TW200950787A - Controlled release corticosteroid compositions and methods for the treatment of otic disorders - Google Patents

Controlled release corticosteroid compositions and methods for the treatment of otic disorders Download PDF

Info

Publication number
TW200950787A
TW200950787A TW98116106A TW98116106A TW200950787A TW 200950787 A TW200950787 A TW 200950787A TW 98116106 A TW98116106 A TW 98116106A TW 98116106 A TW98116106 A TW 98116106A TW 200950787 A TW200950787 A TW 200950787A
Authority
TW
Taiwan
Prior art keywords
formulation
dehydrocortisol
ear
methyl
dehydrocorticosterol
Prior art date
Application number
TW98116106A
Other languages
Chinese (zh)
Other versions
TWI382839B (en
Inventor
Jay Lichter
Benedikt Vollrath
Andrew M Trammel
Sergio G Duron
Fabrice Piu
Luis A Dellamary
Qiang Ye
Carl Lebel
Michael Christopher Scaife
Jeffrey P Harris
Original Assignee
Otonomy Inc
Univ California
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 Otonomy Inc, Univ California filed Critical Otonomy Inc
Publication of TW200950787A publication Critical patent/TW200950787A/en
Application granted granted Critical
Publication of TWI382839B publication Critical patent/TWI382839B/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed herein are compositions and methods for the treatment of otic disorders with steroid, NSAID, and/or adenosine triphosphatase ("ATPase") modulator agents. In these methods, the steroidal, NSAID, and/or ATPase compositions and formulations are administered locally to an individual afflicted with an otic disorder, through direct application of these compositions and formulations onto or via perfusion into the targeted auris structure(s).

Description

φΦ

200950787 六、發明說明: 【發明所屬之技術領域】 舍椎動物具有一對耳朵’其對稱位於頭部相對側。耳做 為谓測聲音感受器官及維持身體位置平衡的器官。耳大致 可分為三部份:外耳、中耳(耳中部)及内耳(耳内部)。 【發明内容】 本發明揭露-種用於至少—皮質類固醇至耳的至少一姓 構或區域的控制釋放之組成物、配方、製造方法、治療: 法使用、套件、及傳送裝置。本發明揭露用於傳送皮質 類固醇至耳的控制釋放配方。在某些實施财,耳的標的 部份為中耳或耳中部。在某些實施财,耳的標的部份為 内耳’或耳内部。在其他實施例中,耳的標的部份為中耳 及内耳一者。在某些實施例中,此控制釋放配方更包含一 用於傳送皮質類m醇至標的耳結構之快速或立即釋放成 份。所有配方包含耳可接受的賦形劑。 本發明亦揭露藉由含有皮質類固醇的控制釋放配方之投 藥以治療耳部失調的方法與組成物。在某些實施例中,耳 部失調為梅尼爾氏症、梅尼爾氏症候群或感音神經性聽力 喪失。在另外的實施例中,耳部失調為一自體免疫内耳失 調(AIED) 〇本發明亦揭露控制釋放類固醇組成物及配方的 局部傳送以壓抑或改善因AIED的聽覺與前庭的受損,該 受扣可能由其他免疫狀況激起,包括僵直性脊椎炎、全身 性紅斑狼瘡(SLE)系、统、休格連氏⑼咖叫症候群、柯剛 氏(c〇gan,s)症、潰瘍性大腸炎、華格納氏肉芽病(wegener s 140447.doc 200950787 granulomatosis)、風濕性關節炎、發炎腸道疾病及皮硬化症 及貝赛特氏病disease ’亦已知為Bechet's disease-adamantiades)。在其他實施例中,此耳部失調為 中耳。在另外的實施例中’耳部失調為前庭的神經元炎、 姿勢性暈眩、雷氏症候群(Ramsay Hum's Syndrome)(帶狀 疱疹感染)、梅毒感染、藥物引起之内耳傷害、聽覺神經 腫瘤、老年失聰、耳硬化症或顳頷關節疾病。 本發明揭露用於治療耳部失調的控制釋放組成物,其包 含一治療有效量之一皮質類固醇、一控制釋放耳可接受賦 形劑及一耳可接受載劑。在一態樣中,此控制釋放耳可接 受賦形劑為選自耳可接受聚合物、耳可接受黏性增進劑、 耳可接受㈣、耳可接受歧膠、耳可接受熱可逆凝膠或 其等之組合。 在某些實施例中,此組成物之pH及實際滲透壓及/或容 積滲透濃度的調配為以確保維持標的耳結構的體内恒定。 例如,周邊淋巴的容積滲透濃度為介於約27〇_3〇()111〇3〇1几 間’且本發明揭露的組成物為可視情況的調製以提供實際 容積渗透濃度為約15〇至約U)00 m〇sm/卜在特定的實施 例中,本發明揭露的配方提供在作用的標的位置(如内 耳及/或周邊淋巴及/或内淋巴)之實際容積滲透濃度為在約 150至約500 m〇sm/L内。在特^的實施例中,本發明揭露 的配方提供在作用的標的位置(如,内耳及/或周邊淋巴及/ 或内淋巴)之實際容積滲透濃度為在約2⑽至約彻心複 内。在特^的實施例中,本發明揭露的配方提供在作用的 140447.doc 200950787 標的位置(如,内耳及/或周邊淋巴及/或内淋巴)之實際容 積滲透濃度為在約250至約350 mOsm/L内。相似地,周邊 淋巴的pH為約7.2-7.4,而本發明配方之pH調配(如,使用 緩衝劑)為在約5.5至約9.0間。在特定的實施例中,此配方 的pH為在约6.0至約7.6間。在特定的實施例中,内淋巴的 的pH為約7.2-7.9,而本發明配方之pH調配(如,使用緩衝 劑)為在約5.5至約9 · 0間,或在約6.5至約8 · 〇間。200950787 VI. Description of the invention: [Technical field to which the invention pertains] A vertebrate animal has a pair of ears' whose symmetry is located on the opposite side of the head. The ear acts as an organ for measuring the sound receptor and maintaining the balance of the body position. The ear can be roughly divided into three parts: the outer ear, the middle ear (middle ear), and the inner ear (inside the ear). SUMMARY OF THE INVENTION The present invention discloses a controlled release composition, formulation, method of manufacture, treatment for at least a corticosteroid to at least one structure or region of the ear: a method of use, a kit, and a delivery device. The present invention discloses a controlled release formulation for delivering corticosteroids to the ear. In some implementations, the ear part of the ear is the middle ear or the middle of the ear. In some implementations, the ear portion of the ear is the inner ear or inside the ear. In other embodiments, the target portion of the ear is the middle ear and the inner ear. In certain embodiments, the controlled release formulation further comprises a fast or immediate release component for delivering a corticosteroid to the target ear structure. All formulations contain an acceptable carrier for the ear. The present invention also discloses methods and compositions for treating ear disorders by administration of a controlled release formulation containing a corticosteroid. In certain embodiments, the ear disorder is Meniere's disease, Meniere's syndrome, or sensorineural hearing loss. In still other embodiments, the ear disorder is an autoimmune inner ear disorder (AIED). The present invention also discloses local delivery of controlled release steroid compositions and formulations to suppress or improve hearing and vestibular damage due to AIED. The detainment may be triggered by other immune conditions, including ankylosing spondylitis, systemic lupus erythematosus (SLE), system, Hugh's (9) coffee, syndrome, c〇gan, s, ulceration Colitis, Wegner's granuloma (wegener s 140447.doc 200950787 granulomatosis), rheumatoid arthritis, inflamed bowel disease and cutaneous sclerosis and Bethel's disease disease 'also known as Bechet's disease-adamantiades). In other embodiments, the ear is dysfunctional to the middle ear. In another embodiment, 'ear disorders are vestibular neuronitis, postural dizziness, Ramsay Hum's Syndrome (herpes zoster infection), syphilis infection, drug-induced inner ear injury, auditory nerve tumor, Old age deafness, otosclerosis or ankle disease. The present invention discloses a controlled release composition for treating an otic disorder comprising a therapeutically effective amount of one of a corticosteroid, a controlled release auris-acceptable excipient, and an auris-acceptable carrier. In one aspect, the controlled release auris-acceptable excipient is selected from the group consisting of an auris-acceptable polymer, an auris-acceptable viscosity enhancer, an auris-acceptable (four), an auris-acceptable gel, an auris-acceptable thermoreversible gel Or a combination thereof. In certain embodiments, the pH of the composition and the actual osmotic pressure and/or volume of the osmotic concentration are formulated to ensure that the body structure of the target otic structure is maintained constant. For example, the volumetric osmotic concentration of the peripheral lymph is between about 27 〇 3 〇 () 111 〇 3 〇 1 ' and the composition disclosed herein is visibly modulated to provide an actual volume osmotic concentration of about 15 〇 to About U) 00 m〇sm/b In a particular embodiment, the formulations disclosed herein provide an actual volumetric osmotic concentration at the target location of action (eg, inner ear and/or peripheral lymph and/or endolymph) of about 150 Up to about 500 m〇sm/L. In a particular embodiment, the formulations disclosed herein provide an actual volumetric osmolality at the target site of action (e.g., inner ear and/or peripheral lymph and/or endolymph) of from about 2 (10) to about 100. In a particular embodiment, the presently disclosed formulation provides an actual volumetric osmolality at a location of 140447.doc 200950787 (eg, inner ear and/or peripheral lymph and/or endolymph) of from about 250 to about 350. Within mOsm/L. Similarly, the pH of the peripheral lymph is about 7.2-7.4, while the pH of the formulation of the invention (e.g., using a buffer) is between about 5.5 and about 9.0. In a particular embodiment, the pH of the formulation is between about 6.0 and about 7.6. In a particular embodiment, the pH of the endolymph is between about 7.2 and 7.9, and the pH of the formulation of the invention (e.g., using a buffer) is between about 5.5 and about 9.0, or between about 6.5 and about 8. · Daytime.

在某些態樣中,此控制釋放耳可接受賦形劑為生物可降 解及/或生物可去除(如,經由尿、糞便或其他排出路徑而 降解及/或移除)。在另一態樣中,此控制釋放組成物更包 含一耳可接受黏膜黏附劑、一耳可接受穿透增進劑或一耳 可接受生物黏附劑。 在一態樣中,此控制釋放組成物使用一藥傳送系統傳 送,其為一針及注射器、泵、微注射裝置、及原處形成海 4物質或其等之組合。在某些實施例中,控制釋放組成物 的皮質類固醇具有限、或為無系統釋放但當以系統投藥時 為系統毒性、具不良pk特性或其等之組合。在又些態樣 中,此皮質類固醇為甲基脫氫皮質固醇、貝皮質醇、去氫 皮質醇、甲基去氫皮質醇、去氧皮質酮、n去氧皮質嗣、 18-經基-11-去氡皮質酮,貝可皮質醇(beci〇methas㈣、特 安皮質醇或其等之組合。在另一態樣中,此皮質類固醇為 類固醇的磷酸i旨或S旨前藥。在另—態樣中,此皮f類固醇 為類固醇的鹽。 其包含使用本發 本發明亦揭露一治療耳部失調的方法 140447.doc 200950787 =露的組絲及配方訂_咖:每 7 、 S 、 9 、 10 ' 】】、】 少-次、每二週至少—二= —次、一週至 ::五週至少一次、每六週至少-次;或-個月至少 I;、?二個月至少—次、每三個月至少-次、每四個月 夕一:人、母五個月至少—次、每⑽月至少 個月至少-次、每八個月至少一次、每九個月至少一次、 每十個月至少一次、每十一個月至少一次或每十二個月至 》一次。在特定的實施例中’本發明揭露之控制釋放配方 ^控制釋放配方的後續刺量間於内耳提供一持續劑量的皮 質類固醇。亦即,如僅例示說明之用範例,若皮質類固醇 控制釋放配方的新劑量每10天經由耳内注射至圓窗膜,則 此控制釋放配方在此10天期間提供一有效劑量的皮質類固 醇至内耳(如,遍及圓窗膜)。 在另一態樣中,投藥此組成物以使此組成物與圓窗膜接 觸。在一態樣中,此組成物經由耳内注射投藥。 本發明提供用於治療一耳疾病或症狀的醫藥組成物,其 經調製以提供一治療有效量的甲基脫氫皮質固醇、曱基去 氫皮質酵或去氫皮質醇’該組成物包含曱基脫氫皮質固 醇、甲基去氫皮質醇或去氫皮質醇之實質低降解產物,該 組成物更包含二或二以上選自下列的特性: (i)介於約0.1 wt°/〇約10 wt°/〇間的曱基脫氫皮質固醇、 曱基去氫皮質醇或去氫皮質酵、或其之醫藥可接受前藥或 鹽; 140447.doc 200950787 (11)介於約16 Wt%約21 wt%間的具通式E106 P70 E106 之聚氧乙稀-聚氧丙烯三嵌共聚合物; (in)滅菌水’適量緩衝以提供介於約5 5至約8·0間之 pH ; (iv)多顆粒化甲基脫氫皮質固醇、曱基去氫皮質醇或去 氫皮質醇; -· (V) —介於約19。(:至約42。(:間之膠化温度; _ (vi)每g配方之微生物劑少於約5 〇菌落形成單位 (cfu),及 (Vii)—個體之每公斤體重為少於約5内毒素單位。 在某些實施例中,本發明描述之醫藥組成物包含: ⑴介於約0.1〜%約10 wt%間的甲基脫氫皮質固醇、 曱基去氫皮質醇或去氫皮質醇、或其之醫藥可接受前藥或 鹽; (Π)介於約16 ”%約21 wt%間的具通式M〇6 p7〇 ei〇6 參 之I氧乙稀-聚氧丙稀三故共聚合物;及 • ㈣多顆粒化甲基脫氫皮質固醇、甲基去氫皮質醇或去 ' 氫皮質酵。 • 在某些實施例中,本發明描述之醫藥組成物包含: ω介於約〇.1心約10 wt%間的甲基脫氯皮質固醇、 曱基去氫皮質醇或去氫皮質醇、或其之醫藥可接受前藥或 鹽; ⑴)介於約16 Wt%約21 Wt%間的具通式E1〇6 P70 E106 之聚氧乙烯-聚氧丙烯三嵌共聚合物; 140447.doc 200950787 (iii) 多顆粒化曱基脫氫皮質固醇、甲基去氫皮質醇或去 氫皮質醇;及 (iv) —介於約19°C至約42°C間之膠化溫度。 在某些實施例中’本發明揭露之醫藥組成物提供介於約 250至3 20 mOsm/L間的傳送容積滲透濃度。 在某些實施例中,此曱基脫氫皮質固醇、曱基去氫皮質 酵或去氫皮質醇由配方在至少3天的期間釋放。在某些實 施例中’此曱基脫氫皮質固醇、曱基去氫皮質醇或去氫皮 質醇由配方在至少5天的期間釋放。此甲基脫氫皮質固 醇、甲基去氫皮質醇或去氫皮質醇由配方在至少1〇天的期 間釋放。 在某些實施例中’此醫藥組成物為耳-可接受熱可逆凝 膠。在某些實施例中,此聚氧乙稀-聚氧丙烯三嵌段共聚 物為可生物降解及/或生物可除的(如,此共聚合物可由身 體經由生物降解過程如,在尿、糞便或相似者中排出而除 去)。在某些實施例中,此組成物更包含黏膜黏附劑。在 某些實施例中,此配方更包含一穿透增進劑。在某些實施 例中,此配方更包含增稠劑。在某些實施例中,此配方更 包含一染料。 在又一實施例中,本發明提供之配方更包含一藥傳遞裝 置,其係選自針及注射器、泵、微注射裝置、芯、一原處 形成海綿物質或其等之組合。 在某些實施例中,本文所述之組成物為其中該甲基脫氫 皮質固醇、曱基去氫皮質醇或去氫皮質醇或其之醫藥可接 140447.doc 200950787 受鹽具有限或為無系統釋放、系統毒性、不良pK特性或 其等之組合的組成物。在本文所述之組成物的某些實施例 中,該曱基脫氫皮質固醇、曱基去氫皮質醇或去氫皮質醇 以自由鹼、自由酸、鹽、前藥、或其等之組合之形式。在 本文所述之組成物的某些實施例中,該曱基脫氫皮質固 醇、曱基去氫皮質醇或去氫皮質醇以磷酸酯或酯前藥的形 : 式投藥。在本文所述之組成物的某些實施例中,此類固醇 ^ 為曱基脫氫皮質固醇磷酸酯或曱基脫氫皮質固醇乙酸酯。 在某些實施例中,本文所述之組成物的包含曱基脫氫皮質 固醇、甲基去氫皮質醇、去氫皮質醇、或其醫藥可接受 鹽、前藥或其之組合為一立即釋放劑。 在某些實施例中,本文所述之組成物為其中該甲基脫氫 皮質固酵、甲基去氫皮質醇或去氫皮質醇包含多粒子的組 成物。在某些實施例中,本文所述之組成物為其中該甲基 脫氫皮質固醇、甲基去氫皮質醇或去氫皮質醇實質為微米 • 化粒子形式的組成物。在本文所述之組成物的某些實施例 中,該甲基脫氫皮質固醇為微-曱基脫氫皮質固醇粉末的 - 形式。 在某些實施例中,本文所述之組成物更包含一額外治療 劑。在某些實施财,此額夕卜治㈣為一Na/K ATPase調 變子、一化療劑、一膠原蛋白、一γ_球蛋白、一干擾素、 一抗微生物劑、一抗生素、一局部作用麻醉劑、一血小板 活化因子拮抗劑、一耳保護劑、一氧化氮合成酶抑制子、 抗暈眩劑、血管加壓素拮抗劑、一抗病毒劑、一抗催吐劑 140447.doc 200950787 或其等之組合。 在某些實施例中’本文所述之組成物為其中組成物之 pH為介於約6.0至約7.6間的組成物。 在本文所述之組成物的某些實施例中,具有通式E106 P70 E106之聚氧乙烯_聚氧丙烯三嵌段共聚物與一增稠劑 的比例為由約40:1至約5:1。在某些實施例中,增稠劑為羧 基甲基纖維素、經基丙基纖維素或羥基丙基甲基纖維素。 在某些實施例中’耳部疾病或症狀為梅尼爾氏症、突生 之感音神經性聽力喪失、噪音引起之聽力喪失、老化_有 關聽力喪失、自體免疫耳疾病或耳鳴。 本發明亦提供一治療耳部疾病或症狀的方法,其包含投 藥予一需要耳内組成物的個體,該組成物包含一治療有效 量之甲基脫氫皮質固醇、曱基去氫皮質醇或去氫皮質醇, 該組成物包含實質曱基脫氫皮質固醇、曱基去氫皮質醇或 去氫皮質醇之低降解產物,該組成物更包含二或二上選自 下列之特性: ⑴介於約0.1 wt%約1〇 wt〇/0間的曱基脫氫皮質固醇、 甲基去氫皮質醇或去氫皮質醇、或其之醫藥可接受前藥或 鹽; (11)介於約16 wt%約21 wt%間的具通式E106 P70 E106 之聚氧乙烯-聚氧丙烯三嵌共聚合物; (iii) 滅菌水’適量緩衝以提供介於約5.5至約8.0間之 pH ; (iv) 每g配方之微生物劑少於約50菌落形成單位(cfu),及 140447.doc 200950787 (V) —個體之每公斤體重為少於約5内毒素單位。In certain aspects, the controlled release auris-acceptable excipient is biodegradable and/or bioremovable (e.g., degraded and/or removed via urine, feces or other drainage routes). In another aspect, the controlled release composition further comprises an Ear Acceptable Mucoadhesive, an Ear Acceptable Penetration Enhancer or an Ear Acceptable Bioadhesive. In one aspect, the controlled release composition is delivered using a drug delivery system which is a needle and syringe, pump, microinjection device, and in situ combination of materials or the like. In certain embodiments, the corticosteroid that controls the release of the composition has a limit, or is systemic, but is systemic, has poor pk characteristics, or the like when administered systemically. In still other aspects, the corticosteroid is methyl dehydrocorticosterol, shell cortisol, dehydrocortisol, methyl dehydrocortisol, deoxycorticosterone, n-deoxycorticosteroid, 18-mercosyl -11-decortisol, a combination of beci〇methas (tetra), triamian cortisol or the like. In another aspect, the corticosteroid is a steroid phosphate or a prodrug. In another aspect, the skin f steroid is a salt of a steroid. The method comprising the use of the present invention also discloses a method for treating an otic disorder 140447.doc 200950787 = exposed silk and formula order _ coffee: every 7 , S , 9 , 10 ' 】,] less - times, every two weeks at least - two = - times, one week to:: at least once every five weeks, at least - every six weeks; or - at least I for months; At least once a month, every three months, at least once, every four months, one at a time: at least five times a month, at least once every (10) months, at least once every eight months, at least once every eight months, every nine At least once a month, at least once every ten months, at least once every eleven months, or once every twelve months. In a specific implementation The subsequent dose of the controlled release formulation disclosed in the present invention is controlled to provide a sustained dose of corticosteroid to the inner ear. That is, as exemplified by the illustrative example, if the new dose of the corticosteroid controlled release formulation is 10 per 10 This controlled release formulation provides an effective dose of corticosteroids to the inner ear (eg, throughout the round window membrane) during the 10 days via intra-auricular injection into the round window membrane. In another aspect, the composition is administered The composition is contacted with a round window membrane. In one aspect, the composition is administered by intra-injection. The invention provides a pharmaceutical composition for treating an ear disease or condition modulated to provide a therapeutically effective amount Methyl dehydrocorticosterol, sulfhydryl dehydrocorticolysis or dehydrocortisol' composition comprising substantially low degradation products of sulfhydryl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol, The composition further comprises two or more properties selected from the group consisting of: (i) sulfhydryl dehydrocorticosterol, decyl dehydrocortisol or between about 0.1 wt[deg.]/about 10 wt[deg.]/〇 Hydrocortisol, or a pharmaceutical acceptable prodrug or salt; 140447.doc 200950787 (11) a polyoxyethylene-polyoxypropylene tri-embedded copolymer of the formula E106 P70 E106 between about 16 Wt% and about 21 wt%; In) sterilized water 'appropriate buffer to provide a pH between about 5 5 and about 8.0; (iv) multiparticulate methyl dehydrocorticosteroid, mercapto dehydrocortisol or dehydrocortisol; · (V) - between about 19. (: to about 42. (: gelatinization temperature; _ (vi) less than about 5 colony forming units (cfu) per g of microbial formulation, and (Vii) - The individual has less than about 5 endotoxin units per kilogram of body weight. In certain embodiments, the pharmaceutical compositions described herein comprise: (1) between about 0.1% to about 10% by weight of methyl dehydrocorticosteroids. Alcohol, mercapto dehydrocortisol or dehydrocortisol, or a pharmaceutically acceptable prodrug or salt thereof; (Π) between about 16"% and about 21% by weight of the formula M〇6 p7〇ei〇 6 Refractory I oxyethylene-polyoxypropylene tri-polymer; and • (iv) multiparticulate methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrogenation. • In certain embodiments, the pharmaceutical compositions described herein comprise: methyl dechlorinated cortisol, deuterated dehydrocortisol or dehydrocortisol, having a ω between about 10% by weight of about 0.1%. Or a pharmaceutically acceptable prodrug or salt thereof; (1)) a polyoxyethylene-polyoxypropylene trimeric copolymer having a formula of E1〇6 P70 E106 of between about 16 Wt% and about 21 Wt%; 140447. Doc 200950787 (iii) Multiparticulate thiol dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol; and (iv) - a gelation temperature between about 19 ° C and about 42 ° C. In certain embodiments, the pharmaceutical compositions disclosed herein provide a delivery volume osmotic concentration of between about 250 and 3 20 mOsm/L. In certain embodiments, the thiol dehydrocorticosteroid, sulfhydryl dehydrocorticosteroid or dehydrocortisol is released from the formulation over a period of at least 3 days. In certain embodiments, the thiol dehydrocorticosteroid, decyl dehydrocortisol or dehydrocortisol is released from the formulation for at least 5 days. This methyl dehydrocorticosteroid, methyl dehydrocortisol or dehydrocortisol is released from the formulation for at least 1 day. In certain embodiments, the pharmaceutical composition is an ear-acceptable thermoreversible gel. In certain embodiments, the polyoxyethylene-polyoxypropylene triblock copolymer is biodegradable and/or bioremovable (eg, the copolymer can be biodegraded by the body, eg, in urine, Excreted in the stool or similar to remove). In certain embodiments, the composition further comprises a mucoadhesive. In certain embodiments, the formulation further comprises a penetration enhancer. In certain embodiments, the formulation further comprises a thickening agent. In certain embodiments, the formulation further comprises a dye. In still another embodiment, the present invention provides a formulation further comprising a drug delivery device selected from the group consisting of a needle and a syringe, a pump, a microinjection device, a core, a gel forming substance or a combination thereof. In certain embodiments, the compositions described herein are wherein the methyl dehydrocorticosterol, decyl dehydrocortisol or dehydrocortisol or a medicinal agent thereof is accommodating to 140447.doc 200950787 A composition that is free of systemic release, systemic toxicity, poor pK characteristics, or the like. In certain embodiments of the compositions described herein, the thiol dehydrocorticosteroid, decyl dehydrocortisol or dehydrocortisol is a free base, a free acid, a salt, a prodrug, or the like The form of the combination. In certain embodiments of the compositions described herein, the thiol dehydrocorticosteroid, decyl dehydrocortisol or dehydrocortisol is administered as a phosphate ester prodrug. In certain embodiments of the compositions described herein, such sterols are decyl dehydrocorticosteroid phosphate or sulfhydryl dehydrocorticosterol acetate. In certain embodiments, the compositions described herein comprise a thiol dehydrocorticosterol, methyl dehydrocortisol, dehydrocortisol, or a pharmaceutically acceptable salt thereof, a prodrug, or a combination thereof, Immediate release agent. In certain embodiments, the compositions described herein are compositions wherein the methyl dehydrocorticolysis, methyl dehydrocortisol or dehydrocortisol comprises multiple particles. In certain embodiments, the compositions described herein are those wherein the methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol is substantially in the form of micronized particles. In certain embodiments of the compositions described herein, the methyl dehydrocorticosterol is in the form of a micro-mercapto dehydrocorticosteroid powder. In certain embodiments, the compositions described herein further comprise an additional therapeutic agent. In some implementations, this Essence (4) is a Na/K ATPase modulator, a chemotherapeutic agent, a collagen, a gamma globulin, an interferon, an antimicrobial agent, an antibiotic, a local An anesthetic, a platelet activating factor antagonist, an ear protectant, a nitric oxide synthase inhibitor, an anti-stun agent, a vasopressin antagonist, an antiviral agent, an anti-emetic agent 140447.doc 200950787 or A combination of the same. In certain embodiments, the compositions described herein are those wherein the pH of the composition is between about 6.0 and about 7.6. In certain embodiments of the compositions described herein, the ratio of polyoxyethylene-polyoxypropylene triblock copolymer having the formula E106 P70 E106 to a thickener is from about 40:1 to about 5: 1. In certain embodiments, the thickening agent is carboxymethylcellulose, propylpropylcellulose or hydroxypropylmethylcellulose. In certain embodiments, the ear disease or condition is Meniere's disease, sudden sensorineural hearing loss, noise-induced hearing loss, aging _ related to hearing loss, autoimmune ear disease, or tinnitus. The invention also provides a method of treating an otic disease or condition comprising administering to a subject in need of an in-ear composition comprising a therapeutically effective amount of methyl dehydrocorticosterol, thiol dehydrocortisol Or dehydrocortisol, the composition comprising a low degradation product of substantially sulfhydryl dehydrocorticosterol, decyl dehydrocortisol or dehydrocortisol, the composition further comprising two or two selected from the following characteristics: (1) thiol-dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol, or a pharmaceutically acceptable prodrug or salt thereof, between about 0.1 wt% and about 1 〇 wt 〇 /0; (11) a polyoxyethylene-polyoxypropylene trimeric co-polymer having a formula of E106 P70 E106 between about 16 wt% and about 21 wt%; (iii) sterilized water 'suitable buffering to provide between about 5.5 and about 8.0 pH; (iv) less than about 50 colony forming units (cfu) per g of formula, and 140447.doc 200950787 (V) - less than about 5 endotoxin units per kilogram of body weight of the individual.

在本發明揭露之方法的某些實施例中,此甲基脫氫皮質 固醇、曱基去氫皮質醇或去氫皮質醇由組成物在至少3天 的期間釋放。在本發明揭露之方法的某些實施例中,此甲 基脫氫皮質H1醇、甲基去氫皮㈣或去氫皮㈣由組成物 在至少5天的期間釋放。在本發明揭露之方法的某些實施 例中,此甲基脫氫皮質固醇、甲基去氫皮f醇或去氯皮質 醇由組成物在至少1 0天的期間釋放。 在此方法的某些實施例中,此組成物投藥遍及内耳圓 窗。在此方法的某些實施例中,耳部疾病或症狀為梅尼爾 氏症、突生之感音神經性聽力喪失、噪音引起之聽力喪 失、老化·有關聽力喪失、自體免疫耳疾病或耳鳴。 【實施方式】 本發明提供控制釋放皮f類固醇組成物及配方以治療耳 疾病’包括梅尼爾氏症及感音神經性聽力喪失。 可取得少數治療產〇 ,、,α 康度°〇以治療耳部失調如AIED ;然而, 經口服、靜脈或肌肉注射 、仏的系統途徑為目前用於傳送 此些治療劑。系統藥之投藥 竣 仅樂在樂濃度上產生潛在的不均 寻在血清中具較而的循援旦工士加 循環里而在標的中耳及内耳器官結 構中具較低量。因此,需尊 莴要大1的樂以克服此不均等以傳 送充分、治療有效量的筚 樂至内耳。此外,系統藥之投藥増 加系統毋性及不良副作用的可能性 以執行充足的局部傳送至輕“…〜要冋血量 〜療劑的肝分解及作用 為 用而發生,其形成有毒代謝物而有致 140447.doc 200950787 消除任何由投藥治療而獲得的優勢。 a為了克服傳送的毒性及附隨副作用,本發明揭露用於局 ^傳送療劑至標的耳結構的方法與組成物。例如,進出 别庭及耳蝎器官將經由中耳,其包括内耳圓窗膜、印形窗/ 鐘月面板、環狀勃帶及通過耳軟骨囊/顳骨。 因此,本發明提供控制釋放皮質類固醇配方及組成物以 局部治療標的耳結構,藉此避免因皮質類 €> Γ:系統投藥之副作用。此局部施用的皮質類固醇 =為!標的耳結構相容,且可直接投藥至預期的標的 耳域、耳道或外耳,或投藥至直接與 區域相通的社禮,甘、' 的、,。構其包括但未限制為圓窗膜、耳 或印形窗膜。藉由特定對準耳結構,可避免來自系奸療 之不良副用。再者,臨床研判顯 , 周邊淋巴之優點,例如“療心夕」時間曝至耳蜗的 == 臨床效能。因此,藉由提供控制 Ο ,放皮質广固醇配方或組成物以治療耳部失調,提供一恒 疋可變及/或延長源之皮質類固醇予遭受 ’、一 體或患者而可減少或除去治療 調的個 露的-實施例為提供一可使至少一二:此’本發明揭 或恒定速率釋放之配…至==量以可變 某些實施例中,本發明揭露的皮質類固醇以續釋出。在 配方或組成物投藥。在其他實施例中醇^即釋放的 調變劑為以-持績性釋出配方給藥 ^醇及/或趟酶 在一脈衝方式或其之變化方式在持續、可變或 在再另一實施例中, 140447.doc 12 200950787 此皮質類固醇配方以立即釋放及控制釋放配方投藥,其係 持續、可變或脈衝或其之變化的釋放◎此釋放可選擇地依 環境或生理症狀而定,例如,外部離子性環境(參閲,例 如 Oros®釋放系統,j〇hnsori & Johnson) 〇In certain embodiments of the methods of the present invention, the methyl dehydrocorticosteroid, decyl dehydrocortisol or dehydrocortisol is released from the composition over a period of at least 3 days. In certain embodiments of the methods disclosed herein, the methyl dehydrocorticoic H1 alcohol, methyl dehydrogenated skin (tetra) or dehydrogenated skin (tetra) is released from the composition over a period of at least 5 days. In certain embodiments of the methods of the present invention, the methyl dehydrocorticosterol, methyl dehydropeno-l-alcohol or de-cortisol is released from the composition by at least 10 days. In certain embodiments of this method, the composition is administered throughout the inner ear window. In certain embodiments of the method, the ear disease or condition is Meniere's disease, sudden sensorineural hearing loss, noise-induced hearing loss, aging, hearing loss, autoimmune ear disease, or tinnitus. [Embodiment] The present invention provides a controlled release steroidal steroid composition and formulation for the treatment of otic diseases including Meniere's disease and sensorineural hearing loss. A small number of therapeutic calves can be obtained to treat otave disorders such as AIED; however, a systemic route of oral, intravenous or intramuscular injection, sputum is currently used to deliver such therapeutic agents. The administration of systemic drugs 潜在 The potential unevenness in the concentration of music is found to be lower in the serum and in the middle ear and inner ear structures in the serum. Therefore, it is necessary to respect the unequalness to overcome this inequality in order to deliver a sufficient, therapeutically effective amount of eucalyptus to the inner ear. In addition, the systemic drug administration increases the possibility of systemic and adverse side effects to perform adequate local delivery to light "...~ to sputum blood volume ~ hepatic breakdown and action of therapeutic agents, which form toxic metabolites致致140447.doc 200950787 Eliminates any advantages obtained by administration. a. In order to overcome the toxicity of delivery and accompanying side effects, the present invention discloses methods and compositions for the delivery of therapeutic agents to the ear structure. For example, in and out The court and the deafness organ will pass through the middle ear, which includes the inner ear round window membrane, the printed window/bellium panel, the annular horn band and the ear cartilage sac/tibia. Thus, the present invention provides a controlled release corticosteroid formulation and composition The topical treatment of the ear structure, thereby avoiding the side effects of the systemic administration of the corticosteroids. This topical application of corticosteroids is compatible with the target ear structure and can be directly administered to the intended ear region, ear. The tract or the outer ear, or the ritual, which is directly connected to the area, is included, but is not limited to a round window film, ear or printed window film. Given the alignment ear structure can be prevented from adverse side effects of the treatment system with rape. Further, judged clinically significant, advantage of the surrounding lymph, such as "heart treatment Xi 'time of exposure to the cochlea == clinical efficacy. Thus, by providing a controlled sputum, a corticosteroid formulation or composition for treating an otic disorder, providing a constant and/or prolonged source of corticosteroids to suffer from 'integration or patient reduction or removal of treatment The present invention provides a corticosteroid as disclosed in the present invention. Released. In the formulation or composition of the drug. In other embodiments, the alcohol-released modulator is administered in a sustained release formulation, in a pulsating manner or in a manner that varies in duration, variable or in another In an embodiment, 140447.doc 12 200950787 The corticosteroid formulation is administered in an immediate release and controlled release formulation which is sustained, variable or pulsed or a modified release thereof. ◎This release may optionally be dependent on environmental or physiological symptoms. For example, an external ionic environment (see, for example, the Oros® release system, j〇hnsori & Johnson) 〇

此外,標的耳結構的局部治療亦提供使用先前不欲使用 的治療劑,包括具有不良PK曲線、不良吸收、低系統釋 放及/或毒性問題的劑。因為皮質類固醇配方配方及組成 物之局部標的,以及在内耳中存在的生物血液屏障,可降 低因先前特有毒性或不具效益的皮質類固醇引起之不良作 用的風險。據此,本文所述之實施例範疇中亦期待使用因 為因為皮質類固醇的不良作用或不具效益而被醫師拒絕使 用的皮貝類固醇用於治療耳部失調。 枣又揭撂之 u⑺领5T斗1相容劑與本 發明揭露之皮質類固醇配方與組成物組合。當使用時,此 劑助於治療因為自體争游生a 目體免疫失调的聽力或平衡喪失或功能異 常,症狀包括暈眩、耳唣、擗六击 斗馬&力喪失、平衡失調、感染或 其等之組合。據此,改善或 =4成/鞏眩、耳鳴、聽力喪失、 平衡失調、感染、發态 ^ 發炎回應或其等之組合作用的劑亦可與 皮質類固醇組合,該劑包括 J匕栝匕括抗-TNF劑、抗催吐劑、 化療劑,包括血液化療劑、 瑩鹼.以賺®疋 瓜坐不7 (azathmfmne)或胺甲 葉酸,乂膠原蛋白、球蛋 龙曰干擾素、克帕松、中卓抽 m部麻醉劑、抗生素、血小板_活= 劑、-氧化氮合成酶抑制子及其等之組合的治療。。 此外,本發㈣述之耳可接受控制釋放皮質類固醇配方 140447.doc •13· 200950787 及治療為提供至需要的個體標的耳區域,此包括内耳,與 需要的個體為額外給予口服劑量的皮質類固醇。在某些實 施例中’此口服劑量之皮質類固醇可在耳之可接受控制釋 放皮質類固醇配方投藥前給予,故口服劑量隨提供耳之可 接受控制釋放皮質類固醇配方的時間減少。可替代地,此 口服劑量之皮質類固醇可在耳之可接受控制釋放皮質類固 醇配方投藥期間給予,故口服劑量隨提供耳之可接受控制 釋放皮質類固醇配方的時間減少。可替代地,此口服劑量 之皮質類固醇可在耳之可接受控制釋放皮質類固醇配方投 藥後給予,故口服劑量隨提供耳之可接受控制釋放皮質類 固酵配方的時間減少。 此外,此皮質類固醇醫藥組成物或配方亦包括載劑、佐 劑,如保存劑、安定劑、濕化劑或乳化劑、溶液促進劑及 調節渗透壓之鹽及/或緩衝液。此載劑、佐劑及其他賦形 劑可與標的耳結構的環境相容。因此,特別期待無毒性或 最小耳毒性的載劑、佐劑及賦形劑以允許本發明預期的耳 部失調之有效治療在標的區域或部份具有最小的副作用。 為了預防耳毒性,本文揭露的皮質類固醇醫藥組成物或配 方視情況可對準不同的標的耳結構之區域,包括但未限制 為耳穴、前庭的骨及膜迷路,耳蜗的骨及膜迷路及其他位 於内耳中的解剖或生理結構。 特定定義 在本文中使用之「耳 」 巧,關一配方 成物或成份,其包括對治療個體不具有持續有害效果 140447.doc 200950787 :及内*纟本文中使用之「耳·醫藥可接受」為指一物 f,如-制或稀釋劑,其不會取消化合物對中耳及内耳 7生物活性或性質,且為相對或減少t耳及内耳的毒性, 有、^此物質投藥至-個體並未造成不期待的效用或以一 的方式與組巾含有料㈣份交互作用。。 在本文中使用藉由一特定化合物或醫藥組成物之投藥以In addition, topical treatment of the subject's ear structure also provides the use of previously undesired therapeutic agents, including agents with poor PK profiles, poor absorption, low systemic release and/or toxicity issues. Because corticosteroid formulations and local targets of the composition, as well as the biological blood barrier present in the inner ear, reduce the risk of adverse effects caused by previously unique or unproductive corticosteroids. Accordingly, it is also contemplated in the context of the examples described herein that the use of a pebbled steroid which is rejected by a physician due to the adverse or non-effective effects of corticosteroids is used to treat ear disorders. The jujube is also disclosed. The u(7) collar 5T bucket 1 compatibilizer is combined with the corticosteroid formulation and composition disclosed in the present invention. When used, this agent helps to treat hearing loss or dysfunction due to autonomic dysfunction, including dizziness, deafness, smashing horses & loss of force, balance disorders, Infection or a combination thereof. Accordingly, an agent that improves or = 4%/distraction, tinnitus, hearing loss, balance disorder, infection, inflammatory response, or the like may also be combined with a corticosteroid, which includes Anti-TNF agent, anti-emetic agent, chemotherapeutic agent, including blood chemotherapeutic agent, acetylcholine. To earn 疋 疋 疋 7 7 7 (azathmfmne) or amine folic acid, 乂 collagen, egg yolk interferon, kepason The treatment of a combination of anesthetic, antibiotics, platelets _ live = agents, nitric oxide synthase inhibitors, and the like. . In addition, the ear can be controlled release corticosteroid formulation 140447.doc •13·200950787 and the treatment is provided to the ear region of the individual subject, which includes the inner ear, and the individual in need is additionally given an oral dose of corticosteroid . In certain embodiments, this oral dose of corticosteroid can be administered prior to administration of an acceptable controlled release corticosteroid formulation for the ear, so that the oral dose is reduced in time with which the controlled release corticosteroid formulation is provided. Alternatively, the oral dose of corticosteroid can be administered during the administration of an acceptable controlled release corticosteroid formulation for the ear, so the oral dose is reduced with the time available to provide acceptable control of the release of the corticosteroid formulation. Alternatively, this oral dose of corticosteroid can be administered after administration of an acceptable controlled release corticosteroid formulation for the ear, so the oral dose is reduced with the time available to provide an acceptable controlled release of the corticoplast formulation. Further, the corticosteroid pharmaceutical composition or formulation also includes a carrier, an adjuvant such as a preservative, a stabilizer, a moisturizing or emulsifying agent, a solution accelerating agent, and a salt and/or a buffer for adjusting the osmotic pressure. The carrier, adjuvant and other excipients are compatible with the environment of the standard ear structure. Accordingly, carriers, adjuvants, and excipients that are non-toxic or minimally toxic are specifically contemplated to allow for effective treatment of the ear disorders contemplated by the present invention with minimal side effects in the target area or portion. To prevent ototoxicity, the corticosteroid pharmaceutical compositions or formulations disclosed herein may be directed to different regions of the ear structure, including but not limited to auricular, vestibular bone and membrane labyrinth, cochlear bone and membrane labyrinth and others. An anatomical or physiological structure located in the inner ear. "Ear" is used in this article to define a formula or composition that does not have a continuing detrimental effect on a treated individual. 140447.doc 200950787 : and * * "Early acceptable for use in medicine" To refer to a substance f, such as a system or a diluent, which does not abolish the biological activity or property of the compound to the middle ear and inner ear 7, and is relatively or less toxic to the ear and inner ear, and the substance is administered to the individual. It does not cause undesired utility or interacts with the tissue containing material in four ways. . As used herein, administration of a particular compound or pharmaceutical composition is used

=或減輕特定耳部疾病的症候群、失調或症狀係意歸因 ;或伴隨化合物或組成物投藥而達到之嚴重性的降低、延 遲症狀開始、緩慢病程的發展或縮短期間,其不論永久或 暫時、持續或瞬間。 / 抗氧化劑」為耳-醫藥可接受抗氧化劑,且包括,例 如丁基化經基甲苯(贿)、抗壞血酸鈉、抗壞血酸、偏二 亞,U·酸納及維生素E。在特定實施例中,抗氧化劑促進需 ^的化學安m氧化劑亦驗反作用特定治療劑之耳 ’!·生效用’其包括與本文所述之耳劑組合使用的劑。 内耳」意指内耳,包括耳蝸及前庭迷徑,與連接的耳 蜗與中耳的内耳圓窗。 「耳生物可用性」或「内耳生物可用性」或「中耳生物 可用性」或「外耳生物可用性」為意指本發明揭露之化合 物的投藥劑!分別在研究之動物或人類的標的耳結構中成 為可用之百分比。 中耳」意指中耳’其包括鼓室、聽骨及卵形窗,其連 通中耳與内耳。 耳外部」為指外耳,包括耳翼、聽管及耳膜,該耳膜 140447.doc 15 200950787 連接外耳與中耳。 血液灰渡濃度」為指本發明提供之化合物在一患者的 血漿之血漿組份中的濃度。= or to alleviate the syndrome, disorder, or symptomatic attribution of a particular ear disease; or the reduction in severity achieved by administration of a compound or composition, the onset of delayed symptoms, the development of a slow course, or a shortened period, whether permanent or temporary , continuous or instantaneous. / Antioxidants are ear-medicinal acceptable antioxidants and include, for example, butylated toluene (bribet), sodium ascorbate, ascorbic acid, meta-dimer, U. acid and vitamin E. In a particular embodiment, the antioxidant promotes the need for a chemical ampoules to also counteract the ear of a particular therapeutic agent'! Effective for use' which includes agents for use in combination with the otic agents described herein. The inner ear means the inner ear, including the cochlea and vestibular labyrinth, and the inner ear round window of the connected cochlea and middle ear. "Auricular bioavailability" or "inner ear bioavailability" or "middle ear bioavailability" or "external ear bioavailability" means the administration of a compound disclosed by the present invention! Percentages are available in the ear structure of the animal or human being studied, respectively. The middle ear means the middle ear' which includes the tympanic cavity, the ossicular bone and the oval window which connects the middle ear and the inner ear. The outer part of the ear refers to the outer ear, including the ear flap, the ear canal and the eardrum. The eardrum 140447.doc 15 200950787 connects the outer ear to the middle ear. The blood ash crossing concentration refers to the concentration of the compound provided by the present invention in the plasma component of the plasma of a patient.

載劑物質」為職形劑,其可與皮質類固醇、標的耳結 構及耳-可接受醫藥配方之釋放曲線性質相容。此載劑物 質包括’例如結合劑、懸浮劑、崩解劑、填充劑、界面活 性劑、溶解劑、安定劑、潤滑劑、濕潤劑、稀釋劑及其相 似者。「耳-醫藥可相容載劑物質」包括但未限制為阿拉伯 膠、明膠、膠質二氧化石夕、鈣甘油磷酸鹽、乳酸鈣、麥芽 糊精、甘油、矽酸鎂、聚乙烯吡咯啶酮(Pvp)、膽固醇、 膽固醇酯、乾酪素鈉、大豆卵磷脂、牛膽酸、磷脂膽鹼、 氣化鈉、三磷酸鹽鈣、二磷酸鉀、纖維素及纖維素共軛 物、糖硬脂酸醯乳酸納、鹿角藻膠、單甘油酯、二甘油 醋、預膠化殿粉及其相似者。 「稀釋劑」一詞為指在皮質類固醇傳遞前用於稀釋之化 學化合物且其與標的耳結構相容。The carrier material is a topical agent that is compatible with the release profile properties of corticosteroids, standard ear structures, and ear-acceptable pharmaceutical formulations. Such carrier materials include, for example, binders, suspending agents, disintegrating agents, fillers, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like. "Aur-medical compatible carrier materials" include, but are not limited to, gum arabic, gelatin, colloidal silica, calcium glycerophosphate, calcium lactate, maltodextrin, glycerin, magnesium citrate, polyvinylpyrrolidine Ketone (Pvp), cholesterol, cholesterol ester, sodium caseinate, soy lecithin, taurocholic acid, phospholipid choline, sodium vapor, calcium triphosphate, potassium diphosphate, cellulose and cellulose conjugate, sugar hard Barium lactate, sodium staghorn, monoglyceride, diglycerin, pregelatinized powder and the like. The term "diluent" refers to a chemical compound that is used for dilution prior to delivery of a corticosteroid and is compatible with the standard ear structure.

「分散劑」及/或「黏性調變劑」為經由液體介質控制 皮質類固醇之擴散與均勻性的物質。擴散促進劑/分散劑 的例示包括但未限制為親水性聚合物、電解質、 Tween®60或80、PEG、聚乙烯吡咯啶酮(PVP ;商業已知 為Plasdone®)、及醣-系分散劑,例如羥基丙基纖維素(例 如HPC、HPC-SL及HPC-L)、羥基丙基曱基纖維素(例如, HPMC K100、HPMC K4M、HPMC K15M及 HPMC K100M)、 羧基甲基纖維素鈉、羧基甲基纖維素、甲基纖維素、羥基 140447.doc -16- 200950787 乙基纖維素、羥基丙基纖維素、羥基丙基曱基纖維素鄰笨 二甲酸、羥基丙基曱基纖維素乙酸硬脂酸酯(HPMCAS)、 非結晶纖維素、矽酸鋁鎂、三乙醇胺、聚乙烯醇(PVA)、 乙烯吡咯啶酮/乙烯乙酸酯共聚物(S630)、4-(1,1,3,3-四甲 基丁基)-酚聚合物及環氧乙烷與曱醛(亦已知為四丁酚 醛)、泊洛沙姆(例如,Pluronic F127、Pluronics F68®、 F88®及F108®,其為環氧乙烯與環氧丙烯的嵌段共聚 物);及 Poloxamines(例如 Tetronic 908®,亦已知為 Poloxamine 908®,其為由環氧丙烧與環氧乙烧依續加至 乙烯二胺衍生的四官功能嵌段共聚物(美國紐澤西洲帕斯 巴尼市BASF公司))、聚乙烯吡咯啶酮K12、聚乙烯吨^各咬 酮K1 7、聚乙烯吡咯啶酮K25、或聚乙烯吡咯咬酮K3 〇、 聚乙烯°比咯啶酮/乙烯醋酸鹽共聚物(S-630)、聚乙二醇(例 如具有分子量為約300至約6000,或約3350至約4〇〇〇,咬 約7000至約5400的聚乙二醇)、羧基曱基纖維素鈉、甲基 纖維素、聚山梨酵酯-80、藻酸鈉、樹膠(例如,黃箸膠及 阿拉伯膠)、胍膠、三仙膠(包括三仙膠樹膠)、糖、纖維素 (例如鈉羧基甲基纖維素、曱基纖維素、鈉羧基甲基纖維 素)、聚山梨醇酯-80、藻酸鈉、聚乙氧化山梨醇酐單月桂 酸醋、聚乙氧化山梨醇酐單月桂酸醋、普維酮、卡波姆 (carbons)、聚乙烯醇(PVA)、藻酸醋、聚葡萄胺糖及龙等 之組合。增塑劑如纖維素或三乙基纖維素亦可用於㈣八 散劑。可選擇之用於本文所述皮f類固醇的脂質 = 與自體乳化分散液之分散劑為二肉該基_膽驗碟= 140447.doc 200950787 鹼(C8-Cl8)、磷脂乙醇胺(c8_cl8)、磷脂甘油(c8_ci8)、來 自蛋或大丑的天然磷脂膽鹼、來自蛋或大豆的天然磷脂甘 油、膽固醇及異丙基肉豆蔻酯。。 「藥吸收作用」4「吸收作用」為指皮質類固醇由投藥 局部位置移動的過程,例示說明為由内耳圓窗膜及通過屏 障(圓窗膜,如後文描述)至内耳或内耳結構。在本文中使 用之「共-投藥」一詞或其相似者意指涵蓋皮質類固醇投 藥予一患者,且欲包括皮質類固醇以相同或不同的投藥路 徑或在相同或不同時間的治療體制。 在本文中使用之「有效量」或r治療有效量」一詞為指 一足量皮質類固醇的投藥,其預期減緩一或一以上被治療 疾病或症狀之症狀群。例如,本發明揭露之皮質類固醇的 投藥結果為降低及/或緩和AIED的症狀、症候群或病源。 例如,一用於治療用的「有效量」為皮質類固醇的量其 需要包括本文揭露之配方提供降低或改善疾病症候群而無 不當之不良副作用。「治療有效量」一詞包括,例如預防 有效量。一本文所述之皮質類固醇組成物的「有效量」為 可有效達到預期之醫藥效果或治療改進且無不當之不良副 作用。在某些實施例中,應瞭解「一有效量」或「一治療 有效量」由個體彼此間不同,此歸因於投藥之化合物的代 謝、年紀、重量、個體之一般症狀、被治療的症狀、被治 療症狀的嚴重性、醫師的判斷。亦應瞭解基於藥物動力學 及藥效學考量,在一延長-釋放劑量形式中的「一有效 量」可不同於在即刻-釋放劑量形式中的「一有效量。 140447.doc 200950787 「增進(enhance)」或「增進(enhancing)」一詞意指皮質 類固醇之預期效用的潛能或作用時間的增加或延長,或任 何不良症狀的降低如因治療劑的投藥的結果而造成之局部 疼痛。例如,有關增進本文揭露之皮質類固醇的效用, 「增進」一詞意指增加或延長的本文揭露之皮質類固醇合 併使用的其他治療劑之效用能力,此不論是在潛能或作用 " 時間上。在本文中使用之「增進-有效量」意指皮質類固 _ 醇或其他治療劑的置,其適於在一預期系統中增進其他治 療劑或皮質類固醇的效用。當用於一患者時,此使用的^ 效量為依的嚴重性及病程、失調或症狀、先前的治療、患 者的健康狀態及對藥的回應,及治療醫師的判斷而定。 「抑制」一詞包括防止、減慢或逆轉症狀的發展,例如 AIED ’或需要治療之患者的症狀進展。 「套件」及「製造物件」等詞為以同義字使用。 「藥效學」意指在標的耳結構内於預期位置測定觀察到 參之相對藥濃度的生物回應之因子。 「藥物動力學」意指在標的耳結構内於預期位置測定藥 之適當濃度的達到及維持之因子。 在預防應用中,含有本發明之皮質類固醇的組成物投藥 予易於遭受或者在一特定疾病、失調或症狀風險的患者, 例如梅尼爾氏症,或遭受AIED相關疾病的患者,包括僅 做為範例的有僵直性脊椎炎、全身性紅斑狼瘡(SLE)、休 格連氏(Sjdgren’s)症候群、柯剛氏⑴叫扣㈨症、潰瘍性大 腸炎、華格納氏肉芽病(Wegener's granulomatosis)、發炎腸 140447.doc -19· 200950787 道疾病' 風濕性關節炎、皮硬化症及貝赛特氏病(Beh9et,! 仏心此量定義為—「預防纽量或劑量」。在此使用 中精準的夏亦依患者的健康狀態、重量、及其相似者而 定。 鈿藥」為私在活體内轉化為母藥的皮質類固醇。在 特定實施财,—前藥藉由至少—步驟或過程酶化代謝至 化合物的生物、醫藥或治療活性形式。為了製造一前驅 藥改冑W藥活性化合物以使活性化合物將依在活體内 投藥而再產生。在一實施例中,前驅藥的設計為改變代謝 安定性或藥的傳送特性、遮蔽副作用或毒性、或改變藥的 其他特性或性質。在某些實施例中,本發明提供之化合物 衍生成合宜之前藥。 「圓窗膜」在人類中為覆蓋耳蝸窗(亦已知圓形窗、圓 窗(fenestrae rotunda)、或圓窗)。在人類中,圓窗膜的厚度 為約70微米。 「>谷解劑」為指耳·可接受化合物如三乙醯甘油、三乙 基檸檬酸酯、乙基油酸酯、乙基辛酸酯、月桂基硫酸鈉、 辛丁醋磺酸鈉、維化命E TPGS、二曱基乙醯胺、N_甲基 比洛咬酮、N·經基乙基η比u各咬酮、聚乙浠„比略咬酮、經基 丙基甲基纖維素、羥基丙基環糊精、乙醇、η· 丁醇、異丙 醇、膽固醇、膽鹽、聚乙二醇200-600、四氫呋。南乙二醇 醚、Transcutol®、丙二醇及二甲基異山梨醇酯及其相似 者。 「安定劑」為指可與標的耳結構環境相容之化合物如任 140447.doc •20- 200950787 何才几化齋f、括,^ 、 液、酸、保存劑及其相似者。安定劑包 括但未限制為可為下列任-之劑:⑴改良賦形劑與容器 〆傳遞系統之相容性,包括注射器或一玻璃瓶,⑺改 良組成物之組份的安定性,或(3)改良配方安定性。 在本文中使用之「穩定態」係當投至標的耳結構的藥量 等於在、、藥的間隔期間消去的藥量以導致在標的結構内 為一平線或恒定的藥量曝露。 Ο Φ 在本文中使用之「個體」一詞為用以指一動物,較佳為 爵札動物’包括人類或非·人類。患者及個體等詞可交 換使用。 厂 界面活性劑」為指耳_可接受化合物,如月桂基硫酸 納、多庫脂納(sodium docusate)、Tween 6〇或 8〇 ' 三乙醯 甘油、維化命E TPGS、麟脂質、印磷脂、鱗脂膽驗(A叫、 1脂乙醇胺(C8-C! 8)、磷脂甘油(c8_cl 8)、山梨醇肝單油酸 酯、聚氧乙烯山梨醇軒單油酸醋、聚山梨醇醋、ρ〇ι_奶、 膽鹽、甘油基單硬脂酸醋、環氧乙炫與環氧丙燒共聚物, 例如P1_ni,(BASF),及其相似者。部份其他界面活性劑 包括聚氧乙稀脂肪酸甘油及蔬菜油,例如聚氧乙烯(6〇)氫 化莲麻油’·及聚氧乙烯烷基醚及烷基苯基醚,例如辛基紛 聚醚10、辛基紛聚謎40。在某些實施例中,包括界面二性 劑以增進物理安定性或用於其他目的。 在本文中使用之「治療(treat)」、「治療(treating)」或 「治療(treatment)」等詞包括減輕、減弱或改善一疾病或 症狀、症候群、防止額外症候群’改善或預防症候群、抑 140447.doc -21 · 200950787 制疾病或症狀之潛在的代謝原因例如制止疾病或症狀的進 展、緩釋疾病或症狀、疾病或症狀退化、緩和由疾病或症 狀造成的症狀、或預防及/或治療的控制或停止疾病或症 狀之症候群。 耳解剖學 如下圖顯示,外耳為器官的外部份且由耳翼(耳廓)、耳 道(外耳道)及鼓膜面向外的部份,亦已知為鼓膜。此取 翼’為外耳的多肉部份且可見於頭二侧,收集聲波並將其 導向耳道。因此,外耳功能部份為收集及導向聲波至鼓膜 及中耳。 、 中耳為在鼓膜之後的空氣充填之腔室,稱之為鼓室。鼓 膜,亦已知為鼓膜,為一由由耳分隔外耳的薄膜。中耳位 於顳骨内,且包括位於此三耳骨(聽骨)的空間内:鎚骨、 石占骨及鏺骨。聽骨經由微小動帶接合在—起,其形成橫越 豉至空間的橋接《鎚骨,其一端接至鼓膜,在其前端連接 至砧骨,其依序接至鐙骨。鏺骨接合至卵形窗,二窗之一 位於鼓室内。一纖維組織層,已知為環狀韌帶接接鐙骨至 卵形窗。由外耳的聲波先造成鼓膜振動。此振動傳送經耳 蝸經過聽骨及卵形窗,其轉換在内耳中的液體。因此,配 置的聽骨提供一鼓膜與在液體充填内耳之卵形窗間的機械 連接,其中聲音轉形且傳導至内耳以進一步處理。聽骨、 鼓膜或卵形窗的硬化、僵化或移動喪失將導致聽力喪失, 例如耳硬化症、或鐘骨硬化。 鼓室亦經由耳咽管連接至喉部。耳咽管提供介於外部空 140447.doc •22- 。内耳圓窗為内耳的—"Dispersant" and / or "viscous modifier" are substances that control the diffusion and homogeneity of corticosteroids via a liquid medium. Examples of diffusion promoters/dispersants include, but are not limited to, hydrophilic polymers, electrolytes, Tween® 60 or 80, PEG, polyvinylpyrrolidone (PVP; commercially known as Plasdone®), and sugar-based dispersants. For example, hydroxypropyl cellulose (such as HPC, HPC-SL and HPC-L), hydroxypropyl decyl cellulose (for example, HPMC K100, HPMC K4M, HPMC K15M and HPMC K100M), sodium carboxymethyl cellulose, Carboxymethylcellulose, methylcellulose, hydroxyl 140447.doc -16- 200950787 ethylcellulose, hydroxypropylcellulose, hydroxypropyl decyl cellulose o-dicarboxylic acid, hydroxypropyl fluorenyl cellulose acetate Stearate (HPMCAS), amorphous cellulose, aluminum magnesium niobate, triethanolamine, polyvinyl alcohol (PVA), vinylpyrrolidone/ethylene acetate copolymer (S630), 4-(1,1, 3,3-Tetrabutylbutyl)-phenol polymers and ethylene oxide and furfural (also known as tetrabutyl phenolic), poloxamers (eg, Pluronic F127, Pluronics F68®, F88®, and F108) ®, which is a block copolymer of ethylene oxide and propylene oxide; and Poloxamines (eg Tetronic 908®) Also known as Poloxamine 908®, which is a tetrakid functional block copolymer derived from ethylene propylene oxide and ethylene bromide, which is derived from ethylene diamine (BASF, Pasbany, New Jersey, USA) , polyvinylpyrrolidone K12, polyethylene tons of each biting ketone K1 7, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K3 〇, polyethylene ° pyrrolidone / ethylene acetate copolymer (S- 630), polyethylene glycol (for example, polyethylene glycol having a molecular weight of from about 300 to about 6000, or from about 3,350 to about 4, biting from about 7,000 to about 5,400), sodium carboxymethyl cellulose, methyl Cellulose, Polysorbate-80, sodium alginate, gum (for example, tragacanth and gum arabic), silicone, tri-sac (including Sanxian gum), sugar, cellulose (eg sodium carboxymethyl) Cellulose, mercapto cellulose, sodium carboxymethyl cellulose), polysorbate-80, sodium alginate, polyethoxylated sorbitan monolaurate, polyethoxylated sorbitan monolaurate, pu A combination of ketones, carbons, polyvinyl alcohol (PVA), alginic acid vinegar, polyglucosamine, and dragons. Plasticizers such as cellulose or triethyl cellulose can also be used in (iv) eight powders. Lipids that can be selected for use in the skin steroids described herein = dispersants with autoemulsion dispersions are bismuth. The base is succinct = 140447.doc 200950787 base (C8-Cl8), phospholipid ethanolamine (c8_cl8), Phospholipid glycerol (c8_ci8), natural phospholipid choline from egg or ugly, natural phospholipid glycerol from egg or soybean, cholesterol and isopropyl myristyl ester. . "Pharmaceutical absorption" 4 "Absorption" refers to the process by which corticosteroids are moved from the local position of the administration, exemplified by the inner ear round window membrane and through the barrier (round window membrane, as described later) to the inner or inner ear structure. The term "co-administration" or its like as used herein is meant to encompass the administration of a corticosteroid to a patient and to include a treatment regimen of corticosteroids with the same or different routes of administration or at the same or different times. The term "effective amount" or "therapeutically effective amount" as used herein refers to a dose of a corticosteroid that is expected to slow down one or more symptomatic groups of the disease or condition being treated. For example, the administration of corticosteroids disclosed herein results in a reduction and/or alleviation of symptoms, syndromes or pathogens of AIED. For example, an "effective amount" for therapeutic use is the amount of corticosteroids which need to include the formulations disclosed herein to provide a reduction or amelioration of disease syndrome without undue adverse side effects. The term "therapeutically effective amount" includes, for example, a prophylactically effective amount. An "effective amount" of a corticosteroid composition as described herein is effective to achieve the desired pharmaceutical effect or therapeutic improvement without undue adverse side effects. In certain embodiments, it is to be understood that "an effective amount" or "a therapeutically effective amount" is different from one another by the metabolism of the compound administered, the age, the weight, the general symptoms of the individual, the symptoms being treated. The severity of the symptoms being treated and the judgment of the physician. It should also be understood that based on pharmacokinetic and pharmacodynamic considerations, an "effective amount" in an extended-release dosage form may differ from an "effective amount" in an immediate-release dosage form. 140447.doc 200950787 "Promotion ( The term "enhancement" or "enhancing" means the increase or prolongation of the potential or duration of action of a corticosteroid, or the reduction of any adverse symptoms, such as local pain resulting from the administration of a therapeutic agent. For example, with regard to enhancing the utility of the corticosteroids disclosed herein, the term "promoting" means increasing or prolonging the utility of other therapeutic agents used in conjunction with the corticosteroids disclosed herein, whether in terms of potential or function. As used herein, "promoting-effective amount" means a setting of a corticosteroid or other therapeutic agent that is suitable for enhancing the utility of other therapeutic agents or corticosteroids in a contemplated system. When used in a patient, the amount of efficacy used is dependent on the severity and duration of the disease, disorders or symptoms, prior treatment, the patient's state of health and response to the drug, and the judgment of the treating physician. The term "inhibition" includes preventing, slowing or reversing the development of symptoms, such as AIED' or the progression of symptoms in a patient in need of treatment. The terms "kit" and "manufactured item" are used synonymously. "Pharmacodynamic" means a factor that determines the biological response of a relative drug concentration observed at a desired location within the target ear structure. "Pharmacokinetics" means the factor at which the appropriate concentration of the drug is determined and maintained at the desired location within the target ear structure. In prophylactic applications, a composition comprising a corticosteroid of the invention is administered to a patient susceptible to or at risk of a particular disease, disorder or symptom, such as Meniere's disease, or a patient suffering from an AIDD-related disease, including Examples of ankylosing spondylitis, systemic lupus erythematosus (SLE), Sjdgren's syndrome, Ke Gangshi (1) called deduction (nine), ulcerative colitis, Wegener's granulomatosis, inflammation Intestine 140447.doc -19· 200950787 Diseases 'Rheumatoid arthritis, cutaneous sclerosis and Bethel's disease (Beh9et,! This amount is defined as “preventive amount or dose”. Accurate summer in this use It is also dependent on the patient's state of health, weight, and the like. "Powder" is a corticosteroid that is converted into a parent drug in vivo. In a specific implementation, the prodrug is enzymatically metabolized by at least a step or process. To a biological, pharmaceutical or therapeutically active form of the compound. In order to produce a prodrug, the active compound is reconstituted so that the active compound will be reconstituted in vivo. In the examples, the prodrug is designed to alter metabolic stability or drug delivery characteristics, mask side effects or toxicity, or alter other properties or properties of the drug. In certain embodiments, the compounds provided herein are derivatized into suitable prodrugs. "Circular window film" covers the cochlear window in humans (also known as round window, round window (fenestrae rotunda), or round window). In humans, the thickness of the round window film is about 70 microns. "> "Glutamine" refers to the ear · acceptable compounds such as triethylene glycol glycerol, triethyl citrate, ethyl oleate, ethyl octanoate, sodium lauryl sulfate, sodium octyl sulfonate, vitamin Life E TPGS, dimercaptoacetamide, N-methyl pirone, N. thioethyl η ratio, ketone, polyethyl hydrazine, acetophenone, propyl methacrylate , hydroxypropyl cyclodextrin, ethanol, η·butanol, isopropanol, cholesterol, bile salts, polyethylene glycol 200-600, tetrahydrofuran, southern glycol ether, Transcutol®, propylene glycol and dimethyl Isosorbide and its analogs. "Stabilizer" means a compound that is compatible with the standard structure of the ear structure. 140447.doc •20- 200950787 Hecaijihuazhai, including, liquid, acid, preservatives and the like. Stabilizers include, but are not limited to, the following agents: (1) Modified excipients and containers The compatibility of the sputum delivery system, including syringes or a glass bottle, (7) improving the stability of the components of the composition, or (3) improving the stability of the formulation. The "steady state" used herein is when the target is applied to the ear. The dose of the structure is equal to the amount of drug removed during the interval between the drug and the drug to cause a flat or constant dose exposure within the target structure. Φ Φ The term "individual" as used herein is used to refer to an animal. Preferably, the singular animal 'includes human or non-human. Words such as patients and individuals can be exchanged. "Factory surfactant" refers to the ear _ acceptable compounds, such as sodium lauryl sulfate, sodium docusate, Tween 6 〇 or 8 〇 'triacetyl glycerol, vitamin E TPGS, Lin lipid, printed Phospholipids, serotonin test (A, 1 lipoethanolamine (C8-C! 8), phospholipid glycerol (c8_cl 8), sorbitol liver monooleate, polyoxyethylene sorbitol oleic acid oleic acid, polysorbate Vinegar, ρ〇ι_milk, bile salts, glyceryl monostearate, epoxy epoxies and propylene propylene copolymers, such as P1_ni, (BASF), and the like. Some other surfactants include Polyoxyethylene fatty acid glycerin and vegetable oil, such as polyoxyethylene (6 〇) hydrogenated lotus oil '· and polyoxyethylene alkyl ether and alkyl phenyl ether, such as octyl polyether 10, octyl convergence 40. In certain embodiments, an intersex agent is included to enhance physical stability or for other purposes. "treat," "treating," or "treatment" as used herein. Words include reducing, attenuating or ameliorating a disease or symptom, syndrome, preventing additional syndromes' improvement or prevention Symptoms, depression 140447.doc -21 · 200950787 Potential metabolic causes of diseases or symptoms such as stopping the progression of a disease or condition, slow-release disease or symptoms, degenerative disease or symptoms, alleviating symptoms caused by disease or symptoms, or preventing and / or the syndrome that controls or stops the disease or symptoms of treatment. The anatomy of the ear shows that the outer ear is the external part of the organ and is covered by the ear (auricle), the ear canal (outer ear canal) and the tympanic membrane. Known as the tympanic membrane. This wing 'is the fleshy part of the outer ear and can be seen on the first two sides, collecting sound waves and guiding it to the ear canal. Therefore, the outer ear function part collects and directs the sound waves to the tympanic membrane and the middle ear. The ear is a chamber filled with air after the tympanic membrane, called the tympanic cavity. The tympanic membrane, also known as the tympanic membrane, is a membrane that separates the outer ear by the ear. The middle ear is located in the tibia and includes the three ear bones. In the space of the bone: the hammer bone, the stone occupying the bone and the tibia. The ossicle is joined by a tiny moving belt, which forms a bridge that crosses the raft to the space. The hammer bone is connected to the tympanic membrane at one end. Connected to the anvil, which is sequentially connected to the tibia. The tibia is joined to the oval window, one of the two windows is located in the tympanic cavity. A fibrous tissue layer, known as an annular ligament, connects the tibia to the oval window. The sound waves of the outer ear first cause the tympanic membrane to vibrate. This vibration is transmitted through the cochlea through the ossicular and ovate windows, which converts the liquid in the inner ear. Therefore, the configured ossicle provides a tympanic membrane and a machine between the liquid-filled inner ear and the oval window. a connection in which the sound is transformed and transmitted to the inner ear for further processing. The loss of hardening, rigidity or movement of the ossicle, tympanic membrane or oval window will result in hearing loss, such as otosclerosis, or hardening of the talus. The tympanic cavity is also connected via the eustachian tube The larynx. The Eustachian tube is provided between the outer space 140447.doc • 22- . The inner ear round window is the inner ear -

❹ 體内耳圓_的移動允許在耳堝中的液體移動,其依序 導致耳堝内毛細胞的移動,允許聽力訊號轉換。内耳圓 _的僵化或移動導致聽力喪失,因為耳蝸液體缺少移動 的能力。目前的研討會已聚焦於植入機械轉換器於内耳 200950787 氣及中耳腔穴間相等之壓力之能力 部伤但亦可在鼓室内接近,該内耳圓窗開口至内耳之耳 蝸。内耳圓窗由一膜覆蓋,其由三層組成:一外或黏膜 層、-令間或纖維層及一内膜,其直接與耳蝸液體相通。 因此,内耳圓窗經由内膜直接與内耳相通。 卵形及内耳圓窗内的移動為為互相連接,亦即當鐙骨 由鼓膜傳送移動至卵形窗以向内移動抵住内耳液體,内 耳圓®(更正確為圓窗膜)對應向外推出並遠離耳蝸液 圓窗上,其繞道正常傳導路徑通過印形窗並提供放大輸 入至耳堝室。 聽覺訊號傳導發生於内耳。液體_充填内耳(内耳)由二主 要組份組成:耳蝸及前庭器官。内耳内部位於骨或骨迷徑 内,其為一位於顱骨之顳骨内之複雜交錯系列。前庭器官 為平衡器官且由三半規管及前庭組成。此三半規管彼此配 置,故可藉由液體的移動偵測頭在沿三正交平面移動及後 續由半規管的感覺器的訊號處理,其稱為聽脊。聽脊含有 毛細胞與支持細胞,且由一稱為頂帽的半穹形明膠物質覆 蓋。此毛細胞的毛包埋於頂帽。此半規管偵測動態平衡、 旋轉或角度移動平衡。 當頭快速轉動,半規管與頭移動,但位於膜狀半規管人 140447.doc -23- 200950787 的内淋巴液體仍保持穩定。内淋巴液體推抵頂帽'其傾向 -側。當頂帽傾斜’其f折在聽脊之毛細胞上的毛,其起 動一感覺脈衝。因為每一半規為位於不同半面,每一半規 管的對應聽脊不同的回應至頭的相同移動。此產生一的脈 衝的鑲嵌’其傳送至在前庭耳蝎神經之前庭支上的中央神 ’”里系統中央神經系統言全釋此些訊息並啟動適當的回以維 持平衡。中央神經系統的重要性者為小腦,其媒介平衡的 感覺及平衡。 · 前庭為内耳的中央部份且含有帶有毛細胞的機械受體,〇 其確定靜態平衡’或頭相對重力的位置。靜態平衡於當頭 /不移動或在-直線移動時扮演一角色。此在前庭内的膜迷 徑區分為二似囊-結構,橢圓囊及球囊。每一結構依序含 有一稱為黃斑的小結構,其回應靜態平衡的維持。此黃斑 由包埋在膠狀物質(相似於頂帽)之感覺毛細胞組成,該膠 狀物物質覆蓋黃斑。碳酸鈣粒子,稱為聽骨,包埋狀 層的表面。 、 當頭在正上位置時,此毛細胞沿黃斑伸直。當頭歪斜 0 時’此膠狀物質及聽骨對應的歪斜’在黃斑上的毛細胞冑 · 份毛彎折。此變折作用啟動-訊號脈衝至中央神經系统, 其前庭耳堝神經的前庭支行進,其接著轉達機動脈衝至— - 適當肌肉以維持平衡。 I40447.doc •24· 200950787移动 The movement of the inner ear circle _ allows the movement of liquid in the deafness, which in turn causes the movement of hair cells in the deafness, allowing hearing signal conversion. The rigidity or movement of the inner ear circle _ causes hearing loss because the cochlear fluid lacks the ability to move. The current seminar has focused on the ability to implant mechanical transducers in the inner ear 200950787 between the gas and the middle ear cavity. The injury can also be accessed in the tympanic cavity, which opens to the inner ear. The inner ear round window is covered by a membrane consisting of three layers: an outer or mucosal layer, an intergranular or fibrous layer, and an inner membrane that is in direct fluid communication with the cochlea. Therefore, the inner ear round window communicates directly with the inner ear via the inner membrane. The movement of the oval and the inner ear round window is interconnected, that is, when the tibia is transported by the tympanic membrane to the oval window to move inward against the inner ear fluid, the inner ear circle® (more correctly the round window membrane) corresponds to the outward It is pushed out and away from the round window of the cochlear fluid, which bypasses the normal conduction path through the print window and provides an amplified input to the deafness chamber. Auditory signal conduction occurs in the inner ear. The liquid _ filling inner ear (inner ear) consists of two main components: the cochlea and the vestibular organ. The inner ear is located inside the bone or bone labyrinth, which is a complex interlaced series located within the tibia of the skull. The vestibular organ is a balanced organ and consists of a three-north tube and a vestibule. The three semicircular canals are configured to each other so that they can be moved by the motion detecting head of the liquid in three orthogonal planes and subsequently by the sensor of the semicircular canal, which is called the auditory ridge. The auditory ridge contains hair cells and supporting cells and is covered by a semi-穹 gelatin substance called a top cap. The hair cells of this hair cell are buried in the top cap. This semi-regulator detects dynamic balance, rotation or angular movement balance. When the head rotates rapidly, the semicircular canal moves with the head, but the endolymphatic fluid located in the membranous semicircular canal is still stable. The endolymphatic fluid pushes against the top cap's tendency - side. When the top cap is tilted, its f is folded over the hair on the hair cells of the ridge, which initiates a sensory pulse. Because each semi-regular is located on a different half, the corresponding audible ridge of each semi-regulator has a different response to the same movement of the head. This produces a pulsed mosaic 'which is transmitted to the central god on the vestibular branch of the vestibular sacral nerve'. The central nervous system of the system fully releases this message and initiates appropriate back to maintain balance. The central nervous system is important The sex is the cerebellum, the sense of balance and balance of the media. · The vestibule is the central part of the inner ear and contains mechanical receptors with hair cells, which determine the static equilibrium 'or the position of the head relative to gravity. Static balance in the head / Does not move or play a role in the - linear movement. This membrane in the vestibule is divided into two capsule-like structures, elliptical sacs and balloons. Each structure sequentially contains a small structure called the macula, which responds Maintenance of static balance. This macula consists of sensory hair cells embedded in a gelatinous substance (similar to the top cap) that covers the macula. Calcium carbonate particles, called the ossicles, are the surface of the embedding layer. When the head is in the up position, the hair cells are straightened along the macula. When the head is skewed 0, the gelatinous substance and the sacral corresponding to the ossicle are bent on the macular hairs of the macula. Move - pulse signals to the central nervous system, its vestibular nerve vestibular cochleafollowing into sub-branches, which are then conveyed to a motor pulse - - appropriate muscles to maintain balance I40447.doc • 24 · 200950787.

耳堝為内耳中有關聽力的部份。耳蝸為一似錐形管結 構,其螺圈為一似蜗牛形狀組件。耳蜗内部分為三區域, 其進一步由前庭膜與腦底膜位置界定。在前庭膜上的部份 為前庭階,其由卵形窗延伸至耳蝸頂並含有周邊淋巴液 體,其為一低鉀但高鈉含量的水性液體。腦底膜定義一鼓 階區域,其由耳蝸頂部延伸至内耳圓窗且亦含有周邊淋 巴。此腦底膜含有數千個硬纖維,其由内耳圓窗至耳蜗頂 部在長度逐漸延伸。當以聲音活化時,底部膜的纖維振 動。在前庭階與鼓階間為耳蝸導管,其如一密閉囊結束於 耳蜗頂部。耳蝸導管含有内淋巴液體,其相似腦脊的液體 且為鉀量高。 τ聽力感覺器官-科氏螺旋聽器位於腦底膜且向上延伸入 耳媧導管。科氏螺旋聽器含有毛細胞,其具有似毛髮突出 物由自由表面延伸,且與一凝膠化表面接觸,其稱為覆 140447.doc -25- 200950787 膜。雖然毛細胞不具有軸索,其由形成前庭耳蝸神經(腦 神經VIII)耳蝸枝的感覺神經纖維包圍。 如討論,卵形窗亦已知為與鐙骨相通的橢圓窗以傳達由 鼓膜振動的聲音。傳送至卵形窗的振動藉由周邊淋巴及前 庭階/鼓階增加在液體充填耳堝内的壓力,其依序引起在 内耳圓1¾上的膜在回應上的擴展。此一致之卵形窗的向内Deafness is the part of the inner ear that is related to hearing. The cochlea is a conical tube structure with a spiral-like snail-like component. The interior of the cochlea is divided into three regions, which are further defined by the position of the vestibular membrane and the basement membrane. The part on the vestibular membrane is the vestibular step, which extends from the oval window to the cochlear apex and contains peripheral lymphoid fluid, which is a low potassium but high sodium content aqueous liquid. The basement membrane defines a tympanic region that extends from the top of the cochlea to the inner ear round window and also contains peripheral lymphocytes. This brain base film contains thousands of hard fibers that extend gradually from the inner ear round window to the top of the cochlea. When activated by sound, the fibers of the bottom film vibrate. Between the vestibular step and the tympanic catheter is a cochlear catheter that terminates at the top of the cochlea as a closed capsule. The cochlear duct contains endolymph fluid, which is similar to the fluid in the cerebrospinal ridge and has a high potassium content. The τ hearing sensory organ - the Coriolis spiral is located in the basement membrane and extends upward into the deafness catheter. Coriolis loudspeakers contain hair cells that have hair-like projections that extend from a free surface and are in contact with a gelled surface, which is referred to as a coating 140447.doc -25-200950787. Although the hair cells do not have an axon, they are surrounded by sensory nerve fibers that form the vestibular cochlear nerve (brain nerve VIII) cochlear branches. As discussed, the oval window is also known as an elliptical window that communicates with the tibia to convey the sound of vibration by the tympanic membrane. The vibration transmitted to the oval window increases the pressure in the liquid-filled deaf by the peripheral lymph and vestibular steps/drums, which in turn causes the membrane on the inner ear circle 126 to expand in response. Inward of this consistent oval window

壓制/内耳圓窗的向外擴張允許在耳蝎内的液體移動而未 改變内耳㈣力。《而,當振動在前庭階徑行經周邊淋 巴,其在前庭膜產生對應的共振。此些對應的共振行經耳 蜗管的内淋巴’且傳送至腦底膜。當腦底膜振M、或向上 及向下移動,科氏料聽m軸。在科㈣旋聽器的 毛細胞受體接著抵住覆腹移動,造成在覆財的機械變 形。此機械變形啟動神經脈衝,其行經前庭耳蜗神經至中 系:,機械傳送接收到聲波為訊號,此訊 中央神經系統處理。 疾病 〇 …耳、中耳及外耳失調的耳部失調產生的症狀包括 但不限制為聽力喪失、眼球震顫、暈眩 、 脹、感染及充血。此些失調可能具有許多原因,2二腫 外傷、發炎、腫瘤及對藥或其他化 有:辭力 及/或平衡受損或炎症的許多原因彳。反應。聽力 及/或細胞素媒介的炎症反應。在A於自體免疫失調 調為梅尼爾氏症。在-實_中,此2例中,此耳部失 性聽力喪失。在-實施财,此錢調為感音神經 調為自體免疫内耳 140447.doc •26· 200950787 失調(AIED)。在-實施例中,此耳部失調為梅尼爾氏症。 在又一實施例中,此耳部失調為梅尼爾氏症候群、前庭神 經兀炎、姿勢性暈眩、雷氏症候群(帶狀疱疹感染)、梅毒 感染、藥物引起之内耳傷害、聽覺神經腫瘤、過度噪音的 聽力喪失、老年失聰、耳硬化症或顳頷關節疾病。 本文所述之疾病,包括在後文中陳述者為使用本發明揭 *· 露之類固醇醫藥組成物治療。 .· 梅尼爾氏症 梅尼爾氏症為一自發性症狀,特徵在於暈眩、噁心及噁 吐的突然發作’其可維持3至24小時’且可能漸漸消失。 漸進聽力喪失、耳鳴及耳内的壓力感覺經由疾病的時間而 發生。梅尼爾氏症的病因可能與内耳液體體内恒定失衡有 關,包括内耳液體產生的增加或内耳液體分解的降低。 可使用外科手術以緩和梅尼爾氏症症狀,包括破壞前庭 功能以緩和暈眩症候群。此些手術目的在於減輕在内耳的 參液體壓力及/或破壞内耳平衡功能。一内琳巴引流手術, .Λ缓和液體壓力,係在内耳中進行以缓和前庭功能異常症 狀。亦可使用前庭神經繞道,其可控制章眩同時保留聽 .力。 一梅尼爾氏症的標準照護需要患者遵循一低鹽飲食。在 特疋例子中,低鹽飲食補充一抗生素投藥。在一特定例 子中,低鹽飲食補充一健他黴素投藥。在一特定例子中, 低鹽飲食補充一口服類固酵投藥。在-特定例子中,低鹽 飲食補充口服普賴松(25-50 mg PO/IM/PR q4-6h)投藥。 140447.doc -27- 200950787 在一組實施例中,一使用前文所述之標準照護治療梅尼 爾氏症的患者,以本文所述之控制釋放皮質類固醇的耳可 接受配方及方法替代。在一組實施例中,一使用前文所述 之標準照護治療梅尼爾氏症的患者,但其對此一治療有抗 性或不回應則以本文所述之控制釋放皮質類固醇的耳可接 受配方及方法替代。 梅尼爾氏症候群 梅尼爾氏症候群,其呈現與梅尼爾氏症的相似症狀’其 係因其他疾病病程造成的第二困擾,如因梅毒感染的甲狀 0 腺疾病或内耳發炎。因此,梅尼爾氏症候群為内淋巴正常 產生或再吸收作用受干擾之病程之第二作用,包括内分液 異常、電解質失衡'自體免疫功能障礙、藥製劑、感染 (例如寄生感染)或高脂質症。受梅尼爾氏症候群折磨患者 的治療為相似於梅尼爾氏症。 感音神經聽力喪失 感曰神經聽力喪失在當内耳組份或附隨神經組份受影燮 曰 d 且其可能包含一神經,亦即當在腦部的聽覺神經或聽覺神 罈 、左通道受影響’或感覺組份時發生。感覺聽力喪失為遺傳 ·· 性’或其可能由藉由聽覺創傷(亦即非常大聲的噪音)、一 病毒感染、藥-誘發或梅尼爾氏症而引起。神經聽力喪失 可因腦腫瘤、感染、或多種腦及神經失調,如中風引起。 有些遺傳性疾病,如Refsum氏疾病(分支脂肪酸的缺陷堆 積)’亦可造成影響聽力喪失的神經失調。聽覺神經受脫 趙勒疾病破壞,例如自發性發炎脫髓鞘疾病(包括多發性 140447.doc *28- 200950787 硬化)、橫貝性脊髓炎、Devic氏疾病、漸進進行性多處腦 白質病 '格巴二氏(Guniain-Barre)症候群、慢性發炎脫髓 鞘聚神經病變及抗MAG周邊神經病變。 瞬間失聰、或感音神經聽力喪失發生率為在每5〇〇〇人約 1人中發生,且由病毒或細菌感染引起,例如腮腺炎、麻 疹、感冒、水痘、巨細胞病毒、梅毒或傳染單核白血球增 多症或内耳器g的生理傷害。在某些病例中,未能辨識 ^ 原因。耳鳴及暈眩可能伴隨瞬間失聰,其逐漸消退。口服 皮質類固醇常用於處方以治療感音神經聽力喪失。在某些 病例中,需要手術治療。 本文所描述之配方及方法包括感音神經聽力喪失的治 療,此包括瞬間感音神經聽力喪失的治療,包括自發性瞬 間感音神經聽力喪失。對SSHL,目前治療選項包括以甲 基脫氫皮質固醇(4-10 mg/ml)或甲基_去氫皮質醇(4〇_62 5 mg/ml)的高劑量口服類固醇2週治療(4_7天療程+7_【〇天減 © 量)。如本文註明,高劑量的口服類固醇附帶有不期待的 ㈤仙及不利狀況。因此,可預期本文所描述之有關類固 • 醇之持續釋放、局部傳送至内耳的配方及方法造成比口服/ •系統類固醇使用的顯著較少副作用。在一實施例中, ISSHL特性在於發作少於72小時期間的單側感音神經性聽 力喪失,其中HL定義為在至少3接近之測試頻率>3〇犯。 一自發性瞬間感音神經性聽力喪失(ISSHL)的標準照護 為以高劑量口服類固醇治療。在特定例子中,一個體以^ 劑量口服類固醇治療約二週。在特定例子中,一個體以高 140447.doc •29- 200950787 劑量口服類固醇治療約二週,接著在約7至約1〇天逐漸減 少口服類固醇。在特定例子中,此口服類固醇為甲基脫氫 皮質固醇(4-10 mg/ml)。在特定例子中,此口服類固醇為 甲基-去氫皮質酵(40-62.5 111§/1111)。 在一組實施例中,在一組貫施例中,一使用前文所述之 標準照護治療ISSHL的患者,以本文所述之控制釋放皮質 類固醇的耳可接受配方及方法替代。在一組實施例中,一 使用A文所述之標準照護治療IS S HL的患者,但其對此一 治療有抗性或不回應則以本文所述之控制釋放皮質類固醇 的耳可接受配方及方法替代。 過度°喿音引起聽力喪失 聽力喪失可能因延長曝露至大聲噪音而引起,如大聲音 樂、重型儀器或機具、飛機 '搶聲或其他人性化噪音。此 聽力喪失的發生為内耳毛細胞受體的破壞結果。此種聽力 喪失常伴隨耳鳴。通常珍斷出對聽力喪失的永久傷害。 雖然目前無法治療噪音-誘發的聽力喪失,已實驗性發 展數種治療,包括以治療以胰島素_生長因子 療。(Lee等人之Otol. Neurotol. (2007) 28:976-981)。 老年失聰 年失聰或老化-有關聽力喪失為正常老化的一部份,且 因為在内耳中的科氏螺旋聽器(Corti器官受體細胞退化而 發生)。其他原因亦可構成在前庭耳堝神經中數種神經纖 維的減少,以及耳蝸中腦底膜喪失彈性。目前未知因為老 年失聰或過度噪音造成的永久聽力受損之治療。 140447.doc •30· 200950787 藥物引起的内耳損害 由藥的投藥造成的損害,包括特定抗生素、利尿劑(例 如埃西克林酸(ethacrynie邮⑷及弗西邁咖⑽mide))、阿 斯匹靈、似阿斯匹靈物質(例如水楊酸鹽)及奎寧。内耳器 B的退化可因損傷的腎功能而加速,損傷的腎功能導致影 響藥及其代謝物的減少清除。藥可影響聽力及平衡,但不 * 可此影響聽力至一較大範圍。 0 例如,新黴素、康黴素、丁胺卡那黴素在聽力比在平衡 上具較大的效用。抗生素紫黴素、紫菌素及泰百徽素同時 影響聽力與平衡。鏈黴素,另一常見投藥的抗生素,其引 起除暈眩外的聽力喪失,且可能造成Dandy氏症候群’其 黑暗中订走困難且在每一步引發環境移動的感覺。阿斯匹 靈,當以高劑量服用時,可能導致暫時性聽力喪失與耳 鳴,其係在無外部聲音時感知聲音的症狀。相似地,奎 f、埃西克林酸及弗西邁可導致暫時或永久聽力喪失。 φ 自體免疫内耳疾病 自體免疫内耳疾病(AIED)為感音神經聽力喪失之少數可 逆原因之一。其為二較少出現在成人與孩童二者的失調, - 其通常為内耳的聽及前庭功能雙側障礙。AIED源可能為 自體抗體及/或免疫細胞攻擊内耳結構,但伴隨其他自體 免疫症狀。在許多例子中,AIEDS生但無系統自體免疫 症狀,但高達三分之一患者亦遭受系統自體免疫疾病,如 發炎、腸道疾病、風濕性關節炎、僵直性脊椎炎、全身性 紅斑狼瘡(SLE)、休格連氏症候群、柯剛氏疾病、潰瘍性 140447.doc -31 - 200950787 大腸炎、華格納氏肉芽病(Wegener's granulomatosis)及硬 皮病。貝赛特氏疾病,多重系統疾病亦常具有聽,亦常見 有聽前庭問題。已有部份證據顯示食物有關過敏為耳蝸及 前庭自體免疫性的病因,但目前在疾病病原學上未對其重 要性的認同。已發展出一 AIED的分類表(Harris與Keithley 著,(2002)Autoimmune inner ear disease,in Otorhinolaryngology Head and Neck Surgery. 91,18-32)。 以皮質類固醇治療緩和AIED症狀。皮質類固醇普賴松 (60 mg/天計4週)的口服給藥顯示在純音及語音聽力結果上 的改良。皮質類固醇的調節作用經皮質類固醇受體或鹽皮 質素受體發生。 發炎失調 耳部的發炎失調包括但未限制為中耳炎、外耳炎、乳突 炎、大泡性鼓膜炎、耳咽管炎(Eustachian tubal catarrh)、 耳咽管炎(Eustachian salpingitis)、迷徑炎或其相似者。範 例包括症狀影響成人及孩童的中耳炎(OM),此包括急性 中耳炎(AOM),溢流中耳炎(OME)與慢性中耳炎。OM感 受性為多重因子及複合性,包括環境、微生物及寄主因 子。在遭受OM個體之排放液介質中已觀察到細胞激素產 生的增加,包括間白素與TNF。以抗炎症之類固醇的治療 緩釋耳部的炎症失調的症狀(如,中耳炎、耳咽管炎 (Eustachian tubal catarrh)或相似者)。在某些例子中,炎 症失調為細菌感染(例如OM)。在某些例子中,一抗生素 與抗炎症之皮質類固醇的組合投藥可緩和OM症狀。 140447.doc -32- 200950787 藥劑 本發明提供改善或減輕耳部失調的皮質類固醇組成物或 配方’包括梅尼爾氏症、感音性聽力喪失及/或發炎失調 與伴隨的症狀,其包括但不限制為聽力喪失、眼球震顏、 • *眩、耳鳴、炎症、腫脹、感染及充血。包括細或梅 • $爾氏症及/或炎症失調的耳部失調具有回應本文所揭露 . <藥劑或其他藥劑的病因及症狀。未於本文描述但可用於 φ 彳部失調之改#或消㈣皮質_醇係顯見的包括及欲涵 括於本發明實施例之範疇中。 再者’先前已顯示在其他器官系統H統性或局部應用 期間為毒性、有害或無效用之藥劑,例如經由肝作用後形 成的毒性代謝物、對特定器官、組織或系統為毒性的藥、 需要高量以達到效用、不能經由系統路徑或經由ρκ不良 特性釋出的藥劑亦可用於本文的某些實施例中。例如,甲 基脫氫皮質固酵的副作用包括:納滞留、過量水滯留、易 ❹感染患者的繫血性心衰竭、高血壓、肌肉無力、肌肉萎 縮、骨質疏鬆症、腱破裂、胃潰瘍、潰瘍性食道炎、皮膚 • 薄化、皮膚反應、傷口癒合能力減弱、痙攣、暈眩、頭 痛、心理失調、庫興氏症候群(Cushing,s synd_e)、兒童 延遲生長、糖尿症、多毛症、白内障、f光眼、體重增 加、增加食慾及噁心。具有限或無系統釋出、系統毒性: 不良ρκ性質或其等組合之醫藥劑係顯見期待包括於本發 明實施例之範_。 本發明㈣的皮質類固醇配方可選擇的直接對$需要治 140447.doc -33· 200950787 療的耳結構;例如,一預期的實施例為直接施用本發明揭 露的皮質類固醇配方在内耳圓窗膜上或耳蝸窩冠部,允許 直接進人內耳或内耳組份及治療。在其他實施财,本發 明揭露的皮質類固醇配方直接施用至圓形窗。在其他實施 例中,直接進入可藉由直接微注射入内耳而達到例如經 由耳堝微灌注。此實施例亦可選擇地包含一藥傳遞裝置, 其中該藥傳遞裝置經由針與注射器、泵、微注射裝置、一 原位形成的海綿狀物質或其等之任何組合傳遞皮質~類固醇 配方。在又其他實施例中,皮質類固醇配方的施用為經由 内耳膜的穿孔而對準中耳且皮質類固醇配方直接施用至受 影響的中耳結構,包括鼓室壁或聽骨而對準至中耳。藉 此,本文揭露之皮質類固醇配方可侷限於標的中耳結構, 且不會經例如經由耳咽管或穿孔的鼓膜之擴散或滲2而損 失。 皮質類固醇/抗炎症類固醇 皮質類固醇的特性在於鹽皮質素與糖皮質素效用,其係 依藥劑的藥理學而定。鹽皮質素的特性在於其與醛固嗣的 相似性及其在電解質量及水平衡間的影響。糖皮質素,如 内生糖皮質素皮質醇,其控制代謝作用且藉由預防細胞素 的釋放而有抗炎症性。許多試劑具有鹽皮質素與糖皮質素 活性的程度。數個合成的糖皮質素之相對效能及活性顯示 於下表。 ” 140447.doc • 34- 200950787 類固醇 皮質醇 普賴松 去氫皮質醇 甲基脫氫皮質固醇 貝皮質醇 特安皮質醇 普林咬(prednylidene) 醛固酮 糖皮質素效能 1 4 1.7 21 45 0.35 182 0.07 鹽皮質素效能 0.054 0,002 0.013 0.0094 0.0038 0.0002 0.0011 1.0The outward expansion of the pressing/inner ear window allows the liquid within the deaf to move without changing the inner ear (four) force. "And, when the vibration travels through the periphery of the vestibular step, it produces a corresponding resonance in the vestibular membrane. These corresponding resonances travel through the endolymph of the cochlear implant and are transmitted to the basement membrane of the brain. When the brain basement vibrates M, or moves up and down, the Coriolis is expected to listen to the m-axis. In the section (4), the hair cell receptors of the tuner then move against the abdomen, causing mechanical deformation in the fortune. This mechanical deformation initiates a nerve impulse that travels through the vestibular cochlear nerve to the middle system: the mechanical transmission receives the sound wave as a signal, which is processed by the central nervous system. Symptoms of the ear, middle ear and outer ear disorders caused by, but not limited to, hearing loss, nystagmus, dizziness, swelling, infection and congestion. These disorders may have many causes, such as 2 swollen trauma, inflammation, tumors, and medication or other factors: many reasons for loss of speech and/or balance or inflammation. reaction. Hearing and / or cytokine mediators of inflammation. In A, the autoimmune disorder is adjusted to Meniere's disease. In the two cases, the ear loss hearing loss was observed in the two cases. In the implementation of the money, the money was transferred to the sensorinegic tone to autoimmune the inner ear 140447.doc •26· 200950787 Disorder (AIED). In an embodiment, the ear disorder is Meniere's disease. In still another embodiment, the ear disorder is Meniere's syndrome, vestibular neuritis, postural dizziness, Ray's syndrome (herpes zoster infection), syphilis infection, drug-induced inner ear injury, auditory nerve tumor Excessive noise hearing loss, old age deafness, otosclerosis or ankle joint disease. The diseases described herein, including those set forth hereinafter, are treated with the steroid medical compositions of the present invention. Meniere's disease Meniere's disease is a spontaneous symptom characterized by a sudden onset of dizziness, nausea, and vomiting - which can last for 3 to 24 hours and may gradually disappear. Progressive hearing loss, tinnitus, and pressure perception in the ear occur through the time of the disease. The cause of Meniere's disease may be related to a constant imbalance in the fluid in the inner ear, including an increase in fluid production in the inner ear or a decrease in fluid decomposition in the inner ear. Surgery can be used to alleviate Meniere's symptoms, including disrupting vestibular function to alleviate dizziness syndrome. The purpose of these procedures is to reduce the fluid pressure in the inner ear and/or to disrupt the inner ear balance function. An inner linba drainage surgery, relief and fluid pressure, is performed in the inner ear to alleviate vestibular dysfunction. A vestibular nerve bypass can also be used, which controls the glare and preserves the hearing force. A standard care of Meniere's disease requires the patient to follow a low-salt diet. In a special case, a low-salt diet is supplemented with an antibiotic. In a specific example, a low salt diet is supplemented with a pentamycin administration. In a specific example, the low-salt diet is supplemented with an oral solid-state fermentation. In a specific example, a low-salt diet supplemented with oral Prysone (25-50 mg PO/IM/PR q4-6h) was administered. 140447.doc -27- 200950787 In one set of embodiments, a patient treated with standard care as described above for the treatment of Meniere's disease is replaced with an acceptable formulation and method for the release of corticosteroids as described herein. In one set of embodiments, a patient treated with Meniere's disease using standard care as described above, but which is resistant or non-responsive to this treatment, is acceptable for the release of corticosteroids as described herein. Formula and method alternatives. Meniere's syndrome Meniere's syndrome, which exhibits similar symptoms to Meniere's disease, is the second cause of illness caused by other diseases, such as thyroid-infected thyroid disease or inflammation of the inner ear. Therefore, Meniere's syndrome is the second role of the pathogenesis of normal production or resorption of endolymph, including internal dissociation abnormalities, electrolyte imbalance 'autoimmune dysfunction, drug preparations, infections (eg parasitic infections) or High lipidosis. Treatment with patients suffering from Meniere's syndrome is similar to Meniere's disease. Sensorineural hearing loss, sacral nerve hearing loss is affected by the inner ear component or the accompanying nerve component and may contain a nerve, that is, when the auditory or auditory altar in the brain, the left channel is affected Occurs when 'or feelings'. Hearing loss is perceived as hereditary • or may be caused by auditory trauma (i.e., very loud noise), viral infection, drug-induced or Meniere's disease. Neurological hearing loss can be caused by brain tumors, infections, or multiple brain and nerve disorders, such as stroke. Some hereditary diseases, such as Refsum's disease (defective accumulation of branched fatty acids) can also cause neurological disorders that affect hearing loss. The auditory nerve is destroyed by the demented Zhaole disease, such as spontaneous inflammation and demyelinating diseases (including multiple 140447.doc *28- 200950787 hardening), transverse benign myelitis, Devic's disease, progressive progressive multiple leukoencephalopathy' Guniain-Barre syndrome, chronic inflammatory demyelinating polyneuropathy and anti-MAG peripheral neuropathy. The incidence of transient deafness, or sensorineural hearing loss occurs in about 1 in every 5 people and is caused by a viral or bacterial infection, such as mumps, measles, colds, chickenpox, cytomegalovirus, syphilis or infection. Physiological damage to mononuclear leukocytosis or inner ear g. In some cases, the cause of ^ was not recognized. Tinnitus and dizziness may be accompanied by an instant deafness, which gradually subsides. Oral corticosteroids are often prescribed to treat sensorineural hearing loss. In some cases, surgery is required. The formulations and methods described herein include treatment of sensorineural hearing loss, including treatment of transient sensorineural hearing loss, including spontaneous transient sensorineural hearing loss. For SSHL, current treatment options include high-dose oral steroids with methyl dehydrocorticosterol (4-10 mg/ml) or methyl-dehydrocortisol (4〇_62 5 mg/ml) for 2 weeks ( 4_7 days of treatment +7_[〇天减©量). As noted herein, high doses of oral steroids are accompanied by undesired (five) cents and adverse conditions. Thus, it is contemplated that the formulations and methods described herein for sustained release of the steroids, local delivery to the inner ear, result in significantly fewer side effects than oral/system steroid use. In one embodiment, the ISSHL is characterized by a unilateral sensorineural hearing loss during a seizure of less than 72 hours, wherein HL is defined as being at least 3 close to the test frequency > A standard care for spontaneous transient sensorineural hearing loss (ISSHL) is treatment with high dose oral steroids. In a specific example, one body is treated with a dose of oral steroid for about two weeks. In a particular example, one body is treated with oral steroids at a dose of 140447.doc • 29-200950787 for about two weeks, followed by a gradual reduction in oral steroids from about 7 to about 1 day. In a particular example, the oral steroid is methyl dehydrocorticosterol (4-10 mg/ml). In a particular example, the oral steroid is methyl-dehydrocorticin (40-62.5 111§/1111). In one set of embodiments, in a set of embodiments, a patient who is treated with standard care as described above for the treatment of ISSHL is replaced with an auris-acceptable formulation and method for controlling the release of corticosteroids as described herein. In one set of embodiments, a patient who is treated with IS S HL using the standard care described in A, but which is resistant or non-responsive to this treatment, is an auris-acceptable formulation that controls the release of corticosteroids as described herein. And method alternatives. Hearing loss due to excessive 喿 听力 Hearing loss may be caused by prolonged exposure to loud noise, such as loud sounds, heavy instruments or implements, aircraft 'snapping sounds or other human noise. This hearing loss occurs as a result of destruction of the inner ear hair cell receptor. This type of hearing loss is often accompanied by tinnitus. Permanent damage to hearing loss is usually cherished. Although currently unable to treat noise-induced hearing loss, several treatments have been experimentally developed, including treatment with insulin-growth factor therapy. (Lee et al., Otol. Neurotol. (2007) 28: 976-981). Deafness or aging in the elderly - hearing loss is part of normal aging, and because of the Coriolis spiral in the inner ear (the Corti organ receptor cells degenerate). Other causes may also constitute a reduction in several nerve fibers in the vestibular deafness and loss of elasticity in the midbrain of the cochlea. There is currently no treatment for permanent hearing loss due to deafness or excessive noise in the elderly. 140447.doc •30· 200950787 Drug-induced damage to the inner ear caused by the administration of the drug, including specific antibiotics, diuretics (such as escicolic acid (ethacrynie mail (4) and Fushimai (10) mide)), aspirin Like aspirin (such as salicylate) and quinine. Degeneration of the inner ear B can be accelerated by impaired renal function, which results in reduced clearance of the drug and its metabolites. Medicine can affect hearing and balance, but not * can affect hearing to a larger extent. 0 For example, neomycin, kenmycin, and amikacin have a greater utility in hearing ratio balance. The antibiotics ziosporin, mycotoxin and taiwansu affect both hearing and balance. Streptomycin, another commonly administered antibiotic, causes hearing loss in addition to dizziness and may cause Dandy's syndrome to be difficult to set in the dark and to trigger a sense of environmental movement at each step. Aspirin, when taken at high doses, may cause temporary hearing loss and tinnitus, which is a symptom of the sound when there is no external sound. Similarly, quetiapine, esciclamic acid, and foximab can cause temporary or permanent hearing loss. φ Autoimmune inner ear disease Autoimmune inner ear disease (AIED) is one of the few reversible causes of sensorineural hearing loss. It is two disorders that are less common in both adults and children, and it is usually a bilateral disorder of the inner ear and vestibular function. The AIED source may attack the inner ear structure with autoantibodies and/or immune cells, but with other autoimmune symptoms. In many cases, AIEDS is born without systemic autoimmune symptoms, but up to one-third of patients also suffer from systemic autoimmune diseases such as inflammation, intestinal disease, rheumatoid arthritis, ankylosing spondylitis, systemic erythema Lupus (SLE), Hugh's syndrome, Kegang's disease, ulceration 140447.doc -31 - 200950787 Colitis, Wegener's granulomatosis and scleroderma. Bessel's disease, multiple system diseases are also often heard, and there are also vestibular problems. There is some evidence that food-related allergies are the cause of autoimmune in the cochlea and vestibule, but there is currently no recognition of its importance in disease etiology. A classification of AIED has been developed (Harris and Keithley, (2002) Autoimmune inner ear disease, in Otorhinolaryngology Head and Neck Surgery. 91, 18-32). Treatment with corticosteroids relieves symptoms of AIED. Oral administration of the corticosteroid Prysson (60 mg/day for 4 weeks) showed an improvement in pure tone and speech hearing results. The regulation of corticosteroids occurs via corticosteroid receptors or salt receptors. Inflammatory disorders Inflammatory disorders of the ear include, but are not limited to, otitis media, otitis externa, mastoiditis, bullous tympanitis, Eustachian tubal catarrh, Eustachian salpingitis, vaginitis or the like. Examples include symptoms that affect otitis media (OM) in adults and children, including acute otitis media (AOM), otitis media with otitis (OME), and chronic otitis media. OM sensibility is multifactorial and complex, including the environment, microbes and host factors. An increase in cytokine production, including interleukin and TNF, has been observed in effluent mediators suffering from OM individuals. Treatment with anti-inflammatory steroids Sustained-release symptoms of inflammatory disorders in the ear (eg, otitis media, Eustachian tubal catarrh or similar). In some instances, the inflammatory disorder is a bacterial infection (e.g., OM). In some instances, a combination of an antibiotic and an anti-inflammatory corticosteroid can alleviate OM symptoms. 140447.doc -32- 200950787 Pharmacy The present invention provides corticosteroid compositions or formulations that improve or alleviate otic disorders including Meniere's disease, sensorineural hearing loss and/or inflammatory disorders and concomitant symptoms, including but Not limited to hearing loss, eye damage, * glare, tinnitus, inflammation, swelling, infection, and congestion. Ear disorders including fine or plum • Er's disease and/or inflammatory disorders are reported in response to this article. <The etiology and symptoms of a drug or other agent. Modifications that are not described herein but that are useful for φ 彳 失 或 或 或 或 或 或 或 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显 显Furthermore, 'agents which have previously been shown to be toxic, harmful or ineffective during the general or topical application of other organ systems, such as toxic metabolites formed by liver action, drugs which are toxic to specific organs, tissues or systems, Agents that require high amounts to achieve utility, are not liberated via the system pathway, or are released via ρK poor properties can also be used in certain embodiments herein. For example, side effects of methyl dehydrocorticolysis include: retention, excessive water retention, bloody heart failure in patients with susceptibility to infection, hypertension, muscle weakness, muscle atrophy, osteoporosis, sputum rupture, gastric ulcer, ulcerative Esophagitis, skin • Thinning, skin reaction, weakened wound healing, convulsions, dizziness, headache, psychological disorders, Cushing, s synd_e, delayed growth of children, diabetes, hirsutism, cataracts, f Light eyes, weight gain, increased appetite and nausea. Pharmaceutical agents with limited or no systemic release, systemic toxicity: poor ρκ properties or combinations thereof are expected to be included in the examples of the present invention. The corticosteroid formulation of the invention (iv) may optionally be in the form of an ear structure that is in need of treatment 140447.doc -33.200950787; for example, a contemplated embodiment is the direct application of the corticosteroid formulation disclosed herein to the inner ear window membrane. Or the crown of the cochlear fossa allows direct access to the inner or inner ear components and treatment. In other implementations, the corticosteroid formulations disclosed herein are applied directly to a round window. In other embodiments, direct access can be achieved by, for example, direct microinjection into the inner ear to achieve microinfusion, for example, via deafness. This embodiment can also optionally include a drug delivery device wherein the drug delivery device delivers a corticosteroid formulation via a needle and syringe, pump, microinjection device, an in situ formed sponge material, or the like. In still other embodiments, the administration of the corticosteroid formulation is directed to the middle ear via perforation of the inner eardrum and the corticosteroid formulation is applied directly to the affected middle ear structure, including the tympanic wall or ossicular bone, to the middle ear. Thus, the corticosteroid formulations disclosed herein can be limited to the target middle ear structure and are not lost by diffusion or osmosis of the tympanic membrane, for example, via the eustachian tube or perforation. Corticosteroids/Anti-inflammatory Steroids Corticosteroids are characterized by the effects of mineralocorticoids and glucocorticoids, depending on the pharmacology of the agent. The characteristic of mineralocorticoid is its similarity to aldosterone and its effect on the amount of electrolyte and water balance. Glucocorticoids, such as the endogenous glucocorticoid cortisol, which control metabolism and have anti-inflammatory properties by preventing the release of cytokines. Many agents have the degree of mineralocorticoid and glucocorticoid activity. The relative potency and activity of several synthetic glucocorticoids are shown in the table below. 140447.doc • 34- 200950787 steroid cortisol Prysone dehydrocortisol methyl dehydrocorticosterol beta cortisol prednylidene aldosterone cortisol efficacy 1 4 1.7 21 45 0.35 182 0.07 Mineralocorticoid efficacy 0.054 0,002 0.013 0.0094 0.0038 0.0002 0.0011 1.0

系統性之糖皮質素治療為目前用於自體免疫聽力喪失的 治療。基本的療料行數月且具實質的“㈣療的副作 用。對於f基脫氫皮質_的副❹包m留n 水滯留、易感染患者的鬱血性心衰竭、高血壓、肌肉無 力、肌肉萎縮、骨質疏鬆症、腱破裂、胃潰癌、潰癌性食 道炎、皮膚薄化、皮膚反應、傷口癒合能力減弱、痙攣: 軍眩、頭痛 '心理失調、庫興氏症候群(Cushing,s㈣贿十 兒童延遲生長、糖尿症、多毛症、白内障、青光眼、體重Systemic glucocorticoid therapy is currently used for the treatment of autoimmune hearing loss. The basic treatment line lasts for several months and has a substantial "(four) treatment side effects. For the f-based dehydrogenated cortex _ the secondary sputum m left n water retention, susceptible to septic heart failure, high blood pressure, muscle weakness, muscle Atrophy, osteoporosis, sputum rupture, gastric ulceration, ulcerative esophagitis, skin thinning, skin reaction, weakened wound healing, sputum: military glare, headache 'psychological disorder, Cushing's syndrome (Cushing, s (four) bribe Ten children with delayed growth, diabetes, hirsutism, cataract, glaucoma, weight

增加、增加食慾及噁心。使用本文所描述之配方的一優 為大大減少系統性曝露至抗炎症的糖皮質素類固醇。 去氫皮質醇為一具有主要為糖皮質素且低鹽皮質素活性 的皮質類固醇藥。其具有内生皮質醇的效能之約4-5倍。 其為一口服投藥普賴松的活性代謝物。甲基脫氫皮質固酵 為具有糖皮質素活性的皮質類固醇。其具有内生皮質醇的 效能之約25-30倍。甲基脫氫皮質固醇磷酸鈉為一曱基脫 氯皮質固醇的水可溶磷酸酯前藥。已公開一分析測定甲基 脫氯皮質固醇磷酸酯在耳堝周邊淋巴液體中的方法(Liu et 140447.doc -35- 200950787 al,J. of Chromatography B (2004),805(2):255-60)。特安皮質 醇為一合成糖皮質素藥,其可經由口服、注射、吸入、戋 局部乳霜或軟膏投藥。特安皮質醇丙縮酮為更有效能的同 類物。特安皮質醇六縮酮為一特安皮質醇丙縮酮的比弗基 酯(pivolyl ester)六丙縮酮。貝可皮質醇二丙酸酯為一非常 有效能的糖皮質素藥,亦稱為貝氯美松(becl〇metas〇ne)。貝 皮質醇S用於局部配方時為一非常有效能的皮質類固醇。 其具有抗炎症、止癢劑、管收縮及免疫調變性質。 在一實施例中’本文所描述的配方之活性醫藥成份為去 氫皮質醇。在另一實施例中在另一實施例中’本文所描述 的配方之活性醫藥成份為曱基脫氫皮質固醇。在另一實施 例中’本文所描述的配方之活性醫藥成份為甲基脫氫皮質 固醇磷酸酯。在又一實施例中,本文所描述的配方之活性 醫藥成份為貝可皮質醇。在又一實施例中,本文所描述的 配方之活性醫藥成份為貝皮質醇。在又一實施例中,本文 所描述的配方之活性醫藥成份為特安皮質醇。在又一實施 例中’本文所描述的配方之活性醫藥成份為特安皮質醇丙 縮酮。在又一實施例中’本文所描述的配方之活性醫藥成 份為氣貝皮質醇。 在又一實施例中,本文所描述的配方之活性醫藥成份為 糖皮質素類固醇的磷酸酯前藥。在又一實施例中,本文所 描述的配方之活性醫藥成份為糖皮質素類固醇的酯前藥。 在又一實施例中,本文所描述的配方之活性醫藥成份為選 自21-乙酿氧孕稀醇嗣、氣皮質醇、阿結斯_ (aigest〇ne)、 140447.doc -36 - 200950787 ΦIncrease, increase appetite and nausea. An advantage of using the formulations described herein greatly reduces systemic exposure to anti-inflammatory glucocorticoid steroids. Dehydrocortisol is a corticosteroid with a predominantly glucocorticoid and low mineralocorticoid activity. It has about 4-5 times the potency of endogenous cortisol. It is an active metabolite of oral administration of Prysone. Methyl dehydrocorticolysis is a corticosteroid with glucocorticoid activity. It has about 25-30 times the potency of endogenous cortisol. Sodium methyl dehydrocorticosterone phosphate is a water-soluble phosphate prodrug of monodecyl chlorocorticosterol. An analytical method for the determination of methyl dechlorinated cortisol phosphate in the peripheral lymphatic fluid of deafness has been disclosed (Liu et 140447.doc-35-200950787 al, J. of Chromatography B (2004), 805(2): 255 -60). Tetracortisol is a synthetic glucocorticoid that can be administered orally, by injection, by inhalation, in the form of a topical cream or ointment. Teancortidol is a more potent isoform. Teancortisol hexagram is a pivolyl ester of hexavolone. Beco Cortisol dipropionate is a very potent glucocorticoid, also known as beclomethasone (becl〇metas〇ne). Shell Cortisol S is a very potent corticosteroid for topical formulation. It has anti-inflammatory, antipruritic, tube contraction and immunomodulatory properties. In one embodiment, the active pharmaceutical ingredient of the formulation described herein is dehydrocortisol. In another embodiment, in another embodiment, the active pharmaceutical ingredient of the formulation described herein is a mercapto dehydrocorticosterol. In another embodiment, the active pharmaceutical ingredient of the formulation described herein is methyl dehydrocorticosteroid phosphate. In yet another embodiment, the active pharmaceutical ingredient of the formulations described herein is becocortisol. In yet another embodiment, the active pharmaceutical ingredient of the formulation described herein is festibrol. In yet another embodiment, the active pharmaceutical ingredient of the formulations described herein is steanocortisol. In yet another embodiment, the active pharmaceutical ingredient of the formulation described herein is triamcinol acetal. In yet another embodiment, the active pharmaceutical ingredient of the formulation described herein is gas shell cortisol. In yet another embodiment, the active pharmaceutical ingredient of the formulations described herein is a phosphate prodrug of a glucocorticoid steroid. In yet another embodiment, the active pharmaceutical ingredient of the formulations described herein is an ester prodrug of a glucocorticoid steroid. In yet another embodiment, the active pharmaceutical ingredient of the formulation described herein is selected from the group consisting of 21-B-oxygenated sterol, gas cortisol, Azeus _ (aigest〇ne), 140447.doc-36 - 200950787 Φ

阿西諾财((amcinonide)、貝可皮質醇、貝皮質醇、亞丁皮 質醇、氯普賴松、氣貝皮質醇、可洛貝他松、可洛脫酮 (clocortolone)、克普醇(cloprednol)、皮質固酮、皮質 _、 可堤凡醇(cortivazol)、得拉雜克(deflazacort)、德松耐 (desonide)、德松西酮(desoximetasone)、曱基脫氫皮質固 醇、二氟拉松酯、二氣脫酮(diflucortolone)、二氟普納 (difluprednate)、依諾嗣(enoxolone)、佛阿柯特(fluazacort)、 氟克耐(flucloronide)、氟皮質醇(flumethasone)、氟尼索里 (flunisolide)、丙嗣氟洛皮質醇(fluocinolone acetonide)、 氟洛奈皮質醇(fluocinonide)、丁基氟洛柯汀(fluocortin butyl)、氟柯脫酮(fluocortolone)、氟美拉酮、氟特嗣乙酸鹽 (fluperolone acetate)、敗比耐定乙酸鹽(fluprednidene acetate).、 氟去氫皮質醇、氟喃諾定(flurandrenolide)、丙酸氟替皮質 醇、氟馬柯越(formocortal)、海辛諾財熱(halcinonide)、鹵 貝松丙酸鹽(halobetasol propionate)、鹵美他松(halometasone)、 鹵普酮乙酸醋(halopredone acetate)、氫柯他美 (hydrocortamate)、氫皮質酮(hydrocortisone)、洛特醇乙他 普鹽(loteprednol etabonate)、馬斯普酮(mazipredone)、經孕酮 (medrysone)、美普賴松、甲基去氫皮質醇、皚膚美得 (mometasone furoate)、派拉美沙松(paramethasone)、普寧 卡貝(prednicarbate)、去氫皮質醇、去氫皮質醇25-二乙基 胺基-乙酸鹽、去氫皮質醇磷酸鈉、普賴松、普列寧弗 (prednival)、普财力定(prednylidene)、林馬索酮(rimexolone)、 堤索卡多(tixocortol)、特安皮質醇、丙鲷特安皮質醇、班 140447.doc •37· 200950787 納特安皮質醇(特安皮質醇benetonide)、六丙酮特安皮質 醇及其等之磷酸前藥或酯前藥組合。Ascinoide (amcinonide, beco Cortisol, betcor Cortisol, buta Cortisol, clprepresin, spirulina cortisol, colobetasone, clocortolone, ketol ( Cloprednol), corticosterone, cortex _, cortivazol, declazacort, desonide, desoximetasone, sulfhydryl dehydrocorticosterol, difluoro Larson ester, diflucortolone, difluprednate, enoxolone, fluazacort, flucloronide, flumethasone, fluoride Flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, flumethazone , fluperolone acetate, fluprednidene acetate, fluorodehydrocortisol, flurandonolide, fluticaginol propionate, fluococcal (formocortal) ), Haixinnuo (halcinonide), halogen Halobetasol propionate, halometasone, halopredone acetate, hydrocortamate, hydrocortisone, lotatrol (loteprednol etabonate), mazipredone, meridone, mepredone, methyl dehydrocortisol, mometasone furoate, paramethasone, Puning Prednicarbate, dehydrocortisol, dehydrocortisol 25-diethylamino-acetate, dehydrocortisol sodium phosphate, predisin, prednival, prednylidene , rimexolone, tixocortol, triamian cortisol, acetaminophen cortisol, ban 140447.doc •37· 200950787 natalan cortisol (teanol corte benetonide), A combination of hexaacetone triamcinol and its phosphate prodrug or ester prodrug.

clobetasol propionate 在某些實施例中,本文所描述的配方具有佔配方之活性 成份的介於約〇.〇 1°/。-約20%間,介於約0.01 %-約1 0%間, 介於約0.01%-約8%間,介於約0.05-6°/。間,介於約0.1-5% 間,介於約0.2-約3%間,或介於約0.1-約2%間之活性醫藥 成份濃度。在某些實施例中,本文所描述的配方具有佔配 方之體積的介於約〇.卜約70 mg/mL間,介於約1 mg-約 70 mg/mL間,介於約1 mg-約50 mg/mL間,介於間約1 mg/mL 至約20 mg/mL,介於約1 mg/mL至約10 mg/mL間,介於間 140447.doc -38- 200950787 勺1邮㈣至約5 mg/mL,或介於約0.5 mg/mL至約5 mg/mL 間之活性醫藥成份濃度。 在某些實施例中,本發明揭露之配方更包含一抗生素且 可用於/σ療本文描之耳部疾病或症狀。抗生素包括但未限 為丁胺卡那黴素、紫菌素、康黴素、新黴素、耐特徽 f、鏈黴素、泰百黴素、巴龍黴素、凝膠達那黴素,除筹 • 射、羅拉卡倍芙、厄他培南、多利培南、亞胺培南、亞 φ 胺培南、美羅培南、頭孢卓西、頭孢坐林、頭孢洛林 (cefalotin)、頭孢氨节、頭孢可若、頭孢孟多、頭孢西 丁、去普集(defprozil)、頭孢福辛、希復欣敏、頭孢地 尼、頭孢妥侖、頭孢哌酮、頭孢唑肟、頭孢泊肟、頭孢他 汀、頭孢布烯、頭孢唑肟、頭孢曲松鈉、頭孢吡肟、替考 拉寧、萬古黴素、亞茲索黴素、克拉黴素、地紅黴素、紅 徽素、羅紅黴素、桃黴素、替利黴素、觀黴素、氮烯内醯 胺、胺經苄青黴素、胺苄青黴素、阿洛西林、卡本西林、 # 氯嘆青黴素、二氣噻青黴素、氟氣噻青黴素、美洛西林、 • 美地西林(meticillin)、乙氧萘青黴素、扼噻青黴素、盤尼 西林、必倍西林、三卡西林(ticarcillan)、枯草菌素、柯利 ,黴素、多黏菌素B、塞普沙辛、伊諾沙星、加替沙星、洛 弗沙星(levofloxacin)、洛美沙星、莫西沙星、諾弗洒欣、 歐弗》西欣、特洛弗沙星(trovfloxacin)、項胺米隆、普脫西 (prontosil)、磺乙醯胺、磺胺甲二唑、硫芬依米啶 (sulfanimilimde)、硫沙拉嗪(sulfsalazine)、硫西雜唾 (sulfsioxazole)、曲美普林(trimethoprim)、地美環素 140447.doc -39· 200950787 (demeclocycline)、去氧經四環黴素、美諾四環素、四環黴 素、歐四環黴素(oxtetracycline)、石申凡納明、氣徽素、氯 林絲菌素、林可黴素、乙二胺二丁醇、弗司弗黴素、梭鏈 孢酸、富來頓、異菸酸肼、雷奈佐利、硝基曱嘧唑乙酵、 莫螢菌素、硝基呋喃妥因、平板黴素、°比嗪甲醯胺、奎奴 普丁/達福普汀、立況黴素、AL-15469A (Alcon Research)、 AL-38905 (Alcon Research)及其等之組合。 滅菌作用的一般方法 本發明提供耳部組成物以改善或減輕本文所述之耳部 失調。本發明更提供包含該耳部組成物投藥的方法。在某 些實施例中,組成物為滅菌的。在本文揭示的實施例中揭 露的醫藥組成物滅菌作用之裝置與方法為用於人類。此目的 為提供一安全的醫藥產品,相對無引起感染的微生物。美國 食物暨藥品管理局提供公開之規範手冊「Guidance for Industry: Sterile Drug Products Produced by Aseptic Processing」, 可見於:http//www.fda.gov/cder/guidance/5882fnl.htm,其全 文併入本案做為參考。 在本文中使用之滅菌作用意指破壞或除去存在於產品或 包裝之微生物的方法。可使用任何可取得之合宜物件及組 成物之滅菌方法。可利用的微生物不活作用之方法包括但 未限制為過熱、致死化學品、或γ輻射的應用.。在某些實 施例中,製備耳部治療配方的方法包含將配方進行一選自 熱滅菌作用、化學滅菌作用、輻射滅菌作用或過濾滅菌作 用的滅菌方法。使用的方法主要依被滅菌的裝置或組成物 140447.doc •40- 200950787 之性質而定。滅菌作用的許多方法之詳細描述可見於 Remington : The Science and Practice of Pharmacy published by Lippincott之第40章,Williams & Wilkins,該内容併入本案 此一主題做為參考。 加熱的滅菌作用 許多方法可用於藉由極熱應用的滅菌作用。一方法為藉 由使用飽和蒸氣高壓爸。在此方法中,在至少121 温卢 飽和泰風允_·與被滅菌的物件接觸。在物件被滅菌的例子 中,熱直接傳送至微生物,或藉由加熱被滅菌之水溶液的 容積而間接至微生物。此方法廣泛的使用,因為其在滅菌 製程中允許使用彈性、安全及經濟考量。 乾熱滅菌作用為一在增温用於殺死微生物及進行去熱原 作用。此製程包括在一 適於加熱HEPA-過濾之無微生物空 氣達至少13(M8(TC温度以用於滅菌作用及達至少23〇· 25(TC温度以用於去熱原作用的裝置中進行。重建濃縮或 粉末配方之水亦藉由高壓釜滅菌。在某些實施例中,本文 所描述的配方包含微米化之藥劑(如,皮質類固醇,(如, 微-曱基脫氫皮質固醇)),該藥劑藉由乾熱,如在内部粉末 温度為13(M4(TC加熱約7_u小肖,或在内部温度為H 180 C加熱約1 -2小時而滅菌。 化學滅菌作用Clobetasol propionate In certain embodiments, the formulations described herein have an active ingredient in the formulation of between about °.〇 1°/. - between about 20%, between about 0.01% and about 10%, between about 0.01% and about 8%, between about 0.05-6°/. The concentration of active pharmaceutical ingredient is between about 0.1 and 5%, between about 0.2 and about 3%, or between about 0.1 and about 2%. In certain embodiments, the formulations described herein have a volume of the formulation of between about 70 mg/mL, between about 1 mg to about 70 mg/mL, and between about 1 mg- Between 50 mg/mL, between about 1 mg/mL and about 20 mg/mL, between about 1 mg/mL and about 10 mg/mL, between 140447.doc -38-200950787 scoop 1 post (d) to a concentration of active pharmaceutical ingredient of between about 5 mg/mL, or between about 0.5 mg/mL and about 5 mg/mL. In certain embodiments, the formulations disclosed herein further comprise an antibiotic and can be used to treat the ear disease or condition described herein. Antibiotics include but are not limited to amikacin, mycotoxin, oxytetracycline, neomycin, refuge f, streptomycin, tambamycin, paromomycin, gelatin , in addition to Philippine, Lolacabe, ertapenem, doripenem, imipenem, imipenem, meropenem, cefotaxime, cephalosporin, cefalotin, cephalosporin Ammonia, cefotaxime, cefmenudene, cefoxitin, defprozil, cefotaxime, xifuxinmin, cefdinir, cefoperon, cefoperazone, ceftizoxime, cefpodoxime , ceftazidime, ceftibuten, ceftizoxime, ceftriaxone sodium, cefepime, teicoplanin, vancomycin, azithromycin, clarithromycin, dirithromycin, red fluorescein, ro Erythromycin, citrillin, telithromycin, spectinomycin, nitrene, amine benzylpenicillin, ampicillin, azlocillin, carbencillin, # leucine, dithiacillin, Fluorothiacillin, mezlocillin, meticillin, ethoxypenicillin, acesulfame, penicillin , must be cillin, ticarcillan, cumin, clopidogrel, polymyxin B, cepsporin, enofloxacin, gatifloxacin, lofofloxacin , lomefloxacin, moxifloxacin, norfloxacin, oufuxixin, tlovfloxacin, mimetamine, prontosil, sulfacetamide, sulfamethoxazole , sulfanimilimde, sulfsalazine, sulfsioxazole, trimethoprim, dimecycline 140447.doc -39· 200950787 (demeclocycline), deoxygenation By tetracycline, menocycline, tetracycline, oxtetracycline, oxhenfanamine, chlorfenapyr, lincomycin, lincomycin, ethylenediamine dibutanol, Fosfomycin, fusidic acid, fulton, bismuth nicotinate, renazol, nitropyrimidine, ferromycin, nitrofurantoin, plateau, pyrazin Amine, quinupristin/dalofopine, oxymycin, AL-15469A (Alcon Research), AL-38905 (Alcon Research), etc. . General Method of Sterilization The present invention provides an ear composition to improve or alleviate the otic disorders described herein. The invention further provides a method of administering the otic composition. In some embodiments, the composition is sterilized. The apparatus and method for sterilizing a pharmaceutical composition disclosed in the embodiments disclosed herein are for use in humans. The aim is to provide a safe pharmaceutical product with relatively no infection-causing microorganisms. The US Food and Drug Administration provides a published specification manual "Guidance for Industry: Sterile Drug Products Produced by Aseptic Processing", which can be found at: http//www.fda.gov/cder/guidance/5882fnl.htm, which is fully incorporated into the case. As a reference. Sterilization as used herein means a method of destroying or removing microorganisms present in a product or package. Any sterilizable method of the appropriate article and composition can be used. Methods of inactivating microorganisms that may be utilized include, but are not limited to, applications of superheat, lethal chemicals, or gamma radiation. In certain embodiments, a method of preparing an ear treatment formulation comprises subjecting the formulation to a sterilization process selected from the group consisting of heat sterilization, chemical sterilization, radiation sterilization, or filtration sterilization. The method used is primarily dependent on the nature of the device or composition being sterilized 140447.doc •40- 200950787. A detailed description of many of the methods of sterilization can be found in Remington: The Science and Practice of Pharmacy published by Lippincott, Chapter 40, Williams & Wilkins, incorporated herein by reference. Sterilization by heating Many methods are available for sterilization by extreme heat applications. One method is to use a saturated steam high pressure dad. In this method, at least 121 liters of saturated typhoon is allowed to come into contact with the sterilized article. In the case where the article is sterilized, the heat is directly transferred to the microorganism or indirectly to the microorganism by heating the volume of the sterilized aqueous solution. This method is widely used because it allows for flexibility, safety, and economic considerations in the sterilization process. Dry heat sterilization is used to increase the temperature to kill microorganisms and perform depyrogenation. The process comprises performing a HEPA-filtered non-microbial air for at least 13 (M8 (TC temperature for sterilization and up to at least 23 〇 25 (TC temperature for depyrogenation). Reconstituted concentrated or powdered formula water is also autoclaved. In certain embodiments, the formulations described herein comprise micronized agents (eg, corticosteroids (eg, micro-mercapto dehydrocorticosterol) The agent is sterilized by dry heat, such as at an internal powder temperature of 13 (M4 (TC heating about 7 _ Xiao Xiao, or heating at an internal temperature of H 180 C for about 1-2 hours. Chemical sterilization)

蒸氣如環氧乙烷、二氧化氯、 140447.doc w人阶㈣囷忭用之極端 使用具有殺菌性質之多 氯、甲醛或臭氧做為抗 -41- 200950787 屑亡劑。環氧乙燒之殺菌活性,例如,來自其能做為一反 應性烧基化劑。因此,滅菌作用需要環氧乙烧蒸氣以直接 與被滅函的產品接觸。 輻射滅菌作用 輻射滅菌作用之-優點為滅菌許多形式產物的能力且 無熱降解或其他破壞。使用的一般輻射為P輻射或可替代 之來自6〇C〇源的γ輻射。γ輻射的穿透能力允許用於許多產 品型式之滅菌作用,包括溶液、組成物及異質混合物。輻 射之殺菌效用來自於γ輻射與生物巨分子之交互作用。此 父互作用產生帶電物種及自由基。後續的化學反應,如重 組及交聯作用,導致此些生物巨分子的正常功能喪失。本 文所述之配方亦可選擇使用Ρ輻射滅菌。 .過濾 過渡滅菌作用為一用於由溶液除去但不會破壞微生物的 方法。使用膜濾網以過濾對熱敏性溶液。此濾網為一具有 孔大小範圍在〇.丨至〇 22 μπι間之混合纖維酯(MCE)、聚氟 化亞乙烯(P VF ;亦已知為p VDF)、或聚四氟乙烯(PTFE)的 薄、強、均質聚合物。可使用不同過濾以可選擇地過濾不 同特性溶液。例如,PVF及PTFE膜已知為適用於過濾有機 溶劑’同時水溶液以PVF或MCE膜過濾。可取得用於不同 規模的遽網裝置範圍由接在注射器上的單點使用之可抛棄 式滤網至用於製造工廠使用的商業規模。此膜濾網可以高 壓爸或化學滅菌作用滅菌。膜過濾系統的有效性係依下列 標準平台進行(Microbiological Evaluation of Filters for U〇447d〇C -42- 200950787Vapor such as ethylene oxide, chlorine dioxide, 140447.doc w human (4) extreme use of bactericidal properties of chlorine, formaldehyde or ozone as anti-41-200950787 scraping agent. The bactericidal activity of ethylene bromide, for example, can be used as a reactive alkylating agent. Therefore, the sterilization requires an ethylene bromide vapor to directly contact the product to be extinguished. Radiation Sterilization - The advantage of radiation sterilization is the ability to sterilize many forms of products without thermal degradation or other damage. The general radiation used is P radiation or alternatively gamma radiation from a 6 〇 C source. The penetrating power of gamma radiation allows for sterilization of many product types, including solutions, compositions, and heterogeneous mixtures. The bactericidal effect of radiation comes from the interaction of gamma radiation with biological macromolecules. This parent interaction produces charged species and free radicals. Subsequent chemical reactions, such as recombination and cross-linking, result in the loss of normal function of these biomacromolecules. Formulations described herein may also be selected for sterilization with xenon radiation. Filtration Transition sterilization is a method for removing from a solution without destroying the microorganism. A membrane filter was used to filter the heat sensitive solution. The screen is a mixed fiber ester (MCE) having a pore size ranging from 〇.丨 to 〇22 μπι, polyvinylidene fluoride (P VF; also known as p VDF), or polytetrafluoroethylene (PTFE). Thin, strong, homogeneous polymer. Different filters can be used to selectively filter different characteristic solutions. For example, PVF and PTFE membranes are known to be suitable for filtering organic solvents' while aqueous solutions are filtered with PVF or MCE membranes. The range of webs available for use on a wide range of sizes can be achieved by a single point disposable filter on a syringe to a commercial scale for use in manufacturing plants. This membrane filter can be sterilized by high pressure dad or chemical sterilization. The effectiveness of membrane filtration systems is based on the following standard platforms (Microbiological Evaluation of Filters for U〇447d〇C -42- 200950787)

Sterilizing Liquids > Vol 4 > No. 3. Washington » D.C. · HealthSterilizing Liquids > Vol 4 > No. 3. Washington » D.C. · Health

Industry Manufacturers Association,1981)且涉及以已知 量(約107/cm2)之不尋常小微生物挑戰膜濾網,如以 Brevundimonas diminuta (ATCC 19146) 0 醫藥組成物可選擇地藉由通過膜濾網而滅菌。含有奈米 粒子之配方(美國專利第6,139,870號)或多層氣孔(Richard 等人,International Journal of Pharmaceutics (2006), 312(1-2)·· 144-50)適於藉由經Ο·22 μπι濾網過濾而未破壞其 組織結構的滅菌作用。 在某些實施例中,本發明揭露之方法包含藉由過濾滅菌 作用以滅菌此配方(或其之組份)。在另一實施例中,耳_可 接受耳部治療劑配方包含一粒子,其中該粒子配方為適宜 過濾滅菌作用。在又一實施例中,該粒子配方包含之粒 子少於300 nm大小之粒子,少於200 nm大小之粒子,少於 10 0 nm之粒子。另一實施例中,耳-可接受耳部治療劑配 方包含一粒子,其中該粒子之無菌性可藉由先驅物質之組 份溶液的滅菌過濾而保證。另一實施例中,耳-可接受配 方包含一粒子配方’其中該粒子之無菌性可藉由低温滅菌 過濾保證。在又一實施例中’該低温滅菌過濾在介於〇與 3(TC間温度進行’或介於〇與2〇°C間’或介於0與i〇°c間, 或介於10與20°C間,或介於20與30°C間。 在另一實施例為一製備耳-可接受粒子配方的方法,其 包含:在低温經由一滅菌濾網過濾含有粒子配方的水溶 液;凍乾該滅菌溶液;及在投藥前以滅菌水重建粒子配方 140447.doc -43· 200950787 在另一實施例中。在某些實施例中’本文所描述之一配方 以一含有微米化的單一小瓶配方中活性醫藥成份的懸浮液 製造。一單一小瓶配方藉由混合一滅菌泊洛沙姆溶液與滅 菌微米化活性成份(如,甲基脫氫皮質固醇)的無菌混合並 傳送此配方至無菌醫藥容器而製備。在某些實施例中,一 含有本文所描述的配方之單一小瓶為在分配及/或投藥前 再懸浮^ 在特定實施例中,過濾及/或充填步驟在低於本文所述 之配方的凝膠温度(T凝膠)約5。(:下進行,該凝膠配方具有 低於理論值100 cP之黏性以允許使用一蠕動泵在一合理時 間過渡。 在另些其他實施例中,耳-可接受耳部治療劑配方包含 一奈米粒子配方,其中該奈米粒子配方為適宜過濾滅菌作 用。在又一實施例中,奈米粒子配方包含奈米粒子大小為 少於300 nm ’大小少於200 nm,或大小少於1〇〇 nm。在另 些其他實施例’耳-可接受配方包含一微球體配方,其中 該微球體的無菌性可藉由先驅物質之有機溶液及水溶液的 滅菌過濾而保證。在另些其他實施例,耳-可接受配方包 含—熱可逆凝膠配方,其中該凝膠配方的無菌性可藉由低 温滅菌過濾保證。在又一實施例中,該低温滅菌過濾在介 於〇與30。(:間溫度進行’或介於〇與2〇〇c間,或介於〇與 10 C間’或介於1〇與2〇。(3間,或介於2〇與30。(:間。在另些 其他實施例為一製備耳-可接受熱可逆凝膠配方的方法, 其包含:在低温經由一滅菌濾網過濾含有熱可逆凝膠組份 H0447.doc -44 - 200950787 的水溶液;凍乾該 可逆凝膠配方。 及在投藥前以滅11水重建熱 在特定實施例中, 緩衝液)並分別的、p 份溶解於合宜之載劑(例如一 '囷(例如藉由熱處理、過濾、γ輻射). 宜方法在—分別液體凝膠組份)以一合 的步驟中滅菌(例如賦形劑的冷卻混合物 參 提供、H或輻射)’此二分別滅_溶液接著無菌混合以 挺供一最終耳配方。 在某些例子中’減菌作用(例如,熱處理(例如,在—高 )γ輻射、過濾)之傳統使用的方法導致此配方中 聚合物組份(例如,熱固化、膠化或黏膜黏附劑聚合物組 份)及/或活化劑的不可逆降解作用。在某些例子中,若此 配方包含在過據期間膝化的搖變減黏性聚合物,則藉由膜 過濾(例如,0.2 _膜)之耳配方的滅菌作用為不可能。、 據此,本發明提供用於耳配方滅菌的方法,其預防聚合 物、’且伤(例如,熱固化及/或膠化及,或黏膜黏附劑聚合物 組份)及/或活化劑在滅菌作用期間的降解。在某些實施例 中,活化劑(例如,本文所述之任何治療耳劑)的降解可藉 由在此配方中緩衝液組份的特定範圍ρ Η之使用與膠化劑 的特定比例範圍使用而減少或去除。在某些實施例中,適 當膠化劑及/或熱固性聚合物的選擇可允許本文所述之配 方藉由過濾的滅菌作用。在某些實施例中,適當熱固性聚 合物及適當共聚物(例如,一膠化劑)使用與此配方之特定 ΡΗ範圍組合容許本文所述配方之高温滅菌作用且實質無 140447.doc •45· 200950787 治療劑或聚合物賦形劑的降解。在特定例子中,本發明提 供之滅菌彳法為將此配方經高壓爸進行終滅菌作用而在減 囷步驟期間無活化劑及/或賦形劑及/或聚合物組份的任何 喪失且產生實質無微生物及/或熱原。 微生物 本發明提供用於改善或減輕本文所述之耳部失調的耳_ 可接受組成物。更提供包含該耳部組成物的投藥方法。在 某些實施例中,該組成物實質無微生物。可接受無菌性量 為基於定義之治療可接受耳部組成物的施用標準,其其包 括但未限制於美國藥典第1 11 i章。例如,可接受無菌性量 包括每克配方10菌落形成單位(cfu),每克配方5〇 cfu,每 克配方100 cfU ’每克配方500 cfu或每克配方1〇〇〇 cfu。此 外,可接受無菌性量包括排除特定不利的微生物劑。例示 之特定不悅微生物劑包括但未限制為大腸桿菌(Escherichia coli ’ E_ coli)、沙門氏菌(Salmonella sp.)、綠膿桿菌(pseudom〇nas aeruginosa,P. aeruginosa)及/或其他特定微生物劑。 耳-可接受耳部療劑配方的無菌性可由美國藥典第 <61>、<62>及<71>的程序確定。無菌性品質控制保證、 品質保證及有效方法的一要件為無菌性測試的方法。無菌 性測試的僅用於例示之範例為以二方法進行。第一為直接 培育’其中被測試的組成物試樣加至培養基中並培育達2 i 天的時間。生長培養基的濁度顯示污染。此方法的缺點包 括大量物質之小試樣大小降低敏感度且係基於目視法檢測 微生物的生長。一可替代方法為膜過濾無菌性測試。在此 140447.doc •46· 200950787 方法中,產品的一體積通過一小膜濾網紙。此濾網紙接著 置於培養基中以促進微生物的生長。此方法當取樣全部質 量產品時具有較大敏感性的優點。可選擇使用商業可得的 Millipore Steritest無菌性測試系統以用於膜過遽無菌性測 試的測定。對於乳霜或軟膏的過濾測試,使用Steritest過 濾系統編號TLHVSL210。對於乳化液或黏性產品的過濾 測試,使用Steritest過濾系統編號TLAREM210或 TDAREM210 〇對於預充填注射器的過滤測試,使用 Steritest過濾系統編號TTHASY210。對於懸浮於氣霧劑或 發泡體物質的過渡測試,使用Steritest;過濾系統編號 TTHVA210。對於在安瓿或小瓶之可溶粉末的過濾測試, 使用 Steritest過濾系統編號TTHADA210或 TTHADV210。 用於大腸桿菌(E. coli)及沙門氏菌(Salmonella)的測試包 括使用乳糖培養液在30-35°C培育24-72小時,包括培育於 MacConkey及/或EMB洋菜中18-24小時,及/或使用Rappap0rt 培養基。檢測Ρ· aeruginosa的測試包括使用NAC洋菜。美 國藥典(USP)第62章更進一步舉出對於特定不愉悦之微生 物的測試步驟。 在特定實施例中’本文所述之任何控制釋放配方具有每 克配方少於約60菌落形成單位(Cfu),少於約50菌落形成 單位,少於約40菌落形成單位,或少於約3〇菌落形成單位 之微生物劑。在特定實施例中,調配之本文所述的耳部配 方為與内淋巴及/或周邊淋巴等張。 内毒素 140447.doc • 47· 200950787 本發明提供耳部組成物以改善或減輕本文所述之耳部失 調。更提供包含該耳部組成物投藥的方法。在某些實施例 中’該組成物實質無内毒素。一滅菌製程之一額外態樣為 除去源自殺死微生物的副產物(後文稱為「產物」)。去熱 原的方法由試樣中除去熱原。熱原為内毒素或外毒素,其 將誘發一免疫回應。内毒素之一範例為在格蘭氏陰性細菌 的細胞壁發現之脂聚醣(LPS)分子。當滅菌製程如高壓釜 或以環氧乙烷處理以殺死細菌,LPS殘餘物誘發前發炎免 疫回應,如敗血性休克。因為内毒素的分子大小非常廣, 内毒素的存在以「内毒素單位」(EU)表示β — EU相當於 100皮克之E. coll LPS。人類可生成一如體重之5 EU/kg少 的回應。此無菌性以此技中已認知的任何單位表示。在特 定實施例中,本文所述之耳部組成物當與傳統的可接受内 毒素量(例如,5 EU/kg體重)相比含較低内毒素量(例如 <4EU/kg體重)。在某些實施例争,在某些實施例中,此 耳可接受耳部治療劑配方具有少於個體之約5 EU/kg體 重。在其他實施例中,此耳_可接受耳部治療劑配方具有 少於個體之約4 EU/kg體重。在額外實施例中,此耳-可接 又耳部治療劑配方具有少於個體之約3 EU/kg體重。在額 外實施例中,此耳-可接受耳部治療劑配方具有少於個體 之約2 EU/kg體重。 在某些實施例中,此耳-可接受耳部治療劑配方具有少 於配方之約5 EU/kg。在其他實施例中,此耳-可接受耳部 治療劑配方具有少於配方之約4 Eu/kg。在額外實施例 140447.doc 200950787 中此耳可接又耳。P冶療劑配方具有少於配方之約3即心。 在某些實施例中,此耳_可接受耳部治療劑配方具有少於 產时之約5 EU/kg。在其他實施例中此耳_可接受耳部治 療劑配方具有少於產品之約1 EU/kg。在額外實施例中, 此耳-可接受耳部治療劑配方具有少於產品之約G.2 EU/kg。 f某些實施例中’此耳·可接受耳部治療劑配方具有少於 ^ 早兀或產品之約5 EU/kg。在其他實施例中,此耳-可接受 0 彳部治療劑配方具有少於單元或產品之約4 EU/kg。在另 外的實施例中,此耳_可接受耳部治療劑配方具有少於單 元或產之約3 EU/kg。在某些實施例中,此耳_可接受耳 部治療劑配方具有少於單元或產品之約5 Eu/kg。在其他 實施例中,此耳-可接受耳部治療劑配方具有少於單元或 產品之約4 EU/kg。在另外的實施例中,此耳_可接受耳部 治療劑配方具有少於單元或產品之約3 EU/kg。在特定的 實施例中,本發明揭露之耳組成物含有配方的約1至約 參 5 EU/mL。在特定的實施例中,本發明揭露之耳組成物含 • 有配方的約2至約5 EU/mL,配方的約3至約5 EU/mL,或 '配方的約4至約5 EU/mL。 在特定實施例中,|文所述之耳部組成⑱含有與傳統可 接受内毒素量(例如,配方之0.5 EU/mL)相比含有較低的 内毒素量(例如<〇配方之5 EU/mL)。在某些實施例中,此 耳-可接交耳部治療劑配方具有少於配方之約0.5 EU/mL。 在其他實施例中,此耳_可接受耳部治療劑配方具有少於 配方之約0.4 EU/mL。在額外實施例中,此耳-可接受耳部 140447.doc -49- 200950787 治療劑配方具有少於配方之約0.2 EU/mL。 做為範例說明,熱原檢測藉由數種方法進行。無菌性的 合宜測試包括述於美國藥典(USP)第71章無菌性測試(第23 版,1995)的測試。兔熱原測試及美洲鱟試驗法(Limulus amebocyte lysate test)為特定述於美國藥典(USP)第71及 151 章(USP23/NF 18,Biological Tests,The United States Pharmacopeial Convention,Rockville,MD,1995)。可替 代之熱原檢測已基於單細胞活化作用-細胞激素檢測發 展。已發展出用於品質控制應用之合宜均勻細胞系且已證 明其檢測在試樣中發熱性的能力,該試樣已通過兔熱原測 試及美洲鱟試驗法(Taktak等人之J_ Pharm. Pharmacol. (1990),43:578-82)。在又一實施例中,耳-可接受耳部治 療劑配方為獨立去熱原。在又一實施例中,製造耳-可接 受耳部治療劑配方的方法包含測試此配方的發熱性。在特 定實施例中,本文所述之配方實質無熱原。 pH及容積滲透濃度 本文使用之「實際容積滲透濃度」意指配方的容積滲透 濃度,其藉由包括活性劑與所有賦形劑但排除膠化及/或 增稠劑(如,聚氧乙烯-聚氧丙烯共聚合物、羧基曱基纖維 素或其相似者)而測量。本發明描述之配方的實際容積滲 透濃度可藉由任何合宜方法測量,如一凝固點下降方法, 如描述於 Viegas 等人著之 Int. J. Pharm·,1998,160,157-162。 本發明描述之耳配方的實際滲透壓為約100 mOsm/kg至約 1000 mOsm/kg,約 200 mOsm/kg至約 800 mOsm/kg,約 250 140447.doc -50- 200950787 mOsm/kg 至約 500 mOsm/kg,或約 250 mOsm/kg 至約 350 mOsm/kg或約 280 mOsm/kg至約 320 mOsm/kg。在某些實 施例中,本發明描述之配方具有實際容積滲透濃度為約 100 mOsm/L 至約 1000 mOsm/L,約 200 mOsm/L 至約 800 mOsm/L,約 250 mOsm/L至約 500 mOsm/L,約 250 mOsm/L 至約 350 mOsm/L,或約 280 mOsm/L至約 320 mOsm/L。 在某些實施例中,在作用之標的位置的容積滲透濃度 (如,周邊淋巴)為約相同於本發明描述之任何配方的可傳 送容積滲透濃度(即,材料通過或穿透圓窗膜的容積滲透 濃度)。在某些實施例中,本發明描述之配方的可傳送容 積滲透濃度為約150 mOsm/L至約500 mOsm/L,約250 mOsm/L至約 500 mOsm/L,約 250 mOsm/L至約 350 mOsm/L ’ 約 280 mOsm/L 至約 370 mOsm/L 或約 250 mOsm/L 至約 320 mOsm/L。 在内淋巴的主要存在的陽離子為鉀。此外,内淋巴具有 高濃度之正價胺基酸。在周邊淋巴的主要存在的陽離子為 鈉。在特定例子中,内淋巴及周邊淋巴的離子性組成物調 節毛細胞的電化學脈衝。在特定例子中,内淋巴或周邊淋 巴之離子性平衡的任何改變造成聽力喪失,此歸因於沿耳 部毛細胞的化學脈衝之傳導改變。在某些實施例中,本文 揭露之組成物未干擾周邊淋巴的離子性平衡性。在某些實 施例中,本文揭露之組成物具有相同或實質相同於周邊淋 巴之離子性平衡。在某些實施例中,本文揭露之組成物未 干擾内淋巴的離子性平衡。在某些實施例中,本文揭露之 140447.doc 200950787 組成物具有相同或實質相同於内淋巴的離子性平衡。在某 些實施例中,調配本文所述之耳部配方以提供離子性^ 衡,其與内耳液體(亦即,内淋巴及/或周邊淋巴)相容。 内淋巴及周邊淋巴具有的抑接近止液之生理pH。内淋 巴具有約7.2-7.9的pH範圍;周邊淋巴具有約72_74的邱 範圍。相鄰内淋巴之原位pH為約74而遠離内淋巴 約 7.9。 在某些實施例中,本文所述組成物之pH調整(例如,使Industry Manufacturers Association, 1981) and involves the challenge of membrane filters with unusual amounts of microbes in a known amount (about 107/cm2), such as with Brevendomonas diminuta (ATCC 19146) 0 pharmaceutical composition, optionally by membrane mesh Sterilize. Formulations containing nanoparticles (U.S. Patent No. 6,139,870) or multi-layered pores (Richard et al., International Journal of Pharmaceutics (2006), 312(1-2)··144-50) are suitable for use by Ο·22 μπι The filter is filtered without damaging the sterilization of its tissue structure. In certain embodiments, the methods disclosed herein comprise sterilizing the formulation (or a component thereof) by filtration sterilization. In another embodiment, the ear-acceptable ear therapeutic formulation comprises a particle, wherein the particle formulation is suitable for filtration sterilization. In yet another embodiment, the particle formulation comprises particles having a particle size of less than 300 nm, particles less than 200 nm in size, and particles less than 100 nm. In another embodiment, the ear-acceptable ear treatment formulation comprises a particle wherein the sterility of the particle is ensured by sterile filtration of the component solution of the precursor material. In another embodiment, the ear-acceptable formulation comprises a particle formulation wherein the sterility of the particles is ensured by cryogenic sterilization. In yet another embodiment, the cryogenic sterilization filter is between 〇 and 3 (between TC temperatures or between 〇 and 2〇°C) or between 0 and i〇°c, or between 10 and Between 20 ° C, or between 20 and 30 ° C. In another embodiment is a method of preparing an ear-acceptable particle formulation comprising: filtering an aqueous solution containing a particle formulation at a low temperature via a sterile filter; Drying the sterilization solution; and reconstituting the particle formulation with sterile water prior to administration 140447.doc -43. 200950787 In another embodiment. In certain embodiments, one of the formulations described herein is a single vial containing micronization. A suspension of active pharmaceutical ingredients in a formulation. A single vial formulation is aseptically mixed with a sterile poloxamer solution and a sterile micronized active ingredient (eg, methyl dehydrocorticosterol) and delivered to the sterility Prepared in a pharmaceutical container. In certain embodiments, a single vial containing a formulation described herein is resuspended prior to dispensing and/or administration. In a particular embodiment, the filtration and/or filling steps are below this disclosure. Gel temperature of the formulation T gel) about 5. (under: the gel formulation has a viscosity of less than the theoretical value of 100 cP to allow for a reasonable time transition using a peristaltic pump. In still other embodiments, the ear - acceptable The otic therapeutic formulation comprises a nanoparticle formulation wherein the nanoparticle formulation is suitable for filter sterilization. In yet another embodiment, the nanoparticle formulation comprises a nanoparticle size of less than 300 nm 'the size is less than 200. Nm, or less than 1 〇〇 nm in size. In still other embodiments, the 'ear-acceptable formulation comprises a microsphere formulation wherein the sterility of the microspheres can be sterilized by filtration of an organic solution of the precursor material and an aqueous solution. In still other embodiments, the ear-acceptable formulation comprises a thermoreversible gel formulation wherein the sterility of the gel formulation is ensured by cryogenic sterilization filtration. In yet another embodiment, the cryostat filtration is Between 〇 and 30. (: between temperatures 'either between 〇 and 2〇〇c, or between 〇 and 10 C' or between 1〇 and 2〇. (3, or between 2〇 With 30. (: between. In other embodiments, one A preparation method for a thermoreversible gel formulation comprising: filtering an aqueous solution containing a thermoreversible gel component H0447.doc-44 - 200950787 via a sterile filter at a low temperature; lyophilizing the reversible gel formulation. Reconstitution of heat in the specific example, buffer solution) and separate, p parts are dissolved in a suitable carrier (eg, by a heat treatment, filtration, gamma irradiation) prior to administration. - separate liquid gel components) sterilized in a combined step (eg, the cooling mixture of the excipients are supplied, H or radiation). The two solutions are then separately sterilized to provide a final ear formulation. The traditionally used methods of 'bacteriolysis (eg, heat-up (eg, high) gamma radiation, filtration) in some examples result in polymer components in this formulation (eg, heat-curing, gelling, or mucoadhesive polymers) Irreversible degradation of the component and/or activator. In some instances, sterilization of the ear formulation by membrane filtration (e.g., 0.2 _ membrane) is not possible if the formulation contains a rocky visbreaking polymer that is kneeized during the passage. Accordingly, the present invention provides a method for sterilizing an ear formulation that prevents polymer, 'and injury (eg, heat curing and/or gelation and/or mucoadhesive polymer component) and/or activator at Degradation during sterilization. In certain embodiments, degradation of an activating agent (eg, any of the therapeutic ear agents described herein) can be utilized by the use of a particular range of buffer components in the formulation and a specific ratio of gelling agent in the formulation. Reduce or remove. In certain embodiments, the selection of a suitable gelling agent and/or thermoset polymer may allow for the sterilization of the formulations described herein by filtration. In certain embodiments, the use of a suitable thermoset polymer and a suitable copolymer (e.g., a gelling agent) in combination with a particular range of the formulation allows for high temperature sterilization of the formulations described herein and is substantially free of 140447.doc • 45· 200950787 Degradation of therapeutic agents or polymeric excipients. In a particular example, the present invention provides a method of sterilizing such that the formulation is subjected to final sterilization by high pressure dad without any loss of activator and/or excipients and/or polymer components during the depressing step. Substantially free of microorganisms and/or pyrogens. Microorganisms The present invention provides an oat-acceptable composition for ameliorating or ameliorating the otic disorders described herein. A method of administration comprising the otic composition is further provided. In certain embodiments, the composition is substantially free of microorganisms. The acceptable amount of sterility is a standard of administration based on a defined therapeutically acceptable ear composition, which includes, but is not limited to, the United States Pharmacopoeia Chapter 1 11 i. For example, the acceptable amount of sterility includes 10 colony forming units (cfu) per gram of formulation, 5 〇 cfu per gram of formulation, 100 c cf' per gram of formulation, 500 cfu per gram of formulation or 1 〇〇〇 cfu per gram of formulation. In addition, the amount of sterility acceptable includes the exclusion of specific adverse microbial agents. Exemplary disappointing microbial agents include, but are not limited to, Escherichia coli 'E_ coli, Salmonella sp., pseudodom〇nas aeruginosa (P. aeruginosa), and/or other specific microbial agents. The sterility of the ear-acceptable otic therapeutic formulation can be determined by the procedures of the U.S. Pharmacopoeia <61>, <62> and <71>. One of the requirements for aseptic quality control assurance, quality assurance and effective methods is the method of sterility testing. The exemplification example of the sterility test is carried out in two ways. The first is direct incubation, in which a sample of the composition to be tested is added to the medium and incubated for a period of 2 i days. The turbidity of the growth medium showed contamination. Disadvantages of this method include the small sample size reduction sensitivity of a large number of materials and the detection of microbial growth based on visual inspection. An alternative method is the membrane filtration sterility test. In the 140447.doc • 46· 200950787 method, a volume of the product passes through a small membrane mesh paper. This screen paper is then placed in the medium to promote the growth of microorganisms. This method has the advantage of greater sensitivity when sampling all quality products. A commercially available Millipore Steritest Aseptic Test System can be optionally used for the determination of the membrane sterility test. For filtration testing of creams or ointments, use the Steritest Filtration System number TLHVSL210. For filtration testing of emulsions or viscous products, use Steritest Filtration System No. TLAREM210 or TDAREM210. For filtration testing of prefilled syringes, use Steritest Filtration System No. TTHASY210. For transition tests suspended in aerosol or foam materials, use Steritest; filtration system number TTHVA210. For filtration testing of soluble powder in ampoules or vials, use the Steritest Filtration System number TTHADA210 or TTHADV210. Tests for E. coli and Salmonella include incubation with lactose broth at 30-35 ° C for 24-72 hours, including incubation in MacConkey and/or EMB wean for 18-24 hours, and / or use Rappap0rt medium. Testing for Ρ· aeruginosa includes the use of NAC agar. Chapter 62 of the US Pharmacopoeia (USP) further cites the testing procedures for specific unpleasant micro-organisms. In a particular embodiment, any of the controlled release formulations described herein have less than about 60 colony forming units (Cfu) per gram of formulation, less than about 50 colony forming units, less than about 40 colony forming units, or less than about 3 A microbial agent that forms a unit of colony colonies. In a particular embodiment, the formulated ear formulation described herein is isotonic with the endolymph and/or peripheral lymph nodes. Endotoxin 140447.doc • 47· 200950787 The present invention provides an ear composition to improve or alleviate the otic disorders described herein. A method of administering the composition of the ear is further provided. In certain embodiments the composition is substantially endotoxin free. An additional aspect of a sterilization process is the removal of by-products (hereinafter referred to as "products") derived from killing microorganisms. The method of depyrogenation removes pyrogen from the sample. The pyrogen is an endotoxin or exotoxin that will elicit an immune response. An example of an endotoxin is a lipopolysaccharide (LPS) molecule found on the cell wall of a Gram-negative bacterium. When a sterilization process such as autoclaving or treatment with ethylene oxide to kill bacteria, the LPS residue induces a pre-inflammatory response, such as septic shock. Because the molecular size of endotoxin is very broad, the presence of endotoxin is expressed as "endotoxin unit" (EU). β - EU is equivalent to 100 picograms of E. coll LPS. Humans can generate a response that is less than 5 EU/kg of body weight. This sterility is expressed in any unit that is known in the art. In a particular embodiment, the otic compositions described herein contain a lower amount of endotoxin (e.g., <4 EU/kg body weight) when compared to conventional acceptable levels of endotoxin (e.g., 5 EU/kg body weight). In certain embodiments, in certain embodiments, the auris-acceptable otic therapeutic formulation has less than about 5 EU/kg body weight of the individual. In other embodiments, the ear-acceptable otic therapeutic formulation has less than about 4 EU/kg body weight of the individual. In additional embodiments, the ear-accommodating and otic therapeutic formulation has less than about 3 EU/kg body weight of the individual. In additional embodiments, the ear-acceptable otic therapeutic formulation has less than about 2 EU/kg body weight of the individual. In certain embodiments, the ear-acceptable otic therapeutic formulation has less than about 5 EU/kg of the formulation. In other embodiments, the ear-acceptable otic therapeutic formulation has less than about 4 Eu/kg of the formulation. In an additional embodiment 140447.doc 200950787 the ear can be attached to the ear. The P treatment formulation has less than about 3 of the formula. In certain embodiments, the ear-acceptable otic therapeutic formulation has less than about 5 EU/kg at the time of delivery. In other embodiments, the ear-acceptable ear treatment formulation has less than about 1 EU/kg of product. In additional embodiments, the ear-acceptable otic therapeutic formulation has less than about G.2 EU/kg of the product. f In certain embodiments, the ear-acceptable otic therapeutic formulation has less than ^ 兀 or about 5 EU/kg of the product. In other embodiments, the ear-acceptable 0 sputum therapeutic formulation has less than about 4 EU/kg of the unit or product. In still other embodiments, the ear-acceptable otic therapeutic formulation has less than a unit or yields about 3 EU/kg. In certain embodiments, the ear-acceptable otic therapeutic formulation has less than about 5 Eu/kg of the unit or product. In other embodiments, the ear-acceptable otic therapeutic formulation has less than about 4 EU/kg of the unit or product. In other embodiments, the ear-acceptable otic therapeutic formulation has less than about 3 EU/kg of the unit or product. In a particular embodiment, the disclosed ear compositions contain from about 1 to about 5 EU/mL of the formulation. In a particular embodiment, the disclosed ear compositions comprise from about 2 to about 5 EU/mL of the formulation, from about 3 to about 5 EU/mL of the formulation, or from about 4 to about 5 EU of the formulation. mL. In a particular embodiment, the ear composition 18 described herein contains a lower endotoxin amount than a conventional acceptable amount of endotoxin (e.g., 0.5 EU/mL of the formulation) (e.g. < EU/mL). In certain embodiments, the ear-accessible ear therapeutic formulation has less than about 0.5 EU/mL of the formulation. In other embodiments, the ear-acceptable otic therapeutic formulation has less than about 0.4 EU/mL of the formulation. In additional embodiments, the ear-acceptable ear 140447.doc -49- 200950787 therapeutic formulation has less than about 0.2 EU/mL of the formulation. As an example, pyrogen detection is performed by several methods. Suitable tests for sterility include those described in the United States Pharmacopoeia (USP) Chapter 71 Asperity Test (23rd Edition, 1995). The rabbit pyrogen test and the Limulus amebocyte lysate test are described in the USP 23/151 (USP23/NF 18, Biological Tests, The United States Pharmacopeial Convention, Rockville, MD, 1995). . Alternative pyrogen detection has been based on single cell activation - cytokine detection. A suitable homogeneous cell line for quality control applications has been developed and demonstrated its ability to detect exothermicity in a sample that has passed the rabbit pyrogen test and the American cockroach test method (Taktak et al. J_Pharm. Pharmacol) (1990), 43:578-82). In yet another embodiment, the ear-acceptable ear treatment formulation is an independent depyrogen. In yet another embodiment, a method of making an ear-acceptable ear treatment formulation comprises testing the fecality of the formulation. In a particular embodiment, the formulations described herein are substantially pyrogen free. pH and Volume Osmotic Concentration As used herein, "actual volume osmolality" means the volumetric osmolality of a formulation by including the active agent with all excipients but excluding gelling and/or thickening agents (eg, polyoxyethylene- Measured by polyoxypropylene copolymer, carboxymercaptocellulose or the like. The actual volumetric permeation concentration of the formulations described herein can be measured by any convenient method, such as a freezing point depressing method, as described in Viegas et al., Int. J. Pharm, 1998, 160, 157-162. The actual osmotic pressure of the ear formulations described herein is from about 100 mOsm/kg to about 1000 mOsm/kg, from about 200 mOsm/kg to about 800 mOsm/kg, from about 250 140447.doc -50 to 200950787 mOsm/kg to about 500. mOsm/kg, or from about 250 mOsm/kg to about 350 mOsm/kg or from about 280 mOsm/kg to about 320 mOsm/kg. In certain embodiments, the formulations described herein have an actual volumetric osmolality of from about 100 mOsm/L to about 1000 mOsm/L, from about 200 mOsm/L to about 800 mOsm/L, from about 250 mOsm/L to about 500. mOsm/L, from about 250 mOsm/L to about 350 mOsm/L, or from about 280 mOsm/L to about 320 mOsm/L. In certain embodiments, the volumetric osmolality (eg, peripheral lymph) at the site of action is about the same as the deliverable volume osmotic concentration of any of the formulations described herein (ie, the material passes through or penetrates the round window membrane) Volume osmotic concentration). In certain embodiments, the formulations described herein have a deliverable volume osmotic concentration of from about 150 mOsm/L to about 500 mOsm/L, from about 250 mOsm/L to about 500 mOsm/L, from about 250 mOsm/L to about 350 mOsm/L 'about 280 mOsm/L to about 370 mOsm/L or about 250 mOsm/L to about 320 mOsm/L. The main cation present in the endolymph is potassium. In addition, the endolymph has a high concentration of a positive valency amino acid. The main cation present in the peripheral lymph is sodium. In a particular example, the ionic composition of the endolymph and peripheral lymph nodes regulates the electrochemical pulse of the hair cells. In a particular example, any change in the ionic balance of endolymph or peripheral lymphocytes results in hearing loss due to conduction changes in chemical pulses along the hair cells of the ear. In certain embodiments, the compositions disclosed herein do not interfere with the ionic balance of the peripheral lymph. In certain embodiments, the compositions disclosed herein have the same or substantially the same ionic balance as the peripheral lymph. In certain embodiments, the compositions disclosed herein do not interfere with the ionic balance of the endolymph. In certain embodiments, the 140447.doc 200950787 compositions disclosed herein have the same or substantially the same ionic balance as the endolymph. In some embodiments, the otic formulations described herein are formulated to provide an ionic balance that is compatible with the inner ear fluid (i.e., the endolymph and/or peripheral lymph). The endolymph and peripheral lymph nodes have a physiological pH close to that of the stop solution. The inner lymph has a pH range of about 7.2-7.9; the peripheral lymph has a range of about 72-74. The in situ pH of the adjacent endolymph is about 74 and is about 7.9 away from the endolymph. In certain embodiments, the pH adjustment of the compositions described herein (eg,

用-缓衝液)至一内淋巴-可相容之約55至9〇的阳範圍。QUse -buffer) to an endolymph-compatible range of about 55 to 9 angstroms. Q

在特定實施例中,本文所述組成物之阳調整至約5.5至U 的PH範圍。在某些實施例中,本文所述組成物之阳調整 至約6.0至7.6的pH範圍。 ,在某些實施例中,有用之配方亦包括—或—以上的阳 節劑或緩衝液。合宜之阳調㈣或緩衝液包括但未限制 為乙酸孤碳酸氫鹽、氣化銨、檸檬酸鹽、鱗酸鹽、其之 醫藥可接受鹽與其之組合或混合物在某些實施例中。 在實施例中,當在本發明揭露之配方中使用一或一卩 〇 上的緩衝劑時,其與如—醫藥可接受載劑組合且在最終I 方中的存在量如,約〇1%至約鳩,約G5%至約1G%間。 在本揭露之特定實施例中,包括在凝膠配方中緩衝劑的# ^ 為可使,膠配方的pH不會干擾身體的天然緩衝系統。 在-實施例中,因為稀釋劑可提供—更安定環境,亦使 用稀釋劑安定化合物。在此技中利用溶解在緩衝溶液中的 (/、’、長:供pH控制或維持)為稀釋劑,其包括但未限制為 140447.doc •52· 200950787 磷酸酯緩衝生理食鹽水溶液。 在某些實施例中,本文所述之任何凝膠配方具有一 pH 可利於凝膠配方的滅菌作用(例如藉由過濾或無菌混合或 熱處理及/或高壓釜(如,最終滅菌作用))而無藥劑(如, 類固醇)或含有凝膠聚合物的降解。為了在滅菌作用期間 減少耳劑及/或凝膠聚合物的水解及/或降解,設計緩衝劑 * 的滅菌作用期間維持此配方之pH在7-8範圍間(如,高 . 温高壓釜作用)。 . 〜 在特定貫施例中,本文所述之任何凝膠配方具有一 以允許凝膠配方的最終滅菌作用(例如,藉由熱處理及/或 高壓釜)而未降解含有凝膠的藥劑(如,皮質類固醇)或聚合 物。例如,為了在滅菌作用期間減少耳劑及/或凝膠聚合 物的水解及/或降解,設計緩衝劑的pH在增温時維持此配 方之pH在7-8範圍間。依在此配方中使用的耳劑使用任何 合宜之緩衝液。在某些例子中,因為當温度增加大約_〇〇3/艽 ❹ 時TRIS的pKa降低,當温度增加大約〇 〇〇3/〇c時卩則之卩心 增加度,在250°F(12rC )高壓釜導致在TRIS緩衝液中顯著 的pH向下移動(亦即更酸化),因此在pBS緩衝液中一相對 較少pH向上的移動,因此更增加耳劑在TRJS比在pBs中更 增加的水解及/或降解。耳劑的降解藉由使用本文所述的 適當之緩衝液與聚合物添加劑的組合而減少(例如p4〇7, CMC)。 在某些實施例中,介於約5.0與約9.0間,介於約5.5與約 8.5間,介於約6.〇與約7 6間,介於約7與7 8間介於約7〇 140447.doc 53- 200950787 與約7.6間,介於約7.2與7.6間,介於約72與74間的^^為 適宜用於本發明描述之配方的滅菌作用(例如藉由過濾或 無菌尾合或熱處理及/或高温加壓滅菌(例如,終滅菌作 用))。在特定的實施例中’為約6 〇、約6 5、約7 〇、約 7.1、約7.2、約7.3、約7.4、約7.5、或約7 6之配方pH為適 宜用於本發明描述之任何組成物之滅菌作用(例如藉由過 濾或無菌混合或熱處理及/或高温加壓滅菌(例如,終滅菌 作用))。 在某些實施例中,配方具有一如本文所述之pH,並包 括一增稠劑(亦即一黏性增進劑),其非限制例示如本文所 之纖維素系增稠劑。在某些例子中,第二聚合物(例如, 增稠劑)的加入及本發明所述之配方的pH允許本發明所述 之配方的滅菌作用而無造成在耳部配方中耳劑及/或聚合 物組份的實質降解。在某些實施例中,在一具有如本文所 述之pH的配方中熱可逆泊洛沙姆對增稠劑的比例為約 40:1、約 35:1、約 30:1、約 25:1、約 20:1、約 15:1 或約 10:1。例如’在特定實施例中’本文所述之持續及/或延長 釋放配方包含一以約40:1、約35:1、約30:1、約25:1、約 20:1、約1 5:1或約1 〇: 1比例之泊洛沙姆4〇7(pluronic F127) 及羧基甲基纖維素(CMC)組合。在某些實施例中,在任何 本文所述之配方中的熱可逆聚合物的量為配方之總重的約 10%、約15%、約20%、約25%、約30%、或約5%。在某些 實施例中’在任何本文所述之配方中的熱可逆聚合物的量 為配方之總重的約約14%、約15%、約16%、約17%、約 I40447.doc -54· 200950787 。% 約 19%、約 20%、約 21%、約 22%、約 23%、約 24% 3、 /〇在某些實施例中,在任何本文所述之配方中的 j稠劑(例如’凝膠舞"的量為配方之總重的約”/〇、5%、 、勺1〇/°或約15%。在某些實施例中,在任何本文所述之配 方f的増稠劑(例如,凝膠劑)的量為配方之總重的約 • 〇·5%、約1%、約 h5%、約 2%、約 2.5%、約 3%、約 3.5%、 • 約4%、約4.5%、或約5%。 'Φ 在某些實%例中,本文所述之醫藥相對於pH在任何下 列週期間為安定的方:至少約i天、至少約2天、至少約 3天、至少約4天、至少約5天、至少約6天、至少約】週、 至〉、約2週、至少約3週、至少約4週、至少約5週、至少約 6週至少約7週、至少約8週、至少約i個個月、至少約2 個個月、至少約3個個月、至少約4個個月、至少約5個個 月、或至少約6個個月。在其他實施例中,本文所述之配 方相對於在至少約1週期間為安定的。且,本文所述之配 φ 方相對於在至少約1個個月間為安定的。 張力劑 . 通常,内淋巴具有比周邊淋巴為較高的滲透壓。例如, 内淋巴具有約304 mOsm/kg H2〇之滲透壓,同時周邊淋巴 具有約294 mOsm/kg HW的滲透壓。在某些實施例中,調 配本文所述之耳組成物以提供一容積滲透濃度為約1〇〇至 約 1000 mOsm/kg,約 200 至約 800 m〇sm/kg,約 25〇 至約 500 mOsm/kg滲透壓;約250至約350 m〇sm/kg或約28〇至 約320 mOsm/kg滲透壓。在某些實施例中,本文所述之具 140447.doc -55· 200950787 有一實際容積滲透濃度為約100 m〇sm/L至約1000 mOsm/L,約 200 mOsm/L至約 800 mOsm/L,約 250 mOsm/L 至約 500 mOsm/L,約 250 mOsm/L至約 350 mOsm/L,或約 280 mOsm/L至約 320 mOsm/L。 在某些實施例中,設計本文所述之任何配方的傳送之容 積滲透濃度與標的耳結構(如,内淋巴、周邊淋巴或其相 似者)為等張。在特定實施例中,配製本文所述之耳組成 物以提供傳送之容積滲透濃度在用的標的位置為約250至 約 320 mOsm/L(為約 250至約 320 mOsm/kg H20之滲透壓); 且較佳為約270至約320 mOsm/L(約270至約320 mOsm/kg H20之滲透壓)。在特定實施例中,該配方的傳送容積滲透 濃度/滲透壓(即,在無膠化或增稠劑(如,熱可逆凝膠聚合 物)之配方的容積滲透濃度/滲透壓)例如藉由使用適當的鹽 濃度(如,鉀或鈉鹽濃度)或使用等張劑調整,以使此當傳 送至標的位置時為内淋巴-可相容及/或周邊淋巴-可相容 (即與内淋巴及/或周邊淋巴等張)。含有熱可逆凝膠聚合物 的配方之容積滲透濃度為一不可靠的方式,因為伴隨水與 聚合物之單體單元的變化量。此配方的實際容積滲透濃度 (即,在無膠化或增稠劑(如,熱可逆凝膠聚合物)之容積滲 透濃度)為一可信賴的量測且可藉由任何合宜的方法(如, 凝固點下降方法)測量。在某些例子中,本文描述的配方 提供造成對内耳環境最小干擾且當投藥時造成哺乳動物的 最小不適(如,暈眩及/或噁心)之可傳送的容積滲透濃度 (如,至一標的位置(如,周邊淋巴))。 140447.doc -56- 200950787 在某些實施例中,在本文中所述的任何配方為與周邊淋 巴及/或内淋巴等張。藉由加入一等張劑而提供等張配 方。適宜的等張劑包括但不限制為任何醫藥可接受糖、鹽 或其等之組合或混合物’如但未限制於葡萄糖'甘油、甘 露醇、山梨糖醇、氣化納及其他電解質。 有效的耳組成物包括一或一以上的鹽,其量為引導組成 物的滲透壓至一可接受範圍所需要的量》此鹽包括具有 _ 鈉、鉀或銨陽離子與氣化物、檸檬酸鹽、抗壞血酸鹽、硼 酸鹽、磷酸鹽、碳酸氫鹽,硫酸鹽,硫代硫酸鹽或亞硫酸 氫鹽陰離子;合宜之鹽包括氯化鈉、氯化鉀、硫化碳酸 鈉、硫酸氫鈉及硫酸銨。 在某些實施例中,本文所述之配方具有如本文所述之的 pH及谷積滲透濃度,與具有介於約丨μΜ與約丨〇 間,介 於約1 mM與約10〇mM間,介於間約〇 ! mM與約1〇〇mM, 介於約0.1 mM與約1〇〇 nM之活性醫藥成份濃度。在某些 ❹ 實施例中,本文所述之配方具有如本文所述之的pH及容 積滲透濃度,且具有一活性醫藥成份濃度為配方重量之介 • 於約0·1-約20%間,介於約0.1-約10%間,介於約o.i-約 7.5%間,介於約〇· 1 _5%間,介於約〇 2_約3%間,介於約 0.1-約2%間的活性成份。在某些實施例中,本文所述之配 方具有如本文所述之的pH及容積滲透濃度,且具有一活 性醫藥成份濃度為配方體積之〇丨_約7〇 mg/mL間,介於約 1 mg-約70 mg/mL間,介於約i „^_約5〇 mg/mL間,介於 約1 mg/mL與約20 mg/mL間,介於約! mg/mL至約1〇 140447.doc •57- 200950787 mg/mL間,介於約! mg/mL至約5吨/灿間,或介於約〇5 mg/mL至約5 mg/mL間之活化劑。 粒子大小 使用降低大小以增加表面積及/或調變配方溶解性質。 亦用以維持本文所述之任何配方的一致平均粒子大小分佈 (PSD)(例如,微米大小粒子,奈米·大小粒子或其才刀目似 者)。在某些例子中,本文所述之任何配方包含多重粒 子,亦即,複數個粒子大小(例如,微米粒子,奈米-大小 粒子,無-大小粒子、膠體粒子);亦即,此配方為多重粒 子配方。在某些貫施例中,本文所述之任何配方包含一或 一以上之多重粒子(例如,微米)治療劑。微米作用為減少 固體物質之粒子的平均直徑的方法。微米粒子由直徑為約 微米-大小至直徑為約奈米-大小。在某些實施例中,微米 化固體之粒子的平均直徑在約0.5 μϊη至約5〇〇 。在某 些實施例中,微米化固體之粒子的平均直徑在約i μη1至約 1 00 μηι間。在某些實施例中,微米化固體之粒子的平均直 徑在約2 μΐη至約1〇〇 μη^Β]。在某些實施例中,微米化固體 之粒子的平均直徑在約3 μιη至約5〇 。在某些實施例 中,一粒子之微米化固體含有少於約5微米,少於約2〇微 米及/或少於約1 00微米的粒子大小。在某些實施例中,皮 質類固醇的粒子(例如,微米化粒子)使用允許本文所述之 任何配方之皮質類固醇相對於含有非_多重粒子(例如,無 微米化)皮質類固醇耳劑配方為延長及/或持續釋放。在某 些例子中,含有多重粒子(例如微米)皮質類固醇的配方由 140447.doc -58- 200950787 適於27G針之lmL注射而無任何堵塞或結塊。 在某些例子中,本文所述之任何配方的任何粒子為—塗 覆粒子(例如,一塗覆的微米粒子、奈米粒子)及/或一微球 體及/或一脂質體粒子。粒子大小降低技術包括之例示為 研磨、磨粉(例如,空氣磨耗研磨機(噴霧研磨機)、球 . 磨)、膠粒凝聚、高壓均質作用、噴霧乾燥及/或超臨界值 • 液體結晶。在某些例子中’粒子大小化係藉由機械衝擊 φ (例如,藉由鍵碎機、球磨機及/或梢研磨機)完成。在某些 例子中’粒子尺寸大小經液態能量(例如’藉由螺旋噴霧 研磨機、迴圈噴霧研磨機、及/或流體化床喷霧研磨機)。 在某些實施例中,本文所述之配方包含結晶粒子及/或等 向粒子。在某些實施例中,本文所述之配方包含非晶形粒 子及7或非等向粒子。在某些實施例中,本文所述之配方 包含治療劑粒子,其中該治療劑為一自由驗、或治療劑之 鹽或前藥、或其等之任何組合。 • 在某些實施例令,本文所述之配方包含一或一以上的皮 • f類固醇’其中該皮質類固酵包含奈米粒子。在某些實施 • 例中,本文所述之配方包含一視情況以控制釋放賦形劑塗 • 覆的皮質類固醇珠(如,甲基脫氫皮質固醇珠p在某些實 鉍例中,本文所述之配方包含為粒狀及/或減小大小且以 控制釋放職形劑塗覆之皮質類固醇;此顆粒態塗覆皮質類 固醇之粒子接著可視情況微米化及/或以任何本文所述之 組成物配製。 在某些例子中,使用為自由酸或自由鹼之皮質類固醇與 140447.doc •59- 200950787 -皮質類固醇鹽組合依本文所述之製程製備脈衝釋放耳劑 配方:在某些配方中,一微米化皮質類固醇(及/或其之鹽 或刖藥)與塗覆之粒子(例如奈米粒子、脂質球、微球體)的 組合為使用本文所述之任何製程製備脈衝釋放耳劑配方。 或者,一脈衝釋放曲線可藉由在環糊精、界面活性劑(例 如,泊洛沙姆(407、338、188)、Tween(8〇、6〇、2〇、川、 PEG·虱化說蔚油、共溶劑如N_甲基_2吼略咬嗣或其相似 者幫助下洛解面達20%的傳送劑量之皮質類固醇⑽如,微 米化皮質類固SI、或其等之自由醇、自由酸或鹽或前藥;· :粒子皮質類固醇、或其等之自由鹼、自由酸或鹽或前 藥)而獲得,其使用本文所述之任何製程製備脈衝釋放配 方。 在特定實施例中,本文所述之任何耳可相容的配方包含 一或—以上的微米化藥劑(如,類固醇)。在某些此種實施 例中,一微米化的藥劑包含微米化粒子、經塗覆(如,以 延長釋出塗層)微米化粒子或其等之組合。在某些此種實 施例中’ 一包含微米化粒子、經塗覆(如以延長釋出塗 Q 層)微米化粒子或其等之組合的微米化的藥劑包含—皮質 .. =固醇為自由酸、自由驗、—鹽、一前藥或其等之任何组 0、。在特定的實施例中,本文所述之一醫藥組成物包含為 微2化粉末的曱基脫氫皮質固醇、曱基去氫皮質醇或去氫 =貝醇。在特定實施例中,本文所述之一醫藥組成物包 為微甲基脫氫皮質固醇粉末的曱基脫氫皮質固醇。 本文所述之多顆粒及/或微米化之皮質類固醇藉由任何 140447.doc -60- 200950787 型式的包含固體、液體或凝膠基體之基體傳送至一耳結構 (如’内耳)。在某些實施财’本文所述之多顆粒及/或微 米化之皮質類固醇藉由任何型式的包含固體、液體或凝膠 基體之基體經由耳内注射傳送至一耳結構(如,内耳)。" 醫藥配方 . I文提供之醫藥組成物包括至少—纟質類固醇及_醫藥 .· 彳接受稀釋劑、賦形劑或載劑。在某些實施例中,此醫藥 0 組成物包括其他藥或醫藥劑、載劑、佐劑,如保存劑、安 定劑、濕化劑或乳化劑、溶液促進劑及調節滲透壓之鹽、 及/或緩衝液。在其他實施例中,此醫藥組成物亦含有其 他治療物質。 在本文所述之耳可接受控制釋放皮質類固醇醫藥配方的 一特定實施例中,此皮質類固醇於一凝膠基體中提供,且 本文亦稱為「耳可接受凝膠配方」、「内耳可接受凝膠配 方」、「中耳可接受凝膠配方」、「耳可接受凝膠配方」、「耳 9 凝膠配方」或其等之變化。此凝膠配方的所有成份必需與 標的耳結構相容。再者,此凝膠配方在標的耳結構内提供 • 皮質類固醇的控制釋放至預期位置;在某些實施例中,此 - 凝膠配方亦具有一立即或快速釋放組份以利於皮質類固醇 傳送至預期位置。在其他實施例中,此凝膠配方具有一持 續釋放成份用於皮質類固醇的傳送。在某些實施例中,凝 膠配方包含一多顆粒(如,微米化)皮質類固醇。在某些實 施例中,此耳凝膠配方為可生物降解的。在其他實施例 中,此耳凝膠配方包括一黏膜黏附劑賦形劑以允許黏附至 140447.doc -61 - 200950787 圓窗膜的外黏膜層。在又一其他實施例中,此耳凝膠配方 包括一穿透增進劑賦形劑;在又一實施例,此耳凝膠配方 s有黏丨生增進劑,其足以提供一黏性為介於約5〇〇至 ι,οοο,οοο厘泊間’介於約75〇至1〇〇〇〇〇〇厘泊Μ ;介於約 1000及400,000厘泊間;介於約2〇〇〇至1〇〇〇〇〇厘泊間;介 於約3000至50,〇〇〇厘泊間;介於約4〇〇〇至25〇〇〇厘泊間;介 於約5000至20,〇〇〇厘泊間;或介於約6〇〇〇至15〇〇〇厘泊間。 在另外或可替代的實施例中,此耳凝膠配方能夠經耳内 注體投藥至圓窗膜或接近圓窗膜。在其他實施例中,此耳 凝膠配方為經後耳後切口及外科手術投藥至圓窗膜或接近 圓窗膜或耳蜗窩冠部區域。可替代地,此耳凝膠配方以針 及注射器施用,其中針插人耳膜並導引至圓窗媒或耳蜗寫 冠部的區域。此耳凝膠配方接著沉積於圓窗膜上或接近圓 窗膜或耳堝寓冠部以局部治療自體免疫耳部失調。在其他 實施例中,此耳凝膠配方由移植入患者的導管施用,且在 再一實施例中,此配方由泵施用至或接近圓窗膜。在仍為 實施例巾’此耳凝膠配方由微注射裝置㈣至或接近圓窗 膜。在又其他的實施例中,此耳凝膠配方施用至耳穴中。 在仍為實施例中,此耳凝膠配方施用耳膜。在仍為實施例 中,此耳凝膠配方施用在耳道上或耳道内。 在又-特別實施例中,本文所描述之任何醫藥組成物包 含一在液體基體中的多粒子皮質類固醇(如,—用於耳内 注射或耳滴劑的液體組成物)。在特定實施例中,本文所 描述之任何醫藥組成物包含—在固體基體中的多粒子皮質 I40447.doc •62- 200950787 類固醇。 控制釋放配方 通常’控制釋放藥配方賦予在身體内相對釋放位置與釋 放時間控制藥釋放。如本文討論,控制釋放為延遲釋放、 延長釋放、持續釋放及/或脈衝釋放(例如,延長釋放與立 即釋放的組合)或其等之組合。藉由控制釋放可提供許多 ' 優點。第一,醫藥劑之控制釋放允許較少給藥頻率且因此 Φ 使重複治療最少化。第二,控制釋放治療造成更有效的藥 利用且較少的化合物殘留。第三,控制釋放藉由在疾病位 置放置傳遞裝置或配方以提供局部藥傳遞的可能性。再 者,控制釋放可經由單一劑量單元提供投藥及釋放至少二 不同藥,每一者具有一獨特的釋放曲線,或在不同速率或 不同期間釋放相同的藥。 因此,本文所描述之一實施例的態樣為提供用於治療自 體免疫失調及/或發炎失調的控制釋放皮質類固醇的耳可 © 接受組成物或配方。本文所揭露之組成物及/或配方的控 ㈣放態樣係經由多種試劑所賦予,包括但未限制為可接 • 受用於内或其化耳結構的賦形劑、試劑或材料。 耳-可接受凝膠 凝膠,有時稱為康膠’其已由多種方式定義。例如,美 國藥典定義凝谬為一半固體系統,其係由小無機粒子或較 大有機分子由-液體内穿透组成的懸浮液。凝膠可更由單In a particular embodiment, the cation of the compositions described herein is adjusted to a pH range of about 5.5 to U. In certain embodiments, the cation of the compositions described herein is adjusted to a pH range of about 6.0 to 7.6. In some embodiments, useful formulations also include - or - a cation or buffer. Suitable cations (iv) or buffers include, but are not limited to, lysine acetate, ammonium sulphate, citrate, sulphate, pharmaceutically acceptable salts thereof, combinations or mixtures thereof, in certain embodiments. In an embodiment, when one or more buffers are used in the formulations disclosed herein, they are combined with, for example, a pharmaceutically acceptable carrier and are present in the final I, for example, about 1%. To about 鸠, about G5% to about 1G%. In a particular embodiment of the present disclosure, the #^ included in the gel formulation is such that the pH of the gel formulation does not interfere with the body's natural buffer system. In the examples, the diluent is used to stabilize the compound because the diluent can provide a more stable environment. In this technique, a diluent (/, ', long: for pH control or maintenance) dissolved in a buffer solution is used, which includes but is not limited to 140447.doc • 52·200950787 Phosphate buffered physiological saline solution. In certain embodiments, any of the gel formulations described herein have a pH that facilitates sterilization of the gel formulation (eg, by filtration or aseptic mixing or heat treatment and/or autoclave (eg, terminal sterilization)). No drug (eg, steroid) or degradation with a gel polymer. In order to reduce the hydrolysis and/or degradation of the otic agent and/or gel polymer during sterilization, the pH of the formulation is maintained during the sterilization of the design buffer* between 7 and 8 (eg, high temperature autoclave action) ). ~ In a particular embodiment, any of the gel formulations described herein have an agent that allows for the final sterilization of the gel formulation (eg, by heat treatment and/or autoclave) without degrading the gel-containing agent (eg, , corticosteroids) or polymers. For example, to reduce hydrolysis and/or degradation of the otic agent and/or gel polymer during sterilization, the pH of the buffer is designed to maintain the pH of the formulation between 7-8 when warming. Use any suitable buffer for the ear used in this formulation. In some cases, because the pKa of the TRIS decreases as the temperature increases by approximately _〇〇3/艽❹, the increase in the enthalpy of the 当 when the temperature increases by approximately 〇〇〇3/〇c, at 250°F (12rC) The autoclave causes a significant pH shift (ie, more acidification) in the TRIS buffer, so a relatively small pH shift in the pBS buffer, thus increasing the ear agent more in the TRJS than in the pBs Hydrolysis and/or degradation. Degradation of the otic agent is reduced by the use of a suitable buffer as described herein in combination with a polymeric additive (e.g., p4 〇 7, CMC). In certain embodiments, between about 5.0 and about 9.0, between about 5.5 and about 8.5, between about 6. and about 76, between about 7 and 7 between about 7 140447.doc 53- 200950787 and about 7.6, between about 7.2 and 7.6, between about 72 and 74 are suitable for sterilization of the formulations described herein (eg by filtration or sterile tailing) Or heat treatment and / or high temperature pressure sterilization (for example, terminal sterilization). In a particular embodiment, a formulation having a pH of about 6 〇, about 65, about 7 〇, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, or about 7.6 is suitable for use in the description of the present invention. Sterilization of any composition (for example by filtration or aseptic mixing or heat treatment and/or high temperature pressure sterilization (eg, terminal sterilization)). In certain embodiments, the formulation has a pH as described herein and includes a thickening agent (i.e., a viscous enhancer) which is, without limitation, a cellulosic thickener as described herein. In certain instances, the addition of a second polymer (e.g., a thickener) and the pH of the formulation of the present invention allow for the sterilization of the formulations described herein without causing earrings in the ear formulation and/or Or substantial degradation of the polymer component. In certain embodiments, the ratio of thermally reversible poloxamer to thickener in a formulation having a pH as described herein is about 40:1, about 35:1, about 30:1, about 25: 1. About 20:1, about 15:1 or about 10:1. For example, 'in a particular embodiment' the sustained and/or extended release formulation described herein comprises a ratio of about 40:1, about 35:1, about 30:1, about 25:1, about 20:1, about 15 : 1 or about 1 〇: 1 ratio of ploxamic 4 〇 7 (pluronic F127) and carboxymethyl cellulose (CMC) combination. In certain embodiments, the amount of thermoreversible polymer in any of the formulations described herein is about 10%, about 15%, about 20%, about 25%, about 30%, or about the total weight of the formulation. 5%. In certain embodiments, the amount of thermoreversible polymer in any of the formulations described herein is about 14%, about 15%, about 16%, about 17%, about I40447.doc - of the total weight of the formulation. 54· 200950787. % about 19%, about 20%, about 21%, about 22%, about 23%, about 24% 3, / in certain embodiments, a thickening agent in any of the formulations described herein (eg ' The amount of gel dance " is about 〇, 5%, 勺1〇/° or about 15% of the total weight of the formula. In certain embodiments, the formula f of any of the formulations described herein is thick. The amount of the agent (e.g., gel) is about 5%·5%, about 1%, about h5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4% of the total weight of the formulation. %, about 4.5%, or about 5%. 'Φ In some real cases, the pharmaceuticals described herein are stable relative to pH over any of the following periods: at least about i days, at least about 2 days, at least About 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 5 weeks, to >, about 2 weeks, at least about 3 weeks, at least about 4 weeks, at least about 5 weeks, at least about 6 weeks At least about 7 weeks, at least about 8 weeks, at least about i months, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, or at least about 6 Months. In other embodiments, the formulations described herein are relative to at least It is stable for 1 week. Moreover, the φ side described herein is stable relative to at least about 1 month. Tension agent. Usually, the endolymph has a higher osmotic pressure than the peripheral lymph. For example, The lymph has an osmotic pressure of about 304 mOsm/kg H2, while the peripheral lymph has an osmotic pressure of about 294 mOsm/kg HW. In certain embodiments, the ear compositions described herein are formulated to provide a volumetric osmotic concentration of about 1 〇〇 to about 1000 mOsm/kg, about 200 to about 800 m〇sm/kg, about 25 〇 to about 500 mOsm/kg osmotic pressure; about 250 to about 350 m 〇sm/kg or about 28 〇 to about 320 mOsm/kg osmotic pressure. In certain embodiments, 140447.doc -55· 200950787 described herein has an actual volumetric osmotic concentration of from about 100 m〇sm/L to about 1000 mOsm/L, about 200 mOsm/L. Up to about 800 mOsm/L, from about 250 mOsm/L to about 500 mOsm/L, from about 250 mOsm/L to about 350 mOsm/L, or from about 280 mOsm/L to about 320 mOsm/L. In certain embodiments , the volume osmotic concentration of the delivered design of any of the formulations described herein is the same as the target otic structure (eg, endolymph, peripheral lymph, or the like) In particular embodiments, the ear compositions described herein are formulated to provide a volumetric osmotic concentration for delivery at a target position of from about 250 to about 320 mOsm/L (from about 250 to about 320 mOsm/kg H20). Osmotic pressure; and preferably from about 270 to about 320 mOsm/L (osmotic pressure of from about 270 to about 320 mOsm/kg H20). In a particular embodiment, the delivery volume osmotic concentration/osmotic pressure of the formulation (ie, the volume osmotic concentration/osmotic pressure of the formulation without gelling or thickening agent (eg, thermoreversible gel polymer)) is utilized, for example, by Use an appropriate salt concentration (eg, potassium or sodium salt concentration) or use an isotonic agent to adjust for endolymph-compatible and/or peripheral lymph-compatible (ie, internal) when delivered to the target location Lymphatic and / or peripheral lymphatic isotonic). The volumetric osmolality of a formulation containing a thermoreversible gel polymer is an unreliable manner because of the amount of change in monomer units accompanying water and polymer. The actual volumetric osmolality of the formulation (i.e., the volumetric osmolality of the gel-free or thickener (e.g., thermoreversible gel polymer)) is a reliable measure and can be by any convenient method (e.g., , freezing point drop method) measurement. In certain instances, the formulations described herein provide a deliverable volume osmotic concentration that causes minimal disruption to the inner ear environment and causes minimal discomfort (eg, dizziness and/or nausea) in the mammal when administered (eg, to a target) Location (eg, peripheral lymph)). 140447.doc -56- 200950787 In certain embodiments, any of the formulations described herein are isotonic with peripheral lymphocytes and/or endolymph. Isotonic formulations are provided by the addition of an isotonic agent. Suitable isotonic agents include, but are not limited to, any pharmaceutically acceptable sugar, salt or combination or mixture thereof such as, but not limited to, glucose 'glycerol, mannitol, sorbitol, gasified sodium, and other electrolytes. An effective ear composition includes one or more salts in an amount necessary to direct the osmotic pressure of the composition to an acceptable range. The salt includes sodium, potassium or ammonium cations and vapors, citrate , ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anion; suitable salts include sodium chloride, potassium chloride, sodium carbonate, sodium hydrogen sulfate and ammonium sulfate . In certain embodiments, the formulations described herein have a pH and grain osmotic concentration as described herein, and have a relationship between about 丨μΜ and about 丨〇, between about 1 mM and about 10 〇 mM. , between about mM and about 1 mM, between about 0.1 mM and about 1 〇〇 nM of active pharmaceutical ingredient concentration. In certain embodiments, the formulations described herein have a pH and volume osmotic concentration as described herein, and have an active pharmaceutical ingredient concentration of from about 0. 1 to about 20% by weight of the formulation. Between about 0.1 and about 10%, between about oi and about 7.5%, between about 〇·1 _5%, between about _2_about 3%, between about 0.1-about 2% Active ingredient. In certain embodiments, the formulations described herein have a pH and volume osmotic concentration as described herein, and have an active pharmaceutical ingredient concentration of between about 7 〇 mg/mL of the formulation volume, between about Between 1 mg and about 70 mg/mL, between about i „^_about 5〇mg/mL, between about 1 mg/mL and about 20 mg/mL, between about ! mg/mL to about 1 〇140447.doc • 57- 200950787 mg/mL, between about ! mg/mL to about 5 tons/can, or between about 5 mg/mL to about 5 mg/mL activator. Use reduced size to increase surface area and/or modulate formulation dissolution properties. Also used to maintain consistent average particle size distribution (PSD) for any of the formulations described herein (eg, micron size particles, nano size particles or their knives) In some instances, any of the formulations described herein comprise multiple particles, that is, a plurality of particle sizes (eg, microparticles, nano-sized particles, non-sized particles, colloidal particles); That is, the formulation is a multi-particle formulation. In some embodiments, any of the formulations described herein comprise one or more multiples. Particle (e.g., micron) therapeutic agent. Micron is a method of reducing the average diameter of particles of a solid material. The microparticles are from about micrometers in size to a diameter of about nanometers in diameter. In some embodiments, micronization The solid particles have an average diameter of from about 0.5 μϊη to about 5 μ. In certain embodiments, the micronized solid particles have an average diameter between about i μη 1 and about 100 μηι. In certain embodiments, micron. The particles of the solid solid have an average diameter of from about 2 μΐη to about 1 μμηΒ. In some embodiments, the particles of the micronized solid have an average diameter of from about 3 μηη to about 5 。. In certain embodiments The micronized solid of a particle contains a particle size of less than about 5 microns, less than about 2 microns, and/or less than about 100 microns. In certain embodiments, particles of a corticosteroid (eg, micronized) Particles) use a corticosteroid that allows any of the formulations described herein to be extended and/or sustained release relative to a formulation containing a non-multiple particle (eg, no micronized) corticosteroid ear. In some instances, The formulation of heavy particle (eg micron) corticosteroids is suitable for lmL injection of 27G needles without any clogging or agglomeration of 140447.doc -58- 200950787. In some instances, any of the particles of any of the formulations described herein are - Coating particles (eg, a coated microparticle, nanoparticle) and/or a microsphere and/or a liposome particle. Particle size reduction techniques include, for example, grinding, milling (eg, air abrasion mills) (spray mill), ball. Grinding, colloidal agglomeration, high pressure homogenization, spray drying and/or supercritical value • liquid crystallization. In some instances 'particle size is accomplished by mechanical impact φ (e.g., by a ball breaker, ball mill, and/or tip mill). In some instances, the particle size is via liquid energy (e.g., by a spiral spray mill, a loop spray mill, and/or a fluidized bed spray mill). In certain embodiments, the formulations described herein comprise crystalline particles and/or isotropic particles. In certain embodiments, the formulations described herein comprise amorphous particles and 7 or non-isotropic particles. In certain embodiments, the formulations described herein comprise a therapeutic agent particle, wherein the therapeutic agent is a free test, or a salt or prodrug of a therapeutic agent, or any combination thereof. • In certain embodiments, the formulations described herein comprise one or more pico-f steroids wherein the corticosome comprises nanoparticle. In certain embodiments, the formulations described herein comprise a corticosteroid bead coated with a controlled release excipient (e.g., methyl dehydrocorticosteroid beads), in some embodiments, The formulations described herein comprise a corticosteroid that is granulated and/or reduced in size and coated with a controlled release agent; the particulate coated corticosteroid particles are then micronized as appropriate and/or as described herein The composition is formulated. In some instances, a corticosteroid that is a free acid or a free base is used in combination with a steroidal salt of 140447.doc • 59-200950787 - a corticosteroid to prepare a pulse release ear formulation according to the process described herein: in some In the formulation, a combination of a micronized corticosteroid (and/or a salt thereof or a peony) and a coated particle (eg, a nanoparticle, a lipid globule, a microsphere) is used to prepare a pulsed release ear using any of the processes described herein. Alternatively, a pulse release profile can be obtained by cyclodextrin, surfactant (eg, poloxamer (407, 338, 188), Tween (8〇, 6〇, 2〇, Sichuan, PEG· Suihua said that oyster oil, Solvents such as N-methyl-2-guanidine or similar agents help to deliver a 20% delivery dose of corticosteroids (10), such as micronized cortical solid SI, or its free alcohol, free acid or Salt or prodrug;: a particulate corticosteroid, or a free base thereof, a free acid or a salt or a prodrug thereof, which is prepared using any of the processes described herein to prepare a pulsed release formulation. In a particular embodiment, Any of the ear compatible formulations include one or more micronized agents (eg, steroids). In some such embodiments, a micronized agent comprises micronized particles, coated (eg, Extended release coating) micronized particles or combinations thereof, etc. In some such embodiments 'a micronized particle, coated (eg, extended release Q coated) micronized particle, or the like The combined micronized agent comprises - cortex.. = sterol is a free acid, a free test, a salt, a prodrug or any other group thereof. In a particular embodiment, one of the pharmaceutical compositions described herein Containing thiol dehydrocorticosterol as a micronized powder, Dehydrocortisol or dehydrogenation = betitol. In a particular embodiment, one of the pharmaceutical compositions described herein is a thiol dehydrocorticosterol of a micromethyl dehydrocorticosteroid powder. Multiparticulate and/or micronized corticosteroids are delivered to an ear structure (eg, 'inner ear) by any matrix comprising a solid, liquid or gel matrix of the type 140447.doc-60-200950787. In some implementations The multiparticulate and/or micronized corticosteroid is delivered to an ear structure (eg, inner ear) via an in-the-ear injection by any type of matrix comprising a solid, liquid or gel matrix. " Pharmaceutical Formulation. I The pharmaceutical compositions provided include at least - steroidal steroids and pharmaceuticals. 彳 accept diluents, excipients or carriers. In certain embodiments, the pharmaceutical composition comprises other drugs or pharmaceutical agents, carriers, adjuvants such as preservatives, stabilizers, wetting or emulsifying agents, solution promoters, and salts for regulating osmotic pressure, and / or buffer. In other embodiments, the pharmaceutical composition also contains other therapeutic substances. In a specific embodiment of the auris-acceptable controlled release corticosteroid pharmaceutical formulation described herein, the corticosteroid is provided in a gel matrix and is also referred to herein as an "early acceptable gel formulation", "the inner ear is acceptable Gel Formulation, Middle Ear Acceptable Gel Formulation, Ear Acceptable Gel Formulation, Ear 9 Gel Formula, or the like. All ingredients of this gel formulation must be compatible with the standard ear structure. Furthermore, the gel formulation provides controlled release of the corticosteroid to the intended location within the target ear structure; in certain embodiments, the gel formulation also has an immediate or rapid release component to facilitate delivery of the corticosteroid to Expected location. In other embodiments, the gel formulation has a sustained release component for delivery of corticosteroids. In certain embodiments, the gel formulation comprises a multiparticulate (e.g., micronized) corticosteroid. In certain embodiments, the ear gel formulation is biodegradable. In other embodiments, the otic gel formulation includes a mucoadhesive excipient to allow adhesion to the outer mucosal layer of the 140447.doc-61 - 200950787 round window film. In still other embodiments, the otic gel formulation comprises a penetration enhancer excipient; in yet another embodiment, the otic gel formulation s has a viscous promoter which is sufficient to provide a viscous Between about 5 〇〇 to ι, οοο, οοο 泊 间 'between about 75 〇 to 1 〇〇〇〇〇〇 Μ Μ; between about 1000 and 400,000 PCT; between about 2 〇〇〇 1 〇〇〇〇〇 泊 泊; between 3,000 and 50, 〇〇〇 泊 ;; between about 4 〇〇〇 to 25 〇〇〇 ;; between 5,000 and 20, 〇〇〇 Between centipoise; or between about 6 〇〇〇 and 15 〇〇〇. In an additional or alternative embodiment, the otic gel formulation can be administered intraocularly to the round window membrane or to the round window membrane. In other embodiments, the otic gel formulation is administered through the posterior ear incision and surgically to the round window membrane or to the round window membrane or coronal fossa crown region. Alternatively, the otic gel formulation is applied with a needle and a syringe, wherein the needle is inserted into the eardrum and guided to the area of the round window media or the cochlea to write the crown. This otic gel formulation is then deposited on or near the round window membrane or the deaf crown to locally treat autoimmune ear disorders. In other embodiments, the otic gel formulation is administered by a catheter that is implanted into a patient, and in yet another embodiment, the formulation is applied to or near the round window membrane by a pump. In still the embodiment, the ear gel formulation is applied to the micro-injection device (4) to or near the round window membrane. In still other embodiments, the otic gel formulation is applied to the auricular acupoints. In still the examples, the ear gel formulation was applied to the eardrum. In still the examples, the otic gel formulation is applied to the ear canal or within the ear canal. In yet another particular embodiment, any of the pharmaceutical compositions described herein comprise a multiparticulate corticosteroid in a liquid matrix (e.g., a liquid composition for in-ear injection or ear drops). In a particular embodiment, any of the pharmaceutical compositions described herein comprise a multiparticulate cortex in a solid matrix I40447.doc • 62-200950787 Steroid. Controlled Release Formulations A 'controlled release drug formulation imparts controlled release of drug release relative to release time and release time in the body. As discussed herein, controlled release is a combination of delayed release, extended release, sustained release, and/or pulsed release (e.g., a combination of extended release and immediate release), or the like. Many advantages are provided by controlled release. First, controlled release of the pharmaceutical agent allows for less dosing frequency and thus Φ minimizes repeated treatment. Second, controlled release therapy results in more effective drug use and less compound residue. Third, controlled release provides the possibility of local drug delivery by placing a delivery device or formulation at the disease location. Further, controlled release can provide for administration and release of at least two different drugs via a single dosage unit, each having a unique release profile, or releasing the same drug at different rates or during different periods. Thus, one aspect of the embodiments described herein provides for the provision of a controlled release corticosteroid for the treatment of autoimmune disorders and/or inflammatory disorders © Accepting compositions or formulations. Controlled by the compositions and/or formulations disclosed herein (4) The metamorphic sample is conferred by a variety of agents, including but not limited to excipients, reagents or materials that are useful for internal or otic structures thereof. Ear-Acceptable Gel Gel, sometimes referred to as Kangjiao, has been defined in a number of ways. For example, the United States Pharmacopoeia defines a coagulation as a semi-solid system consisting of a small inorganic particle or a larger organic molecule consisting of a liquid-permeating suspension. Gel can be more

相或 "*相系統組成。一單柏I 早相凝膠經由一液體均勻分散的 有機巨分子,在此方式於分散之 刀子及液體間無明顯界 140447.doc .63· 200950787 線存在。單相《通t由合成巨分子(例如,卡波姆⑧)或 由天然樹膠(例如,黃蓍膠)製備。在某些實施例中,單相 凝膠通常為水性’⑯亦可使用醇及油製成。二-相凝膠由 小分散粒子網絡形成。 凝膝亦可區分為疏水性或親水ί在特定實施例中,疏 水性凝穋的基質通常由—含聚乙烯或脂肪油液體石腊與含 膠質氧化⑦m鋅4膠化的脂肪油組成。相反地,疏 水性凝夥的基質通常由水、甘油、或丙二醇與合宜之凝膠 劑(例如,黃蓍膠、澱粉、纖維素衍生物、羧基乙烯聚合 物、及/或矽酸鋁鎂)凝膠而組成。在特定實施例中,本文揭 露之組成物的流變為擬塑性、塑性、流動減黏性或膨服性。 在一實施例中,本文所描述之增進黏性的耳可接受配方 在室温時不為液體。在特定實施例中,此增進黏性的配方 之特性在於介於室温與體温(包括一嚴重發燒的患者,如 南至至約42。(:)間的相轉換。在某些實施例中,此相轉換 在在低於體溫it:發生,低於體温發生,低於體温3t>c 發生,低於體温4。(:發生,低於體温6°c發生,低於體温 8 C發生,低於體温i(TC發生。在某些實施例中,此相轉 換在在低於體温15。(:發生,低於體温2〇°c發生,低於體温 25°C發生。在特定實施例中,本文所描述之配方的膠化温 度(Tgel)為約2(TC,約25t,或約3〇它。在特定實施例 中,本文所描述之配方的膠化温度(Tgel)為約35<)(:,或約 40°C。在一實施例中,本文所描述之任何配方在約體温的 投藥減少或抑制耳配方的耳内投藥附隨的暈眩。體温的定 140447.doc • 64 - 200950787 義包括健康身體或-不健康的體溫,後者包括—發燒的患 者(高至〜42°C )。 由聚氧丙烯與聚氧乙烯組成的聚合物當併入水溶液中 時,形成熱可逆凝膠。此些聚合物具有於接近體温的温度 由液態改變至凝膠態的能力’因此允許做為施用至標的耳 結構的有效配方。&液態-至_疑膠態相轉換為依在溶液内 的聚合物濃度及成份而定。 ΦPhase or "* phase system. A single cypress I early phase gel is uniformly dispersed by a liquid organic macromolecule, in this way there is no obvious boundary between the dispersed knife and the liquid 140447.doc .63· 200950787 line exists. Single phase "through t is prepared from synthetic macromolecules (e.g., carbomer 8) or from natural gums (e.g., tragacanth). In certain embodiments, the single phase gel is typically aqueous <16 and can also be made using alcohols and oils. The two-phase gel is formed from a network of small dispersed particles. Kneeling can also be distinguished as hydrophobic or hydrophilic. In a particular embodiment, the hydrophobically condensed matrix is typically comprised of a fatty oil containing polyethylene or a fatty oil liquid paraffin and a gelatinized gum containing 7m zinc. Conversely, the hydrophobic conjugate matrix is typically comprised of water, glycerin, or propylene glycol with a suitable gel (eg, tragacanth, starch, cellulose derivatives, carboxyvinyl polymers, and/or aluminum magnesium silicate) Composition by gel. In a particular embodiment, the flow of the compositions disclosed herein becomes pseudoplastic, plastic, flow desorbing or bulking. In one embodiment, the viscous auris-acceptable formulations described herein are not liquid at room temperature. In certain embodiments, the viscosity enhancing formulation is characterized by a phase transition between room temperature and body temperature (including a severely feverd patient, such as south to about 42. (:). In certain embodiments, This phase transition occurs at lower than body temperature it: occurs below body temperature, below body temperature 3t>c, below body temperature 4. (: occurs, occurs below 6°c of body temperature, occurs below 8 C of body temperature, low At body temperature i (TC occurs. In some embodiments, this phase transition is at a temperature below 15 ((: occurs, occurs below body temperature 2 〇 °c, occurs below body temperature 25 ° C. In a particular embodiment) The gelatinization temperature (Tgel) of the formulations described herein is about 2 (TC, about 25 t, or about 3 Torr. In a particular embodiment, the gelatinization temperature (Tgel) of the formulations described herein is about 35 < (:, or about 40 ° C. In one embodiment, any of the formulations described herein reduces administration at about body temperature or inhibits the stun of the in-ear administration of the ear formulation. Body temperature setting 140447.doc • 64 - 200950787 Sense includes healthy body or - unhealthy body temperature, the latter includes - fever patients (up to ~ 42 ° C A polymer composed of polyoxypropylene and polyoxyethylene forms a thermoreversible gel when incorporated into an aqueous solution. These polymers have the ability to change from a liquid state to a gel state at a temperature close to body temperature' thus allowing For effective application to the target ear structure. & liquid-to-suspect colloidal phase is converted to the concentration and composition of the polymer in the solution.

泊洛沙姆4〇7(PF-127)為一由聚氧乙婦_聚氧丙浠共聚物 組成的非離子性界面活性劑^其他泊洛沙姆包括18咐 級)、237(F-87級)、338(F_1〇8級)。泊洛沙姆水溶液在酸、 驗及金屬離子存在下安PF_127為—商業可得的聚氧乙 烯-聚氧丙烯三嵌段共聚物,其通式£1〇6 p7〇 ei〇6,具有 -平均莫耳質量為13,〇〇〇。此聚合物可藉由適當的方法進 一步純化,以促進的膠化性質。其含有大約7〇%環氧乙 烯,其為親水性來源。其為一系列泊洛沙姆aba嵌段共聚 物之一,其之成員具有如下顯示的化學式。 hydrophilic hydrophilic Η 汁 O-CH,- ch2}{o-ch-ch2-){〇-ch2-ch2)-oh a CH3 hydrophobic P_F127為特別㈣’因為共聚物的濃縮溶液(>20% w/w) 由低黏性透明溶液在加熱至體温下轉形為固態凝膠。因 此,此現象建議當置於與身體接觸,此凝膠製劑將形成一 半固體結構及-控制釋放貯器。再者,pF_127具有良好溶 140447.doc • 65 - 200950787 解能力、低毒性且因此視為一良好的藥傳遞系統介質。 在一可替代實施例中,此熱凝膠為一PEG-PGLA-PEG三 嵌段共聚物(Jeong etal,Nature (1997),388:860-2 ; Jeong etal, J. Control. Release (2000), 63:155-63; Jeong etal, Adv. Drug Delivery Rev· (2002),54:37-51)。此聚合物在約 5% w/w至約 40% w/w濃度呈現膠溶體-凝膠性質。依所需要的性質,在 PGLA共聚物中乳糖苷/乙内酯莫耳比例可在約1:1至約20:1 範圍間。此生成共聚物在水中可溶且在室温形成自由流動 性液體,但在體温形成一氫凝膠。一商業可得的PEG-PGLA-PEG三嵌段共聚物為由Boehringer Ingelheim製造的 RESOMER RGP t50106。此物質由 50:50 聚(DL-乳糖苷-共-乙内酯)PGLA共聚物及10% w/w PEG組成且具有約6000之 分子量。此物質由50:50聚(DL-乳糖苷-共-乙内酯)PGLA共 聚物組成且為10% w/w PEG並具有約6000之分子量。 「Regel™」為巨Med公司的商標,其指定一具有可逆熱 凝膠性質的低分子量、可生物降解的嵌段共聚物族,如美 國專利第 6,004,573、6,117,949、6,201,072及 6,287,588 號。 亦包括揭露於審查中之美國專利申請案第09/906,041、 09/559,799及10/919,603號的可生物降解的聚合物藥載 劑。此可生物降解的藥載劑包含ΑΒΑ-型或BAB-型三嵌段 共聚物或其等之混合物,其中A-嵌段為相對疏水性且包含 可生物降解的聚酯或聚(正酯),且B-嵌段為相對親水性且 包含聚乙二醇(PEG) ’該共聚物具有疏水性含量為介於 50.1與83 wt%間且親水性含量為介於17與49.9 wt°/。間,且 140447.doc -66 - 200950787 總嵌段共聚物分子量為介於2000與8000道耳吞間。此藥載 劑在低於正常哺乳動物體温的温度呈現水溶解性且進行可 逆熱凝膠作用以接著在相等於生理上哺乳動物體温的凝膠 温度下以凝膠存在。可生物降解的、疏水性A聚合物嵌段 • 包含一聚酯或聚(正酯),其中該聚酯由選自下列組群的單 . 體合成且具有介於約600與3000道耳呑間的平均分子量: ·· D,L-乳糖苷、D-乳糖苷、L-乳糖苷、D,L_乳酸、队乳酸、 碜 L-乳酸、乙内酯、羥乙酸、ε-己内酯、ε_羥基己酸、尸丁 内酯、γ-羥基丁酸、δ-戊内酯、δ-羥基戊酸、羥基丁酸、 羥丁二酸、及其等之共聚物。親水性心嵌段部份較佳為具 有平均分子量為介於約500與22〇〇道耳吞間之聚乙二醇 (PEG)。 額外的可生物降解的熱塑性聚酯包括AtHgelTM(由AWx Laboratories公司提供)及/或揭露於,例如美國專利第 5,324,519 ; 4,938763 ; 5,702,716 ; 5,744,153 ; ^5,990,194^; # 中者,其中揭露之合宜可生物降解的熱塑性聚酯為一熱塑 • 性聚合物。合宜之可生物降解的熱塑性聚酯範例包括聚乳 糖苦、聚乙内醋、聚己内S旨、其等之共聚物、其等之四聚 合物及任何其等之組合,在此些實施例中,合宜之可生物 降解的熱塑性聚酯為聚乳糖苷、聚乙内酯、其等之共聚 物、其等之四聚合物或其等之組合。在一實施例中,此可 生物降解的熱塑性聚酯為具有一羧基端基之5〇/5〇聚(〇^ 礼糖苦_共-乙内醋);且存在量為組成物之約30 wt%至約 4〇 Wt% ’且具有平均分子量為約23,000至約45,000。或 140447.doc •67- 200950787 者,在另些其他實施例,此可生物降解的熱塑性聚酯為不 具有一羧基端基之75/25聚(DL-乳糖苷-共-乙内酯);且存 在量為組成物之約40 wt%至約50 wt°/。;且具有平均分子量 為約15,000至約24,000。在又一或替代實施例,該聚(01^ 乳糖苷-共-乙内酯)的端基依聚合方法為羥基、羧基、或 酯。乳酸或羥乙酸的聚縮合作用提供一具有羥基與羧基的 聚合物。環乳糖苷或乙内酯單體與水、乳酸、或羥乙酸的 環開聚合作用提供具有相同端基的聚合物。然而,環單體 以單官能醇如甲醇、乙醇或1-十二烷醇的環開作用提供一 具有一羥基與酯端基的聚合物。環單體以二醇如1,6-己烷 二醇或聚乙二醇的環開聚合作用提供一僅具有羥基端基的 聚合物。 因為熱可逆凝膠的聚合物系統在降温下溶解更完全,溶 解方法包括在降温下加入需要量的聚合物至使用的水量。 通常,在藉由震盪濕潤聚合物後,此混合物加蓋並置於一 冷室或一在約0-1 〇°c的熱恒定容器中以溶解聚合物。攪拌 或震盪此混合物以達到一約更快速的熱可逆凝膠聚合物溶 解。皮質類固醇及多種添加劑如缓衝劑、鹽及保存劑為依 序加入並溶解。在某些例子中,此皮質類固醇及/或其他 醫藥活性劑若不可溶於水則懸浮於水中。此pH藉由加入 合適的緩衝劑而調變。圓窗膜黏膜黏附劑特性可視情況藉 由併入圓窗膜黏膜黏附劑卡波姆,如卡波莫®934P (Carbopol 934P)至組成物而賦予一熱可逆凝膠(Majithiya 等人之 AAPS PharmSciTech (2006),7(3),p. El ; EP0551626), 140447.doc -68- 200950787 其二者皆併入本案做為參考)。 一貫施例中為不需要一加入黏性增進劑之使用的耳可接 受醫藥凝膠配方。此凝膠配方併入至少一醫藥可接受緩衝 劑。在一態樣中,其為一含有皮質類固醇與醫藥可接受緩 衝劑凝膠配方。在另—實施例中,此醫藥可接受賤形劑或 載劑為膠化劑。 . 在其他實施例中,有效的皮質類固酵之耳可接受醫藥配 譽 方亦包括一或一以上pH調節劑或緩衝劑以提供内淋巴或 周邊淋巴合宜之pH。合宜之pH調節劑或緩衝劑包括但不 限制為乙酸酯、碳酸氫酯、氯化銨、檸檬酸酯、磷酸酯、 其等之醫藥可接受鹽及其等之組合或混合物。此pH調節 劑或緩衝劑的使用量係需要維持組成物的pH介約5至約9 的PH間,在一實施例中,pH介於約6.5至約7.5間,且在另 一實施例的 pH為約 6.5、0.6、0.7、0.8、0.9、7.0、7.1、 7·2、7·3、7·4、7.5。在一實施例中,當在本發明揭露之 Φ 配方中使用一或一以上的緩衝劑時,其與如一醫藥可接受 • 載劑組合且在最終配方中的存在量如,約01%至約20〇/〇, ’ 約〇,5%至約!〇%間。在本發明之特定實施例中,包括在凝 膠配方的緩衝液之量為使凝膠配方之pH*會干擾中耳或 内耳的天然緩衝系統,或不會干擾内淋巴或周邊淋巴之天 然PH:依在耳蝸中質類固醇配方之標的處而定。在某些 實鈀例中,約1〇 μΜ至約2〇〇 mM濃度之緩衝液存在於凝膠 配方。在特定實施例中’存在約2〇 mM至約1〇〇 mM濃度 的緩衝液。在一實施例中,一緩衝液為微酸?11如乙酸鹽 140447.doc •69· 200950787 或檸檬酸鹽。在一實施例中,此緩衝液為一具有PH為約 4.5至約6.5的乙酸納缓衝液。在一實施例中,此緩衝液為一具 有pH為約5.0至約8.0的檸檬酸鈉緩衝液,或約5.5至約7.0。 在一實施例中’緩衝液為如略驗性pH之三(羥基曱基)胺 基甲烷、鹽、碳酸鹽或磷酸鹽。在一實施例中,緩衝液為 一具有pH為約6.5至約8.5或約7·〇至約8·〇之碳酸氫鈉緩衝 液。在一實施例中,緩衝液為一具有ρΗ為約6〇至約9 〇之 磷酸二鈉緩衝液。Poloxamer 4〇7 (PF-127) is a nonionic surfactant composed of polyoxyethylene propylene-polyoxypropylene copolymer. Other poloxamers include 18 )), 237 (F- Level 87), 338 (F_1〇8). An aqueous solution of poloxamer in the presence of acid, test and metal ions PF_127 is a commercially available polyoxyethylene-polyoxypropylene triblock copolymer having the general formula: £1〇6 p7〇ei〇6, with - The average molar mass is 13, 〇〇〇. This polymer can be further purified by a suitable method to promote gelation properties. It contains about 7 % epoxy ethene which is a hydrophilic source. It is one of a series of poloxamer aba block copolymers whose members have the chemical formula shown below. Hydrophilic Η juice O-CH, - ch2}{o-ch-ch2-){〇-ch2-ch2)-oh a CH3 hydrophobic P_F127 is special (four)' because of the concentrated solution of the copolymer (>20% w/w ) Converted from a low viscosity transparent solution to a solid gel upon heating to body temperature. Therefore, this phenomenon suggests that when placed in contact with the body, the gel formulation will form a semi-solid structure and a controlled release reservoir. Furthermore, pF_127 has a good solubility, low toxicity and is therefore considered a good drug delivery system medium. In an alternate embodiment, the thermal gel is a PEG-PGLA-PEG triblock copolymer (Jeong et al, Nature (1997), 388:860-2; Jeong et al, J. Control. Release (2000) , 63: 155-63; Jeong et al, Adv. Drug Delivery Rev. (2002), 54: 37-51). The polymer exhibits a colloid-gel nature at a concentration of from about 5% w/w to about 40% w/w. The lactose/ethyl lactone molar ratio in the PGLA copolymer can range from about 1:1 to about 20:1, depending on the desired properties. This resulting copolymer is soluble in water and forms a free flowing liquid at room temperature, but forms a hydrogen gel at body temperature. A commercially available PEG-PGLA-PEG triblock copolymer is RESOMER RGP t50106 manufactured by Boehringer Ingelheim. This material consists of a 50:50 poly(DL-lactosyl-co-caprolactone) PGLA copolymer and 10% w/w PEG and has a molecular weight of about 6,000. This material consisted of a 50:50 poly(DL-lactosyl-co-caprolactone) PGLA copolymer and was 10% w/w PEG and had a molecular weight of about 6,000. "RegelTM" is a trademark of Giant Med, which specifies a family of low molecular weight, biodegradable block copolymers having reversible thermogel properties, such as U.S. Patents 6,004,573, 6,117,949, 6,201,072 and 6,287,588. . Also included are biodegradable polymeric drug carriers disclosed in U.S. Patent Application Serial Nos. 09/906,041, 09/559,799 and 10/919,603. The biodegradable drug carrier comprises a bismuth-type or BAB-type triblock copolymer or a mixture thereof, wherein the A-block is relatively hydrophobic and comprises a biodegradable polyester or poly(orthoester) And the B-block is relatively hydrophilic and comprises polyethylene glycol (PEG) 'The copolymer has a hydrophobic content between 50.1 and 83 wt% and a hydrophilic content between 17 and 49.9 wt. And 140447.doc -66 - 200950787 The total block copolymer molecular weight is between 2000 and 8000 auricular. The drug carrier exhibits water solubility at a temperature below the body temperature of a normal mammal and undergoes a reversible thermogel action to subsequently be present as a gel at a gel temperature equivalent to the physiological body temperature of the mammal. Biodegradable, hydrophobic A polymer block • comprises a polyester or poly(orthoester) wherein the polyester is synthesized from a single body selected from the group consisting of about 600 and 3000 deaf Average molecular weight between: ·· D,L-lactoside, D-lactoside, L-lactoside, D, L_lactic acid, lactic acid, 碜L-lactic acid, ethyl lactone, glycolic acid, ε-caprolactone a copolymer of ε-hydroxycaproic acid, cadaverine lactone, γ-hydroxybutyric acid, δ-valerolactone, δ-hydroxyvaleric acid, hydroxybutyric acid, hydroxysuccinic acid, and the like. The hydrophilic core block portion is preferably polyethylene glycol (PEG) having an average molecular weight of between about 500 and 22 ampoules. Additional biodegradable thermoplastic polyesters include AtHgelTM (supplied by AWx Laboratories) and/or disclosed in, for example, U.S. Patent Nos. 5,324,519; 4,938,763; 5,702,716; 5,744,153; ^5,990,194^; The biodegradable thermoplastic polyester is a thermoplastic polymer. Examples of suitable biodegradable thermoplastic polyesters include polylactose, polyethyl vinegar, polycaprolactone, copolymers thereof, tetrapolymers thereof, and combinations thereof, in these embodiments. Among the suitable biodegradable thermoplastic polyesters are polyglycosides, polyethyl lactones, copolymers thereof, tetrapolymers thereof, or the like. In one embodiment, the biodegradable thermoplastic polyester is 5 〇/5 〇 poly( 〇 苦 _ 共 共 共 乙 ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; From wt% to about 4 〇 Wt% 'and having an average molecular weight of from about 23,000 to about 45,000. Or 140447.doc •67- 200950787, in still other embodiments, the biodegradable thermoplastic polyester is 75/25 poly(DL-lactosyl-co-lactone) having no carboxyl end group; And is present in an amount from about 40% to about 50 wt% of the composition. And having an average molecular weight of from about 15,000 to about 24,000. In still another or alternative embodiment, the terminal group of the poly(01^-lactosyl-co-lactone) is a hydroxyl group, a carboxyl group, or an ester according to a polymerization method. The polycondensation of lactic acid or glycolic acid provides a polymer having a hydroxyl group and a carboxyl group. The ring-opening polymerization of a cyclic lactose or a lactone monomer with water, lactic acid, or glycolic acid provides a polymer having the same end groups. However, the ring monomer provides a polymer having a hydroxyl group and an ester end group by ring opening of a monofunctional alcohol such as methanol, ethanol or 1-dodecanol. The ring-opening polymerization of a ring monomer with a diol such as 1,6-hexanediol or polyethylene glycol provides a polymer having only hydroxyl end groups. Since the polymer system of the thermoreversible gel dissolves more completely under cooling, the dissolution method involves adding the required amount of polymer to the amount of water used under cooling. Typically, after wetting the polymer by shaking, the mixture is capped and placed in a cold chamber or in a thermo-constant container at about 0-1 ° C to dissolve the polymer. The mixture is agitated or shaken to achieve an approximately faster thermal reversible gel polymer dissolution. Corticosteroids and various additives such as buffers, salts and preservatives are added and dissolved in order. In some instances, the corticosteroid and/or other pharmaceutically active agent is suspended in water if it is insoluble in water. This pH is modulated by the addition of a suitable buffer. The characteristics of the round window membrane mucoadhesive agent can be imparted to a thermoreversible gel by incorporating a round window membrane mucoadhesive carbomer such as Carbomo® 934P (Carbopol 934P) (Majithiya et al. AAPS PharmSciTech) (2006), 7(3), p. El; EP0551626), 140447.doc-68-200950787 both of which are incorporated herein by reference. It has been consistently practiced to accept a pharmaceutical gel formulation that does not require the addition of a viscosity enhancing agent. This gel formulation incorporates at least one pharmaceutically acceptable buffer. In one aspect, it is a gel formulation containing a corticosteroid and a pharmaceutically acceptable buffer. In another embodiment, the pharmaceutically acceptable bismuth or carrier is a gelling agent. In other embodiments, an effective corticosteroid-acceptable pharmaceutical remedy also includes one or more pH adjusting or buffering agents to provide a suitable pH for endolymph or peripheral lymph. Suitable pH adjusting or buffering agents include, but are not limited to, acetates, hydrogencarbonates, ammonium chlorides, citrates, phosphates, pharmaceutically acceptable salts thereof, and the like, or combinations or mixtures thereof. The pH adjusting or buffering agent is used in an amount to maintain a pH of the composition of between about 5 and about 9, in one embodiment, the pH is between about 6.5 and about 7.5, and in another embodiment The pH is about 6.5, 0.6, 0.7, 0.8, 0.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5. In one embodiment, when one or more buffering agents are used in the Φ formulation disclosed herein, they are combined with, for example, a pharmaceutically acceptable carrier and are present in the final formulation, for example, from about 01% to about 20〇/〇, '约〇, 5% to about! 〇%. In a particular embodiment of the invention, the amount of buffer included in the gel formulation is such that the pH of the gel formulation interferes with the natural buffer system of the middle or inner ear, or does not interfere with the natural pH of the endolymph or peripheral lymph. : Depends on the target of the steroid formulation in the cochlea. In some real palladium cases, a buffer of about 1 〇 μΜ to about 2 mM is present in the gel formulation. In a particular embodiment, a buffer is present at a concentration of from about 2 mM to about 1 mM. In one embodiment, a buffer is slightly acidic? 11 such as acetate 140447.doc • 69· 200950787 or citrate. In one embodiment, the buffer is a sodium acetate buffer having a pH of from about 4.5 to about 6.5. In one embodiment, the buffer is a sodium citrate buffer having a pH of from about 5.0 to about 8.0, or from about 5.5 to about 7.0. In one embodiment, the buffer is a tris (hydroxyindenyl) aminomethane, salt, carbonate or phosphate such as a slightly acidic pH. In one embodiment, the buffer is a sodium bicarbonate buffer having a pH of from about 6.5 to about 8.5 or from about 7% to about 8%. In one embodiment, the buffer is a disodium phosphate buffer having a pH of from about 6 Torr to about 9 Torr.

本文亦描述包含皮質類固醇與一黏性增進劑的控制釋另 配方。合宜的黏性·增進劑包括僅為例示之勝化劑及❸ 齊卜在-實施例中,此增進黏性的配方並不包括一❹ 劑。在其他實施例中’此增進黏性的配方包括—醫藥可專 受緩衝劑。氣化鈉或其他張力劑為 、 整張力性。 *要可視情況_ 僅為例示之用,耳-可垃典 了接爻黏性劑包括羥基丙基甲基 維素、羥基乙基纖維素、聚Also described herein are controlled release formulations comprising a corticosteroid and a viscosity enhancing agent. Suitable viscosifying agents include, by way of example only, exemplifying agents and bismuth. In the examples, the viscosity enhancing formulation does not include a sputum. In other embodiments, the formulation to enhance adhesion includes - the drug may be specifically buffered. Gasified sodium or other tonicity agent, and tension. * Depending on the situation _ for illustrative purposes only, the ear - can be used as an adhesive, including hydroxypropylmethyl vegan, hydroxyethyl cellulose, poly

眾乙烯吡咯啶酮、羧其甲其縫 素、聚乙烯醇、軟骨辛垆 土甲土鐵 素硫酸鈉、玻尿酸鈉。 文所述之醫荦组忐鉍用於 I晉樂、.且成物的黏性劑包括但未 (gum arabic)、洋菜 為阿拉伯 /毁IS鎂、藻酸鋼、 角藻、膨土、卡波^ 硬知酸鈉、 莫)、三仙膠、纖維音/ 鹿角屬膠、卡波莫(卡 ^ V 、,微結晶纖維素(MCC)、& -各 杉藻膠、葡萄糖、富寒“ )、角旦素 潤土 (heCt〇rite)、乳糖 p度膠、胍膠、 庶糖,麥芽糊精、廿 糖醇、蜂蜜、玉米澱粉、小麥 甘讀、山 J麥歲粉玉米職粉、馬鈐薯i I40447.doc -70- 200950787 粉、明膠、梧桐膠、黃樹膠、聚乙二醇(例如pEG 2〇〇_45〇〇)、 η蓍膠、乙基纖維素、己基髮基乙基纖維素、乙基甲基纖 維素、甲基纖維素、經基乙基纖維素、經基乙基曱基纖維 素、羥基丙基纖維素、聚(羥基乙基胺基甲酸酯)、氧基聚 日月踢、果膠、聚明膠、普維剩、丙稀碳酸醋、甲基乙 順丁烯二酸酐共聚物(PVM/MA)、聚(甲氧基乙基胺基子酸 ·· 酯)、聚(曱氧基乙氧基乙基胺基曱酸酯)、羥基丙基纖維 φ 素、羥基丙基甲基-纖維素(HPMC),鈉羧基甲基-纖維素 (CMC)、二氧化矽、聚乙浠°比咯咬酮(PVP :普維酮)、 Splenda®(葡萄糖麥芽糊精及蔗糖素)或其等之組合。在特 定實施例中,此黏性-增進賦形劑為甲基纖維素(MC)與 CMC的組合。在另些其他實施例,此黏性_增進劑與羧基 甲基化聚葡萄胺糖、或幾丁質及藻酸酯組合。本文揭露之 幾丁質與藻酸酯與本文揭露的皮質類固醇之組合做為一控 制釋放配方’限制皮質類固醇由此配方擴散。再者,缓基 φ 曱基化聚葡萄胺糖與藻酸酯的組合可選擇地用於幫助增加 . 皮質類固醇對内耳圓窗膜的穿透性。 •某些實施例為一增稠配方,其包含約01 Mm至約100 mM 的皮質類固醇、一醫藥可接受黏性劑、及用於注射的水, 黏性劑在水的濃度為足以提供增稠配方之最終呈現黏性係 約100至約100,000 cP。在特定實施例中,凝膠黏性在約 100至約50,000 cP範圍間,約100至約!,〇〇〇 cP範圍間,約 5〇〇至約1,500 cP範圍間,約!,〇〇〇至約3,000 cp範圍間,約 2,000至8,000 cP範圍間’約4,〇〇〇至約50,000 cP範圍間, 140447.doc •71· 200950787 約 10,000至約 500,000 cP範圍間,約15,000 cp至約 1,000,000 cp。 在另一實施例中’當期待一甚至更黏稠的介質時,此生物 相容凝膠包含至少約35 wt%,至少約45 wt%,至少約55 wt%,至少約65 wt% ’至少約70 wt%,至少約75 wt°/〇,或 更至少約80 wt%等之皮質類固醇。在高濃縮試樣中,此生 物相容增稍配方包含至少約25 wt%,至少約35 wt%,至少 約45 wt% ’至少約55 wt%,至少約65 wt%,至少約75 wt%, 至少約85 wt%,至少約90 wt%或至少約95 wt%或更多之 皮質類固醇。 某些實施例中,本文所述之凝膠配方的黏性藉由本文所 述之任何方式測量。例如’在某些實施例中,使用— LVDV-II+CP Cone板黏度計與Cone心轴CPE-40計算本文所 述之凝膠配方的黏性。在其他實施例中,使用一 Br〇〇kfield (心軸及杯)黏度計計算本文所述之凝膠配方的黏性。在某 些實施例中,本文所指的黏性範圍為在室温測量。在其他 實施例中,本文所指的黏性範圍為在體溫測量(如,健康 人體的平均體温)。 在一實施例中,此醫藥可接受增進黏性耳可接受配方包 含至少-皮質類固醇及至少一膠化劑。用⑨製備凝膠配方 的合宜膠化劑包括但未限制為纖維素、纖維素衍生物、纖 維素醚(例如,羧基甲基纖維素、乙基纖維素、羥基乙基 纖維素、羥基甲基纖維素、羥基丙基甲基纖維素、羥基丙 基纖維*、甲基纖維素)、脈谬、三仙滕樹膠、刺槐豆 膠、藻酸酯(例如,藻酸)、矽酸鹽、澱粉、黃蓍膠、羧美 140447.doc •72· 200950787 乙烯聚合物、鹿角藻膠、石臘、凡士林及其等之任何解合 或混合物。在某些其他實施例中,利用羥基丙基甲基纖維 素(Methocel®)為膠化劑。在特定實施例中,亦利用+久 所述之增稠劑做為本文所述之凝膠配方的膠化劑。 在某些實施例中,其他凝膠配方可依使用的特定皮質_ 固醇、其他醫藥劑或賦形劑添加劑而使用,且其等視為屬 於本發明揭露之範疇。例如,其他目前商業可得之甘油— 系凝膠、甘油-衍生化合物、共軛或交聯凝膠、基體、^ 凝膠及聚合物,以及明膠及其等之衍生物、藻酸酯、及療 酸酯-系凝膠、且甚至天然及合成氫凝膠與氫凝膠-衍生化 合物皆可預期用於本文所述之皮質類固醇配方。在某瘦實 施例中,耳可接受之凝膠包括但未限制為藻酸酯氫棘# SAF®-凝膠(美國紐澤西州普林斯頓市之ConvaTec公司)、 Duoderm® Hydro活性凝膝(ConvaTec)、Nu-凝膠® (美圈 <意 州阿靈頓市 Johnson & Johnson Medical) ; Carrasyn®(V) Acemannan 氫凝膠(美國德州爾凡市 Carrington Laboratories 公司);甘油凝膠Elta®氫凝膠(美國德州達拉斯市Swiss-American Products公司)及K-Y® Sterile (Johnson & Johnson)。 在又一實施例中’可生物降解的生物相容凝膠亦為可存在 於本文所述及揭露之可接受配方的化合物。 在某些發展用於哺乳動物投藥及用於人類投藥之組成物 配方的配方中’耳可接受凝膠包含實質所有組成物重量。 在其他實施例中,此耳可接受凝膠包含組成物之如約 98 wt%或約99 wt%多。當需要一實質無-液體,或實質黏 140447.doc •73- 200950787 性配方時,此為需要的。在又一實施例中,當需要稍較低 黏性或稍較高液體耳可接受醫藥凝膠配方時,此配方的生 物相容凝膠部份包含至少約5〇 wt%,至少約60 wt%,至少Vinyl pyrrolidone, carboxy ketone, polyvinyl alcohol, cartilage sulphate, sodium sulphate, sodium hyaluronate. The medical group described in the article is used for I Jinle, and the adhesives of the composition include but not (gum arabic), acacia is Arab / ruined IS magnesium, alginic acid steel, hornweed, benton, Kappo ^ Sodium sulphate, Mo), Sanxian gum, fiber sound / staghorn gum, carbomer (card ^ V, microcrystalline cellulose (MCC), & - each cedar gum, glucose, rich Cold "), horny sulphate (heCt 〇rite), lactose p-degree rubber, tannin extract, sucrose, maltodextrin, sucrose, honey, corn starch, wheat, reading, mountain J wheat powder corn Powder, horse yam i I40447.doc -70- 200950787 powder, gelatin, paulownia gum, gums, polyethylene glycol (eg pEG 2〇〇_45〇〇), η蓍 gum, ethyl cellulose, hexyl hair Ethyl ethyl cellulose, ethyl methyl cellulose, methyl cellulose, transethyl ethyl cellulose, transethyl ethyl fluorenyl cellulose, hydroxypropyl cellulose, poly (hydroxyethyl urethane) ), oxy-poly-day kick, pectin, poly-gelatin, puwei, propylene carbonate, methyl ethyl maleic anhydride copolymer (PVM/MA), poly(methoxyethylamine basic acid) ·· Ester), poly(methoxyethoxyethylamino decanoate), hydroxypropyl fiber φ, hydroxypropylmethyl-cellulose (HPMC), sodium carboxymethyl-cellulose (CMC), a combination of cerium oxide, polyethyl hydrazine, ketamine (PVP: pucanone), Splenda® (glucose maltodextrin and sucralose), or the like. In a particular embodiment, the viscosity-promoting The agent is a combination of methyl cellulose (MC) and CMC. In still other embodiments, the viscosity-enhancing agent is combined with carboxymethylated polyglucamine, or chitin and alginate. The combination of chitin and alginate with the corticosteroids disclosed herein as a controlled release formulation to limit the diffusion of corticosteroids from this formulation. Furthermore, the combination of suspending φ thiolated polyglucamine and alginate Optionally used to help increase the permeability of corticosteroids to the inner ear round window membrane. • Some embodiments are a thickening formulation comprising from about 01 Mm to about 100 mM of corticosteroids, a pharmaceutically acceptable viscosity And the water used for injection, the viscosity of the adhesive is sufficient to provide the thickening formula The adhesive presently exhibits from about 100 to about 100,000 cP. In a particular embodiment, the gel viscosity ranges from about 100 to about 50,000 cP, from about 100 to about !, between 〇〇〇cP, about 5 〇〇 to Between the range of about 1,500 cP, about!, 〇〇〇 to the range of about 3,000 cp, between the range of about 2,000 to 8,000 cP 'about 4, 〇〇〇 to about 50,000 cP, 140447.doc •71· 200950787 Between 10,000 and about 500,000 cP, from about 15,000 cp to about 1,000,000 cp. In another embodiment, the biocompatible gel comprises at least about 35 wt%, at least about 45 wt%, at least about 55 wt%, at least about 65 wt% 'at least about when an even more viscous medium is desired. 70 wt%, at least about 75 wt ° / 〇, or at least about 80 wt%, etc., a corticosteroid. In a highly concentrated sample, the biocompatible extender formulation comprises at least about 25 wt%, at least about 35 wt%, at least about 45 wt% 'at least about 55 wt%, at least about 65 wt%, at least about 75 wt% At least about 85 wt%, at least about 90 wt% or at least about 95 wt% or more of the corticosteroid. In certain embodiments, the viscosity of the gel formulations described herein is measured by any of the methods described herein. For example, in some embodiments, the viscosity of the gel formulations described herein is calculated using the LVDV-II+CP Cone plate viscometer and the Cone mandrel CPE-40. In other embodiments, a Br〇〇kfield (mandrel and cup) viscometer is used to calculate the viscosity of the gel formulations described herein. In some embodiments, the viscosity range referred to herein is measured at room temperature. In other embodiments, the range of viscosity referred to herein is measured at body temperature (e.g., the average body temperature of a healthy human). In one embodiment, the pharmaceutically acceptable viscous auricular acceptable formulation comprises at least a corticosteroid and at least one gelling agent. Suitable gelling agents for formulating gel formulations with 9 include, but are not limited to, cellulose, cellulose derivatives, cellulose ethers (eg, carboxymethylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxymethyl) Cellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose*, methylcellulose), cerium, sylvestre, locust bean gum, alginate (eg alginic acid), citrate, starch , tragacanth, carboxymei 140447.doc • 72· 200950787 Any dissolving or mixture of ethylene polymer, cerulein, paraffin, petrolatum and the like. In certain other embodiments, hydroxypropyl methylcellulose (Methocel®) is utilized as the gelling agent. In a particular embodiment, a thickening agent as described above is also utilized as a gelling agent for the gel formulations described herein. In certain embodiments, other gel formulations may be used depending on the particular corticosteroid, other pharmaceutical agent or excipient additive used, and are considered to be within the scope of the present disclosure. For example, other currently commercially available glycerin-based gels, glycerin-derived compounds, conjugated or crosslinked gels, matrices, gels and polymers, as well as gelatin and its derivatives, alginate, and Therapeutic acid-based gels, and even natural and synthetic hydrogen gels and hydrogen gel-derived compounds, are contemplated for use in the corticosteroid formulations described herein. In a lean embodiment, the gel acceptable for the ear includes, but is not limited to, alginate hydrogen spine # SAF®-gel (ConvaTec, Princeton, New Jersey, USA), Duoderm® Hydro active knee (ConvaTec) ), Nu-Gel® (US Circle &Johnson; Johnson Medical, Arlington); Carrasyn® (V) Acemannan Hydrogel (Carrington Laboratories, Inc.); Glycerin Gel Elta® Hydrogen gel (Swiss-American Products, Dallas, TX) and KY® Sterile (Johnson & Johnson). In yet another embodiment, the biodegradable biocompatible gel is also a compound that can be present in the acceptable formulations described and disclosed herein. In certain formulations for the formulation of compositions for mammalian administration and for human administration, the 'Ace Acceptable Gel contains substantially all of the composition weight. In other embodiments, the auris-acceptable gel comprises, for example, about 98 wt% or more than 99 wt% of the composition. This is required when a substantially non-liquid, or substantially viscous formula is required. In yet another embodiment, the biocompatible gel fraction of the formulation comprises at least about 5 wt%, at least about 60 wt when a slightly lower viscosity or slightly higher liquid ear acceptable pharmaceutical gel formulation is desired. %,at least

約70 wt%,或甚至至少約80 wt%或90 wt%之化合物》所 有在此範圍的中間質的整數皆視為屬於本發明揭露之範 疇’且在某些實施例中,甚至調配更液態(且因此較不黏 性)的耳可接受凝膠組成物’如此混合物之凝膠或基體組 份包含不超過組成物之約50 wt°/。,不超過約40 wt%,不大 於約3 0 wt%,或甚至含有不超過約15 wt%或約20 wt%。 圓形窗膜黏膜黏附劑About 70 wt%, or even at least about 80 wt% or 90 wt% of the compound "all intermediates in this range are considered to be within the scope of the present disclosure" and in some embodiments, even more liquid. (and therefore less viscous) an auris-acceptable gel composition 'The gel or matrix component of such a mixture contains no more than about 50 wt.% of the composition. , no more than about 40 wt%, no more than about 30 wt%, or even no more than about 15 wt% or about 20 wt%. Round window film mucoadhesive

在實施例的範疇中亦預期加入圓窗膜黏膜黏附劑至本發 明揭露之皮質類固醇配方與組成物。「黏膜黏合劑」一詞 為泛指用於可黏合至生物膜的黏蛋白層之物質,如3_層圓 形窗膜的外膜《做為一圓形窗膜黏膜黏附劑聚合物,此聚 η物應具有某些一般物化特徵如主要為具有數個氫鍵形成 基的陰離子性親水性,合宜之表面性質以濕潤黏液/黏膜 組織表面及足夠的彈性以穿透黏膜網絡。 可與耳可接受方使用的圓形窗膜黏膜黏附劑包括但未 限制於至可溶聚乙稀°比11各咬酮聚合物(pvp); 一水_可 膨脹但水-不可溶、纖維、交聯縣·官能聚合物;一交聯 聚(丙稀酸)(例如卡波莫947P);—卡波姆均聚物;一卡波 姆共聚物;-親水性聚醣樹膠、麥芽糊精、一交聯褐藻酸 樹膠凝膠、水·可分散聚羧酷乙烯聚合物、至少二選自二 氧化鈦、二氧化碎、及黏土或其等之混合物的顆粒組份。 140447.doc 74· 200950787 圓®膜黏膜黏附劑可與耳可接受黏性增加賦形劑組合使 用’或單獨使用以增加組成物與黏膜層標的耳組份的交互 作用。在一非限制實施例中,此黏膜黏附劑為麥芽糊精及/ 或藻酸酯樹膠。當使用時,此圓形窗膜賦予組成物的黏膜 黏附劑特性應在一足以傳遞有效量之皮質類固醇組成物至 例如内耳圓窗黏膜或耳蝸窩冠部,且其量為可塗覆黏膜, • 並接著可傳遞組成物至受影響的區域,包括如例示說明的 •參 内耳之#庭及/或耳堝結構。熟於是項技術人士可決定本 文所述之組成物的黏膜黏附劑特性,且可因此決定適當的 使用量。一決定足夠黏膜黏附性之方法可包括監控組成物 與黏膜層的交互作用改變,包括但未限制於量測在沒有或 存在該賦形劑時此組成物停留或維持時間。 在一非限制實施例中,此圓窗膜黏膜黏附劑為麥芽糊 精。麥芽糊精為-水解由玉米、馬鈴薯、小麥或其他植物 衍生之澱粉所製造的碳水化合物。麥芽糊精可單獨使用或 ❷ 與其他圓窗膜黏膜黏附劑一起使用以賦予本文揭露之組成 . 物的黏膜黏附劑特性。在一實施例中,麥芽糊精與卡波莫 ·. 聚合物的組合使用可增加本文揭露之組成物的圓窗膜黏膜 黏附劑。 在另一實施例中,此圓窗膜黏膜黏附劑為一烷基-糖苷 及/或一餹烷基酯。在本文中使用之,一「烷基-糖苷音 指一含有連接至疏水性烷基之任何親水性醣(例如葡萄 糖、果糖、簾糖、或麥芽糖)的化合物。在某些實施例 中,此圓窗膜黏膜黏附劑為一含有烷基_糖苷的界面活性 140447.doc •75· 200950787 劑’其中該烷基-糖苷包含一連接至疏水性烷基(例如,— 含有約6至約25碳原子的烷基)的糖,其係藉由一醯胺連 接、一胺連接、一胺曱酸鹽連接、一趟連接、一硫喊連 接、一S旨連接、一硫酯連接、一苷連接、一硫普連接、及/ 或一酿基尿素連接。在某些實施例中,此圓窗膜黏膜黏附 劑為己基-、庚基-、辛基_、壬基-、癸基-、十一烷基… 十二烷基_、十三烷基_、十四烷基、十五烷基_、十六烷 基-、十七烷基-及十八烷基α_或β—D-麥芽糖苷;己基_、庚 基-、辛基-、壬基_、癸基_、十一烷基_、十二烷基_、十 三烷基_、十四烷基、十五烷基_、十六烷基_、十七烷基_ 、及十八烧基-α-或β-D-葡萄糖苷;己基_、庚基-、辛基_ 、壬基-、癸基-、十一烷基_、十二烷基-、十三烷基-、十 四烷基、十五烷基_、十六烷基_、十七烷基_、及十八烷基 α-或β-D-蔗糖苷;己基_、庚基·、辛基-、十二烷基_、十 二烧基-及十四烧基-β-D-硫麥芽糖苷;庚基_或辛基_丨_硫_ α-或β-D-葡萄喃糖苷;烷基硫蔗糖;烷基麥芽糖三苷;蔗 糖β-胺基-烷基醚之長鏈脂族碳酸醯胺;由一醯胺連接至 一院基鏈的巴糖或異麥芽胺(is〇maltarnine)衍生物及由尿 素連接至一院基鏈的異麥芽胺衍生物;蔗糖β_胺基-烷基 _的長鏈脂族碳酸醯基尿素及蔗糖β_胺基_烷基醚之長鏈 脂族碳酸醢胺。在某些實施例中,此圓窗膜黏膜黏附劑為 一烧基-糖苷,其中該烧基糖苷為麥芽糖、蔗糖、葡萄糖 或其等之組合以一苷連接至一具9至16碳原子的烷基鏈(例 如壬基-、癸基-、十二烷基-及十四烷基蔗糖苷;壬基_、 140447.doc -76- 200950787 癸基-、十二烷基_及十四烷基葡萄糖苷;及壬基_、癸基_ 、十二烷基-及十四烷基麥芽糖苷在某些實施例中,此 圓窗膜黏膜黏附劑為一烧基-糖聲中該燒基糖普為十 二烷基麥芽糖苷、十三烷基麥芽糖苷及十四烷基麥芽糖 誓。在某些實施你j中,料可接受穿透增進劑為一含有烧 基-糖苷的界面活性劑,其中該烷基糖苷為十四烷基-β—D-麥芽糖音此圓固膜黏膜黏附劑為一院基-糖苦,其中該 烧基糖普為—具有至少-葡萄糖的雙膽。在某些實施例 中,此耳可接受穿透增進劑為一界面活性劑,其包含一 α_ D-葡萄喃糖苦基_β_^糖普、η_十二烷基_4_〇_a_D_葡萄喃 糖苷基-β-喃糖苷、及/或心十四烷基_4_〇_a_D_葡萄喃糖苷 基_β-喊糖普。在某些實施例中,此圓窗膜黏膜黏附劑為It is also contemplated in the context of the examples to incorporate a round window film mucoadhesive to the corticosteroid formulations and compositions disclosed herein. The term "mucosal binder" refers to a substance used for binding to the mucin layer of a biofilm, such as the outer membrane of a 3-layer circular window membrane. "As a circular window film mucoadhesive polymer, this The poly(n) material should have certain general physicochemical characteristics such as anionic hydrophilicity with predominantly several hydrogen bond forming groups, suitable surface properties to wet the mucus/mucosal tissue surface and sufficient elasticity to penetrate the mucosal network. Round window film mucoadhesives that can be used with acceptable ears include, but are not limited to, soluble ketone ratios of 11 ketone polymers (pvp); one water _ swellable but water-insoluble, fiber , cross-linked county · functional polymer; a cross-linked poly(acrylic acid) (such as carbomer 947P); - carbomer homopolymer; a carbomer copolymer; - hydrophilic glycan gum, malt Dextrin, a crosslinked alginate gel, a water-dispersible polycarboxylate ethylene polymer, at least two particulate components selected from the group consisting of titanium dioxide, sulphur dioxide, and clay or mixtures thereof. 140447.doc 74· 200950787 Round® Membrane Adhesives can be used in combination with an acceptable viscous excipient for use in the ear or alone to increase the interaction of the composition with the ear component of the mucosal layer. In a non-limiting embodiment, the mucoadhesive agent is maltodextrin and/or alginate gum. When used, the circular window film imparts a mucoadhesive property to the composition that is sufficient to deliver an effective amount of the corticosteroid composition to, for example, the inner ear round window mucosa or the cochlear cap crown, and the amount of which is a coatable mucosa, • and then deliver the composition to the affected area, including the example of the inner ear and/or the deaf structure as illustrated. Those skilled in the art can determine the characteristics of the mucoadhesive agent of the compositions described herein and can therefore determine the appropriate amount of use. A method of determining adequate mucoadhesiveness can include monitoring changes in the interaction of the composition with the mucosal layer, including but not limited to measuring the duration or duration of retention of the composition in the absence or presence of the excipient. In a non-limiting embodiment, the round window membrane mucoadhesive agent is maltodextrin. Maltodextrin is a carbohydrate produced by hydrolyzing starch derived from corn, potato, wheat or other plants. Maltodextrin can be used alone or in combination with other round window film mucoadhesives to impart the properties of the mucosal adhesives disclosed herein. In one embodiment, the combination of maltodextrin and carbomer polymer enhances the round window film mucoadhesive of the compositions disclosed herein. In another embodiment, the round window membrane mucoadhesive agent is an alkyl-glycoside and/or a monoalkyl ester. As used herein, an "alkyl-glycoside" refers to a compound containing any hydrophilic sugar (eg, glucose, fructose, curtain sugar, or maltose) attached to a hydrophobic alkyl group. In certain embodiments, this The round window membrane mucoadhesive agent is an interfacial activity containing an alkyl-glycoside 140447.doc • 75· 200950787 'wherein the alkyl-glycoside comprises a linkage to a hydrophobic alkyl group (eg, containing from about 6 to about 25 carbons) a sugar of an atom of an alkyl group, which is linked by a monoamine, an amine linkage, an amine citrate linkage, a hydrazine linkage, a thiol linkage, a thiol linkage, a thioester linkage, a glycosidic linkage a monothiol linkage, and/or a urethral urea linkage. In certain embodiments, the round window membrane mucoadhesive agent is hexyl-, heptyl-, octyl-, decyl-, fluorenyl-, ten Monoalkyl...dodecyl-, tridecyl-, tetradecyl, pentadecyl-, hexadecyl-, heptadecyl- and octadecyl alpha- or beta-D- Maltoside; hexyl-, heptyl-, octyl-, decyl-, decyl-, undecyl-, dodecyl-, tridecyl-, tetradecyl, ten Alkyl-, hexadecyl-, heptadecyl-, and octadecanthyl-α- or β-D-glucoside; hexyl-, heptyl-, octyl-, fluorenyl-, fluorenyl- , undecyl-, dodecyl-, tridecyl-, tetradecyl, pentadecyl-, hexadecyl-, heptadecyl-, and octadecyl-- or β-D-sucrose glycosides; hexyl-, heptyl, octyl-, dodecyl-, dodecanyl- and tetradecyl-β-D-thiomaltoside; heptyl- or octyl-丨_Sulfur_α- or β-D-gluconoside; alkyl sucrose; alkyl maltoside trisaccharide; sucrose β-amino-alkyl ether long-chain aliphatic decylamine; linked by monoamine a base chain of a sugar or is malartenine derivative and an isomalt derivative linked to a home base chain by urea; a long chain aliphatic group of sucrose β-amino-alkyl- Long-chain aliphatic guanidinium carbonate of guanidinium carbonate and sucrose β-amino-alkyl ether. In some embodiments, the round window membrane mucoadhesive agent is an alkyl-glycoside, wherein the alkyl glucoside is a combination of maltose, sucrose, glucose, or the like, linked to a single alkyl group of 9 to 16 carbon atoms with a glycoside Chains (eg, decyl-, decyl-, dodecyl-, and tetradecyl sucralose; thiol _, 140447.doc -76- 200950787 thiol-, dodecyl- and tetradecyl glucose Glycosides; and fluorenyl-, decyl-, dodecyl-, and tetradecyl maltosides. In certain embodiments, the round-window film mucoadhesive agent is a burnt-sweet It is a dodecyl maltoside, a tridecyl maltoside, and a tetradecyl maltose. In some implementations, it is acceptable to use a penetration enhancer as a thio-glycoside-containing surfactant, wherein The alkyl glycoside is tetradecyl-β-D-maltose. The round-solid film mucoadhesive agent is a hospital-sugar bitter, wherein the alkyl saccharide is a bile having at least glucose. In certain embodiments, the auris-acceptable penetration enhancer is a surfactant comprising an alpha-D-glucomannan-based peptide, η-dodecyl_4_〇_a_D _ glucopyranosyl-β-glycoside, and/or cardiac tetradecyl _4_〇_a_D_ glucopyranosyl _β- shout sugar. In some embodiments, the round window membrane mucoadhesive agent is

Φ 一烷基-糖苷,其中該烷基糖苷在純水或水溶液中具有一 臨界谬束濃度(CMC)為少於約! mMe在某些實施例中,此 圓窗膜黏膜黏附劑為-烷基-糖苷,在烷基-糖苷中的 氧原子由一硫原子取代。在某些實施例中’此圓窗膜黏膜 黏附劑為-烧基-糖苷,其中該炫基糖芽為^變旋異構物。 在某些實施例巾’此圓窗膜黏膜黏附劑為-烷基-糖苷, 其中該烧基糖苷包含 90〇/〇,91〇/〇,92〇/。,93〇/。,94%, 95% ’ 96% ’ 97% ’ 98% ’ 99%,99 1%,99 5%,或 99 9〇外 變旋異構物。 耳可接受環糊精及其他安定配方 在一特定實施例中’此耳可接受配方亦可替代地包含一 環糊精。此環糊精為含有6、7、或8葡萄喝糖單位環狀募 140447.doc •77- 200950787 酷’分別稱為〇c - ί辰糊精、β - ί哀糊精、或γ - ί哀糊精。壤糊精 具有親水性外層,其增進水-可溶性,及疏水性内層以形 成一腔。在一水性環境中,其他分子疏水性部份通常進入 環糊精的疏水性穴以形成内涵物化合物。此外,環糊精亦 能夠以其他型式與不在疏水性穴内的分子未鍵結交互作 用。環糊精之每一葡萄喃糖單元具有三自由羥基或在α-環 糊精上18羥基、在β-環糊精上21羥基,及在γ-環糊精上24 羥基。此些羥基之一與一以上可與多個試劑之一反應以形 成環糊精衍生物的一較大物種。某些更一般環糊精的衍生 物為羥基丙基醚、磺酸鹽及磺烷基醚。下列顯示為β-環糊 精與羥基丙基β-環糊精(HPpCD)的結構。Φ-Alkyl-glycoside, wherein the alkyl glycoside has a critical enthalpy concentration (CMC) of less than about in pure water or aqueous solution! mMe In certain embodiments, the round window membrane mucoadhesive is an -alkyl-glycoside, and the oxygen atom in the alkyl-glycoside is replaced by a sulfur atom. In certain embodiments, the round window film mucoadhesive agent is an alkyl group-glycoside, wherein the steosyl bud is a spirulina isomer. In some embodiments, the round window film mucoadhesive agent is an alkyl-glycoside wherein the alkyl glycoside comprises 90 Å/〇, 91 〇/〇, 92 〇/. , 93〇/. , 94%, 95% ‘ 96% ‘ 97% ‘ 98% ’ 99%, 99 1%, 99 5%, or 99 9 〇 external rotation isomers. Auricularly Acceptable Cyclodextrins and Other Stabilizing Formulations In a particular embodiment, this ear acceptable formulation may alternatively comprise a cyclodextrin. This cyclodextrin is a sugar-containing unit containing 6, 7, or 8 grapes. 140447.doc •77- 200950787 Cool' respectively called 〇c- ί 糊 糊, β- 哀 糊 糊, or γ - ί Mourning. The soil dextrin has a hydrophilic outer layer which enhances the water-soluble, and hydrophobic inner layer to form a cavity. In an aqueous environment, the hydrophobic portion of other molecules typically enters the hydrophobic pockets of the cyclodextrin to form the intrinsic compound. In addition, cyclodextrins can also interact with other types of molecules that are not in hydrophobic pockets. Each glucomannan unit of the cyclodextrin has three free hydroxyl groups or 18 hydroxyl groups on the α-cyclodextrin, 21 hydroxyl groups on the β-cyclodextrin, and 24 hydroxyl groups on the γ-cyclodextrin. One of these hydroxyl groups reacts with more than one of a plurality of reagents to form a larger species of cyclodextrin derivative. Some of the more general cyclodextrin derivatives are hydroxypropyl ethers, sulfonates and sulfoalkyl ethers. The following shows the structure of β-cyclodextrin and hydroxypropyl β-cyclodextrin (HPpCD).

R = H β-cyclodextrin R = CH2CH(OH)CH3 hydroxypropyl β-cyclodextrin 某些實施例中,在醫藥組成物中使用環糊精以改良藥的 安定性。内涵性化合物有關許多促進溶解度的例子;然 而,其他環糊精與不可溶化合物間的交互作用亦可改良溶 解性。羥基丙基-β-環糊精(HPPCD)為商業可得之無熱原產 品。其為一已可溶解於水的防濕白色粉末。HPPCD為熱安 140447.doc -78- 200950787 在中j·生pH不會降解。因此,環糊精改良組成物或配 的…療山合解性。據此,在某些實施例中,包括環糊 、曰進耳可接$皮質類固醇在本文所述之配方中的溶解 ㈣㈣中’環糊精劑更做為本文所述之配方的 控制釋放賦形劑在。R = H β-cyclodextrin R = CH2CH(OH)CH3 hydroxypropyl β-cyclodextrin In certain embodiments, cyclodextrin is used in the pharmaceutical composition to improve the stability of the drug. Intrinsic compounds are associated with many examples of promoting solubility; however, interactions between other cyclodextrins and insoluble compounds can also improve solubility. Hydroxypropyl-β-cyclodextrin (HPPCD) is a commercially available pyrogen-free product. It is a moisture-proof white powder that is soluble in water. HPPCD is a heat-resistant 140447.doc -78- 200950787 in which the pH does not degrade. Therefore, the cyclodextrin improves the composition or the ... Accordingly, in certain embodiments, the inclusion of a cyclodextrin, a sputum ear, and a dissolution of a corticosteroid in a formulation described herein (4) (d), a cyclodextrin is further used as a controlled release formulation of the formulation described herein. The agent is in.

可使用的較佳之環糊精衍生物包括α_環糊精、卜環糊 精、Y-環糊精、經基乙基β_環糊精、經基丙基γ-環糊精、 硫酸化β環糊精、硫酸切·環_、硫了㈣環糊精。 用於本文料之組絲及^㈣環糊精濃度可依組成中 …療活化劑或其等之鹽或前藥的物化性質、藥物動力學性 質、副作用或不良狀況、配方考量、或伴隨的其他因子、 或其他賦形劑性質而變化。因此,在特定狀況中’本文所 述之組成物及方法的環糊精濃度或量可依需要變化。當使 用時,在本文所述之任一配方中增加皮質類固醇的溶解度 及/或做為控制釋放賦形劑之環糊精的量為使用本說明書 描述之原則、實施例及教示。 ㈢ 可用於本文揭露之耳可接受配方的其他安定劑為選自例 如脂肪酸、脂肪醇、醇、長鏈脂肪酸酯、長鏈醚、脂肪酸 之親水性衍生物、聚乙烯吡咯啶酮、聚乙烯醚、聚乙烯 醇、烴、疏水性聚合物、吸濕性聚合物及其等之組人。 、σ °在 某些實施例中’亦使用安定劑醯胺同類物。在又_實施例 中’選用之安定劑改變此配方的疏水性(例如,油酸 腊),或改良此配方之多種組份的混合(例如,乙醇),押制 配方中的濕度量(例如,PVP或聚乙烯吡咯啶酮),控制相 140447.doc -79- 200950787 的移動(、熔點高於室温的物質如長鏈脂肪酸、醇、醋、 醚、醯胺等、或其等之混合物;腊),及/或改良具有封囊 物質之配方的可相容性(例如,油酸、腊)。在另些其他實 施例中,使用某些安定劑做為溶劑/共_溶劑(例如乙醇)。 在又一實施例中,安定劑的存在量為足以抑制皮質類固醇 的降解。此安定劑的例示包括但未限制為:(a)約〇 5%至 約2% w/v甘油,(b)約〇.1%至約1% w/v胺基甲硫醇丁酸, (c)約0.1%至約2% w/v單硫甘油’(d)約i mM至約i〇 EDTA,(e)約〇‘〇ι%至約2% w/v抗壞血酸,⑴〇 〇〇3%至約 0.〇2% w/v聚山梨醇酯8〇,(g)〇 〇〇1%至約〇 〇5% w/v聚山梨 醇酯2〇,(h)精胺酸,⑴肝素,⑴聚葡糖硫酸鹽,(k)環糊 精,⑴聚戊糖聚硫酸鹽及其他類肝素,(m)二價陽離子如 鎮及鋅;或(η)其等之組合。 額外有用的皮質類固醇耳-可接受配方包括至少一抗凝 聚添加劑以藉由減少蛋白質凝聚以促進皮質類固醇配方的 女疋性。抗凝聚添加劑的選用係依曝露之皮質類固醇,例 如皮質類固醇抗體的條件性質而定。例如,進行授拌及熱 應力的特定配方比進行冷凍乾燥及重組的配方需要不同的 抗凝聚添加劑。有用的抗凝聚添加劑包括僅為例示之用的 腺、氯化胍、簡單胺基酸如甘胺酸或精胺酸、糖、聚醇、 聚山梨醇酯、聚合物如聚乙二醇及聚葡糖、烷基醣、如烷 基糖苷、與界面活性劑。 其他有用的組成物可選擇地包括至少一耳可接受抗氧化 劑以促進需要的化學安定性。合宜之抗氧化劑包括,僅為 140447.doc 200950787 例不之用的抗壞血酸、甲硫胺酸、硫代硫酸鈉與偏二亞硫 酸鈉。在一實施例中,抗氧化劑為選自金屬螯合劑、含硫 醇化合物及其他通用的安定劑。 另外其他有用的組成物包括至少一耳可接受界面活性劑 . X促進物理女疋性或用於其他目的。合宜之非離子性界面 • 活性劑包括聚氧乙烯脂肪酸甘油酯及蔬菜油,例如聚氧乙 ‘· 烯(60)氫化讓蔴油;及聚氧乙烯烷基醚與烷基苯基醚,例 如辛基酚聚醚10、辛基酚聚醚40。 在某些實施例中,本文所述之耳可接受醫藥配方在下列 任何之期間於化合物降解上為安定的:至少約丨天至少 約2天’至少約3天’至少約4天,至少約5天,至少約6 天,至少約1週,至少約2週,至少約3週,至少約4週,至 少約5週,至少約6週’至少約7週,至少約㈣,至少約3 個月,至少約4個月,至少約5個月,至少約6個月。在其 他實施例中,本文所述之配方在至少約旧之期間於化合 ❹ ㈣解上為安定的。且,本文所述之配方在至少約!個月 . 之期間於化合物降解上為安定的。 • 在其他實施财,—額外界面活性劑(共_界面活性劑)及/ 或緩衝劑與至少-前文述所述之醫藥可接受載劑組合使 用,故界面活性劑及/或緩衝劑維持此產品於一安定性的 合適pH。合宜之共-界面活性劑包括但未限制為:⑷天然 及合成親脂劑,例如鱗脂質、膽固醇及膽固醇脂肪酸輯與 其等的衍生物,非離子性界面活性劑,其包括例如聚 氧乙稀脂肪醇醋、山梨醇針脂肪酸酿(Spans) '聚氧乙缔山 H0447.doc -81 . 200950787 梨醇酐脂肪酸酯(例如,聚氧乙烯(20)山梨醇酐單油酸酯 (Tween 80)、聚氧乙烯(20)山梨醇酐單硬脂酸酯(Tween 60)、聚氧乙烯(20)山梨酵酐單月桂酸酯(Tween 20)及其他 Tweens、山梨醇if §旨、甘油醋例如,Myrj及甘油三乙酸醋 (三乙醯甘油)、聚乙二醇、十六烧醇、十六基脂酸基醇、 硬脂酸基醇、聚山梨醇酯80、泊洛沙姆、波洛胺 (poloxamine)、聚氧乙烯篦藏油衍生物(例如,克莫弗® RH40 ' Cremphor A25、Cremphor A20、克莫弗 ®EL)及其 他克莫弗、磺琥珀酸酯、硫酸烷酯(SLS) ; PEG甘油基脂 肪酸醋如PEG-8甘油基辛酸醋/癸酸醋(Labrasol)、PEG-4甘 油基辛酸酯 /癸酸酯(Labrafac Hydro WL 1219)、PEG-32甘 油基月桂酸酯(gelucire 444/14)、PEG-6甘油基單油酸酯 (Labrafil Μ 1944 CS)、PEG-6甘油基亞麻油酸酯(Labrafil Μ 2125 CS);丙二醇單-及二-脂肪酸酯如丙二醇月桂酸 酯、丙二醇辛酸酯/癸酸酯;Brij®700、抗壞血酸基-6-棕 櫚酸酯、硬脂酸基胺、月桂基硫酸鈉、聚氧亞乙基甘油三 衣希油酸(triiricinoleate)及其等之任何組合或混合物;(c) 陰離子性界面活性劑包括但未限制為羧基曱基纖維素鈣、 羧基曱基纖維素鈉、磺琥珀酸鈉、二辛基酯、藻酸鈉、烷 基聚氧乙烯硫酸酯、月桂基硫酸鈉、三乙醇胺硬脂酸酯、 月桂酸鉀、膽鹽及其等之任何組合或混合物;及d)陽離子 界面活性劑如四級錢化合物、氯化苯二曱烴鍵、十六基三 甲基溴化銨及月桂基二曱基笨甲基-氣化銨。 在又一實施例中,當至少一共-界面活性劑用於本發明 140447.doc -82- 200950787 之耳可接受配方時’其與例如’醫藥可接受载劑組合且於 最後的配方中存在量,例如在由約G1%至約織,由約 0.5%至約10%範圍間。 在-實施例中,因為稀釋劑可提供—更安㈣境,亦使 用稀釋劑安定皮質類固醇或其他醫藥化合物。利用溶解在 緩衝溶液中的鹽(其亦提供pH控制或維持)為稀釋劑,其包 括但未限制為構酸醋緩衝生理食鹽水溶液。在其他實施例 Ό φ 中’此凝耀配方為與周邊淋巴及/或内淋巴等張:依皮質 類固醇配方的猫準之標定的耳蜗而定。藉由加入一等張劑 而提供等張配方。適宜的等張劑包括但不限制為任何醫藥 °接又糖1或其等之組合或混合物’如但未限制於葡萄 糖及氯化納。張力劑的量依醫藥配方之標的結構而定,如 本文所描述。 有效的張力組成物包括一或一以上的鹽,其量為引導組 成物的滲透麼至周邊淋巴或内淋巴之可接受範圍所需要的 4 °此鹽包括具有納、卸或録陽離子與氯化物、檸檬酸 鹽、抗壞血酸鹽、蝴酸鹽、罐酸鹽、碳酸氫鹽,硫酸 -硫代硫酸鹽或亞硫酸氯鹽陰離子;合宜之鹽包括氣化 納氯化鉀、硫化碳酸鈉、硫酸氯納及硫酸錢。 在某些實施例中,本發明揭露之耳可接受凝膠配可替代 地或額外含有保存劑以預防微生物生長。可用於本文所述 =增進黏性配方合宜的耳_可接受保存劑包括但未限制為 苯甲^爛酸、p-經基苯曱醋、醇、四級化合物、安定之 氣有機汞,如馬芬(merfen)及硫柳汞、前述之混 140447.doc -83 - 200950787 合物及其相似者。 © 在又一實施例中,僅為例示之用,本文所述之耳可接受 配方中的保存劑為-抗菌劑。在一實施例中,此配方包括 一保存劑如僅為例示之用的甲基對羥基苯曱酸酯、亞硫酸 氫鈉、硫代硫酸納、抗壞血酸鹽、氣化丁醇、乙录硫柳酸 鈉、對羥基苯甲酸酯、苯甲基醇、苯基乙醇及其他者。在 另實細*例中,甲基對羥基苯甲酸酯的濃度為約0 05%至 約1.0%,約0.1%至約〇·2%。在又一實施例中凝膠藉由 此合水、甲基對經基苯曱酸西旨、經基乙基纖維素及摔樣酸 鈉而製備。在又一實施例中,凝膠藉由混合水、甲基對羥 基苯甲酸醋、經基乙基纖維素及乙酸鈉而製備。在又一實 轭例中,混合物在高壓釜中於12〇它滅菌約2〇分鐘且在 與適當量之本文揭露之皮f類固醇混合前測試阳、甲基 對經基苯甲酸酯濃度及黏性。 合且之可用於藥傳遞載劑的水可溶保存劑為亞硫酸氫Preferred cyclodextrin derivatives which may be used include α-cyclodextrin, cyclodextrin, Y-cyclodextrin, transethylidene β-cyclodextrin, transpropyl γ-cyclodextrin, sulfation环cyclodextrin, sulfuric acid cut · ring _, sulfur (four) cyclodextrin. The concentration of the silk and the (4) cyclodextrin used in the present invention may depend on the physicochemical properties, pharmacokinetic properties, side effects or adverse conditions, formulation considerations, or concomitant effects of the therapeutic activator or its salt or prodrug. Other factors, or other excipients, vary in nature. Thus, the concentration or amount of cyclodextrin in the compositions and methods described herein can vary as desired in a particular situation. When used, the solubility of the corticosteroid and/or the amount of cyclodextrin as a controlled release excipient in any of the formulations described herein is the use of the principles, examples, and teachings described herein. (iii) Other stabilizers useful in the auris-acceptable formulations disclosed herein are selected from, for example, fatty acids, fatty alcohols, alcohols, long chain fatty acid esters, long chain ethers, hydrophilic derivatives of fatty acids, polyvinylpyrrolidone, polyethylene A group of ethers, polyvinyl alcohols, hydrocarbons, hydrophobic polymers, hygroscopic polymers, and the like. σ ° In some embodiments ' a stabilizer decyl congener is also used. In another embodiment, the stabilizer selected is used to change the hydrophobicity of the formulation (eg, oleic acid wax), or to improve the mixing of various components of the formulation (eg, ethanol), and to compress the amount of moisture in the formulation (eg, , PVP or polyvinylpyrrolidone), the movement of the control phase 140447.doc -79- 200950787 (a substance having a melting point higher than room temperature such as a long-chain fatty acid, an alcohol, a vinegar, an ether, a guanamine or the like, or a mixture thereof; Wax, and/or improve the compatibility of formulations with encapsulated materials (eg, oleic acid, wax). In still other embodiments, certain stabilizers are used as the solvent/co-solvent (e.g., ethanol). In yet another embodiment, the stabilizer is present in an amount sufficient to inhibit degradation of the corticosteroid. Illustrative of such stabilizers include, but are not limited to, (a) from about 5% to about 2% w/v glycerol, (b) from about 0.1% to about 1% w/v aminomethyl mercaptan butyric acid, (c) from about 0.1% to about 2% w/v monothioglycerol '(d) from about i mM to about i 〇 EDTA, (e) from about 〇 '〇ι% to about 2% w/v ascorbic acid, (1) 〇〇 〇3% to about 0.〇2% w/v polysorbate 8〇, (g)〇〇〇1% to about 〇〇5% w w/v polysorbate 2〇, (h) arginine (1) heparin, (1) polyglucose sulfate, (k) cyclodextrin, (1) polypentose polysulfate and other heparinoids, (m) divalent cations such as town and zinc; or (η) combinations thereof. An additional useful corticosteroid ear-acceptable formulation includes at least one anti-agglomeration additive to promote the virginity of the corticosteroid formulation by reducing protein aggregation. The choice of anti-agglomeration additives is based on the conditional nature of the exposed corticosteroid, such as a corticosteroid antibody. For example, specific formulations for mixing and thermal stress require different anti-agglomeration additives than formulations for freeze-drying and reconstitution. Useful anti-agglomeration additives include glands, cesium chloride, simple amino acids such as glycine or arginine, sugars, polyalcohols, polysorbates, polymers such as polyethylene glycol, and poly-only for illustrative purposes. Glucose, alkyl sugars, such as alkyl glycosides, and surfactants. Other useful compositions optionally include at least one ear acceptable antioxidant to promote the desired chemical stability. Suitable antioxidants include ascorbic acid, methionine, sodium thiosulfate and sodium metabisulfite, which are not used in the 140,470. In one embodiment, the antioxidant is selected from the group consisting of metal chelating agents, thiol containing compounds, and other versatile stabilizers. Still other useful compositions include at least one ear acceptable surfactant. X promotes physical virginity or for other purposes. Suitable non-ionic interface • Active agents include polyoxyethylene fatty acid glycerides and vegetable oils, such as polyoxyethylene olefin (60) hydrogenated to make sesame oil; and polyoxyethylene alkyl ethers and alkyl phenyl ethers, such as xin Polyphenol 10, octylphenol polyether 40. In certain embodiments, an auris-acceptable pharmaceutical formulation described herein is stable on compound degradation during any of the following periods: at least about 2 days 'at least about 3 days' for at least about 4 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 1 week, at least about 2 weeks, at least about 3 weeks, at least about 4 weeks, at least about 5 weeks, at least about 6 weeks 'at least about 7 weeks, at least about (four), at least about 3 Months, at least about 4 months, at least about 5 months, at least about 6 months. In other embodiments, the formulations described herein are stable on the compound 四 (d) during at least about the old period. And, the formula described in this article is at least about! During the month, it is stable on the degradation of the compound. • In other implementations, the additional surfactant (co-interacting agent) and/or buffer is used in combination with at least the pharmaceutically acceptable carrier described above, so the surfactant and/or buffer maintains this The product is at a suitable pH for stability. Suitable co-surfactants include, but are not limited to, (4) natural and synthetic lipophilic agents, such as squamous lipids, cholesterol and cholesterol fatty acid complexes, and the like, nonionic surfactants including, for example, polyoxyethylene Fatty alcohol vinegar, sorbitol needle fatty acid brewing (Spans) 'Polyoxygen Bhd. H0447.doc -81 . 200950787 Pear fatty acid ester (for example, polyoxyethylene (20) sorbitan monooleate (Tween 80 ), polyoxyethylene (20) sorbitan monostearate (Tween 60), polyoxyethylene (20) sorbic acid monolaurate (Tween 20) and other Tweens, sorbitol if §, glycerin For example, Myrj and triacetin (triethyl glycerol), polyethylene glycol, hexadecanol, hexadecanol, stearic acid alcohol, polysorbate 80, poloxamer, Poloxamine, polyoxyethylene sulphonic oil derivatives (eg, CMV® RH40 'Cremphor A25, Cremphor A20, CMUV® EL) and other grammore, sulfosuccinate, alkyl sulfate (SLS); PEG glyceryl fatty acid vinegar such as PEG-8 glyceryl octoate / vinegar (Labrasol), PEG -4 glyceryl caprylate/caprate (Labrafac Hydro WL 1219), PEG-32 glyceryl laurate (gelucire 444/14), PEG-6 glyceryl monooleate (Labrafil® 1944 CS), PEG -6 glyceryl linoleate (Labrafil® 2125 CS); propylene glycol mono- and di-fatty acid esters such as propylene glycol laurate, propylene glycol caprylate/caprate; Brij® 700, ascorbyl-6-palmitic acid Any combination or mixture of ester, stearic acid amine, sodium lauryl sulfate, triiricinoleate, and the like; (c) anionic surfactants including but not limited to carboxyl groups Mercapto cellulose calcium, sodium carboxymethyl cellulose, sodium sulfosuccinate, dioctyl ester, sodium alginate, alkyl polyoxyethylene sulfate, sodium lauryl sulfate, triethanolamine stearate, potassium laurate , any combination or mixture of bile salts and the like; and d) cationic surfactants such as quaternary acid compounds, chlorinated phenyl dihydrocarbyl bonds, hexadecyl trimethyl ammonium bromide and lauryl dimercapto Base - vaporized ammonium. In yet another embodiment, when at least one co-surfactant is used in the inventive acceptable formulation of 140447.doc-82-200950787, it is combined with, for example, a 'pharmaceutically acceptable carrier and is present in the final formulation. , for example, from about G1% to about woven, from about 0.5% to about 10%. In the examples, since the diluent can provide a more secure environment, the diluent is also used to stabilize the corticosteroid or other pharmaceutical compound. The salt dissolved in the buffer solution (which also provides pH control or maintenance) is used as a diluent, which includes, but is not limited to, an acid vinegar buffered physiological saline solution. In other embodiments Ό φ 'this radiant formula is determined by the cochlea of the peripheral lymph and/or endolymphatic isotonic: corticosteroid-based cats. An isotonic formulation is provided by the addition of an isotonic agent. Suitable isotonic agents include, but are not limited to, any combination of pharmaceuticals, sugars, or the like, such as, but not limited to, glucose and sodium chloride. The amount of tonicity agent will depend on the identity of the pharmaceutical formulation as described herein. An effective tonic composition comprising one or more salts in an amount to provide for penetration of the composition to an acceptable range of peripheral lymphoid or endolymphs. The salt comprises sodium, unloaded or recorded cations and chlorides. , citrate, ascorbate, fosphate, pot acid, bicarbonate, sulfuric acid-thiosulfate or sulfite chlorate anion; suitable salts include gasified sodium chloride, sodium carbonate, chlorine sulfate And sulfuric acid money. In certain embodiments, the auris-acceptable gels disclosed herein may alternatively or additionally contain a preservative to prevent microbial growth. Can be used as described herein = suitable for improving the viscosity of the formula _ acceptable preservatives include but not limited to benzoic acid, p- phenyl hydrazine vinegar, alcohol, quaternary compounds, stable gas, organic mercury, such as Merfen and thiomersal, the aforementioned mixtures of 140447.doc-83 - 200950787 and the like. © In still another embodiment, for illustrative purposes only, the preservative in the auris-acceptable formulations described herein is an antibacterial agent. In one embodiment, the formulation includes a preservative such as methyl p-hydroxybenzoate, sodium bisulfite, sodium thiosulfate, ascorbate, gasified butanol, and sulphur Sodium, paraben, benzyl alcohol, phenylethyl alcohol and others. In another example, the methyl paraben concentration is from about 05% to about 1.0%, from about 0.1% to about 0.2%. In still another embodiment, the gel is prepared by the combined water, methyl p-benzoic acid, transethyl cellulose, and sodium sulphate. In still another embodiment, the gel is prepared by mixing water, methyl p-hydroxybenzoate, base ethyl cellulose, and sodium acetate. In yet another embodiment, the mixture is sterilized in an autoclave at 12 Torr for about 2 minutes and tested for cation and methyl p-benzoic acid ester concentrations prior to mixing with an appropriate amount of the skin steroids disclosed herein. Sticky. The water-soluble preservative that can be used as a drug delivery carrier is hydrogen sulfite

鈉、硫代硫酸鈉、抗壞血酸鹽、氯化丁醇、乙汞硫柳酸 鈉、對經基笨f酸醋、苯甲基醇、丁基㈣基二甲苯 (BHT)、苯基乙醇及其他者。此些劑通常的存在量為約 〇顧wm至約5 wt%且較佳為約〇 〇1 wt%至約2 wt%。在 某些實施” ’本文所述之耳可相容配方為無保存劑。 圓窗膜穿透增進劑 另一實施例中,此配方更 進劑。圓窗膜的穿透可藉由 窗膜穿透增進劑為化學實體 包含一或一以上圓窗膜穿透增 穿透增進劑的存在而增進。圓 ,其可促進共投藥物質傳送通 140447.doc • 84 - 200950787 過圓窗膜。圓窗膜穿透增進劑可依化學結構而區分。界面 活性劑,離子性及非_離子性二者,如月桂基硫酸納、月 桂酸鈉、聚氧乙稀-20-十六基醚、Laureth_9、十二烷基硫 酸鈉、二辛基磺基琥珀酸鈉、聚氧乙烯_9_月桂基醚 (PLE)、Tween 80、壬基苯氧基聚乙烯(Npp〇E)、聚山梨 醇醋及其相似者可做為圓窗膜穿透增進劑。膽鹽(如甘膽 酸鈉、去氧膽酸鈉、牛磺膽酸鈉、牛磺二氫梭鏈孢酸鈉、 苷二氫梭鏈孢酸鈉及其相似者)、脂肪酸及衍生物(如油 酸、辛酸、單-及二甘油酯、月桂酸、醯基膽鹼、辛酸、 醯基肉驗、辛酸納及其相似者)、螯合劑(如edta、摔檬 酸、水揚酸鹽及其相似者)、亞颯(如二甲基亞碾 (DMSO),癸基甲基亞石風及其相似者)、及醇(如乙醇、異 丙醇、甘油、丙二醇、聚乙二醇、甘油、丙烧二醇及其相 似者)且做為圓窗膜穿透增進劑。 圓窗膜可穿透脂質體 亦可使用脂質球或脂質粒子以囊化皮質類固冑配方或 組成物。温和分散於水性介質的磷脂質形成多層囊泡,其 具有分離脂質層的圈圍住水性媒介區域。此些多層囊泡的 超音波、或旋渦揽動導致單層囊泡的形成,-般為1約 I0,00·大小的腊質球。此些脂質球具有如皮質類固醇 或其他藥劑載劑般的優點。其為生物惰性、可生物降解 的、無-毒性及無抗原。脂質球可在不同大小形成且在具 有不同的組成物與表面性f。此外,其能夠圈圍住多種= 分子藥且在脂質球破裂的位置釋放藥。 140447.doc -85- 200950787 用於本發明之耳可接受脂質的合宜磷脂質為例如磷脂膽 鹼、乙酵胺及絲胺酸、抱合髓磷脂、心磷脂、縮搭構脂、 磷脂酸及腦苷脂,特別是可用與皮質類固醇溶於無-毒 性、醫藥可接受有機溶劑者。較佳之磷脂質為例如,磷脂 膽鹼、磷脂乙醇胺、磷脂絲胺酸、磷脂環己六醇、脫磷脂膽 鹼、磷脂甘油及其相似者,及其等之混合物,尤其是卵磷 脂,例如大豆卵磷脂。用於本發明配方之磷脂質量可在約i 〇 至約30%,較佳為由約15至約25%範圍間且尤其是約2〇%。 可有利的使用親脂添加劑以選擇性改質脂質球的特性。 此添加劑的範例包括,例如硬脂酸基胺、磷脂酸、維生素 E、膽固醇、膽固醇半水揚酸酯及綿羊油萃取物。使用的 親脂添加劑量範圍由0.5至8%,較佳為由丨5至4%且特別是 約2%。通常,親脂添加劑的量對磷脂質的量之比例範圍 由約1:8至約1:12且特別是約1:1〇。該磷脂質、親脂添加劑 及皮負類固醇及其他醫藥化合物與一無_毒性、醫藥可接 受有機溶劑系統,其可溶解該成份一起使用。該溶劑系統 不僅必需完全溶解該皮質類固醇,且亦必需允許安定單一 雙層脂質球的配方。此溶劑系統包含二曱基異山梨醇酯與 甘醇(四虱吱喃乙一醇趟、四氫π夫喃醇聚乙二醇謎),其 量為約8至約30%。在該溶劑系統中,二甲基異山梨醇酯 與四甘醇量的比例為在由約2:1至約1:3_間,尤其由約 1:1至約1:2.5範圍間且較佳為約1:2範圍間。因此,四甘醇 在最終組成物中的量可在5至2〇%間變化,尤其由5至15% 間且較佳為大約10%。因此,二甲基異山梨醇酯在最終組 140447.doc -86 - 200950787 尤其由3至7 %間且較佳為 成物令的量可在3至ι〇%間變化 大約5% 〇 在後文中使用之"有機組份,•一 為扣含有該磷脂質、親 月曰添加劑及有機溶劑的混合物。 皮質類固醇可溶解於有機 、,且伤或其他方式中以維持此劑 角的凡王活性。皮質類固醇在 最〜配方中的量為在〇1至 。軏圍間。此外,其他成份 如^氧化劑可加至有機組料。範例包括維生”、丁基Sodium, sodium thiosulfate, ascorbate, chlorobutanol, sodium thiomethic acid, sulfonate, benzyl alcohol, butyl (tetra) xylene (BHT), phenylethyl alcohol and others By. Such agents are typically present in amounts ranging from about wm to about 5 wt% and preferably from about 1% to about 2% by weight. In certain embodiments, the ear compatible formulation described herein is a non-preservative. Round window film penetration enhancer In another embodiment, the formulation is more advanced. The penetration of the round window film can be achieved by window film. The penetration enhancer is enhanced by the presence of one or more round window membrane penetration enhancement enhancers by the chemical entity. The circle promotes the co-administration of drug delivery. 140447.doc • 84 - 200950787 Over-window film. Window film penetration enhancers can be distinguished by chemical structure. Surfactants, both ionic and non-ionic, such as sodium lauryl sulfate, sodium laurate, polyoxyethylene -20-hexadecyl ether, Laureth_9 , sodium lauryl sulfate, sodium dioctylsulfosuccinate, polyoxyethylene _9_ lauryl ether (PLE), Tween 80, nonylphenoxy polyethylene (Npp〇E), polysorbate And similar can be used as a round window membrane penetration enhancer. Bile salts (such as sodium glycocholate, sodium deoxycholate, sodium taurocholate, sodium taurostanosporin, dihydrochloride Sodium sphaate and its similarities), fatty acids and derivatives (such as oleic acid, octanoic acid, mono- and diglycerides, lauric acid, guanidine bile , octanoic acid, thiol-based meat, octanoic acid and its similar), chelating agents (such as edta, citric acid, salicylic acid and similar), Aachen (such as dimethyl sub-milling (DMSO), 癸Methyl sulphite and its similarities), and alcohols (such as ethanol, isopropanol, glycerol, propylene glycol, polyethylene glycol, glycerol, propylene glycol and the like) and as a round window membrane penetration Promoters. Round window membranes can penetrate liposomes. Lipid spheres or lipid particles can also be used to encapsulate corticosteroid formulations or compositions. Phospholipids gently dispersed in aqueous mediators form multilamellar vesicles with isolated lipid layers. The circle encloses the aqueous medium region. Ultrasonic waves, or vortexing of such multilamellar vesicles, result in the formation of a single layer of vesicles, typically a waxy ball of about 1 00 00 size. These lipid globules have a cortex like a cortex. A steroid or other pharmaceutical carrier-like advantage. It is biologically inert, biodegradable, non-toxic, and antigen-free. Lipid globules can be formed in different sizes and have different compositions and surface properties f. In addition, they can Enclose a variety of = molecular drugs and where the lipid globules rupture 140447.doc -85- 200950787 Suitable phospholipids for use in the auris-acceptable lipids of the present invention are, for example, phospholipid choline, ethylamine and serine, medullary myelin, cardiolipin, condensate, phospholipid Acids and cerebrosides, especially those which can be dissolved in a non-toxic, pharmaceutically acceptable organic solvent with corticosteroids. Preferred phospholipids are, for example, phospholipid choline, phospholipid ethanolamine, phospholipid serine, phospholipid cyclohexanol, Dephospholipid choline, phospholipid glycerol and the like, and mixtures thereof, especially lecithin, such as soy lecithin. The quality of the phospholipid used in the formulations of the invention may range from about i 〇 to about 30%, preferably by It is between about 15 and about 25% and especially about 2%. It may be advantageous to use a lipophilic additive to selectively modify the properties of the lipid globule. Examples of such additives include, for example, stearic acid amines, phosphatidic acid, vitamin E, cholesterol, cholesterol hemi-salicylate, and lanolin extracts. The amount of lipophilic additive used ranges from 0.5 to 8%, preferably from 5 to 4% and especially about 2%. Generally, the ratio of the amount of lipophilic additive to the amount of phospholipid ranges from about 1:8 to about 1:12 and especially about 1:1 Torr. The phospholipid, lipophilic additive and picosteroid and other pharmaceutical compounds are compatible with a non-toxic, pharmaceutically acceptable organic solvent system which can be used in combination with the component. The solvent system must not only completely dissolve the corticosteroid, but must also allow for the formulation of a single bilayer lipid globule. The solvent system comprises dimercaptoisosorbide and glycol (tetramethylene glycol oxime, tetrahydro π-pentanol polyethylene glycol mystery) in an amount of from about 8 to about 30%. In the solvent system, the ratio of the amount of dimethyl isosorbide to tetraethylene glycol ranges from about 2:1 to about 1:3, especially from about 1:1 to about 1:2.5. Good for about 1:2 range. Thus, the amount of tetraethylene glycol in the final composition can vary from 5 to 2%, especially from 5 to 15% and preferably about 10%. Thus, the amount of dimethyl isosorbide in the final group 140447.doc -86 - 200950787, especially from 3 to 7%, and preferably the amount of the composition, can vary from about 3 to about 10% by 5% 〇 〇 The organic component used in the text, one is a mixture containing the phospholipid, pro-monthly additives and organic solvents. Corticosteroids can be dissolved in organic, and injuring or otherwise to maintain the activity of the king of the agent. The amount of corticosteroid in the most ~ formulation is in 〇1 to . Around the room. In addition, other ingredients such as oxidizing agents can be added to the organic ingredients. Examples include vitamins, butyl

參 ㈣基苯m純基甲笨、抗壞血酸基棕搁酸^ 抗壞血酸基油酸酯及其相似者。 在其他實施例中,此包括凝膠配方與黏性_增進配方之 耳可接受配方更包括賦形劑、其他製藥或藥劑、載劑、佐 劑如保存劑、安定劑、濕化劑或乳化劑、溶液促進劑、 鹽、溶解劑、一抗發泡劑、一抗氧化劑、一分散劑、一濕 化劑、一界面活性劑及其等之組合。 用於本文所述耳可接受配方方之合宜載劑包括但未限制 為任何可與標的耳結構之生理環境相容的醫藥可接受溶 劑。在其他實施例中’此鹼為醫藥可接受界面活性劑與溶 劑的組合。 在某些實施例中,其他賦形劑包括硬脂酸基反丁烯二酸 鈉、一乙醇胺十六基硫酸酯、異硬脂酸酯、聚乙氧化篦蔴 油、壬基氧-l〇(nonoxyM〇)、辛基酚聚醚9、月桂基硫酸 鈉、山梨醇酐酯(山梨醇酐單月桂酸酯、山梨醇酐單油酸 酉曰山籴醇軒單標摘酸酯、山梨醇酐單硬脂酸酯、山梨醇 酐倍半油酸酯、山梨醇酐三油酸酯、山梨醇酐三硬脂酸 140447.doc • 87 · 200950787 醋、山梨醇酐月桂酸酯、山犁 未醇酐油酸酯、山梨醇酐棕櫚 酸酯、山梨醇酐硬脂酸酯、 #醇肝二油酸醋、山梨醇軒 倍半-異硬脂酸酯、山梨醇 卿針倍+硬脂酸酯、山梨醇酐三 異硬脂酸s旨)、其等之印破月 丨嶙知醫樂可接受鹽與其等之組合 或混合物。 在其他實施例中’此载劑為聚山梨醇醋。聚山梨醇醋為 山梨醇㈣的非離子性界面活性劑。可用於本發明揭露之 聚山梨醇醋包括但未限制為聚山梨醇㈣、聚山梨醇醋 40聚山4醇^曰60、聚山梨醇酯8〇(Tween 80)及其等之任 何組合或混合物。在又—實施例中,聚山梨醇醋8〇用於做 為醫藥可接受載劑。 在實施例+,用於製備含有至少—皮質類固醇的醫藥 傳遞載劑之水-可溶甘油-系耳可接受增稠配方含有至少約 0.1%之水-可溶甘油化合物或更多。在某些實施例中皮 質類固醇的百分比在醫藥配方之總重量或總體積為介於約 1%與約95%間,介於約5%與約8〇%間,介於約1〇%與約 60%或更多間變化。在某些實施例中,在每一治療有效皮 質類固醇配方的化合物量之製備方式為可使化合物的任何 特定劑量達到合宜的劑量。在本文考慮的因子如溶解性、 生物可用性 '生物半衰期、投藥路徑、產品保存期及其他 醫藥考量且此醫藥配方的製備。 若需要,此耳可接受醫藥凝膠亦含有共溶劑及緩衝劑。 合宜之水可溶緩衝劑為鹼金屬或鹼土金屬之碳酸鹽、磷酸 鹽、碳酸氫鹽、檸檬酸鹽、硼酸鹽、醋酸鹽、水楊酸鹽及 140447.doc • 88 - 200950787 其相似者,如鋼麟酸鹽、禪檬酸納、蝴酸納、贈酸納、石山 酸氫鈉、碳酸納及三木甲胺納(TRIS)。此些劑的存在量= 足以維持系統之pH在7.4士0.2且較佳為74。故此,緩 可如總組成物之重量計的5%般多β Θ • 然而’有些皮質類_或其他醫藥化合物為不可溶的, · ❹共溶劑以增進皮質類固醇溶解度。此些在合直懸浮助 * 或黏性增進劑幫助下通常懸浮於聚合物載劑中。 .再者,有些醫藥賦形劑、稀釋劑或載劑具潛在耳毒性。 例如丄—常用保存劑,氯化苯二甲烴錄為耳毒性且因此若 用至前庭或耳蜗結構具有潛在的傷害。在調配控制釋放耳 部配方冑議避免或組合適當的醫藥賦形劑、稀釋劑或载 劑以減輕或除去此配方的潛在耳毒性組份,或減少此賦形 劑、稀釋劑或載劑的量。一控制釋放皮質類固醇配方視情 況可包括耳保護劑,如抗氧化劑、α類脂酸、弼、弗司弗 黴素或鐵螯合劑,以抵消因使用特定治療劑或賦形劑、稀 ❹ #劑或載劑而可能引起之潛在耳毒性作用。 治療模式 劑量方法及藥程 傳遞至内耳的藥以口服、靜脈或皮下肌肉系統投藥。然 用於内耳病理位置的系統投藥增加系統毒性及不利副 ===可能性且產生—無成效的藥分布,其在血清中發現 同藥量但在内耳發現為相對較低藥量。 化療兒1的耳内注射為注射-治療劑至耳膜後以進入中耳 或内耳的技術。在—實施例中,本文所描述的配方經 140447.doc -89· 200950787 由耳内注射直接投藥至圓窗膜。在另一實施例中,本文所 描述的皮質類固醇之耳可接受配方經由非_耳内注射方式 投藥至内耳◊在另外的實施例中,本文所描述的配方經由 外科手術方式投藥至圓窗膜,其包含蝸窗脊矯正。 在一實施例中’傳送系統為能夠穿透耳膜並直接進入圓 窗膜或内耳蝎窗脊的注射器及針裝置。在某些實施例中, 此針寬於18號針頭。在另些其他實施例,針頭由18號針至 31號針。在又一實施例中,此針為25號針至3〇號針。依本 文揭硌之皮質類固醇或組成物配方的稍度或黏性,注射器 或皮下注射針的針頭號依此變化。 在另實施例中,此針為一用於凝膠配方立即傳遞的皮 下射針此皮下/主射針為單一使用針或一可抛棄針。在 某二實施例中,一注射器為用於傳遞含本文揭露之組成物 的醫藥可接受凝膠-系含皮質類固醇組成物,其中該注射 器具有一壓配合(Luer)或轉上(Luer_1〇ck)配件。在一實施 例中,此注射器為一皮下注射器。在另些其他實施例,此 /主射器由塑膠或玻璃製成。在又一中,此皮下注射器為一 單使用注射器。在又一實施例中,此玻璃注射器能進行 滅菌作用。在另一實施例中,滅菌作用在高壓釜進行。在 另些其他實施例,此注射器包含一筒狀注射器體,其中貯 存使用前的凝膠配方。在其他實施例中,此注射器包含— 筒狀/主射器體,其中貯存使用前的本文揭露之皮質類固醇 醫藥可接受耳部凝膠-系組成物,其適宜與一合宜之醫藥 可接受緩衝液混合。在其他實施例中,此注射器可含有其 140447.doc 200950787 他賦形劑、安定劍 % 疋幻、懸序劑、稀釋劑或其等之組合以滅 菌,或則可安定目中i 存匕S在其中的皮質類固醇或其他醫藥 化合物。 在某:實施例中’此注射器包含一筒狀注射器體,其中 該/主射11體隔間’且每一隔間可貯存耳-可接受皮質類固 酵凝膠配方的至少-崎。在又—實施财,此具有隔間 :/主射器體的 >主射器在注射入中耳或内耳前允許混合組Reference to (tetra) phenyl benzene m pure base, ascorbyl palmoside acid ascorbyl oleate and the like. In other embodiments, this includes gel formulations and viscous-promoting formulations, and the acceptable formulations of the ear include excipients, other pharmaceutical or pharmaceutical agents, carriers, adjuvants such as preservatives, stabilizers, humidifiers or emulsifiers. A combination of a agent, a solution accelerator, a salt, a solvent, an anti-foaming agent, an antioxidant, a dispersing agent, a wetting agent, a surfactant, and the like. Suitable carriers for use in the auris-acceptable formulations described herein include, but are not limited to, any pharmaceutically acceptable solvent that is compatible with the physiological environment of the standard otic structure. In other embodiments, the base is a combination of a pharmaceutically acceptable surfactant and a solvent. In certain embodiments, other excipients include sodium stearate fumarate, monoethanolamine hexadecane sulfate, isostearate, polyethoxylated castor oil, decyloxy-l ( nonoxyM〇), octylphenol polyether 9, sodium lauryl sulfate, sorbitan ester (sorbitol monolaurate, sorbitan monooleate, saponin, single-labeled acid ester, sorbitan Monostearate, sorbitan sesquioleate, sorbitan trioleate, sorbitan tristearate 140447.doc • 87 · 200950787 vinegar, sorbitan laurate, sorbitol Anhydride oleate, sorbitan palmitate, sorbitan stearate, #alcoholic dioleic acid vinegar, sorbitol xanthene-isostearate, sorbitol needles + stearate And sorbitan triisostearic acid s), and the like, or a combination or mixture thereof. In other embodiments, the carrier is polysorbate. Polysorbate vinegar is a nonionic surfactant of sorbitol (IV). Polysorbate vinegars useful in the present disclosure include, but are not limited to, polysorbate (tetra), polysorbate 40 polysorbate 4, polysorbate 8 (Tween 80), and the like or mixture. In yet another embodiment, polysorbate 8 is used as a pharmaceutically acceptable carrier. In Example +, the water-soluble glycerin-tether acceptable thickening formulation for the preparation of a pharmaceutical delivery vehicle containing at least a corticosteroid comprises at least about 0.1% water-soluble glycerin compound or more. In certain embodiments, the percentage of corticosteroids is between about 1% and about 95% of the total weight or total volume of the pharmaceutical formulation, between about 5% and about 8%, and between about 1% and about Approximately 60% or more varies. In certain embodiments, the amount of the compound in each therapeutically effective dermal steroid formulation is prepared in such a way that a particular dosage of the compound will result in a suitable dosage. Factors considered herein such as solubility, bioavailability, biological half-life, route of administration, product shelf life, and other medical considerations and preparation of this pharmaceutical formulation. If desired, the ear acceptable pharmaceutical gel also contains a cosolvent and a buffer. Suitable water soluble buffers are alkali metal or alkaline earth metal carbonates, phosphates, hydrogencarbonates, citrates, borates, acetates, salicylates and 140447.doc • 88 - 200950787 Such as steel sulphate, sodium citrate, sodium sulphate, sodium sulphate, sodium hydrogen sulphate, sodium carbonate and triclosan (TRIS). These agents are present in an amount sufficient to maintain the pH of the system at 7.4 ± 0.2 and preferably 74. Therefore, it is as much as 5% of the total composition of the weight of β Θ • However, some corticals or other pharmaceutical compounds are insoluble, ❹ co-solvent to promote corticosteroid solubility. These are typically suspended in a polymeric carrier with the aid of a colloidal suspension aid or a viscosity enhancer. Furthermore, some pharmaceutical excipients, diluents or carriers have potential ototoxicity. For example, sputum, a commonly used preservative, is expected to be ototoxic and therefore potentially harmful if applied to the vestibular or cochlear structures. In the formulation of a controlled release ear formulation, it is contemplated to avoid or combine a suitable pharmaceutical excipient, diluent or carrier to reduce or remove the potential ototoxic component of the formulation, or to reduce the excipient, diluent or carrier. the amount. A controlled release corticosteroid formulation may optionally include an ear protectant such as an antioxidant, alpha-fat, bismuth, forskolin or an iron chelator to counteract the use of a particular therapeutic agent or excipient, dilute # Potential ototoxic effects that may be caused by agents or carriers. Treatment Mode Dose Method and Pharmacy The drug delivered to the inner ear is administered orally, intravenously or subcutaneously. However, systemic administration for the pathological location of the inner ear increases systemic toxicity and adverse side effects === possibility and produces - ineffective drug distribution, which is found in serum but is found to be relatively low in the inner ear. The intra-oral injection of Chemotherapy 1 is an injection-therapeutic agent to the eardrum to enter the middle or inner ear. In the examples, the formulations described herein are administered directly to the round window membrane by intralesional injection via 140447.doc -89.200950787. In another embodiment, the corticosteroid-acceptable formulations described herein are administered to the inner ear sputum via a non-intra-injection. In additional embodiments, the formulations described herein are surgically administered to the round window membrane. It contains vortex ridge correction. In one embodiment, the delivery system is a syringe and needle device that is capable of penetrating the eardrum and directly into the round window membrane or inner ear ridge. In some embodiments, the needle is wider than the 18 gauge needle. In still other embodiments, the needle is from the 18 gauge needle to the 31 gauge needle. In yet another embodiment, the needle is a 25 gauge to a 3 gauge needle. The needle number of the syringe or hypodermic needle varies depending on the mildness or viscosity of the corticosteroid or composition formulation disclosed herein. In another embodiment, the needle is a subcutaneous needle for immediate delivery of the gel formulation. The subcutaneous/primary needle is a single use needle or a disposable needle. In a second embodiment, a syringe is a pharmaceutically acceptable gel-containing corticosteroid composition for delivering a composition comprising the present invention, wherein the syringe has a press fit (Luer) or turn on (Luer_1〇ck ) Accessories. In one embodiment, the syringe is a hypodermic syringe. In still other embodiments, the /ejector is made of plastic or glass. In still another, the hypodermic syringe is a single use syringe. In yet another embodiment, the glass syringe is capable of being sterilized. In another embodiment, the sterilization is carried out in an autoclave. In still other embodiments, the syringe comprises a cylindrical syringe body in which the gel formulation prior to use is stored. In other embodiments, the syringe comprises a cylindrical/main emitter body, wherein the corticosteroid pharmaceutically acceptable ear gel-based composition disclosed herein is stored prior to use, which is suitable for a suitable pharmaceutical acceptable buffer Liquid mixing. In other embodiments, the syringe may contain a combination of its excipients, diazepam, suspending agent, diluent, or the like, for sterilization, or may be stabilized in the eye. Among them are corticosteroids or other pharmaceutical compounds. In a certain embodiment, the syringe comprises a cylindrical syringe body, wherein the/main shot 11 body compartment' and each compartment stores at least an acne of the ear-acceptable corticosteroid formulation. In the case of the implementation of the money, this compartment with a : / main body * > the main emitter is allowed to mix before injection into the middle or inner ear

份。在其他實施例中,此傳遞系統包含多個注射器,多個 注射器的每-注射器含有凝膠配方的至少—組份,故每一 組份在注射前預混合或在接著注射後混合。在又—實施例 中本文揭露之注射器含有至少_貯器,其中至少—貯器 包含-皮質類固醇、或一醫藥可接受緩衝液、或一黏性增 進劑如-膠化劑或其等之組合。可選擇使用商業可得之注 射裝置可以其最簡單形式之已可使用的塑膠注射器,其具 有注射H筒、具針的針總成件、具柱塞桿的柱塞、及握持 緣,以進行耳内注射。 #在某些實施例中’此傳遞裝置為―設計用於將治療劑投 藥至中耳及/或内耳的裝置。僅為例示說明之用:gyrusShare. In other embodiments, the delivery system comprises a plurality of syringes, each syringe of the plurality of syringes containing at least a component of the gel formulation such that each component is premixed prior to injection or mixed after subsequent injection. In still another embodiment, the syringe disclosed herein contains at least a reservoir, wherein at least the reservoir comprises a corticosteroid, or a pharmaceutically acceptable buffer, or a viscous enhancer such as a gelling agent or a combination thereof. . Alternatively, a commercially available injection device can be used in its simplest form, a plastic syringe having an injection H-tube, a needle assembly with a needle, a plunger with a plunger rod, and a gripping edge to Intrathecal injection. # In certain embodiments, the delivery device is a device designed to administer a therapeutic agent to the middle ear and/or inner ear. For illustrative purposes only: gyrus

Medi^ GmbH提供冑_耳内視鏡以用於觀測及藥傳遞至内 耳圓窗室;Arenberg在美國專利第5,421,818 ; 5,474,529 ; 及5,476,446號描述-醫療用治療裝置以傳遞液體至内耳結 構其等s併入本案做為參考。美國專利申請案第 〇8/874,208號描述一外科手術方法,其植入一傳送導管傳 送以傳送治療劑至内耳,其揭露併入本案做為參考。美國 -9J - 140447.doc 200950787 專利申請案公開第2007/0167918號進一步揭露用於耳内液 體取樣及醫療應用的組合耳部吸引器及醫藥分配器,其揭 露併入本案做為參考。 本文所述之配方及投藥模式亦可應用至内耳空間的直接 滴入或灌流方法。因此,本文所述之配方亦可用於外科手 術中,非限制範例包括封閉開口、迷徑切開術、耳乳突鑿 開術、鐙骨足板切除術、内淋巴球囊切開術或其相似者。 含有本文所述之皮質類固醇化合物的耳可接受組成物用 於預防及/或醫療治療給藥。在醫療應用中,皮質類固醇 組成物投藥予已患有疾病、症狀或失調的患者,其量為足 以治癒或或至少部份制止疾病、失調或症狀的病症。在此 些使用的有效量將依疾病、失調或症狀的嚴重性及病程、 先前的治療、患者的健康狀態及對藥的回應,與有經驗的 醫療人員判斷而決定。 在患者症狀並未改進的例子中’依醫生自行判斷,皮質 類固醇化合物投藥可以長期方式給藥,亦即一延長時間, 包括在患者生命期間以改善、或者控制或限制患者之疾病 或症狀的症候群。 在患者症狀確實改進的例子中,依醫生自行判斷,皮質 類固醇化合物投藥可以持續方式給藥;或者,給藥劑量可 暫時降低或暫時停止一特定長度的時間(亦即,一「藥假 期」)。藥假期的長度可在2天至1年間變化,包括僅為例 示之用的範例為2天、3天、4天、5天、6天、7天、10天、 12天、15 天、20天、28 天、35 天、50 天、70天、1〇〇天、12〇 140447.doc -92- 200950787 天、150天、180天、200天、250天、280天、300天、320 天350天及365天。在藥假期減少的劑量由丨, 包括僅為例示之用的範例為1〇%、15%、2〇%、25%、 30%、35%,4G%、45%、5G%、55%、6G%、65%、70%、 75%、80%、85%、90%、95%,及 100%。 一旦已產生患者的自體免疫症狀改進,若需要則給予一 -·、维持劑量。接[依症候群作用T減少投藥的劑量或頻 *® 率,或一者至一維持改善的疾病、失調或症狀。在特定實 細例中心者在長間依症狀的任何再現性可能需要間 隔的治療。 對應此量的特定劑的皮質類固醇量將依因子如特定化合 物、疾病症狀及嚴重性而變化,依病例有關的特定狀況以 熟於此技人士已知的方式常規的決定,包括例如特定劑的 投藥、投藥路徑、治療的症狀’及治療的個體或主體。然 而,通常用於治療成人的治療劑量基本上每投藥在〇 〇2_5〇 ❹ 範圍間,較佳為每投藥在1-15 mg範圍間。預期的劑量可 合宜的以單一劑量或分開劑量同時(或在-短時間)或間隔 的投藥。 在某些實施例中,最初投藥為一特定皮質類固醇且後績 投藥一不同配方或皮質類固醇。 控制釋放配方的藥動力學 在一實施例中,本文揭露之配方額外提供一由此配方的 皮質類固醇立即釋放’或在i分鐘内’或在5分鐘内,或在 10分鐘内’或在15分鐘内’或在3G分鐘内,或在6〇分鐘内 140447.doc -93· 200950787 或在90分鐘内。在其他實施例中,一治療有效量之至少一 皮質類固醇由此配方立即釋放,或在i分鐘内,或在5分鐘 内,或在1〇分鐘内,或在15分鐘内,或在30分鐘内,或在 60分鐘内或在90分鐘内。在特定實施例中此配方包含一 提供立即釋放至少-皮質類㈣的耳·醫藥可接受凝膠配 方。此配方的額外實施例亦可包括—促進本文揭露配方之 黏性的劑。 在其他或另外的實施例中,此配方提供至少_皮質類固 醇的控制釋放配方。在特定實施財,由配方擴散至少一 皮質類固醇在一超過5分鐘,或15分鐘,或30分鐘,或H、 時,或4小時,或6小時,或12小時,或i8小時,或1天, 或2天’或3天,或4天’或5天’或6天,或7天或ι〇天, 或12天,或天’或18天,或21天,或25天或3〇天或 45天,或2個月或3個月或4個月或5個月或6個月或9個月或 1年時間由配方擴散至少-皮質類固醇。在其他實施例 中,由配方釋放至少—皮質類㈣的治療有效量在-超過 5分鐘’或15分鐘’或3〇分鐘,或*寺,或4小時,或6小 時,或12小時,或18小時,或1天,或2天,或3天,或4天, 或5天’或6天’或7天’或1()天,或12天,或14天或μ 天或21天或25天,或3〇天,或45天或2個月或3個月 或4個月或5個月或6個月或9個月或丨年時間由配方釋放至 少一皮質類固醇的治療有效量。 在其他實施例中’此配方提供—皮f類固醇之立即釋放 及-延長釋放的配方。在又另—實施例中,此配方含有 140447.doc •94- 200950787 0.25:1比例,或一 〇.5:1比例,或一 1:1比例,或一丨:2比 例,或一 1:3比例’或一 1:4比例,或一 ! :5比例,或—1 ·7 比例,或一 1:10比例,或一 1:15比例,或一 1:2〇比例之立 即釋放及延長釋放的配方。在又一實施例中,此配方提供 . 第—皮質類固醇之立即釋放及第二皮質類固醇或其他治療 . 劑的延長釋放。在又另一實施例中,此配方提供至少一耳 ·· 劑之立即釋放及一延長释放的配方,與至少一治療劑。在 φ 某些實施例中,此配方提供一 〇.25」比例,或一 〇.5:1比 例,或一 1··1比例,或一 1:2比例,或一 1:3比例,或—卜4 比例,或一1:5比例,或一 1:7比例,或一 1:1〇比例,或一 1:15比例,或一 1:2〇比例之第一皮質類固醇與第二治療劑 之分別立即釋放與延長釋放配方。 在又一實施例中,此配方於疾病位置提供一治療有效量 的至少一皮質類固醇且實質上無系統曝露。在又一實施例 中,此配方在疾病位置提供一治療有效量的至少一皮質類 ® 固醇而無可測得的系統曝露。在其他實施例中,在疾病位 £提供—治療有效量的至少—耳劑而具少量或無可測得的 系統曝露。 皮質類固醇組成物或配方的立即釋放、延遲釋放及/或 延長釋放之組合可與本文揭露之其他醫藥劑、以及賦形 劑 '稀釋劑、安定劑、張力劑及其他組份組合。故此,依 使用的皮質類固醇、預期的稠度及或黏性、或選擇的傳遞 模式可依本文揭露之實施例的另一態樣與立即釋放,延 遲釋放及/或延長釋放之實施例結合。 140447.doc -95- 200950787 在特定實施例中’本文所述之皮質類固醇配方的藥物動 力學係藉由注入此配方於或接近測試動物(範例包天竺鼠 或粟鼠)的内耳圓窗膜而測定。在一量測時間(例如在長達 1週期間於6小時、12小時、1天、2天、3天、4天、5天、6 天、及7天測試配方的藥物動力學),此動物安樂死並以 5 ml周邊淋巴液體試樣測試。移出内耳並測試皮質類固醇 的存在。如需要,測定皮質類固醇劑在其他器官的量。此 . 外,皮質類固醇的系統量由測試動物中取出血液試樣而測 疋。為了測疋此配方是否妨礙聽力,測試動物聽力可選擇 © 的測試。 了替代也^供内耳(如由測試動物取出)及測量皮質 類固酵的移動。如另一可卷抑本 IB .u 力 j督代者,提供一内耳圓窗膜的活 體外模型及測量皮質類固醇的移動。 製造套件/物件 本發明亦提供一預防、户、麻弋奸至士, ^ /α療或改善哺乳動物的疾病或 調之症候群的套件。此套件Medi^ GmbH provides a 胄 _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ Etc. s incorporated into this case for reference. U.S. Patent Application Serial No. 8/874,208, the disclosure of which is incorporated herein by reference in its entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire entire all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all all A combined ear aspirator and medical dispenser for in-the-ear liquid sampling and medical applications is further disclosed in U.S. Patent Application Publication No. 2007/0167918, the disclosure of which is incorporated herein by reference. The formulations and modes of administration described herein can also be applied to direct instillation or perfusion methods in the inner ear space. Thus, the formulations described herein can also be used in surgery, and non-limiting examples include closed openings, blunt incisions, ear papillary severing, humeral pedicleectomy, endolymphatic balloon incision, or the like. . An auris-acceptable composition comprising a corticosteroid compound as described herein is for use in prophylactic and/or medical treatment. In medical applications, the corticosteroid composition is administered to a patient already suffering from a disease, condition or disorder in an amount sufficient to cure or at least partially arrest the condition, disorder or condition. The effective amount to be used in these settings will be determined by the judgment of an experienced medical professional, depending on the severity and duration of the disease, disorder or condition, prior treatment, patient health and response to the medication. In the case where the patient's symptoms have not improved, 'at the discretion of the physician, the administration of the corticosteroid compound can be administered in a long-term manner, that is, for an extended period of time, including syndromes that improve, or control or limit the disease or symptom of the patient during the life of the patient. . In the case where the patient's symptoms are indeed improved, the corticosteroid compound administration can be administered in a continuous manner at the discretion of the doctor; or, the dose can be temporarily lowered or temporarily stopped for a certain length of time (ie, a "drug holiday"). . The length of the drug holiday can vary from 2 days to 1 year, including the examples for example only 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 Day, 28 days, 35 days, 50 days, 70 days, 1 day, 12〇140447.doc -92- 200950787 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days 350 days and 365 days. The doses reduced during the drug holiday are based on 丨, including examples for illustrative purposes only 1%, 15%, 2%, 25%, 30%, 35%, 4G%, 45%, 5G%, 55%, 6G%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%. Once the patient's autoimmune symptoms have improved, a dose of -, maintenance is given if needed. Depending on the symptom of the syndrome T, reduce the dose or frequency of the drug, or one to maintain an improved disease, disorder or symptom. Any reproducibility of symptoms in the center of a particular case may require a treatment of the interval. The amount of corticosteroid corresponding to a particular amount of this agent will vary depending on factors such as the particular compound, the condition and severity of the disease, and will be conventionally determined in a manner known to those skilled in the art, including, for example, a particular agent. Administration, route of administration, symptoms of treatment' and the individual or subject of treatment. However, the therapeutic dose usually used to treat an adult is generally in the range of 〇2_5〇 每 per dose, preferably between 1 and 15 mg per dose. The intended dose may conveniently be administered simultaneously (or at - short time) or at intervals in a single dose or in divided doses. In certain embodiments, the initial administration is a specific corticosteroid and a different formulation or corticosteroid is administered. Pharmacokinetics of a Controlled Release Formulation In one embodiment, the formulations disclosed herein additionally provide an immediate release of a corticosteroid from the formulation 'either within 1 minute' or within 5 minutes, or within 10 minutes' or at 15 Within minutes or within 3G minutes, or within 140 minutes of 140447.doc -93.200950787 or within 90 minutes. In other embodiments, a therapeutically effective amount of at least one corticosteroid is immediately released by the formulation, or within i minutes, or within 5 minutes, or within 1 minute, or within 15 minutes, or at 30 minutes. Inside, or within 60 minutes or within 90 minutes. In a particular embodiment, the formulation comprises an o-medical acceptable gel formulation that provides immediate release of at least - cortex (tetra). Additional embodiments of this formulation may also include agents that promote the viscosity of the formulations disclosed herein. In other or additional embodiments, the formulation provides a controlled release formulation of at least a _corticosteroid. In a specific implementation, the formulation diffuses at least one corticosteroid for more than 5 minutes, or 15 minutes, or 30 minutes, or H, hour, or 4 hours, or 6 hours, or 12 hours, or i8 hours, or 1 day. , or 2 days 'or 3 days, or 4 days' or 5 days' or 6 days, or 7 days or ι〇 days, or 12 days, or days 'or 18 days, or 21 days, or 25 days or 3 weeks At least - corticosteroids are diffused from the formulation by day or 45 days, or 2 months or 3 months or 4 months or 5 months or 6 months or 9 months or 1 year. In other embodiments, the therapeutically effective amount of at least - cortic (IV) is released by the formulation at - more than 5 minutes 'or 15 minutes' or 3 minutes, or *Temple, or 4 hours, or 6 hours, or 12 hours, or 18 hours, or 1 day, or 2 days, or 3 days, or 4 days, or 5 days 'or 6 days' or 7 days' or 1 () days, or 12 days, or 14 days or μ days or 21 days Or 25 days, or 3 days, or 45 days or 2 months or 3 months or 4 months or 5 months or 6 months or 9 months or leap years, the treatment of at least one corticosteroid is effective the amount. In other embodiments, this formulation provides an immediate release and extended release formulation of the skin steroid. In still another embodiment, the formulation contains 140447.doc •94-200950787 0.25:1 ratio, or a .5:1 ratio, or a 1:1 ratio, or a 丨:2 ratio, or a 1: 3 ratio 'or a 1:4 ratio, or one! : 5 ratio, or -1 · 7 ratio, or a 1:10 ratio, or a 1:15 ratio, or a 1:2 〇 ratio of immediate release and extended release formulations. In yet another embodiment, the formulation provides for immediate release of a first-corticosteroid and a second corticosteroid or other treatment. Extended release of the agent. In yet another embodiment, the formulation provides immediate release and an extended release formulation of at least one of the agents, with at least one therapeutic agent. In certain embodiments of φ, the formulation provides a ratio of .25", or a ratio of .5:1, or a ratio of 1·1, or a ratio of 1:2, or a ratio of 1:3, or - Bu 4 ratio, or a 1:5 ratio, or a 1:7 ratio, or a 1:1 ratio, or a 1:15 ratio, or a 1:2 ratio of the first corticosteroid and the second treatment The immediate release and extended release formulations of the agents. In yet another embodiment, the formulation provides a therapeutically effective amount of at least one corticosteroid at the disease site and is substantially free of systemic exposure. In yet another embodiment, the formulation provides a therapeutically effective amount of at least one corticosteroid at the disease site without measurable systemic exposure. In other embodiments, a therapeutically effective amount of at least an auricular agent is provided at the disease site with little or no measurable systemic exposure. Combinations of immediate, delayed, and/or extended release of a corticosteroid composition or formulation can be combined with other pharmaceutical agents disclosed herein, as well as excipients, diluents, stabilizers, tonicity agents, and other components. Thus, depending on the corticosteroid used, the desired consistency and/or viscosity, or the selected mode of delivery, another aspect of the embodiments disclosed herein can be combined with embodiments of immediate release, delayed release, and/or extended release. 140447.doc -95- 200950787 In a particular embodiment, the pharmacokinetics of the corticosteroid formulations described herein are determined by injecting this formulation into or near the inner ear round window membrane of a test animal (example scorpion or squirrel). . The pharmacokinetics of the formulation are tested at a measurement time (eg, 6 hours, 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, and 7 days for up to 1 week). Animals were euthanized and tested with 5 ml peripheral lymphatic fluid samples. Remove the inner ear and test for the presence of corticosteroids. Determine the amount of corticosteroid in other organs, if needed. In addition, the systemic amount of corticosteroids was measured by taking blood samples from the test animals. To test whether this formula interferes with hearing, test the animal's hearing to select the test for ©. The replacement is also provided for the inner ear (as removed from the test animal) and for measuring the movement of the corticosteroid. If another can suppress this IB.u force, provide an in vitro model of the inner ear window membrane and measure the movement of corticosteroids. Manufacturing Kit/Item The present invention also provides a kit for preventing, warding, numbing, or treating a disease or a symptom of a mammal. This kit

安仟逋*包含至少—本文揭露之 質類固醇可控制釋放的έ 孜的組成物,及使用此套件的指示 明。本發明亦預期將—或一 ^ 乂上之皮質類固醇可控制釋 的組成物用於治療、減少、 ..,At β ^ 降低及改善哺乳動物如人類 疾病、功能異常、或类裀+十Μ f 乳動物如人i§l古·症候群的醫療製劑的製造,該 礼動物如人類具有、懷疑 ^ ^ , , ^ X仕赞展為内耳失調的風險。 在某些實施例中,套杜— 娶件包括一載劑、包装、 隔間以容納一或—以 衣 A合益 -容器包括-分離元件 及其相似者 件以用於本文所述之方法。合宜之 140447.doc •96· 200950787 器包括例如,瓶、小瓶、注射器及測試管。在其他實施例 中,此容器由多種物質如玻璃或塑膠形成。Ampoule* contains at least the composition of the steroid-controlled release oxime disclosed herein and instructions for using the kit. The present invention also contemplates the use of a corticosteroid-controlled release composition of - or a sputum for the treatment, reduction, . . . , At β ^ reduction and improvement in mammals such as human diseases, dysfunction, or genital genus f The manufacture of medical preparations for milk animals such as human i§l ancient syndromes, such as human beings, suspected of ^ ^ , , ^ X praised as the risk of inner ear disorders. In certain embodiments, the sleeve includes a carrier, a package, a compartment to accommodate one or - a garment A - the container includes - a separation element and the like for use in the methods described herein . Suitable 140447.doc • 96· 200950787 includes, for example, bottles, vials, syringes, and test tubes. In other embodiments, the container is formed from a variety of materials such as glass or plastic.

本發明提供之製造物件包含包裝物質。用於包裝本發明 醫藥產品的包裝物質。參閱,例如美國專利第5,323,907、 5,052,558及5,033,252號。醫藥包裝物質的範例包括但未 限制為罩板包裝、瓶、管、吸入器、泵、袋、藥瓶、容 器、注射器、瓶、及任何適宜選擇之配方及投藥與治療的 所用模式之包裝物質。本發明提供之皮質類固醇組成物的 廣序列配方為預期做為任何疾病、失調、或症狀之多種治 療,其優點在於藉由皮質類固醇的控制釋放投藥至内耳。 在某些實施例中,一套件基本上包括一或一以上額外容 器,其每一者具有由商業及使用者觀點為用於本文所述配 方的不同物質(如試劑,可選擇為濃縮型式及/或裝置)。此 物質的非限制性範例包括但未限制於缓衝液、稀釋劑、濾 器、針、注射器、載劑、包裝、容器、小瓶及/或管標籤 標明含量及/或使用指示、及具有使用指示的包裝插入。 基本上包括一組說明指示。 實施例 實施例1-熱可逆凝膠甲基脫氫皮質固醇配方的製備 量(配方之mg/g ) 20.0 1.0 10.0 180.0 789.0 成份The article of manufacture provided by the present invention comprises a packaging material. A packaging material for packaging the pharmaceutical product of the present invention. See, for example, U.S. Patent Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any suitable formulation and packaging materials for the mode of administration and treatment. . The broad sequence formulation of the corticosteroid composition provided by the present invention is intended to be a multitude of treatments for any disease, disorder, or condition, which has the advantage of being administered to the inner ear by controlled release of corticosteroids. In certain embodiments, a kit substantially comprises one or more additional containers, each having a different substance (eg, a reagent, optionally in a concentrated form, and used in the formulations described herein from a commercial and user perspective) / or device). Non-limiting examples of such materials include, but are not limited to, buffers, diluents, filters, needles, syringes, carriers, packs, containers, vials, and/or tube labels indicating levels and/or instructions for use, and instructions for use. The package is inserted. Basically includes a set of instructions. EXAMPLES Example 1 - Preparation of a Thermoreversible Gel Methyl Dehydrocorticosterol Formulation (Formulation mg/g) 20.0 1.0 10.0 180.0 789.0 Ingredients

甲基脫氫皮質固醇 曱基對羥基苯曱酸酯 HPMC 泊洛沙姆407 TRISHC1 缓衝劑(0.1M) 140447.doc -97 200950787 一含有2.0%之甲基脫氫皮質固醇的凝膠配方之10-g批料 藉由將1.80 g泊洛沙姆407(BASF公司)懸浮於5.00 g TRIS HC1緩衝劑(0.1 Μ)中並在4°C下振動過夜以確保完全溶解 而製備。加入曱基脫氫皮質固醇(200·0 mg)、羥基丙基甲 基纖維素(100.0 mg)、曱基對羥基苯甲酸酯(10 mg)與額外 的TRIS HC1緩衝劑(0.1 M)(2_89 g)並再攪拌直至觀察到完 全溶解。此混合物維持低於室温直至使用。 實施例2 -黏膜黏附劑、熱可逆凝膠去氫皮質醇配方的製備 量(配方之mg/g ) 30 1.0 10.0 2.0 180.0 787.0 成份 去氫皮質醇 曱基對羥基苯曱酸酯 HPMC 卡波莫934P 泊洛沙姆407 TRIS HC1 緩衝劑(0.1 M) 一含有2.0%之去氫皮質醇之黏膜黏附劑、凝膠配方之 ΙΟ-g批料藉由將2.0 mg卡波莫934P與1.80 g泊洛沙姆 407(BASF公司)懸浮於5.00 g TRIS HC1緩衝劑(0.1 M)中並 在4°C下振動過夜以確保完全溶解而製備。加入去氫皮質 醇、羥基丙基曱基纖維素(100.0 mg)、曱基對羥基苯曱酸 酯(10 mg)與額外的TRIS HC1緩衝劑(0.1 M)(2_87 g)並再 攪拌直至觀察到完全溶解。此混合物維持低於室温直至使 用。 實施例3 -含環糊精之熱可逆凝膠2.5%甲基脫氫皮質固醇 之配方的製備 140447.doc •98- 200950787 量(配方之mg/g) 500.0 1.0 180.0 317.0 成份 5% CD溶液 曱基對羥基苯曱酸酯 泊洛沙姆407 TRISHC1 缓衝劑(0.1M) 泊洛沙姆407(BASF公司)懸浮於TRIS HC1緩衝劑(0.1 Μ) 中且組份在4°C下振動混合過夜以確保完全溶解。加入環 糊精溶液及甲基對經基苯甲酸g旨並再搜拌直至觀察到完全 溶解。此混合物維持低於室温直至使用。 實施例4 -含環糊精之黏膜黏附劑、熱可逆凝膠甲基脫氫 皮質固醇之配方的製備 量(配方之mg/g) 500.0 1.0 180.0 2.0 317.0Methyl dehydrocorticosterol hydroxybenzoate HPMC Poloxamer 407 TRISHC1 Buffer (0.1M) 140447.doc -97 200950787 A gel containing 2.0% methyl dehydrocorticosterol The 10-g batch of the formulation was prepared by suspending 1.80 g of poloxamer 407 (BASF) in 5.00 g TRIS HC1 buffer (0.1 Torr) and shaking at 4 °C overnight to ensure complete dissolution. Add decyl dehydrocorticosterol (200·0 mg), hydroxypropyl methylcellulose (100.0 mg), decyl paraben (10 mg) and additional TRIS HC1 buffer (0.1 M) (2_89 g) and stir again until complete dissolution was observed. This mixture was kept below room temperature until use. Example 2 - Preparation of mucoadhesive, thermoreversible gel dehydrocortisol formulation (mg/g of formula) 30 1.0 10.0 2.0 180.0 787.0 Ingredients dehydrocortisol thiol p-hydroxybenzoate HPMC Kapomo 934P Poloxamer 407 TRIS HC1 Buffer (0.1 M) A mucoadhesive containing 2.0% dehydrocortisol, a gel formulation of ΙΟ-g batch by 2.0 mg of Carbomo 934P and 1.80 g Losham 407 (BASF Corporation) was suspended in 5.00 g of TRIS HC1 buffer (0.1 M) and shaken at 4 ° C overnight to ensure complete dissolution. Add dehydrocortisol, hydroxypropyl decyl cellulose (100.0 mg), decyl p-hydroxybenzoate (10 mg) and additional TRIS HC1 buffer (0.1 M) (2_87 g) and stir until observed To completely dissolve. This mixture was kept below room temperature until use. Example 3 - Preparation of a cyclodextrin-containing thermoreversible gel 2.5% methyl dehydrocorticosterol formulation 140447.doc •98- 200950787 Amount (formulation mg/g) 500.0 1.0 180.0 317.0 Ingredient 5% CD solution Mercapto-p-hydroxybenzoate poloxamer 407 TRISHC1 buffer (0.1M) Poloxamer 407 (BASF) was suspended in TRIS HC1 buffer (0.1 Μ) and the component vibrated at 4 ° C Mix overnight to ensure complete dissolution. A cyclodextrin solution and methyl p-benzoic acid g were added and re-mixed until complete dissolution was observed. This mixture was kept below room temperature until use. Example 4 - Preparation of a cyclodextrin-containing mucoadhesive, thermoreversible gel methyl dehydrogenation Corticol formulation (mg/g of formula) 500.0 1.0 180.0 2.0 317.0

5% CD溶液 甲基對羥基苯甲酸酯 泊洛沙姆407 卡波莫934P TRISHC1 緩衝劑(0.1M) 卡波莫934P及泊洛沙姆407(BASF公司)懸浮於TRIS HC1 緩衝劑(0.1 Μ)中且組份在4°C下振動混合過夜以確保完全 溶解。加入環糊精溶液及甲基對羥基苯甲酸酯並再攪拌直 至觀察到完全溶解。此混合物維持低於室温直至使用。 實施例5 -含微米化甲基脫氫皮質固醇之熱可逆凝膠甲基 脫氫皮質固醇之配方的製備 成份5% CD solution methyl paraben poloxamer 407 carbomer 934P TRISHC1 buffer (0.1M) Carbomo 934P and poloxamer 407 (BASF) suspended in TRIS HC1 buffer (0.1 The mixture was shaken and mixed at 4 ° C overnight to ensure complete dissolution. The cyclodextrin solution and methyl paraben were added and stirred until complete dissolution was observed. This mixture was kept below room temperature until use. Example 5 - Preparation of a Thermoreversible Gel Methyl Dehydrocorticosterol Formulation Containing Micronized Methyl Dehydrocorticosterol

甲基脫氫皮質W BHT 泊洛沙姆407 PBS緩衝劑(0.1 M) 量(配*方之mg/g) 20.0 0.002 160.0 9.0 140447.doc -99· 200950787 一 10-g批料的2.0%微米化甲基脫氫皮質固醇、13.8 mg 鱗·酸一鈉一水合物USP (Fisher Scientific公司)+3.1 mg構酸 納單水合物USP (Fisher Scientific公司)+74 mg氣化鈉USP (Fisher Scientific公司)的凝膠配方溶解於8.2 g滅菌過濾DI 水中並以1 M NaOH將pH調節至7.4。冰涑此緩衝劑溶液並 於攪拌下灑入1.6 g泊洛沙姆407(BASF公司,其含有約1〇〇 ppm BHT)至此冰凍PBS溶液,混合此溶液直至所有泊洛沙 姆溶解。此泊洛沙姆使用33 mm PVDF 0.22 μιη滅菌注射 過濾器(Millipore公司)滅菌過濾並於一無菌環境傳輸至2 ml 滅菌的玻璃小瓶(Wheaton),此小瓶以滅菌丁基橡膠栓 (Kimble)密封並以13 mm A1封(Kimble)捲邊。20 mg之微米 化甲基脫氫皮質固酵(Spectrum chemicals)置於一分離的乾 淨去熱源小瓶中,此小瓶以滅菌丁基橡膠栓(Kimble)密封 並以13 mm A1封(Kimble)捲邊,小瓶於140。〇進行乾熱滅 菌處理(Fisher Scientific Isotemp烘箱)7小時。在進行本文 所描述的實驗投藥前,使用附在1 mL滅菌注射器(Bect〇nMethyl dehydrogenated cortex W BHT poloxamer 407 PBS buffer (0.1 M) Amount (with mg/g of * square) 20.0 0.002 160.0 9.0 140447.doc -99· 200950787 2.0% micron of a 10-g batch Methyl dehydrocorticosterol, 13.8 mg Sodium citrate monohydrate USP (Fisher Scientific) + 3.1 mg sodium hydrate monohydrate USP (Fisher Scientific) + 74 mg sodium hydride USP (Fisher Scientific The company's gel formulation was dissolved in 8.2 g of sterile filtered DI water and the pH was adjusted to 7.4 with 1 M NaOH. This buffer solution was iced and sprinkled with 1.6 g of Poloxamer 407 (BASF, containing about 1 ppm of BHT) to this frozen PBS solution, and the solution was mixed until all the poloxamers dissolved. This poloxamer was sterilized and filtered using a 33 mm PVDF 0.22 μιη sterile syringe filter (Millipore) and transferred to a 2 ml sterilized glass vial (Wheaton) in a sterile environment. The vial was sealed with a sterile butyl rubber stopper (Kimble). It is crimped with a 13 mm A1 (Kimble). 20 mg of micronized methyl dehydrocorticochemical (Spectrum chemicals) was placed in a separate clean de-heated vial sealed with a sterilized butyl rubber stopper and crimped with a 13 mm A1 (Kimble) , the vial is at 140. The crucible was subjected to dry heat sterilization (Fisher Scientific Isotemp oven) for 7 hours. Use the 1 mL sterile syringe (Bect〇n) before performing the experimental administration described in this article.

Dickinson)的 21G針頭(Becton Dickinson)傳送 1 ml冷泊洛沙 姆溶液至含有20 mg滅菌之微米化的曱基脫氫皮質固醇的 小瓶中,藉由振盪充分混合懸浮掖以確保懸浮液的均質 性。此懸浮液接著以21G注射器退出且針頭換成27 g針頭 以用於投藥。 實施例6 -含穿透增進劑之熱可逆凝膠的微米化普賴松配 方的製備 140447.doc • 100· 200950787 量(配方之mg/g) 20.0 1.0 1.0 10.0 180.0 789.0 成份 普賴松Dickinson's 21G needle (Becton Dickinson) delivers 1 ml of cold poloxamer solution to a vial containing 20 mg of sterilized micronized sulfhydryl dehydrocorticosterol, and the suspension is thoroughly mixed by shaking to ensure the suspension Homogenization. This suspension was then withdrawn using a 21G syringe and the needle was replaced with a 27 g needle for administration. Example 6 - Preparation of micronized Prysson formulation with a thermoreversible gel containing a penetration enhancer 140447.doc • 100· 200950787 Amount (mg/g of formula) 20.0 1.0 1.0 10.0 180.0 789.0 Ingredients Presson

曱基對羥基苯甲酸酯 十二烷基麥芽糖苷(A3) HPMC 泊洛沙姆407 TRISHC1 緩衝劑(0.1M) 一含有2.0%之微米化普賴松的凝膠配方之ΙΟ-g批料藉由 將1.80 g泊洛沙姆407(BASF公司)懸浮於5.00 g TRIS HC1 φ 缓衝劑(0.1 Μ)中而製備且組份在4°c下振動過夜以確保完 全溶解。加入普賴松(200.0 mg)、經基丙基曱基纖維素 (100.0 mg)、甲基對羥基苯甲酸酯(10 mg)及十二烷基麥芽 糖苷(10 mg)與額外的TRIS HC1緩衝劑(0.1 M)(2.89 g)並 再攪拌直至觀察到完全溶解。此混合物維持低於室温直至 使用。 實施例7 -pH在高壓釜處理之於PBS緩衝劑中17%泊洛沙姆 407 NF/2%甲基脫氫皮質固醇磷酸酯(DSP)的效用 Ο 一 17%泊洛沙姆407/2%甲基脫氫皮質固醇磷酸酯(DSP) . 的原料溶液藉由溶解35 1.4 mg氯化納(Fisher Scientific)、 302.1 mg 填酸二納無水物(Fisher Scientific)、122.1 mg 構 酸鈉無水物(Fisher Scientific)及2.062 g甲基脫氫皮質固醇 磷酸酯(DSP)與一適當量耳劑於79.3 g滅菌過濾DI水而製 備。此溶液在一冰凍水浴中冷卻並接著灑入17.05 g泊洛沙 姆407 NF(Spectrum Chemicals)至冷溶液中同時混合。混合 物進一步混合直至泊洛沙姆完全溶解。測定此溶液的pH。 140447.doc -101 - 200950787 17%泊洛沙姆407/2%曱基脫氫皮質固醇磷酸酯(DSP)於 PBS中,pH 5.3。取前述溶液的一部份(大約30 mL)並加 入1 M HC1以調整pH至5.3。 17%泊洛沙姆407/2%曱基脫氫皮質固醇磷酸酯(DSP)於 PBS中,pH 8.0。取前述原料溶液的一部份(大約30 mL)並 加入1 M NaOH以調整pH至8。 PBS緩衝液(pH 7.3)藉由溶解805·5 mg氯化鈉(Fisher Scientific),606 mg填酸二鈉無水物(Fisher Scientific)、 247 mg鱗酸納無水物(Fisher ScientiHe),接著以滅菌過渡 DI水適量至200 g而製備。 一 pH 7.3之2%於PBS的甲基脫氫皮質固醇磷酸酯(DSP) 溶液藉由溶解適當量的甲基脫氫皮質固醇磷酸酯(DSP)於 PBS緩衝液中並以PBS緩衝液適量至10 g而製備。 1 mL試樣分別置於3 mL螺旋蓋玻璃小瓶中(以橡膠襯裡) 並緊密密封。此小瓶置於Market Forge-Sterilmatic高壓爸 (設定,緩慢液態)並在250°F滅菌1 5分鐘。在高壓釜作用 後,此試樣靜置冷卻至室温接著置於冰箱中。當冷卻時此 試樣藉由混合此小瓶而均質之。 觀察及記錄外觀(例如,脫色及/或沉澱)。2% DSP單獨 於PBS中顯示為脫色(淡黃色)及部份沉澱,同時含有泊洛 沙姆的試樣未顯示脫色現象。在含有泊洛沙姆的試樣中, 僅在具有pH為5.3的試樣中觀察到沉澱。 HPLC分析使用附有Luna C18(2) 3 μιη、ΙΟΟΑ、250x4.6 mm 管柱之Agilent 1200,其使用30-80乙腈梯度(1-10 min)(含有 140447.doc -102- 200950787 0.05% TFA水-乙腈混合物)進行,總試驗15分鐘。記錄主要的峰 如下表所示。試樣藉由取用30 μί試樣並溶解於1.5 ml之1:1丙 腈水混合物中稀釋。試樣的純度在高壓蚤處理前為大於99%。 表1在含有甲基脫氫皮質固醇磷酸鈉(DSP)的試樣之高壓 釜處理後觀察的性質 外觀 % DSP % Dex (RRT=1.27) 1.54 之 %RRT 1.68 之 %RRT 2%DSPpH=7.3 淡黃色 89 6.5 1.41 17% 407/2 %DSP PBS, pH-5.3 沉澱 53 45.9 0.48 0.56 17% 407/2% DSP PBS > pH=7.3 清澈溶 液/凝膠 88 10 0.79 17% 407/2% DSP PBS > pH=8.0 清澈溶 液/凝膠 92 4.9 1.18 - 所有試樣的純度在高壓釜處理前為99+%。 實施例8 -高壓釜處理在PBS緩衝液中17%泊洛沙姆407NF/ 2%甲基脫氫皮質固醇磷酸酯(DSP)的釋放曲線與黏性之效用。 實施例6之試樣(高壓釜處理及未經高壓釜處理)的一分 量用於評估釋放曲線及黏性量測以評估加熱滅菌作用在凝 膠性質上的衝擊。 在Snap well s( 6.5 mm直徑之具有孔洞大小為0.4 μηι的聚 碳酸酯膜)中於37°C進行溶解。0.2 mL凝膠配方置於 Snapwell中並靜置至硬化,接著0.5 mL緩衝液置於貯器中 並使用Labline執道震盪器於在70 rpm震盪。每小時取出試 樣(取出0.1 mL並以温緩衝液替代)。使用硫氰酸鈷方法以 624 nm UV分析試樣的泊洛沙姆濃度,並參考一外部校正 標準曲線。簡言之,20μΕ試樣與1980 pL之15mM硫氰酸鈷 140447.doc • 103 · 200950787 溶液混合並使用Evolution 160 UV/Vis光譜儀(ThermoMercapto-p-hydroxybenzoate dodecyl maltoside (A3) HPMC Poloxamer 407 TRISHC1 buffer (0.1M) A gel formulation containing 2.0% micronized Prysson Prepared by suspending 1.80 g of Poloxamer 407 (BASF) in 5.00 g TRIS HC1 φ buffer (0.1 Torr) and the components were shaken overnight at 4 °C to ensure complete dissolution. Adding Prysson (200.0 mg), propyl propyl decyl cellulose (100.0 mg), methyl paraben (10 mg) and dodecyl maltoside (10 mg) with additional TRIS HC1 Buffer (0.1 M) (2.89 g) and stir again until complete dissolution was observed. This mixture is kept below room temperature until use. Example 7 - pH 17% poloxamer 407 NF/2% methyl dehydrocorticosteroid phosphate (DSP) in autoclave treatment in PBS buffer Ο a 17% poloxamer 407/ 2% methyldehydrocorticosteroid phosphate (DSP). The raw material solution was dissolved by 35 1.4 mg of sodium chloride (Fisher Scientific), 302.1 mg of di-n-hydrate (Fisher Scientific), 122.1 mg of sodium silicate. Anhydrous (Fisher Scientific) and 2.062 g of methyl dehydrocorticosteroid phosphate (DSP) were prepared by sterilizing and filtering DI water with 79.3 g of an appropriate amount of ear. This solution was cooled in a freezing water bath and then sprinkled into 17.05 g of poloxamer 407 NF (Spectrum Chemicals) into a cold solution while mixing. The mixture is further mixed until the poloxamer is completely dissolved. The pH of this solution was measured. 140447.doc -101 - 200950787 17% poloxamer 407/2% decyl dehydrocorticosteroid phosphate (DSP) in PBS, pH 5.3. A portion (about 30 mL) of the above solution was taken and 1 M HCl was added to adjust the pH to 5.3. 17% poloxamer 407/2% decyl dehydrocorticosteroid phosphate (DSP) in PBS, pH 8.0. A portion (about 30 mL) of the above raw material solution was taken and 1 M NaOH was added to adjust the pH to 8. PBS buffer (pH 7.3) was prepared by dissolving 805. 5 mg sodium chloride (Fisher Scientific), 606 mg acid disodium anhydride anhydrate (Fisher Scientific), 247 mg sodium sulphate anhydrate (Fisher Scienti He), followed by sterilization. Prepared by converting the amount of DI water to 200 g. A 2% pH 7.3 solution of methyl dehydrocorticosteroid (DSP) in PBS by dissolving the appropriate amount of methyl dehydrocorticosterol phosphate (DSP) in PBS buffer and in PBS buffer Prepared by appropriate amount to 10 g. The 1 mL sample was placed in a 3 mL screw cap glass vial (lined with rubber) and tightly sealed. This vial was placed in a Market Forge-Sterilmatic high pressure dad (set, slow liquid) and sterilized at 250 °F for 15 minutes. After the autoclave was applied, the sample was allowed to stand to cool to room temperature and then placed in a refrigerator. This sample was homogenized by mixing the vial when cooled. Observe and record the appearance (eg, discoloration and/or precipitation). 2% DSP alone showed discoloration (light yellow) and partial precipitation in PBS, while samples containing poloxamer did not show discoloration. In the sample containing poloxamer, precipitation was observed only in the sample having a pH of 5.3. HPLC analysis using Agilent 1200 with Luna C18(2) 3 μιη, ΙΟΟΑ, 250 x 4.6 mm column using a 30-80 acetonitrile gradient (1-10 min) containing 140447.doc -102- 200950787 0.05% TFA The water-acetonitrile mixture was carried out for a total test time of 15 minutes. Record the main peaks as shown in the table below. The sample was diluted by taking a 30 μL sample and dissolving it in 1.5 ml of a 1:1 propionitrile water mixture. The purity of the sample was greater than 99% prior to high pressure helium treatment. Table 1 Properties observed after autoclaving of a sample containing methyl dehydrocortisol sodium phosphate (DSP). % Appearance % DSP % Dex (RRT = 1.27) 1.5% of RRT 1.68 % RRT 2% DSPpH = 7.3 Light yellow 89 6.5 1.41 17% 407/2 % DSP PBS, pH-5.3 Precipitation 53 45.9 0.48 0.56 17% 407/2% DSP PBS > pH=7.3 Clear solution/gel 88 10 0.79 17% 407/2% DSP PBS > pH = 8.0 Clear Solution / Gel 92 4.9 1.18 - The purity of all samples was 99 + % before autoclaving. Example 8 - Effect of autoclave treatment on the release profile and viscosity of 17% poloxamer 407NF / 2% methyl dehydrocorticosteroid phosphate (DSP) in PBS buffer. A component of the sample of Example 6 (autoclave treatment and no autoclave treatment) was used to evaluate the release profile and viscosity measurement to evaluate the impact of heat sterilization on the properties of the gel. Dissolution was carried out at 37 ° C in a Snap well s (6.5 mm diameter polycarbonate film having a pore size of 0.4 μηι). The 0.2 mL gel formulation was placed in Snapwell and allowed to stand until hardened, then 0.5 mL of buffer was placed in the reservoir and shaken at 70 rpm using a Labline Oscillator. The sample was taken every hour (0.1 mL was taken and replaced with warm buffer). The poloxamer concentration of the sample was analyzed using a cobalt thiocyanate method at 624 nm UV and referenced to an external calibration standard curve. Briefly, a 20 μΕ sample was mixed with 1980 pL of 15 mM cobalt thiocyanate 140447.doc • 103 · 200950787 solution and used with the Evolution 160 UV/Vis spectrometer (Thermo

Scientific)在625 nm測量吸收度。 釋放耳劑曱基脫氫皮質固醇磷酸酯(DSP)符合Korsmeyer-Scientific) The absorbance was measured at 625 nm. Release of the ear thiol dehydrocorticosteroid phosphate (DSP) in accordance with Korsmeyer-

Peppas等式 Q = ktn + bPeppas equation Q = ktn + b

Qx 〇 其中Q為耳劑在時間t釋放的量’ Qa為耳劑的總釋放 量,k為第η次順序的釋放常數,η為一有關於溶解機制的 無因次數且b為軸截距,其特徵在於最初突釋機制,其中 n=l特指一侵蝕控制機制。平均溶解時間(MDT)為藥分子 在釋放前於不同時間期間停留在基體内的總和,除以總分 子數且由下式計算: MDr =心 it + 1 黏性測定使用一附有一水夹套温度控制單元(温度於15-34°C 以 1.6°C /min斜率增加)的 Brookfield黏度計 RVDV-II+P 在0.08 rpm(剪力速率為0.31 s·1)旋轉之CPE-51心轴下進 行。T凝膠(Tgel)定義為因膠溶體-凝膠轉換而產生的黏性 增加之曲線反曲點。 表2高壓釜處理在PBS緩衝液中17%泊洛沙姆407NF/2%甲基 脫氫皮質固酵磷酸鈉(DSP)的釋放曲線舆黏性之效用。 MDT(hr) TgelfC) Max黏性 * (Pas) 未經高壓爸處理 3.2 25 403 高壓爸處理. 3.2 26 341 *於一剪力速率0.31s·1在凝膠態(高至37°C)的最大外觀黏性 140447.doc •104· 200950787 此結果顯示在高壓釜處理於PB S緩衝液中17%泊洛沙姆 407 NF/2%甲基脫氫皮質固醇磷酸鈉(DSP)後釋放曲線與黏 性之微小效用。 實施例9 -加入第二聚合物於含有2%甲基脫氫皮質固醇磷酸 酯(DSP)與17%泊洛沙姆407 NF的配方在熱滅菌作用(高壓釜 處理)後之降解產物舆黏性的效用。 溶液A. — pH 7.0之於PBS緩衝液中含有羧基曱基纖維素 納(CMC)溶液藉由溶解 178.35 mg氯化納(Fisher Scientific)、 300.5 mg麟酸二納無水物(Fisher Scientific)、126.6 mg攝 酸納無水物(Fisher Scientific)於78.4 g滅菌過滤DI水而製 備,接著灑入 1 g Blanose 7M65 CMC (Hercules,黏性 5450cP @ 2°/。)至緩衝液溶液並加熱以助於溶解,且此溶液接著冷 卻下來。 一 pH 7.0 之於 PBS 的 17% 泊洛沙姆 407 NF/1% CMC/2% 曱 基脫氫皮質固醇磷酸鈉(DSP)藉由在冷凍水浴中冷卻8.1 g 溶液A並接著加入205 mg甲基脫氫皮質固醇磷酸鈉(DSP) 且接著混合而製得。1.74g泊洛沙姆407 NF(Spectrum Chemicals)灑入至冷溶液中同時混合。混合物進一步混合 直至泊洛沙姆完全溶解。 2 ml前述試樣置於3 mL螺旋蓋玻璃小瓶中(以橡膠襯裡) 並緊密密封。此小瓶置於Market Forge-Sterilmatic高壓爸 (設定’緩慢液態)並在250°F滅菌25分鐘。在高壓釜作用 後,此試樣靜置冷卻至室温接著置於冰箱中。此試樣藉由 當小瓶冷卻時均質混合。 140447.doc -105- 200950787 在經高壓釜處理後未觀察到沉澱或脫色。進行如實施例 6描述之HPLC分析。檢測到因曱基脫氫皮質固醇產物水解 而少於1%的降解產物,即此配方在經高壓釜處理後安 定。 依實施例7描述進行黏性測量。結果顯示高壓釜處理對 凝膠的黏性或Tgel温度具些微的影響。在含有泊洛沙姆的 配方與對照組(2% DSP於PBS中)比較觀察到為較少的整體 不純物。 依實施例7描述進行溶解測試。結果顯示為11.9 hr之 MDT相較於不含CMC的配方之3.2的MDT。CMC或一二級 聚合物的加入為導入一減少曱基脫氫皮質固醇之釋放速率 (即,增加MDT)的擴散屏蔽。 實施例10緩衝液型式於含有泊洛沙姆407NF的配方在熱滅 菌作用(高壓釜處理)後之降解產物的效用。 一 TRIS緩衝液藉由溶解377.8 mg氣化鈉(Fisher Scientific)及 602.9 mg 三 丁醇醋(Tromethamine,Sigma Chemical公司)接著以滅菌過渡DI水適量至100 g而pH以 1 M HC1調整至7.4。 含有25%於TRIS緩衝液泊洛沙姆407溶液的原液: 稱重45 g TRIS緩衝液,在冷凍浴中冰凍,接著灑入15 g 泊洛沙姆407 NF(Spectrum Chemicals)至緩衝液中同時混 合。此混合物進一步混合直至泊洛沙姆完全溶解。 於PBS緩衝液之含有25%泊洛沙姆407溶液之原液(pH 7.3):溶解 704 mg 氯化鈉(Fisher Scientific)、601.2 mg 填 140447.doc •106- 200950787 酸二納無水物(Fisher Scientific)、242.7 mg磷酸鈉無水物 (Fisher Scientific)於140.4 g滅菌過慮DI水中。此浴液在一冰 凍水浴中冷卻並接著灑入5 0 g泊洛沙姆407 NF (SPECTRUM CHEMICALS)至冷溶液中同時混合。混合物進一步混合直 至泊洛沙姆完全溶解,且可得到一清澈透明的溶液。測量 獲得之溶液的pH為7.3。 / 以上述原液製備一系列配方。於所用實驗中使用得自 . Spectrum chemicals公司的甲基脫氫皮質固醇構酸醋(DSP) 及微米化曱基脫氫皮質固醇USP。 1 ml試樣分別置於3 mL螺旋蓋玻璃小瓶中(以橡膠襯裡) 並緊密密封。此小瓶置於Market Forge-Sterilmatic高壓爸 (設定,緩慢液態)並在250°F滅菌25分鐘。在高壓釜作用 後,此試樣靜置冷卻至室温接著置於冰箱中。此試樣藉由 當小瓶冷卻時均質混合。 進行如實施例6描述之HPLC分析。比較在TRIS與PBS 緩衝劑中的安定性。 表3緩衝液型式於含甲基脫氫皮質固醇與甲基脫氫皮質固醇 磷酸酯的配方在降解的效用 試樣編號 外觀 1 % DSP % Dex (RRT=1.27) 1.54 之 %RRT 1.68 之 %RRT 1%DSP/TRIS 淡黃色 &沉澱 41 54 2.68 0.97 2% DSP/TRIS 淡黃色 &沉殿 41 55 2.4 0.57 4% DSP/TRIS 淡黃色 &沉澱 58 36 2.4 0.23 16%P407/2DSP/TRIS 沉澱 54 41 0.79 0.62 18% P407/2DSP/TRIS 沉澱 52 45 1.21 0.51 140447.doc -107- 200950787 20% P407/2DSP/TRIS 沉澱 55 43 0.86 0.49 18% P407/2DEX/TRIS 懸浮液 - 99 0.22 0.55 2% DEX/TRIS 懸浮液 - 99 - 0.28 2% DSP/PBS 淡黃色 &沉澱 85.6 9.8 2.09 - 16% P407/2DSP/PBS 清澈溶液 78 17.5 1.86 - 18% P407/2DSP/PBS 清澈溶液 81.2 16.2 1.14 - 20% P407/2DSP/PBS 清澈溶液 81.5 16.1 1.04 - 18% P407/2DEX/PBS 懸浮液 - 97 1.06 0.45 2% DEX/PBS 懸浮液 - 94.7 1.34 - 黏性量測依所實施例7描述者進行。 結果顯示為了減少高壓釜處理期間的水解作用,緩衝劑 在增温時需要維持pH在7-8範圍間。在TRIS缓衝劑中觀察 到比在PBS中增加的藥水解性(表3)。其他降解產物的發生 可藉由使用在本文中描述的聚合物添加劑(如P407)而減 少。由含有20%泊洛沙姆407的配方與不含泊洛沙姆407的 配方之比較可觀察到在降解產物上的減少(表7)。 含有懸浮微米化之曱基脫氫皮質固醇的配方在高壓釜處 理時具有比其溶液相對組更大的安定性 實施例11 :脈衝釋放耳部配方 使用曱基脫氫皮質固醇及曱基脫氫皮質固醇磷酸鈉 (DSP)( 1:1比例)的組合依本文所述之的製程製備一脈衝釋 放劑配方。甲基脫氫皮質固醇之可傳送劑量之20%於β環 糊精幫助下溶解於實施例7之17%泊洛沙姆溶液。可傳送 之甲基脫氫皮質固醇劑量之剩餘之80%接著加至混合物中 且依本文所述之任何製程製備最終配方。 Ρ依本文所述之實施例的製程製備含有曱基脫氫皮質固 醇的脈衝釋放配方並使用本文所述之步驟測試以測定脈衝 140447.doc -108- 200950787 釋放曲線。 實施例12: 17%泊洛沙姆407/2% DSP劑/78 ppm伊凡氏藍在 PBS中的製備 伊凡氏藍(5.9 mg/mL)於PBS缓衝液中的原液藉由溶解 5_9 mg伊凡氏藍(Sigma Chemical公司)與1 mL PBS緩衝液 (來自實施例61)製備。 ·' 在此研究中使用一含有25%泊洛沙姆407溶液於得自實 施例8之PBS緩衝液的原液。一適當量的DSP加至實施例8 原液以製備含有2% DSP的配方(表4)。 表4含有25%泊洛沙姆407溶液於得自實施例9之PBS缓 衝液的原液。 試樣ID 25% P407 於PBS(g)中 DSP (mg) PBS缓衝劑 (g) 伊凡氏藍溶液 ㈣_ 17% P407/2DSP/EB 13.6 405.6 6 265 20% P407/2DSP/EB 16.019 407 3.62 265^ 25% P407/2DSP/EB 19.63 407 - 265~~ 前述配方依本文所述之步驟給予劑量至天竺鼠之中耳且 在施用劑量及在給予劑量24小時後,辨識當接觸時配方之 凝膠化能力及凝膠的位置。 實施例13:具可視染料及不具可視染料之泊洛沙姆407配方 的最终滅菌作用。 17%泊洛沙姆407/2% DSP/於磷酸鹽緩衝液,pH 7·3 : 溶解709 mg 氣化納(Fisher Scientific)、742 mg填酸二納 無水物USP(Fisher Scientific)、251.1 mg磷酸鈉單水合物 USP(Fisher Scientific)及一適當量之耳劑與158.1 g的滅菌 過濾DI水。此溶液在冰水浴冷卻並接著喷灑34.13 g泊洛沙 140447.doc -109- 200950787 姆407 NF(Spectrum chemicals)至冷卻的溶液並同時混合。 此混合物進一步混合直至泊洛沙姆完全溶解且可得一清澈 透明溶液。此溶液的pH為7.3。 17。/〇泊洛沙姆407/2% DSP/59ppm伊凡氏藍於雄酸緩衝 液中:取2 mL 17%泊洛沙姆407/2%耳劑/於鱗酸鹽緩衝液 溶液並加入2 mL之5.9 mg/mL伊凡氏藍(sigma-Aldrich chemical公司)於PBS緩衝液的溶液。 25%泊洛沙姆407/2%DSP/磷酸鹽緩衝液:溶劑330.5rng 氣化納(Fisher Scientific)、334.5 mg磷酸二納二水合物 USP(Fisher Scientific)、125.9 mg 填酸鈉單水合物 USP(Fisher Scientific)及一適當量的耳劑於70.5 g的滅菌過 慮DI水。 此溶液在冰水浴冷卻並接著噴灑2 5.1 g泊洛沙姆 407 NF(Spectrum chemicals)至冷卻的溶液並同時混合。此 混合物進一步混合直至泊洛沙姆完全溶解且可得一清澈透 明溶液。此溶液的pH為7.3。 25%泊洛沙姆407/2% DSP/59ppm伊凡氏藍於磷酸緩衝 液中:取2 mL 25%泊洛沙姆407/2% DSP/於磷酸鹽緩衝液 溶液並加入2 mL之5.9 mg/mL伊凡氏藍(Sigma-Aldrich chemical公司)於PBS緩衝液的溶液。 2 ml配方置2 mL玻璃小瓶中(Wheaton血清玻璃小瓶)以 13 mm Butylstr (kimble塞頭)密封並以13 mm紹密封捲 邊。此小瓶置於Market Forge-Sterilmatic高壓蒼(設定,緩 慢液態)並在250°F滅菌25分鐘。在高壓爸作用後,此試樣 140447.doc -110- 200950787 靜置冷卻至室温接著置於冰箱中。此小瓶置於冰箱中並當 冷卻時混合以均質化試樣。在高壓釜處理後記錄試樣脫色 或沉澱。 依實施例6所描述進行HPLC分析。 表5高壓釜處理在具可視染料及不具可視染料之含甲 基脫氫皮質固醇磷酸鈉之配方的效用。 試樣ID 0.68 之 %RRT % DSP % Dex (RRT-1.28) 1.41 之 %RRT 1.76 之 %RRT 17%P407 1.1 84.5 12.0 0.7 0.09 17% P407/伊凡氏藍 1.1 84.4 11.7 0.8 0.09 25% P407 0.8 80.9 16.0 0.7 0.10 25%P407/伊凡氏藍 0.9 80.9 15.3 0.7 0.12 黏性量測依實施例7所描述者進行。結果顯示經高壓蚤 處理之含可視染料的配方在產物降解作用及配方的黏性上 無效用。 測定25°/。泊洛沙姆407配方之平均溶解時間(依實施例7 所描述者測量,由UV @ 245 nm測定釋出的曱基脫氫皮質 φ 固醇磷酸酯之量)為5.6 hr而17%泊洛沙姆407配方顯示為 _ 3.2 hr ° 實施例14:在活體外釋放曲線的比較 在Snapwells(6.5 mm直徑之具有孔洞大小為0.4 μπι的聚 碳酸酯膜)中於37°C進行溶解,0.2 mL本文所述之凝膠配 方置於Snapwell中並靜置至硬化,接著0.5 mL缓衝液置於 貯器中並使用Labline軌道震盪器於在70 rpm震盪。每小時 取出試樣(取出0.1 mL並以温緩衝液取代)。以UV在245nm 分析試樣之耳劑濃度比較一外部檢量標準曲。Pluronic® 140447.doc • 111 - 200950787 濃度使用硫氰酸钻方法在624 nm分析。測定為%p4〇7之函 數的平均溶解時間(MDT)之相對等級序列。在此配方平均 溶解時間(MDT)與P407濃度間的線性關係顯示此甲基脫氫 皮質固醇釋放係歸因於聚合物凝膠(泊洛沙姆)之侵蝕但未 經擴散。一無-線性關係顯示耳劑釋放係經由擴散及/或聚 合物凝膠降解的組合。 - 或者,試樣使用在Li Xin-Yu文獻[Acta Pharmaceutica Sini技2008,43(2):208-203]中描述的方法分析且測定為% P407之函數的平均溶解時間(MDT)之等級序列。 ©' 第1圖說明具不同濃度的泊洛沙姆407之甲基脫氫皮質固 醇配方的活體外釋放曲線。第2圖說明介於配方之平均溶 解時間(MDT)與P407濃度間接近線性關係(1:1關連性)。此 結果指出甲基脫氫皮質固醇釋放係歸因於聚合物凝膠(泊 洛沙姆)之侵蝕但未經擴散。 實施例15:在活體外之凝膠化温度比較 以運用凝膠化温度的目的評估泊洛沙姆1 88與甲基脫氫 〇 皮質固醇在凝膠化温度與泊洛沙姆407配方的黏性之效 用。 - 使用一於PBS緩衝液之25%泊洛沙姆407原液(實施例9) , 與得自實施例6之PBS溶液。使用得自BASF的泊洛沙姆188 NF。 140447.doc •112- 200950787 表6含有泊洛沙姆407/泊洛沙姆188之試樣的製備 試樣 25%P407原液 (8) 泊洛沙姆188 (mg) PBS緩系系s (g) 16%P407/10%P188 3.207 501 1.303?^-- 17%P407/10%P188 3.4089 500 1.1056 ~~~ 18%P407/10%P188 3.6156 502 09072~~~~ 19% P407/10% PI 88 3.8183 500 0.7050-— 20% P407/10% PI 88 4.008 501 05032^ - 20% P407/5% PI 88 4.01 256 0770 20%泊洛沙姆407/10%洛沙姆188的平均溶解時間(在實 施例7中所描述的方法)測定為2.2hr而20%泊洛沙姆407/5% 泊洛沙姆18 8顯示為2 · 6 hr。使用在實施例7中所描述的方 法測定黏性。高壓釜處理在含有泊洛沙姆188的配方之黏 性或Tgel上無效用。 一適合所獲得之數據且可用於估計F127/F68混合物(用 於17_2〇%F127及0-10% F68)的凝膠化温度的等式如下。 T»»=-1.8 (%F127)+1.3 (% F68)+53 一適合所獲得之數據且可用於估計基於F127/F68混合物 (用於17-25% F127及0-10% F68)凝膠化温度之平均溶解 (hr)的等式,使用於實施例12及14獲得之結果。 MDT=-0.2 (T凝膠)+ 8 實施例16:測定用於滅菌過渡的温度範圍 測定在低温的黏性以助於導引滅菌過濾需要發生的温度 範圍以減少阻塞的可能性。 使用一附有一水夾套温度控制單元(溫度於1 〇-25°C以 1.6°C /min斜率增加)的 Brookfield黏度計 RVDV-II+P在 1、5 及10 rpm(剪力速率為7.5、37.5及75 s-1)旋轉之CPE-40心 140447.doc -113- 200950787 軸下進行黏性測定。 一 17% Pluronic P407的Tgel依耳劑增加的濃度函數測 定。1 7% Pluronic配方增加的Tgel依下等式估算: △ T 凝移=0.93[〇/〇 耳齊>1] 表7潛在配方在製造/過濾條件下的黏性。Qx 〇 where Q is the amount of release of the ear agent at time t' Qa is the total release amount of the ear agent, k is the release constant of the nth order, η is a factorless number with respect to the dissolution mechanism and b is the axis intercept , characterized by an initial burst mechanism, where n = 1 refers specifically to an erosion control mechanism. Mean dissolution time (MDT) is the sum of the drug molecules staying in the matrix during different time periods before release, divided by the total number of molecules and calculated by the following formula: MDr = heart it + 1 viscosity measurement using a water jacket The temperature control unit (temperature increased at 15-34 ° C with a slope of 1.6 ° C / min) Brookfield viscometer RVDV-II + P at 0.08 rpm (shear rate of 0.31 s · 1) rotating CPE-51 spindle get on. Tgel is defined as the curve inflection point due to the increase in viscosity due to peptide-gel conversion. Table 2 Effect of Autoclave Treatment on 17% Poloxamer 407NF/2% Methyl Dehydrocorticophosphate Sodium Phosphate (DSP) Release Curve 舆 Viscosity in PBS Buffer. MDT(hr) TgelfC) Max Viscosity* (Pas) Not treated by high pressure dad 3.2 25 403 High pressure dad treatment. 3.2 26 341 * at a shear rate of 0.31 s·1 in gel state (up to 37 ° C) Maximum Appearance Viscosity 140447.doc •104· 200950787 This result shows the release profile after 17% poloxamer 407 NF/2% methyldehydrocorticosterone phosphate (DSP) in autoclave treated with PB S buffer. A small utility with stickiness. Example 9 - Degradation products of a second polymer added to a formulation containing 2% methyldehydrocorticosteroid phosphate (DSP) and 17% poloxamer 407 NF after heat sterilization (autoclave treatment) Viscosity utility. Solution A. — pH 7.0 in a PBS buffer containing a carboxy-mercapto cellulose nano (CMC) solution by dissolving 178.35 mg of sodium chloride (Fisher Scientific), 300.5 mg of di-nano-anhydride (Fisher Scientific), 126.6 Mg acid anhydrate (Fisher Scientific) was prepared by sterilizing and filtering DI water at 78.4 g, then sprinkling 1 g of Blanose 7M65 CMC (Hercules, viscous 5450 cP @ 2°/.) into a buffer solution and heating to help dissolve And the solution is then cooled down. 17% poloxamer 407 NF/1% CMC/2% decyl dehydrocorticosterone sodium phosphate (DSP) at pH 7.0 to PBS by cooling 8.1 g of solution A in a chilled water bath followed by 205 mg Methyl dehydrocortisol sodium phosphate (DSP) is then prepared by mixing. 1.74 g of poloxamer 407 NF (Spectrum Chemicals) was sprinkled into a cold solution while mixing. The mixture is further mixed until the poloxamer is completely dissolved. 2 ml of the previous sample was placed in a 3 mL screw-cap glass vial (lined with rubber) and tightly sealed. This vial was placed in a Market Forge-Sterilmatic high pressure dad (set in slow liquid) and sterilized at 250 °F for 25 minutes. After the autoclave was applied, the sample was allowed to stand to cool to room temperature and then placed in a refrigerator. This sample was homogenized by mixing as the vial was cooled. 140447.doc -105- 200950787 No precipitation or discoloration was observed after autoclaving. HPLC analysis as described in Example 6 was carried out. Less than 1% degradation product was detected due to hydrolysis of the sulfhydryl dehydrocorticosteroid product, i.e., the formulation was stabilized after autoclaving. Viscosity measurements were made as described in Example 7. The results show that the autoclave treatment has a slight effect on the viscosity of the gel or the Tgel temperature. Less overall impurities were observed in the formulation containing the poloxamer compared to the control (2% DSP in PBS). The dissolution test was carried out as described in Example 7. The results show that the MDT of 11.9 hr is compared to the MDT of 3.2 without the CMC formulation. The addition of CMC or a primary polymer is a diffusion barrier that introduces a release rate that reduces the thiol-dehydrocorticosterol (i.e., increases MDT). Example 10 Buffer type The utility of the degradation product of the formulation containing poloxamer 407NF after heat sterilization (autoclave treatment). A TRIS buffer was adjusted to 7.4 with 1 M HCl by dissolving 377.8 mg of sodium hydride (Fisher Scientific) and 602.9 mg of tributanol (Tromethamine, Sigma Chemical) followed by sterilizing the transition of DI water to 100 g. Stock solution containing 25% solution of TRIS buffer poloxamer 407: Weigh 45 g of TRIS buffer, freeze in a freezer bath, then sprinkle 15 g of poloxamer 407 NF (Spectrum Chemicals) into buffer while mixing. This mixture is further mixed until the poloxamer is completely dissolved. Stock solution containing 25% poloxamer 407 solution in PBS buffer (pH 7.3): dissolve 704 mg sodium chloride (Fisher Scientific), 601.2 mg fill 140447.doc •106- 200950787 acid dihydrate anhydrate (Fisher Scientific ), 242.7 mg of sodium phosphate anhydrate (Fisher Scientific) was sterilized in 140.4 g of DI water. The bath was cooled in a freezer bath and then sprinkled with 50 g of poloxamer 407 NF (SPECTRUM CHEMICALS) into a cold solution while mixing. The mixture was further mixed until the poloxamer was completely dissolved, and a clear and transparent solution was obtained. The pH of the obtained solution was measured to be 7.3. / Prepare a series of formulations with the above stock solution. The DNA dehydrocorticosteroid vinegar (DSP) and the micronized dehydrocorticosteroid USP from Spectrum chemicals were used in the experiments used. The 1 ml sample was placed in a 3 mL screw cap glass vial (lined with rubber) and tightly sealed. This vial was placed in a Market Forge-Sterilmatic high pressure dad (set, slow liquid) and sterilized at 250 °F for 25 minutes. After the autoclave was applied, the sample was allowed to stand to cool to room temperature and then placed in a refrigerator. This sample was homogenized by mixing as the vial was cooled. HPLC analysis as described in Example 6 was carried out. Compare the stability in TRIS with PBS buffer. Table 3 Buffer type in the formulation containing methyl dehydrocorticosterol and methyl dehydrocorticosteroid phosphate in the degradation of the utility sample number appearance 1% DSP % Dex (RRT = 1.27) 1.54% RRT 1.68 %RRT 1%DSP/TRIS Light Yellow & Precipitation 41 54 2.68 0.97 2% DSP/TRIS Light Yellow & Shen Dang 41 55 2.4 0.57 4% DSP/TRIS Light Yellow & Precipitation 58 36 2.4 0.23 16%P407/2DSP /TRIS Precipitation 54 41 0.79 0.62 18% P407/2DSP/TRIS Precipitation 52 45 1.21 0.51 140447.doc -107- 200950787 20% P407/2DSP/TRIS Precipitation 55 43 0.86 0.49 18% P407/2DEX/TRIS Suspension - 99 0.22 0.55 2% DEX/TRIS suspension - 99 - 0.28 2% DSP/PBS Light yellow & precipitate 85.6 9.8 2.09 - 16% P407/2DSP/PBS Clear solution 78 17.5 1.86 - 18% P407/2DSP/PBS Clear solution 81.2 16.2 1.14 - 20% P407/2DSP/PBS Clear Solution 81.5 16.1 1.04 - 18% P407/2DEX/PBS Suspension - 97 1.06 0.45 2% DEX/PBS Suspension - 94.7 1.34 - Viscosity Measurement According to Example 7 get on. The results show that in order to reduce the hydrolysis during the autoclave treatment, the buffer needs to maintain a pH between 7 and 8 when warming up. An increased drug hydrolyzability was observed in TRIS buffer than in PBS (Table 3). The occurrence of other degradation products can be reduced by using the polymeric additives (e.g., P407) described herein. A reduction in degradation products was observed by comparison of the formulation containing 20% poloxamer 407 with the formulation without poloxamer 407 (Table 7). Formulations containing suspended micronized sulfhydryl dehydrocorticosteroids have greater stability in autoclave treatment than their solution counterparts. Example 11: Pulsed release otic formulations using sulfhydryl dehydrocorticosterol and sulfhydryl groups A combination of dehydrocorticosterol sodium phosphate (DSP) (1:1 ratio) was prepared as described herein for a pulse release agent formulation. 20% of the deliverable dose of methyldehydrocorticosterol was dissolved in the 17% poloxamer solution of Example 7 with the aid of beta cyclodextrin. The remaining 80% of the deliverable methyl dehydrocorticosterol dose is then added to the mixture and the final formulation is prepared according to any of the processes described herein. A pulse release formulation containing sulfhydryl dehydrocorticosteroid was prepared according to the procedure of the examples described herein and tested using the procedure described herein to determine the pulse 140447.doc -108 - 200950787 release profile. Example 12: Preparation of 17% poloxamer 407/2% DSP agent / 78 ppm Evans blue in PBS Stock solution of Ivans blue (5.9 mg/mL) in PBS buffer by dissolving 5_9 mg Ivans Blue (Sigma Chemical) was prepared with 1 mL PBS buffer (from Example 61). - A stock solution containing 25% poloxamer 407 solution in PBS buffer from Example 8 was used in this study. An appropriate amount of DSP was added to the stock solution of Example 8 to prepare a formulation containing 2% DSP (Table 4). Table 4 contains a stock solution of 25% poloxamer 407 solution in PBS buffer from Example 9. Sample ID 25% P407 in PBS (g) DSP (mg) PBS buffer (g) Evans blue solution (4) _ 17% P407/2DSP/EB 13.6 405.6 6 265 20% P407/2DSP/EB 16.019 407 3.62 265^ 25% P407/2DSP/EB 19.63 407 - 265~~ The above formulation was dosed to the middle ear of guinea pigs according to the procedure described herein and the gel of the formula was contacted at the dose administered and 24 hours after the dose was administered. Chemical ability and location of the gel. Example 13: Final sterilization of a poloxamer 407 formulation with a visible dye and no visible dye. 17% poloxamer 407/2% DSP/in phosphate buffer, pH 7.3: Dissolve 709 mg gasification sodium (Fisher Scientific), 742 mg acid dihydrate anhydrate USP (Fisher Scientific), 251.1 mg Sodium phosphate monohydrate USP (Fisher Scientific) and an appropriate amount of ear and 158.1 g of sterile filtered DI water. This solution was cooled in an ice water bath and then sprayed with 34.13 g of poloxamer 140447.doc-109-200950787 NF NF (Spectrum chemicals) to the cooled solution and mixed at the same time. This mixture is further mixed until the poloxamer is completely dissolved and a clear transparent solution is obtained. The pH of this solution was 7.3. 17. / 〇 poloxamer 407/2% DSP / 59ppm Evans blue in maleic acid buffer: take 2 mL 17% poloxamer 407 / 2% ear / in sulphate buffer solution and add 2 A solution of 5.9 mg/mL of Ivan's Blue (sigma-Aldrich chemical) in PBS buffer. 25% poloxamer 407/2% DSP/phosphate buffer: solvent 330.5 rng gasification nano (Fisher Scientific), 334.5 mg diammonium dihydrate USP (Fisher Scientific), 125.9 mg sodium sulphate monohydrate USP (Fisher Scientific) and an appropriate amount of ear are sterilized in 70.5 g of DI water. This solution was cooled in an ice water bath and then sprayed with 2 5.1 g of poloxamer 407 NF (Spectrum chemicals) to the cooled solution while mixing. This mixture is further mixed until the poloxamer is completely dissolved and a clear and transparent solution is obtained. The pH of this solution was 7.3. 25% poloxamer 407/2% DSP/59ppm Evans blue in phosphate buffer: Take 2 mL 25% poloxamer 407/2% DSP/phosphate buffer solution and add 2 mL of 5.9 A solution of mg/mL Ivan's Blue (Sigma-Aldrich Chemical) in PBS buffer. A 2 ml formulation was placed in a 2 mL glass vial (Wheaton serum glass vial) sealed with a 13 mm Butylstr (kimble plug) and sealed at 13 mm. This vial was placed in a Market Forge-Sterilmatic high pressure setting (slow, liquid state) and sterilized at 250 °F for 25 minutes. After the action of the high pressure dad, the sample 140447.doc -110- 200950787 was allowed to cool to room temperature and then placed in a refrigerator. This vial was placed in a refrigerator and mixed while cooling to homogenize the sample. The sample was decolorized or precipitated after autoclaving. HPLC analysis was performed as described in Example 6. Table 5 Effect of autoclave treatment on formulations containing visible dyes and sodium-containing dehydrocorticosterone phosphate without visible dyes. Sample ID 0.68%RRT % DSP % Dex (RRT-1.28) 1.41%RRT 1.76%RRT 17%P407 1.1 84.5 12.0 0.7 0.09 17% P407/Ivan Blue 1.1 84.4 11.7 0.8 0.09 25% P407 0.8 80.9 16.0 0.7 0.10 25% P407/Ivan Blue 0.9 80.9 15.3 0.7 0.12 Viscosity measurement was carried out as described in Example 7. The results show that the dye-containing formulation treated with high pressure ruthenium is ineffective for product degradation and viscosity of the formulation. Determine 25 ° /. The average dissolution time of the poloxamer 407 formulation (measured as described in Example 7, the amount of sulfhydryl dehydrocorticosteroid citrate released by UV @ 245 nm) was 5.6 hr and 17% Polo The sam 407 formulation is shown as _ 3.2 hr ° Example 14: Comparison of in vitro release profiles in Snapwells (6.5 mm diameter polycarbonate membrane with a pore size of 0.4 μm) at 37 ° C, 0.2 mL The gel formulation described herein was placed in Snapwell and allowed to stand until hardened, then 0.5 mL of buffer was placed in the reservoir and shaken at 70 rpm using a Labline orbital shaker. The sample was taken every hour (0.1 mL was taken and replaced with warm buffer). An external calibrated standard curve was compared with the concentration of the ear of the sample analyzed by UV at 245 nm. Pluronic® 140447.doc • 111 - 200950787 Concentration was analyzed at 624 nm using a thiocyanate drilling method. The relative rank sequence of the mean dissolution time (MDT) of the function of %p4〇7 was determined. A linear relationship between the mean dissolution time (MDT) and the P407 concentration of this formulation showed that this methyldehydrocorticosterol release was attributed to the erosion of the polymer gel (poloxamer) but did not spread. A none-linear relationship shows a combination of otic release via diffusion and/or polymer gel degradation. - Alternatively, the sample was analyzed using the method described in Li Xin-Yu [Acta Pharmaceutica Sini Technology 2008, 43(2): 208-203] and determined as a function of % P407 as the average dissolution time (MDT) grade sequence. . © 'Figure 1 illustrates the in vitro release profile of a methyl dehydrocorticosteroid formulation with different concentrations of poloxamer 407. Figure 2 illustrates the near linear relationship (1:1 correlation) between the mean dissolution time (MDT) and the P407 concentration of the formulation. This result indicates that the methyldehydrocorticosterol release is attributed to the erosion of the polymer gel (poloxamer) but not to diffusion. Example 15: Comparison of gelation temperature in vitro. Evaluation of poloxamer 1 88 and methyldehydroquinone cortisol at gelation temperature with poloxamer 407 formulation for the purpose of gelation temperature Viscosity effect. - A 25% poloxamer 407 stock solution in PBS buffer (Example 9) was used with the PBS solution from Example 6. Poloxamer 188 NF from BASF was used. 140447.doc •112- 200950787 Table 6 Preparation of samples containing poloxamer 407/poloxamer 188 25% P407 stock solution (8) Poloxamer 188 (mg) PBS system s (g 16%P407/10%P188 3.207 501 1.303?^-- 17%P407/10%P188 3.4089 500 1.1056 ~~~ 18%P407/10%P188 3.6156 502 09072~~~~ 19% P407/10% PI 88 3.8183 500 0.7050-— 20% P407/10% PI 88 4.008 501 05032^ - 20% P407/5% PI 88 4.01 256 0770 20% poloxamer 407/10% rossam 188 average dissolution time (in implementation The method described in Example 7 was determined to be 2.2 hr and 20% poloxamer 407/5% poloxamer 18 8 was shown to be 2 · 6 hr. The viscosity was measured using the method described in Example 7. Autoclave treatment is not effective on the viscosity or Tgel of formulations containing poloxamer 188. An equation suitable for the data obtained and which can be used to estimate the gelation temperature of the F127/F68 mixture (for 171-2% F127 and 0-10% F68) is as follows. T»»=-1.8 (%F127)+1.3 (% F68)+53 One suitable for the data obtained and can be used to estimate a gel based on F127/F68 mixture (for 17-25% F127 and 0-10% F68) The equation for the average dissolution (hr) of the temperature was used for the results obtained in Examples 12 and 14. MDT = -0.2 (T-gel) + 8 Example 16: Determination of the temperature range for sterilization transitions The viscosity at low temperatures is measured to help guide the temperature range that is required for sterilization filtration to reduce the likelihood of blockage. Use a Brookfield viscometer RVDV-II+P with a water jacketed temperature control unit (temperature increase at 1 〇 -25 ° C with a slope of 1.6 ° C / min) at 1, 5 and 10 rpm (shear rate 7.5) , 37.5 and 75 s-1) Rotating CPE-40 heart 140447.doc -113- 200950787 Viscosity measurement under the shaft. A 17% Pluronic P407 Tgel oxime agent was tested for increasing concentration function. The Tgel increased by 1 7% Pluronic formula is estimated by the following equation: △ T condensate = 0.93 [〇 / 〇 ear &> 1] Table 7 Viscosity of potential formulations under manufacturing/filtration conditions.

試樣 外觀黏* \ia (cP) 温度@ lOOcP 低於 Tgel5°C 20°C 安慰劑 52cP@ 17°C 120 cP 19°C 17% P407/2% DSP 90cP@ 18°C 147 cP 18.5〇C 17% P407/6% DSP 142 cP@ 22〇C 105 cP 19.7 V 在3 7.5 s·1剪力速率下測量的黏性 此結果顯示在本文所描述之配方的滅菌過濾可在約19°C 下進行。 實施例17 :製造條件的測定 製備8升批料之17% P407安慰劑以評估製造/過濾條件。 安慰劑之製備係藉由將6_4升DI水置於3加侖SS壓力容器 中,並在冰箱中靜置冷卻過夜。在次晨取出容器並加入 (水温度5°C,RT 18°C)及48 g氯化鈉、29.6 g磷酸二鈉無水 物及10 g磷酸鈉單水合物並以頂部混合器(IKA RW20 @ 1720 rpm)溶解。半小時後,一旦緩衝液溶解(溶液温度 8°C,RT 18°C),1.36kg泊洛沙姆407 NF(Spectrum Chemicals) 在15分鐘間隔缓慢灑入緩衝液溶液(溶液温度12°C,RT 1 8°C ),接著速度增加至2430 rpm。在又1小時混合後,混 合速度降低至1062 rpm (完全溶解)。。 室温維持低於25°C以保持溶液的温度低於19°C。溶液温 140447.doc -114- 200950787 度維持低於19°C達最初製造的3小時,而不需要冰凍/冷卻 容器。 在20 psi及14°C溶液評估具有17.3 cm2表面積之三不同 Sartoscale (Sartorius Stedim)慮網Sample Appearance Viscosity * \ia (cP) Temperature @ lOOcP Below Tgel5°C 20°C Placebo 52cP@ 17°C 120 cP 19°C 17% P407/2% DSP 90cP@ 18°C 147 cP 18.5〇C 17% P407/6% DSP 142 cP@ 22〇C 105 cP 19.7 V Viscosity measured at a shear rate of 3 7.5 s·1 This result shows that the sterilization filtration of the formulation described in this article can be performed at approximately 19 ° C get on. Example 17: Determination of manufacturing conditions A 7 liter batch of 17% P407 placebo was prepared to evaluate manufacturing/filtration conditions. Placebo was prepared by placing 6_4 liters of DI water in a 3 gallon SS pressure vessel and allowed to stand in the freezer for overnight cooling. Remove the container in the next morning and add (water temperature 5 ° C, RT 18 ° C) and 48 g sodium chloride, 29.6 g disodium phosphate anhydrous and 10 g sodium phosphate monohydrate and use a top mixer (IKA RW20 @ 1720 rpm) dissolved. After half an hour, once the buffer was dissolved (solution temperature 8 ° C, RT 18 ° C), 1.36 kg of poloxamer 407 NF (Spectrum Chemicals) was slowly sprinkled into the buffer solution at a 15 minute interval (solution temperature 12 ° C, RT 1 8 ° C), then increase the speed to 2430 rpm. After mixing for another hour, the mixing speed was reduced to 1062 rpm (completely dissolved). . The room temperature was maintained below 25 ° C to keep the temperature of the solution below 19 °C. The solution temperature 140447.doc -114- 200950787 degrees was maintained below 19 °C for 3 hours of initial manufacture without the need for a freeze/cool container. Estimating three different Sartoscale (Sartorius Stedim) sites with a surface area of 17.3 cm2 at 20 psi and 14 °C

Sartopore 2,0.2 μπι 5445307HS-FF (PES),流速 16 mL/ 分鐘Sartopore 2, 0.2 μm 5445307HS-FF (PES), flow rate 16 mL/min

Sartobran P,0·2 μπι 5235307HS-FF (纖維素 ester),流 速12 mL/分鐘Sartobran P, 0·2 μπι 5235307HS-FF (cellulose ester), flow rate 12 mL/min

Sartopore 2 XLI,0.2 μπι 5445307IS-FF (PES),流速 15 mL/分鐘 使用Sartopore 2濾網5441307H4-SS,在溶液温度使用具 有表面積為 0.015 m2之 0.45、0·2 μιη Sartopore 2 150滅菌 囊(Sartorius Stedim)於16 psi壓力下進行過遽。在16 psi於 大約100 mL/min下測定流速,當温度維持於6.5-14°C範圍 時,速率未改變。溶液的降低壓力及增加温度造成在流速 的減低,此係因為溶液的黏性增加。在製程中監控溶液的 脫色。 表8 17%泊洛沙姆407安慰劑在溶液温度於6.5-14°C範 圍間使用Sartopore 2,0.2 μιη濾網於16 psi壓力下的預測 過滤時間。 濾網 大小(m2) 估計流速 (mL/分鐘) 過滤8 L的時間 (估計) Sartopore 2,大小4 0.015 lOOmL/分鐘 80分鐘 Sartopore 2,大小7 0.05 330 mL/分鐘 24分鐘 Sartopore 2,大小8 0.1 670 mL/分鐘 12分鐘 140447.doc -115- 200950787 在過濾評估前檢測黏性、Tgel及UV/Vis吸收度。 Pluronic UV/Vis 光譜藉由 Evolution 160 UV/Vis (Thermo Scientific)獲得。在250-300 nm範圍之峰係來自存在於原 物料(泊洛沙姆)的BHT安定劑。 前述製程可用於製造17% P407配方,及包括在周遭條件 的温度分析。温度19°C在製造期間減少冷卻容器的成本。 在某些例子中,使用一夾套容器以進一步控制的溶液温度 以減輕製造的關切度。 實施例18甲基脫氫皮質固醇由高壓釜處理之微米化試樣 的在活體外釋放 17%泊洛沙姆 407/1.5%耳劑於TRIS緩衝液中:250.8 mg 氯化納(Fisher ScientiHc)、及 302.4 mg 三丁 醇酉旨 (Sigma Chemical公司)溶解於39.3 g滅菌過渡DI水,pH以 1 M HC1調整至7.4。使用4.9 g前述溶液並懸浮一適當量的 微米化曱基脫氫皮質固醇USP (Spectrum Scientific)並分 散良好。2 mL之此配方傳送至一 2 mL玻璃小瓶中(Wheaton 血清玻璃小瓶)以13 mm 丁基苯乙烯S旨(kimble塞頭)密封並 以13 mm|S密封捲邊。此小瓶置於Market Forge-Sterilmatic 高壓釜(設定,緩慢液態)並在250°F滅菌25分鐘。在高壓 爸作用後,此試樣靜置冷卻至室温。此小瓶置於冰箱中並 當冷卻時混合以均質化試樣。在高壓釜作用後記錄試樣沉 澱或脫色。 在Snapwells(6.5 mm直徑之具有孔洞大小為0.4 μιη的聚 碳酸酯膜)中於37°C進行溶解,0.2 mL凝膠配方置於 140447.doc -116- 200950787Sartopore 2 XLI, 0.2 μm 5445307IS-FF (PES), flow rate 15 mL/min using Sartopore 2 filter 5441307H4-SS, using a 0.45, 0·2 μη Sartopore 2 150 sterilized capsule with a surface area of 0.015 m2 at the solution temperature (Sartorius Stedim) was simmered at a pressure of 16 psi. The flow rate was measured at 16 psi at approximately 100 mL/min and the rate was unchanged when the temperature was maintained at 6.5-14 °C. The reduced pressure of the solution and the increased temperature cause a decrease in flow rate due to the increased viscosity of the solution. The discoloration of the solution was monitored during the process. Table 8 Predicted filtration time of a 17% poloxamer 407 placebo using a Sartopore 2, 0.2 μιη filter at a pressure of 16 psi at a solution temperature of 6.5-14 °C. Filter size (m2) Estimated flow rate (mL/min) Filter 8 L time (estimated) Sartopore 2, size 4 0.015 lOOmL/min 80 min Sartopore 2, size 7 0.05 330 mL/min 24 min Sartopore 2, size 8 0.1 670 mL/min 12 min 140447.doc -115- 200950787 Viscosity, Tgel and UV/Vis absorbance were measured prior to filtration evaluation. Pluronic UV/Vis spectra were obtained by Evolution 160 UV/Vis (Thermo Scientific). The peak in the 250-300 nm range is derived from the BHT stabilizer present in the raw material (poloxamer). The foregoing process can be used to make 17% P407 formulations and includes temperature analysis in ambient conditions. The temperature of 19 ° C reduces the cost of the cooling vessel during manufacturing. In some instances, a jacketed container is used to further control the temperature of the solution to alleviate manufacturing concerns. Example 18 Methyl dehydrocorticosterol In vitro release of 17% poloxamer 407/1.5% otic agent in TRIS buffer by autoclave-treated micronized sample: 250.8 mg sodium chloride (Fisher ScientiHc And 302.4 mg of tributanol (Sigma Chemical) were dissolved in 39.3 g of sterilized transition DI water and the pH was adjusted to 7.4 with 1 M HCl. 4.9 g of the foregoing solution was used and an appropriate amount of micronized deuterated dehydrocorticosterol USP (Spectrum Scientific) was suspended and well dispersed. 2 mL of this formulation was transferred to a 2 mL glass vial (Wheaton serum glass vial) sealed with a 13 mm butyl styrene S (kimble plug) and sealed with a 13 mm|S seal. This vial was placed in a Market Forge-Sterilmatic autoclave (set, slow liquid) and sterilized at 250 °F for 25 minutes. After the high pressure dad, the sample was allowed to cool to room temperature. This vial was placed in a refrigerator and mixed while cooling to homogenize the sample. The sample was precipitated or decolored after the autoclave was applied. Dissolution in Snapwells (6.5 mm diameter polycarbonate film with a pore size of 0.4 μηη) at 37 ° C, 0.2 mL gel formulation placed at 140447.doc -116- 200950787

Snapwell中並靜置至硬化,接著〇·5 mL PBS緩衝液置於貯 器中並使用Labline執道震盈器於在70 rpm震盈。每小時取 出試樣[取出0.1 mL並以含有2% PEG-40氫化蓖麻油(B ASF) 温PBS緩衝液取代促進甲基脫氫皮質固醇溶解性]。以245 nm UV參考一外部校正標準曲線分析試樣的曱基脫氫皮質 固醇濃度。釋放速率與本文揭露之其他配方比較。計算每 一試樣的MDT時間。 曱基脫氫皮質固醇在17%泊洛沙姆系統中的溶解度藉由 在Eppendorf離心機5424於15,000 rpm離心試樣10分鐘的上 清液中的甲基脫氫皮質固醇濃度而評估。上清液中的甲基 脫氫皮質固醇濃度以245 nm UV參考一外部校正標準曲線 測定。第3圖說明含有17% P407的不同類固醇的釋放曲線 皮質酮。表9描述曱基脫氫皮質固醇在TRIS緩衝劑與17% P407溶液的溶解度。 表9甲基脫氫皮質固醇在TRIS緩衝劑與17%P407溶液 的溶解度 試樣 上清液中的甲基脫氫皮質固醇濃度 _ WmL)___ 17% P407/1.5% DEX/TRIS 580 於TRIS缓衝劑之2% DEX(實施例4) 86 經高壓釜處理的於TRIS緩衝劑之2% DEX (實施例4) 153 實施例19含有羧基甲基纖維素鈉之配方的釋放速率或 MDT與黏性。 17%泊洛沙姆407/2% DSP/1% CMC(Hercules Blanose7M): 於PBS緩衝液中之羧基甲基纖維素鈉(CMC)溶液(pH 7.0)藉 140447.doc -117- 200950787The Snapwell was allowed to stand until it hardened, then 5 mL of PBS buffer was placed in the reservoir and the Labline was used to vibrate at 70 rpm. The sample was taken every hour [0.1 mL was taken out and replaced with 2% PEG-40 hydrogenated castor oil (B ASF) warm PBS buffer to promote methyl dehydrocorticosteroid solubility]. The thiol-dehydrocorticosteroid concentration of the sample was analyzed using a 245 nm UV reference-external calibration standard curve. The release rate is compared to other formulations disclosed herein. Calculate the MDT time for each sample. The solubility of sulfhydryl dehydrocorticosteroid in the 17% poloxamer system was assessed by the concentration of methyl dehydrocorticosterol in the supernatant of the sample centrifuged at 15,000 rpm for 10 minutes in an Eppendorf centrifuge 5424. The methyl dehydrocorticosterol concentration in the supernatant was determined using a 245 nm UV reference-external calibration standard curve. Figure 3 illustrates the release profile of different steroids containing 17% P407. Table 9 describes the solubility of sulfhydryl dehydrocorticosteroid in TRIS buffer and 17% P407 solution. Table 9 Methyl dehydrocorticosteroid concentration in the solubility sample of TRES buffer and 17% P407 solution _ WmL) ___ 17% P407/1.5% DEX/TRIS 580 2% DES of TRIS Buffer (Example 4) 86 2% DEX of TRIS Buffer Treated by Autoclave (Example 4) 153 Example 19 Release Rate or MDT of Formulation Containing Sodium Carboxymethyl Cellulose With stickiness. 17% poloxamer 407/2% DSP/1% CMC (Hercules Blanose 7M): carboxymethylcellulose sodium (CMC) solution (pH 7.0) in PBS buffer. 140447.doc -117- 200950787

由溶解 205·6 mg 氯化納(Fisher Scientific)、372_1 mg 填酸 二納二水合物(Fisher Scientific)、106·2 mg構酸納單水合 物(Fisher Scientific)於78.1 g滅菌過濾DI水而製備。接著 灑入 1 g Blanose 7M CMC(Hercules,黏性533cP @ 2%)至 緩衝液溶液並加熱以助於溶解,且此溶液接著冷卻’並灑 入17.08 g泊洛沙姆407NF(Spectrum Chemicals)至冷溶液中 同時混合。製成一含有17%泊洛沙姆407 NF/1% CMC/2% DSP於PBS緩衝液的配方,加入/溶解一適當量的曱基脫氫 皮質固醇至9.8 g之上述溶液中,並混合直至耳劑完全溶 解。此溶液的pH為7·0。The DI water was sterilized by dissolving 205. 6 mg of sodium chloride (Fisher Scientific), 372_1 mg of di-nanohydrate dihydrate (Fisher Scientific), and 106.2 mg of sodium sulphate monohydrate (Fisher Scientific) at 78.1 g. preparation. Then sprinkle 1 g of Blanose 7M CMC (Hercules, viscous 533 cP @ 2%) to the buffer solution and heat to aid dissolution, and the solution was then cooled 'and sprinkled into 17.08 g of Poloxamer 407NF (Spectrum Chemicals) to Mix in a cold solution at the same time. Prepare a formulation containing 17% poloxamer 407 NF/1% CMC/2% DSP in PBS buffer, add/dissolve an appropriate amount of thiol dehydrocorticosterol to 9.8 g of the above solution, and Mix until the ear is completely dissolved. The pH of this solution was 7.0.

17%泊洛沙姆407/2% DSP/0.5% CMC(Blanose 7M65): 於PBS緩衝液之羧基曱基纖維素鈉(CMC)溶液(pH 7.2)藉由 溶解 257 mg 氯化納(Fisher Scientific)、375 mg 構酸二納二 水合物(Fisher Scientific)、108 mg 填酸鈉單水合物(Fisher Scientific)於78.7 g滅菌過濾DI水而製備。灑入0.502 g Blanose 7M65 CMC (Hercules,黏性 of 5450cP @ 2%)至緩 衝液溶液並加熱以助於溶解,且此溶液接著冷卻,並灑入 17.06 g泊洛沙姆 407 NF (Spectrum Chemicals)至冷溶液中 同時混合。製成一含有17%泊洛沙姆407 NF/1% CMC/2% DSP於PBS緩衝液的溶液,加入/溶解201 mg DSP至9.8 g 之上述溶液中,並混合直至DSP完全溶解。此溶液的PH為 7.2。 170/〇泊洛沙姆 407/2% DSP/0.5% CMC (Blanose 7H9):於 PBS緩衝液中之羧基曱基纖維素鈉(CMC)溶液(pH 7.3)藉 140447.doc -118- 200950787 由溶解256.5 mg氯化納(Fisher Scientific)、374 mg填酸二 納二水合物(Fisher Scientific)、107 mg碗酸納單水合物 (Fisher Scientific)於78.6 g滅菌過濾DI水而製備,接著灑 入 0·502 g Blanose 7H9 CMC(Hercules,黏性 of 5600cP @ 1%)至緩衝液溶液並加熱以助於溶解,且此溶液接著冷 卻,並瀵入 17.03 g 泊洛沙姆 407 NF(Spectrum Chemicals) 至冷溶液中同時混合。製成一含有Π%泊洛沙姆407 NF/l°/〇 CMC/2% DSP於PBS緩衝液的溶液,加入/溶解203 mg DSP 至9.8 g之上述溶液中,並混合直至DSP完全溶解。此溶液 的pH為7.3。 依實施例7所描述者進行黏性測定。依實施例7所描述者 進行溶解。 第4圖說明介於配方之平均溶解時間(MDT)與配方之外 觀黏性間的關聯性。釋放速率為藉由一二級聚合物的併入 而調變。一二級聚合物之等級及濃度的選用可藉由使用顯 示於第5圖及第6圖之一般可得水可溶聚合之一者的圖而促 進0 實施例20-微-甲基脫氫皮質固醇粉末的乾滅菌 10 mg微米化之甲基脫氫皮質固醇粉末(Spectrum lot XD0385)充填入2 ml玻璃小瓶中並以13 mm丁基橡膠栓 (Kimble)密封並置於不同温度之烘箱中7-11小時。 HPLC 分析使用附有 Luna C18(2) 3 μιη、100A、250x4.6 mm 管柱之Agilent 1200,其使用30-95溶液B(溶劑A 35%甲 醇:35°/〇水:30%乙酸酯緩衝劑,溶劑B 70%甲醇:30%乙 140447.doc •119- 200950787 酸酯緩衝劑pH 4)梯度(1-6 min),接著等強度(95%溶劑 B)ll分鐘,總試驗22分鐘。試樣溶解於乙醇中並分析。微 米化之甲基脫氫皮質固醇於高至13 8°C温度的乾熱滅菌不 影響微米化之曱基脫氫皮質固醇的粒子大小分佈。HPLC 分析顯示經乾熱滅菌的微米化之甲基脫氫皮質固醇之99% 純度。 實施例21 -增進黏性之皮質類固醇配方在内耳圓窗膜的施用 依實施例1製備配方並裝入一附有15號針之氣密式可抛 棄式的5 ml矽化玻璃注射器。利多卡因局部施用至鼓膜、 及製作一用於觀看中耳腔室的小切口。引導針尖至置於内 耳圓窗膜上的位置’且直接施用抗炎症之皮質類固醇配方 至圓窗膜上。 實施例22 -在一天竺鼠的皮質類固醇配方之耳内注射之活 艘内測試。 同屬性群的21天大的竺鼠(Charles River,重2〇〇_3〇〇§之 雌性)以2CM20叫之2% DSP配方耳内注射。第7圖顯示在 天竺鼠耳進行耳内注射達到5天後的凝膠結果。增加之注 射體積增加達90 μι之注射體積的凝膠滯留。然而,12〇叫 的注射體積顯示一較低的凝膠滞留。 同屬性群的2丨天大的竺鼠(Charles River,重2〇〇3〇〇§之 雖’!·生)以50 pL之本文所沭的冬古η 5 乂 η。17% poloxamer 407/2% DSP/0.5% CMC (Blanose 7M65): Solvent 257 mg sodium chloride in carboxy buffered sodium methacrylate (CMC) solution (pH 7.2) in PBS buffer (Fisher Scientific ), 375 mg of di-nanohydrate dihydrate (Fisher Scientific), 108 mg of sodium carbonate monohydrate (Fisher Scientific) were prepared by sterilizing and filtering DI water at 78.7 g. Sprinkle 0.502 g of Blanose 7M65 CMC (Hercules, viscous of 5450 cP @ 2%) to the buffer solution and heat to aid dissolution, and then cool the solution and sprinkle with 17.06 g of Poloxamer 407 NF (Spectrum Chemicals) Mix simultaneously into the cold solution. A solution containing 17% poloxamer 407 NF/1% CMC/2% DSP in PBS buffer was prepared, and 201 mg DSP was added/dissolved to 9.8 g of the above solution and mixed until the DSP was completely dissolved. The pH of this solution was 7.2. 170/〇泊洛沙姆407/2% DSP/0.5% CMC (Blanose 7H9): a solution of sodium carboxymethyl cellulose (CMC) in PBS buffer (pH 7.3) by 140447.doc -118- 200950787 Prepared by dissolving 256.5 mg of sodium chloride (Fisher Scientific), 374 mg of di-nanohydrate dihydrate (Fisher Scientific), and 107 mg of sodium citrate monohydrate (Fisher Scientific) in 78.6 g of sterile filtered DI water, followed by sprinkling 0·502 g Blanose 7H9 CMC (Hercules, viscous of 5600 cP @ 1%) to a buffer solution and heated to aid dissolution, and this solution was then cooled and poured into 17.03 g of Poloxamer 407 NF (Spectrum Chemicals) Mix simultaneously into the cold solution. A solution containing Π% poloxamer 407 NF/l°/〇 CMC/2% DSP in PBS buffer was prepared, added/dissolved 203 mg DSP to 9.8 g of the above solution, and mixed until the DSP was completely dissolved. The pH of this solution was 7.3. Viscosity measurements were performed as described in Example 7. Dissolution was carried out as described in Example 7. Figure 4 illustrates the correlation between the mean dissolution time (MDT) of the formulation and the apparent viscosity of the formulation. The rate of release is modulated by the incorporation of a secondary polymer. The grade and concentration of the primary and secondary polymers can be selected by promoting the use of one of the generally available water soluble polymerizations shown in Figures 5 and 6, Example 20 - Micro-methyl dehydrogenation Dry Sterilization of Corticosteroid Powder 10 mg of micronized methyl dehydrocorticosterol powder (Spectrum lot XD0385) was filled into a 2 ml glass vial and sealed with a 13 mm butyl rubber stopper (Kimble) and placed in an oven at different temperatures In 7-11 hours. HPLC analysis using Agilent 1200 with Luna C18(2) 3 μιη, 100A, 250 x 4.6 mm column using 30-95 solution B (solvent A 35% methanol: 35 ° / hydrazine: 30% acetate) Buffer, solvent B 70% methanol: 30% B 140447.doc • 119- 200950787 Acid buffer pH 4) Gradient (1-6 min) followed by isocratic (95% solvent B) ll minutes, total test 22 minutes . The sample was dissolved in ethanol and analyzed. Dry heat sterilization of micro-degraded methyl dehydrocorticosterol at temperatures up to 13 8 °C does not affect the particle size distribution of micronized sulfhydryl dehydrocorticosteroids. HPLC analysis showed 99% purity of the micronized methyl dehydrocorticosteroid sterilized by dry heat. Example 21 - Adhesive corticosteroid formulation for inner ear round window film Formulation was prepared as in Example 1 and loaded into a gas-filled disposable 5 ml deuterated glass syringe with a 15 gauge needle. Lidocaine is topically applied to the tympanic membrane and a small incision for viewing the middle ear chamber is made. The needle tip is guided to the position on the inner round window membrane and the anti-inflammatory corticosteroid formulation is applied directly to the round window membrane. Example 22 - In-the-ear test for intra-oral injection of a corticosteroid formulation of the day of the mole. A 21-day-old mole (Charles River, female weighing 2〇〇_3〇〇§) of the same attribute group was injected intratracheally with 2% of the 20% DSP formula. Figure 7 shows the results of gelation after 5 days of intrathecal injection of guinea pig ears. Increased injection volume increased by up to 90 μιη injection volume of gel retention. However, the 12-injected injection volume showed a lower gel retention. The 2nd-day-old squirrel of the same attribute group (Charles River, weighing 2〇〇3〇〇§ although ‘!·生) is 50 L 之 冬 冬 η 乂 乂 乂 。 η.

DSP 配;DSP distribution;

進程。 度的DS 140447.doc 200950787 解時間(MDT))。再者,當用於6% DSP配方(6% Dex-P(*)) 的P407濃度由17%增加至19%,可觀察到一較快的凝膠消 除,如第8圖所示。因此,測試在本文所描述之配方中的 皮質類固醇的注射體積及濃度以決定臨床前實驗及臨床實 驗的最適參數。已觀察到具有高濃度DSP之耳内配方具有 不同於具較低濃度之DSP的耳内配方之釋放曲線。 實施例23-在活體内延長的釋放動能 同屬性群的21天大的竺鼠(Charles River,重200-300g之 雌性)以50 pL 17%在280 mOsm/kg緩衝且具有配方之1.5 wt%至4.5 wt%曱基脫氫皮質固醇的Pluronic F-127配方耳 内注射。動物在第1天給藥。第9圖顯示基於周邊淋巴分析 測試的配方之釋放曲線。在1.5%曱基脫氫皮質固醇療方 中,在第7-10天的曝露量為具有平均滯留時間約3.5天的 Cmax之約10%。在4.5%甲基脫氫皮質固醇療方中,曝露量 維持在相似或高於在第1天觀察到的量至少10天,且一預 計平均滯留時間為超過18天。 實施例24 -在AIED動物模型中評估皮質類固醇配方 方法及物質 免疫回應的誘發process. Degree of DS 140447.doc 200950787 solution time (MDT)). Furthermore, when the P407 concentration for the 6% DSP formulation (6% Dex-P(*)) was increased from 17% to 19%, a faster gel removal was observed, as shown in Figure 8. Therefore, the injection volume and concentration of corticosteroids in the formulations described herein are tested to determine the optimal parameters for preclinical and clinical trials. It has been observed that an in-ear formula with a high concentration of DSP has a release profile that is different from an in-ear formula with a lower concentration of DSP. Example 23 - 21-day-old zigzag (Charles River, 200-300 g female) with a release kinetic energy in vivo and in vivo was buffered at 50 pL 17% at 280 mOsm/kg and had a formulation of 1.5 wt% Intra-injection of Pluronic F-127 formulation to 4.5 wt% thiol dehydrocorticosteroid. Animals were dosed on day 1. Figure 9 shows the release profile of the formulation based on the peripheral lymphatic analysis. In the 1.5% thiol dehydrocorticosterol treatment, the exposure on days 7-10 was about 10% of the Cmax with an average residence time of about 3.5 days. In the 4.5% methyl dehydrocorticosterol treatment, the exposure was maintained at a similar or higher than the amount observed on Day 1 for at least 10 days, and a predicted average residence time was over 18 days. Example 24 - Evaluation of corticosteroid formulations in animal models of AIED Methods and substances Induction of immune response

使用美國國家衛生院之重20至24 g的雌性白瑞士小鼠 (Swiss鼠)(美國印第安納州印第安納波里市Harlan Sprague-Dawley 公司)。匙孔血藍蛋白(Keyhole limpet hemocyanin,KLH ;加拿大維尼斯市 Pacific Biomarine Supply公司)懸浮於磷酸鹽-緩衝生理食鹽水((PBS) IpH 140447.doc -121 - 200950787 6.4)中,相對PBS無菌滲析並離心二次。沉澱物(附隨KLH) 溶解於PBS並皮下注入動物背部(0.2 mg在Freund完全佐劑 乳化)。動物給予一追加劑(0.2 mg KLH於Freund不完全佐 劑),並接著在10週後以0.1 mg KLH於5 μΐ PBS(pH 6.4) 注入通過耳蝸囊的微孔。使用操作顯微鏡及滅菌技術接近 耳蝸。完成一耳後切口並鑽一孔至氣泡以容許耳蝸底回、 鐙骨動脈及内耳圓窗窩之岬角可視性。燒灼鐙骨動脈並去 除,及鑽一 25 μπι孔通過耳堝囊至側底回的鼓階。KLH或 PBS對照組為緩慢的使用耦接一塑膠管的Hamilton注射器 至注入一填充抗原或對照組之玻璃微滴定管。此孔在注射 後以骨壤密封,且除去過量的液體。每動物僅一耳堝以 KLH處理。 治療 KLH及對照組鼠分為二組(每組n= 10)。含有甲基脫氫皮 質固醇的實施例1之皮質類固醇配方施用至一組動物的内 耳圓窗膜。不含有甲基脫氫皮質固醇的對照配方施用至第 二組。此甲基脫氫皮質固醇及對照配方在最初施用後三天 再施用。在7天治療後殺死此動物。 結果分析 電氣生理測試 在最初及在實驗步驟1週後以敲擊刺激每一動物的每一 耳測量聽覺腦幹回應底限(ABR)的聽力底限。動物置於單 壁環境室(美國紐約州布良克士市Industrial Acoustics公司) 於一加熱墊上。皮下電極(美國羅德島州西瓦沃克市Astro- 140447.doc -122- 200950787Female white Swiss mice (Swiss rats) weighing 20 to 24 g from the National Institutes of Health (Harlan Sprague-Dawley, Inc., Indianapolis, Indiana) were used. Keyhole limpet hemocyanin (KLH; Pacific Biomarine Supply, Vic, Canada) was suspended in phosphate-buffered saline (PBS) IpH 140447.doc -121 - 200950787 6.4, sterile dialysis against PBS And centrifuged twice. The pellet (attached to KLH) was dissolved in PBS and injected subcutaneously into the back of the animal (0.2 mg emulsified in Freund's complete adjuvant). Animals were given a supplement (0.2 mg KLH in Freund incomplete adjuvant) and then injected into the micropores through the cochlear sac with 0.1 mg KLH in 5 μM PBS (pH 6.4) after 10 weeks. Use the operating microscope and sterilization technology to access the cochlea. The incision is made after one ear and a hole is drilled into the bubble to allow visibility of the cochlear gyrus, the radial artery and the rounded window of the inner ear. Cauterize the iliac artery and remove it, and drill a 25 μπι hole through the ear sac to the basal back of the basal ganglia. The KLH or PBS control group was slow to use a Hamilton syringe coupled to a plastic tube to inject a glass microtiter filled with antigen or control. This well is sealed with bone soil after injection and excess liquid is removed. Only one deafness per animal was treated with KLH. Treatment KLH and control mice were divided into two groups (n=10 per group). The corticosteroid formulation of Example 1 containing methyl dehydrocorticosteroid was applied to the inner ear window membrane of a group of animals. A control formulation containing no methyldehydrocorticosterol was administered to the second group. This methyl dehydrocorticosterol and control formulation was administered three days after the initial administration. The animals were killed after 7 days of treatment. Analysis of Results Electrophysiological Testing The hearing floor of the auditory brainstem response bottom (ABR) was measured by tapping each ear of each animal initially and after 1 week of the experimental procedure. The animals were placed in a single walled environmental chamber (Industrial Acoustics, Brooks, NY, USA) on a heating mat. Subcutaneous electrode (Astro-140447.doc -122- 200950787, Siva Walk, Rhode Island, USA)

Med公司Grass Instrument分公司)插入顱頂(作用電極)、 乳突(參考電極)及後腿(接地線)。敲擊刺激(0.1毫秒)以電 腦產生並傳送至附有一耳窺器的Beyer DT 48、200 Ohm 放大器置於外聽道。放大記錄的ABR並以一電池操作之預 放大器數位化並輸入一Tucker-Davis Technologies ABR^ 錄系統,其提供刺激、記錄及平均作用的電腦控制(美國 佛羅里達州甘尼佛爾市Tucker Davis Technology公司)。呈 現為5-dB步階之相繼降低振幅刺激至動物,且平均記錄刺 激-出現活性(n=5 12)並顯示。底限定義為不可視可彳貞測 回應及清楚可辨回應間的刺激量。 組織化學分析 麻醉動物並經心内灌注肝素化温食鹽水死亡及接著以大 約40 ml過碘酸-離胺酸-三聚甲醛(4%三聚甲醛最終濃度)固 定。右側顳骨立即移除並以緩衝之5%亞乙基二胺四-醋酸 鹽(pH 7.2)脫鈣化14天(4°C)。在去鈣化作用後,顳骨接著 浸潰於增加濃度(50%、75%、100%)之冰凍包埋劑(〇ct) 化合物(美國印第安那州伊哈特市Tissue-Tek,Miles公 司)、快速冷凍(_7〇°C ),並平行骨軸冷凍切片(4 μιη)。收 集的切片以蘇木精及伊紅(Η&Ε)染色及免疫組織化學分 析。 以鼓階的細胞滲濾量評量的發炎嚴重性,且每一耳蝸給 予一無偏估計量。分數0表示無發炎,而分數5表示所有的 耳蝎回具有嚴重的發炎細胞滲濾。 實施例25-—皮質類固醇配方配方在中耳炎動物模型的評估 140447.doc •123- 200950787 中耳炎的誘發 於此些研究中使用重量400至600 g具有正常中耳且以耳 視鏡及鼓室圖檢查之健康成粟鼠。在接種前進行耳咽管障 礙24小時以預防接種液由耳咽管流出。1 ml第3型 S.pneumoniae菌株以4-h-log相(含有大約40菌落形成單位 (CFU))直接置於長尾栗鼠二中耳之低鼓膜泡上。對照組鼠 以1 ml滅菌 PBS培育。 治療 S. pneumoniae培育及對照組鼠分為二組(每組n=l 0)。含 有實施例2的去氫皮質醇配方施用至一组動物的鼓室壁。 不含有去氫皮質醇的對照配方施用至第二組。此去氫皮質 醇及對照配方在最初施用後三天再施用。在7天治療後殺 死此動物。 結果分析 中耳液體(MEF)在鏈球菌接種後於1、2、6、12、24、 48及72小時取樣。定量MEF培養基在羊血洋菜上進行,定 量底限設定於50 CFU/ml。發炎細胞以血球計計量,且分 化細胞數Wright氏染色進行。 實施例26-使用甲基脫氫皮質固醇配方的AIED臨床實驗 選出10成人患者,其先前回應系統之曱基脫氫皮質固醇 治療,但目前因不良事件而中斷治療。實施例1之曱基脫 氫皮質固醇熱的可逆凝膠配方經由穿刺鼓膜投藥至每一患 者的圓窗膜。在最初給藥的7天後再次給予曱基脫氫皮質 固醇之凝膠配方,且在治療的2及3週再次給藥。 140447.doc -124- 200950787 對每一患者進行由純音聽力測驗(250-8,000 Hz)及使用 法文雙音節字表的語音測試組成的聽力評估。測試在施用 曱基脫氫皮質固醇配方之前及在最初治療後1、2、3及4週 進行。 實施例27-去氫皮質醇在聽覺受損之鼠模型評估 方法及材料 '' 耳毒性的誘發 使用 12 隻重 20 至 24 克的鼠(Harlan Sprague-Dawley 氣)。 測定在4-20 mHz聽性腦幹誘發反應(ABR)基線。此鼠麻醉 並曝至響度為120 dB之6 kHz的一連續純音中30分鐘。 治療 對照組(n=10)在聽力受損後給予食鹽水。實驗組在聽力 受損後給予於實施例2配製的去氫皮質醇(2.0 mg/kg of體 重)。 電氣生理測試 φ 在最初及在實驗步驟1週後以敲擊刺激每一動物的每一 耳測量聽性腦幹誘發反應低限(ABR)的聽力低限。動物置 於單壁環境室(美國紐約州布良克士市Industrial Acoustics 公司)於一加熱墊上。皮下電極(美國羅德島州西瓦沃克市 Astro-Med公司Grass Instrument分公司)插入顧頂(作用電 極)、乳突(參考電極)及後腿(接地線)。敲擊刺激(0.1毫秒) 以電腦產生並傳送至附有一耳窺器的Beyer DT 48、200 Ohm 放大器置於外聽道。放大記錄的ABR並以一電池操作之預 放大器數位化並輸入一 Tucker-Davis Technologies ABR記 140447.doc -125- 200950787 錄系統,其提供刺激、記錄及平均作用的電腦控制(美國 佛羅里達州甘尼佛爾市Tucker Davis Techn〇l〇a公司)。呈 現為f dB步階之相繼降低振幅刺激至動物,且平於~ h 二卞吟圮錄刺 激-出現活性(η=512)並顯示。底限定義為介不可視可㈣ 回應及清楚可辨回應間的刺激量。。 實施例28-甲基脫氫皮質固醇在梅尼爾氏症患者上的臨 實驗 研究目的 ' 此研究的主要目的為評估曱基脫氫皮質固醇與安慰劑組 ❹- 在人類個體改善受梅尼爾氏症折磨之患者的耳鳴症狀之安 全性及功效。 研究設計 此為一 3相、多中心、雙盲、隨機、安慰劑-對照、3臂 平行研究以比較JB004/A與安慰劑組在耳鳴的治療。約Μ。 患者登記於此研究中,且依贊助人準備的隨機順序以隨機 (1:1)分為3治療組。每一組接受3〇〇 mg以熱可凝膠逆傳送 的甲基脫氫皮質固醇,或控制釋放的安慰劑配方。甲基脫 ❹ 氫皮質固醇的釋放為控制釋放且在30天中發生。投藥路經 - 為耳内注射。 & · 主要成果指標 . 視覺類比量表(VA s )以測量在投藥2小時後量測時感知之 耳鳴響度的改變(或在任何其他時間點相對預_劑量基線)。 可替代地,在健康的耳使用聽力測定以配合在受影響耳的 耳鳴音調。 β 140447.doc •126- 200950787 次要成果指標 VAS係測量耳鳴立 馬a頻、痛苦及焦慮。純音聽力測 神性聽覺評估。睡眠 驗&精 耳鳴問卷藥的安全性、耐受性 物動力學。[時間框靼.+ 又性及樂 在任何其他_點㈣預.㈣基線)。 ^ 選用原則 …者若符σ下列原則的任何之—者則選用: -具有診斷耳鳴的男或女個體。 -個體願意限制酒精攝取。 _有生育可能但同意放棄性交或同意節育的婦女。 •無生育可能的婦女。 排除原則 選用的患者若具下列原則的任何之一者則排除: -間歇或脈衝式耳鳴 -個體具有病理級之焦慮或憂鬱。 ❹ -個體無聽力障礙且具有正常聽力。 -個體對利多卡因注入測試無反應或在預注入值顯示一 較大變異。 -存在任何手術或醫療症狀’其可能干擾藥之pK。 _具肝失能或肝功功能異常病史之個體。 -具腎失能個體。 -具HIV、C型肝炎或Β型肝炎陽性之個體。 -個體具不正常之實驗室、ECG或身體檢查判定。 -個體無甲狀腺。 140447.doc -127· 200950787 -個體具肝、心臟、腎、神經、腦血管、代謝或肺疾 病。 -個體有心肌梗塞。 -具癲癇失調病史之個體。 -具癌症病史之個體。 -具藥或其他過敏之個體。 · -個體對藥使用為陽性及/或物質濫用或依賴病史。 _ -個體在特定時間框加服用精神異常藥或抗憂鬱劑。 ^ -已知會干擾肝酶的藥劑或食物(例如葡萄柚或葡萄柚 ©* 果汁)。 -個體具血清素(serotonergic)作用機制之非精神異常 用藥。 -個體目前已使用一研究用藥或最近參予—臨床實驗。 -女性具懷孕測試為陽性反應。 _欲懷孕女生或將在此研究之最後研究藥投藥後的4週 内當父親的男性。 -個體’在前數個用已捐一單位的血或更多或欲在完成 〇 研究的一個月内捐血。 實施例29 ·甲基脫氫皮質固醇配方在内淋巴積水動物㈣ 中的評估 本步驟為用測定於實施例丨製備之曱基脫氫皮質固醇配 方的效能。 材料與方法 使用35天大的Hartley天竺鼠,其具有正向隨音源轉向特 140447.doc -128- 200950787 性(positive Preyer’s reflex)且重量約3〇〇 g。5隻做為對照 組(正常耳組)在無手術或無治療下餵食5週,而其他3〇隻 做為實驗動物。所有實驗動物接受電燒灼内淋巴囊(Lee 等人之Acta Otolaryngol. (1992) 112 : 658-666 ; Takeda等 人之Equilib. Res. (1993) 9 : 139-143)。在手術後四週,此 些動物分為無-灌注水腫耳、載劑_處理水腫耳及甲基脫氫 皮質固醇-處理水腫耳三組,每組1〇動物。無灌注水腫耳 ❹ 組未接文治療,除了電燒灼内淋巴囊。載劑_處理水腫耳 及甲基脫氫皮質固醇-處理水腫耳組中,施用脂質體配方 至圓窗膜。在組成物投藥1週後,殺死所用動物以評估内 淋巴空間的改變。所有動物在整個實驗期間除了執行實驗 步驟時外,其等皆未受干擾且自由在安靜室中的各自籠中 行動。 在評估内淋巴空間的改變中,所有動物以生理食鹽水在 腹部注射戊巴比妥的深度麻醉下以穿心灌注,並在1〇%福 • 馬林中進固定。移出左顳骨並以10°/。福馬林溶液後固定1〇 • 天或更久。接著,其等以5%三氯乙酸去鈣化12天且在級 度乙醇系列脫水。其包埋於石臘及火棉膠中。此製備的塊 以水平方向下6 μιη切片。切片以蘇木精及伊紅染色並在光 顯微鏡下觀察。依Takeda方法進行内淋巴空間的改變評估 (Takeda 等人之Hearing Res. (2003) 182 : 9-18)。 實施例30-耳内甲基脫氫皮質固醇在自發性瞬間感音神經 性聽力喪失(ISSHL)的評估 研究目的 140447.doc -129- 200950787 此研九的主要目的為做口服類固醇治療或耳内(it)類固 醇治療的安全性及效能。 主要結果指標 平均純音聽力(PTA)及詞彙辨識為相等權重的終點值; 對於語音鑑別評分,使用一 50-字單音節系統;在ρτΑ或全 部或部份頻率的大於20 dB的改良,在該頻率缺失為大於 30 dB,及/或在WDS之20%或更多的改良;除了絕對的改 變外,亦測定有關對侧耳的恢復。 完全恢復-恢復至相對側語音鑑別分數的5%點,或在相 對側的PTA之5 dB9内。 研究設計 此為一多中心、雙盲、隨機、安慰劑-對照、平行組研 究以比較甲基脫氫皮質固醇與安慰劑的ISShl治療。大約 1 40個體在此研究中登記,且依隨機順序以隨機(丨··丨)分為3 治療組。 ••在組I的個體接受口服普賴松(給予1 mg/kg/天普賴松 14天’接著以日劑量減少1〇 mg直至不再給予類固醇) -在組II的個體接受IT曱基脫氫皮質固醇磷酸鈉(以每月 給予0.3-0.5 mL的甲基脫氫皮質固醇/每mL載劑的1次注射 直至最南達到3次注射)及口服普賴松(給予1 mg/kg/天普賴 松14天,接著以日劑量減少1 〇 mg直至不再給予類固醇) •在組III的個體接受安慰劑IT注射(以每月給予0·3-〇·5 mL 載劑的1次注射直最高達到3次注射)及口服普賴松 聽力評估 140447,doc -130- 200950787 聽力評估包含: -平均純音聽力(500 Hz、1&2 kHz ; 4、6&8 kHz)。 測定二PTA值:一低頻值(500 Hz-2kHz)及一高頻值 (4-8 kHz)。 -鐙骨肌反射 •鼓室圖檢查&響音衰減檢查 ' -語音辨識閥值 ❿ 在治療開始前’測量每一個體的聽力喪失(在此研究分 配前二次及在隨機分配前一次)。在治療開始後於1、2、 4&8週、4&6個月評估聽力。 主要選用原則 -介於18至75歲間的男性或女性患者 -單側SHL(感音神經性聽力喪失)在72小時内進展 -個體在任一不超過70 dB的頻率具聽力喪失 排除原則 φ -在先前的30天内因任何原因進行多過10天的居先之口 _ 服類固醇治療 -在先前的14天内因ISSHL因進行5或更多天的居先之 口服類固醇治療 -在任一耳的聽力變動病史 實施例31 -耳内甲基脫氫皮質固醇在梅尼爾氏症投藥的評估 研究目的 此研究的主要目的為評估耳内(IT)曱基脫氫皮質固醇在 人類個體改善梅尼爾氏症的安全性及功效。 140447.doc -131 - 200950787 主要療效指標 暈眩 以下列體系之自我描述系統--無暈眩天數分; -具有温和侵襲的天數-1 ; -中等嚴重侵襲持續大於20分鐘-2; -嚴重侵襲持續1小時或更多或伴有噁心或喔、U ; -至目前為最壞的侵襲attack to date-4 ;Med's Grass Instrument branch) inserts the cranial (active electrode), mastoid (reference electrode) and hind leg (grounding wire). The tapping stimulus (0.1 ms) was generated by the brain and transmitted to a Beyer DT 48, 200 Ohm amplifier with an ear speculum placed in the external auditory canal. Amplify the recorded ABR and digitize it with a battery operated preamplifier and enter a Tucker-Davis Technologies ABR recording system that provides computer control for stimulation, recording, and averaging (Tucker Davis Technology, Inc., Ghana, Florida, USA) ). The amplitude-stimulated stimulation was successively reduced to the animals in a 5-dB step, and the stimuli-activity was recorded on average (n = 5 12) and was shown. The bottom limit is defined as the amount of stimulus that is invisible and can be used to detect responses and to clearly distinguish between responses. Histochemical analysis Anesthetized animals were sacrificed by intracardiac perfusion of heparinized warm saline and then fixed with approximately 40 ml of periodic acid- lysine-trimaldehyde (4% final concentration of paraformaldehyde). The right tibia was removed immediately and decalcified with buffered 5% ethylenediamine tetra-acetate (pH 7.2) for 14 days (4 °C). After decalcification, the tibia is then impregnated with increasing concentrations (50%, 75%, 100%) of frozen embedding agent (〇ct) compound (Tissue-Tek, Miles, Inc., Ihart, USA), fast Freeze (_7〇 °C) and freeze the sections (4 μιη) in parallel with the bone axis. The harvested sections were stained with hematoxylin and eosin (Η & Ε) and immunohistochemically analyzed. The severity of inflammation was assessed by cytosolic diafiltration and each cochlea was given an unbiased estimator. A score of 0 indicates no inflammation, while a score of 5 indicates that all of the deafness has severe inflammatory cell percolation. Example 25 - Evaluation of Corticosteroid Formulations in Animal Models of Otitis Media 140447.doc • 123- 200950787 Induction of Otitis Media In these studies, 400 to 600 g of weight were used with normal middle ear and examined with otoscope and tympanogram Healthy into a rat. The Eustachian tube barrier was performed for 24 hours prior to inoculation to prevent the inoculum from flowing out of the Eustachian tube. 1 ml of type 3 S. pneumoniae strain was placed directly on the low tympanic membrane of the middle ear of the long-tailed chinchilla in a 4-h-log phase (containing approximately 40 colony forming units (CFU)). Control rats were incubated with 1 ml of sterile PBS. Treatment S. pneumoniae culture and control mice were divided into two groups (n=l 0 in each group). The dehydrocortisol formulation containing Example 2 was applied to the tympanic wall of a group of animals. A control formulation containing no dehydrocortisol was administered to the second group. This dehydrocortisol and control formulation were re-administered three days after the initial administration. The animals were killed after 7 days of treatment. Analysis of Results Middle ear fluid (MEF) was sampled at 1, 2, 6, 12, 24, 48 and 72 hours after streptococcal inoculation. Quantitative MEF medium was performed on amniotic fluid, and the quantitative limit was set at 50 CFU/ml. Inflammatory cells were measured by a hemocytometer and the number of differentiated cells was stained by Wright's stain. Example 26 - AIDD Clinical Trial Using Methyl Dehydrocorticosterol Formulation 10 adult patients were selected who had previously responded to the system of thiol dehydrocorticosteroid treatment, but currently discontinued treatment due to adverse events. The thiol dehydrocorticosteroid hot reversible gel formulation of Example 1 was administered via a puncture tympanic membrane to the round window membrane of each patient. A gel formulation of thiol dehydrocorticosterol was administered again 7 days after the initial administration and re-administered at 2 and 3 weeks of treatment. 140447.doc -124- 200950787 A hearing assessment consisting of a pure tone audio test (250-8,000 Hz) and a speech test using a French two-syllable word list was performed for each patient. The tests were performed prior to the administration of the thiol dehydrocorticosterol formulation and at 1, 2, 3 and 4 weeks after the initial treatment. Example 27 - Assessment of dehydrocortisol in a rat model of hearing impairment Methods and materials '' Induction of ototoxicity Twelve mice weighing 20 to 24 grams (Harlan Sprague-Dawley gas) were used. The baseline of the auditory brainstem evoked response (ABR) at 4-20 mHz was determined. The rats were anesthetized and exposed to a continuous pure tone of 6 kHz with a loudness of 120 dB for 30 minutes. Treatment The control group (n=10) was given saline after hearing loss. The experimental group was given dehydrocortisol (2.0 mg/kg body weight) prepared in Example 2 after hearing loss. Electrophysiological testing φ The hearing deficit of the auditory brainstem evoked response low (ABR) was measured by tapping each ear of each animal initially and 1 week after the experimental procedure. The animals were placed in a single wall environmental chamber (Industrial Acoustics, Bryan, NY, USA) on a heating mat. The subcutaneous electrode (Grass Instrument Branch, Astro-Med, Siva Walk, Rhode Island, USA) was inserted into the top (acting electrode), the mastoid (reference electrode), and the hind leg (grounding wire). The tap stimulus (0.1 ms) was generated by a computer and transmitted to a Beyer DT 48, 200 Ohm amplifier with an ear spectrometer placed in the external auditory canal. Amplify the recorded ABR and digitize it with a battery operated preamplifier and enter a Tucker-Davis Technologies ABR record 140447.doc -125- 200950787 recording system that provides computer control for stimulation, recording and averaging (Gani, Florida, USA) Tucker Davis Techn〇l〇a, Foer). The amplitude-stimulated stimuli of the f dB step were successively reduced to the animals, and the activity was observed (n=512). The bottom limit is defined as the amount of stimulus between (4) response and clearly identifiable response. . Example 28 - Experimental study of methyl dehydrocorticosteroids in patients with Meniere's disease Objectives The main objective of this study was to evaluate thiol dehydrocorticosteroids in the placebo group - improvement in human subjects The safety and efficacy of tinnitus symptoms in patients suffering from Meniere's disease. Study Design This was a 3-phase, multicenter, double-blind, randomized, placebo-controlled, 3-arm parallel study to compare the treatment of tinnitus in the JB004/A versus placebo groups. Joel. Patients were enrolled in this study and were randomized (1:1) into 3 treatment groups in a random order prepared by the sponsor. Each group received 3 mg of methyl dehydrocorticosterol, which was reversely delivered by a thermogel, or a controlled release placebo formulation. The release of methyl dehydrogenated cortisol was controlled release and occurred within 30 days. Medication route - for intra-ear injection. & · Major Outcome Indicators. Visual Analog Scale (VA s ) to measure changes in tinnitus loudness (or relative pre-dose baseline at any other time point) measured after 2 hours of dosing. Alternatively, a hearing test is used in a healthy ear to match the tinnitus tone in the affected ear. β 140447.doc •126- 200950787 Secondary Outcome Indicators VAS measures the frequency, pain and anxiety of tinnitus. Pure tone hearing test Divine hearing assessment. Sleep Test & Fine Tinnitus Questionnaire Safety, Tolerance Dynamics. [Time frame 靼.+ sex and music in any other _ point (four) pre. (four) baseline). ^ Principles of Selection ... If any of the following principles are used, then: - Have a male or female individual with a diagnosis of tinnitus. - Individuals are willing to limit alcohol intake. _ Women who have the possibility to give birth but agree to give up sexual intercourse or consent to birth control. • Women without fertility. Exclusion Principles Patients selected if they have any of the following principles are excluded: - Intermittent or pulsed tinnitus - Individuals with pathological grade anxiety or depression. ❹ - The individual has no hearing impairment and has normal hearing. - The individual does not respond to the lidocaine injection test or shows a large variation in the pre-injection value. - There are any surgical or medical symptoms that may interfere with the pK of the drug. _ Individuals with a history of liver disability or abnormal liver function. - Individual with kidney disability. - Individuals with HIV, hepatitis C or sputum hepatitis positive. - The individual has an abnormal laboratory, ECG or physical examination decision. - The individual has no thyroid. 140447.doc -127· 200950787 - Individual with liver, heart, kidney, nerve, cerebrovascular, metabolic or pulmonary disease. - The individual has a myocardial infarction. - Individuals with a history of epilepsy disorders. - Individuals with a history of cancer. - Individuals with medication or other allergies. · Individuals are positive for drug use and/or substance abuse or dependence history. _ - Individuals take a psychotropic or antidepressant at a specific time frame. ^ - Agents or foods known to interfere with liver enzymes (eg grapefruit or grapefruit ©* juice). - Individual non-mental abnormalities with serotonergic mechanism of action. - The individual has currently used a study medication or recent participation - clinical trials. - Females have a positive pregnancy test. _ Pregnant women or men who will be fathers within 4 weeks after the last study of this study. - Individuals donated blood in the first few months with a donation of one unit of blood or more or one month after completing the study. Example 29 - Evaluation of methyl dehydrocorticosterol formulation in endolymphatic water animals (IV) This step is the efficacy of the thiol dehydrocorticosteroid formulation prepared in the Example 测定. MATERIALS AND METHODS A 35-day-old Hartley guinea pig was used with positive forward sounding of 140447.doc -128-200950787 (positive Preyer's reflex) and weighing approximately 3 〇〇 g. Five of them were used as control group (normal ear group) for 5 weeks without or without treatment, while the other 3 groups were only used as experimental animals. All experimental animals received an electrocauterized endolymphatic sac (Ere et al., Acta Otolaryngol. (1992) 112: 658-666; Takeda et al., Equilib. Res. (1993) 9: 139-143). Four weeks after the operation, the animals were divided into three groups: no-perfusion edema ear, carrier _ treatment of edema ear and methyl dehydrocorticosteroid-treated edema ear, one group of animals per group. No perfusion edema ear ❹ group did not receive treatment, in addition to electric cauterization of the endolymphatic sac. In the vehicle-treated edema ear and methyl dehydrocorticosteroid-treated edema ear group, the liposome formulation was applied to a round window membrane. One week after the composition was administered, the animals used were sacrificed to assess changes in endolymphatic space. All animals were undisturbed and free to move in their respective cages in a quiet room, except during the entire experimental period. In the assessment of changes in the endolymphatic space, all animals were perfused with a saline solution under deep anesthesia with pentobarbital in the abdomen and fixed in 1% F. Marlin. Remove the left tibia and set it at 10°/. The formalin solution is fixed for 1 〇 • days or longer. Next, it was decalcified by 5% trichloroacetic acid for 12 days and dehydrated in a grade ethanol series. It is embedded in paraffin and fire cotton glue. The prepared block was sliced in a horizontal direction of 6 μm. Sections were stained with hematoxylin and eosin and observed under a light microscope. Endolymphatic space change assessment was performed according to the Takeda method (Takeda et al., Hearing Res. (2003) 182: 9-18). Example 30 - Assessment of Spontaneous Instantaneous Sensorineural Hearing Loss (ISSHL) in Methyl Dehydrocorticosteroids 140148.doc -129- 200950787 The main purpose of this study is to do oral steroid therapy or ear Safety and efficacy of endogenous (it) steroid therapy. The primary outcome measures were plain tone hearing (PTA) and vocabulary recognition as endpoints of equal weight; for speech discrimination scores, a 50-word monosyllabic system was used; improvements in ρτΑ or all or part of the frequencies greater than 20 dB were The frequency loss is greater than 30 dB, and/or a 20% or more improvement in WDS; in addition to the absolute change, the recovery of the contralateral ear is also determined. Full recovery - restores to 5% of the opposite side speech discrimination score, or within 5 dB9 of the opposite side PTA. Study Design This was a multicenter, double-blind, randomized, placebo-controlled, parallel group study comparing ISShl treatment with methyldehydrocorticosterol and placebo. Approximately 1 40 individuals were enrolled in this study and were randomized into 3 treatment groups in random order (丨··丨). • Individuals in group I received oral Prysson (administered 1 mg/kg/day of prednisone for 14 days followed by daily dose reduction of 1 〇mg until no longer administered steroids) - Individuals in group II received IT thiol Dehydrocorticosterone sodium phosphate (administered with 0.3-0.5 mL of methyl dehydrocorticosterol per month/one injection per mL of carrier until up to 3 injections) and oral Prysson (1 mg administered) /kg/day Pleisson for 14 days, followed by a daily dose reduction of 1 〇mg until steroids are no longer administered) • Individuals in group III receive placebo IT injection (to a monthly dose of 0·3-〇·5 mL vehicle) One injection up to 3 injections) and oral Pryson hearing assessment 140447, doc -130- 200950787 Hearing assessment includes: - Average pure tone hearing (500 Hz, 1 & 2 kHz; 4, 6 & 8 kHz). The two PTA values were determined: a low frequency value (500 Hz-2 kHz) and a high frequency value (4-8 kHz). - Tibial muscle reflexes • Tympanogram examination & loudness attenuation check - - Speech recognition threshold ❿ Measure the hearing loss of each individual before the start of treatment (two times before the study is assigned and before the random assignment). Hearing was assessed at 1, 2, 4 & 8 weeks, 4 & 6 months after the start of treatment. The main selection principle - male or female patients between the ages of 18 and 75 - unilateral SHL (sensorineural hearing loss) progresses within 72 hours - the individual has a hearing loss exclusion principle at any frequency not exceeding 70 dB φ - In the previous 30 days for more than 10 days for any reason _ steroid treatment - in the previous 14 days due to ISSHL for 5 or more days of oral steroid treatment - hearing in either ear Change History Example 31 - Assessment of the administration of methyl dehydrocorticosteroids in the treatment of Meniere's disease The purpose of this study was to evaluate the improvement of the in vivo (IT) thiol-dehydrocorticosteroids in human individuals. The safety and efficacy of Niel's disease. 140447.doc -131 - 200950787 The main efficacy indicators are dizzy with the following system of self-description system - no dizziness days; - days with mild invasion -1; - moderate severe attacks lasting more than 20 minutes -2; - severe attack Sustained for 1 hour or more or accompanied by nausea or convulsions, U; - to the present worst attack attack to date-4;

-當月暈眩分數連續二個月為50或大於50定義為治療失效 選用原則 依1995 AAO-HNS原則的MD臨床診斷: •至少二決定性的暈眩侵襲。 · -一決定性長時間為維持至少20分鐘的自發性(旋轉)暈 眩0 排除原則- The monthly dizziness score is defined as 50 or more than 50 consecutive months for treatment failure. Principles of selection According to the 1995 AAO-HNS principle of MD clinical diagnosis: • At least two decisive dizziness attacks. · - A decisive long time to maintain a spontaneous (rotating) dizziness of at least 20 minutes 0 Elimination principle

-以胺基糖苷或巨環内酯抗生素; -以抗癌藥治療 -始化合物, -二氟曱基鳥胺酸。 研究設計 此為-多中心、雙盲、隨機、安慰劑·對照 :比較耳内曱基脫氫皮f固醇與安慰劑⑽狐治療。 約140個體在此研究中登記,且 為3治療組。 隨機順序以隨機㈤ 140447.doc -132- 200950787 在組1的個體接受標準照護(nmt 1500mg/天之鈉飲 食’戒斷二經基噪呤吸收及/或利尿劑) -在組11的個體接受IT甲基脫氫皮質固醇磷酸鈉(以每 月給予0.3-0.5 mL的甲基脫氫皮質固醇/每„^載劑的1次 . 注射直至最高達到3次注射)及標準照護 -在組111的個體接受安慰劑IT注射(以每月給予0.3-0.5 mL載劑的1次注射直最高達到3次注射)及標準照護 • 評估 ❹ ― 在治療開始前,測量每一個體的梅尼爾氏症的嚴重性 (在此研究分配前二次及在隨機分配前一次) 在治療開始後於1、2、4&8週、4&6個月進行梅尼爾氏症 評估聽力 評估 -暈眩及耳鳴的發生曰期、頻率、時間及嚴重性; -降低耳壓感知,使用標準VAS問卷及有效評比協定 φ -血清血管增壓素的量測 雖然在本文已呈現及描述本發明之較佳實施例,此些提 供的實施例僅為例示之用。可選擇本文所述之實施例的多 種變化用於貫施本發明。欲以後文之申請專利範圍界定本 發明且因而涵括在該申請專利範圍之範疇中的方法與結構 及其等效物。 【圖式簡單說明】 第1圖說明具不同濃度的泊洛沙姆4〇7(p〇i〇xamer 407)之 曱基脫氫皮質固醇配方的活體外釋放曲線。 140447.doc -133· 200950787 第2圖說明介於配方之平均溶解時間(]V1DT)與P4〇7濃度 間的關聯性。 第3圖說明含有17% P407的不同類固醇的釋放曲線皮質 酮0 第4圖說明介於配方之平均溶解時間(MDT)與配方之外 觀黏性間的關聯性。 第5圖說明濃度在Blanose精製之CMC的水溶液之黏性上 的效用。 第6圖說明濃度在Methocel的水溶液之黏性上的效用。 第7圖顯示在天竺鼠耳進行耳内注射達到5天後的凝膠結 果。 第8圖顯示本文所描述的配方之凝膠消除時間進程。 第9圖顯示本文所描述的配方之釋放曲線。 140447.doc 134-- using an aminoglycoside or macrolide antibiotic; - treating with an anticancer drug - starting compound, - difluorodecylguanine. Study Design This was a multi-center, double-blind, randomized, placebo-controlled: comparison of urinary dehydrogenated skin sterols with placebo (10) fox treatment. Approximately 140 individuals were enrolled in this study and were 3 treatment groups. Randomized randomized (five) 140447.doc -132- 200950787 Individuals in group 1 received standard care (nmt 1500 mg/day sodium diet 'without two-base sputum absorption and/or diuretic) - accepted in group 11 IT methyl dehydrocorticosterone sodium phosphate (administered with 0.3-0.5 mL of methyl dehydrocorticosterol per month / 1 carrier per injection. Injection up to 3 injections) and standard care - at Individuals in group 111 received a placebo IT injection (up to 3 injections with 0.3-0.5 mL of vehicle per month for up to 3 injections) and standard care • Assessment ― - Measurement of each individual's Meni before treatment begins The severity of the disease (second time before the study was assigned and before the randomization). Meniere's assessment hearing assessment was performed at 1, 2, 4 & 8 weeks, 4 & 6 months after the start of treatment - Dizziness and tinnitus occurrence cycle, frequency, time and severity; - reduction of ear pressure perception, using standard VAS questionnaire and effective rating agreement φ - serum vasopressin measurement although the present invention has been presented and described herein Preferred embodiments, such provided embodiments are merely examples A variety of variations of the embodiments described herein may be employed to implement the present invention. The method and structure of the present invention, and thus the scope of the scope of the patent application, and the like [Ease of the diagram] Figure 1 illustrates the in vitro release profile of thiol dehydrocorticosterol formulations with different concentrations of poloxamer 4〇7 (p〇i〇xamer 407). -133· 200950787 Figure 2 illustrates the correlation between the average dissolution time of the formulation (]V1DT) and the concentration of P4〇7. Figure 3 illustrates the release profile of different steroids containing 17% P407. Cortisol 0 Figure 4 illustrates The correlation between the average dissolution time (MDT) of the formulation and the apparent viscosity of the formulation. Figure 5 illustrates the effect of the concentration on the viscosity of the aqueous solution of BMC that is refined by Blanose. Figure 6 illustrates the concentration of the aqueous solution in Methocel. Viscosity effect. Figure 7 shows the results of gelation after 5 days of intrathecal injection of guinea pig ears. Figure 8 shows the gel elimination time course of the formulations described herein. Figure 9 shows the Release of formula Curve. 140447.doc 134-

Claims (1)

200950787 七、申請專利範圍: 1. 一種用於治療一耳疾病或症狀的醫藥組成物,其經調製 以提供一治療有效量的甲基脫氫皮質固醇、甲基去氫皮 質醇或去氫皮質醇’該組成物包含甲基脫氫皮質固醇、 甲基去氫皮質醇或去氫皮質醇之實質低降解產物,,該 組成物更包含二或二以上選自下列的特性: ⑴介於約〇. 1 wt%約1 〇 Wt%間的甲基脫氫皮質固醇、 甲基去氫皮質醇或去氫皮質醇、或其之醫藥可接受前藥 或鹽; (ii) 介於約16 wt%約21 wt°/〇間的具通式E106 P70 E106之聚氧乙烯-聚氧丙烯三嵌共聚合物; (iii) 滅菌水,適量緩衝以提供介於約5 5至約8.〇間之 pH ; (iv) 多顆粒化甲基脫氫皮質固醇、甲基去氫皮質醇或 去氫皮質醇; (v) —介於約19°C至約40°C間之膠化温度; (VI) 每g配方之微生物劑少於約5〇菌落形成單位 (cfu),及 (VII) —個體之每公斤體重為少於約5内毒素單位。 2. 如請求項1所述之醫藥組成物,其中該組成物包含: (1)介於約〇. 1 wt%約10 wt%間的甲基脫氫皮質固醇、 曱基去氫皮質醇或去氫皮質酵、或其之醫藥可接受前藥 或鹽; (11)介於約16 wt%約21 wt%間的具通式ei〇6 P70 140447.doc 200950787 E106之聚氧乙烯_聚氧丙烯三嵌共聚合物;及 (ϋ〇多顆粒化甲基脫氫皮質固醇、甲基去氫皮質醇或 去氫皮質醇。 3.如請求項1所述之醫藥組成物,其中該組成物包含: ⑴介於約0.1 wt%約1〇 wt%間的曱基脫氫皮質固醇、 甲基去氫皮質醇或去氫皮質醇、或其之醫藥可接受前藥 或鹽; (11)介於約16 wt%約21 wt%間的具通式E106 P70 E106之聚氧乙烯_聚氧丙烯三嵌共聚合物; (出)多顆粒化曱基脫氫皮質固醇、曱基去氫皮質醇或 去氫皮質醇 (iv) —介於約i9°C至約4(rc間之膠化温度。 4·如明求項1至3項任一項所述之醫藥組成物,其中該組成 物提供介於約250至320 mOsm/L間的傳送容積滲透濃 度。 5. 如請求項丨至4項任一項所述之醫藥組成物,其中該曱基 脫鼠皮質固醇、曱基去氫皮質醇或去氫皮質醇由配方在 至少3天的期間釋放。 6. 如求項1至4項任一項所述之醫藥組成物,其中該甲基 脫氫皮質固醇、曱基去氫皮質醇或去氫皮質醇由配方在 至少5天的期間釋放。 7. 如印求項1至4項任一項所述之醫藥組成物,其中該曱基 脫氫皮質固醇、曱基去氫皮質醇或去氫皮質醇由配方在 至少10天的期間釋放。 140447.doc 200950787 8. 如請求項!至7項任一項所述之醫藥組成物,其中該醫藥 配方為一耳可接受之熱可逆凝膠。 9. 如請求項⑴項任-項所述之醫藥組成物,其中該聚氧 乙烯-聚氧丙烯三嵌段共聚合物為 爪如請求項⑴項任—項所述之醫藥組更包含黏 膜黏附劑。200950787 VII. Scope of Application: 1. A pharmaceutical composition for treating an ear disease or condition modulated to provide a therapeutically effective amount of methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrogenation Cortisol' The composition comprises a substantially low degradation product of methyl dehydrocorticosteroid, methyl dehydrocortisol or dehydrocortisol, and the composition further comprises two or more properties selected from the group consisting of: (1) Methyl dehydrocorticosteroid, methyl dehydrocortisol or dehydrocortisol, or a pharmaceutically acceptable prodrug or salt thereof, between about 1 %% 〇Wt%; (ii) A polyoxyethylene-polyoxypropylene triadlocene copolymer having a formula of E106 P70 E106 of about 16 wt% and about 21 wt% per day; (iii) sterilized water, suitably buffered to provide between about 5 5 and about 8 (iv) multiparticulate methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol; (v) - a gel between about 19 ° C and about 40 ° C (VI) less than about 5 colony forming units (cfu) per g of formula, and (VII) - less than 0.5 kg per kg of body weight 5 endotoxin units. 2. The pharmaceutical composition according to claim 1, wherein the composition comprises: (1) methyl dehydrocorticosterol, mercapto dehydrocortisol, between about 1 wt% and about 10 wt% Or dehydrocorticulin, or a pharmaceutically acceptable prodrug or salt thereof; (11) between about 16 wt% and about 21 wt% of a polyoxyethylene having a formula ei〇6 P70 140447.doc 200950787 E106 An oxypropylene tri-embedded copolymer; and (a multiparticulate methyl dehydrocorticosteroid, methyl dehydrocortisol or dehydrocortisol. 3. The pharmaceutical composition according to claim 1, wherein The composition comprises: (1) between about 0.1 wt% and about 1% by weight of decyl dehydrocorticosteroid, methyl dehydrocortisol or dehydrocortisol, or a pharmaceutically acceptable prodrug or salt thereof; 11) a polyoxyethylene-polyoxypropylene trimeric copolymer having a formula of E106 P70 E106 between about 16 wt% and about 21 wt%; (out) multiparticulate sulfhydryl dehydrocorticosterol, fluorenyl Dehydrocortisol or dehydrocortisol (iv) - a pharmaceutical composition according to any one of items 1 to 3, wherein the pharmaceutical composition of any one of items 1 to 3, Where the composition The osmotic concentration of the delivery volume of between about 250 and 320 mOsm/L. The pharmaceutical composition according to any one of claims 4 to 4, wherein the thiol de-corticosteroid, sulfhydryl dehydrogenation The pharmaceutical composition of any one of the above items 1 to 4, wherein the methyl dehydrocorticosterol, sulfhydryl dehydrogenation, is released from the formulation for at least 3 days. The medicinal composition of any one of items 1 to 4, wherein the thiol dehydrocorticosterol, thiol Hydrogen cortisol or dehydrocortisol is released from the formulation for at least 10 days. 140447.doc 200950787. The pharmaceutical composition of any one of clauses, wherein the pharmaceutical formulation is acceptable for one ear. The medicinal composition according to any one of the preceding claims, wherein the polyoxyethylene-polyoxypropylene triblock copolymer is a claw as described in the item (1) The medical group also contains mucoadhesives. 其更包含 其更包含 11. 如請求項1至10項任—項所述之醫藥組成物, 一穿透增進劑。 12. 如請求項1至!1項任一項所述之醫藥組成物, 一增稠劑。 13.如請求項1至12項任一 一染料。 項所述之醫藥組成物, 其更包含 14.如請求項1至13項任一項所述之醫藥組成物,其更包含 一藥傳送系統,其選自—針及注射器、栗、微注射二 置、及原處形成海綿物質或其等之組合。 〇 I5.如請求項1至14項任一項所述之醫藥組成物,其中該甲 基脫氫皮質㈣、甲基去氫皮質醇或去氫皮質醇^醫 藥可接受鹽具有限或為無系統釋放、系統毒性、不良叹 特性或其等之組合。 16. 如請求項丨至15項任—項所述之醫藥組成物,其中該甲 基脫氫皮質㈣、甲基去氫皮f醇或去氫皮㈣為^由 ^自由醇、鹽、如藥、或其等之組合之形式。 17. 如請求項16所述之醫藥組成物,該甲基脫氫皮質固醇、 曱基去氫皮質醇或去氫皮質酵以磷酸酯或酯前藥的形式 140447.doc 200950787 投藥。 18. 19. 20. 21. 22. 23. 24. 25. 如。月求項17所述之醫藥組成物,其中該類固醇為曱基脫 風皮質固醇麟酸酯或曱基脫氫皮質固醇乙酸酯。 如吻求項1至18項任一項所述之醫藥組成物,其更包含 曱基脫氫皮質固醇、曱基去氫皮質醇'去氫皮質醇、或 其醫藥可接文鹽、前藥或其之組合為一立即釋放劑。 凊求項1至19項任一項所述之醫藥組成物,其中該甲基 脫氫皮質固醇、曱基去氫皮質酵或去氫皮質醇包含多粒 子。 如明求項20所述之醫藥組成物,其中該曱基脫氫皮質固 醇、曱基去氫皮質醇或去氫皮㈣實質為微米化粒子形 式。 如叫求項1至2 1項任一項所述之醫藥組成物,其中該甲 基脫氫皮質固醇為微-曱基脫氫皮質固醇粉末的形式。 如明求項1至22項任一項所述之醫藥組成物,其更包含 一額外治療劑。 如請求項23所述之醫藥組成物,其中額外治療劑為— Na/K ATPase調變子、—化療劑、一膠原蛋白、一球蛋 白、一干擾素、一抗微生物劑、一抗生素、一局部作用 麻醉劑、一血小板活化因子拮抗劑、一耳保護劑、—氧 化氮合成酶抑制子、抗暈眩劑、血管加壓素结抗劑、= 抗病毒劑、一抗催吐劑或其等之組合。 如請求項丨至24項任一項所述之醫藥組成物,其中組成 物之pH為介於約6.〇至約7 6間。 I40447.doc 200950787 26. 如請求項〗至25項任一項所述之醫藥組成物,其中該具 有通式E106 P70 E106之聚氧乙烯_聚氧丙烯三嵌段=聚 物與一增稠劑的比例為由約4〇:丨至約5 · 1。 27. 如請求項26所述之醫藥組成物,其令該增稠劑為羧基甲 基纖維素、羥基丙基纖維素或羥基丙基甲基纖维素。 28. 如請求項1所述之醫藥組成物,其中該耳部疾病或症狀 •為梅尼爾氏症、突生之感音神經性聽力喪失、噪音引起 m 之聽力喪失、老化-有關聽力喪失、自體免疫耳疾病或耳 鳴。 29. —種治療耳部疾病或症狀的方法,其包含投藥予一需要 耳内組成物的個體,該組成物包含一治療有效量之甲基 脫氫皮質固醇、甲基去氫皮質醇或去氫皮質醇,該組成 物包含實質甲基脫氫皮質固醇、甲基去氫皮質醇或去氫 皮質醇之低降解產物,該組成物更包含二或二上選自下 列之特性: ❷ ⑴介於約0·1 wt%約10 Wt%間的曱基脫氫皮質固醇、 甲基去氫皮質醇或去氫皮質醇、或其之醫藥可接受前藥 或鹽; ⑻介於約16 wt%約21 wt0/〇間的具通式E106 P70 E106之聚氧乙烯_聚氧丙烯三嵌共聚合物; (in)滅菌水’適量緩衝以提供介於約5 5至約8.〇間之 pH ; (iv)多粒子之曱基脫氫皮質固醇、曱基去氫皮質醇或 去氫皮質醇; 140447.doc 200950787 (V)每g配方之微生物劑少於約5〇菌落形成單位 (cfu),及 (Vi) —個體之每公斤體重為少於約5内毒素單位。 30. 如請求項29項之方法,其中該甲基脫氫皮質固醇、甲基 去氫皮質醇或去氫皮質醇由組成物在至少3天的期間釋 放。 31. 如請求項29之方法,其中該甲基脫氫皮質固醇、甲基去 氫皮質醇或去氫皮質醇由組成物在至少5天的期間释 放。 32. 如請求項29之方法,其中該甲基脫氫皮質固醇、甲基去 氫皮質醇或去氫皮質醇由組成物在至少1〇天的期間釋 放。 33. 如請求項29之方法’其中該組成物投藥遍及圓窗。 34. 如請求項29之方法,其中該耳部疾病或症狀為梅尼爾 症、突生之感音神經性聽力喪失、噪音引起 I力喪 失、老化-有關聽力喪失、自體免疫耳疾病或耳。 140447.docFurthermore, it further comprises 11. The pharmaceutical composition according to any one of claims 1 to 10, a penetration enhancer. 12. If request item 1 to! A pharmaceutical composition according to any one of the preceding claims, a thickening agent. 13. A dye according to any one of claims 1 to 12. The pharmaceutical composition according to any one of claims 1 to 13, further comprising a drug delivery system selected from the group consisting of a needle and a syringe, a chestnut, and a microinjection The second set, and the original place to form a sponge substance or a combination thereof. The pharmaceutical composition according to any one of claims 1 to 14, wherein the methyl dehydrocortex (tetra), methyl dehydrocortisol or dehydrocortisol pharmaceutically acceptable salt is limited or absent System release, system toxicity, bad sag characteristics, or a combination thereof. 16. The pharmaceutical composition according to any one of the preceding claims, wherein the methyl dehydrocortex (tetra), methyl dehydrogenated furanol or dehydrogenated skin (tetra) is a free alcohol, a salt, or the like The form of the drug, or a combination thereof. 17. The pharmaceutical composition according to claim 16, wherein the methyl dehydrocorticosterol, decyl dehydrocortisol or dehydrocorticolysis is administered as a phosphate ester prodrug 140447.doc 200950787. 18. 19. 20. 21. 22. 23. 24. 25. The pharmaceutical composition according to Item 17, wherein the steroid is sulfhydryl corticosteroid or sulfhydryl dehydrocorticosterol acetate. The pharmaceutical composition according to any one of claims 1 to 18, which further comprises a mercapto dehydrocorticosteroid, a mercapto dehydrocortisol dehydrocortisol, or a medically acceptable salt thereof, The drug or a combination thereof is an immediate release agent. The pharmaceutical composition according to any one of items 1 to 19, wherein the methyl dehydrocorticosterol, sulfhydryl dehydrocorticolysis or dehydrocortisol comprises a plurality of particles. The pharmaceutical composition according to claim 20, wherein the sulfhydryl dehydrocorticosteroid, decyl dehydrocortisol or dehydrogenated skin (IV) is substantially in the form of micronized particles. The pharmaceutical composition according to any one of claims 1 to 2, wherein the methyl dehydrocorticosterol is in the form of a micro-mercapto dehydrocorticosteroid powder. The pharmaceutical composition according to any one of items 1 to 22, which further comprises an additional therapeutic agent. The pharmaceutical composition according to claim 23, wherein the additional therapeutic agent is - Na/K ATPase modulator, - chemotherapeutic agent, a collagen, a globulin, an interferon, an antimicrobial agent, an antibiotic, a Topical anesthetic, a platelet activating factor antagonist, an ear protectant, a nitric oxide synthase inhibitor, an anti-stun agent, a vasopressin antagonist, an antiviral agent, an anti-emetic agent, or the like combination. The pharmaceutical composition according to any one of the preceding claims, wherein the pH of the composition is from about 6.〇 to about 76. The pharmaceutical composition according to any one of claims 1 to 25, wherein the polyoxyethylene-polyoxypropylene triblock=polymer and a thickener having the formula E106 P70 E106 The ratio is from about 4 〇: 丨 to about 5 · 1. 27. The pharmaceutical composition of claim 26, wherein the thickening agent is carboxymethylcellulose, hydroxypropylcellulose or hydroxypropylmethylcellulose. 28. The pharmaceutical composition according to claim 1, wherein the ear disease or symptom is Meniere's disease, sudden sensorineural hearing loss, noise-induced hearing loss, aging-related hearing loss Autoimmune ear disease or tinnitus. 29. A method of treating an ear disease or condition comprising administering to a subject in need of an in-ear composition comprising a therapeutically effective amount of methyl dehydrocorticosterol, methyl dehydrocortisol or Dehydrocortisol, the composition comprising a low degradation product of substantially methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol, the composition further comprising two or two selected from the group consisting of: (1) thiol-dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol, or a pharmaceutically acceptable prodrug or salt thereof, between about 0.1% by weight and about 10% by weight; (8) between about 16 wt% of about 21 wt0 / day of polyoxyethylene _ polyoxypropylene tri-embedded copolymer of formula E106 P70 E106; (in) sterilized water 'moderate buffer to provide between about 5 5 to about 8. (iv) multiparticulate thiol dehydrocorticosterol, sulfhydryl dehydrocortisol or dehydrocortisol; 140447.doc 200950787 (V) less than about 5 colonies per microgram of microbial formulation Units (cfu), and (Vi) - the individual has less than about 5 endotoxin units per kilogram of body weight. 30. The method of claim 29, wherein the methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol is released from the composition for at least 3 days. 31. The method of claim 29, wherein the methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol is released from the composition for at least 5 days. 32. The method of claim 29, wherein the methyl dehydrocorticosterol, methyl dehydrocortisol or dehydrocortisol is released from the composition by at least one day. 33. The method of claim 29 wherein the composition is administered throughout a round window. 34. The method of claim 29, wherein the ear disease or symptom is Meniere, sudden sensorineural hearing loss, noise-induced I loss, aging-related hearing loss, autoimmune ear disease or ear. 140447.doc
TW98116106A 2008-05-14 2009-05-14 Controlled release corticosteroid compositions and methods for the treatment of otic disorders TWI382839B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12771308P 2008-05-14 2008-05-14
US8794008P 2008-08-11 2008-08-11
US9524808P 2008-09-08 2008-09-08

Publications (2)

Publication Number Publication Date
TW200950787A true TW200950787A (en) 2009-12-16
TWI382839B TWI382839B (en) 2013-01-21

Family

ID=44871489

Family Applications (1)

Application Number Title Priority Date Filing Date
TW98116106A TWI382839B (en) 2008-05-14 2009-05-14 Controlled release corticosteroid compositions and methods for the treatment of otic disorders

Country Status (2)

Country Link
CL (1) CL2009001178A1 (en)
TW (1) TWI382839B (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070178051A1 (en) * 2006-01-27 2007-08-02 Elan Pharma International, Ltd. Sterilized nanoparticulate glucocorticosteroid formulations

Also Published As

Publication number Publication date
CL2009001178A1 (en) 2010-06-04
TWI382839B (en) 2013-01-21

Similar Documents

Publication Publication Date Title
KR101477329B1 (en) Controlled release corticosteroid compositions and methods for the treatment of otic disorders
JP5903119B2 (en) Controlled release antimicrobial compositions and methods for the treatment of ear disorders
JP7023461B2 (en) TRKB or TRKC agonist compositions and methods for the treatment of ear diseases
JP2021510685A (en) Growth Factor Ear Formulation
EP3174525A1 (en) Otic formulations for the treatment of ceruminosis
KR20160047490A (en) Treatment of pediatric otic disorders
KR20170021353A (en) Sterilization of ciprofloxacin composition
GB2459910A (en) Sustained release corticosteroid compositions for treatment of otic disorders
WO2017100576A1 (en) Ciprofloxacin otic composition and kits and method for using same
TWI382839B (en) Controlled release corticosteroid compositions and methods for the treatment of otic disorders
GB2461961A (en) Sterile anti-apoptotic agent for treatment of ear diseases
TWI450732B (en) Controlled release antimicrobial compositions and methods for the treatment of otic disorders
GB2461962A (en) NMDA receptor antagonists for the treatment of ear disorders

Legal Events

Date Code Title Description
MM4A Annulment or lapse of patent due to non-payment of fees