ribivirin)、阿糖腺苷(vidarabine)、阿昔洛韋(acycl〇vir)及 更昔洛韋(ganciclovir))亦可組合於本發明組合物中。反義 化合物及其他非反義藥物之組合亦屬於本發明範疇内。兩 種或更多種組合之化合物可一起或依序使用。 在另一相關實施例中,本發明組合物可含有一或多種靶 向第一核酸之反義化合物(尤其募核苷酸)及一或多種靶向 第二核酸靶之額外反義化合物。舉例而言,第一靶可為 BCL2結合部分3(BBC3)之特定反義序列,且第二靶可為來 自另-核苷酸序列之區$。另—選擇為,I發明組合物可 含有兩種或更多種乾向同一 BCL2結合部分3⑽c3)核酸起 之不同區域的反義化合物。反義化合物之諸多實例闇釋於 本文中且其他實例可選自業内已知之適宜化合物。兩種或 更多種組合之化合物可一起或依序使用。 投藥: 據信,彼等熟習此項技術者應瞭解治療性組合物之調配 及其隨後之投與(投藥)。投藥取決於欲治療疾病狀態之嚴 重程度及反應性,其中治療過程可持續數天至數月,或直 至實現治癒或減輕疾病狀態為止。可藉由量測患者體内之 樂物積累來計算最適投藥方案。孰 π 呆热各此項技術者可容易地 確疋农適劑量、投藥方法及重 吸疋手。敢適劑量可端視個 1575l4.doc -82- 201209163 別寡核普酸之相對功效而有所變化,且通常可基於發現在 活體外及活體内動物模型中有效之£(:5〇進行估計。一般而 :,劑量為0.01 叫/kg體重至100 mg/kg體重,且可每曰、 母週、每月或每年給予一次或更多次,或甚至每2至20年 給予一次。熟習此項技術者可容易地基於體液或組織中藥 物之所量測滯留時間及濃度來估計投藥的重複速率。成功 治療後,可期望使患者維持該療法以預防疾病狀態復發, 〇 其中以介於0.01 pg/kg體重至100 mg/kg體重之間之維持劑 量投與寡核苷酸,且每日投與一次或更多次至每20年投與 一次。 在實施例中,使用以下劑量之藥物來治療患者:至少約 1 mg/kg體重、至少約2 mg/kg體重、至少約3 mg/kg體重、 至少約4 mg/kg體重、至少約5 mg/kg體重、至少約6 mg/kg 體重、至少約7 mg/kg體重、至少約8 mg/kg體重、至少約9 mg/kg體重、至少約1〇 mg/kg體重、至少約μ mg/kg體重、 Q 至少約20 mg/kg體重、至少約25 mg/kg體重、至少約3〇 mg/kg體重、至少約35 mg/kg體重、至少約40 mg/kg體重、 至少約45 mg/kg體重、至少約50 mg/kg體重、至少約6〇 mg/kg體重、至少約70 mg/kg體重、至少約80 mg/kg體重、 至少約90 mg/kg體重或至少約1〇〇 mg/kg體重。反義寡核芽 酸之某些注射劑量闡述於(例如)美國專利第7,563,884號之 「Antisense modulation of PTP1B expression」中,其全部 内容以引用方式併入本文中。 儘管上文已闡述本發明之各個實施例,然而,應理解, 157514.doc -83 - 201209163 該等實施例僅係以實例方式而非限制方式提供。可根據本 文之揭示内容對所揭示實施例作出諸多改變,此並不背離 本發明之精神或料。因此,本發明之廣度及料不應受 上述實施例中之任一者限制。 又 本文所提及之所有文件皆以引用方式併入本文中。出於 所有目的’在本中請案中引用之所有出版物及專利文件皆 係乂引用方式併入,其併入程度如同每一個別出版物或專 利文件係單獨闡述一般。對於此文件中引用之各個參考文 獻,申請者不承認任一特定參考文獻係其發明之「先前技 術」。本發明組合物及方法之實施例闡釋於下列實例中。 實例 以下非限制性實例用於闡釋本發明之所選實施例。應瞭 解,所示組份之元素之比率變化及代替物已為彼等熟習此 項技術者所明瞭且屬於本發明實施例之範疇内。 實例1 :對BCL2結合部分3(BBC3)之反義核酸分子及,或 BBC3聚核苷酸之正義鏈具有特異性之反義寡核苷酸的設計 如上所述,術語「對…具有特異性之募核苷酸」或「無 向…之寡核苷酸」係指具有如下序列之募核苷酸··(i)能 夠與乾向基因之一部分形成穩定複合物,或(ii)能夠與乾 向基因之mRNA轉錄物之一部分形成穩定雙鏈體。(cytosine arabinoside), bischloroethyl-nitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, Prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethylmelamine, Wujia Pent ame thy lmel amine, mitoxantrone, amsacrine, chlorambucil, methylcyclohexylnitrosurea, nitrogen Nitrogen mustards, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, arabinose Cytarabine), 5-azacytidine, hydroxyurea, deoxycoformycin, 4-peroxy-cyclohexylamine, 5-fluorourine bite (5- Fluorouracil) (5-FU), 5-deoxyuridine (5-fluorodeoxyuridine) (5-FUdR), guanamine pterin (MTX), colchicines, taxol, vincristine, vinblastine, etoposide (VP-16) ), trimetrexate, irinotecan, topotecan, gemcitabine, teniposide, cisplatin, and diethylstilbestrol ) (DES). When used with a compound of the invention, the chemotherapeutic agents can be isolated (eg, 5-FU and oligonucleotide), sequentially (eg, 5-FU and oligonucleotide, followed by a period of time 157514. Doc -81 - 201209163 is a combination of MTX and nucleotides, or in combination with one or more other such chemotherapeutic agents (eg, 5-FU, MTX and oligonucleotides, or 5_Fu, radiation therapy, and oligonucleotides) )use. Anti-inflammatory drugs (including but not limited to non-steroidal anti-inflammatory drugs and corticosteroids, as well as antiviral drugs, including but not limited to ribivirin, vidarabine, acyclovir (acycl vir) And ganciclovir may also be combined in the compositions of the invention. Combinations of antisense compounds and other non-antisense drugs are also within the scope of the invention. Compounds of two or more combinations may be used together or sequentially. In another related embodiment, the compositions of the invention may contain one or more antisense compounds (especially raised nucleotides) that target the first nucleic acid and one or more additional antisense compounds that target the second nucleic acid target. For example, the first target can be a specific antisense sequence of BCL2 binding portion 3 (BBC3) and the second target can be region $ from the other-nucleotide sequence. Alternatively, the inventive composition may contain two or more antisense compounds which are dried to different regions of the same BCL2 binding moiety 3(10)c3) nucleic acid. Many examples of antisense compounds are disclosed herein and other examples may be selected from suitable compounds known in the art. Compounds of two or more combinations may be used together or sequentially. Dosing: It is believed that those skilled in the art should be aware of the formulation of the therapeutic composition and its subsequent administration (administration). Administration depends on the severity and responsiveness of the condition to be treated, where the course of treatment can last from days to months, or until a cure or amelioration of the disease is achieved. The optimal dosing regimen can be calculated by measuring the accumulation of music in the patient's body.孰 π Staying hot with this technology can easily determine the appropriate dosage, method of administration and re-sucking. Dare doses can vary depending on the relative efficacy of the 1575l4.doc -82- 201209163 oligo-nucleotide, and can usually be estimated based on the £(:5〇) found to be effective in in vitro and in vivo animal models. Generally, the dose is from 0.01 call/kg body weight to 100 mg/kg body weight, and can be given once or more per week, maternal week, monthly or yearly, or even every 2 to 20 years. The skilled artisan can easily estimate the repetition rate of administration based on the measured residence time and concentration of the drug in the body fluid or tissue. After successful treatment, it may be desirable to have the patient maintain the therapy to prevent recurrence of the disease state, wherein A maintenance dose between pg/kg body weight to 100 mg/kg body weight is administered to the oligonucleotide and administered once or more times per day to every 20 years. In the examples, the following doses of the drug are used. To treat a patient: at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg Weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, Less than about 9 mg/kg body weight, at least about 1 mg/kg body weight, at least about μ mg/kg body weight, Q at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 3 mg/kg body weight At least about 35 mg/kg body weight, at least about 40 mg/kg body weight, at least about 45 mg/kg body weight, at least about 50 mg/kg body weight, at least about 6 mg/kg body weight, at least about 70 mg/kg body weight, At least about 80 mg/kg body weight, at least about 90 mg/kg body weight, or at least about 1 mg/kg body weight. Certain injection doses of antisense oligonucleotides are described, for example, in "Antisense", U.S. Patent No. 7,563,884. Modulation of PTP1B expression, the entire contents of which are incorporated herein by reference. Although the various embodiments of the present invention have been described above, it is understood that 157514.doc -83 - 201209163 these embodiments are merely examples The present invention is not limited by the spirit and scope of the present invention. The breadth and scope of the present invention should not be limited by the above embodiments. One limit. All the documents mentioned in this article are In this way, all publications and patent documents cited in this application are hereby incorporated by reference inso- In general, the applicant does not recognize that any particular reference is a "prior art" of the invention of the invention. Examples of compositions and methods of the invention are illustrated in the following examples. EXAMPLES The following non-limiting examples are illustrative of selected embodiments of the invention. It is to be understood that the change in the ratio of the elements of the components shown and the alternatives are known to those skilled in the art and are within the scope of the embodiments of the invention. Example 1: Design of an antisense nucleic acid molecule that binds to an antisense nucleic acid molecule of BCL2 binding moiety 3 (BBC3) and, or a sense strand of a BBC3 polynucleotide, as described above, the term "specific to "Nuclear nucleotide" or "undirected oligonucleotide" refers to a nucleotide having the following sequence: (i) capable of forming a stable complex with a part of the dry gene, or (ii) capable of A portion of the mRNA transcript of the dry gene forms a stable duplex.

藉由使用電腦程式(例如IDT AntiSense Design, IDTBy using a computer program (eg IDT AntiSense Design, IDT)

OligoAnalyzer)來促進適當募核苷酸之選擇,該等電腦程 式自動鑑別每一給定序列中之19-25個核苷酸之子序列, 該等子序列與靶聚核苷酸序列形成雜合體且具有期望熔融 157514.doc • 84 - 201209163 溫度(通常為50°C至60°C ),且不會形成自身二聚體或其他 複雜二級結構。 藉由使用自動對準核酸序列並指示一致性或同源性區域 之電腦程式來進一步促進適當寡核苷酸的選擇。使用該等OligoAnalyzer) to facilitate the selection of appropriate nucleotides that automatically identify sub-sequences of 19-25 nucleotides in each given sequence, which form a hybrid with the target polynucleotide sequence and It has the desired melting temperature of 157514.doc • 84 - 201209163 (usually 50 ° C to 60 ° C) and does not form self-dimers or other complex secondary structures. The selection of appropriate oligonucleotides is further facilitated by the use of computer programs that automatically align nucleic acid sequences and indicate regions of identity or homology. Use these

程式藉由(例如)搜索諸如GenBank等數據庫或藉由對PCR 產物測序來比較所獲得的核酸序列。對來自給定基因組中 一定範圍基因及基因間區域之核酸序列進行比較使得可選The program compares the obtained nucleic acid sequences by, for example, searching a database such as GenBank or by sequencing the PCR products. Comparing nucleic acid sequences from a range of genes and intergenic regions in a given genome makes optional

擇對目標基因顯示適當特異性程度的核酸序列。該等程序 使得可選擇對給定基因組中之靶核酸序列顯示高互補性程 度且對其他核酸序列顯示較低互補性程度絲核苦酸。熟A nucleic acid sequence that exhibits an appropriate degree of specificity for the target gene. Such procedures allow for the selection of a degree of high complementarity for a target nucleic acid sequence in a given genome and a lower degree of complementarity to other nucleic acid sequences. Cooked

習此項技術者應認識到,可在較大範圍中選擇適用於本發 明中之基因區域。 X 可特異性雜交」:化合物 在以下情形時反義化合物係 與乾核酸之結合可干擾乾核酸之正常功能以調節功能及/ 或活性’且存在足夠互補性程度以避免反義化合物斑非乾 核酸序列在期望發生特異性結合之條件下(亦即,在活體 内分析或治療性治療情形中之生理條件下,及在活體外分 析情形中實施分析之條件下)發生非特異性結合。 ^ 本文所述寡料酸之雜交性f可藉由業内已°知之 個活體外分析進行測^。舉例而言 八iff、&丨—蜡由使用炼融曲線 刀^測疋無天錢義分子與潛在藥物分子間之結合強度來 獲得本文所述寡核苷酸之性質。 又 =量測分子間相互作用之強度之已確立方法中的任 一者(例如’㈣曲線分析)來估料天然反義分子與潛在 157514.doc •85· 201209163 藥物分子(分子)間之結合強度。 熔融曲線分析可測定對於天然反義分子/分子複合物自 雙鏈快速轉變成單鏈形態時之溫度。此溫度已廣泛接受作 為兩個分子間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拷貝或對應於分 子結合位點之合成DNA或RNA核苷酸來實施熔融曲線分 析。可使用含有所有必需試劑以實施此分析之多個套組 (例如 Applied Biosystems公司,MeltDoctor套組)。該等套 組包含含有雙鏈DNA (dsDNA)結合染料(例如ABI HRM染 料、SYBR Green、SYTO等)中之一者的適宜缓衝溶液。 dsDNA染料之性質在於其幾乎不發射游離形式之螢光,但 在與dsDNA結合時具有高螢光性。 為實施分析,以由特定製造商方案界定之濃度將cDNA 或相應寡核苷酸與分子混合。將混合物加熱至95°C以離解 所有預形成之dsDNA複合物,然後缓慢冷卻至室溫或由套 組製造商界定之其他較低溫度以使DNA分子退火。然後將 新形成複合物缓慢加熱至95°C,同時繼續收集關於反應中 所產生螢光之量的數據。螢光強度與反應中存在之dsDNA 量成反比。可使用與套組相容之實時PCR儀器(例如ABI’s StepOne Plus 實時 PCR 系統或 lightTyper 儀器,Roche Diagnostics, Lewes, UK)來收集數據。 藉由使用適當軟體(例如lightTyper (Roche)或SDS Dissociation Curve, ABI)繪製螢光相對於溫度之負導數 (-d(螢光)/dT),y轴)與溫度(X軸)的圖線來構建熔融峰。分 157514.doc -86- 201209163 析數據以確定自dsDNA複合物快速轉變成單鏈分子時之、田 度。此溫度稱為Tm且與兩個分子間之相互作用強度成= 比。通常,Tm高於40。(:。 實例2 : BBC3聚核苷酸之調節 使用反義募核苷酸處理MCF-7細胞 實例2中所用之所有反義募核普酸皆如實例丨中所述進行 没计。指示製造商(IDT公司,Coralville,IA)來製造所設計 之硫代磷酸酯鍵寡核苷酸並提供表丨中所示之所設計硫代 磷酸酯類似物。核苷酸間之星形符號表示存在硫代磷酸酯 鍵。實例2中之實驗所需的寡核苷酸可使用任一適當技術 狀態之方法來合成’例如IDT所用之方法:在固體載體(例 如5微米定孔玻璃珠(CPG))上使用亞磷醯胺單體(常見核苷 酸,其所有活性基團皆經保護基團保護,例如糖上之三苯 甲基、A及C上之苯曱醢基及G上之N-2-異丁酿基)。保護 基團在寡核苷酸合成期間可防止發生不期望之反應。在合 ◎ 成過程結束時去除保護基團。經由3,碳使初始核苷酸連接 至固體載體且在3'至5,方向上進行合成。在以下4步中向生 長之募核苷酸鏈中添加新鹼基:1)使用三氣乙酸自固定核 苷酸之5’氧處去除保護基團;2)使用四唑使經固定核苷酸 與序列中之下一個核苷酸偶合至一起;經由四唑基亞磷醯 胺中間體使反應繼續進行;3)洗滌除去未反應之游離核苷 酸及反應副產物且將未反應之固定募核苷酸封端以防止其 參與下一輪合成;藉由使用乙酸酐及N-曱基咪唑對游離5, 經基實施乙醯化來達成封端;4)為穩定核苷酸間之鍵,使 157514.doc -87- 201209163 用姨及水(若欲產生構酸二酯鍵)、或Beaucage試劑(3H-1,2-苯并二硫醇-3-酮-1,1-二氧化物)(若期望硫代磷酸酯鍵)來 氧化磷。藉由交替使用兩種氧化劑,可構建嵌合主鏈。對 於序列中之每一核苷酸而言,皆重複上述4步循環。合成 完整序列時,在高溫下使用氫氧化銨自固體載體解離寡核 苷酸並實施去保護。藉由去鹽來洗滌除去保護基團且將剩 餘寡核苷酸凍乾。為實施實例2中所設計之實驗,在37°C 及5% C02下於生長培養基(MEM/EBSS(Hyclone目錄編號: SH30024,或 Mediatech 目錄編號:MT-10-010-CV)+10% FBS(Mediatech 目錄編號:MT35-011-CV)+ 青黴素 (penicillin)/鏈黴素(streptomycin)(Mediatech 目錄編號: MT30-002-CI))中生長來自ATCC之MCF-7細胞(目錄編號: HB-22)。在實驗前一天,以1.5χ105個細胞/ml之密度將細 胞再次平鋪於6孔板中並在37°C及5% C02下培育過夜。在 實驗當天,將6孔板中之培養基更換為新鮮生長培養基。 將由製造商以凍乾形式運送之寡核苷酸在不含 RNAse/DNAse之去離子水中稀釋至濃度為20 μΜ。將2 μΐ 此溶液與400 μΐ OptiMEM培養基(Gibco目錄編號:31985-070)及 4 μΐ Lipofectamine 2000 (Invitrogen 目錄編號: 11668019)在室溫下一起培育20 min,然後逐滴施加至6孔 板中具有MCF-7細胞之一個孔中。使用包含2 μΐ水代替寡 核苷酸溶液之相似混合物作為模擬轉染對照組。在37°C及 5% C02下培育3至18 h之後,將培養基更換為新鮮生長培 養基。添加反義寡核苷酸48 h後,去除培養基,且使用來 157514.doc •88· 201209163 自Promega之SV Total RNA分離系統(目錄編號:Z3105)或 來自Qiagen之RNeasy Total RNA分離套組(目錄編號·· 74181)根據製造商說明書自細胞提取RNA。將6〇〇 ng提取 之RNA添加至使用來自Thermo Scientific之Verso cDNA套 組(目錄編號:AB1453B)或高容量cDNA逆轉錄套組(目錄 編號:4368813)實施的逆轉錄反應中,如製造商方案中所 述。使用來自此逆轉錄反應之cDNA藉由實時PCR使用ABI Taqman Gene Expression Mix(目錄編號:4369510)及由 ABI 設計之引物 / 探針(Applied Biosystems Taqman Gene Expression Assay : Hs00248075_m 1 > Applied Biosystems 公司,Foster City CA)來監測基因表現。使用stepOne Plus 實時PCR機器(Applied Biosystems)來實施下列PCR循環: 在50°C下保持2 min,在95°C下保持10 min,40個循環之 (在95 C下保持15秒,在60°C下保持1 min)。基於經處理及 模擬轉染試樣間18S標準化dCt值之差值來計算使用反義寡 核苷酸處理後基因表現的變化倍數。 潜耒.·實時PCR結果顯示,在使用設計為BBC3反義 mRNA CA306306之寡核苷酸處理48 h之後,MCF-7細胞中 之BBC3 mRNA之含量顯著增加(圖1) 〇 實例3 :用耙向PUMA特異性天然反義轉錄物之反義募核苦 酸處理後原代人類纖維母細胞中的細胞凋亡程度增大 在實例3中’在原代人類纖維母細胞中以20 nM之最終濃 度測試靶向PUMA特異性天然反義轉錄物之反義寡核苷 酸。以下數據確認’經由調節PUMA特異性天然反義轉錄 157514.doc •89· 201209163 物之功能來上調PUMA mRNA與如由TUNEL分析測定之細 胞凋亡程度增大有關。 材料及方法 用乾向#異磔天资及4 #錄勿之及J募核苷酸處 理次托乂庸.鐵襻母鈿虑。於37。(:及5% C02下使由Dr. N-Kenyon (University of Miami)引入培養基中之原代人類 皮膚纖維母細胞在由a-MEM(Gibco,目錄號:12561- 056)+10% FBS(Mediatech,目錄號:35-015 CV)+l〇/〇 Antimycotic-Antibiotic(Gibco,目錄號:15240-062)組成 之生長培養基中生長。使用Next Day方法用反義募核苷酸 處理細胞。在實驗前一天,以約2X1 〇5個細胞/孔之密度將 細胞平鋪於生長培養基中之6孔板中並在37°C及5% C02下 培育過夜。第二天,將6孔板中之培養基更換為新鮮培養 基(1.5 ml/well)並對細胞投與反義募核苷酸。所有反義募 核普酸均係由IDT公司(Coralville,IA)製造。所有寡核苷酸 之序列列於表1中。將寡核苷酸之儲存溶液於不含 DNAse/RNAse之無菌水中稀釋至20 uM之濃度。向一個孔 中投與2 ul此溶液與400 ul 〇pti_MEM培養基(Gibc〇目錄編 號:31985-070)及 4 ul Lip〇fectamine 2000 (Invitrogen 目錄 編號:11668019),將其在室溫下一起培育2〇 min,且逐滴 施加至6孔板中具有細胞之一個孔中。使用包含2 ul水代替 寡核苷酸溶液之相似混合物作為模擬轉染對照組。使用相 同濃度之無活性募核苷酸CUR-1 505作為無活性對照組。 在37 C及5% C〇2下培育約18 h之後,將培養基更換為新鮮 157514.doc •90- 201209163 生長培養基。添加反義寡核苷酸48小時後,去除培養基並 使細胞胰蛋白酶化並以約70,000個細胞/孔之密度重新平鋪 於96孔板中。在37°C及5% C〇2下培育24小時後,使用細胞 進行TUNEL細胞凋亡分析。 . rt/见知虑源亡分於。使用HT Titer TACS分析套組根 據製造商之說明書(Trevigen目錄編號:4822-96-K)檢測細 胞中之細胞凋亡程度。簡言之,將細胞固定於3.7%緩衝曱 醛溶液(Sigma-252549)中,用 PBS 及 100% 甲醇(Sigma_ 〇 厘1775-1〇八)沖洗並在用於分析之前在+4°(:下儲存於80%乙 醇(Sigma-493511)中。為標記DNA斷裂,去除乙醇,且使 用PB S洗務細胞並與蛋白酶K溶液在室溫下一起培育15分 鐘,在水及PBS中沖洗,然後與TACS核酸酶溶液在37°C下 一起培育10分鐘。培育後,使用PBS沖洗細胞,且添加過 氧化氫溶液(Sigma-MKBD1394)。在室溫培育5分鐘後,於 室溫下將細胞與1X TdT標記缓衝液一起培育5分鐘,然後 q 丟棄缓衝液。隨後將細胞與標記反應混合物在37。(:下一起 培育一小時,然後添加lx TdT終止缓衝液並保持5分鐘。 標記反應停止後,使用PBS洗蘇細胞,與Strep-HRP溶液在 ’ 室溫下一起培育10分鐘,使用PBS/Tween洗務且與TACS蘭 寶石在暗處一起培育30分鐘。藉由添加0.2N HC1 (Fluka-343102)來停止反應,且在450 nm下於板讀數儀中讀取吸 光度。除非另有說明,否則上述所有溶液均係以HT Titer TACS分析套組供應。 .结耒,如圖2中所示,在用20 nM靶向PUMA特異性天然 157514.doc •91- 201209163 反義轉錄物之反義寡核苷酸處理後,原代人類纖維母細胞 中之細胞凋亡程度增大超過5倍,該反義寡核苷酸先前顯 示可上調PUMA mRNA含量(圖1,實例2)。與經具有類似 化學性質之無活性寡核苷酸處理的對照組試樣相比,此上 調顯著。結果指示,使用靶向PUMA特異性天然反義轉錄 物之寡核苷酸上調PUMA mRNA可上調功能PUMA蛋白並 增大細胞 >周亡程度。 實例4 :用靶向PUMA特異性天然反義轉錄物之反義寡核普 酸處理的原代人類纖維母細胞中的降低增殖能力 在實例4中’測試靶向PUMA特異性天然反義轉錄物之反 義寡核苷酸抑制原代人類纖維母細胞之菌落形成的能力。 以下數據確認,經由調節PUMA特異性天然反義轉錄物之 功能來上調PUMA mRNA可使寡核苷酸處理之細胞中的增 殖降低。 材料及方法 選產#立分#。於37°C及5% C02下使由Dr. N.Kenyon (University of Miami)引入培養基中之原代人類皮膚纖維母 細胞在由 a-MEM(Gibco,目錄號:12561-056)+10% FBS(Nlediatech ’ 目錄 . 35-015 CV)+1% Antimycotic-Those skilled in the art will recognize that a wide range of gene regions suitable for use in the present invention can be selected. X can specifically hybridize": a compound can bind to a dry nucleic acid when it binds to a dry nucleic acid to interfere with the normal function of the dry nucleic acid to regulate function and/or activity' and to have sufficient complementarity to avoid antisense compound plaque The nucleic acid sequence undergoes non-specific binding under conditions where it is desired to undergo specific binding (i.e., under physiological conditions in the case of in vivo analysis or therapeutic treatment, and under conditions in which the assay is performed in the context of in vitro analysis). ^ The hybridity f of the oligoacid described herein can be measured by an in vitro assay known in the art. For example, VIII, & 丨-wax is obtained by using a smelting curve to determine the binding strength of an innocent molecule to a potential drug molecule to obtain the properties of the oligonucleotides described herein. And = any of the established methods of measuring the strength of the intermolecular interaction (eg '(four) curve analysis) to estimate the combination of natural antisense molecules with potential 157514.doc •85· 201209163 drug molecules (molecules) strength. Melting curve analysis measures the temperature at which a natural antisense molecule/molecular complex rapidly transitions from a double strand to a single stranded form. This temperature has been widely accepted as a reliable measure of the strength of the interaction between two molecules. Melt curve analysis can be performed using a cDNA copy of the actual native antisense RNA molecule or a synthetic DNA or RNA nucleotide corresponding to the molecular binding site. Multiple kits containing all necessary reagents to perform this assay (e.g., Applied Biosystems, MeltDoctor kit) can be used. Such kits comprise a suitable buffer solution containing one of a double stranded DNA (dsDNA) binding dye (e.g., ABI HRM dye, SYBR Green, SYTO, etc.). The nature of the dsDNA dye is that it emits almost no fluorescence in free form, but has high fluorescence when bound to dsDNA. To perform the analysis, the cDNA or corresponding oligonucleotide is mixed with the molecule at a concentration defined by a particular manufacturer's protocol. The mixture was heated to 95 °C to dissociate all preformed dsDNA complexes and then slowly cooled to room temperature or other lower temperatures as defined by the kit manufacturer to anneal the DNA molecules. The newly formed composite was then slowly heated to 95 ° C while continuing to collect data on the amount of fluorescence produced in 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 Real-Time PCR System or lightTyper instrument, Roche Diagnostics, Lewes, UK). Draw a plot of the negative derivative of fluorescence relative to temperature (-d (fluorescence) / dT), y-axis) and temperature (X-axis) by using appropriate software (eg lightTyper (Roche) or SDS Dissociation Curve, ABI) To build a melting peak. 157514.doc -86- 201209163 Analyze the data to determine the field when the dsDNA complex is rapidly converted to a single-stranded molecule. This temperature is called Tm and is proportional to the intensity of interaction between the two molecules. Typically, the Tm is above 40. (Example 2. Example 2: Regulation of BBC3 Polynucleotide Treatment of MCF-7 Cells Using Antisense Nucleotide Nucleotide All antisense priming acids used in Example 2 were performed as described in the Examples. Commercial (IDT, Coralville, IA) to manufacture the designed phosphorothioate-bonded oligonucleotides and provide the designed phosphorothioate analogs shown in the table. The inter-nucleotide star symbol indicates the presence Phosphorothioate linkages. The oligonucleotides required for the experiments in Example 2 can be synthesized using any suitable state of the art method, such as the method used for IDT: in a solid support (eg, 5 micron fixed glass beads (CPG)) The use of a phosphoramidite monomer (common nucleotides, all of which are protected by a protecting group, such as a trityl group on a sugar, a benzoquinone on A and C, and a N on G) 2-Isobutyl-based. The protecting group prevents undesired reactions during oligonucleotide synthesis. The protecting group is removed at the end of the synthesis process. Via 3, the carbon is attached to the initial nucleotide. Solid carrier and synthesis in the direction of 3' to 5, in the following 4 steps Adding a new base to the nucleotide chain: 1) using tri-acetic acid to remove the protecting group from the 5' oxygen of the fixed nucleotide; 2) using tetrazole to immobilize the nucleotide with the next nucleotide in the sequence Coupling together; the reaction is continued via the tetrazolylphosphinamide intermediate; 3) washing removes unreacted free nucleotides and reaction by-products and capping unreacted fixed nucleotides to prevent their participation The next round of synthesis; by using acetic anhydride and N-mercaptopimid for free 5, acetylation of the base to achieve end-capping; 4) to stabilize the bond between nucleotides, so that 157514.doc -87-201209163姨 and water (if you want to produce acid diester bonds), or Beaucage reagent (3H-1,2-benzodithiol-3-one-1,1-dioxide) (if desired phosphorothioate linkage) ) to phosphorus oxide. The chimeric backbone can be constructed by alternately using two oxidizing agents. The above 4-step cycle is repeated for each nucleotide in the sequence. When the complete sequence is synthesized, the oligonucleotide is cleaved from the solid support at a high temperature using ammonium hydroxide and deprotected. The protecting group is removed by washing with desalting and the remaining oligonucleotide is lyophilized. To carry out the experiment designed in Example 2, the growth medium (MEM/EBSS (Hyclone catalog number: SH30024, or Mediatech catalog number: MT-10-010-CV) + 10% FBS at 37 ° C and 5% CO 2 ) (Mediatech catalog number: MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number: MT30-002-CI)) MCF-7 cells grown from ATCC (Catalog No.: HB- twenty two). On the day before the experiment, the cells were plated again in a 6-well plate at a density of 1.5 χ 105 cells/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. Oligonucleotides delivered by the manufacturer in lyophilized form were diluted to a concentration of 20 μM in deionized water without RNAse/DNAse. 2 μΐ of this solution was incubated with 400 μΐ OptiMEM medium (Gibco catalog number: 31985-070) and 4 μL Lipofectamine 2000 (Invitrogen catalog number: 11668019) for 20 min at room temperature, and then applied dropwise to a 6-well plate. One well of MCF-7 cells. A similar mixture containing 2 μM water instead of the oligonucleotide solution was used as a mock transfection control group. After incubation for 3 to 18 h at 37 ° C and 5% CO 2 , the medium was changed to fresh growth medium. After adding antisense oligonucleotides for 48 h, the medium was removed and used to 157514.doc •88· 201209163 from Promega's SV Total RNA Isolation System (Catalog No.: Z3105) or RNeasy Total RNA Isolation Kit from Qiagen (catalog) No. 74181) RNA was extracted from cells according to the manufacturer's instructions. 6 ng of extracted RNA was added to the reverse transcription reaction using the Verso cDNA kit from Thermo Scientific (Catalog No.: AB1453B) or the high-capacity cDNA reverse transcription kit (Catalog No.: 4368813), such as the manufacturer's protocol. Said in the middle. The cDNA from this reverse transcription reaction was used by real-time PCR using ABI Taqman Gene Expression Mix (catalog number: 4369510) and primers/probes designed by ABI (Applied Biosystems Taqman Gene Expression Assay: Hs00248075_m 1 > Applied Biosystems, Foster City CA) to monitor gene expression. The following PCR cycles were performed using a stepOne Plus real-time PCR machine (Applied Biosystems): 2 min at 50 °C, 10 min at 95 °C, 40 cycles (15 seconds at 95 C, at 60 °) Keep 1 min under C). The fold change in gene expression after treatment with antisense oligonucleotides was calculated based on the difference in 18S normalized dCt values between treated and mock transfected samples. The real-time PCR results showed that the content of BBC3 mRNA in MCF-7 cells was significantly increased after 48 h treatment with oligonucleotides designed to be BBC3 antisense mRNA CA306306 (Fig. 1). Example 3: For 耙Increased degree of apoptosis in primary human fibroblasts following antisense nucleoside treatment of PUMA-specific natural antisense transcripts in Example 3 'in final concentration of 20 nM in primary human fibroblasts Antisense oligonucleotides targeting PUMA-specific natural antisense transcripts were tested. The following data confirms that up-regulation of PUMA mRNA by modulating the function of PUMA-specific natural antisense transcription 157514.doc •89·201209163 is associated with an increased degree of apoptosis as determined by TUNEL analysis. Materials and methods Use the dry direction #异磔天资 and 4 #录勿之和J Raised nucleotides to deal with the second sub-Yu Yong. Iron 襻 mother care. At 37. (: and 5% C02 of primary human skin fibroblasts introduced into the medium by Dr. N-Kenyon (University of Miami) by a-MEM (Gibco, catalog number: 12561-056) + 10% FBS ( Mediatech, catalog number: 35-015 CV) + l〇/〇Antimycotic-Antibiotic (Gibco, catalog number: 15240-062) was grown in growth medium. Cells were treated with antisense nucleotides using the Next Day method. One day before the experiment, the cells were plated at a density of about 2×1 〇 5 cells/well in 6-well plates in growth medium and incubated overnight at 37° C. and 5% CO 2 . The next day, 6-well plates were placed. The medium was changed to fresh medium (1.5 ml/well) and the cells were administered antisense nucleotides. All antisense nucleoside acids were manufactured by IDT (Coralville, IA). Sequence of all oligonucleotides Listed in Table 1. The oligonucleotide storage solution was diluted to a concentration of 20 uM in sterile water without DNAse/RNAse. 2 ul of this solution was administered to one well with 400 ul of 〇pti_MEM medium (Gibc〇 catalog No.: 31985-070) and 4 ul Lip〇fectamine 2000 (Invitrogen catalog number: 11668019), which will Incubate for 2 〇 min at room temperature and apply dropwise to one well of the 6-well plate with cells. A similar mixture containing 2 ul of water instead of the oligonucleotide solution was used as a mock transfection control group. Inactive nucleotide CUR-1 505 was used as an inactive control group. After incubation for 18 hours at 37 C and 5% C〇2, the medium was changed to fresh 157514.doc •90- 201209163 growth medium. Add antisense After 48 hours of oligonucleotide removal, the medium was removed and the cells were trypsinized and re-plated in 96-well plates at a density of approximately 70,000 cells/well. After incubation at 37 ° C and 5% C 〇 2 for 24 hours Cells were used for TUNEL apoptosis analysis. rt/see the source of death. The cells were assayed using the HT Titer TACS assay kit according to the manufacturer's instructions (Trevigen catalog number: 4822-96-K). In short, the cells were fixed in 3.7% buffered furfural solution (Sigma-252549), rinsed with PBS and 100% methanol (Sigma_ 1775-1〇8) and used at +4 before analysis. °(: stored in 80% ethanol (Sigma-493511). The labeled DNA was cleaved, the ethanol was removed, and the cells were washed with PB S and incubated with proteinase K solution for 15 minutes at room temperature, rinsed in water and PBS, and then incubated with TACS nuclease solution for 10 minutes at 37 °C. . After the incubation, the cells were washed with PBS, and a hydrogen peroxide solution (Sigma-MKBD1394) was added. After incubation for 5 minutes at room temperature, the cells were incubated with 1X TdT labeling buffer for 5 minutes at room temperature, then q was discarded. The cells were then reacted with the labeling reaction mixture at 37. (: Incubate for one hour, then add lx TdT to stop the buffer for 5 minutes. After the labeling reaction is stopped, wash the cells with PBS and incubate with Strep-HRP solution for 10 minutes at room temperature using PBS/Tween Wash and incubate for 30 minutes with TACS sapphire in the dark. Stop the reaction by adding 0.2 N HCl (Fluka-343102) and read the absorbance at 450 nm in a plate reader unless otherwise stated. All of the above solutions were supplied in the HT Titer TACS assay kit. . As shown in Figure 2, antisense oligos targeting PUMA-specific natural 157514.doc •91-201209163 antisense transcripts at 20 nM After nucleotide treatment, the degree of apoptosis in primary human fibroblasts increased more than 5-fold, and the antisense oligonucleotide was previously shown to upregulate PUMA mRNA content (Fig. 1, Example 2). This up-regulation was significant compared to the chemically inactive oligonucleotide-treated control samples. The results indicate that up-regulation of PUMA mRNA can up-regulate PUMA protein using oligonucleotides targeting PUMA-specific natural antisense transcripts and Increase cell > week Degree 4. Example 4: Reduced Proliferative Capacity in Primary Human Fibroblasts Treated with Antisense Oligonucleotides Targeting PUMA-Specific Natural Antisense Transcripts In Example 4 'Test Targeted PUMA Specific Natural Antisense Antisense oligonucleotides of transcripts inhibit the ability of colonies of primary human fibroblasts. The following data confirm that up-regulation of PUMA mRNA by modulating the function of PUMA-specific natural antisense transcripts allows oligonucleotides to be processed Proliferation in cells is reduced. Materials and Methods Selection #立分#. The primary human skin fibroblasts introduced into the medium by Dr. N. Kenyon (University of Miami) at 37 ° C and 5% C02 a-MEM (Gibco, catalog number: 12561-056) + 10% FBS (Nlediatech ' catalog. 35-015 CV) + 1% Antimycotic-

Antibiotic(Gibco,目錄號:1 5240-062)組成之生長培養基 中生長。使用Next Day方法用反義寡核苷酸處理細胞。在 實驗前一天,以約2x 105個細胞/孔之密度將細胞平鋪於生 長培養基中之6孔板中並在37°C及5% C02下培育過夜。第 -一天’將6孔板中之培養基更換為新鮮培養基(1.5 ml/well) 157514.doc -92- 201209163 並對細胞投與反義寡核。所有反義寡核苦酸均係由 IDT公司(C〇ralviUe,IA)製造。所有寡核苷酸之序列列於表 1中。將寡核苷酸之儲存溶液於不含DNAse/RNAse之無菌 水中稀釋至20 uM之濃度。向一個孔中投與2 ul此溶液與 400 ul Opu-MEM 培養基(Gibco 目錄編號:31985-070)及 4 ui Lipofectamine 2000 (invitrogeng 錄編號:ι1668〇19), 將其在室溫下一起培育2〇 min,且逐滴施加至6孔板中具 有細胞之一個孔中。使用包含2 ul水代替募核苷酸溶液之 相似混合物作為模擬轉染對照組。使用相同濃度之無活性 券核苷酸CUR-1505作為對照組。在37°C及5% C02下培育 約1 8 h之後’將培養基更換為新鮮生長培養基。添加反義 寡核苦酸48小時後,去除培養基並使細胞胰蛋白酶化並以 約5000個細胞/孔之密度重新平鋪於6孔板中。將板於 3 7°C、5% C〇2下培育5至7天直至形成3至10個細胞之菌 落°固定菌落並為具有更好可視性而使用存於95%乙醇中 之0.25 % 1,9-二曱基-亞曱基藍(sigma Aldrich,目錄號 34,108.8)染色並用PBS沖洗。對菌落進行計數並將其數量 與經無活性募核苷酸處理之對照孔及經模擬轉染對照組進 行比較。 .结果’如圖3中所示,與經無活性寡核苷酸及模擬轉染 對照組處理之細胞相比,由經靶向PUMA特異性天然反義 轉錄物之反義募核苷酸處理之原代纖維母細胞形成的菌落 的數量減少。結果指示,由反義募核苷酸上調之PUMA mRNA產生功能蛋白並減少活體外細胞增殖。 157514.doc •93- 201209163 儘管已根據一或多個實施方案闡釋及闡述了本發明,但 熟習此項技術者在閱讀並理解本說明書及附圖後可作出等 效改變及修改。此外,儘管可能只根據多種實施方案中之 一者揭示了本發明之特定特徵,但該特徵可與其他實施方 案之一或多個其他特徵組合,此對於任何給定或特定之應 用而言可能係合乎要求且有利的。 本揭示内容之摘要使得讀者可快速地確定技術揭示内容 之性質。提交本摘要係基於以下理解:其並非用於解釋或 限制下列申請專利範圍之範疇或含義。 【圖式簡單說明】 圖1顯示藉由投·與MCF7細胞靶向PUMA特異性天然反義 轉錄物之反義寡核苷酸的PUMA mRNA之上調。對MCF7乳 腺癌細胞系投與20 nM靶向PUMA特異性天然反義轉錄物 之不同反義寡核苷酸(CUR-1675、CUR-1676、CUR-1677 、 CUR-1678 及 CUR-1679) 。 投與48h後 ,藉由 逆轉錄 (RT)實時聚合酶鏈反應(PCR)對PUMA mRNA進行定量。基 於經投與細胞(向細胞中添加CUR募核苷酸)與模擬細胞(未 向細胞中添加寡核苷酸)之間之1 8S標準化差值計算因 PUMA mRNA之募核苷酸處理的相對表現變化。表示為 CUR-1675、CUR-1676、CUR_1677、CUR-1678 及 CUR-1679之條形 分別對應於經SEQ ID NO: 3 至 7處理之試樣。 圖2顯示用靶向PUMA特異性天然反義轉錄物之反義寡核 苷酸處理後之原代人類纖維母細胞中的細胞凋亡程度的增 大,如由TUNEL分析所測定。CUR-1675-先前顯示上調 157514.doc -94· 201209163 PUMA mRNA之靶向PUMA天然反義轉錄物的寡核苷酸; CUR-1505-具有類似化學性質之無活性寡核苷酸。.表示 為CUR-1675及CUR-1505之條形分別對應於使用SEQ ID NO: 3及8處理之試樣。***指示Ρ<0·001 圖3顯示用靶向PUMA特異性天然反義轉錄物之反義寡核 苷酸處理之原代人類纖維母細胞中的降低增殖能力,如由 選殖發生分析所測定。CUR-1675-先前顯示上調PUMA mRNA之靶向PUMA天然反義轉錄物的寡核苷酸;CUR-1505- 具有 類似化 學性質 之無活 性寡核 苷酸。 表示為 CUR-1675 及 CUR-1509 之條形分別對應於使用 SEQ ID NO: 3及 8 處理之試樣。*指示Ρ<0·05。 序列表說明-SEQ ID NO: 1 :智人(Homo sapiens) BCL2 結合部分3(BBC3),轉錄物變體3,mRNA(NCBI登錄號: NM_001127242); SEQ ID NO: 2:天然 BBC3反義序列 (CA3 06306) ; SEQ ID NO: 3至8 :反義寡核苷酸。*表示硫 代磷酸酯鍵。 157514.doc 95- 201209163 序列表 <110>美商歐科可娜有限責任公司 <120>藉由抑制BCL2結合部分3 (BBC3)之天然反義轉錄物以治療BBC3相關疾病 <130> BBC3 <140> 100124653 <141> 2011-07-12 <150> 61/363,535 <151> 2010-07-12 <160> 8 <170> Patentln version 3.5 <210> 1 <211> 1347 <212> m <213 >智人 <400> 1 gaggegattg cgattgggtg agacccagta aggatggaaa gtgtagagga gacaggaatc 60Growth was carried out in a growth medium consisting of Antibiotic (Gibco, catalog number: 1 5240-062). Cells were treated with antisense oligonucleotides using the Next Day method. On the day before the experiment, cells were plated at a density of about 2 x 105 cells/well in 6-well plates in growth medium and incubated overnight at 37 ° C and 5% CO 2 . Day - Day 'Change the medium in the 6-well plate to fresh medium (1.5 ml/well) 157514.doc -92- 201209163 and administer the antisense oligo to the cells. All antisense oligonucleotides are manufactured by IDT (C〇ralviUe, IA). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 uM in sterile water without DNAse/RNAse. 2 ul of this solution was administered to a well with 400 ul of Opu-MEM medium (Gibco catalog number: 31985-070) and 4 ui Lipofectamine 2000 (invitrogeng number: ι1668〇19), which were incubated together at room temperature 2 〇min, and applied dropwise to one well of a 6-well plate with cells. A similar mixture containing 2 ul of water instead of the nucleotide solution was used as a mock transfection control group. The same concentration of the inactive conjugate nucleotide CUR-1505 was used as a control group. After incubation for about 18 h at 37 ° C and 5% CO 2 , the medium was changed to fresh growth medium. After 48 hours of addition of antisense oligonucleotide, the medium was removed and the cells were trypsinized and re-plated into 6-well plates at a density of about 5000 cells/well. The plate was incubated at 37 ° C, 5% C 〇 2 for 5 to 7 days until colonies of 3 to 10 cells were formed. Fixed colonies and used for 0.25 % in 95% ethanol for better visibility 1 9-Dimercapto-indenyl blue (sigma Aldrich, Cat. No. 34, 108.8) was stained and rinsed with PBS. Colonies were counted and compared to control wells treated with inactive nucleotides and mock transfected controls. The results, as shown in Figure 3, were treated with antisense raised nucleotides targeting PUMA-specific natural antisense transcripts compared to cells treated with inactive oligonucleotides and mock-transfected controls. The number of colonies formed by the primary fibroblasts is reduced. The results indicate that PUMA mRNA upregulated by antisense raised nucleotides produces a functional protein and reduces in vitro cell proliferation. 157514.doc • 93- 201209163 Although the present invention has been illustrated and described in accordance with one or more embodiments of the present invention, it will In addition, although specific features of the invention may be disclosed in accordance with one of the various embodiments, the features may be combined with one or more other features of other embodiments, which may be possible for any given or particular application. It is desirable and advantageous. The Abstract of the Disclosure allows the reader to quickly ascertain the nature of the technical disclosure. The Abstract is submitted with the understanding that it is not intended to limit or limit the scope or meaning of the following claims. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows the up-regulation of PUMA mRNA by antisense oligonucleotides targeting PUMA-specific natural antisense transcripts with MCF7 cells. The MCF7 breast cancer cell line was administered 20 nM of different antisense oligonucleotides targeting CUMMA-specific natural antisense transcripts (CUR-1675, CUR-1676, CUR-1677, CUR-1678 and CUR-1679). After 48 h of administration, PUMA mRNA was quantified by reverse transcription (RT) real-time polymerase chain reaction (PCR). The relative nucleotide processing of PUMA mRNA was calculated based on the 1 8S normalized difference between the administered cells (adding CUR nucleotides to the cells) and the mock cells (no oligonucleotide added to the cells). Performance changes. The bars designated CUR-1675, CUR-1676, CUR_1677, CUR-1678, and CUR-1679 correspond to the samples treated with SEQ ID NOS: 3 to 7, respectively. Figure 2 shows the increase in the degree of apoptosis in primary human fibroblasts treated with antisense oligonucleotides targeting PUMA-specific natural antisense transcripts as determined by TUNEL analysis. CUR-1675 - previously shown upregulated 157514.doc -94 · 201209163 PUMA mRNA targeting oligonucleotides of PUMA natural antisense transcript; CUR-1505 - inactive oligonucleotide with similar chemical properties. The bars indicated as CUR-1675 and CUR-1505 correspond to the samples treated with SEQ ID NOS: 3 and 8, respectively. ***Indication Ρ<0·001 Figure 3 shows the ability to reduce proliferation in primary human fibroblasts treated with antisense oligonucleotides targeting PUMA-specific natural antisense transcripts, as analyzed by selection Measured. CUR-1675 - an oligonucleotide previously shown to up-regulate PUMA mRNA targeting PUMA natural antisense transcript; CUR-1505 - an inactive oligonucleotide with similar chemical properties. The bars denoted as CUR-1675 and CUR-1509 correspond to the samples treated with SEQ ID NOS: 3 and 8, respectively. *Indication Ρ<0·05. BRIEF DESCRIPTION OF THE SEQUENCE LISTING - SEQ ID NO: 1 : Homo sapiens BCL2 binding part 3 (BBC3), transcript variant 3, mRNA (NCBI accession number: NM_001127242); SEQ ID NO: 2: natural BBC3 antisense sequence (CA3 06306); SEQ ID NOS: 3 to 8: antisense oligonucleotides. * indicates a phosphorothioate bond. 157514.doc 95- 201209163 Sequence Listing <110> American Okokena Co., Ltd. <120> By treating BCL2 binding part 3 (BBC3) natural antisense transcript to treat BBC3 related diseases <130> BBC3 <140> 100124653 <141> 2011-07-12 <150> 61/363,535 <151> 2010-07-12 <160> 8 <170> Patentln version 3.5 <210> 1 <211> 1347 <212> m <213 > Homo sapiens <400> 1 gaggegattg cgattgggtg agacccagta aggatggaaa gtgtagagga gacaggaatc 60

Ο cacggctttg gaaaaaggaa ggacaaaact caccaaacca gageagggea ggaagtaaca 120 atgagaaact gaaaaagaaa cggaatggaa agetatgaga caggatgaaa tttggcatgg 180 ggtctgccca ggcatgtcca tgccaggtgc ccagggctgc ttccacgacg tgggtcccct 240 gccagatttg tgagacaaga ggagcagcag cggcaccgcc cctcaccctg gagggtcctg 300 tacaatctca tcat£ggact cctgccctta cccaggggcc acagagcccc egagatggag 360 cccaattagg tgcctgcacc cgcccggtgg acgtcaggga ctcggggggc aggcccctcc 420 cacctcctga caccctggcc agcgcggggg actttctctg caccatgtag catactggac 480 tcccagccct gcctgtcccg ggggcgggcc ggggcagcca ctccagcccc agcccagcct 540 ggggtgcact gaeggagatg cggactcctg ggtccctggc caagaagcca ggagagggac 600 ggctgatgga ctcagcatcg gaaggtggcg gtgaccgagg gggtggggac tgagccgccc 660 gcctctgccg cccaccacca tctcaggaaa ggctgttgtg ctggtgcccg ttccagctgc 720 aggggtgaca ctgggagggg ggggctctcc tctcggtgct ccttcactct gggcctggcc 780 tcaggcccct ggtgcttccc cccctcctcc tgggaggggg cccgtgaaga gcaaatgagc 840 caaacgtgac cactagcctc ctggagccag agagtggggc tcgtttgccg gttgctccag 900 cccggcgccc agccatcttc cctgagccag ccggcgggtg gtgggcatgc ctgcctcacc 960 ttcatcaggg ggtggccagg aggggcccag actgtgaatc ctgtgctctg cccgtgaccg 1020 ccccccgccc catcaatccc attgeatagg tttagagaga gcacgtgtga ccactggcat 1080 tcatttgggg ggtgggagat tttggctgaa gccgccccag ccttagtccc cagggccaag 1140 cgctgggggg aagacgggga gtcagggagg gggggaaatc teggaagagg gaggagtetg 1200 ggagtgggga gggatggccc agcctgtaag atactgtata tgcgctgctg tagataccgg 1260 aatgaatttt ctgtacatgt ttggttaatt ttttttgtac atgatttttg tatgtttcct 1320 tttcaataaa atcagattgg aacagtg 1347 <210> 2 <211> 689 <212> DNA <213>智人 <400> 2 cttttctttt ctttttttag taaagatggg gtctcattat gttgcccagg ttggtctcaa 60 actcctgggc tcaa£tgatc ctccctcctt ggcctcccaa agtgcctgga ttatggtcat 120 157514-序列表.doc 201209163 gaggcaccat gcccagcccc ggaatgttct tatttstttg tttgtttgta gagacagggt 180 ctcactatgt tgcccaggct ggtctcaaat tccagacctc aagcaatcgt tcatccttga 240 cctcccaaaa tgctgggatt acaggtgtga gccaccacac cggcctataa tgttcttttg 300 agaagaaaat gagtgtgaag gatagtg££a atctggcttg gtggctggag gtagaaaaga 360 tggggasagg ccaagagtct atttggccat aagcgttatc actagtcaaa atcctcccca 420 cagctgggcg tggtggctca cgtctgcaat cccagtatca cggaaggccg agatgggtgg 480 atcacctgag cccaggagtt ccagaccagc ctgggcaaca tgcggaaacc taactgggga 540 ggctggggtg ggaggaacgc ttgagcctgt gaggtcgagg ctgcagtagg ctgtgatcgc 600 acattgcact ccagcttggg cgacagagcg agaccctttc tcaaaaaaaa aaaaaacggc 660 atggccgcgg ccctcgtgcc gaattcttg 689 <210> 3 <211> 20 <212> DNA <213>人工序列 <220> <223>反義募核苷 <400> 3 ccaccaagcc agattcccac 20 <210> 4 <211> 20 <212> DNA <213>人工序列 <220> <223>反義募核苷 <400> 4 atccacccat ctcggccttc 20 <210> 5 <211> 20 <212> DNA <213>人工序列 <220> <223>反義寡核苷 <400> 5 aggctcaagc gttcctccca 20 <210> 6 <211> 21 <212> DNA <213>人工序列 <220> <223>反義寡核苷 <400> 6 cccagttagg tttccgcatg t 21 <210> 7 <211> 20 <212> DNA <213>人工序列 <220> <223>反義募核苷 -2 - 157514-序列表.doc 201209163 <4〇0> 7 tctgtcgccc aagctggagt <210> 8 <211> 21 <212> DNA <213>人工序列 <220> <223>反義寡核苷 <400> 8 cctctccacg cgcagtacatΟ cacggctttg gaaaaaggaa ggacaaaact caccaaacca gageagggea ggaagtaaca 120 atgagaaact gaaaaagaaa cggaatggaa agetatgaga caggatgaaa tttggcatgg 180 ggtctgccca ggcatgtcca tgccaggtgc ccagggctgc ttccacgacg tgggtcccct 240 gccagatttg tgagacaaga ggagcagcag cggcaccgcc cctcaccctg gagggtcctg 300 tacaatctca tcat £ ggact cctgccctta cccaggggcc acagagcccc egagatggag 360 cccaattagg tgcctgcacc cgcccggtgg acgtcaggga ctcggggggc aggcccctcc 420 cacctcctga caccctggcc agcgcggggg actttctctg caccatgtag catactggac 480 tcccagccct gcctgtcccg ggggcgggcc ggggcagcca ctccagcccc agcccagcct 540 ggggtgcact gaeggagatg cggactcctg ggtccctggc caagaagcca ggagagggac 600 ggctgatgga ctcagcatcg gaaggtggcg gtgaccgagg gggtggggac tgagccgccc 660 gcctctgccg cccaccacca tctcaggaaa ggctgttgtg ctggtgcccg ttccagctgc 720 aggggtgaca ctgggagggg ggggctctcc tctcggtgct ccttcactct gggcctggcc 780 tcaggcccct ggtgcttccc cccctcctcc tgggaggggg cccgtgaaga gcaaatgagc 840 caaacgtgac cactagcctc ctggagccag agagtggggc tcgtttgccg gttgctccag 900 cccggcgccc agcca tcttc cctgagccag ccggcgggtg gtgggcatgc ctgcctcacc 960 ttcatcaggg ggtggccagg aggggcccag actgtgaatc ctgtgctctg cccgtgaccg 1020 ccccccgccc catcaatccc attgeatagg tttagagaga gcacgtgtga ccactggcat 1080 tcatttgggg ggtgggagat gccgccccag ccttagtccc cagggccaag 1140 cgctgggggg aagacgggga gtcagggagg gggggaaatc teggaagagg gaggagtetg 1200 ggagtgggga gggatggccc agcctgtaag atactgtata tgcgctgctg tagataccgg 1260 aatgaatttt ctgtacatgt ttggttaatt ttttttgtac atgatttttg tatgtttcct 1320 tttcaataaa atcagattgg tttggctgaa Aacagtg 1347 <210> 2 <211> 689 <212> DNA <213> Homo sapiens <400> 2 cttttctttt ctttttttag taaagatggg gtctcattat gttgcccagg ttggtctcaa 60 actcctgggc tcaa£tgatc ctccctcctt ggcctcccaa agtgcctgga ttatggtcat 120 157514-sequence table.doc 201209163 gaggcaccat gcccagcccc ggaatgttct tatttstttg tttgtttgta gagacagggt 180 ctcactatgt tgcccaggct ggtctcaaat tccagacctc aagcaatcgt tcatccttga 240 cctcccaaaa tgctgggatt acaggtgtga gccaccacac cggcctataa tgttcttttg 300 agaagaaaat gagtgtgaag gata gtg ££ a atctggcttg gtggctggag gtagaaaaga 360 tggggasagg ccaagagtct atttggccat aagcgttatc actagtcaaa atcctcccca 420 cagctgggcg tggtggctca cgtctgcaat cccagtatca cggaaggccg agatgggtgg 480 atcacctgag cccaggagtt ccagaccagc ctgggcaaca tgcggaaacc taactgggga 540 ggctggggtg ggaggaacgc ttgagcctgt gaggtcgagg ctgcagtagg ctgtgatcgc 600 acattgcact ccagcttggg cgacagagcg agaccctttc tcaaaaaaaa aaaaaacggc 660 atggccgcgg ccctcgtgcc gaattcttg 689 < 210 & gt 3 <211> 20 <212> DNA <213>Artificial sequence<220><223> Antisense nucleoside <400> 3 ccaccaagcc agattcccac 20 <210> 4 <211> 20 <;212> DNA <213>Artificial sequence<220><223> Antisense nucleoside <400> 4 atccacccat ctcggccttc 20 <210> 5 <211> 20 <212> DNA <213> Artificial sequence <220><223> antisense oligonucleoside <400> 5 aggctcaagc gttcctccca 20 <210> 6 <211> 21 <212> DNA <213> artificial sequence <220>223>antisense oligonucleoside <400> 6 cccag Ttagg tttccgcatg t 21 <210> 7 <211> 20 <212> DNA <213>Artificial sequence <220><223> Antisense nucleoside-2 - 157514 - Sequence Listing.doc 201209163 <4〇0> 7 tctgtcgccc aagctggagt <210> 8 <211> 21 <212> DNA <213> artificial sequence <220><223> antisense oligonucleoside <400> 8 cctctccacg cgcagtacat

157514-序列表.doc157514 - Sequence Listing. doc

Claims (1)

201209163 七、申請專利範圍·· 1. 一種在活體外調節生物系統中BCL2結合部分3(BBC3)聚 核苷酸之功能及/或表現的方法,其包括: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BCL2結合部分3(BBC3)聚核苷 酸之天然反義轉錄物的反向補體具有至少50%的序列一 致性;由此調節該BCL2結合部分3(BBC3)聚核苷酸之功 能及/或表現。 Ο 2.如請求項1之調節生物系統中BCL2結合部分3(BBC3)聚 核苷酸之功能及/或表現的方法,其包括: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苷酸接 觸,其中該至少一種寡核苷酸與聚核苷酸之反向補體具 有至少50%的序列一致性,該聚核苷酸包括SEQ ID NO: 2之天然反義轉錄物核苷酸1至689内之5至30個連續核苷 酸;由此調節該BCL2結合部分3(BBC3)聚核苷酸之功能 及/或表現。 〇 3. —種在活體外調節患者細胞或組織中BCL2結合部分 3(BBC3)聚核苷酸之功能及/或表現的方法,其包括:使 , 該等細胞或組織與至少一種長度為5至30個核苷酸之反 義寡核苷酸接觸,其中該寡核苷酸與該BCL2結合部分 3(BBC3)聚核苷酸之反義寡核苷酸具有至少50%的序列一 致性;由此在活體外調節患者細胞或組織中該BCL2結合 部分3(BBC3)聚核苷酸之功能及/或表現。 4. 如請求項3之調節患者細胞或組織中BCL2結合部分 157514.doc 201209163 3(BBC3)聚核苷酸之功能及/或表現的方法,其包括: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 核苷酸接觸,其中該至少一種寡核苷酸與聚核苷酸之反 向補體具有至少50%的序列一致性,該聚核苷酸包括 SEQ ID NO: 2之天然反義轉錄物核苷酸1至689内之5至30 個連續核苷酸;由此調節該BCL2結合部分3(BBC3)聚核 苷酸之功能及/或表現。 5 _ 一種在活體外調節生物系統中BCL2結合部分3 (BBC3)聚 核苷酸之功能及/或表現的方法,其包括:使該系統與至 少一種乾向該BCL2結合部分3(BBC3)聚核苷酸之天然反 義寡核苷酸中區域之反義募核苷酸接觸;由此調節該 BCL2結合部分3(BBC3)聚核苷酸之功能及/或表現。 6. 如吻求項5之方法,其中該BCL2結合部分3(BBC3)在活 體外之功能及/或表現比對照組增加。 7. 如π求項5之方法,其中該至少一種反義寡核苷酸靶向 BCL2結合部分3(BBC3)聚核苷酸之天然反義序列。 8·如:求項5之方法’纟中該至少一種反義寡核苷酸靶向 核I序列,該核酸序列包括BCL2結合部分3(BBC3)聚核 苷酸之編碼及/或非編碼核酸序列。 9·如响求項5之方法,其中該至少一種反義寡核苷酸靶向 σ。4分3(BBC3)聚核苷酸之重疊及/或非重最 列。 且々' 1 0.如請求項5 $ f、土 ^ . _ 一〆夕 方法,其中该至少一種反義募核苷酸包括 5個k自以下之修飾:至少一個經修飾糖部分、至 157514.doc 201209163 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。 11.如請求項10之方法,其中該 選自以下之經修飾糖部分: 或多個修飾包括至少 經2'-〇-甲氧基乙基修飾 一個 之糖 部分、經2,·甲氧基修飾之糖部分、經2,_q_㈣修飾之糖 部分、二環糖部分及其組合。 12. 如請求項1〇之方法,其中該—或多個修飾包括至少一個 選自以下之經修飾核苦間鍵聯:硫代磷酸酯、2,_〇甲氧 基乙基(ΜΟΕ)、2’-氟、膦酸烷基酯、二硫代磷酸酯、硫 代膦酸烷基醋、胺基磷酸醋、胺基甲酸酿、碳酸醋、磷 酸三酯、胺基乙酸酯、羧甲基酯及其組合。 13. 如請求項10之方法,其中該一或多個修飾包括至少一個 選自以下之經修飾核苦酸:肽核酸(ρΝΑ)、鎖核酸 (LNA)、阿糖核酸(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括至少 一個闡述為SEQ ID NO: 3至8之寡核苷酸序列。 15. —種在活體外調節哺乳動物細胞或組織中BCL2結合部分 3(BBC3)基因之功能及/或表現的方法,其包括: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)募核苷酸接觸,該至少一種siRNA寡核苷酸 對BCL2結合部分3(BBC3)聚核苷酸之反義聚核苷酸具有 特異性,其中該至少一種siRNA寡核苷酸與該BCL2結合 部分3(BBC3)聚核苷酸之反義及/或正義核酸分子中至少 約5個連續核酸之互補序列具有至少5〇%的序列一致性; 157514.doc 201209163 及在活體外調節哺乳動物細胞或組織中之結合部分 3(BBC3)之功能及/或表現。 16. 如味求項15之方法,其中該寡核苷酸與具有至少約$個 連續核酸之序列具有至少8〇%的序列一致性,該序列與 該BCL2結合部分3(BBC3)聚核苦酸之反義及/或正義核酸 分子互補。 17. 種在,¾•體外6周_哺乳動物細胞或、组織中队[2結合部分 3(BBC3)之功施及/或表丨見的方法,丨包括:使該等細胞 或組織與至少一種長度為約5至30個核苷酸之反義寡核 苷馱接觸,δ亥反義寡核苷酸對8(:]^2結合部分3(BBC3)聚 核苷酸之正義及/或天然反義鏈之非編碼及/或編碼序列 具有特異性’其令該至少一種反義寡核苷酸與至少一個 闡述為SEQ ID NO: 1及2之核酸序列具有至少5〇%的序列 一致性,及在活體外調節哺乳動物細胞或組織中該bcL2 結合部分3(BBC3)之功能及/或表現。 18. —種反義券核苷酸之用途,其用以製造用於調節生物系 統中BCL2結合部分3(BBC3)聚核苷酸之功能及/或表現的 藥劑’其中該募核苷酸之長度為5至3 〇個核苷酸且與 BCL2結合部分3(BBC3)聚核苷酸之天然反義轉錄物之反 向補體具有至少50%的序列一致性。 19. 如睛求項18之用途’其中該寡核苷酸與包括SEq id Ν〇· 2之天然反義轉錄物核苷酸1至689内之5至30個連續核苦 酸之聚核苷酸之反向補體具有至少5〇%的序列一致性。 20. —種反義寡核苷酸之用途,其用以製造用於調節患者細 157514.doc 201209163 胞或組織中BCL2結合部分3(BBC3)聚核苷酸之功能及/或 表現的藥劑,其中該反義寡核苷酸之長度為5至30個核 苷酸且與該BCL2結合部分3(BBC3)聚核苷酸之反義寡核 苷酸具有至少50%的序列一致性。 21. 如請求項20之用途,其中該寡核苷酸與包括SEQ ID NO: 2之天然反義轉錄物核苷酸1至689内之5至30個連續核苷 酸之聚核苷酸之反向補體具有至少50%的序列一致性。 22. —種反義寡核苷酸之用途,其用以製造用於調節生物系 統中BCL2結合部分3(BBC3)聚核苷酸之功能及/或表現的 藥劑,其中該反義寡核苷酸靶向該BCL2結合部分 3(BBC3)聚核苷酸之天然反義寡核苷酸的區域。 23. 如請求項22之用途,其中該BCL2結合部分3(BBC3)在活 體内之功能及/或表現比對照組增加。 24. 如請求項22之用途,其中該反義寡核苷酸靶向BCL2結合 部分3(BBC3)聚核苷酸之天然反義序列。 25. 如請求項22之用途,其中該反義寡核苷酸靶向包括BCL2 結合部分3(BBC3)聚核苷酸之編碼及/或非編碼核酸序列 之核酸序列。 26. 如請求項22之用途,其中該反義寡核苷酸靶向BCL2結合 部分3(BBC3)聚核苷酸之重疊及/或非重疊序列。 27. 如請求項22之用途,其中該反義寡核苷酸包括一或多個 選自以下之修飾:至少一個經修飾糖部分、至少一個經 修飾核苷間鍵聯、至少一個經修飾核苷酸及其組合。 28. 如請求項27之用途,其中該一或多個修飾包括至少一個 157514.doc 201209163 選自以下之經修飾糖部分:經2,-〇-甲氧基乙基修飾之糖 部分、經2,-甲ft基修飾之糖部分、經2,_〇_炫基修飾之糖 部分、二環糖部分及其組合。 29. 如請求項27之用途,其中該—或多個修飾包括至少一個 選自以下之經修飾核苦間鍵聯:硫代磷酸醋、2,_〇甲氧 基乙基(ΜΟΕ)、2,-說、膦酸燒基酯、二硫代磷酸醋、硫 代膦酸烷基醋、胺基磷酸輯、胺基甲酸醋、碳酸醋、磷 酸三酿、胺基乙酸醋、羧甲基酯及其組合。 30. 如請求項27之用途,其中該一或多個修飾包括至少一個 選自以下之經修飾核苷酸:肽核酸(ρνα)、鎖核酸 (LNA)、阿糖核酸(FANA)、其類似物、衍生物及組合。 31. 如請求項18之用途,其中該寡核苷酸包括至少一個闡述 為SEQ ID NO: 3至8之寡核苷酸序列。 32. —種短干擾RNA (siRNA)募核苦酸之用途,其用以製造 用於調節哺乳動物細胞或組織中BCL2結合部分3(BBC3) 基因之功能及/或表現的藥劑’其中該siRNA募核苦酸之 長度為5至30個核音酸且對BCL2結合部分3(BBC3)聚核 苷酸之反義聚核苷酸具有特異性,其中該siRNA募核苦 酸與該BCL2結合部分3(BBC3)聚核苷酸之反義及/或正義 核酸分子中之至少約5個連續核酸之互補序列具有至少 5 0 %的序列一致性。 33·如清求項32之用途’其中該养核苦酸與具有至少約$個 連續核酸之序列具有至少80%的序列一致性,該序列與 該BCL2結合部分3(BBC3)聚核苷酸之反義及/或正義核妒 157514.doc 201209163 分子互補。 34. —種反義寡核苷酸之用途,其用以製造用於調節哺乳動 物細胞或組織中BCL2結合部分3(BBC3)之功能及/或表現 的藥劑’其中該反義募核苷酸之長度為約5至30個核苷 酸且對BCL2結合部分3(BBC3)聚核苷酸之正義及/或天然 反義鏈之非編碼及/或編碼序列具有特異性,其中該反義 寡核苷酸與至少一個闡述為SEQ ID N〇: 之核酸序列 具有至少50%的序列一致性。 〇 35. —種包括至少—個修飾之合成的經修飾寡核苷酸,其中 該至少一個修飾選自: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 口,其中β亥券核苷酸係在活體内或活體外與bcl2結合部 分3(BBC3)基因雜交且與正常對照組相比,可調節該 BCL2結合部分3(BBC3)基因之功能及/或表現之反義化合 物’且其中該募核苷酸與至少約5個連續核酸之序列具 ❹ 有至少50°/°的序列一致性,該序列與該BCL2結合部分 3(BBC3)聚核苷酸之反義及/或正義核酸分子及其等位基 因、同系物、同種型、變體、衍生物、突變體、片段或 、組合互補。 36. 如請求項35之募核苷酸,其中該募核苷酸之長度為5至 30個核苦酸,且與該BBC3基因之天然反義轉錄物内5至 30個連續核苷酸之反向補體具有至少5〇%的序列一致 性。 37. 如請求項36之募核苷酸,其中該至少一個修飾包括選自 157514.doc 201209163 由以下組成之群之核苷酸間鍵聯:硫代磷酸酯、膦酸院 基酯、二硫代磷酸酯、硫代膦酸烷基酯、胺基磷酸酯、 胺基曱酸酯、碳酸酯、磷酸三酯、胺基乙酸酯、羧甲基 酯及其組合。 3 8.如請求項36之募核苷酸,其中該寡核苷酸包括至少一個 硫代磷酸酯核苷酸間鍵聯。 39. 如請求項36之募核苷酸,其中該募核苷酸包括硫代磷酸 酯核苷酸間鍵聯之主鏈。 40. 如請求項36之募核苷酸,其中該募核苷酸包括至少一個 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3 6之募核苷酸,其中該寡核苷酸包括複數個修 飾’其中該等修飾包括選自以下之經修飾核苷酸:硫代 磷酸酯、膦酸烧基酯、二硫代磷酸酯、硫代膦酸烷基 酯、胺基磷酸酯、胺基甲酸酯、碳酸酯 '磷酸三酯、胺 基乙酸醋、缓甲基S旨及其乡且合。 42. 如請求項36之募核苷酸,其中該寡核苷酸包括複數個修 飾,其中該等修飾包括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之募核苷酸,其中該寡核苷酸包括至少一個 選自以下之經修飾糖部分:經2,_〇_甲氧基乙基修飾之糖 部分、經2'-曱氧基修飾之糖部分、經2,_〇_烷基修飾之糖 部分、二環糖部分及其組合。 44. 如請求項36之寡核苦酸,其中該寡核苦酸包括複數個修 157514.doc 201209163 飾’其中該等修飾包括選自以下之經修飾糖部分:經2,_ 〇-曱氧基乙基修飾之糖部分、經2,·曱氧基修飾之糖部 分、經2,-〇-烷基修飾之糖部分、二環糖部分及其組合。 45. 如請求項36之寡核苷酸,其中該寡核苷酸之長度為至少 約5至30個核苷酸且與BCL2結合部分3(BBC3)聚核苷酸 之反義及/或正義鏈雜交,其中該寡核苷酸與該BCL2結 合部分3(BBC3)聚核苷酸之反義及/或正義編碼及/或非編 碼核酸序列中至少約5個連續核酸之互補序列具有至少 約60%的序列一致性。 46. 如請求項36之寡核苷酸,其中該寡核苷酸與該8(:1^結合 部分3(BBC3)聚核苷酸之反義及/或正義編碼及/或非編碼 核酸序列之至少約5個連續核酸之互補序列具有至少約 8 0 %的序列一致性。 47. 如請求項36之寡核苷酸,其中該募核苷酸在活體内或活 體外與至少一種BCL2結合部分3(BBC3)聚核苷酸雜交, 並相對於正常對照組,可調節該BCL2結合部分3(BBC3) 聚核苷酸之表現及/或功能。 48. 如請求項36之寡核苷酸,其中該寡核苷酸包括閉述為 SEQ ID NO: 3至8之序列。 49. 一種醫藥組合物,其包括一或多種如請求項35之對一或 多種BCL2結合部分3(BBC3)聚核苷酸具有特異性之寡核 苷酸及醫藥上可接受之賦形劑。 50. 如請求項49之組合物,其中該等寡核苷酸與闡述為seq ID NO: 3至8之核苷酸序列中之任一者比對,具有至少約 157514.doc 201209163 40%的序列一致性。 51. 如請求項49之組合物,其中該等寡核苷酸包括闡述為 SEQ ID NO: 3至8之核苷酸序列。 52. 如請求項51之組合物,其中該等闡述為SEQ ID NO: 3至8 之养核苷酸包括一或多個修倚或取代。 53·如請求項52之組合物,其中該一或多個修飾選自··硫代 磷酸酯、膦酸甲酯、肽核酸、鎖核酸(LNA)分子及其組 合。 54. —種反義募核苷酸之用途,其用以製造用於預防或治療❹ 與至 種BCL2結合部分3(BBC3)聚核普酸及/或其至少 一種編碼產物有關之疾病的藥劑,其中該反義寡核苷酸 與5亥至少一種BCL2結合部分3(BBC3)聚核苷酸之天然反 義序列結合並調節該至少一種BCL2結合部分3⑺BC3)聚 核苷酸的表現。 55. 如请求項54之用途,其中與該至少一種8〇^2結合部分 3(BBC3)聚核苷酸有關之疾病選自:與BBC3之異常功能 及/或表現有關之疾病或病症、癌症、增殖性疾病或病 症' 與細胞增殖有關或特徵在於細胞增殖之疾病或病 狀、與BBC3之突變或異常表現或功能有關之疾病或病 症、神經疾病或病症、發炎、以發炎關節成為病變之— · 部分的病狀(例如,骨關節炎、關節炎、牛皮癬性關節 . 炎、幼年型關節炎、萊特爾氏症候群 syndrome)、與潰瘍性結腸炎有關之關節炎、惠伯爾病 (Wlnpple’s disease)、與肉芽腫回腸結腸炎有關之關節 157514.doc -10- 201209163 56. Ο 57. 58. 59. 〇 60. 61. 炎貝切特氏病(Behcet's disease)、全身性紅斑狼瘡、 乾燥症候群(Sjogrenis syndr〇me)及混合型結締组織病 等)、與粒線體細胞凋亡途徑損傷有關之疾病或病症、自 體免疫性疾病或病症,及與神經元細胞死亡有關之疾病 或病症、衰老或特徵在於不期望細胞損失之其他病狀。 一種活體外誘導生物系統中之細胞凋亡的方法,其包括 向该系統投與募核苷酸,該募核苷酸之長度為5至30個 核苷酸且與BBC3聚核苷酸之天然反義轉錄物之反向補體 内的5至30個連續核苷酸至少50。/。一致。 如咕求項56之方法,其中該天然反義轉錄物具有犯〇 IDNO·· 2 〇 如β求項56之方法,其中該生物系統係患者細胞或組 織。 種活體外誘導生物系統中之細胞凋亡的方法,其包括 向该系統投與長度為約5至30個核苷酸之募核苷酸,其 中邊寡核苦酸藉由靶向靶基因之天然反義轉錄物來上調 δ亥基因’且其中該募核苷酸與該經上調基因之天然反義 轉錄物的反向補體至少5〇%一致。 一種募核苷酸之用途,其用以製造用於誘導生物系統中 之、、’田胞〉周亡之藥劑’其中該募核苷酸之長度為5至3 0個 核苦酸且與BBC3聚核苷酸之天然反義轉錄物之反向補體 内的5至3〇個連續核苷酸至少5〇%一致。 如β求項60之用途’其中該天然反義轉錄物具有SEq IDNO: 2。 157514.doc -11 - 201209163 Μ.如請求項6G之用途,其中該生物系統係患者細胞或組 織。 63· -種寡核苦酸之用途,其用以製造用於誘導生物系統中 之、、田胞/周亡之帛劑,其中該募核普酸之長度為約5至W 個核苷酸且藉由靶向靶基因之天然反義轉錄物來上調該 基因且其中該养核苷酸與該經上調基因之天然反義轉 錄物之反向補體至少5〇% —致。 64. —種鑑別及選擇至少一種對作為選定靶聚核苷酸之 基因之天然反義轉錄物具有選擇性之寡核苷酸供於活體 内投與之方法,該方法包括:鑑別至少一種包括至少5 個連續核苷酸的寡核苷酸,該等至少5個連續核苷酸與 "亥選疋乾聚核皆酸之反義聚核苦酸至少部分互補;在嚴 格雜交條件下量測反義募核苷酸與該靶聚核苷酸或該選 疋乾聚核苷酸之反義聚核苷酸之雜合體的熱熔點;及基 於所獲得資訊選擇至少一種寡核苷酸用於活體内投與。 157514.doc 12-201209163 VII. Patent Application Range 1. A method for regulating the function and/or performance of a BCL2 binding portion 3 (BBC3) polynucleotide in a biological system in vitro, comprising: making the system with at least one length of 5 Up to 30 nucleotide antisense oligonucleotide contacts, wherein the at least one oligonucleotide has at least 50% of the reverse complement of the native antisense transcript of the BCL2 binding portion 3 (BBC3) polynucleotide Sequence identity; thereby modulating the function and/or performance of the BCL2 binding moiety 3 (BBC3) polynucleotide. Ο 2. A method of modulating the function and/or expression of a BCL2 binding portion 3 (BBC3) polynucleotide in a biological system according to claim 1, comprising: bringing the biological system to at least one nucleoside of 5 to 30 in length An acid antisense oligonucleotide contact, wherein the at least one oligonucleotide has at least 50% sequence identity to the reverse complement of the polynucleotide, the polynucleotide comprising the natural inverse of SEQ ID NO: 5 to 30 contiguous nucleotides within the transcript nucleotides 1 to 689; thereby modulating the function and/or expression of the BCL2 binding portion 3 (BBC3) polynucleotide. 〇3. A method of modulating the function and/or expression of a BCL2 binding portion 3 (BBC3) polynucleotide in a patient cell or tissue in vitro, comprising: causing the cells or tissues to have at least one length of 5 Contacting to a 30 nucleotide antisense oligonucleotide having at least 50% sequence identity to the antisense oligonucleotide of the BCL2 binding portion 3 (BBC3) polynucleotide; The function and/or expression of the BCL2 binding portion 3 (BBC3) polynucleotide in the patient's cells or tissues is thereby regulated in vitro. 4. A method of modifying the function and/or expression of a BCL2 binding portion 157514.doc 201209163 3 (BBC3) polynucleotide in a patient cell or tissue according to claim 3, comprising: bringing the biological system to at least one length of 5 Contacting to a 30 nucleotide antisense oligonucleotide, wherein the at least one oligonucleotide has at least 50% sequence identity to the reverse complement of the polynucleotide, the polynucleotide comprising SEQ ID NO : 5 to 30 contiguous nucleotides within nucleotides 1 to 689 of the natural antisense transcript; thereby modulating the function and/or expression of the BCL2 binding portion 3 (BBC3) polynucleotide. 5 _ A method for modulating the function and/or expression of a BCL2 binding portion 3 (BBC3) polynucleotide in a biological system in vitro, comprising: polymerizing the system with at least one stem to the BCL2 binding portion 3 (BBC3) The antisense raised nucleotides of the region of the nucleotide's natural antisense oligonucleotide are contacted; thereby modulating the function and/or expression of the BCL2 binding portion 3 (BBC3) polynucleotide. 6. The method of claim 5, wherein the function and/or performance of the BCL2 binding moiety 3 (BBC3) is increased in vitro compared to the control group. 7. The method of claim 5, wherein the at least one antisense oligonucleotide targets a natural antisense sequence of a BCL2 binding portion 3 (BBC3) polynucleotide. 8. The method of claim 5, wherein the at least one antisense oligonucleotide targets a nuclear I sequence, the nucleic acid sequence comprising a BCL2 binding portion 3 (BBC3) polynucleotide encoding and/or non-coding nucleic acid sequence. 9. The method of claim 5, wherein the at least one antisense oligonucleotide targets σ. The overlap and/or non-weight of the 4 points 3 (BBC3) polynucleotides. And 々 '1 0. such as claim 5 $ f, 土 ^ . _ 〆 方法 method, wherein the at least one antisense nucleotide comprises 5 k from the following modifications: at least one modified sugar moiety, to 157514 .doc 201209163 One less modified internucleoside linkage, at least one modified nucleotide, and combinations thereof. 11. The method of claim 10, wherein the modified sugar moiety is selected from the group consisting of: or a plurality of modifications comprising modifying at least one sugar moiety, 2, methoxyl via 2'-fluorene-methoxyethyl Modified sugar moiety, sugar moiety modified by 2,_q_(tetra), bicyclic sugar moiety, and combinations thereof. 12. The method of claim 1 , wherein the one or more modifications comprise at least one modified internuclear linkage selected from the group consisting of: phosphorothioate, 2,-methoxyethyl (ΜΟΕ), 2'-fluoro, alkyl phosphonate, dithiophosphate, thiophosphonic acid alkyl vinegar, amino citrate, amino carboxylic acid brewing, carbonated vinegar, phosphate triester, amino acetate, carboxymethyl Base esters and combinations thereof. 13. The method of claim 10, wherein the one or more modifications comprise at least one modified nucleotide acid selected from the group consisting of peptide nucleic acid (ρΝΑ), locked nucleic acid (LNA), arabinic acid (FANA), and the like Things, derivatives and combinations. 14. The method of claim 1, wherein the at least one oligonucleotide comprises at least one oligonucleotide sequence set forth as SEQ ID NOs: 3 to 8. 15. A method of modulating the function and/or expression of a BCL2 binding portion 3 (BBC3) gene in a mammalian cell or tissue in vitro, comprising: bringing the cells or tissues to at least one length of 5 to 3 a short interfering RNA (siRNA) nucleotide contact, the at least one siRNA oligonucleotide specific for an antisense polynucleotide of a BCL2 binding portion 3 (BBC3) polynucleotide, wherein the at least one An siRNA oligonucleotide having at least 5% sequence identity to an antisense of the BCL2 binding portion 3 (BBC3) polynucleotide and/or a complement of at least about 5 contiguous nucleic acids in a sense nucleic acid molecule; 157514. Doc 201209163 and in vitro regulation of the function and/or performance of binding moiety 3 (BBC3) in mammalian cells or tissues. 16. The method of claim 15, wherein the oligonucleotide has at least 8% sequence identity to a sequence having at least about $ contiguous nucleic acid, the sequence and the BCL2 binding portion 3 (BBC3) polynuclear Acid antisense and/or sense nucleic acid molecules are complementary. 17. Seeding, 3⁄4• in vitro 6 weeks_mammalian cells or tissue squadrons [2 binding part 3 (BBC3) functions and/or methods of seeing, including: making such cells or tissues with at least An antisense oligodeoxynucleotide conjugate having a length of about 5 to 30 nucleotides, and a sigma antisense oligonucleotide pair 8 (:] 2 binding moiety 3 (BBC3) polynucleotide for justice and/or The non-coding and/or coding sequence of the natural antisense strand is specific in that it aligns the at least one antisense oligonucleotide with at least one sequence of at least 5 % of the nucleic acid sequences set forth as SEQ ID NOS: 1 and 2 And the function and/or expression of the bcL2 binding moiety 3 (BBC3) in mammalian cells or tissues in vitro. 18. Use of an antisense nucleus for the manufacture of a biological system for regulation An agent for the function and/or expression of a BCL2 binding moiety 3 (BBC3) polynucleotide wherein the nucleotide is 5 to 3 nucleotides in length and binds to the BCL2 binding moiety 3 (BBC3) The reverse complement of the acid natural antisense transcript has at least 50% sequence identity. 19. The use of the term 18 The reverse complement of a polynucleotide comprising 5 to 30 contiguous nucleotides of nucleotides 1 to 689 of the natural antisense transcripts of SEq id 具有 2 has a sequence identity of at least 5%. An use of an antisense oligonucleotide for the manufacture of a medicament for modulating the function and/or expression of a BCL2 binding moiety 3 (BBC3) polynucleotide in a cell or tissue of a patient 157514.doc 201209163, wherein The antisense oligonucleotide is 5 to 30 nucleotides in length and has at least 50% sequence identity to the antisense oligonucleotide of the BCL2 binding portion 3 (BBC3) polynucleotide. The use of claim 20, wherein the oligonucleotide is inverted complement to a polynucleotide comprising 5 to 30 contiguous nucleotides within nucleotides 1 to 689 of the natural antisense transcript of SEQ ID NO: 2. Having at least 50% sequence identity. 22. Use of an antisense oligonucleotide for the manufacture of a function and/or expression for modulating a BCL2 binding portion 3 (BBC3) polynucleotide in a biological system An agent, wherein the antisense oligonucleotide targets a region of the natural antisense oligonucleotide of the BCL2 binding portion 3 (BBC3) polynucleotide. The use of claim 22, wherein the function and/or performance of the BCL2 binding member 3 (BBC3) is increased in vivo compared to the control group. 24. The use of claim 22, wherein the antisense oligonucleotide targets BCL2 binding The natural antisense sequence of a portion 3 (BBC3) polynucleotide. 25. The use of claim 22, wherein the antisense oligonucleotide targets a coding comprising a BCL2 binding portion 3 (BBC3) polynucleotide and/ Or a nucleic acid sequence that does not encode a nucleic acid sequence. 26. The use of claim 22, wherein the antisense oligonucleotide targets overlapping and/or non-overlapping sequences of a BCL2 binding portion 3 (BBC3) polynucleotide. 27. 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 157514.doc 201209163 modified sugar moiety selected from the group consisting of 2,-〇-methoxyethyl modified sugar moiety, via 2 , a ft-modified sugar moiety, a sugar moiety modified by 2, _ 〇 炫, a bicyclic sugar moiety, and combinations thereof. 29. The use of claim 27, wherein the one or more modifications comprise at least one modified internuclear linkage selected from the group consisting of: thiophosphoric acid vinegar, 2, 〇 methoxyethyl (ΜΟΕ), 2 ,- said, phosphonic acid ester, dithiophosphoric acid vinegar, thiophosphonic acid alkyl vinegar, amino phosphoric acid, amino carboxylic acid vinegar, carbonated vinegar, triglyceride, aminoacetic acid vinegar, carboxymethyl ester And their combinations. 30. The use of claim 27, wherein the one or more modifications comprise at least one modified nucleotide selected from the group consisting of a peptide nucleic acid (ρνα), a locked nucleic acid (LNA), an arabinic acid (FANA), and the like Things, derivatives and combinations. 31. The use of claim 18, wherein the oligonucleotide comprises at least one oligonucleotide sequence set forth as SEQ ID NOs: 3 to 8. 32. Use of a short interfering RNA (siRNA) to recruit a bitter acid for the manufacture of a medicament for modulating the function and/or expression of a BCL2 binding portion 3 (BBC3) gene in a mammalian cell or tissue The nucleus acid is 5 to 30 nucleotide acids in length and specific for the antisense polynucleotide of the BCL2 binding portion 3 (BBC3) polynucleotide, wherein the siRNA raises the binding moiety of the bitter acid to the BCL2 The antisense of the 3 (BBC3) polynucleotide and/or the complement of at least about 5 contiguous nucleic acids in the sense nucleic acid molecule has a sequence identity of at least 50%. 33. The use of claim 32, wherein the nutrient acid has at least 80% sequence identity to a sequence having at least about $ contiguous nucleic acid, the sequence and the BCL2 binding portion 3 (BBC3) polynucleotide Antisense and/or Justice Nuclear 157514.doc 201209163 Molecular complementarity. 34. Use of an antisense oligonucleotide for the manufacture of an agent for modulating the function and/or expression of BCL2 binding portion 3 (BBC3) in a mammalian cell or tissue, wherein the antisense nucleotide a length of about 5 to 30 nucleotides and specific for a non-coding and/or coding sequence of a sense and/or natural antisense strand of a BCL2 binding portion 3 (BBC3) polynucleotide, wherein the antisense oligo The nucleotide has at least 50% sequence identity to at least one of the nucleic acid sequences set forth as SEQ ID N:. 35. A modified oligonucleotide comprising at least one modified synthesis, wherein the at least one modification 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 its group, wherein the β-coupon nucleotide line hybridizes with the bcl2 binding part 3 (BBC3) gene in vivo or in vitro and can regulate the BCL2 binding part 3 (BBC3) compared with the normal control group. An antisense compound of the function and/or expression of a gene and wherein the nucleotide of the nucleotide has at least 50°/° sequence identity with a sequence of at least about 5 contiguous nucleic acids, the sequence and the BCL2 binding moiety 3 ( BBC3) Antisense and/or sense nucleic acid molecules of a polynucleotide and their alleles, homologs, isoforms, variants, derivatives, mutants, fragments or combinations are complementary. 36. The nucleotide of claim 35, wherein the nucleotide is 5 to 30 nucleotides in length and 5 to 30 contiguous nucleotides in the natural antisense transcript of the BBC3 gene. The reverse complement has a sequence identity of at least 5%. 37. The nucleotide of claim 36, wherein the at least one modification comprises an internucleotide linkage selected from the group consisting of 157514.doc 201209163: phosphorothioate, phosphonic acid ester, disulfide Phosphate, alkyl thiophosphonate, amino phosphate, amino phthalate, carbonate, phosphate, amino acetate, carboxymethyl ester, and combinations thereof. 3. The nucleotide of claim 36, wherein the oligonucleotide comprises at least one phosphorothioate internucleotide linkage. 39. The nucleotide of claim 36, wherein the nucleotide comprises a backbone of a phosphorothioate internucleotide linkage. 40. The nucleotide of claim 36, wherein the nucleotide comprises at least one modified nucleotide selected from the group consisting of: a peptide nucleic acid, a locked nucleic acid (LNA), an analog thereof, and a derivative. Things and combinations. 41. The nucleotide of claim 3, wherein the oligonucleotide comprises a plurality of modifications wherein the modifications comprise modified nucleotides selected from the group consisting of phosphorothioates, phosphonic acid esters, Dithiophosphates, alkyl thiophosphonates, amino phosphates, urethanes, carbonates, phosphotriesters, aminoacetic acid vinegars, and slow methyl groups are intended to be combined. 42. The nucleotide 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 nucleotide of claim 36, wherein the oligonucleotide comprises at least one modified sugar moiety selected from the group consisting of a 2,_〇-methoxyethyl modified sugar moiety, via 2'- a methoxy-modified sugar moiety, a 2,_〇-alkyl-modified sugar moiety, a bicyclic sugar moiety, and combinations thereof. 44. The oligonucleotide according to claim 36, wherein the oligonucleotide comprises a plurality of 157514.doc 201209163 ornaments wherein the modifications comprise a modified sugar moiety selected from the group consisting of 2, _ 〇-曱 oxygen a thioethyl-modified sugar moiety, a 2, decyloxy-modified sugar moiety, a 2,-fluorene-alkyl modified sugar moiety, a bicyclic sugar moiety, and combinations thereof. 45. The oligonucleotide of claim 36, wherein the oligonucleotide is at least about 5 to 30 nucleotides in length and is antisense and/or sensed with a BCL2 binding portion 3 (BBC3) polynucleotide. Chain hybridization wherein the oligonucleotide has at least about an antisense to the BCL2 binding portion 3 (BBC3) polynucleotide and/or a complementary sequence of at least about 5 contiguous nucleic acids in a sense encoding and/or non-coding nucleic acid sequence. 60% sequence identity. 46. The oligonucleotide of claim 36, wherein the oligonucleotide is antisense and/or sense encoding and/or non-coding nucleic acid sequence of the 8 (:1^ binding moiety 3 (BBC3) polynucleotide The complement of at least about 5 contiguous nucleic acids has a sequence identity of at least about 80%. 47. The oligonucleotide of claim 36, wherein the nucleotide is conjugated to at least one BCL2 in vivo or ex vivo A portion 3 (BBC3) polynucleotide hybridizes and modulates the expression and/or function of the BCL2 binding portion 3 (BBC3) polynucleotide relative to a normal control group. 48. The oligonucleotide of claim 36 Wherein the oligonucleotide comprises the sequence SEQ ID NOs: 3 to 8. 49. A pharmaceutical composition comprising one or more of one or more BCL2 binding moiety 3 (BBC3) as claimed in claim 35 Nucleotide-specific oligonucleotides and pharmaceutically acceptable excipients. 50. The composition of claim 49, wherein the oligonucleotides are nucleated as seq ID NO: 3 to 8 Alignment of any of the nucleotide sequences, having a sequence identity of at least about 157514.doc 201209163 40%. 51. The oligonucleotides include the nucleotide sequences set forth as SEQ ID NOS: 3 to 8. 52. The composition of claim 51, wherein the exemplified are SEQ ID NOs: 3 to 8 The nucleotide includes 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 phosphorothioate, methyl phosphonate, peptide nucleic acid, and locked nucleic acid ( LNA) Molecules and combinations thereof 54. Use of an antisense nucleotide to produce or prevent at least one of BCL2 binding moiety 3 (BBC3) polynucleotide and/or at least one thereof An agent encoding a product-related disease, wherein the antisense oligonucleotide binds to a natural antisense sequence of at least one BCL2 binding portion 3 (BBC3) polynucleotide and modulates the at least one BCL2 binding portion 3(7)BC3) polynuclei The performance of glycosides. 55. The use of claim 54, wherein the disease associated with the at least one 8 〇 2 binding portion 3 (BBC3) polynucleotide is selected from the group consisting of: a disease or condition associated with abnormal function and/or performance of BBC3, cancer , a proliferative disease or condition 'a disease or condition characterized by cell proliferation or characterized by cell proliferation, a disease or condition associated with a mutation or abnormal expression or function of BBC3, a neurological disease or condition, inflammation, an inflammation of the joint becoming a disease — · Part of the condition (for example, osteoarthritis, arthritis, psoriasis joints, inflammation, juvenile arthritis, Lyttle syndrome syndrome), arthritis associated with ulcerative colitis, and Whirlple's disease (Wlnpple's Disease), joints associated with granulomatous ileitis 157514.doc -10- 201209163 56. Ο 57. 58. 59. 〇60. 61. Behcet's disease, systemic lupus erythematosus, dry Symptoms (Sjogrenis syndr〇me) and mixed connective tissue diseases, etc., diseases or conditions associated with mitochondrial apoptosis pathway damage, autoimmune diseases or conditions, and with God A disease or condition associated with death of a cell, aging, or other condition characterized by undesired cell loss. A method for inducing apoptosis in a biological system in vitro, comprising administering to the system a nucleotide that is 5 to 30 nucleotides in length and is naturally associated with a BBC3 polynucleotide At least 50 of 5 to 30 contiguous nucleotides in the reverse complement of the antisense transcript. /. Consistent. The method of claim 56, wherein the natural antisense transcript has a method of 〇 IDNO·· 2 〇, such as β, 56, wherein the biological system is a patient cell or tissue. A method for inducing apoptosis in a biological system in vitro, comprising administering to the system a nucleotide of about 5 to 30 nucleotides in length, wherein the oligonucleotide is targeted by a target gene The natural antisense transcript up-regulates the delta gene' and wherein the nucleotide is at least 5% identical to the reverse complement of the native antisense transcript of the up-regulated gene. A use of a nucleotide for the manufacture of a medicament for inducing a 'cell' in a biological system, wherein the nucleotide is 5 to 30 nucleotides in length and BBC3 At least 5% of the 5 to 3 contiguous nucleotides in the reverse complement of the natural antisense transcript of the polynucleotide are at least 5%. For example, the use of beta 60 wherein the natural antisense transcript has SEq IDNO: 2. 157514.doc -11 - 201209163 Μ. The use of claim 6G, wherein the biological system is a patient cell or tissue. 63. The use of an oligo-nucleic acid for the manufacture of a sputum for inducing a cytoplasmic/peripheral death in a biological system, wherein the nucleocapnic acid is about 5 to W nucleotides in length And upregulating the gene by targeting a natural antisense transcript of the target gene and wherein the nucleotide is at least 5% identical to the reverse complement of the natural antisense transcript of the upregulated gene. 64. A method of identifying and selecting at least one oligonucleotide that is selective for a natural antisense transcript of a gene of a selected target polynucleotide for in vivo administration, the method comprising: identifying at least one of Oligonucleotides of at least 5 contiguous nucleotides, at least 5 contiguous nucleotides at least partially complementary to an antisense poly(picoic acid) of "Hypergic acid; and under stringent hybridization conditions Measuring the thermal melting point of the antisense raised nucleotide and the hybrid of the target polynucleotide or the antisense polynucleotide of the selected dry polynucleotide; and selecting at least one oligonucleotide based on the obtained information Invested in the body. 157514.doc 12-
TW100124653A 2010-07-12 2011-07-12 Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3 TW201209163A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US36353510P 2010-07-12 2010-07-12

Publications (1)

Publication Number Publication Date
TW201209163A true TW201209163A (en) 2012-03-01

Family

ID=45470031

Family Applications (1)

Application Number Title Priority Date Filing Date
TW100124653A TW201209163A (en) 2010-07-12 2011-07-12 Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3

Country Status (2)

Country Link
TW (1) TW201209163A (en)
WO (1) WO2012009347A2 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2638163T3 (en) 2010-11-12 2017-07-24 Massachusetts Gen Hospital POLYCOMB-ASSOCIATED NON-CODING RNAs
US9920317B2 (en) 2010-11-12 2018-03-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
DK2850186T3 (en) 2012-05-16 2019-04-08 Translate Bio Ma Inc COMPOSITIONS AND PROCEDURES FOR MODULATING SMN GENFAMILY EXPRESSION
JP2015518710A (en) 2012-05-16 2015-07-06 ラナ セラピューティクス インコーポレイテッド Compositions and methods for regulating hemoglobin gene family expression
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
KR20160074368A (en) 2012-05-16 2016-06-28 라나 테라퓨틱스, 인크. Compositions and methods for modulating utrn expression
WO2013173608A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating mecp2 expression
WO2013173598A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating atp2a2 expression
WO2016070060A1 (en) 2014-10-30 2016-05-06 The General Hospital Corporation Methods for modulating atrx-dependent gene repression
US10195213B2 (en) 2015-03-13 2019-02-05 Unity Biotechnology, Inc. Chemical entities that kill senescent cells for use in treating age-related disease
EP3271460A4 (en) 2015-03-17 2019-03-13 The General Hospital Corporation The rna interactome of polycomb repressive complex 1 (prc1)
CN110290794A (en) 2016-11-01 2019-09-27 纽约州州立大学研究基金会 The microRNA and its purposes in cancer treatment of 5- halo uracil modification

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7202024B2 (en) * 2001-09-17 2007-04-10 Immunogen Inc. bbc3 Gene promoter and methods for identifying modulators of apoptosis and bbc3 gene expression using a bbc3 gene promoter
EP1542016A1 (en) * 2003-12-11 2005-06-15 Institut Gustave Roussy PUMA, phosphorylated p53 and p38 map kinase as markers for HIV-1-induced apoptosis in circulating T cells of infected patient
CA2563074C (en) * 2004-04-09 2014-05-20 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
US20060292695A1 (en) * 2005-06-22 2006-12-28 Roslin Institute Methods and kits for drug screening and toxicity testing using promoter-reporter cells derived from embryonic stem cells

Also Published As

Publication number Publication date
WO2012009347A3 (en) 2012-04-26
WO2012009347A2 (en) 2012-01-19

Similar Documents

Publication Publication Date Title
JP6049623B2 (en) Treatment of IDUA-related diseases by inhibition of natural antisense transcripts to α-L-iduronidase (IDUA)
JP5993744B2 (en) Treatment of nuclear respiratory factor 1 related diseases by inhibition of natural antisense transcripts against nuclear respiratory factor 1 (NRF1)
JP6188686B2 (en) Treatment of FXN-related diseases by inhibition of natural antisense transcripts to frataxin (FXN)
JP6027893B2 (en) Treatment of sex hormone binding globulin (SHBG) related diseases by inhibition of natural antisense transcripts against sex hormone binding globulin (SHBG)
JP6025567B2 (en) Treatment of MBTPS1-related diseases by inhibition of the natural antisense transcript against the membrane-bound transcription factor peptidase, site 1 (MBTPS1)
JP5976548B2 (en) Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcripts against PYCR1
JP6073795B2 (en) Treatment of IFRD1-related diseases by inhibition of natural antisense transcripts to interferon-related developmental regulator 1 (IFRD1)
KR101805213B1 (en) Treatment of 'c terminus of hsp70-interacting protein' (chip) related diseases by inhibition of natural antisense transcript to chip
JP5886757B2 (en) Treatment of interferon regulatory factor 8 (IRF8) related diseases by inhibition of natural antisense transcripts against interferon regulatory factor 8 (IRF8)
JP6031356B2 (en) Treatment of uncoupling protein 2 (UCP2) -related diseases by inhibition of natural antisense transcripts against UCP2.
JP5986998B2 (en) Treatment of NEU4-related diseases by inhibition of natural antisense transcripts to sialidase 4 (NEU4)
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)
JP5981850B2 (en) Treatment of RNase H1-related diseases by inhibition of natural antisense transcripts against RNase H1
JP5964232B2 (en) Treatment of IQGAP-related diseases by inhibition of natural antisense transcripts against 'IQ motif-containing GTPase-activating protein' (IQGAP)
TWI573871B (en) Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
KR101886452B1 (en) Treatment of discs large homolog(dlg) related diseases by inhibition of natural antisense transcript to dlg
JP5973419B2 (en) Treatment of CSF3-related diseases by inhibition of natural antisense transcripts against colony stimulating factor 3 (CSF3)
JP5982362B2 (en) Treatment of PAR4-related diseases by inhibition of natural antisense transcripts to PAR4
JP2016185148A (en) Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
TW201209163A (en) Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3
JP5917497B2 (en) Treatment of MSRA-related diseases by inhibition of natural antisense transcripts against methionine sulfoxide reductase A (MSRA)
TW201210611A (en) Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT
TW201201819A (en) Treatment of BCL2-like 11 (BCL2L11) related diseases by inhibition of natural antisense transcript to BCL2L11
TW201143782A (en) Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2