TW201138831A - Modified granulocyte colony stimulating factor (G-CSF) - Google Patents

Modified granulocyte colony stimulating factor (G-CSF) Download PDF

Info

Publication number
TW201138831A
TW201138831A TW099133189A TW99133189A TW201138831A TW 201138831 A TW201138831 A TW 201138831A TW 099133189 A TW099133189 A TW 099133189A TW 99133189 A TW99133189 A TW 99133189A TW 201138831 A TW201138831 A TW 201138831A
Authority
TW
Taiwan
Prior art keywords
csf
pegylated
peg
protein
composition
Prior art date
Application number
TW099133189A
Other languages
Chinese (zh)
Inventor
Lihsyng S Lee
Abraham Abuchowski
Gerry Gitlin
Original Assignee
Prolong Pharmaceuticals Inc
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43826620&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=TW201138831(A) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Prolong Pharmaceuticals Inc filed Critical Prolong Pharmaceuticals Inc
Publication of TW201138831A publication Critical patent/TW201138831A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid

Landscapes

  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

This invention relates to novel protein conjugates, in particular, to novel pegylated proteins, and their methods of making and use. One aspect of the present invention relates to pegylated- G-CSF having unexpected efficacy and stability than current G-CSF formulations.

Description

201138831 六、發明說明: 【發明所屬之技術領域】 本發明係關於一種新穎之蛋白質接合物,特定言之一種 經聚乙二醇化之蛋白質,並關於其製造及使用方法。本發 明之一態樣係關於一種聚乙二醇化_G_CSF ,其具有出乎竟 • 料之效力及安定性。 〜 【先前技術】 許多類型之化學治療藥物的其中一最嚴重之潛在副作 用係使白血球數降低(嗜中性球減少症(neutr〇penia))。嗜 中I·生球減少症可使患者有受嚴重感染之風險,且可能中斷 化學療法治療。事實上,與低白血球數相關之併發症為進 行化學療法時劑量減少或延遲其進行t最常見原因(參見 Link,等人(2〇〇l)Cancer· ; 92:1354 1367 ; “爪⑽,等人 (2003)J Clin Oncol. 21:4524 4531 ;及―⑽,等人 (2002)Am J Med. 112:406-411) 〇 顆粒性細胞群落刺激因子(G_CSF)為發展具抗細菌性之 嗜中性顆粒細胞性白細胞(嗜中性白血球)的主要調節劑。 於1983年’自外植體組織中純化出小鼠形式之G-CSF ;且 於1985年,自無意地表現高濃度G-CSF之生長於培養基中 之癌症細胞株中純化出人類同等物(參見例如觀以,等人 (1985)PNAS USA 82 : 1526-30)。發現人類 G-CSF 為約 19kD之糖蛋白’且取決於碳水化合物組分,而呈不同酸 性。後來發現,生物活性視需要有碳水化合物組分之存 在於1984至1986年期間選殖出人類重&g_csf並鑑定出 150941.doc 201138831 其特徵’且於大腸桿菌(E. c〇n)細胞中表現,最終在罹患 由化學療法誘發之嗜中性球減少症的患者中達成測試該化 合物效應的人類臨床試驗。於1991年,美國FDA核准使用 於大腸桿菌中製得之重組體人類G_CSF(稱為惠爾血 (Filgrastim) ’商品名Neupogen®),且於1993年,歐洲核准 相關之自中國倉鼠卵巢細胞中表現之形式(商品名來格司 亭(lenograstim))。已發現,雖然已知存在多種變體,但其 核〜蛋白質包含174個胺基酸(參見例如Ngata,等人(1986)201138831 VI. INSTRUCTIONS OF THE INVENTION: FIELD OF THE INVENTION The present invention relates to a novel protein conjugate, in particular a PEGylated protein, and to methods of making and using same. One aspect of the present invention relates to a PEGylated _G_CSF which has an effect on the effectiveness and stability of the material. ~ [Prior Art] One of the most serious potential side effects of many types of chemotherapeutic drugs reduces the number of white blood cells (neutr〇penia). The hoarding of I. hypogonia may expose the patient to a serious infection and may interrupt chemotherapy. In fact, the complications associated with low white blood cell counts are the most common cause of dose reduction or delay in performing chemotherapy (see Link, et al. (2〇〇l) Cancer·; 92:1354 1367; “claw (10), Et al. (2003) J Clin Oncol. 21:4524 4531; and ―(10), et al. (2002) Am J Med. 112:406-411) 〇granular cell community stimulating factor (G_CSF) for development of antibacterial properties a major regulator of neutrophilic granulocytes (neutrophils). In 1983, 'G-CSF in mouse form was purified from explant tissues; and in 1985, unintentionally expressed high concentrations of G - Human equivalents of CSF grown in cancer cell lines grown in culture medium (see, eg, et al. (1985) PNAS USA 82: 1526-30). Human G-CSF was found to be a glycoprotein of about 19 kD' and Depending on the carbohydrate component, it is different in acidity. It was later found that the biological activity required the presence of a carbohydrate component from 1984 to 1986 to select human heavy & g_csf and identified 150941.doc 201138831. And expressed in E. c〇n cells, A human clinical trial to test the effects of this compound was finally achieved in patients suffering from chemotherapy-induced neutropenia. In 1991, the US FDA approved the use of recombinant human G_CSF produced in E. coli (called Hui Filgrastim 'trade name Neupogen®', and in 1993, Europe approved the relevant form of expression from Chinese hamster ovary cells (trade name lenograstim). It has been found that although there are many known Variant, but its core ~ protein contains 174 amino acids (see for example Ngata, et al. (1986)

Nature 319 : 415-18 ; Souza,等人(1986)Science 232 : 61- 5 ;美國專利案第4,999,291號)。 主張於1985年8月23曰申請之美國專利申請案07/768,959 優先權的授權予Amgen公司之美國專利案第4,81〇,643、 4,999,291、5,5 82,823及5,5 80,755號提供某些人類多效用 G-CSF分子及其製造方法。該等分子構成已核准之 Neupogen及Neulastin產品之基礎。於該等案例中,未討論 該分子之聚乙二醇化可能性。 近年來,已使用諸如聚乙二醇(「PEG」)之無抗原性之 水/谷性聚合物來共價修飾具治療及診斷重要性之多肽。 PEG為聚合物,其無毒、無免疫原性、高度水溶性,且很 容易自身體清除。PEG具有許多應用,且常用於食品、化 妝品、飲料、及處方藥中。FDA已核准在美國使用醫藥級 PEG ’且其係廣泛用作生物醫藥載劑,此係由於彼等之高 度生物相容性。PEG化可修飾生物醫藥之某些特性,而不 會改變其功能,藉此增強治療效力。 150941.doc 201138831 通常’藉由蛋白質中之反應性基團使聚乙二醇分子與蛋 白質鍵連。諸如彼等於離胺酸殘基上或於N端上之胺基適 用於該種附接。採用具有不同反應部份之經曱氧基化之 PEG(「mPEG」),使peg分子透過多肽中之胺基附接。該 等聚合物包括mPEG-琥珀醯亞胺基琥珀酸酯、mPEG-琥珀 酸亞胺基碳酸酯、mPEG-亞胺酯、及mPEG-氰尿醯氯。 可採用多種化學方法,使PEG透過位於聚合物鏈末端之 羥基與活性生物醫藥偶聯。例如,已報導使如下治療多肽 共價附接PEG可延長其活體内半衰期,及/或降低其免疫原 性及抗原性:諸如介白素(Knauf,M. J.等人,J. Biol.Nature 319: 415-18; Souza, et al. (1986) Science 232: 61-5; U.S. Patent No. 4,999,291). U.S. Patent Application Serial No. 07/768,959, filed on Aug. 23, 1985, to the U.S. Patent Nos. 4,81,643, 4,999,291, 5,5,82,823 and 5,5,80,755. Some human multi-purpose G-CSF molecules and their manufacturing methods. These molecules form the basis of approved Neupogen and Neulastin products. In these cases, the possibility of PEGylation of the molecule was not discussed. In recent years, antigen-free water/gluten polymers such as polyethylene glycol ("PEG") have been used to covalently modify polypeptides of therapeutic and diagnostic importance. PEG is a polymer which is non-toxic, non-immunogenic, highly water-soluble, and easily cleared by itself. PEG has many applications and is commonly used in foods, cosmetics, beverages, and prescription drugs. The FDA has approved the use of pharmaceutical grade PEG' in the United States and is widely used as a biopharmaceutical carrier due to their high biocompatibility. PEGylation can modify certain properties of biopharmaceuticals without altering their function, thereby enhancing therapeutic efficacy. 150941.doc 201138831 Typically polyethylene glycol molecules are bound to proteins by reactive groups in proteins. An amine group such as that which is equivalent to an amine acid residue or at the N-terminus is suitable for such attachment. The peg molecule is attached to the amine group in the polypeptide by a methoxylated PEG ("mPEG") having a different reaction moiety. Such polymers include mPEG-succinimide succinate, mPEG-succinic acid carbonate, mPEG-imidate, and mPEG-cyanuridine chloride. A variety of chemical methods can be employed to couple the PEG to the active biopharmaceutical through the hydroxyl group at the end of the polymer chain. For example, it has been reported that covalent attachment of PEG to a therapeutic polypeptide can extend its in vivo half-life and/or reduce its immunogenicity and antigenicity: such as interleukin (Knauf, M. J. et al., J. Biol.

Chem. 1988,263,15,064 ; Tsutsumi,Υ·等人,J. Controlled Release 1995, 33, 447)、干擾素(Kita,Y.等人,Drug Des. Delivery 1990,6,157)、過氧化氫酶(Abuchowski,A.等 人,J. Biol. Chem. 1977,252,3,582)、超氧化物歧化酶 (Beauchamp, C· 0·等人,Anal. Biochem. 1983,131, 25)、及腺苷脫胺酶(Chen, R.等人,Biochim. Biophy. Acta 1981, 660, 293)。 美國專利案第4,002,53 1號闡述使鏈結酸之peg附接至 轉--騰凝乳蛋白酶。酸基係經醯化反應隨機附接,導致在 - PEG與酶之間產生醯胺鏈結。類似地,於歐洲專利公開案 EP 0 154 3 16中闡述於中性PH下,還原性烷基化淋巴細胞 活素,其中說明該分子之十一個離胺酸殘基中約33〇/〇經修Chem. 1988, 263, 15, 064; Tsutsumi, Υ· et al., J. Controlled Release 1995, 33, 447), Interferon (Kita, Y. et al., Drug Des. Delivery 1990, 6, 157), Hydrogen Peroxide Enzymes (Abuchowski, A. et al., J. Biol. Chem. 1977, 252, 3, 582), superoxide dismutase (Beauchamp, C. 0 et al., Anal. Biochem. 1983, 131, 25), And adenosine deaminase (Chen, R. et al, Biochim. Biophy. Acta 1981, 660, 293). U.S. Pat. The acid group is randomly attached via a deuteration reaction, resulting in a guanamine linkage between the -PEG and the enzyme. Similarly, in the European Patent Publication EP 0 154 3 16, reductive alkylated lympholysin is described at neutral pH, which indicates about 13 〇/〇 of the eleven amino acid residues of the molecule. Repair

飾。歐洲公開案EP 0 335 423闡述利用peg衍生物使g-CSF 醯基化。 I50941.doc 201138831 爲了改善狀之安定性及生物可利用度、及確保活性,需 要進行位點特異性而不是隨機之聚乙二醇化。已在生長激 素釋放因子之強力類似物的N端、側鏈及c-末端處,透過 固相合成法進行位點特異性聚乙二醇化(Felix,A. M等 人 ’ Int. J. Peptide Protein Res· 1995, 46,253)。 授權予Siegried有限公司之PCT WO 2006/094530闡述某 些經一聚乙二醇化之蛋白質接合物及其製備方法。提供經 —聚乙二醇化之G-CSF實例,其中該等產物係於Ν端及 乙>^17處或於>1端及1^535處經二聚乙二醇化。 由於惠爾血在活體内很容易降解,因此必須在進行化學 療法過程中重複每日注射。因此,需要增強惠爾血之藥物 動力學性質。已利用經醛活化之PEG,使〇-(:卯之Ν端位 點特異性聚乙二醇化,發展出PEG_惠爾血(參見pCT公開 案第WO 96/1 1953號,及美國專利案第5,824,784及 7’〇90,835號)。於2002年,美國FDA已核准該化合物,其 商品名為Neulasta®。 雖然已透過胺與N端鏈結,採用位點特異性附接,發展 出PEG-惠爾血,但是該等反應所需要之反應時間長,且嚴 重依賴於酸性pH。WO 96/11953闡述,反應條件之設計在 於允許聚合物選擇性附接至蛋白質之^^端。該特性顯示, 於所述條件下(其中pH調整至4.〇),组合物主要包含經單聚 乙二醇化之產物,其中PEG係附接至N端、Lys35或 Lys4i。利用該等方法製得之經聚乙二醇化之g_csf組合物 已於例如 Cindric,等人(2〇〇7)J pham Bi〇med Analys· 150941.doc 201138831 44 : 3 88-3 95中得到證實。 已提出一些可提供安定G-CSF分子之替代性策略。使 PEG鏈結至半胱胺酸殘基已在靶向方面提供某些改善。具 酼基反應性之PEG(包括PEG-馬來醯亞胺)已於G-CSF之游 離半胱胺酸殘基處鏈結至G-CSF。Veronese,等人(2007) Bioconjugate Chem. 18: 1824-1830 闡述,於 Cysl7 處使 G- CSF聚乙二醇化,已顯示其增加凝集,雖然凝集物並非經 共價凝集。類似地,Hao,等人(2006)Biodrugs 20 : 357-363闡述使PEG-馬來醯亞胺與Cysl7接合,已顯示其增加該 分子之半衰期。 位點特異性突變為另一種已用於製備供位點特異性聚合 物附接之多肽之方法。例如’美國專利案第6,646,11〇號闡 述一種多肽接合物,其顯示出G-CSF活性,且具有含有供 插入PEG或养糖部份的附接基團的胺基酸殘基。該等胺基 酸殘基可包括離胺酸、麵胺酸、半胱胺酸或天冬胺酸。 就該點而論,仍然需要一種經聚乙二醇化之組合物、及/ 或其製備方法,其產生一種可預測且一致之產物,沒有複 雜之N端靶向問題’並保留原始分子之活性。 【發明内容】 本發明係基於製備一種具出乎意料之活性的新穎形式之 經早聚乙二醇化之蛋白質,例如顆粒性細胞群落刺激因子 (G-CSF」)。於藉由還原性烷基化法製備與pEG鏈結之 蛋白質的方法中,使用二甲基亞礙(「DMS〇」)會使得 PEG接合僅限於N端或鄰近位點。該方法大體上可使得 150941.doc 201138831 PEG鏈結至最接近N端之離胺酸殘基。特定言之,本發明 提供一種G-CSF分子,其包括與Lysl6(若包含末端之甲硫 胺酸,則為Lys 17)鏈結之PEG分子。本發明亦提供一種於 DMSO存在下,使醛-反應性PEG與蛋白質鏈結之方法。 於某些實施例中,本發明係關於一種組合物,其包含至 少一類G-CSF蛋白質,其中各G-CSF分子係與至少一個聚 乙二醇分子共價鏈結,且至少30%之該組合物並非於N端 處經聚乙二醇化。於一項特定實施例中,各G-CSF分子係 與單個聚乙二醇分子共價鏈結(「經單聚乙二醇化之G-CSF」),且至少30%之該組合物並非於N端處經聚乙二醇 化。於另一項實施例中,該組合物包括至少80%經單聚乙 二醇化之G-CSF,其中至少30%之該組合物並非於N端處經 聚乙二醇化。於另一項實施例中,該組合物包括至少 85%、至少90%或至少95%經單聚乙二醇化之G-CSF分子, 其中至少30%之該組合物並非於N端處經聚乙二醇化。 於該組合物之特定實施例中,各G-CSF分子係透過特定 離胺酸殘基與至少一個聚乙二醇分子共價鏈結。於某些實 施例中,本發明包括一種組合物,其具有至少一個G-CSF 分子,經由G-CSF蛋白質之胺基端,利用胺鏈結與至少一 個聚乙二醇分子共價鏈結。於其他實施例中,本發明包括 一種組合物,其具有至少一個G-CSF分子,透過Lysl6(若 計算N端之甲硫胺酸殘基,則為Lysl 7)與至少一個聚乙二 醇分子共價鏈結。 於組合物之一些實施例中,N端處經聚乙二醇化之分子 150941.doc 201138831 對第二群類(尤其係於Lys 1 6處經聚乙二醇化之第二群類)的 比例範圍可為小於約1比約1 00、約10比約90、約20比約 80、約30比約70、約40比約60、約50比約50、約60比約 40、約70比約30,其中小於約1包括利用相關技術中已知 之標準方法不可測得之數量。 於某些實施例中’提供一種實質上均質之組合物,其包 括經單聚乙二醇化之蛋白質,其中至少30%之蛋白質分子 並非於N端處經聚乙二醇化。於某些特定實施例中,該組 合物包含至少30%經單聚乙二醇化之蛋白質,其係於距離 N端100個胺基酸以内之離胺酸殘基處經聚乙二醇化。於特 定實施例中,該離胺酸係距離N端80個以内、70個以内、 60個以内、50個以内、40個以内、30個以内、20個以内、 19個以内、18個以内、17個以内、16個以内、15個以内、 14個以内、13個以内、12個以内、1丨個以内、1〇個以内、 9個以内、8個以内、7個以内、6個以内、5個以内、4個以 内、3個以内或2個以内之胺基酸殘基。 於某些實施例中,本發明係關於一種醫藥調配物,其包 含至少一類G-CSF蛋白質,其中各G_CSF分子係與至少— 個聚乙二醇分子共價鏈結,且其視需要含於醫藥上可接受 之載劑中。於某些實施例中,該載劑實質上不含蛋白質。 於某些實施例中’該醫藥調配物包含至少一類g_csf蛋白 夤,其中各G-CSF分子係與至少一個聚乙二醇分子共價鏈 結,且至少30%之該組合物並非於N端處經聚乙二醇化。 於一項特定實施例中,各G_CSF分子係與單個聚乙二醇分 150941.doc 201138831 子共價鏈結(「經單聚乙二醇化之G-CSF」),且至少30〇/〇 之該組合物並非於N端處經聚乙二醇化。於另一項實施例 中’調配物包含至少80%經單聚乙二醇化之G-CSF,其中 至少3 0%之該組合物並非於n端處經聚乙二醇化。於另一 項實施例中’調配物包含至少85%、至少9〇%或至少95〇/〇 經單聚乙二醇化之G-CSF分子,其中至少30%之該組合物 並非於N端處經聚乙二醇化。 於醫藥調配物之特定實施例中,各G_CSF分子係透過特 定離胺酸殘基’與至少一個聚乙二醇分子共價鏈結。於某 些貫施例中’調配物包含至少一個G-CSF分子,其經由G-CSF蛋白質胺基端,利用胺鏈結與至少一個聚乙二醇分子 共價鏈結。於其他實施例中’調配物包含至少一個G_csf 分子’透過Lys 1 6(若計算N端之甲硫胺酸殘基,則為 Lysl7)與至少一個聚乙二醇分子共價鏈結。 於調配物之一些實施例中,於1^端處經聚乙二醇化之分 子對第二群類(尤其於Lys丨6處經聚乙二醇化之第二群類)之 比例可為小於約1比約100、約10比約9〇、約2〇比約8〇、約 30比約70、約40比約60、約50比約50、約60比約40、約7〇 比、·、勺3 0,其中小於約【包括利用相關技術中已知之標準方 法不可測得之數量。 希望本發明之G-CSF分子及組合物於標準貯藏條件下顯 示出延長的安定性,亦即於標準溫度(例如約25。(:)下可貯 藏至少三個月。於某些實施例中,該不含蛋白質之載劑為 不含血清、不含白蛋白或不含人類血清白蛋白(「不含血 150941.doc -10- 201138831 清、不含白蛋白或不含人類血清白蛋白」(「hsa_ free」))^於某些實施例中,醫藥調配物可貯存更長時 間,且如文中所述之方法及/或於相關技術中已知之方法 測定,G-CSF沒有實質上及/或可測得之降解。於某些實施 例中,於約-20°C或下貯藏至少15個月之後測定,本發 月之4藥s周配物具安定性(亦即不顯示出可測得之降解及/ 或不顯示出實質上之降解)。於其他實施例中,於約25。〇 或約37 C下貯藏至少1G個月之後測定,本發明之醫藥調配 物具安定性(亦即不顯示出可測得之降解及/或不顯示出實 質上之降解)。可藉由相關技術中已知之任一方法測定本 龟明醫藥調配物之安定性β於某些實施例中,本發明醫藥 調配物文疋性之測定方法為:採用二喹啉甲酸(「BCa」) 蛋白遂分析法,監測蛋白質濃度隨時間之變化。於其他實 包例中本發明醫藥調配物安定性之測定方法為:如SDS PAGE分析法所測,指示蛋白質降解(亦即G-CSF接合物降 解)隨時間之變化。於某些其他實施例中,安定性之測定 方法為.藉由HPLC分析產物之降解。於其他實施例中, 本發明醫藥調配物安定性之測定方法為:監測該調配物之 $ 1±隨時間之變化’其中該活性係藉由測定該調配物之活 ^ (例如G-CSF活性)技術中已知之活體外或活體内方法測 又。於根據該實施例之一項特定實例中,包含複數個G_ 接5物之本發明醫藥調配物之活性的評定方法為:利 CSF依賴性鼠類32D細胞株’評定該醫藥調配物於活 卜生物松尺中之能力。於其他實施例中,調配物活性之 150941.doc 201138831 檢測方法為.於活體内測定其改變實驗動物(諸如倉鼠)之 白血球細胞數之能力。 於某些實施例中,提供一種醫藥調配物,其包含經單聚 乙二醇化之蛋白質,其中至少30%之蛋白質分子並非於N 端處經聚乙二醇化。於某些特定實施例中,調配物包含至 少30%經單聚乙二醇化之蛋白質,其於距離>^端1〇〇個胺基 酸以内之離胺酸殘基處經聚乙二醇化。於特定實施例中, 離胺酸為距離N端80個以内、7〇個以内、6〇個以内、5〇個 以内、40個以内、30個以内、2〇個以内、19個以内、以個 以内、17個以内、16個以内、15個以内、⑷固以内、⑽ 以内、12個以内、η個以内、1〇個以内、9個心、請以 内、7個以内、6個以内、5個以内、4個以内、3個以内或2 個以内之胺基酸殘基。 於本發明之另一態樣中,提供一種增加接受者之白血球 數之方法’其包括對有需要之接受者投與本發明醫藥調配 物。於某些實施例中,接受者為人類。於某些實施例中, 接受者羅患嗜中性球減少症或處於罹患風險中。於某些其 他實施例中,接受者正接受會使其白血球數減少之藥劑之 治療。於某些實施例中,接受者之内源性度已經 降低。於某些其他實施例中,接受者正接受放射治療。接 爻者可能正罹患肺癌、淋巴瘤、乳癌、骨髓移植、睾丸 癌、與AIDS相關之惡性腫瘤、骨髓造血不良症、急性白 血病、先天性及週期性嗜中性球減少症或再生障礙性貧企 (參見 M〇rtsyn,等人(1998)Filgrastim(r_metHuG_csF,& 150941.doc 201138831Decoration. European Publication EP 0 335 423 describes the thiolation of g-CSF using peg derivatives. I50941.doc 201138831 In order to improve the stability and bioavailability of the form and to ensure activity, site-specific rather than random PEGylation is required. Site-specific PEGylation by solid phase synthesis at the N-terminus, side chain and c-terminus of potent analogs of growth hormone releasing factor (Felix, A. M et al' Int. J. Peptide Protein Res· 1995, 46, 253). PCT WO 2006/094530, issued to Siegried, Inc., describes certain PEGylated protein conjugates and methods for their preparation. An example of a PEGylated G-CSF is provided, wherein the products are dipegylated at the terminus and B>^17 or at >1 and 1^535. Since Whirlpool blood is easily degraded in vivo, daily injections must be repeated during chemotherapy. Therefore, there is a need to enhance the pharmacokinetic properties of Whirlpool blood. The aldehyde-activated PEG has been utilized to specifically PEGylate the 〇-(:卯 位 位 , , , ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( ( Nos. 5,824,784 and 7'〇90,835. In 2002, the FDA approved the compound under the trade name Neulasta®. Although PEG- has been developed through site-specific attachment through amine and N-terminal linkages. Whirlpool blood, but the reaction time required for such reactions is long and relies heavily on acidic pH. WO 96/11953 states that the reaction conditions are designed to allow selective attachment of the polymer to the ends of the protein. Under the conditions described (wherein the pH is adjusted to 4. 〇), the composition consists essentially of a mono-pegylated product wherein the PEG is attached to the N-terminus, Lys35 or Lys4i. The PEGylated g_csf composition has been confirmed, for example, in Cindric, et al. (2〇〇7) J pham Bi〇med Analys 150941.doc 201138831 44 : 3 88-3 95. Some proposals have been made to provide stability G An alternative strategy for CSF molecules. Linking PEG to cysteine residues Some improvement in targeting is provided. thiol-reactive PEG (including PEG-maleimide) has been linked to G-CSF at the free cysteine residue of G-CSF. Veronese, et al. Human (2007) Bioconjugate Chem. 18: 1824-1830 states that G-CSF is PEGylated at Cysl7, which has been shown to increase agglutination, although agglutination is not covalently coagulated. Similarly, Hao, et al. (2006) Biodrugs 20: 357-363 illustrates the binding of PEG-maleimine to Cysl7, which has been shown to increase the half-life of the molecule. Site-specific mutations have been used to prepare site-specific polymer attachments. A method of polypeptides, for example, in U.S. Patent No. 6,646,11, the disclosure of which is incorporated herein incorporated by reference in its entirety in its entirety in its entirety in the the the the the the the the the the the the the the the the the Residues. The amino acid residues may include a per-amine acid, a face acid, a cysteine or an aspartic acid. As such, there is still a need for a PEGylated composition, and / Or a method for its preparation that produces a predictable and consistent product without complex N-terminal targets The problem 'and retains the activity of the original molecule. The present invention is based on the preparation of an unexpectedly active novel form of an early PEGylated protein, such as a particulate cell community stimulating factor (G-CSF). In the method of preparing a protein linked to pEG by a reductive alkylation method, the use of dimethyl occlusion ("DMS") limits the PEG junction to the N-terminus or adjacent sites. This method generally allows 150941.doc 201138831 PEG to be linked to the amino acid residue closest to the N-terminus. In particular, the present invention provides a G-CSF molecule comprising a PEG molecule linked to Lysl6 (or Lys 17 if a terminal methionine is included). The invention also provides a method of linking an aldehyde-reactive PEG to a protein in the presence of DMSO. In certain embodiments, the present invention relates to a composition comprising at least one class of G-CSF proteins, wherein each G-CSF molecule is covalently linked to at least one polyethylene glycol molecule, and at least 30% of the The composition is not PEGylated at the N-terminus. In a specific embodiment, each G-CSF molecule is covalently linked to a single polyethylene glycol molecule ("mono-PEGylated G-CSF"), and at least 30% of the composition is not The N-terminus is PEGylated. In another embodiment, the composition comprises at least 80% mono-glycolated G-CSF, wherein at least 30% of the composition is not PEGylated at the N-terminus. In another embodiment, the composition comprises at least 85%, at least 90%, or at least 95% mono-PEGylated G-CSF molecules, wherein at least 30% of the composition is not polymerized at the N-terminus Ethylene glycolation. In a particular embodiment of the composition, each G-CSF molecule is covalently linked to at least one polyethylene glycol molecule via a specific lysine residue. In certain embodiments, the invention includes a composition having at least one G-CSF molecule covalently linked to at least one polyethylene glycol molecule via an amine linkage via an amine end of the G-CSF protein. In other embodiments, the invention includes a composition having at least one G-CSF molecule that transmits Lysl6 (Lysl 7 if the N-terminal methionine residue is calculated) and at least one polyethylene glycol molecule Covalent chain. In some embodiments of the composition, the ratio of the ratio of the PEGylated molecule at the N-terminus 150941.doc 201138831 to the second group (especially the second group PEGylated at Lys 16) Can be less than about 1 to about 100, about 10 to about 90, about 20 to about 80, about 30 to about 70, about 40 to about 60, about 50 to about 50, about 60 to about 40, about 70 to about 30, wherein less than about 1 comprises an amount that is not measurable using standard methods known in the related art. In certain embodiments, a substantially homogeneous composition is provided comprising a mono-pegylated protein wherein at least 30% of the protein molecules are not PEGylated at the N-terminus. In certain specific embodiments, the composition comprises at least 30% mono-pegylated protein PEGylated at a distance from an amine acid residue within 100 amino acids from the N-terminus. In a specific embodiment, the lysine is 80 or less, 70 or less, 60 or less, 50 or less, 40 or less, 30 or less, 20 or less, 19 or less, and 18 or less. Within 17, within 16, within 15, within 14, within 13, within 12, within 1丨, within 1〇, within 9, within 8, within 7, within 6, within Amino acid residues within 5, within 4, within 3 or within 2 amino acids. In certain embodiments, the present invention relates to a pharmaceutical formulation comprising at least one class of G-CSF proteins, wherein each G_CSF molecule is covalently linked to at least one polyethylene glycol molecule, and is optionally included A pharmaceutically acceptable carrier. In certain embodiments, the carrier is substantially free of protein. In certain embodiments, the pharmaceutical formulation comprises at least one class of g_csf peptones, wherein each G-CSF molecule is covalently linked to at least one polyethylene glycol molecule, and at least 30% of the composition is not N-terminally It is PEGylated. In a specific embodiment, each G_CSF molecule is covalently linked to a single polyethylene glycol 150941.doc 201138831 ("mono-PEGylated G-CSF"), and at least 30 Å/〇 The composition is not PEGylated at the N-terminus. In another embodiment, the formulation comprises at least 80% monoPEGylated G-CSF, wherein at least 30% of the composition is not PEGylated at the n-terminus. In another embodiment, the formulation comprises at least 85%, at least 9%, or at least 95 Å per gram of mono-pegylated G-CSF molecules, wherein at least 30% of the composition is not at the N-terminus It is pegylated. In a particular embodiment of the pharmaceutical formulation, each G_CSF molecule is covalently linked to at least one polyethylene glycol molecule via a particular lysine residue'. In some embodiments, the formulation comprises at least one G-CSF molecule that is covalently linked to at least one polyethylene glycol molecule via an amine chain via the amine end of the G-CSF protein. In other embodiments, the formulation comprises at least one G_csf molecule that is covalently linked to at least one polyethylene glycol molecule through Lys 16 (or Lysl7 if the N-terminal methionine residue is calculated). In some embodiments of the formulation, the ratio of the PEGylated molecule at the end of the second group to the second group (especially the second group PEGylated at Lys丨6) may be less than about 1 to about 100, about 10 to about 9 〇, about 2 to about 8 〇, about 30 to about 70, about 40 to about 60, about 50 to about 50, about 60 to about 40, about 7 to 、, , spoon 30, wherein less than about [includes quantities that are not measurable using standard methods known in the related art. It is contemplated that the G-CSF molecules and compositions of the present invention exhibit extended stability under standard storage conditions, i.e., can be stored for at least three months at a standard temperature (e.g., about 25. (:). In some embodiments) The protein-free carrier is serum-free, albumin-free or free of human serum albumin ("blood free 150941.doc -10- 201138831 clear, albumin-free or human serum albumin free" ("hsa_free")) In certain embodiments, the pharmaceutical formulation can be stored for a longer period of time, and as determined by methods described herein and/or methods known in the related art, G-CSF is not substantially / or measurable degradation. In some embodiments, after 4 months of storage at or below -20 ° C, the formulation of the 4th s of the month is stable (ie, does not show Degradable and/or does not exhibit substantial degradation. In other embodiments, the pharmaceutical formulation of the present invention has stability after storage for at least 1 G month at about 25 Torr or about 37 C. (ie does not show measurable degradation and/or does not show substantial decline The stability of the present veterinary drug formulation can be determined by any method known in the related art. In some embodiments, the method for determining the medicinal properties of the pharmaceutical formulation of the present invention is: using quinolinic acid ( "BCa") peptone assay to monitor changes in protein concentration over time. In other practical examples, the stability of the pharmaceutical formulations of the present invention is determined by: SDS PAGE analysis, indicating protein degradation (ie, G) - CSF conjugate degradation) as a function of time. In certain other embodiments, the stability is determined by HPLC analysis of the degradation of the product. In other embodiments, the method of determining the stability of the pharmaceutical formulation of the invention To: monitor the change in the formulation for $1 ± over time 'where the activity is determined by an in vitro or in vivo method known in the art for determining the activity of the formulation (eg, G-CSF activity). In a specific example of this embodiment, the method for assessing the activity of a pharmaceutical formulation of the invention comprising a plurality of G_5 substances is: a CSF-dependent murine 32D cell line 'Assessing the pharmaceutical formulation to live In other embodiments, the activity of the formulation is 150941.doc 201138831 is determined by measuring its ability to alter the number of white blood cells in an experimental animal, such as a hamster, in vivo. Therein is provided a pharmaceutical formulation comprising a mono-pegylated protein, wherein at least 30% of the protein molecules are not PEGylated at the N-terminus. In certain particular embodiments, the formulation comprises at least 30 % PEGylated protein which is PEGylated at a distance from the amino acid at the end of the amino acid. In a particular embodiment, the lysine is a distance N 80 or less, 7〇 or less, 6〇 or less, 5〇 or less, 40 or less, 30 or less, 2〇 or less, 19 or less, Within, Less than 17, Within 16, 15 Within, (4) within, (10), within 12, within η, within 1〇, 9 hearts, within, within 7, within 6, within 5, within 4, within 3 or 2 amino acid residues. In another aspect of the invention, a method of increasing the number of white blood cells of a recipient is provided which comprises administering to a recipient in need thereof a pharmaceutical formulation of the invention. In certain embodiments, the recipient is a human. In certain embodiments, the recipient suffers from or is at risk of developing neutropenia. In some other embodiments, the recipient is receiving treatment for an agent that reduces the number of white blood cells. In some embodiments, the endogenous degree of the recipient has decreased. In certain other embodiments, the recipient is receiving radiation therapy. The recipient may be suffering from lung cancer, lymphoma, breast cancer, bone marrow transplantation, testicular cancer, AIDS-related malignancies, bone marrow hematopoietic disorder, acute leukemia, congenital and periodic neutropenia or aplastic deprivation Enterprise (see M〇rtsyn, et al. (1998) Filgrastim (r_metHuG_csF, & 150941.doc 201138831

Clinical Practice,第二版,Marcel Dekker 公司,New York, NY)。於某些實施例中,對處於感染風險中之患者 投與該調配物。 於某些實施例中’於化學療法過程中提供呈單一劑量之 調配物。於一些實施例中,於化學療法過程中提供呈多劑 量之調配物。於某些實施例中,調配物之投與頻率為每日 一次、每週一次、每兩週一次或每月一次。調配物可於一 劑化學療法二十四小時内投與。於某些實施例中,調配物 係於接受一劑化學療法之前至少14天投與。 於某些實施例中,調配物係呈注射液投與。於一些實施 例中,調配物適於經皮下投與。於其他實施例中,調配2 適於經靜脈内投與。調配物亦可呈口服形式提供。 於其他實施财,患者為至少約每兩 -週一次、或至少約每月一次接受一 每週一次接受一劑◊於其他實 週一次、至少約每三週一次、 劑。. w κ力一態稞係關於—種製備蛋白質接合物之太 本發明之另一態樣係關於一Clinical Practice, Second Edition, Marcel Dekker, New York, NY). In certain embodiments, the formulation is administered to a patient at risk of infection. In certain embodiments, a single dose of formulation is provided during chemotherapy. In some embodiments, a multi-dose formulation is provided during the chemotherapy process. In certain embodiments, the formulation is administered once daily, once a week, once every two weeks, or once a month. The formulation can be administered within 24 hours of a single dose of chemotherapy. In certain embodiments, the formulation is administered at least 14 days prior to receiving a dose of chemotherapy. In certain embodiments, the formulation is administered as an injection. In some embodiments, the formulation is suitable for subcutaneous administration. In other embodiments, Formulation 2 is suitable for intravenous administration. Formulations may also be provided orally. In other implementations, the patient receives a dose of at least once every two weeks, or at least about once a month, once a week, at least once every three weeks, at least once every three weeks. w κ 力 稞 关于 关于 关于 关于 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备 制备

’使蛋白質與該活化之 方法可亦包括去除實質上所有未鏈結 心得°亥蛋白質接合物。於某些實施命 物為: ,令蛋白質與活化之聚乙二醇-醛化 兹活化之水溶性聚合物共價鏈結。該 f上所有未鏈結之水溶性聚合物,以 。於某些實施例中,聚乙二醇-醛化The method of making the protein and the activation may also include removing substantially all of the unlinked protein conjugate. In some implementations, the protein is covalently linked to an activated polyethylene glycol-formaldehyde-activated water-soluble polymer. All unlinked water-soluble polymers on the f. In certain embodiments, polyethylene glycol-hydroformylation

。於某些實施例中, 150941.doc •13- 201138831 PEG為至少10 kD。於某些其他實施例中,PEG為至少20 kD。於特定實施例中,PEG為3〇 kD或4〇 kD。可利用任一 連接子’包括具1至3〇、1至20、1至15、1至10、9、8、 7 6、5、4、3、2、或1個碳原子之烷基鏈結,使PEG與 蛋白質鏈結。 本發明方法可採用於高於其他相關技術中已知方法之pH 下進行的反應’諸如實質上於中性pH(約pH 7.0)下(例如利 用PEG醛化物使蛋白質聚乙二醇化,參見例如美國專利案 第5,824,784號)。對相關技術中已知之方法所作之該等修 改可提供於成本、製造效率、及/或製程之簡易程度方面 之製造優勢。 於某些實施例中,反應係於pH為3至8下進行。於某些其 他實施例中,反應係於pH為約4至約7、或約4.5至約6、或 約5下進行。於某些其他實施例中,反應係於pH為約中性 下進行。於某些其他實施例中,反應係於微酸性之pH下 (諸如例如約5或約6)進行。 於另一項實施例中,反應緩衝液包括蛋白質對活化之水 溶性聚合物之莫耳比為約1比約3至約1比約6〇。於其他實 施例中,反應緩衝液包括蛋白質對活性水溶性聚合物之莫 耳比為約1比約4、約1比約5、約1比約6、約1比約7、約1 比約8、約1比約9、約1比約1 〇、約1比約15、約1比約2〇、 約1比約25、約1比約30、約1比約35、約1比約40、約1比 約45、約1比約50、約1比約55、約1比約60。於某些實施 例中,反應緩衝液包括蛋白質對活化之水溶性聚合物之莫 150941.doc •14· 201138831 耳比為約1比約7。於另一項實施你丨中,可茲 貝她例甲可碏由相關技術中 已知之方法(諸如例如透析術或層析術),去除實質上所有 未反應之水溶性聚合物。 【實施方式】 彼等熟習此項技術者很容易自下列詳細闡述部份明瞭本 發明之其他優點,其中僅說明並闡述本發明之某些實施 例。將會明瞭,於不脫離本發明下,本發明可具有其他及 不同之實施例,且對其中之若干細節可進行許多態樣之例 打修改。本發明可在未進行一些或所有該等特定細節下進 行。因此,該闡述部份應視作具闡述性質,而非限制。 本發明係基於製備新穎形式之經單聚乙二醇化之蛋白 質,例如顆粒性細胞群落刺激因子(「G_CSF」),其具有 出人意料之活性。於藉由還原烷基化法製備與pEG鏈結之 蛋白質之本發明方法中,使用二甲基亞砜(「dms〇」)會 驅使PEG結合僅限於]^端或其鄰近位點。特定言之,本發 明提供一種G-CSF分子,其包括與Lysl6(若包含末端之甲 硫胺酸,則為LySl7)鏈結之PEG分子。本發明亦提供一種 於DMSO存在下,使醛反應性pEG與蛋白質鏈結之方法。 不同之修改仍可保留原始分子之活性,且改善分子之醫藥 性質。 定義 如文中所用術6吾「N端」、「胺基端」或類似術語當用 於闡述使蛋白質與另一分子共價鏈結之内容中時,其係指 經由蛋白質胺基端之α_胺基共價鏈結。 150941.doc 15 201138831. In certain embodiments, 150941.doc • 13-201138831 PEG is at least 10 kD. In certain other embodiments, the PEG is at least 20 kD. In a particular embodiment, the PEG is 3 〇 kD or 4 〇 kD. Any linker can be used to include an alkyl chain having 1 to 3 Å, 1 to 20, 1 to 15, 1 to 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 carbon atom. The knot links the PEG to the protein. The process of the invention may be employed at reactions above the pH of methods known in the related art, such as at substantially neutral pH (about pH 7.0) (e.g., PEGylation of proteins using PEG aldehydes, see for example U.S. Patent No. 5,824,784). Such modifications to the methods known in the related art can provide manufacturing advantages in terms of cost, manufacturing efficiency, and/or ease of process. In certain embodiments, the reaction is carried out at a pH of from 3 to 8. In certain other embodiments, the reaction is carried out at a pH of from about 4 to about 7, or from about 4.5 to about 6, or about 5. In certain other embodiments, the reaction is carried out at a pH of about neutral. In certain other embodiments, the reaction is carried out at a slightly acidic pH (such as, for example, about 5 or about 6). In another embodiment, the reaction buffer comprises a molar ratio of protein to activated water soluble polymer of from about 1 to about 3 to about 1 to about 6 Torr. In other embodiments, the reaction buffer comprises a molar ratio of protein to active water soluble polymer of from about 1 to about 4, from about 1 to about 5, from about 1 to about 6, from about 1 to about 7, and about 1 to about 8. about 1 to about 9, about 1 to about 1 〇, about 1 to about 15, about 1 to about 2, about 1 to about 25, about 1 to about 30, about 1 to about 35, about 1 to about. 40, about 1 to about 45, about 1 to about 50, about 1 to about 55, and about 1 to about 60. In certain embodiments, the reaction buffer comprises a protein to activated water soluble polymer of from 150941.doc •14·201138831 in an ear ratio of from about 1 to about 7. In another implementation, Cobbe may remove substantially all of the unreacted water soluble polymer by methods known in the art, such as, for example, dialysis or chromatography. [Embodiment] Other advantages of the present invention will become apparent from the following detailed description of the invention. It will be apparent that the invention may be embodied in other and different embodiments, and many of the details may be modified. The invention may be practiced without some or all of the specific details. Therefore, the elaboration should be considered as illustrative rather than limiting. The present invention is based on the preparation of novel forms of mono-pegylated proteins, such as particulate cell community stimulating factor ("G_CSF"), which have unexpected activity. In the process of the invention for preparing a pEG-linked protein by reductive alkylation, the use of dimethyl sulfoxide ("dms") drives PEG binding to the end or its adjacent sites. In particular, the present invention provides a G-CSF molecule comprising a PEG molecule linked to Lysl6 (LySl7 if a terminal thioacetate is included). The invention also provides a method of linking an aldehyde-reactive pEG to a protein in the presence of DMSO. Different modifications can retain the activity of the original molecule and improve the medical properties of the molecule. Definitions As used herein, when the term "N-terminal", "amine-based" or the like is used to describe the content of a protein that is covalently linked to another molecule, it refers to α_ via the amino terminus of the protein. Amine covalent linkage. 150941.doc 15 201138831

如文中所EB /夕 用術語「野生型」或「天然型」係指蛋白質 或夕肽呈其操作形式或功能形式,通常在身體内發現天然 之功能形式。#楚^ 、 4術s吾亦指呈未經人工修飾或改變之蛋白 質形式。該箸;} 寻術S吾亦可關於重組蛋白質。因此,該等術語 可指相對於該蛋白質之核酸及/或胺基酸序列之起源動物 中所產生之在"/~y frfr 蛋白質,具已改變之醣基化形式(包括缺少醣 基化)的蛋白質。 如文中所用’除非另外指*,否則術語「G-CSF」或顆 粒丨生細胞群落刺激因子係指具有如SEQ ID NO : 1(圖1)所 不之胺基酸序列的蛋白質,或具有實質上同質之胺基酸序 列且具有與刺激白血球產生相關的生物性質之蛋白質。如 文中所用,該等術語包括該等經有意修飾之蛋白質(例 如、’’至4日疋位置突變(site directed mutagenesis)或隨機突 變(accidentally through mutations));從而使該等蛋白質相 對於天然G-CSF具有胺基酸殘基之添加、缺失、或取代。 該等術語包括天然及經重組產生之人類G_CSF。G-CSF係 指天然存在之蛋白質或重組蛋白質,通常為人類,其係獲 自任一 S知來源,诸如組織、蛋白質合成、天然或重組細 胞之細胞培養物。 於本文中,胺基酸序列「實質上相同」之定義為:如根 據Pearson及Lipman,Proc. Natl. Acad. Sci. USA 85, 2444-2448 (1988)之FASTA搜索方法所測,一序列與另一胺基酸序列 有至少70% ’通常至少約80%,且更通常至少約90%相 同0 150941.doc • 16 - 201138831 如文中所用,術語「接合物」係指蛋白質或多肽係一種 與共仏鍵附接之一個或多個其他化學基團相互作用而發揮 作用之蛋白質或多肽或其集群。 G-CSF分子 可與根據本發明聚合物單元接合之蛋白質包括未經突變 及經突變之昼"白暂,士私J# y 贫曰貝邊如但不限於生長因子、抗體、激 素特定5之具治療活性之蛋白質,諸如但不限於促红血 球生成素、干擾素α、干擾素P、干擾素丫、複合干擾素、 G CSF GM-CSF、血紅素、介白素(諸如介白素_2及介白 素-6)、腫瘤壞死因子、多種細胞激素、生長因子(諸如人 類生長因子及表皮生長因子)' 免疫球蛋白(諸如賊、As used herein, the term "wild type" or "natural type" refers to a protein or a peptide that is in its operational form or functional form and usually finds a natural functional form in the body. #楚^, 4术 s I also refers to a protein form that has not been artificially modified or altered. The 箸;} 寻 S I can also be about recombinant protein. Thus, the terms may refer to a "/~y frfr protein produced in an animal of origin relative to the nucleic acid and/or amino acid sequence of the protein, having altered glycosylation forms (including lack of glycosylation) ) protein. As used herein, unless otherwise indicated, the term "G-CSF" or a particulate neoplastic cell stimulating factor refers to a protein having an amino acid sequence as set forth in SEQ ID NO: 1 (Figure 1), or has substantial A homogenous amino acid sequence and having a biological property associated with stimulation of white blood cell production. As used herein, the terms include such intentionally modified proteins (eg, '''''''''''''''''''' -CSF has the addition, deletion, or substitution of an amino acid residue. These terms include natural and recombinantly produced human G_CSF. G-CSF refers to naturally occurring proteins or recombinant proteins, usually humans, obtained from any known source, such as tissue cultures, protein synthesis, natural or recombinant cell cultures. As used herein, the amino acid sequence is "substantially identical" as defined by the FASTA search method according to Pearson and Lipman, Proc. Natl. Acad. Sci. USA 85, 2444-2448 (1988), a sequence and Another amino acid sequence has at least 70% 'typically at least about 80%, and more typically at least about 90% identical. 0 150941.doc • 16 - 201138831 As used herein, the term "conjugate" refers to a protein or polypeptide line and A protein or polypeptide or a cluster thereof in which one or more other chemical groups attached to the conjugate bond interact to function. The G-CSF molecule can be conjugated to the polymer unit according to the present invention, including unmutated and mutated 昼"白暂,士私 J# y barren beak, such as but not limited to growth factors, antibodies, hormones specific 5 A therapeutically active protein such as, but not limited to, erythropoietin, interferon alpha, interferon P, interferon 丫, consensus interferon, G CSF GM-CSF, heme, interleukin (such as interleukin-2) And interleukin-6), tumor necrosis factor, various cytokines, growth factors (such as human growth factors and epidermal growth factor) 'immunoglobulin (such as thieves,

IgE IgM IgA、IgD)、及/或其結構及/或功能變體及/或 片段、及其類蛋白質或合成之類似蛋白質形式。 通常’適用於進行本發明之諸如G_CSF之蛋白質可呈自 哺礼動物中早離出之任—形式;由基因組或CD·選殖術 或由DNA σ成*所付到表現外源性dna序列之原核或真核 宿主的產物;4由化學合成製程或由内源性基因活化所得 之產物。因此’蛋白當可办&丄 皮曰貝了來自由組織、哺乳動物·微生物 細胞培養物、植物細胞培養物、轉殖基因動物、酵母菌、 真菌及/或轉殖基因植物得到之天然或重組來源。適宜原 核宿主包括多種細g ’諸如大腸桿g(E. eGli);適宜真核 宿主包括酵母菌,諸如釀酒 .· 、乂 f u畔母® (s· cerevisiae)或畢氏酵 母菌(pichia pastoris) ’·。君乳動物細胞,諸如中國倉鼠卵巢 細胞或猴細胞;諸如小宣、& , ^兔、山平、綿羊之轉殖基因動 150941.doc 17 201138831 物;植物細胞培養物及轉殖基因植物,諸如球翰薛 (Physcomitrellapatens)(—種蘚類)。取決於所使用之宿 主’蛋白質表現產物可經來自哺乳動物、植物或其他真核 生物之碳水化合物醣基化,或可未經醣基化。 若蛋白質為G-CSF ’則G-CSF表現產物可亦包括於第工位 處之起始曱硫胺酸胺基酸殘基。雖然通常使用重組G_ CSF,尤其係獲自大腸桿菌者,但本發明涵蓋一種以任一 及所有該等形式之G-CSF之用途。已報導某些g_csf類似 物具生物學功能’且其亦可根據本發明進行接合。該等G_ CSF類似物可包括彼等與根據SEq ID N〇. 1之G-CSF胺基酸 序列比對而具有胺基酸加成、缺失及/或取代者。於某些 實施例中’與SEQ ID No. 1,序列比對,該序列包括插入 之胺基酸,如,例如於SEQ ID No. 1之第36、37及38位處 插入VSE。於某些實施例中,序列係如SEq id No. 2(圖 2)。 通常’蛋白質為具有G-CSF活性者,包括G-CSF突變 體、經醣基化之G-CSF、未經醣基化之G-CSF、及/或G-C SF之經修飾結構及/或功能之變體。於另一實施例中,蛋 白質具有如SEQ ID NO. 1之G-CSF胺基酸序列,其對應於 細菌中產生之重組G-CSF,具有174個胺基酸及額外之N端 曱硫胺醯殘基。亦包括不同於SEQ ID NO. 1(不含第1位置 之甲硫胺醯殘基)但具G-CSF生物學活性之胺基酸序列。 G-CSF接合物 於某些實施例中,本發明係關於一種組合物,其包含至 150941.doc • 18 - 201138831 少一類G-CSF蛋白質,其中& G_CSF分子係與至少一個聚 乙二醇分子共價鏈結,且至少30%之該組合物並非於N端 處經聚乙二醇化。於一項特定實施例中,各G_CSF分子係 與單個聚乙二醇分子共價鏈結(「經單聚乙二醇化之G_ CSF」),且至少30%之該組合物並非於N端處經聚乙二醇 化。於另一項實施例中,組合物包含至少8〇%經單聚乙二 醇化之G-CSF,其中至少30%之該組合物並非於N端處經聚 乙二醇化。於其他實施例中,組合物包含至少85%、至少 900/。或至少95°/。經單聚乙二醇化之G_CSF分子,其中至少 30%之該組合物並非於n端處經聚乙二醇化。 於組合物之特定實施例中,各G_CSF分子係與至少一個 聚乙二醇分子透過特定之離胺酸殘基共價鏈結。於某些實IgE IgM IgA, IgD), and/or structural and/or functional variants and/or fragments thereof, and protein-like or synthetic analogous protein forms thereof. Usually, 'a protein suitable for carrying out the present invention, such as G_CSF, may be in the form of early leave from a feeding animal; form an exogenous DNA sequence by genomic or CD-selection or by DNA σ* a product of a prokaryotic or eukaryotic host; 4 a product obtained by a chemical synthesis process or by activation of an endogenous gene. Therefore, 'protein can be & suede mussels derived from tissues, mammals, microbial cell cultures, plant cell cultures, transgenic animals, yeasts, fungi and/or transgenic plants, or Restructuring sources. Suitable prokaryotic hosts include a variety of fine g 'such as the large intestine g (E. eGli); suitable eukaryotic hosts include yeast, such as wine. ·, 乂 cerevisiae or pichia pastoris '·. Jungle animal cells, such as Chinese hamster ovary cells or monkey cells; such as Xiaoxuan, &, rabbit, Shanping, sheep, transgenic genes, 150941.doc 17 201138831; plant cell cultures and transgenic plants, such as Physcomitrellapatens (-species). Depending on the host' protein performance product used, it may be glycosylated by carbohydrates from mammals, plants or other eukaryotes, or may be unglycosylated. If the protein is G-CSF ' then the G-CSF performance product may also be included in the starting thiol amino acid residue at the station. While recombinant G_CSF is commonly used, especially from E. coli, the invention encompasses the use of any and all such forms of G-CSF. It has been reported that certain g_csf analogous organisms have biological functions' and they can also be joined in accordance with the present invention. Such G_CSF analogs may include those having an amino acid addition, deletion and/or substitution as compared to the G-CSF amino acid sequence according to SEq ID N〇. In certain embodiments, the sequence is aligned with SEQ ID No. 1, which includes an inserted amino acid, such as, for example, at positions 36, 37, and 38 of SEQ ID No. 1, inserted into VSE. In certain embodiments, the sequence is such as SEq id No. 2 (Fig. 2). Typically, the protein is a G-CSF-active, including a G-CSF mutant, a glycosylated G-CSF, an unglycosylated G-CSF, and/or a modified structure and/or function of GC SF. a variant. In another embodiment, the protein has the G-CSF amino acid sequence of SEQ ID NO. 1, which corresponds to recombinant G-CSF produced in bacteria, has 174 amino acids and additional N-terminal guanamine醯 residue. Also included are amino acid sequences other than SEQ ID NO. 1 (containing the methionine residue at position 1) but having G-CSF biological activity. G-CSF Conjugates In certain embodiments, the present invention is directed to a composition comprising less than one type of G-CSF protein to 150941.doc • 18 - 201138831, wherein & G_CSF molecule is associated with at least one polyethylene glycol The molecule is covalently linked and at least 30% of the composition is not PEGylated at the N-terminus. In a specific embodiment, each G_CSF molecule is covalently linked to a single polyethylene glycol molecule ("mono-glycolized G_CSF"), and at least 30% of the composition is not at the N-terminus It is pegylated. In another embodiment, the composition comprises at least 8% by weight of mono-glycolylated G-CSF, wherein at least 30% of the composition is not PEGylated at the N-terminus. In other embodiments, the composition comprises at least 85%, at least 900/. Or at least 95°/. A mono-PEGylated G_CSF molecule wherein at least 30% of the composition is not pegylated at the n-terminus. In a particular embodiment of the composition, each G_CSF molecule is covalently linked to at least one polyethylene glycol molecule through a particular lysine residue. In some real

施例中,本發明包含一種組合物,其具有至少一個G_CSF 分子,經由G-CSF蛋白質胺基端之胺鏈結與至少一個聚乙 二醇分子共價鏈結。於其他實施例中,本發明包含一種組 &物其具有至少一個G-CSF分子,透過LySi 6(若計算n 立而之曱V胺酸殘基,則為Lys 17)與至少一個聚乙二醇分子 共價鏈結。 於些組合物之實施例中,於N端處經聚乙二醇化之分 子對第二類群(尤其係於Lys 16處經聚乙二醇化之分子類群) 之比例為小於約1比約1 〇〇、約丨〇比約9〇、約2〇比約8〇、約 0比約70、約40比約60、'約50比約50、約60比約40、約70 比約30,其中小於約!包括利用相關技術中已知之標準方 法不可測得之數量。 '50941.doc -19- 201138831 於某二貫施例中’提供一種實質上均質之組合物,其包 含單聚乙二醇化之I白晳,。 玄白負其中至少30%之該蛋白質分子 並非於N端聚乙二醇化。於某些特定實施例中,組合物包 含至少30〇/〇早聚乙二醇化之蛋白質,其係於個胺基 酸以内之離胺酸殘基經聚乙二醇化。於特定實施例中,離 胺酸為N端80個以内、7〇個以内、6〇個以内、5〇個以内、 40個以内、30個以内、2〇個以内、”個以内、以個以内、 17個以内、16個以内、15個以内' 14個以内、⑶固以内、 12個以内、_以内、1〇個以内、9個以内、8個以内、7 個以内、6個以内、5個以内、4個以内、3個以内或?個以 内之胺基酸。 大多數多肽具有多個潛在的PEG鏈結位點。因此,雖然 1PEG-蛋白質接合物之均質群具有一個pEG分子與各蛋白 質刀子鏈結,但疋該鏈結不一定位於該群中各蛋白質上之 相同位置。類似地,雖然經二聚乙二醇化之蛋白質接合物 之均質群具有兩個peg分子與各蛋白質分子鏈結,但是該 等鏈結不一定位於該群中之各蛋白質上之相同位置。 一些研究已經指出’於Lysl7聚乙二醇化可能損害重組 G-CSF 之 /舌性。例如,Reidhaar 〇ls〇n 等人((ο%) Biochemistry 35 ·· 9034-9041)闡述之掃描誘變揭示離胺酸 35及離胺酸41為生物活性關鍵之殘基,且結構模擬研究揭 示G-CSF於離胺酸π及離胺酸24聚乙二醇化可能導致蛋白 質之生物學活性降低’由於其位於蛋白質受體相互作用之 區域内。聚合物部份(諸如聚乙二醇)連接於該等位置可因 150941.doc -20- 201138831 此外,Aritomi等人 此於空間上阻礙受體結合 (1999)Nature 401 : 713-717^1 ^ W假疋Lys 1 7修飾對蛋白質生物學 活性之負面影響。 PEG分子 爲本發明之目的’通常藉由還原性烷基化製備聚合物_ 蛋白質接合物。因此,本發明提供聚合物-蛋白質接合 物,其中蛋白質胺基中之至少一個氮原子各與聚合物單元 經胺鏈結接合。於本發明中,術語胺基包括第一及第二胺 基,特別是胺基酸側鏈中之而_或顺2_基目,諸如離胺酸 側鏈中之NH2·基團 '精胺酸胍基中之NH^tNH2_基團,或 組胺酸咪唑側鏈中之NH-基團。 聚合物單元通常包含至少一個聚合物部份及連接子部 份,其係位於該至少一個聚合物部份與胺鏈結之間。連接 子部份可為直鏈或分支鏈。若連接子部份為分支鏈,則聚 合物單元可包含一個以上之聚合物部份。連接子部份通常 為脂族連接子部份。適宜之脂族連接子部份亦包括經取代 之烧基二胺及三胺、離胺酸酯及丙二酸酯衍生物。連接子 部份通常為非平面型’因此聚合物鏈並非固定不動。通 常’連接子部份包含經多官能基化之含至多丨8個,且更通 书1至10個奴原子之烧基。諸如氮、氧或硫之雜原子亦可 包含於烷基鏈中。連接子部份可於例如碳原子或氮原子處 刀支。为支鍵連接子部份貫例及所得之分支鍵聚合物單元 及其製備方法闡述於W0 95/ll 1924及W0 03/1049699中。 本發明一項實施例中’該鍵連接子部份包含至少一個亞 150941.doc 21 201138831 曱基附接在胺鏈結之氮原子上,例如:i至12個、1至10 個、1至8個、1至7個、1至6個、1至5或更少個,如:4、 3、2或1個,及最常為2個亞甲基直接附接在胺鏈結之氮原 子上。 本發明所涵蓋之聚合物包括(但不限於)聚乙二醇及其衍 生物’包括PEG、經甲氧基化之PEG(「mPEG」)、PEG均 聚物、聚乙二醇均聚物、乙二醇與丙二醇之共聚物,其中 該均聚物及共聚物係於末端處經烷基取代或未經取代。於 某些實施例中,聚合物為mPEG且最常見為經單甲氧基化 之PEG。水溶性聚合物可為分子重量範圍廣泛之直鏈、分 支鏈、或星形聚合物。PEG之大小可為丨〇至約10〇 kD。於 特定實施例中,PEG之大小為約10至約% kD。於一些實 加例中,附接至胺基之聚乙二醇部份之分子重量為2至1〇〇 kDa ’更通常為5至60 kDa,且最通常為1〇至3〇 kDa。於本 發明之另一實施例中,聚乙二醇部份中之伸乙基氧化物殘 基之數量η為約40至約2270,更通常為約11〇至約137〇,且 最通常為約225至約680。 聚合物部份通常為實質上無抗原性或無免疫原性之聚合 物鏈❶此外,所採用之聚合物部份通常係選自水溶性聚合 物部份。如此選擇具有如下優點:即附接有或接合有水溶 性聚合物部份之蛋白質不會在諸如生理環境之水性環境中 沉殿。爲了進行還原性院基化反應,所選擇之聚合物應另 外具有單個具反應性之酿’以便如本發明方法提供控制聚 合度。聚合物單元及聚合物部份可為分支鏈或不分支鍵。 150941.doc -22· 201138831 r。爲了所製得之終產物用於治療用途,聚合物應為醫 #上:接文。熟f此項技術者將能夠基於如下考慮因素選 、品之聚合物部份.諸如聚合物_蛋白質接合物是否會 用於/α療’且若是’則考慮所需劑量、循環時間、耐蛋白 水解性質、及其他考慮因素。 立通吊’聚合物部份可自以下組成之群中選出:聚院二醇 ί5伤’夕部份,諸如葡聚糖及其衍生物、多_及其衍生 各定酮部份,諸如聚乙浠基吡咯啶酮;纖維素部 份,諸如羥甲基纖維素、聚乙烯基醇、聚-1,3-二噁戊烷、 水-1,3,6-二噁烷、乙烯_馬來酸酐共聚物·,聚胺基酸部份; 及/或聚丙烯醯胺部份;及/或其他類似之無免疫原性之聚 合物部份(均聚物或隨機共聚物)及/或其衍生物。該等聚合 物包含於本發明中時亦能夠發揮作用或具活性。 特疋α之,聚合物部份為聚烷二醇部份。術語聚烷二醇 意指聚烷二醇基或聚烷基二醇部份,其中伸烷基為直鏈或 为支鏈基團。術語聚烷二醇亦包括由烷二醇混合物形成之 聚烷二醇,諸如含有聚伸乙基及聚伸丙基混合物之聚合 物、及含有聚伸異丙基、聚伸乙基及聚伸異丁基混合物之 聚合物。根據本發明之聚合物蛋白質接合物中之聚烷二醇 部份通常為藉由去除兩個末端羥基所形成之聚乙二醇部份 或聚乙二醇殘基。 屬於該群者為a-經取代之聚伸烷基氧化物衍生物,諸如 甲氧基聚乙二醇(mPEG);或其他經適宜烷基取代之聚伸 烷基氧化物衍生物,諸如彼等含有單-或雙-末端crC4基團 I50941.doc -23· 201138831 者。通常為諸如單曱基聚乙二醇均聚物之直鏈無抗原性聚 合物。亦適用之替代性聚伸烷基氧化物,諸如其他聚乙二 醇均聚物、聚乙二醇雜聚物、其他烷基聚伸烷基氧化物嵌 段共聚物及聚伸烷基氧化物嵌段共聚物之共聚物。 為使聚乙二醇(PEG)及類似之聚(伸烧基氧化物)共價附 接至分子(特定言之蛋白質),該聚合物之羥基末端必須首 先轉化成具反應性之官能基。該製程於文中係稱為「活 化」,且產物稱為「經活化之PEG」。例如,可活化經甲氧 基化之PEG(「mPEG」)’以使其隨後經相關技術中已熟知 之方法,共價附接至胺基,亦即mPEG可經修飾而含有不 同反應性部份,適於隨後經由包含可利用之胺基殘基(例 如離胺醯基殘基)之胺基酸殘基附接至蛋白質。 於根據本發明之方法中使用之peg試劑通常為具有如下 通式之試劑:.丨_CH0,其 中R為Η、低碳數烷基、芳基或任一適宜保護基團;n為整 數,其表示聚乙二醇部份中之伸乙基氧化物殘基之數量; m為整數’其表示亞甲基之數量;Llg〇、ν、s及/或分支 或不分支之連接子部份,其可不存在或存在;L2為分支或 不为支之連接子部份,其可不存在或存在;且丫為整數, 其限制條件為:當不存在L2時y為i,且當存在L2時,y為 至少1。於特定實施例中,該PEG為PEG乙醛化物,最通常 為甲氧基-PEG乙醛化物。聚乙二醇_醛化物之結構如下: 150941.doc •24· 201138831In one embodiment, the invention comprises a composition having at least one G_CSF molecule covalently linked to at least one polyethylene glycol molecule via an amine linkage at the amine end of the G-CSF protein. In other embodiments, the invention comprises a group & matter having at least one G-CSF molecule, permeating through LySi 6 (or Lys 17 if the ruthenium V amine acid residue is calculated) and at least one polyethylene Covalent linkage of diol molecules. In some embodiments of the composition, the ratio of the PEGylated molecule at the N-terminus to the second group (especially the PEGylated molecular group at Lys 16) is less than about 1 to about 1 〇. 〇, about 〇 about 9 〇, about 2 〇 to about 8 〇, about 0 to about 70, about 40 to about 60, 'about 50 to about 50, about 60 to about 40, about 70 to about 30, of which Less than about! This includes quantities that are not measurable using standard methods known in the art. '50941.doc -19- 201138831 In a second embodiment, a substantially homogeneous composition comprising a mono-pegylated I white is provided. At least 30% of the protein molecules are not PEGylated at the N-terminus. In certain specific embodiments, the composition comprises at least 30 〇/〇 early PEGylated protein which is PEGylated by an amine acid residue within the amino acid. In a specific embodiment, the amine acid is 80 or less in the N-terminus, within 7 〇, within 6 、, within 5 、, within 40, within 30, within 2 、, within, within Within, within 17, within 16, within 15, within 14, within (3), within 12, within _, within 1 within 9, within 8, within 8, within 6 or within 6 Amino acids within 5, within 4, within 3 or within. Most polypeptides have multiple potential PEG-linked sites. Thus, although a homogenous group of 1PEG-protein conjugates has a pEG molecule Each protein knife is linked, but the chain is not necessarily located at the same position on each protein in the group. Similarly, although a homogenous group of diPEGylated protein conjugates has two peg molecules and each protein molecule Chains, but these chains are not necessarily located at the same position on each protein in the population. Some studies have indicated that 'PEGylation of Lysl7 may impair the / G-CSF of the recombinant G-CSF. For example, Reidhaar 〇ls〇 n et al ((ο%) Biochemistry 35 ·· 9034-9041) The scanning mutagenesis revealed that lysine 35 and lysine 41 are the key residues of biological activity, and structural simulation studies revealed that G-CSF may be PEGylated in cis-amino acid and lysine 24. Resulting in a decrease in the biological activity of the protein 'because it is located in the region of protein receptor interaction. Polymeric parts (such as polyethylene glycol) are attached to these positions due to 150941.doc -20- 201138831 In addition, Aritomi et al. This sterically hinders receptor binding (1999) Nature 401: 713-717^1 ^ W negative effects of Lys 1 7 modification on protein biological activity. PEG molecules are the object of the invention 'usually by reducing alkane The polymerase is prepared as a protein conjugate. Accordingly, the present invention provides a polymer-protein conjugate in which at least one nitrogen atom of a protein amine group is each bonded to a polymer unit via an amine chain. In the present invention, the term amine The group includes the first and second amine groups, particularly in the side chain of the amino acid, or the cis- 2 group, such as the NH 2 group in the side chain of the amine acid, NH 2 in the arginine group a tNH2_ group, or a histidine imidazole side chain The NH-group. The polymer unit usually comprises at least one polymer moiety and a linker moiety between the at least one polymer moiety and the amine chain. The linker moiety may be a straight chain or a branched chain. If the linker moiety is a branched chain, the polymer unit may comprise more than one polymer moiety. The linker moiety is typically an aliphatic linker moiety. Suitable aliphatic linker moieties also include substituted Alkyl diamines and triamines, leucoates and malonate derivatives. The linker moiety is usually non-planar type so the polymer chain is not immobile. Typically, the linker moiety comprises a polyfunctional group containing up to 8 and more than 1 to 10 slave atoms. Heteroatoms such as nitrogen, oxygen or sulfur may also be included in the alkyl chain. The linker moiety can be cleavable at, for example, a carbon atom or a nitrogen atom. The branched bond moiety and the resulting branched bond polymer unit and the preparation method thereof are described in WO 95/11924 and WO 03/1049699. In one embodiment of the invention, the bond linker moiety comprises at least one sub-150941.doc 21 201138831 thiol attached to the nitrogen atom of the amine chain, for example: i to 12, 1 to 10, 1 to 8, 1 to 7, 1 to 6, 1 to 5 or less, such as: 4, 3, 2 or 1, and most often 2 methylene directly attached to the nitrogen of the amine chain On the atom. Polymers encompassed by the present invention include, but are not limited to, polyethylene glycol and its derivatives 'including PEG, methoxylated PEG ("mPEG"), PEG homopolymer, polyethylene glycol homopolymer And a copolymer of ethylene glycol and propylene glycol, wherein the homopolymer and the copolymer are alkyl-substituted or unsubstituted at the terminal. In certain embodiments, the polymer is mPEG and most commonly mono-methoxylated PEG. The water soluble polymer can be a linear, branched, or star polymer having a wide range of molecular weights. The size of the PEG can range from about 10 〇 kD. In a particular embodiment, the size of the PEG is from about 10 to about % kD. In some embodiments, the molecular weight of the polyethylene glycol moiety attached to the amine group is from 2 to 1 〇〇 kDa ', more typically from 5 to 60 kDa, and most typically from 1 〇 to 3 〇 kDa. In another embodiment of the invention, the amount of pendant ethylenoxide residues in the polyethylene glycol moiety is from about 40 to about 2270, more typically from about 11 to about 137, and most typically About 225 to about 680. The polymer portion is typically a polymer chain that is substantially non-antigenic or non-immunogenic. In addition, the polymer portion employed is typically selected from the group of water soluble polymers. Such selection has the advantage that the protein to which the water-soluble polymer portion is attached or bonded does not settle in an aqueous environment such as a physiological environment. In order to carry out the reductive densification reaction, the selected polymer should additionally have a single reactive brew to provide a controlled degree of polymerization as in the method of the invention. The polymer unit and the polymer portion may be branched or unbranched. 150941.doc -22· 201138831 r. In order for the final product to be used for therapeutic purposes, the polymer should be in the form of: Those skilled in the art will be able to select the polymer portion of the product based on considerations such as whether the polymer-protein conjugate will be used for /alpha therapy' and if so, consider the required dose, cycle time, protein resistance Hydrolyzed properties, and other considerations. The Litong Hang's polymer part can be selected from the group consisting of: 聚聚diol ί5 injured 'xi part, such as dextran and its derivatives, poly- and its derived ketone parts, such as poly Acetylpyrrolidone; cellulose fraction such as hydroxymethylcellulose, polyvinyl alcohol, poly-1,3-dioxolane, water-1,3,6-dioxane, ethylene_ma An anhydride copolymer, a polyamino acid moiety; and/or a polypropylene guanamine moiety; and/or other similar non-immunogenic polymer moiety (homopolymer or random copolymer) and/or Its derivatives. These polymers can also function or be active when included in the present invention. In particular, the polymer portion is a polyalkylene glycol moiety. The term polyalkylene glycol means a polyalkylene glycol group or a polyalkylene glycol moiety in which the alkylene group is a straight chain or a branched chain group. The term polyalkylene glycol also includes polyalkylene glycols formed from a mixture of alkanediols, such as polymers containing a mixture of polyethylene and poly(propyl) propyl groups, and poly(extended isopropyl), polyethylidene and polycondensation. A polymer of an isobutyl mixture. The polyalkylene glycol portion of the polymer protein conjugate according to the present invention is usually a polyethylene glycol moiety or a polyethylene glycol residue formed by removing two terminal hydroxyl groups. The group belonging to the group is an a-substituted polyalkylene oxide derivative such as methoxypolyethylene glycol (mPEG); or other polyalkylene oxide derivative substituted by a suitable alkyl group, such as Etc. containing a mono- or bi-terminal crC4 group I50941.doc -23· 201138831. Typically, it is a linear, non-antigenic polymer such as a monodecyl polyethylene glycol homopolymer. Alternative polyalkylene oxides, such as other polyethylene glycol homopolymers, polyethylene glycol heteropolymers, other alkyl polyalkylene oxide block copolymers, and polyalkylene oxides a copolymer of block copolymers. In order to covalently attach polyethylene glycol (PEG) and similar poly(alkylene oxide) to a molecule (specifically, a protein), the hydroxyl end of the polymer must first be converted to a reactive functional group. This process is referred to as "activation" in the text and the product is called "activated PEG". For example, the methoxylated PEG ("mPEG")' can be activated to be covalently attached to the amine group by methods well known in the art, i.e., the mPEG can be modified to contain different reactive moieties. Part, suitable for subsequent attachment to a protein via an amino acid residue comprising an available amine residue (eg, an amine sulfhydryl residue). The peg reagent used in the method according to the invention is generally a reagent having the formula: 丨_CH0, wherein R is hydrazine, a lower alkyl group, an aryl group or any suitable protecting group; n is an integer, It represents the number of ethyl epoxide residues in the polyethylene glycol moiety; m is an integer 'which indicates the number of methylene groups; Llg〇, ν, s and/or the branched or unbranched linker moiety L2 is a branch or a non-branched linker part, which may or may not exist; and 丫 is an integer, with the constraint that y is i when L2 is absent, and when L2 is present , y is at least 1. In a particular embodiment, the PEG is a PEG acetaldehyde, most typically a methoxy-PEG acetaldehyde. The structure of polyethylene glycol_formaldehyde is as follows: 150941.doc •24· 201138831

H,CH, C

接合方法 提供-種製備蛋白f接合物之方法、及由該方法所製得 之接合物,該方法包括:令蛋白質與經活化之聚乙二醇于 链化物於包含DMSO之反應緩衝液中反應,使該蛋白―質盘 該經活化之水溶性聚合物共價鏈結。該方法可亦包括去除 實質上所有未經鏈結之水溶性聚合物,以獲得該蛋白質二 合物。於某些實施例中,聚乙二醇-醛化物為 、The ligation method provides a method of preparing a protein f conjugate, and a conjugate prepared by the method, the method comprising: reacting a protein with an activated polyethylene glycol in a reaction buffer containing DMSO The protein-quality plate is covalently linked to the activated water-soluble polymer. The method can also include removing substantially all of the unchained water soluble polymer to obtain the protein dimer. In certain embodiments, the polyethylene glycol-formaldehyde is

。於某些實施例中,peg為 至少10 kD。於某些其他實施例中,pEG為至少2〇比。於 特定實施例中,PEG為30 kD或40 kD。 、 用於反應性烷基化之還原試劑通常係選自但不限於. In some embodiments, peg is at least 10 kD. In certain other embodiments, the pEG is at least 2 turns ratio. In a particular embodiment, the PEG is 30 kD or 40 kD. The reducing agent for reactive alkylation is usually selected from, but not limited to,

NaCNBH4 或 NaBH4。 通韦,該反應在蛋白質濃度為〇 5至1〇〇 mg/ml,更通常 為1至10 mg/m卜且最通常為3至7 mg/ml下進行。該反應 =可於蛋白質务聚合物之莫耳比為1:1至1:400,且通 常為1 : : 30 ’且最通常為】:i5y : 3〇下進行。於 另一項實施例中’反應緩衝液包含蛋白質對經活化之水溶 性聚合物之莫耳比為約1比約3至約1比約60。於其他實施 例中’反應緩衝液包含蛋白質對經活化之水溶性聚合物之 4耳比為約1比約4、約1比約5、m比約6、W比約7、約 I50941.doc •25· 201138831 1比約8、約!比約9、約!比約1〇、約“匕㈣、約i比約 20、約U匕約25、約!比約30、約】比約35、約以約扣、約 1比約45、約!比約50、約U^55、^比約⑼。於某些實 施例中,聽緩衝;夜包含蛋白質對經活化之水溶性聚合物 之莫耳比為約1比約7。於另-項實施例中,可藉由相關技 術中已知之方法,諸如例如透析術或層析術而去除實質上 所有未反應之水溶性聚合物。 化 雖然所# S自質均可根據文中所述之方法經聚乙二醇 ’然而本發明特定言之涵蓋使治療性多肽聚乙二醇化。NaCNBH4 or NaBH4. Tongwei, the reaction is carried out at a protein concentration of 〇 5 to 1 mg/ml, more usually 1 to 10 mg/m b and most usually 3 to 7 mg/ml. The reaction = can be carried out at a molar ratio of the protein-forming polymer of from 1:1 to 1:400, and usually 1: 1: 30 and most usually: i5y: 3 Torr. In another embodiment, the reaction buffer comprises a molar ratio of protein to activated water-soluble polymer of from about 1 to about 3 to about 1 to about 60. In other embodiments, the 'reaction buffer comprises a protein to the activated water soluble polymer at a ratio of about 4 to about 4, about 1 to about 5, m to about 6, W to about 7, about I50941.doc •25·201138831 1 than about 8, about! More than about 9, about! Approximately 1 〇, about "匕 (4), about i is about 20, about U is about 25, about! About 30, about] about 35, about about deduction, about 1 to about 45, about! , about U^55, ^ about (9). In some embodiments, the hearing buffer; the night contains protein to the activated water-soluble polymer molar ratio of about 1 to about 7. In another embodiment Substantially all of the unreacted water-soluble polymer can be removed by methods known in the art, such as, for example, dialysis or chromatography. Although the #S self-quality can be obtained by the method described herein. Glycols 'However, the invention specifically encompasses pegylation of therapeutic polypeptides.

於某些實施财,根據本發明方法所使用之治療性蛋白】 可為例如蛋白酶、垂體激素蛋白酶抑制劑、生成^ (P 一、群落刺激因子、激素、凝血因+、抗凝血@ 子、神經營養因子、類風濕因子、CD蛋白f、骨誘發廷 子、介白素、生長因子、干擾素 '細胞激素、胰島素樣片 長因子、趨化激素、免疫球蛋白、促性腺激素、介白素、E 趨化素(chemotactin)、干擾素、脂質結合蛋白質過敏原、 或别述物質之組合。該等治療性蛋白質之具體非限制性實 例包括.干擾素_α2Α、干擾素_α2Β、干擾素β、干擾素 γ、胰島素樣生長因子_I(iGF_1}、胰島素樣生長因子-2(IGF-2)、胰島素、人類生長激素(hGH)、轉形生長因子 (TGF)、促紅血球生成素(Ep〇)、睫狀神經轉形因子 (CNTF)、血小板生成素(τρ〇)、腦源性神經突因子 (BDNF) IL-1、促胰島素釋放激素(insuiintr〇pin)、〖I 2 神經勝質源性神經因子(GDNF)、IL_丨RA、組織纖維蛋白 I5094I.doc •26- 201138831 溶酶原活化因子g u ,, (}超氧化物歧化酶(S〇D)、尿激酶、 過氧化氫酶、鏈激酶、成纖維細胞生長因子(FGF)、血紅 素神經犬生長因子、腺苦脫醯胺酶(NGF)、顆粒細胞性 巨嗟細胞群落刺激因子(GM_CSF)、牛生長激素(bgh)、顆 粒性細胞群落刺激因子(G_CSF)、降^素、血小板源性 生長因子(PDGF)、加強殺細菌力/通透性之蛋白質(Βρι)、 L-天冬醯胺酸酶、精胺酸酶、尿酸酶、丫_干擾素、苯丙胺 酸氨解酶、即泡增生激素、胰島素原、表皮生長因子、成 纖維細胞生長因?、神經生長因子(NGF)、腫瘤壞死因 子降血妈素、甲狀旁腺激素(pTH,包括人類p丁H)、骨 形態發生蛋白、造血性生長因子、促黃體生成激素、膜高For some implementations, the therapeutic protein used in the method of the present invention may be, for example, a protease, a pituitary hormone protease inhibitor, a production (P1, a community stimulating factor, a hormone, a coagulation factor +, an anticoagulation@子, Neurotrophic factor, rheumatoid factor, CD protein f, osteoinductive, interleukin, growth factor, interferon' cytokine, insulin-like long-term factor, chemokine, immunoglobulin, gonadotropin, interleukin , Echemotactin, interferon, a lipid-binding protein allergen, or a combination of other substances. Specific non-limiting examples of such therapeutic proteins include interferon-α2Α, interferon_α2Β, interferon β, interferon gamma, insulin-like growth factor _I (iGF_1}, insulin-like growth factor-2 (IGF-2), insulin, human growth hormone (hGH), transforming growth factor (TGF), erythropoietin ( Ep〇), ciliary neurotransformation factor (CNTF), thrombopoietin (τρ〇), brain-derived neurite outgrowth factor (BDNF) IL-1, insulin releasing hormone (insuiintr〇pin), I 2 nerve victory Primitive god Factor (GDNF), IL_丨RA, tissue fibrin I5094I.doc •26- 201138831 lysogen activator gu ,, (} superoxide dismutase (S〇D), urokinase, catalase, chain Kinase, fibroblast growth factor (FGF), heme neuron canine growth factor, adenine deaminase (NGF), granulosa cell giant cell community stimulating factor (GM_CSF), bovine growth hormone (bgh), granular Cell community stimulating factor (G_CSF), vasopressin, platelet-derived growth factor (PDGF), protein that enhances bactericidal/permeability (Βρι), L-aspartate prolylase, arginase, uric acid Enzyme, 丫_interferon, phenylalanine aminolysis enzyme, ie, vesicle proliferating hormone, proinsulin, epidermal growth factor, fibroblast growth factor, nerve growth factor (NGF), tumor necrosis factor, blood serotonin, thyroid Glandular hormone (pTH, including human p-butyl H), bone morphogenetic protein, hematopoietic growth factor, luteinizing hormone, membrane high

血糖素、胰高血糖素樣肽-HGLP」)、Y^ypYY)、VIIIC 因子、ιχ因子、組織因子、及溫韋伯氏因子Glucagon, glucagon-like peptide-HGLP", Y^ypYY), VIIIC factor, ιχ factor, tissue factor, and Wen Weber's factor

Willebrand factor)、蛋白質c、心房利鈉因子、肺表面活 性蛋白、蛙皮素、凝血素、腦啡肽酶、繆勒式抑制性物質 (mullerian-inhibiting agent)、恥骨鬆弛激素A鏈、恥骨鬆 弛激素B鏈、前恥骨鬆弛激素、脫氧核糖核酸酶、抑制 素、活化素、血管表皮生長因子、整合素、蛋白質八或 D、骨源性神經營養因子(BDNF)、神經營養素·3、_4、 -5、或 _6(NT-3、NT-4、NT-5、或 NT-6)、CD-3、CD-4、 CD-8、CD-19、M-CSF、GM-CSF、G-CSF、任一前述物質 之生物活性片段、或前述物質之組合。 爲了維持pH處於較佳範圍内,反應可於缓衝液存在下進 行。例如,緩衝液可選自磷酸鹽緩衝液、乙酸鹽緩衝液、 150941.doc •27· 201138831 HEPES、MES、或其他類似緩衝液。反應緩衝液可通常為 不含胺組分之標準緩衝液,例如磷酸鹽缓衝生理食鹽水 (PBS)。反應緩衝液通常包括鹽(例如Na鹽),其濃度為約 0.1 mM至約100 mM,最通常約1 mM至約50 mM,且更通 常為約10 mM至約20 mM。於某些實施例中,於攪拌下使 多肽與乾燥之經活化之水溶性聚合物混合。反應緩衝液之 pH通常為約6.5至約8.5或約6.6至約7.5。於某些實施例 中,反應緩衝液具有約7.0之中性pH。 反應緩衝液另外包含有機溶劑,特別是二甲基亞砜 (「DMSO」)。DMSO可以濃度 5-80%,通常 10-40%(v/v)存 在於反應混合物中。DMSO廣泛用作通用溶劑,但通常不 用於影響聚乙二醇化反應,特別不用於優先影響該反應遠 離蛋白質N端之聚乙二醇化產生位點β ρ〇τ公開案w〇 08/0192 14闡述一種於含dMS〇緩衝液中選擇性製備^^端經 修飾之紅血球生成素之方法。PEG鏈結提供胺基f酸酯選 擇性鏈結於蛋白質N端之PEG-EPO。 相反地,本文所述之反應條件,包括使用醛接合之 PE^,似乎特異性驅使活化之pEG與特定離胺酸位點共價 接δ此外,現已發現,向反應緩衝液添加DMSO可改變 聚乙二醇化之位1。特別如I文所㉛,向反應緩衝液添加 _會驅使反應朝向使蛋白質中之〜17優先聚乙二醇 2。因此,本發明方法可以選擇性修飾有關蛋白質中之特 疋胺基’特別是修飾i白質胺基端附近之離胺酸殘基。該 種選擇性修飾該等蛋白質係有益的,因為—般已瞭解二 150941.doc -28 · 201138831 水浴性聚合物(例如PEG)與蛋白質經胺基相連會導致活性 喪失。咸信通常與聚乙二醇化相關之活性喪失係由於先前 技術之隨機靶向離胺酸反應。隨機修飾離胺酸殘基可能不 利地改變蛋白質功能,由於實質上改變該蛋白質之三級結 構或形態。雖然不希望以任何方式受限於任一理論,但本 文所揭示之反應條件似乎可選擇性修飾蛋白質於所選殘基 或於胺基端(通常認為其不促成蛋白質活性)。 先前已報導利用醛活化之PEG對蛋白質進行特異性單聚 乙二醇化。然而,該種反應僅於低?11具特異性,而於pH 7 或以上喪失特異性。因&,本發明方法特別可用於pH敏感 性蛋白質之聚乙二醇化及/或接合。 進行反應之溫度通常為至5〇t,更通常之溫度為2。〇 至8°C,且最通常為約4t: ^對特定溫度之選擇可影響反應 時間,所選之溫度通常彳製得的聚合物·蛋白質接:物應 具有利用胺鏈結與聚合物單位接合之蛋白f之兩個胺基氣 原子。 於本文所述之反應中,經活化之水溶性聚合物之存在量 為超過莫耳量’且因此’將需要自新形成之蛋白質接合物 中去除未反應之過剩經活化之水溶性聚合物。如文中所 用’「去除實質上所有未鏈結之水溶性聚合物」係指用於 進行該種分離法之通常已知之方法,例如透過透析二… 般去除約80%未經鏈結之水溶性聚合物,通常去除約 90%,更通常去除約95%,且最通常去除約99%。 示、’ 醫樂調配物 150941.doc -29- 201138831 於某些實施例中,本發明係關於一種醫藥調配物,其包 含至少一類G-CSF蛋白質,其中各G-CSF分子係與至少一 個聚乙二醇分子共價鏈結’其視需要含於醫藥上可接受載 劑中。於某些實施例中,載劑實質上不含蛋白質。於某些 實施例中,醫藥調配物包含至少一類G-CSF蛋白質,其中 各G-CSF分子係與至少一個聚乙二醇分子共價鏈結,且至 少30%之組合物並非於n端處經聚乙二醇化。於一項特定 實施例中’各G-CSF分子係與單個聚乙二醇分子共價鏈結 (「經單聚乙二醇化之G-CSF」),且至少30%之組合物並 非於N端處經聚乙二醇化。於另一項實施例中,調配物包 含至少80%經單聚乙二醇化之g-CSF,其中至少30%之組 合物並非於N端處經聚乙二醇化。於其他實施例中,調配 物包含至少85°/。,至少90%或至少95°/。經單聚乙二醇化之 G-CSF分子’其中至少3〇%之該組合物並非於n端處經聚乙 二醇化。 於醫藥調配物之特定實施例中,各G-CSF分子係透過特 定之離胺酸殘基,與至少一個聚乙二醇分子共價鏈結。於 某些實施例中’調配物包含至少一個G-CSF分子,經由G-CSF蛋白質胺基端’利用胺鏈結與至少一個聚乙二醇分子 共價鍵結。於其他實施例中,調配物包含至少一個G_CSF 分子’透過Lysl6(若計算n端之甲硫胺酸殘基,則為 Lys 17)與至少一個聚乙二醇分子共價鏈結。 於調配物之一些實施例中,於N端處經聚乙二醇化之分 子對第二類群(尤其係於Lysl6處經聚乙二醇化之第二類群) 150941.doc 201138831 之比例為小於約1比約100、約10比約90、約20比約80、約 3〇比約7〇、約4〇比約60、約比約50、約60比約40、約7〇 比約3〇,纟中小於約1包括利用相關技術中已知之標準方 法不可測得之數量。 希望本&明之G CSF分子及组合物顯示出於標準貯藏條 件下之長期安线’亦即於標準溫度下(例如約25。〇例如 貯藏至少三個月。於某些實施例中,不含蛋白質之載劑為 不含血清、不含白蛋白或不含人類血清白蛋白(「hsa-free」)。於某些貫施例中,醫藥調配物可貯藏更長時間, 且如本文所述之方法及/或相關技術中已知之方法測定, 無實質上及/或可測得之G_CSF降解。於某些實施例中如 於約-2〇t或4t下貯藏至少15個月後測定,本發明醫藥調 配物=安定性(亦即不會顯示出可測得之降解及/或不會顯 ,、出貫質上之降解)。於其他實施例+,如於約Μ。。或約 37°C下貯藏至少10個月後測定’本發明醫藥調配物具安定 性(亦即不會顯示出可測得之降解及/或不會顯示出實質上 ^降解)。可藉由相關技術中已知之任一方法測定本發明 j藥調配物之安定性。於某些實施例中,本發明醫藥調配 物之安定性之測定方法為:如藉由二喹啉甲酸(「bca」) 蛋白質分析法敎,監測蛋白質濃度隨時間之變化。於其 他實施例中’纟發明f藥調配物之安定性之測冑方法為: 藉由SDS PAGE分析法測定蛋白質降解(亦即g_csf接合 物降解)隨時間之變化。於某些其他實施例令,安定性之 測定方法可為:由HpLC分析產物之降解。於其他實施例 150941.doc 31 201138831 中’本發明醫藥調配物之安定性之測定方法為:監測該調 配物活性隨時間之變化,其中該活性之測定方法為相關技 術中已知用於測定該調配物活性(例如G-CSF活性)之任— 活體外或活體内方法。於根據該實施例之特定實例中,包 含複數個G-CSF-接合物之本發明醫藥調配物之活性的評定 方法為:利用G-CSF依賴性鼠類32D細胞株,於活體外生 物學分析法中評定該醫藥調配物之能力,於其他實施例 中,調配物活性之測定方法為:於活體内測定其改變實驗 動物(諸如倉鼠)之血細胞數之能力。 於某些實施例中,提供一種醫藥調配物,其包含經單聚 乙二醇化之蛋白質,其中至少3〇%之該等蛋白質分子並非 於N端處經聚乙二醇化。於某些特定實施例中,調配物包 含至少鄕經單聚乙二醇化之蛋白質,其係於距離心 個胺基酸以内之離胺酸殘基處經聚乙二醇化。於某些實施 例中,該離胺酸距離N端80個以内、7〇個以内、個以 内、50個以内、40個以内、3〇個以内、2〇個以内' μ個以 内、18個以内、17個以内、16個以内、15個以内' μ個以 内、13個以内、12個以内、u個以内、1〇個以内、9個以 内、8個以内、7個以内、6個以内、5個以内、4個以内、3 個以内或2個以内胺基酸殘基。 可藉由相關技術中已知之方法’混合或組合本發明調配 物與其他醫藥上可接受載劑或媒劑,使其適於注射。其中 一些用於調配本發明產品之醫藥上可接受載劑為生理食鹽 水、人類金清白蛋白、人類血毁蛋白質等。本發明亦關於 150941.doc -32· 201138831 :種醫藥Μ合物,其包含如上所述之 接受之賦形劑及/或载劑。 樂上了 # -¾ ik li W ’人 樂上可接受載劑可為水 液、懸浮液、及乳液。非水性溶液實例為丙 (;Γ二二醇、植物油(諸如橄欖油)、及注射用有機酯 :水:载劑包括水、醇溶液/水溶液、乳液 匕生理食鹽水及經緩衝之介質。非經腸媒劑 鋼溶液、林格氏葡萄糖Wnger.s deXtrose)、葡萄 t與氣化納、乳酸化林格氏溶液⑽⑽Ringer,s)或不揮 U 生油。經靜脈内用媒劑包括流體及營養補充液、電解質 補充液(諸如彼等以林格式葡萄糖為底質之補充液)等。亦 可存在防腐劑或其他添加劑,諸如例如抗微生物劑、抗氧 化劑、螯合劑、惰性氣體等。 醫藥組合物包含有效量之本發明聚合物-蛋自質接合 物、及醫藥上可接受之稀釋劑、防腐劑、助溶劑、乳化 ^佐^及/或載劑。該等組合物包括各種不同緩衝内容 物之稀釋液(諸如Tris_Hci緩誠、乙酸鹽緩衝液、碟酸鹽 緩衝液、PH緩衝液及離子強度緩衝液);添加劑(諸如清潔 劑及助溶劑,諸如Tween 8〇、聚山梨醇酯8〇)、抗氧化劑 (諸如抗壞血酸及偏亞硫酸氫鈉)、防腐劑(諸如苯甲醇)及 填充物質(諸如乳糖或甘露醇);並使材料併入聚合性化合 物之顆粒製劑(諸如聚乳酸、聚乙醇酸等),或併入脂質 體。該等組合物可影響根據本發明之聚合物蛋白質接合物 的物理狀態、安定性、活體内釋放速率及活體内清除率。 可藉由相關技術中已知之方法,調配根據本發明製得之 150941.doc •33- 201138831 蛋白f接合物與醫藥上可接受之載劑或媒劑,形成適於注 ^ 藥,,且合物。參見例如 W097/09996、W097/40850、 8660、及w〇99/〇74〇1。可於7之含有例如 mM氣化鈉之滲透劑的1〇 mM磷酸鈉/磷酸鉀緩衝液中調配 本,:月化合物。醫藥組合物可視需要包含防腐劑。 醫藥組合物實質上包含接合物、含於適於保持溶液pH處 於’勺5.5至約7〇範圍内之醫藥上可接受緩衝液中之多電荷 '”、機陰離子、及視需要選用之—種或多種醫藥上可接受之 載劑及/或賦形劑。 使用方法 於本發明之另一態樣中,提供一種使接受者體内之白細 肊數增加的方法’包括對有需要之接受者投與本發明醫藥 調配物。於某些實施例中,接受者為人類。於某些實施例 中,接受者罹患嗜中性球減少症或處於罹患風險中。於某 些其他實施例中’接受者正接受會使其白血球數減少之藥 d之⑺療。於某些貫施例中,接受者之内源性濃度 降低。於某些其他實施例中,接受者正接受輻射治療。接 爻者正罹患肺癌、淋巴瘤、乳癌、骨髓移植、睾丸癌、與 AIDS相關之惡性腫瘤、骨髓造血不良症、急性白血病、 先天性及週期性嗜中性球減少症或再生障礙性貧血(參見 Mortsyn,等人(1998)Filgrastim(r_metHuG_CSF)。於」臨床 操作(Clinical Practice)」,第二版,Marcel Dekker公司,Willebrand factor), protein c, atrial natriuretic factor, pulmonary surfactant protein, bombesin, prothrombin, enkephalinase, mullerian-inhibiting agent, pubic relaxin A chain, pubic relaxation Hormone B chain, pubic flaccid relaxin, deoxyribonuclease, inhibin, activin, vascular epidermal growth factor, integrin, protein VIII or D, bone-derived neurotrophic factor (BDNF), neurotrophin 3, _4, -5, or _6 (NT-3, NT-4, NT-5, or NT-6), CD-3, CD-4, CD-8, CD-19, M-CSF, GM-CSF, G - CSF, a biologically active fragment of any of the foregoing, or a combination of the foregoing. In order to maintain the pH in a preferred range, the reaction can be carried out in the presence of a buffer. For example, the buffer may be selected from the group consisting of phosphate buffer, acetate buffer, 150941.doc • 27·201138831 HEPES, MES, or other similar buffer. The reaction buffer may typically be a standard buffer containing no amine component, such as phosphate buffered saline (PBS). The reaction buffer typically comprises a salt (e.g., a Na salt) at a concentration of from about 0.1 mM to about 100 mM, most typically from about 1 mM to about 50 mM, and more typically from about 10 mM to about 20 mM. In certain embodiments, the polypeptide is mixed with the dried activated water soluble polymer with agitation. The pH of the reaction buffer is typically from about 6.5 to about 8.5 or from about 6.6 to about 7.5. In certain embodiments, the reaction buffer has a neutral pH of about 7.0. The reaction buffer additionally contains an organic solvent, particularly dimethyl sulfoxide ("DMSO"). DMSO can be present in the reaction mixture at a concentration of 5-80%, usually 10-40% (v/v). DMSO is widely used as a general-purpose solvent, but it is generally not used to affect the pegylation reaction, and is not particularly useful for preferentially affecting the reaction away from the N-terminal PEGylation site of the protein. β ρ〇τ disclosure case w〇08/0192 14 A method for selectively preparing a modified erythropoietin in a dMS containing buffer. The PEG chain provides a selective attachment of the amine-based ester to the PEG-EPO at the N-terminus of the protein. Conversely, the reaction conditions described herein, including the use of aldehyde-bonded PE^, appear to specifically drive the covalent attachment of activated pEG to a particular lysine site. Furthermore, it has been found that the addition of DMSO to the reaction buffer can be altered. Position of PEGylation 1. In particular, the addition of _ to the reaction buffer will drive the reaction toward the preferential concentration of polyethylene glycol 2 in the protein. Thus, the method of the present invention can selectively modify the amino acid residues in the related protein, particularly the lyophilic acid residues near the terminal end of the modified white amine. Such selective modification of such proteins is beneficial because it is generally known that two water-soluble polymers (e.g., PEG) are linked to proteins via amine groups, resulting in loss of activity. The loss of activity normally associated with PEGylation is due to the previously targeted random targeted amide acid reaction. Random modification of the lysine residue may adversely alter protein function due to substantially altering the tertiary structure or morphology of the protein. While not wishing to be bound by any theory in any way, the reaction conditions disclosed herein appear to selectively modify the protein at the selected residue or at the amino terminus (generally believed to not contribute to protein activity). Specific pegylation of proteins with aldehyde activated PEG has previously been reported. However, this kind of reaction is only low? 11 specific, but lost specificity at pH 7 or above. The <amp;> method is particularly useful for PEGylation and/or ligation of pH sensitive proteins. The temperature at which the reaction is carried out is usually up to 5 Torr, and more usually the temperature is 2. 〇 to 8 ° C, and most usually about 4 t: ^ The choice of a specific temperature can affect the reaction time, the selected temperature is usually prepared by the polymer · protein connection: should use the amine chain and polymer units The two amine-based gas atoms of the joined protein f. In the reactions described herein, the activated water soluble polymer is present in excess of the molar amount ' and thus will require removal of unreacted excess activated water soluble polymer from the newly formed protein conjugate. As used herein, "removing substantially all of the unlinked water soluble polymer" refers to a generally known method for carrying out such separation methods, such as removal of about 80% unlinked water solubility by dialysis. The polymer, typically removed by about 90%, more typically by about 95%, and most typically by about 99%. Illustrative, 'Pharmaceutical Formulations 150941.doc -29- 201138831 In certain embodiments, the present invention relates to a pharmaceutical formulation comprising at least one class of G-CSF proteins, wherein each G-CSF molecule is associated with at least one The covalent linkage of the ethylene glycol molecule is optionally included in a pharmaceutically acceptable carrier. In certain embodiments, the carrier is substantially free of protein. In certain embodiments, the pharmaceutical formulation comprises at least one class of G-CSF proteins, wherein each G-CSF molecule is covalently linked to at least one polyethylene glycol molecule, and at least 30% of the composition is not at the n-terminus It is pegylated. In a particular embodiment, 'each G-CSF molecule is covalently linked to a single polyethylene glycol molecule ("mono-PEGylated G-CSF"), and at least 30% of the composition is not N The end is PEGylated. In another embodiment, the formulation comprises at least 80% monoPEGylated g-CSF, wherein at least 30% of the composition is not PEGylated at the N-terminus. In other embodiments, the formulation comprises at least 85°/. , at least 90% or at least 95°/. The mono-PEGylated G-CSF molecule' wherein at least 3% of the composition is not polyethylene glycolated at the n-terminus. In a particular embodiment of the pharmaceutical formulation, each G-CSF molecule is covalently linked to at least one polyethylene glycol molecule via a particular lysine residue. In certain embodiments, the formulation comprises at least one G-CSF molecule covalently bonded to at least one polyethylene glycol molecule via an amine linkage at the amine end of the G-CSF protein. In other embodiments, the formulation comprises at least one G_CSF molecule 'covalently linked to at least one polyethylene glycol molecule through Lysl6 (or Lys 17 if the n-terminal methionine residue is calculated). In some embodiments of the formulation, the ratio of the PEGylated molecule at the N-terminus to the second group (especially the second group of PEGylated at Lysl6) 150941.doc 201138831 is less than about 1 More than about 100, about 10 to about 90, about 20 to about 80, about 3 to about 7, about 4 to about 60, about 50, about 60 to about 40, about 7 to about 3, Less than about 1 in the sputum includes quantities that are not measurable using standard methods known in the related art. It is intended that the present & G GSF molecules and compositions exhibit long-term safety lines under standard storage conditions, i.e., at standard temperatures (e.g., about 25. 〇 for example, at least three months of storage. In some embodiments, The protein-containing carrier is serum-free, albumin-free or free of human serum albumin ("hsa-free"). In some embodiments, the pharmaceutical formulation can be stored for a longer period of time, and as herein The method described and/or the method known in the related art determines that there is no substantial and/or measurable degradation of G_CSF. In some embodiments, it is determined after storage for at least 15 months at about -2 Torr or 4 Torr. The pharmaceutical formulation of the present invention = stability (i.e., does not exhibit measurable degradation and/or does not show, and degradation in the periplasm). In other embodiments +, such as about Μ. The pharmaceutical formulation of the present invention has stability (i.e., does not exhibit measurable degradation and/or does not exhibit substantial degradation) after storage at about 37 ° C for at least 10 months. Any method known in the art for determining the stability of the formulation of the j drug of the present invention. In one embodiment, the method for determining the stability of a pharmaceutical formulation of the present invention is to monitor the change in protein concentration over time, such as by bisquinolinecarboxylic acid ("bca") protein analysis. In other embodiments, 'inventive f The method for determining the stability of a pharmaceutical formulation is as follows: The change in protein degradation (i.e., g_csf conjugate degradation) over time is determined by SDS PAGE analysis. In some other embodiments, the stability can be determined by: The degradation of the product is analyzed by HpLC. In other examples 150941.doc 31 201138831, the stability of the pharmaceutical formulation of the present invention is determined by monitoring the activity of the formulation over time, wherein the activity is determined by the related art. Any in vitro or in vivo method for determining the activity of the formulation (e.g., G-CSF activity) is known. In a particular example according to this embodiment, the invention comprises a plurality of G-CSF-conjugates. The method for assessing the activity of a formulation is to assess the ability of the pharmaceutical formulation in an in vitro biological assay using a G-CSF-dependent murine 32D cell line. In an embodiment, the assay activity is determined by measuring its ability to alter the number of blood cells in an experimental animal, such as a hamster, in vivo. In certain embodiments, a pharmaceutical formulation comprising a monomeric ethylene is provided. Alcoholized protein, wherein at least 3% of the protein molecules are not PEGylated at the N-terminus. In certain particular embodiments, the formulation comprises at least a PEGylated protein that is ligated to The lyophilic acid residue is PEGylated from within the amino acid. In some embodiments, the lysine is within 80, less than 7, within, within 50, within the N-terminus, 40 or less, 3〇 or less, 2〇 or less, within , within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within, within Within 1 、, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 internal amino acid residues. The formulations of the present invention can be mixed or combined with other pharmaceutically acceptable carriers or vehicles by methods known in the art to render them suitable for injection. Some of the pharmaceutically acceptable carriers for formulating the products of the present invention are physiological saline, human gold albumin, human blood-damaged proteins and the like. The present invention also relates to a pharmaceutical composition comprising 150941.doc-32·201138831 comprising an excipient and/or a carrier as described above. Happy to be # -3⁄4 ik li W ‘People The acceptable carrier can be water, suspension, and lotion. Examples of non-aqueous solutions are C (?? didiol, vegetable oil (such as olive oil), and organic ester for injection: water: carrier includes water, alcohol solution / aqueous solution, emulsion, physiological saline and buffered medium. Enteral vehicle steel solution, Ringer's dextrose Wnger.s deXtrose), grape t and gasified sodium, lactated Ringer's solution (10) (10) Ringer, s) or no U oil. The intravenous vehicle includes a fluid and a nutrient replenisher, an electrolyte replenisher (such as a supplemental solution in which the forest format is used as a substrate), and the like. Preservatives or other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, and the like. The pharmaceutical compositions comprise an effective amount of a polymer-egg self-contained conjugate of the invention, and a pharmaceutically acceptable diluent, preservative, co-solvent, emulsifying agent and/or carrier. Such compositions include dilutions of various buffer contents (such as Tris_Hci, acetate buffer, discate buffer, pH buffer, and ionic strength buffer); additives such as detergents and co-solvents, such as Tween 8〇, polysorbate 8〇), antioxidants (such as ascorbic acid and sodium metabisulfite), preservatives (such as benzyl alcohol) and fillers (such as lactose or mannitol); and the incorporation of materials into the polymerizability A granular preparation of the compound (such as polylactic acid, polyglycolic acid, etc.), or incorporated into a liposome. Such compositions can affect the physical state, stability, in vivo release rate, and in vivo clearance of the polymer protein conjugates according to the present invention. The 150941.doc •33-201138831 protein f-conjugate and the pharmaceutically acceptable carrier or vehicle prepared according to the present invention may be formulated by a method known in the related art to form a drug suitable for injection, and Things. See, for example, W097/09996, W097/40850, 8660, and w〇99/〇74〇1. The compound can be formulated in a 1 mM sodium phosphate/potassium phosphate buffer containing 7, for example, mM sodium hydride penetrant. The pharmaceutical composition may contain a preservative as needed. The pharmaceutical composition substantially comprises a conjugate, a multi-charge contained in a pharmaceutically acceptable buffer suitable for maintaining a solution pH in the range of 5.5 to about 7 Å, a machine anion, and optionally Or a plurality of pharmaceutically acceptable carriers and/or excipients. Method of Use In another aspect of the invention, a method of increasing the number of white fines in a recipient is provided 'including acceptance of need The pharmaceutical formulation of the invention is administered. In certain embodiments, the recipient is a human. In certain embodiments, the recipient suffers from or is at risk of developing neutropenia. In certain other embodiments The recipient is receiving a drug that reduces the number of white blood cells (7). In some embodiments, the endogenous concentration of the recipient is reduced. In some other embodiments, the recipient is receiving radiation therapy. The recipient is suffering from lung cancer, lymphoma, breast cancer, bone marrow transplantation, testicular cancer, malignant tumor associated with AIDS, bone marrow hematopoietic disorder, acute leukemia, congenital and periodic neutropenia or aplastic anemia See Mortsyn, et al. (1998) Filgrastim (r_metHuG_CSF). In "clinical practice (Clinical Practice)," second edition, Marcel Dekker company,

New York,NY)中。於某些實施例中,向處於感染風險中 之患者投與本發明調配物。 150941.doc •34· 201138831 於某些實施例申, _ 、化予療法過程中提供呈單一劑量之 調配物。於一也訾奸办丨士 量之^ 於化學療法難中投與呈多劑 2 於某些實施财,調配物之投與頻率為每日 人母週一次、每兩週-次或每月-次。調配物可於一 劑化學痒法-4* m + '、 、日,内知·與。於某些實施例中’調配物 係於進行-劑化學療法之前至少14天投與。 於某些實施例,’調配物係呈注射液投與。於一些實施 例卜調配物適於經皮下投與。於其他實施例中,調配物 適^經靜脈内投與。亦可提供口服形式之調配物,且至少 約每週-次接H於其他實施财,患者為至少約每 兩週一次、至少約每三週一次、或至少約每月一次接受一 劑。 治療有效量為可產生導致骨髓細胞增加產生白血球之活 to·内生物活性所必須之接合物數量。接合物之確切數量需 要參照如下因素:諸如待治療之病症之確切種類、待治療 患者的病症、及組合物中之其他成份。所調配之含接合物 之"J·藥s周配物強度應可有效藉由多種方式投與正經歷特徵 為白血球產生量低或產生缺陷之病症的人類患者。接合物 之平均治療有效數量可能不同,且特定言之應基於有資格 之醫生之建議及處方。例如,可例如於每一化學療法週 期’投與一次每公斤體重〇.〇丨至10 pg,通常每公斤體重 〇. 1至3 pg。或者’本發明醫藥組合物可含有固定劑量之接 合物,例如於適用於45 kg以上之接受者的固定劑量調配 物中為1至10 mg ’或2至9或約6 mg。然而,熟習此項技術 150941.doc 35· 201138831 者咸明瞭,含本發明接合物之醫藥組合物之調配強度應可 有效藉由多種方式投與正經歷特徵為白血球產生量低或產 生缺陷之病症的人類患者。接合物之平均治療有效數量可 能不同,且應基於有資格之醫生的建議及處方。 實例 實例1 :利用醛-PEG(30K),於DMSO存在下產生不同 PEG-GCSF(d-PEG-GCSF)之製程。 取經單曱氧基化之PEG(分子重量30,000道爾頓)活化為 醛化物(Ald-PEG-30K),並用作翠乙二醇化試劑。使用於 大腸桿菌中產生之GCSF作為多肽。由GCSF-蛋白質(7 mg) 於含15% DMSO、10 mM乙酸鈉-MES緩衝液,pH 5之反應 緩衝液中製成濃度0.37 mg/ml。於攪拌下,使多肽與PEG 試劑(含於0· 5 mM HCL中之30 mg/mL溶液)混合,達到最終 之PEG/GCSF莫耳比為16 : 1。添加NaCNBH3,並於4°C 下,維持濃度22 mM達24小時。純化反應產物,並於10 mM乙酸鈉緩衝液中,於pH 4下,以SP-管柱層析術去除未 反應之PEG分子。藉由如下分步梯度溶離出結合之經聚乙 二醇化之分子:(1)50 mM NaCl,pH 4 ; (2)25 mM NaCl, pH 5 ; (3)40 mM NaCl,pH 5.5 ; (4)70 mM NaCl,pH 5.5 ;及(5)50 mM NaCl,pH6.5。於第一步與第二步期間 溶離出經聚乙二醇化之GCSF產物。圖3顯示純化後樣本的 PAGE(聚丙烯醯胺凝膠電泳)圖像,其係呈單一條帶之物 質。 於本發明方法中使用DMSO產生單一種經單聚乙二醇化 150941.doc •36· 201138831 之產物’其總分子重量為約80 KD。於交聯丙烯醯胺中之 直鏈30K PEG的流體動力學大小為約60 kD’且球形GCSF 蛋白質之分子重量為19 kD。不採用DMSO時所產生之類似 產物亦顯示為具相同分子重量之單一物質種類(參見圖4)。 實例中之縮寫’ d-PEG-GCSF係指採用包含DMSO之方法, 及酸-PEG(3 0K)所製得之產物。且NeulastaTM係指採用不包 含DMSO之方法’及醛-PEG(2〇k)所製得之產物。Neulasta 為Amgen的商標名稱。 實例2 :利用兩個樣本(DM_82〇5及82〇1ICi6組),說明d_ PEG-GCSF等同於Neu丨asta之活體外研究。 利用實例1所示之方法製得兩個樣本(DM8205組及 8210K16組)。利用需要GCSF用於生長之細胞株,於細胞 增殖分析中比較d-PEG-GCSF相對於對照組peg-GCSF(於習 知方法中製得,名稱為Neulasta®,組編號:p〇8〇〇98)之生 物活性。 於微孔分析板中,使組織培養細胞NFS-60呈單層生長於 由含於RPMI培養基中之1〇% FBS(胎牛血清)及1%青黴素_ 鏈黴素所組成之培養基中。使固定數量之細胞(每孔1〇,〇〇〇 個)與各種不同濃度之測試化合物接觸,並由接觸到藥物 之細胞生長情形與僅含培養基之各孔中之細胞生長情形比 較。於5% C〇2之潮濕培養箱中,於3rc下,培養72小時之 後,使細胞與含有5 mg/ml Μττ(*)之培養基接觸 ,並於相 同土。養和中再培養4.5小時。藉由添加經酸化之25% SDS水 溶液’中止細胞對MTT之代謝作用。分析板留置於室溫 150941.doc -37· 201138831 下,使細胞溶解。溫和振盪溶解之材料5分鐘,使其均質 化,並於設定於波長570 nm下之分光光度計中讀出各孔中 内容物的吸光值。以吸光度OD相對於細胞所接觸對應測 試化合物之濃度作圖。(圖5)。確定ED5〇劑量。結果示於表 1中。活體外活性之一單位係基於ED5〇=0.01 ng/ml之 Neupogen活性 0 統計學分析顯示,觀察到之差異具有大於95%可信度 (P<0.001)之顯著性。數據說明,8201K16組之生物活性係 與Neulasta相當,而DM8205之生物活性優於Neulasta。 表1 :藉由MTT分析之d-PEG-GCSF與Neulasta於刺激 NFS-60細胞生長時之ED50濃度。(*)MTT為3-(4,5-二甲基-2-噻唑基)-2,5-二苯基-2H-四唑鏽溴化物之縮寫;其係獲自 Sigma(MO,目錄號M2128)。 樣本 ED5〇(ng/mL) 對照(Neulasta) 0.0177 DM8205 0.0163 8201K16 0.0175 Neupogen 0.01 實例 3 :說明 d-PEG-GCSF(lPN7019組)等同於Neulasta之 活體外研究。 依實例2所述之相同方式,於細胞增殖分析中測試第二 組d-PEG-GCSF(lPN7019)之活體外活性。結果示於圖6 中。PEG-GCSF(7019 組)之 ED5〇 濃度為 Neulasta之 ED50濃度 150941.doc -38 - 201138831 的99% »因此,之活性相當;其差異 屬於統計學誤差範圍内。 實例4 :於活體外及於活體内比較d_PI:G-GCSF(lPN7606 組)與 Neulasta。 藉由包含DMSO之方法(Prol〇ng方法)製得—組d_pEG_ GCSF(1PN7606)。採用與上述實例中所出示方法相同之方 法,於活體外NFS-60細胞增殖分析中比較該等樣本。數據 示於圖7中。比較ED50濃度,7606組之ED5(^Neuiasta之 97%。因此,d-PEG-GCSF(lPN7606組)之活體外活性稍優 於Neulasta。該等樣本係於進行測試前3個月以上製備。 隨後於活體内分析中比較兩種化合物刺激嗜中性白血球 絕對數量(ANC)的生物活性。於該分析中,於第〇天, C57BL/6N小鼠接受注射單一劑量1〇〇 mg/kg之環磷醯胺 (CPA) ’以誘發嗜中性球減少症。隨後,於投與cpA之後 24小時,以d-PEG-GCSF(測試)或Neulasta(對照)處理小 鼠。於隨後連續4天内,每天每隔24小時投與測試物或對 照物之注射液。於第〇、3〇、54、78、1〇2、及126小時採 集血液樣本。採用零時間點時之樣本作為基線讀數。每一 處理組有1〇隻動物。利用獸用細胞計數計分析血液樣本, • 以估算WBC(白血球)總數。檢查周邊血液抹片,以估算分 類細胞數。利用血液中之嗜中性白血球百分比計算嗜中性 白血球絕對數量(ANC)。利用梯形法則’計算各動物之 ANC相對於時間之曲線的曲線下面積(AUC)。以AUC數據 相對於劑量作圖,如圖8所示。 I50941.doc -39- 201138831New York, NY). In certain embodiments, a formulation of the invention is administered to a patient at risk of infection. 150941.doc • 34· 201138831 In certain embodiments, _, chemotherapeutic therapy provides a single dose of the formulation. In the case of Yu Yi, the number of gents is too high. In the case of chemotherapy, it is difficult to invest in multiple doses. 2 In some implementations, the frequency of the formulation is once a week, every two weeks, once or every month. - times. The formulation can be used in a chemical itch method - 4* m + ', , day, and inside. In certain embodiments, the formulation is administered at least 14 days prior to the on-agent chemotherapy. In certain embodiments, the formulation is administered as an injectable solution. In some embodiments, the formulation is suitable for subcutaneous administration. In other embodiments, the formulation is administered intravenously. Formulations in oral form may also be provided, and at least about weekly to other times, the patient receives at least once every two weeks, at least about once every three weeks, or at least about once a month. A therapeutically effective amount is one that produces the amount of conjugate necessary to cause an increase in the production of white blood cells by the bone marrow cells. The exact number of conjugates needs to be referenced to factors such as the exact species of condition to be treated, the condition of the patient to be treated, and other ingredients in the composition. The formulated conjugated "J</RTI> s weekly ligand strength should be effective in administering a human patient who is experiencing a condition characterized by low white blood cell production or defects. The average therapeutically effective amount of the conjugate may vary and is specifically based on the advice and prescription of a qualified physician. For example, it may be administered, for example, once per kilogram of body weight per gram of body weight to 10 pg, typically 1 to 3 pg per kilogram of body weight. Alternatively, the pharmaceutical compositions of the present invention may contain a fixed dose of the compound, for example, from 1 to 10 mg' or from 2 to 9 or about 6 mg in a fixed dose formulation suitable for recipients above 45 kg. However, it is well known that the formulation of the pharmaceutical composition comprising the conjugate of the present invention should be effective in administering a variety of ways to treat a condition characterized by low white blood cell production or defects. Human patient. The average therapeutically effective amount of the conjugate may vary and should be based on the advice and prescription of a qualified physician. EXAMPLES Example 1: A process for producing different PEG-GCSF (d-PEG-GCSF) in the presence of DMSO using aldehyde-PEG (30K). The monoethoxylated PEG (molecular weight 30,000 Daltons) was activated to the aldehyde compound (Ald-PEG-30K) and used as a acetylation reagent. The GCSF produced in E. coli is used as a polypeptide. A concentration of 0.37 mg/ml was prepared from GCSF-protein (7 mg) in a reaction buffer containing 15% DMSO, 10 mM sodium acetate-MES buffer, pH 5. The polypeptide was mixed with PEG reagent (30 mg/mL solution in 0.5 mM HCL) with stirring to achieve a final PEG/GCSF molar ratio of 16:1. NaCNBH3 was added and maintained at a concentration of 22 mM for 24 hours at 4 °C. The reaction product was purified, and unreacted PEG molecules were removed by SP-column chromatography at pH 4 in 10 mM sodium acetate buffer. The bound PEGylated molecule is eluted by a stepwise gradient of: (1) 50 mM NaCl, pH 4; (2) 25 mM NaCl, pH 5; (3) 40 mM NaCl, pH 5.5; 70 mM NaCl, pH 5.5; and (5) 50 mM NaCl, pH 6.5. The PEGylated GCSF product is dissolved during the first and second steps. Figure 3 shows a PAGE (polyacrylamide gel electrophoresis) image of the purified sample, which is a single band of material. The use of DMSO in the process of the invention produces a single mono-pegylated product of 150941.doc • 36·201138831 having a total molecular weight of about 80 KD. The linear 30K PEG in the cross-linked acrylamide has a hydrodynamic size of about 60 kD' and the spherical GCSF protein has a molecular weight of 19 kD. Similar products produced without DMSO were also shown as a single species with the same molecular weight (see Figure 4). The abbreviation 'd-PEG-GCSF' in the examples refers to a product obtained by a method comprising DMSO and acid-PEG (30K). And NeulastaTM refers to a product obtained by a method which does not contain DMSO and aldehyde-PEG (2〇k). Neulasta is the trade name for Amgen. Example 2: Two samples (DM_82〇5 and 82〇1 ICi6 groups) were used to demonstrate that d_PEG-GCSF is equivalent to the in vitro study of Neu丨asta. Two samples (DM8205 group and 8210K16 group) were prepared by the method shown in Example 1. Using cell strains that require GCSF for growth, d-PEG-GCSF was compared to the control group peg-GCSF in a cell proliferation assay (obtained in a conventional method, the name is Neulasta®, group number: p〇8〇〇) 98) Biological activity. Tissue culture cells NFS-60 were monolayer grown in a medium consisting of 1% FBS (fetal calf serum) and 1% penicillin-streptomycin contained in RPMI medium in a microwell assay plate. A fixed number of cells (one per well, one per well) were contacted with various concentrations of test compound and compared to the growth of cells in the wells containing only the medium in the case of cell growth in contact with the drug. After incubation for 72 hours at 3 rc in a humidified incubator at 5% C 2 , the cells were contacted with a medium containing 5 mg/ml of Μττ (*) and in the same soil. The culture was further incubated for 4.5 hours. The metabolism of MTT was stopped by the addition of acidified 25% SDS aqueous solution. The assay plate was left at room temperature 150941.doc -37·201138831 to allow the cells to dissolve. The dissolved material was gently shaken for 5 minutes to homogenize, and the absorbance of the contents in each well was read in a spectrophotometer set at a wavelength of 570 nm. The absorbance OD is plotted against the concentration of the corresponding test compound to which the cells are exposed. (Figure 5). Determine the ED5 〇 dose. The results are shown in Table 1. One unit of in vitro activity was based on ED5 〇 = 0.01 ng/ml of Neupogen activity. 0 Statistical analysis showed that the difference observed was greater than 95% confidence (P < 0.001) significance. The data indicates that the biological activity of the 8201K16 group is comparable to that of Neulasta, while the biological activity of DM8205 is superior to that of Neulasta. Table 1: ED50 concentration of d-PEG-GCSF and Neulasta stimulated by mM-well cells by MTT assay. (*) MTT is an abbreviation for 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazole rust bromide; it is obtained from Sigma (MO, catalog number M2128). Sample ED5〇 (ng/mL) Control (Neulasta) 0.0177 DM8205 0.0163 8201K16 0.0175 Neupogen 0.01 Example 3: Description d-PEG-GCSF (group 1PN7019) is equivalent to an in vitro study of Neulasta. The in vitro activity of the second group of d-PEG-GCSF (lPN7019) was tested in a cell proliferation assay in the same manner as described in Example 2. The results are shown in Figure 6. The ED5〇 concentration of PEG-GCSF (Group 7019) was 99% of the ED50 concentration of Neulasta 150941.doc -38 - 201138831 » Therefore, the activity was comparable; the difference was within the statistical error range. Example 4: d_PI: G-GCSF (group 1PN7606) and Neulasta were compared in vitro and in vivo. The group d_pEG_GCSF (1PN7606) was prepared by a method comprising DMSO (Prol〇ng method). The samples were compared in an in vitro NFS-60 cell proliferation assay using the same method as the method presented in the above examples. The data is shown in Figure 7. The ED50 concentration was compared with the ED5 of the 7606 group (97% of the Neuaasta. Therefore, the in vitro activity of the d-PEG-GCSF (group 1PN7606) was slightly better than that of Neulasta. These samples were prepared more than 3 months before the test. The in vivo assay was used to compare the biological activity of two compounds to stimulate the absolute number of neutrophils (ANC). In this analysis, on day ,, C57BL/6N mice received a single dose of 1 〇〇mg/kg ring. Phospholipid (CPA)' to induce neutropenia. Subsequently, mice were treated with d-PEG-GCSF (test) or Neulasta (control) 24 hours after administration of cpA. For the next 4 consecutive days, The test substance or control injection was administered every 24 hours every day. Blood samples were taken at 〇, 3〇, 54, 78, 1, 2, and 126 hours. Samples at zero time point were used as baseline readings. One treatment group has 1 animal. The blood sample is analyzed using a veterinary cytometer, • to estimate the total number of WBCs (white blood cells). Peripheral blood smears are examined to estimate the number of cells sorted. The percentage of neutrophils in the blood is calculated. Absolute neutrophils (ANC). Trapezoidal rule 'calculating each ANC animals relative to the area under the curve of the time curve (AUC). AUC to dose plotted with respect to the data, as shown in FIG. I50941.doc -39- 201138831

數據顯示’在治療期間,d_PEG_GCSF及Neulasta之AUC 皆南於基線值。d-peg-GCSF(lPN7606)之平均值看起來較 佳’然而’於該實例中,測試化合物與Neulasta具有類似 之活體内效力。 實例5:聚乙二醇化位置比較 檢查於先前實例中所研究之樣本的聚乙二醇化位置。利 用蛋白酶V8消化樣本,並如相關技術中所熟知,利用標準 方法,藉由HPLC分離出含PEG之片段。隨後如相關技術 中所熟知’採用愛德曼降解法(Edman Degradation method),測定經聚乙二醇化之片段的序列。取樣本添加 至Prosorb 膜,並利用購自Applied Biosystem公司之494The data shows that during treatment, the AUC of d_PEG_GCSF and Neulasta are both above the baseline value. The average of d-peg-GCSF (lPN7606) appears to be better 'however' in this example, the test compound has similar in vivo efficacy as Neulasta. Example 5: Comparison of PEGylation positions The PEGylation positions of the samples studied in the previous examples were examined. The sample is digested with protease V8 and the PEG-containing fragment is separated by HPLC using standard methods as is well known in the art. The sequence of the PEGylated fragment is then determined using the Edman Degradation method as is well known in the art. The sample was added to the Prosorb membrane and utilized from 494 from Applied Biosystem.

Procise protein Sequencer蛋白質定序儀/140C分析儀測定 其序列》由所得序列與理論序列比對。發現自N端至第19 位殘基之片段含有PEG。因此,PEG僅可與N端(α胺)或離 胺酸16接合《進一步藉由胰凝乳蛋白酶消化該片段。若測 得如XXLE或LXXLE之序列,則確定聚乙二醇化位點為Ν 端。若未測得離胺酸,或測得其濃度比亮胺酸低得多,則 確定聚乙二醇化位點為Lys-16。請注意,吾人所計算之第 1唬序列位點為Thr。若吾人所計算之第丨號位點為Met,則 離胺酸16應稱為離胺酸17。結果概括於下表2中。 150941.doc -40- 201138831 表2 : d-peg_GCVSF相對於NeiHasta之序列分配 產物中各群類之百分比 與 Neulasta 相比 樣本組 PEG-Ald DMSO N端 離胺酸-16 活性 DM8205 30K 有 52% 48% 更佳 8201K16 3 OK 有 33% 67% 等同 1PN7019 30K 混合 18% 75% 等同 1PN7606 3 OK 有 42% 58% 等同或更佳 Neulasta 20K 無 100% 0% 等同 實例6 :測定活體外活性。 G-CSF活體外生物學分析法為利用G-CSF依賴性鼠32D細 胞株之有絲分裂分析法。細胞保存於含有5% FBS及20 ng/ml G-CSF之Iscoves培養基中。於添加樣本之前,以缺 少G-CSF之生長培養基兩次沖洗,製得細胞。製備擴展之 十二個點G-CSF標準曲線,其係自48至0.5 ng/ml(等同於 4800-5 0 IU/ml)。依落在標準曲線直線部份之估算,製備 各樣本之四種稀釋液(1000至3000 IU/ml),並重複三次。 可將經聚乙二醇化之G-CSF樣本稀釋成未經聚乙二醇化之 G-CSF的約4分之一至10分之一。向含10,000個細胞/孔之 96孔微滴定分析板中之適宜孔添加體積為40 μΐ之樣本之各 稀釋液或標準液。經於37°C及5.5% C02下培養四十八小時 之後,向各孔添加0.5 pmCi曱基-3H-胸苷。經十八小時之 後,收集分析板並計數。產生劑量效應曲線(G-CSF濃度 150941.doc 41 201138831 log值相對於背景CPM) ’並對屬於標準曲線直線部份的點 進行線性回歸分析。利用所得之線性等式,並利用稀釋倍 數校正,確定未知測試樣本之濃度。 實例7 :測定活體内活性。 藉由對雄性金色倉鼠單次經皮下注射〇」mg/kg之樣本, 進行活體内測試。對各動物進行尾端採血,且於採集樣本 之同一天,對血清樣本進行完全血液計數。計算白血球數 平均值。根據CRC標準數學表格,第^版⑺町% w. h.編 輯)CRC出版公司,Boca Rat〇n,Fla 1981第125頁,計算 絰單次經皮下注射後之曲線下方之淨WBC平均值面積。 實例8 :安定性研究。 可如利用SEC-HPLC所觀察,由產物降解來評定安定 性。於4t下,於兩種PH值下(pH 4.0及pH 6.0),研究經聚 乙二醇化G-CSF長達16天。使pH升高至6 〇可提供加快安定 性分析之環境。對於ρΗ 6.0之樣本,取如上所述製得之經 單聚乙二醇化之G-CSF置於含有2〇 mM磷酸鈉、5 mM乙酸 鈉、2.5%甘露醇、〇.〇〇5%1[^刪_8〇之緩衝液_6〇) 中使最終蛋白質濃度為0.25 mg/ml ^取1 ml等分試樣貯 藏於3 mi無菌注射玻璃小瓶中。各玻璃小瓶係貯藏於斗七 及29 C下至多16天。藉由SEC_HpLC追蹤法評定安定性。 右隨後之測量值保持與初始(時間=〇)測量值相同(如藉由肉 眼觀察所確定),認為樣本於該時間期内安定》 【圖式簡單說明】 圖1主要之經完全加工之人類顆粒性細胞群落刺激因子 15094I.doc -42· 201138831 (「G-CSF」)的胺基酸序列(SEQ m N〇 : 〇。 圖2 κ例中所使用之G_CSF蛋白質的序列數據。 圖3對根據本發明製得之經純化之d_peg_GcsF進行 烯醯胺凝膠電泳。第1行:分子重量大小標記(以kD表 聚内示, 200、116.3、97.4、66.3、55.4、36.5、31、21.5、第2行:經純化之d_peg_GCSF ;第3行:未經 GCSF。 14.4);改質之 圖4對經不採用DMS〇,但採用樣本pEG及反應條件之現 有方法所製得之經純化之1^8_(3(:817進行聚丙烯醯胺凝膠電 泳。第1行:分子重量大小標記(以kD表示,200、116.3、 97.4、66.3 ' 55.4、36.5、31、21.5、14.4),第 2行:經純 化之peg-GCSF。 圖5(^丑〇-〇€8?與對照(仏111&31&)對則860細胞之增生致 應。 圖6d-PEG-GCSF與對照(Neulasta)對NFS60細胞之增生欵 應。 圖7d-PEG-GCSF與Neulasta對NFS60細胞之增生效應。 圖8於注射測試化合物(d_P]EG-GCSF與對照)後5天内之對 ANC之活體内分析。 150941.doc •43· 201138831 序列表 <11〇>美商普隆藥品公司 <120>經修飾之顆粒性細胞群落刺激因子(G-CSF) <130> 13939-105002 <140> 099133189 <141> 2010-09-29 <150> 61/247,389 <151> 2009-09-30 <160> 4 <170> Patentln version 3.5 <210> 1 <211> 175 <212> PRT <2]3>人類 <400> 1The Procise Protein Sequencer/140C Analyzer determines its sequence by comparing the resulting sequence to the theoretical sequence. A fragment from the N-terminus to the 19th residue was found to contain PEG. Thus, PEG can only be conjugated to the N-terminus (alphaamine) or to the amino acid 16 "further digesting the fragment by chymotrypsin. If a sequence such as XXLE or LXXLE is determined, the PEGylation site is determined to be the terminus. If no lysine is detected, or if the concentration is much lower than that of leucine, the PEGylation site is determined to be Lys-16. Please note that the first sequence sequence we calculated is Thr. If the nickname site calculated by us is Met, the lysine 16 should be referred to as lysine 17. The results are summarized in Table 2 below. 150941.doc -40- 201138831 Table 2: Percentage of groups in d-peg_GCVSF relative to NeiHasta's sequence distribution product compared to Neulasta sample group PEG-Ald DMSO N-terminal lysine-16 activity DM8205 30K 52% 48 % Better 8201K16 3 OK 33% 67% Equivalent 1PN7019 30K Mixed 18% 75% Equivalent 1PN7606 3 OK 42% 58% Equivalent or better Neulasta 20K No 100% 0% Equivalent Example 6: Determination of in vitro activity. The in vitro biological assay of G-CSF is a mitotic assay using G-CSF-dependent murine 32D cell strain. The cells were stored in Iscoves medium containing 5% FBS and 20 ng/ml G-CSF. Cells were prepared by rinsing twice with growth medium lacking G-CSF prior to sample addition. An extended twelve point G-CSF standard curve was prepared from 48 to 0.5 ng/ml (equivalent to 4800-5 0 IU/ml). Four dilutions (1000 to 3000 IU/ml) of each sample were prepared and evaluated three times in accordance with the estimation of the straight line portion of the standard curve. The PEGylated G-CSF sample can be diluted to about one-quarter to one-tenth of the unpegylated G-CSF. Each dilution or standard of a sample having a volume of 40 μM was added to a suitable well in a 96-well microtiter assay plate containing 10,000 cells/well. After incubation for 48 hours at 37 ° C and 5.5% C02, 0.5 pmCi thiol-3H-thymidine was added to each well. After 18 hours, the assay plates were collected and counted. A dose-response curve (G-CSF concentration 150941.doc 41 201138831 log value versus background CPM) was generated and a linear regression analysis was performed on the points belonging to the straight line portion of the standard curve. The concentration of the unknown test sample is determined using the linear equation obtained and using dilution factor correction. Example 7: Determination of in vivo activity. In vivo testing was performed by a single subcutaneous injection of a mg"mg/kg sample of male golden hamsters. Blood was collected from the tails of each animal, and the blood samples were subjected to complete blood count on the same day as the samples were collected. Calculate the average number of white blood cells. According to the CRC standard math table, the second edition (7) is written by the CRC Publishing Company, Boca Rat〇n, Fla 1981, page 125, to calculate the average area of the net WBC below the curve after a single subcutaneous injection. Example 8: Stability study. The stability can be assessed by product degradation as observed by SEC-HPLC. The pegylated G-CSF was studied for 16 days at 4 t under two pH values (pH 4.0 and pH 6.0). Increasing the pH to 6 〇 provides an environment for accelerated stability analysis. For the sample of ρΗ 6.0, the mono-pegylated G-CSF prepared as described above was placed in a solution containing 2 mM sodium phosphate, 5 mM sodium acetate, 2.5% mannitol, 〇.〇〇 5% 1 [ ^ Delete _8 〇 buffer _6 〇) The final protein concentration was 0.25 mg / ml ^ 1 ml aliquots were stored in 3 mi sterile injection glass vials. Each glass vial was stored at Dou and 29 C for up to 16 days. Stability was assessed by the SEC_HpLC tracking method. The subsequent measured value of the right remains the same as the initial (time = 〇) measurement (as determined by visual observation), and the sample is considered to be stable during the time period. [Simplified illustration] Figure 1 Mainly fully processed human The amino acid sequence of the granulocyte community stimulating factor 15094I.doc -42· 201138831 ("G-CSF") (SEQ m N〇: 〇. Figure 2 Sequence data of the G_CSF protein used in the κ case. Figure 3 The purified d_peg_GcsF prepared according to the present invention is subjected to eneamine gel electrophoresis. Line 1: molecular weight size labeling (indicated by kD, 200, 116.3, 97.4, 66.3, 55.4, 36.5, 31, 21.5, Line 2: purified d_peg_GCSF; line 3: without GCSF. 14.4); modified Figure 4 for purified 1^ without using DMS〇, but using the existing method of sample pEG and reaction conditions 8_(3(:817) for polyacrylamide gel electrophoresis. Line 1: molecular weight size label (in kD, 200, 116.3, 97.4, 66.3 '55.4, 36.5, 31, 21.5, 14.4), line 2 : Purified peg-GCSF. Figure 5 (^ ugly - 〇 € 8? and control (仏 111 & 31 &);) The proliferation of 860 cells. Figure 6d-PEG-GCSF and control (Neulasta) on NFS60 cells proliferation. Figure 7d-PEG-GCSF and Neulasta proliferation effect on NFS60 cells. Figure 8 in the injection In vivo analysis of ANC within 5 days after test compound (d_P]EG-GCSF and control) 150941.doc •43· 201138831 Sequence Listing <11〇>American Pune Pharmaceutical Company <120> Modified Granular cell community stimulating factor (G-CSF) <130> 13939-105002 <140> 099133189 <141> 2010-09-29 <150> 61/247,389 <151> 2009-09-30 <160> 4 <170> Patentln version 3.5 <210> 1 <211> 175 <212> PRT <2]3>Human<400>

Met Thr Pro Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu 15 10 15Met Thr Pro Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu 15 10 15

Lys Cys Leu Glu Gin Va) Arg Lys !le Gin Gly Asp Gly Ala Ala Leu 20 25 30Lys Cys Leu Glu Gin Va) Arg Lys !le Gin Gly Asp Gly Ala Ala Leu 20 25 30

Gin Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu 35 40 45Gin Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu 35 40 45

Val Leu Leu Gly His Ser Leu Gly He Pro Trp Ala Pro Leu Ser Ser 50 55 60Val Leu Leu Gly His Ser Leu Gly He Pro Trp Ala Pro Leu Ser Ser 50 55 60

Cys Pro Ser Gin Ala Leu Gin Leu Ala Gly Cys Leu Ser Gin Leu His 65 70 75 80Cys Pro Ser Gin Ala Leu Gin Leu Ala Gly Cys Leu Ser Gin Leu His 65 70 75 80

Ser Gly Leu Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly lie 85 90 95Ser Gly Leu Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly lie 85 90 95

Ser Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu G)n Leu Asp Val Ala 100 105 110Ser Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu G)n Leu Asp Val Ala 100 105 110

Asp Phe Ala Thr Thr lie Trp Gin Gin Met Glu Glu Leu Gly Met Ala 115 120 125Asp Phe Ala Thr Thr lie Trp Gin Gin Met Glu Glu Leu Gly Met Ala 115 120 125

Pro Ala Leu Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala 130 135 140Pro Ala Leu Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala 130 135 140

Phe Gin Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser 145 150 155 160Phe Gin Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser 145 150 155 160

Phe Leu Glu Val Ser Tyr Arg Val Leu Arg His Leu Ala Gin Pro 165 170 175 <210> 2 <211〉 531 <212> DNA <213>人類 150941.doc <220> 48 201138831 <221> CDS <222> (1)..(525) <400> 2 atg act cca tta ggt cct get tet tet ctg ccg caa age ttt ctg ctgPhe Leu Glu Val Ser Tyr Arg Val Leu Arg His Leu Ala Gin Pro 165 170 175 <210> 2 <211> 531 <212> DNA <213> Human 150941.doc <220> 48 201138831 <221&gt CDS <222> (1)..(525) <400> 2 atg act cca tta ggt cct get tet tet ctg ccg caa age ttt ctg ctg

Met Thr Pro Leu Gly Pro Aia》er Ser Leu Pro Gin Ser Phe Leu 1 5 H) 15 aaa tgt ctg gaa cag gtt cgt aaa ate cag ggt gac ggt get gca ctgMet Thr Pro Leu Gly Pro Aia"er Ser Leu Pro Gin Ser Phe Leu 1 5 H) 15 aaa tgt ctg gaa cag gtt cgt aaa ate cag ggt gac ggt get gca ctg

Lys Cys Leu Glu Gin Val Arg Lys lie Gin Gly Asp Gly Ala Ala Leu 20 25 30 caa gaa aaa ctg tgc get act tac aaa ctg tgc cat ccg gaa gag ctgLys Cys Leu Glu Gin Val Arg Lys lie Gin Gly Asp Gly Ala Ala Leu 20 25 30 caa gaa aaa ctg tgc get act tac aaa ctg tgc cat ccg gaa gag ctg

Gin Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Uu 35 40 45 gta ctg ctg ggt cat tet ett ggg ate ccg tgg get ccg ctg tet tetGin Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Uu 35 40 45 gta ctg ctg ggt cat tet ett ggg ate ccg tgg get ccg ctg tet tet

Val Leu Leu Gly His Ser Leu Gly lie Pro Trp Ala Pro Uu Ser Ser 50 55 60 igt cca tet caa get ett cag ctg get ggt tgt ctg tet caa ctg catVal Leu Leu Gly His Ser Leu Gly lie Pro Trp Ala Pro Uu Ser Ser 50 55 60 igt cca tet caa get ett cag ctg get ggt tgt ctg tet caa ctg cat

Cys Pro Ser Gin Ala Leu Gin Uu Ala Gly Cys Leu Ser Gin Leu His 65 70 75 S〇 tet ggt ctg itc ctg tat cag ggt ett ctg caa get ctg gaa ggt ateCys Pro Ser Gin Ala Leu Gin Uu Ala Gly Cys Leu Ser Gin Leu His 65 70 75 S〇 tet ggt ctg itc ctg tat cag ggt ett ctg caa get ctg gaa ggt ate

Ser Gly Leu Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly He 85 90 . 95 tet ccg gaa ctg ggt ccg act ctg gac act ctg cag eta gat gta getSer Gly Leu Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly He 85 90 . 95 tet ccg gaa ctg ggt ccg act ctg gac act ctg cag eta gat gta get

Ser Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu Gin Leu Asp Val Ala 100 105 Π0 gac ttt get act act att tgg caa cag atg gaa gag etc ggt atg gcaSer Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu Gin Leu Asp Val Ala 100 105 Π0 gac ttt get act act att tgg caa cag atg gaa gag etc ggt atg gca

Asp Phe Ala Thr Thr lie Trp Gin Gin Met Glu Clu Leu Gly Met Ala 115 120 125 cca get ctg caa ccg act caa ggt get atg ccg gca ttc get tet gcaAsp Phe Ala Thr Thr lie Trp Gin Gin Met Glu Clu Leu Gly Met Ala 115 120 125 cca get ctg caa ccg act caa ggt get atg ccg gca ttc get tet gca

Pro Ala Leu Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala 130 135 140 ttc cag cgt cgt gca gga ggt gta ctg gtt get tet cat ctg caa tetPro Ala Leu Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala 130 135 140 ttc cag cgt cgt gca gga ggt gta ctg gtt get tet cat ctg caa tet

Phe Gin Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser 145 150 155 160 lie ctg gaa gta tet tac cgt gtt ctg cgt cat ctg gee cag ccgPhe Gin Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser 145 150 155 160 lie ctg gaa gta tet tac cgt gtt ctg cgt cat ctg gee cag ccg

Phe Leu Clu Val Ser Tyr Arg Val Leu Arg His Leu Ala Gin Pro 165 170 175 taatag <210> 3 <211> 207 <212> PRT <213>人類 <400> 3Phe Leu Clu Val Ser Tyr Arg Val Leu Arg His Leu Ala Gin Pro 165 170 175 taatag <210> 3 <211> 207 <212> PRT <213> Human <400> 3

Met Ala Gly Pro Ala Thr Gin Ser Pro Met Lys Leu Mel Ala Leu Gin ! 5 10 15Met Ala Gly Pro Ala Thr Gin Ser Pro Met Lys Leu Mel Ala Leu Gin ! 5 10 15

Leu Leu Leu Trp His Ser Ala Leu Trp Thr Val Gin Clu Ala Thr Pro 20 25 30Leu Leu Leu Trp His Ser Ala Leu Trp Thr Val Gin Clu Ala Thr Pro 20 25 30

Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu Lys Cys Leu 35 40 45Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu Lys Cys Leu 35 40 45

Glu Gin Val Arg Lys He Gin Gly Asp Gly Ala Ala Leu Gin Glu Lys 50 55 60Glu Gin Val Arg Lys He Gin Gly Asp Gly Ala Ala Leu Gin Glu Lys 50 55 60

Leu Val Ser Glu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu 65 70 75 80 150941.doc 96 144 192 240 288 336 384 432 480 525 531 201138831Leu Val Ser Glu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu 65 70 75 80 150941.doc 96 144 192 240 288 336 384 432 480 525 531 201138831

Val Leu Leu Gly His Ser Leu Gly He Pro Trp Ala Pro Leu Ser Ser 85 90 95Val Leu Leu Gly His Ser Leu Gly He Pro Trp Ala Pro Leu Ser Ser 85 90 95

Cys Pro Ser G)n Ala Leu Gin Leu Ala Gly Cys Leu Ser Gin Leu His 100 105 110Cys Pro Ser G)n Ala Leu Gin Leu Ala Gly Cys Leu Ser Gin Leu His 100 105 110

Ser Gly Leu Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly Me 115 120 125Ser Gly Leu Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly Me 115 120 125

Ser Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu Gin Leu Asp Val Ala 130 135 140Ser Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu Gin Leu Asp Val Ala 130 135 140

Asp Phe Ala Thr Thr Me Trp Gin Gin Met Glu Glu Leu Gly Met Ala 145 150 155 160Asp Phe Ala Thr Thr Me Trp Gin Gin Met Glu Glu Leu Gly Met Ala 145 150 155 160

Pro Ala Leu Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala 165 170 175Pro Ala Leu Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala 165 170 175

Phe Gin Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser 180 185 190Phe Gin Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser 180 185 190

Phe Leu Glu Val Ser Tyr Arg Va) Leu Arg His Leu Ala Gin Pro 195 200 205 <210> 4 <211> 204 <212> PRT <213>人類 <400> 4Phe Leu Glu Val Ser Tyr Arg Va) Leu Arg His Leu Ala Gin Pro 195 200 205 <210> 4 <211> 204 <212> PRT <213> Human <400>

Met Ala Gly Pro Ala Thr Gin Ser Pro Met Lys Leu Met Ala Leu Gin 15 10 15Met Ala Gly Pro Ala Thr Gin Ser Pro Met Lys Leu Met Ala Leu Gin 15 10 15

Leu Leu Leu Trp His Ser Ala Leu Trp Thr Val Gin Glu Ala Thr Pro 20 25 30Leu Leu Leu Trp His Ser Ala Leu Trp Thr Val Gin Glu Ala Thr Pro 20 25 30

Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu Lys Cys Leu 35 40 45Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu Lys Cys Leu 35 40 45

Glu Gin Val Arg Lys lie Gin Gly Asp Gly Ala Ala Leu Gin Glu Lys 50 55 60Glu Gin Val Arg Lys lie Gin Gly Asp Gly Ala Ala Leu Gin Glu Lys 50 55 60

Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu Val Leu Leu 65 70 75 80Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu Val Leu Leu 65 70 75 80

Gly His Ser Leu Gly lie Pro Trp Ala Pro Leu Ser Ser Cys Pro Ser 85 90 95Gly His Ser Leu Gly lie Pro Trp Ala Pro Leu Ser Ser Cys Pro Ser 85 90 95

Gin Ala Leu Gin Leu Ala Gly Cys Leu Ser Gin Leu His Ser Gly Leu 100 105 110Gin Ala Leu Gin Leu Ala Gly Cys Leu Ser Gin Leu His Ser Gly Leu 100 105 110

Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly lie Ser Pro Glu 115 120 125Phe Leu Tyr Gin Gly Leu Leu Gin Ala Leu Glu Gly lie Ser Pro Glu 115 120 125

Leu Gly Pro Thr Leu Asp Thr Leu Gin Leu Asp Va) Ala Asp Phe Ala 130 135 140 150941.doc 201138831Leu Gly Pro Thr Leu Asp Thr Leu Gin Leu Asp Va) Ala Asp Phe Ala 130 135 140 150941.doc 201138831

Thr Thr lie Trp Gin Gin Met Giu Glu Leu Gly Met Ala Pro Ala Leu 145 150 155 160Thr Thr lie Trp Gin Gin Met Giu Glu Leu Gly Met Ala Pro Ala Leu 145 150 155 160

Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala Phe Gin Arg 165 170 175Gin Pro Thr Gin Gly Ala Met Pro Ala Phe Ala Ser Ala Phe Gin Arg 165 170 175

Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser Phe Leu Glu 180 185 190Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gin Ser Phe Leu Glu 180 185 190

Val Ser Tyr Arg Val Leu Arg His Leu Ala Gin Pro 195 200 J 5094】.docVal Ser Tyr Arg Val Leu Arg His Leu Ala Gin Pro 195 200 J 5094].doc

Claims (1)

201138831 七、申請專利範圍: 1. 一種包含顆粒性細胞群落刺激因子(「G-CSF」)分子之 組合物,該等分子包括聚乙二醇(「PEG」)分子共價鏈 結於最接近N端之離胺酸殘基,其中至少30%之G-CSF分 子並非於N端聚乙二醇化。 2·如請求項1之組合物,其中各G-CSF分子係共價鏈結於單 一 PEG分子。 3·如請求項1之組合物,其中至少80%之該等G-CSF分子係 單聚乙二醇化。 4·如1求項1之組合物’其中至少85%之該等G-CSF分子係 單聚乙二醇化。 5. 如叫求項1之組合物’其中至少90%之該等G-CSF分子係 單聚乙二醇化。 6. 如請求組合物,其中至少95%之該等gcsf分子係 單聚乙二醇化。 7·如吻求項丨之組合物,其中該離胺酸殘基為6殘基。 如。月求項1之組合物,其中若計數N端之甲硫胺酸殘基, 則該離胺酸殘基為Lys 17殘基。 9’種組合物,其包含一群單聚乙二醇化蛋白質,其中至 v 3 0 /〇之該等蛋白質分子並非於N端聚乙二醇化。 1〇·如π求項9之組合物,其中至少3〇%之該等單聚乙二醇化 蛋白質係於Ν端1〇〇個胺基酸以内之離胺酸殘基聚乙二醇 化。 士 π求項9之組合物,其中至少3〇%之該等單聚乙二醇化 150941.doc 201138831 蛋白質係於N端50個胺基酸以内之離胺酸殘基聚乙二醇 化。 12. 如請求項9之組合物,其中至少30%之該等單聚乙二醇化 蛋白質係於N端20個胺基酸以内之離胺酸殘基聚乙二醇 化。 13. 如請求項i之組合物,其中n端經聚乙二醇化之G-CSF分 子相對於最接近N端之離胺酸經聚乙二醇化之G-CSF分 子的比例為小於約1比約1 00。 14. 如請求項!之組合物,其中n端經聚乙二醇化之G-CSF分 子相對於最接近N端之離胺酸經聚乙二醇化之G-CSF分 子的比例為約1 〇比約90。 15. 如請求項!之組合物,其中n端經聚乙二醇化之G-CSF分 子相對於最接近N端之離胺酸經聚乙二醇化之G-CSF分 子的比例為小於約20比約80。 16_如請求項組合物,其中n端經聚乙二醇化之G-CSF分 子相對於最接近N端之離胺酸經聚乙二醇化之G-CSF分 子的比例為小於約3〇比約70。 17. —種醫藥調配物,其包含如請求項1之組合物及無蛋白 質載劑。 18. 如請求項π之醫藥調配物,其中該調配物於約_2(TC貯藏 至少15個月後仍安定。 19. 如請求項17之醫藥調配物,其中該調配物於約4°C貯藏 至少15個月後仍安定。 20. 如請求項丨7之醫藥調配物,其中該調配物於約251貯藏 150941.doc 201138831 至少ίο個月後仍安定。 21·如請求項丨7之醫藥調配物,其中該調配物於約37。〇貯藏 至少10個月後仍安定。 22. —種使接受者(host)之白血球數量增加之方法包含對 有此需要之接受者投與如請求項丨至2丨中任一項之組人 物。 23_如請求項22之方法’其中該接受者為人類。 24·如請求項22之方法,其中該接受者有罹患嗜中性球減少 症(neutropenia)風險或罹患嗜中性球減少症。 25. 如請求項22之方法,其中該接受者正接受使白血球數減 少之藥劑治療。 26. 如請求項22之方法,其中該接受者之内源性G_CSF濃度 降低。 27_如請求項22之方法’其中該接受者正接受輻射治療。 28. 如請求項22之方法’其中該接受者正罹患肺癌、淋巴 瘤、乳癌、骨髓移植、睾丸癌、與AIDS相關之惡性腫 瘤、骨髓造血不良症(myelodysplastic disorders)、急性 白血病、先天性及週期性嗜中性球減少症或再生障礙性 貧血(aplastic anemia) ° 29. 如請求項22之方法,其中該調配物係注射投與。 30. 如請求項22之方法,其中該調配物係經皮下投與。 3 1.如請求項22之方法,其中該調配物係經口投與。 32.如請求項22之方法,其中該調配物係經靜脈内投與。 3 3 _ —種製備蛋白質接合物之方法,包含使蛋白質與經活化 150941.doc 201138831 之PEG-醛於包含DMSO之反應緩衝液中反應,藉此使該 蛋白質與該PEG聚合物共價鏈結。 34. —種由如請求項33之方法形成之接合物。 3 5.如請求項33之方法,其中實質上去除所有未鏈結之PEG 聚合物,以獲得該蛋白質接合物。 3 6.如請求項33之方法,其中該PEG為至少10 kD。 3 7.如請求項33之方法,其中該PEG為至少20 kD。 3 8.如請求項33之方法,其中該PEG為30 kD。 3 9.如請求項33之方法,其中該PEG為至少40 kD。 40. —種製備G-CSF蛋白質接合物之方法,包含使G-CSF蛋 白質與經活化之PEG-醛於包含DMSO之反應缓衝液中反 應,藉此使該G-CSF蛋白質與該PEG聚合物共價鏈結。 41. 一種由如請求項40之方法形成之接合物。 42. 如請求項40之方法,其中實質上去除所有未鏈結之PEG 聚合物,以獲得該G-CSF蛋白質接合物。 43. 如請求項40之方法,其中該PEG為至少10 kD。 44. 如請求項40之方法,其中該PEG為至少20 kD。 45. 如請求項40之方法,其中該PEG為30 kD。 46. 如請求項40之方法,其中該PEG為至少40 kD。 150941.doc201138831 VII. Scope of Application: 1. A composition comprising granulosa cell community stimulating factor ("G-CSF") molecules, including the covalent linkage of polyethylene glycol ("PEG") molecules in the closest An amino acid residue at the N-terminus, wherein at least 30% of the G-CSF molecules are not PEGylated at the N-terminus. 2. The composition of claim 1, wherein each G-CSF molecule is covalently linked to a single PEG molecule. 3. The composition of claim 1 wherein at least 80% of the G-CSF molecules are mono-PEGylated. 4. The composition of claim 1 wherein at least 85% of the G-CSF molecules are mono-PEGylated. 5. The composition of claim 1 wherein at least 90% of the G-CSF molecules are mono-PEGylated. 6. If the composition is claimed, at least 95% of the gcsf molecules are mono-PEGylated. 7. A composition according to the formula, wherein the lytic acid residue is 6 residues. Such as. The composition of claim 1, wherein if the N-terminal methionine residue is counted, the lysine residue is a Lys 17 residue. A 9' composition comprising a population of mono-pegylated proteins, wherein the protein molecules to v30/〇 are not PEGylated at the N-terminus. The composition of claim 9, wherein at least 3% by weight of the monopegylated protein is PEGylated with an amino acid residue within one of the amino acids of the quinone. The composition of π § 9, wherein at least 3% by weight of the mono-pegylated 150941.doc 201138831 protein is PEGylated at the N-terminal 50 amino acids. 12. The composition of claim 9, wherein at least 30% of the monopegylated proteins are PEGylated with an amino acid residue within the N-terminal 20 amino acids. 13. The composition of claim i, wherein the ratio of the p-PEGylated G-CSF molecule at the n-terminus to the G-CSF molecule PEGylated to the most proximal N-terminal is less than about 1 ratio About 100. 14. As requested! The composition wherein the ratio of the p-PEGylated G-CSF molecule at the n-terminus to the G-CSF molecule which is pegylated by the amino acid closest to the N-terminus is about 1 Torr to about 90. 15. As requested! The composition wherein the proportion of the G-CSF molecule at the n-end of the pegylated G-CSF molecule relative to the G-CSF molecule which is pegylated at the N-terminus is less than about 20 to about 80. 16_, as in the claim composition, wherein the ratio of the p-PEGylated G-CSF molecule at the n-terminus to the G-CSF molecule that is pegylated with respect to the amino acid closest to the N-terminus is less than about 3 Å. 70. 17. A pharmaceutical formulation comprising the composition of claim 1 and a protein-free carrier. 18. The pharmaceutical formulation of claim π, wherein the formulation is stable at about _2 (TC remains stable for at least 15 months. 19. The pharmaceutical formulation of claim 17, wherein the formulation is at about 4 ° C The drug is still stable after at least 15 months of storage. 20. The pharmaceutical formulation of claim 7, wherein the formulation is stable at about 251 storage 150941.doc 201138831 at least ίο months. 21·If the request is 丨7 Formulation wherein the formulation is stable at about 37. The sputum remains stable after at least 10 months of storage. 22. A method of increasing the number of white blood cells of the recipient comprises administering to the recipient of the need, such as a request 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 Neutropenia) The risk of or suffering from neutropenia. 25. The method of claim 22, wherein the recipient is receiving a treatment for reducing the number of white blood cells. 26. The method of claim 22, wherein the recipient The concentration of source G_CSF is reduced. 27_If requested The method of item 22 wherein the recipient is receiving radiation therapy. 28. The method of claim 22 wherein the recipient is suffering from lung cancer, lymphoma, breast cancer, bone marrow transplantation, testicular cancer, AIDS-related malignancy, bone marrow Myelodysplastic disorders, acute leukemia, congenital and periodic neutropenia or aplastic anemia. 29. The method of claim 22, wherein the formulation is administered by injection. 30. The method of claim 22, wherein the formulation is administered subcutaneously. 3 1. The method of claim 22, wherein the formulation is administered orally. 32. The method of claim 22, wherein The formulation is administered intravenously. 3 3 _ - A method of preparing a protein conjugate comprising reacting a protein with a PEG-aldehyde activated by 150941.doc 201138831 in a reaction buffer comprising DMSO, thereby allowing the protein to be Covalently linked to the PEG polymer. 34. A conjugate formed by the method of claim 33. 3. 5. The method of claim 33, wherein substantially all unlinked PEG poly is removed 6. The method of claim 33, wherein the PEG is at least 10 kD. The method of claim 33, wherein the PEG is at least 20 kD. The method of item 33, wherein the PEG is 30 kD. 3. The method of claim 33, wherein the PEG is at least 40 kD. 40. A method of preparing a G-CSF protein conjugate comprising reacting a G-CSF protein with an activated PEG-aldehyde in a reaction buffer comprising DMSO, thereby rendering the G-CSF protein and the PEG polymer Covalent chain. 41. A conjugate formed by the method of claim 40. 42. The method of claim 40, wherein substantially all of the unlinked PEG polymer is removed to obtain the G-CSF protein conjugate. 43. The method of claim 40, wherein the PEG is at least 10 kD. 44. The method of claim 40, wherein the PEG is at least 20 kD. 45. The method of claim 40, wherein the PEG is 30 kD. 46. The method of claim 40, wherein the PEG is at least 40 kD. 150941.doc
TW099133189A 2009-09-30 2010-09-29 Modified granulocyte colony stimulating factor (G-CSF) TW201138831A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US24738909P 2009-09-30 2009-09-30

Publications (1)

Publication Number Publication Date
TW201138831A true TW201138831A (en) 2011-11-16

Family

ID=43826620

Family Applications (1)

Application Number Title Priority Date Filing Date
TW099133189A TW201138831A (en) 2009-09-30 2010-09-29 Modified granulocyte colony stimulating factor (G-CSF)

Country Status (4)

Country Link
AR (1) AR078205A1 (en)
CL (1) CL2010001049A1 (en)
TW (1) TW201138831A (en)
WO (1) WO2011041376A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2659990C (en) 2006-08-04 2016-03-22 Prolong Pharmaceuticals, Inc. Polyethylene glycol erythropoietin conjugates
EP2399572A1 (en) * 2010-06-22 2011-12-28 Sandoz AG Long-term storage of non-glycosylated recombinant human G-CSF
WO2016201448A2 (en) * 2015-06-11 2016-12-15 Prolong Pharmaceuticals, LLC Pegylated granulocyte colony stimulating factor (gcsf)
CA3171491A1 (en) * 2020-03-20 2021-09-23 Amgen, Inc. Determination of free n-terminus of pegfilgrastim using an acid protease

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2599502T3 (en) * 2003-04-11 2017-04-18 Antriabio Inc Process for Preparation of Site-Specific Protein Conjugates
EP1674113A1 (en) * 2004-12-22 2006-06-28 F. Hoffmann-La Roche Ag Conjugates of insulin-like growth factor-1 (IGF-1) and poly(ethylene glycol)
CN101193658B (en) * 2005-06-01 2011-08-31 马克西根控股公司 Pegylated G-CSF polypeptides and methods of producing same
JP2009503111A (en) * 2005-08-04 2009-01-29 ネクター セラピューティックス エイエル,コーポレイション G-CSF moiety and polymer composite

Also Published As

Publication number Publication date
AR078205A1 (en) 2011-10-19
CL2010001049A1 (en) 2011-04-15
WO2011041376A1 (en) 2011-04-07

Similar Documents

Publication Publication Date Title
US7893019B2 (en) G-CSF site-specific mono-conjugates
CN1729018B (en) G-CSF conjugates
KR100203824B1 (en) Compositions and methods for stimulating megakaryocyte growth and differentiation
KR100961859B1 (en) G-csf conjugate
KR100773323B1 (en) G-csf conjugates
TWI364295B (en) Polymer conjugates of cytokines, chemokines, growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
US7074755B2 (en) Erythropoietin conjugate compounds with extended half-lives
KR101330338B1 (en) Conjugates of a gm-csf moiety and a polymer
JPH09506116A (en) N-terminal chemically modified protein compositions and methods
CN104689333B (en) The liquid preparation of G-CSF conjugates
JP2011507913A (en) Y-type polyethylene glycol-modified G-CSF and its production method and use
TW201138831A (en) Modified granulocyte colony stimulating factor (G-CSF)
WO2004020468A2 (en) Interferon beta-like molecules for treatment of cancer
US10653752B2 (en) Methods and use of growth hormone supergene family protein analogs for treatment of radiation exposure
JP2006521372A (en) 1: 1 conjugate of biologically active substance and biocompatible polymer, method for producing the same, and pharmaceutical composition containing the same
US20050281778A1 (en) Human growth hormone conjugated with biocompatible polymer
WO2001076639A2 (en) Chemically-modified myelopoietin conjugates
JP2704214B2 (en) Cytokine having unpaired cysteine residue and conjugate thereof
CN103096916B (en) For the preparation of bG-CSF and variant thereof
US20060052291A1 (en) Chemically-modified progenipoietin conjugates
KR20180017104A (en) PEGylated granulosa cell colony stimulating factor (GCSF)
KR20040086521A (en) Biologically Active Material Conjugated With Biocompatible Polymer with 1:1 complex, Preparation Method Thereof And Pharmaceutical Composition Comprising The Same
CN101172160A (en) Interferon gamma conjugates