TW201002821A - Method of producing agent containing single useful material that has polypeptide - Google Patents

Method of producing agent containing single useful material that has polypeptide Download PDF

Info

Publication number
TW201002821A
TW201002821A TW098114515A TW98114515A TW201002821A TW 201002821 A TW201002821 A TW 201002821A TW 098114515 A TW098114515 A TW 098114515A TW 98114515 A TW98114515 A TW 98114515A TW 201002821 A TW201002821 A TW 201002821A
Authority
TW
Taiwan
Prior art keywords
virus
preparation
stabilizer
polypeptide
monoclonal antibody
Prior art date
Application number
TW098114515A
Other languages
Chinese (zh)
Inventor
Terumichi Tonooka
Original Assignee
Asahi Kasei Medical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Asahi Kasei Medical Co Ltd filed Critical Asahi Kasei Medical Co Ltd
Publication of TW201002821A publication Critical patent/TW201002821A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention provides a method of producing a high-concentration agent solution containing a single useful material that has polypeptides, characterized in that the virus removal step removes the virus originated from the formation of the single useful material having polypeptides, and originated from the formation of the stabilizer, wherein the virus removal capability of the virus removal step is (LRV, logarithmic reduction value) ≥4, the concentration of the single useful material having polypeptides during the virus removal step is between the range of 3.0-10 wt%, and the average permeability of the single useful material having polypeptides against the virus removal membrane used in the virus removal step is 1.0(kg/m.sup.2/hour) or more both in 0-10 minutes and 2-3 hours after the start of filtration.

Description

201002821 六、發明說明: 【發明所屬之技術領域】 本發明係關於一種具有多肽之單一有用物質、尤其是單 株抗體製劑或重組製劑之製造方法。 【先前技術】 作為蛋白製劑中之-種的單株抗體製劑或重組製劑等具 有使用基因重組細胞等而生成之單一成分。藉由利用該等201002821 VI. Description of the Invention: [Technical Field of the Invention] The present invention relates to a method for producing a single useful substance having a polypeptide, particularly a monoclonal antibody preparation or a recombinant preparation. [Prior Art] A single antibody preparation or a recombinant preparation which is a species in a protein preparation has a single component produced by using a recombinant cell or the like. By using these

製劑’可將誘發某種病狀之特定蛋白質或癌細胞等特異地 識別而引起抗體反應,因而被廣泛用於風濕、癌症、白血 病等疑難病症之治療。 以單株抗體製劑或重組製劑等為代表之蛋白製劑之製迭 步驟係視所欲產生之有用物f之種類而有㈣各樣,通常 至少需要如下步驟:使用基因經重組之細胞或動物來產生 上述有用物質之培養步驟;利用除菌膜、吸附柱、層析儀 等將上述有用物質純化之純化步驟;#包含上述有用物質 之溶液中之病毒去除或鈍化之病毒去除步驟;添加抑制上 述有用物質之凝聚之穩定劑的穩定劑添加㈣;以„去 除了病毒之上述有用物質㈣之密封步驟。 對於以單株抗體製劑為代表之包含具有多肽之單 物質的製劑整體而言, 有用 之病毒…畫, 大里報告有源自原料、源自步驟 亞^ 列’因此將病毒純化或去除之步驟伟非常 重要之步驟。作為病毒之鈍化方法 或利用化學藥品之處理等,但若僅單獨進==處理 毒之鈍化不充分,品 却進仃忒寻處理則病 而且該等方法有使蛋白製劑中之有用物 139962.doc 201002821 質自身亦發生變性之虞。出於上述背景,作為未伴有化學 變性之物理性的病毒去除方法,係利用病毒去除膜來實施 過濾。病毒去除膜係基於病毒尺寸之去除方法,因此與加 熱處理或利用化學藥品之處理等其他方法相比,具有不易 受到病毒特性影響之優點,若錢具有20 nm左右之孔徑The preparation 'specifically recognizes a specific protein or cancer cell which induces a certain condition and causes an antibody reaction, and is therefore widely used for the treatment of a difficult condition such as rheumatism, cancer, and leukemia. The step of forming the protein preparation represented by the monoclonal antibody preparation or the recombinant preparation or the like depends on the kind of the desired substance f to be produced. (4) Usually, at least the following steps are required: using the recombinant cell or animal of the gene a step of cultivating the above-mentioned useful substance; a purification step of purifying the above-mentioned useful substance by using a sterilization membrane, an adsorption column, a chromatograph, etc.; a virus removal step of removing or inactivating a virus in a solution containing the above-mentioned useful substance; a stabilizer for the stabilizer of agglomerates of a useful substance (IV); a sealing step of the above-mentioned useful substance (IV) with a virus removed. For a preparation comprising a single substance having a polypeptide represented by a monoclonal antibody preparation as a whole, useful Virus...Drawing, Dali reports that there are steps from the raw material, which are derived from the steps of the step, so the process of purifying or removing the virus is very important. It is used as a passivation method for viruses or treatment with chemicals, etc. == The passivation of the poison is not sufficient, but the product is processed and the disease is treated. Useful material 139962.doc 201002821 The quality itself is also denatured. From the above background, as a physical virus removal method without chemical degeneration, the virus removal membrane is used for filtration. The virus removal membrane is based on the size of the virus. The removal method is therefore less susceptible to the influence of virus characteristics than other methods such as heat treatment or treatment using chemicals, and if the money has a pore diameter of about 20 nm

之病毒去除膜,則可將目帝· p A j將目别已知之所有病毒去除至過濾前 之1萬分之1左右。 使用以單株抗體製劑為代表之蛋白製劑時,有直接使用 液體製劑之情形、及將冷殊乾燥之製劑溶於蒸餾水等中而 使用之情形,就降低醫療現狀貞擔及醫療錯誤之風險的 觀點考慮,而期望製劑之液狀化、及用以減少投”劑之 液量而降低患者負擔之高濃度化、然而,以單株抗體製劑 為代表之蛋白製劑與以其成分、比例而原本自然地存在於 血液中由此利用Cohn分離而純化所得之多株抗體不同,由 於其係藉由培養基因重組細胞等方法而產生'亦包括诀合 抗體等天然不存在者、而且為單一成分等原因,其物性: 多株抗體相比較為多種多樣,其中亦存在性狀不穩定者。、 關於以單株抗體製劑為代表之蛋白製劑之穩定性,特別 重要的要素之一係製劑溶液之濃度。即,當濃度較高^例 如超過最終製劑濃度即2重量%時,大多情況下難以確^ 蛋白質自身於溶液中之穩定性,而導致多聚體、凝聚體等 混入。因此,必須於純化步驟中進行了抗體濃縮後之階段 中使用穩定劑等,該穩定劑係用於使具有不穩定性狀之蛋 白製劑變得穩定(專利文獻1}。已知多聚體、凝聚體等會導 139962.doc 201002821 致過敏性休克等嚴重之副作用, + ή 即便於可生產出單株抗體 本身之情形時,有時亦無法完全防止純化步驟中之蛋白質 之變性而放棄最終製劑化。 2前利用病毒去除膜之過渡係於〇 w重量%左右之較低 =下進行’故係保持相對較難形成多聚體、凝聚體等之 浪度的狀態而進行直至純化之濃縮為止之步驟。因此,將 抑制蛋白質之多聚體形成之穩定劑添加於系統内的需 :又ν ’通常係利用病毒去除膜進行過據後利用仙⑽ra F伽,超濾、)膜進行濃縮,並添加穩定劑而進行最終 化。 然而’對以上述單株抗體製劑為代表之蛋白製劑之凝 多聚體形成發揮抑制效果之穩定劑,係大量使用源自 勿、源自酸酵產物而製造者(非專利文獻卜2、3、4' 1)1該情料’雖可確保將作為製狀單株抗體自身之病 毋-全去除Μ旦難以確保將源自上述穩定劑之病毒完全去 除=此’穩定劑難以使用源自生物、源自酸酵者,或者 走用才存在穩疋劑本身之純化步驟變複雜等問題。 又:於相當於上述最終製劑之濃度之抗體、及其中所添 加之穩定劑濃度之溶液中進行病毒去除步驟於先前技術中 較為困難。即,若使用此種成分之溶液,則無法進行兼罝 病毒去除之完全性與溶液之透過性的過濾、,因此,尤以 =用可將小病毒之類的小型病毒去除的病毒去除膜進行過 i重量%以下來進;上;1之“中間物綱釋成最高為 進订過濾。而且如上所述,先前技術中係 139962.doc 201002821 將其後經過利用UF膜之濃縮及穩定劑添加步驟而進行最終 製劑化作為前提。此外’大多情況下若其中混入穩定劑等 成分則透過性進一步下降。 上述限制除了會引起上述病毒安全性之相關問題以外, 亦會導致如下許多問題之產生:主體直至下游無法精簡 化’結果無法避免製造步驟之管理上之複雜化、高成本 化’或具有不穩定性狀之蛋白質之情形時無法確保此後之 濃縮步驟之穩定性,而難以進行製劑化等。 [專利文獻1]曰本專利特表2001-503781號公報 [非專利文獻1 ]「最新微生物手冊」(丨986) [非專利文獻 2]「Bio Industry」4,554(1984) [非專利文獻3]「醱酵手冊」((財團法人)Bi〇industry協會 醱酵與代謝研究會編)(ISBN 4-320-05575-6) [非專利文獻4]「14303之化學商品」(化學工業曰報 社)(ISBN 4-87326-404-9) [非專利文獻5]獨立行政法人科學技術振興機構(JST)主 頁 http//ww. go.jp/pr/report/report68/details.html [非專利文獻6 ]病毋貫驗學總論(國立預防衛生研究所學 友會編) 【發明内容】 [發明所欲解決之問題] 鑒於上述問題,本發明之課題在於提供一種單株抗體之 製造方法,其可實現高濃度地包含具有多肽之單一有用物 139962.doc 201002821 質之製劑溶液的較高之透過性與病毒去除性兩方,且可利 Z包含源自纟物或源㈣酵產⑯而存在病毒混入風險之穩 定劑的製劑,可簡便地用於各種製劑之製造製程。 鑒於該課題,最難解決者係病毒去除步驟。就利用尺寸 去除之機構(參照下述[實施方式])來去除病毒之膜之特性 方面而言,通常病毒去除膜之透過性較低,而通常難以適 應於較高之製劑濃度之過渡’因此成為製造步驟方面之最 大=題。進而,病毒去除步驟之透過性係與病毒去除膜之 緻密層之孔徑成正比例,病毒去除性係與緻密層之孔徑成 反比例’因此以較高性能而兼具該等相悖性質之兩方成為 技術上之課題。 [解決問題之技術手段] …,發明者們為了解決上述問題而進行了潛心研究,結果 獲得了本發明。即,本發明包括以下内容。 ⑴-種包含具有多肽之單一有用物質之製劑之製造方法, 其特徵在於:其係至少包括如下步驟者:產生具有多肽之 早一有用物質之培養步驟’將具有多肽之單一有用物質純 :之純化步驟,添加至少一種抑制具有多狀之單一有用物 =之凝聚的穩定劑之穩定劑添加步驟,藉由病毒去除膜將 匕合具有多肽之單-有用物質之溶液中的病毒去除之病毒 絲步驟,以及將去除了病毒之具有多肽之單一有用物質 被封之密封步驟’ ·並且上述病毒去除步驟係將源自具有多 肽之單_有用物質之太 ± 座生手之病t、與源自穩定劑之產生 病母去除’病毋去除步驟之病毒去除能力為LRvy, 139962.doc 201002821 病毒去除步驟時之具有多肽之單一有用物質的濃度為 3.0〜10.0重量%之範圍,溶液中所含之具有多肽之單:有 用物質相對於上述病毒去除步驟中所使用之病毒去:膜之 過遽開始後㈡0分#、及經過2〜3小時之時的平均透過性 均為 1.0(kg/m2/hour)以上。 ° [2]如[1]之包含具有多肽之單一有用 男代物貝之製劑之製造方 法,其中上述穩定劑係源自生物或源自酸酵產物者。 m如Π]或[2]之包含具有多肽之單—有用物f之製劑之製 造方法’其中上述穩定劑係選自糖類、胺基酸、胺基糖、 有機酸、或該等之衍生物中。 [4] 如⑴至[3]中任一項之包含具有多肽之單一有用物質之 製劑之製造方法’ #中上述病毒去除膜為包含合成高分子 之中空纖維膜。 [5] 如[1 ]至[4]中任一項之包含具有多 。 另夕肽之早一有用物質之 製劑之製造方法,盆中上成、忘基土队. 上迷病毋去除臈為多層微多孔膜。 [6] —種包含單株抗體之製劑萝 ^ 衣剞<衣k方法,其特徵在於:其 係至少包括如下步驟者:產生單姓 八 株抗體之培養步驟,將單 株抗體純化之純化步驟,添加至少— λ 種抑制早株抗體之凝 聚之穩定劑的穩定劑添加步驟,藉 稽由病毒去除膜將包含單 株抗體之溶液十之病毒去降的在主丄 〇〇 涡毋去除的病勢去除步驟,以及將去除 了病毒之單株抗體密封之密封步. . 了 ^驟,亚且上述病毒去除步 驟係將源自單株抗體之產生時 了心届t、與源自穩定劑之產 生時之病毒去除’病毒去除步 τ ν鄉之病毒去除能力為 LRV = 4,病毒去除步驟時單 平体彳几體濃度為3.0〜10重量% 139962.doc 201002821 之範圍’溶液中所含之單株抗體相對於上述病毒去除步驟 中所使用之病毒去除臈之過濾開始後〇分鐘、及經過 2〜3小時之時的平均透過性均為1.0(kg/m2/h〇ur)以上。 [7] 如[6]之包含單株抗體之製劑之製造方法,其中上述穩 定齊彳係源自生物或源自醱酵產物者。 [8] 如[6]或[7]之包含單株抗體之製劑之製造方法,其中上 述穩疋劑係選自糖類、胺基酸、胺基糖、有機酸、或該等 之衍生物中。When the virus is removed from the membrane, all the viruses known to the target can be removed to about 1 in 10,000 before filtration. When a protein preparation represented by a single antibody preparation is used, the case where the liquid preparation is used as it is, and the case where the cold-dried preparation is dissolved in distilled water or the like is used, and the risk of medical care and medical errors is reduced. In view of the above, it is desirable to liquefy the preparation and to reduce the amount of the administration agent to reduce the burden on the patient. However, the protein preparation represented by the monoclonal antibody preparation is originally composed of the components and ratios thereof. Naturally, it exists in the blood, and the multi-strain antibody obtained by Cohn isolation and purification is different, and it is produced by a method such as culturing a genetic recombinant cell, and includes a naturally occurring non-existent antibody such as a chelate antibody, and is a single component. The reason is that the physical properties of the multi-strain antibody are various, and there are also unstable traits. One of the most important factors regarding the stability of the protein preparation represented by the monoclonal antibody preparation is the concentration of the preparation solution. That is, when the concentration is higher, for example, more than 2% by weight of the final formulation concentration, it is often difficult to confirm that the protein itself is in solution. Qualitative, resulting in the incorporation of multimers, aggregates, etc. Therefore, it is necessary to use a stabilizer or the like in the stage after concentration of the antibody in the purification step, and the stabilizer is used to stabilize the protein preparation having the unstable state. (Patent Document 1). It is known that multimers, aggregates, and the like may cause serious side effects such as anaphylactic shock, and ή ή ή 便于 便于 便于 便于 便于 便于 便于 便于 便于 便于 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单 单Degradation of the protein in the purification step and abandonment of the final formulation. 2 The transition of the membrane to remove the virus before is lower than the weight of the 〇w by the lower = lower, it is relatively difficult to form a polymer, agglomerate, etc. The step of concentrating until the purification is concentrated. Therefore, the stabilizer for inhibiting the formation of multimers of proteins is added to the system: ν 'usually using the virus removal membrane and using the sage (10)ra F gamma, ultrafiltration, membranes are concentrated, and stabilizers are added for finalization. However, 'coagulation of protein preparations represented by the above-mentioned monoclonal antibody preparations A stabilizer which exhibits an inhibitory effect on the formation of a body, and is produced by a large amount of a manufacturer derived from an acid-derived product (Non-Patent Document 2, 3, 4' 1) 1 It is difficult to ensure the complete removal of the virus derived from the above stabilizers. It is difficult to use the stabilizer derived from the organism, from the acidifier, or the stabilizer itself. The purification step becomes complicated, etc. Further, it is difficult in the prior art to carry out the virus removal step in a solution corresponding to the concentration of the above-mentioned final preparation and the concentration of the stabilizer added thereto, that is, if such a component is used. In the case of the solution, the filtration of the completeness of the virus and the permeability of the solution cannot be performed. Therefore, the virus removal film which can remove the small virus such as the small virus is subjected to i% by weight or less; Above; 1 "Intermediate class is released into the highest order for filtering. Further, as described above, the prior art 139962.doc 201002821 is premised on the final formulation by the concentration and stabilizer addition step of the UF film. In addition, in many cases, the permeability is further lowered if a stabilizer or the like is mixed therein. In addition to the above-mentioned limitations that may cause problems related to the above-mentioned virus safety, many of the following problems may arise: the main body cannot be simplified until the downstream; the result cannot avoid the complexity of the management of the manufacturing steps, and the cost is high or unstable. In the case of the protein of the trait, the stability of the subsequent concentration step cannot be ensured, and formulation or the like is difficult. [Patent Document 1] Japanese Patent Laid-Open Publication No. 2001-503781 [Non-Patent Document 1] "Latest Microbiology Manual" (丨 986) [Non-Patent Document 2] "Bio Industry" 4, 554 (1984) [Non-Patent Document 3] "The Fermentation Handbook" (edited by the Institute of Fermentation and Metabolism of the Bi〇industry Association) (ISBN 4-320-05575-6) [Non-Patent Document 4] "Chemical Products of 14303" (Chemical Industry News) (ISBN 4-87326-404-9) [Non-Patent Document 5] Homepage of the Independent Administrative Corporation Science and Technology Promotion Agency (JST) http//ww.go.jp/pr/report/report68/details.html [Non-Patent Document 6 In general, the problem of the problem is to solve the problem of the invention. In view of the above problems, an object of the present invention is to provide a method for producing a monoclonal antibody, which can be Achieving a high concentration of a single useful substance having a polypeptide, 139962.doc 201002821, a high permeability and virus removal of the preparation solution, and the extract Z contains a source derived from the substance or the source (4) Formulations that incorporate risky stabilizers can be easily used for each The formulation manufacturing processes. In view of this issue, the most difficult to solve is the virus removal step. In terms of the characteristics of the membrane for removing the virus by the mechanism for removing the size (refer to the following [Embodiment]), the permeability of the virus removal membrane is generally low, and it is generally difficult to adapt to the transition of a higher concentration of the preparation. Become the biggest = problem in the manufacturing steps. Furthermore, the permeability of the virus removal step is proportional to the pore size of the dense layer of the virus removal membrane, and the virus-removing system is inversely proportional to the pore size of the dense layer. Therefore, the two technologies having the high performance and the relative properties become technologies. The topic. [Technical means for solving the problem] ... The inventors conducted intensive studies to solve the above problems, and as a result, obtained the present invention. That is, the present invention includes the following. (1) A method for producing a preparation comprising a single useful substance having a polypeptide, characterized in that it comprises at least the following steps: a step of cultivating a useful substance having a polypeptide early, 'a single useful substance having a polypeptide is pure: a purification step of adding at least one stabilizer addition step for inhibiting a cohesive stabilizer having a polymorphic single substance = a virus-removed virus in a solution of a single-useful substance having a polypeptide by a virus removal membrane a step of sealing the single useful substance having the polypeptide with the virus removed, and the above-mentioned virus removal step is derived from the disease of the single-useful substance having the polypeptide Stabilizer production The disease removal ability of the disease removal step is LRvy, 139962.doc 201002821 The concentration of a single useful substance having a polypeptide in the virus removal step is in the range of 3.0 to 10.0% by weight, and is contained in the solution. A single substance having a polypeptide: the useful substance is removed relative to the virus used in the above virus removal step: after the start of the membrane (2) The average permeability of the fraction # and the passage of 2 to 3 hours was 1.0 (kg/m2/hour) or more. [2] A method of producing a preparation comprising a single useful male meringue having a polypeptide, wherein the stabilizer is derived from a living organism or derived from an acid yeast product, as in [1]. Or a method for producing a preparation comprising a single-useful substance f of a polypeptide, wherein the stabilizer is selected from the group consisting of a saccharide, an amino acid, an amino sugar, an organic acid, or a derivative thereof in. [4] The method for producing a preparation comprising a single useful substance having a polypeptide according to any one of (1) to [3], wherein the virus removal membrane is a hollow fiber membrane comprising a synthetic polymer. [5] As included in any of [1] to [4], there are many. In addition, the preparation method of the preparation of the early useful substance of the peptide, the upper layer of the pot, and the base of the forgotten soil. The diseased mites are removed into a multi-layer microporous membrane. [6] A preparation comprising a monoclonal antibody, wherein the method comprises at least the following steps: a step of culturing a single antibody, and a purification step of purifying the individual antibody Adding at least a lambda stabilizer addition step for inhibiting the aggregation of the early antibody, and removing the virus containing the monoclonal antibody solution from the virus removal membrane in the main vortex removal a removal step, and a sealing step of sealing the virus-removed monoclonal antibody. The virus removal step is derived from the production of the monoclonal antibody, and the production of the stabilizer. When the virus is removed, the virus removal ability of the virus removal step τ ν 乡 is LRV = 4, and the concentration of the monoterpenoid oxime is 3.0 to 10% by weight in the virus removal step. 139962.doc The range of the solution in the range of '02102821' The average permeability of the strain antibody was 1.0 (kg/m 2 /h 〇 ur) or more after two minutes and two hours after the start of filtration of the virus removal enthalpy used in the virus removal step. [7] The method for producing a preparation comprising a monoclonal antibody according to [6], wherein the above-mentioned stable Qishen is derived from a living organism or derived from a fermentation product. [8] The method for producing a preparation comprising a monoclonal antibody according to [6] or [7], wherein the above-mentioned stabilizer is selected from the group consisting of a saccharide, an amino acid, an amino sugar, an organic acid, or the like .

[9] 如[6]至[8]中任一項之包含單株抗體之製劑之製造方 法’其甲上述病毒去除膜為包含合成高分子之中空纖維 膜。 [10] 如[6]至[9]巾任-項之包含單株抗體之製劑之製造方 法,其中上述病毒去除膜為多層微多孔膜。 [11] 如[6]至[1G]中任_項之包含單株抗體之製劑之製造方 法’其中上述穩定劑添加步驟中各穩定劑之添加量分別為 0.1〜15重量。/〇之範圍,且穩定劑添加步驟結束時及密封步 驟結束時之單株抗體純度為8〇%以上。 [發明之效果] 藉由使用本發明’可製造實現較高之透過性及病毒去除 性兩者之高品質的高濃度單株抗體溶液製劑或重組製劑之 製造中間物。並且,即便利用七人Ή A L u 1 從W用包含源自生物或源自醱酵產 物之存在病毒混人風險的穩定劑之製劑,亦可製造上述^ 品質製劑或製造中間物。因挤,可担# ^ 口此,可提供一種能夠簡便地用 於各種製劑之製造製程、包含以罝矬P触4 ± 13以早株抗體為代表的具有多 139962.doc 201002821 肽之單-有用物質之製劑之製造方法,藉此可總括地 製造步驟之簡略化、精簡化、低成本化等。 【實施方式】 本發明t提出之所謂包含具有多肽之單—有用物質之^ 劑之製造方法,係指至少包括如下步驟者:藉由細菌衣 ra〇(chlnese hamster 〇vary,中國倉鼠印巢)細胞、融合瘤 等來產生具有多肽之單一有用物質之培養步驟;利用層析 儀、超濾膜、精緻純化(polishing)等將具有多肽之單二有 質純化之純化步驟;添加抑制具有多肽之單一有用物 貝單株抗體之凝聚的穩定劑之穩定劑添加步驟;利用病毒 去除膜將包含單株抗體之蛋白質溶液中之病毒去除的病: 去除步驟;以及將去除了病毒之單株抗體密封之密封步 :。所謂上述培養步驟中培養之具有多肽之單一有用物 質’係指藉由將基因重組技術應用於細菌或ch〇細胞並進 行培養而產生之重組蛋白f或單株抗體、或者利用細胞融 合等技術對融合瘤等本質上產生單—㈣物質之細胞進行 培養而產生之單株抗料,其應用並㈣別限制,更具體 可列舉:&因重組菌落刺激因+、基因重組紅血球生成 素基因重組干擾素、基因重組融合蛋白質、單株抗體、 人源化抗體、嵌合抗體等。上述病毒去除步驟中重要之評 價項目係製劑中間產品於病毒去除步驟中之過渡速度與病 毒去除能力。該步驟之目的在於將源自單株抗體之產生時 之病毒、與源自穩定劑之產生時之病毒去除。因此,尤其 係病毒混人風險較高之源自生物或源自醱酵產物之添加物 139962.doc 201002821 的添加步驟必須於南丨田 利用病毒去除膜之病毒去除步驟之前、 行。就無機鹽或石油產品、或^之則進 自生物或源自醋酵產物之D成者非源 並無特別限制。於病毒二,其添加步驟之順序 '病毒過濾後進行利用UF膜等 步驟之情形時,通常隨著溶液成分之調整而 辰= 定劑,因此較理想的是不具有此步驟。 貞再-加穩 如上所述,作為本方法之目的之-,可列舉:藉由在勺 含具有多肽之單-有用物質之製劑之最終步驟中2 物或源自酸酵產物之穩定劑與製劑一併進行過據,而對制 造稳定劑時混入病毒之風險實現更本質之安全化。所謂:: 自生物或源自醱酵產物之穩定劑,係指與「藉由 醱 酵而獲得之葡聚糖」等真菌、細菌酸酵產物或者「由疋Γ 來源物經過酵素反應而獲得之神經胺糖酸」等將自生= 接提取所得之產物料原料而獲得之產㈣相當者(專利 文獻1)。關於該等穩定劑之利用醱酵法之具體生產方法, 已由大量文獻等而公知,且已以工業規模進行生產(非專 利文獻1〜5)。所謂該等敎劑中混人病毒之風險係指:例 如使用醱酵法之情形時’培養牛金清等源自生物或源自醱 酵產物之穩定劑時使用源自生物之原料之情形時,或者於 將蛋黃等源自生物之材料(利用酵素等)分解而獲得目標穩 定劑等,於使用任何源自生物之材料製造穩定劑之步= 源自原料、人會感染之病毒混入,該病毒於穩定劑之製造 步驟中並未被充分去除或鈍化之情形時,源自該等原料之 病毒混入之風險存在。 139962.doc -11 - 201002821 無=:12:,:自_產物之穩定劑之種類並 更理'-的疋糖類(作為代表性者,有單糖、 :或;寡糖、糖醇等)、胺基酸、胺基糖、有機 萄糖二Γ ’進而理想的是,作為糖類,有葡 讀、半乳糖、果糖、山梨糖1糖、乳糖、海 ^糖/搪、棉子糖、葡聚糖、甘露糖醇、山梨糖醇、 醇;作為胺基酸,有白胺酸、異白胺酸、丙胺酸、 :::、麵胺酸、天冬胺酸、苯基丙胺酸、路胺酸、色胺 二脯:酸、經基脯胺酸、瓜胺酸;作為胺基糖,有葡萄 士礼胺糖、唾液酸;作為有機酸,有α-酮戊二酸、 2’:甸糖酸、5_酮葡萄糖酸、抗壞血酸、異抗壞血酸、 別異檸檬酸、異檸檬酸、衣庵 缺, 义衣康酸、己酸、檸檬酸、葡萄糖 -夂、琥賴、丙S同酸、反丁烯二酸、甲基二經戊酸、薄果 酸、麴酸、酒石酸、乙酸、乳酸、草酸、2,5_二嗣葡萄糖 酸、2-酮古洛糖酸。此處所謂衍生物等,係指缓基及經基 之醋化物、胺基及經基之貌基取代物或酿基取代物等,若 為具有酸性或鹼性部位者則係指該等之鹽’或該等中之複 數個之經衍生者的組合。關於鹽之種類,只要在作為: 品、醫藥品之添加物而法律上允許之範圍内則無特別限 制。 所謂具有多肽之單-有用物質之多聚體、凝聚體等,係 指具有多肽之單-有用物質科學鍵結而形成二聚物或三聚 物以上之凝聚體者、或者2個以上之且右夕& 〇α ,、’夕肽之早一有用 物質由於分子間力或任何原因(變性等)而雜礼凝聚者。 139962.doc 12 201002821 時本=二:當穩定劑添加步驟中穩定劑之添加量過低 果,=得具有多肽之單-有用物質之充分之穩定效 因此::二’病毋過濾步驟中可能會引起透過性之下降, ==添加步驟中各敎劑之添加量較理想的是分別 為0.1〜15重量%之範圍, 相 進而理想的是。 時具有多肽之單一有用物, @ ’右控封步驟結束 A '彳 貝之純度為8()%以上,則可看作 -于V刀之穩定效果,更理想的是85%以上,進而理相 的是9_上,最理想的是9制上。 心 肽之單—有用物質溶液之透過性通常依存於抗體 〜夜派度、及入口壓力。即,透過性與具有多狀之單一有 用物質之濃度為負相關’若蛋白濃度變高則有過渡速度下 降之傾向’另-方面,無論蛋白濃度如何,透過性與對且 有多肽之單-有用物質溶液進行過濾時之入口壓力為正相 關。因此,為保持具有多肽之單一有用物質之濃度為高濃 度之狀態進行過遽而不使過濾、速度下降,較理想的是病毒 去除膜本身之㈣性為_咖以上。作為具有_咖以 上之财壓性之膜的材質’例如較理想的是將聚偏二氟乙烯 (PVDF,P〇lyvinylidene flu〇ride)或聚趟石風(pEs,驗 sulfone) '聚硬(PS,polysulf_)之類的合成高分子作為原 材料且具有親水性之膜。關於膜之形狀,可利用平板膜姓 構或中空纖維結構者,較好的是以中空纖維結構為宜。 又’膜之結構較理想的是如下多層微多孔膜:其係具有開 孔率較大之粗大結構層、及開孔率較小之緻密結構層之包 139962.doc 201002821 含熱塑性樹脂的微多孔膜,並且該粗大結構層存在於至少 方之膜表面且其厚度為5.0 μιη以上,該緻密結構層之厚 度為膜厚整體之5〇%以上,並且該粗大結構層與該緻密結 構層成-體化。進而好的是,上絲大結構層係開孔率為 膜厚整體之平均開孔率+2.〇%以上之層,上述緻密結構層 係開孔率未滿膜厚整體之平均開孔率+2〇%、且冑於[未滿 膜厚整體之平均開孔率+ 2狀層之部分開孔率的平均 值]销(包括兩端)之範圍内的層。上述粗大結構層較理 想的疋部分開孔率自膜表面朝向緻密結構層連續地減少之 傾斜結構、且僅存在於一方之膜表面。 如上所述,透過性與具有多肽之單—有用物質之濃度為 負相關,若蛋白濃度變高則有透過性(過濾速度)下降之傾 向病幸過濾步驟係於具有多肽之單一有用物質溶液的濃 度為3.〇 10重量%之範圍内實行,更理想的是3.0〜7.5重量 /〇之範圍,進而理想的是3 〇〜5 〇重量%之範圍最理想的 疋3 _0〜4_〇重量%之範圍。於滿足以上壓力及單株抗體濃度 之範圍内,單株抗體之單位膜面積及單位時間之透過性較 理心的疋1.0(kg/m2/hour)以上,更理想的是! 5(kg/m2/h〇ur) 以上進而理想的是2.0(kg/m2/hour)以上。 對於利用病毒去除膜進行之病毒去除步驟之入口壓力, 高壓具有可於短時間内迅速進行過濾之優點,但若壓力過 问則C力對裝置之損傷或洩漏之風險增大,故病毒去除 乂驟係於入口壓力為1 50 kPa以上、600 kPa以下而實施, 較理心的疋196 kPa以上、500 kPa以下,更理想的是245 139962.doc -14 - 201002821 kPa以上、400 kPa以下,進而理想的是294 kPa以上、4〇0 kPa以下’最理想的是294〜300 kPa。 於本1明中,作為用以顯示病毒過據步驟中具有充 分之透過性、且未產生急遽之透過性下降之指標,過濾開 始後經過10分鐘時、與過濾開始後經過2〜3小時之時的包 έ具有夕肽之單一有用物質之製劑溶液的平均透過性必須 均為1.0(kg/m2/h〇ur)以上,較理想的是丨25(kg/m2/h〇ur)以 上,進而理想的是1 ·5(1^/Γη2/1ι〇ιΙΓ)以上。上述過濾開始後 經過1 〇分鐘時(或過濾開始後經過3小時之時)之平均透過性 的计算,可根據〇〜1〇分鐘整體(或2〜3小時整體)之透過量 進行倒算而求出。即,測定1〇分鐘(或2〜3小時)時間點之 透過畺之重畺,算出膜之每單位面積之重量,將其除以10 分鐘(或60分鐘),由此計算出IgG透過量[kg/m2/h]。 為了達成此種所需之透過性,可列舉如下方法:由於蛋 白之堵塞而使用耐蛋白附著性能較高之病毒去除膜,使用 不會因蛋白之堵塞而引起透過性下降之膜,抑制堵塞之最 大要因即包含具有多肽之單一有用物質之製劑之凝聚體的 形成,控制用以維持膜之透過性之透過壓,在增大壓力以 提高透過性時使用具有耐壓性之膜等。 關於病毒去除膜之病毒去除能力,應根擄ICH (International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for[9] The method for producing a preparation comprising a monoclonal antibody according to any one of [6] to [8] wherein the virus removal membrane is a hollow fiber membrane comprising a synthetic polymer. [10] A method for producing a preparation comprising a monoclonal antibody according to [6] to [9], wherein the virus removal membrane is a multilayer microporous membrane. [11] A method for producing a preparation comprising a monoclonal antibody according to any one of [6] to [1G] wherein the amount of each stabilizer added in the stabilizer addition step is 0.1 to 15% by weight. The range of /〇 is, and the purity of the individual antibody at the end of the stabilizer addition step and at the end of the sealing step is 8% or more. [Effects of the Invention] By using the present invention, it is possible to produce a high-quality single-concentration antibody solution preparation or a recombinant preparation of a recombinant preparation which achieves both high permeability and virus-removability. Further, even if a seven-person Ή A L u 1 is used for the preparation of a stabilizer containing a stabilizer derived from a living organism or a virulence-derived product, the above-mentioned quality preparation or intermediate can be produced. Because of the squeezing, it can provide a simple process for the preparation of various preparations, including a single 139962.doc 201002821 peptide represented by 罝矬P touch 4 ± 13 as an early strain antibody - A method for producing a preparation of a useful substance, whereby the steps of the manufacturing step can be simplified, simplified, reduced in cost, and the like. [Embodiment] The method for producing a so-called "substance-containing substance having a polypeptide" as proposed by the present invention refers to a method comprising at least the following steps: by bacterium clothing ra (chlnese hamster 〇vary, Chinese hamster printing) a step of culturing a single useful substance having a polypeptide by using a cell, a fusion tumor, or the like; and purifying the single-purity purification of the polypeptide by using a chromatograph, an ultrafiltration membrane, a polishing, or the like; Stabilizer addition step of a stabilizer for agglomeration of a single useful monoclonal antibody; removal of a virus in a protein solution containing a monoclonal antibody by a virus removal membrane: a removal step; and sealing of the individual antibody from which the virus has been removed Sealing step: The single useful substance having a polypeptide cultured in the above-mentioned culture step refers to a recombinant protein f or a monoclonal antibody produced by applying a genetic recombination technique to a bacterial or ch〇 cell and cultured, or a technique using cell fusion or the like. A single tumor resistant material produced by culturing a cell containing a single-(four) substance in nature, and its application is (4) not limited, and more specifically: & recombinant colony stimulating factor +, recombinant erythropoietin gene recombination Interferon, gene recombinant fusion protein, monoclonal antibody, humanized antibody, chimeric antibody, and the like. An important evaluation item in the above virus removal step is the transition speed and virus removal ability of the intermediate product of the preparation in the virus removal step. The purpose of this step is to remove the virus from the production of the monoclonal antibody and the virus from the generation of the stabilizer. Therefore, in particular, the addition step of the organism-derived organism-derived additive derived from the organism or from the fermented product 139962.doc 201002821 must be performed before the virus removal step of the virus removal membrane in Nantun. There is no particular limitation on the inorganic salt or petroleum product, or the non-source of the D-forming agent which is derived from the organism or derived from the vinegar-derived product. In the case of virus 2, the order of the addition step is as follows: when the virus is filtered and the step of using a UF membrane or the like is performed, it is usually adjusted with the composition of the solution, and therefore it is preferable to have no such step. Further, as described above, as the object of the method, there may be mentioned a stabilizer or a stabilizer derived from an acid yeast product in a final step of a preparation containing a single-useful substance having a polypeptide in a spoon. The preparation has been carried out together, and the risk of mixing the virus in the manufacture of the stabilizer is more essentially safe. The so-called:: a self-biological or stabilizer derived from a fermented product refers to a fungus, a bacterial acid yeast product such as a "glucan obtained by fermentation," or a "reacted from an enzyme by an enzyme reaction." The production of the "neuraminic acid" or the like is obtained by the production of the raw material of the product obtained by the extraction of the raw material (four) (Patent Document 1). The specific production method for the use of the stabilizers by the fermentation method has been known from a large number of documents and the like, and has been produced on an industrial scale (Non-Patent Documents 1 to 5). The risk of mixing human viruses in such tinctures means, for example, when using a fermentation method, when cultivating a raw material derived from living organisms derived from a biological or a stabilizer derived from a fermented product, or A step of decomposing a biologically derived material such as egg yolk (using an enzyme or the like) to obtain a target stabilizer, etc., and using a stabilizer derived from a material derived from a living organism = a virus derived from a raw material and infected by a human, the virus is mixed with In the case where the stabilizer is not sufficiently removed or passivated in the manufacturing step, there is a risk of virus incorporation from the raw materials. 139962.doc -11 - 201002821 No =: 12:,: The type of stabilizer from the product and more '------------------------------------------------------------------------------------------------------------- Amino acid, amino sugar, and organic sugar bismuth ' Further desirable as sugar, there are Portuguese, galactose, fructose, sorbose 1 sugar, lactose, sea sugar / cockroach, raffinose, Portuguese Glycan, mannitol, sorbitol, alcohol; as amino acid, there are leucine, isoleucine, alanine, :::, face acid, aspartic acid, phenylalanine, road Amine acid, tryptamine diterpene: acid, trans-proline glutamic acid, citrulline; as amino sugar, grape glucosamine, sialic acid; as organic acid, α-ketoglutaric acid, 2': Dyrosic acid, 5-ketone gluconic acid, ascorbic acid, isoascorbic acid, iso-citric acid, iso-citric acid, sputum deficiency, prohexitol, hexanoic acid, citric acid, glucose-glucone, amber, C-acid , fumaric acid, methyl divaleric acid, thin fruit acid, citric acid, tartaric acid, acetic acid, lactic acid, oxalic acid, 2,5-dioxalic acid, 2-ketogulonic acid. Here, the term "derivative" or the like refers to a base of a buffer or a base of a buffer, an amine group, and a base group, or a base substituent, etc., and if it has an acidic or basic moiety, it means such a Salt' or a combination of a plurality of such derivatives. There is no particular limitation on the type of the salt as long as it is legally permitted as an additive to the product or the pharmaceutical product. The term "polymer, agglomerate, and the like of a single-useful substance having a polypeptide" means a substance in which a single-use substance having a polypeptide is scientifically bonded to form a dimer or a trimer or more, or two or more Right eve & 〇α,, 'Early peptide, a useful substance, due to intermolecular forces or any cause (denaturation, etc.). 139962.doc 12 201002821 时本=二: When the stabilizer addition step is too low in the stabilizer addition step, = sufficient stability of the single-useful substance with the polypeptide is obtained. Therefore: the second 'sickness filter step may be The decrease in permeability is caused, and the addition amount of each of the tanning agents in the addition step is preferably in the range of 0.1 to 15% by weight, and further preferably. When there is a single useful substance of the polypeptide, @ 'right control sealing step ends A 'mussel purity is 8 ()% or more, it can be regarded as - the stability effect of the V knife, more desirably 85% or more, and further The phase is 9_, the most ideal is the 9 system. The permeability of a single peptide-soluble substance solution usually depends on the antibody to the nighttime and the inlet pressure. That is, the permeability is negatively correlated with the concentration of a single useful substance having a polymorphism. 'If the protein concentration becomes high, there is a tendency for the transition velocity to decrease'. In addition, regardless of the protein concentration, the permeability and the peptide are mono- The inlet pressure when filtering with a useful substance solution is positively correlated. Therefore, in order to maintain the concentration of a single useful substance having a polypeptide at a high concentration without causing filtration and a decrease in speed, it is preferable that the (four) property of the virus removing film itself is _ or more. As a material having a film having a financial property of more than _ café, for example, it is preferable to use poly(vinylidene fluoride) (PVDF, P〇lyvinylidene flu〇ride) or poly sapphire (pEs, test sulfone) to gather hard ( A synthetic polymer such as PS or polysulf_) is used as a raw material and has a hydrophilic film. Regarding the shape of the film, a flat film structure or a hollow fiber structure may be used, and a hollow fiber structure is preferred. Further, the structure of the film is preferably a multilayer microporous film having a large structural layer having a large opening ratio and a dense structural layer having a small opening ratio. 139962.doc 201002821 Microporous containing a thermoplastic resin a film, and the coarse structural layer is present on at least the surface of the film and has a thickness of 5.0 μm or more, the thickness of the dense structural layer being more than 5% by weight of the entire film thickness, and the coarse structural layer and the dense structural layer are - Physicalization. Further preferably, the large-structure layer of the upper filament is an average opening ratio of the film thickness of +2. 〇% or more, and the average opening ratio of the dense structure layer is less than the entire film thickness. +2〇%, and 胄 [the average opening ratio of the entire film thickness + the average of the partial opening ratio of the 2-layer layer] the layer within the range of the pin (including both ends). The above-mentioned coarse structural layer is preferably an inclined structure in which the opening ratio of the crucible portion continuously decreases from the film surface toward the dense structural layer, and exists only on one of the film surfaces. As described above, the permeability is negatively correlated with the concentration of the single-useful substance having the polypeptide, and if the protein concentration becomes high, the permeability (filtration rate) is lowered. The filtering step is based on a single useful substance solution having a polypeptide. The concentration is in the range of 〇10% by weight, more preferably in the range of 3.0 to 7.5 wt/〇, and further preferably in the range of 3 〇~5 〇% by weight. The most ideal 疋3 _0~4_〇 weight The range of %. Within the range of satisfying the above pressure and the concentration of the individual antibody, the permeation of the unit membrane area per unit time and the per unit time is more than 1.0 (kg/m2/hour), more preferably! 5 (kg/m2/h〇ur) or more is preferably 2.0 (kg/m2/hour) or more. For the inlet pressure of the virus removal step using the virus removal membrane, the high pressure has the advantage of being able to rapidly filter in a short time, but if the pressure is excessive, the risk of damage or leakage of the C force to the device increases, so the virus is removed. The system is carried out at an inlet pressure of 1 50 kPa or more and 600 kPa or less, and is more preferably 196 kPa or more and 500 kPa or less, more preferably 245 139962.doc -14 - 201002821 kPa or more and 400 kPa or less. It is ideally 294 kPa or more and 4 〇 0 kPa or less 'the most ideal is 294 to 300 kPa. In the present invention, as an indicator for indicating that the virus has sufficient permeability in the passing step and that there is no rapid decrease in permeability, 10 minutes after the start of filtration and 2 to 3 hours after the start of filtration. The average permeability of the preparation solution of the single useful substance having the oxime peptide must be 1.0 (kg/m 2 /h 〇 ur) or more, and more preferably 丨 25 (kg/m 2 /h 〇 ur) or more. Further, it is preferably 1·5 (1^/Γη2/1ι〇ιΙΓ) or more. The calculation of the average permeability after 1 minute of the above filtration (or 3 hours after the start of filtration) can be calculated based on the total amount of 〇~1〇 minutes (or 2 to 3 hours overall). Out. That is, the weight per unit area of the film was measured at a time point of 1 minute (or 2 to 3 hours), and the weight per unit area of the film was calculated and divided by 10 minutes (or 60 minutes) to calculate the IgG permeation amount. [kg/m2/h]. In order to achieve such a desired permeability, a virus removal membrane having high protein adhesion resistance is used due to clogging of the protein, and a membrane which does not cause a decrease in permeability due to clogging of the protein is used, and clogging is suppressed. The most important factor is the formation of agglomerates of a preparation containing a single useful substance of a polypeptide, controlling the permeation pressure for maintaining the permeability of the membrane, and using a membrane having pressure resistance when the pressure is increased to improve the permeability. Regarding the virus removal ability of the virus removal membrane, it should be based on ICH (International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for

Human Use ’日美EU(European Union,歐盟)國際醫藥法 規協合會)準則等公知之要求事項而決定。即規定,出於 139962.doc •15- 201002821 以下目的而進行人為地添知古、产主λ咖士 巧c办、加有两毋之製劑溶液之過濾試 驗:分析製造步驟於病毒之去除或純化方面通常具有何種 程度之能力,即分析步驟於媒實地發揮病毒清除能力方面 之特性(robustness,穩健性)。病毒去除率lrv係以對數值 來表示以病毒去除膜進行過渡後之溶液之病毒量相對於過 濾丽之溶液之病毒量的下降率者,通常係於將實際之製造 步驟準確地料之實驗室水平之小規模過濾操作中進行該 評價。 作為通吊所使用之病毒之定量法,可列舉溶斑法、 丁 CID50法等(非專利文獻6)。5一方自,本方法中所使用 之病毒去除膜係依據單株抗體與病毒尺 機構將病毒去除。因此,目前公知且最難以利用筛: 構去除之病毒係具有最小之大小的小病毒科的病毒(直徑 為1 8 25 nm)。本方法中所使用之病毒去除膜可使用豬小 病毒(PPV,pig parvovirus)或小鼠微小病毒(mvm, ☆us 〇f mice)、犬小病毒(cpv’咖心pa削心㈣等來作 為刀析上述特性(robustness)方面之相當於最壞情況之病 毋於本發明中,作為一例,示出使用由豬小病毒(ppv) 所仔之病毒的病毒去除性試驗。 豬小病毒(PPV)可利用TCID50法進行定量。以下表示詳 細順序之一例。將會感染豬小病毒之任意細胞之加入有3 體積。/。牛血清(Upstate公司製造’於56。〇之水浴中加熱儿分 鐘而失活後使用)iD_MEM(Gibc〇製造,Mgh_gluc〇se)懸浮 ' 與s有豬小病毒之包含具有多肽之單一有用物質之製 139962.doc -16- 201002821 劑溶液以1 . 1之比例加以混合。同樣,對於相同之細胞懸 浮液’將相同之含有豬小病毒之包含具有多肽之單一有用 物質之製劑溶液的10倍稀釋液以丨:i之比例混合。以下, 以同樣之順序’對相同之含有則、病毒之包含具有多狀之 單一有用物質之製劑溶液的102倍、103倍、104倍、105 倍、1〇6倍、107倍稀釋液亦進行混合。就各病毒溶液或稀 釋液分別準備8份該等混合液,分別於37它、5%二氧化碳 氣體環境下之培養箱中進行培養。繼而,對經細胞培養之 各懸浮液利用紅血球吸附法(非專利文獻6)進行 TCID50(5〇%感染價)之測定。即,以pBS㈠(日水製藥股份 有限公司製造,利用商品隨附之方法而製備)將雞紅血球 稀釋成5倍,以2500(rpm)進行5分鐘離心分離,去除上清 液。再次以PBS(-)將所獲得之保存血稀釋液之殘渣稀釋成 200倍後,以與上述細胞懸浮液相等之量添加至經細胞培 養之各培養液中,靜置1〜2小時。目測確認經培養之細胞 組織之表面有無紅血球之吸附,將確認到吸附者作為引起 了病毋感染之培養液而進行計數。關於所得各培養液之病 毒感染之有無’就最初添加至細胞懸浮液中之各含有緒小 病毒之包含具有多肽之單一有用物質的製劑溶液或稀釋液 濃度分別確認看到感染之培養液的比例,利用Reed_Munch 法(非專利文獻6)計算出log(TCID5〇/mL)作為感染價。對利 用病毒去除膜進行過濾前後之兩方之溶液進行上述操作, 利用下述式計算LRV :The Human Use ‘Japan-EU (European Union) International Medicine Law Association) guidelines are determined by known requirements. That is to say, for the following purposes of 139962.doc •15-201002821, artificially add the filtration test of the preparation solution of the ancient, the owner λ café, and the addition of two preparations: analysis of the manufacturing steps in the removal of the virus or The extent to which purification generally has the ability to analyze the steps to achieve virality in terms of viral clearance. The virus removal rate lrv is a logarithmic value indicating the rate of decrease in the amount of virus in the solution after the transition of the virus removal membrane relative to the amount of virus in the filtered solution, usually in a laboratory that accurately measures the actual manufacturing steps. This evaluation was carried out in a horizontal small-scale filtration operation. The quantitative method of the virus used for the sling is, for example, a speckle method, a DID CID50 method, or the like (Non-Patent Document 6). 5 From one, the virus-removing membrane used in the method removes the virus according to the monoclonal antibody and the virus ruler mechanism. Therefore, it is currently known and most difficult to utilize a sieve: the virus removed by the structure is the virus of the small virus family (the diameter is 1 8 25 nm) having the smallest size. The virus-removing membrane used in the method can be used as a pig virus (PPV, pig parvovirus) or a mouse parvovirus (mvm, ☆us 〇f mice), a canine parvovirus (cpv' café heart-shaving heart (four), etc. A disease equivalent to the worst case in terms of the above-mentioned characteristics is described in the present invention, and as an example, a virus removal test using a virus vaccinated with porcine virus (ppv) is shown. Pig prion (PPV) The TCID50 method can be used for quantification. An example of a detailed sequence is shown below. Any cell that infects porcine virulence virus is added in a volume of 3. The bovine serum (manufactured by Upstate Inc.) is heated in a water bath of 56. After inactivation, iD_MEM (Mig_gluc〇se) suspension is mixed with s. Similarly, for the same cell suspension, the same 10-fold dilution of the preparation solution containing the porcine parvovirus containing a single useful substance having the polypeptide is mixed in the ratio of 丨:i. The procedure is as follows: 102, 103, 104, 105, 1, 6 and 107 times dilutions of the preparation solution containing the same useful substance as the virus, and the virus is mixed. Each of the solution or the diluent is prepared by separately preparing 8 such mixtures, and culturing them in an incubator under a 5% carbon dioxide atmosphere, respectively, and then using the red blood cell adsorption method for each suspension of the cell culture (Non-Patent Document 6) Measure the TCID50 (5〇% of the infection price). That is, the chicken red blood cells are diluted 5 times with pBS (1) (manufactured by Rishui Pharmaceutical Co., Ltd., using the method attached to the product), and 2500 (rpm) is used. The supernatant was removed by centrifugation in a minute, and the residue of the obtained preserved blood dilution was diluted to 200-fold with PBS (-), and then added to each culture medium of the cell culture in an amount equivalent to the above-mentioned cell suspension liquid phase or the like. In the middle, it was allowed to stand for 1 to 2 hours, and it was confirmed by visual observation whether or not the adsorption of the red blood cells on the surface of the cultured cell tissue was observed, and it was confirmed that the adsorbent was counted as a culture solution causing the disease infection. The presence or absence of a virus infection is determined by the ratio of the concentration of the preparation solution or the diluent containing the single useful substance having the polypeptide, which is initially added to the cell suspension, and the concentration of the culture solution in which the infection is observed, using the Reed_Munch method (non- Patent Document 6) Calculates log (TCID5〇/mL) as the infection price. The above operation is performed on both solutions before and after filtration using the virus removal membrane, and LRV is calculated by the following formula:

LRV=l〇g10A-l〇g10B 139962.doc •17· 201002821 之溶液之感染價 其中,A=利用病毒去除膜過遽、前 (TCID50/mL); 渡後之溶液之感染價 本發明並不限定於利用 B =利用病毒去除臈過 (TCID50/mL)。 然而,此係病毒定量法之一例, §亥方法進行之病毒定量法。 作為本方法中所使用之病毒去除膜於分析上述特相 (robustness)之試驗中表現出充分之病毒去除性之值,對汽 有種類之病毒而言’於相當於進行了 , 疋仃ί 3小時製劑溶液之發 濾時之總過濾量的溶液之總量混合液中 0Υ,右乙1〇/24則為交 分,更理想的是LRVS5。 本方法亦可容易地應用於具有上述製造步驟之任何生物 醫藥品之製造步驟,較好的是可應用於單株抗體、人源化 抗體、嵌合抗體、基因重組菌落刺激因子、基因重組紅血 球生成素、基因重組干擾素、基因重組融合蛋白質之製造 步驟,更好的是可應用於人類單株抗體,進而好的是可應 用於人類γ球蛋白G。 關於病毒去除膜之膜面積之調整,已廣泛知悉其於規模 放大、規模縮小時亦無較大之干擾因素,故具有如下特 徵:可容易地進行規模放大、規模縮小,⑹而容易獲得可 調能力。即,若膜面積改變則僅單位膜面積之過濾體積改 變,可認為若其為固定則過濾性能得以維持,因此可利用 與貫際生產規模成比例之任意之膜面積。 本方法中’利用病毒去除膜進行單株抗體溶液之過濾之 139962.doc •18- 201002821 介曰、、…特別限制。於即便病毒去除過遽器之膜面積 亦欲於短時間内完成製造步驟中之病毒去除步驟之LRV=l〇g10A-l〇g10B 139962.doc •17· 201002821 The infection price of the solution, where A=use the virus to remove the membrane over the sputum, before (TCID50/mL); the infection price of the solution after the ferry is not Limited to use B = use virus to remove 臈 (TCID50 / mL). However, this is an example of a virus quantification method, and the virus quantification method by the § Hai method. The virus-removing film used in the method exhibits sufficient value of virus removal in the test for analyzing the above-mentioned particularity, and is equivalent to the virus of the steam type, 疋仃ί 3 The total amount of the solution in the filtration solution of the hourly preparation solution is 0 Υ in the total mixture, and the right 乙 〇 / 24 is a cross, more preferably LRVS5. The method can also be easily applied to the manufacturing steps of any biopharmaceutical having the above manufacturing steps, and is preferably applicable to monoclonal antibodies, humanized antibodies, chimeric antibodies, recombinant colony stimulating factors, and recombinant red blood cells. The production steps of the genomicin, the recombinant interferon, and the recombinant fusion protein are more preferably applied to human monoclonal antibodies, and thus can be applied to human gamma globulin G. Regarding the adjustment of the membrane area of the virus-removing membrane, it has been widely known that there is no large disturbance factor in scale-up and scale-down, so it has the following characteristics: scale-up and scale reduction can be easily performed, and (6) can be easily adjusted. ability. That is, if the membrane area is changed, only the filtration volume per unit membrane area is changed, and it is considered that if the filtration performance is maintained, the membrane area can be maintained in any ratio proportional to the production scale. In the present method, the filtration of a single antibody solution is carried out by using a virus removing membrane. 139962.doc • 18- 201002821 Introduction, ..., particularly limited. In order to complete the virus removal step in the manufacturing step in a short time even if the virus removes the membrane area of the device

% ’可u為短時間’於即便時間稍長亦欲減小病毒切 過遽器之膜面積之情形時,可設定為長時間。X,作為利 用病毒去除臈進行單株抗體料之過濾之條件,過料、·容 液之溫度、室溫、無機鹽等並非源自生物或醱酵物之添加 物之有無、溶劑之種類、並非源自包含具有多肽之單 用物質之製劑的失雜物之有無並無特別限制。 [實施例] 以下實施例中,使用如下過渡襄置、SUS304製槽及石夕 管mgers P〇lymer製造),上述過遽裝置係使用將親水化聚 偏二氟乙烯作為#質之中空纖維膜來作為病毒去除膜,且 於合器内工間申’巾空纖維膜被隔在入口側空間與出口側 空間之間;上述SUS304製槽及矽管係用於向過濾裝置輸 送單株抗體溶液,已於121。(:下進行了 15分鐘蒸氣殺菌。 作為單株抗體製劑中間產品之模型,係恢據 W02004/087761中記載之方法製備單株抗體(以下記載為抗 體A)並使用。 作為包含具有多肽之單一有用物質之製劑的模型,係依 據曰本專利特開平7-173 196中記载之方法製備基因重組伽 馬干擾素(以下記載為製劑A)並使用。 作為以下實施例中所使用之豬小病毒(ppv)溶液(自社團 法人動物用生物學製劑協會獲取,90HS株,Lot N〇.VS0201,感染價:8.67 l〇g(TCID為 50/mL),以下記載 139962.doc 19- 201002821 為PPV)、或PP V溶液之製備及LRV之測定所用之細胞而使 用之 PK-13細胞(自 ATCC獲取 ’ Lot No.ATCC CRL-6489), 係利用75(cm2)組織培養用燒瓶(BD Falcon製造)以及加入 有10體積牛血清(Upstate公司製造,於56它之水浴中加熱 30分鐘而失活後使用)及1體積。/〇青黴素/鏈黴素(+1〇〇〇〇 Units/mL 青黴素,+10000 黴素,Invitr〇gen 製造) 之D-MEM(Invitr〇gen製造,高葡萄糖)反覆培養並使用。 [實施例1病毒去除膜之製造法] 使用亨舍爾混合機(三井礦山(股)製造,型號20B),將熔 融流動指數(MFI,Melt Flow Index)為 2.5(g/l〇 mL)之由 49 wt%聚偏二氟乙稀樹脂(吳羽化學(股)製造,T#1300)及51 wt%鄰苯二甲酸二環己酯(大阪有機化學工業(股)製造工 業品)構成之組合物於70°C下攪拌混合後,進行冷卻而製 成粉體狀,利用漏斗將該粉體狀者投入玄雙軸同向螺旋式 擠出機(Technovel(股)製造 KZW25TW-5〇MG-NH(-600)) 中,於21 〇°C下熔融混合,均勻熔解。繼而,一面於中空 内部流動溫度為130。(:之鄰苯二曱酸二丁 S旨(大八化學工業 (股)製造工業品)’ 一面從由内直徑為〇·8 mm、外直徑為 1.05 mm之環狀孔口形成的紡絲嘴分別將均勻熔解物擠出 成中空纖維狀,於溫度調整為10、20、3〇、40°C之冷卻水 浴中進行冷卻固化,以5 0 m/分鐘之速度、纏繞於金屬框 上。其後,利用58%異丙醇水溶液(大八化學工業(股)製造 工業品)將鄰苯二甲酸二環己酯及鄰苯二甲酸二丁醋萃取 去除,以水將附著之58%異丙醇水溶液置換後’以浸潰於 139962.doc -20- 201002821 水中之狀態使用高壓蒸氣殺菌裝置(平山製作所(股)製造 HV-85)於125。〇下實施4小日夺熱處理。其後,利用異丙醇 (大八化车工業(版)製造工業品)將附著之水置換後,利用 真空乾炼機(Stec(股)製造)於6〇°c之溫度下進行乾燥,藉此 獲得中空纖維狀微多孔膜。再者,自纏繞起直至乾燥為止 之所有步驟中,中空纖維係以固定長度狀態固定並進行處 理。 繼而’對上述微多孔膜利用接枝法進行親水化處理。反 應液係使用如下者:使丙烯酸羥丙酯(大阪有機化學(股)製 造工業品)以達到8體積%之方式溶解於3_ 丁醇(純正化學 (股)製造工業品)之2 5體積%水溶液中,於保持於4 5之 狀態下進行30分鐘氮氣鼓泡。首先,於氮氣環境下,一面 利用乾冰將έ亥微多孔膜冷卻至_ 6 〇 ,一面以c 〇 6 0為輕射 源對該微多孔膜照射25 kGy之γ射線。將照射後之微多孔 膜於13.4 Pa以下之減壓下靜置15分鐘後,使上述反應液與 s亥微多孔膜於60°C下接觸,靜置1小時。其後,利用μ體 積%異丙醇水溶液清洗該微多孔膜,於60。(:下真空乾燥4小 時,獲得具有親水性之微多孔膜。可確認該微多孔膜與水 接觸時水自發地浸透至微孔内。以聚胺酯來填塞12根該微 多孔膜之絲束之兩端,接合於聚苯乙烯製之中空纖維膜被 隔在入口側空間與出口侧空間之間的匣,而製成過濾裝置 (有效膜面積為0.001 m2)。 [實施例2] [單株抗體之製造步驟] 139962.doc 21 201002821 將藉由深度過濾器及〇.2(μπι)薄膜過濾器澄清化之包含 人類單株抗體(人類IgGl)之CHO細胞無血清培養上清液(表 現量:700 mg/L)1500(mL)添加至以10(mmol/L)石粦酸鈉缓 衝液(pH值為6.0)平衡化之Protein A管柱(Amercham Biosciences公司製造 Mabselect 20 mm ID><20 cm)中(線速 度為500 cm/h)。繼而,利用5管柱容量之20(mmol/L)擰檬 酸鈉緩衝液(pH值為3.4)將人類單株抗體溶出(線速度為500 cm/h)。以1 0(mmol/L)石粦酸鈉緩衝液(pH值為8_2)將該溶出 液中和,進而以1.5(mol/L)Tris-HCl調整成pH值為8.0後, 添加至以1 〇(mmol/L)Tris-HCl平衡化之陰離子交換管柱 (Amercham Biosciences 公司製造 Q Sepharose XL 10 mm IDx 15 cm)中(線速度為300 cm/h)。添加結束後,使3管柱 容量之平衡化緩衝液於管柱中通過(線速度為300 cm/h), 以1.0(mol/L)乙酸將未吸附於管柱之成分調整成pH值為5.0 後,添加至以20(mmol/L)乙酸納緩衝液(pH值為5.0)平衡化 之陽離子交換管柱(Amercham Biosciences公司製造 SP Sepharose FF 26 mm IDx 1 5 cm)中(線速度為 300 cm/h)。添 加結束後,以5管柱容量之平衡化緩衝液進行清洗(線速度 為300 cm/h),進而使5管柱容量之20(mmol/L)乙酸納/ 0.30(mol/L)氯化鈉缓衝液(pH值為5.0)通過,作為人類單株 抗體溶液而溶出(線速度為300 cm/h)。利用超濾膜 (Millipore公司製造Biomax 30 50 cm2)將該溶出液濃縮直 至抗體濃度為1〇重量%。 使用藉由上述方法獲得之抗體A溶液、穩定劑(參照下述 139962.doc -22- 201002821 表1)、氯化鈉及注射用水(大塚製藥製造),分別製備以下 述表1中記載之比例包含抗體A及穩定劑之〇_ 1 (mol/L)氯化 納水溶液。繼而,使用少量之1_〇至〇.1〇(m〇l/L)鹽酸(和光 純藥製造)或者1.0至〇.10(m〇l/L)氫氧化鈉水溶液(和光純藥 製造)’將該等抗體A及穩定劑溶液之pH值調整成5 ·5。繼 而 將δ亥穩疋劑浴液靜置1小時,其後,測定該階段之單 株抗體純度,並利用HPLC(島津製作所製造Prominence, 官柱:Tosoh GPC用管柱 TSK gel G3000SWXL,移動相: 磷酸緩衝液(pH值為6.9)/0.3(mol/L)氯化鈉水溶液)根據峰 值面積比來測定製劑密封步驟結束時之單株抗體製劑純 度,結果如以下[表1]所示。 對於上述各抗體A及穩定劑溶液(包含ppV),分別準備藉 由[實施例1]之方法製造之過濾膜(〇 〇〇1 m2)及上述過濾裝 置,為測定LRV,取5(mL)之各抗體a及穩定劑溶液(以下 記載為原液)放入至已殺菌之PP製螺旋樣本瓶(BD Fak〇n製 造)中,立即於-78t:下冷凍保存,然後以294 kpa之壓力實 施Dead-end(端點)式過遽。各時間之單株抗體溶液之過遽 量、及♦艮魏體濃度計算出《單株抗體之透過量如下述 [表1]所示’可滿足所要求之單株抗體透過性。將上述各滤 液分配至50 mL之已殺菌之螺旋樣本航中,分別獲得目標 抗體A製劑。 139962.doc •23- 201002821 [表i] 項 (entry) 抗體A 濃度 (重量%) 穩定劑 ------ 穩定劑 濃度 (重量%) — 抗體製 劑純度 (%) 過濾0〜10分鐘 之抗體Α之平 均透過性 (kg/m2/hour) 過遽2〜3小時 之抗體A之平 均透過性 (kg/m2/hour) 過慮0〜3小時 之抗體A之平 均透過性 (kg/m^our) 1 3.0 d-甘露ΐϋ 3.0 97.5 2.44 2.15 2.23 — 2 3.0 D-棉 ___3.0 — 93.5 2.38 1.95 2.06 3 3.0 L-丙胺酸鹽 酸鹽观 3.0 94.8 2.77 2.43 2,53 4 3,0 D-葡萄糖胺 鹽酸鹽 3.0 -------- 92.0 1.89 1.62 1,69 5 3.0 檸檬酸二鈉 3.0 92.4 3.08 2.60 2.77 ο 5.0 D-葡萄糖 —5.0 96.5 2.92 2.44 卜 2.58 7 5.0 D-甘露 —5.0 90.6 2.25 1,81 1.90 8 5.0 L-丙胺酸鹽 酸鹽 5.0 ---- 98.1 2.34 1.95 2.05 9 5.0 D-葡萄糖胺 鹽酸鹽 5.0 88.9 L95 1.69 1.76 1U 5.0 檸檬酸二鈉 5.0 91.9 2.85 2.39 2.56 [實施例3] [病毒去除性試驗及單株抗體純度測定] 以加入有3體積%牛血清(Upstate公司製造,於56〇c之水 浴中加熱30分鐘而失活後使用)及1體積%青黴素/鏈黴素 (+10000 Units/mL Penicillin ’ +10000 pg/mL Streptomycin, Invitrogen 製造)之 D-MEM(Invitrogen 製造,high-glucose) (下文中將該混合液記載為3% FBS/D_ME]y[)將經培養之PK-13細胞稀釋’製備細胞濃度為2 〇xl〇5(cells/mL)之稀釋懸 浮液。準備10片96孔圓底細胞培養板(Falcon公司製造), 於所有孔中各分配1 〇〇(μί)之該細胞懸浮液。 繼而’對於上述進行了 3小時過濾之濾液之總量混合 液’利用3% FBS/D-MEM製備該等之1〇倍、1〇2倍、1〇3 倍、1 04倍、1 〇5倍稀釋液。進而,對於即將過濾之前所採 集之各原液,利用3% FBS/D-MEM製備該等之102倍、1〇3 -24- 139962.doc 201002821 倍、10倍、l〇5倍、106倍、1〇7倍稀釋液。於分配有上述 細胞懸浮液之96孔細胞培養板上,將各濾液及濾液之1〇 倍、102倍、1〇3倍、104倍、1〇5倍稀釋液以及原液之1〇2 倍、10倍、104倍、1〇5倍、1〇6倍、1〇7倍稀釋液於8孔中 各分配刚㈣,於3rc、5%二氧化碳氣體環境下於培養 箱t培養1 0天。% ' can be u for a short time'. When it is desired to reduce the membrane area of the virus cutting device even if the time is long, it can be set to a long time. X, as a condition for filtering a single antibody material by removing cockroaches by a virus, the temperature of the material, the temperature of the liquid, the room temperature, the inorganic salt, and the like are not derived from the addition of the organism or the mash, the type of the solvent, or the source The presence or absence of the inclusions from the preparation containing the substance alone with the polypeptide is not particularly limited. [Examples] In the following examples, the following transition chambers, SUS304 tanks, and Shishi tube mills P〇lymer were used, and the above-mentioned ruthenium apparatus was a hollow fiber membrane using hydrophilicized polyvinylidene fluoride as the #质. The membrane is removed as a virus, and the hollow fiber membrane is interposed between the inlet side space and the outlet side space in the interior of the fitting; the SUS304 groove and the manifold are used to transport the single antibody solution to the filtering device. , already at 121. (: The steam sterilization was carried out for 15 minutes. As a model of the intermediate product of the monoclonal antibody preparation, a monoclonal antibody (hereinafter referred to as antibody A) was prepared and used according to the method described in WO2004/087761. A model for the preparation of a useful substance is prepared by using the method described in Japanese Patent Laid-Open No. Hei 7-173 196, and is used as a recombinant gamma interferon (hereinafter referred to as Formulation A). Virus (ppv) solution (obtained from the Association of Biological Agents for Society Animals, 90HS strain, Lot N〇.VS0201, infection price: 8.67 l〇g (TCID 50/mL), 139962.doc 19-201002821 PK), or PK-13 cells used for the preparation of PP V solution and cells for the determination of LRV (obtained from ATCC ' Lot No. ATCC CRL-6489), using 75 (cm 2 ) tissue culture flask (BD Falcon Manufactured) and added 10 volumes of bovine serum (manufactured by Upstate, used in a water bath of 56 for 30 minutes and inactivated) and 1 volume. / patulin/streptomycin (+1 〇〇〇〇 Units/mL) Penicillin, +10000 D-MEM (manufactured by Invitr〇gen) (manufactured by Invitr〇gen, high glucose) was repeatedly cultured and used. [Example 1 Method for Producing Virus Removal Membrane] Using Henschel Mixer (Mitsui Mining Co., Ltd., Model 20B), a melt flow index (MFI, Melt Flow Index) of 2.5 (g/l 〇 mL) of 49 wt% polyvinylidene fluoride resin (Wu Yu Chemical Co., Ltd., T#1300) and A composition comprising 51 wt% of dicyclohexyl phthalate (made by Osaka Organic Chemical Industry Co., Ltd.) was stirred and mixed at 70 ° C, and then cooled to obtain a powder, which was powdered by a funnel. The body is put into a mysterious twin-axis co-rotating extruder (manufactured by Technovel KZW25TW-5〇MG-NH(-600)), melt-mixed at 21 〇 ° C, and uniformly melted. The internal flow temperature of the hollow is 130. (: The product of bis-phthalic acid dibutyl S (Da-8 Chemical Industry Co., Ltd.)) has a ring from the inner diameter of 〇·8 mm and the outer diameter of 1.05 mm. The spinning nozzle formed by the orifices respectively extrudes the uniform melt into a hollow fiber shape, and the temperature is adjusted to 10, 20 The mixture was cooled and solidified in a 3 °, 40 ° C cooling water bath, and wound on a metal frame at a speed of 50 m / min. Thereafter, an industrial product was manufactured using a 58% aqueous solution of isopropyl alcohol (Da Ba Chemical Industry Co., Ltd.). The dicyclohexyl phthalate and dibutyl phthalate are extracted and removed, and the attached 58% aqueous solution of isopropyl alcohol is replaced with water to be immersed in water of 139962.doc -20-201002821. The high-pressure steam sterilization device (Hirayama Manufacturing Co., Ltd. manufactures HV-85) is at 125. The armpit was subjected to heat treatment for 4 days. Thereafter, the adhered water was replaced with isopropyl alcohol (manufactured by Daigo Kasei Co., Ltd.), and then dried by a vacuum dryer (manufactured by Stec Co., Ltd.) at a temperature of 6 ° C. Thereby, a hollow fiber-like microporous film was obtained. Further, in all the steps from the winding up to the drying, the hollow fiber is fixed and treated in a fixed length state. Then, the above microporous membrane was hydrophilized by a grafting method. The reaction liquid was used in the following manner: 5% by volume of 3 -butanol (a pure chemical (manufactured) industrial product) was dissolved in a hydroxypropyl acrylate (manufactured by Osaka Organic Chemical Co., Ltd.) at a volume of 8 vol%. In the aqueous solution, nitrogen gas was bubbled for 30 minutes while being kept at 45 °C. First, the microporous membrane was cooled to _ 6 利用 by dry ice under a nitrogen atmosphere, and the microporous membrane was irradiated with γ-rays of 25 kGy with c 〇 60 as a light source. After the irradiated microporous membrane was allowed to stand under a reduced pressure of 13.4 Pa or less for 15 minutes, the reaction solution was brought into contact with a microporous membrane at 60 ° C, and allowed to stand for 1 hour. Thereafter, the microporous membrane was washed with a μ volume% aqueous solution of isopropyl alcohol at 60. (: vacuum drying for 4 hours to obtain a hydrophilic microporous film. It was confirmed that the microporous film spontaneously permeated into the micropores when it was in contact with water. The tow of 12 microporous membranes was filled with polyurethane. At both ends, a hollow fiber membrane made of polystyrene was interposed between the inlet side space and the outlet side space to form a filtration device (effective membrane area of 0.001 m2). [Example 2] [Single plant Antibody manufacturing procedure] 139962.doc 21 201002821 CHO cell serum-free culture supernatant containing human monoclonal antibody (human IgG1) clarified by a depth filter and a 〇.2 (μπι) membrane filter (expression amount) : 700 mg/L) 1500 (mL) was added to a Protein A column (Mabselect 20 mm ID manufactured by Amercham Biosciences Co., Ltd.) equilibrated with 10 (mmol/L) sodium citrate buffer (pH 6.0). 20 cm) (linear velocity of 500 cm/h). Then, human monoclonal antibody was eluted using a 20-column capacity of 20 (mmol/L) sodium citrate buffer (pH 3.4) (linear velocity was 500 cm/h). The solution was neutralized with 10 (mmol/L) sodium citrate buffer (pH 8_2), and further After 1.5 (mol/L) Tris-HCl was adjusted to pH 8.0, it was added to an anion exchange column equilibrated with 1 〇 (mmol/L) Tris-HCl (Q Sepharose XL 10 mm ID x 15 cm manufactured by Amercham Biosciences) Medium) (linear velocity 300 cm/h). After the addition, the equilibration buffer of the 3-column capacity is passed through the column (linear velocity 300 cm/h), with 1.0 (mol/L) acetic acid. The component which was not adsorbed on the column was adjusted to a pH of 5.0, and then added to a cation exchange column (A Seclose FF 26 manufactured by Amercham Biosciences Co., Ltd.) equilibrated with 20 (mmol/L) sodium acetate buffer (pH 5.0). Mm IDx 1 5 cm) (line speed 300 cm/h). After the addition, clean it with a 5-bar column balance buffer (linear speed 300 cm/h), and then make 5 column capacity 20 (mmol/L) sodium acetate / 0.30 (mol/L) sodium chloride buffer (pH 5.0) was passed as a human monoclonal antibody solution (linear velocity 300 cm / h). UF membrane was used. (Biomax 30 50 cm2 manufactured by Millipore Co., Ltd.) The eluate was concentrated until the antibody concentration was 1% by weight. The antibody A solution obtained by the above method was used. , Stabilizer (refer to Table 139962.doc -22-201002821 Table 1 below), sodium chloride and water for injection (manufactured by Otsuka Pharmaceutical Co., Ltd.), respectively, to prepare 〇 1 containing antibody A and a stabilizer in the ratios shown in Table 1 below. (mol/L) aqueous sodium chloride solution. Then, a small amount of 1_〇 to 〇.1〇(m〇l/L) hydrochloric acid (made by Wako Pure Chemical Industries) or 1.0 to 10.10 (m〇l/L) aqueous sodium hydroxide solution (made by Wako Pure Chemical Industries, Ltd.) 'The pH of these antibody A and stabilizer solutions was adjusted to 5 · 5. Then, the δ 疋 疋 疋 浴 浴 bath was allowed to stand for 1 hour, and thereafter, the purity of the individual antibody at this stage was measured, and HPLC was used (Prominence manufactured by Shimadzu Corporation, column: Tosoh GPC column TSK gel G3000SWXL, mobile phase: Phosphate buffer (pH 6.9) / 0.3 (mol/L) aqueous sodium chloride solution) The purity of the individual antibody preparation at the end of the preparation sealing step was measured based on the peak area ratio, and the results are shown in the following [Table 1]. For each of the above antibody A and the stabilizer solution (including ppV), a filtration membrane (〇〇〇1 m2) produced by the method of [Example 1] and the above filtration apparatus were prepared, and 5 (mL) was measured for LRV. Each antibody a and a stabilizer solution (hereinafter referred to as a stock solution) were placed in a sterilized PP spiral sample bottle (manufactured by BD Fak〇n), immediately frozen at -78 t:, and then subjected to a pressure of 294 kPa. Dead-end is too late. The amount of permeation of the monoclonal antibody solution at each time and the concentration of 艮 艮 体 计算 《 《 《 《 《 《 《 《 《 《 《 单 单 单 单 单 单 单 单 单 单 单 单 。 。 。 。 。 。 。 。 。 。 Each of the above filtrates was dispensed into 50 mL of the sterilized spiral sample to obtain the target antibody A preparation. 139962.doc •23- 201002821 [Table i] entry (entry) Antibody A concentration (% by weight) Stabilizer ------ Stabilizer concentration (% by weight) - Antibody preparation purity (%) Filter 0~10 minutes Average permeability of antibody kg (kg/m2/hour) Average permeability of antibody A over 2 to 3 hours (kg/m2/hour) Average permeability of antibody A over 0 to 3 hours (kg/m^ Our) 1 3.0 d-甘露ΐϋ 3.0 97.5 2.44 2.15 2.23 — 2 3.0 D-cotton ___3.0 — 93.5 2.38 1.95 2.06 3 3.0 L-Alanine hydrochloride view 3.0 94.8 2.77 2.43 2,53 4 3,0 D - Glucosamine hydrochloride 3.0 -------- 92.0 1.89 1.62 1,69 5 3.0 Disodium citrate 3.0 92.4 3.08 2.60 2.77 ο 5.0 D-glucose - 5.0 96.5 2.92 2.44 Bu 2.58 7 5.0 D-mannose - 5.0 90.6 2.25 1,81 1.90 8 5.0 L-Alanine hydrochloride 5.0 ---- 98.1 2.34 1.95 2.05 9 5.0 D-Glucosamine hydrochloride 5.0 88.9 L95 1.69 1.76 1U 5.0 Disodium citrate 5.0 91.9 2.85 2.39 2.56 [Example 3] [Vacuum removal test and determination of individual antibody purity] 3% by volume of bovine serum (manufactured by Upstate Co., Ltd., added in a water bath of 56 ° C) D-MEM (manufactured by Invitrogen, high-glucose) of 1% by volume of penicillin/streptomycin (+10000 Units/mL Penicillin ' + 10000 pg/mL Streptomycin, manufactured by Invitrogen) (after use) The mixture was recorded as 3% FBS/D_ME]y [) and the cultured PK-13 cells were diluted to prepare a diluted suspension having a cell concentration of 2 〇xl〇5 (cells/mL). Ten 96-well round bottom cell culture plates (manufactured by Falcon Co., Ltd.) were prepared, and 1 〇〇 (μί) of the cell suspension was dispensed into each well. Then, 'Total mixture of the filtrates which were filtered for 3 hours as described above' was prepared by using 3% FBS/D-MEM to prepare 1〇 times, 1〇2 times, 1〇3 times, 104 times, 1〇5 Double dilution. Further, for each stock solution collected immediately before filtration, 102 times, 1〇3 -24-139962.doc 201002821 times, 10 times, l〇5 times, 106 times, etc. were prepared by using 3% FBS/D-MEM. 1〇 7 times dilution. In a 96-well cell culture plate to which the above cell suspension is dispensed, 1×2, 102 times, 1〇3 times, 104 times, 1〇5 times dilution of each filtrate and filtrate, and 1〇2 times of the stock solution, 10 times, 104 times, 1〇5 times, 1〇6 times, and 1〇7 times dilutions were dispensed in each of the 8 wells, and cultured in an incubator for 10 days in a 3rc, 5% carbon dioxide atmosphere.

繼而,對於經10天培養之上述細胞培養板,利用紅血球 吸附法(非專利文獻6)進行TCID50(50%感染價)之測定。以 PBS()(日水製藥股份有限公司製造,利用商品隨附之方法 而製備)將雞保存血(日本生物測試製造)稀釋成5倍後,以 25 00(rpm)、於4C下進行5分鐘離心分離而使紅血球沈澱, 然後抽吸去除上清液,再次以PBS㈠將所獲得之包含紅血 球之沈殿物稀釋成2 〇 〇倍。 繼而,於上述細胞培養板之所有孔(well)中各分配 100(μΙ〇的所製備之紅血球沈澱物之pBs(_)稀釋液,靜置2 小時後,目測確認經培養之細胞組織之表面有無紅血球之 吸附 將確認到吸附者作為引起了病毒感染之孔、未择認 到吸附者作為未感染之孔而進行計數。關於所獲得之各培 養液之病毒感染之有無,就濾液或其稀釋液、或者原液之 稀釋液分別確認比例’利用Reed_Muneh法(非專利文㈣ 計算出i〇g(TCID 5〇/mL)作為感染價,求出病毒去除率 LRV,結果如以下[表2]所示。 進而’利用HPLC(島津製作所製造ρΓ〇ηιίη_β,管 柱· T〇s〇h GPC用管柱 TSKgel G3〇〇〇swxl,移動相:磷 139962.doc -25· 201002821 酸緩衝液(pH值為6.9)/0.3(mol/L)氣化鈉水溶液),根據峰 值面積比來測定製劑密封步驟結束時之單株抗體製劑純 度’結果如以下[表2]所示。結果,就entry 1〜1〇而言,所 要求之病毒去除性、單株抗體製劑純度、以及製造步驟中 之單株抗體於溶液中之穩定性確保全部可滿足。 [表2] 項 (entry) 抗體A 濃度 (重量%) 穩定劑 穩定劑 濃度 (重量%) 原液 log(TCID50) 渡液 log(TCID50) LRV 抗體製 劑純度 (%) 1 3.0 D-甘露糖醇 3.0 6.00 <0.5 >5.50 97.2 2 3.0 D-棉子糖 3.0 5.67 <0.5 >5.17 92.4 3 3.0 L-丙胺酸鹽酸鹽 3.0 5.80 <0.5 >5.30 95.7 4 3.0 D-葡萄糖胺鹽酸鹽 3.0 5.67 <0.5 >5.17 90.3 5 3.0 檸檬酸二納 3.0 5.80 <0.5 >5.30 92.5 6 5.0 D-甘露糖酵 5.0 6.12 <0.5 >5.62 95.5 7 5.0 D-棉子糖 5.0 6.20 <0.5 >5.70 89.5 8 5.0 L-丙胺酸鹽酸鹽 5.0 6.00 <0.5 >5.50 95.6 9 5.0 D-葡萄糖胺鹽酸鹽 5.0 6.00 <0.5 >5.50 88.6 10 5.0 檸檬酸二納 5.0 5.80 <0.5 >5.30 91.9 [實施例4 ] [基因重組伽馬干擾素之製造步驟] 於含有150〜500 mL無菌培養基之無菌附擋板培養燒瓶中 製備儲備培養物,該無菌培養基具有如下成分:胰化蛋白 脒為10 g/L、酵母萃取物為5 g/L、氯化鈉為5〜1〇 mg/;L。 其次,將旦·_ W3110/py_CYC5之一次培養物接種於培養 基中。 < 繼而,將接種之燒瓶於振盪器上於25〜37。〇下培養直至 5 50 nm之吸光度達到約i 〇為止。將3〇%(v/v)之二甲基亞 砜約50%(V/V)添加於培養液卡。立即將i mL之等分試樣 139962.doc •26- 201002821Then, the TCID50 (50% infection price) was measured by the red blood cell adsorption method (Non-Patent Document 6) on the cell culture plate cultured for 10 days. The chicken preserved blood (manufactured by Nippon Biotest Co., Ltd.) was diluted to 5 times with PBS() (manufactured by Nissui Pharmaceutical Co., Ltd., and prepared by the method attached to the product), and then subjected to 2500 (rpm) at 4C. After centrifugation, the red blood cells were precipitated, and then the supernatant was removed by suction, and the obtained erythrocyte-containing sediment was diluted to 2 times with PBS (1). Then, 100 (μΙ〇 of the prepared pBs(_) dilution of the prepared red blood cell pellet was dispensed into all the wells of the above cell culture plate, and after standing for 2 hours, the surface of the cultured cell tissue was visually confirmed. The presence or absence of red blood cell adsorption will confirm that the adsorber is counted as a hole causing the virus infection, and the adsorbent is not recognized as an uninfected well. Regarding the presence or absence of the virus infection of each culture solution obtained, the filtrate or its dilution The dilution ratio of the liquid or the stock solution was confirmed by the Reed_Muneh method (Non-patent (4), i〇g (TCID 5〇/mL) was calculated as the infection price, and the virus removal rate LRV was obtained. The results are as follows [Table 2] Further, 'Using HPLC (made by Shimadzu Corporation, ρΓ〇ηιίη_β, column, T〇s〇h GPC column TSKgel G3〇〇〇swxl, mobile phase: phosphorus 139962.doc -25· 201002821 acid buffer (pH value) The purity of the individual antibody preparation at the end of the sealing step of the preparation was determined according to the peak area ratio of 6.9)/0.3 (mol/L) of the sodium carbonate aqueous solution. The results are shown in the following [Table 2]. 1〇, what you want The virus removal property, the purity of the monoclonal antibody preparation, and the stability of the monoclonal antibody in the solution in the production step are all satisfied. [Table 2] entry Antibody A concentration (% by weight) Stabilizer stabilizer Concentration (% by weight) stock log (TCID50) travertine log (TCID50) LRV antibody preparation purity (%) 1 3.0 D-mannitol 3.0 6.00 <0.5 > 5.50 97.2 2 3.0 D-raffinose 3.0 5.67 < 0.5 >5.17 92.4 3 3.0 L-Alanine hydrochloride 3.0 5.80 <0.5 > 5.30 95.7 4 3.0 D-Glucosamine hydrochloride 3.0 5.67 <0.5 > 5.17 90.3 5 3.0 Citric acid di-nano 3.0 5.80 <;0.5>5.30 92.5 6 5.0 D-mannose fermentation 5.0 6.12 <0.5 >5.62 95.5 7 5.0 D-raffinose 5.0 6.20 <0.5 > 5.70 89.5 8 5.0 L-alanine hydrochloride 5.0 6.00 <;0.5>5.50 95.6 9 5.0 D-Glucosamine hydrochloride 5.0 6.00 <0.5 > 5.50 88.6 10 5.0 Citric acid di-nano 5.0 5.80 <0.5 > 5.30 91.9 [Example 4] [Genetic recombination gamma interference a manufacturing step] preparing a stock culture in a sterile baffle culture flask containing 150 to 500 mL of sterile medium, Sterile medium having the following composition: tryptone amidine of 10 g / L, yeast extract was 5 g / L, sodium chloride 5~1〇 mg /; L. Next, a primary culture of denier _ W3110/py_CYC5 was inoculated into the medium. < In turn, the inoculated flask was placed on a shaker at 25 to 37. The culture was carried out under the armpit until the absorbance at 5 50 nm reached about i 〇. About 35% (v/v) of dimethyl sulfoxide was added to the culture solution card at about 50% (V/V). Immediately aliquot the i mL 139962.doc •26- 201002821

分配於益_婵+ ,,丄 Q …囷衩本瓶中,關上蓋子。 保存。各醱觖γ 樣本瓶係於-60°C以下 各醱酵係使用接種用複製儲備 % ^ ., 角心蚕物而開始。於振 =瓶或切_槽时用上述培養基(LBb_h)製備接 由 約听下培養約8小時後,將接種物移至醱酵槽 中。接種物之容量係撥酵容量之2至1()%。重組干擾素_丫之 產生係於具有約10至1000 L之作用容積之醱酵槽内進行。 醱酵培養基係每丨L具有如下成分:Assigned to the benefits of _婵+, 丄Q ... 囷衩 in the bottle, close the lid. save. Each 酦觖 γ sample bottle is below -60 ° C. Each fermentation system is started using the inoculation replication reserve % ^ . In the case of Yu Zhen = bottle or cut-slot, the medium (LBb_h) was used to prepare for about 8 hours of incubation, and the inoculum was transferred to a fermentation tank. The inoculum capacity is 2 to 1% of the fermented capacity. The recombinant interferon _ 丫 is produced in a fermentation tank having an action volume of about 10 to 1000 L. The fermentation medium has the following composition per liter:

葡萄糖(*) : 50-100 g 硫酸銨:4.0-8.0 g 磷酸二氫鉀:3.0-5.0 g 磷酸氫二鉀:5.0-8.0 g 硫酸鎂七水合物:〇.5-5.1 g 擰檬酸鈉二水合物:0.5-2.0 g UCON LB-625 : 0.5-2.0 mL 氣化鐵六水合物:0.005-0.15 gGlucose (*) : 50-100 g Ammonium sulfate: 4.0-8.0 g Potassium dihydrogen phosphate: 3.0-5.0 g Dipotassium hydrogen phosphate: 5.0-8.0 g Magnesium sulfate heptahydrate: 〇.5-5.1 g sodium citrate Dihydrate: 0.5-2.0 g UCON LB-625 : 0.5-2.0 mL gasified iron hexahydrate: 0.005-0.15 g

氣化钻六水合物:0.001-0.005 g 鉬酸鈉二水合物:〇.〇〇1_〇.〇〇5 g 硫酸銅五水合物:0.001-0.005 g 硼酸:0.001-0.005 g 硫酸錳一水合物:0.001-0.005 g 鹽酸:0.0-1.0 mL 噻胺-HC1 : 0.0-0.1 g 四環素-HC1 : 0.001-0.01 g 139962.doc •27· 201002821 L-色胺酸(*) : 0.1-0.5 g 酵母萃取物:2.0-8.0 g 3-β-吲哚丙烯酸:0.02-0.1〇g (*)葡萄糖及色胺酸係最初將— 部分於醱酵過程中供給。 部分放入至醱酵槽中 剩餘 培養基中之成分係於用於醱酵之前藉由熱處理或過濾進 行殺菌。酸酵係於25〜贼下進行。其他操作條件如下: 攪拌(rpm) : 100〜1〇〇〇 通氣(vvm) : 0_5〜1.5(必要時補充氧) pH值:6.5〜7.5(藉由添加氫氧化銨而調節) 使細胞懸浮於含有鹽及pH值為6至9、冑好的是^9 之適當緩衝液的培養基中。於Gaulin磨機之類的高壓膠體 磨機中使細胞懸浮液均質化,提取重組伽馬干擾素。將充 分之聚乙烯亞胺添加至上述溶液中,製備成 之溶液。上清液中包含伽馬干擾素。使上述上清液吸附於 以PH值$6至9之範圍之適當的鹽溶液進行清洗而去除了雜 質之以二氧切為基質之吸附劑上。使用含有Q 5至m 之氯化四甲基銨之溶液將重組伽馬干擾素溶出。本步驟之 所有操作係於PH值為7至9下進行。對上述溶出液進行透 析,使其吸附於陰離子交換層析介質,繼而進行清洗而去 除雜質。以增大之鹽梯度將重組伽馬干擾素溶出。本步驟 中可使用之典型陰離子交換樹脂有緩甲基纖維素及碏乙基 纖維素。所有操作係於pH值為7至9下進行。使上述溶出^ 吸附於《_介質,繼而進行清洗而絲雜^於碟酸鹽 139962.doc •28- 201002821 濃度梯度令使鹽濃度增加而將重組伽馬干擾素溶出。本步 驟之所有操作係於pH值為7至9下實施。對上述溶出物進行 透析’使其吸附於陰離子交換層析介質,繼而進行清洗而 去除雜質。自陰離子交換介質中以增大鹽濃度之梯度將重 組伽馬干擾素溶出。典型之陰離子交換層析介質係羧甲基 纖維素及確乙基纖維素。本步驟之所有操作係於pH值為7 至9下實施。將上述溶出物應用於凝膠渗透介質,於管柱 中以含鹽溶劑展開。將含有重組伽馬干擾素之適當之組分 彙集,利时子篩管柱(Phannaeia)進行脫錢冷;東乾燥, &得純化主體。本步驟之所有操作係於值為7至9下進 行。Gasified drill hexahydrate: 0.001-0.005 g sodium molybdate dihydrate: 〇.〇〇1_〇.〇〇5 g copper sulfate pentahydrate: 0.001-0.005 g boric acid: 0.001-0.005 g manganese sulfate monohydrate 0.001-0.005 g Hydrochloric acid: 0.0-1.0 mL Thiamine-HC1 : 0.0-0.1 g Tetracycline-HC1 : 0.001-0.01 g 139962.doc •27· 201002821 L-Tryptophanic acid (*) : 0.1-0.5 g Yeast Extract: 2.0-8.0 g 3-β-indole acrylic acid: 0.02-0.1 〇g (*) Glucose and tryptophan are initially supplied in part during the fermentation process. The components which are partially placed in the fermentation tank are left to be sterilized by heat treatment or filtration before being used for fermentation. The acid yeast is carried out under 25~ thief. Other operating conditions are as follows: Stirring (rpm): 100~1〇〇〇 aeration (vvm): 0_5~1.5 (addition of oxygen if necessary) pH: 6.5~7.5 (adjusted by adding ammonium hydroxide) It is contained in a medium containing a salt and a suitable buffer having a pH of 6 to 9, preferably ^9. The cell suspension was homogenized in a high pressure colloid mill such as a Gaulin mill to extract recombinant gamma interferon. A sufficient solution of polyethyleneimine was added to the above solution to prepare a solution. The supernatant contains gamma interferon. The supernatant is adsorbed to an appropriate salt solution having a pH of from $6 to 9 to remove impurities on the dioxosite-based adsorbent. The recombinant gamma interferon is eluted using a solution containing Q 5 to m tetramethylammonium chloride. All operations in this step were performed at a pH of 7 to 9. The above-mentioned eluate is diafiltered, adsorbed to an anion exchange chromatography medium, and then washed to remove impurities. The recombinant gamma interferon is eluted with an increased salt gradient. Typical anion exchange resins which can be used in this step are slow methyl cellulose and decyl ethyl cellulose. All operations were carried out at a pH of 7 to 9. The above-mentioned dissolution was adsorbed to the "medium, and then washed and the silk was mixed with the acid salt. 139962.doc • 28-201002821 The concentration gradient caused the concentration of the salt to increase and the recombinant gamma interferon was eluted. All of the operations in this step were carried out at a pH of 7 to 9. The above-mentioned eluate is dialyzed to be adsorbed to an anion exchange chromatography medium, followed by washing to remove impurities. The recombinant gamma interferon is eluted from the anion exchange medium with a gradient of increasing salt concentration. Typical anion exchange chromatography media are carboxymethyl cellulose and indeed ethyl cellulose. All of the operations in this step were carried out at a pH of 7 to 9. The above-mentioned eluted matter is applied to a gel permeation medium, and is developed in a column with a salt-containing solvent. Appropriate components containing recombinant gamma interferon are pooled, and the Phinenae column is subjected to decoholization; the east is dried, & All operations in this step are performed at values of 7 to 9.

使用藉由上述方法獲得之製劑A、穩定劑(參照下述[表 3])、氯化鈉及注射用水(大塚製藥製造),分別製備以下述 [表3]中§己載之比例包含製劑a及穩定劑的〇♦舰)氯化 鈉水溶液。繼而,使用少量之以至㈣一叫鹽酸(和光 純藥製造)或者1 ·〇至0.! 〇(m()1/L)氫氧化納水溶液(和光純藥 製造),將該等製劑八及穩定劑溶液之pH值調整成5.5。 對於上述各製劑A及穩定劑溶液(包含PPV),分別準備藉 由[實施例1]之方法製造之過渡膜(G•⑼i m2)及上述過滤裝 置’為測定LRV,取5(mL)之各製劑a及穩定劑溶液(以下 。己載為原液)放入至已殺菌之pp製螺旋樣品瓶(BD Fak〇n製 、)中立即於C下冷束保存’然後以294 kPa之壓力實 施Dead-end式過濾。各時間之單株抗體溶液之過濾量、及 根據製劑濃度計算出之製劑A之透過量如下述[表3]所示, 139962.doc •29· 201002821 可滿足所要求之重組伽馬干擾素透過性。將上述各〉慮液分 配於5 0 mL已殺菌之螺旋樣品瓶中’分別獲得目標製劑 A 〇 [表3] 項 (entry) 抗體A 濃度 (重量%) 穩定劑 穩定劑 濃度 (重量%) 過濾〇~1〇分鐘 之抗體A之平 均透過性 (kg/m2/hour) 過濾2〜3小時之 抗體A之平均透 過性 (kg/m2/hour) 過濾、〇~3小時之 抗體A之平均 透過性 (kg/m2/hour、 1 3.0 D-甘露糖醇 3.0 3.07 2.58 2.77 2 3.0 卜丙胺酸鹽 酸鹽 3.0 2.93 2.47 2.65 [實施例5] [病毒去除性試驗] 以加入有3體積%牛血清(Upstate公司製造’於56°C之水 浴中加熱30分鐘而失活後使用)及1體積%青黴素/鏈黴素 (+10000 Units/mL Penicillin ’ +10000 pg/mL Streptomycin ’ Invitrogen 製造)之 D-MEM(Invitrogen 製造 ’ high-glucose) (下文中將該混合液記載為3% FBS/D-MEM)將經培養之PK-13細胞稀釋,製備細胞濃度為2.0xl05(cells/mL)之稀釋懸 浮液。準備10片96孔圓底細胞培養板(Falcon公司製造), 於所有孔中各分配1 之該細胞懸浮液。 繼而,對於上述進行了 3小時過濾之濾液之總量混合 液,利用3% FBS/D-MEM製備該等之10倍、1〇2倍、1〇3 倍、1 04倍、1 05倍稀釋液。進而,對即將過渡之前所採集 之各原液,利用3% FBS/D-MEM製備該等之1〇2倍、1〇3 倍、104倍、1〇5倍、1〇6倍、1〇7倍稀釋液。於分配有上述 細胞懸浮液之96孔細胞培養板上,將各濾液及濾液之10 139962.doc •30- 201002821 倍、ίο2倍、ίο3倍、104倍、105倍稀釋液以及原液之ίο2 倍、103倍、104倍、1〇5倍、1〇6倍、107倍稀釋液於8孔中 各分配100(μΙ〇,於37°c、5%二氧化碳氣體環境下於培養 箱中培養10天。 繼而’對於經10天培養之上述細胞培養板,利用紅血球 吸附法(非專利文獻6)進行TCID50(50%感染價)之測定。以 PBS㈠(曰水製藥股份有限公司製造,利用商品隨附之方法 而製備)將雞保存血(日本生物測試製造)稀釋成5倍後,以 2500(rpm)、於4°C下進行5分鐘離心分離而使紅血球沈澱, 然後抽吸去除上清液,再次以PBS(-)將所獲得之包含紅血 球之沈澱物稀釋成200倍。 繼而,於上述細胞培養板之所有孔中各分配1〇〇(μί)的 所製備之紅血球沈澱物之PBS㈠稀釋液’靜置2小時後, 目測確認經培養之細胞組織之表面有無紅血球之吸附,將 確δ忍到吸附者作為引起了病毒感染之孔、未確認到吸附者 〇 作為未感染之孔而進行計數。對於所獲得之各培養液之病 f感染之有無,就濾液或其稀釋液、或原液之稀釋液分別 確認比例,利用Reed_Munch法(非專利文獻6)計算出 l〇g(TCID50/mL)作為感染價,求出病毒去除率LRv,結果 如以下[表4]所示。結果,就_y i〜⑼言,可滿足所要求 之病毒去除性。 139962.doc -31· 201002821 [表4] 項 (entry) 抗體A濃度 (重量%) 穩定劑 穩定劑濃度 (重量%) 原液 log(TCID50) 渡液 log(TCID50) LRV 1 3.0 D-甘露糖醇 3.0 6.20 <0.5 >5.70 2 3.0 L-丙胺酸鹽酸鹽 3.0 6.20 <0.5 >5.70 [產業上之可利用性] 藉由使用本發明,於製造包含具有多肽之單一有用物質 之製劑時,可於維持較高之透過性及較高之病毒去除性之 狀態下利用病毒去除膜對高濃度製劑溶液進行過濾,且亦 可使用源自生物或源自醱酵產物之存在病毒混入風險之穩 定劑。藉此可總括地達成上述製劑之製造步驟之簡略化、 精簡彳έ、低成本化等。 139962.doc -32-Formulation A, a stabilizer (refer to the following [Table 3]), sodium chloride, and water for injection (manufactured by Otsuka Pharmaceutical Co., Ltd.) obtained by the above method were separately prepared to contain the preparation in the ratio of § contained in the following [Table 3]. a and stabilizer 〇 ♦ ship) aqueous sodium chloride solution. Then, use a small amount of (4) one called hydrochloric acid (made by Wako Pure Chemical Industries) or 1 · 〇 to 0.! 〇 (m () 1 / L) aqueous sodium hydroxide solution (made by Wako Pure Chemical Industries), and these preparations and The pH of the stabilizer solution was adjusted to 5.5. For each of the above-mentioned preparation A and the stabilizer solution (including PPV), a transition film (G•(9)i m2) manufactured by the method of [Example 1] and the above-mentioned filtration device were prepared for measuring LRV, and 5 (mL) was used. Each of the preparation a and the stabilizer solution (hereinafter referred to as the stock solution) was placed in a sterilized pp spiral sample vial (manufactured by BD Fak〇n), and immediately stored under a cold bundle at C. Then, the pressure was carried out at a pressure of 294 kPa. Dead-end filtering. The filtration amount of the monoclonal antibody solution at each time and the permeation amount of the preparation A calculated according to the concentration of the preparation are as shown in the following [Table 3], 139962.doc •29·201002821 can satisfy the required recombinant gamma interferon transmission. Sex. Distribute each of the above solutions into a 50 mL sterilized spiral sample vial to obtain the target preparation A 分别 [Table 3] entry (investigation) A concentration (% by weight) Stabilizer stabilizer concentration (% by weight)平均~1 minute average permeability of antibody A (kg/m2/hour) Filtered for 2 to 3 hours of average permeability of antibody A (kg/m2/hour) Filtration, 〇~3 hours of average transmission of antibody A Sex (kg/m2/hour, 1 3.0 D-mannitol 3.0 3.07 2.58 2.77 2 3.0 amphetamine hydrochloride 3.0 2.93 2.47 2.65 [Example 5] [Virus removal test] Adding 3 vol% bovine serum (Upstate manufactured by 'heating in a 56 ° C water bath for 30 minutes and inactivated) and 1% by volume of penicillin/streptomycin (+10000 Units/mL Penicillin ' + 10000 pg/mL Streptomycin 'Invitrogen) -MEM (manufactured by Invitrogen 'high-glucose) (hereinafter, the mixture is described as 3% FBS/D-MEM). The cultured PK-13 cells were diluted to prepare a dilution of a cell concentration of 2.0 x 105 (cells/mL). Suspension. Prepare 10 96-well round bottom cell culture plates (manufactured by Falcon) in all wells The cell suspension of 1 was dispensed. Then, the total mixture of the filtrates which were filtered for 3 hours as described above was prepared by using 3% FBS/D-MEM to prepare 10 times, 1〇2 times, 1〇3 times, 1 04 times, 1.00 times the dilution solution. Further, for each stock solution collected immediately before the transition, 1 〇 2 times, 1 〇 3 times, 104 times, 1 〇 5 were prepared using 3% FBS/D-MEM. Multiple, 1 〇 6 times, 1 〇 7-fold dilution. 10 139962.doc • 30- 201002821 times, ίο2 times, ίο3 times of each filtrate and filtrate in a 96-well cell culture plate to which the above cell suspension is dispensed , 104 times, 105 times dilution, and ίο2 times, 103 times, 104 times, 1〇5 times, 〇6 times, 107 times dilution of the stock solution are each dispensed in 8 wells (μΙ〇 at 37 ° C, The cells were cultured in an incubator for 10 days in a 5% carbon dioxide atmosphere. Then, for the above-mentioned cell culture plates cultured for 10 days, TCID50 (50% infection price) was measured by a red blood cell adsorption method (Non-Patent Document 6). (manufactured by Surabaya Pharmaceutical Co., Ltd., prepared by the method attached to the product) to preserve blood from chicken (Japan Biotest Manufacturing) After diluting to 5 times, the red blood cells were pelleted by centrifugation at 2500 (rpm) for 5 minutes at 4 ° C, and then the supernatant was removed by suction, and the obtained pellet containing red blood cells was again obtained with PBS (-). Dilute to 200 times. Then, 1 〇〇 (μί) of the prepared erythrocyte pellet PBS (1) dilution was dispensed in each well of the above cell culture plate for 2 hours, and visually confirmed whether the surface of the cultured cell tissue was adsorbed by red blood cells. It is confirmed that the δ endorses the adsorber as a hole causing the virus infection, and the adsorbent is not confirmed as the uninfected hole. Regarding the presence or absence of the disease f infection of each culture solution obtained, the ratio of the filtrate or the dilution thereof or the dilution of the stock solution was confirmed, and 1 〇g (TCID50/mL) was calculated by the Reed_Munch method (Non-Patent Document 6). The infection price was determined, and the virus removal rate LRv was determined. The results are shown in the following [Table 4]. As a result, as far as _y i~(9) is concerned, the required virus removal property can be satisfied. 139962.doc -31· 201002821 [Table 4] Item (entry) Antibody A concentration (% by weight) Stabilizer stabilizer concentration (% by weight) Stock log (TCID50) Drain log (TCID50) LRV 1 3.0 D-mannitol 3.0 6.20 <0.5 > 5.70 2 3.0 L-Alanine hydrochloride 3.0 6.20 <0.5 > 5.70 [Industrial Applicability] By using the present invention, a preparation comprising a single useful substance having a polypeptide is produced. The virus removal membrane can be used to filter the high concentration preparation solution while maintaining high permeability and high virus removal, and the risk of virus incorporation from the organism or from the fermentation product can also be used. Stabilizer. Thereby, the simplification, reduction, cost reduction, and the like of the manufacturing steps of the above-described preparations can be collectively achieved. 139962.doc -32-

Claims (1)

201002821 七、申請專利範圍: 1· 一種包含具有多肽之單— 盆特在π . #, 用物質之製劑之製造方法, 之單-有用ρ 括如下步驟者:產生具有多肽 心早有用物質之培養步 質吨化t "字具有多肽之單一有用物 貝'、,屯化之純化步驟,添加至 右田私•新 種抑制具有多肽之單一 有用物貝之凝聚的穩定劑之 才心疋Μ添加步驟,蘇出、忘基 去除膜將包含具有多肽h冑精由病毋 去除之疒主土队 有用物質之溶液中的病毒 覃一古田“ 以及將去除了病毒之具有多肽之 早-有用物質密封的密封 將源自具有多肽之單—有用物曾上述病毋去除步驟係 自韁—主 有用物負之產生時之病毒、與源 自穩疋劑之產生時之病毒 险处七…' 6毋㈣病毋去除步驟之病毒去 ” .、、、$4 ’病毒去除步驟時之具有多肽之單一有 用物質的濃度為3.0〜10.0重量%之範圍,溶液中所含之且 有多肽之單-㈣㈣㈣於上述病毒去除步驟中所使 用之病毒去除膜之過遽開始後〇〜10分鐘、及經過2〜3小 時之時的平均透過性均為1 _G(kg/m2/h_)以上。 2.:巧求項1之包含具有多肽之單-有用物質之製劑之製 &方法’其中上述穩定劑係源自生物或源自醱酵產物 者。 3·:請求項2之包含具有多肽之單一有用物質之製劑之製 k方法其中上述穩定劑係選自糖類、胺基酸、胺基 糖、有機酸、或該等之衍生物中。 4.如凊求項1至3中任-項之包含具有多肽之單-有用物質 之製劑之製造方法,其中上述病毒去除膜為包含合成高 139962.doc 201002821 分子之t空纖維膜。 5. 如請求項1至3中任一項夕—A 貝之包含具有多肽 α 之製劑之製造方法,其中u、、 之早—有用物質 T上述病毒去略 膜。 膜為多層微多孔 6. 如請求項4之包含具有多肽之 造方法,其中上述病毒去除膜…有用物質之製劑之製 7. 方降膜為多層微多 一種包含單株抗體之製劑之 、。 係至少包括如下步驟者:彦 L其特徵在於:其 單株抗體純化之純化步賢,Μ 。養步驟,將 驟添加至少—種抑制單株抗體 之凝象的穩定劑之穩定劑 勺人⑽< Μ添加步驟’藉由病毒去除膜將 包3早株抗體之溶液中之痣主 a ^. χ f去除的病毒去除步驟,以 及將去除了病毒之單株抗體 主丄 几肢在封之抢封步驟;且上述病 毋去除步驟係將源自單株抗體 , 卞休机頫之產生時之病毒、與源自 穩定劑之產生時之病毒去主 、 ^ ^ 病母去除步驟之病毒去除 能力為LRV24、病毒去除步驟時之單株抗體的濃度為 3.0〜H)重量%之範圍’溶液中所含之單株抗體相對於上 述病毒去除步驟中所使用之病毒去除膜之過遽開始後 〇〜1〇分鐘、及經過2〜3小時之時的平均透過性均為 LCKkg/m'hour)以上。 8.如β求項7之包含單株抗體之製劑之製造方法,其中上 述穩定劑係源自生物或源自醋酵產物者。 9_如請求項8之包含單株抗體之製劑之製造方法,其中上 述穩疋劑係選自糖類、胺基酸、胺基糖、有機酸、或該 等之衍生物中。 139962.doc 201002821 10·如請求項7至9中 法,其之包含單株抗體之製劑之製h 膜。 *域為包含合成高分子之中空纖維 11. 如請求項7至9中任— 土 ^ 項之包含單株抗體之製劑之贺1 12. 如珠::中上述病毒去除膜為多層微多孔膜。、"方 二項10之包含單株抗體之製劑之製造方法,t中 迷病毒去除臈為多層微多孔膜。 其中上 13 _如請求項7至9 φ〆工 甲任—項之包含單株抗體之製劑之 法,其中上述穩定#丨、& 4 i x k方 ^ “加步财之各穩定劑之添力α量八 内”、、.54ϊ%之範圍,且穩定劑添加步驟結束日士习 在封步驟結束時之單株抗體純度為 80%以上^ 、:长貝1 G之包含單株抗體之製劑之製造方法, 述疋為加步驟中之各穩定劑之添加量分別為〇、上 軏圍且夂劑添加步驟結束時及密封弗 束時之單株抗體純度為80%以上。 夕結 15·如請求項11之包含單株抗體之製劑之製造方法,其 述穩定劑添加步驟中之各穩定劑之添加量分別為二〜上 重量%之範圍,且穩定劑添加步驟結束時及密封步2 束時之單株抗體純度為80%以上。 。 16.如請求項12之包含單株抗體之製劑之製造方法,其中 述穩定劑添加步驟尹之各穩定劑之添加量分別為〇 ]上 重量%之範圍’且穩定劑添加步驟結束時㈣封步= 束時之單株抗體純度為80%以上。 ' ° 139962.doc 201002821 四、指定代表圖: (一) 本案指定代表圖為:(無) (二) 本代表圖之元件符號簡單說明: 五、本案若有化學式時,請揭示最能顯示發明特徵的化學式: (無) 139962.doc201002821 VII. Scope of application for patents: 1. A method for the preparation of a preparation comprising a polypeptide having a polypeptide in the form of a π. #, using a substance, the single-use ρ includes the following steps: producing a culture with a polypeptide early useful substance The step-tanning t " word has a single useful substance of the polypeptide, and the purification step of the deuteration is added to the right-handed addition step of the stabilizer of the new species inhibiting the aggregation of a single useful substance of the polypeptide. , Su-extraction, forgetting to remove the membrane will contain the virus in the solution of the useful substance of the main soil team with the polypeptide h胄 fine removed from the disease, and the old-useful substance with the polypeptide removed from the virus. The seal will be derived from a single-utility with a polypeptide. The above-mentioned path removal step is based on the fact that the virus is produced when the main useful substance is produced, and when the virus is derived from the production of the stabilizer (7) (4) The virus of the disease removal step goes to the range of 3.0 to 10.0% by weight of the single useful substance having the polypeptide at the time of the virus removal step, and is contained in the solution. And the polypeptide has a single-(four) (four) (d). The average permeability of the virus-removing membrane used in the above-mentioned virus removal step is 〇10 minutes, and after 2 to 3 hours, the average permeability is 1 _G (kg/m2/ H_) above. 2. A method of preparing a preparation comprising a single-useful substance having a polypeptide, wherein the above stabilizer is derived from a living organism or derived from a fermentation product. 3. The method of claim 2, wherein the stabilizer is selected from the group consisting of a saccharide, an amino acid, an amino sugar, an organic acid, or a derivative thereof. 4. The method for producing a preparation comprising a single-useful substance having a polypeptide according to any one of items 1 to 3, wherein the virus-removing film is a hollow fiber membrane comprising a synthetic high 139962.doc 201002821 molecule. 5. The method of producing a preparation comprising a polypeptide α according to any one of claims 1 to 3, wherein the u-, early-use substance T is a virus-depleted membrane. The film is a multi-layered microporous body. 6. The method of claim 4, comprising the method for producing a polypeptide, wherein the virus removing film is prepared by a preparation of a useful substance. 7. The falling film is a multilayered one, and comprises a preparation of a monoclonal antibody. The system includes at least the following steps: Yan L is characterized by: purification of the individual antibody purification step, Μ. In the step of cultivating, at least one stabilizer for inhibiting the coagulation of the monoclonal antibody is added to the stabilizer (10) < Μ adding step 'by removing the membrane from the virus to remove the sputum in the solution of the early antibody.病毒 f removal of the virus removal step, and the removal of the virus from the individual antibody main limbs in the sealing step; and the above-mentioned path removal step will be derived from the individual antibody, when the sputum is taken The virus removal ability of the virus, the virus removal from the generation of the stabilizer, the removal of the virus is LRV24, and the concentration of the monoclonal antibody in the virus removal step is 3.0~H)% by weight in the solution. The average permeability of the contained monoclonal antibody relative to the virus removal membrane used in the above-mentioned virus removal step is 〇~1〇 minutes and the average permeability after 2 to 3 hours is LCKkg/m'hour) the above. 8. A method of producing a preparation comprising a monoclonal antibody according to the method of claim 7, wherein the above stabilizer is derived from a living organism or a product derived from a vinegar yeast product. A method for producing a preparation comprising a monoclonal antibody according to claim 8, wherein the above-mentioned stabilizer is selected from the group consisting of a saccharide, an amino acid, an amino sugar, an organic acid, or a derivative thereof. 139962.doc 201002821 10. The method of claims 7 to 9, which comprises a h film of a preparation of a monoclonal antibody. *The domain is a hollow fiber containing a synthetic polymer. 11. The preparation of a monoclonal antibody according to any of claims 7 to 9 is as follows: 12. The above-mentioned virus removal membrane is a multilayer microporous membrane. . And the method for producing a monoclonal antibody comprising a monoclonal antibody, wherein the prion removal enthalpy is a multilayer microporous membrane. Wherein the above 13 _ as claimed in items 7 to 9 φ 〆 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲 甲The amount of α is within the range of ”, .54ϊ%, and the stabilizer addition step is completed. At the end of the sealing step, the purity of the individual antibody at the end of the sealing step is 80% or more. ^: The preparation of the monoclonal antibody containing the monoclonal antibody In the production method, the amount of each of the stabilizers added in the step is 〇, 軏 軏, and the purity of the individual antibody at the end of the step of adding the elixirs and at the time of sealing the bundle is 80% or more. The method for producing a preparation comprising a monoclonal antibody according to claim 11, wherein the amount of each stabilizer added in the stabilizer addition step is in the range of ii to wt%, respectively, and the stabilizer addition step is completed. And the purity of the individual antibody at the time of sealing step 2 is 80% or more. . 16. The method for producing a preparation comprising a monoclonal antibody according to claim 12, wherein the stabilizer addition step of each of the stabilizers is in the range of 重量]% by weight and at the end of the stabilizer addition step (four) The purity of the individual antibody at step = bundle is 80% or more. ' ° 139962.doc 201002821 IV. Designated representative map: (1) The representative representative of the case is: (none) (2) The symbolic symbol of the representative figure is simple: 5. If there is a chemical formula in this case, please reveal the best display invention. Characteristic chemical formula: (none) 139962.doc
TW098114515A 2008-07-02 2009-04-30 Method of producing agent containing single useful material that has polypeptide TW201002821A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2008173135A JP2011184299A (en) 2008-07-02 2008-07-02 Method for producing preparation containing monoclonal antibody

Publications (1)

Publication Number Publication Date
TW201002821A true TW201002821A (en) 2010-01-16

Family

ID=41465762

Family Applications (1)

Application Number Title Priority Date Filing Date
TW098114515A TW201002821A (en) 2008-07-02 2009-04-30 Method of producing agent containing single useful material that has polypeptide

Country Status (3)

Country Link
JP (1) JP2011184299A (en)
TW (1) TW201002821A (en)
WO (1) WO2010001659A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016154290A1 (en) 2015-03-23 2016-09-29 Alexion Pharmaceuticals, Inc. Virus filtration

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998030230A1 (en) * 1997-01-09 1998-07-16 Yoshitomi Pharmaceutical Industries, Ltd. Protein-containing compositions and process for producing the same
KR101251902B1 (en) * 2003-02-25 2013-04-08 메디뮨 엘엘씨 Methods of producing inflenza vaccine compositions

Also Published As

Publication number Publication date
WO2010001659A1 (en) 2010-01-07
JP2011184299A (en) 2011-09-22

Similar Documents

Publication Publication Date Title
CN105051067B (en) CD 20-binding immunotoxins for causing cellular internalization and methods of use thereof
RU2661764C2 (en) Compositions and methods based on dac hyp
JP5755136B2 (en) Virus removal method in high concentration monoclonal antibody solution
CN1195577C (en) Filter membranes for physiologically active substances
CN107109337A (en) The double bag assemblies of T-shaped cell culture
CN1200742C (en) Methods and compositions for use in perfusion applications
JP2017505118A (en) Fusion protein that suppresses neovascularization or growth of blood vessel and use thereof
CN113430174B (en) High-lytic salmonella bacteriophage RDP-SA-19009 and application thereof
CN102858975A (en) Antibody against serotype a lipopolysaccharide of pseudomonas aeruginosa
JP6768012B2 (en) Wide spectrum anti-infective peptide
Shtreimer Kandiyote et al. Synergy on surfaces: Anti-biofouling interfaces using surface-attached antimicrobial peptides PGLa and magainin-2
TW201002821A (en) Method of producing agent containing single useful material that has polypeptide
Aksu et al. Nanobodies to multiple spike variants and inhalation of nanobody-containing aerosols neutralize SARS-CoV-2 in cell culture and hamsters
JP6883102B2 (en) Filtration method for protein-containing liquid
CN108424874A (en) A kind of cell culture medium and preparation method thereof
CN110872345B (en) Preparation method of high-purity immunoglobulin G
Madaeni et al. Removal of mixtures of viruses using microfiltration membrane
JP6731055B2 (en) Method for treating solution contaminated with porcine circovirus
RU2190220C1 (en) Method for finding tuberculosis micobacteria
RU2415922C1 (en) Nutrient medium for lactic acid bacilli cultivation
RU2538624C1 (en) Method for preparing tuberculin for mass diagnosis and prevention of tuberculosis
TW202039836A (en) Viral inactivation methods for continuous manufacturing of antibodies
RU2525637C2 (en) Nutrient medium for cultivation of listeriosis causative agent
KR100561688B1 (en) Revers osmotic membrane filtration method of concentration and lowering salt concentration of korean traditional soy sauce
RU2528101C2 (en) Nutrient medium for legionella culture