TW201143782A - Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2 - Google Patents

Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2 Download PDF

Info

Publication number
TW201143782A
TW201143782A TW100117637A TW100117637A TW201143782A TW 201143782 A TW201143782 A TW 201143782A TW 100117637 A TW100117637 A TW 100117637A TW 100117637 A TW100117637 A TW 100117637A TW 201143782 A TW201143782 A TW 201143782A
Authority
TW
Taiwan
Prior art keywords
lhx2
antisense
polynucleotide
sequence
oligonucleotide
Prior art date
Application number
TW100117637A
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 TW201143782A publication Critical patent/TW201143782A/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • C12N2310/11Antisense
    • C12N2310/113Antisense targeting other non-coding nucleic acids, e.g. antagomirs

Abstract

The present invention relates to antisense oligonucleotides that modulate the expression of and/or function of LIM homeobox 2 (LHX2), in particular, by targeting natural antisense polynucleotides of LIM homeobox 2 (LHX2). The invention also relates to the identification of these antisense oligonucleotides and their use in treating diseases and disorders associated with the expression of LHX2.

Description

201143782 六、發明說明: 【發明所屬之技術領域】 本發明實施例包括調節LHX2之表現及/或功能之寡核苷 酸及相關分子。 本申請案主張2010年5月19曰提出申請之美國臨時專利 申請案第61/346,284號之優先權,其全部内容以引用方式 併入本文中。 【先前技術】 O DNA-RNA及RNA-RNA雜交對於包含DNA複製、轉錄、 及轉譯在内之許多核酸功能態樣甚為重要。雜交對於檢測 特定核酸或改變其表現之各種技術亦至關重要。舉例而 言,反義核苷酸藉由與靶RNA雜交來破壞基因表現,由此 干擾RNA剪接、轉錄、轉譯、及複製。反義DNA具有額外 特性,亦即DNA-RNA雜合體可用作核糖核酸酶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,例如寡去氧核苷酸(ODN)之情形,或其可自内源 〇 基因表現為RNA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 性質及大意。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圍或含義。 在一實施例中,本發明提供抑制天然反義轉錄本之作用 156342.doc 201143782 之方法,其係藉由使用靶向天然反義轉錄本之任一區域的 反義寡核苷酸從而上調相應有義基因來達成。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 本,此視為屬於本發明範圍内。 一實施例提供在活體内或活體外調節患者細胞或組織中 LHX2聚核普酸之功能及/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, 其中該养核苷酸與聚核苷酸之反向互補序列具有至少5〇% 的序列一致性,該聚核苷酸包括SEQ m NO: 2中核苷酸1 至 2145、SEQ ID NO: 3 中核苷酸 1 至 6〇6、及 SEQ ID NO: 4 中核苷酸1至480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LHX2聚核苷酸之功能及/ 或表現。 在一實施例中,寡核苷酸靶向LHX2聚核苷酸之天然反 義序列(例如,SEQ ID NO: 2至4中所述之核苷酸)、及其任 變體、等位基因、同系物、突變體、衍生物、片段及互 補序列。反義寡核苷酸之實例係如SEq ID NO: 5至16所 7f> ° 另—實施例提供在活體内或活體外調節患者細胞或組織 中LHX2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’其中該募核苷酸與LHX2聚核苷酸反義轉錄本之反向 互補序列具有至少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LHX2聚核苷酸之功能及/或表 156342.doc 201143782 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LHX2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LHX2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LHX2聚核苷酸之功能及/或表現。 在一實施例中,組合物包括一或多種結合至有義及/或 反義LHX2聚核苷酸之反義寡核苷酸。 在一實施例中’寡核苷酸包括一或多個經修飾或取代之 核苦酸。 在一實施例中,寡核苷酸包括一或多個經修飾鍵。 在又一貫施例中,經修飾核苷酸包括含有以下之經修飾 驗基.硫代構酸酯、膦酸甲醋、肽核酸、2,曱基、氟_ 或碳、亞甲基或其他鎖核酸(LNA)分子。較佳地,經修飾 核音酸係鎖核酸分子,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寡 核苦酸投與患者。 在只施例中,以醫藥組合物形式投與募核苷酸。治療 方案包括至少一次向患者投與反義化合物;然而,此治療 可經改良以在一定時間内包含多個劑量。該治療可與一或 多個其他類型之療法組合。 在-實施例中’將寡核魏囊封於脂質體中或附接至載 劑分子(例如膽固醇、TAT肽)。 156342.doc 201143782 其他態樣闡述於下文中。 【實施方式】 下文參照闡釋用實例廡田水 頁例應用來闞述本發明之若干態樣。 理解’本文列舉各種具#心銘 ^ 具體細π、關似Μ吨供對本發 明之完全理解。“ m目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 -些作用可以不同順序發生及/或與其他動作或事件同時 發生n未必需要所有所闡釋動作或事件來實施 明方法。 本文所揭示之所有基因、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H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’此冑示内容意欲僅具有實例性,且不應解釋為限制 意義,除非上下文明確指明。因此,舉例而言,在某些關 於哺乳動物核酸及胺基酸序列之實施例中,本文所揭示基 因思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、及鳥。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 156342.doc 201143782 (an)」及「該(the)」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(inciudes)」、「具有 (having)」、「具有(has)」、「具有(with)」、或其變化形式 時,該等術語意欲以類似於術語「包括(c〇mprising)」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,量測系統之侷限性。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2〇%、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」意指乾向基因之當前已知 mRNA轉錄本、及可闡釋之任一其他轉錄本。 「反義券核♦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 156342.doc 201143782 含可用於治療、診斷、或其他方面之任何外來RNA或DNA 分子。該等分子包括(例如)反義RNA或DNA分子、干擾 RNA (RNAi)、微小 RNA、誘餌 RNA 分子、siRNA、酶 RNA、治療性編輯RNA及RNA激動劑與拮抗劑、反義寡聚 化合物、反義寡核苷酸、外部引導序列(EGS)寡核苷酸、 交替剪接、引物、探針、及舆靶核酸之至少一部分雜交之 其他寡聚化合物。因此,該等化合物可呈單鏈、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,術語「寡核苷酸」係指核糖核酸 (RNA)或去氧核糖核酸(DNA)之寡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)、硫代磷酸酯、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特異性結合至靶 聚核苦酸,例如Watson-Crick型驗基配對、Hodgsteen型驗 基配對或反向Ho6gsteen型驗基配對、或諸如此類。 募核苷酸可為「嵌合」寡核苷酸,亦即,由不同區域組 成。在本發明之上下文中,「嵌合」化合物係寡核苷酸, 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。在寡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 核苷酸通常包括至少一個對寡核苷酸進行修飾以顯示一或 多個期望性質之區域。寡核苷酸之期望性質包含但不限於 156342.doc 201143782 (例如):增加對於核酸酶降解之抗性、增加細胞攝取、及/ 或加對於乾核酸之結合親和力。寡核苷酸之不同區域可 因此具有不同性質。本發明之嵌合募核苷酸可形成為兩種 或更夕種上述募核苷酸、經修飾寡核苷酸、寡核苷及/或 • 募核苷酸類似物的混合結構。 • 募核苦酸可由可在「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 ◎ 冑連接。間隔體意欲在區域之間構成共價「橋」且在較佳 匱形下具有不超過約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 純質(嵌合劑、槽溝黏合劑、毒素、螢光團等),具有親 月曰)·生誘導特異性二級結構(例如,誘導以螺旋之含丙胺酸 之肽)。 本文所用之「LHX2」及「咖同源序列2」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 Q 序列、有義及反義聚核苷酸鏈等。 本文所用之詞語UM同源序列2、LHX2、hLhx2、同源 序 2 LH2、LH-2、LIM/同源序列 Lhx2、LIM/同源 序列2、MGC138390在文獻中視為相同,且在本中請案中 - 可互換使用。 本文所用之術語Γ料 H . 〜 對·.·具有特異性之募核苷酸」或「靶 向…之寡核苷酸俜指且 保知具有如下序列之寡核苷酸:(i)能夠 與乾向基因之一部分形成穩定複合物,或⑴)能夠與把向 ^ RNA轉錄本的一部分形成穩定雙鏈體。複合物及 156342.doc 201143782 雙鏈體之穩定性可藉由理論計算及/或活體外分析進行測 定。測定雜交複合物及雙鏈體之穩定性的實例性分析闡述 於下文實例中。 本文所用之術語「靶核酸」涵蓋自該DNA轉錄之DNA、 RNA(包括mRNA前體及mRNA)、以及源自該RNA、編碼序 列、非編碼序列、有義或反義聚核苷酸之cDNA。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 蛋白質轉譯位點之移位、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可調節編 碼產物或寡核苷酸之表現。 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,介質係具有5-25個核苷酸之「小干擾」 RNA雙鏈體(siRNA) 〇 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之多蛋 白siRNA複合物(RNA誘導之沉默複合物)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 156342.doc -10- 201143782 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4〇個討、約5至約儿個加、 約10至約30個nt、約15至約25個拊、或約2〇_25個核苷酸。 ' 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(s〇uthern 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 〇 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(Cech, (1988) J.201143782 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 LHX2. The present application claims priority to U.S. Provisional Patent Application Serial No. 61/346,284, filed on Jan. [Prior Art] O DNA-RNA and RNA-RNA hybridization are important for many nucleic acid functional aspects including DNA replication, transcription, and translation. Hybridization is also critical 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 property that DNA-RNA hybrids can be used as receptors 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 oligodeoxynucleotide (ODN), or it can be expressed as an RNA molecule from an endogenous 〇 gene. The FDA recently approved the antisense drug VITRAVENETM (for the treatment of cytomegalovirus retinitis), indicating that this antisense drug has therapeutic utility. BRIEF DESCRIPTION OF THE DRAWINGS The Summary is provided to introduce a summary of the invention This Summary is submitted with the understanding that it is not 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 of inhibiting the action of a natural antisense transcript 156342.doc 201143782 by upregulating an antisense oligonucleotide by targeting any region of a natural antisense transcript The righteous gene is reached. It is also encompassed herein that inhibition of natural antisense transcripts by siRNA, ribozymes and small molecules is considered to be within the scope of the invention. One embodiment provides a method of modulating the function and/or expression of LHX2 polynucleotide in a patient's cells or tissues in vivo or in vitro, comprising subjecting such cells or tissues to a length of 5 to 30 nucleotides The oligonucleotide is contacted, wherein the nucleotide has at least 5% sequence identity with the reverse complement of the polynucleotide, and the polynucleotide comprises nucleotides 1 to 2145 of SEQ m NO: , nucleotides 1 to 6〇6 in SEQ ID NO: 3, and 5 to 30 contiguous nucleotides in nucleotides 1 to 480 of SEQ ID NO: 4, thereby modulating the patient's cells in vivo or ex vivo The function and/or performance of the LHX2 polynucleotide in the tissue. In one embodiment, the oligonucleotide targets a natural antisense sequence of the LHX2 polynucleotide (eg, the nucleotides set forth in SEQ ID NOS: 2 to 4), and any variants, alleles thereof , homologs, mutants, derivatives, fragments and complementary sequences. Examples of antisense oligonucleotides are, for example, SEq ID NO: 5 to 16 7f> ° Further embodiments provide for the function and/or expression of LHX2 polynucleotides in a patient's cells or tissues in vivo or in vitro. A method comprising contacting the cells or tissues with an antisense oligonucleotide of 5 to 30 nucleotides in length, wherein the nucleotide and the reverse complement of the LHX2 polynucleotide antisense transcript Having at least 50% sequence identity; thereby modulating the function of LHX2 polynucleotides in a patient's cells or tissues in vivo or in vitro and/or Table 156342.doc 201143782. Another embodiment provides a method of modulating the function and/or expression of an LHX2 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 An antisense oligonucleotide contact, wherein the oligonucleotide has at least 50% sequence identity to an antisense oligonucleotide of an LHX2 antisense polynucleotide; thereby modulating a patient cell or in vivo or in vitro The function and/or performance of the LHX2 polynucleotide in the tissue. In one embodiment, the composition comprises one or more antisense oligonucleotides that bind to a sense and/or antisense LHX2 polynucleotide. In one embodiment the 'oligonucleotide comprises one or more modified or substituted nucleotides. In one embodiment, the oligonucleotide comprises one or more modified linkages. In a consistent embodiment, the modified nucleotide comprises a modified test group comprising the following: a thioformatate, a phosphonic acid methyl vinegar, a peptide nucleic acid, a 2, a fluorenyl group, a fluorine _ or a carbon, a methylene group or the like. Locked nucleic acid (LNA) molecules. Preferably, the modified nuclear acid-locking nucleic acid molecule comprises a_L-LNA. In one embodiment, the oligonucleotide is administered to the patient subcutaneously, intramuscularly, intravenously or intraperitoneally. In only the examples, nucleotides are administered in the form of a pharmaceutical composition. The treatment regimen includes administering the antisense compound to the patient at least once; however, the treatment can be modified to include multiple doses over a period of time. The treatment can be combined with one or more other types of therapies. In an embodiment, the oligonucleoside is encapsulated in a liposome or attached to a carrier molecule (e.g., cholesterol, TAT peptide). 156342.doc 201143782 Other aspects are set out below. [Embodiment] Several aspects of the present invention will be described hereinafter with reference to the application of the example of the 庑田水. Understand that the texts listed in this article have a complete understanding of the present invention. The present invention may be practiced without the use of any specific details or other methods. The invention is not limited to the order of the various acts or events, which may be The occurrence of different sequences and/or simultaneous occurrences with other actions or events does not necessarily require all of the illustrated actions or events to implement the methods. All of the genes, gene names, and gene products disclosed herein are intended to correspond to the compositions disclosed herein. And methods of homologs from any species H. These terms include, but are not limited to, human and mouse genes and gene products. It should be understood that when revealing genes or gene products from a particular species, this indication is intended to have only The examples are not to be construed as limiting, unless the context clearly indicates. Thus, for example, in certain embodiments relating to mammalian nucleic acid and amino acid sequences, the genes disclosed herein are intended to encompass other animals. Homologous and/or orthologous genes and gene products 'These other animals include, but are not limited to, other mammals, Fish, amphibians, reptiles, and birds. In one embodiment, the gene or nucleic acid sequence is human. Definitions The terms used herein are used for the purpose of describing particular embodiments only and are not intended to limit the invention. The singular forms "a", "a", "a", "a", "the", "the", "the" In addition, the terms "including", "inciudes", "having", "has", "with", or "the" are used in the detailed description and/or the scope of the application. In the case of variations, the terms are intended to mean a range of inclusions in a manner similar to the term "comprising". The term "about" or "approximately" means an acceptable margin of error for a particular value determined by those skilled in the art 'which depends in part on the measurement or measurement of the value', ie, the limitations of the measurement system. . For example, according to industry practice '"about" can mean within one or more standard deviations. Another option of "about" may mean a difference of up to 2%, preferably up to 10°/. More preferably up to 5%, and even more preferably up to 1%. Another option is that, particularly for a biological system or process, the term can 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 dry gene, and any other transcript that can be elucidated. "Antisense nucleus acid" or "antisense compound" means an RNA or DNA molecule that binds to another RNA or DNA (target RNA, DNA). For example, if an RNA oligonucleotide is used, it binds to another RNA target via rnA-RNA interaction and alters the activity of the target RNA. Antisense oligonucleotides can up-regulate or down-regulate the expression and/or function of a particular polynucleotide. This definition is intended to encompass 156342.doc 201143782 containing any foreign RNA or DNA molecule that can be used therapeutically, diagnostically, or otherwise. 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 oligomeric compounds, Antisense oligonucleotides, external leader sequence (EGS) oligonucleotides, alternative splicing, primers, probes, and other oligomeric compounds that hybridize to at least a portion of the target nucleic acid. Thus, the compounds can be introduced as single-stranded, double-stranded, partially single-stranded, or cyclic oligomeric compounds. In the context of the present invention, the term "oligonucleotide" refers to an oligo 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, including deoxyribonucleosides, ribonucleosides, substituted and alpha-rotated Format, peptide nucleic acid (PNA), locked nucleic acid (LNA), phosphorothioate, decyl phosphonate, and the like. Oligonucleotides can specifically bind to a target polynucleic acid by means of a regular pattern of monomer-to-monomer interactions, such as Watson-Crick type base pairing, Hodgsteen type base pairing or reverse Ho6gsteen type base pairing, or And so on. The raised nucleotides can be "chimeric" oligonucleotides, i.e., composed of different regions. In the context of the present invention, a "chimeric" compound is an oligonucleotide comprising two or more chemical regions, such as a DNA region, an RNA region, a PNA region, and the like. In the case of an oligonucleotide compound, each chemical region consists of at least one monomer unit (i.e., nucleotide). Such oligonucleotides typically include at least one region that modifies the oligonucleotide to reveal one or more desired properties. Desirable properties of the oligonucleotide include, but are not limited to, 156342.doc 201143782 (for example): increasing resistance to nuclease degradation, increasing cellular uptake, and/or adding binding affinity for dry nucleic acids. Different regions of the oligonucleotide may thus have different properties. The chimeric nucleotides of the present invention can be formed into a mixed structure of two or more of the above-mentioned nucleotides, modified oligonucleotides, oligonucleosides, and/or nucleotide analogs. • The nucleus acid can be composed of a region that can be joined in a “register”, that is, a single system continuous connection (such as in natural DNA), or a gap ◎ 胄. The spacers are intended to form a covalent "bridge" between the regions and have a length of no more than about one carbon atom in the preferred shape. The spacer may have different functions, such as having a positive or negative charge, having a specific nucleic acid purity (chimeric agent, groove binder, toxin, fluorophore, etc.), having pro-monthly) Grade structure (eg, induction of alanine-containing peptides in a helix). As used herein, "LHX2" and "coffee homologous sequence 2" encompass all family members, mutants, alleles, fragments, species, coding and non-coding Q sequences, sense and antisense polynucleotide chains, and the like. As used herein, the terms UM homologous sequence 2, LHX2, hLhx2, homologous sequence 2 LH2, LH-2, LIM/homologous sequence Lhx2, LIM/homologous sequence 2, MGC138390 are considered identical in the literature, and In case - interchangeable use. As used herein, the term "H. ~.." has specific nucleotides" or "targeted oligonucleotides" and refers to oligonucleotides having the following sequences: (i) capable of Forming a stable complex with a portion of the stem gene, or (1)) capable of forming a stable duplex with a portion of the RNA transcript. The stability of the complex and 156342.doc 201143782 duplex can be calculated by theory and / Or in vitro assays are performed. 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, RNA (including mRNA precursors) transcribed from the DNA. And mRNA), and cDNA derived from the RNA, coding sequence, non-coding sequence, sense or antisense polynucleotide. 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 includes, for example, copying and transcription. The RNA function to be interfered with includes all important functions, such as shifting 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 the regulation of 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 medium is a "small interference" RNA duplex (siRNA) of 5-25 nucleotides. The 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 (RNA-induced silencing complex) called RISC. 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) where the siRNA duplex interacts in a sequence-specific manner to catalyze cleavage in a catalytic manner. Small interfering RNAs which can be used in the present invention can be synthesized and used according to procedures well known in the art and known to those skilled in the art, 156342. doc -10- 201143782. The small interfering RNA used in the methods of the invention suitably comprises 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 4, about 5 to about 3,000, about 10 to about 30 nt, about 15 to about 25 拊, or about 2 〇. _25 nucleotides. The selection of appropriate oligonucleotides is facilitated by the use of computer programs that automatically align nucleic acid sequences and indicate 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 measuring the pCR product. 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 blotting is performed to determine the degree of agreement between 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 with enzymatic activity (Cech, (1988) J.

American. Med· Assoc. 26〇,3030-3035)。酶性核酸(核糖 ,酶)首先藉由結合至靶RNA而發揮作用。該結合經由酶性 核酸之乾結合部分來進行,該靶結合部分緊鄰分子中用於 裂解乾RNA之酶性部分。因此,酶性核酸首先識別靶RNA 且然後經由驗基配對與靶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RN A。 156342.doc 201143782 「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 至在HIV RNA中編碼之TAR序列。此意欲係一具體實例。 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」通常表示藉由磷酸二酯鍵或其 類似物連接以形成尺寸介於數個單體單元(例如,約3-4個) 至約數百個單體單元之寡核苷酸的單體。磷酸二酯鍵聯之 類似物包含:硫代磷酸酯、二硫代磷酸酯、膦酸甲酯、硒 代磷酸酯、胺基磷酸酯、及諸如此類,如下文更全面所 述。 術語「核苷酸」涵蓋天然存在之核苷酸以及非天然存在 之核苷酸。熟習此項技術者應明瞭,先前視為「非天然存 在」之各種核苷酸後來已發現於自然界中。因此,「核苷 酸」不僅包含含有已知°票呤及°密π定雜環之分子,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-曱基腺嘌呤、7-去氮黃嘌呤、7-去氮鳥嘌呤、 N4,N4-乙醇胞嘧啶、N6,N6-乙醇-2,6-二胺基嘌呤、5-曱基 胞°密咬、5-(C3-C6)-炔基胞。密。定、5-氟尿α密咬、5-溴尿嘴 啶、假異胞嘧啶、2-羥基-5-甲基-4-三唑并吡啶、異胞嘧 156342.doc -12- 201143782 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「非天然存在」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,其係視為與人類之治療及診斷應用 有關之天然存在之核苷酸。核苷酸包含天然2’-去氧及2·-羥 基糖(例如,如Kornberg及Baker,DNAReplication,第2版 (Freeman, San Francisco, 1992)中所述)以及其類似物0 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 ;American. Med· Assoc. 26〇, 3030-3035). The enzymatic nucleic acid (ribose, enzyme) first acts by binding to the target RNA. The binding is carried out via a dry binding portion of an enzymatic nucleic acid that is immediately adjacent to the enzymatic portion of the molecule used to lyse the dry RNA. Thus, the enzymatic nucleic acid first recognizes the target RNA and then binds to the target rNA via the genomic pairing and acts enzymatically to cleave the target RN A upon binding to the exact site. 156342.doc 201143782 "Decoy RNA" means an RNA molecule that mimics the natural binding domain of a ligand. The decoy RNA thus competes with the natural binding target for binding to a specific ligand. For example, it has been shown that overexpression of HIV transactivation (TAR) RNA can be used as a "bait" and effectively binds to the HIV tat protein, thereby preventing its binding to the TAR sequence encoded in HIV RNA. This is intended to be a specific example. Those skilled in the art will recognize that this is only an example and that other embodiments can be readily derived using techniques generally known in the art. The term "monomer" as used herein generally means attached by a phosphodiester bond or an analog thereof to form an oligomer having a size ranging from a plurality of monomer units (for example, about 3-4) to about several hundred monomer units. A monomer of a nucleotide. Phosphodiester linked analogs include: phosphorothioates, dithiophosphates, methyl phosphonates, phosphatidyl phosphates, amine phosphates, and the like, as described more fully below. The term "nucleotide" encompasses naturally occurring nucleotides as well as non-naturally occurring nucleotides. Those skilled in the art should be aware that the various nucleotides previously considered "non-natural" have subsequently been discovered in nature. Therefore, "nucleotide" includes not only molecules containing a known quaternary ring and a heterocyclic hexacyclic ring, but also heterocyclic analogs and tautomers thereof. An illustrative example of other types of nucleotides are molecules that contain: adenine, guanine, thymine, cytosine, uracil, guanidine, xanthine, diamine guanidine, 8-sided oxy-Ν6-曱Adenine, 7-deazapurine, 7-deazaguanine, N4, N4-ethanol cytosine, N6, N6-ethanol-2,6-diaminopurine, 5-quinone cytosolic, 5-(C3-C6)-alkynyl group. dense. 5-, 5-fluorouridine α-bite, 5-bromoindoline, pseudoisopyrimidine, 2-hydroxy-5-methyl-4-triazolopyridine, isocytosine 156342.doc -12- 201143782 "Non-naturally occurring" nucleotides as described in U.S. Patent No. 5,432,272 to Benner et al. The term "nucleotide" is intended to cover every and all such examples as well as the analogs and tautomers thereof. Nucleotides of particular interest include adenine, guanine, thymine, cytosine, and uracil, which are considered naturally occurring nucleotides associated with therapeutic and diagnostic applications in humans. Nucleotides comprise native 2'-deoxy and 2-hydroxyl sugars (for example, as described in Kornberg and Baker, DNA Replication, 2nd Edition (Freeman, San Francisco, 1992)) and analogs thereof are related to nucleotides. An "analog" 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, J.J., (2001) Nature Biotechnology 19:17-18 ; Manoharan M·,(1999) Biochemica et Biophysica Acta 1489:117-139 i Freier S. M., (1997) Nucleic Acid Research, 25:4429-4443, Uhlman, E·,(2000) Drug Discovery & Development, 3: 203-213, Herdewin P., (2000) Antisense & Nucleic Acid Drug Dev” 10:297-310) ; 2’-0、3'-C連接之 [3.2.0]雙環阿糖核苷。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。 本文所用之「雜交」意指寡聚化合物之實質上互補鏈之 配對。一種配對機制涉及募聚化合物鏈之互補核苷或核苷 酸驗基(核芽酸)之間的氫鍵,其可為Watson-Crick、 156342.doc •13- 201143782 HWgsteen或反向H〇dgSteen氫鍵。舉例而言,腺嘌呤及胸 腺嘧啶係經由形成氫鍵配對之互補核苷酸。雜交可在不 情形下發生。 5 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, 或活性’且存在足夠互補性程度以避免反義化合物與非乾 核酸序列在期望發生肖異性結合之條件下(亦n舌體 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。 本文所用之片語「嚴格雜交條件」《「嚴格條件」係指 本發明化合物與其靶序列發生雜交、但與最少數量之其: 序列發生雜交的條件°嚴格條件具有序列依賴性且在不同 情況及本發明上下文中有所不同,藉由募聚化合物之性質 及組成及研究其之分析來測定寡聚化合物與靶序列進行雜 父的「嚴格條件」。一般而言,嚴格雜交條件包括低濃度 (<0.15 M)含有無機陽離子(例如Na++或κ++)之鹽 低離子強度),溫度高於奶七以旦低於寡聚化合物:靶 序列複合物之Tm,及存在變性劑(例如甲醯胺、二甲基甲 醯胺一甲基亞楓)或洗滌劑十二烷基硫酸鈉(SDS)。舉例 而5 ’對於每-1%甲醯胺而言’雜交速率降低。高 嚴格度雜交條件之實例係O.i χ氯化鈉-檸檬酸鈉緩衝液 (ssc)/o.i% (w/v) SDS(在6crc 下,保持3〇分鐘)。 本文所用之「互補」係指一或兩個寡聚鏈上之兩個核苷 酸之間精確配對的能力。舉例而言,若反義化合物某一位 156342.doc -J4- 201143782 置處之核鹼基能夠與靶核酸(該把核酸係DNA、RNA、戍 寡核苷酸分子)某一位置處之核驗基發生氫鍵結,則該寡 核苷酸及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、或寡核芽酸 ‘ 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 I 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,募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)。本發明募聚化合物包括與 其所乾向靶核酸序列内靶區域之至少約7〇%、或至少約 75%、或至少約8〇%、或至少約85%、或至少約9〇%、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18個與靶區域互補且由此特異性 雜交之反義化合物將代表9〇%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18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 接)之反義化合物與靶核酸具有77·8%之總體互補性且由此 屬於本發明範圍内。具有靶核酸區域之反義化合物的互補 !生百刀比通常可使用業内已知之BLAST程式(鹼基局部對 156342.doc -15- 201143782 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(Wisc〇nsin 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 Appl. Math., (1981) 2, 482-489) 〇 本文所用之術語「熱力學熔點(Tm)」係指在界定離子強 度、pH、及核酸濃度下與乾序列互補之寡核普酸中之 在平衡下與靶序列雜交的溫度。通常,對於短募核皆酸 (例如,具有1 〇至5 〇個核苷酸)而言,嚴格條件係彼等以下 條件:鹽濃度在pH 7.0至8.3下為至少約⑴㈦至^ MiNa 離子濃度(或其他鹽)且溫度為至少約3(TC ^嚴格條件亦可 藉由添加諸如曱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芽酸序列。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處理 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 本發月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,且可產生改變之mRNA或 156342.doc -16 - 201143782 ^生:構或功能可改變或不改變的多肽。任'給定天然或 =因:不具有等位基因形式、具有1或許多等位基 產生變體之常見突變變化通常歸因於核紐之天 :、失、添加'或取代。該等變化_中之每—者可在給 足序列中單獨、或與其他者組合發生—或多次。 所得多肽通常彼此之間具有顯著之胺基酸—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Freier & Altmann, (1997) Nucl. Acid. Res., 25(22), 4429-4443, Toulme, JJ, (2001) Nature Biotechnology 19:17-18; Manoharan M·, (1999) Biochemica et Biophysica Acta 1489:117-139 i Freier SM, (1997) Nucleic Acid Research, 25:4429-4443, Uhlman, E., (2000) Drug Discovery & Development, 3: 203-213, Herdewin P., (2000) Antisense & Nucleic Acid Drug Dev" 10: 297-310); 2'-0, 3'-C linked [3.2.0] bicyclic glucosinolates. These analogs are designed to enhance binding properties (eg, Synthetic nucleotides of duplex or triplex stability, specificity, or the like. As used herein, "hybridization" means the pairing of substantially complementary strands of an oligomeric compound. A pairing mechanism involves hydrogen bonding between complementary nucleosides or nucleotides (nuclear acid) of a compound chain, which can be Watson-Crick, 156342.doc • 13-201143782 HWgsteen or reverse H〇dgSteen Hydrogen bond. For example, adenine and thymidine are complementary nucleotides that form a hydrogen bond pair. Hybridization can occur without circumstances. 5 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 the function and/or activity' and is sufficiently complementary to avoid antisense compounds The non-native nucleic acid sequence undergoes non-specific binding under conditions where it is desired to undergo heterosexual binding (also under physiological conditions in the case of in vivo or therapeutic treatment, and under conditions in which the analysis is performed in the case of in vitro analysis). As used herein, the phrase "stringent hybridization conditions" and "stringent conditions" refer to the conditions under which a compound of the invention hybridizes to its target sequence, but with a minimum number of: sequence hybridizations. Strict conditions are sequence dependent and in different cases and In the context of the present invention, the "stringent conditions" of the heteropoly compound and the target sequence are determined by the nature and composition of the polycondensation compound and the analysis thereof. In general, stringent hybridization conditions include low concentrations (<0.15 M) of low ionic strengths of salts containing inorganic cations (e.g., Na++ or κ++), temperatures higher than that of milk, and less than oligomeric compounds: target sequence complexes Tm of the substance, and the presence of a denaturant (such as formamidine, dimethylformamide monomethyl sulfoxide) or detergent sodium dodecyl sulfate (SDS). For example, 5' decreases the rate of hybridization for every -1% metformin. An example of high stringency hybridization conditions is O.i χ sodium chloride-sodium citrate buffer (ssc) / o.i% (w/v) SDS (3 sec at 6 crc). As used herein, "complementary" refers to the ability to precisely pair between two nucleotides on one or two oligomeric chains. For example, if the antisense compound 156342.doc -J4- 201143782 is placed in a nucleobase and can be verified at a position near the target nucleic acid (the nucleic acid DNA, RNA, or oligodeoxynucleotide molecule) When hydrogen bonding occurs, the position at which hydrogen bonding occurs between the oligonucleotide and the target nucleic acid is regarded as a complementary position. When a sufficient number of complementary positions in each molecule are occupied by a nucleoside that can be hydrogen bonded to each other, the 'oligomer compound and other DNA, RNA, or oligonucleotide' molecules are complementary 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 such that I is stable between the oligomeric compound and the target nucleic acid and Specific binding. 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. Alternatively, the raised nucleotides can hybridize in one or more regions such that the inserted or adjacent segments do not participate in hybridization events (e.g., loop structures, mismatches, or hairpin structures). The polycondensation compound of the present invention comprises at least about 7%, or at least about 75%, or at least about 8%, or at least about 85%, or at least about 9%, or about the target region of the target nucleic acid sequence to which it is dried, or To ◎ about 95% less, or at least about 99% sequence complementarity. For example, an antisense compound of 18 of the 20 nucleotides of the antisense compound that is complementary to the target region and thereby specifically hybridizes will represent a 99% complementarity. In this example, the remaining non-complementary nucleotides may 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 of 18 nucleotides in length and having 4 (four) non-complementary nucleotides flanked by two regions that are fully complementary to the target nucleic acid has 77.8% overall complementarity to the target nucleic acid. It is within the scope of the invention. The complementation of antisense compounds with a target nucleic acid region can generally be measured using the BLAST program known in the art (base partial pair 156342.doc -15-201143782 quasi-search tool) and PowerBLAST program. The homology, sequence identity, or percent complementarity can be used by, for example, the Gap program (Wisc〇ns Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.) using default settings (its use) Smith and Waterman algorithm) (Adv Appl. Math., (1981) 2, 482-489) As used herein, the term "thermodynamic melting point (Tm)" means drying under defined ionic strength, pH, and nucleic acid concentration. The temperature of the sequence complementary to the oligonucleotide that hybridizes to the target sequence under equilibrium. Generally, for short-collar acid (for example, having 1 〇 to 5 核苷酸 nucleotides), stringent conditions are the following: salt concentration is at least about (1) (seven) to ^ MiNa ion concentration at pH 7.0 to 8.3. (or other salt) and at a temperature of at least about 3 (TC ^ stringent conditions can also be achieved by the addition of a destabilizing agent such as guanamine. As used herein, "modulating" means increasing (stimulating) or reducing (inhibiting) a gene. In the context of a polynucleotide sequence, the term "variant" may encompass a polynucleotide sequence associated with a wild-type gene. This definition may also include, for example, "alleles", Splice, "species" or "polymorphism" variants. Splice variants can be significantly consistent with a reference molecule, but usually have a larger or smaller number of aggregates due to alternate splicing of exons during mRNA processing. Nucleotide. The corresponding polypeptide may have an additional functional domain or a non-existing domain. The sequence of the variant of the species varies from species to species. It is particularly useful in this month to be a variant of the wild-type gene product. Body At least one mutation in the nucleic acid sequence, and can produce altered mRNA or a polypeptide whose structure or function may or may not be altered. Any 'given natural or = cause: does not have an allele Gene forms, common mutational changes with 1 or many allelic variants are usually attributed to the day of the nucleus: loss, addition, or substitution. Each of these changes can be alone in the sequence of the foot. Or in combination with others—or multiple times. The resulting polypeptides typically have significant amino acid-likeness with each other. Polymorphisms change in the nucleotide sequence of a particular gene between individuals in a given species.

多態性變體亦可涵蓋「單一核芽酸多態性」(sNp)或 單驗基犬變,其中聚核芽酸序列之—個驗基有所變化。 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)β 何生聚核苷酸包含經受化學修飾(例如,氫由烷基、醯 土或胺基代替)之核酸。衍生物(例如,衍生寡核苷酸)可 。括非天然存在之部分,例如改變之糖部分或糖間鍵聯。 μ等實例f生衍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、辅因子、抑制劑、磁 性顆粒、及諸如此類。 衍生」夕肽或肽係(例如)藉由以下方式進行修飾者: 糖基化聚乙一醇化、磷醯化、硫酸化、還原/烷基化、 醯基化、化學偶合、或輕度福爾馬林(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「動物」或「患者」意欲包含(例如)人 156342.doc •17· 201143782 哺乳動物、狼子、 鼠、鳥、雞、爬行 類、綿羊、麋鹿、鹿、長耳鹿、水紹 馬、牛、豬、山羊、狗、貓、大鼠、 動物、魚、昆蟲及缺j蛛。 「哺乳動物」涵蓋通常處於醫學護理下之溫企哺乳動物 (例如,人類及家養動物)。實例包含貓、 八 馬、牛、及 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「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 2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(位於中樞及周邊神經系統二者中之 邛7刀)之疾病或病症。神經疾病或病症包含但不限於後天 性癲癎失語症;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 syndr〇me);亞歷山大病(Alexander . )’阿爾拍斯病(Alpers' disease);交叉性肢體癱 疾阿爾次海默氏病(Alzheimer's disease);血管性癡呆; 156342.doc -18- 201143782 肌萎縮側索硬化;無腦畸形;天使症候群(Angeiman syndrome),·血管瘤病;缺養症;失語症;失用症;蛛網 膜囊腫,蛛網膜炎;阿·蔡二氏畸形(Anr〇ni_chiaH maiformation);動靜脈畸形;阿斯佩格症候群(Asperger ' Syndr〇me);運動失調性毛細血管擴張症;注意力缺陷伴 • 乡動病症;自閉症;自主神經功能障礙;背痛;巴登氏病 (Batten disease),貝切特氏病(Behcet,sdisease);貝爾麻痺 〇 (BeU'S Palsy);良性本質瞼痙攣;良性局部肌萎縮;良性 顱内冋壓,賓斯旺格病(Binswanger,s disease);眼瞼痙 攀;布洛克-蘇茲貝克症候群(B1〇ch 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); 腕迢症候群;灼性神經痛;中樞性疼痛症候群;腦橋中央 髓鞘溶解,碩部病症;腦動脈瘤;腦動脈硬化症;大腦萎 〇 縮;大腦性巨人症;大腦性麻痺;夏-馬-圖三氏病 (Charcot-Marie-Tooth disease);化學療法誘導性神經病及 神經性疼痛;恰裏畸形(Chiad malformati〇n);舞蹈病;慢 性炎症性脫髓鞘性多發性神經病;慢性疼痛;慢性區域性 疼痛症侯群’科_勒二氏症候群(以出^ 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’顧動脈炎,顱縫早閉;克雅氏病(Creutzfeldt-Jakob dlSeaSe);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— 156342.doc •19- 201143782 舞蹈足症候群;丹-沃二氏症候群(Dandy Walker 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; 腦三叉神經丘管瘤病;癲癇症;歐勃麻痒(Erb's palsy); 特發性震顏;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r 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 相關性脊髓病;哈_斯 二氏病(Hallervorden-Spatz disease);頭部損傷;頭痛;半 面痙攣;遺傳性痙攣性截癱;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相關性癡呆及神經病(以及八11)8之神經表 現);前腦無裂畸形;亨廷頓病(Huntington,s仙⑽)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;2 素失調症;嬰兒植烧酸貯積病;嬰兒雷弗蘇姆病㈣咖以 refsum disease) ·’嬰兒痙攣;炎性肌病;顱内囊腫;顱内 156342.doc -20- 201143782 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);延髓外側(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;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 156342.doc -21- 201143782 陣攣性腦病;肌陣攣;肌病;肌強直;嗜眠症;神經纖維 瘤病;神經阻滯劑惡性症候群;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-麥克勞德病症(O'Sullivan-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(Ohtahara syndrome);撖欖體腦橋小腦萎縮;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)、于廷頓病、阿爾茨海默氏病、肌萎縮侧索硬化 (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 R〇mberg 佩-梅一氏病(Pelizaeus-Merzbacher disease);週期性癱 瘓’周邊神經病;疼痛性神經病及神經性疼痛;持續性植 物人狀態;全身性發育遲緩;旋光性喷嚏反射;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;小兒麻痒症後期症候群;帶狀皰疹後神 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);原發性側索硬化症;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;進行性核上麻痒;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 156342.doc •22· 201143782 症候群;雷夫敘姆病(Refsum disease);重複性運動病症; 重複性壓迫損傷;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);謝耳德病(Schilder’s disease);腦裂; 透明隔·視神經發育不良;驚嚇嬰兒症候群;帶狀皰疹; 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-Person 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 disease); 顳動脈炎;脊髓牽扯症候群;湯姆森病(Thomsen disease); 胸廓出口症候群;三叉神經痛症(Tic Douloureux);托德氏 麻痺(Todd's paralysis);多動穢語症候群;短暫性腦缺血 發作;傳播性海绵狀腦病;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;血管性癡呆(多發梗塞性癡呆);血管炎,包含顳動脈 炎;希林二氏病(Von Hippel-Lindau disease);瓦偷伯格氏 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 156342.doc -23- 201143782 病(Wildon’s disease);及澤韋格症候群 syndrome) ° 「A液疾病或病症」包含影響造血細胞或組織之疾病、 病症、或病狀。血液病症包含與異常血液含量或功能有關 之疾病、病症、或病狀。血液病症之實例包含源自骨髓輻 照或癌症化學療法治療之病症、諸如以下病症:惡性貧 血、出血性貧血、溶血性貧血、再生障礙性貧血、鐮狀細 胞性貧血、鐵粒幼細胞貧血、與慢性感染(例如瘧疾、錐 蟲病、HI V、肝炎病毒或其他病毒)有關之貧血、由骨髓缺 失引起之骨髓病性貧血、源自貧血之腎衰竭、貧血、紅細 胞增多症、感染性單核細胞增多症(IM)、急性非淋巴細胞 性白血病(ANLL)、急性髓性白血病(AML)、急性早幼粒細 胞白血病(APL)、急性粒-單核細胞白血病(AMMoL)、真性 紅細胞增多症、淋巴瘤、急性淋巴細胞白血病(ALL)、慢 性淋巴細胞白血病、維爾姆斯腫瘤(wilm,s turn〇r)、尤因 氏肉瘤(Ewing’s sarcoma)、視網膜母細胞瘤、血友病、與 血检形成風險增加有關之病症、胞療、地中海貧血、抗體 誘導之病症(例如輸血反應及成紅細胞增多病)、紅血細胞 之機械性創傷(例如微血管病性溶血性貧血、血栓性血小 板減少性紫癜及彌漫性血管内凝血)、由寄生蟲(例如遽原 蟲)引起之感染、來自(例如)鉛中毒之化學性損傷、及脾功 能充進。淋巴系統疾病包含但不限於淋巴結炎、淋巴管擴 張、淋巴管炎、淋巴水腫、淋巴囊腫、淋巴組織增殖性病 症、皮膚黏膜淋巴結症候群、網狀内皮組織增殖、脾疾 156342.doc -24- 201143782 病、胸腺增生、胸腺腫瘤、肺結核、淋巴結、假性淋巴 瘤、及淋巴異常。淋巴造血系統之病症包含但不限於非霍 奇金氏淋巴瘤、慢性淋巴細胞白血病、及反應性淋巴樣組 織增生。 • 聚核苷酸及募核苷酸組合物及分子 • 靶:在一實施例中,靶包括LIM同源序列2(LHX2)之核 酸序列,包含(不限於)與LHX2有關之有義及/或反義非編 碼及/或編碼序列。 〇 LIM-同源結構域(LIM-HD)蛋白可調節靶基因之組織特 異性表現’且為許多器官之正常發育及維持所需。在脊椎 動物及無脊椎動物之胚胎發育期間,LIM同源結構域轉錄 因子家族成員藉由控制諸如細胞不對稱分裂、組織特異化 及特定細胞類型之分化等過程來發揮重要作用。此家族之 種成員Lhx2基於其在若干不同組織發育中之功能而尤其 令人關注’其功能係經由間質·上皮相互作用及對幹細胞/ Q 祖細胞之調節來達成。Lhx2係轉錄因子之此家族成員,其 由12個已知基因組成,該等基因之結構特徵在於具有兩個 胺基末端鋅指基序(LIM結構域)及羧基末端同源結構域。 Lhx2在小鼠中之功能失活會導致大腦皮質尺寸減小以及外 皮稽邊、端腦、嗅覺系統之異常發育、及外皮模式形成缺 陷。Polymorphic variants may also cover "single nuclear bud acid polymorphism" (sNp) or single-killer canine changes, in which the base of the polymorphic acid sequence changes. The presence of a SNP may indicate, for example, that a population has a predisposition to disease state (i.e., susceptibility to resistance). Beta Polynucleotide comprises undergoing chemical modification (eg, hydrogen is replaced by an alkyl, alumina or amine group) Nucleic acid. Derivatives (e.g., derived oligonucleotides) can be used. Non-naturally occurring parts, such as altered sugar moieties or intersaccharide linkages. Examples of μ, such as derivative phosphorothioates, and other sulfur-containing material-derived nucleic acids known in the art may also contain a label, including radionucleotides, enzymes, phosphor chemiluminescent agents, color formers, receptors, Cofactors, inhibitors, magnetic particles, and the like. Derivatives of peptides or peptides, for example, are modified by: glycosylation, polyethylation, phosphating, sulfating, reduction/alkylation, thiolation, chemical coupling, or mild flu Marlin (formalin) processing. The derivative may also be modified to contain a detectable label (directly or indirectly) including, but not limited to, a radioisotope, a fluorescent, and an enzymatic label. The term "animal" or "patient" as used herein is intended to include, for example, human 156342.doc. •17· 201143782 Mammals, wolves, rats, birds, chickens, reptiles, sheep, elk, deer, mule deer, water, horse, cow, pig, goat, dog, cat, rat, animal, fish, Insects and J. Spiders. "Mammals" include warm-blooded mammals (eg, humans and domestic animals) that are usually under medical care. Examples include cats, horses, cows, and humans, and only humans. Or treatment" encompasses treatment of a disease state in a mammal, and comprises: (4) preventing the disease state in a mammal, particularly when the mammal is susceptible to a disease state but has not been diagnosed with 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. It also includes improving the symptoms of the disease (for example, alleviating pain or discomfort), where the improvement may directly affect or may not directly affect the disease (eg, cause, infection, performance, etc.). "Neural disease or condition" as used herein means Any of the diseases or conditions of the nervous system and/or the visual system. "Nervous diseases or conditions" include 2 central nervous systems (brain, brainstem and cerebellum), peripheral nervous system (including cranial nerves), and autonomic nervous system (located A disease or condition of the central and peripheral nervous system. The neurological disease or condition includes, but is not limited to, acquired epilepsy aphasia; acute disseminated encephalomyelitis; adrenal leukodystrophy, age-related macular degeneration Symptoms; dysplasia; aphasia; Aicardi syndr〇me; Alexander disease; Alpers' disease; crossed limb dysentery Alzheimer's disease (Alzheimer's disease) Vascular dementia; 156342.doc -18- 201143782 Amyotrophic lateral sclerosis; no brain malformation; angel syndrome (Angeiman) Syndrome), hemangiopathy; dystrophy; aphasia; apraxia; arachnoid cyst, arachnoiditis; An 〇 _ _ ( ( ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; 'Syndr〇me); ataxia telangiectasia; attention deficit with • motility disorder; autism; autonomic dysfunction; back pain; Batten disease, Beckett's disease Behcet, sdisease); BeU'S Palsy; benign essential sputum; benign local muscle atrophy; benign intracranial pressure, Binswanger disease (Binswanger, s disease); eyelid climbing; Brock-Suz B1〇ch Sulzberger syndrome; brachial plexus injury; brain abscess; brain injury; brain tumor (including glioblastoma multiforme); spinal cord tumor; Brown Sequard syndrome; Canavan disease; wrist pain syndrome; burning neuralgia; central pain syndrome; central medullary cerebral lysis, cerebral aneurysm; cerebral arteriosclerosis; cerebral atrophy; brain giant Cerebral palsy; Charcot-Marie-Tooth disease; chemotherapy-induced neuropathy and neuropathic pain; Chiad malformati〇n; chorea; chronic inflammatory degeneration Myelinating polyneuropathy; chronic pain; chronic regional pain syndrome group 'ke _ er 2 syndrome (to produce ^ L〇 Wry syndrome); coma 'contains persistent vegetative state; congenital facial paralysis; cortical basal degeneration' Gu arteritis, craniosynostosis; Creutzfeldt-Jakob dlSeaSe; accumulated traumatic disease; Cushing's syndrome; giant cell inclusion disease; cytomegalovirus infection; dance eye - 156342. Doc •19- 201143782 Dance foot syndrome; Dandy Walker syndrome; Dawson disease; De Morsier's syndrome; cloned Peck-Clump's syndrome (Dejerine- Klumke palsy); dementia; dermatomyositis; diabetic neuropathy; diffuse sclerosis; autonomic dysfunction; difficulty in writing; dyslexia; dystonia; early childhood epileptic brain ; empty sella syndrome; encephalitis; cerebral palsy; brain trigeminal ganglion tumor; epilepsy; Erb's palsy; idiopathic seismic; Fabry's disease; (Fahr's syndrome); fainting; familial spastic paralysis; febrile seizures; Fisher syndrome; Friedreich's ataxia; frontotemporal dementia and other r tau lesions Gaucher's disease; Gerstmann's syndrome; giant cell arteritis; giant cell inclusion disease; globular white matter dystrophy; Guillain-Barre Syndrome); HTLV-1 associated myelopathy; Hallervorden-Spatz disease; head injury; headache; hemifacial spasm; hereditary spastic paraplegia; hereditary polyneuritis-like ataxia; Herpes zoster; herpes zoster; Hirayama syndrome; HIV-associated dementia and neuropathy (and eight 11) neurological manifestations; forebrain non-cracking malformation; Huntington's disease (Huntington, s (10) )and Other polyglutamine indole repeat disease; water-free no brain malformation; hydrocephalus; hypercortisolism; hypoxia; immune-mediated encephalomyelitis; inclusion body myositis; 2 dysregulated disease; Disease; infant reefsum disease (four) coffee to refsum disease) · 'baby sputum; inflammatory myopathy; intracranial cyst; intracranial 156342.doc -20- 201143782 high pressure; Jubbert syndrome (Joubert syndrome); Keams-Sayre syndrome; Kennedy disease; Kinsboume syndrome; Klippel Feil syndrome; Krabbe disease Kugelberg-Welander disease; Kuru disease; Lafora disease; Lambert-Eaton myasthenic syndrome; Landa - Landau-Kleffner syndrome; lateral medullary (Wallenberg) syndrome; learning disability; Leigh's disease; Lennox-Gastau syndrome (Lennox-Gustaut Syndrome); Lai-naphthalene Lesch-Nyhan syndrome; leukodystrophy; Lewy body dementia; no cerebral gyrus; atresia syndrome; Lou Gehrig's disease (ie, motor neuron disease or Amyotrophic lateral sclerosis), lumbar disc disease; Lyme disease - neurological sequelae, Machado-Joseph disease; brain hypertrophy, giant brain; Mel-Rosenthal syndrome Menieres disease; Meningitis; Menkes disease; Metachromatic leukodystrophy; Microcephaly; Migraine; Miller Fisher syndrome ; small stroke; mitochondrial myopathy; Mobius syndrome; single limb muscle atrophy; motor neuron disease; abnormal vascular network disease of the brain; mucopolysaccharidosis; multiple infarct dementia; Focal motor neuropathy; multiple sclerosis and other demyelinating disorders; multiple systemic atrophy with positional hypotension; muscular dystrophy; myasthenia gravis; demyelinating diffuse sclerosis; infant muscle 156342.doc -21- 201143782 Clonic encephalopathy; myoclonus; myopathy; myotonia; narcolepsy; neurofibromatosis; neuroleptic malignant syndrome; neurological manifestations of AIDS; neurological sequelae of lupus; neuromuscular rigidity; neuronal waxy lipid Leptin; abnormal brain neuron migration; Niemann_pick disease; O'Sullivan-McLeod syndrome; occipital neuralgia; recessive spinal canal insufficiency sequence Ohtahara syndrome; cerebral atrophy of cerebral palsy; strabismic cerebral palsy; optic neuritis; orthostatic hypotension; overuse syndrome; paresthesia' neurodegenerative disease or disorder (Parkinsen's disease) 〇n, s disease), utton's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), dementia, multiple sclerosis and other diseases and conditions associated with neuronal cell death); congenital deputy Myotonia; paraneoplastic disease; paroxysmal episode, Pary R〇mberg (Pelizaeus-Merzbacher disease); periodic 瘫痪 'periphery Trans disease; 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; polio pruritus syndrome; postherpetic neuralgia; post-infectious encephalomyelitis; orthostatic hypotension; Prader-Willi syndrome; primary lateral sclerosis ; prion disease; progressive one-sided atrophy; progressive multifocal leukoencephalopathy; progressive sclerosing gray matter atrophy; progressive nucleus itching; pseudocephaloma; Ramsay-Hunt syndrome (Types I and II); Rasmussen's encephalitis; Reflex sympathetic dystrophy 156342.doc •22· 201143782 Syndrome; Refsum disease; Repetitive motor disorder; Reproducibility Compression injury; restless leg syndrome; retrovirus-associated myelopathy; Rett syndrome; Rey's syndrome; chorea (Saint Vitus dance) ); Sandhoff disease; Schilder's disease; cerebral palsy; clear septum optic nerve dysplasia; scare infant syndrome; herpes zoster; Shay-Dreig syndrome (Shy -Drager syndrome); Sjogren's syndrome; sleep apnea; Soto's syndrome; spasticity; spina bifida; spinal cord injury; spinal cord tumor; spinal muscular atrophy; stiff syndrome (Stiff-Person Syndrome; stroke; Sturge-Weber syndrome; subacute sclerosing panencephalitis; subcortical arteriosclerotic encephalopathy; Sydenham chorea; syncope; syringomyelia Delayed dyskinesia; Tay-Sachs disease; Temporal arteritis; Spinal cord syndrome; Thomsen disease; Thoracic outlet syndrome; Tic Douloureux; Todd Todd's paralysis; hyperactive slang syndrome; transient ischemic attack; disseminated spongiform encephalopathy; transverse myelitis; traumatic brain injury; Trigeminal neuralgia; tropical spastic paraplegia; tuberous sclerosis; vascular dementia (multiple infarct dementia); vasculitis, including temporal arteritis; Von Hippel-Lindau disease; Wallenberg's syndrome; Werdnig-Hoffman disease; West syndrome; cervical spine; Williams syndrome; Wilson 156342.doc -23- 201143782 Disease (Wildon's disease); and Zweiger syndrome syndrome) ° "A fluid disease or condition" contains diseases, conditions, or conditions that affect hematopoietic cells or tissues. Blood disorders include diseases, conditions, or conditions associated with abnormal blood levels or function. Examples of blood disorders include conditions derived from bone marrow irradiation or cancer chemotherapy, such as the following conditions: pernicious anemia, hemorrhagic anemia, hemolytic anemia, aplastic anemia, sickle cell anemia, iron granule anemia, Anemia associated with chronic infections (eg malaria, trypanosomiasis, HI V, hepatitis virus or other viruses), myeloid anemia caused by bone marrow loss, renal failure from anemia, anemia, polycythemia, infectious singles Nuclear cell hyperplasia (IM), acute non-lymphocytic leukemia (ANLL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), acute myelomonocytic leukemia (AMMoL), true erythrocytosis Disease, lymphoma, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia, wilms, s turn〇r, Ewing's sarcoma, retinoblastoma, hemophilia, and Symptoms associated with increased blood test risk, cytotherapy, thalassemia, and antibody-induced conditions (eg, transfusion reactions and erythroblastia) ) mechanical trauma of red blood cells (eg, microangiopathic hemolytic anemia, thrombotic thrombocytopenic purpura, and disseminated intravascular coagulation), infections caused by parasites (eg, protozoa), from, for example, lead poisoning Chemical damage, and spleen function. Lymphatic diseases include, but are not limited to, lymphadenitis, lymphatic vessel dilatation, lymphangitis, lymphedema, lymphocystosis, lymphoproliferative disorders, cutaneous mucosal lymph node syndrome, reticuloendothelial proliferation, spleen 156342.doc -24- 201143782 Disease, thymic hyperplasia, thymic tumor, tuberculosis, lymph nodes, pseudo lymphoma, and lymphoid abnormalities. Conditions of the lymphoid hematopoietic system include, but are not limited to, non-Hodgkin's lymphoma, chronic lymphocytic leukemia, and reactive lymphoid tissue hyperplasia. • Polynucleotide and Nucleotide Compositions and Molecules • Targets: In one embodiment, the target comprises a nucleic acid sequence of LIM homolog 2 (LHX2), including (without limitation) senses associated with LHX2 and/ Or antisense non-coding and / or coding sequences. The LIM-homology domain (LIM-HD) protein regulates the tissue specificity of the target gene and is required for the normal development and maintenance of many organs. During embryonic development of vertebrates and invertebrates, members of the LIM homology domain transcription factor family play an important role by controlling processes such as asymmetric cell division, tissue specificity, and differentiation of specific cell types. The member of this family, Lhx2, is particularly interesting based on its function in the development of several different tissues. Its function is achieved through interstitial-epithelial interactions and regulation of stem/Q progenitor cells. A member of this family of Lhx2 lineage transcription factors, which consists of 12 known genes, which are structurally characterized by two amino-terminal zinc finger motifs (LIM domains) and a carboxy-terminal homology domain. Inactivation of Lhx2 in mice results in a decrease in cerebral cortex size and abnormal development of the striate, telencephalon, olfactory system, and dermal pattern formation.

Lhx2亦為正常四肢及肝形成所需;Lhx2-/-小鼠會發生 肝纖維化且紅細胞生成不完全,從而在出生前因嚴重貧血 而導致死亡。Lhx2表現之已知位點與該等表現型極其吻 156342.doc •25- 201143782 合。Lhx2表現於成為肝組成部分之肝相關原始橫隔間充質 中,且在肝發育期間可維持其表現直至肝星形細胞之成熟 體階段為止。Lhx2-/-胚胎顯示已顯示於E1〇5處之肝的尺 寸有所減小,此表明Lhx2為胎兒肝之擴增所需。突變體表 現型係源於存在活化肝星形細胞從而產生含有表現型異常 内胚層細胞之纖維變性及分裂肝。Lhx2_/_小鼠之肝中之間 質缺陷會引起具有細胞非自主性之致命貧血,此乃因 Lhx2-/-造血細胞表現為正常,此表明突變體微環境不能支 持造血發育。該等發現表明,肝星形細胞中之Lhx2表現參 與間質-上皮細胞相互作用,該等相互作用對於胎兒肝中 造血微環境之肝擴增、組織、分化及形成較為重要。 在一實施例中,使用反義寡核苷酸來預防或治療與 LHX2家族成貢有關之疾病或病症。可使用自利用反義化 合物所獲得幹細胞再生之細胞/組織治療之實例性類um同 源序列2(LHX2)介導的疾病及病㈣括:與匕肋之異常功 月b及/或表現有關之疾病或病症、血液疾病或病症、毛髮 生長受損、神經疾病或病症、視網膜疾病或病症、與免疫 ί·生又扣有關之疾病或病症、垂體疾病或病症、肝疾病或病 症、嗅覺“錢或病症、紅細胞生成受損、造&疾病或 病症月趟增生病及脊椎肢體突長。 貫%例中,對有需要之患者實施藉由一或多種反義 寡核苷酸來調節LHX2,從而預防或治療與LHX2異常表 見功、活性(與正常對照組相比)有關的任—疾病或病 症。 156342.doc -26 - 201143782 在一實施例中,募核苷酸對LHX2之聚核苷酸具有特異 性’其包含(不限於)非編碼區域。LHX2靶包括LHX2之變 體;LHX2之突變體,包含SNp ; LHX2i非編碼序列;等 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LHX2聚核 苷酸而是擴展至LHX2之任一同功型、受體、同系物、非 編碼區域及諸如此類。 〇 在一實施例中,寡核苷酸靶向LHX2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’募核苷酸係反義RNA或DNA分子。 在一貫施例中’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 Q 普、鳥普、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,反義化合物及靶之間之同源性、序列 致性或互補性為約5 〇 %至約6 〇 %。在一些實施例中,同 源性、序列一致性或互補性為約6〇%至約7〇%。在一些實 施例中’同源性、序列一致性或互補性為約7〇%至約 80%。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 156342.doc -27- 201143782 或互補性為約90%、約92%、約94%、約95°/。、約96%、約 97%、約 98%、約 99%或約 100%。 在以下情形時反義化合物係可特異性雜交:化合物與乾 核酸之結合可干擾靶核酸之正常功能而導致活性損失,且 存在足夠互補性程度以避免反義化合物與非靶核酸序列在 期望發生特異性結合之條件下發生非特異性結合。該等條 件包含(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:該化合物與靶 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LHX2(包含(不限於)使用(例 如)PCR、雜交等鑑別及擴展之反義序列、如SEQ ID NO: 2 至4所示之序列之一或多者及諸如此類)可調節LHX2之表 現或功能。在一實施例中,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,寡核苷酸包括如SEQ ID NO: 5至16所示 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。經修飾鍵或核苷 156342.doc -28 - 201143782 二硫代磷酸s旨或諸如此 衍生物。可附接至本發 酸間鍵聯之實例包括硫代蹲酸醋、二硫代^ 類。在—實施例中,核苷酸包括磷衍生物。 明紅修錦寡核I酸中之糖或糖類似物部分之磷衍生物(或 經修飾磷酸酯基團)可為單磷酸酯、二磷酸酯、三磷酸 西曰椒駚烷基酯、烷磷酸酯、硫代磷酸酯及諸如此類。上 巧酸0旨類似物之製備、及其進人核普酸、經修飾核苦酸 及寡核苦酸中之方法本身亦已知且無需閣述於本文中。 ❹ v等•為i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。反義寡核芽酸已安全且有效地 投與人類且當前正實施許多臨床試驗。由此確定,募核苷 酸可為可經設置用於治療細胞、組織及動物(尤其人類 治療方案中的有用治療方式。 在本發明實施例中,寡聚反義化合物、尤其募核苦酸會 結合至靶核酸分子並調節由靶基因編碼之分子的表現及/ 〇 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 譯位點之移位、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。 可端視期望功能來上調或抑制功能。 化兮物。因此,該等 或環狀募聚化合物形 反義化合物包含反義寡聚化合物、反義募核苦酸 '外部 引導序列(EGS)寡核苷酸、交替剪接、引物、探針、及其 他與靶核酸之至少一部分雜交的募聚化合物。 =Lhx2 is also required for normal limbs and liver formation; Lhx2-/- mice develop liver fibrosis and incomplete erythropoiesis, leading to death from severe anemia before birth. The known locus of Lhx2 expression is extremely close to these phenotypes 156342.doc •25- 201143782. Lhx2 is expressed in the liver-associated primitive transverse mesenchyme that is part of the liver and maintains its performance during liver development until the mature stage of hepatic stellate cells. The Lhx2-/- embryo showed a decrease in the size of the liver that had been shown at E1〇5, indicating that Lhx2 is required for fetal liver expansion. Mutant expression is derived from the presence of activated hepatic stellate cells to produce fibrotic and schizophrenic cells containing phenotypically abnormal endoderm cells. The interstitial defects in the liver of Lhx2_/_ mice cause fatal anemia with cell involuntaryness, as Lhx2-/- hematopoietic cells appear normal, suggesting that the mutant microenvironment does not support hematopoietic development. These findings indicate that Lhx2 in hepatic stellate cells interacts with mesenchymal-epithelial cells, and these interactions are important for liver expansion, organization, differentiation, and formation of hematopoietic microenvironments in fetal liver. In one embodiment, an antisense oligonucleotide is used to prevent or treat a disease or condition associated with a tribute to the LHX2 family. An example um homologous sequence 2 (LHX2) mediated disease and disease mediated by stem cell regeneration obtained from an antisense compound can be used (4): associated with an abnormal uterine b and/or performance of the rib Disease or condition, blood disease or condition, impaired hair growth, neurological disease or condition, retinal disease or condition, disease or condition associated with immunization, pituitary disease or condition, liver disease or condition, smell " Money or illness, impaired erythropoiesis, creation & disease or condition, meningococcal disease and vertebral limb length. In % of cases, one or more antisense oligonucleotides are used to regulate LHX2 in patients in need , thereby preventing or treating any disease or condition associated with the LHX2 abnormality table, activity (compared to the normal control group). 156342.doc -26 - 201143782 In one embodiment, the nucleotides are gathered for LHX2 Nucleotides are specific 'which include, without limitation, non-coding regions. LHX2 targets include variants of LHX2; mutants of LHX2, including SNp; LHX2i non-coding sequences; alleles, fragments and the like. 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 LHX2 polynucleotide but extends to any isoform, receptor, homologue, non-LHX2 A coding region and the like. In one embodiment, the oligonucleotide targets a natural antisense sequence (a natural sequence encoding a non-coding region) of the LHX2 target, including, without limitation, variants, alleles thereof, Homologs, mutants, derivatives, fragments and complementary sequences. Preferably, the nucleotides are antisense RNA or DNA molecules. In a consistent application, the oligomeric compound of the invention is also included in the compound. Or a variant of a different base at a plurality of nucleotide positions. For example, if the first nucleotide is adenine, it may be produced at this position containing a chest, a bird, a cytidine or other natural or non- Variants of natural nucleotides. This can occur anywhere in the antisense compound. These compounds are then tested using the methods described herein to determine their ability to inhibit the performance of a target nucleic acid. In some embodiments, antisense compounds and The same between targets The sex, sequence or complementarity is from about 5% to about 6%. In some embodiments, the homology, sequence identity or complementarity is from about 6% to about 7%. In some embodiments 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 In some embodiments, homology, sequence identity 156342.doc -27- 201143782 or complementarity is about 90%, about 92%, about 94%, about 95°/, about 96%, about 97%, about 98%, about 99% or about 100%. The antisense compound can specifically hybridize in the following cases: the binding of the compound to the dry nucleic acid can interfere with the normal function of the target nucleic acid resulting in loss of activity, and there is sufficient complementarity to avoid Non-specific binding of the antisense compound to the non-target nucleic acid sequence under conditions where it is desired to specifically bind. Such conditions include (i.e., physiological conditions in the case of in vivo analysis or therapeutic treatment, and conditions under which the analysis is performed in the context of in vitro analysis. An antisense compound (whether DNA, RNA, chimeric compound, or substituted compound, etc.) can specifically hybridize in the following situations: binding of the compound to a 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 the case of in vivo analysis or therapeutic treatment, or in vivo) Non-specific binding occurs under conditions in which the analysis is performed in the case of an external analysis. In one embodiment, targeting LHX2 (including, without limitation, antisense sequences identified and expanded using, for example, PCR, hybridization, etc., one or more of the sequences set forth in SEQ ID NOs: 2 to 4, and the like ) can adjust the performance or function of LHX2. In one embodiment, the performance or function is up-regulated compared to the control group. In one embodiment, the performance or function is downregulated as compared to a control group. In one embodiment, the oligonucleotide comprises a nucleic acid sequence as set forth in SEQ ID NOS: 5 to 16, comprising an antisense sequence identified and expanded using, for example, PCR, hybridization, and the like. Such oligonucleotides can include one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. Modified bond or nucleoside 156342.doc -28 - 201143782 dithiophosphoric acid or such a derivative. Examples of linkages that can be attached to the present acid include thiophthalic acid vinegar, dithiophenes. In an embodiment, the nucleotide comprises a phosphorus derivative. The phosphorus derivative (or modified phosphate group) of the sugar or sugar analog moiety of the bright red nucleoside I acid may be a monophosphate, a diphosphate, a sulfonium triphosphate, an alkane Phosphates, phosphorothioates, and the like. The preparation of the analogs of the above-mentioned analogs, as well as the methods of their introduction into nucleotides, modified nucleotides and oligonucleotides, are also known per se and need not be described herein. ❹ v et al. For this technology, the specificity and sensitivity of antisense oligonucleotides are also utilized 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 is thus determined that the raised nucleotides can be a useful therapeutic modality that can be used to treat cells, tissues, and animals, particularly in human therapeutic regimens. In embodiments of the invention, oligomeric antisense compounds, particularly nucleus Binding to a target nucleic acid molecule and modulating the expression and/or function of the molecule encoded by the target gene. The DNA function to be interfered with includes, for example, replication and transcription, and the RNA function to be interfered with includes all important functions, such as RNA to protein. Translocation of translation sites, translation of proteins from RNA, splicing of one or more mRNA species by Rna, and catalytic activity that RNA can participate in or promote. Up-regulation or inhibition of function can be performed depending on the desired function. , or a cyclic polymeric compound antisense compound comprising an antisense oligomeric compound, an antisense scorpion acid 'external leader sequence (EGS) oligonucleotide, an alternative splicing, a primer, a probe, and others A polymeric compound that hybridizes to at least a portion of a nucleic acid.

156342.doc -29- 201143782 式引入。 在本發明之上下文中’反義化合物針對特定核酸分子之 靶向可為多步過程。該過程經常始於鑑別所調節功能之乾 核酸。舉例而言’此乾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 原之核酸分子。在本發明巾,㈣酸編竭um同源序列2 (LHX2)。 乾向過程經常亦包含測定把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)的至少一個靶區域、區 段、或位點。在本發明之上下文内,術語「區域」定義為 靶核酸中具有至少一個可鏗別結構、功能、或特性的一部 分。靶核酸之區域内具有區段。「區段」定義為靶核酸内 區域之較小或子部分。本發明所用之「位點」定義為乾核 酸内之位置。 在一實施例中,反義寡核苷酸結合至UM同源序列 2(LHX2)之天然反義序列並調節LHX2 (SEq m N〇: u之表 現及/或功能。反義序列之實例包含沾〇 ID N〇: 2至16。 在一實施例中’反義寡核苷酸結合至LIM同源序列2 (LHX2)聚核苷酸之一或多個區段並調節LHX2i表現及/或 功月b。區#又包括LHX2有義或反義聚核苦酸之至少5個連續 核苷酸。 在一實施例中,反義寡核苷酸對LHX2之天然反義序列 具有特異性,其中募核苷酸與LHX2之天然反義序列之結 合可調節LHX2的表現及/或功能。 156342.doc •30· 201143782 在一實施例中,寡核苷酸化合物包括如SEQ ID NO: 5至 16所示之序列、使用(例如)pcR、雜交等鑑別及擴展之反 義序列。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。經修飾鍵或核苷酸 間鍵聯之實例包括硫代磷酸酯、二硫代礙酸酯或諸如此 類。在一實施例中,核苷酸包括磷衍生物。可附接至本發156342.doc -29- 201143782 Introduction. The targeting of an antisense compound to a particular nucleic acid molecule can be a multi-step process in the context of the present invention. This process often begins with the identification of the dry nucleic acid of the regulated function. For example, the dry nucleic acid can be a nucleic acid molecule that exhibits a cellular gene (or mRNA transcribed from the gene) associated with a particular disorder or disease state or from an infectious agent. In the present invention, (iv) acid-encoded um homologous sequence 2 (LHX2). The dry-direction process also often includes the determination of at least one target region, region, or site that produces an antisense interaction within the nucleic acid to produce a desired effect (e.g., modulate expression). Within the context of the present invention, the term "region" is defined as a portion of a target nucleic acid having at least one distinguishable structure, function, or property. There is a segment within the region of the target nucleic acid. A "segment" is defined as a small or sub-portion of a region within a 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 oligonucleotide binds to the natural antisense sequence of UM homolog 2 (LHX2) and regulates the expression and/or function of LHX2 (SEq m N〇: u. Examples of antisense sequences include ID N〇: 2 to 16. In one embodiment, the 'antisense oligonucleotide binds to one or more segments of the LIM homolog 2 (LHX2) polynucleotide and modulates LHX2i expression and/or功月 b.区# further comprises at least 5 contiguous nucleotides of LHX2 sense or antisense polynucleic acid. In one embodiment, the antisense oligonucleotide is specific for the natural antisense sequence of LHX2, The binding of the raised nucleotide to the natural antisense sequence of LHX2 can modulate the expression and/or function of LHX2. 156342.doc • 30· 201143782 In one embodiment, the oligonucleotide compound comprises SEQ ID NO: 5 to The sequence of 16 is identified and expanded using, for example, pcR, hybridization, etc. The oligonucleotides may include one or more modified nucleotides, shorter or longer fragments, modified keys. And the like. Examples of modified bonds or internucleotide linkages include phosphorothioates, dithiocarbamate or the like. In one embodiment, the nucleotide comprises a phosphorus derivative. It can be attached to the hair

明經修飾募核苷酸中之糖或糖類似物部分之磷衍生物(或 經修飾磷酸酯基團)可為單磷酸酯、二磷酸酯、三罐酸 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、及其在核苷酸、經修飾核苷酸及 寡核苷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5I_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「AUG起始 禮馬子」、數基因.具有轉譯起動密碼子,其具有rn a序 列 5,-GUG、5i_UUG5l5i_CUG;且已顯 ^_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-情形中通常係甲硫胺酸(在真核生物中)或甲酿甲硫胺 酸(在原核生物中)’術語「轉譯起動密碼子」及「起始密 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,1中之任一者 可優先用於在特定細胞類型或組、織中或在特定條件组下之 轉譯起動。在本發明之上下文中,「起始密碼子」及「轉 潭起動密碼子」健用於在㈣^動自編碼L糊源序 156342.doc -31- 201143782 列2(LHX2)之基因轉錄之mRNA之轉譯的一或多個密碼 子’不管該等密碼子之序列如何。基因之轉譯終止密碼子 (或「停止密碼子」)可具有三種序列(亦即,5, Uaa、 5’-UAG及 5’-UGA,相應 DNA序列分別係 5,-TAA、5,-TAg 及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」係 指此一 mRNA或基因中在來自轉譯起動密碼子之任—方向 (亦即,5·或3,)涵蓋約25至約5〇個鄰近核苷酸的一部分。同 樣,術語「停止密碼子區域」及「轉譯終止密碼子區域」 係指此一 mRNA或基因中在來自轉譯終止密碼子之任—方 向(亦即,5’或3’)涵蓋約25至約5〇個鄰近核苷酸的一部分。 因此,「起始密碼子區域」(或「轉譯起動密碼子區域」)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」係指轉譯 起動密碼子及轉譯終止密碼子之間之區域,其亦係可有效 靶向之區域。在本發明之上下文内,靶向區域係涵蓋基因 之開放讀碼框(ORF)之轉譯起動或終止密碼子的基因内區 域。 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’加帽位點及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 156342.doc -32- 201143782 向上之mRNA部分’且由此包含mRNA之轉譯終止密碼子 及3'端間之核苷酸(或基因上之相應核苷酸)。mRNA之5'加 帽位點包括經由5’-5'三磷酸酯鍵聯接合至mRNA之5,-最殘 基之N7 -曱基化鳥苷殘基。mRNA之5 ’加帽區域視為包含5' 加帽結構本身以及與加帽位點相鄰之前50個核苷酸。本發 明之另一靶區域係5·加帽區域。 儘管一些真核生物mRNA轉錄本可直接轉譯,但許多真 核生物mRNA轉錄本含有一或多個在轉譯之前自轉錄本切 割且稱為「内含子」的區域。剩餘(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。在一實施 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乾位點之另一實施例。經由自不同基 因源剪接兩個(或更多)mRNA之過程產生之mRNA轉錄本稱 為「融合轉錄本」。可使用輕向(例如)DNA或mRNA前體之 反義化合物來有效契*向内含子。 在一實施例中’反義募核苷酸結合至乾聚核苷酸之編碼 及/或非編碼區域並調節乾分子之表現及/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,反義寡核苷酸結合至有義聚核苷酸並調 節靶分子之表現及/或功能。 替代性RNA轉錄本可自DNA之相同基因組區域產生。該 156342.doc •33· 201143782 等替代性轉錄本通稱為「變體」。更特定而言,「mRNA前 體變體」係自相同基因組DNA產生之轉錄本,其與自相同 基因組DNA產生之其他轉錄本在起始或停止位置方面有所 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前體及mRNA可具有一個以上之起始密碼子或停止 密碼子。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本稱為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「P〇lyA變體」,其中所產生之多個轉錄本源自轉 錄機對於「P〇lyA停止信號」中之一者之替代性選擇,由 此產生在唯一 P〇lyA位點終止之轉錄本。在本發明之上下 文内,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至少定義為靶區域中 由活性反義化合物乾向之長5個核苦酸的部分。 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 156342.doc • 34· 201143782 明範圍内之特定實施例。熟習此項技術者根據本揭示内容 可容易地鑑別其他起區段β 人們認為長度為5-1 00個核苷酸且包括一段至少五(5)個 選自較佳闡釋性靶區段之連續核苷酸之靶區段亦適於靶 向0 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 苷酸係同一 DNA或RNA中之連續的一段,其緊接輕區段之 5·-末端上游開始並持續至該〇ΝΑ或RNA含有約5至約1〇〇個 核苷酸為止)。同樣,較佳靶區段表示為DNA或RNA序 列,其包括至少5個來自較佳闡释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100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 佳靶區段。 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,寡核苷酸結合至特定靶之反義股。 募核苷酸之長度為至少5個核苷酸且可經合成以使每一寡 核皆酸皆㈣重疊序列,從而寡核純經合成以覆蓋把聚 核芽酸之整個長度4亦包含編碼m纟扁碼區域。 在一實施財,反義寡核㈣較錄向特異性核酸。反 156342.doc -35- 201143782 義化合物至特定核酸之靶向係多步過程。該過程經常始於 鑑別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、 反義轉錄本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 之聚腺苷酸化mRNA。最近,顯示非聚腺苷酸化核RNA之 組可能極大,且許多該等轉錄本源自所謂的基因間區域。 ncRNA可調控基因表現之機制係與把轉錄本之鹼基配對。 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 鏈處編碼且由此顯示與其靶之完全互補性;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,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有義信使RNA之表現。然而,此調 控可不協調(反義弱化(knockdown)造成信使RNA增加)或協 調(反義弱化造成伴隨信使RNA減少)。在該等情形下,反 156342.doc •36- 201143782 義寡核苷酸可靶向反義轉錄本之重疊或非重疊部分以產生 弱化或隔絕(sequestration)。可以相同方式靶向編碼以及非 編碼反義轉錄本,且任一種類皆能以協調或不協調方式調 控相應有義轉錄本。在鑑別針對靶使用之新寡核苷酸時採 - 用的策略可基於藉由反義寡核苷酸來弱化反義RNA轉錄本 或調節期望乾的任一其他方式。 農略i :在不協調調控之情形下,弱化反義轉錄本會增 0 加習用(有義)基因之表現。若後一基因編碼已知或假定藥 物靶,則可設想弱化其反義對等部分來模擬受體激動劑或 酶刺激物之作用。 農略在協調調控之情形下,可同時弱化反義及有義 轉錄本且由此協同性降低習知(有義)基因表現。舉例而 曰,右使用反義募核苷酸來達成弱化,則可使用此策略來 應用一種靶向有義轉錄本之反義寡核苷酸及靶向相應反義 轉錄本之另一反義寡核苷酸、或同時靶向重疊有義及反義 〇 轉錄本之單一能量對稱性反義寡核苷酸。 根據本發明,反義化合物包含反義寡核苷酸、核糖酶、 外部引導序列(EGS)寡核苷酸、siRNA化合物、單-或雙鏈 RNA干擾(RNAi)化合物(例如siRNA化合物)、及與乾核酸 之至少一部分雜交並調節其功能之其他募聚化合物。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 鍵雙鏈、環狀或髮夾募聚化合物,且可含有諸如内部或 末^膨服、失配或環路等結構要素。反義化合物通常製成 156342.doc •37· 201143782 直鏈n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含諸如以下構築體: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 I· '進行雜交並&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 髮夾。髮夾型結構可在M3’末端含有懸垂 P刀^延長單鏈特徵。雙鏈化合物視需要可在末端包含懸 垂部刀λ他修飾可包含附接至一個末端、所選核苦酸位 置、糖位置或附接至—個核芽間鍵聯之偶聯基團。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 邛刀雙鏈可藉由在轉基因細胞系中穩定表現dsRNA髮夾 5L來特異性調@基因表現,然而,在_些實施例中,基因 表現或功此經上調。在自兩條鏈、或呈自體互補髮夾型分 子开v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Wats〇n_Crick方式鹼基配對 之互補RNA鏈。 引入系統中之後,本發明化合物可引發一或多種酶或結 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(包含寡核皆 酸)可闡述為「類DNA」(亦即,通常具有一或多個2,_去氧 糖及(通* )T而非u鹼基)或「類rnA」(亦即,通常具有一 或多個2’-羥基或2,_修飾糖及(通常}υ而非τ鹼基)。核酸螺 156342.doc -38- 201143782 旋可採用一種以上之結構類型,最通常係A型及B型。據 信,一般而言,具有B型樣結構之募核苷酸係「類DNA」 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);或短髮夾型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可誘導相 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。當前尚未暸 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 寡核苷酸可增加LIM同源序列2(LHX2)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,藉由把向基因啟動子中之 序列,saRNA可誘導靶基因表現,此現象稱為dsRNA誘導 156342.doc -39- 201143782 之轉錄活化(RNAa)。 在另一實施例中,本文所鑑別之「較佳乾區段」可用於 篩選調節LIM同源序列2(LHX2)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 LHX2之核酸分子之表現’且至少包括與較佳把區段互補 之5-核苷酸部分。篩選方法包括以下步驟:使編碼lhx2 之有義或天然反義聚核苷酸之核酸分子的較佳把區段與一 或多種候選調節劑接觸,且選擇一或多種可降低或增加編 碼LHX2聚核苷酸之核酸分子(例如SEq m N〇: 5至16)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LHX2聚核苷酸之核酸分子的表現, 則該調節劑可用於LHX2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。 靶向天然反義序列較佳地可調節靶基因之功能。例如, LHX2基因(例如,登錄號為NM_0(M789)。在一實施例中, 乾係LHX2基因之反義聚核苦酸。在一實施例中,反義寡 核苷酸靶向LHX2聚核苷酸(例如,登錄號為 _004789)之 有義及/或天然反義序列、其變體、等位基因、同功型、 同系物、犬變體、街生物、片段及互補序列。較佳地,募 核苷酸係反義分子且靶包含反義及/或有義[Ηχ2聚核苷酸 之編碼及非編碼區域。 本發明之較佳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)寡核普酸。 業内已顯不’ 3亥等雙鏈寡核苷酸部分可調節耙表現並經 156342.doc •40- 201143782 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 體之反義股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LIM同源序列2 (LHX2)聚核苷酸(例如,登錄號為NM_004789))、其變 體、等位基因、同功型、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LHX2而是 擴展至LHX2分子之任一同功型、受體、同系物及諸如此 類。 在一實施例中,寡核苷酸靶向LHX2聚核苷酸之天然反 義序列(例如,如SEQ ID NO: 2至4所示之聚核苷酸)、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。反義寡核苷酸之實例係如SEQ ID NO: 5至16所 示。 在一實施例中,寡核苷酸與LHX2反義分子之核酸序列 (包含(不限於)與LHX2聚核苷酸有關之非編碼有義及/或反 義序列)互補或結合並調節LHX2分子之表現及/或功能。 在一實施例中,寡核苷酸與LHX2天然反義分子之核酸 序列(如SEQ ID NO: 2至4所示)互補或結合並調節LHX2分 子之表現及/或功能。 在一實施例中,募核苷酸包括SEQ ID NO: 5至16中至少 5個連續核苷酸之序列並調節LHX2分子之表現及/或功 156342.doc -41 - 201143782 聚核苷酸靶包括LHX2(包含其家族成員)、LHX2之變 體;LHX2之突變體,包含SNP ; LHX2之非編碼序列; LHX2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LHX2聚核苷酸之寡核苷酸包括: 反義RNA、干擾RNA (RNAi)、短干擾RNA (siRNA);微小 干擾RNA (miRNA);小時序rna (stRNA);或短髮夾型 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LIM同源序列2 (LHX2)聚核苷酸(例 如SEQ ID NO : 2至16)可調節該等靶之表現或功能。在一 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,反義化合物包括如SEQ ID NO: 5至16所 示之序列。該等募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: 5至16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寡核苷 酸。 表1 ·· 序列編號 反義序列名 稱 序列 SEQ ID NO: 5 CUR-1560 (^*T*(^*(^*(^*T*G*G*G*As|iC!*T*G*T*A*A*G*G*A SEQ ID NO: 6 CUR-1561 156342.doc •42· 201143782The phosphorus derivative (or modified phosphate group) of the sugar or sugar analog moiety in the modified modified nucleotide may be a monophosphate, a diphosphate, a trican acid ester, an alkyl phosphate, an alkanoic acid. Esters, phosphorothioates, and the like. The preparation of the above-described phosphate vinegar 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 5I_AUG (in the transcriptional mRNA molecule; in the corresponding DNA molecule, 5, _ATG), the translation initiation codon is also called "AUG codon", "start codon" Or "AUG starter horse", number gene. Has a translation start codon, which has rn a sequence 5, -GUG, 5i_UUG5l5i_CUG; and has been shown ^_AUA, 5, _acg and 5, -CUG can be played in vivo effect. Therefore, even if the starting amino acid is in each case, it is usually methionine (in eukaryotes) or methionine (in prokaryotes), the term "translation start codon" and "starting" Codons can also cover a variety of codon sequences. Eukaryotic and prokaryotic genes may have two or more alternative initiation codons, one of which may be preferentially used for translational initiation in a particular cell type or group, in a tissue, or under a particular set of conditions. . In the context of the present invention, the "starting codon" and "turning-start codon" are used to transcribe the gene in the (4)-moving self-encoding L paste source sequence 156342.doc -31- 201143782 column 2 (LHX2). One or more codons of the translation of the mRNA 'regardless of the sequence of the codons. The gene translation stop codon (or "stop codon") can have three sequences (ie, 5, Uaa, 5'-UAG, and 5'-UGA, and the corresponding DNA sequences are 5, -TAA, 5, -TAg, respectively). And one of the 5'-TGA). The terms "start codon region" and "translation initiation codon region" refer to any mRNA or gene in the direction from the translation initiation codon (ie, 5 or 3) covering from about 25 to about 5 A portion of a neighboring nucleotide. Similarly, the terms "stop codon region" and "translatation stop codon region" refer to the orientation or orientation (ie, 5' or 3') of the mRNA or gene from the translation stop codon that covers about 25 to about A portion of 5 adjacent nucleotides. Therefore, the "start codon region" (or "translation start codon region") and the "stop codon region" (or "transition stop codon region") are all regions in which the antisense compound of the present invention can effectively be used. An open reading frame (〇RF) or "encoding region" known in the art refers to the region between the translation start codon and the translation stop codon, which is also an area that can be effectively targeted. Within the context of the present invention, the targeting region encompasses the intragenic region of the translational start or stop codon of the open reading frame (ORF) of the gene. Another target region comprises the 5, non-translated region (5, utr) known in the art, which refers to the portion of the mRNA from the direction 5 of the translation initiation codon, and thus comprises the 5' capping site of the mRNA. And translating the nucleotides (or corresponding nucleotides in the gene) between the initiation codons. Another target region comprises the 3, non-translated region (3'UTR) known in the art, which refers to the mRNA portion from the translation stop codon 3, 156 342. doc -32 - 201143782 up and thus contains mRNA Translate the stop codon and the nucleotide between the 3' ends (or the corresponding nucleotide on the gene). The 5' capping site of the mRNA includes a 5'-most residue N7-mercaptoguanosine residue joined to the mRNA via a 5'-5' triphosphate linkage. The 5' capped region of the mRNA is considered to comprise the 5' capping structure itself and 50 nucleotides immediately adjacent to the capping site. Another target area of the present invention is a 5. capping area. Although some eukaryotic mRNA transcripts are directly translatable, many eukaryotic mRNA transcripts contain one or more regions that are transcribed and translated as "introns" prior to translation. The remaining (and thus translated) regions are referred to as "exons" and are spliced together to form a continuous 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 for excessive splicing products Produce a situation related to the disease. Another embodiment of an abnormal fusion node that is due to rearrangement or loss is a dry site. An mRNA transcript produced by splicing two (or more) mRNAs from different gene sources is referred to as a "fusion transcript." An antisense compound that is light, for example, to a DNA or mRNA precursor, can be used to efficiently bind to an intron. In one embodiment, the antisense raised nucleotide binds to the coding and/or non-coding region of the dry polynucleotide and modulates the performance and/or function of the dry 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 oligonucleotide binds to a sense polynucleotide and modulates the expression and/or function of the target molecule. Alternative RNA transcripts can be produced from the same genomic region of DNA. Alternative transcripts such as 156342.doc •33· 201143782 are commonly referred to as “variants”. More specifically, "mRNA precursor variants" are transcripts produced from the same genomic DNA that differ from the other transcripts produced from the same genomic DNA in terms of starting or stopping positions and contain introns and Proton sequence. 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 precursor and mRNA may have more than one start codon or stop codon. Variants derived from mRNA precursors or mRNAs using alternative initiation codons are referred to as "alternative initiation variants" of mRNA precursors or mRNA. Their transcripts using alternative stop codons are referred to as "alternative stop variants" of mRNA precursors or mRNA. A specific type of alternative stop variant is a "P〇lyA variant" in which multiple transcripts are derived from an alternative to the transcription machine for one of the "P〇lyA stop signals", resulting in A transcript that terminates at the unique P〇lyA site. In the context of the present invention, the types of variants described herein are also examples of target nucleic acids. The position at which the target nucleic acid hybridizes to the antisense compound is defined at least as a portion of the target region which is 5 nucleotides long 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 elucidate specific implementations within the scope of the disclosure 156342.doc • 34 201143782. example. Those skilled in the art can readily identify other segments 7 according to the present disclosure that are considered to be 5-1 00 nucleotides in length and include a segment of at least five (5) consecutive segments selected from preferred interpretive target segments. The target segment of the nucleotide is also suitable for targeting 0. The target segment may comprise a DNA or RNA sequence comprising at least 5 contiguous nucleotides from the 5 terminus of one of the preferred interpretable target segments (remaining nucleoside The acid is a contiguous stretch of the same DNA or RNA that begins immediately upstream of the 5'-end of the light segment and continues until the sputum or RNA contains from about 5 to about 1 nucleotide. Likewise, preferred target segments are represented as DNA or RNA sequences comprising at least 5 contiguous nucleotides from the 3, _ terminus of one of the preferred interpretable target segments (remaining nucleotides are the same DNA) Or a continuous segment of the RNA that begins immediately downstream of the 3, _ terminal of the target segment and continues until the DNA or RNA contains from about 5 to about 100 nucleotides. The skilled artisan can identify other preferred target segments without undue experimentation. After identifying one or more target regions, segments or sites, the target can be selected to be sufficiently complementary to the target (ie, the hybridization is sufficiently sufficient and sufficiently specific). Antisense compounds to achieve the desired effect. In an embodiment of the invention, the oligonucleotide binds to an antisense strand of a particular target. The nucleotides are at least 5 nucleotides in length and can be synthesized to make each oligo Nucleic acid (4) overlapping sequences, so that the oligonuclear pure synthesis to cover the entire length of the polynuclear acid 4 also contains the m纟 flat code region. In one implementation, the antisense oligo (4) is more specific to the specific nucleic acid 156342.doc -35- 201143782 Sense compound to specific nucleic acid target A multi-step process that often begins with the identification of a nucleic acid sequence that regulates function. This can, for example, be a cellular gene (or mRNA transcribed from a gene) associated with a particular disorder or disease state, or a non-coding polynucleoside. Acids (eg, non-coding RNA (ncRNA)) RNA can be classified into (1) messenger RNA (mRNA), which translates into protein; and (2) non-protein-encoding RNA (ncRNA). ncRNA includes microRNA, 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 in the 3' non-translated region of the locus encoding the protein (3 The ntRNA is usually rare and at least half of the ncRNAs sequenced by the FANTOM Alliance do not appear to be polyadenylated. Most researchers focus on processing and export to the cytoplasm for obvious reasons. Polyadenylation of mRNA. Recently, the group showing non-polyadenylated nuclear RNA may be extremely large, and many of these transcripts are derived from so-called intergenic regions. ncRNA regulates the mechanism of gene expression and transcripts Base pairing. RNAs that function by base pairing can be grouped into: (1) cis-encoded RNA, which encodes at the same genetic position in which it functions, but encodes at the opposite strand of RNA and thus displays its target Complete complementarity; and (2) trans-encoded RNA, which encodes at a different chromosomal location than the RNA it functions with and generally does not exhibit complete base-pairing potential with its target. Unexpectedly limited by theory, this article Interference with an antisense oligonucleotide against an antisense polynucleotide can alter the expression of the corresponding sense messenger RNA. However, this regulation can be uncoordinated (an increase in messenger RNA caused by knockdown) or coordination (antisense weakening) Causes a decrease in accompanying messenger RNA). In such cases, the anti-156342.doc •36-201143782 sense oligonucleotide can target overlapping or non-overlapping portions of the antisense transcript to create a weakening or sequestration. Both coding and non-coding antisense transcripts can be targeted in the same manner, and any species can modulate the corresponding sense transcript in a coordinated or uncoordinated manner. The strategy employed in identifying new oligonucleotides for use with a target can be based on any other way of attenuating the antisense RNA transcript by antisense oligonucleotides or modulating the desired stem. Nongluo i: In the case of uncoordinated regulation, weakening antisense transcripts will increase the performance of the learned (sense) gene. If the latter gene encodes a known or putative drug target, it is conceivable to attenuate its antisense equivalent to mimic the effects of the receptor agonist or enzyme stimulator. In the case of coordinated regulation, the agricultural strategy can simultaneously weaken the antisense and sense transcripts and thereby reduce the conventional (sense) gene expression. For example, if the anti-sense nucleotide is used to achieve attenuation, then this strategy can be used to apply an antisense oligonucleotide targeting a sense transcript and another antisense targeting the corresponding antisense transcript. Oligonucleotides, or a single energy symmetric antisense oligonucleotide that simultaneously targets overlapping sense and antisense transcripts. According to the invention, an antisense compound comprises an antisense oligonucleotide, a ribozyme, an external leader sequence (EGS) oligonucleotide, an siRNA compound, a single- or double-stranded RNA interference (RNAi) compound (eg, an siRNA compound), and Other polymeric compounds that hybridize to at least a portion of the dry nucleic acid and modulate its function. 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-stranded double-stranded, cyclic or hairpin poly-compounding compounds and may contain structural elements such as internal or terminal, mismatch or loop. Antisense compounds are typically made 156342.doc • 37· 201143782 Linear n but can be joined or otherwise made into a cyclic and/or branched form. Antisense compounds can comprise, for example, the following constructs: hybridized to form complete Or a double strand of a partially double-stranded compound, or a single strand having sufficient auto-complementary I' to hybridize and & into a fully or partially double-stranded compound. The two chains can be joined internally to create a free 3, or 5, end or connectable to form a continuous hairpin. The hairpin type structure may have a pendant P knife at the M3' end to extend the single chain feature. The double-stranded compound may optionally comprise a pendant knife at the end. The modification may comprise a coupling group attached to one end, a selected nucleotide acid site, a sugar site or attached to a internuclear bud 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 may be in the form of an auto-complementary hairpin-type molecule that folds in itself to form a duplex. Thus, the dsRNA can be either full or sickle double-stranded to specifically modulate @gene expression by stably expressing the dsRNA hairpin 5L in the transgenic cell line, however, in some embodiments, the gene expression or function is upregulated. When a single strand is formed from two strands, or a self-complementary hairpin-type molecule is opened (self-folded to form a duplex), the two strands (or regions in which the duplex forms a duplex) are Wats〇n_Crick mode base pairing complementary RNA strand. Following introduction into the system, the compounds of the invention may elicit the action of one or more 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 oligonucleic acids) can be described as "DNA-like" (ie, usually having one or more 2,_deoxy sugars and (through*)T instead of u bases) or "classes" rnA" (ie, usually having one or more 2'-hydroxy or 2,-modified sugars and (usually} υ instead of τ bases). Nucleic acid snail 156342.doc -38- 201143782 can be used in more than one structure Types, most commonly type A and type B. It is believed that, in general, nucleotides having a B-like structure are "like DNA" and those having a type A structure are "like RNA". (Chimeric) In embodiments, the antisense compound may contain A- and B-type regions. In one embodiment, the desired oligonucleotide or antisense compound comprises at least one of the following: antisense RNA, antisense DNA , chimeric antisense oligonucleotides, including modified linkages of antisense oligonucleotides, 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); small activating RNA (saRNA), or a combination thereof. dsRNA can also activate gene expression, a mechanism called Small RNA-induced gene activation" or RNAa. The dsRNA targeting the gene promoter induces efficient transcriptional activation of related genes. RNAa is displayed in human cells using synthetic dsRNA (called "small activating RNA" (saRNA). It is not known whether RNAa is conserved in other organisms. It has been found that small double-stranded RNA (dsRNA) (such as small interfering RNA (siRNA) and microRNA (miRNA)) is triggered by the evolutionary conservation mechanism of RNA interference (RNAi). RNAi typically causes gene silencing by remodeling chromatin to thereby repress transcription, thereby degrading complementary mRNA, or blocking protein translation. However, in the context of the examples section below, oligonucleotides may be displayed. Increase the expression and/or function of the LIM homolog 2 (LHX2) polynucleotide and its encoded product. The dsRNA can also be used as a small activating RNA (saRNA). It is not expected to be limited by theory, by the gene promoter In the sequence, saRNA can induce target gene expression, a phenomenon known as dsRNA-induced transcriptional activation (RNAa) of 156342.doc-39-201143782. In another embodiment, the "better dry segment" identified herein is available Screening for additional compounds that modulate the expression of LIM homolog 2 (LHX2) polynucleotides. "Modulators" are compounds that reduce or increase the performance of a nucleic acid molecule encoding LHX2' and include at least a complementary 5-nucleotide portion of the segment. The screening method comprises the steps of: contacting a preferred segment of a nucleic acid molecule encoding a lhx2 sense or natural antisense polynucleotide with one or more candidate modulators, and selecting One or more candidate modulators that reduce or increase the performance of a nucleic acid molecule encoding an LHX2 polynucleotide (eg, SEq m N〇: 5 to 16). 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 an LHX2 polynucleotide, the modulator can be used in other investigative studies of LHX2 polynucleotide function' or as a A research, diagnostic, or therapeutic agent of the invention. Targeting a natural antisense sequence preferably modulates the function of the target gene. For example, the LHX2 gene (for example, accession number is NM_0 (M789). In one embodiment, the antisense polynucleotide of the LHX2 gene is killed. In one embodiment, the antisense oligonucleotide targets the LHX2 polynucleus. Sense and/or natural antisense sequences, variants, alleles, isoforms, homologs, canine variants, street organisms, fragments and complementary sequences of a nucleoside (for example, accession number _004789). Preferably, the nucleotide is raised as an antisense molecule and the target comprises an antisense and/or a sensed [coding and non-coding region of the Ηχ2 polynucleotide. The preferred segment of the invention may also be complementary to the present invention. Antisense compounds are combined to form stable double-stranded (duplex) oligonucleotides. It has been shown in the industry that the double-stranded oligonucleotides such as 3H can regulate the expression of 耙 and pass 156342.doc •40- 201143782 by The sense mechanism regulates translation and RNA processing. Additionally, the double-stranded portion can undergo chemical modification. For example, it has been shown that the double-stranded portion can inhibit the target by typical hybridization of the antisense strand of the duplex with the target, thereby Initiating enzymatic degradation of the target. In one embodiment, the antisense oligonucleotide targets the LIM homolog 2 (LHX2) polynucleotides (e.g., accession number NM_004789)), variants, 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 LHX2 but extends to any of the isoforms, receptors, homologs, and the like of the LHX2 molecule. In one embodiment, the oligonucleotide targets a natural antisense sequence of the LHX2 polynucleotide (eg, the polynucleotides set forth in SEQ ID NOS: 2 to 4), any variant thereof, etc. Genes, homologs, mutants, derivatives, fragments and complementary sequences. Examples of antisense oligonucleotides are set forth in SEQ ID NOS: 5 to 16. In one embodiment, the oligonucleotide complements or binds to the nucleic acid sequence of the LHX2 antisense molecule (including, without limitation, non-coding sense and/or antisense sequences associated with the LHX2 polynucleotide) and modulates the LHX2 molecule Performance and / or function. In one embodiment, the oligonucleotide complements or binds to the nucleic acid sequence of the LHX2 natural antisense molecule (as set forth in SEQ ID NOS: 2 to 4) and modulates the expression and/or function of the LHX2 molecule. In one embodiment, the raised nucleotide comprises the sequence of at least 5 contiguous nucleotides of SEQ ID NOs: 5 to 16 and modulates the expression and/or work of the LHX2 molecule. 156342.doc -41 - 201143782 Polynucleotide Target These include LHX2 (including its family members), variants of LHX2; mutants of LHX2, including SNPs; non-coding sequences of LHX2; alleles of LHX2; species variants, fragments and the like. Preferably, the oligonucleotide is an antisense molecule. In one embodiment, the oligonucleotides that target the LHX2 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 LIM homologous sequence 2 (LHX2) polynucleotides (e. g., SEQ ID NOs: 2 to 16) can modulate the expression or function of such targets. In one embodiment, the performance or function is up-regulated compared to the control group. In one embodiment, the performance or function is downregulated as compared to a control group. In one embodiment, the antisense compound comprises the sequences set forth in SEQ ID NOs: 5 to 16. Such nucleotides may include one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. In one embodiment, SEQ ID NOS: 5 to 16 comprise one or more LNA nucleotides. Table 1 shows exemplary antisense oligonucleotides for use in the methods of the invention. Table 1 ·· Sequence number Antisense sequence name sequence SEQ ID NO: 5 CUR-1560 (^*T*(^*(^*(^*T*G*G*G*As|iC!*T*G* T*A*A*G*G*A SEQ ID NO: 6 CUR-1561 156342.doc •42· 201143782

SEQ ID NO: 7 CUR-1562 T*了*γ*(^*γ*(^*Τ*0*(^*€Ι*0*0*(^*Τ*ΟΙ*Τ*0*0*0^*Τ SEQ ID NO: 8 CUR-1563 SEQ ID NO: 9 CUR-1588 q*T*T*G*T*A*T*G*T申 G*G*G*T*TT*T*G*G*G*T*G*G SEQ ID NO: 10 CUR-1589 As,eC*A*T*C*C*C*T*G3|£C*T*Ti,:T*C*T*C*Cs|iC*T*G SEQ ID NO: 11 CUR-1590 T*T*T*A*G*T*A*G*T*A*C*C*A*C*C*G*G*A*G*C*C SEQ ID NO: 12 CUR-1591 C*C*C*A*G*C*T*T*C*A*T*C*C*T*C*A*G*A*G*C SEQ ID NO: 13 CUR-1592 申T*T*G*G*G*T*T'*Gs|err*G*A*G*G*G*G*A*G 申 G*A SEQ ID NO: 14 CUR-1593 T*C*C*A*T*A*T*C*C*A*G*C*C*G*T*G*C*C*A*C*T SEQ ID NO: 15 CUR-1594 T*c*C*A*T*T*T*G*T*C*A*C*C*A*C*C*A*G*C*A*C SEQ ID NO: 16 CUR-1595 G*G*C*T*T*C*T*G*G*G*T*G*TsliT*T*C*G1,iT*T5|eTSEQ ID NO: 7 CUR-1562 T*了*γ*(^*γ*(^*Τ*0*(^*€Ι*0*0*(^*Τ*ΟΙ*Τ*0*0*0 ^*Τ SEQ ID NO: 8 CUR-1563 SEQ ID NO: 9 CUR-1588 q*T*T*G*T*A*T*G*T申G*G*G*T*TT*T*G *G*G*T*G*G SEQ ID NO: 10 CUR-1589 As,eC*A*T*C*C*C*T*G3|£C*T*Ti,:T*C*T* C*Cs|iC*T*G SEQ ID NO: 11 CUR-1590 T*T*T*A*G*T*A*G*T*A*C*C*A*C*C*G*G *A*G*C*C SEQ ID NO: 12 CUR-1591 C*C*C*A*G*C*T*T*C*A*T*C*C*T*C*A*G* A*G*C SEQ ID NO: 13 CUR-1592 T*T*G*G*G*T*T'*Gs|err*G*A*G*G*G*G*A*G G *A SEQ ID NO: 14 CUR-1593 T*C*C*A*T*A*T*C*C*A*G*C*C*G*T*G*C*C*A*C* T SEQ ID NO: 15 CUR-1594 T*c*C*A*T*T*T*G*T*C*A*C*C*A*C*C*A*G*C*A*C SEQ ID NO: 16 CUR-1595 G*G*C*T*T*C*T*G*G*G*T*G*TsliT*T*C*G1,iT*T5|eT

Ο 可以業内已知之若干方式來調節期望靶核酸。舉例而 言,使用反義寡核苷酸、siRNA等。酶性核酸分子(例如, 核糖酶)係能夠催化一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 轉錄本。 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RN A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#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 156342.doc -43- 201143782 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(〇rgel, (1979) pr〇c. R. Soc· London,B 205, 435)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。 對於具有最適催化活性之核糖酶之研發將顯著有助於採 用RNA裂解性核糖酶來調控基因表現的任一策略。舉例而 言,錘頭狀核糖酶在飽和(10 mM)濃度之Mg2+辅因子存在 下以約1 min-丨之催化速率仏以〇發揮作用。已顯示人工 「RNA連接酶」核糖酶可以約1〇〇瓜丨^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糖酶具有由 DNA構成之受質結合臂,其可以接近之多倍周轉 速率催化RNA裂解。最後,制某些核㈣類似物代替鐘 頭狀核糖酶催化核心内之特定殘基可產生經亀糖酶, 其催化速率顯示提高多達1〇倍。該等發現表明,核糖酶可 以顯著大於大部分天然自裂解核糖酶在活體外所顯示之摧 化速率的催化速率來促進化學轉化。則可優化某些自裂解 核糖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⑽enbeck,〇 c (1987) 328. 596_6〇〇)mNA觸媒並使其與乡個rna分子進行 156342.doc -44 - 201143782 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已用於裂解特異性靶 序列,其係藉由在催化RNA中作出適當鹼基改變以維持與 靶序列之必需鹼基配對來達成。此使得可使用催化RNA來 裂解特異性靶序列,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 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之小髮夾型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)之募聚物或聚合物、或其 模擬物、後合體、類似物或同系物。此術語包含由天然存 在之核苷酸、糖及共價核苷間(主鏈)鍵聯組成之寡核苷 酸;以及具有非天然存在部分且以相似方式發揮作用之寡 核苷酸。該等經修飾或經取代寡核苷酸經常優於天然形 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)、核糖酶、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 156342.doc -45- 201143782 物)、saRNA、aRNA、及其他與靶核酸之 六姑)苗々々甘A 主v —個部分雜 父並凋即其功能之寡聚化合物。因此其 RNA、類 DNA、類 rNa、或 i 混合物 *' 〇ΝΑ ' A,、成口物,或可為該等物 -或多者之模擬物。該等化合物可為單鍵、雙鏈、環狀或 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 合物可包含諸如以下構㈣: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3’或5’末端或可連接形成連續髮夾型結構或環 路。髮夾型結構可在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 接至個核音間鍵聯之偶聯基團。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 以开> 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髮夾型來特異性調節 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 RNA 鏈。 引入系統中之後,本發明化合物可引發一或多種酶或結 156342.doc • 46· 201143782 Ο 構蛋白質之作用以實現t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(包含募核皆 )可闡述為「類DNA」(亦即,通常具有一或多個去氧 糖及(通常)τ而非u驗基)或「類RNA」(亦即,通常具有一 或多個2、經基或2’·修飾糖及(通常)U而非T驗基)。核酸螺 =可私用-種以上之結構類$,最通常係A型及B型。據 般而5,具有B型樣結構之募核苷酸係「類DNa」 且彼等具有A型樣結構者係「類RNA」n(|合)實施 例中,反義化合物可含有a_&b型區域。 本發明之反義化合物可包括長度為約5至約8〇個核苷酸 (亦即’約5至約8〇個連接核芽)的反義部分。此係指反義化 口物之反義股或部分之長度。換言之,本發明之單鍵反義 化合物包括5至約8G個核㈣,且本發明之雙鏈反義化合 (幻如dsRNA)包括長度為5至約8〇個核苷酸之有義及反 義股或部分。熟習此項技術者應瞭解,此涵蓋長度為5、 Ο 6 19 31 43 55 67 79 20 32 44 56 68 、9 21 33 45 57 69 10 22 34 46 58 70 11 23 35 47 59 71 12 24 36 48 60 72 13 25 37 49 61 73 14 26 38 50 62 74 15 27 39 51 63 75 16 28 40 52 64 76 17 29 41 53 65 77 18 30 42 54 66 78 、或80個核苷酸、或其任一範圍之反義部分。在-實施例中,本發明之反義化合物具有長度為1〇至5〇 個核替酸之反義部分。熟f此項技術者應瞭解,此展現反 156342.d〇( -47· 201143782 義部分長度為 10、11、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 個核苷酸。 在一實施例中’本發明之反義或寡核苷酸化合物具有長 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30 個核皆酸、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 吕,若第一核苷酸係腺嘌呤,則可產生在此位置含有胸 苷、鳥苷或胞苷之變體。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 定其抑制靶核酸表現之能力。 在一些實施例中,反義化合物及靶之間之同源性、序列 一致性或互補性為約40%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 156342.doc -48· 201143782 97%、約 98°/。、約 99%或約 100%。 在一實施例中,反義寡核苷酸(例如,SEQ ID NO: 5至 16中所述之核酸分子)包括一或多個取代或修飾。在一實 施例中,核苷酸經鎖核酸(LNA)取代。 • 在一實施例中,寡核苷酸靶向與LHX2有關之編碼及/或 非編碼序列及如SEQ ID NO: 1至4所示之序列之有義及/或 反義核酸分子的一或多個區域。寡核苷酸亦靶向SEQ ID NO: 1至4之重疊區域。 〇 U 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「欲合寡核苦酸」或「彼合體」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 個核苷酸構成。該等寡核苷酸通常含有至少一個經修飾核 普酸區域,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,增加與靶之結合親和力);及係 能夠裂解RNA:DNA或RNA:RNA雜合體之酶受質的區域。 0 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 Η可裂解RNA靶,由 此大大增強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 - 寡核苷酸時利用較短寡核苷酸通常可獲得可比性結果。通 常可藉由凝膠電泳及(若需要)業内已知之相關核酸雜交技 術來檢測RNA靶之裂解。在一實施例中,嵌合寡核苷酸包 括至少一個經修飾以增加靶結合親和力之區域、及(經常) 用作RNAse Η受質之區域。通常藉由量測寡核苷酸/靶配對 156342.doc -49- 201143782 之Trn來測定寡核苷酸與其靶(在此情形下係編碼ras之核 酸)之親和力,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,則寡核苷酸與靶之親和力 愈大。 本發明之嵌合反義化合物可以兩個或更多個如上所述之 寡核苷酸、經修飾寡核苷酸、寡核苷及/或寡核苷酸模擬 物之複合結構形式形成。因此,化合物在業内亦稱為雜合 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5,〇13,8:3〇號、第^丨竹,7”號、 第 5,220,〇〇7 冑、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、f 5,652,356 號、及第 5,700,922號,每一者皆以引用方式併入本文中。 在一實施例中,寡核苷酸之經修飾區域包括至少—個在 糖之2'位處修飾的核苷酸,最佳係經2,-0烷基、2,-0-烷 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、無鹼基殘基或中3,端之反 向鹼基之2’-氟、2,_胺基及2,〇_甲基修飾。通常將該等修飾 納入寡核苷酸中且已顯示該等寡核苷酸相對於2,_去氧寡核 苷I對於給定靶具有較高Tm(亦即,較高靶結合親和力” 该增加之親和力效應大大增強了 RNAi寡核苷酸對基因表 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乾,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 156342.doc •50· 201143782 凝膠電泳來顯示。在一實施例中,亦對嵌合募核苷酸進行 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 物或分離核酸酶溶液一起培育並在一段時間後經常藉由凝 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募核苷 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募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、膦酸曱酯、短鏈烷基或環烷基糠間鍵聯或 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 曱基(曱基亞胺基)或MMI主鏈]、CH2--0--N(CH3)--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0--P--0--CH)。由De Mesmaeker等人(19'95) 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 156342.doc -51· 201143782 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。較佳募核苷酸在2'位包括下列基團中之一者: OH、SH、SCH3、F、OCN、0CH30CH3、OCH3 0(CH2)n CH3、0(CH2)n NH2 或 0(CH2)n CH3(n 為 1 至約 10); C1-C10低碳烷基、烷氧基、經取代低碳烷基、烷芳基或芳 烷基;Cl ; Br ; CN ; CF3 ; OCF3 ; Ο—、S--、或 N-烷基; 〇__、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;用於改良募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'-曱氧基乙氧基 [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(2'-OCH2 CH2CH3)及2'-氟(2’-F)。亦可在募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1位及5'末端核苷 酸之5'位。募核苷酸亦可具有糖模擬物,例如使用環丁基 來代替戊呋喃酿基。 募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 嘧啶(T)、胞嘧啶(C)及尿嘧啶(U)。經修飾核苷酸包含僅偶 156342.doc -52- 201143782 爾或短暫在天然核酸中發現之核苷酸,例如次黃過吟 6 甲基腺嘌呤、5-Me嘧啶、尤其5-甲基胞嘧啶(亦稱為5曱 基去氧胞嘧啶且業内通常稱為5_Me-C)、5_經甲義於嘴 咬(HMC)、糖基HMC及龍膽二糖基HMC ;以;5人^外 夂σ成核芽 • 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、弘 ◎ 羥甲基尿嘧啶、8-氮雜鳥嘌呤、7_去氮鳥嘌呤、Ν6 (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0.6-1.2。(:且係當前之較佳鹼基取代。 對本發明募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募核苷酸、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。 給定募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 所疋義欲合募核普酸之募核苦酸。 在另一實施例中’本發明之核酸分子與另一部分偶聯, 156342.doc -53· 201143782 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 酸醋基團上之若干位置包括核酸分子之任一核苦酸中的一 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固相 合成技術來製備。包含Applied Biosystems在内之若干供 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填酸S旨及烧基化衍生物 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(amidite)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 156342.doc • 54· 201143782 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(包括胺基碟 酸3胺基酯及胺基填酸胺基烧基酯)、硫羰基胺基破酸 S旨、膦酸硫幾基烧基醋、硫獄基烧基麟酸三醋、及具有常 見3'-5'鍵聯之蝴院填酸醋、該等主鏈之2,_5'連接類似物、 C3 及彼專具有反極性者(其中相鄰核普單元對之連接由變 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。 教示上述含破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 4,476,301 號、第 5,023,243 號、第 5,177,196 號、第 5,188,897 號、第 5,264,423 號、第 5,276,019 號、第 5,278,302 號、第 Q 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 5,55〇,ηι 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募核苷酸主鏈具有藉由以下形 成之主鏈.短鏈烧基或環烧基核苷間鍵聯、混合雜原子及 烷基或環烷基核苷間鍵聯、或一或多個短鍵雜原子或雜環 156342.doc -55- 201143782 核苷間鍵聯。該等主鏈包括彼 ώ _ 傲寺具有嗎啉基鍵聯者(部分 自核皆之糖部分形幻4氧μ鏈;硫化物、亞硬及礙 主鏈,甲醯乙醯基及硫代甲酿乙醢基主鏈;亞甲基甲酿乙 醯基及硫代甲醯乙醯基主鏈;含 蟪3有烯蛵之主鏈;胺基磺酸 s旨主鏈;亞曱基亞胺基及亞甲其 f基肼基主鏈;磺酸酯及磺醯 月女主鍵,酿胺主鍵;及:i:他且古..¾人Χτ 他,、有混合Ν、〇、S及CH2組成 部分者。 教广、述养核苷之代表性美國專利包括但不限於美國專 利第 5’〇34,506 號、第 5,166,315 號、第 5,185,444 號、第 5,214,134 號、帛 5,216,141 號、帛 5,235,〇33 號、第 5’264,562 號、第 5,264,564 冑、帛 5,4〇5,938 號、第 5,434,257 號、第 5,偏,677 冑、帛 5,47〇,967 號、第 5’489’677 號、帛 5,541,307 號、帛 5,561,225 號、第 5,596,086 號、第 5 602 240 冑、帛 5 61〇 289 號第 5’602’240 號、第 5,608,04·6 1、帛 5,610,289 號、第 5,618’704 號、第 5,623,〇7〇 冑、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。 在其他較佳募核苷酸模擬物中,核苷酸單元中之糖及核 苦間鍵聯(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。一種該寡聚化合物(已顯示具 有極佳雜交性質之募核苷酸模擬物)稱為肽核酸(ΡΝΑ)。在 ΡΝΑ化合物中’募核苷酸中之糖-主鍵經含醯胺主鏈、特 定而言胺基乙基甘胺酸主鏈代替。保留核鹼基且其與主鏈 156342.doc -56- 201143782 中醯胺部分之氮雜氮原子直接或間接結合。教示PNA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,每一者 皆以引用方式併入本文中。PNA化合物之其他教示内容可 參見 Nielsen 等人,(1991) Science 254, 1497-1500。 本發明一實施例係具有硫代磷酸酯主鏈之寡核苷酸及具 有雜原子主鏈之募核苷,且特定而言係上文所提及美國專 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-0-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 主鏈結構之寡核苷酸。 經修飾寡核苦酸亦可含有一或多個經取代糖部分。較佳 寡核苷酸在2'位包括以下中之一者:OH ; F ; 0-、S-或N-烷基、Ο-、S-或N-烯基;0-、S-或N-炔基;或Ο烷基-Ο-烷 基,其中烷基、烯基及炔基可係經取代或未經取代之C1至 C6烷基或C2至C10烯基及炔基。尤佳者係〇 (CH2)n OmCH3、0(CH2)n、OCH3、0(CH2)nNH2、0(CH2)nCH3、 0(CH2)n0NH2、及 〇(CH2nON(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C6低碳烷基 '經取代低碳烷基、烷芳基、芳烷基、 〇-烷芳基或0-芳烷基、SH、SCH3、OCN、C卜Br、CN、 156342.doc -57- 201143782 CF3、OCF3、SOCH3、S02CH3、0N02、N02、N3、 NH2、雜環烷基、雜環烷芳基、胺基烷基胺基、聚烷基胺 基、經取代曱矽烷基、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 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。較佳修飾包括2’-曱氧基乙氧基(2’-0-CH2CH20CH3,亦稱為 2,-0-(2-甲氧基乙基)或 2'-MOE), 亦即,烷氧基烷氧基。其他較佳經修飾包括2’-二曱基胺基 氧基乙氧基(亦即,〇(CH2)20N(CH3)2基團,亦稱為T-DMAOE,如下文實例中所述)、及2’-二曱基胺基乙氧基乙 氧基(業内亦稱為2'-0-二曱基胺基乙氧基乙基或2’-DMAEOE),亦即,2'-0-CH2-0-CH2-N(CH2)2)。 其他較佳修飾包括2'-曱氧基(2'-0-CH3)、2'-胺基丙氧基 (2'-OCH2CH2CH2NH2)及2'-氟(2'-F)。亦可在寡核苷酸上 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 5,359,044 號、第 5,393,878 號、第 5,446,137 號、第 5,466,786 號、第 5,514,785 號、第 5,519,134 號、第 5,567,811 號、第 5,576,427 號、第 5,591,722 號、第 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 156342.doc •58- 201143782 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驗基(業内通常簡寫為「驗基」)修 飾或取代。本文所用之「未經修飾」或「天然」核苷酸包 括嘌呤鹼基(腺嘌呤(A)及鳥嘌呤及嘧啶鹼基(胸腺嘧啶 (T)、胞嘴咬(C)及尿峨咬(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-羥甲基 胞嘴咬、黃噪吟、次黃嗓岭、2 -胺基腺嗓吟、腺°票吟及鳥 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_硫代屎嘧啶、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-偶氣尿定、胞嘧啶及胸腺嘧啶、5-尿嘧啶 (假尿鳴唆)、4-硫代尿嘴咬、8-鹵代、8-胺基、8-硫醇、8-硫代烧基、8-經基及其他8-取代腺嘌呤及鳥嘌呤、5-鹵 代、尤其5-;臭、5 -三氟曱基及其他5 -取代尿嘴咬及胞嘴 咬、7-曱基鳥嘌吟及7-曱基腺嘌呤、8 -氮雜鳥嘌呤及8-氮 雜腺嘌呤、7-去氮鳥嘌呤及7-去氮腺嘌呤及3-去氮鳥嘌呤 及3 -去氮腺嘌吟。 另外,核苷酸包括彼等揭示於美國專利第3,687,808號中 者、彼等揭示於「The Concise Encyclopedia of Polymer 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, 156342.doc -59- 201143782 「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寡聚化合物之結合親 和力。該等核苷酸包括5-取代嘴咬、6-氮雜。密咬及N-2、 Ν-6及0-6取代嗓吟’包括2-胺基丙基腺嗓吟、5-丙快基尿 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 0.6-1.2°C(Sanghvi,Y.S.,Crooke,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 之鹼基取代,甚至更尤其在與2’-〇曱氧基乙基糖修飾組合 時。 教不上述經修飾核普酸以及其他經修飾核皆酸之製備之 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、及第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 學方式連接至募核苷酸。 *亥等部分包括但不限於脂質部分(例如膽固醇部分)、膽 156342.doc •60- 201143782 已基-s-三苯曱基硫醇)、硫代膽固醇、脂 二烷二酵或十一烷基殘基)、磷脂(例如, 酸、硫鱗(例如, 肪族鍵(例如,十 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-甘油-3-H-膦酸醋)、聚胺或聚乙二醇鏈、或金剛院 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。 教示該等寡核普酸偶聯物之製備之代表性美國專利包括 ΟThe 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. Enzymatic nucleic acid molecules (e.g., ribozymes) are nucleic acid molecules capable of catalyzing one 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 RN A . This binding is carried out via a target binding portion of an enzymatic nucleic acid which is in close proximity to the enzymatic portion of the molecule used to cleave #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 by 156342.doc-43-201143782. 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 (〇rgel, (1979) pr〇c. R. Soc. London, B 205, 435), have been used to generate various reactions (eg, phosphodiester linkages and hydrazines). A new nucleic acid catalyst for the cleavage and linkage of amine linkages. 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 of about 1 min-丨 in the presence of a saturated (10 mM) concentration of Mg2+ cofactor. Artificial "RNA ligase" ribozymes have been shown to catalyze the corresponding self-modification reactions at a rate of about 1 〇〇. In addition, certain modified hammerhead ribozymes are known to have a binding arm composed of DNA that catalyzes RNA cleavage by a multiple of turnover rate. Finally, the preparation of certain nuclear (iv) analogs in place of the specific residues in the nucleus catalyzed core of the head-like ribozyme produces carboxylase, which shows a catalytic rate that is increased by up to a factor of 1. These findings indicate that ribozymes can significantly promote chemical conversion by significantly greater than the catalytic rate of the rate of destruction exhibited by most native self-cleaving ribozymes in vitro. The structure of certain self-cleaving ribozymes can 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 the rna-bearing catalyzed by the RNA catalyst catalyzed by the "hammerhead" model was first shown in 1987 (10) enbeck, 〇c (1987) 328. 596_6〇〇) mNA catalyst and made it with the rural rna molecule 156342 .doc -44 - 201143782 Reaction, thus indicating that it is indeed catalytic. Catalytic RNA based on the "hammerhead" motif design has been used to cleave specific target sequences by making appropriate base changes in the catalytic RNA to maintain the necessary base pairing with the target sequence. This allows the use of catalytic RNA to cleave specific target sequences and demonstrates that catalytic RNA designed according to the "hammerhead" model can cleave specific receptor RNA in vivo. RNA interference (RNAi) has become an effective tool for regulating gene expression in mammalian and mammalian cells. This approach requires the delivery of small interfering RNA (siRNA) in the form of RNA itself or in the form of DNA using expression plasmids or viruses and coding sequences for small hairpin RNA for processing 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 a polymer or polymer of ribonucleic acid (RNA) and/or deoxyribonucleic acid (DNA), or a mimetic, conjugate, analog or homolog thereof Things. The term encompasses oligonucleotides consisting of naturally occurring nucleotides, sugars, and covalent internucleoside (backbone) linkages; and oligonucleotides having non-naturally occurring portions and functioning in a similar manner. Such modified or substituted oligonucleotides are often preferred over natural 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 compound 156342.doc-45-201143782), saRNA, aRNA, and other six-nucleus with target nucleic acid) A main v is an oligomeric compound that is partially heterozygous and has its function. Therefore, its RNA, DNA, rNa, or i mixture *' 〇ΝΑ 'A, a mouthpiece, or may be a mimic of - or more of these. The compounds may be single bond, double chain, cyclic 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. The antisense compound may comprise, for example, the following (four): two strands that are hybridized to form a fully or partially double-stranded compound, or a single strand that has sufficient auto-complementarity to hybridize and form a fully or partially double-stranded compound. The two strands can be joined internally to create a free 3' or 5' end or can be joined to form a continuous hairpin type structure or loop. The hairpin type structure may have an overhang at the end of 5, or 3 to extend the single-strand character. The double-stranded compound may contain an overhang at the end as needed. Other modifications may include a coupling group attached to one end, a selected nucleotide position, a sugar position, or a linkage to a nuclear tone. Alternatively, the two strands can be joined via a non-nucleic acid moiety or a linker group. When formed 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. Gene expression can be specifically regulated by stably expressing the dsRNA hairpin type in the transgenic cell line. When formed from a single strand of two strands, or in the form of an autorecognition hairpin-type molecule (self-folding to form a duplex), the two strands (or regions in which the duplex forms a duplex) are Watson- Crick-type base pairing complementary RNA strand. Following introduction into the system, the compounds of the invention may elicit one or more enzymes or cleaves to effect cleavage or other modification of the t-nucleic acid or may act via a mechanism based on occupancy. In general, nucleic acids (including nucleus) can be described as "DNA-like" (ie, usually with one or more deoxy sugars and (usually) τ instead of u-test) or "RNA-like" (ie, , usually with one or more 2, a trans- or 2'-modified sugar and (usually) U instead of a T-test). Nucleic acid spiro = privately available - more than one type of structural class, most commonly type A and type B. As a general example, in the case of a nucleotide-like "DNa" having a B-like structure and having an A-like structure, the "antigen-like compound" may contain a_& Type b area. Antisense compounds of the invention can include an antisense portion of from about 5 to about 8 nucleotides in length (i.e., ' from about 5 to about 8 such linked nuclear buds). This refers to the length of the antisense stock or part of the antisense. In other words, the single-bond antisense compound of the present invention comprises 5 to about 8G cores (four), and the double-stranded antisense compound (phantom of dsRNA) of the present invention comprises a sense and a counter of 5 to about 8 nucleotides in length. A stock or part. Those skilled in the art should understand that this cover length is 5, Ο 6 19 31 43 55 67 79 20 32 44 56 68 , 9 21 33 45 57 69 10 22 34 46 58 70 11 23 35 47 59 71 12 24 36 48 60 72 13 25 37 49 61 73 14 26 38 50 62 74 15 27 39 51 63 75 16 28 40 52 64 76 17 29 41 53 65 77 18 30 42 54 66 78 , or 80 nucleotides, or any of them The antisense part of the scope. In an embodiment, the antisense compound of the invention has an antisense portion of from 1 to 5 nucleotides in length. Those skilled in the art should understand that this shows the reverse 156342.d〇 (-47· 201143782 meaning part length is 10, 11, 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 An oligonucleotide of 48, 49, or 50 nucleotides, or any range thereof. In some embodiments, the oligonucleotide is 15 nucleotides in length. In one embodiment, the invention The antisense or oligonucleotide compound has an antisense portion of 12 or 13 to 30 nucleotides in length. Those skilled in the art will appreciate that the length of the antisense portion is 12, 13, 14, 15, 16 , 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nuclear 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, it may be produced. The position contains a variant of thymidine, guanosine or cytidine. This can occur anywhere in the antisense or dsRNA compound. These compounds are then tested using the methods described herein to determine their ability to inhibit the performance of the target nucleic acid. In embodiments, the homology, sequence identity or complementarity between the antisense compound and the target is from about 40% to about 60%. In some embodiments, the homology, sequence identity or complementarity is about 6 〇% 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 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 156,342.doc -48· 201143782 97%, about 98°/, about 99% or about 100%. In one embodiment, antisense oligonucleotides (for example, the nucleic acid molecules described in SEQ ID NOS: 5 to 16) Included in one or more substitutions or modifications. In one embodiment, the nucleotide is substituted with a locked nucleic acid (LNA). • In one embodiment, the oligonucleotide One or more regions of a sense and/or antisense nucleic acid molecule that encodes a coding and/or non-coding sequence associated with LHX2 and sequences as set forth in SEQ ID NOs: 1 to 4. Oligonucleotides also target SEQ ID NO: overlap region of 1 to 4. 〇U Certain preferred oligonucleotides of the invention are chimeric oligonucleotides. In the context of the present invention, "oligonucleotide" or "complex" is an oligonucleotide comprising two or more chemically distinct regions, each region consisting of at least one nucleotide. The oligonucleotides typically contain at least one modified nucleotide acid region that confers one or more beneficial properties (eg, increased nuclease resistance, increased cellular uptake, increased binding affinity to the target); and is capable of cleaving RNA : DNA or RNA: the region of the RNA heterozygote that is regulated by the enzyme. 0 For example, RNase Η is an endonuclease that cleaves the RNA strand of an RNA:DNA duplex. Thus, activation of RNase Η cleaves RNA targets, thereby greatly enhancing the efficiency of antisense regulation of gene expression. Thus, comparable results are generally obtained with shorter oligonucleotides when using chimeric-oligonucleotides compared to phosphorothioate deoxyoligonucleotides that hybridize to the same 'target region'. Cleavage of the RNA target can typically be detected by gel electrophoresis and, if desired, related nucleic acid hybridization techniques known in the art. In one embodiment, the chimeric oligonucleotide comprises at least one region modified to increase target binding affinity, and (often) used as a region for RNAse Η receptor. The affinity of an oligonucleotide to its target (in this case, a nucleic acid encoding ras), Tm-based oligonucleotides, is typically determined by measuring the Trn of the oligonucleotide/target pair 156342.doc-49-201143782. The temperature at which the target dissociates; spectrophotometry is used to detect dissociation. The higher the Tm, the greater the affinity of the oligonucleotide to the target. The chimeric antisense compounds of the invention may be formed as a composite structure of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Thus, compounds are also known in the art as hybrids or combinations. Representative U.S. patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Patent Nos. 5, 〇13, 8:3 、, 丨^, 77, 5, 220, 〇〇7 胄, 5, 256, 775. No. 5,366,878, 5,403,711, 5,491,133, 5,565,35, 5,623,065, 5,652,355, 5,652,356, and 5,700,922, each of which is incorporated herein by reference. In one embodiment, the modified region of the oligonucleotide comprises at least one nucleotide modified at the 2' position of the sugar, preferably via a 2,-0 alkyl, 2,-0-alkyl group. - 〇-alkyl or 2'-fluoro modified nucleotide. In another embodiment, the RNA modification comprises 2' to the ribose, abasic residue or the reverse base of the 3' end of the pyrimidine Fluorine, 2,-amino group and 2, 〇-methyl modification. These modifications are usually incorporated into the oligonucleotide and the oligonucleotides have been shown relative to the 2,-deoxyoligonucleoside I for a given The target has a higher Tm (i.e., higher target binding affinity). This increased affinity effect greatly enhances the inhibition of gene expression by RNAi oligonucleotides. An endonuclease that cleaves an RNA strand in an rNA:dna duplex: thus, activation of this enzyme cleaves the rna stem, and thereby greatly enhances the efficiency of RNAi inhibition. The cleavage of the RNA target is usually performed by 156342. Doc •50· 201143782 Gel electrophoresis for display. In one embodiment, chimeric nucleotides are also modified to enhance nuclease resistance. Cells contain a variety of degradable nucleic acid exonuclease and endonuclease A variety of nucleotide and nucleoside modifications have been shown to allow oligonucleotides incorporating such modifications to be more resistant to nuclease digestion than native oligodeoxynucleotides. Nuclease resistance is typically achieved by oligonucleosides The acid is incubated with the cell extract or the isolated nuclease solution and is often measured by gel electrophoresis over a period of time to measure the amount of remaining intact oligonucleotide. Oligonucleotides that have been modified to enhance their nuclease resistance Glycosides remain intact for 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 Nucleotide-enhancing nucleotides that enhance target binding affinity can also independently enhance nuclease resistance. Specific examples of some preferred nucleotides to be used in the present invention include those including modified backbones, The modified backbones are, for example, phosphorothioates, phosphotriesters, decyl phosphonates, short chain alkyl or cycloalkyl hydrazine linkages or short chain heteroatoms or heterocyclic saccharide linkages. 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 fluorenylene (mercaptoimine) or MMI backbone], CH2--0--N(CH3)--CH2, CH2-N(CH3)--N (CH3)--CH2 and 0--N(CH3)--CH2--CH2 backbone, wherein the natural phosphodiester backbone is represented by 0--P--0--CH). The guanamine backbone disclosed by De Mesmaeker et al. (19'95) 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 example, 156, 342. doc - 51, 201143, 782 (for example, peptide nucleic acid (PNA) master Chain), the phosphodiester backbone of the oligonucleotide is replaced by a polyamine backbone, which binds directly or indirectly to the aza nitrogen atom of the polyamine backbone. The raised nucleotides may also include one or more substituted sugar moieties. Preferred nucleotides include one of the following groups at the 2' position: OH, SH, SCH3, F, OCN, 0CH30CH3, OCH3 0(CH2)n CH3, 0(CH2)n NH2 or 0(CH2) n CH3 (n is from 1 to about 10); C1-C10 lower alkyl, alkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF3; OCF3; , S--, or N-alkyl; 〇__, S--, or N-alkenyl; SOCH3; S02 CH3; 0N02; N02; N3; NH2; heterocycloalkyl; heterocycloalkylaryl; Alkylamino group; polyalkylamino group; substituted decyl group; RNA cleavage group; reporter group; immigration agent; group for improving the pharmacokinetic properties of nucleotide recruitment; A group that improves the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A preferred modification comprises 2'-methoxyethoxyethoxy [2LO-CH2 CH2 OCH3, also known as 2-0-(2-decyloxyethyl)]. Other preferred modifications include 2'-decyloxy (2'-0--CH3), 2'-propoxy (2'-OCH2 CH2CH3) and 2'-fluoro (2'-F). Similar modifications can be made at other positions on the nucleotide, particularly at the 1' position of the 3' terminal nucleotide and at the 5' position of the 5' terminal nucleoside. The nucleotides may also have a sugar mimetic, for example, using cyclobutyl instead of the pentofuran. Nucleotides may also be modified or substituted, in addition or alternatively as a nucleobase (commonly abbreviated as "base"). As used herein, "unmodified" or "natural" nucleotides include adenine (A), guanine (G), thymine (T), cytosine (C), and uracil (U). The modified nucleotide comprises only 156342.doc -52 - 201143782 or a nucleotide found in the natural nucleic acid, such as hypoxanthine 6 methyl adenine, 5-Me pyrimidine, especially 5-methyl Pyrimidine (also known as 5-mercapto-deoxycytosine and commonly referred to in the industry as 5_Me-C), 5-based on mouth-bite (HMC), glycosyl-based HMC, and gentio-disaccharide-based HMC; ^External 夂 成 nucleation bud • Acid, for example, 2_amino adenine, 2-(methylamino) adenine, 2_(imidazolylalkyl) adenine, 2_(alkylalkylamino) Adenine or other hetero-substituted alkyl adenine, 2-thiouracil, 2-thiothymidine, 5-bromouracil, Hong ◎ hydroxymethyl uracil, 8-azaguanine, 7-azepine guanine , 6 (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 0.6-1.2. (: and is currently a preferred base substitution. Another modification to the nucleotides of the present invention involves chemically linking one or more of the conjugates that enhance the activity or cellular uptake of the raised nucleotide to the recruitment Nucleotides "These moieties include, but are not limited to, lipid moieties (eg, cholesterol moieties), cholesteryl moieties, aliphatic chains (eg, dodecanediol or ◎ undecyl residues), polyamines or poly(ethylene) Alcohol chains, or adamantane acetic acid. Nucleotides comprising lipophilic moieties, and methods of making such oligonucleotides are known in the art, for example, U.S. Patent Nos. 5,138, No. 45, No. 5,218,1 〇5 and 5,459,255. All positions in a given nucleotide 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 nucleoside In the single nucleoside of the acid, the present invention also encompasses the nucleus acid which is conjugated to the above-mentioned nucleotides. In another embodiment, the nucleic acid molecule of the present invention is coupled to another moiety, 156342. Doc -53· 201143782 This other part contains but not limited to Abasic nucleotides, polyethers, polyamines, polyamines, peptides, carbohydrates, lipids, or polyhydrocarbon compounds. Those skilled in the art will recognize that such molecules can be attached to sugars, The number of positions on the base or phosphate vine group includes one or more of any of the nucleic acid molecules of the nucleic acid molecule. The oligonucleotides used in the present invention can be prepared in a convenient conventional manner via well-known solid phase synthesis techniques. Equipment for carrying out the synthesis is sold by a number of suppliers, including Applied Biosystems. 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 for preparation. Other oligonucleotides such as thioacids S and alkylated derivatives are well known in the art using similar techniques and commercially available modified DNA synthetic amidite and fixed pore glass (CPG) products (eg, Fluorescently labeled, biotinylated by biosynthetic, luciferin, acridine or psoralen modified DNA synthesis nucleotides and / or CPG (available from Glen Research, Sterling, VA) Or other modifications Nucleotides, such as cholesterol-modified oligonucleotides. According to the present invention, the use of modifications (e.g., using LNA monomers) to enhance the efficacy, specificity, and duration of action and broaden the administration of oligonucleotides includes, for example, Chemical methods such as MOE, ANA, FANA, PS, etc. This can be achieved by using LNA monomers instead of some of the monomers in the current oligonucleotide. LNA-modified oligonucleotides can have dimensions similar to the parent compound or Preferably, the LNA-modified oligonucleotide has less than about 70%, more preferably less than about 60%, most preferably less than about 50% of the LNA monomer, and the size thereof Between about 5 and 25 nucleotides, more preferably about 12 and 20 cores 156342.doc • 54·201143782. Preferred modified oligonucleotide backbones include, but are not limited to, phosphorothioates, palmitic phosphorothioates, dithiophosphates, phosphotriesters, aminoalkyl phosphates, methyl phosphonates, and Other alkyl phosphonates (including phosphonic acid 3-alkyl esters and palmitic phosphonates), phosphinates, amino phosphates (including amine-based acid 3-amino esters and amine-based amines) Alkyl ester), thiocarbonylamine-based acid-decomposing S, phosphonic acid sulfur-based sulphuric acid vinegar, sulfur-based ketone-based linoleic acid triacetate, and a common 3'-5' linkage 2, _5' connection analogs of these main chains, C3 and those with opposite polarity (where the connection of adjacent nuclear unit pairs becomes 5'-3' or 2'-5' becomes 5 '-2'). Also included are various salts, mixed salts and free acid forms. Representative U.S. patents including the above-described broken bonds include, but are not limited to, U.S. Patent Nos. 3,687,808, 4,469,863, 4,476,301, 5,023,243, 5,177,196, 5,188,897, 5,264,423, 5,276,019, 5,278,302, Q 5,286,717, 5,321,131, 5,399,676, 5,405,939, 5,453,496, 5,455,233, 5,466,677, 5,476,925, 5,519,126, 5,536,821 No. 5,541,306, 5,55, η, pp. 5,563,253, 5,571, 799, 5,587, 361, and 5, 625, 050, each incorporated herein by reference. Preferred modified nucleotide backbones containing no phosphorus atoms have a backbone formed by the following: short chain alkyl or cycloalkyl nucleoside linkages, mixed heteroatoms, and alkyl or cycloalkyl nucleosides Inter-bonding, or one or more short-bond heteroatoms or heterocycles 156342.doc -55- 201143782 Internucleoside linkages. These main chains include those of ώ ώ 寺 具有 吗 ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( The main chain of the methyl ketone group; the main chain of the methylene ketone and the thioformamyl group; the main chain containing the oxime 3 with an olefin; the main chain of the amino sulfonic acid s; Amine and methylene, its f-base group main chain; sulfonate and sulfonate, the main bond of the amine, and the main bond of the amine; and: i: he and the ancient.. 3⁄4 people Χτ, he has mixed Ν, 〇, S and Representative of CH2. Representative US patents for cultivating and nucleating nucleosides include, but are not limited to, U.S. Patents 5'〇34,506, 5,166,315, 5,185,444, 5,214,134, 帛5,216,141 No. 5, 235, 〇 33, 5'264, 562, 5, 264, 564 胄, 帛 5, 4 〇 5, 938, 5, 434, 257, 5, partial, 677 胄, 帛 5, 47 〇, 967, 5 '489'677, 帛5,541,307, 帛5,561,225, 5,596,086, 5 602 240 胄, 帛5 61〇289, 5'602'240, 5,608,04·6 1帛5, 610, 289, 5, 618 '704, 5, 623, 〇 7 〇胄, 5, 663, 312, 5, 633, 360, 5, 677, 437, and 5, 677, 439, each of which is incorporated herein by reference. In the nucleotide mimetic, both the sugar and the internucleosis linkage (ie, the backbone) in the nucleotide unit are replaced by a new group. The base unit is retained to hybridize with a suitable nucleic acid target compound. A poly compound (a nucleotide mimetic that has been shown to have excellent hybridization properties) is called a peptide nucleic acid (ΡΝΑ). In a ruthenium compound, the sugar-primary bond in the nucleotide is contained in the guanamine-containing backbone, specifically The aminoethylglycine backbone is substituted. The nucleobase is retained and directly or indirectly bonded to the aza nitrogen atom of the guanamine moiety of the backbone 156342.doc-56-201143782. A representative US representative of the preparation of PNA compounds Patents include, but are not limited to, U.S. Patent Nos. 5,539,082, 5, 714, 331, and 5, 719, 262, each of which is incorporated herein by reference. 4, 1497-1500. An embodiment of the invention is an oligonucleotide having a phosphorothioate backbone and a nucleoside having a hetero atom backbone, and in particular, the above-mentioned U.S. Patent No. 5,489,677 -CH2-NH-0-CH2-, -CH2-N(CH3)-0-CH2-[called 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 as -0-P-0-CH2- And the guanamine backbone of U.S. Patent No. 5,602,240, incorporated herein by reference. Also preferred are the oligonucleotides having the morpholino backbone structure of U.S. Patent No. 5,034,506, the disclosure of which is incorporated herein by reference. The modified oligonucleotide may also contain one or more substituted sugar moieties. Preferred oligonucleotides include one of the following in the 2' position: OH; F; 0-, S- or N-alkyl, Ο-, S- or N-alkenyl; 0-, S- or N - alkynyl; or decyl-fluorenyl-alkyl, wherein the alkyl, alkenyl and alkynyl groups may be substituted or unsubstituted C1 to C6 alkyl or C2 to C10 alkenyl and alkynyl groups. Particularly preferred are 〇(CH2)n OmCH3, 0(CH2)n, OCH3, 0(CH2)nNH2, 0(CH2)nCH3, 0(CH2)n0NH2, and 〇(CH2nON(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: a C1 to C6 lower alkyl 'substituted lower alkyl, alkaryl, aralkyl, fluorenyl-aryl or 0-aryl Alkyl, SH, SCH3, OCN, C, Br, CN, 156342.doc -57- 201143782 CF3, OCF3, SOCH3, S02CH3, 0N02, N02, N3, NH2, heterocycloalkyl, heterocycloalkylaryl, amine Alkylamino group, polyalkylamine group, substituted alkylene group, RNA cleavage group, acceptor group, intercalator, group of pharmacokinetic properties of modified oligonucleotide, or modified oligo A group of pharmacodynamic properties of a glycoside, and other substituents having similar properties. Preferred modifications include 2'-methoxyethoxyethoxy (2'-0-CH2CH20CH3, also known as 2,-0-(2-methoxyethyl) or 2'-MOE), ie, alkoxy Alkoxy group. Other preferred modifications include 2'-didecylaminooxyethoxy (i.e., 〇(CH2)20N(CH3)2 group, also known as T-DMAOE, as described in the Examples below), And 2'-didecylaminoethoxyethoxy (also known in the art as 2'-0-didecylaminoethoxyethyl or 2'-DMAEOE), ie, 2'-0 -CH2-0-CH2-N(CH2)2). Other preferred modifications include 2'-decyloxy (2'-0-CH3), 2'-aminopropoxy (2'-OCH2CH2CH2NH2) and 2'-fluoro (2'-F). Similar modifications can be made at other positions on the oligonucleotide, especially at the 3' end nucleotide or the 2'-5' ligation oligonucleotide in the 3' and 5' end nucleotides of the sugar. '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, 5,359,044, 5,393,878, 5,446,137, 5,466,786, Nos. 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, 156,342.doc •58 - 201143782 5,658,873, 5,670,633, and 5,700,920 each incorporated herein by reference. Oligonucleotides can also be modified or replaced by a verification base (often abbreviated as "test base" in the industry). As used herein, "unmodified" or "natural" nucleotides include purine bases (adenine (A) and guanine and pyrimidine bases (thymine (T), beetle bite (C), and urinary bites ( U)). Modified nucleotides include other synthetic and natural nucleotides, such as 5-methylcytosine (5-me_C), 5-hydroxymethyl beetle, yellow noise, hypoxanthine, 2 - Amino adenine, glandular sputum and 6-methyl and other alkyl derivatives of guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2 thiopyrimidine, 2-thiothymidine and 2-thiocytosine, 5-protonated uracil and cytosine, 5-propynyl uracil and cytosine, 6-oxyuracil, cytosine and thymine, 5- Uracil (false urinary sputum), 4-thiourine bite, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-carbyl and other 8-substituted adenine And guanine, 5-halogen, especially 5--; odor, 5-trifluoromethyl and other 5-substituted urinary mouth bites, 7-mercapto guanine and 7-mercapto adenine, 8 -azaguanine and 8-azadenine, 7-deazaguanine and 7-deaza adenine and 3- Denitrified guanine and 3 - deaza adenine. In addition, the nucleotides include those disclosed in U.S. Patent No. 3,687,808, the disclosure of which is incorporated herein by reference to the " The Concise Encyclopedia of Polymer Science And Engineering, pp. 858-859 Page, Kroschwitz, JI Editor, John Wiley & Sons, 1990, by Englisch et al., "Angewandle Chemie, International Edition", 1991, 30, p. 613, and by Sanghvi, YS, Chapter 15, 156342.doc -59-201143782 "Antisense Research and Applications", pp. 289-302, Crooke, ST and Lebleu, B. ea., CRC Press, 1993. Some of these nucleotides are especially useful. To increase the binding affinity of the oligomeric compound of the present invention. The nucleotides include a 5-substituted mouth bite, a 6-aza. The bite and N-2, Ν-6 and 0-6 substituted 嗓吟' include 2- Aminopropyl adenine, 5-propionyl uracil and 5-propynyl cytosine. 5-methylcytosine substitution has been shown to increase the stability of nucleic acid duplexes by 0.6-1.2 ° C (Sanghvi, YS, Crooke, ST and Lebleu, B. Editor, "Antisense Research and A Pplications, CRC Press, Boca Raton, 1993, pp. 276-278) and are currently preferred base substitutions, even more particularly when combined with 2'-methoxyethyl saccharide modifications. Representative U.S. patents which teach the preparation of the above-described modified nucleotide acid and other modified nucleic acids include, but are not limited to, U.S. Patents 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, 5,587,469 No. 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 nucleotide. *Hai and other parts include, but are not limited to, lipid parts (such as cholesterol part), biliary 156342.doc • 60-201143782 hexyl-s-triphenylmercaptothiol), thiocholesterol, lipodiene diacetate or undecane Base residues), phospholipids (eg, acid, sulfur scales (eg, aliphatic bonds (eg, dodeca-hexadecyl-racemic-glycerol or triethylammonium 1,2,di-anthracene-hexa) Alkyl--racemic-glycerol-3-H-phosphonic acid vinegar, polyamine or polyethylene glycol chain, or diamondine acetic acid, palmitoyl moiety, or octadecylamine or hexylamino-carbonyl-oxycholesterol Representative US patents that teach the preparation of such oligonucleotide primers include Ο

但不限於美國 5,218,105 號、 5.545.730 號、 5.580.731 號、 5,109,124 號、 5,414,077 號、 5,578,718 號、 4,605,735 號、 4,789,737 號、 4,876,335 號、 5.082.830 號、 5.082.830 號、 5,245,022 號、 5,262,536 號、 5,317,098 號、 5,416,203 號、 5,512,667 號、 卜利第4,828,979號、 第 5,525,465 號、第 第 5,552,538 號、第 第 5,580,731 號、第 第 5,118,802 號、第 第 5,486,603 號、第 第 5,608,046 號、第 第 4,667,025 號、第 第 4,824,941 號、第 第 4,904,582 號、第 第 5,112,963 號、第 第 5,112,963 號、第 第 5,254,469 號、第 第 5,272,250 號、第 第 5,371,241 號、第 第 5,451,463 號、第 第 5,514,785 號、第 第 4,948,882 號、第 5,541,313 號、第 5,578,717 號、第 5,591,584 號、第 5,138,045 號、第 5,512,439 號、第 4,587,044 號、第 4,762,779 號、第 4,835,263 號、第 4,958,013 號、第 5.214.136 號、第 5.214.136 號、第 5,258,506 號、第 5,292,873 號、第 5,391,723 號、第 5,510,475 號、第 5,565,552 號、第 156342.doc -61- 201143782 5,567,810 號、第 5,574,142 號、第 5,585,481 號、第 5,587,371 號、第 5,595,726 號、第 5,597,696 號、第 5,599,923 號、第 5,599,928號、及第 5,688,941 號,每一者 皆以引用方式併入本文中。 藥物研究.亦可將本發明化合物加至藥物研究及乾驗證 之區域中。本發明涵蓋本文所鑑別化合物及較佳靶區段在 藥物研究中之用途,該藥物研究試圖闡釋UM同源序列2 (LHX2)聚核苷酸與疾病狀態、表現型、或病狀之間所存在 之關係。該等方法包含檢測或調節LHX2聚核苷酸,其包 括使試樣、組織、細胞、或有機體與本發明化合物接觸, 在治療後某一時間量測LHX2聚核苷酸之核酸或蛋白質含 置及/或相關表現型或化學端 未處理試樣或經本發明中另一 點’及視需要比較量測值與 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t驗證過程 之未知基因的功能’或用以測定特定基@產物作為用於治 療或預防特定疾病、病狀、或表現型之㈣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。該檢測可藉由業内熟 知之若干方法來達成。例如,可藉由南方印跡或藉由聚合 酶鏈反應(明技術使㈣異性擴增與核酸有關之核皆酸 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 J56342.doc -62- 201143782 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來外源核 酸中RNA之表現。舉例而言,可根據靶核酸表現之降低或 增加來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 編碼區域。藉由將每一編碼區域插入報導子編碼區域及其 r\ ^ P〇ly(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3’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)、胭脂鹼合酶(NOS)、章魚肉鹼合 酶(OCS)、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)、慶大黴素 (gentamycin)、潮黴素(hygromycin)、卡那黴素 (kanamycin)、林可徽素(lincomycin)、甲胺蝶呤 (methotrexate)、草胺膦(phosphinothricin)、°票呤黴素 (puromycin)、及四環素(tetracycline)。測定報導子基因之 156342.doc -63- 201143782 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LHX2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。lhx2 ELISA分 析套組市面有售’例如,購自R&D Systeins (Minneap〇Us, MN)。 , 在實施例中,藉由與對照組試樣中之Lhx2表現進行比 較來評估使用本發明反義寡核苷酸處理之試樣(例如,活 體内或活體外中之細胞或組織)中的LHX2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 理試樣與對照反義寡核苷酸(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, 持家基因)表現之差異來比較經處理試樣與未處理試樣中 LHX2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 異以用於與對照組進行比較。在實施例中,相對於未處理 試樣或經對照核酸處理之試樣,經本發明反義募核苦酸處 理之試樣中LHX2 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,LHX2 mRNA或蛋 156342.doc -64- 201143782 白質之含量增加或降低至少約1.25倍、至少約1.3倍、至少 約1.4倍、至少約1.5倍、至少約1.6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7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10倍或更高。However, it is not limited to US 5,218,105, 5.545.730, 5.580.731, 5,109,124, 5,414,077, 5,578,718, 4,605,735, 4,789,737, 4,876,335, 5.082.830, 5.082.830 , 5, 245, 022, 5, 262, 536, 5, 317, 098, 5, 416, 203, 5, 512, 667, Bliss 4, 828, 979, 5, 525, 465, 5, 552, 538, 5, 580, 731, 5, 118, 802, 5, 486, 603, pp. 5,608,046, 4,667,025, 4,824,941, 4,904,582, 5,112,963, 5,112,963, 5,254,469, 5,272,250, 5,371,241, 5,451,463 No. 5,514,785, 4,948,882, 5,541,313, 5,578,717, 5,591,584, 5,138,045, 5,512,439, 4,587,044, 4,762,779, 4,835,263, 4,958,013, 5.214.136, 5.214.136, 5,258,506, 5,292,873, 5, 391,723, 5,510,475, 5,565,552, 156,342.doc-61-201143782 5,567,810, 5,574,142, 5,585,481, 5,587,371, 5,595,726, 5,597,696, 5,599,923, Nos. 5, 599, 928, and 5,688, 941 each incorporated herein by reference. Drug Research. The compounds of the invention may also be added to the area of drug research and dry validation. The present invention encompasses the use of the compounds identified herein and preferred target segments for drug research, which attempts to elucidate the relationship between a UM homologous sequence 2 (LHX2) polynucleotide and a disease state, phenotype, or condition. The relationship of existence. The methods comprise detecting or modulating an LHX2 polynucleotide comprising contacting a sample, tissue, cell, or organism with a compound of the invention, and measuring the nucleic acid or protein of the LHX2 polynucleotide at a time after treatment. And/or related phenotype or chemically untreated sample or another sample in the present invention' and a comparatively measured value and compound treated sample as needed. The methods may also be performed in parallel or in combination with other experiments to determine the function of an unknown gene for the t-validation process' or to determine a particular base@product for use in treating or preventing a particular disease, condition, or phenotype (4) Effectiveness. 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 well known in the art. For example, the presence of an exogenous nucleic acid can be detected by Southern blotting or by polymerase chain reaction (better technique for the amplification of a nucleic acid-related nucleotide sequence by a heterosexual amplification). Conventional methods for measuring the performance of exogenous nucleic acids. For example, Northern blot and reverse transcription pcR (RT_pcR) can be used to detect mRNA produced from an exogenous nucleic acid by J56342.doc-62-201143782. Activity or reporter protein activity to detect the expression of RNA in an exogenous nucleic acid. For example, the antisense modulating activity can be indirectly measured according to the decrease or increase in the performance of the target nucleic acid, thereby indicating that the exogenous nucleic acid is in the effector-subRNA Based on sequence conservation, primers can be designed and used to amplify the coding region of the target gene. Initially, the highest expression coding region from each gene can be used to create a control gene model, but any coding or non-coding region can be used. Each control gene is assembled by inserting each coding region between the reporter coding region and its r\^P〇ly(A) signal. These plasmids will be generated on the gene. The swim portion has a reporter gene and has a potential RNAi target mRNA in the 3' non-coding region. The effectiveness of the individual antisense oligonucleotides is analyzed by modulating the reporter gene. Reporter genes for use in the methods of the invention Containing acetaminolate synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucuronidase (GUS), chloramphenicol acetyltransferase (CAT), Green fluorescent protein (GFP), red fluorescent protein (RFP), yellow ^ fluorescent protein (YFP), cyan fluorescent protein (CFP), horseradish peroxidase (HRP), luciferase (Luc) , nopaline synthase (NOS), octopine carnitine synthase (OCS), and its derivatives. A variety of markers can be used to confer resistance to: • Selectable markers: ampicillin (ampicillin), bleomycin ( Bleomycin), chloramphenicol, gentamycin, hygromycin, kanamycin, lincomycin, methotrexate, grass Phosphothosicin, puromycin, and tetracycline. Determination of reporters Gene 156342.doc -63- 201143782 Methods of modulation 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 LHX2 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 lhx2 ELISA assay kit is commercially available', for example, from R&D Systeins (Minneap® Us, 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 Lhx2 expression in the control sample. LHX2 behaves (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 treated sample can be compared to a control antisense oligonucleotide (e.g., with altered or different sequences) depending on the desired information. In another embodiment, the treated sample and the untreated test can be compared using differences in the performance of different nucleic acids (including any criteria deemed suitable by the investigator, eg, housekeeping genes) in the treated and untreated samples. The difference in the expression of LHX2 protein or nucleic acid in the sample. The observed difference can be expressed, for example, in ratios or parts as needed for comparison with a control group. In an embodiment, the amount of LHX2 mRNA or protein in the sample treated with the antisense nucleus acid of the present invention is increased or decreased by about 1.25 times to about 1 with respect to the untreated sample or the sample treated with the control nucleic acid. 〇 times or higher. In embodiments, the LHX2 mRNA or egg 156342.doc-64-201143782 white matter is increased or decreased by at least about 1.25 times, at least about 1.3 times, at least about 1.4 times, at least about 1.5 times, at least about 1.6 times, at least about 1.7.倍, at least about 1.8 times, at least about 2 times, at least about 2.5 times, at least about 3 times, at least about 3.5 times, at least about 4 times, at least about 4.5 times, at least about 5 times, at least about 5.5 times, at least about 6 Multiplier, at least about 6.5 times, at least about 7 times, at least about 7.5 times, at least about 8 times, at least about 8.5 times 'at least about 9 times, at least about 9.5 times, or at least about 10 times or more.

套組、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 組之研究試劑及組份。另外,彼等熟習此項技術者通常使 用能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路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σ物(單獨或與其他化合物或治療劑組合)可用作差值 及/或組合分析中之工具以闡釋細胞及組織内所表現基因 之一部分或整個互補序列的表現模式。 本文所用之術語「生物系統」或「系統」定義為表現、 或使其足以表現LIM同源序列2 (LHX2)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、&織 植 物、移植物及其組合。 、種移植 理之 照組 病相 π Μ坰性霄例,對經一或多種反義 細胞或組織内 的表現核式與未經反義化合物 細胞或組織逸彡ϋ & 進仃比較,且分析所得模式關於 156342.doc -65- 201143782 關性、信號傳導路徑、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)、READS(消化⑺财之 限制性酶擴增)、TOGA(總體基因表現分析)、蛋白質陣列 及蛋白組學、已表現序列標誌(EST)測序、消減RNa指紋 技術(Surf)、消減選殖、差異顯示(DD)、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LIM同源序列2(LHX2)之核酸雜交。舉例而言, 在如本文所揭示可用作有效LHX2調節劑之效率及條件下 雜交之寡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m丨物及探針可用於需要特異性檢測 編碼LHX2之核酸分子的方法中及擴增該等核酸分子以用 於檢測或用於其他LHX2研究。本發明之反義寡核替酸(尤 其引物及探針)與編碼LHX2之核酸的雜交可藉由f内 方式進行檢測。該等方式可包含使酶與募核㈣偶聯、放 射性標記寡核苷酸、或任一其他適宜檢測方式。亦可製造 使用邊等檢測方式檢測試樣中之⑶幻含量的套組。 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 物已用作治療部分來治療動物 (已3人類)之疾病狀態。反蠢寞 汉義养核苷酸樂物已安全且有效 156342.doc -66 - 201143782 地投與人類且當前正實施許多臨床試驗。由此確定,反義 化合物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。 在治療時,藉由投與本發明反義化合物來治療懷疑患有 可藉由調節LHX2聚核芽酸之表現進行治療之疾病或病症 的動物(較佳係人類)。舉例而言,在一非限制性實施例 中,該等方法包括向需要治療之動物投與治療有效量之 LHX2調節劑之步驟。本發明之LHX2調節劑可有效調節 LHX2之活性或調節LHX2蛋白質之表現。在一實施例中, 與對照組相比,使LHX2在動物中之活性或表現抑制約 10%。較佳地,使LHX2在動物中之活性或表現抑制約 3 0%。更佳地,使LHX2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LIM同源序 列2(LHX2)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%。 在一實施例中,與對照組相比,使LIM同源序列2 (LHX2)在動物中之活性或表現增加約10%。較佳地,使 LHX2在動物中之活性或表現增加約30%。更佳地,使 LHX2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LHX2 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 156342.doc -67· 201143782 85/〇、至少90%、至少95%、至少98。/。、至少99%、或 100%。 舉例而s,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UM同源序列2(LHX2)之表 現的減小。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LHX2肽及/或LHX2蛋白質本身的核酸分 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 對本發明募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、細胞分佈或細胞攝取之部分或偶聯物以化 學方式連接至募核苷酸。該等部分或偶聯物可包含共價結 合至諸如一級或二級羥基等功能基團之偶聯基團。本發明 之偶聯基團包含嵌入劑、報導子分子、聚胺、聚醯胺、聚 乙二醇、聚謎、增強募聚物之藥效動力學性質之基團、及 增強寡聚物之藥物代謝動力學性f之基團。典型偶聯基團 包含膽固醇、脂質、磷脂、生物素、吩嗪、葉酸酯、菲 啶、蒽醌、吖啶、螢光素、若丹明(rh〇damine)、香豆素、 及染料。在本發明之上下文中’增強藥效動力學性質之基 團包含可改良攝取、增強降解抗性、及/或增強與乾核酸 之序列特異性雜交的基團。在本發明之上下文中,增強藥 物代謝動力H生質之基團&含可改良本發明化合物之攝 156342.doc -68- 201143782 取、分佈、代謝或分泌的基團。代表性偶聯基團揭示於 1992年10月23曰提出申請之國際專利申請案第 PCT/US92/09196號、及美國專利第6,287,860號中,其以引 用方式併入本文中。偶聯部分包含但不限於脂質部分(例 如膽固醇部分)、膽酸、硫醚(例如,己基_5_三苯甲基硫 醇)、硫代膽固醇、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-Ο-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》本發明之募核苷酸亦可偶聯 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)、酮洛芬 (ketoprofen)、(S)-(+)-普拉洛芬((S)-(+)-pranoprofen)、卡 洛芬(carprofen)、丹肌胺酸(dansylsarcosine)、2,3,5-三碘 苯曱酸、氟芬那酸(flufenamic acid)、亞葉酸、苯并售二 喚、氯嗔D秦、二氮呼、°引D朵美辛(indomethicin)、巴比妥酸 鹽、頭孢菌素(cephalosporin)、續胺藥、抗糖尿病藥、抗 菌劑或抗生素。 教示該等募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 156342.doc -69- 201143782 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 X 第 5,112,963 號、 第 5,214,136 號 第 5,245,022 號 第 5,254,469 號、 .第 5,258,506 號 、 第 5,262,536 號 第 5,272,250 號、 .第 5,292,873 號 第 5,317,098 號 % 第 5,371,241 號、 .第 5,391,723 號 、 第 5,416,203 號 > 第 5,451,463 號、 .第 5,510,475 號 、 第 5,512,667 號 第 5,514,785 號、 ,第 5,565,552 號 、 第 5,567,810 號 > 第 5,574,142 號、 ,第 5,585,481 號 、 第 5,587,371 號 > 第 5,595,726 號、 第 5,597,696 號 第 5,599,923號、第 5,599,928號及第 5,688,941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5,108,921號、第5,354,844號、第5,416,016 號、第 5,459,127號、第 5,521,291 號 '第 5,543,165號、第 156342.doc •70· 201143782 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 5,534,259 號、第 5,543,152 號、第 5,556,948 號、第 5,580,5<75號、及第5,595,756號,每一者皆以引用方式併入 〇 W 本文中。 儘管為調節挺表現及/或功能不需要在載體背景中投與 反義券核普酸’但本發明實施例係關於用於表現反義寡核 苦酸之表現載體構築體,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。 在一實施例中,本發明實踐涉及使用適宜核酸遞送系統 Q 投與上述反義寡核苷酸中之至少一者。在一實施例中,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: 5至 16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 腺病毋、腺病毒相關病毒(AAV)、輔助細胞依賴性腺病 母、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 156342.doc -71- 201143782 至聚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’較佳載體包含病毒載體、融合蛋白及化學偶聯 物。逆轉錄載體包含Moloney小鼠白血病病毒及HIV基病 毒。一種較佳HIV基病毒載體包括至少兩個載體,其中gag 及pol基因來自HIV基因組且env基因來自另一病毒。dnA 病毋載體較佳。該等载體包含ρ〇χ载體(例如正痘病毒或烏 痘病毒載體)、皰疹病毒載體(例如單純皰疹ζ病毒(HSV)載 體)、腺病毒载體及腺相關病毒載體。 本發明之義化合物涵蓋任一醫藥上可接受之鹽、酯、或 該等酯之鹽、或在投與動物(包含人類)後能夠提供(直接或 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。對於寡核苷 酸而言,S藥上T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㈣,包含陰道及直腸遞送)、肺部(例 =藉由吸人或噴射粉末或氣溶膠,包含藉由霧化器); 耽官内、鼻内、表皮及經皮、經口或非經腸投與。非經腸 156342.doc -72- 201143782 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,鞘内或心室内)投與。 為治療中樞神經系統中之組織,可藉由(例如)注射或輸 注至腦脊髓流體中來進行投與。反義RNA在腦脊髓流體中 之投與闡述於(例如)美國專利申請公開案第2007/0117772 號之「Methods for slowing familial ALS disease progression」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寡核苷酸投與中樞神經系統之細 胞中時,可投與一或多種能夠促進標題反義寡核苷酸滲透 經過血腦障壁之藥劑。可在(例如)内嗅皮層或海馬區中進 行注射。藉由向肌肉組織中之運動神經元投與腺病毒載體 來遞送神經營養因子闡述於(例如)美國專利第6,632,427號 之 「 Adenoviral-vector-mediated gene transfer into medullary motor neurons」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)中並闡述於(例如)美國專利第 6,756,523 號之「Adenovirus vectors for the transfer of foreign genes into cells of the central nervous system particularly in brain」中,其以引用方式併入本文中。可 藉由注射快速投與或經一定時間藉由緩慢輸注或投與緩釋 調配物進行投與。 標題反義寡核苷酸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,反義寡核苷酸可與業内 已知促進在血腦障壁中之滲透或輸送之任一物質(例如鐵 156342.doc -73- 201143782 傳遞蛋白受體之抗體)偶合,並藉由靜脈内注射投與。反 義化合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/或增加反義化合物在血腦障壁中之輸 送。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、D(+)半 乳糖、D(+)乳糖、D(+)木糖、衛矛醇、肌-肌醇、L(-)果 糖、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、D(-)核糖、核糖醇、D(+)阿糖醇、L(-) 阿糖醇、D(+)岩藻糖、L(-)岩藻糖、D㈠來蘇糖、L(+)來 蘇糖、及L(-)來蘇糖;或胺基酸,包含但不限於麩胺醯 胺、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、亮胺酸、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。 標題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、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。舉例而言,陽離子型脂質可 156342.doc -74- 201143782 包含於調配物中以促進寡核芽酸攝取。一種顯示可促進攝 取之該組合物係 LIPOFECTIN(購自 GIBCO-BRL,Bethesda, MD)。 據信’至少一個2'-0-曱氧基乙基修飾之募核苷酸尤其可 • 用於經口投與。用於局部投與之醫藥組合物及調配物可包 . 含經皮貼片、軟膏、洗劑、乳膏、凝膠、滴劑、栓劑、噴 霧劑、液體及粉末。可能需要或期望習用醫藥載劑、水 性、粉末或油性基質、增稠劑及諸如此類。亦可使用包覆 〇 避孕套、手套及諸如此類。 可便利地以單位劑型呈現之本發明醫藥調配物可根據醫 藥工業中熟知之習用技術製得。該等技術包含使活性成份 與醫藥載劑或賦形劑結合之步驟。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、'膝囊、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 含有可增加懸浮液之黏度之物質,其包含(例如)羧曱基纖 維素鈉、山梨糖醇及/或右旋糖酐。懸浮液亦可含有穩定 劑。 本發明之醫藥組合物包含但不限於溶液、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 包括一或多種滲透增強劑、載劑、賦形劑或其他活性或惰 156342.doc •75- 201143782 性成份。 2㈣㈣分散於另―液滴形式液體(直徑經 Μ 異質系統。乳液除分散相外亦可含有額外 組份、及可呈現為存於次相、 子⑦水相、油相中之溶液形式或自身作 為分離相的活性藥物。本發 本 赞月θ施例包含微乳液。乳液 及其應用在業内已眾所用L n 斤周知且進一步闡述於美國專利第 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」意指由怖晉协—- 於一或夕個球形雙層中之兩親性脂 質組成的囊泡。脂質體係單層或多層囊泡,其具有自親脂 !生材料形成之膜及含有擬遞送組合物之水性内部結構。陽 離子里月日質體係帶正電之脂質體,據信其可與帶負電之 舰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職而非與其複合1離子型及非陽 離子型脂質體皆可用於向細胞遞送dna。 $脂㈣亦包含「空間穩定」之脂㈣,本文所用之該術 ”。係扣包括一或多種特定脂質之脂質體。在納入脂質體中 ^該等特疋脂質會產生相對於缺乏該等特定脂質之脂質 體具有增強之循環壽命的脂質體。空間穩定脂質體之實例 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 行生自或夕種親水性聚合物(例如聚乙二醇(peg)部分) 者。脂質體及其應用進一步闌述於美國專利第6,287,860號 中。 本I月之省藥調配物及組合物亦可包含表面活性劑。表 156342.doc -76- 201143782 面活性劑在藥物產物、調配物及乳液中之應用在業内已眾 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。 在一實施例中,本發明採用各種滲透增強劑來實現核 ' 酸、尤其募核苷酸之有效遞送。除有助於非親脂性藥物在 • 細胞膜中之擴散外,滲透增強劑亦增強親脂性藥物之滲透 性。滲透增強劑可歸類為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闡述於美國專利第 6,287,860號中,其以引用方式併入本文中。 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募核苷酸與 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、脂肪酸酯、類固醇、螯合劑及表面活性劑。 〇 較佳脂質及脂質體包含中性脂質及脂質體(例如二油醯基_ 磷脂酿基D〇PE乙醇胺、二肉豆謹醯基磷脂醯基膽1 DMPC、一硬脂醯基磷脂醯基膽鹼)、負脂質及脂質體(例 如一肉丑蔻醯基磷脂醯基甘油DMpG)及陽離子型脂質及脂 f體(例如二油酿基四甲基胺基丙基D〇TAp及二油酿基^ 脂醯基乙醇胺DOTMA)。 對於局部或其他投與而言,本發明之寡核普酸可囊封於 脂質體内或可與其形成複合物(尤其陽離子型脂質體卜另 一選擇為,募核普酸可與脂質、尤其陽離子型脂質複合。 156342.doc -77- 201143782 較佳脂肪酸及酯、其醫藥上可接受之鹽、及其應用進一步 闡述於美國專利第6,287,860號中。 用於經口投與之組合物及調配物包含粉末或粒子、微顆 粒、奈米顆#、存於水或非水性介料之懸浮液或溶液、 膠囊、凝膠膠囊、藥囊、錠劑或微錠劑。可能期望使用增 稠劑、矯味劑、稀釋劑、乳化劑、分散助劑或黏合劑。較 佳口服調配物係彼等本發明募核苷酸與一或多種滲透增強 劑、表面活性劑及螯合劑聯合投與者。較佳表面活性劑包 含脂肪酸及/或其酯或鹽、膽汁酸及/或其鹽。較佳膽汁酸/ 鹽及脂肪酸及其應用進-步闡述於美國專利第6,287,86〇號 令,其以引用方式併入本文中。亦較佳者係渗透增強劑之 組合,例如,脂肪酸/鹽與膽汁酸/鹽之組合。尤佳之組合 係月桂酸之納鹽、癸酸及UDCA。其他滲透增強劑包含^ 氧乙烯冬月桂基醚、聚氧乙歸,_鯨蠟基醚。本發明之募 核苦酸可以粒子形式(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Μ複合劑及其應 用進一步闡述於美國專利第6,287,咖號中,其以引用方式 併入本文中。 用於非經腸、鞘内或心室内投與之組合物及調配物可包 含亦可含有緩衝劑、稀釋劑及其他適宜添加劑之I菌水容 液,該等其他適宜添加劑係(例如)但不限於渗透增強劑’、 載劑化合物及其他醫藥上可接受之載劑或賦形叫。 本發明某些實施例提供含有—或多種寡聚“物及一或 多種藉由非反義機制發揮_之其他化學治療劑之醫藥組 156342.doc -78· 201143782 ΟKits, Research Reagents, Diagnosis, and Therapy The compounds of the present invention are useful in the diagnosis, treatment, and prevention, and are useful as kits for research reagents and components. In addition, those skilled in the art typically employ antisense nucleotides that are capable of inhibiting gene expression with strong specificity to elucidate the function of a particular gene or to distinguish the function of each member of the biological pathway. For use in kits and diagnostics and in a variety of biological systems, the present sigma sigma (alone or in combination with other compounds or therapeutic agents) can be used as a tool in differential and/or combinatorial analysis to elucidate cells and The pattern of expression of a portion of the gene expressed in the tissue or the entire complementary sequence. The term "biological system" or "system" as used herein is defined to mean any organism, cell, cell culture or tissue that exhibits, or is sufficient to represent, the LIM homolog 2 (LHX2) gene product. Such systems include, but are not limited to, classes, transgenic animals, cells, cell cultures, & plants, grafts, and combinations thereof.移植 移植 理 , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , Analyze the resulting pattern for the degree of difference in genetic performance of 156342.doc -65- 201143782 correlation, signaling pathway, cell localization, degree of performance, size, structure or function of the gene being tested. Such assays 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), READS (digestion amplification of digestive (7), TOGA (general gene expression analysis), protein arrays and Proteomics, sequenced display (EST) sequencing, subtractive RNA fingerprinting (Surf), subtractive selection, differential display (DD), 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 can hybridize to nucleic acids encoding LIM homolog 2 (LHX2). For example, oligonucleotides that hybridize under the efficiency and conditions of an effective LHX2 modulator as disclosed herein are effective primers or probes and probes under conditions that facilitate gene amplification or detection, respectively. Needles can be used in methods that require specific detection of nucleic acid molecules encoding LHX2 and amplification of such nucleic acid molecules for detection or for use in other LHX2 studies. Hybridization of the antisense oligonucleotide (especially primers and probes) of the present invention with a nucleic acid encoding LHX2 can be detected by means of f. Such means may include coupling the enzyme to the nucleus (4), a radiolabeled oligonucleotide, or any other suitable means of detection. It is also possible to manufacture a kit that detects the (3) phantom content in the sample using an edge detection method. Those skilled in the art also utilize the specificity and sensitivity of antisense compounds in therapeutic applications. Antisense compounds have been used as therapeutic components to treat disease states in animals (3 humans). Anti-Stupidity Hanyi Nutrient Music is safe and effective 156342.doc -66 - 201143782 The field is being administered to humans and many clinical trials are currently being implemented. It is thus determined that the antisense compound can be a useful therapeutic modality that can be used in the treatment of cells, tissues, and animals, particularly humans. At the time of treatment, an animal (preferably a human) suspected of having a disease or condition treatable by modulating the expression of LHX2 polymorphic acid is treated by administering an antisense compound of the present invention. For example, in one non-limiting embodiment, the methods comprise the step of administering to a subject in need of treatment a therapeutically effective amount of an LHX2 modulator. The LHX2 modulator of the present invention is effective for regulating the activity of LHX2 or regulating the performance of LHX2 protein. In one embodiment, the activity or performance of LHX2 in the animal is inhibited by about 10% compared to the control. Preferably, the activity or performance of LHX2 in the animal is inhibited by about 30%. More preferably, the activity or performance of LHX2 in the animal is inhibited by 50% or more. Thus, an oligomeric compound can modulate the expression of LIM homolog 2 (LHX2) mRNA by at least 10%, at least 50%, at least 25%, at least 30%, at least 40%, at least 50%, at least as compared to a control group. 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 LIM homolog 2 (LHX2) in an animal is increased by about 10% compared to a control group. Preferably, the activity or performance of LHX2 in the animal is increased by about 30%. More preferably, the activity or performance of LHX2 in animals is increased by 50% or more. Thus, the oligomeric compound can modulate the performance of the LHX2 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 156,342.doc -67.201143782 85/〇, at least 90%, at least 95%, at least 98. /. At least 99%, or 100%. For example, the reduction in the expression of UM homologous sequence 2 (LHX2) 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 contain nucleic acid molecules encoding the LHX2 peptide and/or the LHX2 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 Another modification of the nucleotides of the present invention involves the chemical attachment of one or more moieties or conjugates that enhance the activity, cellular distribution or cellular uptake of the nucleotide to the nucleotide. The moieties or conjugates may comprise a coupling group covalently bonded to a functional group such as a primary or secondary hydroxyl group. The coupling group of the present invention comprises an intercalating agent, a reporter molecule, a polyamine, a polyamine, a polyethylene glycol, a polymymy, a group for enhancing the pharmacodynamic properties of the polymer, and an oligo reinforced group. a group of pharmacokinetic properties f. Typical coupling groups include cholesterol, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, luciferin, rh〇damine, coumarin, and dyes. . In the context of the present invention, a group that enhances pharmacodynamic properties comprises a group that improves uptake, enhances degradation resistance, and/or enhances sequence-specific hybridization with dry nucleic acids. In the context of the present invention, the group which enhances the metabolic kinetics of the drug H contains <RTI ID=0.0>>>>> A representative coupling group is disclosed in International Patent Application No. PCT/US92/09196, filed on Oct. 23, 1992, and hereby incorporated herein by reference. Coupling moieties include, but are not limited to, lipid moieties (eg, cholesterol moieties), cholic acid, thioethers (eg, hexyl-5-trityl mercaptan), thiocholesterol, aliphatic chains (eg, dodecanediol) Or undecyl residue), phospholipid (for example, di-hexadecyl-racemic-glycerol or triethylammonium 1,2-di-indole-hexadecyl-racemic-glycerol_3_H Phosphonate), polyamine or polyethylene glycol chain, or adamantane acetic acid, palmitoyl moiety, or octadecylamine or hexylamino-carbonyl-oxycholesterol moiety To active drug substances 'eg aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen ), (S)-(+)-pranoprofen ((S)-(+)-pranoprofen), carprofen, dansylsarcosine, 2,3,5-triiodobenzene Sulfonic acid, flufenamic acid, leucovorin, benzophenone, chlorinated D-Qin, diazep, ° D-omeemine (indomethicin), barbiturate, Cephalosporin, a tranexamine, an antidiabetic, an antibacterial or an antibiotic. Representative U.S. patents which teach the preparation of such nucleotide conjugates include, but are not limited to, U.S. Patent Nos. 4,828,979, 4,948,882, 5,218,105, 5,525,465, 5,541,313, 5.545. 730, 5, 552, 538, 5, 578, 717, 5.580.731, 5, 580, 731, 5, 591, 584, 156, 342. doc - 69 - 201143782 5, 109, 124, 5, 118, 802, 5, 138, 045, 5, 414, 077, 5, 486, 603, , No. 5, 512, 439, No. 5, No. 5, 598, pp. No. 5, 082, 013, No. 5, 082, 830, No. 5, 112, 963, No. 5, 214, 136, No. 5, 082, 830, No. 5,112, 963, No. 5, 214, 136, No. 5, 245, 022, No. 5, 258, 506, No. 5, 258, 506, No. 5, 262, 536, No. 5, 272, 250, No. 5, 292, 873 No. 5,317,098% Nos. 5, 371, 241, 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, Nos. 5,587,371 & 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. 5,108,921, 5,354,844, 5,416,016, 5,459,127, 5,521,291. '5, 543, 165, 156 342. doc • 70 · 201143782 5, 547, 932, 5, 583, 020, 5, 591, 721, 4, 426, 330, 4, 534, 899, 5, 013, 556, 5, 108, 921, 5, 213, 804, Nos. 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, 5,534,259, 5,543,152 No. 5,556,948, 5,580, 5 <75, and 5,595,756 each incorporated herein by reference. Although it is not necessary to administer antisense nucleoside acid in the context of the vector in order to modulate performance and/or function, embodiments of the invention relate to expression vector constructs for expression of antisense oligonucleotides, including promoters , 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 Q. 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 NO: 5 to 16) or in combination with a suitable protein, polysaccharide or lipid formulation. Further, a suitable nucleic acid delivery system comprises a viral vector, typically from adenosis, adeno-associated virus (AAV), helper cell-dependent adenosis, retrovirus, or Japanese liposome-hemagglutinating virus (HVJ) complex. A sequence of at least one of them. Preferably, the viral vector comprises a strong eukaryotic promoter (e.g., a cytomegalovirus (CMV) promoter) operably linked to 156342.doc-71-201143782 to a polynucleotide. Further preferred carriers comprise viral vectors, fusion proteins and chemical conjugates. The retroviral vector comprises Moloney mouse leukemia virus and HIV-based virus. A preferred HIV-based viral vector comprises at least two vectors, wherein the gag and pol genes are from the HIV genome and the env gene is from another virus. The dnA disease vector is preferred. Such vectors comprise a ρ〇χ vector (e.g., orthopoxvirus or acne virus vector), a herpesvirus vector (e.g., herpes simplex prion (HSV) vector), an adenoviral vector, and an adeno-associated viral vector. A compound of the invention encompasses any pharmaceutically acceptable salt, ester, or salt of such an ester, or is capable of providing (directly or indirectly) 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. For oligonucleosides, the preferred examples of the salts of the T-accepting salts and their use 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 (4), including vaginal and rectal delivery), lung (eg, by inhaling or spraying powder or aerosol, including by atomizer); , intranasal, epidermal and percutaneous, oral or parenteral administration. Parenteral 156342.doc -72- 201143782 administration includes intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial (eg, intrathecal or intraventricular) administration. For treatment of tissue in the central nervous system, administration can be by, for example, injection or infusion into the cerebrospinal fluid. The administration of antisense RNA in cerebrospinal fluid is described, for example, in "Methods for slowing familial ALS disease progression", U.S. Patent Application Publication No. 2007/0117772, the entire contents of which is incorporated herein by reference. When it is intended to administer an antisense oligonucleotide of the present invention to a cell of the central nervous system, one or more agents capable of promoting penetration of the subject antisense oligonucleotide through the blood-brain barrier may be administered. Injections can be made, for example, in the entorhinal cortex or hippocampus. The delivery of a neurotrophic factor by administering an adenoviral vector to a motor neuron in muscle tissue is described, for example, in "Adenoviral-vector-mediated gene transfer into medullary motor neurons", U.S. Patent No. 6,632,427, incorporated herein by reference. Incorporated herein. 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 in, for example, U.S. Patent No. 6,756,523, "Adenovirus vectors for the transfer of foreign genes into cells". In the central nervous system particularly in brain", which is incorporated herein by reference. Administration can be by rapid administration by injection or by slow infusion or administration of a sustained release formulation over a period of time. The title antisense oligonucleotide can also be linked or coupled to an agent that provides the desired pharmaceutical or pharmacodynamic properties. For example, an antisense oligonucleotide can be coupled to any substance known in the art to promote permeation or delivery in the blood-brain barrier (eg, an antibody to the iron-protein 156342.doc-73-201143782 transfer protein receptor), and It is administered by intravenous injection. The antisense compound can be linked to a viral vector which can, for example, render the antisense compound more effective and/or increase the delivery of the antisense compound in 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(+) galactose, D(+) lactose, D (+) xylose, dulcitol, muscle-inositol, L(-) fructose, D(-) mannitol, D(+) glucose, D(+) arabinose, D(-) arabinose, fiber two Sugar, D(+) maltose, D(+) raffinose, L(+) rhamnose, D(+) melibiose, D(-) ribose, ribitol, D(+) arabitol, L (-) arabitol, D(+) fucose, L(-) fucose, D (one) to threose, L(+) to threose, and L(-) tosuose; or amino acid, Including but not limited to glutamine, lysine, arginine, aspartame, aspartic acid, cysteine, glutamic acid, glycine, histidine, leucine, methyl sulfide Amine acid, phenylalanine, valine, 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. 6,294,042, "Material for passage through the blood-brain". "Blocker" and "Parotheral delivery systems", No. 6,936,589, the entire contents of each of which are incorporated herein by reference. The title antisense compound can be mixed, encapsulated, coupled or otherwise combined with other molecules, molecular structures or mixtures of compounds (eg, liposomes, molecules that target receptors, oral, rectal, topical or other formulations). To help ingest, distribute and / or absorb. For example, cationic lipids may be included in the formulation 156342.doc-74-201143782 to promote oligonucleoic acid uptake. One of the compositions shown to promote capture is LIPOFECTIN (available from GIBCO-BRL, Bethesda, MD). It is believed that at least one 2'-0-methoxyethyl modified nucleotide is particularly useful for oral administration. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids, and powders. Pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be required or desired. You can also use a wrap 避孕 condom, gloves, and the like. The pharmaceutical formulations of the present invention which may be conveniently presented in unit dosage form can be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing the active ingredient into association with a pharmaceutical carrier or excipient. In general, the formulations may be prepared by uniformly and intimately bringing the active ingredient into the liquid carrier or the fine solid carrier or both, and then, if necessary, shaping the product. The compositions of the present invention may be formulated into any of a number of possible dosage forms, such as, but not limited to, lozenges, 'knee pockets, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. The aqueous suspension may further contain a substance which increases the viscosity of the suspension, and contains, for example, sodium carboxymethyl cellulose, sorbitol and/or dextran. The suspension may also contain a stabilizer. The pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, foams, and formulations containing liposomes. The pharmaceutical compositions and formulations of the present invention may include one or more penetration enhancers, carriers, excipients or other active or inert ingredients of 156342.doc • 75- 201143782. 2 (4) (4) Dispersed in another liquid in the form of droplets (diameter through a heterogeneous system. The emulsion may contain additional components in addition to the dispersed phase, and may be present as a solution in the secondary phase, sub 7 aqueous phase, oil phase or itself As an active drug for the separation phase, the present application contains a microemulsion. The emulsion and its use are well known in the art and are described in U.S. Patent No. 6,287,860. The formulation of the present invention comprises a lipid. The term "liposome" as used in the present invention means a vesicle composed of two amphiphilic lipids in one or one spherical double layer. A lipid system monolayer or multilamellar vesicle a membrane having a self-lipophilic! raw material formation and an aqueous internal structure containing the composition to be delivered. The positively charged liposomes of the cationic oligosaccharide system are believed to interact with negatively charged ship molecules to form a stable complex. Liposomes that are pH-sensitive or negatively charged are believed to be captureable rather than complexed. Both ionic and non-cationic liposomes can be used to deliver DNA to cells. $Fat (4) also contains "spatial-stable" lipids. (d), the technique used herein." A button comprising one or more specific lipids. In the inclusion of liposomes, such characteristic lipids produce enhanced cycle life relative to liposomes lacking the particular lipids. Liposomes. Examples of sterically stabilized liposomes are those in which the vesicle-forming lipid portion of the liposome comprises one or more glycolipids or a hydrophilic or polymeric polymer (eg, polyethylene glycol (peg) moiety). Liposomes and their use are further described in U.S. Patent No. 6,287,860. The pharmaceutical formulations and compositions of this month may also contain surfactants. Table 156342.doc -76- 201143782 Surfactants in pharmaceutical products, The use of formulations and emulsions is well known in the art. Surfactants and their use are further described in U.S. Patent No. 6,287,860, which is incorporated herein by reference in its entirety herein in Agent to achieve effective delivery of nuclear 'acids, especially nucleotides. In addition to contributing to the diffusion of non-lipophilic drugs in the cell membrane, penetration enhancers also enhance lipophilic drugs Permeability of substances. Penetration enhancers can be classified into one of five categories: that is, surface active agents, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants ° penetration enhancers and Its use is further described in U.S. Patent No. 6,287,860, the disclosure of which is incorporated herein by reference in its entirety in its entirety in the the the the the the the the Preferred formulations for topical administration comprise a mixture of the present invention and a local delivery agent, for example, lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents, and surface active agents. 〇Preferred lipids and liposomes comprise neutral lipids and liposomes (eg diterpenoids _ phospholipids D〇PE ethanolamine, digestive phosphatidylcholine 1 DMPC, monostearyl phospholipids) Choline choline), negative lipids and liposomes (eg, a ugly phosphatidyl glycerol DMpG) and cationic lipids and lipids (eg, dioleyltetramethylaminopropyl D〇TAp and Two oil base Alcohol amines DOTMA). For topical or other administration, the oligonucleotides of the present invention may be encapsulated in or may form a complex with the liposome (especially cationic liposomes. Alternatively, the nucleophilic acid may be associated with lipids, especially Cationic lipid complexes. 156342.doc -77- 201143782 Preferred fatty acids and esters, pharmaceutically acceptable salts thereof, and uses thereof are further described in U.S. Patent No. 6,287,860. Compositions and formulations for oral administration The composition comprises a powder or a particle, a microparticle, a nanoparticle, a suspension or solution in a water or non-aqueous medium, a capsule, a gel capsule, a sachet, a lozenge or a micro-tablet. It may be desirable to use a thickener. A flavoring agent, a diluent, an emulsifier, a dispersing aid or a binder. Preferred oral formulations are those in which the present invention is administered in combination with one or more penetration enhancers, surfactants, and chelating agents. Preferred surfactants comprise fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Preferred bile acids/salts and fatty acids and their use are further described in U.S. Patent No. 6,287,86, which is incorporated herein by reference. Reference method incorporated into this Also preferred is a combination of penetration enhancers, for example, a combination of a fatty acid/salt and a bile acid/salt. Particularly preferred combinations are sodium laurate, citric acid and UDCA. Other permeation enhancers include oxyethylene Winter lauric ether, polyoxyethylene, _ cetyl ether. The nucleus acid of the present invention can be orally delivered in the form of particles (including spray dried granules, or composited to form micro or nano granules). The ruthenium complexing agent and its use are further described in U.S. Patent No. 6,287, the disclosure of which is hereby incorporated by reference herein in its entirety herein in I bacteria aqueous solution containing buffers, diluents and other suitable additives, such as, but not limited to, penetration enhancers', carrier compounds and other pharmaceutically acceptable carriers or excipients Certain embodiments of the present invention provide a pharmaceutical group containing one or more oligomeric species and one or more other chemotherapeutic agents that are exerted by a non-antisense mechanism 156342.doc -78· 201143782 Ο

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)、道諾徽素 (daunomycin)、更生黴素(dactinomycin)、多柔比星 (doxorubicin)、表柔比星(epirubicin)、伊達比星 (idarubicin)、依索比星(esorubicin)、博來黴素、馬填醢胺 (mafosfamide)、異環磷醯胺(ifosfamide)、胞嘴咬阿糖核苦 (cytosine arabinoside)、亞硝脲氮芥(bischloroethyl-nitrosurea)、白消安(busulfan)、絲裂黴素 C(mitomycin C)、放線菌素D(actinomycin D)、光輝徽素(mithramycin)、 潑尼松(prednisone)、經孕酬(hydroxyprogesterone)、睪酮 (testosterone)、他莫昔芬(tamoxifen)、達卡巴嗪(dacarbazine)、 丙卡巴肼(procarbazine)、六甲三聚氣胺(hexamethylmelamine)、 五甲三聚氰胺(pentamethylmelamine)、米托蒽西昆 (mitoxantrone)、安0丫唆(amsacrine)、苯丁酸氮芥· (chlorambucil)、環己亞石肖腺(methylcyclohexylnitrosurea)、 氮芬(nitrogen mustards)、美法舍(melphalan)、環磷醢胺 (cyclophosphamide)、6-疏嗓吟(6-mercaptopurine)、6-硫鳥 °票吟(6-thioguanine)、阿糖胞苷(cytarabine)、5-氮雜胞苷 (5-azacytidine)、經基腺(hydroxyurea)、喷司他丁 (deoxycoformycin) 、4-經基過氧環填酿胺(4-hydroxyperoxycyclo-phosphoramide) 、 5-氟尿0密咬(5- fluorouracil)(5-FU)、5- IL 去氧尿普(5-fluorodeoxyuridine) (5-FUdR)、甲胺蝶呤(MTX)、秋水仙鹼(colchicines)、紫杉 醇(taxol)、長春新驗(vincristine)、長春驗(vinblastine)、 156342.doc -79- 201143782 依託泊甙(etoposide)(VP-16)、三甲曲沙(trimetrexate)、伊 立替康(irinotecan)、托泊替坎(topotecan)、吉西他濱 (gemcitabine)、替尼泊甙(teniposide)、順鉑(cisplatin)及己 稀雌酌 (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及寡核苦 酸)、依序(例如,5-FU及寡核苷酸,隨後在一定時間之後 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 *(vidarabine)、阿昔洛韋(acyci〇v⑴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组合亦屬於本發明範圍内兩種 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(尤其寡核芽酸)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 篇同源序列2(LHX2)之特定反義序列,以:㈣為來 自另一核苷酸序列之區域。另__選 … ^为選擇為,本發明組合物可 :不同區域的反義化合物。反義化合物之諸多實例閣釋於 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—起或依序使用。 投藥: 156342.doc 201143782 據信,彼等熟f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’丨中治療過程可持續數天至數月,或直 士實現治癒或減輕疾病狀態為止。可藉由量測患者體内之 藥物積累來計算最適投藥方案。熟習此項技術者可容易地 確,最適劑量、投藥方法及重複速率。最適劑量可端視個 別寡核《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㈣物模财有效U⑽進行枯計。一般而 言,劑量為〇.〇1吨/kg體重至1〇〇g/kg體重,且可每日、每 週、每月或每年給予—次或更多次,或甚至每2至20年給 予-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,其 中以’I於0.01 pg/kg體重至100 g/k^重之間之維持劑量投 與募核苦酸,且每日投與-次或更多次至每20年投與_ 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2mg/kg體重、至少約3 mg/kg體重、 至v約4 mg/kg體重、至少約5 mg/kg體重、至少約6 mg/kg 體重至約7 mg/kg體重、至少約8 mg/kg體重、至少約9 mg/kg體重、至少約10 mg/kg體重、至少約15體重、 至/、勺20 mg/kg體重' 至少約25 體重、至少約 mg/kg體重、至少約35 mg/kg體重、至少約4〇 mg/kg體重、 至y,勺45 mg/kg體重、至少約5〇 mg/kg體重、至少約⑽ 156342.docCompound. Examples of such chemotherapeutic agents include, but are not limited to, cancer chemotherapeutic drugs, such as daunorubicin, daunomycin, dactinomycin, doxorubicin, and softness. Epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, sputum bite (cytosine arabinoside), bischloroethyl-nitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, splatter Prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethylmelamine, five Pentamethylmelamine, mitoxantrone, amsacrine, chlorambucil, methylcyclohexylnitrosurea, nitrogen mustar Ds), melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-azacytidine, hydroxyurea, deoxycoformycin, 4-hydroxyperoxycyclo-phosphoramide, 5-fluorourine 5-fluorouracil (5-FU), 5-IL-deoxyuronium (5-FUdR), methotrexate (MTX), colchicines, taxol , Changchun new test (vincristine), Changchun test (vinblastine), 156342.doc -79- 201143782 etoposide (VP-16), trimetrexate (trimetrexate), irinotecan (irinotecan), topotem 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 oligo-acidic acid), sequentially (eg, 5-FU and oligonucleotides, followed by MTX after a certain period of time) 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). 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, aciclovir (acyci〇v (1) 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 taken together or sequentially In another related embodiment, the compositions of the present invention may contain one or more antisense compounds (especially oligonucleotides) 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 may be a specific antisense sequence of the homologous sequence 2 (LHX2), wherein: (4) is a region from another nucleotide sequence. __Select... ^ is selected as the present invention The composition may be: antisense compounds of different regions. 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 or sequentially Use. Dosing: 156342. Doc 201143782 It is believed that those skilled in the art should be aware of the formulation of therapeutic compositions and their subsequent administration (administration). The administration depends on the severity and responsiveness of the condition to be treated. It lasts for several days to several months, or until the right to cure or reduce the disease state. The optimal drug administration plan can be calculated by measuring the drug accumulation in the patient. Those skilled in the art can easily determine the optimal dosage and dosage method. And the rate of repetition. The optimum dose may vary depending on the relative efficacy of the individual oligonuclei, and can usually be calculated based on the discovery of U(10) in vivo and in vivo (four). In general, the dose is 〇.〇 1 ton / kg body weight to 1 〇〇 g / kg body weight, and can be given daily, weekly, monthly or yearly - one or more times, or even every 2 to 20 years - times. Those skilled in the art The repetition rate and concentration of the drug can be easily estimated based on the measured residence time and concentration of the drug in the body fluid or tissue. After successful treatment, it can be expected that the patient can maintain the therapy to prevent recurrence of the disease state, wherein 'I is 0.01 A maintenance dose between pg/kg body weight and 100 g/k^ weight is administered to raise the bitter acid, and is administered _ times per day to - or more times to every 20 years. In the examples, the following is used Dosage of the drug to treat the 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, to v about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 Mg/kg body weight to about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 body weight, to/, spoon 20 mg/kg body weight At least about 25 body weight, at least about mg/kg body weight, at least about 35 mg/kg body weight, at least about 4 mg/kg body weight, to y, spoon 45 mg/kg body weight, at least about 5 mg/kg body weight, at least About (10) 156342.doc

•8U 201143782 mg/kg體重、至少約7〇 mg/kg體重、至少約8〇 mg/kg體重、 至少約90 mg/kg體重、或至少約1〇〇,體重。反義寡核 普酸之某些注射劑量闡述於(例如)美國專利第7,563观 之「Antisense m〇dulati〇n 〇fpTpiB up·—」中其全 部内容以引用方式併入本文中。 儘官上文已闡述本發明之各個實施例,然而,應理解, 該等實施例僅係、以實例方式而非限制方式提供。可根據本 文之揭不内容對所揭示實施例作出諸多改變,此並不背離 本發:之精神或範圍。因此,本發明之廣度及範圍不應受 上述實施例中之任一者限制。 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,在本申請案中引用之所有出版物及專利文件皆 係以引用方式併入,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圍内。 實例1 :對L1M同源序列2(LHX2)之反義核酸分子及/或 LHX2聚核普酸之有義股具有特異性之反義寡核苷酸的設計 如上所述,術語「對…具有特異性之寡核苷酸」或「靶 向…之寡核苷酸」係指具有如下序列之寡核苷酸:能夠 156342.doc •82- 201143782 與靶向基因之一部分形成穩定複合物,或(ϋ)能夠與靶向 基因之mRNA轉錄本之一部分形成穩定雙鏈體。 藉由使用電腦程式(例如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i9-25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 溫度(通常為50-60。〇 ’且不會形成自身二聚體或其他複雜 二級結構。 〇 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募核苷酸的選擇。使用該等• 8 U 201143782 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 90 mg/kg body weight, or at least about 1 inch, body weight. Some of the injectable doses of antisense oligonucleotides are described, for example, in "Antisense m〇dulati〇n 〇fpTpiB up." in U.S. Patent No. 7,563, the entire contents of which is incorporated herein by reference. The various embodiments of the present invention have been described hereinabove, however, it is understood that the embodiments are provided by way of example and not limitation. Many changes may be made to the disclosed embodiments without departing from the spirit and scope of the invention. Therefore, the breadth and scope of the present invention should not be limited by any of the above embodiments. All documents mentioned herein are incorporated herein by reference. All publications and patent documents cited in this application are hereby incorporated by reference in their entirety in the extent of the extent The Applicant does not recognize that any particular reference is a "previous technique" of its invention for each reference cited in this document. 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 ratios of the elements of the components shown and the substitutions thereof are within the scope of the embodiments of the present invention. Example 1: Design of antisense oligonucleotides specific for the antisense nucleic acid molecule of L1M homologous sequence 2 (LHX2) and/or the sense strand of LHX2 polynucleotide, as described above, the term "has "Specific oligonucleotide" or "targeted oligonucleotide" refers to an oligonucleotide having the following sequence: capable of forming a stable complex with one of the target genes, 156342.doc •82-201143782, or (ϋ) is capable of forming a stable duplex with a portion of the mRNA transcript of the targeted gene. Promote the selection of appropriate nucleotides by using a computer program (eg IDT AntiSense Design, IDT OligoAnalyzer) that automatically identifies sub-sequences of i9-25 nucleotides in each given sequence, such The sequence forms a hybrid with the target polynucleotide sequence and has a desired melting temperature (typically 50-60. 〇' and does not form a self-dimer or other complex secondary structure. 〇 by using an auto-alignment nucleic acid sequence and Computer programs indicating areas of homology or homology to further facilitate the selection of appropriate nucleotides. Use of 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 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 〇 度且對其他核酸序列顯示較低互補性程度的寡核苷酸。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。 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/ 或活性,且存在足夠互補性程度以避免反義4匕合物與非乾 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。 156342.doc -83· 201143782 本文所述寡核苷酸之雜交性質可藉由業内已知之一或多 個活體外分析進行測定。舉例而言,可藉由使用溶融曲線 分析測定靶天然反義分子及潛在藥物分子間之結合強度來 獲得本文所述募核苷酸之性質。 可使用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,溶融曲線分析)來估計粗天然反義分子及潛在 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)中之一者的適宜緩衝溶液。 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。The program compares 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 a range of genes and intergenic regions in a given genome allows for the selection of nucleic acid sequences of the appropriate degree of specificity for the target gene. Such procedures allow for the selection of oligonucleotides that exhibit a high degree of complementarity to a target nucleic acid sequence in a given genome and a lower degree of complementarity to other nucleic acid sequences. Those skilled in the art will recognize that a wide range of gene regions suitable for use in the present invention can be selected. 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/or activity, and is sufficiently complementary to avoid antisense 4 conjugate Non-specific with non-dry nucleic acid sequences under conditions where specific binding is desired to occur (ie, under physiological conditions in the case of in vivo analysis or therapeutic treatment, and under conditions of analysis in the context of in vitro analysis) Combine. 156342.doc -83· 201143782 The hybridization properties of the oligonucleotides described herein can be determined by one or more in vitro assays known in the art. For example, the nature of the nucleotides described herein can be obtained by measuring the binding strength between the target natural antisense molecule and the potential drug molecule using a melting curve analysis. Any of the established methods of measuring the strength of the intermolecular interaction (e.g., melting curve analysis) can be used to estimate the binding strength between the crude natural antisense molecule and the potential drug molecule (molecule). 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.

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 新形成複合物緩慢加熱至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 156342.doc •84- 201143782 量成反比。可使用與套組相容之實時PCR儀器(例如ABI's StepOne Plus Real Time PCR System 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軸)與溫度(X軸)的圖線來構建熔融峰。分析 數據以確定自dsDNA複合物快速轉變成單鏈分子時之溫 度。此溫度稱為Tm且與兩個分子間之相互作用強度成正 比。通常,Tm高於40°C。 實例2 : LHX2聚核苷酸之調節 使用反義募核苦酸處理HepG2細胞 實例2中所用之所有反義寡核苷酸皆如實例1中所述進行 設計。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1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 曱基、A及C上之苯曱醯基及G上之N-2-異丁醯基)。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 長之寡核苷酸鏈中添加新鹼基:1)使用三氣乙酸自固定核 156342.doc -85- 201143782 苦酸之5’氧處去除保護基團;2)使用四嗤使經固定核苷酸 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)洗務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寡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—甲基咪唑對游離5, 羥基實施乙醯化來達成封端;4)為穩定核苷酸間之鍵,使 用埃及水(若欲產生攝酸二酯鍵)、或以叫以#試劑(3H1,2_ 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募核 苦酸並實施去保護。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-10-〇l〇-CV)+l〇% FBS(MediateCh 目錄編號:MT35-011-CV)+青黴素(penicillin)/鏈黴素 (streptomycin)(Mediatech目錄編號:Mt3〇-〇〇2-CI))中生長 來自ATCC之HepG2細胞(目錄編號:HB-8065)。在實驗前 一天,以0.5xl04/ml之密度將細胞再次平鋪於6孔板中並在 3 7°C及5% C〇2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;東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2〇 μΜ。將2 μΐ 156342.doc -86 - 201143782 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HepG2細胞之一個孔中。使用包含2 μΐ水代替募 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3-18 h之後,將培養基更換為新鮮生長培養 基。添加反義募核苷酸48 h後,去除培養基,且使用來自 Promega 之 SV Total RNA Isolation System (目錄編號: Z3105)或來自 Qiagen之RNeasy Total RNA Isolation套組(目 錄編號:74181)根據製造商說明書自細胞提取RNA。將 600 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:43688 13)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 : Hs00180351_ml(Human) 5 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 值來計算使用反義募核苷酸處理後基因表現的變化倍數。 .敏肩:實時PCR結果顯示,在使用設計為LHX2反義 156342.doc -87- 201143782 smaplerbu.aApr07之寡聚物處理48 h之後,HepG2細胞中之 LHX2 mRNA之含量顯著增加(圖1)。 使用反義募核苷酸處理373細胞 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/鏈黴素(Mediatech目錄編號:MT30-002-CI))中生長來 自ATCC之3T3細胞(目錄編號:CRL-1658)。在實驗前一 天,以1.5xl05/ml之密度將細胞再次平鋪於6孔板中並在 37°C及5°/。C〇2下培育。在實驗當天,將6孔板中之培養基 更換為新鮮生長培養基。將所有反義寡核苷酸稀釋至濃度 為20 μΜ。將2 μΐ此溶液與4〇〇 μΐ 〇pti-MEM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,且施加至 6孔板中具有3T3細胞之每一孔中。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。(:及 5% C02下培育3-18 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,且使用來自 Promega之 SV Total RNA Isolation System (目錄編號: Z3 105)或來自 Qiagen之 RNeasy Total RNA Isolation套組(目 錄編號:741 81)根據製造商說明書自細胞提取RNA。將 600 ng 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:43 688 13)實施的逆轉錄反應中,如製造商方 156342.doc *· 88 · 201143782 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: 43695 10)及由 ABI 設計之引物 / 探針(Applied Biosystems Taqman Gene Expression Assay : Mm008 39783_ml (Mouse),Applied Biosystems 公司,Foster City CA)來監 測基因表現。使用StepOne Plus實時PCR機器(Applied Bio systems)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)。 ,结耒:實時PCR結果顯示,在使用設計為LHX2反義分子 之寡聚物處理48 h之後,3T3細胞中之LHX2 mRNA之含量 顯著增加(圖2)。 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圍或含義。 【圖式簡單說明】 圖1展示與對照組相比,在使用利用Lipofectamine 2000 引入之硫代磷酸酯寡聚物處理後48小時時,HepG2細胞中 156342.doc -89- 201143782 LHX2 mRNA之變化倍數及標準偏差《實時PCR結果顯 示,在使用設計為LHX2反義smaplerbu.aApr07之募聚物中 之一者處理後48 h時,HepG2細胞中之LHX2 mRNA之含量 顯著增加。表示為CUR-1560至CUR-1563之條形分別對應 於使用SEQ ID NO: 5至8處理之試樣。 圖2展示與對照組相比,在使用利用Lipofectamine 2000 引入之硫代磷酸酯寡聚物處理後48小時時,3T3細胞中 LHX2 mRNA之變化倍數及標準偏差《實時PCR結果顯 示,在使用設計為LHX2小鼠反義分子之募聚物中之一者 處理後48 h時,3T3細胞中之LHX2 mRNA之含量顯著增 加。表示為CUR-1588至CUR-1595之條形分別對應於使用 SEQ ID NO: 9至16處理之試樣。 序列表說明-SEQ ID NO: 1 :智人LIM同源序列2 (LHX2),mRNA(NCBI登錄號:NM_004789) ; SEQ ID NO: 2 :天然LHX2反義序列(smaplerbu.aApr07) ; SEQ ID NO: 3 :小鼠天然 LHX2反義序列(fl〇ysla.aSep07) ; SEQ ID NO·· 4 :小鼠天然LHX2反義序列(CJ098619) ; SEQ ID NO: 5至 16 :反義寡核苷酸。*表示硫代磷酸醋鍵。 156342.doc 90- 201143782 序列表 <11〇>美商歐科可娜有限貴任公司 <120>以針對LIM同源序列2(LHX2)之天然反義轉錄本之抑制作用治療LHX2相關疾病 <130> LHX2 <140〉100117637 <141〉2011/05/19 <150> 61/346,284 <151〉2010/05/19 <160> 16 <170> Patent In version 3.5 <210> 1 <21!> 2416 <212> DNA <213>智人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 of the 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 fluorescence intensity is inversely proportional to the amount of dsDNA present in the reaction 156342.doc •84- 201143782. 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. The data was analyzed to determine the temperature at which the dsDNA complex rapidly converted to a single-stranded molecule. This temperature is called Tm and is proportional to the strength of the interaction between the two molecules. Typically, the Tm is above 40 °C. Example 2: Modulation of LHX2 Polynucleotide HepG2 cells were treated with antisense nucleoside acid. All antisense oligonucleotides used in Example 2 were designed as described in Example 1. The manufacturer (IDT Corporation, Coralville, IA) was instructed to manufacture the designed phosphorothioate-bonded oligonucleotides and to provide the designed phosphorothioate analogs shown in Table 1. The star symbol between the nucleotides indicates the presence of a phosphorothioate bond. The oligonucleotides required for the experiments in Example 2 can be synthesized using any suitable state of the art, such as the method used in IDT: the use of phosphorous ruthenium on solid supports (e.g., 5 micron fixed glass beads (CPG)) Amine monomer (common nucleotides, all of which are protected by protecting groups such as triphenylsulfonyl groups on sugars, benzoquinones on A and C, and N-2-isobutyl) on G) . The protecting group prevents undesired reactions from occurring during oligonucleotide synthesis. The protecting group is removed at the end of the synthesis process. The initial nucleotide is attached to the solid support via 3' carbon and synthesized in the 3' to 5' direction. Add a new base to the growing oligonucleotide strand in the following 4 steps: 1) Remove the protecting group from the 5' oxygen of the bitter acid using a tri-glycolic acid from the immobilized core 156342.doc -85- 201143782; 2) Use The tetranucleotide couples the immobilized nucleotide to the next nucleotide in the sequence; the reaction is continued via the tetrazolylphosphinamide intermediate; 3) the unreacted free nucleotide and the reaction are removed by washing By-product and capping the unreacted fixed oligonucleotide to prevent its participation in the next round of synthesis; by using acetic anhydride and N-methylimidazole, the free 5, hydroxyl group is acetylated to achieve capping; 4) Stabilize the bond between nucleotides, use Egyptian water (if you want to produce acid diester bonds), or use # reagent (3H1,2_ benzodithiol-3-keto-1,1-dioxide) (If a phosphorothioate bond is desired) to oxidize phosphorus. 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. Upon synthesis of the complete sequence, the acid is removed from the solid support using ammonium hydroxide at elevated temperature 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-10-〇l〇-CV) + l at 37 ° C and 5% CO 2 〇% FBS (MediateCh catalog number: MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number: Mt3〇-〇〇2-CI)) grew HepG2 cells from ATCC (catalog) No.: HB-8065). One day before the experiment, the cells were plated again in a 6-well plate at a density of 0.5 x 104 / ml and incubated overnight at 37 ° C and 5% C 〇 2 . On the day of the experiment, the medium in the 6-well plate was replaced with fresh growth medium. Oligonucleotides delivered by the manufacturer in the form of Donggan were diluted to a concentration of 2 〇 μΜ in deionized water without RNAse/DNAse. 2 μΐ 156342.doc -86 - 201143782 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, then drop by drop Apply to a well of a 6-well plate with HepG2 cells. A similar mixture containing 2 μM water instead of the nucleotide solution was used as a mock transfection control group. After incubation for 3-18 h at 37 ° C and 5% CO 2 , the medium was changed to a fresh growth medium. After 48 h of antisense nucleotide addition, the medium was removed and the SV Total RNA Isolation System from Promega (catalog number: Z3105) or the RNeasy Total RNA Isolation kit from Qiagen (catalog number: 74181) was used according to the manufacturer's instructions. RNA is extracted from cells. 600 ng of extracted RNA was added to the reverse transcription reaction using the Verso cDNA kit from Thermo Scientific (catalog number: AB1453B) or the high capacity cDNA reverse transcription kit (catalog number: 43688 13), as in the manufacturer's protocol. Said. 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: Hs00180351_ml (Human) 5 Applied Biosystems, Foster City CA) to monitor gene expression. The following PCR cycle was 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 °C) Hold at °C for 1 min). The fold change in gene expression after treatment with antisense nucleotides was calculated based on the difference between the 18S normalized dCt values between the treated and mock transfected samples. Sensitive shoulder: Real-time PCR results showed that the content of LHX2 mRNA in HepG2 cells was significantly increased after 48 h treatment with an oligomer designed to be LHX2 antisense 156342.doc -87-201143782 smaplerbu.aApr07 (Fig. 1). 373 cells were treated with antisense nucleotides at 37 ° C and 5 ° /. C02 under growth medium (MEM/EBSS (Hyclone catalog number: SH30024, or Mediatech catalog number: MT-10-010-CV) + 10% FBS (Mediatech catalog number: MT35-011_CV) + penicillin/streptomycin (Mediatech) Catalog number: MT30-002-CI)) 3T3 cells from ATCC (Catalog No.: CRL-1658) were grown. One day before the experiment, the cells were plated again in a 6-well plate at a density of 1.5 x 105/ml at 37 ° C and 5 ° /. C〇2 is cultivated. On the day of the experiment, the medium in the 6-well plate was changed to fresh growth medium. All antisense oligonucleotides were diluted to a concentration of 20 μΜ. 2 μL of this solution was incubated with 4 μμΐ 〇pti-MEM medium (Gibco Cat. No. 31985-070) and 4 μL Lipofectamine 2000 (Invitrogen Cat. No. 11668019) at room temperature for 20 min and applied to 6 wells. The plate has 3T3 cells in each well. A similar mixture containing 2 μM water instead of the oligonucleotide solution was used as a mock transfection control group. At 37. (: and after culturing for 3-18 h at 5% C02, the medium was changed to fresh growth medium. After adding antisense oligonucleotide for 48 h, the medium was removed and the SV Total RNA Isolation System from Promega was used. Z3 105) or RNeasy Total RNA Isolation kit from Qiagen (Catalog No. 741 81) Extract RNA from cells according to the manufacturer's instructions. Add 600 ng of RNA to the Verso cDNA kit from Thermo Scientific (Catalog No.: AB1453B) Or the reverse transcription reaction carried out by the high-capacity cDNA reverse transcription kit (catalog number: 43 688 13), as described in the manufacturer's section 156342.doc *· 88 · 201143782. The cDNA from this reverse transcription reaction is used. Real-time PCR uses ABI Taqman Gene Expression Mix (catalog number: 43695 10) and ABI designed primer/probe (Applied Biosystems Taqman Gene Expression Assay: Mm008 39783_ml (Mouse), Applied Biosystems, Foster City CA) to monitor gene expression The following PCR cycle was performed using a StepOne Plus real-time PCR machine (Applied Bio systems): kept at 50 ° C for 2 min, Hold at 95 ° C for 10 min, 40 cycles (15 seconds at 95 ° C, 1 min at 60 ° C)., Crust: Real-time PCR results show that the design is LHX2 antisense molecule After 48 h of oligomer treatment, the amount of LHX2 mRNA in 3T3 cells was significantly increased (Fig. 2). Although the invention has been illustrated and described in terms of one or more embodiments, those skilled in the art are reading and understanding Equivalent changes and modifications may be made in the specification and the drawings. Further, 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. This disclosure may be desirable and advantageous for any given or specific application. The Abstract of the Disclosure allows the reader to quickly ascertain the nature of the technical disclosure. This summary is based on the understanding that it is not intended to be construed Or limit the scope or meaning of the following patent application scope. [Simplified Schematic] Figure 1 shows the use of phosphorothioate oligosaccharide introduced using Lipofectamine 2000 compared to the control group. The fold change and standard deviation of 156342.doc-89-201143782 LHX2 mRNA in HepG2 cells at 48 hours after treatment. Real-time PCR results showed that one of the concentrators designed using LHX2 antisense smaplerbu.aApr07 was treated. At 48 h after the hour, the content of LHX2 mRNA in HepG2 cells increased significantly. The bars denoted as CUR-1560 to CUR-1563 correspond to the samples treated with SEQ ID NOS: 5 to 8, respectively. Figure 2 shows the fold change and standard deviation of LHX2 mRNA in 3T3 cells at 48 hours after treatment with the phosphorothioate oligomer introduced by Lipofectamine 2000 compared with the control group. Real-time PCR results show that the design is The content of LHX2 mRNA in 3T3 cells was significantly increased 48 h after treatment of one of the LHX2 mouse antisense molecules. The bars denoted as CUR-1588 to CUR-1595 correspond to the samples treated with SEQ ID NOS: 9 to 16, respectively. BRIEF DESCRIPTION OF THE SEQUENCE LISTING - SEQ ID NO: 1 : Homo sapiens LIM homolog 2 (LHX2), mRNA (NCBI accession number: NM_004789); SEQ ID NO: 2: native LHX2 antisense sequence (smaplerbu.aApr07); SEQ ID NO : 3 : mouse native LHX2 antisense sequence (fl〇ysla.aSep07); SEQ ID NO.. 4: mouse native LHX2 antisense sequence (CJ098619); SEQ ID NOs: 5 to 16: antisense oligonucleotide . * indicates thiophosphate vinegar bond. 156342.doc 90- 201143782 Sequence Listing <11〇> American Okokena Limited Company <120> Treatment of LHX2 by Inhibition of Natural Antisense Transcript Against LIM Homologous Sequence 2 (LHX2) Disease <130> LHX2 <140>100117637 <141>2011/05/19 <150> 61/346,284 <151>2010/05/19 <160> 16 <170> Patent In version 3.5 <;210> 1 <21!> 2416 <212> DNA <213> Homo sapiens

<300> <3〇8> mjmm3 <309> 2011-03-12 <313> (1)..(2416) <400> 1 gactgcagag ccggggctgg gctaggcgcg cgcttggaga gcattgcgcg cggctgggcc 60 cgcggccggc ggctcctcct cccactctgc tcctcctctt ttttctcctc ctccacctcc 120 tcctccgcct cctcctcctc ctcttcctcc icctcttcaa ttctcccggt ggctcgactc 180 ggctcgcagg cttcggagaa acccctactc cagtcgccga ctcagcgccc aagagggtcg 240 ccttgggctg ggggcgcacc ccagggaggg gaggggtcca ggcagctggg ccgccgcgga 300 cacctagcgg cttcagggtg aaccccgacc gcagccgtcg ccgcctcggg cagagtttgc 360 gcccttgctt tgcgccccgg gcgctgaagc cgggcgggcg atgcccgcgg cgtgaaagcg 420 cccgcggcgg gcgccgacct ctgtcctagt ctcctgctcc ccccgccccg cttgtcccgt 480 gcccttgtga ccctggcttt ggcgccgtcg cccaggcgcc ccgcaatgta gctgcccctg 540 cgcctcggcg ggaggcgtcc tgccccgcga gcgcccgggg cccggagccc ggcctggggg 600 ctcagccgag ctcgggcggg gccggggccg cggtggcgat gcaccgggcc cgttagcgcc 660 aggagcgcca ggcagctgag gcggggggca agccctccct cggaggagcc gcgcccccgg 720 ccccgccggt cccgccgcga tgctgttcca cagtctgtcg ggccccgagg tgcacggggt 780 catcgacgag atggaccgca gggccaagag cgaggctccc sccatcagct ccgccatcga 840 ccgcggcgac accgagacga ccatgccgtc catcagcagt gaccgcgccg cgctgtgcgc 900 cggctgcggg ggcaagatct cggaccgota ctacctgctg gcggtggaca agcagtggca 960 catgcgctgc ctcaagtgct gcgagtgcaa gctcaacctg gagtcggagc tcacctgttt 1020 cagcaaggac ggtagcatct actgcaagga agactactac aggcgcttct ctgtgcagcg 1080 ctgcgcccgc tgccacctgg gcatctcggc ctcggagatg gtgatgcgcg ctcgggactt 1140 ggtttatcac ctcaactgct tcacgtgcac cacgtgtaac aagatgctga ccacgggcga 1200 ccacttcggc atgaaggaca gcctggtcta ctgccgcttg cacttcgagg cgctgctgca 1260 gggcgagtac cccgcacact tcaaccatgc cgacgtggca gcggcggccg ctgcagccgc 1320 ggcggccaag agcgcggggc tgggcgcagc aggggccaac cctctgggtc ttccctacta 1380 caatggcgtg ggcactgtgc agaaggggcg gccgaggaaa cgtaagagcc cgggccccgg 1440 156342-序列表.doc 201143782 tgcggatctg gcggcctaca acgctgcgct aagctgcaac gaaaacgacg cagagcacct 1500 ggaccgtgac cagccatacc cgagcagcca gaagaccaag cgcatgcgca cgtccttcaa 1560 gcaccaccag cttcggacca tgaagtctta ctttgccatt aaccacaacc ccgacgccaa 1620 ggacttgaag cagctcgcgc aaaagacggg cctcaccaag cgggtcctcc aggtctggtt 1680 ccagaacgcc cgagccaagt tcaggcgcaa cctcttacgg caggaaaaca cgggcgtgga 1740 caagtcgaca gacgcggcgc tgcagacagg gacgccatcg ggcccggcct cggagctctc 1800 caacgcctcg ctcagcccct ccagcacgcc caccaccctg acagacttga ctagccccac 1860 cctgccaact gtgacgtccg tcttaacttc tgtgcctggc aacctggagg gccatgagcc 1920 tcacagcccc tcacaaacga ctcttaccaa ccttttctaa tgactcgcaa cccctcaccc 1980 cacaatttct ttaaaaaaga aattatcttt agttgaattc caagtgtalt ttaaaataga 2040 ggctttgagc aactaaciaa ccacatttta ggztctcgcc tggaaacaga ggtaaaaaaa 2100 agaagtgtgc gcccggctaa tgcagcggtg t£gaccgag£ aacaacitgg aagatctacc 2160 tgcaacacaa catttgtgtc actgtacagt ittgtggact gagcgaggaa aaacaacaaa 2220 taatttaagt tggctagagc ttctgtattt tcaaagactg ccacgtgcct taggaatact 2280 gttttatctc catactttgg atgacttgtt catttttctc tccctctttt tctctgtata 2340 ttiatgacca gagcaaaaat gtaaaaaaca aaaaaaacaa caaaaaaagt ttgttacttt 2400 gaatagtcct aaaaag 2416 <210> 2 <211> 2145 <212> DMA <^13>智人 <400> 2 tacatatagg cacgcgattc cgtcgccacc tctcggacca gcagcccacg tccaacgctg 60 ggccgacccc agatacacag gcagtcggga ttcccgcccg gtgcgcttgt ctattcatcc 120 ctctgcgtca ggctgggacg cgctccgtct gtaaaaggct caaacgcatc tcccgccgcg 180 gggcgggatc taggggccca ggccccgggg tccagaggcg ggtaactttg ctaatctccc 240 ccagcggcgg ggaacgtcgc gcaaccgctg agccctgtcc gccgagacta aacaagcaga 300 ggaacaggct gtaaacacac atcctgacac gcaggsatgt tgctcttgga gaatgtgaag 360 acagtttgct tcttgacaag cgagcgaacg ggcgcccaga tttttgcagc ctttccgagt 420 ctcccccgag ggagaggcgg cagagaaaac gccggatttg ggagccacca gggaaggatc 480 cgcgcagggg agccgccctc cttggcccca gacccgcctg cctggggccc cctttgctca 540 ctgtcaatgt atggtctgaa gctctgagga tggtgctggg gactggggtc gggggaagcc 600 tcttgaataa taactgcaaa gaagaaagag gcgagaacgt ctccctaacc ttgaagcaga 660 aaggactgtg ttcttaaagc tgttggctgc agtcacaggg ccagttgccc gcctctgttc 720 cctgagtaaa gtgtaacatc ttctgtcctc ctggctt|:ct tgcaccattc a2caaattat 780 actccttcct taccaaagtg ggaatgctca gggaagtgtg igtgtgtgtg tgtgtgtgtg 840 tgtgtcccct ctgcactgag gctgctgtta gagatgttac caatttaaac cttccagaat 900 cctggaggtt tacattttgt aaagggaggg gacactcctg gactgtcaca atcccaattc 960 tggttagagt ggcagggact gaacaaagcc accaacatgc aaaagtccat ctccagagta 1020 -2· 156342-序列表.doc 201143782 ❹ gcatgcatgc taacctctgt tgcagggtta aaaagctctg tcattcagca tcagttcagg cacagaaaat ccgaagagga cagagagaag cgcctccctg cctgttaaat atgcatcagg gtattaatat atgcaatttt gagggaaatg caaagtggtg iaggtcagag gtatgtatct ttttctgaaa ccccaggaaa ctcctgtttg gggttatctc tatcttcaat aagcagggag caugctatc ctgcttatca accccgtgtc cccaattcct ggcgtcagct gcactgaact aaacgtggag tstattaaat gtcaaggttt aggggctgct acaaaatglt attgttcacc aggtcctgct gaacaaitat cccccagtgg taaggcagag caagaaaggg gaggacccac tgttttatat tctttcttta ccgctatcga tgtgacatta cctcctcttc gagcaggttg taaaatgaaa gttggcagcc gttatgagaa ttatctgtga ggcatgtgac caattttatt tcagcaaaag taaatcacat aacaaaatac gatatgcttc gaagggtgat tggggtgagg acacacacac tgatgcgasa cgaaatcagc caataaaaat gaatagataa iccctgcagc gggaactaac cacgaagtgt cttttcctcc agtaaaggct ttatgattcc atgtgtttta acagggaatt gatttaccat tgtcaacaat actatagcta ctgcactttc tccttgccac ctgagtcaca ctttcccagg ggctgtcagt tcattgcctt<300><3〇8> mjmm3 <309> 2011-03-12 <313> (1)..(2416) <400> 1 gactgcagag ccggggctgg gctaggcgcg cgcttggaga gcattgcgcg cggctgggcc 60 cgcggccggc ggctcctcct cccactctgc tcctcctctt ttttctcctc ctccacctcc 120 tcctccgcct cctcctcctc ctcttcctcc icctcttcaa ttctcccggt ggctcgactc 180 ggctcgcagg cttcggagaa acccctactc cagtcgccga ctcagcgccc aagagggtcg 240 ccttgggctg ggggcgcacc ccagggaggg gaggggtcca ggcagctggg ccgccgcgga 300 cacctagcgg cttcagggtg aaccccgacc gcagccgtcg ccgcctcggg cagagtttgc 360 gcccttgctt tgcgccccgg gcgctgaagc cgggcgggcg atgcccgcgg cgtgaaagcg 420 cccgcggcgg gcgccgacct ctgtcctagt ctcctgctcc ccccgccccg cttgtcccgt 480 gcccttgtga ccctggcttt ggcgccgtcg cccaggcgcc ccgcaatgta gctgcccctg 540 cgcctcggcg ggaggcgtcc tgccccgcga gcgcccgggg cccggagccc ggcctggggg 600 ctcagccgag ctcgggcggg gccggggccg cggtggcgat gcaccgggcc cgttagcgcc 660 aggagcgcca ggcagctgag gcggggggca agccctccct cggaggagcc gcgcccccgg 720 ccccgccggt cccgccgcga tgctgttcca cagtctgtcg ggccccgagg tgcacggggt 78 0 catcgacgag atggaccgca gggccaagag cgaggctccc sccatcagct ccgccatcga 840 ccgcggcgac accgagacga ccatgccgtc catcagcagt gaccgcgccg cgctgtgcgc 900 cggctgcggg ggcaagatct cggaccgota ctacctgctg gcggtggaca agcagtggca 960 catgcgctgc ctcaagtgct gcgagtgcaa gctcaacctg gagtcggagc tcacctgttt 1020 cagcaaggac ggtagcatct actgcaagga agactactac aggcgcttct ctgtgcagcg 1080 ctgcgcccgc tgccacctgg gcatctcggc ctcggagatg gtgatgcgcg ctcgggactt 1140 ggtttatcac ctcaactgct tcacgtgcac cacgtgtaac aagatgctga ccacgggcga 1200 ccacttcggc atgaaggaca gcctggtcta ctgccgcttg cacttcgagg cgctgctgca 1260 gggcgagtac cccgcacact tcaaccatgc cgacgtggca gcggcggccg ctgcagccgc 1320 ggcggccaag agcgcggggc tgggcgcagc aggggccaac cctctgggtc ttccctacta 1380 caatggcgtg ggcactgtgc agaaggggcg gccgaggaaa cgtaagagcc cgggccccgg 1440 156342- sequence Listing .doc 201143782 tgcggatctg gcggcctaca acgctgcgct aagctgcaac gaaaacgacg cagagcacct 1500 ggaccgtgac cagccatacc cgagcagcca gaagaccaag cgcatgcgca cgtccttcaa 1560 gcaccaccag Cttcggacca tgaagtctta ctttgccatt aac cacaacc ccgacgccaa 1620 ggacttgaag cagctcgcgc aaaagacggg cctcaccaag cgggtcctcc aggtctggtt 1680 ccagaacgcc cgagccaagt tcaggcgcaa cctcttacgg caggaaaaca cgggcgtgga 1740 caagtcgaca gacgcggcgc tgcagacagg gacgccatcg ggcccggcct cggagctctc 1800 caacgcctcg ctcagcccct ccagcacgcc caccaccctg acagacttga ctagccccac 1860 cctgccaact gtgacgtccg tcttaacttc tgtgcctggc aacctggagg gccatgagcc 1920 tcacagcccc tcacaaacga ctcttaccaa ccttttctaa tgactcgcaa cccctcaccc 1980 cacaatttct ttaaaaaaga aattatcttt agttgaattc caagtgtalt ttaaaataga 2040 ggctttgagc aactaaciaa ccacatttta ggztctcgcc tggaaacaga ggtaaaaaaa 2100 agaagtgtgc gcccggctaa tgcagcggtg t £ gaccgag £ aacaacitgg aagatctacc 2160 tgcaacacaa catttgtgtc actgtacagt ittgtggact gagcgaggaa aaacaacaaa 2220 taatttaagt tggctagagc ttctgtattt tcaaagactg ccacgtgcct taggaatact 2280 gttttatctc catactttgg atgacttgtt catttttctc tccctctttt tctctgtata 2340 ttiatgacca gagcaaaaat gtaaaaaaca aaaaaaacaa caaaaaaagt ttgttacttt 2400 gaatagtcct aaaaag 2416 <210> 2 <211> 21 45 < 212 > DMA < ^ 13 > Homo sapiens < 400 > 2 tacatatagg cacgcgattc cgtcgccacc tctcggacca gcagcccacg tccaacgctg 60 ggccgacccc agatacacag gcagtcggga ttcccgcccg gtgcgcttgt ctattcatcc 120 ctctgcgtca ggctgggacg cgctccgtct gtaaaaggct caaacgcatc tcccgccgcg 180 gggcgggatc taggggccca ggccccgggg tccagaggcg ggtaactttg ctaatctccc 240 ccagcggcgg ggaacgtcgc gcaaccgctg agccctgtcc gccgagacta aacaagcaga 300 ggaacaggct gtaaacacac atcctgacac gcaggsatgt tgctcttgga gaatgtgaag 360 acagtttgct tcttgacaag cgagcgaacg ggcgcccaga tttttgcagc ctttccgagt 420 ctcccccgag ggagaggcgg cagagaaaac gccggatttg ggagccacca gggaaggatc 480 cgcgcagggg agccgccctc cttggcccca gacccgcctg cctggggccc cctttgctca 540 ctgtcaatgt atggtctgaa gctctgagga tggtgctggg gactggggtc gggggaagcc 600 tcttgaataa taactgcaaa gaagaaagag gcgagaacgt ctccctaacc ttgaagcaga 660 aaggactgtg ttcttaaagc tgttggctgc agtcacaggg ccagttgccc Gcctctgttc 720 cctgagtaaa gtgtaacatc ttctgtcctc ctggctt|:ct tgcaccattc a2caaattat 780 actccttcct taccaaagtg ggaatgctca ggga agtgtg igtgtgtgtg tgtgtgtgtg 840 tgtgtcccct ctgcactgag gctgctgtta gagatgttac caatttaaac cttccagaat 900 cctggaggtt tacattttgt aaagggaggg gacactcctg gactgtcaca atcccaattc 960 tggttagagt ggcagggact gaacaaagcc accaacatgc aaaagtccat ctccagagta 1020 -2 · 156342- Sequence Listing .doc 201143782 ❹ gcatgcatgc taacctctgt tgcagggtta aaaagctctg tcattcagca tcagttcagg cacagaaaat ccgaagagga cagagagaag cgcctccctg cctgttaaat atgcatcagg gtattaatat atgcaatttt gagggaaatg caaagtggtg iaggtcagag gtatgtatct ttttctgaaa ccccaggaaa ctcctgtttg gggttatctc tatcttcaat aagcagggag caugctatc ctgcttatca accccgtgtc cccaattcct ggcgtcagct gcactgaact aaacgtggag tstattaaat gtcaaggttt aggggctgct acaaaatglt attgttcacc aggtcctgct gaacaaitat cccccagtgg taaggcagag caagaaaggg gaggacccac tgttttatat tctttcttta ccgctatcga tgtgacatta cctcctcttc gagcaggttg taaaatgaaa gttggcagcc gttatgagaa ttatctgtga ggcatgtgac caattttatt tcagcaaaag taaatcacat aacaaaatac gatatgcttc gaagggtgat tggggtgagg acacacacac tgatgcgasa cgaaatcagc caataaaaat Gaatagataa ic Cctgcagc gggaactaac cacgaagtgt cttttcctcc agtaaaggct ttatgattcc atgtgtttta acagggaatt gatttaccat tgtcaacaat actatagcta ctgcactttc tccttgccac ctgagtcaca ctttcccagg ggctgtcagt tcattgcctt

Ugtggagcc gtggcttggc gatctsaaca cagggctctc gaatttc^gg ctcctgcttt gcggagagga agatgtagaa aggtgtttgg SStaaaagaa tattgattag ataaatctgt attgc cccttctctc tgcacaggaa gggagagcag cttgtgggcc cagcagtaaa ggtcacacta ttagttgtti ctgttgcagg attctgaaac tctctagggc tttgaacatg tcagtagcag aigtgctcag actcccggag cagtgtttct atatgaatac ggtgcagaaa cagatcagat 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 I860 1920 1980 2040 2100 2145 Ο <210> 3 <211> 606 <212> DNA <213>小家鼠 <400> 3 actggaccat gagtgttgtg gatgaaagct ttgctaigta tgggtccaag tgacaatccc taactcctgg cttcattact tataaaataa ggggggggag ttaacatacg tgcatggatc gcatccgccg agcacagagt caglggcacg gctggatatg gcagtggatt gaaccttgga taafttcaag agcagcagtg tcctta gctgtaatgt ttgccggaac gcgctttgaa aactgtaaac gatgttgaaa caatcacatc atcaagttgt gagaaaatga aaacaaaacg gagtcttttt gctggtggtg cctcctgctc agatccctgg caaagctcac iccctaccaa atttcatttt cacccacatc agctcactga aaacacccag ggatgccaga acaaatggaa tgtcttctgc actcaagaac tgaacctcag agacacaatg ctccttttga acctcagtag aaatgggacc aagccatcat ggctgcacta tmtgacgt attctgcagc agtctgggct tttccccaac ctatgtgaca tatccagtaa cacctcccat tgtctaggaa gtcgcaagaa aatglctttc 60 120 180 240 300 360 420 480 540 600 606 0>1>2>3> 4 480 DNA 小家鼠 <220> 156342-序列表.d〇c 201143782 <221> misc feature <222> (355)..(355) <223> n為 a、c、g、或t <400> 4 gtgcggagat gccgagcgca aacttcccgg ggctcccggg gcaggcgggg ggggccgggg agggaggaag cgcccagccc aggsagaaag cagggatgtt tatgtgaaga cagtttgctt tttgacaagc gagcaagcgg gcgcccagat tttggagtct ccccggaggg aaaggcagat agaacaaacc tcggactggg ggaagggctc tgcgcagggg aaccgcccca ccctttggcc ccagactctc ctccggtsgt actactaaag gattgcctca agctctgagg atgaagctgg tccggaaacc tctcgaataa caactgtggg ggagaaagag actctgccaa tggcctcaca acggaaagga ctgttttctt aaagctgttg acttcagtca <210> 5 <211> 19 <212> 腿 <213>人工序列 <220> <223>反義寡核苷酸 <400> 5 gtccctgcca ctctaacca <210> 6 <211> 20 <212> m <213>人工序列 <220> <223>反義募核苷酸 <400> 6 ttctgctctc cctgtgactcUgtggagcc gtggcttggc gatctsaaca cagggctctc gaatttc ^ gg ctcctgcttt gcggagagga agatgtagaa aggtgtttgg SStaaaagaa tattgattag ataaatctgt attgc cccttctctc tgcacaggaa gggagagcag cttgtgggcc cagcagtaaa ggtcacacta ttagttgtti ctgttgcagg attctgaaac tctctagggc tttgaacatg tcagtagcag aigtgctcag actcccggag cagtgtttct atatgaatac ggtgcagaaa cagatcagat 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 I860 1920 1980 2040 2100 2145 Ο < 210 > 3 < 211 > 606 < 212 > DNA < 213 > Mus musculus < 400 > 3 actggaccat gagtgttgtg gatgaaagct ttgctaigta tgggtccaag tgacaatccc taactcctgg cttcattact tataaaataa ggggggggag ttaacatacg tgcatggatc gcatccgccg agcacagagt caglggcacg gctggatatg gcagtggatt gaaccttgga taafttcaag agcagcagtg Tcctta gctgtaatgt ttgccggaac gcgctttgaa aactgtaaac gatgttgaaa caatcacatc atcaagttgt gagaaaatga aaacaaaacg gagtcttttt gctggtggtg cctcctgctc agatccctgg caaagctcac iccctaccaa atttcatttt cacccacatc agctcactga aaacacccag ggatgccaga acaaatggaa tgtcttctgc actcaagaac tgaacc Tcag agacacaatg ctccttttga acctcagtag aaatgggacc aagccatcat ggctgcacta tmtgacgt attctgcagc agtctgggct tttccccaac ctatgtgaca tatccagtaa cacctcccat tgtctaggaa gtcgcaagaa aatglctttc 60 120 180 240 300 360 420 480 540 600 606 0>1>2>3> 4 480 DNA Mus musculus <220> 156342 - Sequence Listing .d〇c 201143782 <221> misc feature <222> (355)..(355) <223> n is a, c, g, or t <400> 4 gtgcggagat gccgagcgca aacttcccgg ggctcccggg gcaggcgggg ggggccgggg agggaggaag cgcccagccc aggsagaaag cagggatgtt tatgtgaaga cagtttgctt tttgacaagc gagcaagcgg gcgcccagat tttggagtct ccccggaggg aaaggcagat agaacaaacc tcggactggg ggaagggctc tgcgcagggg aaccgcccca ccctttggcc ccagactctc ctccggtsgt actactaaag gattgcctca agctctgagg atgaagctgg tccggaaacc tctcgaataa caactgtggg ggagaaagag actctgccaa tggcctcaca acggaaagga ctgttttctt aaagctgttg acttcagtca < 210 > 5 < 211 > 19 < 212 > leg < 213 > artificial sequence <220><223> antisense oligonucleotide <400> 5 gtccctgcca ctctaacca <210≫ 6 <211> 20 <212> m < 213 > artificial sequence <220><223> antisense nucleotide <400> 6 ttctgctctc cctgtgactc

<210> Ί <211> 20 <212> DNA <213>人工序列 <220> <223>反義募核苷酸 <400> 7 tttctctgcc gcctctccct <210> 8 <211> 20 <212> DNA <213>人工序列 <220> <223>反義募核苷酸 <400> 8 cttccctgag cattcccact <210> 9 <211> 21 <212> m 人工序列 -4 - ccacccggcc actcttggag ttctacagcc gagccatcct ctgcctgggg ggttnagggt catttcttcc cagagccagt 156342-序列表.doc 201143782 <220> <223>反義寡核苷酸 <400> 9 gttctatctg cctttccctc c <210> 10 <211> 20 <212> ΪΜΚ <213>人工序列 <220> <223>反義寡核苷酸 <400> 10 acatccctgc tttctccctg <210> 11 <211> 21 <212><210> Ί <211> 20 <212> DNA <213> artificial sequence <220><223> antisense nucleotide <400> 7 tttctctgcc gcctctccct <210> 8 <211> 20 <212> DNA <213>Artificial sequence<220><223> Antisense nucleotides <400> 8 cttccctgag cattcccact <210> 9 <211> 21 <212> m artificial Sequence-4 - ccacccggcc actcttggag ttctacagcc gagccatcct ctgcctgggg ggttnagggt catttcttcc cagagccagt 156342 - Sequence Listing.doc 201143782 <220><223> Antisense Oligonucleotide <400> 9 gttctatctg cctttccctc c <210> 10 <211> 20 <212> ΪΜΚ <213> artificial sequence <220><223> antisense oligonucleotide <400> 10 acatccctgc tttctccctg <210> 11 <211> 21 <212>

<213>人工序列 <220><213>Artificial sequence <220>

<223>反義寡核苷酸 <400> II tttagtagta ccaccggagc c <210> <211> <212> <213> 12 20<223> Antisense Oligonucleotide <400> II tttagtagta ccaccggagc c <210><211><212><213> 12 20

DNA 人工序列 <220> 反義寡核苷酸 <400> 12 cccagcttca tcctcagagc <210> 13 <211> 21 <212〉腿 <213>人工序列 <220> <^3>反義募核苷酸 <400> 13 ctttccgttg tgaggccagg a <210> 14 <2Π> 21 <212> Wk <213>人工序列 <220> <223>反義寡核苷酸 <400> 14 tccatatcca gccgtgccac t <210> 15 <211> 21 <212> 腿 <2】3>人工序列 <220> <223>反義寡核苷酸 156342-序列表.doc 201143782 <400> 15 tccatttgtc accaccagca c 21 <210〉 16 <211> 20 <212> Wk <213>人工序列 <220〉 <223>反義寡核苷酸 <400> 16 ggcttctggg tgtttcgttt 20 156342-序列表.docDNA artificial sequence <220> antisense oligonucleotide <400> 12 cccagcttca tcctcagagc <210> 13 <211> 21 <212>leg <213> artificial sequence <220><^3> Antisense nucleotides <400> 13 ctttccgttg tgaggccagg a <210> 14 <2Π> 21 <212> Wk <213> artificial sequence <220><223> antisense oligonucleotide <223>;400> 14 tccatatcca gccgtgccac t <210> 15 <211> 21 <212> Leg <2]3>Artificial sequence <220><223> Antisense oligonucleotide 156342 - Sequence Listing.doc 201143782 <400> 15 tccatttgtc accaccagca c 21 <210> 16 <211> 20 <212> Wk <213>Artificial sequence<220><223> Antisense oligonucleotide <400> Ggcttctggg tgtttcgttt 20 156342 - Sequence Listing.doc

Claims (1)

201143782 七、申請專利範圍: 1. 一種長度為5至30個核苷酸之反義寡核苷酸的用途,其 用以製造用於調節生物系統中LIM同源序列2(LHX2)聚 核普酸之功能及/或表現的藥劑’其中該募核苷酸與LIM 同源序列2(LHX2)聚核苷酸之天然反義轉錄本之反向互 補序列具有至少50%的序列一致性。 2. 如請求項1之用途,其中該寡核苷酸與包括天然反義轉 錄本SEQ ID NO: 2之核苷酸1至2145、SEQ ID NO: 3之核 普酸1至606及SEQ ID NO: 4之核苷酸1至480内的5至30個 連續核苷酸之聚核苷酸之反向互補序列具有至少5〇。/〇的 序列一致性》 3. 一種長度為5至30個核苷酸之反義募核苷酸的用途,其 用以製造用於調節患者細胞或組織中LIM同源序列 2(LHX2)聚核苷酸之功能及/或表現的藥劑,其中該寡核 苷酸與該LIM同源序列2(LHX2)聚核苷酸之反義寡核苷 酸具有至少50%的序列一致性。 4. 如請求項3之用途,其中該募核苷酸與包括天然反義轉 錄本SEQ IDNO: 2之核苷酸1至2145、SEQ ID NO: 3之核 苷酸1至606及SEQ ID NO: 4之核苷酸1至480内的5至30個 連續核苷酸之聚核苷酸之反向互補序列具有至少50°/〇的 序列一致性。 5. —種靶向LIM同源序列2(LHX2)聚核苷酸之天然反義寡 核苷酸中區域之反義寡核苷酸的用途,其用以製造用於 調節生物系統中LIM同源序列2(LHX2)聚核苷酸之功能 156342.doc 201143782 及/或表現的藥劑。 6·如請求項5之用途,其中該LIM同源序列2(LHX2)之功能 及/或表現比對照組增強。 7.如請求項5之用途,其中該反義募核苷酸靶向LIM同源序 列2(LHX2)聚核苦酸之天然反義序列。 8,如請求項5之用途,其中該反義寡核苷酸靶向包括LIM同 源序列2(LHX2)聚核苷酸之編碼及/或非編碼核酸序列的 核酸序列。 9. 如請求項5之用途,其中該反義募核苷酸靶向LIM同源序 列2 (LHX2)聚核普酸之重疊及/或非重叠序列。 10. 如請求項5之用途,其中該反義寡核苷酸包括一或多個 選自以下之修倚: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。 11_如請求項10之用途,其中該一或多個修飾包括至少一個 選自以下之經修飾糖部分:經2,_o_甲氧基乙基修飾之糖 部分、經2’-甲氧基修飾之糖部分、經2,_〇_烷基修飾之糖 部分、雙環糖部分、及其組合。 12.如請求項10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間鍵聯:硫代磷酸酯、2,_〇甲氧 基乙基(MOE)、2’-氟、膦酸烷基酯、二硫代磷酸酯、硫 代脎酸炫基酯、胺基磷酸酯、胺基甲酸酯、破酸酯、鱗 酸三酯、胺基乙酸酯、羧曱基酯、及其組合。 13_如請求項10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苷酸:肽核酸(pNA)、鎖核酸 156342.doc 201143782 (LNA)、阿糖核酸(FANA)、其類似物、衍生物、及組 合。 14. 如請求項!之用途,其中該寡核苷酸包括至少一個如seq ID NO: 5至16所示之寡核苷酸序列。 15. —種以長度為5至3〇個核苷酸之短干擾11]^八(siRNA)募核 苷酸在製造藥劑中之用途,該藥劑用於調節哺乳動物細 胞或組織中LIM同源序列2 (LHX2)基因之功能及/或表 現,其中該siRNA寡核苷酸對LIM同源序列2 (LHX2)聚 核苷酸之反義聚核苷酸具有特異性,且其中該至少一種 siRNA募核苷酸與該LIM同源序列2 (LHX2)聚核苷酸之 反義及/或有義核酸分子之至少約5個連續核酸之互補序 列具有至少50%的序列一致性。 16. 如請求項15之用途,其中該寡核苷酸與該um同源序列2 (LHX2)聚核苷酸之反義及/或有義核酸分子之互補序列 之至少約5個連續核酸之序列具有至少80%的序列一致 性。 17. —種長度為約5至3〇個核苷酸之反義寡核苷酸之用途, 該反義寡核苷酸對LIM同源序列2(LHX2)聚核苷酸中之 有義及/或天然反義股之非編碼及/或編碼序列具有特異 性,該反義募核苷酸用以製造用於調節哺乳動物細胞或 組織中LIM同源序列2(LHX2)之功能及/或表現的藥劑, 其中4至少一種反義寡核苷酸與至少一個如SEq ID N〇: 1至4所示之核酸序列具有至少5〇%的序列一致性。 18. —種包括至少一個修飾之經合成與修飾之寡核苷酸,其 156342.doc 201143782 中該至少一個修挪選自··至.— 至^個經修飾糖部分、至少 一個經修飾核苷酸間鍵聯、 夕個經修飾核苷酸、及 其、.且σ,其中該寡核苷酸係 于在,舌體内或活體外與LIM同 源序列2(Lhx2)基因雜交且與相對於正常對照組可以調 節該讀同源序列2 (LHX2)基因之魏及/或表現之反義 化合物’且其中該募核苷酸與該UM同源序列2 (lhx2) 聚核苷酸之反義及/或有義核酸分子及其等位基因、同系 物、同功型、變體、衍生物、突變體、片段、或組合之 互補序列中至少約5個連續核酸之序列具有至少5〇%的序 列一致性。 19.如請求項18之募核苷酸,其中該寡核苷酸之長度為5至 30個核苷酸’且與該LHX2基因之天然反義轉錄本内5至 3 0個連續核普酸之反向互補序列具有至少5〇%的序列一 致性。 20. 如請求項19之寡核苷酸,其中該至少一個修飾包括選自 由以下組成之群之核苷酸間鍵聯:硫代磷酸酯、膦酸烷 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、胺基乙酸酯、羧曱基 醋、及其組合。 21. 如請求項19之募核苷酸,其中該募核苷酸包括至少一個 硫代磷酸酯核苷酸間鍵聯。 22. 如請求項19之寡核苷酸’其中該募核苷酸包括硫代磷酸 酯核苷酸間鍵聯之主鏈。 23. 如請求項19之寡核苷酸,其中該募核苷酸包括至少一個 156342.doc 201143782 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。 24. 如請求項19之募核苷酸,其中該募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、膦酸烷基酯、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S旨、缓曱基g旨、及其組合。 25. 如請求項19之寡核苷酸,其中該募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。 26. 如請求項19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’-甲氧基修飾之糖部分、經2,_〇_烷基修飾之糖 部分、雙環糖部分、及其組合。 27. 如請求項19之寡核苷酸,其中該募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'-0-烷基修飾之糖部分、雙環糖部分、及其組 合0 28. 如請求項19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LIM同源序列2(LHX2)聚核苷酸之 反義及/或有義股雜交,其中該寡核苷酸與該LIM同源序 列2(LHX2)聚核苷酸中之反義及/或有義編碼及/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 156342.doc 201143782 60%的序列一致性。 29. 如請求項19之募核苷酸,其中該募核苷酸與該lim同源 序列2 (LHX2)聚核苷酸之反義及/或有義編碼及/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80%的序列一致性。 30. 如請求項19之募核苷酸,其中該募核苷酸在活體内或活 體外與至少一種LIM同源序列2 (LHX2)聚核苷酸雜交並 相對於正常對照組可以調節該聚核苷酸之表現及/或功 能。 31. 如请求項19之募核苷酸’其中該寡核苷酸包括如犯卩ID NO: 5至16所示之序列。 32. —種醫藥組合物’其包括—或多種如請求項18之對一或 多種LIM同源序列2 (LHX2)聚核苷酸具有特異性之寡核 普酸及醫藥上可接受之賦形劑。 33. 如請求項32之組合物’其中該等寡核苷酸與如seq N〇: 5至16所示之任一種核苷酸序列比對,具有至少約 40°/。的序列一致性。 34. 如請求項32之組合物,其中該等寡核苷酸包括如seq NO · 5至16所不之核普酸序列。 35. 如印求項34之組合物,其中該等如seq id no: 5至16所 示之I核苷酸包括一或多個修飾或取代。 36. 如請求項35之組合物,其中該一或多個修飾選自··硫代 磷酸知、膦酸曱酯、肽核酸、鎖核酸(LNA)分子、及其 組合。 156342.doc 201143782 37. 38.t) 39. Ο 40. 一種反義寡核苷酸之用途,其用以製造用於預防或治療 與至少一種LIM同源序列2 (LHX2)聚核苷酸及/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 與該至少一種類UM同源序列2 (LHX2)聚核苷酸之天然 反義序列結合並調節該至少一種UM同源序列2 (LHX2) 聚核普酸的表現。 如請求項37之用途,其中與該至少一種LIM同源序列2 (LHX2)聚核苷酸有關之疾病選自:與[11又2之異常功能 及/或表現有關之疾病或病症、血液疾病或病症、毛髮生 長受扣、神經疾病或病症、視網膜疾病或病症、與免疫 性受損有關之疾病或病症、垂體疾病或病症、肝疾病或 病症、嗅覺系統疾病或病症、紅細胞生成受損、造血疾 病或病症、骨髓增生病及脊椎肢體突長。 一種鑑別及選擇至少一種對LHX2基因之天然反義轉錄 本具有選擇性之募核苷酸作為選定靶聚核苷酸用於活體 内投與之方法,該方法包括:鑑別至少一種包括與該選 定靶聚核苷酸之反義聚核苷酸至少部分互補之至少5個 連續核苷酸的募核苷酸;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定靶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 種募核皆酸用於活體内投與。 一種在活體外調節生物系統中UM同源序列2 (LHX2)聚 核苷酸之功能及/或表現之方法,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 156342.doc 201143782 中該至少一種寡核苷酸與LIM同源序列2 (LHX2)聚核普 酸之天然反義之反向互補序列具有至少5〇%的序列一致 性,·由此調節該LIM同源序列2 (LHX2)聚核苷酸之功能 及/或表現。 41. 如請求項40之在活體外調節生物系統中LIM同源序列2 (LHX2)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3〇個核苷酸之反義寡 核苷酸接觸,其中該至少一種寡核苷酸與包括天然反義 轉錄本SEQ ID NO: 2之核普酸1至2145、SEQ ID NO: 3之 核苷酸1至606及SEQ ID NO: 4之核苷酸1至480内之5至30 個連續核苷酸之聚核苷酸之反向互補序列具有至少5〇0/〇 的序列一致性;由此調節該LIM同源序列2 (LHX2)聚核 苷酸之功能及/或表現。 42. —種在活體外調節患者細胞或組織中LIM同源序列2 (LHX2)聚核苷酸之功能及/或表現之方法,其包括:使 s亥等細胞或組織與至少一種長度為5至3〇個核苷酸之反 義募核苷酸接觸,其中該寡核苷酸與該LIM同源序列2 (LHX2)聚核苷酸之反義寡核苷酸具有至少5〇%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該 LIM同源序列2 (LHX2)聚核苷酸之功能及/或表現。 43. 如晴求項42之調節患者細胞或組織中LIM同源序列2 (LHX2)聚核苷酸之功能及/或表現的方法,其包括:使 §亥生物系統與至少一種長度為5至3〇個核苷酸之反義募 核苷酸接觸,其中該至少一種募核苷酸與包括天然反義 156342.doc 201143782 轉錄本SEQ ID NO: 2之核苷酸1至2145、SEQ ID NO: 3之 核苷酸1至606及SEQ ID NO: 4之核苷酸1至480内之5至30 個連續核苷酸之聚核苷酸之反向互補序列具有至少50% 的序列一致性;由此調節該LIM同源序列2 (LHX2)聚核 苷酸之功能及/或表現。 44. 一種在活體外調節生物系統中LI]y[同源序列2 (LHX2)聚 核苷酸之功能及/或表現的方法,其包括:使該系統與至 少一種靶向該LIM同源序列2 (LHX2)聚核苷酸之天然反 義募核苷酸中區域之反義寡核苷酸接觸;由此調節該 LIM同源序列2 (LHX2)聚核苷酸之功能及/或表現。 45. 如請求項44之方法,其中該LIM同源序列2(LHX2)在活 體外之功能及/或表現比對照組增強。 46. 如請求項44之方法,其中該至少一種反義募核苷酸靶向 LIM同源序列2(LHX2)聚核苷酸之天然反義序列。 47_如請求項44之方法,其中該至少一種反義募核苷酸靶向 包括LIM同源序列2(LHX2)聚核苷酸之編碼及/或非編碼 核酸序列之核酸序列。 48.如吻求項料之方法,其中該至少一種反義募核苷酸靶向 LIM同源序列2(LHX2)聚核苷酸之重疊及/或非重疊序 列0 49·如請求項44之方法,其中該至少一種反義募核苦酸包括 一或多個選自以下之修飾··至少-個經修飾糖部分、至 少一個經修飾核㈣鍵聯、至少—個經修飾核㈣ 其組合。 156342.doc 201143782 50.如請求項49之方法,其中該—或多個㈣包括至少一個 選自以下之經修娜糖郁八· ^刀·座2-0-曱氧基乙基修飾之糖 〇P刀、& 2 ·曱氧基修飾之糖部分、經2,_〇_烷基修飾之糖 部分 '雙環糖部分、及其組合。 51 ·如”月求項49之方法,其中該_或多個修飾包括至少—個 選自以下之經修飾核苷間鍵聯:硫代磷酸酯' 2'-0甲氧 基乙基(MOE)、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、胺基曱酸醋、碳酸酯、磷 酸三酯、胺基乙酸酯、羧曱基酯、及其組合。 52.如請求項49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普酸:肽核酸(pNA)、鎖核酸 (LNA)、阿糖核酸(FANA)、其類似物、衍生物、及組 合0 53. 如請求項40之方法,其中該至少一種募核苷酸包括至少 一個如SEQ ID NO: 5至I6所示之寡核苷酸序列。 54. —種在活體外調節哺乳動物細胞或組織中同源序列 2(LHX2)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 0個核苦酸之短干擾 RNA(siRNA)券核普酸接觸’該至少一種siRNA寡核苦酸 對LIM同源序列2(LHX2)聚核苷酸之反義聚核苦酸具有 特異性’其中該至少一種siRNA寡核苷酸與該lim同源 序列2(LHX2)聚核苷酸中反義及/或有義核酸分子之至少 約5個連續核酸之互補序列具有至少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之LIM同源序列 156342.doc -10- 201143782 2(LHX2)之功能及/或表現。 55. 如請求項54之方法,其中該募核苷酸與該LIM同源序列 2(LHX2)聚核苷酸之反義及/或有義核酸分子之互補序列 之至少約5個連續核酸之序列具有至少80%的序列一致 性。 56. —種在活體外調節哺乳動物細胞或組織中UM同源序列 2(LHX2)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約5至30個核苷酸之反義寡核 普酸接觸’該反義寡核苷酸對LIM同源序列2(LHX2)聚 核苦酸之有義及/或天然反義股之非編碼及/或編碼序列 具有特異性,其中該至少一種反義寡核苷酸與至少一種 如SEQ ID NO: 1至4所示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LIM 同源序列2(LHX2)之功能及/或表現。 156342.doc201143782 VII. Patent Application Range: 1. Use of an antisense oligonucleotide of 5 to 30 nucleotides in length for the production of LIM homologous sequence 2 (LHX2) polynucleotide in a biological system An agent that functions and/or exhibits acidity wherein the nucleotide has at least 50% sequence identity to the reverse complement of the natural antisense transcript of the LIM homolog 2 (LHX2) polynucleotide. 2. The use of claim 1, wherein the oligonucleotide comprises nucleotides 1 to 2145 comprising SEQ ID NO: 2 of the natural antisense transcript, nucleotides 1 to 606 of SEQ ID NO: 3, and SEQ ID NO: The reverse complement of the 5 to 30 contiguous nucleotides of nucleotides 1 to 480 of 4 has at least 5 Å. Sequence Consistency of 〇/ 3. 3. The use of an antisense nucleotide of 5 to 30 nucleotides in length to modulate LIM homologous sequence 2 (LHX2) aggregation in a patient's cells or tissues An agent that functions and/or exhibits a nucleotide, wherein the oligonucleotide has at least 50% sequence identity to an antisense oligonucleotide of the LIM homolog 2 (LHX2) polynucleotide. 4. The use of claim 3, wherein the nucleotide and nucleotides 1 to 2145 comprising SEQ ID NO: 2 of the natural antisense transcript, nucleotides 1 to 606 of SEQ ID NO: 3, and SEQ ID NO The reverse complement of a polynucleotide of 5 to 30 contiguous nucleotides within nucleotides 1 to 480 of 4 has a sequence identity of at least 50°/〇. 5. Use of an antisense oligonucleotide targeting a region of a natural antisense oligonucleotide of a LIM homolog 2 (LHX2) polynucleotide for use in the regulation of LIM in a biological system Function of source sequence 2 (LHX2) polynucleotide 156342.doc 201143782 and/or agent of expression. 6. The use of claim 5, wherein the function and/or performance of the LIM homolog 2 (LHX2) is enhanced compared to the control group. 7. The use of claim 5, wherein the antisense raised nucleotide targets a natural antisense sequence of LIM homolog 2 (LHX2) polynucleotide. 8. The use of claim 5, wherein the antisense oligonucleotide targets a nucleic acid sequence comprising a coding and/or non-coding nucleic acid sequence of a LIM homolog 2 (LHX2) polynucleotide. 9. The use of claim 5, wherein the antisense raised nucleotide targets an overlapping and/or non-overlapping sequence of LIM homologous sequence 2 (LHX2) polynucleotide. 10. The use of claim 5, 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 Nucleotides, and combinations thereof. 11. The use of claim 10, wherein the one or more modifications comprise at least one modified sugar moiety selected from the group consisting of 2, _o-methoxyethyl modified sugar moieties, 2'-methoxy Modified sugar moiety, sugar moiety modified with 2,_〇-alkyl, bicyclic sugar moiety, and combinations thereof. 12. The use of claim 10, wherein the one or more modifications comprise at least one modified internucleoside linkage selected from the group consisting of: phosphorothioate, 2,-methoxyethyl (MOE), 2 '-Fluorine, alkyl phosphonate, dithiophosphate, thioester thiodecanoate, amino phosphate, urethane, acid ester, glycerate, amino acetate, Carboxynonyl esters, and combinations thereof. 13_ The use of claim 10, wherein the one or more modifications comprise at least one modified nucleotide selected from the group consisting of: peptide nucleic acid (pNA), locked nucleic acid 156342.doc 201143782 (LNA), arabinic acid (FANA) ), analogs, derivatives, and combinations thereof. 14. As requested! Use of the oligonucleotide, wherein the oligonucleotide comprises at least one oligonucleotide sequence as shown in seq ID NO: 5 to 16. 15. Use of a short interfering 11]^8 (siRNA) nucleotide of 5 to 3 nucleotides in length for the manufacture of a medicament for modulating LIM homology in mammalian cells or tissues The function and/or expression of the sequence 2 (LHX2) gene, wherein the siRNA oligonucleotide is specific for an antisense polynucleotide of a LIM homolog 2 (LHX2) polynucleotide, and wherein the at least one siRNA The raised nucleotide has at least 50% sequence identity to the antisense of the LIM homolog 2 (LHX2) polynucleotide and/or the complement of at least about 5 contiguous nucleic acids of the sense nucleic acid molecule. 16. The use of claim 15, wherein the oligonucleotide is at least about 5 contiguous nucleic acids of the antisense of the um homologous sequence 2 (LHX2) polynucleotide and/or the complement of the sense nucleic acid molecule. The sequence has a sequence identity of at least 80%. 17. Use of an antisense oligonucleotide of about 5 to 3 nucleotides in length, which is sensed in a LIM homolog 2 (LHX2) polynucleotide and And/or a non-coding and/or coding sequence of a natural antisense strand, which is used to produce a function for modulating LIM homolog 2 (LHX2) in a mammalian cell or tissue and/or An agent for expression, wherein 4 at least one antisense oligonucleotide has at least 5% sequence identity with at least one nucleic acid sequence as set forth in SEq ID N〇: 1 to 4. 18. An oligonucleotide comprising at least one modified synthesis and modification, wherein at least one of the 156342.doc 201143782 is selected from the group consisting of a modified sugar moiety, at least one modified core An internucleotide linkage, a modified nucleotide, and a σ thereof, wherein the oligonucleotide is hybridized with the LIM homolog 2 (Lhx2) gene in the tongue or in vitro and The antisense compound of the read homologous sequence 2 (LHX2) gene and/or the expressed antisense compound can be modulated relative to the normal control group and wherein the nucleotide and the UM homologous sequence 2 (lhx2) polynucleotide are A sequence of at least about 5 contiguous nucleic acids of an antisense and/or sense nucleic acid molecule and its alleles, homologs, isoforms, variants, derivatives, mutants, fragments, or combinations of complementary sequences having at least 5 〇% sequence consistency. 19. The nucleotide of claim 18, wherein the oligonucleotide is 5 to 30 nucleotides in length and 5 to 30 consecutive nucleotides in the natural antisense transcript of the LHX2 gene The reverse complement sequence has a sequence identity of at least 5%. 20. The oligonucleotide of claim 19, wherein the at least one modification comprises an internucleotide linkage selected from the group consisting of phosphorothioate, alkyl phosphonate, dithiophosphate, sulfur An alkyl phosphonate, an amino phosphate, a urethane, a carbonate, a phosphate, an amino acetate, a carboxy thioglycol, and combinations thereof. 21. The nucleotide of claim 19, wherein the nucleotide comprises at least one phosphorothioate internucleotide linkage. 22. The oligonucleotide of claim 19, wherein the nucleotide comprises a backbone of a phosphorothioate internucleotide linkage. 23. The oligonucleotide of claim 19, wherein the raised nucleotide comprises at least one 156342.doc 201143782 modified nucleotide selected from the group consisting of: a peptide nucleic acid, a locked nucleic acid (LNA), Analogs, derivatives, and combinations. 24. The nucleotide of claim 19, wherein the nucleotide comprises a plurality of modifications wherein the modifications comprise modified nucleotides selected from the group consisting of phosphorothioates, alkyl phosphonates, A phosphorothioate, an alkyl thiophosphonate, an amino phosphate, an amino phthalate, a carbonate, a phosphate, an aminoacetic acid, a buffer, and combinations thereof. 25. The oligonucleotide of claim 19, wherein the nucleotide 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. 26. The nucleotide of claim 19, 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. 27. The oligonucleotide of claim 19, wherein the raised nucleotide comprises a plurality of modifications wherein the modifications comprise a modified sugar moiety selected from the group consisting of 2'-〇-methoxyethyl modified a sugar moiety, a 2,0-oxy-modified sugar moiety, a 2'-0-alkyl modified sugar moiety, a bicyclic sugar moiety, and combinations thereof. 28. The oligonucleotide of claim 19, wherein The oligonucleotide is at least about 5 to 30 nucleotides in length and hybridizes to an antisense and/or sense strand of a LIM homolog 2 (LHX2) polynucleotide, wherein the oligonucleotide is associated with the LIM An antisense and/or sense encoding in a homologous sequence 2 (LHX2) polynucleotide and/or a complementary sequence of at least about 5 contiguous nucleic acids in a non-coding nucleic acid sequence having at least about 156342.doc 201143782 60% sequence identity Sex. 29. The nucleotide of claim 19, wherein the nucleotide is at least in the antisense and/or sense encoding and/or non-coding nucleic acid sequence of the lim homologous sequence 2 (LHX2) polynucleotide A complementary sequence of about 5 contiguous nucleic acids has a sequence identity of at least about 80%. 30. The nucleotide of claim 19, wherein the nucleotide is hybridized in vivo or in vitro to at least one LIM homolog 2 (LHX2) polynucleotide and can be modulated relative to a normal control group. The performance and/or function of nucleotides. 31. The nucleotide of claim 19, wherein the oligonucleotide comprises a sequence as set forth in ID NO: 5 to 16. 32. A pharmaceutical composition comprising: or a plurality of oligonucleotides specific for one or more LIM homolog 2 (LHX2) polynucleotides of claim 18 and a pharmaceutically acceptable form Agent. 33. The composition of claim 32, wherein the oligonucleotides are aligned with any one of the nucleotide sequences, such as seq N〇: 5 to 16, having at least about 40°. Sequence consistency. 34. The composition of claim 32, wherein the oligonucleotides comprise nucleotide sequences such as seq NO 5 to 16. 35. The composition of claim 34, wherein the I nucleotides as indicated by seq id no: 5 to 16 comprise one or more modifications or substitutions. 36. The composition of claim 35, wherein the one or more modifications are selected from the group consisting of thiophosphoric acid, phosphonium phosphonate, peptide nucleic acid, locked nucleic acid (LNA) molecules, and combinations thereof. 156342.doc 201143782 37. 38.t) 39. Ο 40. Use of an antisense oligonucleotide for the manufacture of a polynucleotide for preventing or treating at least one LIM homologous sequence 2 (LHX2) and Or an agent thereof, or at least one agent encoding a disease, wherein the antisense oligonucleotide binds to a natural antisense sequence of the at least one UM-like homologous sequence 2 (LHX2) polynucleotide and modulates the at least one UM Expression of homologous sequence 2 (LHX2) polynucleotide. The use of claim 37, wherein the disease associated with the at least one LIM homolog 2 (LHX2) polynucleotide is selected from the group consisting of: a disease or condition associated with abnormal function and/or performance of [11 and 2, a blood disease Or a condition, a hair growth buckle, a neurological disease or condition, a retinal disease or condition, a disease or condition associated with impaired immunity, a pituitary disease or condition, a liver disease or condition, an olfactory system disease or condition, impaired erythropoiesis, Hematopoietic diseases or conditions, myeloproliferative diseases, and long limbs of the spine. A method for identifying and selecting at least one nucleotide that is selective for a natural antisense transcript of the LHX2 gene as a selected target polynucleotide for in vivo administration, the method comprising: identifying at least one of the inclusions and the selection An oligonucleotide of at least 5 contiguous nucleotides at least partially complementary to an antisense polynucleotide of a target polynucleotide; measuring the antisense nucleotide and the target polynucleotide under stringent hybridization conditions or The thermal melting point of the hybrid of the antisense polynucleotide of the selected target polynucleotide; and selecting at least one nucleoside acid for in vivo administration based on the information obtained. A method of modulating the function and/or expression of a UM homologous sequence 2 (LHX2) polynucleotide in a biological system in vitro comprising: rendering the system antisense with at least one 5 to 30 nucleotides in length Oligonucleotide contact, wherein the at least one oligonucleotide of 156342.doc 201143782 has at least 5% sequence identity with the reverse complement of the natural antisense of LIM homolog 2 (LHX2) polynucleotide, - thereby modulating the function and/or expression of the LIM homolog 2 (LHX2) polynucleotide. 41. The method of claim 40, wherein the biological system is in vitro modulating the function and/or expression of a LIM homolog 2 (LHX2) polynucleotide, comprising: causing the biological system to have at least one length of 5 to 3 Nucleotide antisense oligonucleotide contacts, wherein the at least one oligonucleotide is nucleoprotein 1 to 2145, SEQ ID NO: 3 comprising the natural antisense transcript of SEQ ID NO: 2 The reverse complement of the polynucleotide of 1 to 606 and 5 to 30 contiguous nucleotides within nucleotides 1 to 480 of SEQ ID NO: 4 has a sequence identity of at least 5〇0/〇; The function and/or expression of the LIM homolog 2 (LHX2) polynucleotide is modulated. 42. A method of modulating the function and/or expression of a LIM homolog 2 (LHX2) polynucleotide in a patient cell or tissue in vitro, comprising: cultivating a cell or tissue such as shai with at least one length of 5 Contacting an antisense oligonucleotide of up to 3 nucleotides, wherein the oligonucleotide has at least 5% sequence with the antisense oligonucleotide of the LIM homolog 2 (LHX2) polynucleotide Consistency ' thereby modulating the function and/or expression of the LIM homolog 2 (LHX2) polynucleotide in a patient's cells or tissues in vivo or in vitro. 43. A method of modulating the function and/or expression of a LIM homolog 2 (LHX2) polynucleotide in a patient cell or tissue, comprising: causing a biological system with at least one length of 5 to Contacting 3 nucleotides of antisense nucleotides, wherein the at least one nucleotide comprises nucleotides 1 to 2145, SEQ ID NO of SEQ ID NO: 2, including natural antisense 156342.doc 201143782 transcript The reverse complement of the nucleotides of nucleotides 1 to 606 of 3 and 5 to 30 contiguous nucleotides of nucleotides 1 to 480 of SEQ ID NO: 4 has at least 50% sequence identity Thereby modulating the function and/or performance of the LIM homolog 2 (LHX2) polynucleotide. 44. A method of modulating the function and/or expression of a LI]y [homologous sequence 2 (LHX2) polynucleotide in a biological system, comprising: targeting the system to at least one of the LIM homologous sequences 2 (LHX2) The antisense oligonucleotide of the region of the natural antisense nucleotide of the polynucleotide is contacted; thereby modulating the function and/or expression of the LIM homolog 2 (LHX2) polynucleotide. 45. The method of claim 44, wherein the LIM homolog 2 (LHX2) is enhanced in function and/or performance in vitro compared to a control group. 46. The method of claim 44, wherein the at least one antisense nucleotide targets a natural antisense sequence of a LIM homolog 2 (LHX2) polynucleotide. 47. The method of claim 44, wherein the at least one antisense oligonucleotide targets a nucleic acid sequence comprising a coding and/or non-coding nucleic acid sequence of a LIM homolog 2 (LHX2) polynucleotide. 48. A method of claim, wherein the at least one antisense nucleotide targets an overlapping and/or non-overlapping sequence of a LIM homolog 2 (LHX2) polynucleotide. The method, wherein the at least one antisense nucleoside acid comprises one or more modifications selected from the group consisting of: at least one modified sugar moiety, at least one modified core (four) linkage, at least one modified core (four) combination thereof . The method of claim 49, wherein the one or more (four) comprises at least one sugar selected from the group consisting of the following: saccharin, yoghurt, turmeric, 2-0-decyloxyethyl modified sugar 〇P knife, & 2 曱oxy modified sugar moiety, 2,_〇-alkyl modified sugar moiety 'bicyclic sugar moiety, and combinations thereof. 51. The method of claim 49, wherein the or more modifications comprise at least one modified internucleoside linkage selected from the group consisting of phosphorothioate '2'-0 methoxyethyl (MOE) , 2'-fluoro, alkyl phosphonate, dithiophosphate, thiophosphonic acid alkyl vinegar, amino citrate, amino citrate, carbonate, phosphate triester, amino acetate 52. The method of claim 49, wherein the one or more modifications comprise at least one modified nucleotide acid selected from the group consisting of: peptide nucleic acid (pNA), locked nucleic acid (LNA) And a method of claim 40, wherein the at least one raised nucleotide comprises at least one of the oligos shown in SEQ ID NOS: 5 to I6. Nucleotide sequence 54. A method of modulating the function and/or expression of a homologous sequence 2 (LHX2) gene in a mammalian cell or tissue in vitro, comprising: equating the cell or tissue with at least one length 5 to 30 nucleotides of short acid interfering RNA (siRNA) nucleoside acid exposure' of at least one siRNA oligonucleotide to LIM homologous sequence 2 (LHX2) The antisense polynucleotide of the polynucleotide is specific in that at least one siRNA oligonucleotide and at least one of the antisense and/or sense nucleic acid molecules in the lim homologous sequence 2 (LHX2) polynucleotide a complement of about 5 contiguous nucleic acids having a sequence identity of at least 5%; and modulating the function of the LIM homologue 156342.doc -10- 201143782 2 (LHX2) in a mammalian cell or tissue in vitro and/or 55. The method of claim 54, wherein the raised nucleotide is at least about 5 consecutive to the antisense of the LIM homolog 2 (LHX2) polynucleotide and/or the complement of the sense nucleic acid molecule. The sequence of the nucleic acid has a sequence identity of at least 80%. 56. A method of modulating the function and/or expression of UM homologous sequence 2 (LHX2) in a mammalian cell or tissue in vitro, comprising: constituting the cells Or the tissue is contacted with at least one antisense oligonucleotide having a length of about 5 to 30 nucleotides. 'The antisense oligonucleotide is sensed for LIM homolog 2 (LHX2) polynucleic acid and/or a non-coding and/or coding sequence of a natural antisense strand having specificity, wherein the at least one antisense oligonucleotide And the at least one nucleic acid sequence of SEQ ID NOS: 1 to 4 has a sequence identity of at least 5%; and modulating the function of the LIM homolog 2 (LHX2) in a mammalian cell or tissue in vitro And / or performance. 156342.doc
TW100117637A 2010-05-19 2011-05-19 Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2 TW201143782A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US34628410P 2010-05-19 2010-05-19

Publications (1)

Publication Number Publication Date
TW201143782A true TW201143782A (en) 2011-12-16

Family

ID=44992329

Family Applications (1)

Application Number Title Priority Date Filing Date
TW100117637A TW201143782A (en) 2010-05-19 2011-05-19 Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2

Country Status (3)

Country Link
AR (1) AR081295A1 (en)
TW (1) TW201143782A (en)
WO (1) WO2011146675A2 (en)

Families Citing this family (13)

* 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
KR20160074368A (en) 2012-05-16 2016-06-28 라나 테라퓨틱스, 인크. Compositions and methods for modulating utrn 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
DK2850186T3 (en) 2012-05-16 2019-04-08 Translate Bio Ma Inc COMPOSITIONS AND PROCEDURES FOR MODULATING SMN GENFAMILY EXPRESSION
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
WO2013173608A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating mecp2 expression
CN104583398A (en) 2012-05-16 2015-04-29 Rana医疗有限公司 Compositions and methods for modulating gene expression
WO2013173598A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating atp2a2 expression
US10174328B2 (en) 2013-10-04 2019-01-08 Translate Bio Ma, Inc. Compositions and methods for treating amyotrophic lateral sclerosis
WO2016070060A1 (en) 2014-10-30 2016-05-06 The General Hospital Corporation Methods for modulating atrx-dependent gene repression
EP3271460A4 (en) 2015-03-17 2019-03-13 The General Hospital Corporation The rna interactome of polycomb repressive complex 1 (prc1)
CN114917367A (en) * 2021-06-02 2022-08-19 中国科学院动物研究所 Application of LHX2 in promoting injury repair and regeneration of central system neurons

Also Published As

Publication number Publication date
WO2011146675A3 (en) 2012-03-08
AR081295A1 (en) 2012-08-01
WO2011146675A2 (en) 2011-11-24

Similar Documents

Publication Publication Date Title
JP6259029B2 (en) Treatment of SCNA-related diseases by inhibition of natural antisense transcripts on sodium channels, voltage-gated, alpha subunit (SCNA)
JP6049623B2 (en) Treatment of IDUA-related diseases by inhibition of natural antisense transcripts to α-L-iduronidase (IDUA)
JP6027893B2 (en) Treatment of sex hormone binding globulin (SHBG) related diseases by inhibition of natural antisense transcripts against sex hormone binding globulin (SHBG)
JP5993744B2 (en) Treatment of nuclear respiratory factor 1 related diseases by inhibition of natural antisense transcripts against nuclear respiratory factor 1 (NRF1)
US20190040394A1 (en) Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
JP6073795B2 (en) Treatment of IFRD1-related diseases by inhibition of natural antisense transcripts to interferon-related developmental regulator 1 (IFRD1)
JP6031356B2 (en) Treatment of uncoupling protein 2 (UCP2) -related diseases by inhibition of natural antisense transcripts against UCP2.
JP5998131B2 (en) DISCSLARGEHOMOLOG (DLG) Treatment of DLG-related diseases by inhibition of natural antisense transcripts on DLG1
TWI573871B (en) Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
TW201143782A (en) Treatment of LIM homeobox 2 (LHX2) related diseases by inhibition of natural antisense transcript to LHX2
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)
TW201200138A (en) Treatment of Atonal homolog 1 (ATOH1) related diseases by inhibition of natural antisense transcript to ATOH1
JP5982362B2 (en) Treatment of PAR4-related diseases by inhibition of natural antisense transcripts to PAR4
TWI575068B (en) Treatment of methionine sulfoxide reductase a (msra) related diseases by inhibition of natural antisense transcript to msra
JP2014519333A (en) Treatment of FXN-related diseases by inhibition of natural antisense transcripts to frataxin (FXN)
TW201210611A (en) Treatment of Nicotinamide Phosphoribosyltransferase (NAMPT) related diseases by inhibition of natural antisense transcript to NAMPT
JP2013515459A (en) Treatment of MBTPS1-related diseases by inhibition of the natural antisense transcript against the membrane-bound transcription factor peptidase, site 1 (MBTPS1)
KR20120086282A (en) Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
TW201201819A (en) Treatment of BCL2-like 11 (BCL2L11) related diseases by inhibition of natural antisense transcript to BCL2L11
TW201209163A (en) Treatment of BCL2 binding component 3 (BBC3) related diseases by inhibition of natural antisense transcript to BBC3