JP2004527472A - Neurotrophic tacrolimus analog - Google Patents

Neurotrophic tacrolimus analog Download PDF

Info

Publication number
JP2004527472A
JP2004527472A JP2002554109A JP2002554109A JP2004527472A JP 2004527472 A JP2004527472 A JP 2004527472A JP 2002554109 A JP2002554109 A JP 2002554109A JP 2002554109 A JP2002554109 A JP 2002554109A JP 2004527472 A JP2004527472 A JP 2004527472A
Authority
JP
Japan
Prior art keywords
nerve
compound
injury
disease
spinal cord
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
JP2002554109A
Other languages
Japanese (ja)
Other versions
JP2004527472A5 (en
Inventor
信也 松岡
隆之 山路
ゴールド、ブルース
Original Assignee
藤沢薬品工業株式会社
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 藤沢薬品工業株式会社 filed Critical 藤沢薬品工業株式会社
Publication of JP2004527472A publication Critical patent/JP2004527472A/en
Publication of JP2004527472A5 publication Critical patent/JP2004527472A5/ja
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Abstract

高レベルの神経栄養活性と低レベルの免疫抑制活性を有するタクロリムス誘導体。これらの化合物は、神経栄養剤、特に、神経傷害/機能不全の予防又は治療用の神経栄養剤として有用である。Tacrolimus derivatives with high levels of neurotrophic activity and low levels of immunosuppressive activity. These compounds are useful as neurotrophic agents, especially for the prevention or treatment of nerve injury / dysfunction.

Description

【0001】
発明の背景
発明の分野
本発明は、高レベルの神経栄養性活性と低レベルの免疫抑制活性を有するタクロリムス誘導体に関する。
【0002】
背景の説明
ある種のマクロライド化合物、例えばタクロリムス(tacrolimus)及び関連化合物は、脳虚血の予防又は治療に役立つことが知られている(WO94/14443)。タクロリムスなどの、FKBP型イムノフィリンに対し親和性を有する特定のピペコリン酸誘導体は、損傷した末梢神経の成長を刺激すること、又は神経再生を促進することが知られている(WO96/40140)。ある種の非免疫抑制化合物、即ち、ゲルダナマイシン及びそのアナログは、ステロイドレセプター複合体を分解し、神経成長を促進することが示されている(WO99/21552)。
【0003】
発明の概要
本発明者らは、以下に記載の特定のタクロリムスアナログ、即ち、化合物(I)が優れた神経栄養性 (neurotrophic) 活性を有するものの、タクロリムスとは異なり免疫抑制活性を殆ど又は全く有しないことを見出した。以下に示すように、例えば、神経突起の長さを増加させる能力により測定すると、化合物(I)は、タクロリムスと比較して優れたレベルの向神経性 (neurotropic) 活性を発揮する。同様に、化合物(I)の投与が、軸索再生を誘導し、神経挫滅又は脊髄損傷からの回復速度を増加させることが示される。更に、化合物(I)はこれらの利点のある向神経性効果を発揮するが、タクロリムスと比べて免疫抑制活性を殆ど又は全く有しない。
それ故、本発明は、優れた神経栄養剤としての、並びに殆ど又は全く免疫抑制活性を有しない向神経剤としての、化合物(I)の新規使用を提供する。
更に、本発明は、化合物(I)を含む神経栄養剤又は組成物を提供する。
更に尚、本発明は、哺乳動物に化合物(I)を投与することを含む、神経傷害/機能不全の予防又は治療の方法を提供する。
【0004】
発明の詳細な説明
予想外なことに、本発明者らは、化合物(I)が、神経系への損傷若しくは傷害、神経系の悪化又は神経系の疾患によって引き起こされる神経傷害又は機能不全の改善、予防又は治療に有用であり、同時に、有利なことに免疫抑制効果を殆ど又は全く有しないことを発見した。
【0005】
化合物(I)は、身体傷害、栄養性疾患、虚血、変性疾患、悪性腫瘍疾患、感染症、並びに薬物相互作用、毒素若しくは毒物によって引き起こされる損傷、悪化、又は機能不全の治療に有用である。例えば、化合物(I)は、神経手術、末梢神経傷害、火傷、脳脊髄炎、HIV、ヘルペス、癌、放射線治療、薬物相互作用、葉酸若しくはビタミンB12欠乏症、並びに、神経毒若しくは鉛などの化学物質への暴露によって引き起こされる神経学的損傷又は機能不全の治療に有用である。
【0006】
従って、化合物(I)は、例えば、多発性筋炎、ギラン−バレー症候群、メニエール病、多発性神経炎、単発神経炎(孤立性神経炎)、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症(ALS)、ハンチントン病、神経根障害、神経障害(例えば、糖尿病性神経障害、化学療法誘発神経障害など)、脊髄損傷、老人性痴呆症、血管性痴呆症、多発性硬化症、身体麻痺などの神経傷害及び機能不全の予防又は治療に有用である。
【0007】
本発明で使用するタクロリムスアナログである化合物(I)は、以下の化学式を有する:
【0008】
【化3】

Figure 2004527472
【0009】
本化合物は、米国特許第5,376,663号の実施例29に記載の通りに製造してもよい。本発明で使用する化合物(I)については、立体配座体(conformer)、並びに、不斉炭素原子又は二重結合に起因する光学異性体や幾何異性体などの1個以上の立体異性体があり得、このような立体配座体及び異性体もまた、本発明の化合物の範囲に包含されることを理解すべきである。
【0010】
化合物(I)は、医薬として許容できる塩、誘導体、溶媒和物又はプロドラッグの形態であり得、それらは全て本発明の範囲に包含される。溶媒和物としては好ましくは、水和物及びエタノール和物が挙げられる。
【0011】
化合物(I)の好適な形態は、以下である:
【0012】
【化4】
Figure 2004527472
【0013】
本発明での化合物(I)は、純粋な化合物として、又は他の化合物若しくは別の成分との混合物として、好ましくは医薬用溶媒若しくは担体中で投与してもよい。化合物(I)が医薬製剤若しくは組成物の形態で使用されるとき、化合物(I)は、外用(局所)、経口、経腸、皮下、静脈内、筋肉内又は非経口適用に適した、有機若しくは無機の担体、溶媒又は賦形剤と混合してもよい。例えば、化合物(I)は、活性成分としての化合物(I)と1つ以上の担体、溶媒又は賦形剤を含む、固形、半固形又は液状の組成物中に存在していてもよい。典型的な担体、溶媒又は賦形剤としては、通常の医薬担体、医薬剤、緩衝剤、分散剤、乳化剤及びアジュバントが挙げられるがそれらに限定されない。
【0014】
化合物(I)はまた、錠剤、ペレット剤、カプセル剤、点眼剤、坐剤、液剤(例えば、生理食塩水)、乳剤、懸濁剤(例えば、オリーブ油)、軟膏剤、エアロゾルスプレー剤、クリーム剤、皮膚プラスター剤、パッチ剤、及び使用に適した任意の他の剤形のための慣用の無毒性の、医薬として許容できる担体と複合化してもよい。
【0015】
適切な担体としては、水、生理食塩水及びデキストロース水溶液、油(動物油、植物油及び合成油を含む)、並びに石油製品が挙げられる。他の有用な担体としては、グルコース、ラクトース、アカシアゴム、ゼラチン、マンニトール、澱粉ペースト、三ケイ酸マグネシウム、タルク、コーンスターチ、ケラチン、コロイドシリカ、ジャガイモデンプン、尿素、並びに、固形、半固形若しくは液状形態の製剤の製造における使用に適した他の担体が挙げられる。更に、補助剤、安定化剤、乳化剤、増粘剤、着色剤、フレーバー剤、及び香料を使用してもよい。
【0016】
化合物(I)は、特定の疾患プロセス又は状態に対し所望の効果を生じるのに有効な量で医薬組成物に含まれる。化合物(I)は、向神経性効果の提供又は神経細胞成長の刺激に十分な量で含まれる。
【0017】
本発明の方法を用いて治療できる哺乳動物としては、牛、馬、豚などの家畜用哺乳動物、犬、猫、ラット、マウス、ウサギ、ハムスターなどの家庭用動物、霊長類、及びヒトが挙げられる。
ヒトへの、化合物(I)を含む製品又は組成物の投与又は適用の好適な様式としては、注射又は経口投与が挙げられる。
化合物(I)の治療有効量又は治療有効投与量は、個々の患者で変動してもよく、また、治療する個々の患者の年齢や状態に依存するが、約0.0001−1000mg、好ましくは0.001−500mg、より好ましくは0.01−100mgの範囲に及ぶ1日投与量の活性成分が、一般的に、疾患の治療に与えられ、約0.001−0.01mg、0.2−0.5mg、1mg、5mg、10mg、50mg、100mg、250mg、500mgの平均単回投与量が一般的には投与される。ヒトでの慢性的投与の1日投与量は、約0.1−30mg/kg/日の範囲であろう。化合物(I)はまた、神経栄養性活性又は神経細胞成長刺激活性を有する他の薬剤と、同時に、別々に、又は順次、投与又は適用されてもよい。
【0018】
本発明の医薬組成物は、神経再生と機能回復を促進し、神経突起成長を刺激し、それによって種々の神経病理的状態を治療するために、このような治療の必要性のある哺乳動物被験体(例えば、ヒト患者)に定期的に投与できる。種々の神経病理的状態としては、身体傷害によって引き起こされる末梢神経及び中枢神経系への損傷(例えば、脊髄損傷及び外傷、坐骨若しくは顔面神経障害若しくは傷害、切断後四肢移植);疾患(例えば、糖尿病性神経障害);癌化学療法(例えば、アクリルアミド、タキソール、ビンカアルカロイド及びドキソルビシンによって誘起される神経障害);後遺症−例えば、脳梗塞、出血性梗塞などに伴う言語障害(例えば、構音障害)、意識混濁、ジスキネジアなど;並びに、三叉神経痛、舌咽頭神経痛、顔面麻痺、重症筋無力症、筋ジストロフィー、筋萎縮性側索硬化症、進行性筋萎縮症、進行性延髄性遺伝性筋萎縮症、椎間板ヘルニア症候群(herniated, ruptured or prolapsed vertebral disk syndrome)、頚椎症、叢症、胸郭口破壊症候群、末梢神経障害(例えば、鉛、アクリルアミド、ガンマ−ジケトン(接着剤中毒(glue−sniffer’s)神経障害)、二硫化炭素、ダプソン、チック (ticks)、ポルフィリン症、ギラン−バレー症候群、アルツハイマー病、パーキンソン病及びハンチントン舞踏病を含むがそれらに限定されない種々の末梢神経障害及び神経学的疾患を含むがそれらに限定されない神経学的疾患が挙げられる。
【0019】
末梢神経の断裂 (transsection) 又は脊髄損傷は、哺乳動物に神経成長刺激量の薬剤を投与し、末梢神経又は脊髄に、同種異系移植片(allograft)などの神経移植片(Osawaら,J.Neurocytol. 19:833−849, 1990;Buttemeyerら, Ann.Plastic Surgery 35:396−401,1995)又は人工神経移植片(Madison及び Archibald, Exp. Neurol. 128:266−275,1994;Wellsら,Exp.Neurol.146:395−402,1997)を移植することによって治療できる。末梢神経又は脊髄の断裂末端間の空間は、好ましくは、コラーゲン、メチルセルロースなどの非細胞性間隙充填物質;又は、シュワン細胞(Xuら,J.Neurocytol.26:1−16,1997)、臭覚細胞及び鞘細胞(Liら,Science 277:2000−2002,1997)などの、神経細胞成長を促進する細胞の懸濁液によって充填される。本神経成長促進剤は、このような細胞性又は非細胞性間隙充填物質と共に含ませることができ、又は、神経移植方法の前、間、又はその後に全身的に投与できる。
【0020】
特に、化合物(I)は、神経傷害/機能不全、多発性筋炎、ギラン−バレー症候群、メニエール病、多発性神経炎、単発神経炎(孤立性神経炎)、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症(ALS)、ハンチントン病、神経根障害、糖尿病性神経障害、化学療法誘発神経障害、老人性痴呆症、血管性痴呆症、多発性硬化症、身体麻痺、又は脊髄損傷の治療又は予防に有用である。
【0021】
以下の実施例は、更に詳細に本発明を説明する。これらの実施例は、本発明のある局面又は実施態様を記載するが、本発明の範囲の限定を意図するものではないことを理解すべきである。
【0022】
実施例1:化合物(I)による処理は、海馬ニューロンの神経突起の長さを有意に増加させる
細胞培養物の調製:
Banker 及び Cowan (Brain Research, 1977, 126: 397−425)に従い、胎仔海馬ニューロンを、胎生期18.5日目(「E18.5」)の仔ラットから得た。簡潔には、海馬を取り出し、細かく切断し、100I.U.パパインとともに37℃で45分間インキュベートし、細胞を完全ニューロン培地(L−グルタミン非含有の最小必須培地(GIBCO,Grand Island, NY)、高グルコース最小必須培地(GIBCO)1.5ml/100ml、血清増量培地(Hito+Tm;Collaborative Research Inc, Lexington, MA)0.1ml/100ml、グルタミン(GIBCO)、5%胎仔ウシ血清(GIBCO))に再懸濁した。
【0023】
細胞を、ポリ−L−リジンで被覆したカバースリップに播種した(500細胞/カバースリップ)。カバースリップをディッシュの上で逆さにした。ディッシュは、皮質アストロサイトの単層で予め被覆しておいた。
【0024】
海馬ニューロン軸索の長さの解析:
海馬ニューロン(それらの特徴的な極性及び樹状突起で識別)を毎日検査し、72時間で無作為に写真をとった(9−12フレーム/カバースリップ)。軸索(最長の突起として定義)の長さを、適切なソフトウエア(Bioquant IV, R & M Biometrics, Nashville, TN)を備えたIBM XTコンピューターと連結したHouston Instrument HI−PADデジタイジングタブレットを用い、写真印刷物で測定した(細胞体の長さの3倍を超える突起のみを測定した)。同様に処理したカバースリップからのデータ(1群当り3又は4)は異ならず、従って一緒にした。平均値を計算し、1要因((化合物(Ia)又はタクロリムス処理群)対(未処理対照群))ANOVA、次いでNewman−Kuels多重比較テスト(WINKS 4.62 プロフェッショナル版)を用い、比較した。
【0025】
結果:
72時間では、未処理対照群と10nMタクロリムス処理群との間に有意差はなかった。しかし、化合物(Ia)10nM濃度処理群は、神経突起の長さが統計的に有意に増加した。下記の表1の結果を参照のこと。
【0026】
【表1】
Figure 2004527472
【0027】
実施例2:化合物(I)による処理は、SH−SYSYヒト神経芽細胞腫の神経突起の長さの平均値を増加させる
SH−SY5Y神経芽細胞腫培養物の調製:
10%胎仔ウシ血清(SIGMA)、ペニシリン50I.U./ml、50μg/mlストレプトマイシン(GIBCO)を補充したDMEM培地(GIBCO)中で、7%CO中37℃でSH−SY5Yヒト神経芽細胞腫を維持した。15,000細胞/ウエルで6ウエルプレートに細胞を蒔き、0.4μMアフィディコリン(SIGMA)で処理した。5日目に細胞を洗浄し、タクロリムス(10nM)又は化合物(Ia)(1nM)の存在又は非存在下、10ng/mlの神経成長因子(NGF)で処理した(突起成長を誘導するため)。培地を96時間経過後に交換し、更なる72時間経過後に、新鮮培地で置き換えた(合計時間168時間)。全ての実験は、duplicateで行い、各処理群につき、データを平均化した。
【0028】
SH−SY5Y神経芽細胞腫の神経突起の長さの解析
突起の長さの解析のために、168時間で細胞を無作為に撮影した(1ウエル当り20視野)。神経突起の長さを、適切なソフトウエア(Bioquant IV, R & M Biometrics, Nashville, TN)を備えたIBM XTコンピューターと連結したHouston Instrument HI−PADデジタイジングタブレットを用い、写真印刷物で測定した(細胞体の長さの2倍を超える突起のみを測定した)。同様に処理したウエルからのデータは異ならず、従って一緒にした。平均値を計算し、1要因((化合物(Ia)又はタクロリムス処理サンプル)対(NGF単独の処理サンプル))ANOVA、次いでNewman−Kuels多重比較テスト(WINKS4.62プロフェッショナル版)を用い、比較した。
【0029】
結果:
神経突起の長さの測定により、化合物(Ia)(1nM)とタクロリムス(10nM)の両方が、NGF(10ng/ml)のみと比較して、168時間で神経突起の長さを有意に増加させることが実証された。しかし、NGFとの組み合わせにおける1nM化合物(Ia)の効果は、NGFとの組み合わせにおける10nMタクロリムスの効果より大きかった。
【0030】
【表2】
Figure 2004527472
【0031】
実施例3:化合物(I)による処理は、ラット坐骨神経挫滅モデルの機能回復を促進する
動物及び手術方法:
9匹の6週齢雄性Sprague−Dawleyラットを2%ハロタンで麻酔し、右坐骨神経を露出させ、股関節のレベルで神経を2度押しつぶした(No.7 Dumont宝石職人用鉗子を用い合計60秒間)。神経鞘に滅菌済の9−O縫合糸を結びつけることにより、挫滅部位に印を付けた。
【0032】
化合物(Ia)の調製及び投与
30%ジメチルスルホキシド(DMSO):70%生理食塩水を含む溶媒に化合物(Ia)を溶解した。3匹の神経挫滅ラットは、化合物(Ia)(1又は5mg/kg)又は等容量の溶媒(生理食塩水中30%DMSO)(5ml/kg)を毎日皮下注射した。
【0033】
行動評価
灌流の日(18日目)まで、動物を毎日試験した。以下の半定量的スケールを用い、動物の機能回復を評価した:
0:足が麻痺し、歩行時に外側に曲がり、つま先が曲がる;
1:足がまっすぐで、つま先を動かすことができる;
2:足で一定に歩くことができる;
3:歩行時につま先が広がっている;
4:かかとを離して歩き、ほぼ正常のつま先の広がりを示す;
中間の能力を示す動物には、部分的スコア:+,0.25;++,0.5;+++,0.75を与えた。
【0034】
組織の固定及び調製:
神経挫滅後18日目に、4%ハロタンでラットを深く麻酔し、ヘパリン処理し、4%パラホルムアルデヒドの0.1Mリン酸ナトリウム緩衝液(pH7.4)で10秒間、次いで5%グルタルアルデヒド(IL)の0.1Mリン酸ナトリウム緩衝液(pH7.4)で灌流し、4℃で24時間固定した。挫滅部位から既知(5mm)の距離にある坐骨神経から組織のサンプルを採取した。本研究では、ヒラメ筋を支配している後方脛骨神経枝のデータのみ報告する。組織を0.1Mリン酸ナトリウム緩衝液(pH7.4)中に置き、1%四酸化オスミウム(0.1Mリン酸緩衝液中)で2.5時間後固定し、エタノール中で脱水し、プラスチックに包埋した。半薄切片を酢酸ウラニルとクエン酸鉛で染色し、フィルムで支持された75メッシュグリッドに載せ、JEOL100CX電子顕微鏡を用いて観察した。
【0035】
形態解析:
軸索直径の解析を脛骨神経で行った。電子顕微鏡を用い、再生有髄軸索の数を計測した。溶媒処理群、化合物(Ia)(1mg/kg)処理群、及び化合物(Ia)(5mg/kg)処理群について、平均値と標準誤差を計算した。
【0036】
統計学的解析:
行動解析に関しては、個々の値の比較のために1要因ANOVA、次いでNewman−Keuls多重比較テストを用いて、機能回復の平均値を比較した。形態解析に関しては、個々の値の比較のために1要因ANOVA、次いでNewman−Keuls多重比較テストを用いて、軸索数の平均値を比較した。
【0037】
結果:
機能回復:
機能回復は、15−17日目に観察し、溶媒処理ラットよりも1mg/kg処理ラットと5mg/kg処理ラットの両方においてより早期に生じた。下記の表3を参照のこと。
【0038】
【表3】
Figure 2004527472
【0039】
電子顕微鏡
形態学的検討を神経挫滅後18日目に行った。
神経面積(5,000μm)当りの再生有髄軸索の数は、溶媒処理ラットの5.5±2.7(平均±SEM)から、1mg/kg処理ラットと5mg/kg処理ラットでそれぞれ19±2.4と20±2.9へと劇的に増加した。下記の表4を参照のこと。
【0040】
【表4】
Figure 2004527472
【0041】
実施例4:化合物(I)による処理は、ラット脊髄損傷モデルの機能回復を促進する
(1)方法
動物及び手術方法
28匹の6週齢雄性Sprague−Dawleyラットを2%ハロタンで麻酔し、T10/T11で椎弓切除を行い、脊髄の片側切断手術をT10/T11脊髄のレベルで行った。
化合物(Ia)の調製及び投与
30%ジメチルスルホキシド(DMSO):70%生理食塩水を含む溶媒に化合物(Ia)を溶解した。脊髄損傷ラットは、手術後7週間にわたって、化合物(Ia)(2mg/kg)又は等容量の溶媒(生理食塩水中30%DMSO)(5ml/kg)を毎日皮下注射した。
【0042】
機能回復の評価
損傷2週間後に、改変Tarlov/Klingerスケール(modified Tarlov/Klinger scale)、narrow beamテスト及びフットプリントテスト(footprint test)を用いて、機能回復を評価した。
A.改変 Tarlov/Klinger スケール
ラットを、1分間オープンフィールドで自由に歩かせ、下記のスケールに基づき0−6に分類した。
0:損傷した後肢の動き無し
1:損傷した後肢の動きをかろうじて確認できる
2:損傷した後肢関節(股関節、膝関節又は足関節)は活発に動くが、連携は無く、体重を支えられない
3:損傷した後肢を交互に使って歩き推進力を伴うが、体重は支えられない
4:損傷した後肢で体重を支えることができる
5:若干の歩行障害が見られる
6:正常な歩行
【0043】
B. narrow beam テスト
木製角材(長さ1.5m)を用いて、7.7cm、4.7cm、2.7cm、1.7cmと幅の広いものから順に試験を行なった。ラットを棒上で歩行させ、滑ることなく歩行可能な最も狭い棒を、少なくとも2回の試行において記録した。
0:如何なる角材でも歩行不可
1:7.7cm角材で歩行可能
2:4.7cm角材で歩行可能
3:2.7cm角材で歩行可能
4:1.7cm角材で歩行可能
【0044】
C.フットプリントテスト
ラットの後肢にインクを付け、長さ60cm、幅7.5cmの狭い走路を覆う紙に足跡を付けた。1連の少なくとも6個の連続したフットプリントを用い、5ポイントフットプリントスコアを測定した。
0:後肢をひきずり、足跡は全く観察されない
1:少なくとも3個の足跡において、少なくとも3個のつま先の跡が見られる
2:個々のラットの神経挫滅前の値と比べて、2倍以上の足が外側又は内側に曲がっている
3:つま先を引きずる兆候は示さないが、足が曲がっている兆候を示す
4:外側にも内側にも足が曲がっていないが(ベースライン値の角度の2倍未満)、1個以上のかかと跡が観察される
5:かかと跡が全く観察されない
【0045】
統計学的解析
行動解析に関し、個々の値の比較のために1要因ANOVA、次いでNewman−Keuls多重比較テストを用いて、各機能試験のスコアの平均値を比較した。
【0046】
(2)結果
機能回復
改変Tarlov/Klingerスケール(表5)、narrow beamテスト(表6)及びフットプリントテスト(表7)を用いた3つの機能回復試験において、化合物(Ia)は、改変Tarlov/Klingerスケール(表5)、narrow beamテスト(表6)及びフットプリントテスト(表7)における運動機能不全を改善した。
【0047】
【表5】
Figure 2004527472
【0048】
【表6】
Figure 2004527472
【0049】
【表7】
Figure 2004527472
【0050】
実施例5:化合物(Ia)はFKBP12と結合するが、タクロリムスとは異なり免疫抑制効果を殆ど又は全く発揮しない
(1)FKBP12との結合実験
Tamura,K.ら(Biochemical and Biophysical Research Communications, Vol.202, No.1, 437−499,1994)と同様な方法に基づき、結合実験を行った。結果を表8に示す。
【0051】
(2)混合リンパ球反応(MLR)
米国特許第4,929,611号と同様な方法に基づき、MLRテストを行った。
結果を表8に示す。
【0052】
【表8】
Figure 2004527472
【0053】
上記結果は、化合物(Ia)がFKBP12と結合できるが、免疫抑制活性を有しないことを示す。
上記結果は、インビトロとインビボモデルの両方を用いる化合物(I)の強力な神経栄養効果を示す。2つの細胞培養モデルにおいて、化合物(I)は低濃度でさえも神経突起成長を増加させた。更に、低用量の化合物(I)の全身投与は、坐骨神経の軸索再生の速度を増加させることにより神経挫滅損傷後の機能回復を促進し、脊髄損傷からの機能回復を促進した。
更に、上記のように、化合物(I)は、免疫抑制活性を有しないものの、強力な神経栄養活性又は神経細胞成長刺激活性を提供する。従って、本発明は、神経成長又は再生の刺激又は促進に、特に免疫抑制効果が有利でないとき又は所望されないときに、有用な神経栄養剤を提供する。
【0054】
本発明の他の局面は以下を含む:
包装物と、包装物内に含まれる上記化合物(I)とを含む製品であって、化合物(I)が、神経機能不全の予防又は治療に治療的に有効である製品であり、包装物が、神経傷害/機能不全の予防又は治療に化合物(I)を使用できること又は使用すべきであることを指示しているラベル又は記載物を含む、製品。
上記の化合物(I)を含む医薬組成物と、それに添付される記載物とを含む商業パッケージであって、記載物が、神経傷害/機能不全の予防又は治療に化合物(I)を使用できること又は使用すべきであることを記述している、商業パッケージ。
化合物(I)で処理された細胞を含む、細胞懸濁液、組織又は移植片などの組成物。このような組成物は、神経系への損傷を修復するのに有用である。このような組成物はまた、神経細胞成長を促進又は支援する細胞懸濁液の他のタイプ(例えば、シュワン細胞又はオリゴデンドロサイト、グリア細胞及び鞘細胞などのミエリン産生細胞、コラーゲンなどの細胞外基質物質、又はサイトカイン、分裂促進因子、イムノフィリン、及びニューロトロフィン(例えば、NGF−1、BDNF、CNTF、NT−3、NT−4、及びNT−5)などの他の特定の神経調節剤)の他の神経細胞成長刺激剤を含んでいてもよい。
【0055】
同種同系移植片 (homograft)、同種異系移植片又は異種移植片などの移植片もまた、神経成長並びに移植片としての、及びその他の適用のための使用を容易にするため、化合物(I)で処理してもよい。
【0056】
参照による援用
本開示で引用された又は言及された各文献、特許出願又は特許刊行物の内容は、参照によりその全体が援用される。本出願が優先権を主張するいずれの特許文献の内容もまた、参照によりその内容の全体が援用される。具体的には、米国仮出願第60/258,500号の内容が、参照により援用される。
【0057】
改変及び他の実施態様
明らかに、本発明の多数の改変やバリエーションが、上記教示により可能である。従って、添付した特許請求の範囲の範囲内で、本発明は、具体的に本明細書に記載した発明以外にも実施されうることを理解すべきである。記載した組成物及び方法の種々の改変やバリエーション並びに本発明の概念が、本発明の範囲及び思想から逸脱しないことは当業者にとって明白であろう。特定の好適な実施態様と関連して本発明を記載したが、請求項に係る発明は、このような特定の実施態様への限定を意図しないことを理解すべきである。本発明の実施のために記載された様式の種々の改変が、医学、生物学、化学、又は薬理学の技術分野又は関連分野の当業者にとって自明であり、本発明の範囲内であることが意図される。[0001]
Background of the Invention
Field of the invention
The present invention relates to tacrolimus derivatives having high levels of neurotrophic activity and low levels of immunosuppressive activity.
[0002]
Background description
Certain macrolide compounds, such as tacrolimus and related compounds, are known to be useful in preventing or treating cerebral ischemia (WO 94/14443). Certain pipecolic acid derivatives, such as tacrolimus, which have an affinity for FKBP-type immunophilins are known to stimulate the growth of damaged peripheral nerves or promote nerve regeneration (WO 96/40140). Certain non-immunosuppressive compounds, geldanamycin and its analogs, have been shown to degrade steroid receptor complexes and promote nerve growth (WO 99/21552).
[0003]
Summary of the Invention
The present inventors have determined that certain tacrolimus analogs described below, ie, compound (I), have excellent neurotrophic activity, but have little or no immunosuppressive activity, unlike tacrolimus. I found it. As shown below, for example, as measured by the ability to increase neurite length, compound (I) exerts superior levels of neurotropic activity compared to tacrolimus. Similarly, administration of compound (I) is shown to induce axonal regeneration and increase the rate of recovery from nerve crush or spinal cord injury. Furthermore, compound (I) exerts these advantageous neurotrophic effects, but has little or no immunosuppressive activity compared to tacrolimus.
The present invention therefore provides a novel use of compound (I) as an excellent neurotrophic agent and as a neurotrophic agent with little or no immunosuppressive activity.
Further, the present invention provides a neurotrophic agent or composition comprising Compound (I).
Still further, the present invention provides a method for preventing or treating nerve injury / dysfunction, comprising administering compound (I) to a mammal.
[0004]
DETAILED DESCRIPTION OF THE INVENTION
Unexpectedly, we have found that compound (I) is useful for ameliorating, preventing or treating nerve injury or dysfunction caused by damage or injury to the nervous system, deterioration of the nervous system or diseases of the nervous system. It has been found to be useful, while at the same time advantageously having little or no immunosuppressive effect.
[0005]
Compound (I) is useful for the treatment of personal injury, nutritional disease, ischemia, degenerative disease, malignant disease, infectious disease, and damage, aggravation, or dysfunction caused by drug interactions, toxins or toxins. . For example, Compound (I) is useful for neurosurgery, peripheral nerve injury, burns, encephalomyelitis, HIV, herpes, cancer, radiation therapy, drug interaction, folate or vitamin B12 deficiency, and chemicals such as neurotoxins or lead. It is useful for treating neurological damage or dysfunction caused by exposure to.
[0006]
Thus, compound (I) is, for example, polymyositis, Guillain-Barre syndrome, Meniere's disease, polyneuritis, solitary neuritis (isolated neuritis), Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS), Huntington's disease, radiculopathy, neuropathy (eg, diabetic neuropathy, chemotherapy-induced neuropathy, etc.), spinal cord injury, senile dementia, vascular dementia, multiple sclerosis, physical paralysis, etc. It is useful for preventing or treating nerve injury and dysfunction.
[0007]
Compound (I), a tacrolimus analog used in the present invention, has the following chemical formula:
[0008]
Embedded image
Figure 2004527472
[0009]
This compound may be prepared as described in Example 29 of US Pat. No. 5,376,663. The compound (I) used in the present invention has a conformer (conformer) and one or more stereoisomers such as an optical isomer or a geometric isomer resulting from an asymmetric carbon atom or a double bond. It is to be understood that, possibly, such conformations and isomers are also within the scope of the compounds of the present invention.
[0010]
Compound (I) may be in the form of a pharmaceutically acceptable salt, derivative, solvate or prodrug, all of which are included within the scope of the present invention. Preferably, the solvates include hydrates and ethanolates.
[0011]
Preferred forms of compound (I) are:
[0012]
Embedded image
Figure 2004527472
[0013]
Compound (I) according to the present invention may be administered as a pure compound or as a mixture with other compounds or other ingredients, preferably in a pharmaceutical solvent or carrier. When compound (I) is used in the form of a pharmaceutical preparation or composition, compound (I) is an organic, suitable for topical (topical), oral, enteral, subcutaneous, intravenous, intramuscular or parenteral application Alternatively, it may be mixed with an inorganic carrier, solvent or excipient. For example, compound (I) may be present in a solid, semi-solid or liquid composition comprising compound (I) as an active ingredient and one or more carriers, solvents or excipients. Typical carriers, solvents or excipients include, but are not limited to, conventional pharmaceutical carriers, pharmaceutical agents, buffers, dispersants, emulsifiers, and adjuvants.
[0014]
Compound (I) can also be used as tablets, pellets, capsules, eye drops, suppositories, liquids (eg, physiological saline), emulsions, suspensions (eg, olive oil), ointments, aerosol sprays, creams May be complexed with conventional non-toxic, pharmaceutically acceptable carriers for skin plasters, patches, and any other dosage form suitable for use.
[0015]
Suitable carriers include water, saline and aqueous dextrose solutions, oils (including animal, vegetable and synthetic oils), and petroleum products. Other useful carriers include glucose, lactose, gum acacia, gelatin, mannitol, starch paste, magnesium trisilicate, talc, corn starch, keratin, colloidal silica, potato starch, urea, and solid, semi-solid or liquid forms Other carriers suitable for use in the manufacture of a formulation of the invention. In addition, auxiliary, stabilizing, emulsifying, thickening, coloring, flavoring, and perfuming agents may be used.
[0016]
Compound (I) is included in the pharmaceutical composition in an amount effective to produce the desired effect on a particular disease process or condition. Compound (I) is included in an amount sufficient to provide a neurotrophic effect or stimulate nerve cell growth.
[0017]
Mammals that can be treated using the methods of the present invention include domestic mammals such as cows, horses, pigs, domestic animals such as dogs, cats, rats, mice, rabbits, hamsters, primates, and humans. Can be
Suitable modes of administration or application of the product or composition comprising compound (I) to a human include injection or oral administration.
The therapeutically effective amount or dosage of Compound (I) may vary from patient to patient and will depend on the age and condition of the individual patient being treated, but will range from about 0.0001-1000 mg, preferably Daily dosages of active ingredient ranging from 0.001 to 500 mg, more preferably 0.01 to 100 mg, are generally given for the treatment of disease and are administered in the range of about 0.001 to 0.01 mg, 0.2 Average single doses of -0.5 mg, 1 mg, 5 mg, 10 mg, 50 mg, 100 mg, 250 mg, 500 mg are generally administered. The daily dosage for chronic administration in humans will be in the range of about 0.1-30 mg / kg / day. Compound (I) may also be administered or applied simultaneously, separately or sequentially with other agents having neurotrophic activity or nerve cell growth stimulating activity.
[0018]
The pharmaceutical compositions of the present invention may be used in mammalian subjects in need of such treatment to promote nerve regeneration and functional recovery, stimulate neurite outgrowth, and thereby treat various neuropathological conditions. It can be administered periodically to the body (eg, a human patient). Various neuropathological conditions include damage to peripheral and central nervous systems caused by physical injury (eg, spinal cord injury and trauma, sciatic or facial nerve injury or injury, limb transplantation after amputation); disease (eg, diabetes) Cancer chemotherapeutics (eg, neuropathy induced by acrylamide, taxol, vinca alkaloids and doxorubicin); sequelae—language disorders (eg, dysarthria) associated with cerebral infarction, hemorrhagic infarction, etc., consciousness Opacity, dyskinesia, etc .; and trigeminal neuralgia, glossopharyngeal neuralgia, facial paralysis, myasthenia gravis, muscular dystrophy, amyotrophic lateral sclerosis, progressive muscular atrophy, progressive hereditary medulla oblongata, herniated disc Herniated, ruptured or propagated vertebral isk syndrome, cervical spondylopathy, plexopathy, thoraco-oral destruction syndrome, peripheral neuropathy (eg, lead, acrylamide, gamma-diketone (glue-sniffer's neuropathy), carbon disulfide, dapsone, tic (Ticks), various peripheral neuropathies including but not limited to porphyria, Guillain-Barre syndrome, Alzheimer's disease, Parkinson's disease and Huntington's disease, and neurological diseases including but not limited to neurological diseases. No.
[0019]
Transection of the peripheral nerve or spinal cord injury can be achieved by administering a nerve growth stimulating amount of the drug to a mammal and injecting a nerve graft, such as an allograft, into the peripheral nerve or spinal cord (Osawa et al., J. Am. Neurocytol. 19: 833-849, 1990; Buttemeyer et al., Ann. Plastic Surgery 35: 396-401, 1995) or artificial nerve grafts (Madison and Archivald, Exp. Neurol. 128: 266-275, 1994; Wel). Exp. Neurol. 146: 395-402, 1997). The space between the tear ends of the peripheral nerve or spinal cord is preferably a non-cellular gap-filling material such as collagen, methylcellulose; or Schwann cells (Xu et al., J. Neurocytol. 26: 1-16, 1997), olfactory cells. And a suspension of cells that promote nerve cell growth, such as sheath cells (Li et al., Science 277: 2000-2002, 1997). The nerve growth promoting agent can be included with such cellular or non-cellular gap filling materials, or can be administered systemically before, during, or after the nerve transplantation method.
[0020]
In particular, Compound (I) is useful for treating neurological injuries / dysfunction, polymyositis, Guillain-Barre syndrome, Meniere's disease, polyneuritis, solitary neuritis (isolated neuritis), Alzheimer's disease, Parkinson's disease, muscular atrophy Treatment of lateral sclerosis (ALS), Huntington's disease, radiculopathy, diabetic neuropathy, chemotherapy-induced neuropathy, senile dementia, vascular dementia, multiple sclerosis, physical paralysis, or spinal cord injury or Useful for prevention.
[0021]
The following examples illustrate the invention in more detail. While these examples describe certain aspects or embodiments of the present invention, it should be understood that they are not intended to limit the scope of the present invention.
[0022]
Example 1 Treatment with Compound (I) Significantly Increases Neurite Length of Hippocampal Neurons
Preparation of cell culture:
Banker as well as Cowan Fetal hippocampal neurons were obtained from embryonic day 18.5 ("E18.5") rat pups according to (Brain Research, 1977, 126: 397-425). Briefly, the hippocampus was removed, minced and 100 I. U. Incubate with papain at 37 ° C. for 45 minutes and cells were cultured in complete neuronal medium (minimal essential medium without L-glutamine (GIBCO, Grand Island, NY), high glucose minimal essential medium (GIBCO) 1.5 ml / 100 ml, serum expansion) The cells were resuspended in a medium (Hito + Tm; Collaborative Research Inc, Lexington, Mass.) 0.1 ml / 100 ml, glutamine (GIBCO), 5% fetal bovine serum (GIBCO).
[0023]
Cells were seeded on poly-L-lysine-coated coverslips (500 cells / coverslip). The coverslip was inverted on the dish. The dishes were pre-coated with a monolayer of cortical astrocytes.
[0024]
Analysis of hippocampal neuron axon length:
Hippocampal neurons (identified by their characteristic polarity and dendrites) were examined daily and photographed randomly at 72 hours (9-12 frames / coverslip). The length of axons (defined as the longest protrusion) is determined using a Houston Instrument HI-PAD digitizing tablet coupled to an IBM XT computer equipped with appropriate software (Bioquant IV, R & M Biometrics, Nashville, TN). Measured on photographic prints (only protrusions greater than three times the length of the cell body were measured). Data from similarly treated coverslips (3 or 4 per group) were not different and were therefore combined. Mean values were calculated and compared using one factor ((Compound (Ia) or tacrolimus-treated group) versus (untreated control group)) ANOVA followed by the Newman-Kuels multiple comparison test (WINKS 4.62 Professional Edition).
[0025]
result:
At 72 hours, there was no significant difference between the untreated control group and the 10 nM tacrolimus treated group. However, the neurite length was statistically significantly increased in the compound (Ia) 10 nM treatment group. See the results in Table 1 below.
[0026]
[Table 1]
Figure 2004527472
[0027]
Example 2: Treatment with compound (I) increases mean neurite length of SH-SYSY human neuroblastoma
Preparation of SH-SY5Y neuroblastoma culture:
10% fetal calf serum (SIGMA), penicillin 50I. U. / Ml, 7% CO 2 in DMEM medium (GIBCO) supplemented with 50 μg / ml streptomycin (GIBCO).2SH-SY5Y human neuroblastoma was maintained at 37 ° C in medium. Cells were seeded at 15,000 cells / well in 6-well plates and treated with 0.4 μM aphidicolin (SIGMA). On day 5, cells were washed and treated with 10 ng / ml nerve growth factor (NGF) in the presence or absence of tacrolimus (10 nM) or compound (Ia) (1 nM) (to induce process growth). The medium was changed after 96 hours and replaced with fresh medium after a further 72 hours (total time 168 hours). All experiments were performed in duplicate and data were averaged for each treatment group.
[0028]
Analysis of neurite length in SH-SY5Y neuroblastoma
Cells were randomly photographed at 168 hours (20 fields per well) for projection length analysis. Neurite length was measured on photographic prints using a Houston Instrument HI-PAD digitizing tablet coupled to an IBM XT computer equipped with appropriate software (Bioquant IV, R & M Biometrics, Nashville, TN) ( Only those protrusions that were more than twice the length of the cell body were measured). Data from wells treated similarly did not differ and were therefore combined. Mean values were calculated and compared using one factor ((Compound (Ia) or tacrolimus treated sample) vs. (NGF alone treated sample)) ANOVA followed by the Newman-Kuels multiple comparison test (WINKS 4.62 Professional Edition).
[0029]
result:
Neurite length measurements indicate that both compound (Ia) (1 nM) and tacrolimus (10 nM) significantly increase neurite length at 168 hours compared to NGF (10 ng / ml) alone. This has been proven. However, the effect of 1 nM compound (Ia) in combination with NGF was greater than the effect of 10 nM tacrolimus in combination with NGF.
[0030]
[Table 2]
Figure 2004527472
[0031]
Example 3: Treatment with compound (I) promotes functional recovery in a rat sciatic nerve crush model
Animals and surgical methods:
Nine 6-week-old male Sprague-Dawley rats were anesthetized with 2% halothane, the right sciatic nerve was exposed, and the nerve was crushed twice at the level of the hip joint (for a total of 60 seconds using No. 7 Dumont jewelry forceps). ). The crush site was marked by tying a sterile 9-O suture to the nerve sheath.
[0032]
Preparation and administration of compound (Ia)
Compound (Ia) was dissolved in a solvent containing 30% dimethyl sulfoxide (DMSO): 70% saline. Three nerve crushed rats were injected subcutaneously daily with compound (Ia) (1 or 5 mg / kg) or an equal volume of solvent (30% DMSO in saline) (5 ml / kg).
[0033]
Behavior evaluation
Animals were tested daily until the day of perfusion (day 18). The following semi-quantitative scale was used to assess animal functional recovery:
0: foot paralyzed, bends outward when walking, toes bend;
1: The feet are straight and the toes can move;
2: You can walk with your feet constantly;
3: Toes open when walking;
4: walking away from heel, showing near normal toe spread;
Animals with intermediate abilities received partial scores: +, 0.25; ++, 0.5; ++, 0.75.
[0034]
Fixation and preparation of tissue:
Eighteen days after nerve crush, rats were deeply anesthetized with 4% halothane, treated with heparin, treated with 4% paraformaldehyde in 0.1 M sodium phosphate buffer (pH 7.4) for 10 seconds, then with 5% glutaraldehyde ( (IL) in 0.1 M sodium phosphate buffer (pH 7.4) and fixed at 4 ° C. for 24 hours. Tissue samples were taken from the sciatic nerve at a known (5 mm) distance from the crush site. In this study, we report only data on the posterior tibial nerve branch that controls the soleus muscle. Tissues are placed in 0.1 M sodium phosphate buffer (pH 7.4), fixed with 1% osmium tetroxide (in 0.1 M phosphate buffer) for 2.5 hours, dehydrated in ethanol, plastic Embedded. Semi-thin sections were stained with uranyl acetate and lead citrate, mounted on a 75 mesh grid supported by a film, and observed using a JEOL100CX electron microscope.
[0035]
Morphological analysis:
Axon diameter analysis was performed on the tibial nerve. The number of regenerating myelinated axons was counted using an electron microscope. The average value and standard error were calculated for the solvent-treated group, the compound (Ia) (1 mg / kg) treated group, and the compound (Ia) (5 mg / kg) treated group.
[0036]
Statistical analysis:
For behavioral analysis, the mean value of functional recovery was compared using a one-way ANOVA followed by a Newman-Keuls multiple comparison test for comparison of individual values. For morphological analysis, the average values of axon numbers were compared using a one-way ANOVA followed by a Newman-Keuls multiple comparison test for comparison of individual values.
[0037]
result:
Functional recovery:
Functional recovery was observed on days 15-17 and occurred earlier in both 1 mg / kg and 5 mg / kg treated rats than in vehicle treated rats. See Table 3 below.
[0038]
[Table 3]
Figure 2004527472
[0039]
electronic microscope
Morphological examination was performed 18 days after nerve crush.
Nerve area (5,000 μm2The number of regenerating myelinated axons per vehicle was 5.5 ± 2.7 (mean ± SEM) in the vehicle-treated rats, and 19 ± 2.4 and 20 in the 1 mg / kg-treated and 5 mg / kg-treated rats, respectively. Dramatically increased to ± 2.9. See Table 4 below.
[0040]
[Table 4]
Figure 2004527472
[0041]
Example 4: Treatment with compound (I) promotes functional recovery in a rat spinal cord injury model
(1) Method
Animals and surgical methods
Twenty-eight male Sprague-Dawley rats, 6 weeks old, were anesthetized with 2% halothane, laminectomy was performed at T10 / T11, and hemisection of the spinal cord was performed at the level of the T10 / T11 spinal cord.
Preparation and administration of compound (Ia)
Compound (Ia) was dissolved in a solvent containing 30% dimethyl sulfoxide (DMSO): 70% saline. Spinal cord injured rats were injected daily subcutaneously with compound (Ia) (2 mg / kg) or an equal volume of solvent (30% DMSO in saline) (5 ml / kg) for 7 weeks after surgery.
[0042]
Evaluation of functional recovery
Two weeks after injury, functional recovery was assessed using a modified Tarlov / Klinger scale, modified beam test, and footprint test.
A. Alteration Tarlov / Klinger scale
Rats were allowed to walk freely in the open field for one minute and were classified as 0-6 based on the scale below.
0: No hind limb movement
1: Bare hind limb movement can be barely confirmed
2: Damaged hind limb joints (hip, knee or ankle) move actively, but do not cooperate and cannot support weight
3: Walking with alternating hind limbs is accompanied by propulsion, but cannot support weight
4: Can support weight with injured hind limb
5: Some walking disorders are seen
6: Normal walking
[0043]
B. narrow beam test
The test was performed in order from 7.7 cm, 4.7 cm, 2.7 cm, and 1.7 cm in width using a wooden timber (length 1.5 m). Rats were allowed to walk on the stick and the narrowest stick that could walk without slipping was recorded in at least two trials.
0: Unable to walk with any square timber
1: Walkable with 7.7cm square timber
2: Can be walked with 4.7cm square lumber
3: Walkable with 2.7cm square timber
4: Walkable with 1.7cm square lumber
[0044]
C. Footprint test
Rat hind limbs were inked and footprints were made on paper covering a narrow runway 60 cm long and 7.5 cm wide. A 5-point footprint score was determined using a series of at least six consecutive footprints.
0: Dragging hind limb, no footprint is observed
1: At least three toe marks are seen in at least three footprints
2: More than twice the paw is bent outward or inward compared to the value before nerve crush of individual rat
3: No sign of toe dragging, but sign of bent leg
4: The foot is not bent outside or inside (less than twice the angle of the baseline value), but one or more heel marks are observed
5: No trace of heel is observed
[0045]
Statistical analysis:
For behavioral analysis, one-way ANOVA for comparison of individual values was used, followed by the Newman-Keuls multiple comparison test to compare the mean scores of each functional test.
[0046]
(2) Result
Functional recovery
In three functional recovery tests using the modified Tarlov / Klinger scale (Table 5), the narrow beam test (Table 6), and the footprint test (Table 7), compound (Ia) was converted to the modified Tarlov / Klinger scale (Table 5). Improved motor dysfunction in the narrow beam test (Table 6) and the footprint test (Table 7).
[0047]
[Table 5]
Figure 2004527472
[0048]
[Table 6]
Figure 2004527472
[0049]
[Table 7]
Figure 2004527472
[0050]
Example 5: Compound (Ia) binds to FKBP12, but unlike tacrolimus, exerts little or no immunosuppressive effect
(1) Binding experiment with FKBP12
Tamura, K .; (Biochemical and Biophysical Research Communications, Vol. 202, No. 1, 437-499, 1994), and a binding experiment was performed. Table 8 shows the results.
[0051]
(2) Mixed lymphocyte reaction (MLR)
The MLR test was performed based on a method similar to that of US Pat. No. 4,929,611.
Table 8 shows the results.
[0052]
[Table 8]
Figure 2004527472
[0053]
The above results indicate that compound (Ia) can bind to FKBP12 but has no immunosuppressive activity.
The above results indicate a strong neurotrophic effect of compound (I) using both in vitro and in vivo models. In two cell culture models, compound (I) increased neurite growth even at low concentrations. In addition, systemic administration of low doses of Compound (I) promoted functional recovery following nerve crush injury by increasing the rate of axonal regeneration of the sciatic nerve and promoted functional recovery from spinal cord injury.
Further, as described above, compound (I) provides potent neurotrophic activity or nerve cell growth stimulating activity, although it has no immunosuppressive activity. Thus, the present invention provides useful neurotrophic agents for stimulating or promoting nerve growth or regeneration, especially when the immunosuppressive effect is not advantageous or desired.
[0054]
Other aspects of the invention include:
A product comprising a package and the compound (I) contained in the package, wherein the compound (I) is a product which is therapeutically effective in preventing or treating neurological dysfunction. A product comprising a label or statement indicating that Compound (I) can or should be used for the prevention or treatment of nerve injury / dysfunction.
A commercial package comprising a pharmaceutical composition comprising Compound (I) as described above and a description attached thereto, wherein the description allows the use of Compound (I) for the prevention or treatment of nerve injury / dysfunction; or A commercial package stating that it should be used.
A composition, such as a cell suspension, tissue or implant, comprising cells treated with compound (I). Such compositions are useful for repairing damage to the nervous system. Such compositions may also include other types of cell suspensions that promote or support nerve cell growth (eg, Schwann cells or oligodendrocytes, myelin-producing cells such as glial and sheath cells, extracellular cells such as collagen, etc.). Substrate substances or other specific neuromodulators such as cytokines, mitogens, immunophilins, and neurotrophins (eg, NGF-1, BDNF, CNTF, NT-3, NT-4, and NT-5) ) Other nerve cell growth stimulating agents may be included.
[0055]
Grafts, such as homografts, allografts or xenografts, may also be used to facilitate the use of compounds (I) for nerve growth and for use as grafts and for other applications. May be processed.
[0056]
Incorporation by reference
The contents of each document, patent application or patent publication cited or referred to in this disclosure are incorporated by reference in their entirety. The contents of any patent document to which this application claims priority is also incorporated by reference in its entirety. Specifically, the contents of US Provisional Application No. 60 / 258,500 are incorporated by reference.
[0057]
Modifications and other embodiments
Obviously, many modifications and variations of the present invention are possible in light of the above teachings. Therefore, it is to be understood that, within the scope of the appended claims, the invention may be practiced other than as specifically described herein. It will be apparent to those skilled in the art that various modifications and variations of the described compositions and methods, and the concepts of the present invention, do not depart from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be construed as limited to such specific embodiments. Various modifications of the described modes for carrying out the invention will be obvious to those skilled in the medical, biological, chemical, or pharmacology arts or related fields and are within the scope of the invention. Is intended.

Claims (42)

神経栄養剤の製造のための、以下の式:
Figure 2004527472
の化合物の使用。
The following formula for the production of a neurotrophic agent:
Figure 2004527472
Use of compounds.
神経栄養剤が神経傷害/機能不全の予防又は治療用である、請求項1に記載の使用。The use according to claim 1, wherein the neurotrophic agent is for prevention or treatment of nerve injury / dysfunction. 向神経剤が神経細胞成長又は再生の刺激又は促進用である、請求項1に記載の使用。The use according to claim 1, wherein the neurotrophic agent is for stimulating or promoting nerve cell growth or regeneration. 向神経剤が神経傷害からの機能回復の促進用である、請求項1に記載の使用。The use according to claim 1, wherein the neurotrophic agent is for promoting the recovery of function from nerve injury. 神経傷害/機能不全が、多発性筋炎、ギラン−バレー症候群、メニエール病、多発性神経炎、単発神経炎(孤立性神経炎)、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症(ALS)、ハンチントン病、神経根障害、糖尿病性神経障害、化学療法誘発神経障害、老人性痴呆症、血管性痴呆症、多発性硬化症、身体麻痺又は脊髄損傷である、請求項1に記載の使用。Nerve injury / dysfunction may include polymyositis, Guillain-Barre syndrome, Meniere's disease, polyneuritis, solitary neuritis (isolated neuritis), Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS) The use according to claim 1, which is Huntington's disease, radiculopathy, diabetic neuropathy, chemotherapy-induced neuropathy, senile dementia, vascular dementia, multiple sclerosis, paralysis or spinal cord injury. 神経傷害/機能不全が、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症、糖尿病性神経障害、化学療法誘発神経障害又は脊髄損傷である、請求項1に記載の使用。The use according to claim 1, wherein the nerve injury / dysfunction is Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, diabetic neuropathy, chemotherapy-induced neuropathy or spinal cord injury. 化合物(I)が、その医薬として許容できる塩、誘導体、プロドラッグ又は溶媒和物の形態である、請求項1に記載の使用。The use according to claim 1, wherein compound (I) is in the form of a pharmaceutically acceptable salt, derivative, prodrug or solvate thereof. 以下の式:
Figure 2004527472
を有する化合物(I)を含む組成物。
The following formula:
Figure 2004527472
A composition comprising compound (I) having the formula:
化合物(I)が、その医薬として許容できる塩、誘導体、プロドラッグ又は溶媒和物の形態である、請求項8に記載の組成物。9. The composition according to claim 8, wherein compound (I) is in the form of a pharmaceutically acceptable salt, derivative, prodrug or solvate thereof. 化合物(I)が化合物(Ia)の形態である、請求項8に記載の組成物。9. The composition according to claim 8, wherein compound (I) is in the form of compound (Ia). 少なくとも1つの他の神経栄養剤を更に含む、請求項8に記載の組成物。9. The composition of claim 8, further comprising at least one other neurotrophic agent. 包装物と包装物内に含まれる化合物(I)とを含む製品又はキットであって、包装物が、神経傷害/機能不全の予防、改善又は治療に化合物(I)を使用できること又は使用すべきであることを指示しているラベル又は記載物を含む、製品又はキット。A product or kit comprising a package and a compound (I) contained within the package, wherein the package is capable of or should use compound (I) for the prevention, amelioration or treatment of nerve injury / dysfunction. A product or kit comprising a label or statement indicating that 化合物(I)を含む医薬組成物とそれに添付される記載物とを含む商業パッケージ又はキットであって、記載物が、神経傷害/機能不全の予防、改善又は治療に化合物(I)を使用できること又は使用すべきであることを記述している、商業パッケージ又はキット。A commercial package or kit comprising a pharmaceutical composition comprising Compound (I) and a description attached thereto, wherein the description is capable of using Compound (I) for prevention, amelioration or treatment of nerve injury / dysfunction. Or a commercial package or kit stating that it should be used. 神経栄養剤の生産又は製造方法であって、化合物(I)を、医薬として許容できる担体、溶媒又は賦形剤と混合することを含む方法。A method for producing or producing a neurotrophic agent, comprising mixing Compound (I) with a pharmaceutically acceptable carrier, solvent or excipient. 神経傷害/機能不全の予防、改善又は治療の方法であって、その必要のある被験体に有効量の化合物(I)を投与することを含む方法。A method of preventing, ameliorating or treating nerve injury / dysfunction, comprising administering to a subject in need thereof an effective amount of Compound (I). 被験体が哺乳動物である、請求項15に記載の方法。16. The method according to claim 15, wherein the subject is a mammal. 被験体がヒトである、請求項15に記載の方法。16. The method of claim 15, wherein the subject is a human. 神経傷害/機能不全が、多発性筋炎、ギラン−バレー症候群、メニエール病、多発性神経炎、単発神経炎(孤立性神経炎)、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症(ALS)、ハンチントン病、神経根障害、糖尿病性神経障害、化学療法誘発神経障害、老人性痴呆症、血管性痴呆症、多発性硬化症、身体麻痺又は脊髄損傷である、請求項15に記載の方法。Nerve injury / dysfunction may include polymyositis, Guillain-Barre syndrome, Meniere's disease, polyneuritis, solitary neuritis (isolated neuritis), Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS) 16. The method of claim 15, wherein the disease is Huntington's disease, radiculopathy, diabetic neuropathy, chemotherapy-induced neuropathy, senile dementia, vascular dementia, multiple sclerosis, paralysis or spinal cord injury. 神経傷害/機能不全が、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症、糖尿病性神経障害、化学療法誘発神経障害又は脊髄損傷である、請求項15に記載の方法。16. The method of claim 15, wherein the nerve injury / dysfunction is Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, diabetic neuropathy, chemotherapy-induced neuropathy or spinal cord injury. 神経細胞成長又は再生の刺激又は促進の方法であって、神経細胞を化合物(I)と接触させることを含む方法。A method of stimulating or promoting nerve cell growth or regeneration, comprising contacting a nerve cell with compound (I). 軸索成長若しくは再生の速度、又は神経細胞の長さを増加させる方法であって、神経細胞を化合物(I)と接触させることを含む方法。A method of increasing the rate of axonal growth or regeneration, or the length of a nerve cell, comprising contacting the nerve cell with Compound (I). その必要のある組織で神経細胞成長を増加させる方法であって、組織に化合物(I)を投与することを含む方法。A method of increasing nerve cell growth in a tissue in need thereof, comprising administering compound (I) to the tissue. 組織が、脳組織、脊髄組織又は末梢神経組織からなる群より選択される、請求項22に記載の方法。23. The method of claim 22, wherein the tissue is selected from the group consisting of brain tissue, spinal cord tissue, or peripheral nerve tissue. 神経傷害からの機能回復の促進方法であって、その必要のある被験体に有効量の化合物(I)を投与することを含む方法。A method of promoting functional recovery from a nerve injury, comprising administering to a subject in need thereof an effective amount of compound (I). 神経傷害が、火傷、外傷性傷害、機械的傷害、手術性傷害、生理的傷害、病理的傷害及び免疫的傷害からなる群より選択される、請求項24に記載の方法。25. The method of claim 24, wherein the nerve injury is selected from the group consisting of burns, traumatic injuries, mechanical injuries, operative injuries, physiological injuries, pathological injuries and immunological injuries. 断裂した末梢神経又は脊髄の修復方法であって、末梢神経又は脊髄の断裂末端を有効量の化合物(I)と接触させることを含む方法。A method for repairing a torn peripheral nerve or spinal cord, comprising contacting the torn end of the peripheral nerve or spinal cord with an effective amount of compound (I). 被験体における断裂した末梢神経又は脊髄の修復方法であって、被験体に神経成長刺激量の化合物(I)を投与し、末梢神経又は脊髄に移植することを含む方法。A method for repairing a torn peripheral nerve or spinal cord in a subject, comprising administering to the subject a nerve growth stimulating amount of compound (I) and transplanting the compound into the peripheral nerve or spinal cord. 移植片が同種異系移植片である、請求項27に記載の方法。28. The method of claim 27, wherein the graft is an allograft. 移植片が人工神経移植片である、請求項27に記載の方法。28. The method of claim 27, wherein the implant is an artificial nerve implant. 末梢神経又は脊髄の断裂末端間の空間を、非細胞性間隙充填物質で充填することを更に含む、請求項27に記載の方法。28. The method of claim 27, further comprising filling the space between the tear ends of the peripheral nerve or spinal cord with a non-cellular gap filling material. 末梢神経又は脊髄の断裂末端間の空間を細胞懸濁液で充填することを更に含む、請求項27に記載の方法。28. The method of claim 27, further comprising filling the space between the tear ends of the peripheral nerve or spinal cord with a cell suspension. 化合物(I)で処理された神経細胞を含む組成物。A composition comprising a nerve cell treated with compound (I). 化合物(I)で処理された神経細胞を含む組織。Tissue containing nerve cells treated with compound (I). 化合物(I)で処理された神経細胞を含む移植片。An implant containing nerve cells treated with compound (I). 移植片が、同種同系移植片、同種異系移植片又は異種移植片である、請求項34に記載の移植片。35. The graft of claim 34, wherein the graft is an allograft, an allograft or a xenograft. 神経傷害からの機能回復の促進方法であって、その必要のある被験体に、化合物(I)で処理された細胞を含む組成物の有効量を投与することを含む方法。A method of promoting functional recovery from a nerve injury, comprising administering to a subject in need thereof an effective amount of a composition comprising cells treated with compound (I). 神経傷害が、外傷性傷害、機械的傷害、手術性傷害、生理的傷害、病理的傷害及び免疫的傷害からなる群より選択される、請求項36に記載の方法。37. The method of claim 36, wherein the nerve injury is selected from the group consisting of traumatic injury, mechanical injury, surgical injury, physiological injury, pathological injury, and immune injury. 断裂した末梢神経又は脊髄の修復方法であって、末梢神経又は脊髄の断裂末端を、化合物(I)で処理された細胞を含む組成物の有効量と接触させることを含む方法。A method for repairing a torn peripheral nerve or spinal cord, comprising contacting the torn peripheral nerve or spinal cord end with an effective amount of a composition comprising cells treated with compound (I). 化合物(I)及び少なくとも1つの神経細胞成長促進剤で処理された細胞、組織又は移植片を含む組成物。A composition comprising cells, tissues or grafts treated with compound (I) and at least one nerve cell growth promoter. 神経成長促進剤が、神経成長を促進する細胞懸濁液、非細胞性間隙充填物質又は神経成長因子からなる群より選択される、請求項39に記載の組成物。40. The composition of claim 39, wherein the nerve growth promoting agent is selected from the group consisting of a cell suspension that promotes nerve growth, a non-cellular gap filling material, or a nerve growth factor. 神経成長促進剤としてコラーゲン又はメチルセルロースを含む、請求項39に記載の組成物。The composition according to claim 39, comprising collagen or methylcellulose as a nerve growth promoter. 神経成長促進剤として細胞懸濁液を含む、請求項39に記載の組成物。40. The composition according to claim 39, comprising a cell suspension as a nerve growth promoter.
JP2002554109A 2000-12-29 2001-12-31 Neurotrophic tacrolimus analog Ceased JP2004527472A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25850000P 2000-12-29 2000-12-29
PCT/US2001/050419 WO2002053159A1 (en) 2000-12-29 2001-12-31 Neurotrophic tacrolimus analogs

Publications (2)

Publication Number Publication Date
JP2004527472A true JP2004527472A (en) 2004-09-09
JP2004527472A5 JP2004527472A5 (en) 2005-12-22

Family

ID=22980816

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2002554109A Ceased JP2004527472A (en) 2000-12-29 2001-12-31 Neurotrophic tacrolimus analog

Country Status (18)

Country Link
EP (1) EP1353671A4 (en)
JP (1) JP2004527472A (en)
KR (2) KR100794204B1 (en)
CN (1) CN1293877C (en)
AR (1) AR035411A1 (en)
AU (1) AU2002231277B2 (en)
BR (1) BR0116762A (en)
CA (1) CA2433384A1 (en)
CZ (1) CZ20032060A3 (en)
HU (1) HUP0302521A3 (en)
IL (1) IL156664A0 (en)
MX (1) MXPA03005941A (en)
NO (1) NO20032913D0 (en)
NZ (1) NZ527209A (en)
PL (1) PL366301A1 (en)
RU (1) RU2288716C2 (en)
WO (1) WO2002053159A1 (en)
ZA (1) ZA200305806B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007094166A1 (en) * 2006-02-14 2007-08-23 Meiji Dairies Corporation Therapeutic agent for spinal cord injury
JP2009523763A (en) * 2006-01-18 2009-06-25 アンスティテュート キュリー Method for treating Huntington's disease by inhibiting dephosphorylation of huntingtin in S421
JP2016528171A (en) * 2013-05-24 2016-09-15 クロノス セラピューティクス リミテッドChronos Therapeutics Limited Tacrolimus for use in the treatment of diseases characterized by the deposition of protein aggregates in neurons

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003901023A0 (en) * 2003-03-04 2003-03-20 Fujisawa Pharmaceutical Co., Ltd. New use
WO2005067928A1 (en) * 2004-01-20 2005-07-28 Astellas Pharma Inc. Method for treating erectile dysfunction
US9149459B2 (en) 2008-07-23 2015-10-06 Novartis Ag Sphingosine 1 phosphate receptor modulators and their use to treat muscle inflammation
EP2817009A1 (en) * 2012-02-23 2014-12-31 INSERM - Institut National de la Santé et de la Recherche Médicale Calcineurin inhibitors for use in the treatment of lesional vestibular disorders

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2799208B2 (en) * 1987-12-09 1998-09-17 フアイソンズ・ピーエルシー Macrocyclic compound
GR1001225B (en) * 1989-09-14 1993-06-30 Fisons Plc Novel macrocyclic compositions and new application method thereof
US5541193A (en) 1991-09-05 1996-07-30 Abbott Laboratories Heterocycle-containing macrocyclic immunomodulators
GB9202196D0 (en) * 1992-02-01 1992-03-18 Fisons Plc Method of treatment
GB9218027D0 (en) * 1992-08-25 1992-10-14 Fisons Plc Novel method of treatment
GB9917158D0 (en) * 1999-07-21 1999-09-22 Fujisawa Pharmaceutical Co New use

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009523763A (en) * 2006-01-18 2009-06-25 アンスティテュート キュリー Method for treating Huntington's disease by inhibiting dephosphorylation of huntingtin in S421
WO2007094166A1 (en) * 2006-02-14 2007-08-23 Meiji Dairies Corporation Therapeutic agent for spinal cord injury
JP2016528171A (en) * 2013-05-24 2016-09-15 クロノス セラピューティクス リミテッドChronos Therapeutics Limited Tacrolimus for use in the treatment of diseases characterized by the deposition of protein aggregates in neurons

Also Published As

Publication number Publication date
NZ527209A (en) 2005-09-30
CA2433384A1 (en) 2002-07-11
RU2288716C2 (en) 2006-12-10
HUP0302521A2 (en) 2003-11-28
BR0116762A (en) 2004-08-10
NO20032913D0 (en) 2003-06-24
CN1538843A (en) 2004-10-20
HUP0302521A3 (en) 2007-03-28
MXPA03005941A (en) 2005-02-14
KR20040007431A (en) 2004-01-24
EP1353671A1 (en) 2003-10-22
ZA200305806B (en) 2005-01-26
WO2002053159A1 (en) 2002-07-11
KR100794204B1 (en) 2008-01-14
PL366301A1 (en) 2005-01-24
CN1293877C (en) 2007-01-10
RU2003123493A (en) 2005-01-20
CZ20032060A3 (en) 2004-01-14
AR035411A1 (en) 2004-05-26
KR20070030331A (en) 2007-03-15
EP1353671A4 (en) 2004-07-14
IL156664A0 (en) 2004-01-04
AU2002231277B2 (en) 2006-11-30

Similar Documents

Publication Publication Date Title
KR100298808B1 (en) Pharmaceutical Compositions Including Lilusol for Treatment of Neurological Lesions Associated with Traumatic Injury
Gaviria et al. Neuroprotective effects of a novel NMDA antagonist, Gacyclidine, after experimental contusive spinal cord injury in adult rats
Cui et al. CNTF, not other trophic factors, promotes axonal regeneration of axotomized retinal ganglion cells in adult hamsters.
RU2197240C2 (en) Methods and compositions for growth stimulation of neuritis
JP7082186B2 (en) How to treat amyotrophic lateral sclerosis with pridopidin
TWI452039B (en) Anti-neurodegenerative diseases agents
Yin et al. Taxol-modified collagen scaffold implantation promotes functional recovery after long-distance spinal cord complete transection in canines
KR20010032357A (en) Application of Substituted Aminomethyl Chromans in order to Prevent Neural Degeneration and to Promote Neural Regeneration
JP6708869B2 (en) Nervous system therapeutic agent
DE69635754T9 (en) MEDICAMENTS FOR PREVENTING CONDITIONAL STENOSIS AS A RESULT OF NON-BYPASS INVASIVE INTERVENTION
Cui et al. Synergistic effect of Nogo-neutralizing antibody IN-1 and ciliary neurotrophic factor on axonal regeneration in adult rodent visual systems
AU2002231277B2 (en) Neurotrophic tacrolimus analogs
AU2002231277A1 (en) Neurotrophic tacrolimus analogs
y Ribotta et al. Strategies for regeneration and repair in spinal cord traumatic injury
US6284771B1 (en) Method for treating schizophrenia
JP6835738B2 (en) Parthenolide and its derivatives for the treatment of axonal disorders
WO2017057562A1 (en) Therapeutic agent and/or prophylactic agent for peripheral nerve disorder or spinal injury
Hitzenberger et al. Pharmacological properties of piracetam: Rationale for use in stroke patients
US20040077676A1 (en) Neurotrophic tacrolimus analogs
JP2009504636A (en) Pharmaceutical composition for treatment of nerve injury comprising plasma or serum
Itoh et al. The effect of neurotrophic pyrimidine heterocyclic compounds, MS-818 and MS-430, on the regeneration of injured peripheral nerves
US6852496B1 (en) Methods of screening for agents that promote nerve cell growth
Rodinskii et al. Effects of systemic injections of gamma-hydroxybutyrate on the recovery of functions of the distal hindlimb muscles in rats after compression of the sciatic nerve
CN117530937A (en) Application of Idebenone in preparation of drugs for preventing and/or treating tendon injury
Kim et al. on Functional Recovery after Sciatic Nerve Injury

Legal Events

Date Code Title Description
A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20041124

A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20041124

A711 Notification of change in applicant

Free format text: JAPANESE INTERMEDIATE CODE: A712

Effective date: 20050520

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20080701

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20080828

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20081202

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20091208

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20100112

A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20100202

A045 Written measure of dismissal of application [lapsed due to lack of payment]

Free format text: JAPANESE INTERMEDIATE CODE: A045

Effective date: 20100622