TW201731523A - Composition comprising long-acting erythropoietin - Google Patents

Composition comprising long-acting erythropoietin Download PDF

Info

Publication number
TW201731523A
TW201731523A TW105132540A TW105132540A TW201731523A TW 201731523 A TW201731523 A TW 201731523A TW 105132540 A TW105132540 A TW 105132540A TW 105132540 A TW105132540 A TW 105132540A TW 201731523 A TW201731523 A TW 201731523A
Authority
TW
Taiwan
Prior art keywords
epo
solution
composition
mol
sialic acid
Prior art date
Application number
TW105132540A
Other languages
Chinese (zh)
Other versions
TWI654991B (en
Inventor
金閔友
鍾宥坤
韓尚叡
成永喆
梁世煥
禹晶媛
楊尚仁
Original Assignee
綠十字股份有限公司
格納西尼有限公司
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 綠十字股份有限公司, 格納西尼有限公司 filed Critical 綠十字股份有限公司
Publication of TW201731523A publication Critical patent/TW201731523A/en
Application granted granted Critical
Publication of TWI654991B publication Critical patent/TWI654991B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Analytical Chemistry (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Engineering & Computer Science (AREA)
  • Water Supply & Treatment (AREA)

Abstract

The present invention provides a composition containing long-acting erythropoietin, and a method for preparation thereof. More particularly, there is provided an EPO-Fc fusion protein composition with excellent bio-sustainability and high purity, wherein sialic acid content of EPO-Fc is 17 mol/mol or more, and host-cell derived protein (HCP) impurity is included in an amount of 100 ng/mg or less.

Description

包含長效紅血球生成素之組成物 a composition comprising long-acting erythropoietin

本發明是關於一種包括長效紅血球生成素的醫藥組成物及其製備方法。 The present invention relates to a pharmaceutical composition comprising long-acting erythropoietin and a process for the preparation thereof.

包括165個胺基酸的紅血球生成素(EPO)是一種糖蛋白以促進骨髓內紅血球母細胞的分化且產生紅血球。EPO最多於腎臟產生且已知亦少量於肝臟產生。例如,如於慢性腎衰竭(CRF)所示,腎臟功能的損失典型地引起EPO產生的下降以及因此造成相關紅血球的下降。業已發現EPO對於因CRF引起的貧血和因多元成因造成的其他貧血的治療具有優異功效,以及具有如外科手術輔助之不同的造血功能。 Erythropoietin (EPO), which includes 165 amino acids, is a glycoprotein that promotes differentiation of red blood cells in the bone marrow and produces red blood cells. EPO is produced at most in the kidney and is known to be produced in small amounts in the liver. For example, as shown by chronic renal failure (CRF), loss of renal function typically causes a decrease in EPO production and thus a decrease in associated red blood cells. EPO has been found to have excellent efficacy in the treatment of anemia caused by CRF and other anemia caused by multiple causes, as well as having different hematopoietic functions such as surgical assistance.

一般而言,如EPO之多肽具有低穩定度且容易被蛋白質水解酶,亦即血液中的蛋白酶,所退化及分解,由此經由腎臟或肝臟容易地排除。因此,多肽有短的生物半衰期間(半衰期)。所以,為了維持含有多肽作為藥學組成的蛋白質醫藥的血液濃度及效價,該蛋白質醫藥需要經常投藥給患者。然而,在該欲投藥的蛋白質醫藥為注 射調配物類型的情況中,經常注射以保持活性多肽的血液濃度通常可能引起患者疼痛。 In general, polypeptides such as EPO have low stability and are easily degraded and decomposed by proteolytic enzymes, that is, proteases in the blood, thereby being easily eliminated via the kidney or liver. Thus, a polypeptide has a short biological half-life period (half-life). Therefore, in order to maintain the blood concentration and potency of a protein medicine containing a polypeptide as a pharmaceutical composition, the protein medicine needs to be frequently administered to a patient. However, in the protein medicine to be administered, it is a note. In the case of a type of priming formulation, frequent injections to maintain the blood concentration of the active polypeptide may often cause pain in the patient.

為了增加EPO多肽的半衰期,目前已開發使用免疫球蛋白(Ig)所製備的融合蛋白質。Ig是血液的重要構造性組成。人類Ig(hIg)可包括多種類別如IgG、IgM、IgA、IgD和IgE。 In order to increase the half-life of EPO polypeptides, fusion proteins prepared using immunoglobulin (Ig) have been developed. Ig is an important structural component of blood. Human Ig (hIg) can include a variety of classes such as IgG, IgM, IgA, IgD, and IgE.

免疫球蛋白含有四個多肽鏈,且更具體而言,兩個重鏈和兩個輕鏈,其中該等鏈是經由雙硫鍵一起組合以形成四聚體。各鏈由變異區和恆定區所組成。重鏈的恆定區(重鏈恆定區)根據其同型(isotype)可再分為三或四個區段(site)如CH1、CH2、CH3和CH4。重鏈恆定區的Fc區依Ig的同型可包括鉸鏈(hinge)、CH2、CH3和/或CH4功能域(domain)。 An immunoglobulin contains four polypeptide chains, and more specifically two heavy chains and two light chains, wherein the chains are combined together via disulfide bonds to form a tetramer. Each chain consists of a variant region and a constant region. The constant region of the heavy chain (the heavy chain constant region) can be subdivided into three or four sites such as CH1, CH2, CH3 and CH4 depending on its isotype. The Ig isotype of the Fc region of the heavy chain constant region may include a hinge, CH2, CH3, and/or CH4 domain.

關於半衰期,融合於免疫球蛋白之Fc區域的嵌合蛋白質已知顯示增加安定性與延長的血清半衰期。 Regarding half-life, chimeric proteins fused to the Fc region of an immunoglobulin are known to exhibit increased stability and prolonged serum half-life.

再者,已知黏附唾液酸至EPO蛋白質的表面可於肝臟中誘發蛋白質降解的預防。唾液酸於去唾液酸糖蛋白受體中保護第二個半乳糖基因而因此影響活體中的EPO活性(生物活性)。已知存在於紅血球生成素的糖鏈的唾液酸在紅血球生成素半衰期的延長扮演重要角色,最終地,影響其功效。再者,由於已發現紅血球生成素的生物活性藉由唾液酸酶的反應而失活,更活躍地進行唾液酸對紅血球生成素生物活性的功能的研究。在多數情況下,使用於治療劑製造中的糖在表現糖蛋白的本質功效上具有基礎及不可 少的角色。尤其是,對於人衍生的紅血球生成素,糖的角色是絕對需要的。換言之,假如不包括糖,紅血球生成素不顯示活性,根據糖的結構組成或於終端的唾液酸的含量,EPO的生物活性顯著地受到影響。 Furthermore, it is known that adhesion of sialic acid to the surface of EPO protein can induce prevention of protein degradation in the liver. The sialic acid protects the second galactose gene in the asialoglycoprotein receptor and thus affects the EPO activity (biological activity) in the living body. It is known that sialic acid present in the sugar chain of erythropoietin plays an important role in prolonging the half-life of erythropoietin and, ultimately, affects its efficacy. Furthermore, since the biological activity of erythropoietin has been found to be inactivated by the reaction of sialidase, the function of sialic acid on the biological activity of erythropoietin is more actively studied. In most cases, the sugar used in the manufacture of therapeutic agents has a basis and does not represent the essential efficacy of glycoproteins. Less characters. In particular, for human-derived erythropoietin, the role of sugar is absolutely necessary. In other words, if blood sugar is not included, erythropoietin does not exhibit activity, and the biological activity of EPO is significantly affected depending on the structural composition of the sugar or the content of sialic acid at the terminal.

宿主細胞表現期間,EPO-Fc中的唾液酸含量可藉由添加N-乙醯基甘露糖胺至培養溶液而增加。上述過程後所得到的EPO-Fc通常是高唾液酸含量的EPO-Fc和低唾液酸含量的EPO-Fc的混合物。在此,已知高唾液酸含量的EPO-Fc較低唾液酸含量的EPO-Fc具有較長的半衰期。因此,有需要開發出選擇性地製造具有增加的唾液酸含量的EPO-Fc的技術。 The sialic acid content in EPO-Fc during host cell expression can be increased by the addition of N-ethinylmannosamine to the culture solution. The EPO-Fc obtained after the above process is usually a mixture of a high sialic acid content EPO-Fc and a low sialic acid content EPO-Fc. Here, EPO-Fc having a high sialic acid content of EPO-Fc having a lower sialic acid content has a longer half life. Therefore, there is a need to develop a technique for selectively producing EPO-Fc having an increased sialic acid content.

EPO-Fc製造時,需要培養經轉形的宿主細胞,表現大量的EPO-Fc,崩解細胞以及純化EPO-Fc。因此,對於大量生產高品質EPO-Fc,最適化培養和純化技術為非常重要。於目前已開發的培養和純化技術仍有大的改良空間,因此,迫切需要的是經由開發上述技術以最適化EPO-Fc製備過程。韓國專利公告案號897938(2009年5月8日登錄)揭露免疫球蛋白融合蛋白質。 In the production of EPO-Fc, it is necessary to culture transfected host cells, express a large amount of EPO-Fc, disintegrate cells, and purify EPO-Fc. Therefore, it is very important to optimize the culture and purification techniques for mass production of high quality EPO-Fc. There is still much room for improvement in the culture and purification techniques that have been developed so far, and it is therefore highly desirable to optimize the EPO-Fc preparation process by developing the above techniques. Korean Patent Publication No. 897938 (registered on May 8, 2009) discloses an immunoglobulin fusion protein.

因此,本發明的目的是提供一種在活體中具有優異持續性(生物持續性)的EPO-Fc融合蛋白質組成物。 Accordingly, an object of the present invention is to provide an EPO-Fc fusion protein composition having excellent persistence (biosustainability) in a living body.

此外,本發明的另一個目的是提供一種具有高唾液酸含量的EPO-Fc融合蛋白質組成物。 Further, another object of the present invention is to provide an EPO-Fc fusion protein composition having a high sialic acid content.

再者,本發明的另一個目的是提供一種具有高純度的EPO-Fc融合蛋白質組成物。 Furthermore, another object of the present invention is to provide an EPO-Fc fusion protein composition having high purity.

進一步地,本發明的另一個目的是提供一種具有降低含量的宿主細胞衍生的雜質的EPO-Fc融合蛋白質組成物。 Further, it is another object of the present invention to provide an EPO-Fc fusion protein composition having a reduced amount of host cell-derived impurities.

本發明的上述目的藉由下述特徵達成: The above object of the present invention is achieved by the following features:

(1)一種EPO-Fc組成物,包括EPO-Fc,該EPO-Fc具有唾液酸的含量是17mol/mol或者更多,以及宿主細胞衍生的蛋白質雜質的量是100ng/mg或者更少。 (1) An EPO-Fc composition comprising EPO-Fc having a sialic acid content of 17 mol/mol or more, and a host cell-derived protein impurity amount of 100 ng/mg or less.

(2)如上述(1)之EPO-Fc組成物,其中唾液酸含量範圍從20至28mol/mol。 (2) The EPO-Fc composition according to (1) above, wherein the sialic acid content ranges from 20 to 28 mol/mol.

(3)如上述(1)之EPO-Fc組成物,其中EPO-Fc具有pI6.0。 (3) The EPO-Fc composition according to (1) above, wherein the EPO-Fc has a pI 6.0.

(4)如上述(1)之EPO-Fc組成物,其中EPO-F具有4.5pI5.3。 (4) The EPO-Fc composition according to (1) above, wherein the EPO-F has 4.5 pI 5.3.

(5)如上述(1)之EPO-Fc組成物,其中宿主細胞衍生的蛋白質雜質是EPO-Fc的凝集物或片段。 (5) The EPO-Fc composition according to (1) above, wherein the host cell-derived protein impurity is an agglutinant or a fragment of EPO-Fc.

(6)如上述(1)之EPO-Fc組成物,其中宿主細胞衍生的蛋白質雜質的量是60ng/mg或者更少。 (6) The EPO-Fc composition according to (1) above, wherein the amount of the host cell-derived protein impurity is 60 ng/mg or less.

(7)如上述(1)之EPO-Fc組成物,進一步包括0.5ng/mg或者更少的宿主細胞衍生的DNA雜質。 (7) The EPO-Fc composition according to (1) above, which further comprises 0.5 ng/mg or less of host cell-derived DNA impurities.

(8)一種製備EPO-Fc組成物的方法,包括:於溫度範圍35℃至39℃以及6.5pH7.5的條件下以4vvd或更低的流動速率流動EPO-Fc細胞培養溶液,以製備灌注培養溶液; 從灌注培養液得到EPO-Fc純溶液;以及EPO-Fc純溶液吸附至陰離子交換樹脂以製備Q流出物,其包括具有唾液酸含量是17mol/mol或者更多的EPO-Fc。 (8) A method of preparing an EPO-Fc composition comprising: at a temperature ranging from 35 ° C to 39 ° C and 6.5 pH The EPO-Fc cell culture solution is flowed at a flow rate of 4 vvd or lower under conditions of 7.5 to prepare a perfusion culture solution; an EPO-Fc pure solution is obtained from the perfusion culture solution; and the EPO-Fc pure solution is adsorbed to the anion exchange resin to prepare Q effluent comprising EPO-Fc having a sialic acid content of 17 mol/mol or more.

(9)如上述(8)的方法,其中Q流出物的製備包括:EPO-Fc吸附至陰離子交換樹脂;以及使用含有0.005到0.02M的磷酸鈉、0.05到0.2M的L-精胺酸和0.02到0.1M的氯化鈉,且具有6.7pH7.1的緩衝液,洗脫出經吸附的EPO-Fc。 (9) The method according to (8) above, wherein the preparation of the Q effluent comprises: adsorption of EPO-Fc to the anion exchange resin; and use of sodium phosphate containing 0.005 to 0.02 M of sodium phosphate, 0.05 to 0.2 M, and 0.02 to 0.1 M sodium chloride with 6.7 pH The buffer of 7.1 elutes the adsorbed EPO-Fc.

(10)如上述(8)的方法,其中得到EPO-Fc純溶液的步驟包括將灌注培養溶液通過POD過濾器。 (10) The method of (8) above, wherein the step of obtaining an EPO-Fc pure solution comprises passing the perfusion culture solution through a POD filter.

(11)如上述(8)的方法,其中宿主細胞衍生的蛋白質(HCP)雜質的量於EPO-Fc組成物中為100ng/mg或者更少。 (11) The method according to (8) above, wherein the amount of host cell-derived protein (HCP) impurities is 100 ng/mg or less in the EPO-Fc composition.

(12)如上述(8)的方法,其中EPO-Fc細胞培養溶液,係以2.0x105細胞/mL或更多的量接種至培養器。 (12) The method according to the above (8), wherein the EPO-Fc cell culture solution is inoculated to the incubator in an amount of 2.0 x 10 5 cells/mL or more.

(13)如上述(10)的方法,進一步包括,灌注培養溶液通過POD過濾器之後,將上述溶液通過經填充有蛋白質A-結合樹脂的管柱。 (13) The method of (10) above, further comprising, after the perfusion culture solution passes through the POD filter, passing the solution through a column packed with the protein A-binding resin.

(14)如上述(8)的方法,其中Q流出物中所含有的EPO-Fc具有pI是6.0或者更低的。 (14) The method according to (8) above, wherein the EPO-Fc contained in the Q effluent has a pI of 6.0 or less.

(15)如上述(8)的方法,進一步包括分別使用POD過濾器、超濾膜以及奈米過濾器過濾Q流出物。 (15) The method of (8) above, further comprising filtering the Q effluent using a POD filter, an ultrafiltration membrane, and a nanofilter, respectively.

根據本發明的EPO-Fc組成物的特徵是糖鏈末端的常現部份是用唾液酸封端,因此增加半衰期以及增強患者的便利性。 The EPO-Fc composition according to the present invention is characterized in that the normally occurring portion of the sugar chain end is capped with sialic acid, thereby increasing half-life and enhancing patient convenience.

根據本發明的EPO-Fc組成物具有降低含量的宿主細胞衍生的蛋白質雜質和DNA雜質,因此具有高的EPO-Fc純度。 The EPO-Fc composition according to the present invention has a reduced content of host cell-derived protein impurities and DNA impurities, and thus has high EPO-Fc purity.

根據本發明之用於製備EPO-Fc組成物的方法可能適合用於具有上述特徵之EPO-Fc的選擇性純化。 The method for preparing an EPO-Fc composition according to the present invention may be suitable for selective purification of EPO-Fc having the above characteristics.

根據本發明之用於製備EPO-Fc組成物的方法可能顯著地改良唾液酸的附著至EPO-Fc和增強宿主細胞的存活,以及增強過程效率。 The method for preparing an EPO-Fc composition according to the present invention may significantly improve the attachment of sialic acid to EPO-Fc and enhance the survival of host cells, as well as enhance process efficiency.

由後文的詳細說明以及隨附的圖式使本發明之上述及其他的目的、特徵和其他優點更清楚地被了解。 The above and other objects, features and other advantages of the present invention will become apparent from the <RTIgt;

第1圖是顯示EPO-Fc等電點聚焦(isoelectric focusing)檢查的檢測結果圖。 Fig. 1 is a view showing the detection results of an EPO-Fc isoelectric focusing inspection.

本案發明者已基於用於改良製造性及唾液酸含量的宿主細胞培養技術、高唾液酸含量EPO-Fc選擇技術以及用於移除宿主細胞衍生的雜質而完成本發明。 The inventors of the present invention have completed the present invention based on host cell culture techniques for improving manufacturability and sialic acid content, high sialic acid content EPO-Fc selection techniques, and for removing host cell derived impurities.

本發明揭露一種EPO-Fc組成物,其含有具17mol/mol或者更多唾液酸,以及100ng/mg或者更少的宿主細胞衍生的蛋白質(宿主細胞蛋白質,HCP)雜質的EPO-Fc,和用於其製備的方法。 The present invention discloses an EPO-Fc composition comprising EPO-Fc having 17 mol/mol or more of sialic acid, and 100 ng/mg or less of host cell-derived protein (host cell protein, HCP) impurities, and The method for its preparation.

以下,本發明將更詳細說明。 Hereinafter, the present invention will be described in more detail.

EPO-Fc融合蛋白質是藉由將人類免疫球蛋白重鏈恆定區的Fc區融合到紅血球生成素(EPO)而形成, 且具有較EPO為更長的半衰期。該增加的半衰期是指增加功效的生物可持續性。 The EPO-Fc fusion protein is formed by fusing the Fc region of the human immunoglobulin heavy chain constant region to erythropoietin (EPO). And has a longer half-life than EPO. This increased half-life refers to the biological sustainability of increased efficacy.

唾液酸存在於EPO的糖鏈,保護去唾液酸糖蛋白受體的第二個半乳糖基團,如此大幅地影響EPO的生物活性。進一步地,唾液酸可於肝臟中誘發EPO降解的預防因而增加半衰期。 Sialic acid is present in the sugar chain of EPO, protecting the second galactose group of the asialoglycoprotein receptor, thus greatly affecting the biological activity of EPO. Further, sialic acid can induce prevention of EPO degradation in the liver and thus increase half-life.

因此,除了起因於Fc融合的半衰期增加功效外,EPO-Fc融合蛋白質中所含有的唾液酸的附著可提供增加半衰期的功效。 Thus, in addition to the increased half-life due to Fc fusion, attachment of sialic acid contained in the EPO-Fc fusion protein provides increased half-life efficacy.

在EPO-Fc宿主細胞的培養期間,可將N-乙醯基甘露糖胺(NAM)添加於培養溶液以增加EPO-Fc的唾液酸含量。上述過程後所得的EPO-Fc通常包括高唾液酸含量的EPO-Fc和低唾液酸含量的EPO-Fc的混合物,其中,已知高唾液酸含量的EPO-Fc較低唾液酸含量的EPO-Fc,具有較長的半衰期。 During the culture of the EPO-Fc host cells, N-ethylmercaptomannosamine (NAM) can be added to the culture solution to increase the sialic acid content of the EPO-Fc. The EPO-Fc obtained after the above process generally comprises a mixture of a high sialic acid content of EPO-Fc and a low sialic acid content of EPO-Fc, wherein a high sialic acid content of EPO-Fc is known to have a lower sialic acid content of EPO- Fc has a longer half-life.

根據本發明的一具體例,附著至EPO-Fc的唾液酸數目係增加,pI值係降低。基於此面向,使用陰離子交換樹脂可選擇性地純化高唾液酸含量的EPO-Fc。較佳的是使用含有交聯瓊脂糖的陰離子交換樹脂。根據一具體例,可以使用GE醫療公司(GE Healthcare Co.)所製造的Q-Sepharose樹脂。 According to a specific example of the present invention, the number of sialic acid attached to EPO-Fc is increased, and the pI value is decreased. Based on this aspect, the high sialic acid content of EPO-Fc can be selectively purified using an anion exchange resin. It is preferred to use an anion exchange resin containing crosslinked agarose. According to a specific example, Q-Sepharose resin manufactured by GE Healthcare Co. can be used.

藉由使用陰離子交換樹脂的純化過程,可選擇性地純化具有pI6的EPO-Fc。特別地,具有唾液酸含量是17mol/mol或者更多的EPO-Fc可經由此過程獲得。 根據一具體例,在4.5pI6.0的情況,可以獲得唾液酸含量是17到28mol/mol之含有EPO-Fc的組成物。根據另一具體例,在4.5pI5.3的情況,可以獲得唾液酸含量是20到28mol/mol之含有EPO-Fc的組成物。 Selective purification with pI by purification process using anion exchange resin 6 EPO-Fc. In particular, EPO-Fc having a sialic acid content of 17 mol/mol or more can be obtained through this process. According to a specific example, at 4.5 pI In the case of 6.0, an EPO-Fc-containing composition having a sialic acid content of 17 to 28 mol/mol can be obtained. According to another specific example, at 4.5 pI In the case of 5.3, an EPO-Fc-containing composition having a sialic acid content of 20 to 28 mol/mol can be obtained.

在EPO-Fc製造期間,可能混有宿主細胞衍生的蛋白質(HCP)雜質。HCP雜質可包括,例如,含有如可能衍生自宿主細胞及其他材料之不同凝集物及片段的不正常胜肽的蛋白質雜質、DNA雜質、固有或外來的病毒和其他的顆粒或類似物。 Host cell-derived protein (HCP) impurities may be mixed during the manufacture of EPO-Fc. HCP impurities can include, for example, protein impurities, DNA impurities, intrinsic or foreign viruses, and other particles or the like that contain abnormal peptides such as may be derived from different agglutinates and fragments of host cells and other materials.

消除這些雜質可能是一個重要的過程而直接影響EPO-Fc融合蛋白質的品質。當這個組成物是經由根據本發明的一系列過程而製備時,HCP可調控在100ng/mg或者更少的量,以及較佳是60ng/mg或者更少。宿主細胞衍生的DNA雜質可能可以0.5ng/mg或更少的量含有。 Elimination of these impurities may be an important process that directly affects the quality of the EPO-Fc fusion protein. When this composition is prepared via a series of processes according to the present invention, the HCP can be adjusted in an amount of 100 ng/mg or less, and preferably 60 ng/mg or less. Host cell-derived DNA impurities may be contained in an amount of 0.5 ng/mg or less.

用於製備本發明的EPO-Fc組成物的方法,可依下述完成: The method for preparing the EPO-Fc composition of the present invention can be accomplished as follows:

用於製備本發明的EPO-Fc組成物可包括:(1)於溫度範圍35℃至39℃以及6.5pH7.5的條件下,以4vvd或更低的流動率流動EPO-Fc細胞培養溶液以製備灌注培養溶液;(2)從灌注培養溶液得到EPO-Fc純溶液;(3)EPO-Fc純溶液吸附至陰離子交換樹脂,以製備具有唾液酸含量17mol/mol或者更多之含有EPO-Fc的Q流出物。 The EPO-Fc composition used to prepare the present invention may comprise: (1) at a temperature in the range of 35 ° C to 39 ° C and 6.5 pH Under conditions of 7.5, the EPO-Fc cell culture solution is flowed at a flow rate of 4 vvd or lower to prepare a perfusion culture solution; (2) an EPO-Fc pure solution is obtained from the perfusion culture solution; (3) the EPO-Fc pure solution is adsorbed to An anion exchange resin was prepared to prepare an EPO-Fc-containing Q effluent having a sialic acid content of 17 mol/mol or more.

步驟(1)的灌流培養溶液可以如下述製備。 The perfusion culture solution of the step (1) can be prepared as follows.

首先,EPO-Fc主細胞庫(master cell bank,MCB)可從用於EPO-Fc研究的細胞庫(RCB)來建立,以及EPO-Fc工作細胞庫(working cell bank,WCB)可從EPO-Fc主細胞庫(MCB)來建立。 First, the EPO-Fc master cell bank (MCB) can be established from the cell bank (RCB) for EPO-Fc studies, and the EPO-Fc working cell bank (WCB) can be obtained from EPO- The Fc master cell bank (MCB) was established.

用於建立MCB和WCB的培養可包括於3至7% CO2和37℃條件下,於包括L-穀胺醯胺及甲胺蝶呤的EX-cell CHO DHFR(-)的液體培養基中,次培養細胞,然後,將產物分配在用於冷凍和儲存的低溫小瓶。 The culture for establishing MCB and WCB may be included in a liquid medium of EX-cell CHO DHFR (-) including L-glutamine and methotrexate at 3 to 7% CO 2 and 37 ° C. The cells are cultured and the product is then dispensed into cryo vials for freezing and storage.

在經冷凍及儲存的WCB細胞株解凍之後,經解凍的細胞株係提供且懸浮在培養基中以於培養基中增殖細胞。例如,次培養可使用搖瓶於經設定為3到7% CO2和37℃的CO2培養箱,以1:2到1:6的比率,間隔50到90小時進行。該培養中使用數個搖瓶可使培養的細胞數達到足以接種到細胞培養箱的足量。 After thawing of the frozen and stored WCB cell line, the thawed cell line is provided and suspended in the medium to proliferate the cells in the medium. For example, the secondary culture can be carried out using a shake flask in a CO 2 incubator set to 3 to 7% CO 2 and 37 ° C at a ratio of 1:2 to 1:6 at intervals of 50 to 90 hours. The use of several shake flasks in this culture allows the number of cells cultured to be sufficient to be inoculated into the cell culture incubator.

在這種情況下,可使用EPO-Fc培養基。EPO-Fc培養基可包括,例如,EX-cell CHO DHFR(-)的粉末培養基、L-穀胺醯胺、甲胺蝶呤和碳酸氫鈉。 In this case, EPO-Fc medium can be used. The EPO-Fc medium may include, for example, a powder medium of EX-cell CHO DHFR (-), L-glutamine, methotrexate, and sodium hydrogencarbonate.

當用於接種的足量宿主細胞係經由搖瓶培養確保時,該等細胞可接種到主要培養箱,以便進行灌注培養。此處使用的主要培養箱可為30公升生物培養箱。 When a sufficient number of host cell lines for inoculation are ensured via shake flask culture, the cells can be seeded into a primary incubator for perfusion culture. The main incubator used here can be a 30 liter biological incubator.

在接種到主要培養箱後,可獲得細胞濃度為2.0x105細胞/mL或更高。 After inoculation into the main incubator, a cell concentration of 2.0 x 10 5 cells/mL or higher can be obtained.

主要培養箱的培養溫度範圍可從35到39℃或從36到38℃等。 The culture temperature of the main incubator can range from 35 to 39 ° C or from 36 to 38 ° C.

主要培養箱的酸度可維持在範圍6.5pH7.5(在pH調控後),6.8pH7.2(在pH調控後)等。 The acidity of the main incubator can be maintained in the range of 6.5 pH 7.5 (after pH regulation), 6.8 pH 7.2 (after pH control) and so on.

在灌流培養期間,若有需要,培養溶液可以取樣且以顯微鏡觀察,以便監控細胞的情況,過程檢測可以藉由分析pH、細胞數、細胞活性、葡萄糖濃度、穀胺醯胺濃度、氨濃度、滲透壓等而實施。 During perfusion culture, if necessary, the culture solution can be sampled and observed under a microscope to monitor the condition of the cells. The process can be analyzed by pH, cell number, cell activity, glucose concentration, glutamine concentration, ammonia concentration, It is implemented by osmotic pressure or the like.

灌流率可調控為4vvd或更低。灌流率範圍可從0到4vvd、0到2vvd、1到3vvd、1到2vvd等。 The perfusion rate can be adjusted to 4vvd or lower. The perfusion rate can range from 0 to 4 vvd, 0 to 2 vvd, 1 to 3 vvd, 1 to 2 vvd, and the like.

用於此處的灌流培養基可包括藉由在EPO-Fc培養基添加N-乙醯基甘露糖胺所製備的培養基(即是經添加N-乙醯基甘露糖胺的培養基),在EPO-Fc培養基添加N-乙醯基甘露糖胺及葡萄糖所製備的培養基(製造培養基),或這兩種培養基可依序使用。 The perfusion medium used herein may include a medium prepared by adding N-ethylmercaptomannamine in EPO-Fc medium (that is, a medium to which N-ethylmercaptomannamine is added) in EPO-Fc. A medium (manufacturing medium) prepared by adding N-acetylmercaptoamine and glucose to the medium, or both of the mediums may be used in sequence.

例如,根據每天40公升灌流培養4天,可得到總共140至180公斤的用於EPO-Fc製造的細胞培養溶液。重複上述收集細胞培養溶液一次到十次或二次到五次,可提供更增量的用於EPO-Fc製造的細胞培養溶液。 For example, a total of 140 to 180 kg of a cell culture solution for EPO-Fc production can be obtained according to a 40-liter perfusion per day culture for 4 days. Repeating the above collection of the cell culture solution once to ten or two to five times provides a more incremental cell culture solution for EPO-Fc production.

如上述所得到的細胞培養溶液,可成為本發明的步驟(1)的灌流培養溶液。 The cell culture solution obtained as described above can be a perfusion culture solution of the step (1) of the present invention.

根據本發明之製備灌流培養溶液的步驟,17mol/mol或更多、17至28mol/mol、或20至28mol/mol的唾液酸可黏附至EPO-Fc。進一步地,細胞的存活可經改良因而增強EPO-Fc組成物的生產性。 According to the step of preparing the perfusion culture solution of the present invention, 17 mol/mol or more, 17 to 28 mol/mol, or 20 to 28 mol/mol of sialic acid can adhere to EPO-Fc. Further, the survival of the cells can be modified to enhance the productivity of the EPO-Fc composition.

用於製備本發明之EPO-Fc組成物的方法可 進一步包括(2)從灌流培養溶液得到EPO-Fc純溶液。 A method for preparing the EPO-Fc composition of the present invention Further included is (2) obtaining a pure EPO-Fc solution from the perfusion culture solution.

上述的步驟具有主要目的在於移除除了步驟(1)的灌流培養溶液中所含有的EPO-Fc以外的其他成份。這種移除可包括用於細胞培養的任何傳統方法。這種移除方法可與如上所述的過濾和純化過程的至少一者組合。 The above steps have a main purpose of removing the components other than the EPO-Fc contained in the perfusion culture solution of the step (1). Such removal can include any conventional method for cell culture. This removal method can be combined with at least one of the filtration and purification processes described above.

在回收步驟(1)的灌注培養溶液後,這個溶液可接受用於過濾及純化的多個步驟,因而移除宿主細胞衍生的雜質。該等宿主細胞衍生的雜質可包括,例如,含有如不同的凝集物和片段的異常胜肽的蛋白質雜質、DNA雜質、固有的病毒、外來的病毒和其他顆粒。 After recovering the perfusion culture solution of step (1), this solution can be subjected to various steps for filtration and purification, thereby removing host cell-derived impurities. Such host cell-derived impurities may include, for example, protein impurities, DNA impurities, intrinsic viruses, foreign viruses, and other particles containing anomalous peptides such as different agglutinates and fragments.

宿主細胞是指步驟(1)中用於EPO-Fc表現的所有細胞。 The host cell refers to all cells used in the step (1) for EPO-Fc expression.

根據一具體例,為了移除用於EPO-Fc製造的宿主細胞培養溶液中所含有的宿主細胞衍生的蛋白質(HCP)雜質或DNA雜質,可以使用POD深度濾器。POD深度濾器通常用於移除細胞,然而,本發明用於移除蛋白質。 According to a specific example, in order to remove host cell-derived protein (HCP) impurities or DNA impurities contained in a host cell culture solution for EPO-Fc production, a POD depth filter can be used. POD depth filters are commonly used to remove cells, however, the invention is used to remove proteins.

使用POD深度濾器以及過濾器,可移除如蛋白質凝集物、片段和其他的顆粒的宿主細胞衍生的蛋白質(HCP)雜質。 Host cell-derived protein (HCP) impurities such as protein agglutinates, fragments and other particles can be removed using POD depth filters and filters.

例如,如POD濾器,可以使用Merck Millipore製造的Millistak+(MA1HC10FS1)。進一步地,此處使用的過濾器可為Sartorius Co.製造的Sartobran P無菌等級膠囊。 For example, as a POD filter, Millistak+ (MA1HC10FS1) manufactured by Merck Millipore can be used. Further, the filter used herein may be a Sartobran P aseptic grade capsule manufactured by Sartorius Co.

根據另一具體例,蛋白質A純化,低pH病毒失活和/或羥基磷灰石純化等,可適用於EPO-Fc純化。 According to another specific example, protein A purification, low pH virus inactivation and/or hydroxyapatite purification, etc., are applicable to EPO-Fc purification.

根據另一具體例,超濾裝置,亦即切向流過濾(tangential flow filtration,TFF)膜系統,可用於濃縮和緩衝液更換。若導電率和酸度經由緩衝液更換而在參考範圍內,則完成濃縮和緩衝液更換的過程(參考:導電率9.0±1.0mS/cm,酸度pH 6.9±0.1)。 According to another embodiment, an ultrafiltration unit, i.e., a tangential flow filtration (TFF) membrane system, can be used for concentration and buffer exchange. If conductivity and acidity are replaced by buffers within the reference range, the process of concentration and buffer exchange is completed (reference: conductivity 9.0 ± 1.0 mS/cm, acidity pH 6.9 ± 0.1).

步驟(2)中的EPO-Fc純溶液可如上所述製備。 The EPO-Fc pure solution in the step (2) can be prepared as described above.

用於製備本發明之EPO-Fc組成物的方法可包括(3)EPO-Fc純溶液吸附到陰離子交換樹脂,以製備Q流出物,該Q流出物包括具有唾液酸含量17mol/mol或者更多的EPO-Fc。 The method for preparing the EPO-Fc composition of the present invention may comprise (3) adsorbing a pure solution of EPO-Fc to an anion exchange resin to prepare a Q effluent comprising a sialic acid content of 17 mol/mol or more. EPO-Fc.

上述步驟具有主要的目的在於選出組成物中所含有之EPO-Fc具有唾液酸含量17mol/mol或者更高的特異者。 The above steps have a main purpose of selecting a specific one in which the EPO-Fc contained in the composition has a sialic acid content of 17 mol/mol or more.

這一個步驟可以藉由將步驟(2)中的EPO-Fc純溶液,通過經填充含有交聯瓊脂糖的陰離子交換樹脂的管柱進行。 This step can be carried out by passing the EPO-Fc pure solution in the step (2) through a column packed with an anion exchange resin containing crosslinked agarose.

根據一具體例,可使用GE醫療公司的Q Sepharose樹脂,例如,可使用下表1所示的Q Sepharose Fast Flow樹脂。 According to a specific example, GE Sepharose resin of GE Healthcare can be used, for example, Q Sepharose Fast Flow resin shown in Table 1 below can be used.

製備Q流出物的過程可以包括:藉由使用含有0.005至0.02M磷酸鈉的平衡緩衝液平衡Q-Sepharose樹脂;使EPO-Fc純溶液通過Q-Sepharose樹脂以吸附EPO-Fc;藉由使用上述平衡緩衝液重新平衡Q-Sepharose樹脂;和將含有0.005至0.02M磷酸鈉、0.05至0.2M L-精胺酸和0.02至0.1M氯化鈉且具有酸度為6.7pH7.1的流出液通過該管柱以收集EPO-Fc的Q流出物。 The process of preparing the Q effluent may include: equilibrating the Q-Sepharose resin by using an equilibration buffer containing 0.005 to 0.02 M sodium phosphate; passing the EPO-Fc pure solution through the Q-Sepharose resin to adsorb EPO-Fc; The equilibration buffer rebalances the Q-Sepharose resin; and will contain 0.005 to 0.02 M sodium phosphate, 0.05 to 0.2 M L-arginine and 0.02 to 0.1 M sodium chloride with an acidity of 6.7 pH The effluent from 7.1 was passed through the column to collect the Q effluent of EPO-Fc.

可以適當地重複洗脫。例如,當洗脫重複進行4次時,在第一至第三次純化中獲得的Q流出物No.1至3被儲存在超低溫冷凍冰箱中,並且在完成第四次洗脫後,所有的Q流出物No.1至4可以進行匯集。 The elution can be repeated as appropriate. For example, when the elution is repeated 4 times, the Q effluents No. 1 to 3 obtained in the first to third purifications are stored in an ultra-low temperature freezer, and after the completion of the fourth elution, all Q effluents No. 1 to 4 can be collected.

視需要地,在洗脫後,可以使用POD濾器進行過濾。此處使用的POD濾器可以包括,例如,由Merck Millipore Co.製造的Millistak+(MA1HC10FS1)。 Optionally, after elution, a POD filter can be used for filtration. The POD filter used herein may include, for example, Millistak+ (MA1HC10FS1) manufactured by Merck Millipore Co.

除了上述過程之外,視需要地,可以進一步包括在管柱安裝過濾器的過程和藉由使樹脂與CIP接觸來殺菌和洗滌Q-Sepharose樹脂的過程。根據一具體例,此處使用的過濾器可包括,例如,由Sartorius Co.製造的Sartobran P無菌等級膠囊。 In addition to the above process, as needed, a process of installing a filter on the column and a process of sterilizing and washing the Q-Sepharose resin by bringing the resin into contact with the CIP may be further included. According to a specific example, the filter used herein may include, for example, a Sartobran P aseptic grade capsule manufactured by Sartorius Co.

步驟(3)中得到的Q流出液中所含的EPO-Fc可以具有pI6.0且唾液酸含量為17mol/mol或者更多。例如,EPO-Fc可以為4.5pI6.0且唾液酸含量可為範圍17至28mol/mol,或EPO-Fc可以為4.5pI5.3且唾液酸含量可為範圍20至28mol/mol。 The EPO-Fc contained in the Q effluent obtained in the step (3) may have a pI 6.0 and the sialic acid content was 17 mol/mol or more. For example, EPO-Fc can be 4.5 pI 6.0 and the sialic acid content may range from 17 to 28 mol/mol, or EPO-Fc may be 4.5. pI 5.3 and the sialic acid content may range from 20 to 28 mol/mol.

用於製備本發明的EPO-Fc組成物的方法還可以包括(4)分別經由POD濾器、超濾膜和/或奈米濾器過濾Q流出物。 The method for preparing the EPO-Fc composition of the present invention may further comprise (4) filtering the Q effluent via a POD filter, an ultrafiltration membrane, and/or a nanofilter, respectively.

根據上述步驟,EPO-Fc可以具有增加的濃度,可以更換緩衝液,並且可以消除病毒。 According to the above steps, EPO-Fc can have an increased concentration, the buffer can be exchanged, and the virus can be eliminated.

此處使用的POD濾器可以是由Merck Milipore Co.製造的Millistak+(MA1HC 10FS1)。 The POD filter used herein may be Millistak+ (MA1HC 10FS1) manufactured by Merck Milipore Co.

此處使用的超濾膜可以包括,例如,切向流過濾(TFF)膜系統。例如,在使用注射緩衝液洗滌膜(截留:30K)後,可以使用製劑緩衝液使膜平衡。製劑緩衝液可以含有0.01M檸檬酸鈉、0.1M甘胺酸和0.1M氯化鈉,並且具有pH 6.2±0.2的酸度。 The ultrafiltration membranes used herein may include, for example, a tangential flow filtration (TFF) membrane system. For example, after washing the membrane with an injection buffer (interception: 30K), the formulation buffer can be used to equilibrate the membrane. The formulation buffer may contain 0.01 M sodium citrate, 0.1 M glycine and 0.1 M sodium chloride, and has an acidity of pH 6.2 ± 0.2.

完成平衡後,可將目的蛋白質濃縮至約1.5±0.3mg/mL。如果回收的溶液具有約1.5±0.3mg/mL的蛋白 質濃度,則可以藉由以相同體積連續加入用於製備粗製溶液的緩衝液來進行緩衝液交換。如果最終回收的溶液的導電率和酸度在其參考範圍內,則完成濃度和緩衝液交換過程(參考:導電率12.0±2.0mS/cm,酸度pH6.2±0.2)。 After the equilibration is completed, the protein of interest can be concentrated to about 1.5 ± 0.3 mg/mL. If the recovered solution has a protein of about 1.5 ± 0.3 mg / mL For the concentration, buffer exchange can be carried out by continuously adding a buffer for preparing a crude solution in the same volume. If the conductivity and acidity of the finally recovered solution are within its reference range, the concentration and buffer exchange process is completed (reference: conductivity 12.0 ± 2.0 mS/cm, acidity pH 6.2 ± 0.2).

當最終回收的溶液的濃縮和緩衝液交換過程完成時,可以使用奈米濾器(PALL(NT6DV20P1G))進行過濾,以消除可能源自宿主細胞或在該過程中使用的額外的材料的病毒。 When the concentration of the final recovered solution and the buffer exchange process are complete, a nano filter (PALL (NT6DV20P1G)) can be used for filtration to eliminate viruses that may be derived from host cells or additional materials used in the process.

根據一具體例,在洗滌用於病毒過濾的濾器之後,進行濾器的完整性測試,並且可藉由將用於粗製製造的緩衝液通過病毒過濾器來達成平衡。完成平衡後,濃縮和緩衝液更換的過程完成後的最終回收的溶液可以在30±3psi的壓力下通過濾器。因此,可以回收無病毒的病毒濾液。過濾完成後,可以使用注射緩衝液洗滌病毒過濾器,然後進行完整性測試。 According to a specific example, after washing the filter for virus filtration, the integrity test of the filter is performed, and the balance can be achieved by passing the buffer for the crude manufacturing through the virus filter. Upon completion of the equilibration, the final recovered solution after completion of the concentration and buffer exchange process can be passed through the filter at a pressure of 30 ± 3 psi. Therefore, the virus-free virus filtrate can be recovered. Once the filtration is complete, the virus filter can be washed with injection buffer and then tested for integrity.

對病毒濾液加入濃度為0.12g/Kg的聚山梨醇酯20和製劑緩衝液可以使蛋白質濃度調節至1.1±0.3mg/mL,然後使用滅菌過濾器可以製造EPO-Fc粗製溶液。 The addition of the polysorbate 20 and the formulation buffer at a concentration of 0.12 g/kg to the virus filtrate allowed the protein concentration to be adjusted to 1.1 ± 0.3 mg/mL, and then a crude filter of EPO-Fc was produced using a sterile filter.

根據一具體例,製劑緩衝液可以包括0.01M檸檬酸鈉、0.1M甘胺酸和0.1M氯化鈉,並且具有pH 6.2±0.2的酸度。根據一具體例,此處使用的滅菌過濾器可以是由Sartorius Co.製造的Sartobran P300。所製備的EPO-Fc粗製溶液可以分配並儲存在超低溫冷凍冰箱中。 According to a specific example, the formulation buffer may include 0.01 M sodium citrate, 0.1 M glycine, and 0.1 M sodium chloride, and has an acidity of pH 6.2 ± 0.2. According to a specific example, the sterilization filter used herein may be Sartobran P300 manufactured by Sartorius Co. The prepared EPO-Fc crude solution can be dispensed and stored in an ultra-low temperature freezer.

藉由對EPO-Fc粗製溶液加入製劑緩衝液,然後通過滅菌過濾器過濾,將蛋白質濃度調節至0.5±0.1mg/mL,得到最終的EPO-Fc粗製溶液。此處使用的該滅菌過濾器可以包括例如由Sartorius Co.製造的Sartobran P midicap。 The final EPO-Fc crude solution was obtained by adding the formulation solution to the crude EPO-Fc solution and then filtering through a sterile filter to adjust the protein concentration to 0.5 ± 0.1 mg/mL. The sterilization filter used herein may include, for example, Sartobran P midicap manufactured by Sartorius Co.

在下文中,將參考實施例更詳細地描述本發明。然而,提出這些實施例僅用於說明本發明的較佳具體例,並且本發明的範圍不特別限於此。 Hereinafter, the present invention will be described in more detail with reference to the embodiments. However, the embodiments are merely described to illustrate preferred embodiments of the present invention, and the scope of the present invention is not particularly limited thereto.

實施例1:EPO-Fc組成物的製備 Example 1: Preparation of EPO-Fc composition

<過程1:EPO-Fc主細胞庫(MCB)的製備> <Process 1: Preparation of EPO-Fc Master Cell Bank (MCB)>

對EX-細胞CHO DHFR(-)液體培養基中加入L-穀胺醯胺和甲胺蝶呤以形成培養基並將用於EPO-Fc研究的細胞庫(RCB)接種到培養基後,經由次培養增加總細胞數和總培養體積,從而構建MCB。培養在5.0±2.0%CO2培養箱中進行,直至培養溶液達到37℃的溫度。 Adding L-glutamine and methotrexate to EX-cell CHO DHFR(-) liquid medium to form a medium and inoculating the cell bank (RCB) for EPO-Fc study into the medium, increasing by subculture The total number of cells and the total culture volume were used to construct MCB. The cultivation was carried out in a 5.0 ± 2.0% CO 2 incubator until the culture solution reached a temperature of 37 °C.

<過程2:EPO-Fc工作細胞庫(WCB)的製備> <Process 2: Preparation of EPO-Fc Working Cell Bank (WCB)>

對EX-細胞CHO DHFR(-)液體培養基中加入L-穀胺醯胺和甲胺蝶呤以形成培養基並將過程1所製備的MCB接種到培養基後,經由次培養增加總細胞數和總培養體積,從而構建WCB。培養在5.0±2.0%CO2培養箱中進行,直至培養溶液達到37℃的溫度。 Adding L-glutamine and methotrexate to EX-cell CHO DHFR(-) liquid medium to form medium and inoculating MCB prepared in process 1 into medium, increasing total cell number and total culture through subculture Volume to build WCB. The cultivation was carried out in a 5.0 ± 2.0% CO 2 incubator until the culture solution reached a temperature of 37 °C.

<過程3:WCB融合> <Process 3: WCB Fusion>

在將過程2中經冷凍及儲存的WCB解凍後,經解凍的WCB懸浮在EPO-Fc培養基(包括EX-細胞CHO DHFR(-)粉 末培養基、L-穀胺醯胺、甲胺蝶呤和碳酸氫鈉)中,然後將其培養在設置有5%CO2的37℃的CO2培養箱中。 After thawing the frozen and stored WCB in Process 2, the thawed WCB is suspended in EPO-Fc medium (including EX-cell CHO DHFR (-) powder medium, L-glutamine, methotrexate and carbonic acid In sodium hydrogenate, it was then cultivated in a 37 ° C CO 2 incubator set with 5% CO 2 .

<過程4:搖瓶細胞的培養> <Process 4: Culture of shake flask cells>

將過程3的培養溶液以64至80小時的間隔進行次培養,以確保足以將其接種到細胞培養箱所需的細胞數。使用EPO-Fc培養基在設置有5%CO2和37℃的CO2培養箱中以1:3至1:4的比率進行次培養。 The culture solution of Process 3 was subcultured at intervals of 64 to 80 hours to ensure the number of cells required to inoculate it into the cell culture incubator. The subculture was carried out at a ratio of 1:3 to 1:4 in a CO 2 incubator provided with 5% CO 2 and 37 ° C using EPO-Fc medium.

<過程5:在主培養箱(30L生物培養箱)中的製造和培養> <Process 5: Manufacturing and culture in a main incubator (30 L biological incubator)>

在獲得足量的過程4的培養溶液後,收集細胞並接種到主培養箱中以達到2.0×105細胞/mL。然後,使用其中進一步添加有N-乙醯基甘露糖胺的EPO-Fc培養基培養上述溶液。在培養中保持培養溫度為37±1℃和pH 7.0±0.2(pH調控後),進行灌注培養。如果需要,藉由取樣培養溶液進行過程檢測。根據過程檢測的結果,將灌注率調控在0至2vvd的範圍。為了製備培養溶液,藉由新的EPO-Fc製造培養基(於EPO-Fc培養基中加入N-乙醯基甘露糖胺和碳酸氫鈉所製備)更換所用的培養基,然後進一步進行灌注培養。以約每天40L持續4天收集140至180kg培養溶液,以獲得一個子批次。藉由重複上述過程總共4次,持續16天,進行培養直到收集總共四(4)個子批次。結果,獲得用於EPO-Fc製造的宿主細胞的培養溶液(灌注培養溶液)。 After a sufficient amount of the culture solution of the process 4 was obtained, the cells were collected and inoculated into a main incubator to reach 2.0 × 10 5 cells/mL. Then, the above solution was cultured using an EPO-Fc medium to which N-ethylmercaptomannamine was further added. Perfusion culture was carried out while maintaining the culture temperature at 37 ± 1 ° C and pH 7.0 ± 0.2 (after pH control). Process detection is performed by sampling the culture solution if necessary. The perfusion rate was adjusted in the range of 0 to 2 vvd based on the results of the process detection. To prepare a culture solution, the medium used was replaced with a new EPO-Fc production medium (prepared by adding N-ethylmercaptoamine and sodium hydrogencarbonate to EPO-Fc medium), and then further subjected to perfusion culture. 140 to 180 kg of the culture solution was collected for about 4 days per day for about 4 days to obtain a sub-batch. The culture was carried out by repeating the above process for a total of 4 times for 16 days until a total of four (4) sub-lots were collected. As a result, a culture solution (perfusion culture solution) for a host cell produced by EPO-Fc was obtained.

<過程6:培養溶液的回收和過濾> <Process 6: Recovery and filtration of culture solution>

使用POD濾器(MA1HC10FS1)和過濾器(Sartobran P-無 菌等級膠囊)過濾過程5的用於EPO-Fc製造的宿主細胞培養溶液,然後獲得灌注培養溶液的濾液並儲存。 Use POD filter (MA1HC10FS1) and filter (Sartobran P-None Bacterial grade capsule) The host cell culture solution for EPO-Fc production of Process 5 was filtered, and then the filtrate of the perfusion culture solution was obtained and stored.

<過程7:蛋白質A的純化> <Process 7: Purification of Protein A>

在做成使用平衡緩衝液(0.01M磷酸鈉)以平衡狀態填充蛋白A鍵合樹脂的管柱並將過程6的濾液吸附到管柱之後,使用洗滌緩衝液(0.7M L-精胺酸,pH 5.7±0.05,導電率37.0±1.0mS/cm)清洗管柱,然後使用洗脫緩衝液(0.02M乙酸鈉無水物、0.2M L-精胺酸、7.94%(v/v)甘油,pH 3.7±0.05)洗脫目標蛋白質(EPO-Fc),然後儲存。 After making the column packed with Protein A bonding resin in equilibrium with equilibration buffer (0.01 M sodium phosphate) and adsorbing the filtrate of Process 6 to the column, wash buffer (0.7 M L-arginine, pH 5.7 ± 0.05, conductivity 37.0 ± 1.0 mS / cm) clean the column, then use the elution buffer (0.02 M sodium acetate anhydrate, 0.2 M L-arginine, 7.94% (v / v) glycerol, pH 3.7 ± 0.05) The target protein (EPO-Fc) was eluted and then stored.

<過程8:低pH病毒失活> <Process 8: Low pH virus inactivation>

如果過程7的純溶液測量其pH的結果為不適合作為參考,則對其加入pH調節緩衝液(1M氫氧化鈉(NaOH),10%乙酸)以調節pH(3.7±0.05),接著進行此過程同時攪拌溶液持續2小時。在完成該過程之後,使用pH調節緩衝液(1M氫氧化鈉,10%乙酸)將pH調節至約pH 6.9±0.1。 If the pure solution of Process 7 measures its pH as unsuitable for reference, add pH adjustment buffer (1M sodium hydroxide (NaOH), 10% acetic acid) to adjust the pH (3.7 ± 0.05), followed by this process. The solution was stirred for 2 hours at the same time. After completing the process, the pH was adjusted to about pH 6.9 ± 0.1 using pH adjustment buffer (1 M sodium hydroxide, 10% acetic acid).

<過程9:羥基磷灰石的純化> <Process 9: Purification of Hydroxyapatite>

在填充有固定相(羥基磷灰石)並且用平衡緩衝液(0.01M磷酸鈉)將其平衡的管柱安裝過濾器(Sartobran P-無菌等級膠囊)之後,將過程8的純溶液吸附到管柱,然後,移除過濾器(Sartobran P-無菌等級膠囊),隨後使用洗脫緩衝液(0.1M磷酸鈉和0.1M L-精胺酸,pH 6.9±0.1)洗脫目標蛋白質,然後儲存。 After installing a filter (Sartobran P-sterile grade capsule) filled with a stationary phase (hydroxyapatite) and equilibrating it with equilibration buffer (0.01 M sodium phosphate), the pure solution of process 8 is adsorbed to the tube The column was then removed (Sartobran P-sterile grade capsule) and the target protein was eluted using an elution buffer (0.1 M sodium phosphate and 0.1 M L-arginine, pH 6.9 ± 0.1) and stored.

<過程10:超濾濃縮和緩衝液更換1> <Process 10: Ultrafiltration Concentration and Buffer Replacement 1>

在使用超濾裝置(膜:截斷30K,包括0.5m2)濃縮過程 9的純溶液後,藉由連續加入平衡緩衝液(0.01M磷酸鈉)進行緩衝液更換。當導電率和pH在其參考範圍內時,該過程完成且收集濃縮和緩衝液更換1的滯留溶液,得到EPO-Fc純溶液。 After the pure solution of Process 9 was concentrated using an ultrafiltration apparatus (membrane: cut off 30K, including 0.5 m 2 ), buffer exchange was performed by continuously adding an equilibration buffer (0.01 M sodium phosphate). When the conductivity and pH are within its reference range, the process is complete and the retentate solution of the concentrate and buffer exchange 1 is collected to obtain a pure EPO-Fc solution.

<過程11:Q Sepharose的純化> <Process 11: Purification of Q Sepharose>

在填充有固定相(Q Sepharose)的管柱安裝過濾器(Sartobran P-無菌等級膠囊)並以包含0.01M磷酸鈉的平衡緩衝液平衡Q Sepharose樹脂後,將過程10的EPO-Fc純溶液吸附到管柱,然後,除去過濾器,即Sartobran P無菌等級膠囊,並將Q Sepharose樹脂重新平衡,隨後使用洗脫緩衝液(0.01M磷酸鈉及0.1ML-精胺酸、0.05M氯化鈉,pH 6.9±0.1)洗脫目標蛋白質,以獲得並儲存EPO-Fc的Q流出物。 EPO-Fc pure solution adsorption of process 10 was carried out after installing a filter (Sartobran P-sterile grade capsule) filled with a stationary phase (Q Sepharose) and equilibrating Q Sepharose resin with an equilibration buffer containing 0.01 M sodium phosphate. To the column, then remove the filter, the Sartobran P aseptic grade capsule, and re-equilibrate the Q Sepharose resin, followed by the elution buffer (0.01 M sodium phosphate and 0.1 ML-arginine, 0.05 M sodium chloride, The target protein was eluted at pH 6.9 ± 0.1) to obtain and store the Q effluent of EPO-Fc.

<過程12:流出物的解凍和過濾> <Process 12: Thawing and Filtration of Effluent>

將過程11的經儲存的Q流出物解凍並進行匯集。使用製劑緩衝液(0.01M檸檬酸鈉、0.1M甘胺酸、0.1M氯化鈉,pH 6.2±0.2)平衡POD濾器(MA1HC054H1),隨後藉由使Q流出液通過濾器以獲得濾液。 The stored Q effluent from process 11 is thawed and pooled. The POD filter (MA1HC054H1) was equilibrated using formulation buffer (0.01 M sodium citrate, 0.1 M glycine, 0.1 M sodium chloride, pH 6.2 ± 0.2), and then the filtrate was obtained by passing the Q effluent through a filter.

<過程13:超濾濃縮和緩衝液更換2> <Process 13: Ultrafiltration Concentration and Buffer Replacement 2>

在使用超濾裝置(膜:截留30K,包括0.1M2)濃縮過程12的Q匯集濾液以達到目的蛋白質濃度後,藉由連續添加製劑緩衝液(0.01M檸檬酸鈉、0.1M甘胺酸、0.1M氯化鈉,pH 6.2±0.2)進行緩衝液更換。當導電率和pH在其參考範圍內時,該過程完成,並收集濃縮和緩衝液更換2的滯留溶液。 After the Q collection filtrate of the process 12 was concentrated using an ultrafiltration apparatus (membrane: 30K, including 0.1 M 2 ) to achieve the target protein concentration, a continuous addition of the formulation buffer (0.01 M sodium citrate, 0.1 M glycine, Buffer replacement with 0.1 M sodium chloride, pH 6.2 ± 0.2). When the conductivity and pH are within its reference range, the process is complete and the retention and buffer exchange 2 retention solution is collected.

<過程14:奈米濾器過濾> <Process 14: Nano filter filtration>

將製劑緩衝液(0.01M檸檬酸鈉、0.1M甘胺酸、0.1M氯化鈉,pH 6.2±0.2)流入奈米濾器(Pall(NT6DV20P1G))以使其平衡後,過程13的濃縮和緩衝液更換2的滯留溶液流入其中,從而消除可能存在於濃縮和緩衝液更換2滯留溶液中的病毒。 Concentration and buffering of Process 13 after the formulation buffer (0.01 M sodium citrate, 0.1 M glycine, 0.1 M sodium chloride, pH 6.2 ± 0.2) was flowed into a nano filter (Pall (NT6DV20P1G)) to equilibrate it. The retentate solution of the liquid exchange 2 flows therein, thereby eliminating viruses that may be present in the concentration and buffer exchange 2 retention solution.

<過程15:EPO-Fc粗製溶液的製備> <Process 15: Preparation of EPO-Fc crude solution>

過程14中獲得的病毒濾液中加入0.12g/kg聚山梨醇酯20,並使用製劑緩衝液調節蛋白質濃度。然後,使用滅菌及過濾器(Sartobran P300)進行滅菌和過濾以製備EPO-Fc粗製溶液,同時符合粗製蛋白質濃度(參考:1.1±0.3mg/mL),隨後將其分注至儲存容器並將該容器儲存在超低溫冷凍冰箱中。 0.12 g/kg of polysorbate 20 was added to the virus filtrate obtained in Process 14, and the protein concentration was adjusted using the formulation buffer. Then, sterilization and filtration were carried out using a sterilization and filter (Sartobran P300) to prepare a crude EPO-Fc solution while meeting the crude protein concentration (reference: 1.1 ± 0.3 mg/mL), which was then dispensed into a storage container and The container is stored in an ultra-low temperature freezer.

<過程16:EPO-Fc最終粗製溶液的製備> <Process 16: Preparation of final crude solution of EPO-Fc>

在水浴中解凍過程15的EPO-Fc粗製溶液後,對其加入製劑緩衝液(0.01M檸檬酸鈉、0.1M甘胺酸、0.1M氯化鈉,pH 6.2±0.2,聚山梨醇酯20 0.12g/Kg)以達到0.5±0.1mg/mL的蛋白質濃度,使用滅菌及過濾器(Sartobran P midicap)進行滅菌和過濾,從而製備最終的EPO-Fc粗製溶液。 After thawing the crude EPO-Fc solution of Process 15 in a water bath, it was added to the formulation buffer (0.01 M sodium citrate, 0.1 M glycine, 0.1 M sodium chloride, pH 6.2 ± 0.2, polysorbate 20 0.12). g/Kg) To achieve a protein concentration of 0.5 ± 0.1 mg/mL, sterilized and filtered using a sterilization and filter (Sartobran P midicap) to prepare a final crude EPO-Fc solution.

實施例2:EPO-Fc組成物的純度檢測 Example 2: Purity detection of EPO-Fc composition

<檢測1:宿主細胞衍生的蛋白質(肽)(HCP)雜質含量的測試 <Detection 1: Test for impurity content of host cell-derived protein (peptide) (HCP)

為了檢測最終粗製溶液中的HCP雜質含量,使用CHO宿主細胞蛋白質試劑盒進行以下測試:1)稀釋並使用試劑 盒中提供的標準溶液;2)測試溶液係使用稀釋緩衝液製備為稀釋狀態;3)藉由將標準溶液加入到上述測試溶液中製備添加測試(spiked test)溶液;4)在使標準溶液、測試溶液和添加測試溶液與抗-CHO(磷酸酶連接物)反應之後,使該混合物在抗-CHO包被的微量滴定板中反應;和5)在洗滌板後,對其加入PNPP受質,並使用ELISA讀板器測量吸光度。此處使用的樣品是批號747R0001和747R0002。檢測結果總結在下表2。從測試結果可以看出,宿主細胞衍生的蛋白質雜質的含量為60ng/mg。 To test the HCP impurity content in the final crude solution, the following tests were performed using the CHO Host Cell Protein Kit: 1) Dilution and use of reagents a standard solution provided in the cartridge; 2) the test solution is prepared in a diluted state using a dilution buffer; 3) a spiked test solution is prepared by adding a standard solution to the above test solution; 4) in the standard solution, After the test solution and the addition of the test solution are reacted with an anti-CHO (phosphatase conjugate), the mixture is reacted in an anti-CHO coated microtiter plate; and 5) after washing the plate, a PNPP substrate is added thereto, Absorbance was measured using an ELISA plate reader. The samples used here are lot numbers 747R0001 and 747R0002. The test results are summarized in Table 2 below. It can be seen from the test results that the host cell-derived protein impurity content is 60 ng/mg.

<檢測2:宿主細胞衍生的DNA雜質含量的測試> <Detection 2: Test for DNA content of DNA derived from host cells>

為了檢測EPO-Fc最終粗製溶液中宿主細胞衍生的DNA雜質的含量,進行以下測試:1)將試劑盒中提供的高校標準劑稀釋並使用作為標準溶液;2)將試劑盒中提供的零校標準劑稀釋並用作測試溶液;3)藉由將標準溶液添加到測試樣品中製備添加測試溶液;4)此處使用的零校標準劑是試劑盒中提供的零校標準劑;5)藉由將標準溶液加入 零校標準劑以製備此處使用的添加零校標準劑;6)所製備的標準溶液、測試溶液、添加測試溶液、零校標準劑和添加零校標準劑進行DNA標記;和7)使用閾值系統濾器單元進行測試桿(stick)結合,然後讀取測試桿以確認結果。此處使用的樣品是批號Nos.GC1113-PUR-0901-PR、GC1113-PUR-0902-PR、747R0001和747R0002。檢測結果總結在下表3。從測試結果可以看出,宿主細胞衍生的DNA雜質的含量為0.5ng/mg或更低,最大為0.215ng/mg,平均為0.143ng/mg。 In order to detect the content of host cell-derived DNA impurities in the final crude solution of EPO-Fc, the following tests were performed: 1) diluting and using the standard reagent provided in the kit as a standard solution; 2) setting the zero calibration provided in the kit The standard agent is diluted and used as a test solution; 3) the test solution is prepared by adding a standard solution to the test sample; 4) the zero calibration standard used herein is the zero calibration standard provided in the kit; 5) by Add the standard solution Zero calibration standard to prepare the zero calibration standard used herein; 6) prepared standard solution, test solution, added test solution, zero calibration standard and zero calibration standard for DNA labeling; and 7) use threshold The system filter unit performs a test stick combination and then reads the test rod to confirm the result. The samples used here were batch Nos. GC1113-PUR-0901-PR, GC1113-PUR-0902-PR, 747R0001, and 747R0002. The test results are summarized in Table 3 below. As can be seen from the test results, the host cell-derived DNA impurity content was 0.5 ng/mg or less, and the maximum was 0.215 ng/mg, and the average was 0.143 ng/mg.

實施例3:EPO-Fc組成物中的EPO-Fc的糖分布檢測 Example 3: Detection of sugar distribution of EPO-Fc in EPO-Fc composition

<檢測3:EPO-Fc糖基化位置的分析> <Detection 3: Analysis of EPO-Fc glycosylation position>

EPO-Fc是指同型二聚體形式的融合蛋白,其中包括由IgD鉸鏈組合的EPO和Fc區域的兩種單體已經藉由鉸鏈區 中的雙硫鍵組合。EPO序列中有三個N-糖基化位置(N24、N38和N83)和一個O-糖基化位置(S126)。另外,在Fc序列的IgD CH2結構域中存在另外的N-糖基化位置(N261)。推測EPO-Fc可具有總共八個N-糖基化位置和兩個O-糖基化位置。 EPO-Fc refers to a fusion protein in the form of a homodimer, in which two monomers including the EPO and Fc regions combined by the IgD hinge have been passed through the hinge region. The combination of disulfide bonds. There are three N-glycosylation positions (N24, N38 and N83) and one O-glycosylation position (S126) in the EPO sequence. In addition, an additional N-glycosylation position (N261) is present in the IgD CH2 domain of the Fc sequence. It is speculated that EPO-Fc may have a total of eight N-glycosylation sites and two O-glycosylation sites.

實際的糖基化位置已經由接受請求的Protagen公司確認。 The actual glycosylation site has been confirmed by the requesting Protagen.

在處理DTT和碘乙醯胺以由此還原/烷基化蛋白質後,使用PNGase F處理蛋白質以除去N-糖基化位置中的糖。使用胰蛋白酶和GluC/胰蛋白酶處理不含糖的蛋白質和仍然含有糖的其它蛋白質以形成多個肽,隨後使用MALDI-MS比較這些肽的大小,從而確定N-糖基化位置。 After treating DTT and iodoacetamide to thereby reduce/alkylate the protein, the protein is treated with PNGase F to remove sugar from the N-glycosylation site. The sugar-free protein and other proteins still containing sugar were treated with trypsin and GluC/trypsin to form a plurality of peptides, and then the size of these peptides was compared using MALDI-MS to determine the N-glycosylation position.

為了確認O-糖基化位置,在標記HexNAc後,將其用PNGas F處理以除去N-糖基化位置中的所有糖。藉由使用Arg C、胰蛋白酶/Glu C和Asp N/Tripsin處理,獲得多個肽,然後使用MALDI-MS比較這些肽的大小,從而確定O-糖基化位置。 To confirm the O-glycosylation position, after labeling HexNAc, it was treated with PNGas F to remove all sugars in the N-glycosylation position. A plurality of peptides were obtained by treatment with Arg C, trypsin/Glu C and Asp N/Tripsin, and then the size of these peptides was compared using MALDI-MS to determine the O-glycosylation position.

分析結果總結在下表4。鑑定了存在於EPO序列中的三個N-糖基化位置(N24、N38和N83)和一個O-糖基化位置(S126),並且在Fc序列的IgD CH2結構域中還發現了另一個N-糖基化位置(N261)。 The results of the analysis are summarized in Table 4 below. Three N-glycosylation positions (N24, N38 and N83) and one O-glycosylation position (S126) present in the EPO sequence were identified, and another was found in the IgD CH2 domain of the Fc sequence. N-glycosylation position (N261).

<檢測4:EPO-Fc單醣的構成組成的分析> <Detection 4: Analysis of the composition of EPO-Fc monosaccharide>

糖蛋白的糖鏈通常由單醣如岩藻糖、N-乙醯基葡糖胺(GluNAc)、N-乙醯基半乳糖胺(GalNAc)、半乳糖、甘露糖、唾液酸等組成。或者,也可包括葡萄糖、木糖、甘露糖-6-磷酸或類似物等等。 The sugar chain of the glycoprotein is usually composed of a monosaccharide such as fucose, N-ethyl glucosamine (GluNAc), N-ethyl galactosamine (GalNAc), galactose, mannose, sialic acid or the like. Alternatively, glucose, xylose, mannose-6-phosphate or the like may be included.

接受請求的Protagen公司分析了單醣的構成組成。用4M鹽酸(最終濃度)將EPO-Fc在100℃水解4小時後,然後經由真空離心將其乾燥,所得材料溶於第三次蒸餾水中且經由液相色譜法(高效陰離子交換色譜法採用脈衝式安培法檢測;HPAEC-PAD)。藉由在相同條件下分析具有已知濃度的標準單醣物質並比較相同和各單醣之間的面積,估計各單醣與糖蛋白莫耳的莫耳比。使用標準物質的各單醣的莫耳比總結在下表5。從測試結果可以看出,在1莫耳EPO-Fc中分別存在約2.2莫耳的岩藻糖,約42.1莫耳的GluNAc,約9.3莫耳的半乳糖,約7.0莫耳的 甘露糖。 Protagen, which accepted the request, analyzed the composition of the monosaccharide. EPO-Fc was hydrolyzed at 4 ° C for 4 hours with 4 M hydrochloric acid (final concentration), then dried by vacuum centrifugation, and the resulting material was dissolved in a third distilled water and subjected to liquid chromatography (high-efficiency anion exchange chromatography using a pulse) Amperometric detection; HPAEC-PAD). The molar ratio of each monosaccharide to glycoprotein moule was estimated by analyzing a standard monosaccharide material having a known concentration under the same conditions and comparing the area between the same and each monosaccharide. The molar ratios of the individual monosaccharides using the standard materials are summarized in Table 5 below. As can be seen from the test results, there were about 2.2 moles of fucose, about 42.1 moles of GluNAc, about 9.3 moles of galactose, about 7.0 moles, respectively, in 1 mole EPO-Fc. Mannose.

實施例4:EPO-Fc組成物中EPO-Fc的唾液酸含量的檢測 Example 4: Detection of sialic acid content of EPO-Fc in EPO-Fc composition

<檢測5:EPO-Fc的等電聚焦測試> <Detection 5: Isoelectric focusing test of EPO-Fc>

為了確定所製備的EPO-Fc的pI值,進行如下之等電聚焦(IEF)凝膠電泳測試:1)將標準溶液和測試溶液各者與樣品緩衝液(2X)混合並製備;2)所製備的樣品使用IEF pH 3-7凝膠進行電泳;3)使用三氯乙酸溶液固定完成電泳後獲得的凝膠;和4)使用混合在一起的包括Brilliant Blue R、甲醇和乙酸的染色溶液將凝膠染色,使用混合在一起的包含甲醇和乙酸的漂白溶液脫色,並乾燥之。此處使用的是批號GC1113-PUR-0902-PR。檢測結果總結在第1圖。 To determine the pI value of the prepared EPO-Fc, the following isoelectric focusing (IEF) gel electrophoresis test was performed: 1) mixing the standard solution and the test solution with the sample buffer (2X) and preparing; 2) The prepared sample was electrophoresed using an IEF pH 3-7 gel; 3) the gel obtained after electrophoresis was fixed using a trichloroacetic acid solution; and 4) the dyeing solution including Brilliant Blue R, methanol and acetic acid mixed together was used. The gel was stained, destained using a mixed bleach solution containing methanol and acetic acid, and dried. The batch number GC1113-PUR-0902-PR is used here. The test results are summarized in Figure 1.

如第1圖的測試結果所示,可以看出所製造的EPO-Fc的主條帶分佈在pI 4.5至6.0的範圍。 As shown by the test results of Fig. 1, it can be seen that the main band of the produced EPO-Fc is distributed in the range of pI 4.5 to 6.0.

<檢測6:唾液酸含量的測試> <Test 6: Test for sialic acid content>

為了檢測所製造的EPO-Fc的唾液酸含量,進行以下測試:1)於蒸餾水中藉由稀釋N-乙醯基神經胺酸製備標準溶液;2)使用蒸餾水稀釋製備測試溶液於稀釋狀態;3)使用間苯二酚試劑處理標準溶液和測試溶液後加熱;4)使用萃取溶液(1-丁醇,乙酸丁酯)回收反應產物;和5)在580nm測量回收的反應產物的吸光度以估計測試溶液的唾液酸含量,然後計算每1莫耳EPO-Fc的唾液酸含量(mol/mol)。此處使用的樣品是批號Nos.GC1113-PUR-0901-PR、GC1113-PUR-0902-PR、747R0001和747R0002。檢測結果總結在下表6。從測試結果可以看出,所製造的EPO-Fc具有唾液酸含量為20mol/mol或更多。 In order to detect the sialic acid content of the produced EPO-Fc, the following tests were carried out: 1) preparing a standard solution by diluting N-acetyl thioglycolic acid in distilled water; 2) preparing a test solution in a diluted state by diluting with distilled water; Reheating using a resorcinol reagent to treat the standard solution and the test solution; 4) recovering the reaction product using an extraction solution (1-butanol, butyl acetate); and 5) measuring the absorbance of the recovered reaction product at 580 nm to estimate the test The sialic acid content of the solution was then calculated as the sialic acid content (mol/mol) per 1 mol of EPO-Fc. The samples used here were batch Nos. GC1113-PUR-0901-PR, GC1113-PUR-0902-PR, 747R0001, and 747R0002. The test results are summarized in Table 6 below. As can be seen from the test results, the produced EPO-Fc had a sialic acid content of 20 mol/mol or more.

Claims (15)

一種EPO-Fc組成物,其包含EPO-Fc,該EPO-Fc具有唾液酸含量為17mol/mol或者更多,以及100ng/mg或者更少的宿主細胞衍生的蛋白質雜質。 An EPO-Fc composition comprising EPO-Fc having a sialic acid content of 17 mol/mol or more, and 100 ng/mg or less of host cell-derived protein impurities. 如申請專利範圍第1項所述的EPO-Fc組成物,其中,該唾液酸含量範圍為20至28mol/mol。 The EPO-Fc composition according to claim 1, wherein the sialic acid content ranges from 20 to 28 mol/mol. 如申請專利範圍第1項所述的EPO-Fc組成物,其中,該EPO-Fc具有pI6.0。 The EPO-Fc composition of claim 1, wherein the EPO-Fc has a pI 6.0. 如申請專利範圍第1項所述的EPO-Fc組成物,其中,該EPO-Fc具有4.5pI5.3。 The EPO-Fc composition according to claim 1, wherein the EPO-Fc has 4.5 pI 5.3. 如申請專利範圍第1項所述的EPO-Fc組成物,其中,該宿主細胞衍生的蛋白質雜質是EPO-Fc的凝集物或片段。 The EPO-Fc composition of claim 1, wherein the host cell-derived protein impurity is an agglomerate or fragment of EPO-Fc. 如申請專利範圍第1項所述的EPO-Fc組成物,其中,該宿主細胞衍生的蛋白質雜質的量為含有60ng/mg或者更少。 The EPO-Fc composition according to claim 1, wherein the host cell-derived protein impurity is contained in an amount of 60 ng/mg or less. 如申請專利範圍第1項所述的EPO-Fc組成物,其還包含0.5ng/mg或者更少的宿主細胞衍生的DNA雜質。 The EPO-Fc composition of claim 1, which further comprises 0.5 ng/mg or less of host cell-derived DNA impurities. 一種製備EPO-Fc組成物的方法,包括:在溫度範圍35℃至39℃和6.5pH7.5的條件下,以4vvd或更低的流動率流動EPO-Fc細胞培養溶液以製備灌注培養溶液;從該灌注培養溶液獲得EPO-Fc純溶液;和將該EPO-Fc純溶液吸附到陰離子交換樹脂以製備 Q流出物,該Q流出物包括具有唾液酸含量為17mol/mol或者更多的EPO-Fc。 A method of preparing an EPO-Fc composition comprising: at a temperature in the range of 35 ° C to 39 ° C and 6.5 pH The EPO-Fc cell culture solution is flowed at a flow rate of 4 vvd or lower to prepare a perfusion culture solution under conditions of 7.5; an EPO-Fc pure solution is obtained from the perfusion culture solution; and the EPO-Fc pure solution is adsorbed to the anion exchange Resin to prepare a Q effluent comprising EPO-Fc having a sialic acid content of 17 mol/mol or more. 如申請專利範圍第8項所述的方法,其中該Q流出物的製備包括:將該EPO-Fc吸附到該陰離子交換樹脂;並使用含有0.005至0.02M磷酸鈉、0.05至0.2M L-精胺酸和0.02至0.1M氯化鈉,並且具有6.7pH7.1的緩衝液洗脫所吸附的EPO-Fc。 The method of claim 8, wherein the preparation of the Q effluent comprises: adsorbing the EPO-Fc to the anion exchange resin; and using 0.005 to 0.02 M sodium phosphate, 0.05 to 0.2 M L-fine Amino acid and 0.02 to 0.1 M sodium chloride with 6.7 pH The adsorbed EPO-Fc was eluted with a buffer of 7.1. 如申請專利範圍第8項所述的方法,其中獲得EPO-Fc純溶液的步驟包括使該灌注培養溶液通過POD濾器。 The method of claim 8, wherein the step of obtaining an EPO-Fc pure solution comprises passing the perfusion culture solution through a POD filter. 如申請專利範圍第8項所述的方法,其中,該EPO-Fc組成物中的宿主細胞衍生蛋白質(HCP)雜質的量為100ng/mg或者更少。 The method of claim 8, wherein the amount of host cell-derived protein (HCP) impurities in the EPO-Fc composition is 100 ng/mg or less. 如申請專利範圍第8項所述的方法,其中,將該EPO-Fc細胞培養溶液以2.0×105細胞/mL或者更多的量接種到培養箱。 The method of claim 8, wherein the EPO-Fc cell culture solution is inoculated to the incubator in an amount of 2.0 × 10 5 cells/mL or more. 如申請專利範圍第10項所述的方法,還包括,使該灌注培養溶液通過該POD濾器之後,使該上述溶液通過經填充有蛋白A鍵合樹脂的管柱。 The method of claim 10, further comprising, after passing the perfusion culture solution through the POD filter, passing the solution through a column packed with a protein A bonding resin. 如申請專利範圍第8項所述的方法,其中,該Q流出液中所含的EPO-Fc具有pI為6.0或者更低。 The method of claim 8, wherein the EPO-Fc contained in the Q effluent has a pI of 6.0 or less. 如申請專利範圍第8項所述的方法,還包括分別使用POD濾器、超濾膜和奈米濾器過濾該Q流出物。 The method of claim 8, further comprising filtering the Q effluent using a POD filter, an ultrafiltration membrane, and a nanofilter, respectively.
TW105132540A 2015-10-07 2016-10-07 Composition comprising long-acting erythropoietin TWI654991B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
??10-2015-0140753 2015-10-07
KR1020150140753A KR101847169B1 (en) 2015-10-07 2015-10-07 Composition comprising long-acting Erythropoietin

Publications (2)

Publication Number Publication Date
TW201731523A true TW201731523A (en) 2017-09-16
TWI654991B TWI654991B (en) 2019-04-01

Family

ID=58488013

Family Applications (1)

Application Number Title Priority Date Filing Date
TW105132540A TWI654991B (en) 2015-10-07 2016-10-07 Composition comprising long-acting erythropoietin

Country Status (5)

Country Link
KR (1) KR101847169B1 (en)
CN (1) CN108137665A (en)
AU (1) AU2016334954B2 (en)
TW (1) TWI654991B (en)
WO (1) WO2017061780A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3145382A1 (en) * 2019-07-08 2021-01-14 Gi Innovation, Inc. Polypeptide dimer with high sialic acid content, comprising extracellular domain of alpha subunit of ige fc receptor, and pharmaceutical composition comprising same
CU24704B1 (en) * 2019-09-05 2024-04-08 Ct Inmunologia Molecular METHOD FOR OBTAINING HYPOSIALYLATED RECOMBINANT HUMAN ERYTHROPOIETIN FOR THE TREATMENT OF ALTERATIONS OF THE NERVOUS SYSTEM

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0417916A (en) * 2003-12-31 2007-04-10 Merck Patent Gmbh fc-erythropoietin fusion protein with improved pharmacokinetics
US7625564B2 (en) * 2006-01-27 2009-12-01 Novagen Holding Corporation Recombinant human EPO-Fc fusion proteins with prolonged half-life and enhanced erythropoietic activity in vivo
PL1961821T3 (en) * 2007-02-22 2010-01-29 Polymun Scient Immunbiologische Forschung Gmbh Erythropoietin fusion protein
TWI554520B (en) * 2007-07-20 2016-10-21 諾華健控股公司 Recombinant human epo-fc fusion proteins with prolonged half-life and enhanced erythropoietic activity in vivo
DE102008002209A1 (en) * 2008-06-04 2009-12-10 Evonik Degussa Gmbh Process for the purification of erythropoietin
WO2013002330A1 (en) * 2011-06-29 2013-01-03 協和発酵キリン株式会社 Method for purifying protein
CN103820514B (en) * 2014-03-10 2017-10-31 深圳赛保尔生物药业有限公司 A kind of method of efficient production hematopoietin

Also Published As

Publication number Publication date
CN108137665A (en) 2018-06-08
TWI654991B (en) 2019-04-01
WO2017061780A1 (en) 2017-04-13
KR101847169B1 (en) 2018-04-09
AU2016334954A1 (en) 2018-05-24
KR20170041384A (en) 2017-04-17
AU2016334954B2 (en) 2020-02-27

Similar Documents

Publication Publication Date Title
EP2412817B2 (en) Method for removing viruses in high-concentration monoclonal antibody solution
CN106536565A (en) Process for the purification of TNFR:Fc fusion protein
CN107988192A (en) For purify restructuring ADAMTS13 and other oroteins method with and combinations thereof
CN108524929B (en) A kind of production method of rabies vacciness
JP2014210796A (en) Purification of erythropoietin
EP2943582A1 (en) Methods for improved production and recovery of recombinant proteins from eukaryotic cell cultures
JP2017526649A (en) New process for purification of rHu-GCSF
CN101268181A (en) Protein A production and purification without using animal derived components
EP2141178A1 (en) Method of producing antithrombin composition
TW201731523A (en) Composition comprising long-acting erythropoietin
JP2009502117A (en) Recombinant method for erythropoiesis stimulating protein production
CN109196099A (en) Method for producing the active Hepatocyte Growth factor (HGF)
KR20210116467A (en) How to transfer a batch production process to a continuous production process
CN109776675A (en) The method of two-step solution chromatography preparation dog immunoglobulin
KR20180026688A (en) Composition comprising long-acting Erythropoietin
US8461302B2 (en) Purification of recombinant human factor XIII
WO2018222723A1 (en) C3 fusion protein and methods of making and using thereof
CN117126832B (en) Recombinant human blood coagulation factor IX fusion protein and preparation method thereof
BR112012004054B1 (en) METHOD FOR PRODUCING AN ANTI-HER2 ANTIBODY
JP7396997B2 (en) Complete flow-through method for purifying recombinant proteins
EP2565269B1 (en) Method for producing high-purity soluble thrombomodulin
KR20220110555A (en) VEGF mini-traps and methods of use thereof
CN115215940A (en) Production method of TNFR-Ig fusion protein
EA046459B1 (en) METHODS FOR OBTAINING USTEKINUMAB
CN112980796A (en) Liquid medium for reducing oxidation of recombinant protein expressed in CHO cells