TW201305341A - Production of low fucose antibodies in H4-II-E rat cells - Google Patents

Production of low fucose antibodies in H4-II-E rat cells Download PDF

Info

Publication number
TW201305341A
TW201305341A TW100134655A TW100134655A TW201305341A TW 201305341 A TW201305341 A TW 201305341A TW 100134655 A TW100134655 A TW 100134655A TW 100134655 A TW100134655 A TW 100134655A TW 201305341 A TW201305341 A TW 201305341A
Authority
TW
Taiwan
Prior art keywords
cells
cell
antibody
cell line
rat hepatoma
Prior art date
Application number
TW100134655A
Other languages
Chinese (zh)
Inventor
Kristina Ellwanger
Lore Florin
Hitto Kaufmann
Original Assignee
Boehringer Ingelheim Int
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 Boehringer Ingelheim Int filed Critical Boehringer Ingelheim Int
Publication of TW201305341A publication Critical patent/TW201305341A/en

Links

Abstract

The invention concerns the field of cell culture technology. It specifically concerns a rat hepatoma cell, preferably a H4-II-E rat hepatoma cell, carrying a DNA encoding an antibody or Fc-fusion protein and having low fucosylation activity for adding fucose to glycosidic structures such as biantennary glycans, e.g. N-acetylglucosamine. The invention furthermore concerns a method for producing low fucose glycoproteins especially antibodies or Fc-fusion proteins in rat hepatoma cells, preferably in H4-II-E rat hepatoma cells. It further concerns the identification and generation of new host cell lines which are capable of synthetizing glycoproteins with beneficial properties, improving the therapeutic efficacy and/or serum half-life of the product compared to products from commonly used host cell lines.

Description

H4-II-E大鼠細胞中低海藻糖抗體之生產Production of low trehalose antibodies in H4-II-E rat cells

本發明係關於細胞培養技術領域。其係關於在H4-II-E大鼠肝瘤細胞中產生低海藻糖糖蛋白、尤其抗體及Fc-融合蛋白之方法。其係關於新宿主細胞系之鑑別及生成,該等細胞系能夠合成具有有益性質之糖蛋白,增進產物相對於來自常用宿主細胞系之產物之治療效果及/或血清半衰期。本發明係關於此等細胞系、尤其大鼠肝瘤細胞系H4-II-E用於表現重組蛋白之用途及優化及其作為高活性生物醫藥治療劑之應用。The present invention relates to the field of cell culture technology. It relates to a method for producing low trehalose glycoproteins, particularly antibodies and Fc-fusion proteins, in H4-II-E rat hepatoma cells. It relates to the identification and production of new host cell lines capable of synthesizing glycoproteins having beneficial properties, enhancing the therapeutic effect and/or serum half-life of the product relative to products from commonly used host cell lines. The present invention relates to the use and optimization of such cell lines, particularly the rat hepatoma cell line H4-II-E, for the expression of recombinant proteins and their use as therapeutic agents for highly active biopharmaceuticals.

供人類療法使用之生物醫藥市場持續高速增長,其中在臨床研究中已評估270種新生物醫藥且估計2003年的銷售額為300億(Werner,2004)。可自不同宿主細胞系統(包括細菌細胞、酵母細胞、昆蟲細胞、植物細胞及哺乳動物細胞以及源自人類之細胞系)產生生物醫藥。當前,由於真核細胞能夠正確加工並轉譯後修飾重組人類蛋白,因此越來越來的生物醫藥係其產生。因此,影響對用於製造過程之細胞系進行選擇的關鍵問題係以均一轉譯後修飾模式一致地產生產物之能力(產生高生物活性、穩定性及批次間一致性)。The biopharmaceutical market for human therapy continues to grow at a rapid rate, with 270 new biopharmaceuticals evaluated in clinical studies and estimated sales of 30 billion in 2003 (Werner, 2004). Biopharmaceuticals can be produced from different host cell systems, including bacterial cells, yeast cells, insect cells, plant cells, and mammalian cells, as well as cell lines derived from humans. Currently, as eukaryotic cells are able to properly process and translate and recombine recombinant human proteins, more and more biopharmaceuticals are produced. Thus, a key issue affecting the selection of cell lines for use in the manufacturing process is the ability to consistently produce products with uniform post-translational modification patterns (resulting in high biological activity, stability, and batch-to-batch consistency).

當前經許可用於治療應用之最大部分抗體係在中國倉鼠卵巢(CHO)細胞系中製造的。其他生產系統係鼠類淋巴樣細胞(包括NS0及Sp2/0-Ag 14)。該等親本細胞系亦係在臨床試驗中最常用於產生抗體者。另外,使用鼠類及諸如人類細胞系PER.C6等其他細胞系。The largest part of the anti-system currently licensed for therapeutic applications is manufactured in the Chinese hamster ovary (CHO) cell line. Other production systems are murine lymphoid cells (including NS0 and Sp2/0-Ag 14). These parental cell lines are also the most commonly used in antibody production in clinical trials. In addition, murines and other cell lines such as the human cell line PER.C6 are used.

單株抗體之療法已成為生物技術產業的主要焦點之一。儘管到目前為止已存在經食品及藥物管理局(Food and Drug Administration)批准之有效單株抗體,但仍需要甚至更有效且更相容之藥物來不僅降低製造成本,而且有助於應用於更多患者。此外,可減少優化治療劑之應用劑量,由此產生較高耐受及較低不良效應。The treatment of monoclonal antibodies has become one of the main focuses of the biotechnology industry. Although effective monoclonal antibodies approved by the Food and Drug Administration have been available to date, there is still a need for even more effective and compatible drugs to not only reduce manufacturing costs, but also help to apply more Many patients. In addition, the dosage of the optimized therapeutic agent can be reduced, thereby resulting in higher tolerance and lower adverse effects.

在5種哺乳動物抗體類別IgA、IgD、IgE、IgG及IgM中,IgG類別主要用於治療、預防及診斷各種疾病。此歸因於其有利功能特性(例如在血液中之長半衰期)及各種效應子功能(例如抗體依賴性細胞介導之細胞毒性(ADCC)及補體依賴性細胞毒性(CDC))。在人類IgG類別中,亞類(同種型)IgG1及IgG3具有最高ADCC及CDC活性,但與半衰期長達21天之IgG1相比,IgG3僅為7至8天。鑒於上文,若出於最佳治療功效需要高活性效應子功能來去除在表面上帶有抗原之細胞,則主要使用人類IgG1子類抗體。Among the five mammalian antibody classes IgA, IgD, IgE, IgG and IgM, the IgG class is mainly used for the treatment, prevention and diagnosis of various diseases. This is due to its advantageous functional properties (eg long half-life in blood) and various effector functions (eg antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC)). Among the human IgG classes, subclass (isotype) IgG1 and IgG3 have the highest ADCC and CDC activities, but IgG3 is only 7 to 8 days compared to IgG1 with a half-life of up to 21 days. In view of the above, human IgG1 subclass antibodies are predominantly used if high activity effector functions are required for optimal therapeutic efficacy to remove cells bearing antigen on the surface.

轉譯後修飾不僅對於正確的蛋白質摺疊、細胞內轉運、溶解性及穩定性至關重要,而且對於分泌蛋白的生物活性及免疫原性具有重要的功能影響。在生物醫藥蛋白(例如治療抗體)中,轉譯後修飾對產品之治療效能、藥物代謝動力學、藥效動力學及免疫原性具有特定影響。Post-translational modifications are important not only for proper protein folding, intracellular transport, solubility and stability, but also for the biological activity and immunogenicity of secreted proteins. In biopharmaceutical proteins (eg, therapeutic antibodies), post-translational modifications have a specific effect on the therapeutic efficacy, pharmacokinetics, pharmacodynamics, and immunogenicity of the product.

糖基化代表在天然分泌蛋白以及經批准生物醫藥中發現的最廣泛之轉譯後修飾。幾乎50%人類蛋白經糖基化。天冬醯胺酸(N)-及絲胺酸(O)-連接聚糖係在源自哺乳動物細胞之分泌糖蛋白上形成的兩種主要聚糖類別。糖結構至分泌蛋白之轉移在內質網(ER)及高爾基器中發生且代表由眾多基因之活性調節的複雜酶促過程。糖基化途徑所涉及許多基因之缺陷會引起具有嚴重醫學後果之先天性病症,此證實正常糖基化之重要性。Glycosylation represents the most extensive post-translational modification found in natural secreted proteins as well as in approved biopharmaceuticals. Almost 50% of human proteins are glycosylated. Aspartate (N)- and serine (O)-linked glycans are the two major glycocatelines formed on secreted glycoproteins derived from mammalian cells. The transfer of glycostructure to secreted protein occurs in the endoplasmic reticulum (ER) and Golgi apparatus and represents a complex enzymatic process that is regulated by the activity of numerous genes. Defects in many genes involved in the glycosylation pathway cause congenital conditions with severe medical consequences, which confirms the importance of normal glycosylation.

在真核生物中,N-連接聚糖以預合成寡糖形式附著至ER腔中之蛋白上,該等預合成寡糖由具分支寡糖單元(由3個葡萄糖(Glc)、9個甘露糖(Man)及2個N-乙醯葡糖胺(GlcNAc)構成)組成。該等核心聚糖(Glc3Man9GlcNAc2)經由脂質載體(ER膜中之多萜醇-焦磷酸鹽)轉移至移位至ER腔中之分泌蛋白上。將聚糖轉移至適當序列(即Asn-X-Ser/Thr)上,其中X係初生糖蛋白中除脯胺酸外之任一胺基酸。正確摺疊糖蛋白主動轉運至高爾基器中,其中其N-聚糖由葡糖苷酶、甘露糖苷酶及糖基轉移酶修飾以產生含有唾液酸、海藻糖及半乳糖之複合結構。葡糖苷酶及甘露糖苷酶在N-聚糖加工之最早階段自聚糖去除葡萄糖(Glc)及甘露糖單糖(Man)。然後,N-乙醯基葡糖胺酶催化將N-乙醯葡糖胺(GlcNAc)添加至附著至N-聚糖之保守核心結構上之甘露糖上,此對聚糖上形成之分支或枝結構之數量具有決定作用。海藻糖基轉移酶將海藻糖添加至靠近蛋白之N-乙醯葡糖胺上且半乳糖基轉移酶及唾液酸轉移酶分別將半乳糖及唾液酸添加至N-聚糖分支之末端上。該等酶之反應通常係不可逆的,從而生成穩定的N-糖基化蛋白。In eukaryotes, N-linked glycans are attached to proteins in the ER lumen in the form of pre-synthesized oligosaccharides consisting of branched oligosaccharide units (by 3 glucose (Glc), 9 nectars) The composition of sugar (Man) and two N-acetylglucosamine (GlcNAc). These core glycans (Glc 3 Man 9 GlcNAc 2 ) are transferred via a lipid carrier (polysterol-pyrophosphate in the ER membrane) to secreted proteins that are translocated into the ER lumen. The glycan is transferred to the appropriate sequence (i.e., Asn-X-Ser/Thr), wherein any of the amino acids other than proline in the X-linked nascent glycoprotein. The correctly folded glycoprotein is actively transported into the Golgi apparatus, wherein its N-glycan is modified by glucosidase, mannosidase and glycosyltransferase to produce a complex structure containing sialic acid, trehalose and galactose. Glucosidase and mannosidase remove glucose (Glc) and mannose monosaccharide (Man) from glycans at the earliest stages of N-glycan processing. N-Ethyl Glucosaminease then catalyzes the addition of N-acetylglucosamine (GlcNAc) to the mannose attached to the conserved core structure of the N-glycan, which forms a branch on the glycan or The number of branch structures has a decisive role. The trehalose transferase adds trehalose to the N-acetylglucosamine adjacent to the protein and the galactosyltransferase and sialyltransferase add galactose and sialic acid to the ends of the N-glycan branch, respectively. The reaction of these enzymes is generally irreversible, resulting in a stable N-glycosylated protein.

經由生成核心寡糖並對其實施差式修飾且可變添加外臂糖,蛋白糖基化可引入顯著異質性。Protein glycosylation can introduce significant heterogeneity by generating core oligosaccharides and performing differential modifications thereto and variable addition of exo-arm sugars.

已證明不正確糖基化或非糖基化抗體展示不受控制之功能。因此,選擇能夠一致地生成具有所期望轉譯後修飾模式之產物的適當生產系統對於成功的藥物製造及應用而言至關重要。糖蛋白之糖型特徵及因此功能活性可端視生產系統而顯著不同。原核生產系統(例如大腸桿菌(Escherichia coli))中之生物醫藥蛋白產生可獲得非糖基化蛋白產物,其在活體外必須以經溶解並再摺疊之包涵體形式回收。相比之下,酵母表現系統添加高甘露糖含量之糖側鏈,在昆蟲細胞中產生後獲得之糖基化模式亦與特徵性哺乳動物模式顯著不同。植物可將蛋白差式糖基化,但一致地添加經報導在人類中具有免疫原性或過敏性之α1,3-海藻糖及β1,3-木糖。Incorrect glycosylated or non-glycosylated antibodies have been shown to exhibit uncontrolled functions. Therefore, the selection of a suitable production system capable of consistently generating a product with the desired post-translational modification pattern is critical to successful drug manufacturing and application. The glycoform characteristics of glycoproteins and thus their functional activity can vary significantly depending on the production system. Biopharmaceutical proteins in prokaryotic production systems (e.g., Escherichia coli ) produce non-glycosylated protein products that must be recovered in vitro as dissolved and refolded inclusion bodies. In contrast, the yeast expression system adds a high mannose content sugar side chain, and the glycosylation pattern obtained after production in insect cells is also significantly different from the characteristic mammalian pattern. Plants can differentially glycosylate proteins, but consistently add alpha 1,3-trehalose and beta 1,3-xylose, which are reported to be immunogenic or allergenic in humans.

不同哺乳動物宿主細胞能夠進行差式N-聚糖加工。對所產生糖蛋白之分析顯示異質糖型,此端視產生細胞所源自之組織或物種而定。因此,重要的是,確保產生用於臨床應用之糖蛋白產物的糖基化模式在整個產生批次中及之間係均一的,且亦確保保持抗體之有利活體內性質。Different mammalian host cells are capable of differential N-glycan processing. Analysis of the glycoprotein produced shows a heterologous glycoform depending on the tissue or species from which the cell is derived. Therefore, it is important to ensure that the glycosylation pattern that produces the glycoprotein product for clinical use is uniform throughout and throughout the production batch, and also ensures that the beneficial in vivo properties of the antibody are maintained.

機體內之天然(固有)抗體以及由重組DNA技術在哺乳動物宿主細胞系中產生之抗體二者皆經由IgG-Fc區內CH2結構域中在進化上保守之Asn297處共價附著寡糖來糖基化。寡糖係IgG-Fc結構之組成部分且係效應子功能所絕對需要的。IgG-Fc上用於效應子配體(例如FcγRI、FcγRII、FcγRIII及C1q)之相互作用位點由蛋白質部分構成;然而,基本IgG-Fc蛋白構象之生成取決於寡糖之存在及化學組成。因此,經由效應子配體之接合介導之效應子機制(清除機制,例如吞噬作用、抗體依賴性細胞毒性(ADCC)及補體依賴性細胞毒性(CDC))會因不正確或非糖基化IgG而嚴重受損或去除。Both natural (inherent) antibodies in the body and antibodies produced by recombinant DNA techniques in mammalian host cell lines are covalently attached to oligosaccharides via the evolutionarily conserved Asn297 in the CH2 domain of the IgG-Fc region. Basic. Oligosaccharides are part of the IgG-Fc structure and are absolutely required for effector function. The interaction sites on IgG-Fc for effector ligands (eg, FcyRI, FcyRII, FcyRIII, and C1q) are composed of protein moieties; however, the formation of a basic IgG-Fc protein conformation depends on the presence and chemical composition of the oligosaccharides. Thus, the effector mechanisms mediated through the junction of effector ligands (cleavage mechanisms such as phagocytosis, antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC)) may be incorrect or non-glycosylated. IgG is severely damaged or removed.

人類FcγRIIIa受體具有多態性且已顯示FcγRIIIa-158V(纈胺酸)形式與IgG1-Fc之親和力高於FcγRIIIa-158F(苯丙胺酸)形式。經活體外證實,IgG1抗體經由帶有純合FcγRIIIa-158V之細胞比純合FcγRIIIa-158F或雜合FcγRIIIa-158V/FcγRIIIa-158F細胞更有效地介導ADCC。The human FcγRIIIa receptor is polymorphic and has been shown to have an affinity for the FcγRIIIa-158V (proline) form to be higher than the FcγRIIIa-158F (phenylalanine) form. It was confirmed in vitro that the IgG1 antibody mediates ADCC more efficiently via homozygous FcγRIIIa-158F or hybrid FcγRIIIa-158V/FcγRIIIa-158F cells via cells bearing homozygous FcγRIIIa-158V.

正常多株人類IgG-Fc之典型寡糖結構係複合二枝結構類型。當糖蛋白經過在核心(海藻糖、N-乙醯葡糖胺(GlcNAc))(或外臂(半乳糖(Gal)及N-乙醯基神經胺酸(Neu5Ac))處具有額外糖殘基之高爾基器時,二枝結構核心七糖受到可變修飾(由(Walsh及Jefferis,2006)綜述)。A typical oligosaccharide structure of a normal multi-plant human IgG-Fc is a complex bi-tower structure type. When the glycoprotein has extra sugar residues at the core (trehalose, N-acetylglucosamine (GlcNAc)) (or the outer arm (galactose (Gal) and N-acetylthione (Neu5Ac)) In the Golgi apparatus, the core heptasaccharide of the two branches is subject to variable modification (reviewed by Walsh and Jefferis, 2006).

來自不同脊椎動物物種之血清IgG抗體共同具有基本二枝結構Fc糖結構,但糖鏈之周圍區域之結構及組成有所不同(Hamako等人,1993)。可變性最可能歸因於存在於不同物種之B-淋巴球中之糖基轉移酶的不同活性及/或每一IgG對於該等轉移酶之可及性。若使用重組DNA技術來在不天然分泌IgG之宿主細胞中穩定表現IgG,則糖基化模式同樣由存在於該細胞系中之糖基轉移酶或聚糖修飾酶之活性規定。因此,在替代生產細胞系中產生之重組蛋白之糖基化模式具有細胞型-、組織-及物種特異性且可顯著不同(Raju等人,2000)。在CHO、Y0骨髓瘤及NS0細胞中產生相同IgG1抗體可產生三種不同產物,其在糖基化模式及生物活性上不同(Lifely等人,1995)。因此,已充分確定,效應子功能之活化強烈地取決於抗體分子之寡糖組成且若存在某些糖基化模式,則某些效應子功能之活化將更為有效。Serum IgG antibodies from different vertebrate species share a basic two-branched Fc sugar structure, but the structure and composition of the surrounding regions of the sugar chain are different (Hamako et al., 1993). The variability is most likely due to the different activities of glycosyltransferases present in B-lymphocytes of different species and/or the accessibility of each IgG to such transferases. If recombinant DNA technology is used to stably express IgG in host cells that do not naturally secrete IgG, the glycosylation pattern is also defined by the activity of the glycosyltransferase or glycan modifying enzyme present in the cell line. Thus, the glycosylation pattern of recombinant proteins produced in alternative production cell lines is cell type-, tissue- and species-specific and can be significantly different (Raju et al., 2000). Production of the same IgGl antibody in CHO, Y0 myeloma and NSO cells produces three different products that differ in glycosylation patterns and biological activities (Lifely et al, 1995). Thus, it has been well established that activation of effector functions is strongly dependent on the oligosaccharide composition of the antibody molecule and that activation of certain effector functions will be more effective if certain glycosylation patterns are present.

採用具有修飾糖基化模式之治療抗體之研究表明,缺乏α1,6-連接核心海藻糖可提高IgG1抗體與FcγRIIIa受體的結合親和力,且因此由天然殺傷(NK)細胞介導之ADCC效果提高高達100倍。Studies using therapeutic antibodies with modified glycosylation patterns have shown that the lack of α1,6-linked core trehalose increases the binding affinity of IgG1 antibodies to the FcγRIIIa receptor, and thus the ADCC effect mediated by natural killer (NK) cells Up to 100 times.

由於補體系統組份與半乳糖基化Fc-聚糖之高結合親和力,因此Fc聚糖之半乳糖基化經提高之抗體可活化補體系統且補體依賴性細胞毒性(CDC)比具有低或非半乳糖基化糖型之抗體更有效。Due to the high binding affinity of the complement system component to galactosylated Fc-glycans, the galactosylation of Fc glycans increases the activation of the complement system and the complement-dependent cytotoxicity (CDC) ratio is low or non- Antibodies to galactosylated glycoforms are more effective.

治療抗體可具有不同的作用方法。一些抗體、抗體片段或Fc-融合蛋白經設計以在活體內中和生物分子(例如細胞因子)。相比之下,癌症療法中之重組抗體經常識別腫瘤細胞上之蛋白且其功效明確地依賴於敏化效應子細胞以供隨後藉由抗體依賴性細胞毒性(ADCC)及/或補體依賴性細胞毒性(CDC)機制實施殺傷。然而,對於其他基於抗體之療法而言,活化發炎性級聯可能有害,其會產生不期望之副作用。Therapeutic antibodies can have different methods of action. Some antibodies, antibody fragments or Fc-fusion proteins are designed to neutralize biomolecules (eg, cytokines) in vivo. In contrast, recombinant antibodies in cancer therapy often recognize proteins on tumor cells and their efficacy is clearly dependent on sensitized effector cells for subsequent antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cells. The toxicity (CDC) mechanism implements killing. However, for other antibody-based therapies, activation of the inflammatory cascade may be detrimental, which can cause undesirable side effects.

因此,常用生產細胞系(例如CHO、NS0及SP2/0)中產生之抗體不顯示最佳Fc糖基化模式係一種挑戰。Therefore, antibodies produced in commonly used cell lines (eg, CHO, NS0, and SP2/0) do not exhibit the best Fc glycosylation pattern as a challenge.

CHO、NS0或人類細胞系中產生之抗體顯示主要(約95%)由二枝結構核心七糖結構構成之Fc糖基化特徵,該二枝結構核心七糖結構在中心GlcNAc處帶有α1,6連接海藻糖殘基。Antibodies produced in CHO, NS0 or human cell lines show predominantly (approximately 95%) Fc glycosylation features consisting of a dimeric structural core heptasaccharide structure with alpha 1 at the central GlcNAc, 6 connect the trehalose residue.

已充分確定,在Fc聚糖中之二枝結構碳水化合物核心結構內存在α1,6-連接海藻糖會顯著損害抗體活化效應子功能(例如抗體依賴性細胞毒性(ADCC))之潛力。已證實另一效應子功能(補體誘導之細胞毒性(CDC))具有半乳糖基化依賴性,其中較高半乳糖含量可使CDC活性增強。因此,具有海藻糖基化聚糖及低度半乳糖之單株抗體在治療實體或非實體腫瘤上尤其有效,此乃因抗體療法之治療效果在此情形下主要取決於抗體在募集並活化腫瘤攻擊免疫細胞、細胞凋亡誘導或ADCC或CDC活化上之效能。因此,CHO中產生之抗體在癌症療法上保持有限益處,且為了克服其低活性,必須以高劑量投與患者。相比之下,具有經改變糖基化模式(例如,缺乏α1,6-連接核心海藻糖)之抗體應具有顯著增強之治療效果。It has been well established that the presence of alpha 1,6-linked trehalose in the dimeric structural carbohydrate core structure in Fc glycans significantly impairs the potential of antibody activation effector functions, such as antibody-dependent cellular cytotoxicity (ADCC). Another effector function (complement-induced cytotoxicity (CDC)) has been shown to be galactosylation dependent, with higher galactose content enhancing CDC activity. Therefore, a monoclonal antibody having a fucosylated glycan and a low galactose is particularly effective in treating a solid or non-solid tumor, because the therapeutic effect of the antibody therapy in this case mainly depends on the antibody recruiting and activating the tumor. The efficacy of attacking immune cells, apoptosis induction or activation of ADCC or CDC. Thus, antibodies produced in CHO maintain a limited benefit in cancer therapy, and in order to overcome their low activity, patients must be administered at high doses. In contrast, antibodies with altered glycosylation patterns (eg, lacking alpha 1,6-linked core trehalose) should have a significantly enhanced therapeutic effect.

儘管存在以下事實:多數經許可治療抗體係在CHO、NS0或Sp2/0細胞中產生,但已知在非最佳條件下,該等細胞可產生許多缺乏效能或潛在免疫原性之異常糖基化產物。除不正確加工寡糖外,亦已知鼠類細胞添加通常不會在人類IgG上發現之糖殘基。此結構(例如Gal-1,3-Gal及N-羥乙醯神經胺酸(NeuGc))可觸發免疫原性或過敏性反應,其對於擬活體內遞送至人類患者之治療劑係不可接受的。Despite the fact that most licensed therapeutic systems are produced in CHO, NS0 or Sp2/0 cells, it is known that under non-optimal conditions, such cells can produce a number of abnormal glycosyl groups that lack potency or potential immunogenicity. Chemical product. In addition to incorrect processing of oligosaccharides, it is also known that murine cells add sugar residues that are not normally found on human IgG. This structure (eg, Gal-1,3-Gal and N-hydroxyethyl cyanokine (NeuGc)) can trigger an immunogenic or allergic response that is unacceptable for therapeutic agents intended for in vivo delivery to human patients. .

迄今為止,獲得能夠產生具有有利糖基化模式之蛋白之細胞系的實驗方法集中於對編碼糖基化機器之酶之基因的遺傳操作,藉此以人工方式改變現有產生宿主細胞系中糖基化途徑之性質(遺傳失活或去調節)。由此方法產生之抗體可顯示某些糖結構或連接之含量降低或完全缺乏。此等經遺傳修飾細胞系可經篩選以獲得能夠合成某些期望糖基化模式之突變體。To date, experimental methods for obtaining cell lines capable of producing proteins with favorable glycosylation patterns have focused on the genetic manipulation of genes encoding enzymes of glycosylation machinery, thereby artificially altering glycosylation in existing host cell lines. The nature of the pathway (genetic inactivation or deregulation). The antibodies produced by this method may show a reduced or complete deficiency in the amount of certain sugar structures or linkages. Such genetically modified cell lines can be screened to obtain mutants capable of synthesizing certain desired glycosylation patterns.

編碼糖基轉移酶GntIII之基因在CHO細胞中之轉基因過表現可使細胞能夠生成具有經改變糖基化模式之抗體產物及具有減少海藻糖基化之最終產物,從而在活化ADCC上比具有未修飾聚糖之抗體強20至100倍(Umana等人,1999)。然而,應考慮,此等經遺傳修飾宿主細胞系需要主動選擇來穩定遺傳改變之效應,此外,遺傳改造可具有不期望之副作用,此會增加工業產生過程之調節負擔。Transgenic overexpression of the gene encoding the glycosyltransferase GntIII in CHO cells allows the cell to produce an antibody product with altered glycosylation patterns and a final product with reduced hyphal glycosylation, thereby having a higher Antibodies that modify glycans are 20 to 100 times stronger (Umana et al., 1999). However, it should be considered that such genetically modified host cell lines require active selection to stabilize the effects of genetic alterations. Furthermore, genetic engineering can have undesirable side effects, which increases the regulatory burden of the industrial production process.

抗凝集素CHO突變細胞系Lec13(Ripka等人,1986)因遺傳缺陷而缺乏將GDP-甘露糖轉化為GDP-海藻糖之酶。若細胞不能代謝來自替代(例如外部)來源之海藻糖,則可使用該等細胞來產生海藻糖基化經降低且ADCC活性經提高之產物(Shields等人,2002)。然而,在用小扁豆凝集素(Lens culinaris agglutinin)(LCA)連續選擇後僅維持Lec13細胞在其基因組中含有之突變。然而,就調節角度而言,在大規模產生過程中添加凝集素或其他選擇物質係非常令人懷疑的。此外,已知Lec13細胞之蛋白產量較低,且海藻糖含量僅部分降低,但高度可變(Kanda等人,2006;Shields等人,2002)。The anti-lectin CHO mutant cell line Lec13 (Ripka et al., 1986) lacks an enzyme that converts GDP-mannose into GDP-trehalose due to genetic defects. If the cells are unable to metabolize trehalose from an alternative (e.g., external) source, the cells can be used to produce products with reduced fucosylation and increased ADCC activity (Shields et al, 2002). However, only the mutations contained in the genome of Lec13 cells were maintained after continuous selection with Lens culinaris agglutinin (LCA). However, in terms of adjustment angles, it is highly doubtful to add lectins or other selected substances during large-scale production. Furthermore, Lec13 cells are known to have lower protein yields and only a partial decrease in trehalose content, but are highly variable (Kanda et al., 2006; Shields et al., 2002).

本發明令人驚奇地證實大鼠肝瘤細胞系H4-II-E係高活性生物醫藥糖蛋白(例如抗體、尤其治療抗體及Fc-融合蛋白)之具有改良糖基化性質的極佳生產細胞系。The present invention surprisingly demonstrates that the rat hepatoma cell line H4-II-E is a highly productive biopharmaceutical glycoprotein (eg, antibodies, particularly therapeutic antibodies and Fc-fusion proteins) with excellent production cells with improved glycosylation properties. system.

本發明另外闡述對所選代表各種物種、組織、細胞類型及分化/腫瘤發生階段之非改造細胞作為宿主細胞系之適用性的分析,該等宿主細胞系用於產生具有有利糖基化模式及因此增進效應子功能之生物醫藥。The invention further sets forth an assay for the suitability of selected non-engineered cells representing various species, tissues, cell types, and differentiation/tumor stages as host cell lines for producing favorable glycosylation patterns and Therefore, the biomedicine that enhances the effector function.

令人驚奇地,大鼠肝瘤細胞系H4-II-E係若干大鼠及/肝源性細胞系中唯一組合眾多用於治療糖蛋白之增進功效之有利糖性質的細胞系。Surprisingly, the rat hepatoma cell line H4-II-E is the only cell line in the rat/hepatocyte-derived cell line that combines a number of advantageous glycan properties for the therapeutic effect of glycoproteins.

本發明首次另外闡述,H4-II-E大鼠肝瘤細胞系可用作產生重組糖蛋白(例如抗體或Fc-融合蛋白)之宿主細胞系。本發明證實,H4-II-E大鼠肝瘤細胞可經編碼抗體或Fc-融合蛋白之輕鏈及重鏈的遺傳元件穩定轉染,且所得產生細胞將高活性功能抗體分子分泌至細胞培養物上清液中,自該上清液可對其實施純化。H4-II-E細胞中產生之重組抗體因細胞糖基化能力而在品質及活性上優於以常規方式產生之治療抗體。The invention is additionally described for the first time that the H4-II-E rat hepatoma cell line can be used as a host cell line for the production of recombinant glycoproteins (e.g., antibodies or Fc-fusion proteins). The present invention demonstrates that H4-II-E rat hepatoma cells can be stably transfected with genetic elements encoding the light chain and heavy chain of an antibody or Fc-fusion protein, and the resulting cells secrete highly active functional antibody molecules to cell culture. In the supernatant, the supernatant can be purified. Recombinant antibodies produced in H4-II-E cells are superior in quality and activity to therapeutic antibodies produced in a conventional manner due to cellular glycosylation ability.

本發明H4-II-E細胞系已於2011年6月28日根據布達佩斯條約(Budapest treaty)以登錄編號DSM ACC3129(H4-II-E)寄存於德國微生物菌種保存中心(Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH,DSMZ),Inhoffenstrasse 7B,D-38124 Braunschweig,Germany。The H4-II-E cell line of the present invention was deposited with the German Collection of Microbial Species (Deutsche Sammlung von Mikroorganismen und) on June 28, 2011 under the Budapest treaty under the accession number DSM ACC3129 (H4-II-E). Zellkulturen GmbH, DSMZ), Inhoffenstrasse 7B, D-38124 Braunschweig, Germany.

H4-II-E細胞中產生之抗體、尤其IgG1抗體之糖模式顯示複合二枝結構聚糖,其主要不含海藻糖且同時半乳糖基化高於通常於CHO中產生之抗體。藉此,H4-II-E細胞中產生之生物治療抗體具有劇烈地且有效地活化抗體介導效應子功能(例如ADCC及CDC)之高潛力。The glycoform pattern of antibodies produced in H4-II-E cells, particularly IgGl antibodies, shows complex dimeric structural glycans that are predominantly free of trehalose and at the same time galactosylation is higher than antibodies normally produced in CHO. Thereby, the biotherapeutic antibodies produced in H4-II-E cells have a high potential to rapidly and efficiently activate antibody-mediated effector functions such as ADCC and CDC.

本發明另外闡述,H4-II-E大鼠肝瘤細胞系可在無血清培養基中懸浮培養,此在該等細胞系用作生物治療劑之生產細胞系時係強制性的。令人驚奇地,在使H4-II-E細胞適應於在無動物組份且無鈣離子之化學限定培養基中懸浮生長後,與含血清培養基中H4-II-E細胞呈貼壁細胞層之常用培養模式相比,生長、活力及加倍時間未受影響。在含血清貼壁培養物中以及在適應於無血清、無動物組份且無鈣離子之培養基後,在適應階段期間,H4-II-E細胞培養物之加倍時間在24小時至32小時之範圍內或在32小時至60小時之範圍內。群加倍時間為24小時至32小時之H4-II-E細胞之懸浮培養的關鍵態樣係使用無鈣培養基。與此相比,在化學限定培養基中無血清培育H4-II-E細胞先前已闡述於文獻中,但群加倍時間為68.5小時(Miyazaki等人,1991)或4天(Niwa等人,1980),此過慢而不能滿足對生物醫藥劑之生產細胞系之需求。The invention further clarifies that the H4-II-E rat hepatoma cell line can be cultured in suspension in serum-free medium, which is mandatory when such cell lines are used as a production cell line for biotherapeutics. Surprisingly, after adapting H4-II-E cells to suspension growth in chemically defined medium without animal components and without calcium ions, adherent cell layers were present in H4-II-E cells in serum-containing medium. Growth, viability, and doubling time were not affected by the usual culture mode. In serum-containing adherent cultures and after adaptation to serum-free, animal-free and calcium-free media, the H4-II-E cell culture doubling time is between 24 hours and 32 hours during the adaptation phase. Within the range or within the range of 32 hours to 60 hours. A key aspect of suspension culture of H4-II-E cells with a population doubling time of 24 hours to 32 hours was the use of calcium-free medium. In contrast, serum-free incubation of H4-II-E cells in chemically defined media has previously been described in the literature, but the population doubling time is 68.5 hours (Miyazaki et al., 1991) or 4 days (Niwa et al., 1980). This is too slow to meet the demand for biopharmaceutical production cell lines.

適應於在闡述於本發明中之無血清無Ca2+培養基中懸浮生長的H4-II-E細胞系已於2011年6月28日根據布達佩斯條約以登錄編號DSM ACC3130(H4-II-Es)寄存於德國微生物菌種保存中心(DSMZ),Inhoffenstrasse 7B,D-38124 Braunschweig,Germany。The H4-II-E cell line adapted for suspension growth in serum-free Ca 2+-free medium described in the present invention was registered under the Budapest Treaty on June 28, 2011 under the accession number DSM ACC3130 (H4-II-Es). Registered at the German Collection of Microorganisms (DSMZ), Inhoffenstrasse 7B, D-38124 Braunschweig, Germany.

本發明首次闡述對源自不同物種及組織之細胞系之選擇,該等細胞系經分析與其用於產生具有有利糖基化模式(降低之海藻糖基化、存在末端NeuAc-殘基)之重組蛋白的適宜性有關。分析顯示,僅大鼠肝瘤細胞系H4-II-E能夠產生若干有益糖基化模式,此可顯著增進治療蛋白、尤其糖蛋白(例如抗體)之活性、功效及穩定性。The present invention is the first to describe the selection of cell lines derived from different species and tissues which are analyzed for their production to produce a recombination with a favorable glycosylation pattern (reduced haylosylation, presence of terminal NeuAc-residues) The suitability of the protein is related. Analysis has shown that only the rat hepatoma cell line H4-II-E is capable of producing several beneficial glycosylation patterns that can significantly enhance the activity, efficacy and stability of therapeutic proteins, particularly glycoproteins (eg, antibodies).

H4-II-E細胞最初源自大鼠肝細胞癌(REUBER,1961)且經分析作為用於適應肝之活體內模型。與此相比,本發明首次闡述H4-II-E細胞作為高活性治療蛋白之生產系統之用途。源自大鼠肝之H4-II-E細胞不會天然產生抗體,且H4-II-E細胞先前尚未用於重組蛋白產生。H4-II-E細胞源自大鼠肝細胞癌(PITOT等人,1964;REUBER,1961)且專門用於毒性學分析且用作模型系統來研究細胞壓力反應(例如(Horikoshi等人,1988;Houser等人,1992))。H4-II-E cells were originally derived from rat hepatocellular carcinoma (REUBER, 1961) and were analyzed as in vivo models for adaptation to the liver. In contrast, the present invention describes for the first time the use of H4-II-E cells as a production system for highly active therapeutic proteins. H4-II-E cells derived from rat liver do not naturally produce antibodies, and H4-II-E cells have not previously been used for recombinant protein production. H4-II-E cells are derived from rat hepatocellular carcinoma (PITOT et al., 1964; REUBER, 1961) and are used exclusively for toxicological analysis and as a model system to study cellular stress responses (eg (Horikoshi et al., 1988; Houser et al., 1992)).

極令人驚奇地,具體而言大鼠細胞且尤其H4-II-E細胞產生具有低海藻糖含量之糖蛋白或糖基化。與其他物種(例如兔或貓)相比,來自正常大鼠血液血清之抗體係顯著海藻糖基化的(Raju等人,2000)。Hamako及同事分析了來自不同物種之血清IgG抗體之糖組成且發現來自大鼠血液血清之內源性抗體之92.8%係海藻糖基化的,此比例高於多數其他物種之血清IgG(Hamako等人,1993)。Surprisingly, in particular rat cells and in particular H4-II-E cells produce glycoproteins or glycosylation with a low trehalose content. Anti-systems from blood serum of normal rats are significantly fucosylated compared to other species (eg rabbits or cats) (Raju et al., 2000). Hamako and colleagues analyzed the sugar composition of serum IgG antibodies from different species and found that 92.8% of endogenous antibodies from rat blood serum were fucosylated, which is higher than serum IgG of most other species (Hamako et al. People, 1993).

因此,預計大鼠H4-II-E細胞會產生具有高海藻糖含量而非低海藻糖含量之糖蛋白或糖基化。Therefore, rat H4-II-E cells are expected to produce glycoproteins or glycosylation with high trehalose content rather than low trehalose content.

但意想不到的是,大鼠肝瘤細胞H4-II-E細胞之情形正好相反。本發明顯示,源自大鼠肝之H4-II-E細胞系(例如以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ之細胞及以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞)可藉由其在產生海藻糖含量顯著低於任一其他細胞系之抗體之能力上令人驚奇地與源自不同物種之許多其他細胞系區別開來。Unexpectedly, the situation of rat hepatoma cells H4-II-E cells is just the opposite. The present invention shows that the H4-II-E cell line derived from rat liver (for example, cells deposited in DSMZ under accession number DSM ACC3129 (H4-II-E) and deposited under the accession number DSM ACC3130 (H4-II-Es) The cells of DSMZ can be surprisingly distinguished from many other cell lines derived from different species by their ability to produce antibodies with significantly lower levels of trehalose than any other cell line.

Shinkawa及同事闡述大鼠骨髓瘤細胞系YB2/0用於抗體產生之用途(Shinkawa等人,2003)。YB2/0細胞係天然生成海藻糖基化降低34-91%之Fc糖結構,藉此在ADCC分析中產生提高最多50倍之活性之抗體生產細胞系。因此,YB2/0細胞與H4-II-E細胞已知產生子細胞不同。然而,作為骨髓瘤細胞系,YB2/0細胞對細胞凋亡相當敏感,且對細胞壓力顯示低穩健性,從而使得此細胞系不適於工業產生過程。如所報導,另外,YB2/0細胞中之海藻糖基化高度可變,從而使得難以將海藻糖基化控制並維持在某些閾值內。相比之下,闡述於本發明中之H4-II-E細胞令人驚奇地顯示,去海藻糖基化程度及一致性高於針對YB2/0細胞所報導者。此外,H4-II-E細胞係穩健的且對壓力或其他細胞凋亡誘導刺激物不敏感,如通常使用H4-II-E細胞作為模型系統之許多毒性研究中所顯示。直接比較H4-II-E細胞及YB2/0細胞對不同細胞壓力(滲透壓力、溫度壓力、機械壓力、化學壓力)之敏感性顯示,H4-II-E細胞在各方面皆優於YB2/0細胞。因此H4-II-E細胞比YB2/0細胞意想不到地更適於用作工業產生過程之宿主細胞系。Shinkawa and colleagues describe the use of the rat myeloma cell line YB2/0 for antibody production (Shinkawa et al., 2003). The YB2/0 cell line naturally produces a 14-91% reduction in the Fc glycostructure of the fucosylation, thereby producing an antibody production cell line that increases activity by up to 50-fold in the ADCC assay. Therefore, YB2/0 cells are different from H4-II-E cells in that they are known to produce daughter cells. However, as a myeloma cell line, YB2/0 cells are quite sensitive to apoptosis and show low robustness to cell pressure, making this cell line unsuitable for industrial production processes. As reported, in addition, the fucosylation in YB2/0 cells is highly variable, making it difficult to control and maintain trehalylation within certain thresholds. In contrast, the H4-II-E cells described in the present invention surprisingly show that the degree and consistency of de-fucosylation is higher than that reported for YB2/0 cells. Furthermore, H4-II-E cell lines are robust and insensitive to stress or other apoptosis-inducing stimuli, as shown in many toxicity studies using H4-II-E cells as a model system. Direct comparison of the sensitivity of H4-II-E cells and YB2/0 cells to different cell pressures (osmotic pressure, temperature pressure, mechanical pressure, chemical pressure) shows that H4-II-E cells are superior to YB2/0 in all respects. cell. Thus, H4-II-E cells are unexpectedly more suitable than YB2/0 cells for use as host cell lines for industrial production processes.

用於獲得具有非海藻糖基化糖結構之產物之另一先前方法係使CHO中編碼海藻糖轉移酶8(Fut8)之基因靶向失活,此可導致分泌蛋白(例如抗體產物)之聚糖中完全損失α1,6-海藻糖基化(Yamane-Ohnuki等人,2004)。應用siRNA敲弱技術來削弱Fut8基因活性之替代方法允許產生部分去海藻糖基化抗體(Mori等人,2004)。此等抗體顯示強50至100倍之ADCC活化。然而,該等與使用H4-II-E細胞作為產生細胞不同之策略之缺陷在於對現有宿主細胞系實施遺傳改造一定會比僅僅對所關注分泌蛋白實施糖基化影響更多細胞過程。除期望突變效應外,與H4-II-E細胞不同之經遺傳改造宿主細胞系亦可顯示一些變化,例如在(例如)產生過程發展之後期階段出現之產量降低或不穩定性或不可預測之問題。此外,經遺傳改造細胞系經常需要主動選擇以穩定遺傳改變之效應,藉此增加工業產生過程之調節負擔。Another prior method for obtaining a product having a non-fucosylated glycostructure is to target the gene encoding the trehalose transferase 8 (Fut8) in CHO, which can result in the aggregation of secreted proteins (eg, antibody products). Alpha 1,6-trehalylation is completely lost in sugar (Yamane-Ohnuki et al., 2004). Alternative methods of using siRNA knockdown techniques to attenuate the activity of the Fut8 gene allow for the production of partially de-fucosylated antibodies (Mori et al., 2004). These antibodies show 50 to 100 times greater ADCC activation. However, a disadvantage of these strategies, which differ from the use of H4-II-E cells as producer cells, is that genetic engineering of existing host cell lines will certainly affect more cellular processes than glycosylation of only the secreted proteins of interest. In addition to the desired mutating effect, genetically engineered host cell lines that differ from H4-II-E cells may also exhibit some changes, such as reduced yield or instability or unpredictable during, for example, post-production development. problem. In addition, genetically engineered cell lines often require active selection to stabilize the effects of genetic alterations, thereby increasing the regulatory burden of the industrial production process.

除顯示降低之海藻糖基化外,H4-II-E細胞亦顯示在Fut8缺陷CHO細胞未發現之額外有益性質,例如增加之半乳糖基化及唾液酸化。In addition to showing reduced fucosylation, H4-II-E cells also showed additional beneficial properties not found in Fut8 deficient CHO cells, such as increased galactosylation and sialylation.

優點advantage

業內認為候選細胞系之以下性質相對於現有生產細胞系係有利的:細胞系應為未經遺傳改造之天然(幼稚)宿主細胞系。The following properties of the candidate cell line are believed to be advantageous over existing production cell lines: the cell line should be a natural (naive) host cell line that has not been genetically engineered.

細胞系應能夠產生具有有利糖基化模式之蛋白。The cell line should be capable of producing a protein with a favorable glycosylation pattern.

細胞系應穩定顯示期望糖模式而無需選擇(例如凝集素抗性)。The cell line should stably display the desired sugar pattern without selection (eg, lectin resistance).

此等細胞系可在無血清培養基中懸浮培養。These cell lines can be cultured in suspension in serum-free medium.

此等細胞系之特徵在於對壓力或細胞凋亡刺激物之高穩健性及低敏感性。These cell lines are characterized by high robustness and low sensitivity to stress or apoptosis stimuli.

闡述於本發明中之大鼠肝瘤細胞系H4-II-E組合多數該等有利屬性。The rat hepatoma cell line H4-II-E described in the present invention combines most of these advantageous properties.

與用於獲得海藻糖基化經降低之抗體之先前方法(其主要基於對現有生產細胞系之遺傳改造)不同,H4-II-E細胞天然地(即,無遺傳操作、誘變或選擇)適合用於產生具有有利糖基化模式之重組蛋白及其治療應用。Unlike previous methods for obtaining hypoglycosylated reduced antibodies, which are primarily based on genetic engineering of existing production cell lines, H4-II-E cells are native (ie, without genetic manipulation, mutagenesis or selection). It is suitable for the production of recombinant proteins with favorable glycosylation patterns and their therapeutic applications.

與當前使用之產生宿主細胞系中製造之蛋白不同,H4-II-E中產生之抗體顯示不同有益糖基化性質之組合,從而顯著增進產生品質:Unlike the proteins produced in the host cell line currently used to produce, the antibodies produced in H4-II-E show a combination of different beneficial glycosylation properties, which significantly enhances the quality of production:

- 低海藻糖基化或缺乏海藻糖:>80%非海藻糖基化二枝結構聚糖- low seawed glycosylation or lack of trehalose: >80% non-fucosylated dimeric structural glycans

 提高活化效應子功能(例如ADCC)之潛力 Increase the potential of activated effector functions such as ADCC

- 高半乳糖基化:>40%半乳糖基化二枝結構聚糖- High galactosylation : >40% galactosylated di- structural glycans

 提高活化效應子功能(例如CDC)之潛力 Increase the potential of activated effector functions such as CDC

H4-II-E細胞中產生之抗體藉此具有活化抗體依賴性效應子功能(例如ADCC及CDC)之高潛力。此活化與抗體與不同Fc受體之結合相關聯。除對FcgRIIIa之較高活化外,H4-II-E中產生之抗體亦具有較低程度地活化抑制受體FcγRIIb之潛力。此有益效應使H4-II-E中產生之抗體能夠增強由巨噬細胞介導之免疫反應,此乃因尤其中性粒細胞亦表現抑制受體。因此,H4-II-E細胞中產生之治療抗體尤其對腫瘤靶標且亦對其他適應症在其治療效率上優於以常規方式產生之抗體。The antibodies produced in H4-II-E cells thus have the high potential to activate antibody-dependent effector functions such as ADCC and CDC. This activation is associated with binding of the antibody to different Fc receptors. In addition to the higher activation of FcgRIIIa, the antibodies produced in H4-II-E also have the potential to activate the inhibitory receptor FcyRIIb to a lesser extent. This beneficial effect enables antibodies produced in H4-II-E to enhance the immune response mediated by macrophages, since neutrophils, in particular, also exhibit inhibitory receptors. Thus, therapeutic antibodies produced in H4-II-E cells are superior to tumor-specific and other indications in their therapeutic efficiency over conventionally produced antibodies.

- 不存在免疫原性殘基:缺乏Gal-α1,3-Gal-連接及NeuGc-殘基- absence of immunogenic residues: lack of Gal-α1,3-Gal-linkage and NeuGc-residue

 無過敏或免疫原性反應 No allergic or immunogenic reactions

缺乏潛在免疫原性糖結構(例如Gal-α1,3-Gal-連接糖或NeuGc-殘基)係H4-II-E細胞中產生之糖蛋白(例如抗體或Fc-融合蛋白)之另一優點。可在小鼠骨髓瘤細胞系NS0或SP2/0中產生之抗體中發現的此等結構若施加於敏感患者可誘導不期望之發炎反應或免疫原性排斥。Another advantage of lacking a potentially immunogenic glycostructure (eg, Gal-α1,3-Gal-linked sugar or NeuGc-residue) is a glycoprotein (eg, an antibody or Fc-fusion protein) produced in H4-II-E cells . Such structures found in antibodies produced in the mouse myeloma cell line NS0 or SP2/0 can induce undesirable inflammatory responses or immunogenic rejection if applied to sensitive patients.

- 唾液酸化:末端α2,3或α2,6-連接NeuAc-殘基- sialylation: terminal α2, 3 or α2,6-linked NeuAc-residue

 提高之血清穩定性(血清半衰期) Improved serum stability (serum half-life)

H4-II-E細胞中所產生抗體之聚糖的末端唾液酸化對治療抗體之血清穩定性及分解代謝半衰期具有額外正面效應。The terminal sialylation of the glycans of the antibodies produced in H4-II-E cells has an additional positive effect on the serum stability and catabolic half-life of the therapeutic antibodies.

H4-II-E大鼠肝瘤係未經遺傳改造且無需在凝集素存在下進行選擇或培養以維持其產生有益糖基化模式之能力的細胞。The H4-II-E rat hepatoma line is not genetically engineered and does not require selection or culture in the presence of lectin to maintain its ability to produce a beneficial glycosylation pattern.

此外,H4-II-E細胞系可在無血清培養基中懸浮培養。即使H4-II-E細胞(例如以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ之細胞)通常使用含血清培養基進行貼壁培養(其亦由美國典型培養物保存中心(American Type Culture Collection)(ATCC,寄存編號CRL-1548)或歐洲細胞培養物保存中心(European Collection of Cell Cultures)(ECACC,寄存編號87031301)所推薦),亦可在本發明中證實,與在含血清培養基中以貼壁培養物之生長相比,H4-II-E細胞可成功地適應於在無血清培養基中懸浮生長而不會損害24至32小時之群加倍時間(例如以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞)。In addition, the H4-II-E cell line can be cultured in suspension in serum-free medium. Even H4-II-E cells (eg, cells deposited with DSMZ under accession number DSM ACC3129 (H4-II-E)) are typically cultured in adherent culture using serum-containing media (also known as American Type Culture). Collection) (ATCC, accession number CRL-1548) or European Collection of Cell Cultures (ECACC, accession number 87031301), can also be confirmed in the present invention, and in serum-containing medium Compared to the growth of adherent cultures, H4-II-E cells can be successfully adapted to suspension growth in serum-free medium without compromising group doubling time of 24 to 32 hours (eg, under the accession number DSM ACC3130 (H4- II-Es) cells deposited in DSMZ).

H4-II-E細胞在無血清培養基中之加倍時間係24至32小時。達成H4-II-E細胞在無血清培養基中懸浮恆定生長及此等有利加倍時間之關鍵因素係在培養基中省略鈣。若在細胞培養基中存在鈣,則H4-II-E細胞不會懸浮生長且加倍時間更長,此使得按照商業規模應用於生物醫藥生產不具吸收力。The doubling time of H4-II-E cells in serum-free medium is 24 to 32 hours. A key factor in achieving constant growth of H4-II-E cells in serum-free medium and such favorable doubling time is the omission of calcium in the medium. If calcium is present in the cell culture medium, the H4-II-E cells will not grow in suspension and will be doubled for a longer period of time, which makes it unabsorbable for biomedical production on a commercial scale.

H4-II-E細胞在生物醫藥生產過程中用作宿主細胞之另一優點係其高穩健性及低壓力或細胞凋亡敏感性。其他生產細胞系(例如骨髓瘤細胞系YB2/0)亦顯示出有益糖模式,但同時對任一種類之壓力皆極敏感且因此不適於在大規模產生過程中培養。Another advantage of H4-II-E cells as host cells in biopharmaceutical production is their high robustness and low pressure or apoptosis sensitivity. Other production cell lines (e.g., myeloma cell line YB2/0) also exhibit a beneficial sugar pattern, but are also extremely sensitive to either type of stress and are therefore not suitable for cultivation in large scale production processes.

除先前提及之優點外,H4-II-E細胞中產生之治療蛋白亦具有以下額外有益性質:In addition to the previously mentioned advantages, the therapeutic proteins produced in H4-II-E cells have the following additional beneficial properties:

較高有效性及穩定性=降低之所需劑量Higher effectiveness and stability = reduced required dose

提高之患者便利性(減小之治療(輸注)頻率)Improved patient convenience (reduced treatment (infusion) frequency)

經由降低之循環劑量所達成降低之副作用風險Reduced risk of side effects through reduced circulating dose

用於供應臨床及市售材料之縮短之時間線及降低之成本Reduced timeline and reduced costs for the supply of clinical and commercial materials

總之:降低之治療成本。 In short: reduced treatment costs.

適用性applicability

H4-II-E大鼠細胞可用於工業生產在治療上具有高活性之蛋白、較佳抗體或Fc-融合蛋白,從而在投與患者後可有效活化效應子功能。H4-II-E rat cells can be used to industrially produce therapeutically highly active proteins, preferably antibodies or Fc-fusion proteins, thereby effectively activating effector functions upon administration to a patient.

具有顯著降低之核心海藻糖含量,H4-II-E產生之抗體或Fc-融合蛋白對多態性受體FcγRIII(CD16-F158,CD16-V158)顯示較高之親和力且較低程度地活化抑制受體FcγRIIb。藉此,與海藻糖基化CHO產生之抗體不同,H4-II-E產生之抗體不僅可快速且有效地募集並活化CD16陽性細胞,而且亦可募集並活化巨噬細胞及中性粒細胞以活化免疫系統之細胞毒性功能。顯示半乳糖基化高於以常規方式產生之抗體,H4-II-E細胞中修飾並分泌之產物可另外更有效地結合補體系統之組份(即Clq)且藉此活化可最終殺傷靶細胞之另一級聯。With a significantly reduced core trehalose content, H4-II-E-producing antibodies or Fc-fusion proteins show higher affinity for the polymorphic receptor FcγRIII (CD16-F158, CD16-V158) and a lower degree of activation inhibition Receptor FcyRIIb. In contrast, unlike the antibodies produced by the fucosylated CHO, the antibodies produced by H4-II-E can not only rapidly and efficiently recruit and activate CD16-positive cells, but also recruit and activate macrophages and neutrophils. Activate the cytotoxic function of the immune system. It is shown that the galactosylation is higher than the antibody produced in a conventional manner, and the product modified and secreted in the H4-II-E cell can additionally bind the component of the complement system (ie, Clq) more effectively and thereby activate to finally kill the target cell. Another cascade.

出於多種原因(參見上述優點),H4-II-E細胞係所選用於產生抗體或Fc-融合蛋白、尤其彼等識別腫瘤靶標者之細胞系。藉由H4-II-E細胞中產生之抗體或Fc-融合蛋白對效應子功能ADCC及CDC實施之增進活化亦允許在抗體治療後對實體腫瘤實施有效治療,該等實體腫瘤通常不受患者免疫系統有效攻擊。For a variety of reasons (see the above advantages), H4-II-E cell lines are selected for the production of antibodies or Fc-fusion proteins, particularly those that recognize tumor targets. Enhanced activation of effector functions ADCC and CDC by antibodies or Fc-fusion proteins produced in H4-II-E cells also allows for effective treatment of solid tumors after antibody treatment, which are usually immune to patient immunity The system is effectively attacked.

H4-II-E細胞中產生之抗體之經增進血清穩定性在許多治療領域、尤其在慢性疾病中有益,其中當前必須將治療物質頻繁地且重複地遞送至患者。The improved serum stability of antibodies produced in H4-II-E cells is beneficial in many therapeutic areas, particularly in chronic diseases, where therapeutic substances must currently be delivered to patients frequently and repeatedly.

已知轉譯後蛋白修飾對蛋白之生物活性、穩定性及免疫原性具有關鍵效應。分泌蛋白之最常見轉譯後修飾係糖基化且所有經批准治療生物醫藥(包括重組抗體或重組Fc-融合蛋白)中之大多數係糖蛋白。It is known that post-translational protein modification has a key effect on the biological activity, stability and immunogenicity of the protein. The most common post-translational modifications of secreted proteins are glycosylation and most of the glycoproteins in all approved biopharmaceuticals (including recombinant antibodies or recombinant Fc-fusion proteins).

由於正確糖基化模式對於治療活性及特異性係不可缺少的,因此多數糖蛋白治療劑係在能夠生成糖基化模式之複雜光譜的真核表現系統中產生。當前,中國倉鼠卵巢(CHO)細胞已成為用於產生重組蛋白之標準哺乳動物宿主細胞。然而,已知不同真核生產細胞系產生不同Fc糖基化模式且藉此對治療抗體之生物活性具有重要影響。此外,已知某些確定糖結構在活化具體下游效應子機制上比其他更有效。帶有多數人類樣糖基化模式之治療抗體在活化抗腫瘤效應子機制上並不尤其有效,但同時,此等抗體不太可能引起患者之不期望之過敏性或免疫原性排斥反應且因此視為完全的。為增進(例如)由抗癌症抗體誘導之細胞殺傷活性,期望產生不同糖基化模式。同樣,經改變糖基化模式可增進其他抗體依賴性效應子功能或其可改變血清半衰期,而不會損害治療產品本身之安全性或穩定性。Since the correct glycosylation pattern is indispensable for therapeutic activity and specificity, most glycoprotein therapeutics are produced in eukaryotic expression systems capable of generating complex spectra of glycosylation patterns. Currently, Chinese hamster ovary (CHO) cells have become standard mammalian host cells for the production of recombinant proteins. However, it is known that different eukaryotic production cell lines produce different Fc glycosylation patterns and thereby have a significant impact on the biological activity of the therapeutic antibodies. In addition, certain certain sugar structures are known to be more effective than others in activating specific downstream effector mechanisms. Therapeutic antibodies with most human-like glycosylation patterns are not particularly effective at activating anti-tumor effector mechanisms, but at the same time, such antibodies are less likely to cause undesirable allergic or immunogenic rejection in patients and therefore Treated as complete. To enhance, for example, cell killing activity induced by anti-cancer antibodies, it is desirable to have different glycosylation patterns. Likewise, altered glycosylation patterns may enhance other antibody-dependent effector functions or may alter serum half-life without compromising the safety or stability of the therapeutic product itself.

在本發明中,具體選擇並分析源自多種物種、組織及細胞譜系之不同細胞系。對暴露於細胞表面上之聚糖以及在細胞內之酶促活性進行了檢驗(圖1)。藉此,可賦予每一細胞系合成某些糖基化模式之能力。選擇及分析顯示細胞系在其糖基化能力上明顯展示差異,此端視其所源自之物種與組織或細胞譜系二者而定。僅少數細胞系天然展示低海藻糖基化能力,此糖基化特性對抗體依賴性細胞毒性途徑(ADCC)之活化具有正面影響。一些細胞系能夠生成可增進治療蛋白之血清穩定性之唾液酸化結構。一些細胞系產生免疫原性糖結構(例如Gal-1,3-Gal及NeuGc),從而引起人類之發炎反應。對本發明之分析顯示,單獨給定細胞系之物種來源或組織、器官或細胞譜系皆不能確定細胞之糖基化能力。因此,遺傳及後生因素影響細胞合成某些糖基化模式之能力。因此,本發明數據證實,僅僅基於對源自相同組織及/或物種之另一細胞系中獲得之糖模式的瞭解不可能預測細胞系之糖基化性質(圖1)。In the present invention, different cell lines derived from a variety of species, tissues, and cell lineages are specifically selected and analyzed. The glycans exposed on the cell surface and the enzymatic activity in the cells were examined (Fig. 1). Thereby, each cell line can be given the ability to synthesize certain glycosylation patterns. Selection and analysis showed that the cell line clearly showed differences in its glycosylation ability depending on the species from which it originated and the tissue or cell lineage. Only a few cell lines naturally exhibit low algal glycosylation properties, which have a positive effect on the activation of the antibody-dependent cellular cytotoxic pathway (ADCC). Some cell lines are capable of producing a sialylation structure that enhances the serum stability of the therapeutic protein. Some cell lines produce immunogenic sugar structures (eg, Gal-1,3-Gal and NeuGc) that cause an inflammatory response in humans. Analysis of the present invention shows that the species source or tissue, organ or cell lineage of a given cell line alone is unable to determine the glycosylation ability of the cell. Thus, genetic and epigenetic factors influence the ability of cells to synthesize certain glycosylation patterns. Thus, the data of the present invention demonstrate that it is not possible to predict the glycosylation properties of a cell line based solely on knowledge of the sugar patterns obtained in another cell line derived from the same tissue and/or species (Fig. 1).

顯示若干性質(例如降低之海藻糖基化、缺乏免疫原性殘基及存在α-2,6連接唾液酸)之唯一細胞系係大鼠肝瘤細胞系H4-II-E。該等性質正面影響重組生物治療劑、尤其抗體或Fc-融合蛋白之活性及穩定性。所選細胞系H4-II-E係所有經分析細胞系在此分析中唯一未顯示可檢測之海藻糖基化跡象者。此令人感到驚奇,此乃因其他大鼠細胞系及其他肝細胞系具有不同性質。此外,據闡述,大鼠血液血清抗體係顯著海藻糖基化的,而根據文獻,其他物種(例如兔或貓)在血清抗體上具有低於大鼠之海藻糖含量(Raju等人,2000)。The only cell line that exhibits several properties (eg reduced hyalosylation, lack of immunogenic residues, and the presence of alpha-2,6 linked sialic acid) is the rat hepatoma cell line H4-II-E. These properties positively affect the activity and stability of recombinant biotherapeutics, particularly antibodies or Fc-fusion proteins. Selected cell lines H4-II-E were the only cells in the assay that showed no detectable signs of fucosylation in this assay. This is surprising because other rat cell lines and other liver cell lines have different properties. Furthermore, it has been demonstrated that the rat serum anti-system is significantly fucosylated, and according to the literature, other species (such as rabbits or cats) have lower serum levels of trehalose than rats (Raju et al., 2000). .

糖結構(例如Gal-1,3-Gal及NeuGc)具有潛在免疫原性。H4-II-E大鼠細胞不顯示此等潛在免疫原性糖結構。H4-II-E大鼠細胞另外不產生具有此等潛在免疫原性糖結構之抗體或Fc-融合蛋白。實際上,具有潛在免疫原性之諸如Gal-1,3-Gal及NeuGc等糖結構在所選人類或大鼠細胞系中皆未出現,而源自小鼠、兔及其他物種之細胞系則一致地產生此等結構,此可誘導人類之免疫原性反應。因此,根據本發明之實驗數據,該等潛在免疫原性糖結構由細胞以物種依賴性方式附著至分泌糖蛋白。Sugar structures such as Gal-1,3-Gal and NeuGc are potentially immunogenic. H4-II-E rat cells do not display these potential immunogenic sugar structures. H4-II-E rat cells additionally do not produce antibodies or Fc-fusion proteins with such potential immunogenic sugar structures. In fact, glycostructures such as Gal-1,3-Gal and NeuGc with potential immunogenicity are not present in selected human or rat cell lines, whereas cell lines derived from mice, rabbits and other species are These structures are produced consistently, which induces an immunogenic response in humans. Thus, according to experimental data of the present invention, these potentially immunogenic sugar structures are attached to the secreted glycoprotein by the cells in a species-dependent manner.

為驗證H4-II-E細胞能夠產生具有預測糖基化性質之分泌糖蛋白,藉由對相應DNA構築體實施轉染並隨後選擇穩定整合產物基因及抗生素抗性標記物之細胞來生成產生穩定重組抗體之H4-II-E細胞。獲得產生抗體之細胞群並將其作為貼壁細胞層培養。H4-II-E細胞另外適應於懸浮生長且可在無血清化學限定培養基中培養(圖5)。藉由蛋白質A層析自細胞培養上清液純化抗體。分析Fc糖基化並將其與在CHO-DG44、CHO-Lec13突變體及YB2/0大鼠骨髓瘤細胞中產生之抗體上獲得之模式進行比較。二枝結構聚糖佔所有4種IgG1製劑中之最大部分。在二枝結構聚糖之比例內,CHO-DG44細胞如先前所報導主要(約95%)產生海藻糖基化形式。相比之下,H4-II-E細胞中表現之IgG1中之80%以上含有無海藻糖之二枝結構聚糖,此比例顯著高於在細胞系YB2/0或CHO突變體Lec13中產生抗體(圖2)。對H4-II-E細胞中產生之抗體上糖基化模式之詳細分析亦顯示,>40%聚糖係半乳糖基化的,此比例高於在CHO-DG44中產生抗體時所獲得者,該CHO-DG44宿主細胞系通常用於生物醫藥蛋白之產生(圖3)。此外,H4-II-E細胞中產生之約8%抗體帶有末端唾液酸殘基,若在CHO-DG44細胞中產生重組抗體,則通常不會發現該等末端唾液酸殘基(圖4)。末端唾液酸殘基可對修飾抗體之活性及穩定性具有不同效應。最近出版物顯示唾液酸可抑制抗體之發炎活性(Burton及Dwek,2006;Scallon等人,2007)。其他數據顯示,不存在唾液酸可提高小鼠肝之代謝速率,從而表明藉由基於肝之受體之清除(Wright等人,2000)。發現唾液酸衍生物(例如N-羥乙醯神經胺酸(NeuGc))在人類中具有免疫原性(Noguchi等人,1995)。然而,不能檢測出H4-II-E細胞中產生之蛋白上之NeuGc修飾之證據(圖1)。總之,H4-II-E源抗體缺乏潛在免疫原性殘基。To verify that H4-II-E cells are capable of producing secreted glycoproteins with predictive glycosylation properties, stable production is achieved by transfecting the corresponding DNA constructs and subsequently selecting cells that stably integrate the product gene and antibiotic resistance markers. Recombinant antibody H4-II-E cells. The antibody-producing cell population is obtained and cultured as an adherent cell layer. H4-II-E cells were additionally adapted for suspension growth and cultured in serum-free chemically defined medium (Figure 5). The antibody was purified from the cell culture supernatant by protein A chromatography. Fc glycosylation was analyzed and compared to the pattern obtained on antibodies produced in CHO-DG44, CHO-Lec13 mutants and YB2/0 rat myeloma cells. The two branched glycans accounted for the largest part of all four IgG1 formulations. Within the ratio of the two structural glycans, CHO-DG44 cells produced a predominantly (approximately 95%) pre-glycosylated form as previously reported. In contrast, more than 80% of the IgG1 expressed in H4-II-E cells contained trehalose-free twig-structured glycans, which was significantly higher than that produced in the cell line YB2/0 or the CHO mutant Lec13. (figure 2). A detailed analysis of the glycosylation pattern of antibodies produced in H4-II-E cells also showed that >40% glycans were galactosylated, a higher ratio than those obtained when antibodies were produced in CHO-DG44, This CHO-DG44 host cell line is commonly used for the production of biopharmaceutical proteins (Figure 3). In addition, about 8% of antibodies produced in H4-II-E cells carry terminal sialic acid residues, and if a recombinant antibody is produced in CHO-DG44 cells, these terminal sialic acid residues are usually not found (Fig. 4). . The terminal sialic acid residue can have different effects on the activity and stability of the modified antibody. Recent publications have shown that sialic acid inhibits the inflammatory activity of antibodies (Burton and Dwek, 2006; Scallon et al., 2007). Other data show that the absence of sialic acid increases the metabolic rate of the liver in mice, indicating clearance by liver-based receptors (Wright et al., 2000). Sialic acid derivatives (e.g., N-hydroxyethyl ceramide (NeuGc)) have been found to be immunogenic in humans (Noguchi et al., 1995). However, evidence of NeuGc modification on proteins produced in H4-II-E cells could not be detected (Fig. 1). In summary, H4-II-E source antibodies lack potential immunogenic residues.

該等表現數據證實了對有利糖基化模式之預測分析中獲得之結果(圖1)。總之,大鼠肝瘤細胞系H4-II-E本身因能生成若干種有益糖基化模式而與其他細胞系區別開來。H4-II-E細胞中在抗體產生後獲得之所述糖基化模式可表現為在ADCC分析中抗體與FcγRIII受體之經增進結合親和力及極佳活性、在CDC分析中與補體系統組份之較強結合及增強之活性、與新生兒Fc受體FcRn之較強結合(對抗體穩定性及血清半衰期具有正面效應)。These performance data confirm the results obtained in the predictive analysis of favorable glycosylation patterns (Figure 1). In conclusion, the rat hepatoma cell line H4-II-E itself is distinguished from other cell lines by the ability to generate several beneficial glycosylation patterns. The glycosylation pattern obtained after antibody production in H4-II-E cells can be expressed as enhanced binding affinity and excellent activity of the antibody to the FcγRIII receptor in the ADCC assay, and in the CDC assay with the complement system component. Strong binding and enhanced activity, strong binding to the neonatal Fc receptor FcRn (positive effect on antibody stability and serum half-life).

除產生有益糖性質外,H4-II-E細胞之特徵亦在於對壓力或細胞凋亡誘導刺激物之高穩健性及低敏感性。據此,H4-II-E細胞可適應於在無血清培養基中懸浮生長。H4-II-E細胞在不同培育模式下生長良好且可在進料分批過程中以高活力懸浮培養10天以上。總之,H4-II-E細胞可對於大規模工業產生效應子活性突出且血清半衰期延長之生物治療劑帶來極佳品質。In addition to producing beneficial glycoprotein properties, H4-II-E cells are also characterized by high robustness and low sensitivity to stress or apoptosis-inducing stimuli. Accordingly, H4-II-E cells can be adapted to grow in suspension in serum-free medium. H4-II-E cells grow well in different incubation modes and can be cultured in suspension for more than 10 days with high vigor during the feed batch process. In conclusion, H4-II-E cells provide excellent quality for large-scale industrial biotherapeutics with outstanding effector activity and prolonged serum half-life.

定義definition

一般實施例「包含(comprising或comprised)」涵蓋更具體之實施例「由...組成」。此外,單數及複數形式並不以限定方式使用。本發明所用術語具有以下含義。The general embodiment "comprising or comprising" encompasses a more specific embodiment "consisting of". In addition, the singular and plural forms are not used in a limiting manner. The terms used in the present invention have the following meanings.

本發明係關於所有源自「Reuber H-35肝瘤」之大鼠肝瘤細胞系((REUBER,1961)及1964(PITOT等人,1964))。「Reuber H-35肝瘤」係AxC大鼠中藉由化學致癌作用所誘導。源自此等「Reuber H-35肝瘤」之細胞系包括(例如)H4-II-E及H4-II-E-C3細胞系及其衍生物或子代。The present invention relates to all rat hepatoma cell lines derived from "Reuber H-35 hepatoma" (REUBER, 1961) and 1964 (PITOT et al., 1964). "Reuber H-35 hepatoma" was induced by chemical carcinogenesis in AxC rats. Cell lines derived from such "Reuber H-35 hepatoma" include, for example, H4-II-E and H4-II-E-C3 cell lines and derivatives or progeny thereof.

更具體而言,「H4-II-E細胞」意指源自歐洲細胞培養物保存中心(ECACC,目錄編號87031301)或美國典型培養物保存中心(ATCC,寄存編號CRL-1548)或源自於1961年(REUBER,1961)及1964年(PITOT等人,1964)分離且首先闡述於文獻中之大鼠肝瘤細胞系之細胞。具體而言,H4-II-E細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。More specifically, "H4-II-E cell" means derived from the European Cell Culture Preservation Center (ECACC, catalog number 87031301) or the American Type Culture Collection (ATCC, accession number CRL-1548) or derived from Cells of the rat hepatoma cell line isolated in 1961 (REUBER, 1961) and 1964 (PITOT et al., 1964) were first described in the literature. Specifically, the H4-II-E cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548.

術語「H4-II-E細胞」另外意指H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112);H4II細胞系(HPACC Nr. 89042702),其亦源自「Reuber H35肝瘤」且與H4-11-E-C3(ECACC目錄編號85061112)相同;H4-TG細胞系(CRL-1578),其係具有HPRT缺陷且恆定表現苯丙胺酸羥化酶之CRL-1600衍生物;H5細胞系(HPACC,Nr. 94101905),其亦係H4-II-E-C3(CRL-1600)之亞純系;及H4-S細胞系(HPACC Nr. 89102001),其係經VS病毒感染之大鼠肝瘤細胞。The term "H4-II-E cell" additionally means the H4-II-E-C3 cell line (CRL-1600 or HPACC No. 85061112); the H4II cell line (HPACC Nr. 89042702), which is also derived from "Reuber H35 hepatoma". And the same as H4-11-E-C3 (ECACC Cat. No. 85061112); H4-TG cell line (CRL-1578), which is a CRL-1600 derivative with HPRT deficiency and constant expression of amphetamine hydroxylase; H5 Cell line (HPACC, Nr. 94101905), which is also a mesogenic line of H4-II-E-C3 (CRL-1600); and H4-S cell line (HPACC Nr. 89102001), which is infected by VS virus. Murine liver tumor cells.

術語「H4-II-E細胞」亦包含最初源自經寄存H4-II-E細胞之細胞及為最初寄存H4-II-E細胞之子代或最初源自經寄存H4-II-E細胞之細胞。The term "H4-II-E cells" also encompasses cells originally derived from deposited H4-II-E cells and are progeny originally deposited with H4-II-E cells or cells originally derived from deposited H4-II-E cells. .

因此,術語「H4-II-E細胞」包含H4-II-E細胞之未經修飾或經修飾後代/形式。H4-II-E細胞之此等未經修飾或經修飾後代/形式亦稱作最初寄存H4-II-E細胞之「衍生物或子代」。Thus, the term "H4-II-E cell" encompasses an unmodified or modified progeny/form of H4-II-E cells. Such unmodified or modified progeny/form of H4-II-E cells are also referred to as "derivatives or progeny" of the originally deposited H4-II-E cells.

H4-II-E細胞之未經修飾形式係所產生之全部細胞,其構成H4-II-E細胞之未經修飾功能亞單位。一些實例包括:未經修飾細胞系之亞純系、其經純化或分級分離亞群。The unmodified form of H4-II-E cells is the entire cell produced by the unconstituted subunit of H4-II-E cells. Some examples include: a sub-pure of an unmodified cell line, a purified or fractionated subpopulation thereof.

H4-II-E細胞之經修飾形式(或衍生物或子代)係經由引入功能DNA序列、尤其彼等賦予各別起始細胞產生重組蛋白、尤其糖蛋白(包括抗體或Fc-融合蛋白)之潛力者自親本H4-II-E細胞生成之全部細胞。Modified forms (or derivatives or progeny) of H4-II-E cells confer recombinant proteins, in particular glycoproteins (including antibodies or Fc-fusion proteins), by introducing functional DNA sequences, in particular, to each of the respective starting cells. The potential of all cells produced by the parental H4-II-E cells.

H4-II-E細胞之修飾形式(或衍生物或子代)係經由誘變或靶向基因修飾或基因整合自親本H4-II-E細胞生成之全部細胞。此等經修飾H4-II-E細胞係經遺傳改造H4-II-E細胞,其包含轉基因(例如脂質轉移蛋白CERT,亦稱為Goodpasture抗原結合蛋白)、轉錄因子(例如上游結合因子UBF)或編碼Sec-1/Munc18蛋白家族成員之基因。Modified forms (or derivatives or progeny) of H4-II-E cells are all cells produced from the parental H4-II-E cells via mutagenesis or targeted gene modification or gene integration. Such modified H4-II-E cell lines are genetically engineered into H4-II-E cells, which comprise a transgene (eg, a lipid transfer protein CERT, also known as Goodpasture antigen binding protein), a transcription factor (eg, upstream binding factor UBF), or A gene encoding a member of the Sec-1/Munc18 protein family.

經修飾H4-II-E細胞之其他實例係敲除或敲弱一或多種基因(例如DHFR(二氫葉酸脫氫酶)、GS(麩醯胺酸合成酶)、TIP-5或SNF 2H)之H4-II-E細胞。尤其敲除DHFR或GS之H4-II-E細胞在重組蛋白表現之情形下因有利選擇選項而可用於生物醫藥生產。Other examples of modified H4-II-E cells knock out or knock down one or more genes (eg, DHFR (dihydrofolate dehydrogenase), GS (glutamate synthase), TIP-5 or SNF 2H) H4-II-E cells. In particular, H4-II-E cells that knock out DHFR or GS can be used in biomedical production due to favorable selection options in the case of recombinant protein expression.

此等敲除或敲弱DHFR或GS之H4-II-E細胞係營養缺陷型細胞之實例。敲除或敲弱DHFR之H4-II-E細胞係次黃嘌呤(H)及胸苷(T)營養缺陷型。敲除或敲弱GS之H4-II-E細胞係麩醯胺酸營養缺陷型。但存在熟習此項技術者可生成之其他營養缺陷型H4-II-E細胞。Examples of such auxotrophic cells that knocked out or knocked down the H4-II-E cell line of DHFR or GS. Knockout or knockdown of HH-II-E cells of DHFR are hypoxanthine (H) and thymidine (T) auxotrophs. Knocking or knocking down the GS H4-II-E cell line glutamate auxotrophy. However, there are other auxotrophic H4-II-E cells that can be generated by those skilled in the art.

H4-II-E細胞之經修飾形式(或衍生物或子代)另外意指經由對特殊培養基、生長、培養模式或選擇物質之適應自親本H4-II-E細胞生成之任一細胞。A modified form (or derivative or progeny) of a H4-II-E cell additionally means any cell produced from a parental H4-II-E cell via adaptation to a particular medium, growth, culture mode or selection of material.

具體而言,術語H4-II-E細胞係關於兩種寄存細胞系。本發明H4-II-E細胞系已於2011年6月28日根據布達佩斯條約以登錄編號DSM ACC3129(H4-II-E)寄存於德國微生物菌種保存中心(DSMZ),Inhoffenstrasse 7B,D-38124 Braunschweig,Germany。In particular, the term H4-II-E cell line is directed to two deposited cell lines. The H4-II-E cell line of the present invention was deposited with the German Collection of Microbial Species (DSMZ), Inhoffenstrasse 7B, D-38124, on June 28, 2011 under the Budapest Treaty under the accession number DSM ACC3129 (H4-II-E). Braunschweig, Germany.

適應於在闡述於本發明中之無血清無Ca2+培養基中懸浮生長的H4-II-E細胞系已於2011年6月28日根據布達佩斯條約以登錄編號DSM ACC3130(H4-II-Es)寄存於德國微生物菌種保存中心(DSMZ),Inhoffenstrasse 7B,D-38124 Braunschweig,Germany。The H4-II-E cell line adapted for suspension growth in serum-free Ca 2+-free medium described in the present invention was registered under the Budapest Treaty on June 28, 2011 under the accession number DSM ACC3130 (H4-II-Es). Registered at the German Collection of Microorganisms (DSMZ), Inhoffenstrasse 7B, D-38124 Braunschweig, Germany.

本文所用術語「細胞」及「細胞系」尤其係指表現細胞/表現細胞系及宿主細胞/宿主細胞系。The terms "cell" and "cell line" as used herein, in particular, refer to a cell/presenting cell line and a host cell/host cell line.

術語鈣減少或較佳無鈣培養基意指限定含有1 μmol/L-500 μmol/L Ca2+離子之培養基,更佳地鈣減少培養基含有1 μmol/L-250 μmol/L Ca2+離子且甚至更佳地鈣減少或無Ca培養基含有0 μmol/L-100 μmol/L或0.5 μmol/L-100 μmol/L Ca2+離子。在AEM培養基中,在培養3天後添加250 μM/L CaCl2或更高CaCl2濃度會顯著損害H4-II-E細胞生長。在補充有250 μM/L CaCl2或更高CaCl2濃度之AEM培養基中,80%以上之細胞形成緊密聚集體。亦可經由將EDTA添加至最初含有鈣之培養基中,藉此藉由與EDTA形成錯合物以降低培養基游離鈣離子之濃度來獲得鈣減少培養基。較佳以介於400 μmol/L-1200 μmol/L或600 μmol/L-1000 μmol/L或700 μmol/L-900 μmol/L之間之濃度添加EDTA。較佳以800 μmol/L左右之濃度添加EDTA。The term calcium reduction or preferably calcium-free medium means to define a medium containing 1 μmol/L to 500 μmol/L Ca 2+ ions, and more preferably the calcium reduction medium contains 1 μmol/L to 250 μmol/L Ca 2+ ions and Even better calcium-reduced or Ca-free medium contains 0 μmol/L to 100 μmol/L or 0.5 μmol/L to 100 μmol/L Ca 2+ ions. In AEM medium, addition of 250 μM/L CaCl 2 or higher CaCl 2 concentration after 3 days of culture significantly impaired H4-II-E cell growth. In AEM medium supplemented with 250 μM/L CaCl 2 or higher CaCl 2 concentration, more than 80% of the cells formed tight aggregates. Calcium-reducing medium can also be obtained by adding EDTA to a medium originally containing calcium, thereby reducing the concentration of free calcium ions in the medium by forming a complex with EDTA. Preferably, EDTA is added at a concentration between 400 μmol/L and 1200 μmol/L or 600 μmol/L to 1000 μmol/L or 700 μmol/L to 900 μmol/L. Preferably, EDTA is added at a concentration of about 800 μmol/L.

市售無鈣培養基之實例係MEM Joklik Modification(Sigma)或MEM Spinner Modification(Sigma)。用於在血清存在下貼壁培育H4-II-E細胞之典型含鈣培養基係含有1360 μmol/L Ca2+離子之鷹氏最低必需培養基(Eagle's Minimum Essential Medium)(Sigma)。用於懸浮培育H4-II-E細胞之無血清鈣減少培養基係AEM(Invitrogen)。An example of a commercially available calcium-free medium is MEM Joklik Modification (Sigma) or MEM Spinner Modification (Sigma). A typical calcium-containing medium for adherent incubation of H4-II-E cells in the presence of serum is Eagle's Minimum Essential Medium (Sigma) containing 1360 μmol/L Ca 2+ ions. The serum-free calcium-reducing medium for suspension incubation of H4-II-E cells is AEM (Invitrogen).

與鈣離子不同,鎂離子對大鼠肝瘤細胞/H4-II-E細胞之聚集比率無影響。因此,大鼠肝瘤細胞/H4-II-E細胞聚集具有鎂(Mg2+)離子獨立性,但具有鈣(Ca2+)離子依賴性。Unlike calcium ions, magnesium ions have no effect on the aggregation ratio of rat hepatoma cells/H4-II-E cells. Therefore, rat hepatoma cells/H4-II-E cells aggregated with magnesium (Mg 2+ ) ion independence but with calcium (Ca 2+ ) ion dependence.

「糖基化位點」係指由真核細胞識別為糖殘基附著位置之胺基酸殘基。術語「糖苷結構」或「聚糖」係指附著至糖基化位點之糖殘基。附著碳水化合物(例如寡糖)之胺基酸通常為天冬醯胺酸(N-連接)、絲胺酸(O-連接)及蘇胺酸(O-連接)殘基。具體附著位點通常由本文稱作「糖基化位點序列」之胺基酸序列界定。"Glycosylation site" refers to an amino acid residue recognized by a eukaryotic cell as a sugar residue attachment site. The term "glycoside structure" or "glycan" refers to a sugar residue attached to a glycosylation site. Amino acids attached to carbohydrates (e.g., oligosaccharides) are typically aspartic acid (N-linked), serine (O-linked), and threonine (O-linked) residues. The specific attachment site is typically defined by an amino acid sequence referred to herein as a "glycosylation site sequence."

用於N-連接糖基化之糖基化位點序列係-Asn-X-Ser-或-Asn-X-Thr-,其中X可係習用胺基酸中除脯胺酸外之任一者。用於O-連接糖基化之主要糖基化位點係-(Thr或Ser)-X-X-Pro-(SEQ ID NO 1),其中X係任一習用胺基酸。術語「N-連接」及「O-連接」係指在糖分子與胺基酸殘基之間用作附著位點的化學基團。N-連接之糖經由胺基附著;O-連接之糖經由羥基附著。然而,並非蛋白中之所有糖基化位點序列皆必須經糖基化。一些蛋白係以糖基化形式與非糖基化形式二者分泌,而其他則在一個糖基化位點序列處經完全糖基化,但含有另一未糖基化之糖基化位點序列。因此,並非多肽中存在之所有糖基化位點序列皆必須為實際附著糖之糖基化位點。在生物合成期間初始N-糖基化插入「核心碳水化合物」或「核心寡糖」。a glycosylation site sequence for N-linked glycosylation - Asn-X-Ser- or -Asn-X-Thr-, wherein X may be any of the amino acids other than proline in the conventional amino acid . The major glycosylation site for O-linked glycosylation - (Thr or Ser)-X-X-Pro- (SEQ ID NO 1), wherein X is any conventional amino acid. The terms "N-linked" and "O-linked" refer to a chemical group that acts as an attachment site between a sugar molecule and an amino acid residue. The N-linked sugar is attached via an amine group; the O-linked sugar is attached via a hydroxyl group. However, not all glycosylation site sequences in the protein must be glycosylated. Some proteins are secreted in both glycosylated and non-glycosylated forms, while others are fully glycosylated at one glycosylation site sequence but contain another unglycosylated glycosylation site. sequence. Therefore, not all glycosylation site sequences present in the polypeptide must be glycosylation sites that actually attach to the sugar. Initial N-glycosylation is inserted into "core carbohydrates" or "core oligosaccharides" during biosynthesis.

在N-連接糖基化中,碳水化合物部分經由GlcNAc附著至多肽鏈中之天冬醯胺酸殘基。N-連接碳水化合物具有不同結構,但全部含有共同核心結構,其中結合天冬醯胺酸之末端GlcNAc稱為還原端且相反側稱為非還原端:In N-linked glycosylation, the carbohydrate moiety is attached to the aspartate residue in the polypeptide chain via GlcNAc. N-linked carbohydrates have different structures, but all contain a common core structure in which the end GlcNAc binding to aspartic acid is referred to as the reducing end and the opposite side is referred to as the non-reducing end:

熟習此項技術者應認識到,根據Kabat EU命名法(Kabat等人,1991),例如,鼠類IgG3、IgG1、IgG2B、IgG2A及人類IgD、IgG3、IgG1、IgA1、IgG2及IgG4CH2結構域中之每一者在第297位胺基酸殘基處皆具有用於N-連接糖基化之單一保守位點。抗體結構域中之殘基係以常規方式根據由Kabat所述之系統(Kabat等人,1991)來編號,此係指對EU抗體之編號(Edelman等人,1969)。應注意,Kabat殘基命名並不總是直接對應於胺基酸殘基之線性編號。與對應於縮短或插入結構組份之嚴格Kabat編號相比,實際線性胺基酸序列可含有更少或額外胺基酸。可藉由用「標準」Kabat經編號序列比對抗體序列中之同源殘基來確定給定抗體殘基之正確Kabat編號。熟習此項技術者應瞭解,該等慣例由免疫球蛋白序列具體區域中之非依序編號組成,使得能正規化參照免疫球蛋白家族中之保守位置。Those skilled in the art will recognize that, according to the Kabat EU nomenclature (Kabat et al., 1991), for example, murine IgG3, IgG1, IgG2B, IgG2A, and human IgD, IgG3, IgG1, IgA1, IgG2, and IgG4 CH2 domains Each has a single conserved site for N-linked glycosylation at the amino acid residue at position 297. Residues in the antibody domain are numbered in a conventional manner according to the system described by Kabat (Kabat et al., 1991), which refers to the numbering of EU antibodies (Edelman et al., 1969). It should be noted that the Kabat residue designation does not always correspond directly to the linear numbering of the amino acid residues. The actual linear amino acid sequence may contain fewer or additional amino acids than the stringent Kabat number corresponding to the shortened or inserted structural component. The correct Kabat numbering for a given antibody residue can be determined by aligning the homologous residues in the antibody sequence with a "standard" Kabat numbered sequence. Those skilled in the art will appreciate that such practices consist of non-sequential numbering in specific regions of the immunoglobulin sequence such that normalized positions in the reference immunoglobulin family can be normalized.

最重要之N-連接碳水化合物係「複合」N-連接碳水化合物。根據本發明,此等複合碳水化合物應具有本文所述「二枝結構」結構。核心二枝結構結構係二枝結構寡糖之典型特點且可示意性表示如下。The most important N-linked carbohydrates are "complex" N-linked carbohydrates. In accordance with the present invention, such complex carbohydrates should have the "two-branched" structure described herein. The core two-branched structure is a typical feature of two-branched oligosaccharides and can be schematically represented as follows.

由於各二枝結構結構可具有二等分N-乙醯葡糖胺(GlcNAc)、在非還原末端側添加至一或二個分支之外側半乳糖及唾液酸糖及在核心還原端添加至GlcNAc之海藻糖,因此可佔據N-連接Asn 297位點之結構獨特複合型寡糖總共有36種。Since each of the two branched structures may have a halved N-acetylglucosamine (GlcNAc), added to one or two branches on the non-reducing end side, galactose and sialic acid sugar, and added to GlcNAc at the core reducing end. The trehalose, therefore, has a total of 36 structurally complex oligosaccharides that occupy the N-linked Asn 297 site.

亦應認識到,在特定CH2結構域內,因附著於二鏈Fc結構域內任一Asn 297殘基之寡糖鏈不同(或轉譯後修整)而使得Asn 297處之糖基化可係不對稱的。舉例而言,儘管在單一抗體分泌細胞內合成之重鏈在其胺基酸序列上可係均勻的,但其通常經差式糖基化,從而產生大量結構獨特免疫球蛋白糖型(具有不同生物活性及生物物理性質)。在IgG之CH2結構域中發現之複合寡糖結構的主要類型顯示如下。It should also be recognized that in a particular CH2 domain, glycosylation at Asn 297 may be due to differences in oligosaccharide chains attached to any Asn 297 residue in the di-chain Fc domain (or post-translational trimming). Symmetrical. For example, although a heavy chain synthesized in a single antibody secreting cell can be homologous in its amino acid sequence, it is usually differentially glycosylated, resulting in a large number of structurally unique immunoglobulin glycoforms (having different Biological activity and biophysical properties). The major types of complex oligosaccharide structures found in the CH2 domain of IgG are shown below.

二枝結構核心(A2G0):Two-piece structure core (A2G0):

二等分GlcNAc核心:Dichotomous GlcNAc core:

典型複合型二枝結構聚糖:A2FG0:Typical complex type two-structured glycan: A2FG0:

A2G0:A2G0:

A2FG1:A2FG1:

or

A2G1:A2G1:

or

A2FG2:A2FG2:

A2G2:A2G2:

唾液酸化A2FG1:Sialylation of A2FG1:

or

唾液酸化A2G1:Sialylation of A2G1:

or

唾液酸化A2FG2:Sialylation of A2FG2:

or

or

唾液酸化A2G2:Sialylation of A2G2:

or

or

除寡糖之複合類型外,N-糖苷連接糖鏈之其他實例亦包括高甘露糖類型,其中僅甘露糖結合核心結構之非還原末端;雜合型,其中核心結構之非還原末端側同時具有高甘露糖N-糖苷連接糖鏈及複合N-糖苷連接糖鏈之分支;及諸如此類。In addition to the complex type of oligosaccharide, other examples of N-glycoside linked sugar chains also include high mannose types in which only mannose binds to the non-reducing end of the core structure; heterozygous, wherein the non-reducing end side of the core structure has both a branch of a high mannose N-glycoside linkage sugar chain and a complex N-glycoside linkage sugar chain; and the like.

可在N-聚糖樹內不同位點上發現海藻糖,但絕大多數附著抗體之N-聚糖上之海藻糖係以α1,6連接至還原端之末端GlcNAc。在源自人類之重組蛋白中未發現以α1,3連接至末端還原GlcNAc之海藻糖,而是在(例如)植物中產生且保持嚴重免疫原性之風險。其他可能存在但極少發現之海藻糖連接係與枝結構定位GlcNAc之α1,3及α1,4連接或與枝結構定位Gal殘基之α1,2(H-抗原,A及B血型抗原之子結構)。Trehalose can be found at different sites in the N-glycan tree, but most of the trehalose on the N-glycan attached to the antibody is linked to the terminal GlcNAc at the end of the reducing end with α1,6. The trehalose which is linked to the terminal-reduced GlcNAc by α1,3 is not found in the recombinant protein derived from human, but is at risk of being produced, for example, in plants and maintaining serious immunogenicity. Other possible but rarely found trehalose linkages and branching structures localize α1,3 and α1,4 of GlcNAc or locale 1,2 of the Gal residue with the branch structure (H-antigen, substructure of A and B blood group antigens) .

本發明中所述H4-II-E細胞產生之大多數糖鏈具有「複合二枝結構類型」且在還原端不含有結合N-乙醯葡糖胺之海藻糖。Most of the sugar chains produced by the H4-II-E cells of the present invention have a "complex dichotomous structure type" and do not contain trehalose which binds N-acetylglucosamine at the reducing end.

術語「低海藻糖基化」意指小於20%之在糖蛋白內含有/在抗體或Fc-融合蛋白內含有之聚糖/糖苷結構含有結合至末端還原GlcNAc之海藻糖。更佳地,小於10%或甚至更佳小於5%之所有複合二枝結構聚糖含有結合至聚糖之末端還原GlcNAc之海藻糖。低海藻糖基化闡述介於0%至20%海藻糖基化、0%至10%海藻糖基化、較佳0%至5%海藻糖基化之間之範圍。具體而言,低海藻糖基化闡述介於0.5%至20%海藻糖基化、0.5%至10%海藻糖基化、較佳0.5%至5%海藻糖基化之間之範圍。低海藻糖基化另外闡述介於1%至20%海藻糖基化、1%至10%海藻糖基化、1%至5%海藻糖基化之間之範圍。The term "low-fucosylation" means that less than 20% of the glycan/glycosidic structure contained in the glycoprotein/containing in the antibody or Fc-fusion protein contains trehalose bound to the terminal-reduced GlcNAc. More preferably, less than 10% or even more preferably less than 5% of all of the composite twig structure glycans contain trehalose bound to the end-reduced GlcNAc of the glycan. Low seawed glycosylation illustrates a range between 0% to 20% trehalylation, 0% to 10% trehalylation, preferably 0% to 5% trehalylation. In particular, low fucosylation illustrates a range between 0.5% to 20% trehalylation, 0.5% to 10% trehalylation, preferably 0.5% to 5% trehalylation. Low seawed glycosylation additionally illustrates a range between 1% to 20% trehalylation, 1% to 10% trehalylation, and 1% to 5% trehalylation.

因此,去海藻糖基化程度係超過80%。此意指超過80%聚糖不含有結合至末端還原GlcNAc之海藻糖。在具體實施例中,超過90%或較佳超過95%聚糖不含有結合至末端還原GlcNAc之海藻糖。因此,去海藻糖基化程度在介於80%至100%、90%至100%、較佳95%至100%之間之範圍內。具體而言,去海藻糖基化程度在介於80%至99.5%、90%至99.5%、較佳95%至99.5%之間之範圍內。較佳地,去海藻糖基化程度在介於80%至99%、90%至99%、較佳95%至99%之間之範圍內。Therefore, the degree of demyosylation is more than 80%. This means that more than 80% of the glycans do not contain trehalose bound to the terminal-reduced GlcNAc. In a particular embodiment, more than 90% or preferably more than 95% of the glycans do not contain trehalose that binds to the terminal-reduced GlcNAc. Thus, the degree of de-cosamination is in the range of between 80% and 100%, between 90% and 100%, preferably between 95% and 100%. Specifically, the degree of demycosylation is in the range of from 80% to 99.5%, from 90% to 99.5%, preferably from 95% to 99.5%. Preferably, the degree of de-cosamination is in the range of between 80% and 99%, between 90% and 99%, preferably between 95% and 99%.

具有「高半乳糖基化」意指超過40%之所有在糖蛋白內含有/在複合型抗體或Fc-融合蛋白含有之聚糖/糖苷結構含有一或兩個連接至核心結構之末端非還原末端處之GlcNAc殘基之半乳糖殘基。更佳地,超過45%或甚至更佳超過50%之所有複合二枝結構型聚糖係半乳糖基化的。高半乳糖基化闡述介於40%至100%半乳糖基化、45%至100%半乳糖基化、較佳50%至100%半乳糖基化或51%至100%半乳糖基化之間之範圍。具體而言,高半乳糖基化闡述介於40%至99.5%半乳糖基化、45%至99.5%半乳糖基化、較佳50%至99.5%半乳糖基化或51%至99.5%半乳糖基化之間之範圍。高半乳糖基化較佳闡述介於40%至99%半乳糖基化、45%至99%半乳糖基化、較佳50%至99%半乳糖基化或51%至99%半乳糖基化之間之範圍。By "high galactosylation" is meant that more than 40% of all glycan/glycosidic structures contained in the glycoprotein/in the complex antibody or Fc-fusion protein contain one or two non-reducing ends linked to the core structure A galactose residue of the GlcNAc residue at the terminus. More preferably, more than 45% or even more preferably more than 50% of all complex dimeric structural glycans are galactosylated. High galactosylation states between 40% to 100% galactosylation, 45% to 100% galactosylation, preferably 50% to 100% galactosylation or 51% to 100% galactosylation. The range between the two. In particular, high galactosylation states between 40% and 99.5% galactosylation, 45% to 99.5% galactosylation, preferably 50% to 99.5% galactosylation or 51% to 99.5% half. The range between lactosylation. High galactosylation is preferably between 40% and 99% galactosylation, 45% to 99% galactosylation, preferably 50% to 99% galactosylation or 51% to 99% galactosyl The scope between the two.

高半乳糖基化較佳闡述在聚糖/糖苷結構中存在至少一個半乳糖殘基(G1)、更佳在聚糖/糖苷結構中存在一或兩個半乳糖殘基(G1或G2)。較佳地,高半乳糖基化意指50%糖苷結構含有至少一個半乳糖殘基。具體而言,高半乳糖基化意指存在G1或G2糖苷結構,但幾乎沒有G0糖苷結構。High galactosylation preferably illustrates the presence of at least one galactose residue (G1) in the glycan/glycosidic structure, more preferably one or two galactose residues (G1 or G2) in the glycan/glycosidic structure. Preferably, high galactosylation means that the 50% glycoside structure contains at least one galactose residue. In particular, high galactosylation means the presence of a G1 or G2 glycoside structure, but almost no G0 glycoside structure.

因此,去半乳糖基化程度小於60%。此意指所有複合型聚糖中60%以下不含有與核心結構之末端非還原端處之GlcNAc連接之殘基。在具體實施例中,所有複合型聚糖55%以下或較佳50%或49%以下不含有與核心結構之末端非還原端處之GlcNAc連接之半乳糖殘基。因此,去半乳糖基化程度在介於60%至0%、55%至0%、較佳50%至0%或49%至0%之間之範圍內。具體而言,去半乳糖基化程度在介於60%至0.5%、55%至0.5%、較佳50%至0.5%或49%至0.5%之間之範圍內。去半乳糖基化程度較佳在介於60%至1%、55%至1%、較佳50%至1%或49%至1%之間之範圍內。Therefore, the degree of degalactosylation is less than 60%. This means that less than 60% of all complex glycans do not contain residues linked to GlcNAc at the non-reducing end of the core structure. In a particular embodiment, less than 55% or preferably 50% or less of all complex glycans do not contain a galactose residue attached to the GlcNAc at the non-reducing end of the core structure. Thus, the degree of degalactosylation is in the range of between 60% and 0%, between 55% and 0%, preferably between 50% and 0% or between 49% and 0%. Specifically, the degree of degalactosylation is in the range of from 60% to 0.5%, from 55% to 0.5%, preferably from 50% to 0.5% or from 49% to 0.5%. The degree of degalactosylation is preferably in the range of from 60% to 1%, from 55% to 1%, preferably from 50% to 1% or from 49% to 1%.

具有高唾液酸化或將唾液酸或神經胺酸殘基添加至半乳糖基化糖苷結構(例如二枝結構聚糖)上之高活性意指超過5%之所有聚糖含有末端唾液酸殘基。更佳地,5-10%或0-8%或1-8%之所有Fc-聚糖含有唾液酸或神經胺酸殘基。在具體實施例中,超過10%或10%-50%或10%-45%之所有Fc-聚糖係唾液酸化的。因此,聚糖不含有末端唾液酸殘基之程度小於95%,或其在介於95-90%或92-100%之間之範圍內。在具體實施例中,聚糖不含有末端唾液酸殘基之程度小於90%,或其在介於50%-90%或55%-90%之間之範圍內。High activity with high sialylation or addition of a sialic acid or a neuratic acid residue to a galactosylated glycoside structure (e.g., a two-structured glycan) means that more than 5% of all glycans contain terminal sialic acid residues. More preferably, 5-10% or 0-8% or 1-8% of all Fc-glycans contain sialic acid or ceramide residues. In a particular embodiment, more than 10% or 10%-50% or 10%-45% of all Fc-glycans are sialylated. Thus, the glycan does not contain terminal sialic acid residues to a degree less than 95%, or it is in the range of between 95-90% or 92-100%. In a particular embodiment, the glycan does not contain terminal sialic acid residues to a degree less than 90%, or it is in the range of between 50% and 90% or between 55% and 90%.

使用H4-II-E細胞作為「宿主細胞」用於產生重組糖蛋白、尤其抗體或Fc-融合蛋白係本發明之標的物。H4-II-E細胞並非標準宿主細胞。The use of H4-II-E cells as "host cells" for the production of recombinant glycoproteins, particularly antibodies or Fc-fusion proteins, is the subject of the present invention. H4-II-E cells are not standard host cells.

用於產生生物醫藥蛋白之標準「宿主細胞」或常用宿主細胞在本發明含義內係(例如)BHK21、BHK TK-、CHO、CHO-K1、CHO-DUKX、CHO-DUKX B1、CHO-DG44、鼠類骨髓瘤細胞、較佳NS0及Sp2/0細胞或此細胞系中任一者之衍生物/子代。尤佳標準宿主細胞係CHO-DG44、CHO-DUKX、CHO-K1及BHK21及甚至更佳CHO-DG44及CHO-DUKX細胞。最佳標準宿主細胞係CHO-DG44細胞。鼠類及倉鼠細胞之實例亦匯總於表1中。然而,彼等細胞之衍生物/子代、其他哺乳動物細胞(包括(但不限於)人類、小鼠、大鼠、猴及鳥類或較佳齧齒類動物細胞系)或真核細胞(包括(但不限於)酵母、昆蟲及植物細胞)亦用作標準宿主細胞、尤其用於產生生物醫藥蛋白。通常,該等細胞能夠將大量所關注特定糖蛋白表現並分泌至培養基中。Standard "host cells" or common host cells for the production of biopharmaceutical proteins are within the meaning of the present invention (for example) BHK21, BHK TK-, CHO, CHO-K1, CHO-DUKX, CHO-DUKX B1, CHO-DG44, Murine myeloma cells, preferably NS0 and Sp2/0 cells or derivatives/progeny of any of these cell lines. The standard host cell lines are CHO-DG44, CHO-DUKX, CHO-K1 and BHK21 and even better CHO-DG44 and CHO-DUKX cells. The best standard host cell line is CHO-DG44 cells. Examples of rodent and hamster cells are also summarized in Table 1. However, derivatives/progeny of their cells, other mammalian cells (including but not limited to human, mouse, rat, monkey and avian or preferred rodent cell lines) or eukaryotic cells (including However, it is not limited to yeast, insects and plant cells) and is also used as a standard host cell, especially for the production of biomedical proteins. Typically, such cells are capable of expressing and secreting a large number of specific glycoproteins of interest into the culture medium.

表1:真核標準宿主細胞/常用生產細胞系Table 1: Eukaryotic Standard Host Cells/Common Production Cell Lines

一般而言,當在無血清條件下且視情況在不含有任一動物來源之蛋白/肽之培養基中建立、適應且完全培育時,宿主細胞係最佳的。諸如以下等市售培養基係實例性適當營養溶液:Ham's F12(Sigma,Deisenhofen,Germany)、RPMI-1640(Sigma)、杜貝克氏改良鷹氏培養基(Dulbecco's Modified Eagle's Medium)(DMEM;Sigma)、最低必需培養基(MEM;Sigma)、伊思柯夫氏改良杜貝克氏培養基(Iscove's Modified Dulbecco's Medium)(IMDM;Sigma)、CD-CHO(Invitrogen,Carlsbad,CA),CHO-S-Invitrogen)、無血清CHO培養基(Sigma)及無蛋白CHO培養基(Sigma)、EX-CELL培養基(SAFC)、CDM4CHO及SFM4CHO(HyClone)。任一培養基皆可視需要補充多種化合物,該等化合物之實例係激素及/或其他生長因子(例如胰島素、轉鐵蛋白、表皮生長因子、胰島素樣生長因子)、鹽(例如氯化鈉、鈣鹽、鎂鹽、磷酸鹽)、緩衝劑(例如HEPES)、核苷(例如腺苷、胸苷)、麩醯胺酸、葡萄糖或其他等效能源、抗生素、微量元素。亦可以彼等熟習此項技術者應已知之適當濃度包括任何其他必需補充物。在本發明中較佳使用無血清培養基,但補充有適宜量之血清之培養基亦可用於培育H4-II-E細胞並用於生長及選擇穩定產生細胞。對於表現所選基因之遺傳修飾細胞之生長及選擇而言,將適宜選擇劑添加至培養基中。In general, host cell lines are optimal when established, adapted, and fully cultured under serum-free conditions and optionally in a medium that does not contain any animal-derived protein/peptide. Commercially available media such as the following are exemplary suitable nutrient solutions: Ham's F12 (Sigma, Deisenhofen, Germany), RPMI-1640 (Sigma), Dulbecco's Modified Eagle's Medium (DMEM; Sigma), lowest Essential medium (MEM; Sigma), Iscove's Modified Dulbecco's Medium (IMDM; Sigma), CD-CHO (Invitrogen, Carlsbad, CA), CHO-S-Invitrogen), serum-free CHO medium (Sigma) and protein-free CHO medium (Sigma), EX-CELL medium (SAFC), CDM4 CHO and SFM4 CHO (HyClone). Any medium may be supplemented with various compounds as needed. Examples of such compounds are hormones and/or other growth factors (eg, insulin, transferrin, epidermal growth factor, insulin-like growth factor), salts (eg, sodium chloride, calcium salts). , magnesium salts, phosphates), buffers (eg HEPES), nucleosides (eg adenosine, thymidine), glutamic acid, glucose or other equivalent energy sources, antibiotics, trace elements. They may also include any other necessary supplements at an appropriate concentration known to those skilled in the art. Preferably, serum-free medium is used in the present invention, but a medium supplemented with a suitable amount of serum can also be used to grow H4-II-E cells and to grow and select stably producing cells. For selection and selection of genetically modified cells expressing the selected gene, a suitable selection agent is added to the culture medium.

為使生長於含血清培養基中之細胞適應於無血清生長並在單細胞懸浮培養物中培養細胞,使用無血清培養基。為促進懸浮生長,無血清且無鈣離子或鈣減少之培養基較佳。根據本發明,使H4-II-E細胞適應無血清且Ca降低之培養基且隨後在該等培養基中懸浮培養該等細胞。使H4-II-E細胞適應懸浮及無血清培養基且在該等培養基中懸浮培養該等細胞之方法包含使用無血清且無鈣離子(Ca2+)或鈣減少之培養基。術語鈣減少或較佳無鈣培養基意指限定含有1 μmol/L-500 μmol/L Ca2+離子之培養基,更佳地鈣減少培養基含有1 μmol/L-250 μmol/L Ca2+離子,且甚至更佳地鈣減少或無Ca培養基含有0 μmol/L-100 μmol/L或0.5 μmol/L-100 μmol/L Ca2+離子。市售無鈣培養基之實例係MEM Joklik Modification(Sigma)或MEM Spinner Modification(Sigma)。用於在血清存在下貼壁培育H4-II-E細胞之典型含鈣培養基係含有1360 μmol/L Ca2+離子之鷹氏最低必需培養基(Sigma)。To adapt cells grown in serum-containing medium to serum-free growth and to culture cells in single cell suspension culture, serum-free medium is used. To promote suspension growth, a serum-free medium free of calcium ions or calcium is preferred. According to the present invention, H4-II-E cells are adapted to a serum-free and Ca-reduced medium and then the cells are cultured in suspension in the medium. The method of adapting H4-II-E cells to suspension and serum-free medium and suspending the cells in such medium comprises using a serum-free medium without calcium ion (Ca 2+ ) or calcium reduction. The term calcium reduction or preferably calcium-free medium means a medium containing 1 μmol/L to 500 μmol/L Ca 2+ ions, and more preferably a calcium reduction medium containing 1 μmol/L to 250 μmol/L Ca 2+ ions. And even better calcium-reduced or Ca-free medium containing 0 μmol/L-100 μmol/L or 0.5 μmol/L-100 μmol/L Ca 2+ ions. An example of a commercially available calcium-free medium is MEM Joklik Modification (Sigma) or MEM Spinner Modification (Sigma). A typical calcium-containing medium for adherent incubation of H4-II-E cells in the presence of serum is Eagle's Minimum Essential Medium (Sigma) containing 1360 μmol/L Ca 2+ ions.

本發明之術語「宿主細胞」涵蓋包含異源核酸序列之細胞以及不(尚未)包含異源核酸序列之細胞。在本發明之具體實施例中,該宿主細胞包含異源核酸序列,其編碼該宿主細胞表現之(重組)蛋白、較佳所關注糖蛋白,例如抗體或Fc-融合蛋白。為此,在允許表現該(重組)蛋白之條件下培育該宿主細胞。The term "host cell" as used in the present invention encompasses cells comprising a heterologous nucleic acid sequence and cells which do not (not yet) comprise a heterologous nucleic acid sequence. In a particular embodiment of the invention, the host cell comprises a heterologous nucleic acid sequence encoding a (recombinant) protein, preferably a glycoprotein of interest, such as an antibody or Fc-fusion protein, expressed by the host cell. To this end, the host cell is cultured under conditions permitting expression of the (recombinant) protein.

術語「蛋白」可與胺基酸殘基序列或多肽互換使用且係指任一長度之胺基酸聚合物。該等術語亦包括經由包括(但不限於)糖基化、乙醯化、磷酸化或蛋白加工在內之反應進行轉譯後修飾的蛋白。可在多肽結構內進行修飾及改變,例如與其他蛋白融合、胺基酸序列取代、缺失或插入,同時分子維持其生物功能活性。舉例而言,可在多肽或其潛在核酸編碼序列內進行某些胺基酸序列取代且可獲得具有相同性質之蛋白。The term "protein" is used interchangeably with an amino acid residue sequence or polypeptide and refers to an amino acid polymer of any length. The terms also include proteins that are post-translationally modified via reactions including, but not limited to, glycosylation, acetylation, phosphorylation, or protein processing. Modifications and alterations can be made within the structure of the polypeptide, such as fusion with other proteins, amino acid sequence substitutions, deletions or insertions, while the molecule maintains its biological functional activity. For example, certain amino acid sequence substitutions can be made within a polypeptide or its potential nucleic acid coding sequence and a protein of the same nature can be obtained.

術語「多肽」意指具有超過10個胺基酸之序列且術語「肽」意指長度為最多10個胺基酸之序列。The term "polypeptide" means a sequence having more than 10 amino acids and the term "peptide" means a sequence of up to 10 amino acids in length.

術語「核酸序列」、「所關注基因」(GOI)、「所選序列」或「產物基因」在本文中具有相同含義且係指編碼「所關注產物」或「所關注蛋白」(亦由術語「期望產物」提及)之任一長度之多核苷酸序列。在本發明較佳實施例中,所關注核酸序列或基因編碼糖蛋白、較佳抗體或Fc融合蛋白。所關注核酸序列或基因可係全長或截短基因、融合基因或加標籤基因,且可係cDNA、基因組DNA或DNA片段,較佳為cDNA。其可係天然序列,即天然形式,或可視需要經突變或以其他方式修飾。該等修飾包括優化密碼子在所選宿主細胞中之使用之密碼子優化、人類化或加標籤。所選序列可編碼經分泌多肽、細胞質多肽、核多肽、膜結合多肽或細胞表面多肽。The terms "nucleic acid sequence", "gene of interest" (GOI), "selected sequence" or "product gene" have the same meaning herein and refer to the code "product of interest" or "protein of interest" (also by terminology) The "desired product" refers to a polynucleotide sequence of any length. In a preferred embodiment of the invention, the nucleic acid sequence or gene of interest encodes a glycoprotein, preferably an antibody or an Fc fusion protein. The nucleic acid sequence or gene of interest may be a full length or truncated gene, a fusion gene or a tagged gene, and may be a cDNA, genomic DNA or DNA fragment, preferably a cDNA. It may be in the native sequence, ie in its native form, or may be mutated or otherwise modified as desired. Such modifications include codon optimization, humanization or tagging of the use of optimized codons in the host cell of choice. The selected sequence may encode a secreted polypeptide, a cytoplasmic polypeptide, a nuclear polypeptide, a membrane-bound polypeptide, or a cell surface polypeptide.

「所關注糖蛋白」包括全部可在H4-II-E宿主細胞中表現之蛋白、多肽、其片段、肽。期望蛋白可係(例如)抗體、酶、細胞因子、淋巴因子、黏附分子、受體及其衍生物或片段、及可用作激動劑或拮抗劑及/或具有治療或診斷用途之任何其他多肽。期望蛋白/多肽之實例亦可參見下文。The "glycoprotein of interest" includes all proteins, polypeptides, fragments thereof, and peptides which can be expressed in H4-II-E host cells. The desired protein may be, for example, an antibody, an enzyme, a cytokine, a lymphokine, an adhesion molecule, a receptor and derivatives or fragments thereof, and any other polypeptide useful as an agonist or antagonist and/or for therapeutic or diagnostic use. . Examples of desired proteins/polypeptides can also be found below.

在更複雜分子(例如單株抗體)之情形下,所關注基因編碼兩條抗體鏈中之一或二者。In the case of more complex molecules (eg, monoclonal antibodies), the gene of interest encodes one or both of the two antibody chains.

術語「抗體」係指由一或多種實質上由免疫球蛋白基因編碼之多肽組成之蛋白。所識別免疫球蛋白基因包括κ、λ、α、γ、δ、ε及μ恆定區基因以及無數免疫球蛋白可變區基因。The term "antibody" refers to a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes as well as myriad immunoglobulin variable region genes.

本文所用術語「抗體」包括多株抗體、單株抗體、雙特異性抗體、多特異性抗體、人類抗體、人類化抗體或嵌合抗體。The term "antibody" as used herein includes a plurality of antibodies, monoclonal antibodies, bispecific antibodies, multispecific antibodies, human antibodies, humanized antibodies or chimeric antibodies.

術語「抗體」及「免疫球蛋白」可互換使用且用於表示具有上文針對免疫球蛋白所述之結構特性的糖蛋白。The terms "antibody" and "immunoglobulin" are used interchangeably and are used to denote a glycoprotein having the structural properties described above for an immunoglobulin.

術語「抗體」係以最廣泛意義使用且具體而言涵蓋單一單株抗體(包括激動劑抗體及拮抗劑抗體)及具有多表位特異性之抗體組合物。術語「抗體」具體而言涵蓋單株抗體(包括全長單株抗體)、多株抗體、多特異性抗體(例如雙特異性抗體)及抗體片段,只要該等抗體及片段含有或經修飾成含有免疫球蛋白重鏈恆定區之CH2結構域中至少包含N-連接糖基化位點之部分即可。實例性抗體在本發明範圍內包括(但不限於)抗CD20、抗CD33、抗CD37、抗CD40、抗CD44、抗CD52、抗HER2/neu(erbB2)、抗EGFR、抗IGF、抗VEGF、抗TNFα、抗IL2或抗IgE抗體。The term "antibody" is used in the broadest sense and specifically encompasses single monoclonal antibodies (including agonist antibodies and antagonist antibodies) and antibody compositions having multi-epitope specificity. The term "antibody" specifically encompasses monoclonal antibodies (including full-length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (eg, bispecific antibodies), and antibody fragments, as long as the antibodies and fragments contain or are modified to contain The CH2 domain of the immunoglobulin heavy chain constant region may comprise at least a portion of the N-linked glycosylation site. Exemplary antibodies within the scope of the invention include, but are not limited to, anti-CD20, anti-CD33, anti-CD37, anti-CD40, anti-CD44, anti-CD52, anti-HER2/neu (erbB2), anti-EGFR, anti-IGF, anti-VEGF, anti- TNFα, anti-IL2 or anti-IgE antibodies.

本文所用術語「單株抗體」(mAb)係指自一群基於胺基酸序列實質上同源之抗體獲得之抗體。單株抗體具有高度特異性,其針對單一抗原性位點。此外,與通常包括針對不同決定簇(表位)之不同抗體之習用(多株)抗體製劑不同,每一mAb針對抗原上之單一決定簇。除特異性外,mAb係有利之原因亦在於其可藉由未被其他免疫球蛋白污染之細胞培養物(融合瘤、重組細胞或諸如此類)合成。mAb在本文中包括嵌合、人類化及人類抗體。The term "monoclonal antibody" (mAb) as used herein refers to an antibody obtained from a population of antibodies that are substantially homologous based on amino acid sequences. Individual antibodies are highly specific and target a single antigenic site. Furthermore, unlike conventional (multi-drug) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each mAb is directed against a single determinant on the antigen. In addition to specificity, mAbs are advantageous because they can be synthesized by cell cultures (fusion tumors, recombinant cells, or the like) that are not contaminated with other immunoglobulins. mAbs include chimeric, humanized, and human antibodies herein.

「嵌合抗體」係其輕鏈及/或重鏈基因已通常藉由遺傳改造自免疫球蛋白可變區及恆定區構築之抗體,該等區域與不同物種(例如小鼠及人類)之對應序列相同或同源。或另一選擇為,其重鏈基因屬於特定抗體類別或子類,而該鏈之其餘部分來自相同物種之另一抗體類別或子類或來自其他物種。其亦涵蓋此等抗體之片段,只要該等片段含有或經修飾成含有至少一個CH2結構域即可。舉例而言,來自小鼠單株抗體之基因之可變區段可接合至人類恆定區段,例如γ1及γ3。因此,典型治療嵌合抗體係由來自小鼠抗體之可變結構域或抗原結合結構域及來自人類抗體之恆定結構域或效應子結構域構成的雜合蛋白(例如ATCC登錄編號CRL 9688分泌抗Tac嵌合抗體),但可使用其他哺乳動物物種。A "chimeric antibody" is an antibody whose light chain and/or heavy chain genes have been normally engineered by genetically engineering variable regions and constant regions of immunoglobulins, corresponding to different species (eg, mice and humans). The sequences are identical or homologous. Or alternatively, the heavy chain gene belongs to a particular antibody class or subclass, and the remainder of the chain is from another antibody class or subclass of the same species or from another species. It also encompasses fragments of such antibodies as long as the fragments contain or are modified to contain at least one CH2 domain. For example, a variable segment of a gene from a mouse monoclonal antibody can be ligated into a human constant segment, such as gamma 1 and gamma 3 . Thus, a typical therapeutic chimeric anti-system consists of a hybrid protein from a variable domain or antigen-binding domain of a mouse antibody and a constant or effector domain derived from a human antibody (eg, ATCC Accession No. CRL 9688). Tac chimeric antibodies), but other mammalian species can be used.

本發明之術語「人類化抗體」係指特異性嵌合抗體、免疫球蛋白鏈或其片段(例如Fv、Fab、Fab'、F(ab)2或抗體之其他抗原結合子序列),只要該等抗體或片段含有或經修飾成含有免疫球蛋白重鏈恆定區之CH2結構域中至少包含N-連接糖基化位點之部分,且含有源自非人類免疫球蛋白之最少序列即可。在極大程度上,人類化抗體係人類免疫球蛋白(接受者抗體),其中來自接受者之互補決定區(CDR)的殘基由來自諸如小鼠、大鼠或兔等非人類物種(供體抗體)之CDR且具有期望特異性、親和力及能力的殘基所替代。在一些情況下,人類免疫球蛋白之Fv框架殘基由對應非人類殘基所替代。此外,人類化抗體可包含在接受者抗體與引入之CDR或框架序列中皆未發現之殘基。實施該等修飾來進一步改善並最大化抗體性能。一般而言,人類化抗體將包含實質上所有至少一個且通常兩個可變結構域,其中所有或實質上所有CDR區對應於非人類免疫球蛋之彼等,且所有或實質上所有區為人類免疫球蛋白共有序列之彼等。人類化抗體最佳亦應包含至少一部分免疫球蛋白恆定區(通常為人類免疫球蛋白恆定區)。The term "humanized antibody" as used in the present invention refers to a specific chimeric antibody, immunoglobulin chain or a fragment thereof (for example, Fv, Fab, Fab', F(ab)2 or other antigen-binding sequence of an antibody), as long as the The antibody or fragment contains or is modified to contain a portion of the CH2 domain comprising the constant region of the immunoglobulin heavy chain comprising at least an N-linked glycosylation site and contains minimal sequence derived from a non-human immunoglobulin. To a large extent, humanized anti-system human immunoglobulins (recipient antibodies) in which residues from the complementarity determining regions (CDRs) of the recipient are derived from non-human species such as mice, rats or rabbits (donors) The CDRs of the antibody) are replaced by residues with the desired specificity, affinity and ability. In some cases, the Fv framework residues of human immunoglobulins are replaced by corresponding non-human residues. Furthermore, humanized antibodies can comprise residues that are not found in the recipient antibody and the introduced CDR or framework sequences. These modifications are implemented to further improve and maximize antibody performance. In general, a humanized antibody will comprise substantially all of at least one and usually two variable domains, wherein all or substantially all of the CDR regions correspond to non-human immunoglobulins, and all or substantially all regions are These are the consensus sequences of human immunoglobulins. The humanized antibody should preferably also contain at least a portion of the immunoglobulin constant region (typically a human immunoglobulin constant region).

人類化抗體:包含人類框架區及一或多個來自非人類(通常為小鼠或大鼠)抗體之CDR。可能需要對框架胺基酸進行調節以保持結構域之抗原結合特異性、親和力及或結構。Humanized antibodies: comprise human framework regions and one or more CDRs from non-human (usually mouse or rat) antibodies. It may be desirable to modulate the framework amino acid to maintain the antigen binding specificity, affinity and or structure of the domain.

本發明之術語「CH2結構域」意欲描述免疫球蛋白重鏈恆定區中包含N-連接糖基化位點之CH2結構域。在定義免疫球蛋白CH2結構域時,一般而言可提及免疫球蛋白且特定而言可提及如由Kabat,E.A.應用於人類IgG1之免疫球蛋白之結構域結構(Kabat,1988;Kabat等人,1991)。因此,免疫球蛋白通常為約150 kDa之異四聚體糖蛋白,由兩條相同輕鏈及兩條相同重鏈構成。每一輕鏈由一共價二硫鍵連接至重鏈上,而在不同免疫球蛋白同種型之重鏈之間二硫連接之數目會有變化。每一重鏈及輕鏈亦具有規則間隔之鏈內二硫橋鍵。每一重鏈具有胺基末端可變結構域(VH)及之後的羧基末端恆定結構域(CH)。每一輕鏈具有可變N-末端結構域(VL)及C-末端恆定結構域(CL)。The term "CH2 domain" of the present invention is intended to describe the CH2 domain comprising an N-linked glycosylation site in the immunoglobulin heavy chain constant region. In the definition of immunoglobulin CH2 domain, immunoglobulins may generally be mentioned and, in particular, the domain structure of immunoglobulins as applied to human IgG1 by Kabat, EA may be mentioned (Kabat, 1988; Kabat et al. People, 1991). Thus, an immunoglobulin is typically a heterotetrameric glycoprotein of about 150 kDa, composed of two identical light chains and two identical heavy chains. Each light chain is linked to the heavy chain by a covalent disulfide bond, and the number of disulfide linkages between the heavy chains of different immunoglobulin isotypes varies. Each heavy and light chain also has a regularly spaced intrachain disulfide bridge. Each heavy chain has an amino terminal variable domain (VH) followed by a carboxy terminal constant domain (CH). Each light chain has a variable N-terminal domain (VL) and a C-terminal constant domain (CL).

端視重鏈中恆定結構域之胺基酸序列,可將抗體歸類為不同類別。存在5個主要類別:IgA、IgD、IgE、IgG及IgM。對應於不同類別之抗體重鏈恆定結構域分別稱為α、δ、ε、γ及μ結構域。IgM之μ鏈含有5個結構域(VH、CHmu1、CHmu2、CHmu3及CHmu4)。IgE之重鏈亦含有5個結構域,而IgA之重鏈具有4個結構域。免疫球蛋白類別可進一步分成亞類(同種型),例如IgG1、IgG2、IgG3、IgG4、IgA1及IgA2。The amino acid sequence of the constant domain in the heavy chain can be classified into different classes. There are five main categories: IgA, IgD, IgE, IgG, and IgM. The heavy chain constant domains corresponding to different classes of antibodies are referred to as the alpha, delta, epsilon, gamma and mu domains, respectively. The μ chain of IgM contains five domains (VH, CHmu1, CHmu2, CHmu3, and CHmu4). The heavy chain of IgE also contains five domains, while the heavy chain of IgA has four domains. The immunoglobulin class can be further divided into subclasses (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2.

不同類別免疫球蛋白之亞單位結構及三維構型已為吾人所熟知。其中IgA及IgM為聚合物且每一亞單位含有兩條輕鏈及兩條重鏈。IgG之重鏈含有一定長度之位於CHγ1與CHγ2結構域之間之多肽鏈(稱作鉸鏈區)。IgA之α鏈具有含有O-連接糖基化位點之鉸鏈區且μ鏈及ε鏈不具有與γ鏈及α鏈之鉸鏈區類似之序列,然而,其含有4個其他鏈中缺乏之恆定結構域。The subunit structure and three-dimensional configuration of different classes of immunoglobulins are well known. Wherein IgA and IgM are polymers and each subunit contains two light chains and two heavy chains. The heavy chain of IgG contains a length of polypeptide chain (referred to as the hinge region) between the CHγ1 and CHγ2 domains. The α chain of IgA has a hinge region containing an O-linked glycosylation site and the μ chain and the ε chain do not have a sequence similar to the hinge region of the γ chain and the α chain, however, it contains a constant lack of 4 other chains. Domain.

因此,CH2結構域係免疫球蛋白重鏈恆定區結構域。完整抗體之Fc區通常包含兩個CH2結構域及兩個CH3結構域。根據本發明,CH2結構域較佳為上述5個免疫球蛋白類別中一者之CH2結構域。較佳為哺乳動物免疫球蛋白CH2結構域,例如靈長類動物或鼠類免疫球蛋白,其中靈長類動物且尤其人類免疫球蛋白CH2結構域較佳。熟習此項技術者已知且通常可獲得免疫球蛋白CH2結構域之胺基酸序列(Kabat等人,1991)。在本發明上下文中,較佳免疫球蛋白CH2結構域係人類IgG且較佳來自IgG1、IgG2、IgG3、IgG4、更佳人類IgG1及IgG3且甚至更佳人類IgG1。使用Edelman編號系統(Edelman等人,1969)時,免疫球蛋白CH2結構域較佳始於相當於人類IgG1之麩醯胺酸233之胺基酸位置且延伸至相當於離胺酸340之胺基酸(Ellison及Hood,1982)。Thus, the CH2 domain is an immunoglobulin heavy chain constant region domain. The Fc region of an intact antibody typically comprises two CH2 domains and two CH3 domains. According to the invention, the CH2 domain is preferably the CH2 domain of one of the five immunoglobulin classes described above. Preferred are mammalian immunoglobulin CH2 domains, such as primate or murine immunoglobulins, of which primate and especially human immunoglobulin CH2 domains are preferred. Amino acid sequences of the immunoglobulin CH2 domain are known and commonly available to those skilled in the art (Kabat et al., 1991). In the context of the present invention, it is preferred that the immunoglobulin CH2 domain is human IgG and preferably from IgGl, IgG2, IgG3, IgG4, better human IgGl and IgG3 and even better human IgGl. When using the Edelman numbering system (Edelman et al., 1969), the immunoglobulin CH2 domain preferably begins at the amino acid position corresponding to the glutamic acid 233 of human IgG1 and extends to an amine group equivalent to the amine acid 340. Acid (Ellison and Hood, 1982).

就人類抗體分子而言,可提及N-連接寡糖附著至β-4中Asn297處之醯胺側鏈之IgG類別,該醯胺側鏈向Fc區之CH2結構域之內面彎曲。本發明之糖蛋白、尤其抗體或Fc-融合蛋白之特徵在於其含有或經修飾成含有至少一個CH2結構域。CH2結構域係具有人類IgG CH2結構域之單一N-連接寡糖之免疫球蛋白之CH2結構域。CH2結構域較佳為人類IgG1之CH2結構域。As far as human antibody molecules are concerned, mention may be made of the attachment of an N-linked oligosaccharide to the IgG class of the indoleamine side chain at Asn297 in β-4, which is bent toward the inner face of the CH2 domain of the Fc region. A glycoprotein, especially an antibody or Fc-fusion protein of the invention is characterized in that it contains or is modified to contain at least one CH2 domain. The CH2 domain is the CH2 domain of a single N-linked oligosaccharide immunoglobulin with a human IgG CH2 domain. The CH2 domain is preferably the CH2 domain of human IgG1.

「所關注糖蛋白」、「所關注多肽」、「所關注蛋白」或「所關注產物」係彼等上文所提及者且包括全部可在H4-II-E宿主細胞中表現之抗體或Fc-融合蛋白。此外,所關注期望蛋白或糖蛋白可為(例如)酶、細胞因子、淋巴因子、黏附分子、受體及其衍生物或片段、及可用作激動劑或拮抗劑及/或具有治療或診斷用途之任何其他多肽,其係糖基化的。具體而言,所關注期望糖蛋白/多肽或蛋白係(例如但不限於)胰島素;胰島素樣生長因子;hGH;tPA;細胞因子,例如白介素(IL)(例如IL-1、IL-2、IL-3、IL-4、IL-5、IL-6、IL-7、IL-8、IL-9、IL-10、IL-11、IL-12、IL-13、IL-14、IL-15、IL-16、IL-17、IL-18)、干擾素(IFN)α、IFNβ、IFNγ、IFNω或IFNτ、腫瘤壞死因子(TNF)(例如TNFα及TNFβ、TNFγ、TRAIL)、G-CSF、GM-CSF、M-CSF、MCP-1、VEGF;及奈米體(nanobody)。亦包括產生紅血球生成素或任何其他激素生長因子及可用作激動劑或拮抗劑及/或具有治療或診斷用途之任何其他多肽。本發明H4-II-E細胞可有利地用於產生諸如以下等抗體:單株、多株、多特異性抗體或其片段,該等片段包含CH2結構域、Fc及Fc'片段、免疫球蛋白重鏈及輕鏈及其恆定片段。此外,該產生本發明(重組)糖蛋白之方法可有利地用於產生諸如以下等抗體:單株、多株、多特異性抗體或其片段,該等片段包含CH2結構域、Fc及Fc'片段、免疫球蛋白重鏈及輕鏈及其恆定片段以及Fc-融合蛋白。"Glycoprotein of interest", "polypeptide of interest", "protein of interest" or "product of interest" are those mentioned above and include all antibodies which are expressed in H4-II-E host cells or Fc-fusion protein. Furthermore, the desired protein or glycoprotein of interest may be, for example, an enzyme, a cytokine, a lymphokine, an adhesion molecule, a receptor and derivatives or fragments thereof, and may be used as an agonist or antagonist and/or have a therapeutic or diagnostic Any other polypeptide of use that is glycosylated. In particular, the desired glycoprotein/polypeptide or protein line of interest (such as, but not limited to) insulin; insulin-like growth factor; hGH; tPA; cytokines, such as interleukin (IL) (eg, IL-1, IL-2, IL) -3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15 , IL-16, IL-17, IL-18), interferon (IFN)α, IFNβ, IFNγ, IFNω or IFNτ, tumor necrosis factor (TNF) (eg TNFα and TNFβ, TNFγ, TRAIL), G-CSF, GM-CSF, M-CSF, MCP-1, VEGF; and nanobody. Also included are erythropoietin or any other hormone growth factor and any other polypeptide useful as an agonist or antagonist and/or for therapeutic or diagnostic use. The H4-II-E cells of the invention can be advantageously used to produce antibodies such as: single, multiple, multispecific antibodies or fragments thereof, which comprise a CH2 domain, an Fc and an Fc' fragment, an immunoglobulin Heavy and light chains and their constant fragments. Furthermore, the method of producing a (recombinant) glycoprotein of the present invention can be advantageously used to produce antibodies such as: single, multiple, multispecific antibodies or fragments thereof, which include a CH2 domain, an Fc and an Fc' Fragments, immunoglobulin heavy and light chains and constant fragments thereof, and Fc-fusion proteins.

「Fc-融合蛋白」定義為含有或經修飾成含有免疫球蛋白重鏈恆定區之CH2結構域中至少包含單一N-連接糖基化位點之部分的蛋白。根據Kabat EU命名法(Kabat等人,1991),在IgG1、IgG2、IgG3或IgG4抗體中,此部分係Asn297。An "Fc-fusion protein" is defined as a protein comprising or modified to comprise a portion of a CH2 domain comprising an immunoglobulin heavy chain constant region comprising at least a single N-linked glycosylation site. According to the Kabat EU nomenclature (Kabat et al., 1991), this portion is Asn297 in IgG1, IgG2, IgG3 or IgG4 antibodies.

融合蛋白之另一部分可係天然或經修飾異源蛋白序列之完整序列或任一部分或天然或經修飾異源蛋白序列之完整序列或任一部分之組合物。免疫球蛋白恆定結構域序列可自任一免疫球蛋白亞型(例如IgG1、IgG2、IgG3、IgG4、IgA1或IgA2亞型)或類別(例如IgA、IgE、IgD或IgM)獲得。優先地,其源自人類免疫球蛋白,更佳源自人類IgG且甚至更佳源自人類IgG1及IgG3。Fc融合蛋白之實例包含MCP1-Fc、ICAM-Fc、EPO-Fc、scFv片段或諸如此類,該等片段偶合至免疫球蛋白重鏈恆定區中包含N-連接糖基化位點之CH2結構域。Fc-融合蛋白可藉由遺傳改造方法藉由以下方式來構築:將免疫球蛋白重鏈恆定區中包含N-連接糖基化位點之CH2結構域引入另一包含(例如)其他免疫球蛋白結構域、酶促活性蛋白部分、效應子結構域之表現構築體中。因此,本發明Fc融合蛋白亦包含連接至免疫球蛋白重鏈恆定區中包含N-連接糖基化位點之CH2結構域的單鏈Fv片段。Another portion of the fusion protein can be the entire sequence or any portion of the native or modified heterologous protein sequence or a complete sequence or a combination of any of the native or modified heterologous protein sequences. The immunoglobulin constant domain sequence can be obtained from any immunoglobulin subtype (eg, IgGl, IgG2, IgG3, IgG4, IgA1, or IgA2 subtype) or class (eg, IgA, IgE, IgD, or IgM). Preferentially, it is derived from human immunoglobulins, more preferably from human IgG and even more preferably from human IgGl and IgG3. Examples of Fc fusion proteins include MCP1-Fc, ICAM-Fc, EPO-Fc, scFv fragments, or the like, which are coupled to a CH2 domain comprising an N-linked glycosylation site in the immunoglobulin heavy chain constant region. An Fc-fusion protein can be constructed by genetic engineering by introducing a CH2 domain comprising an N-linked glycosylation site in an immunoglobulin heavy chain constant region into another comprising, for example, other immunoglobulins. The domain, the enzymatically active protein moiety, and the effector domain are expressed in the construct. Thus, an Fc fusion protein of the invention also comprises a single chain Fv fragment linked to a CH2 domain comprising an N-linked glycosylation site in the immunoglobulin heavy chain constant region.

較佳自培養基以分泌多肽形式回收/分離所關注糖蛋白、尤其抗體或Fc-融合蛋白,或若無分泌信號進行表現,則可自宿主細胞溶解物將其回收/分離。需要以可獲得所關注蛋白之實質上同源製劑之方式自其他重組蛋白及宿主細胞蛋白純化所關注糖蛋白、尤其抗體或Fc-融合蛋白。作為第一步驟,藉由(例如)離心或過濾自培養基或溶解物去除細胞及/或微粒細胞碎片。此後,藉由(例如)以下方式自污染性可溶性蛋白、多肽及核酸純化所關注糖蛋白、尤其抗體或Fc-融合蛋白:在免疫親和力或離子交換管柱實施之分級分離、乙醇沉澱、硫酸銨沉澱、反相HPLC、層析聚焦、Sephadex層析、在二氧化矽或陽離子交換樹脂(例如DEAE)實施之層析、凝膠過濾或具體而言蛋白質A親和力層析。一般而言,教示熟習此項技術者如何純化由宿主細胞表現之異源蛋白的方法為業內所熟知。Preferably, the glycoprotein, particularly the antibody or Fc-fusion protein, is recovered/isolated in the form of a secreted polypeptide from the culture medium, or it can be recovered/separated from the host cell lysate if it is expressed without a secretion signal. It is desirable to purify the glycoprotein of interest, particularly an antibody or Fc-fusion protein, from other recombinant proteins and host cell proteins in a manner that results in a substantially homologous preparation of the protein of interest. As a first step, cells and/or particulate cell debris are removed from the culture medium or lysate by, for example, centrifugation or filtration. Thereafter, the glycoprotein of interest, in particular the antibody or Fc-fusion protein, is purified from contaminating soluble proteins, polypeptides and nucleic acids by, for example, fractionation, ethanol precipitation, ammonium sulfate in immunoaffinity or ion exchange columns. Precipitation, reverse phase HPLC, chromatofocusing, Sephadex chromatography, chromatography on cerium oxide or cation exchange resin (eg DEAE), gel filtration or, in particular, protein A affinity chromatography. In general, it is well known in the art to teach those skilled in the art how to purify heterologous proteins expressed by host cells.

根據定義,將引入宿主細胞中之任何序列或基因稱為宿主細胞之「異源序列」或「異源基因」或「轉基因」,即使所引入序列或基因與宿主細胞中之內源性序列或基因相同。因此,「異源」蛋白係自異源序列表現之蛋白。在本發明通篇中、尤其在蛋白表現之情形下,術語「重組」可與術語「異源」互換使用。因此,「重組」蛋白係自異源序列表現之蛋白。By definition, any sequence or gene introduced into a host cell is referred to as a "heterologous sequence" or "heterologous gene" or "transgene" of the host cell, even if the introduced sequence or gene is associated with an endogenous sequence in the host cell or The genes are the same. Thus, a "heterologous" protein is a protein expressed from a heterologous sequence. In the context of the present invention, particularly in the context of protein expression, the term "recombinant" is used interchangeably with the term "heterologous". Thus, a "recombinant" protein is a protein expressed from a heterologous sequence.

可藉由使用「表現載體」、較佳真核生物且甚至更佳哺乳動物表現載體將異源基因序列引入靶細胞中。用於構築載體之方法為熟習此項技術者所熟知且闡述於各種出版物中。特定而言,用於構築適宜載體之技術(包括對功能組份(例如啟動子、增強子、終止信號及聚腺苷酸化信號、選擇標記物、複製起點及剪接信號)之說明)相當詳細地概述於(Sambrook等人,1989)及其中引用之參考文獻中。載體可包括(但不限於)質粒載體、噬菌粒、黏粒、人工/微型染色體(例如ACE)或病毒載體(例如杆狀病毒、逆轉錄病毒、腺病毒、腺相關病毒、單純皰疹病毒、逆轉錄病毒、噬菌體)。真核表現載體通常亦將含有促進載體在細菌中增殖之原核序列,例如複製起點及用於在細菌中進行選擇之抗生素抗性基因。含有多核苷酸可操作地連接之選殖位點的多種真核表現載體為業內所熟知,且一些可購自諸如以下等公司:Stratagene,La Jolla,CA;Invitrogen,Carlsbad,CA;Promega,Madison,WI或BD Biosciences Clontech,Palo Alto,CA。A heterologous gene sequence can be introduced into a target cell by using a "expression vector", preferably a eukaryotic organism, and even a better mammalian expression vector. Methods for constructing vectors are well known to those skilled in the art and are set forth in various publications. In particular, techniques for constructing suitable vectors (including instructions for functional components (eg, promoters, enhancers, termination signals, and polyadenylation signals, selection markers, origins of replication, and splicing signals) are described in considerable detail. It is outlined in (Sambrook et al., 1989) and references cited therein. Vectors can include, but are not limited to, plasmid vectors, phagemids, cosmids, artificial/minichromosome (eg, ACE) or viral vectors (eg, baculovirus, retrovirus, adenovirus, adeno-associated virus, herpes simplex virus) , retrovirus, phage). Eukaryotic expression vectors will also typically contain prokaryotic sequences that promote the propagation of the vector in bacteria, such as the origin of replication and the antibiotic resistance genes used to select among the bacteria. A variety of eukaryotic expression vectors containing selection sites for operably linked polynucleotides are well known in the art, and some are commercially available from companies such as: Stratagene, La Jolla, CA; Invitrogen, Carlsbad, CA; Promega, Madison , WI or BD Biosciences Clontech, Palo Alto, CA.

在H4-II-E大鼠肝瘤細胞中穩定表現所關注(糖)蛋白、較佳抗體或Fc融合蛋白之前提係該等細胞經編碼所關注蛋白之核酸序列轉染,其中該核酸序列可操作地連接至至少一種允許表現該核酸序列之調節序列。H4-II-E細胞中穩定表現糖蛋白之前提係該等細胞經DNA轉染,其中編碼所關注蛋白之基因功能連接至控制基因轉錄之遺傳元件。Stabilizing the expression of a glycoprotein, a preferred antibody or an Fc fusion protein in a H4-II-E rat hepatoma cell prior to transfection of the nucleic acid sequence encoding the protein of interest, wherein the nucleic acid sequence is It is operatively linked to at least one regulatory sequence that allows for the representation of the nucleic acid sequence. The cells are transfected with DNA prior to the stable expression of glycoproteins in H4-II-E cells, wherein the gene encoding the protein of interest is functionally linked to a genetic element that controls transcription of the gene.

此等控制基因轉錄之元件或「調節序列」係(例如)增強子、啟動子及5'UTR序列。實例包括(但不限於)SV40增強子、CMV增強子、白蛋白增強子、B型肝炎增強子、醛縮酶增強子、Ig增強子、酪胺酸酶增強子、SV40啟動子、EF1-α啟動子、雞β-肌動蛋白啟動子、CMV啟動子、HSV TK啟動子、磷酸甘油激酶(PGK)啟動子、聚合酶II啟動子、泛素C啟動子、白蛋白啟動子、α1-抗胰蛋白酶啟動子、α-甲胎蛋白(AFP)啟動子、醛縮酶啟動子、α1微球蛋白啟動子、磷酸烯醇丙酮酸羧激酶啟動子、RSV啟動子、GAPDH啟動子、β球蛋白啟動子及MT1啟動子。Such elements or "regulatory sequences" that control gene transcription are, for example, enhancers, promoters, and 5' UTR sequences. Examples include, but are not limited to, SV40 enhancer, CMV enhancer, albumin enhancer, hepatitis B enhancer, aldolase enhancer, Ig enhancer, tyrosinase enhancer, SV40 promoter, EF1-α Promoter, chicken β-actin promoter, CMV promoter, HSV TK promoter, phosphoglycerin kinase (PGK) promoter, polymerase II promoter, ubiquitin C promoter, albumin promoter, α1-antibody Trypsin promoter, α-fetoprotein (AFP) promoter, aldolase promoter, α1 microglobulin promoter, phosphoenolpyruvate carboxykinase promoter, RSV promoter, GAPDH promoter, beta globulin Promoter and MT1 promoter.

因此,在較佳實施例中,本發明H4-II-E細胞包含含有至少一種核酸序列之表現載體,該核酸序列為編碼所關注糖蛋白之核酸序列轉錄及轉譯所必需之調節序列。在具體實施例中,表現載體包含至少一種允許編碼所關注糖蛋白之所關注核酸序列或基因轉錄及轉譯(表現)之調節序列,該所關注糖蛋白較佳為抗體或Fc融合蛋白。Thus, in a preferred embodiment, the H4-II-E cells of the invention comprise an expression vector comprising at least one nucleic acid sequence which is a regulatory sequence necessary for transcription and translation of a nucleic acid sequence encoding a glycoprotein of interest. In a particular embodiment, the expression vector comprises at least one regulatory sequence that permits transcription and translation (expression) of the nucleic acid sequence of interest or encoding of the glycoprotein of interest, preferably an antibody or Fc fusion protein.

「調節序列」另外包括啟動子、增強子、終止信號及聚腺苷酸化信號以及其他表現控制元件。業內已知誘導型與組成型調節序列二者在多種細胞類型中發揮功能。轉錄調節元件通常包含位於擬表現基因序列上游之啟動子、轉錄起始及終止位點及聚腺苷酸化信號序列。術語轉錄起始位點係指構築體中對應於納入初級轉錄物(即,mRNA前體)中之第一核酸之核酸;轉錄起始位點可與啟動子序列重疊。術語轉錄終止位點係指通常顯示於所關注基因或擬轉錄序列伸長段之3'端之核苷酸序列,其使RNA聚合酶終止轉錄。聚腺苷酸化信號序列或聚A添加信號為真核mRNA 3'端之特異性位點之解離及在核中於該經解離3'端處轉錄後添加約100至200個腺嘌呤核苷酸(聚A尾)之序列提供信號。聚腺苷酸化信號序列包括位於解離位點上游約10至30個核苷酸處之序列AATAAA及下游序列。業內已知各種聚腺苷酸化元件,例如SV40晚期及早期聚A或BGH聚A。轉譯調節元件包括用於每一擬表現個別多肽之轉譯起始位點(AUG)、終止密碼子及聚A信號。一些構築體中包括內核糖體進入位點(IRES)。IRES定義於下文中。為優化表現,可能需要去除、添加或改變擬表現核酸序列之5'及/或3'未轉譯部分,以消除潛在的多餘的不適當替代轉譯起始密碼子或可在轉錄或轉譯層面干擾或降低表現之其他序列。另一選擇為,可緊鄰起始密碼子的5'插入共有核糖體結合位點以增強表現。為產生分泌多肽,所選序列通常將包括編碼前導肽之信號序列,該前導肽引導新合成多肽到達並穿過ER膜,其中可將多肽定至分泌途徑。前導肽經常但並非普遍位於分泌蛋白之胺基末端,且在蛋白穿過ER膜後藉由信號肽酶解離。所選序列通常(但並非必需)將包括其特有的信號序列。倘若不存在天然信號序列,則可將異源信號序列融合至所選序列。眾多信號序列為業內已知且可自序列數據庫(例如GenBank及EMBL)獲得。"Regulatory sequences" additionally include promoters, enhancers, termination signals, and polyadenylation signals as well as other expression control elements. Both inducible and constitutive regulatory sequences are known in the art to function in a variety of cell types. Transcriptional regulatory elements typically comprise a promoter upstream of the sequence of the gene to be expressed, a transcriptional initiation and termination site, and a polyadenylation signal sequence. The term transcription initiation site refers to a nucleic acid in a construct corresponding to a first nucleic acid that is included in a primary transcript (ie, an mRNA precursor); a transcription initiation site may overlap with a promoter sequence. The term transcription termination site refers to a nucleotide sequence typically displayed at the 3' end of the stretch of interest or a transcribed sequence that causes RNA polymerase to terminate transcription. The polyadenylation signal sequence or the poly A addition signal is a dissociation of a specific site at the 3' end of the eukaryotic mRNA and about 100 to 200 adenine nucleotides are added in the nucleus after transcription at the dissociated 3' end. The sequence of (poly A tail) provides a signal. The polyadenylation signal sequence comprises the sequence AATAAA and the downstream sequence located about 10 to 30 nucleotides upstream of the dissociation site. Various polyadenylation elements are known in the art, such as SV40 late and early poly A or BGH poly A. Translational regulatory elements include a translation initiation site (AUG), a stop codon, and a poly A signal for each of the individual polypeptides to be represented. Some constructs include a nucleosome saccharide entry site (IRES). IRES is defined below. To optimize performance, it may be necessary to remove, add or alter the 5' and/or 3' untranslated portion of the nucleic acid sequence to be expressed in order to eliminate potentially redundant inappropriate replacement translation initiation codons or to interfere at the transcriptional or translational level or Reduce other sequences of performance. Alternatively, a consensus ribosome binding site can be inserted 5' to the start codon to enhance performance. To produce a secreted polypeptide, the selected sequence will typically include a signal sequence encoding a leader peptide that directs the new synthetic polypeptide to and through the ER membrane, wherein the polypeptide can be placed into the secretory pathway. The leader peptide is often, but not exclusively, located at the amino terminus of the secreted protein and is cleaved by signal peptidase after the protein has passed through the ER membrane. The selected sequence will usually (but not necessarily) include its unique signal sequence. If the native signal sequence is absent, the heterologous signal sequence can be fused to the selected sequence. Numerous signal sequences are known in the art and are available from sequence databases such as GenBank and EMBL.

「啟動子」係指控制可操作連接之基因或序列轉錄之多核苷酸序列/核酸序列。啟動子包括用於RNA聚合酶結合及轉錄起始之信號。所用啟動子將在涵蓋表現所選序列之細胞類型之宿主細胞中具有功能,該細胞類型在本發明中為H4-II-E細胞。包括來自多種不同來源之組成型、誘導型及抑制型啟動子之大量啟動子已為業內所熟知(且在諸如GenBank等數據庫中予以鑑別)且可以選殖多核苷酸(來自(例如)諸如ATCC等寄存處以及其他商業或個人來源)或在選殖多核苷酸內獲得。就誘導型啟動子而言,啟動子活性因應信號而提高或降低。舉例而言,含有四環素操縱子序列(tetO)之四環素(tet)啟動子可藉由四環素調節之反式活化蛋白(tTA)誘導。在tet存在下可抑制tTA與tetO之結合。關於其他誘導型啟動子(包括jun、fos、金屬硫蛋白及熱激啟動子)可參見(例如)Sambrook等人,1989。已因高程度表現而鑑別為強啟動子之真核啟動子包括SV40早期啟動子、腺病毒主要晚期啟動子、小鼠金屬硫蛋白-I啟動子、勞氏肉瘤病毒(Rous sarcoma virus)長末端重複序列及人類巨細胞病毒極早期啟動子(CMV)。其他異源哺乳動物啟動子包括(例如)肌動蛋白啟動子、免疫球蛋白啟動子、熱激啟動子。上述啟動子為業內所熟知。"Promoter" refers to a polynucleotide sequence/nucleic acid sequence that controls the transcription of a operably linked gene or sequence. Promoters include signals for RNA polymerase binding and transcription initiation. The promoter used will be functional in a host cell encompassing the cell type exhibiting the selected sequence, which is a H4-II-E cell in the present invention. A large number of promoters including constitutive, inducible and inhibitory promoters from a variety of different sources are well known in the art (and identified in databases such as GenBank) and can be cloned (for example from, for example, ATCC) Such as storage and other commercial or personal sources) or obtained within the selected polynucleotide. In the case of an inducible promoter, the promoter activity is increased or decreased in response to the signal. For example, a tetracycline (tet) promoter containing a tetracycline operator sequence (tetO) can be induced by tetracycline regulated transactivation protein (tTA). The binding of tTA to tetO can be inhibited in the presence of tet. For other inducible promoters (including jun, fos, metallothionein and heat shock promoters) see, for example, Sambrook et al., 1989. Eukaryotic promoters that have been identified as strong promoters due to their high degree of expression include the SV40 early promoter, the adenovirus major late promoter, the mouse metallothionein-I promoter, and the long end of Rous sarcoma virus. Repeat sequences and human cytomegalovirus very early promoter (CMV). Other heterologous mammalian promoters include, for example, actin promoters, immunoglobulin promoters, heat shock promoters. The above promoters are well known in the art.

本文所用「增強子」係指作用於啟動子以增強可操作地連接之基因或編碼序列之轉錄的多核苷酸序列/核酸序列。與啟動子不同,增強子具有方向及位置相對獨立性且已發現其在轉錄單元中位於內含子內以及編碼序列本身內之5'或3'處。因此,增強子可置於轉錄起始位點之上游或下游或距啟動子相當大距離處,但實際上增強子可在物理上及功能上與啟動子重疊。來自多種不同來源之大量增強子為業內所熟知(且在諸如GenBank等數據庫中予以鑑別,例如SV40增強子、CMV增強子、多瘤增強子、腺病毒增強子)且可以選殖多核苷酸序列(來自(例如)諸如ATCC等寄存處以及其他商業或個人來源)或在選殖多核苷酸序列內獲得。許多包含啟動子序列(例如常用CMV啟動子)之多核苷酸亦包含增強子序列。舉例而言,上文所列示之所有強啟動子亦含有強增強子。As used herein, "enhancer" refers to a polynucleotide sequence/nucleic acid sequence that acts on a promoter to enhance transcription of an operably linked gene or coding sequence. Unlike promoters, enhancers have relative orientation and positional independence and have been found to be located within the intron and at the 5' or 3' within the coding sequence itself in the transcription unit. Thus, an enhancer can be placed upstream or downstream of the transcription start site or at a considerable distance from the promoter, but in fact the enhancer can physically and functionally overlap with the promoter. Numerous enhancers from a variety of different sources are well known in the art (and are identified in databases such as GenBank, such as the SV40 enhancer, CMV enhancer, polyoma enhancer, adenovirus enhancer) and can be used to select polynucleotide sequences. (from, for example, a registry such as ATCC and other commercial or personal sources) or obtained within a select polynucleotide sequence. Many polynucleotides comprising a promoter sequence (eg, a common CMV promoter) also contain an enhancer sequence. For example, all of the strong promoters listed above also contain strong enhancers.

術語「可操作地連接」意指兩個或更多個核酸序列或序列元件以允許其以其期望方式發揮功能之方式佈置。舉例而言,若啟動子及/或增強子以順式方式作用以控制或調節所連接序列之轉錄,則該啟動子及/或增強子與編碼序列可操作地連接。通常(但並非必需),可操作地連接之DNA序列係鄰接的,且倘若必需接合兩個蛋白編碼區或在分泌前導序列之情形下,則為鄰接且處於閱讀框內。The term "operably linked" means that two or more nucleic acid sequences or sequence elements are arranged in a manner that allows them to function in their intended manner. For example, a promoter and/or enhancer is operably linked to a coding sequence if the promoter and/or enhancer acts in a cis manner to control or regulate transcription of the ligated sequence. Typically, but not necessarily, the operably linked DNA sequences are contiguous and, if necessary, ligated to the two protein coding regions or in the context of a secretory leader sequence, are contiguous and in reading frame.

然而,儘管可操作地連接之啟動子通常位於編碼序列之上游,但不必與其鄰接。增強子不必鄰接,只要其可增加編碼序列之轉錄即可。為此,其可位於編碼序列之上游或下游且甚至相隔一定距離。若聚腺苷酸化位點位於編碼序列之3'端以使轉錄通過編碼序列進行至聚腺苷酸化序列,則該聚腺苷酸化位點與編碼序列可操作連接。連接係藉由業內已知之重組方法達成,例如,使用PCR方法、藉由在適宜限制位點連接或藉由退火。若不存在適宜限制位點,則可根據習用慣例使用合成寡核苷酸連接體或銜接子。However, although the operably linked promoter is typically located upstream of the coding sequence, it does not have to be contiguous thereto. The enhancer does not have to be contiguous as long as it increases the transcription of the coding sequence. To this end, it can be located upstream or downstream of the coding sequence and even at a distance. A polyadenylation site is operably linked to a coding sequence if the polyadenylation site is located at the 3' end of the coding sequence such that transcription proceeds through the coding sequence to the polyadenylation sequence. Linkages are achieved by recombinant methods known in the art, for example, using PCR methods, by ligation at appropriate restriction sites, or by annealing. If no suitable restriction sites are present, synthetic oligonucleotide linkers or adaptors can be used according to customary practice.

「轉錄單元」界定構築體內含有一或多個擬轉錄基因之區域,其中含於該區段內之基因彼此可操作地連接並自單一啟動子轉錄,且因此,不同基因至少轉錄連接。可自每一轉錄單元轉錄並表現一種以上蛋白或產物。每一轉錄單元將包含該單元中所含任一所選序列之轉錄及轉譯所必需之調節元件。A "transcription unit" defines a region in a construct that contains one or more pseudo-transcribed genes in which genes contained within the segment are operably linked to each other and transcribed from a single promoter, and thus, different genes are at least transcribed. More than one protein or product can be transcribed and expressed from each transcription unit. Each transcription unit will contain the regulatory elements necessary for transcription and translation of any of the selected sequences contained in the unit.

本文所用術語「表現」係指異源核酸序列在宿主細胞內之轉錄及/或轉譯。期望產物/所關注蛋白在宿主細胞內之表現程度可基於存在於細胞中之對應mRNA之量、或由本實例內所選序列編碼之期望多肽/所關注蛋白之量來確定。舉例而言,自所選序列轉錄之mRNA可藉由北方墨點雜交(Northern blot hybridization)、核糖核酸酶RNA保護、原位雜交至細胞RNA或藉由PCR來量化。由所選序列編碼之蛋白可藉由各種方法來量化,例如藉由ELISA、藉由西方墨點(Western blotting)、藉由放射免疫分析、藉由免疫沉澱、藉由分析蛋白之生物活性、藉由對蛋白實施免疫染色且隨後進行FACS分析或藉由均相時間解析螢光(HTRF)分析。The term "expression" as used herein refers to the transcription and/or translation of a heterologous nucleic acid sequence within a host cell. The degree of expression of the desired product/protein of interest in the host cell can be determined based on the amount of corresponding mRNA present in the cell, or the amount of the desired polypeptide/protein of interest encoded by the sequence selected within the example. For example, mRNA transcribed from a selected sequence can be quantified by Northern blot hybridization, ribonuclease RNA protection, in situ hybridization to cellular RNA, or by PCR. The protein encoded by the selected sequence can be quantified by various methods, such as by ELISA, by Western blotting, by radioimmunoassay, by immunoprecipitation, by analyzing the biological activity of the protein, borrowing Immunostaining is performed on the protein and then subjected to FACS analysis or by homogeneous time-resolved fluorescence (HTRF) analysis.

「表現盒」界定構築體內含有一或多個擬轉錄基因之區域,其中含於該區段內之基因彼此可操作地連接並自單一啟動子轉錄,且因此,不同基因至少轉錄連接。可自每一轉錄單元轉錄並表現一種以上蛋白或產物。每一轉錄單元將包含該單元中所含任一所選序列之轉錄及轉譯所必需之調節元件。A "benefit cassette" defines a region in a body that contains one or more pseudo-transcribed genes in which genes contained within the segment are operably linked to each other and transcribed from a single promoter, and thus, different genes are at least transcribed. More than one protein or product can be transcribed and expressed from each transcription unit. Each transcription unit will contain the regulatory elements necessary for transcription and translation of any of the selected sequences contained in the unit.

用多核苷酸或表現載體「轉染」真核宿主細胞以產生遺傳修飾細胞或轉基因細胞,可藉由業內熟知之任一方法來實施(例如參見(Sambrook等人,1989))。轉染方法包括(但不限於)脂質體介導之轉染、磷酸鈣共沉澱、電穿孔、核轉染、核穿孔(nucleoporation)、微穿孔、聚陽離子(例如DEAE-葡聚糖)介導之轉染、原生質體融合、病毒感染及微注射。較佳地,轉染係穩定轉染。提供異源基因在特定宿主細胞系及類型中之最佳轉染頻率及表現的轉染方法係有利的。適宜方法可藉由常規程序確定。對於穩定轉染子而言,構築體係整合至宿主細胞之基因組或人工染色體/微型染色體中或游離定位以穩定維持於宿主細胞內。The "transfection" of a eukaryotic host cell with a polynucleotide or expression vector to produce a genetically modified cell or transgenic cell can be carried out by any method well known in the art (see, for example, (Sambrook et al., 1989)). Transfection methods include, but are not limited to, liposome-mediated transfection, calcium phosphate co-precipitation, electroporation, nuclear transfection, nuclear nucleoporation, microperforation, polycation (eg, DEAE-dextran) mediated Transfection, protoplast fusion, viral infection and microinjection. Preferably, the transfection is stably transfected. Transfection methods that provide optimal transfection frequencies and performance of heterologous genes in a particular host cell line and type are advantageous. Suitable methods can be determined by routine procedures. For stable transfectants, the construction system is integrated into the genome or artificial chromosome/minichromosome of the host cell or freely positioned to stably maintain in the host cell.

「可選標記物基因」或「選擇標記物基因」係允許藉由將對應選擇劑添加至培養基中來特異性選擇含有此基因之細胞之基因。作為說明,抗生素抗性基因可用作陽性可選標記物。僅已經此基因轉化之細胞能夠在對應抗生素存在下生長並因此進行選擇。另一方面,未轉化細胞不能在該等選擇條件下生長或存活。存在陽性、陰性及雙功能可選標記物。陽性可選標記物藉由賦予對選擇劑之抗性或藉由補償宿主細胞之代謝或分解代謝缺陷來允許對轉化細胞進行選擇及(因此)富集。相比之下,已接收用於可選標記物之基因之細胞可藉由陰性可選標記物來選擇性地消除。其實例係單純皰疹病毒之胸苷激酶基因,其在同時添加有阿昔洛維(acyclovir)或丙氧鳥苷(gancyclovir)之細胞中之表現可導致其消除。用於本發明中之可選標記物(包括可擴增可選標記物)包括遺傳修飾突變體及變體、片段、功能等效物、衍生物、同源物及與其他蛋白或肽之融合物,前提為可選標記物保持其選擇性品質。此等衍生物在視為具有選擇性之區域或結構域中之胺基酸序列上展示相當大的同源性。文獻闡述大量可選標記物基因(包括雙功能(陽性/陰性)標記物)(例如參見WO 92/08796及WO 94/28143)。通常用於真核細胞中之可選標記物之實例包括用於以下之基因:胺基糖苷磷酸轉移酶(APH)、潮黴素磷酸轉移酶(hygromycine phosphostransferase)(HYG)、二氫葉酸還原酶(DHFR)、胸苷激酶(TK)、麩醯胺酸合成酶、天冬醯胺酸合成酶;及賦予對以下之抗性之基因:新黴素(neomycin)(G418/遺傳黴素(Geneticin))、嘌呤黴素(puromycin)、組胺醇D、博來黴素(bleomycin)、腐草黴素(phleomycin)及吉歐黴素(zeocin)。The "selectable marker gene" or "selection marker gene" allows for the specific selection of the gene of the cell containing the gene by adding the corresponding selection agent to the medium. As an illustration, an antibiotic resistance gene can be used as a positive selectable marker. Only cells that have been transformed with this gene are able to grow in the presence of the corresponding antibiotic and are therefore selected. On the other hand, untransformed cells cannot grow or survive under these selective conditions. There are positive, negative and bifunctional selectable markers. Positive selectable markers allow selection and (and therefore) enrichment of transformed cells by conferring resistance to the selection agent or by compensating for metabolic or catabolic defects in the host cell. In contrast, cells that have received a gene for a selectable marker can be selectively eliminated by a negative selectable marker. An example of this is the herpes simplex virus thymidine kinase gene, which is manifested in the elimination of cells that are simultaneously added with acyclovir or gancyclovir. Alternative labels (including amplifiable selectable markers) for use in the present invention include genetically modified mutants and variants, fragments, functional equivalents, derivatives, homologs, and fusions with other proteins or peptides The premise is that the optional marker retains its selective quality. Such derivatives exhibit considerable homology on amino acid sequences in regions or domains that are considered to be selective. The literature describes a number of selectable marker genes (including bifunctional (positive/negative) markers) (see, for example, WO 92/08796 and WO 94/28143). Examples of selectable markers commonly used in eukaryotic cells include the following genes: aminoglycoside phosphotransferase (APH), hygromycine phosphostransferase (HYG), dihydrofolate reductase (DHFR), thymidine kinase (TK), glutamate synthase, aspartate synthase; and a gene conferring resistance to neomycin (G418/geneticin) )), puromycin, histamine D, bleomycin, phleomycin and zeocin.

可藉由螢光活化細胞分選(FACS)使用(例如)細胞表面標記物、細菌β-半乳糖苷酶或螢光蛋白(例如來自維多利亞水母(Aequorea victoria)及海腎(Renilla reniformis)或其他物種之綠色螢光蛋白(GFP)及其變體);紅螢光蛋白、來自非生物發光物種(例如香菇珊瑚(Discosoma sp.)海葵(Anemonia sp.)羽珊瑚(Clavularia sp.)、棕綠紐扣珊瑚(Zoanthus sp.)之螢光蛋白及其變體)對重組細胞進行選擇來進行選擇。Fluorescence-activated cell sorting (FACS) can be used, for example, on cell surface markers, bacterial beta-galactosidase or fluorescent proteins (eg, from Aequorea victoria and Renilla reniformis or others). Species of green fluorescent protein (GFP) and its variants; red fluorescent protein, from non-bioluminescent species (eg, Discosoma sp . , Anemonia sp .) , Clavularia sp . , the fluorescent protein of Zoanthus sp . and its variants are selected for selection of recombinant cells.

術語「選擇劑」係指干擾缺乏特定可選基因之H4-II-E細胞之生長或存活的物質。舉例而言,為對抗生素抗性基因(例如APH(胺基糖苷磷酸轉移酶))在轉染細胞中之存在進行選擇,使用抗生素遺傳黴素(G418)。The term "selective agent" refers to a substance that interferes with the growth or survival of H4-II-E cells lacking a particular selectable gene. For example, to select for the presence of an antibiotic resistance gene (eg, APH (aminoglycoside phosphotransferase)) in transfected cells, the antibiotic geneticin (G418) is used.

術語「經修飾新黴素-磷酸轉移酶(NPT)」涵蓋WO2004/050884中所述之所有突變體、尤其突變體D227G(Asp227Gly)(其特徵在於在胺基酸位置227處用天冬胺酸(Asp,D)取代甘胺酸(Gly,G))且尤佳突變體F240I(Phe240Ile)(其特徵在於在胺基酸位置240處用苯丙胺酸(Phe,F)取代異白胺酸(Ile,I))。The term "modified neomycin-phosphotransferase (NPT)" encompasses all mutants described in WO2004/050884, in particular mutant D227G (Asp227Gly) (characterized by the use of aspartic acid at amino acid position 227) (Asp, D) substituted glycine (Gly, G)) and especially preferred mutant F240I (Phe240Ile) (characterized by the replacement of isoleucine with phenylalanine (Phe, F) at position 125 of the amino acid (Ile) , I)).

本發明進一步包括製備及選擇重組H4-II-E細胞之方法,其包含以下步驟:(i)用編碼至少一種所關注糖蛋白/產物及選擇標記物、較佳新黴素-磷酸轉移酶之基因轉染H4-II-E細胞,其中為增強轉錄或表現,將至少所關注基因(或多個基因)連接至驅使基因穩定表現之增強子、啟動子及5'-UTR序列及/或視情況功能連接至至少一種TE元件(參見WO2008/012142,其以引用方式併入本文中);(ii)在能夠表現不同基因之條件下培育該等細胞;及(iii)藉由在選擇劑(例如G418、MTX或MSX)存在下培育該等細胞來選擇該等共整合基因。較佳地,在無血清培養基中培育轉染細胞。較佳地,G418之濃度係至少200 μg/mL。然而,該濃度亦可為至少400 μg/mL。The invention further comprises a method of making and selecting recombinant H4-II-E cells comprising the steps of: (i) encoding at least one glycoprotein/product of interest and a selection marker, preferably neomycin-phosphotransferase. Genes are transfected into H4-II-E cells, wherein to enhance transcription or expression, at least the gene of interest (or genes) is ligated to enhancers, promoters and 5'-UTR sequences and/or visually The conditional function is linked to at least one TE element (see WO 2008/012142, which is incorporated herein by reference); (ii) cultivating the cells under conditions capable of expressing different genes; and (iii) by selecting a reagent ( Such cells are incubated in the presence of, for example, G418, MTX or MSX) to select such co-integrating genes. Preferably, the transfected cells are cultured in serum-free medium. Preferably, the concentration of G418 is at least 200 μg/mL. However, the concentration can also be at least 400 μg/mL.

可擴增可選標記物基因:Amplable selectable marker genes:

另外,本發明細胞亦可視情況經受一或多個基因擴增步驟,其中在選擇劑存在下培育該等細胞,該選擇劑使可擴增可選標記物基因擴增。In addition, the cells of the invention may optionally be subjected to one or more gene amplification steps, wherein the cells are incubated in the presence of a selection agent that amplifies the amplifiable selectable marker gene.

前提為另外用編碼可擴增可選標記物之基因轉染H4-II-E細胞。可設想編碼可擴增可選標記物之基因存在於本發明表現載體中之一者上或藉助其他載體引入宿主細胞中。It is a prerequisite to additionally transfect H4-II-E cells with a gene encoding an amplifiable selectable marker. It is contemplated that the gene encoding the amplifiable selectable marker will be present on one of the expression vectors of the invention or introduced into the host cell by means of other vectors.

可擴增可選標記物基因通常編碼在某些培育條件下生長真核細胞需要之酶。舉例而言,可擴增可選標記物基因可編碼二氫葉酸還原酶(DHFR)或麩醯胺酸合成酶(GS)。在此情形下,若在選擇劑甲胺蝶呤(MTX)或甲硫胺酸磺醯亞胺(MSX)存在下培育用該基因轉染之宿主細胞,則可擴增該基因。The amplifiable selectable marker gene typically encodes an enzyme required to grow eukaryotic cells under certain incubation conditions. For example, the amplifiable selectable marker gene can encode dihydrofolate reductase (DHFR) or glutamate synthase (GS). In this case, the gene can be amplified if the host cell transfected with the gene is cultured in the presence of the selective agent methotrexate (MTX) or sulfinamide imipenem (MSX).

下表2給出可根據本發明使用之可擴增可選標記物基因及相關選擇劑之實例,其由Kaufman進行了概述(Kaufman,1990)。Table 2 below gives examples of amplifiable selectable marker genes and related selection agents that can be used in accordance with the present invention, as outlined by Kaufman (Kaufman, 1990).

表2:可擴增可選標記物基因Table 2: Amplable selectable marker genes

根據本發明,所用可擴增可選標記物基因較佳為編碼具有GS或DHFR功能之多肽之基因。According to the invention, the amplifiable selectable marker gene used is preferably a gene encoding a polypeptide having GS or DHFR function.

本文所用術語「轉化(transformation或to transform)」、「轉染(transfection或to transfect)」意指核酸序列至細胞中之任何引入,以產生遺傳修飾、重組、轉化或轉基因細胞。可藉由業內熟知之任一方法來實施引入。方法包括(但不限於)脂轉染、電穿孔、聚陽離子(例如DEAE-葡聚糖)介導轉染、原生質體融合、病毒感染及微注射,或可藉助鈣法、電休克法、靜脈內/肌內注射、氣溶膠吸入或卵母細胞注射來實施。轉化可瞬時或穩定轉化宿主細胞。術語「轉染(transfection或to transfect)」、「轉化(transformation或to transform)」亦意指以各別病毒之天然引入方式引入病毒核酸序列。病毒核酸序列無需以裸露核酸序列形式存在,而可封裝於病毒蛋白包膜中。因此,該術語不僅係關於通常以術語「轉染(transfection或to transfect)」、「轉化(transformation或to transform)」已知之方法。提供所引入核酸之最佳轉染頻率及表現之轉染方法係有利的。適宜方法可藉由常規程序確定。對於穩定轉染子而言,構築體係整合至宿主細胞之基因組或人工染色體/微型染色體中或游離定位以穩定維持於宿主細胞內。The terms "transformation or to transform", "transfection or to transfect" as used herein, mean any introduction of a nucleic acid sequence into a cell to produce a genetically modified, recombinant, transformed or transgenic cell. The introduction can be implemented by any method well known in the art. Methods include, but are not limited to, lipofection, electroporation, polycation (eg, DEAE-dextran) mediated transfection, protoplast fusion, viral infection, and microinjection, or may be by calcium, electroshock, vein This is carried out by intra/intramuscular injection, aerosol inhalation or oocyte injection. Transformation can transiently or stably transform host cells. The terms "transfection or to transfect", "transformation or to transform" also mean the introduction of a viral nucleic acid sequence by the natural introduction of a particular virus. The viral nucleic acid sequence does not need to be in the form of a naked nucleic acid sequence, but can be encapsulated in a viral protein envelope. Thus, the term is not only about methods that are commonly known by the terms "transfection or to transfect", "transformation or to transform". Transfection methods that provide optimal transfection frequencies and performance of the introduced nucleic acids are advantageous. Suitable methods can be determined by routine procedures. For stable transfectants, the construction system is integrated into the genome or artificial chromosome/minichromosome of the host cell or freely positioned to stably maintain in the host cell.

具體實施例Specific embodiment

本發明闡述包含編碼抗體或Fc-融合蛋白之核酸序列的大鼠肝瘤細胞,其中該核酸序列可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之核酸序列的調節序列。本發明進一步闡述特徵在於帶有編碼抗體或Fc-融合蛋白之核酸序列的大鼠肝瘤細胞,其中該核酸序列可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之核酸序列的調節序列。The invention features a rat hepatoma cell comprising a nucleic acid sequence encoding an antibody or Fc-fusion protein, wherein the nucleic acid sequence is operably linked to at least one regulatory sequence that permits expression of the nucleic acid sequence encoding the antibody or Fc-fusion protein. The invention further describes a rat hepatoma cell characterized by a nucleic acid sequence encoding an antibody or Fc-fusion protein, wherein the nucleic acid sequence is operably linked to at least one nucleic acid sequence which permits expression of the encoding antibody or Fc-fusion protein. Adjust the sequence.

在一具體實施例中,大鼠肝瘤細胞係H4-II-E細胞。在又一具體實施例中,大鼠肝瘤細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。In a specific embodiment, the rat hepatoma cell line H4-II-E cells. In yet another embodiment, the rat hepatoma cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

本發明闡述包含/特徵在於帶有編碼抗體或Fc-融合蛋白之核酸序列的H4-II-E大鼠肝瘤細胞,其中該核酸序列可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之核酸序列的調節序列。The invention encloses H4-II-E rat hepatoma cells comprising/characterized with a nucleic acid sequence encoding an antibody or Fc-fusion protein, wherein the nucleic acid sequence is operably linked to at least one of which allows expression of the encoding antibody or Fc- A regulatory sequence of a nucleic acid sequence of a fusion protein.

本發明闡述藉由以下方式進行遺傳修飾之H4-II-E大鼠肝瘤細胞:引入編碼糖蛋白、較佳抗體或Fc融合蛋白之所關注核酸序列/基因,其可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之所關注核酸序列/基因之調節序列。The present invention describes H4-II-E rat hepatoma cells genetically modified by introducing a nucleic acid sequence of interest/gene encoding a glycoprotein, preferably an antibody or Fc fusion protein operably linked to at least one The regulatory sequence of the nucleic acid sequence/gene of interest encoding the antibody or Fc-fusion protein is allowed to be expressed.

本發明闡述包含/特徵在於帶有編碼抗體或Fc-融合蛋白之核酸序列的H4-II-E大鼠肝瘤細胞或其衍生物或子代,其中該核酸序列可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之核酸序列的調節序列。The invention features H4-II-E rat hepatoma cells or derivatives or progeny thereof comprising/characterized with a nucleic acid sequence encoding an antibody or Fc-fusion protein, wherein the nucleic acid sequence is operably linked to at least one of the permits The regulatory sequence of the nucleic acid sequence encoding the antibody or Fc-fusion protein is expressed.

本發明闡述藉由以下方式進行遺傳修飾之H4-II-E大鼠肝瘤細胞或其衍生物或子代:引入編碼糖蛋白、較佳抗體或Fc融合蛋白之所關注核酸序列/基因,其可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之所關注核酸序列/基因之調節序列。The present invention provides H4-II-E rat hepatoma cells or derivatives or progeny thereof genetically modified by introducing a nucleic acid sequence/gene of interest encoding a glycoprotein, preferably an antibody or an Fc fusion protein, The regulatory sequence is operably linked to at least one nucleic acid sequence/gene of interest that allows expression of the encoding antibody or Fc-fusion protein.

在一具體實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係源自歐洲細胞培養物保存中心(ECACC,目錄編號87031301)或美國典型培養物保存中心(ATCC,寄存編號CRL-1548)之細胞或該細胞係以編號ECACC、目錄編號87031301寄存於歐洲細胞培養物保存中心之細胞或係其衍生物或子代或其中該細胞係以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC或係其衍生物或子代。在又一具體實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。在另一具體實施例中,該大鼠肝瘤細胞係:In a specific embodiment, the rat hepatoma cell or the H4-II-E cell line is derived from the European Cell Culture Preservation Center (ECACC, Cat. No. 87031301) or the American Type Culture Collection (ATCC, Accession Number CRL) The cell of -1548) or the cell line is deposited in the cell of the European Cell Culture Preservation Center under the number ECACC, catalog number 87031301, or a derivative or progeny thereof, or the cell line is deposited in the American typical culture under the accession number CRL-1548. The object preservation center ATCC is either a derivative or a progeny thereof. In still another embodiment, the rat hepatoma cell or the H4-II-E cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548. In another specific embodiment, the rat hepatoma cell line:

a)源自選自由以下組成之群之細胞的細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或a) cells derived from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E -C3 cell line (CRL-1600 or HPACC No. 85061112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) And the H4-S cell line (HPACC Nr. 89102001), or

b)以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或b) deposited in the European Cell Culture Preservation Center under the number ECACC catalog number 87031301 or in the US Type Culture Collection Center ATCC under the accession number CRL-1548, or

c)以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或c) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E), or

d)以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或d) cells deposited in the DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or

e)a)或b)或c)或d)中任一細胞之衍生物或子代。在一具體實施例中,該大鼠肝瘤或該H4-II-E細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。e) a derivative or progeny of any of cells a) or b) or c) or d). In a specific embodiment, the rat hepatoma or the H4-II-E cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

在另一具體實施例中,該大鼠肝瘤細胞或該H4-II-E大鼠肝瘤細胞具有低海藻糖基化活性,該活性用於將海藻糖添加至諸如二枝結構聚糖(例如N-乙醯葡糖胺)等糖苷結構中。In another specific embodiment, the rat hepatoma cell or the H4-II-E rat hepatoma cell has low algal glycosylation activity for adding trehalose to a structure such as a twig For example, in the glycoside structure such as N-acetylglucosamine.

具體而言,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞之特徵進一步在於:i)由該細胞表現之抗體或Fc-融合蛋白中所含糖苷結構含有海藻糖之程度(或比例)為(所有聚糖/糖苷結構之)小於20%、10%或5%或ii)該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有海藻糖之程度在(所有聚糖/糖苷結構之)介於0%至20%、0%至10%、0%至5%、0.5%至20%、0.5%至10%、0.5%至5%、1%至20%、1%至10%或1%至5%之間之範圍內。Specifically, the rat hepatoma cell of the present invention or the H4-II-E rat hepatoma cell is further characterized by: i) the glycoside structure contained in the antibody or the Fc-fusion protein expressed by the cell contains trehalose The degree (or ratio) is less than 20%, 10% or 5% (of all glycan/glycoside structures) or ii) the degree of glycosidic structure contained in the antibody or Fc-fusion protein expressed by the cell is such that the degree of trehalose is ( All glycan/glycosidic structures) between 0% and 20%, 0% to 10%, 0% to 5%, 0.5% to 20%, 0.5% to 10%, 0.5% to 5%, 1% to 20 %, 1% to 10% or between 1% and 5%.

具體而言,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞之特徵進一步在於:該抗體或Fc-融合蛋白之小於20%聚糖/糖苷結構含有結合至末端還原N-乙醯葡糖胺(GlcNAc)殘基之海藻糖。較佳地,該等去海藻糖基化糖苷結構/聚糖係N-連接,最佳地,對於IgG1、IgG2、IgG3及IgG4抗體而言,根據Kabat EU命名法(Kabat等人,1991),該等糖苷結構/聚糖係藉由N-連接糖基化附著於第297位胺基酸殘基。Specifically, the rat hepatoma cell or the H4-II-E rat hepatoma cell of the present invention is further characterized in that less than 20% of the glycan/glycoside structure of the antibody or Fc-fusion protein contains binding to the terminal reduction N - Trehalose of the residue of glucosamine (GlcNAc). Preferably, the de-fucosylated glycoside structure/glycans are N-linked, optimally, for IgGl, IgG2, IgG3 and IgG4 antibodies, according to the Kabat EU nomenclature (Kabat et al., 1991), The glycoside structures/glycans are attached to the amino acid residue at position 297 by N-linked glycosylation.

在一具體實施例中,該大鼠肝瘤細胞或該H4-II-E大鼠肝瘤細胞具有高半乳糖基化活性,該活性用於將半乳糖添加至諸如二枝結構聚糖(例如N-乙醯葡糖胺)等糖苷結構中。In a specific embodiment, the rat hepatoma cell or the H4-II-E rat hepatoma cell has high galactosylation activity for adding galactose to a glycan such as a twig (eg N-acetylglucosamine) and other glycoside structures.

具體而言,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞之特徵進一步在於:i)由該細胞表現之該抗體或Fc-融合蛋白中所含聚糖/糖苷結構、較佳複合型聚糖/糖苷結構含有至少一個、較佳一或兩個或一或多個半乳糖殘基之程度為(所有複合型聚糖/糖苷結構之)超過40%、45%或50%或ii)由該細胞表現之該抗體或Fc-融合蛋白中所含聚糖/糖苷結構、較佳複合型聚糖/糖苷結構含有至少一個、較佳一或兩個或一或多個半乳糖殘基之程度在(所有複合型聚糖/糖苷結構之)介於40%至100%、45%至100%、50%至100%、51%至100%、40%至99.5%、45%至99.5%、50%至99.5%或51%至99.5%、40%至99%、45%至99%、50%至99%或51%至99%之間之範圍內。Specifically, the rat hepatoma cell or the H4-II-E rat hepatoma cell of the present invention is further characterized by: i) a glycan/glycosidic structure contained in the antibody or Fc-fusion protein expressed by the cell Preferably, the complex glycan/glycosidic structure contains at least one, preferably one or two or one or more galactose residues to a degree (all of the complex glycan/glycosidic structures) of more than 40%, 45% or 50% or ii) the glycan/glycosidic structure, preferably the complex glycan/glycosidic structure contained in the antibody or Fc-fusion protein represented by the cell, containing at least one, preferably one or two or one or more The degree of galactose residues is between 40% to 100%, 45% to 100%, 50% to 100%, 51% to 100%, 40% to 99.5%, of all complex glycan/glycosidic structures, It is in the range of 45% to 99.5%, 50% to 99.5% or 51% to 99.5%, 40% to 99%, 45% to 99%, 50% to 99% or 51% to 99%.

較佳地,該等半乳糖基化糖苷結構/聚糖含有一或兩個較佳連接至該等糖苷結構之末端非還原處之N-乙醯葡糖胺(GlcNAc)之半乳糖殘基(G1或G2)。較佳地,對於IgG1、IgG2、IgG3及IgG4抗體而言,根據Kabat EU命名法(Kabat等人,1991),該等糖苷結構/聚糖係N-連接於第297位胺基酸殘基。Preferably, the galactosylated glycoside structures/glycans contain one or two galactose residues of N-acetylglucosamine (GlcNAc) preferably attached to the terminal non-reducing sites of the glycoside structures ( G1 or G2). Preferably, for IgGl, IgG2, IgG3 and IgG4 antibodies, the glycoside structure/glycans are N-linked to the amino acid residue at position 297 according to the Kabat EU nomenclature (Kabat et al., 1991).

在本發明一具體實施例中,聚糖/糖苷結構係G1或G2。聚糖/糖苷結構較佳不為G0。In a particular embodiment of the invention, the glycan/glycosidic structure is G1 or G2. The glycan/glycoside structure is preferably not G0.

在一具體實施例中,該大鼠肝瘤細胞或該H4-II-E大鼠肝瘤細胞具有高唾液酸化活性,該活性用於將唾液酸或神經胺酸殘基添加至諸如半乳糖基化二枝結構聚糖等糖苷結構中。In a specific embodiment, the rat hepatoma cell or the H4-II-E rat hepatoma cell has high sialylation activity for adding a sialic acid or a neuramic acid residue to, for example, a galactosyl group. The structure of glycosides such as dimeric structural glycans.

具體而言,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞之特徵進一步在於:i)由該細胞表現之該抗體或Fc-融合蛋白中所含(半乳糖基化)糖苷結構含有末端唾液酸或神經胺酸殘基之程度為超過5%或超過10%或ii)由該細胞表現之該抗體或Fc-融合蛋白中所含(半乳糖基化)糖苷結構含有末端唾液酸或神經胺酸殘基之程度在介於0-8%、1-8%、5-10%、10-50%或10-45%之間之範圍內。Specifically, the rat hepatoma cells of the present invention or the H4-II-E rat hepatoma cells are further characterized by: i) the antibody or Fc-fusion protein expressed by the cell (galactosylation) a glycoside structure containing a terminal sialic acid or a ceramide residue to a degree of more than 5% or more than 10% or ii) a (galactosylated) glycoside structure contained in the antibody or Fc-fusion protein expressed by the cell The degree of terminal sialic acid or neuramic acid residues is in the range of 0-8%, 1-8%, 5-10%, 10-50% or 10-45%.

較佳地,該等含有末端唾液酸或神經胺酸殘基之糖苷結構/聚糖係N-連接,最佳地,對於IgG1、IgG2、IgG3及IgG4抗體而言,根據Kabat EU命名法(Kabat等人,1991),該等糖苷結構/聚糖藉由N-連接糖基化附著於第297位胺基酸殘基。Preferably, the glycoside structure/glycan N-linkage containing a terminal sialic acid or a neuraminic acid residue, optimally, for the IgG1, IgG2, IgG3 and IgG4 antibodies, according to the Kabat EU nomenclature (Kabat Et al., 1991), the glycoside structures/glycans are attached to the amino acid residue at position 297 by N-linked glycosylation.

在一具體實施例中,分離本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞。In a specific embodiment, the rat hepatoma cells of the invention or the H4-II-E rat hepatoma cells are isolated.

在又一具體實施例中,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞之特徵進一步在於帶有選擇標記物基因(例如新黴素-磷酸轉移酶(NPT))、針對嘌呤黴素、潮黴素或吉歐黴素之抗性基因或可擴增選擇標記物基因(例如二氫葉酸還原酶(DHFR)或麩醯胺酸合成酶(GS))。在一具體實施例中,該NPT係野生型新黴素-磷酸轉移酶。In still another embodiment, the rat hepatoma cell of the invention or the H4-II-E rat hepatoma cell is further characterized by a selection marker gene (eg, neomycin-phosphotransferase (NPT)). A resistance gene for puromycin, hygromycin or genomicin or an amplifiable selectable marker gene (for example, dihydrofolate reductase (DHFR) or glutamate synthase (GS)). In a specific embodiment, the NPT is a wild type neomycin-phosphotransferase.

在本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞之另一具體實施例中,允許表現該編碼抗體或Fc-融合蛋白之核酸序列之調節序列係a)啟動子或b)增強子或c)5'-UTR序列、或d)轉錄增強(TE)元件。In another specific embodiment of the rat hepatoma cell or H4-II-E rat hepatoma cell of the invention, the regulatory sequence that confers the nucleic acid sequence encoding the antibody or Fc-fusion protein is a) a promoter or b) enhancer or c) 5'-UTR sequence, or d) transcription enhancing (TE) element.

在本發明又一實施例中,該抗體或Fc融合蛋白含有包含以下糖鏈之糖苷結構:In still another embodiment of the invention, the antibody or Fc fusion protein comprises a glycoside structure comprising the following sugar chains:

在本發明另一實施例中,該抗體或Fc融合蛋白含有連接至N-天冬醯胺酸(N-Asn)殘基之糖苷結構,其中該糖苷結構包含以下糖鏈:In another embodiment of the invention, the antibody or Fc fusion protein comprises a glycoside structure linked to a N-aspartic acid (N-Asn) residue, wherein the glycoside structure comprises the following sugar chain:

具體而言,糖苷結構包含以下糖鏈:Specifically, the glycoside structure comprises the following sugar chains:

較佳地,根據Kabat EU命名法(Kabat等人,1991),上述實施例中之該N-Asn係N-Asn(297)。Preferably, the N-Asn system N-Asn (297) in the above examples is according to the Kabat EU nomenclature (Kabat et al., 1991).

在一具體實施例中,聚糖還原端之小於20%、較佳小於10%或小於5%之GlcNAc殘基結合有海藻糖,更佳無海藻糖結合至聚糖還原端之GlcNAc殘基,如藉由以下結構所繪示:In a specific embodiment, less than 20%, preferably less than 10% or less than 5% of the GlcNAc residue of the glycan reducing end is bound to trehalose, and more preferably no trehalose is bound to the GlcNAc residue of the reducing end of the glycan, As shown by the following structure:

or

在另一具體實施例中,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞適應於在無血清且鈣減少或較佳無鈣培養基中生長。In another embodiment, the rat hepatoma cells or H4-II-E rat hepatoma cells of the invention are adapted to grow in serum-free and calcium-reduced or preferably calcium-free medium.

在又一具體實施例中,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞適應於在懸浮培養物中生長。在另一具體實施例中,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞另外適應於在不含任一動物來源之蛋白/肽之培養基中生長。在另一具體實施例中,與YB2/0細胞相比,本發明之大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞對細胞凋亡具有低敏感性及/或對細胞壓力具有高穩健性。In yet another embodiment, the rat hepatoma cells or H4-II-E rat hepatoma cells of the invention are adapted to grow in suspension culture. In another embodiment, the rat hepatoma cells or H4-II-E rat hepatoma cells of the invention are additionally adapted to grow in a medium free of protein/peptide of any animal origin. In another specific embodiment, the rat hepatoma cells or H4-II-E rat hepatoma cells of the invention have low sensitivity to apoptosis and/or have cell stress compared to YB2/0 cells. Highly robust.

本發明進一步闡述產生所關注糖蛋白之方法,其特徵在於以下步驟:The invention further describes a method of producing a glycoprotein of interest, characterized by the following steps:

a) 提供大鼠肝瘤細胞,a) providing rat hepatoma cells,

b) 視情況使該步驟a)之細胞適應於在懸浮培養物中生長,b) adapting the cells of step a) to growth in suspension culture, as appropriate,

c) 視情況使該步驟a)及/或步驟b)之細胞適應於在無血清培養基中生長,c) adapting the cells of step a) and/or step b) to growth in serum-free medium, as appropriate,

d) 視情況使該步驟a)及/或步驟b)及/或步驟c)之細胞在鈣減少或無鈣培養基中生長,d) depending on the condition, the cells of step a) and/or step b) and/or step c) are grown in a calcium-reduced or calcium-free medium,

e) 用編碼所關注重組糖蛋白之核酸序列轉染此視情況經適應之大鼠肝瘤細胞,e) transfecting the optionally adapted rat hepatoma cells with a nucleic acid sequence encoding a recombinant glycoprotein of interest,

f) 在允許表現該所關注糖蛋白之條件下培育該步驟e)之轉染細胞,f) cultivating the transfected cells of step e) under conditions permitting expression of the glycoprotein of interest,

g) 視情況分離並純化該所關注(糖)蛋白。g) The desired (sugar) protein is isolated and purified as appropriate.

在又一實施例中,該大鼠肝瘤細胞係H4-II-E細胞,較佳地,該細胞係:In still another embodiment, the rat hepatoma cell line H4-II-E cells, preferably, the cell line:

i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或i) Cells derived from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E -C3 cell line (CRL-1600 or HPACC No. 85061112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) And the H4-S cell line (HPACC Nr. 89102001), or

ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或Ii) deposited in the European Cell Culture Preservation Center under the numbered ECACC catalog number 87031301 or in the US Type Culture Collection Center ATCC under the accession number CRL-1548, or

iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iii) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E), or

iv)以登錄編號DSM ACC3130(H4-II-ES)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iv) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-ES), or

v) i)或ii)或iii)或iv)中任一細胞之衍生物或子代。v) a derivative or progeny of any of i) or ii) or iii) or iv).

較佳地,該細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。在尤佳實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。Preferably, the cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548. In a particularly preferred embodiment, the rat hepatoma cell or the H4-II-E cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

在一具體實施例中,該步驟b)、c)或d)之培養基另外不含任一動物來源之蛋白/肽。In a specific embodiment, the medium of step b), c) or d) is additionally free of any animal derived protein/peptide.

在又一具體實施例中,該方法之特徵進一步在於轉染步驟e)包含將包含編碼該所關注糖蛋白之核酸序列之表現載體引入該大鼠肝瘤細胞中,該核酸序列可操作地連接至至少一種允許表現該編碼所關注糖蛋白之核酸序列之調節序列。In still another embodiment, the method is further characterized in that the transfection step e) comprises introducing into the rat hepatoma cell an expression vector comprising a nucleic acid sequence encoding the glycoprotein of interest, the nucleic acid sequence being operably linked To at least one regulatory sequence that permits expression of the nucleic acid sequence encoding the glycoprotein of interest.

在另一具體實施例中,該產生所關注糖蛋白之方法之特徵進一步在於培育步驟f)包含使該轉染細胞適應於在懸浮培養物中生長,適應於在無血清培養基中生長,或適應於在鈣還原或無鈣培養基中生長,或適應於在無血清且鈣減少/無鈣培養基中懸浮培養生長。In another embodiment, the method of producing a glycoprotein of interest is further characterized in that the culturing step f) comprises adapting the transfected cells for growth in suspension culture, for growth in serum-free medium, or for adaptation Growth in calcium-reduced or calcium-free medium, or in suspension culture in serum-free and calcium-reduced/calcium-free medium.

在又一具體實施例中,本發明方法之特徵進一步在於該所關注糖蛋白係抗體或Fc-融合蛋白、較佳抗體或Fc-融合蛋白,其具有In still another embodiment, the method of the invention is further characterized by the glycoprotein antibody or Fc-fusion protein, preferably antibody or Fc-fusion protein of interest, having

a) FcγRIIIa結合活性且較佳ADCC,或a) FcγRIIIa binding activity and preferably ADCC, or

b) 補體結合活性且較佳CDC,或b) complement binding activity and better CDC, or

c) 與新生兒Fc受體FcRn之結合活性且較佳血清穩定性、具體而言長半衰期。c) binding activity to the neonatal Fc receptor FcRn and preferably serum stability, in particular long half-life.

本發明進一步闡述產生(重組)抗體或Fc融合蛋白之方法,該蛋白具有The invention further describes methods of producing (recombinant) antibodies or Fc fusion proteins having

a) FcγRIIIa結合活性及/或a) FcγRIIIa binding activity and/or

b) 補體結合活性及/或b) complement binding activity and / or

c) 與新生兒Fc受體FcRn之結合活性,c) binding activity to the neonatal Fc receptor FcRn,

該方法包含在大鼠肝瘤細胞中產生該抗體或Fc融合蛋白,其中該大鼠肝瘤細胞較佳為H4-II-E細胞,更佳地,該細胞係:The method comprises producing the antibody or Fc fusion protein in a rat hepatoma cell, wherein the rat hepatoma cell is preferably a H4-II-E cell, more preferably, the cell line:

i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或i) Cells derived from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E -C3 cell line (CRL-1600 or HPACC No. 85061112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) And the H4-S cell line (HPACC Nr. 89102001), or

ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或Ii) deposited in the European Cell Culture Preservation Center under the numbered ECACC catalog number 87031301 or in the US Type Culture Collection Center ATCC under the accession number CRL-1548, or

iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iii) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E), or

iv) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iv) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or

v) i)或ii)或iii)或iv)中任一細胞之衍生物或子代。v) a derivative or progeny of any of i) or ii) or iii) or iv).

較佳地,該細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。在尤佳實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。Preferably, the cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548. In a particularly preferred embodiment, the rat hepatoma cell or the H4-II-E cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

較佳地,(重組)抗體或Fc融合蛋白係藉由核酸序列編碼,該核酸序列可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之核酸序列之調節序列。Preferably, the (recombinant) antibody or Fc fusion protein is encoded by a nucleic acid sequence operably linked to at least one regulatory sequence that permits expression of the nucleic acid sequence encoding the antibody or Fc-fusion protein.

在一具體實施例中In a specific embodiment

i) 該先前步驟a)之抗體或Fc融合蛋白具有(提高)抗體依賴性細胞毒性(ADCC)或i) the antibody or Fc fusion protein of the previous step a) has (increased) antibody-dependent cellular cytotoxicity (ADCC) or

ii) 該先前步驟b)之抗體或Fc融合蛋白具有(提高)補體依賴性細胞毒性(CDC)或Ii) the antibody or Fc fusion protein of the previous step b) has (increased) complement dependent cytotoxicity (CDC) or

iii) 該先前步驟c)之抗體或Fc融合蛋白具有血清穩定性。具體而言,iii)意指H4-II-E細胞中產生之糖苷結構之末端唾液酸化對治療抗體或Fc-融合蛋白之血清穩定性及分解代謝半衰期具有正面效應。因此,與其他細胞(例如CHO細胞)中產生之抗體或Fc融合蛋白相比,iii)之該等抗體或Fc融合蛋白具有延長半衰期/提高血清穩定性/延長分解代謝半衰期。Iii) The antibody or Fc fusion protein of the previous step c) is serum stable. Specifically, iii) means that the terminal sialylation of the glycoside structure produced in H4-II-E cells has a positive effect on the serum stability and catabolic half-life of the therapeutic antibody or Fc-fusion protein. Thus, such antibodies or Fc fusion proteins of iii) have an extended half-life/increased serum stability/prolonged catabolic half-life compared to antibodies or Fc fusion proteins produced in other cells (eg, CHO cells).

ADCC、CDC活性之提高或半衰期之延長可藉由比較大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞中產生之抗體或Fc融合蛋白之各別活性或半衰期與CHO細胞(具體而言,CHO DG44細胞)中產生之相應抗體或Fc融合蛋白之活性來量測。Increased ADCC, CDC activity or half-life can be achieved by comparing the individual activity or half-life of antibodies or Fc fusion proteins produced in rat hepatoma cells or H4-II-E rat hepatoma cells with CHO cells (specifically The activity of the corresponding antibody or Fc fusion protein produced in CHO DG44 cells was measured.

本發明另外係關於產生具有經促進ADCC之抗體或Fc融合蛋白之方法,其包含將編碼該抗體之DNA引入大鼠肝瘤或H4-II-E大鼠肝瘤細胞中,另外包含在該細胞中培育並產生該抗體。The invention further relates to a method for producing an antibody or Fc fusion protein having an promoted ADCC, which comprises introducing a DNA encoding the antibody into a rat liver tumor or a H4-II-E rat liver tumor cell, additionally comprising the cell The antibody is grown and produced.

本發明另外係關於產生具有經促進CDC之抗體或Fc融合蛋白之方法,其包含將編碼該抗體之DNA引入大鼠肝瘤細胞或H4-II-E大鼠肝瘤細胞中,該方法另外包含在該細胞中培育並產生該抗體。The invention further relates to a method for producing an antibody or Fc fusion protein having a promoted CDC, which comprises introducing a DNA encoding the antibody into a rat hepatoma cell or a H4-II-E rat hepatoma cell, the method additionally comprising The antibody is grown and produced in the cell.

本發明進一步係關於產生具有(經促進/提高)血清穩定性及半衰期之抗體或Fc融合蛋白(尤其若與其他細胞(例如CHO細胞)中產生之抗體或Fc融合蛋白相比)之方法,其包含將編碼該抗體之DNA引入H4-II-E大鼠肝瘤細胞中,該方法進一步包含在該細胞中培育並產生該抗體。The invention further relates to a method of producing an antibody or Fc fusion protein having (promoted/improved) serum stability and half-life, especially if compared to an antibody or Fc fusion protein produced in other cells (eg, CHO cells), Including introducing the DNA encoding the antibody into H4-II-E rat hepatoma cells, the method further comprising culturing and producing the antibody in the cell.

具體而言,產生意指在允許表現抗體或Fc融合蛋白之條件下培育大鼠肝瘤細胞且視情況將其純化並分離,其中該大鼠肝瘤細胞較佳為H4-II-E細胞,更佳地,該細胞係:Specifically, production means that rat hepatoma cells are cultured and optionally purified and isolated under conditions permitting expression of an antibody or Fc fusion protein, wherein the rat hepatoma cells are preferably H4-II-E cells, More preferably, the cell line:

i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或i) Cells derived from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E -C3 cell line (CRL-1600 or HPACC No. 85061112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) And the H4-S cell line (HPACC Nr. 89102001), or

ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或Ii) deposited in the European Cell Culture Preservation Center under the numbered ECACC catalog number 87031301 or in the US Type Culture Collection Center ATCC under the accession number CRL-1548, or

iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iii) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E), or

iv) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iv) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or

v)i)或ii)或iii)或iv)中任一細胞之衍生物或子代。v) a derivative or progeny of any of i) or ii) or iii) or iv).

較佳地,該細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。在尤佳實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。Preferably, the cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548. In a particularly preferred embodiment, the rat hepatoma cell or the H4-II-E cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

在本發明(產生)方法之一具體實施例中,所關注糖蛋白之特徵進一步在於i)所關注糖蛋白(例如抗體或Fc-融合蛋白)中所含糖苷結構含有海藻糖之程度(或比例)為(所有聚糖/糖苷結構之)小於20%、小於10%或小於5%或ii)該所關注糖蛋白(例如該抗體或Fc-融合蛋白)中所含糖苷結構含有海藻糖之程度在(所有聚糖/糖苷結構之)介於0%至20%、0%至10%、0%至5%、0.5%至20%、0.5%至10%、0.5%至5%、1%至20%、1%至10%或1%至5%之間之範圍內。具體而言,所關注糖蛋白(例如抗體或Fc-融合蛋白)之特徵進一步在於該所關注糖蛋白(例如該抗體或Fc-融合蛋白)之小於20%聚糖/糖苷結構含有結合至末端還原N-乙醯葡糖胺(GlcNAc)殘基之海藻糖。較佳地,該等去海藻糖基化糖苷結構/聚糖係N-連接,最佳地,對於IgG1、IgG2、IgG3及IgG4抗體而言,根據Kabat EU命名法(Kabat等人,1991),該等糖苷結構/聚糖係藉由N-連接糖基化附著於第297位胺基酸殘基。In a specific embodiment of the method of the present invention, the glycoprotein of interest is further characterized by i) the degree (or proportion) of the glycosidic structure contained in the glycoprotein of interest (eg, antibody or Fc-fusion protein) containing trehalose Is less than 20%, less than 10% or less than 5% of all glycan/glycoside structures or ii) the degree of glycosidic structure contained in the glycoprotein of interest (eg, the antibody or Fc-fusion protein) contains trehalose Between 0% to 20%, 0% to 10%, 0% to 5%, 0.5% to 20%, 0.5% to 10%, 0.5% to 5%, 1% (of all glycan/glycosidic structures) To the range of 20%, 1% to 10% or 1% to 5%. In particular, the glycoprotein of interest (eg, an antibody or Fc-fusion protein) is further characterized by less than 20% glycan/glycoside structure of the glycoprotein of interest (eg, the antibody or Fc-fusion protein) containing binding to terminal reduction Trehalose of the residue of N-acetylglucosamine (GlcNAc). Preferably, the de-fucosylated glycoside structure/glycans are N-linked, optimally, for IgGl, IgG2, IgG3 and IgG4 antibodies, according to the Kabat EU nomenclature (Kabat et al., 1991), The glycoside structures/glycans are attached to the amino acid residue at position 297 by N-linked glycosylation.

在本發明(產生)方法之另一具體實施例中,所關注糖蛋白之特徵進一步在於i)該所關注糖蛋白(例如該抗體或Fc-融合蛋白)中所含聚糖/糖苷結構、較佳複合型聚糖/糖苷結構含有至少一個、較佳一或兩個或一或多個半乳糖殘基之程度為(所有複合型聚糖/糖苷結構之)超過40%、45%或50%或ii)該所關注糖蛋白(例如該抗體或Fc-融合蛋白)中所含聚糖/糖苷結構、較佳複合型聚糖/糖苷結構含有至少一個、較佳一或兩個或一或多個半乳糖殘基之程度在(所有複合型聚糖/糖苷結構之)介於40%至100%、45%至100%、50%至100%、51%至100%、40%至99.5%、45%至99.5%、50%至99.5%或51%至99.5%、40%至99%、45%至99%、50%至99%或51%至99%之間之範圍內。In another embodiment of the method of the present invention, the glycoprotein of interest is further characterized by i) a glycan/glycosidic structure contained in the glycoprotein of interest (eg, the antibody or Fc-fusion protein), Preferably, the complex glycan/glycosidic structure contains at least one, preferably one or two or one or more galactose residues to a degree (all of the complex glycan/glycosidic structures) of more than 40%, 45% or 50% Or ii) the glycan/glycosidic structure, preferably the complex glycan/glycosidic structure contained in the glycoprotein of interest (eg, the antibody or Fc-fusion protein) contains at least one, preferably one or two or one or more The degree of galactose residues is between 40% to 100%, 45% to 100%, 50% to 100%, 51% to 100%, 40% to 99.5% (all complex glycan/glycosidic structures) , in the range of 45% to 99.5%, 50% to 99.5% or 51% to 99.5%, 40% to 99%, 45% to 99%, 50% to 99% or 51% to 99%.

較佳地,該等半乳糖基化糖苷結構/聚糖含有一或兩個較佳連接至該等糖苷結構之末端非還原處之N-乙醯葡糖胺(GlcNAc)之半乳糖殘基(G1或G2)。較佳地,對於IgG1、IgG2、IgG3及IgG4抗體而言,根據Kabat EU命名法(Kabat等人,1991),該等糖苷結構/聚糖係N-連接於第297位胺基酸殘基。在本發明一具體實施例中,聚糖/糖苷結構係G1或G2。聚糖/糖苷結構較佳不為G0。Preferably, the galactosylated glycoside structures/glycans contain one or two galactose residues of N-acetylglucosamine (GlcNAc) preferably attached to the terminal non-reducing sites of the glycoside structures ( G1 or G2). Preferably, for IgGl, IgG2, IgG3 and IgG4 antibodies, the glycoside structure/glycans are N-linked to the amino acid residue at position 297 according to the Kabat EU nomenclature (Kabat et al., 1991). In a particular embodiment of the invention, the glycan/glycosidic structure is G1 or G2. The glycan/glycoside structure is preferably not G0.

在本發明(產生)方法之又一具體實施例中,所關注糖蛋白之特徵進一步在於i)該糖蛋白(例如該抗體或Fc-融合蛋白)中所含(半乳糖基化)糖苷結構含有末端唾液酸或神經胺酸殘基之程度為超過5%或超過10%或ii)該所關注糖蛋白(例如該抗體或Fc-融合蛋白)中所含(半乳糖基化)糖苷結構含有末端唾液酸或神經胺酸殘基之程度在介於0-8%、1-8%、5-10%、10-50%或10-45%之間之範圍內。較佳地,該等含有末端唾液酸或神經胺酸殘基之糖苷結構/聚糖係N-連接、最佳地,對於IgG1、IgG2、IgG3及IgG4抗體而言,根據Kabat EU命名法(Kabat等人,1991),該等糖苷結構/聚糖藉由N-連接糖基化附著於第297位胺基酸殘基。In still another embodiment of the method of the present invention, the glycoprotein of interest is further characterized by i) a (galactosylated) glycoside structure contained in the glycoprotein (eg, the antibody or Fc-fusion protein) The degree of terminal sialic acid or neuramic acid residues is more than 5% or more than 10% or ii) the (galactosylated) glycoside structure contained in the glycoprotein of interest (eg, the antibody or Fc-fusion protein) contains an end The degree of sialic acid or neuramic acid residues is in the range of 0-8%, 1-8%, 5-10%, 10-50% or 10-45%. Preferably, the glycoside structure/glycans N-linked containing terminal sialic acid or ceramide residues, optimally, for IgG1, IgG2, IgG3 and IgG4 antibodies, according to Kabat EU nomenclature (Kabat Et al., 1991), the glycoside structures/glycans are attached to the amino acid residue at position 297 by N-linked glycosylation.

本發明進一步闡述生成用於產生重組糖蛋白之(宿主)細胞之方法,其包含:The invention further clarifies a method of producing a (host) cell for producing a recombinant glycoprotein, comprising:

a) 提供大鼠肝瘤細胞,a) providing rat hepatoma cells,

b) 使該步驟a)之大鼠肝瘤細胞適應於在懸浮培養物中生長,及b) adapting the rat hepatoma cells of step a) to growth in suspension culture, and

c) 使該步驟a)之大鼠肝瘤細胞適應於在無血清培養基中生長,及c) adapting the rat hepatoma cells of step a) to growth in serum-free medium, and

d) 使該步驟a)之大鼠肝瘤細胞適應於在鈣減少或無鈣培養基中生長,及d) adapting the rat hepatoma cells of step a) to growth in a calcium-reduced or calcium-free medium, and

e) 視情況使該步驟a)之大鼠肝瘤細胞適應於在不含任一動物來源之蛋白/肽之培養基中生長,及e) adapting the rat hepatoma cells of step a) as appropriate to growth in a medium free of protein/peptide of any animal origin, and

f) 視情況選擇單細胞純系,f) select a single cell pure line as appropriate,

g) 獲得(產生)(宿主)細胞。較佳地,該大鼠肝瘤細胞係H4-II-E細胞,更佳地,該細胞係:g) Obtain (produce) (host) cells. Preferably, the rat hepatoma cell line H4-II-E cells, more preferably, the cell line:

i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或i) Cells derived from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E -C3 cell line (CRL-1600 or HPACC No. 85061112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) And the H4-S cell line (HPACC Nr. 89102001), or

ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或Ii) deposited in the European Cell Culture Preservation Center under the numbered ECACC catalog number 87031301 or in the US Type Culture Collection Center ATCC under the accession number CRL-1548, or

iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iii) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E), or

iv) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iv) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or

v) i)或ii)或iii)或iv)中任一細胞之衍生物或子代。v) a derivative or progeny of any of i) or ii) or iii) or iv).

較佳地,該細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。在尤佳實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。Preferably, the cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548. In a particularly preferred embodiment, the rat hepatoma cell or the H4-II-E cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

本發明進一步闡述生成用於產生重組糖蛋白之(宿主)細胞之方法,其包含:The invention further clarifies a method of producing a (host) cell for producing a recombinant glycoprotein, comprising:

a) 提供H4-II-E大鼠肝瘤細胞,a) providing H4-II-E rat hepatoma cells,

b) 使該步驟a)之H4-II-E大鼠肝瘤細胞適應於在懸浮培養物中生長,及b) adapting the H4-II-E rat hepatoma cells of step a) to growth in suspension culture, and

c) 使該步驟a)之H4-II-E大鼠肝瘤細胞適應於在無血清培養基中生長,及c) adapting the H4-II-E rat hepatoma cells of step a) to growth in serum-free medium, and

d) 使該步驟a)之H4-II-E大鼠肝瘤細胞適應於在鈣減少或無鈣培養基中生長,及d) adapting the H4-II-E rat hepatoma cells of step a) to growth in a calcium-reduced or calcium-free medium, and

e) 視情況使該步驟a)之H4-II-E大鼠肝瘤細胞適應於在不含任一動物來源之蛋白/肽之培養基中生長,及e) adapting the H4-II-E rat hepatoma cells of step a) to a medium cultured in a protein/peptide free of any animal source, as appropriate, and

f) 視情況選擇單細胞純系,f) select a single cell pure line as appropriate,

g) 獲得(宿主)細胞。g) Obtain (host) cells.

在一具體實施例中,該方法進一步包含:In a specific embodiment, the method further comprises:

h) 用編碼所關注糖蛋白核酸序列轉染該步驟g)之獲得之H4-II-E大鼠肝瘤宿主細胞,及h) transfecting the H4-II-E rat hepatoma host cell obtained in step g) with a glycoprotein nucleic acid sequence encoding the same, and

i) 視情況在允許表現該所關注糖蛋白之條件下培育該步驟h)之轉染細胞。i) The transfected cells of step h) are incubated under conditions permitting expression of the glycoprotein of interest.

較佳地,該所關注糖蛋白係抗體或Fc融合蛋白、最佳抗體或Fc融合蛋白具有ADCC及/或CDC及/或血清穩定性、及/或具體而言長半衰期。Preferably, the glycoprotein antibody or Fc fusion protein of interest, the optimal antibody or Fc fusion protein has ADCC and/or CDC and/or serum stability, and/or specifically a long half-life.

較佳地,該抗體係人類抗體、人類化抗體、人類嵌合抗體或人類CDR-接枝抗體。Preferably, the anti-system human antibody, humanized antibody, human chimeric antibody or human CDR-grafted antibody.

本發明進一步係關於根據該如上文所述用於產生重組糖蛋白之宿主細胞之方法生成的(宿主)細胞。The invention further relates to a (host) cell produced according to the method of the host cell for producing a recombinant glycoprotein as described above.

本發明進一步係關於以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之(大鼠肝瘤)細胞。The present invention further relates to (rat hepatoma) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es).

本發明進一步係關於以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之(大鼠肝瘤)細胞。The present invention further relates to (rat hepatoma) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E).

本發明另外係關於大鼠肝瘤細胞作為宿主細胞用於生物醫藥生產或作為宿主細胞用於產生(重組)糖蛋白、較佳抗體或Fc融合蛋白之用途。較佳地,該大鼠肝瘤細胞係H4-II-E細胞,更佳地,該細胞係:The invention further relates to the use of rat hepatoma cells as host cells for biopharmaceutical production or as host cells for the production of (recombinant) glycoproteins, preferably antibodies or Fc fusion proteins. Preferably, the rat hepatoma cell line H4-II-E cells, more preferably, the cell line:

i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或i) Cells derived from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E -C3 cell line (CRL-1600 or HPACC No. 85061112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) And the H4-S cell line (HPACC Nr. 89102001), or

ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或Ii) deposited in the European Cell Culture Preservation Center under the numbered ECACC catalog number 87031301 or in the US Type Culture Collection Center ATCC under the accession number CRL-1548, or

iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iii) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E), or

iv) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或Iv) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or

v) i)或ii)或iii)或iv)中任一細胞之衍生物或子代。v) a derivative or progeny of any of i) or ii) or iii) or iv).

較佳地,該細胞係具有ECACC目錄編號87031301或ATCC編號CRL-1548之細胞。在尤佳實施例中,該大鼠肝瘤細胞或該H4-II-E細胞係以登錄編號DSM ACC3129(H4-II-E)或DSM ACC3130(H4-II-Es)寄存於DSMZ之細胞。Preferably, the cell line has cells of ECACC catalog number 87031301 or ATCC number CRL-1548. In a particularly preferred embodiment, the rat hepatoma cell or the H4-II-E cell line is deposited in a DSMZ cell with accession number DSM ACC3129 (H4-II-E) or DSM ACC3130 (H4-II-Es).

除非另有說明,否則本發明實踐將採用業內人士所熟習之細胞生物學、分子生物學、細胞培養、免疫學及諸如此類之習用技術。該等技術完全揭示於現行文獻中。Unless otherwise indicated, the practice of the present invention will employ conventional techniques of cell biology, molecular biology, cell culture, immunology, and the like, which are well known in the art. These techniques are fully disclosed in the current literature.

本說明書中所提及之所有出版物及專利皆表示彼等熟習本發明所涉及之技術者之熟練程度。本文所引用所有出版物及專利說明書之全文皆以引用方式併入本文中,以更全面地闡述本發明所涉及之技術狀態。藉由參照以下實例可更容易地理解上文所概述之本發明,本文僅出於闡釋本發明某些實施例之目的包括該實例,而非欲以任何方式限制本發明。All publications and patents referred to in this specification are indicative of the skill of those skilled in the art. The disclosures of all publications and patent specifications cited herein are hereby incorporated by reference in their entirety in their entirety in their entirety herein The invention as outlined above is more readily understood by reference to the following examples, which are intended to be illustrative only.

實驗experiment 材料及方法Materials and methods DNA構築體DNA construct

用於穩定轉染不同細胞系之質粒DNA在病毒或遍在性啟動子、以及選擇標記物neo及dhfr控制下編碼IgG1抗體重鏈及輕鏈。Plasmid DNA for stable transfection of different cell lines encodes the IgGl antibody heavy and light chains under the control of viral or ubiquitous promoters, as well as selection markers neo and dhfr.

細胞系及細胞培育Cell line and cell culture

先前已闡述缺乏二氫葉酸還原酶之CHO細胞系CHO/DG44(Urlaub及Chasin,1980)及缺乏內源性GMD之變體CHO細胞系Pro-Lec13.6A(Ripka等人,1986)。大鼠融合瘤細胞系YB2/0以及大鼠肝瘤細胞系H4-II-E源自歐洲細胞培養物保存中心(ECACC,目錄編號85110501及87031301)。The CHO cell line CHO/DG44 lacking dihydrofolate reductase (Urlaub and Chasin, 1980) and the variant CHO cell line Pro-Lec 13.6A lacking endogenous GMD have been previously described (Ripka et al., 1986). The rat fusion tumor cell line YB2/0 and the rat liver tumor cell line H4-II-E were derived from the European Cell Culture Preservation Center (ECACC, Cat. No. 85110501 and 87031301).

本發明H4-II-E細胞系已於2011年6月28日根據布達佩斯條約以登錄編號DSM ACC3129(H4-II-E)寄存於德國微生物菌種保存中心(DSMZ),Inhoffenstrasse 7B,D-38124 Braunschweig,Germany。在含有5% FCS之MEMalpha(Invitrogen)或EMEM中培養H4-II-E細胞(例如以登錄編號DSM ACC3129寄存於DSMZ之細胞)。每3至4天用胰蛋白酶/EDTA(Invitrogen)將貼壁細胞胰蛋白酶化並以約20,000-30,000個細胞/cm2之細胞密度接種於組織培養物處理板或T形燒瓶(Nunc,Denmark)中之新鮮培養基中。The H4-II-E cell line of the present invention was deposited with the German Collection of Microbial Species (DSMZ), Inhoffenstrasse 7B, D-38124, on June 28, 2011 under the Budapest Treaty under the accession number DSM ACC3129 (H4-II-E). Braunschweig, Germany. H4-II-E cells (e.g., cells deposited with DSMZ under accession number DSM ACC3129) were cultured in MEMalpha (Invitrogen) or EMEM containing 5% FCS. The adherent cells were trypsinized with trypsin/EDTA (Invitrogen) every 3 to 4 days and seeded in tissue culture treatment plates or T-flasks (Nunc, Denmark) at a cell density of about 20,000-30,000 cells/cm 2 . In the fresh medium.

適應於在本發明中所述無血清、無Ca2+培養基中懸浮生長之H4-II-E細胞系已於2011年6月28日根據布達佩斯條約以登錄編號DSM ACC3130(H4-II-Es)寄存於德國微生物菌種保存中心(DSMZ),Inhoffenstrasse 7B,D-38124 Braunschweig,Germany。在37℃之溫度下且在含有5% CO2之氣氛中在振盪燒瓶(Corning)中以100-120 rpm培養適應於在無血清、無Ca2+培養基中懸浮生長之H4-II-E細胞,例如以登錄編號DSM ACC3130寄存於DSMZ之細胞。每3至4天對H4-II-E懸浮培養物進行傳代培育,且接種密度為300 000-400 000個細胞/mL。The H4-II-E cell line adapted for suspension growth in the serum-free, Ca 2+-free medium of the present invention was deposited on June 28, 2011 under the Budapest Treaty under the accession number DSM ACC3130 (H4-II-Es). Registered at the German Collection of Microorganisms (DSMZ), Inhoffenstrasse 7B, D-38124 Braunschweig, Germany. H4-II-E cells adapted to suspension growth in serum-free, Ca 2+-free medium were incubated at 37 ° C in an atmosphere containing 5% CO 2 in a shaking flask (Corning) at 100-120 rpm. For example, the cells deposited in DSMZ with the accession number DSM ACC3130. H4-II-E suspension cultures were subcultured every 3 to 4 days and seeded at a density of 300 000-400 000 cells/mL.

在37℃之溫度下且在含有5% CO2之氣氛中在表面充氣T形燒瓶(Nunc,Denmark)或振盪燒瓶(Corning)中以100-120 rpm在培養器(Thermo,Germany)中在無血清培養基中懸浮培育CHO-DG44細胞以及經適應之CHO-Lec13細胞、YB2/0細胞及H4-II-E細胞。每2至3天對懸浮培養物進行傳代培育,且接種密度為200 000-300 000個細胞/mL細胞/mL。藉由使用血球計來測定所有培養物中之細胞濃度。藉由錐蟲藍排除法來評價活力。在BI-專利培養基或MEM Joklik Modification(Sigma)或MEM Spinner Modification(Sigma)中培養所有懸浮培養物。In a surface aerated T-flask (Nunc, Denmark) or shake flask (Corning) at a temperature of 37 ° C and in a 5% CO 2 atmosphere at 100-120 rpm in an incubator (Thermo, Germany) CHO-DG44 cells and adapted CHO-Lec13 cells, YB2/0 cells and H4-II-E cells were cultured in suspension in serum medium. Suspension cultures were subcultured every 2 to 3 days and seeded at a density of 200 000-300 000 cells/mL cells/mL. Cell concentrations in all cultures were determined by using a hemocytometer. Vitality was assessed by trypan blue exclusion. All suspension cultures were cultured in BI-patent medium or MEM Joklik Modification (Sigma) or MEM Spinner Modification (Sigma).

對於糖基化性質之預測分析Predictive analysis of glycosylation properties

選擇不同細胞系並分析其糖基化機器中所涉及酶之表現程度。進一步分析該等細胞系之凝集素(碳水化合物特異性結合蛋白)與含糖蛋白細胞表面之結合。將對每一細胞系獲得之值正規化至對CHO獲得之值,CHO係重組蛋白之習知生產系統且展示經充分表徵之糖基化模式。在細胞表面上存在某些糖結構係細胞糖基化能力之指標。Different cell lines were selected and analyzed for the degree of performance of the enzymes involved in the glycosylation machinery. The binding of lectins (carbohydrate-specific binding proteins) of these cell lines to the surface of glycoprotein-containing cells was further analyzed. The values obtained for each cell line were normalized to values obtained for CHO, a conventional production system for CHO-based recombinant proteins and exhibiting a well characterized glycosylation pattern. There are indicators of the glycosylation ability of certain sugar structural cells on the cell surface.

穩定轉染細胞群之轉染及分離Transfection and isolation of stably transfected cell populations

根據由製造商提供之導則用LipofectamineTM及PLUSTM試劑(皆為Invitrogen,Germany)或藉由核轉染(Amaxa/Lonza)轉染細胞系。The guide provided by the manufacturer and then with Lipofectamine TM PLUS TM reagent (both Invitrogen, Germany), or by nucleofection (Amaxa / Lonza) transfected cell lines.

為分離穩定轉染之IgG1產生細胞群,48小時後,在選擇性抗生素G418及MTX存在下對細胞實施轉染並平鋪。在約3週選擇後分離存活細胞群並藉由ELISA使用來自細胞培養上清液之試樣分析其產量。To isolate stably transfected IgGl producing cell populations, 48 hours later, cells were transfected and plated in the presence of the selective antibiotics G418 and MTX. Surviving cell populations were isolated after approximately 3 weeks of selection and analyzed by ELISA using samples from cell culture supernatants.

無血清進料分批培養Serum-free feed batch culture

使H4-II-E產生細胞適應於在無血清培養基中懸浮生長。對於無血清進料分批培育而言,以400,000個細胞/ml或600,000個細胞/ml將經適應細胞接種於BI-專利產生培養基或MEM Spinner Modification(Sigma)中且在37℃及5% CO2下以120 rpm攪拌培養物。每日測定包括pH、葡萄糖及乳酸鹽濃度在內之培養參數。視需要使用NaCO3將pH調節至pH 7.0,進給葡萄糖以維持約4 g/L之葡萄糖含量。藉由錐蟲藍排除使用自動化CEDEX細胞量化系統(Innovatis)測定細胞密度及活力。藉由對所述人類IgG具有特異性之酶連接免疫吸附分析(ELISA)來量測培養上清液中之抗體或Fc-融合蛋白濃度。The H4-II-E producing cells were adapted to grow in suspension in serum-free medium. For serum-free fed batch culture, the adapted cells were seeded in BI-patented production medium or MEM Spinner Modification (Sigma) at 400,000 cells/ml or 600,000 cells/ml and at 37 ° C and 5% CO The culture was stirred at 120 rpm under 2 hours. Culture parameters including pH, glucose, and lactate concentrations were measured daily. The pH was adjusted to pH 7.0 using NaCO 3 as needed, and glucose was fed to maintain a glucose content of about 4 g/L. Cell density and viability were determined by trypan blue exclusion using an automated CEDEX cell quantification system (Innovatis). The antibody or Fc-fusion protein concentration in the culture supernatant is measured by an enzyme-linked immunosorbent assay (ELISA) specific for the human IgG.

在懸浮適應之前,可使用貼壁H4-II-E產生純系在無血清培養基中產生固定進料分批培養物中之材料。使H4-II-E IgG1-產生細胞在Hyperflasks(Corning)中在含有5% FCS之MEMalpha中生長至匯合。然後用無血清BI-專利培養基或無血清商業培養基(例如HyClone SFM4(Thermo Fisher Scientific))替代培養基。在培養13天後,使用MabSelect(Amersham)自培養基純化抗體並將其儲存於10 mM檸檬酸鹽/0.15 M NaCl(pH 6.0)中。Adherent H4-II-E can be used to produce materials in a fixed feed batch culture in serum-free medium prior to suspension adaptation. H4-II-E IgG1-producing cells were grown to confluence in Hyperflasks (Corning) in MEMalpha containing 5% FCS. The medium is then replaced with serum-free BI-patented medium or serum-free commercial medium (eg, HyClone SFM4 (Thermo Fisher Scientific)). After 13 days of culture, the antibody was purified from the culture medium using MabSelect (Amersham) and stored in 10 mM citrate / 0.15 M NaCl (pH 6.0).

以300,000個細胞/ml將CHO-DG44、CHO-Lec13及YB2/0 IgG1產生細胞接種於BI-專利產生培養基中且在37℃及5% CO2下以120 rpm攪拌培養物,CO2稍後隨著細胞數量增加而降至2%。每日測定包括pH、葡萄糖及乳酸鹽濃度在內之培養參數。視需要使用NaCO3將pH調節至pH 7.0,進給葡萄糖以維持約4 g/L之葡萄糖含量。每24 hr添加BI-專利進料溶液。藉由錐蟲藍排除使用自動化CEDEX細胞量化系統(Innovatis)測定細胞密度及活力。CHO-DG44, CHO-Lec13 and YB2/0 IgG1 producing cells were seeded in BI-patent producing medium at 300,000 cells/ml and the culture was stirred at 120 rpm at 37 ° C and 5% CO 2 , CO 2 later It decreased to 2% as the number of cells increased. Culture parameters including pH, glucose, and lactate concentrations were measured daily. The pH was adjusted to pH 7.0 using NaCO 3 as needed, and glucose was fed to maintain a glucose content of about 4 g/L. The BI-patented feed solution was added every 24 hr. Cell density and viability were determined by trypan blue exclusion using an automated CEDEX cell quantification system (Innovatis).

藉由對所述人類IgG具有特異性之酶連接免疫吸附分析(ELISA)來量測培養上清液中之抗體濃度。The concentration of the antibody in the culture supernatant was measured by an enzyme-linked immunosorbent assay (ELISA) specific for the human IgG.

ELISAELISA

經由夾心ELISA技術實施細胞培養物上清液中之IgG分子之量化。在4℃下使用山羊抗人類IgGFc片段抗體(Dianova,Germany)塗佈ELISA板過夜。在用1% BSA溶液洗滌並封阻各板後,添加試樣並將其培育1.5小時。在洗滌後,添加檢測抗體(鹼性磷酸酶偶聯山羊抗人類κ輕鏈抗體)並藉由與作為受質之4-硝基苯基磷酸二鈉鹽六水合物(Sigma,Germany)一起培育來實施比色檢測。在黑暗中培育20 min後,終止反應且立即使用吸光度讀數器(Tecan,Germany)以405/492 nm量測吸光度。根據存在於各板上之標準曲線計算濃度。Quantification of IgG molecules in cell culture supernatants was performed via a sandwich ELISA technique. ELISA plates were coated overnight at 4 °C using goat anti-human IgG Fc fragment antibody (Dianova, Germany). After washing and blocking the plates with a 1% BSA solution, samples were added and incubated for 1.5 hours. After washing, a detection antibody (alkaline phosphatase-conjugated goat anti-human kappa light chain antibody) was added and incubated with 4-nitrophenyl phosphate disodium salt hexahydrate (Sigma, Germany) as a substrate. To perform colorimetric detection. After incubation for 20 min in the dark, the reaction was stopped and the absorbance was measured immediately at 405/492 nm using an absorbance reader (Tecan, Germany). The concentration is calculated from the standard curve present on each plate.

自細胞培養上清液純化IgG1Purification of IgG1 from cell culture supernatant

藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體且將其儲存於10 mM檸檬酸鹽/0.15 M NaCl(pH 6.0)中。藉由蛋白質A-HPLC來量測純化抗體之濃度。Recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences) and stored in 10 mM citrate / 0.15 M NaCl (pH 6.0). The concentration of the purified antibody was measured by protein A-HPLC.

對糖基化模式之分析Analysis of glycosylation patterns

為闡明不同細胞系中所產生IgG之Fc-糖基化之結構及組成,在藉由用PNGase F實施酶促消化來還原後自純化抗體釋放聚糖。將聚糖純化,用2-胺基苯甲醯胺(2-AB)進行螢光標記且在用外切糖苷酶(例如α-甘露糖苷酶、神經胺酸酶、β-半乳糖苷酶、α-半乳糖苷酶、β-己糖胺酶及α-海藻糖苷酶)處理之前及之後,在HPLC管柱上進行分級分離。自外切糖苷消化之前及之後之層析峰面積比率來計算海藻糖基化二枝結構聚糖對非海藻糖基二枝結構聚糖、及其他糖苷結構之百分比並對糖結構及組成進行定性及定量驗證。To elucidate the structure and composition of Fc-glycosylation of IgG produced in different cell lines, glycans were released from purified antibodies after reduction by enzymatic digestion with PNGase F. The glycan is purified, fluorescently labeled with 2-aminobenzamide (2-AB) and used with exoglycosidases (eg alpha-mannosidase, neuraminidase, beta-galactosidase, Fractionation was carried out on HPLC columns before and after treatment with α-galactosidase, β-hexosaminidase and α-trehalosidase. Calculate the percentage of non-trehalose-based twig-structured glycans and other glycoside structures from the ratio of chromatographic peak areas before and after digestion with exoglycosides and characterize the sugar structure and composition And quantitative verification.

FcγRIIIa-結合分析FcγRIIIa-binding assay

如下使用BIAcore T100儀器及CM5感測器晶片(BIACORE,Uppsala,Sweden)來量測不同細胞系中產生之IgG1與FcgRIIIa之結合動力學。將可溶性重組FcgRIIIa固定至BIAcore感測器晶片上。以6種不同濃度(自4.17 nM至133.3 nM)將經純化IgG1稀釋於HBS-EP緩衝液(0.01 M HEPES,0.15 M NaC1,3 mM EDTA,0.005%表面活性劑P20,pH 7.4)中,且以5 mL/min之流速將每一經稀釋IgG1注射於受體捕獲感測器表面上。在25℃下利用HBS-EP作為運行緩衝液來實施該等實驗。將無試樣IgG1之緩衝溶液注射於受體捕獲感測器表面上作為空白對照。藉由以10 mL/min之流速將7.5 mM HCl注射30 s來去除結合至感測器表面之可溶性FcgRIIIa及IgG1。在數據分析前針對空白對照校正藉由注射IgG1獲得之數據。藉由穩態分析使用BIAcore T100動力學評估軟體(BIACORE)來計算FcgRIIIa之親和力(KD)。BIAcore T100 instruments and CM5 sensor wafers (BIACORE, Uppsala, Sweden) were used to measure the binding kinetics of IgGl and FcgRIIIa produced in different cell lines as follows. Soluble recombinant FcgRIIIa was immobilized onto a BIAcore sensor wafer. Purified IgG1 was diluted in HBS-EP buffer (0.01 M HEPES, 0.15 M NaC1, 3 mM EDTA, 0.005% Surfactant P20, pH 7.4) at 6 different concentrations (from 4.17 nM to 133.3 nM), and Each diluted IgGl was injected onto the surface of the receptor capture sensor at a flow rate of 5 mL/min. The experiments were carried out using HBS-EP as a running buffer at 25 °C. A buffer solution without sample IgG1 was injected onto the surface of the receptor capture sensor as a blank control. Soluble FcgRIIIa and IgG1 bound to the sensor surface were removed by injecting 7.5 mM HCl for 30 s at a flow rate of 10 mL/min. Data obtained by injection of IgG1 were corrected for blank controls prior to data analysis. Affinity (KD) of FcgRIIIa was calculated by steady state analysis using BIAcore T100 Kinetic Evaluation Software (BIACORE).

FcγRIIb-結合分析FcγRIIb-binding assay

如下使用BIAcore T100儀器及CM5感測器晶片(BIACORE,Uppsala,Sweden)來量測不同細胞系中產生之IgG1與FcgRIIIb之結合動力學。將可溶性重組FcgRIIIa固定至BIAcore感測器晶片上。以6種不同濃度(自4.17 nM至133.3 nM)將經純化IgG1稀釋於HBS-EP緩衝液(0.01 MHEPES,0.15 M NaC1,3 mM EDTA,0.005%表面活性劑P20,pH 7.4)中,且以5 mL/min之流速將每一經稀釋IgG1注射於受體捕獲感測器表面上。在25℃下利用HBS-EP作為運行緩衝液來實施該等實驗。將無試樣IgG1之緩衝溶液注射於受體捕獲感測器表面上作為空白對照。藉由以10 mL/min之流速將7.5 mM HCl注射30 s來去除結合至感測器表面之可溶性FcgRIIb及IgG1。在數據分析前針對空白對照校正藉由注射IgG1獲得之數據。藉由穩態分析使用BIAcore T100動力學評估軟體(BIACORE)來計算對FcgRIIb之親和力(KD)。The binding kinetics of IgGl and FcgRIIIb produced in different cell lines were measured using a BIAcore T100 instrument and a CM5 sensor wafer (BIACORE, Uppsala, Sweden) as follows. Soluble recombinant FcgRIIIa was immobilized onto a BIAcore sensor wafer. Purified IgG1 was diluted in HBS-EP buffer (0.01 MHEPES, 0.15 M NaC1, 3 mM EDTA, 0.005% Surfactant P20, pH 7.4) at 6 different concentrations (from 4.17 nM to 133.3 nM) and A flow rate of 5 mL/min injected each diluted IgGl onto the surface of the receptor capture sensor. The experiments were carried out using HBS-EP as a running buffer at 25 °C. A buffer solution without sample IgG1 was injected onto the surface of the receptor capture sensor as a blank control. Soluble FcgRIIb and IgGl bound to the sensor surface were removed by injecting 7.5 mM HCl for 30 s at a flow rate of 10 mL/min. Data obtained by injection of IgG1 were corrected for blank controls prior to data analysis. Affinity (KD) for FcgRIIb was calculated by steady state analysis using BIAcore T100 Kinetic Evaluation Software (BIACORE).

ADCC分析ADCC analysis

藉由乳酸脫氫酶(LDH)釋放分析來實施ADCC分析,其中使用藉由Lymphoprep(AXIS SHIELD,Dundee,UK)自健康供體製備之人類外周血液單核細胞(PBMC)作為效應子細胞。將靶腫瘤細胞(表現人類CD20之人類伯基特氏淋巴瘤細胞系Ramos(human Burkitt's lymphoma cell line Ramos)或HER2-陽性乳癌細胞系)之等分試樣分配至96孔U形底板(存於50 μl/孔中之10,000個細胞)中且在PBMC(100 μL)存在下以20/1之E/T比率與抗體之連續稀釋液(50 μL)一起培育。在37℃下培育4 h後,使用非放射性細胞毒性分析套組(Promega,Madison,WI)量測上清液LDH活性。根據下式自試樣活性計算特異性細胞溶解%:特異性溶解[%]=100×(E-SE-ST)/(M-ST),其中E係實驗釋放量(來自與抗體及效應子細胞一起培育之靶細胞之上清液中之活性),SE係在效應子細胞存在下之自發釋放量(來自單獨使用培養基之效應子細胞之上清液中之活性),ST係靶細胞之自發釋放量(來自單獨用培養基培育之靶細胞之上清液中之活性),且M係靶細胞之最大釋放量(自用9% Triton X-100溶解之靶細胞釋放之活性)。ADCC analysis was performed by lactate dehydrogenase (LDH) release assay using human peripheral blood mononuclear cells (PBMC) prepared from healthy donors by Lymphoprep (AXIS SHIELD, Dundee, UK) as effector cells. Aliquots of target tumor cells (human Burkitt's lymphoma cell line Ramos or HER2-positive breast cancer cell lines expressing human CD20) to a 96-well U-shaped substrate (stored in Incubate with contiguous dilutions of antibody (50 μL) at an E/T ratio of 20/1 in the presence of PBMC (100 μL) in 10,000 cells in 50 μl/well. Supernatant LDH activity was measured using a non-radioactive cytotoxicity assay kit (Promega, Madison, WI) after incubation for 4 h at 37 °C. Calculate the specific cell lysis % from the sample activity according to the following formula: specific dissolution [%] = 100 × (ES E - S T ) / (MS T ), where E is the experimental release amount (from the antibody and effector cells) The activity of the supernatant in the supernatant of the target cells cultured together, the spontaneous release of the S E in the presence of effector cells (the activity in the supernatant from the effector cells using the medium alone), the S T- target cells The spontaneous release amount (activity from the supernatant above the target cells cultured with the medium alone), and the maximum release amount of the M-type target cells (the activity released from the target cells dissolved with 9% Triton X-100).

Clq-結合分析Clq-binding analysis

藉由流式細胞計數分析使用純化人類補體Clq來研究每一純化IgG結合補體之Clq組份之能力。將表現人類CD20之人類伯基特氏淋巴瘤細胞系Ramos或HER2-陽性乳癌細胞系調節至2×106個細胞/mL且在含有1%(w/v) BSA之PBS中與抗人類CD20 IgG或抗人類HER2之連續稀釋液一起培育30 min。在用含有1%(w/v) BSA之PBS洗滌後,以20 mg/mL之最終濃度添加經純化人類補體Clq(Biogenesis Ltd,Poole,UK)並在37℃下使其與結合細胞之IgG結合30 min。然後洗滌細胞並將其與針對人類Clq之異硫氰酸螢光素偶聯多株抗體(Acris Antibodies GmbH,Hiddenhausen,Germany)一起培育30 min。藉由流式細胞術使用FACSCalibur分析經染色細胞。Purified human complement Clq was used to investigate the ability of each purified IgG to bind to the Clq component of complement by flow cytometric analysis. The human Burkitt's lymphoma cell line Ramos or HER2-positive breast cancer cell line expressing human CD20 was adjusted to 2×10 6 cells/mL and in anti-human CD20 in PBS containing 1% (w/v) BSA. Serial dilutions of IgG or anti-human HER2 were incubated for 30 min. After washing with PBS containing 1% (w/v) BSA, purified human complement Clq (Biogenesis Ltd, Poole, UK) was added at a final concentration of 20 mg/mL and IgG was conjugated to the cells at 37 °C. Combined for 30 min. The cells were then washed and incubated with luciferin isothiocyanate conjugated to human Clq (Acris Antibodies GmbH, Hiddenhausen, Germany) for 30 min. The stained cells were analyzed by flow cytometry using a FACSCalibur.

CDC分析CDC analysis

藉由LDH分析來測定CDC活性。簡言之,在37℃下在96孔平底板(Greiner)中將表現人類CD20之靶人類伯基特氏淋巴瘤細胞系Ramos或HER2-陽性乳癌細胞系、2倍稀釋人類血清補體(Sigma-Aldrich)及抗人類CD20或抗人類HER2-IgG1之連續稀釋液培育3 h。將細胞增殖LDH試劑(Roche Diagnostics,Basel,Switzerland)添加至各孔(15 μL/孔)中且在37℃下培育30 min。使用微量板讀數器(Tecan,Germany)在492 nm下量測各孔中之吸光度且以作為活細胞數指數之相對吸光度單位(RAU)表示。根據下式計算CDC%:CDC活性[%]=100×(RAU背景-RAU測試)/RAU背景CDC activity was determined by LDH analysis. Briefly, the human CD20 target human Burkitt's lymphoma cell line Ramos or HER2-positive breast cancer cell line, 2-fold diluted human serum complement (Sigma-) in a 96-well flat bottom plate (Greiner) at 37 °C. Aldrich) and serial dilutions of anti-human CD20 or anti-human HER2-IgG1 were incubated for 3 h. Cell proliferation LDH reagent (Roche Diagnostics, Basel, Switzerland) was added to each well (15 μL/well) and incubated at 37 ° C for 30 min. The absorbance in each well was measured at 492 nm using a microplate reader (Tecan, Germany) and expressed as relative absorbance units (RAU) as a viable cell number index. CDC% was calculated according to the following formula: CDC activity [%] = 100 x (RAU background - RAU test ) / RAU background .

FcRn-結合分析FcRn-binding assay

在CHO/DG44細胞中表現重組可溶性人類FcRn-b2微球蛋白複合物且藉由Ni-NTA層析(Qiagen)自培養上清液純化。使用BIAcore T100儀器及CM5感測器晶片來量測人類IgG1-FcRn相互作用之動力學。使用胺偶合套組(BIACORE)將抗人類b2-微球蛋白單株抗體(Abcam,Cambridge,UK)固定至晶片上。藉由以5 mL/min之流速注射可溶性FcRn-b2微球蛋白複合物,藉由經固定抗b2-微球蛋白抗體來捕獲該複合物。將無該複合物之緩衝溶液注射於抗體捕獲感測器表面上作為空白對照。以5種不同濃度(自4.17 nM至66.7 nM)將每一經純化IgG稀釋於pH調節至6.0之HBS-EP+緩衝液(0.01 M HEPES,0.15 M NaCl,3 mM EDTA,0.05%表面活性劑P20)中,且以5 mL/min之流速將每一經稀釋IgG1注射於複合物捕獲感測器表面或空白上。藉由以60 mL/min之流速將7.5 mM HCl注射1 min來去除結合至感測器表面上之可溶性FcRn及IgG1。在25℃下利用HBS-EP+作為運行緩衝液來實施該等實驗。使用藉由減去空白獲得之數據進行數據分析。使用BIAcore T100評估軟體藉由二元擬合模型來計算表觀締合速率常數(ka)、解離速率常數(kd)及結合親和力(KD)。The recombinant soluble human FcRn-b2 microglobulin complex was expressed in CHO/DG44 cells and purified from the culture supernatant by Ni-NTA chromatography (Qiagen). The kinetics of the human IgGl-FcRn interaction was measured using a BIAcore T100 instrument and a CM5 sensor wafer. Anti-human b2-microglobulin monoclonal antibody (Abeam, Cambridge, UK) was immobilized onto the wafer using an amine coupling kit (BIACORE). The complex was captured by immobilization of an anti-b2-microglobulin antibody by injecting a soluble FcRn-b2 microglobulin complex at a flow rate of 5 mL/min. A buffer solution without the complex was injected onto the surface of the antibody capture sensor as a blank control. Each purified IgG was diluted in 5 different concentrations (from 4.17 nM to 66.7 nM) to HBS-EP+ buffer (0.01 M HEPES, 0.15 M NaCl, 3 mM EDTA, 0.05% Surfactant P20) adjusted to pH 6.0. Each diluted IgGl was injected onto the composite capture sensor surface or blank at a flow rate of 5 mL/min. Soluble FcRn and IgGl bound to the surface of the sensor were removed by injecting 7.5 mM HCl for 1 min at a flow rate of 60 mL/min. These experiments were carried out using HBS-EP+ as a running buffer at 25 °C. Data analysis was performed using data obtained by subtracting blanks. Apparent association rate constants (ka), dissociation rate constants (kd), and binding affinities (KD) were calculated using a BIAcore T100 evaluation software by a binary fit model.

小鼠之藥物代謝動力學分析Pharmacokinetic analysis of mice

對於CHO或H4-II-E細胞中產生之經純化抗人類CD20IgG1而言,向13週齡雌性ddY小鼠(Charles River Laboratories)之尾靜脈中注射20 mg IgG1。在0.083 h(5 min)、0.5 h、1 h、6 h、24 h、60 h、120 h、216 h、312 h及384 h時自尾靜脈取外周血液試樣,且藉由如先前所述對人類IgG1具有特異性之ELISA來量測血漿中之抗體濃度。自消除β-相之斜率計算所投與IgG1之血清半衰期。For the purified anti-human CD20 IgG1 produced in CHO or H4-II-E cells, 20 mg of IgG1 was injected into the tail vein of 13-week-old female ddY mice (Charles River Laboratories). Peripheral blood samples were taken from the tail vein at 0.083 h (5 min), 0.5 h, 1 h, 6 h, 24 h, 60 h, 120 h, 216 h, 312 h, and 384 h, and as previously An ELISA specific for human IgG1 is described to measure the antibody concentration in plasma. The serum half-life of IgG1 administered was calculated by eliminating the slope of the β-phase.

實例Instance 實例1:預測顯示細胞系之間在分泌蛋白上糖結構之相對含量上具有顯著差異Example 1: Predictions show significant differences in the relative amounts of glycostructures on secreted proteins between cell lines

選擇源自不同物種且在該等物種內源自不同組織或細胞譜系之細胞系並分析其表面結構及酶促活性(圖1)。基於分析結果,可賦予每一細胞系合成某些糖基化模式之能力。分析結果顯示細胞系在其糖基化性質上端視其所源自之物種與組織或細胞譜系二者而有所不同。僅少數細胞系天然顯示低海藻糖基化能力。一些細胞系潛在地生成唾液酸化結構且若干細胞系形成免疫原性糖結構(Gal-1,3-Gal及NeuGc)。給定細胞系之物種來源與組織、器官或細胞譜系皆不能完全決定細胞之糖基化能力。物種來源與組織細胞譜系二者皆可影響細胞合成某些糖基化模式能力。然而,僅僅基於對源自相同組織及/或物種之另一細胞系中獲得之糖模式的瞭解不可能完全預測細胞系之糖基化性質(圖1)。Cell lines derived from different species and derived from different tissues or cell lineages within the species were selected and analyzed for surface structure and enzymatic activity (Figure 1). Based on the results of the analysis, each cell line can be assigned the ability to synthesize certain glycosylation patterns. The results of the analysis show that the cell line differs in its glycosylation properties depending on the species from which it is derived and the tissue or cell lineage. Only a few cell lines naturally show low algal glycosylation ability. Some cell lines potentially produce sialylated structures and several cell lines form immunogenic sugar structures (Gal-1,3-Gal and NeuGc). The source of the species and the tissue, organ or cell lineage of a given cell line cannot fully determine the glycosylation ability of the cell. Both species source and tissue cell lineage can affect the ability of cells to synthesize certain glycosylation patterns. However, it is not possible to fully predict the glycosylation properties of cell lines based solely on the knowledge of the sugar patterns obtained in another cell line derived from the same tissue and/or species (Fig. 1).

大鼠肝瘤細胞系H4-II-E可在其生成具有有利糖性質之抗體之高潛力上與所有經選擇及分析之細胞系區別開來,且因此被選擇用於進一步評估。The rat hepatoma cell line H4-II-E can be distinguished from all selected and analyzed cell lines in its high potential to generate antibodies with favorable saccharide properties and is therefore selected for further evaluation.

細胞系H4-II-E係所有經選擇及分析之細胞系在此分析中唯一未顯示可檢測之海藻糖基化跡象者。此令人驚奇之原因在於其他大鼠細胞系與其他肝細胞系皆不以相同方式累積有益糖性質,例如降低之海藻糖基化、缺乏免疫原性殘基或存在α-2,6連接唾液酸(圖1)。Cell line H4-II-E All selected and analyzed cell lines were the only ones in this analysis that did not show detectable signs of fucosylation. The surprising reason for this is that other rat cell lines do not accumulate beneficial glycochemical properties in the same way as other hepatic cell lines, such as reduced haylosylation, lack of immunogenic residues, or the presence of alpha-2,6 linked saliva. Acid (Figure 1).

尤其大鼠細胞H4-II-E產生具有低海藻糖含量之糖基化係令人驚奇的,此乃因來自大鼠血液血清之抗體係顯著海藻糖基化的,而其他物種(例如兔或貓)中之固有抗體具有低海藻糖含量。In particular, rat cell H4-II-E produces a glycosylation line with a low trehalose content, which is due to the significant fucosylation of the anti-system from rat blood serum, while other species (eg rabbit or The intrinsic antibody in cats has a low trehalose content.

與海藻糖含量不同,某些細胞系中可能存在生成潛在免疫原性糖結構(例如Gal-1,3-Gal及NeuGc)之物種依賴性傾向性。經選擇及分析之人類或大鼠細胞系皆未顯示此等殘基,而與已公佈數據一致,源自小鼠、兔及其他物種之細胞系一致地產生此等結構,且人類中潛在地誘導免疫原性反應(Jenkins等人,1996;Raju等人,2000)。Unlike trehalose content, there may be a species-dependent propensity to generate potentially immunogenic sugar structures (eg, Gal-1,3-Gal and NeuGc) in certain cell lines. None of the human or rat cell lines selected and analyzed showed such residues, and consistent with published data, cell lines derived from mice, rabbits, and other species consistently produced such structures, and potentially in humans. An immunogenic response is induced (Jenkins et al., 1996; Raju et al., 2000).

實例2:與其他已知生產細胞系相比,大鼠肝瘤細胞系H4-II-E中產生之抗體顯示FC聚糖顯著降低之海藻糖基化Example 2: Antibodies produced in the rat hepatoma cell line H4-II-E showed significantly reduced fucosylation of FC glycans compared to other known production cell lines.

為驗證H4-II-E細胞中產生之預測糖基化性質,藉由用編碼IgG1抗體之輕鏈及重鏈之DNA構築體實施轉染且隨後對亦存在於經轉染DNA構築體上之抗性標記物進行抗生素選擇來生成產生穩定IgG1抗體之H4-II-E細胞。獲得IgG1產生細胞群且使其適應於在無血清化學限定培養基中生長。To verify the predicted glycosylation properties produced in H4-II-E cells, transfection is performed by DNA constructs encoding the light and heavy chains of IgG1 antibodies and subsequently also present on transfected DNA constructs. The resistance marker is subjected to antibiotic selection to generate H4-II-E cells that produce a stable IgGl antibody. The IgGl producing cell population was obtained and adapted to grow in serum-free chemically defined medium.

H4-II-E細胞中所產生且藉由蛋白質A層析自細胞培養上清液純化之IgG1抗體係完整的,且在電泳分離後產生重鏈及輕鏈之離散條帶(數據未顯示)。分析H4-II-E細胞中所產生經蛋白質A純化之IgG1抗體上Fc聚糖之結構及組成且將其與CHO-DG44、CHO-Lec13突變體及YB2/0大鼠骨髓瘤細胞中所產生相同抗體上獲得之糖基化模式進行比較。在所有4種來自不同宿主細胞系之IgG1抗體製劑中,二枝結構聚糖佔所有所量測Fc聚糖之最大部分。僅對於YB2/0細胞中產生之抗體而言,其他結構(其主要為不完整雜合聚糖或高甘露糖結構)之比例亦佔約23%之顯著部分(圖2)。就二枝結構聚糖之比例而言,CHO-DG44細胞如先前所報導主要(約95%)產生海藻糖基化結構。與其他細胞不同,H4-II-E細胞中產生之抗體明顯帶有大部分非海藻糖基化二枝結構聚糖。H4-II-E細胞中表現之IgG1中之80%以上含有非海藻糖二枝結構聚糖,此比例顯著高於細胞系YB2/0或CHO突變體Lec13中產生之抗體(圖2)。YB2/0及CHO-Lec13細胞亦產生不含海藻糖之聚糖,然而,不含海藻糖之形式與含有海藻糖之形式間之百分比差不像H4-II-E產生之抗體那樣顯著(圖2)。在初始選擇及分析有利糖結構後,H4-II-E細胞中產生之IgG1抗體儘管未經遺傳改造,但明顯顯示最高比例之非海藻糖基化二枝結構聚糖,藉此證實預測(圖1)。對於低海藻糖基化產生增強之ADCC活化之已知相關性而言,在產生具有高FcγRIII依賴性效應子功能活性之抗體上,H4-II-E細胞優於其他宿主細胞系。The IgG1 anti-system produced in H4-II-E cells and purified from the cell culture supernatant by protein A chromatography was complete and produced discrete bands of heavy and light chains after electrophoretic separation (data not shown) . The structure and composition of Fc glycans on Protein A purified IgG1 antibody produced in H4-II-E cells were analyzed and generated in CHO-DG44, CHO-Lec13 mutants and YB2/0 rat myeloma cells. The glycosylation patterns obtained on the same antibodies were compared. In all four IgGl antibody preparations from different host cell lines, the dimeric structural glycans accounted for the largest portion of all measured Fc glycans. For the antibodies produced in YB2/0 cells alone, the proportion of other structures, which are predominantly incomplete heteropolysaccharides or high mannose structures, also accounted for a significant portion of approximately 23% (Figure 2). In terms of the ratio of the two branched structural glycans, CHO-DG44 cells were mainly reported (about 95%) to produce a fucosylated structure as previously reported. Unlike other cells, antibodies produced in H4-II-E cells clearly carry most of the non-fucosylated dimeric structural glycans. More than 80% of the IgG1 expressed in H4-II-E cells contained non-trehalose dimeric structural glycans, which was significantly higher than that produced in the cell line YB2/0 or the CHO mutant Lec13 (Fig. 2). YB2/0 and CHO-Lec13 cells also produced trehalose-free glycans. However, the percentage difference between the form containing no trehalose and the form containing trehalose was not as significant as the antibody produced by H4-II-E (Fig. 2). After initial selection and analysis of the favorable glycan structure, the IgG1 antibody produced in H4-II-E cells, although not genetically engineered, clearly showed the highest proportion of non-fucosylated dimeric structural glycans, thereby confirming the prediction (Fig. 1). For the known correlation of low algae glycosylation to enhance ADCC activation, H4-II-E cells are superior to other host cell lines in producing antibodies with high FcγRIII-dependent effector functional activity.

實例3:與CHO產生抗體相比,大鼠肝瘤細胞系H4-II-E中產生之抗體顯示FC聚糖顯著提高之半乳糖基化Example 3: Antibodies produced in the rat hepatoma cell line H4-II-E showed significantly increased galactosylation of FC glycans compared to CHO-producing antibodies

對H4-II-E細胞中所產生IgG1之糖基化模式之詳細分析顯示半乳糖基化糖型之含量與CHO-DG44中產生之抗體相比有所升高,CHO-DG44係通常用於生物醫藥蛋白產生之宿主細胞系(圖3)。在H4-II-E中產生之IgG1抗體中,>40%二枝結構聚糖係半乳糖苷基化的,此比率顯著高於CHO細胞中表現之抗體(圖3)。二枝結構聚糖之提高之半乳糖基化可增進抗體活化補體系統之潛力。此與H4-II-E細胞中所產生Fc聚糖降低之海藻糖基化共同使H4-II-E產生之抗體在兩種類型之抗體依賴性效應子功能(ADCC及CDC)上獲得較高效率。二枝結構聚糖之半乳糖基化除增進補體系統組份之Fc-結合及活化外,亦係在末端位置對聚糖進行進一步修飾之基礎。A detailed analysis of the glycosylation pattern of IgG1 produced in H4-II-E cells showed that the content of galactosylated glycoforms was increased compared to the antibody produced in CHO-DG44, which is commonly used in CHO-DG44 lines. Host cell line produced by biomedical proteins (Figure 3). Among the IgG1 antibodies produced in H4-II-E, >40% of the dimeric structural glycans were galactosylated, which was significantly higher than that expressed in CHO cells (Fig. 3). Increased galactosylation of the two structural glycans enhances the potential of the antibody to activate the complement system. This combined with the reduced fucosylation of Fc glycans produced in H4-II-E cells allows H4-II-E-producing antibodies to be higher on both types of antibody-dependent effector functions (ADCC and CDC). effectiveness. In addition to enhancing the Fc-binding and activation of the components of the complement system, galactosylation of the dimeric structural glycans is also the basis for further modification of the glycans at the terminal positions.

實例4:與CHO產生抗體不同,大鼠肝瘤細胞系H4-II-E中產生之抗體顯示FC聚糖之可檢測唾液酸化Example 4: Unlike antibodies produced by CHO, antibodies produced in the rat hepatoma cell line H4-II-E show detectable sialylation of FC glycans

可發現唾液酸或神經胺酸殘基以α-2,3或α-2,6連接至在複合型N-聚糖之枝結構末端處之先前半乳糖殘基。就唾液酸化而言,在分析H4-II-E細胞中所產生IgG1抗體之糖結構後獲得之結果與預測糖結構一致(圖1)。H4-II-E細胞中產生之約8%抗體帶有可藉由用外切糖苷酶神經胺酸酶處理經釋放聚糖來解離之末端唾液酸殘基(圖4)。與H4-II-E細胞中產生之抗體不同,且與預測糖基化模式一致,CHO-DG44細胞中產生之抗體不帶有末端唾液酸殘基(圖4)。末端唾液酸殘基可對修飾抗體之活性及穩定性具有不同效應。最近出版物顯示唾液酸可抑制抗體之發炎活性(Burton及Dwek,2006;Scallon等人,2007)。其他數據顯示,不存在唾液酸可提高小鼠肝之代謝速率,從而表明藉由基於肝之受體之清除(Wright等人,2000)。發現唾液酸衍生物(例如N-羥乙醯神經胺酸(NeuGc))在人類中具有免疫原性(Noguchi等人,1995),然而,不能檢測出H4-II-E細胞中產生之蛋白上之NeuGc修飾之證據(圖1)。It can be found that the sialic acid or neuraminic acid residue is attached to the previous galactose residue at the end of the branched structure of the complex N-glycan with α-2,3 or α-2,6. In terms of sialylation, the results obtained after analyzing the glycostructure of the IgG1 antibody produced in H4-II-E cells were consistent with the predicted glycostructure (Fig. 1). About 8% of the antibodies produced in H4-II-E cells harbor terminal sialic acid residues that can be dissociated by treatment of the released glycans with exoglycosidase neuraminidase (Fig. 4). Unlike antibodies produced in H4-II-E cells, and consistent with the predicted glycosylation pattern, antibodies produced in CHO-DG44 cells do not carry terminal sialic acid residues (Figure 4). The terminal sialic acid residue can have different effects on the activity and stability of the modified antibody. Recent publications have shown that sialic acid inhibits the inflammatory activity of antibodies (Burton and Dwek, 2006; Scallon et al., 2007). Other data show that the absence of sialic acid increases the metabolic rate of the liver in mice, indicating clearance by liver-based receptors (Wright et al., 2000). Sialic acid derivatives (e.g., N-hydroxyethyl cyanokinin (NeuGc)) have been found to be immunogenic in humans (Noguchi et al., 1995), however, proteins produced in H4-II-E cells cannot be detected. Evidence of NeuGc modification (Figure 1).

實例5:適應於在無血清培養基中懸浮生長之H4-II-E大鼠肝瘤細胞Example 5: H4-II-E rat hepatoma cells adapted for suspension growth in serum-free medium

含血清培養基(EMEM(EBSS)+2 mM麩醯胺酸+1%非必需胺基酸+10%胎牛血清(FBS))中培養之H4-II-E細胞以貼壁細胞層形式生長。對於工業生產生物醫藥蛋白而言,在無血清培養基中、較佳在化學限定之無動物組份培養基中且以懸浮培養物形式生長H4-II-E細胞。為使H4-II-E細胞適應於在無血清培養基中懸浮培養,首先在顯示30小時之平均加倍時間之含血清培養基中貼壁擴增細胞。然後藉由用胰蛋白酶/EDTA處理來分開並解離細胞,藉由離心收穫,用無鈣及鎂之杜貝克氏磷酸鹽緩衝鹽水(DPBS)充分洗滌且以200,000個細胞/ml至600,000個細胞/ml之密度懸浮於無血清BI專利培養基(其特別設計用於懸浮培養H4-II-E細胞)或商業AEM(Invitrogen)或PEM(Invitrogen)培養基中。懸浮培養群加倍時間為24小時至32小時之H4-II-E細胞之關鍵態樣係使用無鈣或鈣減少之培養基。在振盪燒瓶(Corning)中在37℃、100-120 rpm及5% CO2下培養細胞懸浮物。每3至4天對細胞進行傳代且將其懸浮於新鮮培養基中。在若干次傳代後,H4-II-E細胞以加倍時間為24小時至32小時之單細胞懸浮物形式恆定生長。H4-II-E細胞在不同培育模式下生長良好且可在進料分批過程中以高活力懸浮培養10天以上。在標準CHO優化培養基中,H4-II-E細胞達到8,000,000個細胞/ml之最大密度,此密度可使用H4-II-E優化培養基來進一步改良(圖5)。總之,經適應之H4-II-E細胞非常適合作為大規模生物醫藥蛋白產生之(產生)宿主細胞(系)。H4-II-E cells cultured in serum-containing medium (EMEM (EBSS) + 2 mM glutamic acid + 1% non-essential amino acid + 10% fetal bovine serum (FBS)) were grown as adherent cell layers. For industrial production of biopharmaceutical proteins, H4-II-E cells are grown in serum-free medium, preferably in chemically defined animal-free medium, and in suspension culture. In order to adapt H4-II-E cells to suspension culture in serum-free medium, cells were first adherently expanded in serum-containing medium showing an average doubling time of 30 hours. The cells were then separated and dissociated by treatment with trypsin/EDTA, harvested by centrifugation, washed thoroughly with calcium and magnesium-free Dubec's phosphate buffered saline (DPBS) and at 200,000 cells/ml to 600,000 cells/ The density of ml is suspended in serum-free BI patented medium (which is specifically designed for suspension culture of H4-II-E cells) or commercial AEM (Invitrogen) or PEM (Invitrogen) medium. The key aspect of H4-II-E cells in which the suspension culture group is doubling from 24 hours to 32 hours is to use a medium without calcium or calcium reduction. The cell suspension was incubated in a shaking flask (Corning) at 37 ° C, 100-120 rpm and 5% CO 2 . Cells were passaged every 3 to 4 days and suspended in fresh medium. After several passages, H4-II-E cells were constantly grown in the form of single cell suspensions with a doubling time of 24 hours to 32 hours. H4-II-E cells grow well in different incubation modes and can be cultured in suspension for more than 10 days with high vigor during the feed batch process. In standard CHO optimized media, H4-II-E cells reached a maximum density of 8,000,000 cells/ml, which was further improved using H4-II-E optimized media (Figure 5). In conclusion, adapted H4-II-E cells are well suited as host cells (lines) for large-scale biopharmaceutical protein production.

實例6:與YB2/0細胞相比,H4-II-E細胞對細胞凋亡具有低敏感性及/或對細胞壓力具有高穩健性Example 6: H4-II-E cells have low sensitivity to apoptosis and/or high robustness to cell stress compared to YB2/0 cells

與YB2/0細胞不同,H4-II-E細胞對藉由高溫、低或高滲透性、pH變化、機械刺激或用化學品或藥物處理誘導之細胞壓力顯示高穩健性。使等細胞數之存於完全培養基(用於H4-II-E細胞之無血清BI培養基(無Ca)、用於YB2/0細胞之RPMI+10% FCS)中之H4-II-E大鼠肝瘤細胞及YB2/0大鼠骨髓瘤細胞暴露於一定劑量之以下壓力源(括號中顯示壓力時間)下:+42℃(2小時,參見圖6A)、暴露於低或高鹽濃度(24小時,參見圖6B)、暴露於2 μg/ml或5 μg/ml嘌呤黴素(48小時,參見圖6C):在溫度刺激後,將細胞在完全培養基中在37℃、5% CO2下培養24小時,隨後藉由錐蟲藍排除染色來分析細胞數及活力。在細胞壓力或細胞凋亡誘導刺激物後,H4-II-E細胞顯示顯著高於YB2/0細胞之穩健性及活力(圖6)。H4-II-E細胞對細胞壓力源之低敏感性及高穩健性使H4-II-E成為用於生物醫藥大規模產生過程之極佳系統,其中在長培養時期內需要高細胞活力及存活。Unlike YB2/0 cells, H4-II-E cells show high robustness to cell pressure induced by high temperature, low or high permeability, pH change, mechanical stimulation or treatment with chemicals or drugs. H4-II-E rats in the complete medium (serum-free BI medium (without Ca) for H4-II-E cells, RPMI + 10% FCS for YB2/0 cells) Hepatoma cells and YB2/0 rat myeloma cells were exposed to a pressure source below a certain dose (pressure time shown in brackets): +42 ° C (2 hours, see Figure 6A), exposure to low or high salt concentrations (24 Hours, see Figure 6B), exposure to 2 μg/ml or 5 μg/ml puromycin (48 hours, see Figure 6C): After temperature stimulation, cells were in complete medium at 37 ° C, 5% CO 2 The cells were cultured for 24 hours, and then the number of cells and viability were analyzed by trypan blue exclusion staining. After cell stress or apoptosis-inducing stimuli, H4-II-E cells showed significantly higher robustness and viability than YB2/0 cells (Fig. 6). The low sensitivity and high robustness of H4-II-E cells to cell stressors make H4-II-E an excellent system for large-scale biopharmaceutical production processes, where high cell viability and survival are required during long culture periods. .

實例7:H4-II-E細胞之單細胞懸浮培育需要CA2+減少或無CA2+之無血清培養基Example 7: Single cell suspension incubation of H4-II-E cells requires serum-free medium with reduced CA 2+ or no CA 2+

懸浮培養群加倍時間為24小時至32小時之H4-II-E細胞之關鍵態樣係使用無鈣或鈣減少之培養基。兩種鈣減少或無鈣之培養基係藉由利用Hitachi 917(Roche)分析Ca-濃度來鑑別(圖7A)。鈣減少培養基AEM與無Ca BI培養基二者允許單細胞懸浮培育H4-II-E細胞(圖7B、C)。若用1 mM CaCl2補充AEM(圖7C)或若將細胞接種於含Ca BI培養基中(圖7E),則細胞形成大聚集體且單細胞懸浮生長受阻。含鈣培養基中游離鈣離子之減少亦可藉由將EDTA添加至該培養基中來達成,以減少H4-II-E細胞在此等補充EDTA之培養基中之聚集(圖7F)。The key aspect of H4-II-E cells in which the suspension culture group is doubling from 24 hours to 32 hours is to use a medium without calcium or calcium reduction. Two calcium-reduced or calcium-free media were identified by analyzing the Ca-concentration using Hitachi 917 (Roche) (Fig. 7A). Both calcium-reducing medium AEM and Ca-free BI medium allowed single cell suspensions to culture H4-II-E cells (Fig. 7B, C). If AEM was supplemented with 1 mM CaCl 2 (Fig. 7C) or if cells were seeded in Ca-containing BI medium (Fig. 7E), the cells formed large aggregates and the growth of single cell suspension was blocked. The reduction of free calcium ions in the calcium-containing medium can also be achieved by adding EDTA to the medium to reduce aggregation of H4-II-E cells in the medium supplemented with EDTA (Fig. 7F).

實例8:H4-II-E細胞聚集具有CA2+濃度依賴性及MG2+獨立性Example 8: H4-II-E cell aggregation has CA 2+ concentration dependence and MG 2+ independence

為解決以下問題,即鈣離子對H4-II-E細胞聚集之效應是否亦可藉由通常存在於細胞培養基中之其他二價陽離子來達成,使H4-II-E細胞之懸浮培養物經受增加濃度之CaCl2或MgCl2(圖8)。若將50 μMol/L CaCl2添加至無CaBI培養基中,則可觀察到H4-II-E細胞聚集之增加。細胞聚集體之聚集比率及緊密度隨著添加至無Ca培養基中之CaCl2之濃度增加而進一步增加(圖8A)。相比之下,將等濃度MgCl2添加至培養基中對單細胞懸浮或細胞聚集無顯著影響(圖8B)。在AEM培養基中,在培養3天後添加250 μM/L CaCl2或更高CaCl2濃度會顯著損害H4-II-E細胞生長。在補充有250 μM/L CaCl2或更高CaCl2濃度之AEM培養基中,80%以上細胞形成緊密聚集體,此係細胞培養發酵過程中所不期望的。與CaCl2不同,增加MgCl2之濃度對H4-II-E細胞之聚集比率無影響。In order to solve the problem that whether the effect of calcium ion on H4-II-E cell aggregation can also be achieved by other divalent cations usually present in the cell culture medium, the suspension culture of H4-II-E cells is subjected to an increase. Concentration of CaCl 2 or MgCl 2 (Figure 8). If 50 μMol/L CaCl 2 was added to the CaBI-free medium, an increase in H4-II-E cell aggregation was observed. The aggregation ratio and tightness of the cell aggregates were further increased as the concentration of CaCl 2 added to the Ca-free medium was increased ( FIG. 8A ). In contrast, the addition of an equal concentration of MgCl 2 to the medium had no significant effect on single cell suspension or cell aggregation (Fig. 8B). In AEM medium, addition of 250 μM/L CaCl 2 or higher CaCl 2 concentration after 3 days of culture significantly impaired H4-II-E cell growth. In AEM medium supplemented with 250 μM/L CaCl 2 or higher CaCl 2 concentration, more than 80% of the cells form tight aggregates, which are undesirable during cell culture fermentation. Unlike CaCl 2 , increasing the concentration of MgCl 2 had no effect on the aggregation ratio of H4-II-E cells.

實例9:H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGG1抗體以高於CHO中產生之抗CD20 IGG1之親和力結合至FC受體CD16-V158及CD16-F158(FCGRIIIA)Example 9: Anti-CD20 IGG1 antibody produced in H4-II-E rat hepatoma cells binds to FC receptors CD16-V158 and CD16-F158 (FCGRIIIA) with higher affinity than anti-CD20 IGG1 produced in CHO

使用抗CD20 IgG1抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG1抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG1抗體之重鏈(SEQ ID NO:2)及輕鏈(SEQ ID NO:3)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG1 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG1 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 2) and the light chain (SEQ ID NO: 3) of the anti-CD20 IgG1 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

使用BIAcore分析來量測H4-II-E細胞及CHO細胞中產生之抗CD20 IgG1與FcγRIIIa受體CD16-V158及CD16-F158之結合動力學。與CHO細胞中產生之抗CD20 IgG1相比,H4-II-E細胞中產生之抗CD20 IgG1顯示與FcγRIIIa之兩種變體顯著更高之親和力(圖9)。BIAcore analysis was used to measure the binding kinetics of anti-CD20 IgG1 and FcγRIIIa receptors CD16-V158 and CD16-F158 produced in H4-II-E cells and CHO cells. The anti-CD20 IgG1 produced in H4-II-E cells showed significantly higher affinity with the two variants of FcγRIIIa compared to the anti-CD20 IgG1 produced in CHO cells (Fig. 9).

實例10:H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGG4抗體以高於CHO中產生之抗CD20 IGG4之親和力結合至FC受體CD16-V158及CD16-F158(FCGRIIIA)Example 10: Anti-CD20 IGG4 antibody produced in H4-II-E rat hepatoma cells binds to FC receptors CD16-V158 and CD16-F158 (FCGRIIIA) with higher affinity than anti-CD20 IGG4 produced in CHO

使用抗CD20 IgG4抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG4抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG4抗體之重鏈(SEQ ID NO:4)及輕鏈(SEQ ID NO:5)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG4 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG4 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 4) and the light chain (SEQ ID NO: 5) of the anti-CD20 IgG4 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

使用BIAcore分析來量測H4-II-E細胞及CHO細胞中產生之抗CD20 IgG4與FcγRIIIa受體CD16-V158及CD16-F158之結合動力學。與CHO細胞中產生之抗CD20 IgG4相比,H4-II-E細胞中產生之抗CD20 IgG4顯示與FcγRIIIa之兩種變體顯著更高之親和力。Binding kinetics of anti-CD20 IgG4 and FcγRIIIa receptors CD16-V158 and CD16-F158 produced in H4-II-E cells and CHO cells were measured using BIAcore analysis. Anti-CD20 IgG4 produced in H4-II-E cells showed significantly higher affinity than the two variants of FcyRIIIa compared to anti-CD20 IgG4 produced in CHO cells.

實例11:H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGG1抗體比CHO中產生之抗CD20 IGG1抗體更有效地活化活體外ADCCExample 11: Anti-CD20 IGG1 antibody produced in H4-II-E rat hepatoma cells activates in vitro ADCC more efficiently than anti-CD20 IGG1 antibody produced in CHO

使用抗CD20 IgG1抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG1抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG1抗體之重鏈(SEQ ID NO:2)及輕鏈(SEQ ID NO:3)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG1 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG1 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 2) and the light chain (SEQ ID NO: 3) of the anti-CD20 IgG1 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

藉由乳酸脫氫酶(LDH)釋放分析來實施ADCC分析,其中使用人類外周血液單核細胞(PBMC)作為效應子細胞。將靶腫瘤細胞(表現人類CD20之人類伯基特氏淋巴瘤細胞系Ramos)之等分試樣分配至96孔U形底板(存於50 μl/孔中之10,000個細胞)中且在PBMC(100 μL)存在下以20/1之E/T比率與分別在H4-II-E細胞或CHO細胞中產生之經純化抗CD20 IgG1之連續稀釋液(50 μL)一起培育。在37℃下培育4 h後,量測上清液LDH活性。計算特異性細胞溶解%且若所用抗CD20 IgG1係自H4-II-E細胞而非自CHO細胞之細胞培養上清液產生並純化,則該值顯著較高,從而表明H4-II-E產生之IgG1在效應子細胞中誘導ADCC之潛力明顯更高(圖10)。ADCC analysis was performed by lactate dehydrogenase (LDH) release assay using human peripheral blood mononuclear cells (PBMC) as effector cells. Aliquots of target tumor cells (human Burkitt's lymphoma cell line Ramos expressing human CD20) were dispensed into 96-well U-shaped bottom plates (10,000 cells in 50 μl/well) and in PBMC ( 100 μL) was incubated with a serial dilution of purified anti-CD20 IgG1 (50 μL) produced in H4-II-E cells or CHO cells at an E/T ratio of 20/1. After incubation at 37 ° C for 4 h, the supernatant LDH activity was measured. The % specific cytolysis was calculated and if the anti-CD20 IgG1 line used was produced and purified from H4-II-E cells but not from cell culture supernatants of CHO cells, the value was significantly higher, indicating H4-II-E production. The potential of IgG1 to induce ADCC in effector cells was significantly higher (Fig. 10).

實例12:H4-II-E大鼠肝瘤細胞中產生之抗HER2 IGG4抗體比CHO中產生之抗HER2 IGG4抗體更有效地活化活體外ADCCExample 12: Anti-HER2 IGG4 antibody produced in H4-II-E rat hepatoma cells activates in vitro ADCC more efficiently than anti-HER2 IGG4 antibody produced in CHO

使用抗HER2 IgG4抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗HER2表現來分離穩定抗HER2生產細胞系。在無血清進料分批培養物中培育抗HER2產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-HER2 IgG4 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The stable anti-HER2 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-HER2 expression of the cell supernatant of the viable cells by ELISA. Anti-HER2 producing cells were grown in serum-free fed batch cultures and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

藉由乳酸脫氫酶(LDH)釋放分析來實施ADCC分析,其中使用人類外周血液單核細胞(PBMC)作為效應子細胞。將HER2-陽性乳癌細胞系之等分試樣分配至96孔U形底板(存於50 μl/孔中之10,000個細胞)且在PBMC(100 μL)存在下以20/1之E/T比率與分別在H4-II-E細胞或CHO細胞中產生之經純化抗HER2 IgG4之連續稀釋液(50 μL)一起培育。在37℃下培育4 h後,量測上清液LDH活性。計算特異性細胞溶解%且若所用抗HER2 IgG4係自H4-II-E細胞而非自CHO細胞之細胞培養上清液產生並純化,則該值顯著較高,從而表明H4-II-E產生之IgG4在效應子細胞中誘導ADCC之潛力明顯更高。ADCC analysis was performed by lactate dehydrogenase (LDH) release assay using human peripheral blood mononuclear cells (PBMC) as effector cells. An aliquot of the HER2-positive breast cancer cell line was dispensed to a 96-well U-shaped bottom plate (10,000 cells in 50 μl/well) and an E/T ratio of 20/1 in the presence of PBMC (100 μL) Incubation was carried out with serial dilutions (50 μL) of purified anti-HER2 IgG4 produced in H4-II-E cells or CHO cells, respectively. After incubation at 37 ° C for 4 h, the supernatant LDH activity was measured. The specific cell lysis is calculated and if the anti-HER2 IgG4 line is produced and purified from H4-II-E cells but not from cell culture supernatants of CHO cells, the value is significantly higher, indicating H4-II-E production. The potential of IgG4 to induce ADCC in effector cells is significantly higher.

實例13:H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGG1抗體以高於CHO中產生之抗CD20 IGG1抗體之親和力結合補體系統之組份Example 13: Anti-CD20 IGG1 antibody produced in H4-II-E rat hepatoma cells binds to components of the complement system with higher affinity than the anti-CD20 IGG1 antibody produced in CHO

使用抗CD20 IgG1抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG1抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG1抗體之重鏈(SEQ ID NO:2)及輕鏈(SEQ ID NO:3)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG1 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG1 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 2) and the light chain (SEQ ID NO: 3) of the anti-CD20 IgG1 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

藉由流式細胞計數分析使用純化人類補體Clq來研究每一純化IgG1結合補體之Clq組份之能力。將表現人類CD20之人類伯基特氏淋巴瘤Ramos細胞與分別在H4-II-E細胞或CHO中產生之抗人類CD20 IgG1之連續稀釋液一起培育。在用含有1%(w/v) BSA之PBS洗滌後,以20 mg/mL之最終濃度添加經純化人類補體Clq並在37℃下與結合細胞之IgG1結合30 min。然後洗滌細胞並將其與針對人類Clq之螢光素異硫氰酸偶聯多株抗體一起培育。藉由流式細胞術使用FACSCalibur來分析染色細胞。與CHO細胞中產生之抗CD20 IgG1相比,H4-II-E細胞中產生之抗CD20 IgG1顯示與補體組份Clq強得多之結合。The ability of each purified IgGl binding complement Clq component was investigated by flow cytometric analysis using purified human complement Clq. Human Burkitt's lymphoma Ramos cells expressing human CD20 were incubated with serial dilutions of anti-human CD20 IgG1 produced in H4-II-E cells or CHO, respectively. After washing with PBS containing 1% (w/v) BSA, purified human complement Clq was added at a final concentration of 20 mg/mL and bound to IgG1 of the bound cells for 30 min at 37 °C. The cells were then washed and incubated with luciferin isothiocyanate-conjugated antibodies against human Clq. The stained cells were analyzed by flow cytometry using a FACSCalibur. The anti-CD20 IgG1 produced in H4-II-E cells showed a much stronger binding to the complement component Clq compared to the anti-CD20 IgG1 produced in CHO cells.

實例14:與CHO細胞中產生之抗CD20 IGG1抗體相比,H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGG1抗體顯示增強之活體外補體活化Example 14: Anti-CD20 IGG1 antibody produced in H4-II-E rat hepatoma cells showed enhanced in vitro complement activation compared to anti-CD20 IGG1 antibody produced in CHO cells

使用抗CD20 IgG1抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG1抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG1抗體之重鏈(SEQ ID NO:2)及輕鏈(SEQ ID NO:3)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG1 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG1 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 2) and the light chain (SEQ ID NO: 3) of the anti-CD20 IgG1 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

藉由WST-1分析來測定CDC活性。簡言之,在37℃下在96孔平底板(Greiner)中將表現人類CD20之靶人類伯基特氏淋巴瘤細胞系Ramos、2倍稀釋人類血清補體(Sigma-Aldrich)及分別在H4-II-E細胞或CHO細胞中產生之抗人類CD20之連續稀釋液培育1 h。將細胞增殖WST-1試劑添加至各孔(15 μL/孔)中且在37℃下培育6 h。使用微量板讀數器(Tecan,Germany)在450 nm下量測各孔中之吸光度且以作為活細胞數指數之相對吸光度單位(RAU)表現。根據下式計算CDC%:CDC活性[%]=100×(RAU背景-RAU測試)/RAU背景。若抗CD20 IgG1係在H4-II-E中而非在CHO細胞中產生,則分析中量測之CDC活性顯著較高。CDC activity was determined by WST-1 analysis. Briefly, the human CD20 target human Burkitt's lymphoma cell line Ramos, 2-fold diluted human serum complement (Sigma-Aldrich), and H4-, respectively, in a 96-well flat bottom plate (Greiner) at 37 °C Serial dilutions of anti-human CD20 produced in II-E cells or CHO cells were incubated for 1 h. Cell proliferation WST-1 reagent was added to each well (15 μL/well) and incubated for 6 h at 37 °C. The absorbance in each well was measured at 450 nm using a microplate reader (Tecan, Germany) and expressed as a relative absorbance unit (RAU) as a viable cell number index. CDC% was calculated according to the following formula: CDC activity [%] = 100 x (RAU background - RAU test ) / RAU background . If the anti-CD20 IgG1 line is produced in H4-II-E but not in CHO cells, the CDC activity measured in the assay is significantly higher.

實例15:H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGGI抗體以高於CHO中產生之抗CD20 IGG1抗體之親和力結合新生兒FC受體FCRN.Example 15: Anti-CD20 IGGI antibody produced in H4-II-E rat hepatoma cells binds to neonatal FC receptor FCRN with higher affinity than anti-CD20 IGG1 antibody produced in CHO.

使用抗CD20 IgG1抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG1抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG1抗體之重鏈(SEQ ID NO:2)及輕鏈(SEQ ID NO:3)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG1 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG1 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 2) and the light chain (SEQ ID NO: 3) of the anti-CD20 IgG1 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

使用BIAcore分析來量測H4-II-E細胞或CHO細胞中產生之抗CD20 IgG1與新生兒Fc受體FcRn之結合。在CHO/DG44細胞中表現重組可溶性人類FcRn-b2微球蛋白複合物且藉由Ni-NTA層析(Qiagen)自培養上清液純化。使用胺偶合套組(BIACORE)將抗人類b2-微球蛋白單株抗體(Abcam,Cambridge,UK)固定至BIAcore T100 CM5感測器晶片上。藉由注射可溶性FcRn-b2微球蛋白複合物,藉由經固定抗b2-微球蛋白抗體來捕獲該複合物。以5種不同濃度(自4.17 nM至66.7 nM)將每一經純化抗CD20 IgG1稀釋於pH調節至6.0之HBS-EP+緩衝液(0.01 M HEPES,0.15 M NaCl,3 mM EDTA,0.05%表面活性劑P20)中,且以5 mL/min之流速將每一經稀釋IgG1注射於複合物捕獲感測器表面或空白上。藉由以60 mL/min之流速將7.5 mM HCl注射1 min來去除結合至感測器表面上之可溶性FcRn及IgG1。在25℃下利用HBS-EP+作為運行緩衝液來實施該等實驗。使用藉由減去空白獲得之數據進行數據分析。使用BIAcore T100評估軟體藉由二元擬合模型來計算表觀締合速率常數(ka)、解離速率常數(kd)及結合親和力(KD)。與CHO細胞中產生之抗CD20 IgG1相比,H4-II-E細胞中產生之抗CD20 IgG1顯示與新生兒Fc受體FcRn強得多之結合(圖11).Binding of anti-CD20 IgGl produced in H4-II-E cells or CHO cells to the neonatal Fc receptor FcRn was measured using BIAcore analysis. The recombinant soluble human FcRn-b2 microglobulin complex was expressed in CHO/DG44 cells and purified from the culture supernatant by Ni-NTA chromatography (Qiagen). Anti-human b2-microglobulin monoclonal antibody (Abeam, Cambridge, UK) was immobilized onto a BIAcore T100 CM5 sensor wafer using an amine coupling kit (BIACORE). The complex is captured by immobilization of an anti-b2-microglobulin antibody by injection of a soluble FcRn-b2 microglobulin complex. Each purified anti-CD20 IgG1 was diluted at 5 different concentrations (from 4.17 nM to 66.7 nM) to HBS-EP+ buffer (0.01 M HEPES, 0.15 M NaCl, 3 mM EDTA, 0.05% surfactant) adjusted to pH 6.0. In each of P20), each diluted IgG1 was injected onto the composite capture sensor surface or blank at a flow rate of 5 mL/min. Soluble FcRn and IgGl bound to the surface of the sensor were removed by injecting 7.5 mM HCl for 1 min at a flow rate of 60 mL/min. These experiments were carried out using HBS-EP+ as a running buffer at 25 °C. Data analysis was performed using data obtained by subtracting blanks. Apparent association rate constants (ka), dissociation rate constants (kd), and binding affinities (KD) were calculated using a BIAcore T100 evaluation software by a binary fit model. The anti-CD20 IgG1 produced in H4-II-E cells showed a much stronger binding to the neonatal Fc receptor FcRn compared to the anti-CD20 IgG1 produced in CHO cells (Fig. 11).

實例16:H4-II-E大鼠肝瘤細胞中產生之MCP1-FC-融合蛋白以高於CHO中產生之MCP1-FC-融合蛋白之親和力結合至FC受體CD16-V158及CD16-F158(FCGRIIIA)Example 16: MCP1-FC-fusion protein produced in H4-II-E rat hepatoma cells binds to FC receptors CD16-V158 and CD16-F158 with higher affinity than MCP1-FC-fusion protein produced in CHO ( FCGRIIIA)

使用MCP1-Fc融合蛋白表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生MCP1-Fc融合蛋白之H4-II-E細胞係藉由用編碼SEQ ID NO:6之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之MCP1-Fc表現來分離穩定MCP1-Fc融合蛋白生產細胞系。在無血清進料分批培養物中培育MCP1-Fc產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The MCP1-Fc fusion protein expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the MCP1-Fc fusion protein was generated by transfection with the DNA construct encoding SEQ ID NO: 6. The stable MCP1-Fc fusion protein producing cell line was isolated by selecting an antibiotic resistance marker and analyzing the MCP1-Fc expression of the cell supernatant of the viable cells by ELISA. MCP1-Fc-producing cells were grown in serum-free fed batch cultures and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

使用BIAcore分析來量測H4-II-E細胞及CHO細胞中產生之MCP1-Fc與FcγRIIIa受體CD16-V158及CD16-F158之結合動力學。與CHO細胞中產生之抗CD20 IgG1相比,H4-II-E細胞中產生之MCP1-Fc顯示與FcγRIIIa之兩種變體顯著更高之親和力。BIAcore analysis was used to measure the binding kinetics of MCP1-Fc and FcγRIIIa receptors CD16-V158 and CD16-F158 produced in H4-II-E cells and CHO cells. MCP1-Fc produced in H4-II-E cells showed significantly higher affinity than the two variants of FcyRIIIa compared to anti-CD20 IgG1 produced in CHO cells.

實例17:H4-II-E大鼠肝瘤細胞中產生之包含EPO-FC之FC-融合蛋白以高於CHO中產生之包含EPO-FC之FC-融合蛋白之親和力結合至FC受體CD16-V158及CD16-F158(FCGRIIIA)Example 17: EPO-FC-containing FC-fusion protein produced in H4-II-E rat hepatoma cells binds to FC receptor CD16 with higher affinity than EPO-FC-containing FC-fusion protein produced in CHO- V158 and CD16-F158 (FCGRIIIA)

使用包含框架內融合至IgG1Fc之核酸序列之EPO之核酸序列的表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生EPO-Fc融合蛋白之H4-II-E細胞係藉由用編碼SEQ ID NO:7之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之Fc-融合蛋白表現來分離產生包含EPO-Fc之穩定Fc融合蛋白之細胞系。在無血清進料分批培養物中培育Fc-融合蛋白產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化EPO-Fc蛋白。The expression vector containing the nucleic acid sequence of EPO fused to the nucleic acid sequence of IgG1 Fc in the framework was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the EPO-Fc fusion protein was generated by transfection with the DNA construct encoding SEQ ID NO: 7. A cell line producing a stable Fc fusion protein comprising EPO-Fc was isolated by selecting an antibiotic resistance marker and analyzing the Fc-fusion protein expression of the cell supernatant of the viable cells by ELISA. Fc-fusion protein producing cells were grown in serum-free fed batch culture and EPO-Fc protein was purified from serum-free culture supernatant by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

使用BIAcore分析來量測H4-II-E細胞及CHO細胞中產生之包含EPO-Fc之Fc融合蛋白與FcγRIIIa受體CD16-V158及CD16-F158之結合動力學。與CHO細胞中產生之抗CD20 IgG1相比,H4-II-E細胞中產生之包含EPO-Fc之Fc融合蛋白顯示與FcγRIIIa之兩種變體顯著更高之親和力。BIAcore analysis was used to measure the binding kinetics of EPO-Fc-containing Fc fusion proteins produced in H4-II-E cells and CHO cells to the FcγRIIIa receptors CD16-V158 and CD16-F158. The EPO-Fc-containing Fc fusion protein produced in H4-II-E cells showed significantly higher affinity to the two variants of FcyRIIIa compared to the anti-CD20 IgG1 produced in CHO cells.

實例18:H4-II-E大鼠肝瘤細胞中產生之抗CD20 IGG1抗體以低於CHO中產生之抗CD20 IGG1抗體之親和力結合FC受體CD32A及CD32BExample 18: Anti-CD20 IGG1 antibody produced in H4-II-E rat hepatoma cells binds FC receptor CD32A and CD32B with lower affinity than anti-CD20 IGG1 antibody produced in CHO

使用抗CD20 IgG1抗體表現載體來分別穩定轉染CHO細胞及H4-II-E細胞。產生抗CD20 IgG1抗體之H4-II-E細胞係藉由用編碼抗CD20 IgG1抗體之重鏈(SEQ ID NO:2)及輕鏈(SEQ ID NO:3)之DNA構築體實施轉染來生成。藉由選擇抗生素抗性標記物並藉由ELISA分析存活細胞之細胞上清液之抗CD20表現來分離穩定抗CD20生產細胞系。在無血清進料分批培養物中培育抗CD20產生細胞且藉由蛋白質A-親和力層析使用MabSelectTM(Amersham Biosciences)自無血清培養上清液純化重組抗體。The anti-CD20 IgG1 antibody expression vector was used to stably transfect CHO cells and H4-II-E cells, respectively. The H4-II-E cell line producing the anti-CD20 IgG1 antibody was generated by transfection with a DNA construct encoding the heavy chain (SEQ ID NO: 2) and the light chain (SEQ ID NO: 3) of the anti-CD20 IgG1 antibody. . The stable anti-CD20 production cell line was isolated by selecting an antibiotic resistance marker and analyzing the anti-CD20 expression of the cell supernatant of the viable cells by ELISA. Anti-CD20 producing cells were grown in serum-free fed batch culture and recombinant antibodies were purified from serum-free culture supernatants by protein A-affinity chromatography using MabSelectTM (Amersham Biosciences).

使用BIAcore分析來量測H4-II-E細胞及CHO細胞中產生之抗CD20 IgG1與FcγRIIa及FcγRIIb之結合動力學。與CHO細胞中產生之抗CD20 IgG1相比,H4-II-E細胞中產生之抗CD20 IgG1顯示與FcγRIIa及FcγRIIb之兩種變體顯著更高之親和力。BIAcore analysis was used to measure the binding kinetics of anti-CD20 IgG1 to FcγRIIa and FcγRIIb produced in H4-II-E cells and CHO cells. Anti-CD20 IgG1 produced in H4-II-E cells showed significantly higher affinity to both variants of FcγRIIa and FcγRIIb compared to anti-CD20 IgG1 produced in CHO cells.

參考文獻列表Reference list

1. Burton,D.R.及Dwek,R.A.(2006). Immunology. Sugar determines antibody activity. Science 313,627-628。1. Burton, DR and Dwek, RA (2006). Immunology. Sugar determined antibody activity. Science 313 , 627-628.

2. Edelman,G.M.,Cunningham,B.A.,Gall,W.E.,Gottlieb,P.D.,Rutishauser,U.及Waxdal,M.J.(1969). The covalent structure of an entire gammaG immunoglobulin molecule.Proc. Natl. Acad. Sci. U. S. A 63,78-85。2. Edelman, GM, Cunningham, BA, Gall, WE, Gottlieb, PD, Rutishauser, U. and Waxdal, MJ (1969). The covalent structure of an entire gammaG immunoglobulin molecule. Proc. Natl. Acad. Sci. US A 63 , 78-85.

3. Ellison,J.及Hood,L.(1982). Linkage and sequence homology of two human immunoglobulin gamma heavy chain constant region genes. Proc. Natl. Acad. Sci. U. S. A 79,1984-1988。3. Ellison, J. and Hood, L. (1982). Linkage and sequence homology of two human immunoglobulin gamma heavy chain constant region genes. Proc. Natl. Acad. Sci. US A 79 , 1984-1988.

4. Fallaux,F.J.,Bout,A.,van,d.V.,I,van den Wollenberg,D.J.,Hehir,K.M.,Keegan,J.,Auger,C.,Cramer,S.J.,van,O.H.,van der Eb,A.J.,Valerio,D.及Hoeben,R.C.(1998).New helper cells and matched early region 1-deleted adenovirus vectors prevent generation of replication-competent adenoviruses. Hum. Gene Ther. 9,1909-1917。4. Fallaux, FJ, Bout, A., van, dV, I, van den Wollenberg, DJ, Hehir, KM, Keegan, J., Auger, C., Cramer, SJ, van, OH, van der Eb, AJ , Valerio, D. and Hoeben, RC (1998). New helper cells and matched early region 1-deleted adenovirus vectors prevent generation of replication-competent adenoviruses. Hum. Gene Ther. 9 , 1909-1917.

5. Hamako,J.,Matsui,T.,Ozeki,Y.,Mizuochi,T.及Titani,K.(1993). Comparative studies of asparagine-linked sugar chains of immunoglobulin G from eleven mammalian species. Comp Biochem. Physiol B 106,949-954。5. Hamako, J., Matsui, T., Ozeki, Y., Mizuochi, T. and Titani, K. (1993). Comparative studies of asparagine-linked sugar chains of immunoglobulin G from eleven mammalian species. Comp Biochem. B 106 , 949-954.

6. Horikoshi,N.,Tashiro,F.,Tanaka,N.及Ueno,Y.(1988). Modulation of hormonal induction of tyrosine aminotransferase and glucocorticoid receptors by aflatoxinB1 and sterigmatocystin in Reuber hepatoma cells. Cancer Res. 48,5188-5192。6. Horikoshi, N., Tashiro, F., Tanaka, N. and Ueno, Y. (1988). Modulation of hormonal induction of tyrosine aminotransferase and glucocorticoid receptors by aflatoxinB1 and sterigmatocystin in Reuber hepatoma cells. Cancer Res. 48 ,5188 -5192.

7. Houser,W.H.,Raha,A.及Vickers,M.(1992). Induction of CYP1A1 gene expression in H4-II-E rat hepatoma cells by benzo[e]pyrene. Mol. Carcinog. 5,232-237。7. Houser, WH, Raha, A. and Vickers, M. (1992). Induction of CYP1A1 gene expression in H4-II-E rat hepatoma cells by benzo[e]pyrene. Mol. Carcinog. 5 , 232-237.

8. Jenkins,N.,Parekh,R.B.及James,D.C.(1996). Getting the glycosylation right: implications for the biotechnology industry. Nat. Biotechnol. 14,975-981。8. Jenkins, N., Parekh, RB and James, DC (1996). Getting the glycosylation right: implications for the biotechnology industry. Nat. Biotechnol. 14 , 975-981.

9. Kabat,E.A.(1988). Antibody complementarity and antibody structure. J. Immunol. 141,S25-S36。9. Kabat, EA (1988). Antibody complementarity and antibody structure. J. Immunol. 141 , S25-S36.

10.Kabat,E.A.,Wu,T.T.,Perry,H.M.,Gottesman,K.S.及Foeller,C.(1991). Sequences of Proteins of Immunological Interest. U. S. Department of Health and Human Services,Natl. Inst. of Health,Bethesda.10. Kabat, E.A., Wu, T.T., Perry, H.M., Gottesman, K.S. and Foeller, C. (1991). Sequences of Proteins of Immunological Interest. U. S. Department of Health and Human Services, Natl. Inst. of Health, Bethesda.

11. Kanda,Y.,Yamane-Ohnuki,N.,Sakai,N.,Yamano,K.,Nakano,R.,Inoue,M.,Misaka,H.,Iida,S.,Wakitani,M.,Konno,Y.,Yano,K.,Shitara,K.,Hosoi,S.及Satoh,M.(2006). Comparison of cell lines for stable production of fucose-negative antibodies with enhanced ADCC. Biotechnol. Bioeng. 94,680-688。11. Kanda, Y., Yamane-Ohnuki, N., Sakai, N., Yamano, K., Nakano, R., Inoue, M., Misaka, H., Iida, S., Wakitani, M., Konno , Y., Yano, K., Shitara, K., Hosoi, S. and Satoh, M. (2006). Comparison of cell lines for stable production of fucose-negative antibodies with enhanced ADCC. Biotechnol. Bioeng. 94 ,680 -688.

12. Kaufman,R.J.(1990). Selection and coamplification of heterologous genes in mammalian cells. Methods Enzymol. 185,537-566。12. Kaufman, RJ (1990). Selection and coamplification of heterologous genes in mammalian cells. Methods Enzymol. 185 , 537-566.

13. Lifely,M.R.,Hale,C.,Boyce,S.,Keen,M.J.及Phillips,J. (1995). Glycosylation and biological activity of CAMPATH-1H expressed in different cell lines and grown under different culture conditions. Glycobiology 5,813-822。13. Lifely, MR, Hale, C., Boyce, S., Keen, MJ and Phillips, J. (1995). Glycosylation and biological activity of CAMPATH-1H expressed in different cell lines and grown under different culture conditions. Glycobiology 5 , 813-822.

14. Miyazaki,M.,Bai,L.,Taga,H.,Hirai,H.,Sato,J.及Namba,M. (1991). Expression of liver-specific functions and secretion of a hepatocyte growth factor by a newly established rat hepatoma cell line growing in a chemically-defined serum-free medium. Res. Exp. Med. (Berl) 191,297-307。14. Miyazaki, M., Bai, L., Taga, H., Hirai, H., Sato, J. and Namba, M. (1991). Expression of liver-specific functions and secretion of a hepatocyte growth factor by a Newly established rat hepatoma cell line growing in a chemically-defined serum-free medium. Res. Exp. Med. (Berl) 191 , 297-307.

15. Mori,K.,Kuni-Kamochi,R.,Yamane-Ohnuki,N.,Wakitani,M.,Yamano,K.,Imai,H.,Kanda,Y.,Niwa,R.,Iida,S.,Uchida,K.,Shitara,K.及Satoh,M. (2004). Engineering Chinese hamster ovary cells to maximize effector function of produced antibodies using FUT8 siRNA. Biotechnol. Bioeng. 88,901-908。15. Mori, K., Kuni-Kamochi, R., Yamane-Ohnuki, N., Wakitani, M., Yamano, K., Imai, H., Kanda, Y., Niwa, R., Iida, S. , Uchida, K., Shitara, K. and Satoh, M. (2004). Engineering Chinese hamster ovary cells to maximize effector function of produced antibodies using FUT8 siRNA. Biotechnol. Bioeng. 88 , 901-908.

16. Niwa,A.,Yamamoto,K.,Sorimachi,K.及Yasumura,Y. (1980). Continuous culture of Reuber hepatoma cells in serum-free arginine-,glutamine- and tyrosine-deprived chemically defined medium. In Vitro 16,987-993。16. Niwa, A., Yamamoto, K., Sorimachi, K. and Yasumura, Y. (1980). Continuous culture of Reuber hepatoma cells in serum-free arginine-, glutamine- and tyrosine-deprived chemically defined medium. In Vitro 16 , 987-993.

17. Noguchi,A.,Mukuria,C.J.,Suzuki,E.及Naiki,M. (1995). Immunogenicity of N-glycolylneuraminic acid-containing carbohydrate chains of recombinant human erythropoietin expressed in Chinese hamster ovary cells. J. Biochem. 117,59-62。17. Noguchi, A., Mukuria, CJ , Suzuki, E. And Naiki, M. (1995). Immunogenicity of N-glycolylneuraminic acid-containing carbohydrate chains of recombinant human erythropoietin expressed in Chinese hamster ovary cells. J. Biochem. 117 , 59-62.

18. PITOT,H.C.,PERAINO,C.,MORSE,P.A,,Jr.及POTTER,V.R.(1964). HEPATOMAS IN TISSUE CULTURE COMPARED WITH ADAPTING LIVER IN VIVO. Natl. Cancer Inst. Monogr 13,229-245。18. PITOT, HC, PERAINO, C., MORSE, PA, Jr. and POTTER, VR (1964). HEPATOMAS IN TISSUE CULTURE COMPARED WITH ADAPTING LIVER IN VIVO. Natl. Cancer Inst. Monogr 13 , 229-245.

19. Raju,T.S.,Briggs,J.B.,Borge,S.M.及Jones,A.J.(2000). Species-specific variation in glycosylation of IgG: evidence for the species-specific sialylation and branch-specific galactosylation and importance for engineering recombinant glycoprotein therapeutics. Glycobiology 10,477-486。19. Raju, TS, Briggs, JB, Borge, SM and Jones, AJ (2000). Species-specific variation in glycosylation of IgG: evidence for the species-specific sialylation and branch-specific galactosylation and importance for engineering recombinant glycoprotein therapeutics. Glycobiology 10 , 477-486.

20. REUBER,M.D.(1961). A transplantable bile-secreting hepatocellular carcinoma in the rat. J. Natl. Cancer Inst. 26,891-899。20. REUBER, MD (1961). A transplantable bile-secreting hepatocellular carcinoma in the rat. J. Natl. Cancer Inst. 26 , 891-899.

21. Ripka,J.,Adamany,A.及Stanley,P.(1986). Two Chinese hamster ovary glycosylation mutants affected in the conversion of GDP-mannose to GDP-fucose. Arch. Biochem. Biophys. 249,533-545。21. Ripka, J., Adamany, A . , And Stanley, P. (1986). Two Chinese hamster ovary glycosylation mutants affected in the conversion of GDP-mannose to GDP-fucose. Arch. Biochem. Biophys. 249, 533-545 .

22. Sambrook,J.,Fritsch,d.F.及Maniatis,T.(1989). Molecular Cloning: A Laboratory Manual.(Cold Spring Harbor: Cold Spring Harbor Laboratory Press).22. Sambrook, J., Fritsch, d. F. and Maniatis, T. (1989). Molecular Cloning: A Laboratory Manual. (Cold Spring Harbor: Cold Spring Harbor Laboratory Press).

23. Scallon,B.J.,Tam,S.H.,McCarthy,S.G.,Cai,A.N.及Raju,T.S.(2007). Higher levels of sialylated Fc glycans in immunoglobulin G molecules can adversely impact functionality. Mol. Immunol. 44,1524-1534。23. Scallon, BJ, Tam, SH, McCarthy, SG, Cai, AN and Raju, TS (2007). Higher levels of sialylated Fc glycans in immunoglobulin G molecules can adversely affect functionality. Mol. Immunol. 44 , 1524-1534.

24. Shields,R.L.,Lai,J.,Keck,R.,O'Connell,L.Y.,Hong,K.,Meng,Y.G.,Weikert,S.H.及Presta,L.G.(2002). Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J. Biol. Chem. 277,26733-26740。24. Shields, RL, Lai, J., Keck, R., O'Connell, LY, Hong, K., Meng, YG, Weikert, SH and Presta, LG (2002). Lack of fucose on human IgG1 N- linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J. Biol. Chem. 277, 26733-26740.

25. Shinkawa,T.,Nakamura,K.,Yamane,N.,Shoji-Hosaka,E.,Kanda,Y.,Sakurada,M.,Uchida,K.,Anazawa,H.,Satoh,M.,Yamasaki,M.,Hanai,N.及Shitara,K.(2003). The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J. Biol. Chem. 278,3466-3473。25. Shinkawa, T., Nakamura, K., Yamane, N., Shoji-Hosaka, E., Kanda, Y., Sakurada, M., Uchida, K., Anazawa, H., Satoh, M., Yamasaki , M., Hanai, N. and Shitara, K. (2003). The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity J. Biol. Chem. 278 , 3466-3473.

26. Umana,P.,Jean-Mairet,J.及Bailey,J.E.(1999). Tetracycline-regulated overexpression of glycosyltransferases in Chinese hamster ovary cells. Biotechnol. Bioeng. 65,542-549。26. Umana, P., Jean-Mairet, J. and Bailey, JE (1999). Tetracycline-regulated overexpression of glycosyltransferases in Chinese hamster ovary cells. Biotechnol. Bioeng. 65 , 542-549.

27. Urlaub,G. and Chasin,L.A.(1980). Isolation of Chinese hamster cell mutants deficient in dihydrofolate reductase activity. Proc. Natl. Acad. Sci. U. S. A 77,4216-4220。27. Urlaub, G. and Chasin, LA (1980). Isolation of Chinese hamster cell mutants deficient in dihydrofolate reductase activity. Proc. Natl. Acad. Sci. US A 77 , 4216-4220.

28. Urlaub,G.,Kas,E.,Carothers,A.M.及Chasin,L.A.(1983). Deletion of the diploid dihydrofolate reductase locus from cultured mammalian cells. Cell 33,405-412。28. Urlaub, G., Kas, E., Carothers, AM and Chasin, LA (1983). Deletion of the diploid dihydrofolate reductase locus from cultured mammalian cells. Cell 33 , 405-412.

29. Walsh,G. and Jefferis,R.(2006). Post-translational modifications in the context of therapeutic proteins. Nat. Biotechnol. 24,1241-1252。29. Walsh, G. and Jefferis, R. (2006). Post-translational modifications in the context of therapeutic proteins. Nat. Biotechnol. 24 , 1241-1252.

30. Werner,R.G.(2004). Economic aspects of commercial manufacture of biopharmaceuticals. J. Biotechnol. 113,171-182。30. Werner, RG (2004). Economic aspects of commercial manufacture of biopharmaceuticals. J. Biotechnol. 113 , 171-182.

31. Wright,A.,Sato,Y.,Okada,T.,Chang,K.,Endo,T.及Morrison,S.(2000). In vivo trafficking and catabolism of IgG1 antibodies with Fc associated carbohydrates of differing structure. Glycobiology 10,1347-1355。31. Wright, A., Sato, Y., Okada, T., Chang, K., Endo, T. and Morrison, S. (2000). In vivo trafficking and catabolism of IgG1 antibodies with Fc associated carbohydrates of differing structure Glycobiology 10 , 1347-1355.

32. Yamane-Ohnuki,N.,Kinoshita,S.,Inoue-Urakubo,M.,Kusunoki,M.,Iida,S.,Nakano,R.,Wakitani,M.,Niwa,R.,Sakurada,M.,Uchida,K.,Shitara,K.及Satoh,M.(2004). Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity. Biotechnol. Bioeng. 87,614-622。32. Yamane-Ohnuki, N., Kinoshita, S., Inoue-Urakubo, M., Kusunoki, M., Iida, S., Nakano, R., Wakitani, M., Niwa, R., Sakurada, M. , Uchida, K., Shitara, K. and Satoh, M. (2004). Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for playing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity. Biotechnol. Bioeng. 87 , 614-622.

序列表說明Sequence table description

SEQ ID NO 1:O-連接糖基化位點之胺基酸序列SEQ ID NO 1: O-linked amino acid sequence of a glycosylation site

SEQ ID NO 2:抗CD20 IgG1 mAb重鏈之胺基酸序列SEQ ID NO 2: Amino acid sequence of the anti-CD20 IgG1 mAb heavy chain

SEQ ID NO 3:抗CD20 IgG1 mAb輕鏈之胺基酸序列SEQ ID NO 3: Amino acid sequence of the anti-CD20 IgG1 mAb light chain

SEQ ID NO 4:抗CD20 IgG4 mAb重鏈之胺基酸序列SEQ ID NO 4: Amino acid sequence of the anti-CD20 IgG4 mAb heavy chain

SEQ ID NO 5:抗CD20 IgG4 mAb輕鏈之胺基酸序列SEQ ID NO 5: Amino acid sequence of the anti-CD20 IgG4 mAb light chain

SEQ ID NO 6:MCP1-Fc融合蛋白之胺基酸序列SEQ ID NO 6: amino acid sequence of MCP1-Fc fusion protein

SEQ ID NO 7:EPO-Fc融合蛋白之胺基酸序列SEQ ID NO 7: amino acid sequence of EPO-Fc fusion protein

<110> 德商百靈佳殷格翰國際股份有限公司<110> Dessert Bailingjia Ingelheim International Co., Ltd.

<120> H4-II-E大鼠細胞中低海藻糖抗體之生產<120> Production of low trehalose antibodies in H4-II-E rat cells

<130> p01-2641 pct-ff<130> p01-2641 pct-ff

<140> 100134655<140> 100134655

<141> 2011-09-26<141> 2011-09-26

<150> 10180321.1;11156848.1<150> 10180321.1; 11156848.1

<151> 2010-09-27;2011-03-03<151> 2010-09-27;2011-03-03

<160> 7<160> 7

<170> PatentIn version 3.3<170> PatentIn version 3.3

<210> 1<210> 1

<211> 4<211> 4

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> o-連接糖基化位點序列<223> o-linked glycosylation site sequence

<220><220>

<221> X<221> X

<222> (1)..(1)<222> (1)..(1)

<223> Thr或ser<223> Thr or ser

<220><220>

<221> X<221> X

<222> (2)..(3)<222> (2)..(3)

<223> 任一習用胺基酸:Ala、Arg、Asn、Asp、Cys、Glu、Gln、Gly、His、Ile、Leu、Lys、Met、Pro、Ser、Thr、Tyr、Val<223> Any of the conventional amino acids: Ala, Arg, Asn, Asp, Cys, Glu, Gln, Gly, His, Ile, Leu, Lys, Met, Pro, Ser, Thr, Tyr, Val

<400> 1<400> 1

<210> 2<210> 2

<211> 451<211> 451

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> 抗CD20 IgG1 mAb重鏈之胺基酸序列<223> Amino acid sequence of the anti-CD20 IgG1 mAb heavy chain

<400> 2<400> 2

<210> 3<210> 3

<211> 213<211> 213

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> 抗CD20 IgG1 mAb輕鏈之胺基酸序列<223> Amino acid sequence of the anti-CD20 IgG1 mAb light chain

<400> 3<400> 3

<210> 4<210> 4

<211> 449<211> 449

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> 抗CD20 IgG4 mAb重鏈之胺基酸序列<223> Amino acid sequence of the anti-CD20 IgG4 mAb heavy chain

<400> 4<400> 4

<210> 5<210> 5

<211> 213<211> 213

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> 抗CD20 IgG4 mAb輕鏈之胺基酸序列<223> Amino acid sequence of the anti-CD20 IgG4 mAb light chain

<400> 5<400> 5

<210> 6<210> 6

<211> 303<211> 303

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> MCP1-FC融合蛋白之胺基酸序列<223> Amino acid sequence of MCP1-FC fusion protein

<400> 6<400> 6

<210> 7<210> 7

<211> 401<211> 401

<212> PRT<212> PRT

<213> 人工<213> Labor

<220><220>

<223> EPO-FC融合蛋白之胺基酸序列<223> Amino acid sequence of EPO-FC fusion protein

<400> 7<400> 7

圖1:不同細胞系中所產生糖蛋白上糖結構之預測性相對含量。Figure 1: Predictive relative amounts of glycostructures on glycoproteins produced in different cell lines.

分析源自不同物種及該等物種內不同組織之已建立細胞系之糖性質。藉由量測細胞中之酶促活性及分析細胞表面上之結構來估計該等細胞系中所產生蛋白上海藻糖、α-2,6唾液酸化結構、N-乙醯基神經胺酸(NeuAc)、半乳糖-1,3-半乳糖(Gal-1,3-Gal)及N-羥乙醯神經胺酸(NeuGc)之相對含量。將所獲得值正規化為在中國倉鼠卵巢細胞系(CHO)中獲得之結果且繪製為各別糖結構之經預測相對含量之曲線。Analysis of the sugar properties of established cell lines derived from different species and different tissues within such species. Estimating the protein produced in these cell lines by measuring the enzymatic activity in cells and analyzing the structure on the cell surface, trehalose, α-2,6 sialylation structure, N-acetylthioneamine (NeuAc) ), the relative content of galactose-1,3-galactose (Gal-1,3-Gal) and N-hydroxyacetamidine neurone (NeuGc). The values obtained were normalized to the results obtained in the Chinese hamster ovary cell line (CHO) and plotted as a predicted relative content of the individual sugar structures.

縮寫:ha,倉鼠;mo,小鼠;gp,豚鼠;rab,兔;go,山羊;sh,綿羊;hu,人類;ch,雞;du,鴨;te,睪丸;ov,卵巢;pa,胰腺;ki,腎;li,肝;carc,肝瘤/癌;eo,食管;lu,肺;br.canc,乳癌;co.carc,結腸癌;my,骨髓瘤;pr.ly,初代淋巴母細胞樣細胞;pr.b.m,骨髓幹細胞;emb.fib,胚胎纖維母細胞。Abbreviations: ha, hamster; mo, mouse; gp, guinea pig; rab, rabbit; go, goat; sh, sheep; hu, human; ch, chicken; du, duck; te, testicle; ov, ovary; pa, pancreas ;ki, kidney; li, liver; carc, hepatoma/cancer; eo, esophagus; lu, lung; br. canc, breast cancer; co.carc, colon cancer; my, myeloma; pr.ly, primary lymphoblast Like cells; pr. bm, bone marrow stem cells; emb. fib, embryonic fibroblasts.

圖2:CHO、Lec13、YB2/0及H4-II-E細胞中產生之IgG1抗體在其糖基化模式:海藻糖基化方面有所不同。Figure 2: IgGl antibodies produced in CHO, Lec13, YB2/0 and H4-II-E cells differ in their glycosylation pattern: fucosylation.

分析CHO-DG44細胞、CHO-Lec13突變體、YB2/0大鼠骨髓瘤細胞及H4-II-E大鼠肝瘤細胞中所產生IgG1抗體之Fc聚糖之結構及組成。在藉由用PNGase F實施酶促消化還原後自純化抗體釋放聚糖。將聚糖純化,用2-胺基苯甲醯胺(2-AB)進行螢光標記且在用外切糖苷酶α-海藻糖苷酶或其他外切糖苷酶處理之前及之後,在HPLC管柱上進行分級分離。大多數糖結構係非唾液酸化二枝結構聚糖。自外切糖苷消化之前及之後之層析峰面積比率來計算海藻糖基化及非海藻糖基化二枝結構聚糖及其他糖苷結構(唾液酸化聚糖、高甘露糖結構或雜合聚糖)之比例。The structure and composition of Fc glycans of IgG1 antibodies produced in CHO-DG44 cells, CHO-Lec13 mutants, YB2/0 rat myeloma cells and H4-II-E rat hepatoma cells were analyzed. The glycan is released from the purified antibody after reduction by enzymatic digestion with PNGase F. The glycan was purified and fluorescently labeled with 2-aminobenzamide (2-AB) and before and after treatment with exoglycosidase alpha-trehalosidase or other exoglycosidases in HPLC column Fractionation is performed on. Most sugar structures are non-sialylated bi-glycans. Calculation of the fucosylated and non-fucosylated dimeric structural glycans and other glycoside structures (sialylated glycans, high mannose structures or hybrid glycans) from the ratio of chromatographic peak areas before and after digestion with exoglycosides The ratio of ).

圖3:CHO及H4-II-E中所產生IgG1抗體之糖模式:半乳糖基化Figure 3: Sugar pattern of IgG1 antibodies produced in CHO and H4-II-E: galactosylation

分析CHO-DG44細胞及H4-II-E大鼠肝瘤細胞中所產生IgG1抗體之Fc聚糖之結構及組成。在藉由用PNGase F實施酶促消化還原後自純化抗體釋放聚糖。將聚糖純化,用2-胺基苯甲醯胺(2-AB)進行螢光標記且在用外切糖苷酶β-半乳糖苷酶或其他外切糖苷酶處理之前及之後,在HPLC管柱上進行分級分離。自外切糖苷消化之前及之後之層析峰面積比率來計算半乳糖基化對非半乳糖基化二枝結構聚糖之百分比。The structure and composition of Fc glycans of IgG1 antibodies produced in CHO-DG44 cells and H4-II-E rat hepatoma cells were analyzed. The glycan is released from the purified antibody after reduction by enzymatic digestion with PNGase F. The glycan was purified, fluorescently labeled with 2-aminobenzamide (2-AB) and before and after treatment with exoglycosidase β-galactosidase or other exoglycosidases, in HPLC tubes Fractionation was carried out on the column. The percentage of galactosylated versus non-galactosylated bi-structured glycans was calculated from the ratio of chromatographic peak areas before and after exo-glycoside digestion.

圖4:CHO及H4-II-E中產生之IgG1抗體之糖模式:唾液酸化Figure 4: Sugar pattern of IgG1 antibodies produced in CHO and H4-II-E: sialylation

分析CHO-DG44細胞及H4-II-E大鼠肝瘤細胞中所產生IgG1抗體之Fc聚糖之結構及組成。在藉由用PNGase F實施酶促消化還原後自純化抗體釋放聚糖。將聚糖純化,用2-胺基苯甲醯胺(2-AB)進行螢光標記且在用外切糖苷酶神經胺酸酶或其他外切糖苷酶處理之前及之後,在HPLC管柱上進行分級分離。自外切糖苷消化之前及之後之層析峰面積比率來計算唾液酸化二枝結構聚糖之百分比。The structure and composition of Fc glycans of IgG1 antibodies produced in CHO-DG44 cells and H4-II-E rat hepatoma cells were analyzed. The glycan is released from the purified antibody after reduction by enzymatic digestion with PNGase F. The glycan was purified, fluorescently labeled with 2-aminobenzamide (2-AB) and applied to the HPLC column before and after treatment with exoglycosidase neuraminidase or other exoglycosidases Perform fractionation. The percentage of sialylated di-branched glycans was calculated from the ratio of chromatographic peak areas before and after digestion with exo glycosides.

圖5:使H4-II-E細胞適應於在無血清培養基中懸浮生長Figure 5: Adapting H4-II-E cells to suspension growth in serum-free medium

(A) H4-II-E細胞在含有10% FCS之MEMalpha培養基中之貼壁生長。(B)H4-II-E細胞在適應在振盪燒瓶中之無血清無Ca培養基中生長後之懸浮培養物。(C)以兩種不同接種物細胞密度接種之BI SFM培養基中之H4-II-E培養物之生長曲線。在37℃、5% CO2及120 rpm下在振盪燒瓶中培育培養物。在所示時間點量測活細胞濃度。(A) Adherent growth of H4-II-E cells in MEMalpha medium containing 10% FCS. (B) Suspension culture of H4-II-E cells after growth in serum-free Ca-free medium in shake flasks. (C) Growth curves of H4-II-E cultures in BI SFM medium inoculated at two different inoculum cell densities. Cultures were incubated in shake flasks at 37 ° C, 5% CO 2 and 120 rpm. The viable cell concentration was measured at the indicated time points.

縮寫:BI-SFM,Boehringer Ingelheim無血清無鈣培養基;FCS,胎牛血清;VCC,活細胞濃度;4×105個細胞/ml=400,000個細胞/ml;6×105個細胞/ml=600,000個細胞/ml。Abbreviations: BI-SFM, Boehringer Ingelheim serum-free calcium-free medium; FCS, fetal bovine serum; VCC, viable cell concentration; 4 × 10 5 cells / ml = 400,000 cells / ml; 6 × 10 5 cells / ml = 600,000 cells/ml.

圖6:H4-II-E細胞對細胞凋亡之低敏感性及對細胞壓力之高穩健性Figure 6: Low sensitivity of H4-II-E cells to apoptosis and high robustness to cellular stress

(A)在將含有等細胞數之細胞懸浮物加熱至42℃並維持2小時且隨後在37℃、5% CO2下培育22小時後,H4-II-E細胞(黑色條形圖)及YB2/0細胞(灰色條形圖)之相對活細胞密度及活力。將對照細胞在37℃、5% CO2下培養24小時。在熱壓力後,H4-II-E細胞顯示顯著較高之活細胞密度及活力。(B)在37℃、5% CO2下在用去礦質水(低離子強度)或10×PBS(高離子強度)稀釋之培養基中培育24小時後,H4-II-E細胞及YB2/0細胞之相對活細胞密度及活力。在未稀釋培養基中培養對照細胞。在低或高滲透性下培育後,H4-II-E細胞顯示顯著較高之活細胞密度及活力。(C)在37℃、5% CO2下用2 μg/ml或5 μg/ml嘌呤黴素將含有等細胞數之細胞懸浮物處理48小時後,H4-II-E細胞及YB2/0細胞之相對活細胞密度及活力。在37℃、5% CO2下在無嘌呤黴素培養基中將對照細胞培養48小時。在藥物處理後H4-II-E細胞顯示顯著較高之活細胞密度及活力。(A) H4-II-E cells (black bars) after heating the cell suspension containing equal cell numbers to 42 ° C for 2 hours and then incubated at 37 ° C, 5% CO 2 for 22 hours Relative living cell density and viability of YB2/0 cells (grey bar graph). Control cells were incubated for 24 hours at 37 ° C, 5% CO 2 . After heat stress, H4-II-E cells showed significantly higher viable cell density and viability. (B) H4-II-E cells and YB2/0 after incubation for 24 hours at 37 ° C, 5% CO 2 in medium diluted with demineralized water (low ionic strength) or 10×PBS (high ionic strength) Relative living cell density and viability of cells. Control cells were cultured in undiluted medium. After incubation under low or high permeability, H4-II-E cells showed significantly higher viable cell density and viability. (C) H4-II-E cells and YB2/0 cells after treatment with cell suspension containing equal cell number for 48 hours at 37 ° C, 5% CO 2 with 2 μg/ml or 5 μg/ml puromycin Relative living cell density and vigor. Control cells were cultured for 48 hours in a puromycin-free medium at 37 ° C, 5% CO 2 . H4-II-E cells showed significantly higher viable cell density and viability after drug treatment.

圖7:單細胞懸浮培育H4-II-E細胞對Ca2+減少或無Ca2+培養基之需要。Figure 7: Single cell suspension incubation of H4-II-E cells for Ca 2+ reduced or no Ca 2+ medium.

(A)使用Hitachi 917(Roche)來分析兩種適於懸浮培育H4-II-E細胞之培養基之Ca含量。含有極低Ca濃度之AEM培養基及BI專利培養基之無Ca形式,二者皆適於單細胞懸浮培育H4-II-E細胞(B、D)。(B)-(E)以3×105個細胞/ml=300,000個細胞/ml之密度將適應於在AEM中或在BI(無Ca)培養基中懸浮生長之H4-II-E細胞接種於存於12孔板中之添加或不添加CaCl2之所示培養基中。接種後3天對生長形態及細胞聚集進行顯微分析。(B) H4-II-E細胞在AEM培養基中以單細胞形式懸浮生長。(C) H4-II-E細胞在補充有1 mM=1 mMol/L CaCl2之AEM中形成大聚集體。(D) H4-II-E細胞在無Ca BI專利培養基中以單細胞形式懸浮生長。(E) H4-II-E細胞在BI培養基(根據(A)中之分析,含有1.38 mM=1.38 mMol/L Ca)中形成大聚集體。(F)以3×105個細胞/ml=300,000個細胞/ml之密度將適應於在BI(無Ca)培養基中懸浮生長之H4-II-E細胞接種於存於12孔板中之BI培養基(含有Ca)中並將所示量之EDTA添加至培養物中。接種後3天對生長形態及細胞聚集進行顯微分析。應注意,培養基中游離Ca2+之劑量依賴性、EDTA介導之消耗可減少細胞聚集並提高懸浮單細胞之比例。(A) Hitachi 917 (Roche) was used to analyze the Ca content of two media suitable for suspension incubation of H4-II-E cells. AEM medium containing very low Ca concentration and no Ca form of BI patent medium, both suitable for single cell suspension incubation of H4-II-E cells (B, D). (B)-(E) Inoculation of H4-II-E cells adapted to suspension growth in AEM or in BI (Ca-free) medium at a density of 3 x 10 5 cells/ml = 300,000 cells/ml Store in a 12-well plate in the medium with or without CaCl 2 added. Microscopic analysis of growth morphology and cell aggregation was performed 3 days after inoculation. (B) H4-II-E cells were grown in suspension in a single cell format in AEM medium. (C) H4-II-E cells formed large aggregates in AEM supplemented with 1 mM = 1 mMol/L CaCl 2 . (D) H4-II-E cells were grown in suspension in a single cell format in a Ca BI-free medium. (E) H4-II-E cells formed large aggregates in BI medium (containing 1.38 mM = 1.38 mMol/L Ca according to the analysis in (A)). (F) Inoculation of H4-II-E cells adapted to suspension growth in BI (Ca-free) medium at a density of 3 × 10 5 cells/ml = 300,000 cells/ml in BI stored in a 12-well plate The medium (containing Ca) was added to the culture in the amount of EDTA indicated. Microscopic analysis of growth morphology and cell aggregation was performed 3 days after inoculation. It should be noted that the dose-dependent, EDTA-mediated consumption of free Ca 2+ in the medium reduces cell aggregation and increases the proportion of suspended single cells.

圖8:經懸浮H4-II-E細胞之Ca2+濃度依賴性及Mg2+獨立性聚集。Figure 8: Ca 2+ concentration-dependent and Mg 2+ independent aggregation of suspended H4-II-E cells.

(A)以4×105個細胞/ml=400,000個細胞/ml之密度將適應於在BI(無Ca)培養基中懸浮生長之H4-II-E細胞接種於存於12孔板中之BI(無Ca)培養基中並將所示量之CaCl2添加至培養物中。接種後2天對生長形態及細胞聚集進行顯微分析。注意經懸浮H4-II-E細胞之濃度依賴性、Ca2+介導之細胞聚集。(B)以4×105個細胞/ml=400,000個細胞/ml之密度將適應於在BI(無Ca)培養基中懸浮生長之H4-II-E細胞接種於存於12孔板中之BI(無Ca)培養基中並將所示量之MgCl2添加至培養物中。接種後2天對生長形態及細胞聚集進行顯微分析。應注意細胞獨立於Mg2+濃度以單細胞形式懸浮生長。(C)以3×105個細胞/ml=300,000個細胞/ml之密度將適應於在AEM培養基中懸浮生長之H4-II-E細胞接種於存於振盪燒瓶中之AEM培養基中且將所示量之CaCl2或MgCl2添加至培養物中。在37℃、5% CO2及120 rpm下培育培養物。利用CEDEX細胞量化系統(Innovatis)來分析活細胞密度及細胞聚集比率。應注意,若培養基中之CaCl2濃度高於100 μM,則培養物之活細胞密度顯著下降。相比之下,增加MgCl2之濃度對活細胞密度無影響。此外,應注意,細胞聚集比率隨著CaCl2濃度增加而增加且在CaCl2濃度>250 μM時達到飽和。相比之下,增加MgCl2之濃度對H4-II-E細胞之聚集比率無影響。(A) Inoculation of H4-II-E cells adapted to suspension growth in BI (Ca-free) medium at a density of 4 × 10 5 cells/ml = 400,000 cells/ml in BI stored in a 12-well plate (Ca-free) medium and the indicated amount of CaCl 2 was added to the culture. Microscopic analysis of growth morphology and cell aggregation was performed 2 days after inoculation. Note the concentration-dependent, Ca 2+ -mediated cell aggregation of suspended H4-II-E cells. (B) Inoculation of H4-II-E cells adapted to suspension growth in BI (Ca-free) medium at a density of 4 × 10 5 cells/ml = 400,000 cells/ml in BI stored in a 12-well plate (Ca-free) medium and the indicated amount of MgCl 2 was added to the culture. Microscopic analysis of growth morphology and cell aggregation was performed 2 days after inoculation. It should be noted that the cells are suspended in a single cell form independently of the Mg 2+ concentration. (C) Inoculation of H4-II-E cells adapted to suspension growth in AEM medium at a density of 3 × 10 5 cells/ml = 300,000 cells/ml in AEM medium stored in a shaking flask and An amount of CaCl 2 or MgCl 2 was added to the culture. The culture was incubated at 37 ° C, 5% CO 2 and 120 rpm. Live cell density and cell aggregation ratio were analyzed using the CEDEX Cell Quantification System (Innovatis). It should be noted that if the CaCl 2 concentration in the medium is higher than 100 μM, the viable cell density of the culture is significantly decreased. In contrast, increasing the concentration of MgCl 2 has no effect on viable cell density. Furthermore, it should be noted that the cell aggregation ratio increases as the CaCl 2 concentration increases and reaches saturation when the CaCl 2 concentration is >250 μM. In contrast, increasing the concentration of MgCl 2 had no effect on the aggregation ratio of H4-II-E cells.

圖9:CHO及H4-II-E中所產生IgG1與FcγRIIIa之結合親和力。Figure 9: Binding affinities of IgG1 and FcγRIIIa produced in CHO and H4-II-E.

如下使用BIAcore T100儀器及CM5感測器晶片(BIACORE,Uppsala,Sweden)來量測不同細胞系中產生之IgG1與FcγRIIIa之結合動力學。將可溶性重組FcγRIIIa固定至BIAcore感測器晶片上。以6種不同濃度(自4.17 nM至133.3 nM)將經純化IgG1稀釋於HBS-EP緩衝液(0.01 M HEPES,0.15 M NaCl,3 mM EDTA,0.005%表面活性劑P20,pH 7.4)中,且以5 mL/min之流速將每一經稀釋IgG1注射於受體捕獲感測器表面上。在25℃下利用HBS-EP作為運行緩衝液來實施該等實驗。將無試樣IgG1之緩衝溶液注射於受體捕獲感測器表面上作為空白對照。藉由以10 mL/min之流速將7.5 mM HCl注射30 s來去除結合至感測器表面之可溶性FcγRIIIb及IgG1。在數據分析前針對空白對照校正藉由注射IgG1獲得之數據。藉由穩態分析使用BIAcore T100動力學評估軟體(BIACORE)來計算對FcγRIIIa之親和力(KD)。應注意,H4-II-E細胞系中產生之IgG1與受體之結合親和力高於CHO中產生之IgG1。BIAcore T100 instruments and CM5 sensor wafers (BIACORE, Uppsala, Sweden) were used to measure the binding kinetics of IgGl and FcyRIIIa produced in different cell lines as follows. Soluble recombinant FcyRIIIa was immobilized onto a BIAcore sensor wafer. Purified IgG1 was diluted in HBS-EP buffer (0.01 M HEPES, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, pH 7.4) at 6 different concentrations (from 4.17 nM to 133.3 nM), and Each diluted IgGl was injected onto the surface of the receptor capture sensor at a flow rate of 5 mL/min. The experiments were carried out using HBS-EP as a running buffer at 25 °C. A buffer solution without sample IgG1 was injected onto the surface of the receptor capture sensor as a blank control. Soluble FcγRIIIb and IgG1 bound to the sensor surface were removed by injecting 7.5 mM HCl for 30 s at a flow rate of 10 mL/min. Data obtained by injection of IgG1 were corrected for blank controls prior to data analysis. Affinity (KD) for FcyRIIIa was calculated by steady state analysis using BIAcore T100 Kinetic Evaluation Software (BIACORE). It should be noted that the binding affinity of IgG1 produced in the H4-II-E cell line to the receptor is higher than that produced in CHO.

圖10:CHO及H4-II-E中產生之IgG1之效應子功能:ADCC。Figure 10: Effector function of IgG1 produced in CHO and H4-II-E: ADCC.

藉由乳酸脫氫酶(LDH)釋放分析來實施ADCC分析,其中使用藉由Lymphoprep(AXIS SHIELD,Dundee,UK)自健康供體製備之人類外周血液單核細胞(PBMC)作為效應子細胞。將靶腫瘤細胞(表現人類CD20之人類伯基特氏淋巴瘤細胞系Ramos)之等分試樣分配至96孔U形底板(存於50 μl/孔中之10,000個細胞)中且在PBMC(100 μL)存在下以20/1之E/T比率與抗體之連續稀釋液(50 μL)一起培育。在37℃下培育4 h後,使用非放射性細胞毒性分析套組(Promega,Madison,WI)量測上清液LDH活性。根據下式自試樣活性計算特異性細胞溶解%:特異性溶解[%]=100×(E-SE-ST)/(M-ST)。FU代表螢光單位。ADCC analysis was performed by lactate dehydrogenase (LDH) release assay using human peripheral blood mononuclear cells (PBMC) prepared from healthy donors by Lymphoprep (AXIS SHIELD, Dundee, UK) as effector cells. Aliquots of target tumor cells (human Burkitt's lymphoma cell line Ramos expressing human CD20) were dispensed into 96-well U-shaped bottom plates (10,000 cells in 50 μl/well) and in PBMC ( Incubate with serial dilutions of antibody (50 μL) at an E/T ratio of 20/1 in the presence of 100 μL). Supernatant LDH activity was measured using a non-radioactive cytotoxicity assay kit (Promega, Madison, WI) after incubation for 4 h at 37 °C. The specific cell lysis % was calculated from the sample activity according to the following formula: specific dissolution [%] = 100 × (ES E - S T ) / (MS T ). FU stands for fluorescent unit.

應注意,與CHO中產生之IgG1相比,H4-II-E中產生之IgG1具有更強ADCC活化。It should be noted that IgG1 produced in H4-II-E has stronger ADCC activation than IgG1 produced in CHO.

圖11:CHO及H4-II-E中所產生IgG1與FcRn之結合親和力。Figure 11: Binding affinities of IgG1 and FcRn produced in CHO and H4-II-E.

使用BIAcore T100儀器及CM5感測器晶片來量測人類IgG1-FcRn相互作用之動力學。使用胺偶合套組(BIACORE)將抗人類b2-微球蛋白單株抗體(Abcam,Cambridge,UK)固定至晶片上。藉由以5 mL/min之流速注射可溶性FcRn-b2微球蛋白複合物,藉由經固定抗b2-微球蛋白抗體來捕獲該複合物。將無該複合物之緩衝溶液注射於抗體捕獲感測器表面上作為空白對照。以5種不同濃度(自4.17 nM至66.7 nM)將每一經純化IgG稀釋於pH調節至6.0之HBS-EP+緩衝液(0.01 M HEPES,0.15 M NaCl,3 mM EDTA,0.05%表面活性劑P20)中,且以5 mL/min之流速將每一經稀釋IgG1注射於複合物捕獲感測器表面或空白上。藉由以60 mL/min之流速將7.5 mM HCl注射1 min來去除結合至感測器表面上之可溶性FcRn及IgG1。在25℃下利用HBS-EP+作為運行緩衝液來實施該等實驗。使用藉由減去空白獲得之數據進行數據分析。使用BIAcore T100評估軟體藉由二元擬合模型來計算表觀締合速率常數(ka)、解離速率常數(kd)及結合親和力(KD)。應注意,H4-II-E中產生之抗體結合FcRn之程度與CHO中產生之抗體相當。The kinetics of the human IgGl-FcRn interaction was measured using a BIAcore T100 instrument and a CM5 sensor wafer. Anti-human b2-microglobulin monoclonal antibody (Abeam, Cambridge, UK) was immobilized onto the wafer using an amine coupling kit (BIACORE). The complex was captured by immobilization of an anti-b2-microglobulin antibody by injecting a soluble FcRn-b2 microglobulin complex at a flow rate of 5 mL/min. A buffer solution without the complex was injected onto the surface of the antibody capture sensor as a blank control. Each purified IgG was diluted in 5 different concentrations (from 4.17 nM to 66.7 nM) to HBS-EP+ buffer (0.01 M HEPES, 0.15 M NaCl, 3 mM EDTA, 0.05% Surfactant P20) adjusted to pH 6.0. Each diluted IgGl was injected onto the composite capture sensor surface or blank at a flow rate of 5 mL/min. Soluble FcRn and IgGl bound to the surface of the sensor were removed by injecting 7.5 mM HCl for 1 min at a flow rate of 60 mL/min. These experiments were carried out using HBS-EP+ as a running buffer at 25 °C. Data analysis was performed using data obtained by subtracting blanks. Apparent association rate constants (ka), dissociation rate constants (kd), and binding affinities (KD) were calculated using a BIAcore T100 evaluation software by a binary fit model. It should be noted that the degree of binding of the antibody produced in H4-II-E to FcRn is comparable to that produced in CHO.

(無元件符號說明)(no component symbol description)

Claims (29)

一種包含編碼抗體或Fc-融合蛋白之核酸序列的大鼠肝瘤細胞,其中該核酸序列可操作地連接至至少一種允許表現該編碼抗體或Fc-融合蛋白之核酸序列的調節序列。A rat hepatoma cell comprising a nucleic acid sequence encoding an antibody or an Fc-fusion protein, wherein the nucleic acid sequence is operably linked to at least one regulatory sequence that permits expression of the nucleic acid sequence encoding the antibody or Fc-fusion protein. 如請求項1之大鼠肝瘤細胞,其中該細胞係H4-II-E細胞。The rat hepatoma cell of claim 1, wherein the cell line is a H4-II-E cell. 如請求項1或2之大鼠肝瘤細胞,其中該細胞係:a) 源自選自由以下組成之群之細胞的細胞:歐洲細胞培養物保存中心(European Collection of Cell Cultures)(ECACC,目錄編號87031301)、美國典型培養物保存中心(American Type Culture Collection)(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或b) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或c) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心(Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH))之細胞,或d) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或e) a)或b)或c)或d)中任一細胞之衍生物或子代。The rat hepatoma cell of claim 1 or 2, wherein the cell line: a) is derived from a cell selected from the group consisting of: European Collection of Cell Cultures (ECACC, catalogue) No. 87031301), American Type Culture Collection (ATCC, accession number CRL-1548), H4-II-E-C3 cell line (CRL-1600 or HPACC number 85601112 or ECACC catalog number 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) and H4-S cell line (HPACC Nr. 89102001), or b) numbered ECACC Catalog No. 87031301 is deposited with the European Cell Culture Preservation Center or with cells deposited with the American Type Culture Collection ATCC under the accession number CRL-1548, or c) deposited with DSMZ under the accession number DSM ACC3129 (H4-II-E) Cells of the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, or d) cells deposited in the DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or e) a) or b) or c) or d) a derivative or progeny of any of the cells. 如請求項1或2之大鼠肝瘤細胞,其中該細胞係以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞或其中該細胞係以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞。The rat hepatoma cell of claim 1 or 2, wherein the cell line is deposited in a cell of DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E) or wherein the cell line is registered with a number DSM ACC3130 (H4-II-Es) is deposited in the cells of the DSMZ (German Microbial Culture Preservation Center). 如請求項1或2之大鼠肝瘤細胞,其中進一步a) 由該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有海藻糖之程度小於20%、10%或5%,或b) 由該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有海藻糖之程度在介於0%至20%、0%至10%、0%至5%、0.5%至20%、0.5%至10%、0.5%至5%、1%至20%、1%至10%或1%至5%之間之範圍內。The rat hepatoma cell of claim 1 or 2, wherein a) the glycoside structure contained in the antibody or the Fc-fusion protein expressed by the cell contains trehalose to a degree of less than 20%, 10% or 5%, or b) the glycoside structure contained in the antibody or Fc-fusion protein expressed by the cell contains trehalose to a degree ranging from 0% to 20%, 0% to 10%, 0% to 5%, 0.5% to 20% , in the range of 0.5% to 10%, 0.5% to 5%, 1% to 20%, 1% to 10% or 1% to 5%. 如請求項1或2之大鼠肝瘤細胞,其中進一步a) 由該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有至少一個半乳糖殘基之程度超過40%、45%或50%,或b) 由該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有至少一個半乳糖殘基之程度在介於40%至100%、45%至100%、50%至100%、51%至100%、40%至99.5%、45%至99.5%、50%至99.5%或51%至99.5%、40%至99%、45%至99%、50%至99%或51%至99%之間之範圍內,其中該等糖苷結構較佳含有一或兩個較佳與該等糖苷結構之末端非還原端處之N-乙醯葡糖胺(GlcNAc)連接之半乳糖殘基(G1或G2)。The rat hepatoma cell of claim 1 or 2, wherein a) the glycoside structure contained in the antibody or the Fc-fusion protein expressed by the cell contains at least one galactose residue in an amount of more than 40%, 45% or 50%, or b) the degree to which the glycoside structure contained in the antibody or Fc-fusion protein expressed by the cell contains at least one galactose residue is between 40% and 100%, 45% to 100%, 50% to 100%, 51% to 100%, 40% to 99.5%, 45% to 99.5%, 50% to 99.5% or 51% to 99.5%, 40% to 99%, 45% to 99%, 50% to 99% Or in the range of from 51% to 99%, wherein the glycoside structures preferably contain one or two, preferably attached to the N-acetylglucosamine (GlcNAc) at the terminal non-reducing end of the glycoside structure. A galactose residue (G1 or G2). 如請求項1或2之大鼠肝瘤細胞,其中進一步a) 由該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有末端唾液酸或神經胺酸殘基之程度超過5%或超過10%,或b) 由該細胞表現之該抗體或Fc-融合蛋白中所含糖苷結構含有末端唾液酸或神經胺酸殘基之程度在介於0至8%、1%至8%、5%至10%、10%至50%或10%至45%之間之範圍內。The rat hepatoma cell of claim 1 or 2, wherein a) the glycoside structure contained in the antibody or the Fc-fusion protein expressed by the cell contains a terminal sialic acid or a neuramic acid residue of more than 5% or More than 10%, or b) the degree of glycosidic structure contained in the antibody or Fc-fusion protein expressed by the cell containing terminal sialic acid or a tranexamic acid residue is between 0 and 8%, 1% to 8%, Between 5% and 10%, 10% to 50% or 10% to 45%. 如請求項1或2之大鼠肝瘤細胞,其中進一步帶有選擇標記物基因,例如新黴素-磷酸轉移酶(NPT);針對嘌呤黴素(puromycin)、潮黴素(hygromycin)或吉歐黴素(zeocin)之抗性基因;或可擴增選擇標記物基因,例如二氫葉酸還原酶(DHFR)或麩醯胺酸合成酶(GS)。The rat hepatoma cell of claim 1 or 2, further comprising a selectable marker gene, such as neomycin-phosphotransferase (NPT); for puromycin, hygromycin or gem A resistance gene for zeocin; or an amplifiable selectable marker gene, such as dihydrofolate reductase (DHFR) or glutamate synthase (GS). 如請求項1或2之大鼠肝瘤細胞,其中該允許表現該編碼抗體或Fc-融合蛋白之核酸序列之調節序列係a) 啟動子,或b) 增強子,或c) 5'-UTR序列。The rat hepatoma cell of claim 1 or 2, wherein the regulatory sequence that allows expression of the nucleic acid sequence encoding the antibody or Fc-fusion protein is a) a promoter, or b) an enhancer, or c) 5'-UTR sequence. 如請求項1或2之大鼠肝瘤細胞,其中該抗體或Fc融合蛋白含有連接至N-天冬醯胺酸(N-Asn)殘基之糖苷結構,其中該糖苷結構包含以下糖鏈: The rat hepatoma cell of claim 1 or 2, wherein the antibody or Fc fusion protein comprises a glycoside structure linked to a N-aspartic acid (N-Asn) residue, wherein the glycoside structure comprises the following sugar chain: 如請求項1或2之大鼠肝瘤細胞,其中該抗體或Fc融合蛋白含有連接至N-天冬醯胺酸(N-Asn)殘基之糖苷結構,其中該糖苷結構包含以下糖鏈: The rat hepatoma cell of claim 1 or 2, wherein the antibody or Fc fusion protein comprises a glycoside structure linked to a N-aspartic acid (N-Asn) residue, wherein the glycoside structure comprises the following sugar chain: 如請求項10之大鼠肝瘤細胞,其中根據Kabat EU命名法,該N-Asn較佳為N-Asn(297)。The rat hepatoma cell of claim 10, wherein the N-Asn is preferably N-Asn (297) according to the Kabat EU nomenclature. 如請求項11之大鼠肝瘤細胞,其中根據Kabat EU命名法,該N-Asn較佳為N-Asn(297)。The rat hepatoma cell of claim 11, wherein the N-Asn is preferably N-Asn (297) according to the Kabat EU nomenclature. 如請求項1或2之大鼠肝瘤細胞,其中使該細胞適應於在無血清且鈣減少或較佳無鈣培養基中生長。A rat hepatoma cell according to claim 1 or 2, wherein the cell is adapted to grow in a serum-free and calcium-reduced or preferably calcium-free medium. 如請求項1或2之大鼠肝瘤細胞,其中使該細胞適應於在懸浮培養物中生長。The rat hepatoma cell of claim 1 or 2, wherein the cell is adapted to grow in suspension culture. 如請求項1或2中任一項之大鼠肝瘤細胞,其中該細胞與YB2/0細胞相比對細胞凋亡具有低敏感性及/或對細胞壓力具有高穩健性。The rat hepatoma cell of any one of claims 1 or 2, wherein the cell has low sensitivity to apoptosis and/or high robustness to cell pressure as compared to YB2/0 cells. 一種產生所關注糖蛋白之方法,其特徵在於以下步驟:a) 提供大鼠肝瘤細胞,b) 視情況使該步驟a)之細胞適應於在懸浮培養物中生長,c) 視情況使該步驟a)及/或步驟b)之細胞適應於在無血清培養基中生長,d) 視情況使該步驟a)及/或步驟b)及/或步驟c)之細胞在鈣減少或無鈣培養基中生長,e) 用編碼所關注重組糖蛋白之核酸序列轉染此視情況經適應之大鼠肝瘤細胞,f) 在允許表現該所關注糖蛋白之條件下培育該步驟e)之轉染細胞,g) 視情況分離並純化該所關注糖蛋白。A method of producing a glycoprotein of interest, characterized by the steps of: a) providing a rat hepatoma cell, b) adapting the cell of step a) to growth in suspension culture, as appropriate, c) optionally The cells of step a) and/or step b) are adapted to grow in serum-free medium, d) the cells of step a) and/or step b) and/or step c) are optionally reduced in calcium or calcium-free medium. Medium growth, e) transfection of the optionally adapted rat hepatoma cell with a nucleic acid sequence encoding a recombinant glycoprotein of interest, f) cultivating the transfection of step e) under conditions permitting expression of the glycoprotein of interest Cells, g) The glycoprotein of interest is isolated and purified as appropriate. 如請求項17之方法,其中該大鼠肝瘤細胞係H4-II-E細胞,較佳地,該細胞係:a) 源自選自由以下組成之群之細胞的細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或b) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或c) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或d) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或e) a)或b)或c)或d)中任一細胞之衍生物或子代,最佳地,該細胞係以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞或以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞。The method of claim 17, wherein the rat hepatoma cell line H4-II-E cells, preferably, the cell line: a) is derived from a cell selected from the group consisting of cells: European cell culture preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E-C3 Cell Line (CRL-1600 or HPACC No. 85061112 or ECACC Catalog No. 85061112), H4II Cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) and H4-S cell line (HPACC Nr. 89102001), or b) numbered ECACC catalog No. 87031301 is deposited with the European Cell Culture Preservation Center or with cells deposited with the American Type Culture Collection Center ATCC under the accession number CRL-1548, or c) deposited with DSMZ under the accession number DSM ACC3129 (H4-II-E) Cells of the Culture Collection Center, or d) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or e) a) or b) or c) or d a derivative or progeny of any of the cells, optimally, the cell line is registered under the accession number DSM ACC3129 (H4-II- E) Cells deposited in DSMZ (German Microbial Culture Preservation Center) or cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es). 如請求項17或18之方法,其中該步驟b)、c)及/或d)之培養基另外不含任一動物來源之蛋白/肽。The method of claim 17 or 18, wherein the medium of steps b), c) and/or d) is additionally free of protein/peptide of any animal origin. 如請求項17或18之方法,其中該轉染步驟e)進一步包含將包含編碼該所關注糖蛋白之核酸序列之表現載體引入該大鼠肝瘤細胞中,該核酸序列可操作地連接至至少一種允許表現該編碼所關注糖蛋白之核酸序列之調節序列。The method of claim 17 or 18, wherein the transfection step e) further comprises introducing into the rat hepatoma cell an expression vector comprising a nucleic acid sequence encoding the glycoprotein of interest, the nucleic acid sequence being operably linked to at least A regulatory sequence that allows expression of the nucleic acid sequence encoding the glycoprotein of interest. 如請求項17或18之方法,其中該所關注糖蛋白係抗體或Fc-融合蛋白,較佳具有以下之抗體或Fc-融合蛋白a) FcγRIIIa結合活性且較佳ADCC,或b) 補體結合活性且較佳CDC,或c) 與新生兒Fc受體FcRn之結合活性且較佳血清穩定性。The method of claim 17 or 18, wherein the glycoprotein antibody or Fc-fusion protein of interest preferably has the following antibody or Fc-fusion protein a) FcγRIIIa binding activity and preferably ADCC, or b) complement binding activity And preferably CDC, or c) binding activity to the neonatal Fc receptor FcRn and preferably serum stability. 一種產生(重組)抗體或Fc-融合蛋白之方法,該(重組)抗體或Fc-融合蛋白具有a) FcγRIIIa結合活性及/或b) 補體結合活性及/或c) 與新生兒Fc受體FcRn之結合活性,該方法包含在大鼠肝瘤細胞中產生該抗體或Fc融合蛋白,其中該大鼠肝瘤細胞較佳為H4-II-E細胞,更佳地,該細胞係:i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或iv) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或v) i)或ii)或iii)或iv)中任一細胞之衍生物或子代,最佳地,該細胞係以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞或以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞。A method for producing a (recombinant) antibody or an Fc-fusion protein, the (recombinant) antibody or Fc-fusion protein having a) FcγRIIIa binding activity and/or b) complement binding activity and/or c) and neonatal Fc receptor FcRn The binding activity, the method comprising producing the antibody or Fc fusion protein in a rat hepatoma cell, wherein the rat hepatoma cell is preferably a H4-II-E cell, more preferably, the cell line: i) Cells from cells selected from the group consisting of: European Cell Culture Preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession No. CRL-1548), H4-II-E-C3 cells Department (CRL-1600 or HPACC No. 85061112 or ECACC Catalog No. 85061112), H4II cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) and H4- S cell line (HPACC Nr. 89102001), or ii) deposited in the European Cell Culture Preservation Center under the number ECACC catalog number 87031301 or cells deposited in the American Type Culture Collection Center ATCC under the accession number CRL-1548, or iii) Registration number DSM ACC3129 (H4-II-E) registered in DSMZ (Germany Cell of the Biological Bacterial Preservation Center), or iv) cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es), or v) i) or ii) or iii) or Iv) a derivative or progeny of any of the cells, optimally, the cell line is deposited in the cell of DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3129 (H4-II-E) or under the accession number DSM ACC3130 (H4-II-Es) is deposited in the cells of DSMZ (German Microbial Culture Preservation Center). 如請求項22之方法,其中i) 該如請求項22 a)之抗體或Fc融合蛋白具有抗體依賴性細胞毒性(ADCC)或ii) 該如請求項22 b)之抗體或Fc融合蛋白具有補體依賴性細胞毒性(CDC)或iii)該如請求項22 c)之抗體或Fc融合蛋白具有血清穩定性。The method of claim 22, wherein i) the antibody or Fc fusion protein of claim 22 a) has antibody-dependent cellular cytotoxicity (ADCC) or ii) the antibody or Fc fusion protein of claim 22 b) has complement Dependent Cytotoxicity (CDC) or iii) The antibody or Fc fusion protein of claim 22 c) is serum stable. 一種生成用於生產重組糖蛋白之宿主細胞的方法,其包含:a) 提供大鼠肝瘤細胞,b) 使該步驟a)之大鼠肝瘤細胞適應於在懸浮培養物中生長,及c) 使該步驟a)之大鼠肝瘤細胞適應於在無血清培養基中生長,及d) 使該步驟a)之大鼠肝瘤細胞適應於在鈣減少或無鈣培養基中生長,及e) 視情況使該步驟a)之大鼠肝瘤細胞適應於在不含任一動物來源之蛋白/肽之培養基中生長,及f) 視情況選擇單細胞純系,g) 獲得宿主細胞。A method of producing a host cell for producing a recombinant glycoprotein, comprising: a) providing a rat hepatoma cell, b) adapting the rat hepatoma cell of step a) to growth in suspension culture, and c Adapting the rat hepatoma cells of step a) to growth in serum-free medium, and d) adapting the rat hepatoma cells of step a) to growth in calcium-reduced or calcium-free medium, and e) The rat hepatoma cells of step a) are optionally adapted to grow in a medium free of protein/peptide of any animal origin, and f) a single cell pure line is selected as appropriate, g) obtaining a host cell. 如請求項24之方法,其中該大鼠肝瘤細胞係H4-II-E細胞,較佳地,該細胞係:i) 源自選自由以下組成之群之細胞之細胞:歐洲細胞培養物保存中心(ECACC,目錄編號87031301)、美國典型培養物保存中心(ATCC,寄存編號CRL-1548)、H4-II-E-C3細胞系(CRL-1600或HPACC編號85061112或ECACC目錄編號85061112)、H4II細胞系(HPACC Nr. 89042702)、H4-TG細胞系(CRL-1578)、H5細胞系(HPACC,Nr. 94101905)及H4-S細胞系(HPACC Nr. 89102001),或ii) 以編號ECACC目錄編號87031301寄存於歐洲細胞培養物保存中心或以寄存編號CRL-1548寄存於美國典型培養物保存中心ATCC之細胞,或iii) 以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或iv) 以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞,或v) i)或ii)或iii)或iv)中任一細胞之衍生物或子代,最佳地,該細胞係以登錄編號DSM ACC3129(H4-II-E)寄存於DSMZ(德國微生物菌種保存中心)之細胞或以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之細胞。The method of claim 24, wherein the rat hepatoma cell line H4-II-E cells, preferably, the cell line: i) is derived from a cell selected from the group consisting of cells: European cell culture preservation Center (ECACC, Cat. No. 87031301), American Type Culture Collection (ATCC, Accession Number CRL-1548), H4-II-E-C3 Cell Line (CRL-1600 or HPACC No. 85061112 or ECACC Catalog No. 85061112), H4II Cell line (HPACC Nr. 89042702), H4-TG cell line (CRL-1578), H5 cell line (HPACC, Nr. 94101905) and H4-S cell line (HPACC Nr. 89102001), or ii) numbered ECACC catalog No. 87031301 is deposited in the European Cell Culture Preservation Center or in cells deposited with the American Type Culture Collection Center ATCC under the accession number CRL-1548, or iii) deposited in DSMZ under the accession number DSM ACC3129 (H4-II-E) Cells of the Culture Collection Center, or iv) cells deposited with DSMZ (HZII-E-S) in DSMZ (German Microbial Culture Preservation Center), or v) i) or ii) or iii) or iv a derivative or progeny of any of the cells, optimally, the cell line is numbered DSM ACC312 9 (H4-II-E) cells deposited in DSMZ (German Microbial Culture Preservation Center) or cells deposited in DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es). 如請求項24或25之方法,其進一步包含h) 用編碼所關注糖蛋白之核酸序列轉染該如請求項24之步驟g)之所獲得宿主細胞,及i) 視情況在允許表現該所關注糖蛋白之條件下培育該步驟h)之轉染細胞,其中該所關注糖蛋白較佳為抗體或Fc融合蛋白、最佳具有ADCC及/或CDC及/或血清穩定性之抗體或Fc融合蛋白。The method of claim 24 or 25, further comprising h) transfecting the host cell obtained according to step g) of claim 24 with a nucleic acid sequence encoding a glycoprotein of interest, and i) permitting expression of the The transfected cells of step h) are incubated under the condition of glycoprotein, wherein the glycoprotein of interest is preferably an antibody or Fc fusion protein, an antibody or Fc fusion optimally having ADCC and/or CDC and/or serum stability. protein. 一種根據如請求項24至26中任一項之方法生成之細胞。A cell produced according to the method of any one of claims 24 to 26. 一種以登錄編號DSM ACC3130(H4-II-Es)寄存於DSMZ(德國微生物菌種保存中心)之(大鼠肝瘤)細胞。A (rat hepatoma) cell deposited with DSMZ (German Microbial Culture Preservation Center) under the accession number DSM ACC3130 (H4-II-Es). 一種如請求項1至16、27及28中任一項之大鼠肝瘤細胞之用途,其作為宿主細胞用於生物醫藥生產。Use of a rat hepatoma cell according to any one of claims 1 to 16, 27 and 28 as a host cell for biomedical production.
TW100134655A 2010-09-27 2011-09-26 Production of low fucose antibodies in H4-II-E rat cells TW201305341A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10018032 2010-09-27
EP11015684 2011-03-03

Publications (1)

Publication Number Publication Date
TW201305341A true TW201305341A (en) 2013-02-01

Family

ID=48169042

Family Applications (1)

Application Number Title Priority Date Filing Date
TW100134655A TW201305341A (en) 2010-09-27 2011-09-26 Production of low fucose antibodies in H4-II-E rat cells

Country Status (1)

Country Link
TW (1) TW201305341A (en)

Similar Documents

Publication Publication Date Title
US20120258496A1 (en) Production of low fucose antibodies in h4-ii-e rat cells
KR101275452B1 (en) Glycosylated antibodies
ES2542885T3 (en) Fusion constructs and use thereof to produce antibodies with greater affinity for Fc receptor binding and effector function
US8025879B2 (en) Modified glycoproteins and uses thereof
KR101667981B1 (en) Methods and vectors for generating asialylated immunoglobulins
CA2773240C (en) Process for producing molecules containing specialized glycan structures
KR101913440B1 (en) Modified antibody composition
KR102182885B1 (en) Compositions and methods for producing glycoproteins
TW201202266A (en) Method for preparing antibodies having improved properties
EP2409989A1 (en) Method to improve glycosylation profile for antibody
ES2349777T3 (en) FUSION CONSTRUCTION AND USE OF THE SAME TO PRODUCE ANTIBODIES WITH GREATER AFFINITY OF UNION TO THE FC RECEIVER AND EFFECTIVE FUNCTION.
TW201305341A (en) Production of low fucose antibodies in H4-II-E rat cells