TW202010844A - Manufacture method of afucosylated antibodies - Google Patents

Manufacture method of afucosylated antibodies Download PDF

Info

Publication number
TW202010844A
TW202010844A TW107130630A TW107130630A TW202010844A TW 202010844 A TW202010844 A TW 202010844A TW 107130630 A TW107130630 A TW 107130630A TW 107130630 A TW107130630 A TW 107130630A TW 202010844 A TW202010844 A TW 202010844A
Authority
TW
Taiwan
Prior art keywords
antibody
cells
defucosylated
protein
cell
Prior art date
Application number
TW107130630A
Other languages
Chinese (zh)
Other versions
TWI748124B (en
Inventor
彭文君
陳惠蓉
Original Assignee
聯合生物製藥股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 聯合生物製藥股份有限公司 filed Critical 聯合生物製藥股份有限公司
Priority to TW107130630A priority Critical patent/TWI748124B/en
Publication of TW202010844A publication Critical patent/TW202010844A/en
Application granted granted Critical
Publication of TWI748124B publication Critical patent/TWI748124B/en

Links

Images

Abstract

The present disclosure is directed to methods for producing an afucosylated antibody, the afucosylated antibodies and composition thereof, and cells for producing antibodies. The method comprises introducing a nucleic acid encoding at least one modified enzyme of the fucosylation pathway to a host cell to produce the afucosylated antibody in the host cell. The disclosure method can easily, stably, and cost-effectively produce afucosylated antibodies. In addition, the afucosylated antibodies produced by the disclosed methods have increased ADCC activity and would not suppress their CDC and safety.

Description

去岩藻醣基化抗體的製造方法 Method for manufacturing defucosylated antibody

本發明係有關於一種去岩藻醣基化蛋白,包括有增強的活性及治療特性之去岩藻醣基化免疫功能分子,以及製備去岩藻醣基化蛋白的方法。 The present invention relates to a defucosylated protein, including defucosylated immune function molecules with enhanced activity and therapeutic properties, and a method for preparing the defucosylated protein.

醣蛋白涉及人類的許多基本功能,包括催化、信號傳導、細胞間的訊息傳遞以及分子辨識別與結合。許多醣蛋白已被用於治療的目的,且在過去的二十年中,天然存在的分泌醣蛋白重組形式一直是生技產業的主要產物。範例包括促紅血球生成素(erythropoietin,EPO)、治療性單株抗體(therapeutic monoclonal antibody,治療性mAb)、組織纖維溶酶原活化劑(tissue plasminogen activator,tPA)、干擾素-β(interferon-β,IFN-β)、顆粒細胞-噬細胞集落刺激因子(granulocyte-macrophage colony stimulating factor,GM-CSF)和人類絨毛膜促性腺激素(human chorionic gonadotrophin,hCG)。 Glycoproteins are involved in many basic human functions, including catalysis, signal transduction, cell-to-cell message transmission, and molecular recognition and binding. Many glycoproteins have been used for therapeutic purposes, and in the past two decades, naturally occurring recombinant forms of secreted glycoproteins have been the main products of the biotechnology industry. Examples include erythropoietin (EPO), therapeutic monoclonal antibody (therapeutic mAb), tissue plasminogen activator (tPA), and interferon-β (interferon-β , IFN-β), granulocyte-macrophage colony stimulating factor (GM-CSF) and human chorionic gonadotrophin (hCG).

哺乳動物中存在5類抗體,即IgM、IgD、IgG、IgA及IgE。人類IgG抗體由於其血液中半衰期長與功能特性,如各種效應子功能等,主要用於各種人類疾病的診斷、預防和治療。人類IgG抗體更可分成以下4種亞類:IgG1、IgG2、IgG3及IgG4。已有許多針對作為IgG類抗體的效應子功 能之抗體依賴性細胞毒性(antibody-dependent cellular cytotoxicity,ADCC)活性與補體依賴性細胞毒性(complement-dependent cytotoxicity,CDC)的研究,且已報導IgG1亞類的抗體在人類IgG抗體中具有最大的ADCC活性和CDC活性。 There are five types of antibodies in mammals, namely IgM, IgD, IgG, IgA and IgE. Human IgG antibodies are mainly used for the diagnosis, prevention and treatment of various human diseases due to their long half-life in blood and functional characteristics, such as various effector functions. Human IgG antibodies can be further divided into the following four subclasses: IgG1, IgG2, IgG3 and IgG4. There have been many studies on antibody-dependent cellular cytotoxicity (ADCC) activity and complement-dependent cytotoxicity (CDC) as effector functions of IgG antibodies, and IgG1 Class antibodies have the largest ADCC activity and CDC activity among human IgG antibodies.

人類IgG1亞類抗體的ADCC活性和CDC活性的表現需要抗體Fc區與效應細胞,如殺手細胞、自然殺手細胞、活化的巨噬細胞或其類似細胞及各種補體的表面上存在的抗體受體(以下稱為「FcγR」)結合。已顯示抗體鉸鏈區及C區(以下稱為「Cγ2結構域(domain)」)中第2結構域之許多胺基酸殘基,以及連結至Cγ2區域的醣鏈對於此結合反應是重要的。 The performance of ADCC activity and CDC activity of human IgG1 subclass antibodies requires antibody Fc region and effector cells, such as killer cells, natural killer cells, activated macrophages or similar cells, and antibody receptors present on the surface of various complements ( Hereinafter referred to as "FcγR") binding. It has been shown that many amino acid residues of the second domain in the hinge region and the C region of the antibody (hereinafter referred to as "Cγ2 domain"), and the sugar chain linked to the Cγ2 region are important for this binding reaction.

降低或抑制抗體或Fc融合蛋白的N-多醣的岩藻醣基化可增強ADCC活性。ADCC一般參與自然殺手(natural killer,NK)細胞的活化,且依賴抗體包覆細胞經NK細胞表面上Fc受體的辨識。Fc區域與NK細胞上Fc受體的結合會受Fc區域醣基化狀態的影響。此外,在Fc區域的N-多醣類型也會影響ADCC活性。因此,對於抗體組成物或Fc融合蛋白組成物,可藉由增加去岩藻醣基之N-多醣的相對量來增加對FcγRIII結合親和力,或組成物的ADCC活性。 Decreasing or inhibiting the fucosylation of N-polysaccharides of antibodies or Fc fusion proteins can enhance ADCC activity. ADCC is generally involved in the activation of natural killer (NK) cells, and relies on antibody-coated cells to recognize Fc receptors on the surface of NK cells. The binding of the Fc region to Fc receptors on NK cells is affected by the glycosylation state of the Fc region. In addition, the type of N-polysaccharide in the Fc region can also affect ADCC activity. Therefore, for an antibody composition or an Fc fusion protein composition, the binding affinity for FcγRIII or the ADCC activity of the composition can be increased by increasing the relative amount of defucosylated N-polysaccharide.

許多可影響醣化的因子,包含物種、組織及細胞類型,均已顯示對於醣化的發生方式非常重要。此外,細胞外環境,通過改變培養條件,如血清濃度,可直接對醣基化產生影響。已提出各種方法,來改變在特定宿主生物體中達到的醣基化模式,包括導入或過度表現與產生寡醣有關之某些酵素(U.S.Pat.No.5,047,335;U.S.Pat.No.5,510,261)。 Many factors that can affect glycation, including species, tissues and cell types, have been shown to be very important for the way glycation occurs. In addition, the extracellular environment can directly affect glycosylation by changing culture conditions, such as serum concentration. Various methods have been proposed to change the glycosylation pattern achieved in a particular host organism, including the introduction or excessive expression of certain enzymes related to the production of oligosaccharides (U.S. Pat. No. 5,047,335; U.S. Pat. No. 5,510,261).

WO98/58964描述抗體組成物,其中大致上所有的N端-連接 的寡醣皆為G2寡醣。G2係指具有兩個終端Gal,且沒有NeuAcs的分歧(biantennary)結構。WO99/22764提到大致上沒有在CH2結構域中具有N端-連接之G1、G0或G-1寡醣的醣蛋白的抗體組成物,其。G1係指具有一個Gal,且不具有NeuAcs的分歧結構,G0係指其中不具有末端NeuAcs或Gals的分歧結構,且G-1係指核心單元少一個GlcNAc。 WO98/58964 describes antibody compositions in which substantially all N-terminally-linked oligosaccharides are G2 oligosaccharides. G2 refers to a structure with two terminals Gal and no NeuAcs (biantennary). WO99/22764 mentions antibody compositions that are substantially free of glycoproteins with N-terminally-linked G1, G0 or G-1 oligosaccharides in the CH2 domain. G1 refers to a bifurcated structure with one Gal and no NeuAcs, G0 refers to a bifurcated structure without terminal NeuAcs or Gals, and G-1 refers to one GlcNAc less in the core unit.

WO00/61739報導的是,與73%的由NSO(小鼠骨髓瘤)細胞表現的抗體相比,47%的由YB2/0(大鼠骨髓瘤)細胞表現的抗-hIL-5R抗體具有α1-6岩藻醣連接的醣鏈。由不同宿主細胞表現的αhIL-5R抗體岩藻醣抗體的岩藻醣相對比例為YB2/0<CHO/d<NSO。 WO00/61739 reports that 47% of anti-hIL-5R antibodies expressed by YB2/0 (rat myeloma) cells have α1 compared to 73% of antibodies expressed by NSO (mouse myeloma) cells -6 fucose linked sugar chains. The relative ratio of fucose of αhIL-5R antibody fucose antibody expressed by different host cells is YB2/0<CHO/d<NSO.

WO02/31140與WO03/85118顯示可藉由利用RNAi抑制α1,6-岩藻醣基轉移酶的功能,來控制與醣鏈結合之岩藻醣的修飾。使用細胞生產抗體組成物的方法,其包括使用抗凝集素的細胞,凝集素辨識醣鏈,其中岩藻醣的第1位置透過複合的N-醣苷連接的醣鏈中的α-鍵,結合至還原端中N-乙醯葡萄醣胺的第6位置。 WO02/31140 and WO03/85118 show that the modification of fucose bound to sugar chains can be controlled by using RNAi to inhibit the function of α1,6-fucosyltransferase. A method for producing an antibody composition using a cell, which includes a cell using an anti-lectin, the lectin identifying a sugar chain, wherein the first position of fucose is bound through an α-bond in a sugar chain linked to a complex N-glycoside To the 6th position of N-acetylglucosamine in the reducing end.

醣鏈的結構在人類IgG1亞類抗體的效應功能上扮演非常重要的角色,且藉由改變醣鏈結構,來製備具有更強效應功能的抗體可以是可能的。然而,醣鏈的結構多樣且複雜,且解決醣鏈的生理角色仍不充分,且昂貴。因此,需要一種生產去岩藻醣基化抗體的方法。 The structure of the sugar chain plays a very important role in the effect function of human IgG1 subclass antibodies, and by changing the structure of the sugar chain, it may be possible to prepare antibodies with stronger effect functions. However, the structure of sugar chains is diverse and complex, and the physiological role of sugar chains is still insufficient and expensive. Therefore, a method for producing defucosylated antibodies is needed.

參考文獻 references

1. PAULSON, James, et al., “Process for controlling intracellular glycosylation of proteins” US Patent No. 5,047,335 (1991) 1. PAULSON, James, et al., "Process for controlling intracellular glycosylation of proteins" US Patent No. 5,047,335 (1991)

2. GOOCHEE, Charles F., et al., “Method of controlling the degradation of glycoprotein oligosaccharides produced by cultured Chinese hamster ovary cells” US Patent No. 5,510,261 (1996) 2. GOOCHEE, Charles F., et al., “Method of controlling the degradation of glycoprotein oligosaccharides produced by cultured Chinese hamster ovary cells” US Patent No. 5,510,261 (1996)

3. WONG, Chi-Huey, et al., “Method and composition for synthesizing sialylated glycosyl compounds” US Patent No. 5,278,299 (1994) 3. WONG, Chi-Huey, et al., "Method and composition for synthesizing sialylated glycosyl compounds" US Patent No. 5,278,299 (1994)

4. RAJU, T., Shantha, “Methods and compositions for galactosylated glycoproteins” WO/1998/058964 (1998) 4. RAJU, T., Shantha, "Methods and compositions for galactosylated glycoproteins" WO/1998/058964 (1998)

5. RAJU, T., Shantha, “Methods and compositions comprising galactosylated glycoproteins” WO/1999/022764 (1999) 5. RAJU, T., Shantha, "Methods and compositions comprising galactosylated glycoproteins" WO/1999/022764 (1999)

6. HANAI, Nobuo, et al., “Method for controlling the activity of immunologically functional molecule” WO/2000/061739 (2000) 6. HANAI, Nobuo, et al., "Method for controlling the activity of immunologically functional molecule" WO/2000/061739 (2000)

7. KANDA, Yutaka, et al., “cells producing antibody compositions” WO/2002/031140 (2002) 7. KANDA, Yutaka, et al., “cells producing antibody compositions” WO/2002/031140 (2002)

8. BLUMBERG, R. S., et al., “Central airway administration for systemic delivery of therapeutics” WO 03/077834 (2002) 8. BLUMBERG, R. S., et al., "Central airway administration for systemic delivery of therapeutics" WO 03/077834 (2002)

9. BLUMBERG, R. S., et al., “Central airway administration for systemic delivery of therapeutics” US Patent Application Publication 2003-0235536 (2003) 9. BLUMBERG, R. S., et al., "Central airway administration for systemic delivery of therapeutics" US Patent Application Publication 2003-0235536 (2003)

10. MÖSSENER, E., et al., “Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity” Blood 115, 4393-4402 (2010) 10. MÖSSENER, E., et al., “Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity” Blood 115, 4393- 4402 (2010)

11. FERRARA, C., et al., “Unique carbohydrate-carbohydrate interactions are required for high affinity binding between FcγRIII and antibodies lacking core fucose” Proc Natl Acad Sci U.S.A. 108, 12669 - 12674 (2011) 11. FERRARA, C., et al., “Unique carbohydrate-carbohydrate interactions are required for high affinity binding between FcγRIII and antibodies lacking core fucose” Proc Natl Acad Sci USA 108, 12669-12674 (2011)

本發明係有關於產生具加強活性之去岩藻醣化蛋白,包括去岩藻醣化抗體的新穎方法。本發明也有關於以本發明方法所產生之去岩藻醣基化蛋白,以及產生去岩藻醣基化蛋白的細胞。本發明的去岩藻醣基化抗體,相較於天然存在的岩藻醣基化抗體,具有增加之抗體依賴性細胞毒性(ADCC)活性。 The present invention relates to a novel method for producing defucosylated proteins with enhanced activity, including defucosylated antibodies. The present invention also relates to afucosylated protein produced by the method of the present invention, and cells producing afucosylated protein. The defucosylated antibodies of the present invention have increased antibody-dependent cytotoxicity (ADCC) activity compared to naturally occurring fucosylated antibodies.

本發明的一範疇係有關於一種在宿主細胞產生去岩藻醣基化蛋白,包括去岩藻醣基化抗體的方法。本發明之方法一般包括將編碼岩藻醣基化途徑中經修飾之酵素的核酸導入至宿主細胞中,以抑制抗體在此宿主細胞中的岩藻醣基化。經修飾的酵素可源自於岩藻醣基化途徑中的酵素。在某些實施例中,經修飾的酵素可源自於GDP-甘露醣4,6-脫水酶(GDP-mannonse 4,6-dehydratase,GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(GDP-4-keto-6-deoxy-D-mannose epinierase-reductase,FX)及/或岩藻醣轉移酶(FUT1至FUT12、POFUT1及POFUT2)。在一些實施例中,經修飾的酵素可源自於GMD或FUT。在特定實施例中,經修飾的酵素可源自於α-1,6-岩藻醣轉移酶(FUT8)。經修飾的酵素可抑制宿主細胞在岩藻醣化途徑中天然存在的酵素的功能,進而抑制宿主細胞中抗體的岩藻醣化。 One category of the invention relates to a method for producing defucosylated proteins, including defucosylated antibodies, in host cells. The method of the present invention generally includes introducing a nucleic acid encoding a modified enzyme in the fucosylation pathway into a host cell to inhibit antibody fucosylation in the host cell. The modified enzyme may be derived from enzymes in the fucosylation pathway. In some embodiments, the modified enzyme may be derived from GDP-mannonse 4,6-dehydratase (GDP-mannonse 4,6-dehydratase, GMD), GDP-4-keto-6-deoxy-D -Mannose isomerase-reductase (GDP-4-keto-6-deoxy-D-mannose epinierase-reductase, FX) and/or fucose transferase (FUT1 to FUT12, POFUT1 and POFUT2). In some embodiments, the modified enzyme may be derived from GMD or FUT. In certain embodiments, the modified enzyme may be derived from α-1,6-fucose transferase (FUT8). The modified enzyme can inhibit the function of the host cell's naturally occurring enzyme in the fucosylation pathway, thereby inhibiting the fucosylation of antibodies in the host cell.

在一些實施例中,生產去岩藻醣基化蛋白(包括去岩藻醣基化抗體)的方法,包括(a)提供宿主細胞,(b)將編碼岩藻醣基化途徑之經修飾的酵素的核酸導入至此宿主細胞中,以及(c)於此宿主細胞中產生去岩藻醣基化蛋白。 In some embodiments, a method of producing defucosylated proteins (including defucosylated antibodies) includes (a) providing a host cell and (b) modifying a modified fucosylation pathway The nucleic acid of the enzyme is introduced into the host cell, and (c) the defucosylated protein is produced in the host cell.

本發明另一範疇係有關於以本發明方法所產生的去岩藻醣基化蛋白,包括去岩藻醣基化抗體。相較於天然存在的岩藻醣基化抗體,去岩藻醣化抗體具有增加及改善的活性。在一些實施例中,抗體具有增加及改善的ADCC。 Another category of the invention relates to defucosylated proteins produced by the method of the present invention, including defucosylated antibodies. Compared to naturally occurring fucosylated antibodies, defucosylated antibodies have increased and improved activity. In some embodiments, the antibody has increased and improved ADCC.

本發明另有關於生產去岩藻醣基化蛋白,包括去岩藻醣基化抗體的細胞。 The invention also relates to cells for producing defucosylated proteins, including defucosylated antibodies.

於以下實施例提供本發明之實施方式,並搭配所附圖式。 The following examples provide the embodiments of the present invention, together with the accompanying drawings.

第1圖為於RC79細胞(表現RITUXAN®之穩定細胞株)及表現F83M、F8M1、F8M2、F8M3或F8D1突變蛋白之RC79重組細胞中所生產之FUT8蛋白的西方墨點法的圖譜。甘油醛-3-磷酸脫氫酶(glyceraldehyde-3-phosphate dehydrogenase,GAPDH)的表現作為蛋白上樣控制組(loading control)。在表現突變FUT8酵素之RC79重組細胞中,FUT8蛋白的表現量類似於,或相同於親源RC79細胞中FUT8蛋白的表現量。 1 is a view of cells in RC79 (RITUXAN® performance of stable cell lines) and performance F83M, F8M1, F8M2, F8M3 F8D1 mutant cell RC79 recombinant proteins produced in the protein profiles of FUT8 or a western blot. The performance of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) serves as a protein loading control group. In RC79 recombinant cells expressing mutant FUT8 enzyme, the expression level of FUT8 protein is similar to, or the same as that of parental RC79 cells.

第2圖為表現F83M突變蛋白之RC79重組細胞和RC79親源細胞的流式細胞分析。長虛線之波峰代表經羅丹明-LCA染色之表現F83M的RC79重組細胞。灰色填滿之波峰代表未經羅丹明-LCA染色之表現F83M的RC79重組細胞(陰性對照組)。短虛線之波峰代表經羅丹明-LCA染色的RC79細胞(未表現F83M的親源細胞)(陽性對照組)。 Flow cytometric analysis showed F83M mutant RC79 RC79 recombinant cell proteins and the parental cell is the second FIG. The long dashed peak represents the RC79 recombinant cells expressing F83M stained with rhodamine-LCA. The gray filled peaks represent RC79 recombinant cells expressing F83M without Rhodamine-LCA staining (negative control group). The peak of the short dashed line represents RC79 cells stained with Rhodamine-LCA (parent cells not expressing F83M) (positive control group).

第3a及3b圖為顯示ADCC

Figure 107130630-A0202-12-0006-69
的圖。第3a及3b圖分別顯示經由來自捐贈者1(第3a圖)及捐贈者2(第3b圖)之PBMC細胞的RITUXAN®與去岩藻醣基化抗-CD20單株抗體的ADCC活性。去岩藻醣基化抗-CD20單株抗體 (R1細胞株)的ADCC活性顯著高於RITUXAN®。 Of FIG. 3a and 3b show ADCC is
Figure 107130630-A0202-12-0006-69
Figure. Figures 3a and 3b show the ADCC activity of RITUXAN® and defucosylated anti-CD20 monoclonal antibodies via PBMC cells from donor 1 ( Figure 3a ) and donor 2 ( Figure 3b ), respectively. The ADCC activity of defucosylated anti-CD20 monoclonal antibody (R1 cell line) was significantly higher than that of RITUXAN®.

第4a至4c圖為顯示用SPR生物感應器(BIACORETM X100)的FcγRIIIa親和力分析的SPR感應圖的圖。將His-標籤FcγRIIIa(1μg/mL)及5-80nM去岩藻醣基化抗-CD20單株抗體(第4a圖)、20-320nM RITUXAN®(第4b圖)或5-80nM GAZYVA®(第4c圖),以30μL/分鐘的流速,依序流過抗-His抗體固定的CM5晶片。相較於RITUXAN®和GAZYVA®,去岩藻醣基化抗-CD20單株抗體(R1細胞株)對FcγRIIIa具有更強的親和力。 4a to 4c is a view of FIG SPR sensorgrams FcγRIIIa affinity analysis by SPR biosensor (BIACORE TM X100) display. Combine His-tag FcγRIIIa (1 μg/mL) and 5-80nM defucosylated anti-CD20 monoclonal antibody ( Figure 4a ), 20-320nM RITUXAN® ( Figure 4b ) or 5-80nM GAZYVA® ( section 4b ) Fig. 4c ), flow through the CM5 wafer immobilized with anti-His antibody in sequence at a flow rate of 30 μL/min. Compared to RITUXAN® and GAZYVA®, the defucosylated anti-CD20 monoclonal antibody (R1 cell line) has a stronger affinity for FcγRIIIa.

第5圖為顯示RITUXAN®與去岩藻醣基化抗-CD20單株抗體的CDC活性的圖。去岩藻醣基化抗-CD20單株抗體(R1細胞株)的CDC活性與RITUXAN®的相當。 FIG 5 is a graph showing anti -CD20 CDC activity monoclonal antibodies with defucosylated RITUXAN® glycosylation. The CDC activity of the defucosylated anti-CD20 monoclonal antibody (R1 cell line) is comparable to that of RITUXAN®.

第6圖為顯示以食鹽水(載體)、RITUXAN®或去岩藻醣基化抗-CD20單株抗體(R1細胞株)治療之老鼠的腫瘤體積的圖。數據點表示腫瘤體積的平均值±SD(每組n=5)。去岩藻醣基化抗-CD20單株抗體(細胞株R1)的抗-腫瘤功效顯著高於RITUXAN®。 FIG. 6 is displayed in saline (vehicle), RITUXAN® or de-fucosylated anti-tumor volumes -CD20 monoclonal antibody (R1 cell line) mice Healing of FIG. Data points represent the mean ± SD of tumor volume (n=5 per group). The anti-tumor efficacy of defucosylated anti-CD20 monoclonal antibody (cell line R1) is significantly higher than that of RITUXAN®.

第7圖為顯示自以食鹽水(載體)、RITUXAN®或去岩藻醣基化抗-CD20單株抗體(R1細胞株)治療之老鼠收集的腫瘤的重量的圖。以去岩藻醣基化抗-CD20單株抗體(R1細胞株)治療的老鼠的腫瘤重量顯著比RITUXAN®和載體組輕。 7 is a view of brine from to (carrier), RITUXAN® or de-fucosylated anti-tumor -CD20 mouse monoclonal antibody (R1 cell line) treating the collected weight of the FIG. The tumor weight of mice treated with defucosylated anti-CD20 monoclonal antibody (R1 cell line) was significantly lighter than that of RITUXAN® and vehicle group.

第8圖為顯示以食鹽水(載體)、RITUXAN®或去岩藻醣基化抗-CD20單株抗體(R1細胞株)治療之老鼠體重的圖。圖中的數據點表示腫瘤體積的平均值±SD(每組n=5)。 8 is shown in FIG saline (vehicle), RITUXAN® or de-fucosylation FIG -CD20 mouse monoclonal antibody anti-weight (R1 cell lines) of the treatment. The data points in the graph represent the mean ± SD of tumor volume (n=5 per group).

本發明係有關於生產具改善活性之去岩藻醣基化抗體的新穎方法。本發明亦有關於以此發明方法所產生的抗體以及產生此去岩藻醣基化抗體的細胞。相較於天然存在的岩藻醣基化抗體,本發明之去岩藻醣基化抗體具有增加的抗體依賴性細胞毒性(ADCC)活性。 The present invention relates to a novel method for producing defucosylated antibodies with improved activity. The invention also relates to the antibody produced by the method of the invention and the cell producing the defucosylated antibody. Compared to naturally-occurring fucosylated antibodies, the defucosylated antibodies of the present invention have increased antibody-dependent cytotoxicity (ADCC) activity.

以下為提供的實施方式,以協助本發明所屬技術領域中具有通常知識者實施本發明。本發明所屬技術領域中具有通常知識者可理解本發明說明明確描述的實施例的修改或變化並不會偏離本發明所含有的資訊的精神或範圍,仍屬於本發明之範疇。於描述中所使用的術語僅用於描述特定實施例,並不限制本發明。以下所使用的標題僅為組織各章節的目的,不應被視為限制所描述的主題。 The following are the provided embodiments to assist those with ordinary knowledge in the technical field to which the present invention belongs to implement the present invention. Those with ordinary knowledge in the technical field to which the present invention pertains can understand that modifications or changes to the embodiments explicitly described in the present description do not deviate from the spirit or scope of the information contained in the present invention, and still belong to the scope of the present invention. The terminology used in the description is only for describing specific embodiments, and does not limit the present invention. The headings used below are for the purpose of organizing the chapters only and should not be considered as limiting the subject matter described.

本發明說明中所提及的所有文獻、申請專利、專利、圖式及其他參考文獻整體(包括部分)可完全併入,如在發明說明中所公開和敘述。 All documents, patent applications, patents, drawings and other references (including parts) mentioned in the description of the present invention can be fully incorporated, as disclosed and described in the description of the invention.

除另外說明,本發明說明所使用的所有技術和科學術語與本發明所屬技術領域中具有通常知識者通常理解具有相同的意義。除非上下文另有說明,發明說明中所述單數「一」和「該」包括複數的意思。類似地,除非上下文另有說明,詞語「或」意圖包括「及」的意思。因此,「包括A或B」係指包括A或B、或A及B。更應了解的是,多胜肽的所有胺基酸大小,所有分子量或分子質量值為近似值,且僅供描述。儘管可使用與本發明說明所述類似或相同的方法和材料,但適合的方法及材料如下所述。本發明說明中所述所有文獻、專利說明書、專利及其它文獻皆可併入本發明中。如果發生衝突,會受到本發說明(包括術語解釋)限制。此外,材料、方法和實施例僅是說明性質,不意圖限制本發明。 Unless otherwise stated, all technical and scientific terms used in the description of the present invention have the same meaning as those generally understood in the technical field to which the present invention belongs. Unless the context indicates otherwise, the singular "a" and "the" in the description of the invention include the plural. Similarly, unless the context indicates otherwise, the word "or" is intended to include the meaning of "and". Therefore, "including A or B" means including A or B, or A and B. It should be further understood that all amino acid sizes, all molecular weights or molecular mass values of Polypeptide are approximate values and are for description only. Although methods and materials similar or identical to those described in the present invention can be used, suitable methods and materials are described below. All documents, patent specifications, patents and other documents mentioned in the description of the present invention can be incorporated into the present invention. In case of conflict, it will be restricted by the instructions (including term explanation). In addition, the materials, methods, and examples are illustrative only and are not intended to limit the invention.

1.細胞中岩醣基化的抑制1. Inhibition of rock glycosylation

本發明之一範疇係有關於抑制或減少細胞中岩藻醣基化的方法。 One category of the invention relates to methods for inhibiting or reducing fucosylation in cells.

a.宿主細胞a. Host cell

任何適當的宿主細胞可用於生產去岩醣基化抗體,包括源自於酵母菌、昆蟲、兩棲類、魚類、爬蟲動物、鳥類、哺乳動物或人類,或融合瘤的宿主細胞。宿主細胞可以是未經修飾的細胞或細胞株,或經遺傳修飾的細胞株(例如,以促進生物產品的生產)。在一些實施例中,宿主細胞為細胞株,其已經修飾,以允許在期望的條件下,例如在無血清培養基中、在細胞懸浮式培養中或在貼壁式細胞培養中生長。 Any suitable host cell can be used to produce defucosylated antibodies, including those derived from yeast, insects, amphibians, fish, reptiles, birds, mammals or humans, or fusion tumors. The host cell may be an unmodified cell or cell strain, or a genetically modified cell strain (eg, to promote the production of biological products). In some embodiments, the host cell is a cell line that has been modified to allow growth under desired conditions, such as in serum-free medium, in cell suspension culture, or in adherent cell culture.

使用哺乳動物宿主細胞有助於給予至人類的抗體。在一些實施例中,宿主細胞為中國倉鼠卵巢(Chinese hamster ovary,CHO)細胞,其為用於表現許多重組蛋白的細胞株。一般用於表現重組蛋白的額外哺乳動物細胞株包括293HEK細胞、HeLa細胞、COS細胞、NIH/3T3細胞、Jurkat細胞、NSO細胞及HUVEC細胞。在另一些實施例中,宿主細胞為表現抗體的重組細胞。 The use of mammalian host cells facilitates the administration of antibodies to humans. In some embodiments, the host cell is a Chinese hamster ovary (CHO) cell, which is a cell line used to express many recombinant proteins. Additional mammalian cell lines commonly used to express recombinant proteins include 293HEK cells, HeLa cells, COS cells, NIH/3T3 cells, Jurkat cells, NSO cells, and HUVEC cells. In other embodiments, the host cell is a recombinant cell expressing antibodies.

於本發明說明提供的方法中有用的人類細胞株的範例包括293T(胚胎腎)、786-0(腎)、A498(腎)、A549(肺泡上皮)、ACHN(腎)、BT-549(乳房)、BxPC-3(胰)、CAKI-1(腎)、Capan-i(胰)、CCRF-CEM(白血病)、COLO 205(結腸)、DLD-1(結腸)、DMS 114(小細胞肺)、DU145(前列腺)、EKVX(非小細胞肺)、HCC-2998(結腸)、HCT-15(結腸)、HCT-1 16(結腸)、HT29(結腸)、S IT-1080(纖維肉瘤)、HEK 293(胚胎腎)、HeLa(子宮頸癌)、 HepG2(肝細胞癌)、HL-60(TB)(白血病)、HOP-62(非小細胞肺)、HOP-92(非小細胞肺)、HS 578T(乳房)、HT-29(結腸腺癌)、IG-OV1(卵巢)、IMR32(神經母細胞瘤)、Jurkat(T淋巴細胞)、K-562(白血病)、KM 12(結腸)、KM20L2(結腸)、LANS(神經母細胞瘤)、LNCap.FGC(高加索前列腺腺癌)、LOX IMV1(黑色素瘤)、LXFL 529(非小細胞肺)、M 14(黑色素瘤)、M19-MEL(黑色素瘤)、MALME-3M(黑色素瘤)、MCFIOA(乳房上皮)、MCI '7(乳腺)、MDA-MB-453(乳房上皮)、MDA-MB-468(乳房)、MDA-MB-231(乳房)、MDA-N(乳房)、MOLT-4(白血病)、NCl/ADR-RES(卵巢)、NCI-1122.0(非小細胞肺)、NCI-H23(非小細胞肺)、NCl-H322M(非小細胞肺)、NCI-H460(非小細胞肺)、NCI-H522(非小細胞肺)、OVCAR-3(卵巢)、QVCAR-4(卵巢)、OVCAR-5(卵巢)、OVCAR-8(卵巢)、P388(白血病)、P388/ADR(白血病)、PC-3(前列腺)、PERC6®(El-變形胚胎視網膜)、RPMI-7951(黑色素瘤)、RPMI-8226(白血病)、RXF 393(腎)、RXF-631(腎)、Saos-2(骨)、SF-268(中央神經系統)、SF-295(中央神經系統)、SF-539(中央神經系統)、SHP-77(小細胞肺)、SH-SY5Y(神經母細胞瘤)、SK-BR3(乳房)、SK-MEL-2(黑色素瘤)、SK-MEL-5(黑色素瘤)、SK-MEL-28(黑色素瘤)、SK-OV-3(卵巢)、SN12K1(腎)、SN12C(腎)、SNB-19(中央神經系統)、SNB-75(中央神經系統)、SNB-78(中央神經系統)、SR(白血病)、SW-620(結腸)、T-47D(乳房)、THP-1(單核細胞之巨噬細胞)、TK-10(腎)、U87(膠質母細胞瘤)、U293(腎)、U251(中央神經系統)、UACC-257(黑色素瘤)、UACC-62(黑色素瘤)、UO-31(腎)、W138(肺)及XF 498(中央神經系統)之細胞株。 Examples of human cell lines useful in the methods provided by the present description include 293T (embryonic kidney), 786-0 (kidney), A498 (kidney), A549 (alveolar epithelium), ACHN (kidney), BT-549 (breast ), BxPC-3 (pancreas), CAKI-1 (kidney), Capan-i (pancreas), CCRF-CEM (leukemia), COLO 205 (colon), DLD-1 (colon), DMS 114 (small cell lung) , DU145 (prostate), EKVX (non-small cell lung), HCC-2998 (colon), HCT-15 (colon), HCT-1 16 (colon), HT29 (colon), S IT-1080 (fibrosarcoma), HEK 293 (embryonic kidney), HeLa (cervical cancer), HepG2 (hepatocellular carcinoma), HL-60 (TB) (leukemia), HOP-62 (non-small cell lung), HOP-92 (non-small cell lung) , HS 578T (breast), HT-29 (colon adenocarcinoma), IG-OV1 (ovarian), IMR32 (neuroblastoma), Jurkat (T lymphocyte), K-562 (leukemia), KM 12 (colon) , KM20L2 (colon), LANS (neuroblastoma), LNCap.FGC (Caucasian prostate adenocarcinoma), LOX IMV1 (melanoma), LXFL 529 (non-small cell lung), M 14 (melanoma), M19-MEL (Melanoma), MALME-3M (melanoma), MCFIOA (breast epithelium), MCI '7 (breast), MDA-MB-453 (breast epithelium), MDA-MB-468 (breast), MDA-MB-231 (Breast), MDA-N (breast), MOLT-4 (leukemia), NCl/ADR-RES (ovary), NCI-1122.0 (non-small cell lung), NCI-H23 (non-small cell lung), NCl-H322M (Non-small cell lung), NCI-H460 (non-small cell lung), NCI-H522 (non-small cell lung), OVCAR-3 (ovary), QVCAR-4 (ovary), OVCAR-5 (ovarian), OVCAR- 8 (ovary), P388 (leukemia), P388/ADR (leukemia), PC-3 (prostate), PERC6® (El-transformed embryonic retina), RPMI-7951 (melanoma), RPMI-8226 (leukemia), RXF 393 (kidney), RXF-631 (kidney), Saos-2 (bone), SF-268 (central nervous system), SF-295 (central nervous system), SF-539 (central nervous system), SHP-77 ( Small cell lung), SH-SY5Y (neuroblastoma), SK-BR3 (breast), SK-MEL-2 (melanoma), SK-MEL-5 (melanoma), SK -MEL-28 (melanoma), SK-OV-3 (ovarian), SN12K1 (kidney), SN12C (kidney), SNB-19 (central nervous system), SNB-75 (central nervous system), SNB-78 ( Central nervous system), SR (leukemia), SW-620 (colon), T-47D (breast), THP-1 (monocyte macrophages), TK-10 (kidney), U87 (glioblastoma ), U293 (kidney), U251 (central nervous system), UACC-257 (melanoma), UACC-62 (melanoma), UO-31 (kidney), W138 (lung) and XF 498 (central nervous system) Cell line.

於本發明說明提供的方法中有用之非人類靈長類細胞株的 範例包括經SV40(COS-7)轉染之經猴腎(CVI-76)、非洲綠猴腎(VERO-76)、綠猴纖維細胞(COS-1)及猴腎細胞之細胞株。額外的哺乳動物細胞株為本發明所屬技術領域中具有通常知識者已知,且為編錄在美國典型培養物保藏中心(American Type Culture Collection,ATCC)目錄(Manassas,VA)中。 Examples of non-human primate cell lines useful in the methods provided by the description of the present invention include monkey kidney (CVI-76), African green monkey kidney (VERO-76), green transfected with SV40 (COS-7) Cell lines of monkey fiber cells (COS-1) and monkey kidney cells. The additional mammalian cell lines are known to those with ordinary knowledge in the technical field to which the present invention belongs, and are cataloged in the American Type Culture Collection (ATCC) catalog (Manassas, VA).

b.修飾岩藻醣基化途徑中的酵素b. Modify enzymes in the fucosylation pathway

本發明之去岩藻醣基化抗體可在宿主細胞中產生,此宿主細胞中的岩藻醣基化途徑已經以減少或抑制蛋白的岩藻醣基化的方式被改變。 The defucosylated antibody of the present invention can be produced in a host cell, and the fucosylation pathway in this host cell has been altered in a manner that reduces or inhibits the fucosylation of the protein.

i.經修飾的酵素i. Modified enzyme

本發明說明中所使用之詞組「經修飾的酵素」係指衍生自在岩藻醣基化途徑中天然存在或野生型酵素的蛋白,其以在修飾後改變或破壞蛋白質的天然酵素活性的方式被改變。經修飾的酵素能夠抑制或干擾其對應的野生型酵素,以改變、抑制或降低宿主細胞中野生型酵素的活性。 The phrase "modified enzyme" used in the description of the present invention refers to a protein derived from a naturally-occurring or wild-type enzyme in the fucosylation pathway, which is modified or modified to destroy the natural enzyme activity of the protein change. The modified enzyme can inhibit or interfere with its corresponding wild-type enzyme to change, inhibit or reduce the activity of the wild-type enzyme in the host cell.

經修飾的酵素可藉由改變天然存在的酵素來生產,例如,藉由改變總蛋白質電荷、共價連接化學或蛋白質部分(moiety)、導入胺基酸置換、插入及/或刪除,及/或其任何組合。在一些實施例中,相較於天然存在的酵素對應物,經修飾的酵素具有胺基酸的取代、添加、及/或刪除。在一些實施例中,相較於天然存在的酵素對應物,經修飾的酵素有介於1至約20個胺基酸取代、添加、及/或刪除。胺基酸的取代、添加及插入可利用天然或非天然胺基酸來完成。非天然存在之胺基酸包括,但不限於,ε-N賴胺酸、ß-丙胺酸、鳥胺酸、正白胺酸、正纈胺酸、羥脯胺酸、甲狀腺素、γ-胺基丁 酸、高絲胺酸、瓜胺酸、胺基苯甲酸、6-氨基己酸(Aca;6-Aminohexanoic acid)、羥脯胺酸、硫醇丙酸(MPA)、3-硝基-酪胺酸、焦谷胺酸及其類似。天然存在之胺基酸包括丙胺酸、精胺酸、天門冬醯胺、天冬胺酸、半胱胺酸、麩胺酸、麩醯胺酸、甘胺酸、組胺酸、異白胺酸、白胺酸、賴胺酸、甲硫胺酸、苯丙胺酸、脯胺酸、絲胺酸、蘇胺酸、色胺酸、酪胺酸及纈胺酸。 Modified enzymes can be produced by changing naturally occurring enzymes, for example, by changing the total protein charge, covalently linking chemical or protein moieties, introducing amino acid substitutions, insertions and/or deletions, and/or Any combination of them. In some embodiments, the modified enzyme has amino acid substitutions, additions, and/or deletions compared to naturally occurring enzyme counterparts. In some embodiments, the modified enzyme has between 1 and about 20 amino acid substitutions, additions, and/or deletions compared to naturally occurring enzyme counterparts. The substitution, addition and insertion of amino acids can be accomplished using natural or unnatural amino acids. Non-naturally occurring amino acids include, but are not limited to, ε-N lysine, ß-alanine, ornithine, n-leucine, n-valine, hydroxyproline, thyroxine, γ-amine Butyric acid, homoserine, citrulline, aminobenzoic acid, 6-aminohexanoic acid (Aca; 6-Aminohexanoic acid), hydroxyproline, thiopropionic acid (MPA), 3-nitro-casein Amino acid, pyroglutamic acid and the like. Naturally occurring amino acids include alanine, arginine, aspartic acid, aspartic acid, cysteine, glutamic acid, glutamic acid, glycine, histidine, isoleucine , Leucine, lysine, methionine, amphetamine, proline, serine, threonine, tryptophan, tyrosine and valine.

經修飾的酵素可源自於岩藻醣基化途徑中的任何天然存在之酵素。例如,經修飾的酵素可源自於GDP-甘露醣4,6-脫水酶(GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(FX)及/或任何岩藻醣轉移酶,包含:2-α-L-岩藻醣轉移酶1(FUT1)、半乳醣苷2-α-L-岩藻醣轉移酶2(FUT2)、半乳醣苷3(4)-L-岩藻醣轉移酶(FUT3)、α(1,3)岩藻醣轉移酶,骨髓特異性(FUT4)、α-(1,3)-岩藻醣轉移酶(FUT5)、α-(1,3)-岩藻醣轉移酶(FUT6)、α-(1,3)-岩藻醣轉移酶(FUT7)、α-(1,6)-岩藻醣轉移酶(FUT8)、α-(1,3)-岩藻醣轉移酶(FUT9)、蛋白O-岩藻醣轉移酶1(POFUT1)、蛋白O-岩藻醣轉移酶2(POFUT2)。 The modified enzyme may be derived from any naturally occurring enzyme in the fucosylation pathway. For example, the modified enzyme may be derived from GDP-mannose 4,6-dehydratase (GMD), GDP-4-keto-6-deoxy-D-mannose isomerase-reductase (FX) and// Or any fucose transferase, including: 2-α-L-fucose transferase 1 (FUT1), galactosides 2-α-L-fucose transferase 2 (FUT2), galactosides 3 ( 4)-L-fucose transferase (FUT3), α(1,3) fucose transferase, bone marrow specific (FUT4), α-(1,3)-fucose transferase (FUT5), α-(1,3)-fucose transferase (FUT6), α-(1,3)-fucose transferase (FUT7), α-(1,6)-fucose transferase (FUT8) , Α-(1,3)-fucose transferase (FUT9), protein O-fucose transferase 1 (POFUT1), protein O-fucose transferase 2 (POFUT2).

在一些實施例中,岩藻醣基化途徑中之一個以上的酵素被修飾。在某些實施例,經修飾的酵素源自於GMD、FX及/或FUT8。 In some embodiments, more than one enzyme in the fucosylation pathway is modified. In certain embodiments, the modified enzyme is derived from GMD, FX, and/or FUT8.

ii.編碼經修飾的酵素的核酸ii. Nucleic acid encoding modified enzyme

本發明的去岩藻醣基化抗體可於宿主細胞中產生,在此宿主細胞中,岩藻醣基化途徑中酵素已經以降低或抑制蛋白的岩藻醣基化的方式被改變。 The defucosylated antibody of the present invention can be produced in a host cell in which the enzyme in the fucosylation pathway has been altered in a manner that reduces or inhibits the fucosylation of the protein.

在一些實施例中,藉由將編碼岩藻醣基化途徑中經修飾之酵 素的核酸導入至宿主細胞中,以改變宿主細胞的岩藻醣基化途徑。例如,將編碼經修飾的酵素的核酸分子插入至表現載體中,並轉染至宿主細胞中。編碼經修飾的酵素的核酸分子可瞬時導入宿主細胞中,或穩定地融合至宿主細胞的基因體中。可使用標準的重組DNA方法,來產生編碼經修飾的酵素的核酸,將核酸併入至表現載體中,以及將載體導入至宿主細胞中。 In some embodiments, the host cell's fucosylation pathway is altered by introducing a nucleic acid encoding a modified enzyme in the fucosylation pathway into the host cell. For example, nucleic acid molecules encoding modified enzymes are inserted into expression vectors and transfected into host cells. The nucleic acid molecule encoding the modified enzyme can be transiently introduced into the host cell or stably fused into the host cell's genome. Standard recombinant DNA methods can be used to generate nucleic acids encoding modified enzymes, incorporate the nucleic acids into expression vectors, and introduce the vectors into host cells.

在一些實施例中,宿主細胞可表現2個或以上的經修飾的酵素。例如,可用編碼2個或以上經修飾的酵素的核酸,來轉染宿主細胞。或者,可用一個以上的核酸,來轉染宿主細胞,各個核酸可編碼一個或以上的經修飾之酵素。 In some embodiments, the host cell may express 2 or more modified enzymes. For example, nucleic acids encoding two or more modified enzymes can be used to transfect host cells. Alternatively, more than one nucleic acid can be used to transfect the host cell, and each nucleic acid can encode one or more modified enzymes.

編碼經修飾的酵素的核酸可含有額外的核酸序列。例如,核酸可含有蛋白標籤、篩選標誌、或控制宿主細胞中蛋白表現的調控序列,例如啟動子、增強子或其它控制核酸(如,聚腺苷酸化信號)的轉錄或轉譯的表現控制片段。此種調控序列已為習知。本發明所屬技術領域中具有通常知識者可理解的是,可依據數個因素,包括要轉形的宿主細胞的選擇、所需蛋白質的表現量等,來選擇表現載體,包括調控片段的選擇。用於哺乳動物宿主細胞表現的示例性調控序列,包括哺乳動物細胞中指引高蛋白質表現量的病毒片段,例如,源自於巨細胞病毒(cytomegalovirus,CMV)(例如,CMV啟動子及/或增強子)、猿猴病毒40(Simian Virus,SV40)(例如,SV40啟動子及/或增強子)、腺病毒(例如,腺病毒主要晚期啟動子(adenovirus major late promoter,AdMLP))與多瘤性病毒的啟動子及/或增強子。 The nucleic acid encoding the modified enzyme may contain additional nucleic acid sequences. For example, the nucleic acid may contain protein tags, selection markers, or regulatory sequences that control the expression of proteins in the host cell, such as promoters, enhancers, or other expression control fragments that control the transcription or translation of nucleic acids (eg, polyadenylation signals). Such regulatory sequences are already known. Those of ordinary skill in the technical field to which the invention pertains can understand that the expression vector, including the selection of regulatory fragments, can be selected based on several factors, including the selection of the host cell to be transformed, the amount of protein expression required, and so on. Exemplary regulatory sequences for mammalian host cell expression, including viral fragments in mammalian cells that direct high protein expression, for example, derived from cytomegalovirus (CMV) (eg, CMV promoter and/or enhanced ), simian virus 40 (Simian Virus, SV40) (for example, SV40 promoter and/or enhancer), adenovirus (for example, adenovirus major late promoter (AdMLP)) and polyoma virus Promoter and/or enhancer.

在某些實施例中,將含有源自於GMD、FX及/或FUT的經修飾的酵素之核酸序列導入宿主細胞。在表現經修飾的酵素的宿主細胞中, 岩藻醣基化途徑會被改變、抑制、或降低。 In certain embodiments, nucleic acid sequences containing modified enzymes derived from GMD, FX, and/or FUT are introduced into host cells. In host cells that express modified enzymes, the fucosylation pathway is altered, inhibited, or reduced.

c.表現經修飾的酵素的宿主細胞c. Host cells expressing modified enzymes

本發明另一範疇有關於表現岩藻醣基化途徑中經修飾的酵素的宿主細胞。宿主細胞中經修飾的酵素的表現會干擾野生型酵素的活性,導致抑制或降低岩藻醣基化途徑。因此,於表現經修飾的酵素之宿主細細胞中所產生的蛋白(如,抗體)被去岩藻醣基化(無岩醣基化)。 Another category of the invention relates to host cells that express modified enzymes in the fucosylation pathway. The performance of the modified enzyme in the host cell interferes with the activity of the wild-type enzyme, resulting in inhibition or reduction of the fucosylation pathway. Therefore, proteins (eg, antibodies) produced in the host fine cells expressing modified enzymes are defucosylated (non-fucosylated).

如本發明說所使用的詞組「低岩藻醣基化細胞」或「低岩藻醣基化宿主細胞」係指,因為細胞表現岩藻醣基化途徑中經修飾的酵素,所以岩藻醣基化途徑已受到抑制或減少的細胞。 The phrase "low-fucosylated cell" or "low-fucosylated host cell" as used in the present invention refers to the fact that since the cell expresses a modified enzyme in the fucosylation pathway, fucose Cells whose basification pathway has been inhibited or reduced.

可藉由將含有編碼岩藻醣基化途徑中經修飾之酵素的核酸序列的表現載體轉染至宿主細胞,以製備低岩藻醣基化細胞。可使用本領域已知的技術進行轉染。例如,可使用以化學為基礎的方法(如,脂質、磷酸鈣、陽離子聚合物、DEAE-葡聚醣、活化的樹枝形大分子,磁珠等)、藉由以儀器為基礎的方法(如,電穿孔、生物噴射技術、顯微注射、雷射/光注射等),或藉由以病毒為基礎的方法,來進行轉染。 Low-fucosylated cells can be prepared by transfecting a expression vector containing a nucleic acid sequence encoding a modified enzyme in the fucosylation pathway into a host cell. Transfection can be performed using techniques known in the art. For example, chemical-based methods (eg, lipids, calcium phosphate, cationic polymers, DEAE-dextran, activated dendrimers, magnetic beads, etc.), and instrument-based methods (eg , Electroporation, biojet technology, microinjection, laser/light injection, etc.), or by virus-based methods for transfection.

可使用存在於表現載體上的篩選標誌,自未轉染細胞篩選並分離出轉染細胞。此外,可進一步藉由各種技術,自具正常岩醣基化途徑的細胞篩選並分離出具有岩藻醣基化途徑被抑制或降低的轉染細胞,。例如,可使用抗體、凝集素、代謝標誌或化學酵素策略來確定岩藻醣基化。此外,可藉由使轉染細胞暴露於小扁豆凝集素(Lens culinaris agglutinin,LCA,Vector laboratories L-1040),來篩選出具有岩藻醣基化途徑被抑制或降低的細胞。LCA辨識N端連接的寡醣的α-1,6-岩藻醣基化三甘露醣核心結 構,並使表現此結構的細胞進入細胞死亡途徑。因此,在接觸LCA後存活的細胞具有經抑制或降低的岩藻醣化途徑,且被視為低岩藻醣基化細胞。 The screening markers present on the expression vector can be used to screen and isolate transfected cells from untransfected cells. In addition, transfected cells with inhibited or reduced fucosylation pathways can be screened and isolated from cells with normal fucosylation pathways by various techniques. For example, antibodies, lectins, metabolic markers, or chemical enzyme strategies can be used to determine fucosylation. In addition, cells with inhibited or reduced fucosylation pathways can be screened by exposing transfected cells to lentil lectin (Lens culinaris agglutinin, LCA, Vector laboratories L-1040). LCA recognizes the α-1,6-fucosylated trimannose core structure of N-linked oligosaccharides and allows cells expressing this structure to enter the cell death pathway. Therefore, cells that survive after exposure to LCA have an inhibited or reduced fucosylation pathway and are considered low-fucosylated cells.

2.去岩藻醣基化蛋白2. Defucosylated protein

本發明另一範疇係有關於一種生產去岩藻醣基化蛋白的方法。在一些實施例中,去岩藻醣基化蛋白為去岩藻醣基化抗體。 Another category of the invention relates to a method for producing defucosylated proteins. In some embodiments, the defucosylated protein is a defucosylated antibody.

a.蛋白a. protein

可被產生為去岩藻醣基化蛋白產生的蛋白的非限制範例包括,GP-73、凝血酶、HBsAg、B型肝炎病毒顆粒、α-酸-醣蛋白、α-1-糜蛋白酶、α-1-糜蛋白酶、α-1-糜蛋白酶組胺酸-脯胺酸-較少(His-Pro-less)、α-1-抗胰蛋白酶、血清轉鐵蛋白(Serotransferrin)、血漿銅藍蛋白、α-2-巨球蛋白、α-2-HS-醣蛋白、α-甲胎蛋白、結合珠蛋白、纖維蛋白原γ鏈前驅物、免疫球蛋白(包括IgG、IgA、IgM、IgD、IgE及其類似)、APO-D、激肽原、富含組胺酸的醣蛋白、補體因子1前驅物、補體因子I重鏈、補體因子I輕鏈、補體C1s、補體因子B前驅物、補體因子B Ba片段、補體因子B Bb片段、補體C3前驅物、補體C3 β鏈、補體C3 α鏈、C3a過敏毒素、補體、C3b α’鏈、補體C3c片段、補體C3dg片段、補體C3g片段、補體C3d片段、補體C3f片段、補體C5、補體C5 β鏈、補體C5 α鏈、C5a過敏毒素、補體C5α’鏈、補體C7、α-1 B醣蛋白、B-2-醣蛋白、維生素D-結合蛋白、Inter-α-胰蛋白酶抑製劑重鏈H2、α-1B-醣蛋白、血管收縮素原前驅物、血管收縮素-1、血管收縮素-2、血管收縮素-3、GARP蛋白、β-2-醣蛋白、聚集素(Clusterin)(Apo J)、整合素α-8前驅物醣蛋白、整合素α-8重鏈、整合素α-8輕鏈、C型肝炎病毒顆粒、elf-5、激肽原、HSP33-同源、離胺醯基內肽酶及富含重複亮胺酸的蛋白質 32前驅物。 Non-limiting examples of proteins that can be produced as defucosylated proteins include GP-73, thrombin, HBsAg, hepatitis B virus particles, α-acid-glycoprotein, α-1-chymotrypsin, α -1-chymotrypsin, α-1-chymotrypsin histidine-proline-less (His-Pro-less), α-1-antitrypsin, serum transferrin (Serotransferrin), plasma ceruloplasmin , Α-2-macroglobulin, α-2-HS-glycoprotein, α-fetoprotein, binding globin, fibrinogen γ chain precursor, immunoglobulin (including IgG, IgA, IgM, IgD, IgE And similar), APO-D, prokinin, histidine-rich glycoprotein, complement factor 1 precursor, complement factor I heavy chain, complement factor I light chain, complement C1s, complement factor B precursor, complement Factor B Ba fragment, complement factor B Bb fragment, complement C3 precursor, complement C3 β chain, complement C3 α chain, C3a allergic toxin, complement, C3b α′ chain, complement C3c fragment, complement C3dg fragment, complement C3g fragment, complement C3d fragment, complement C3f fragment, complement C5, complement C5 β chain, complement C5 α chain, C5a allergic toxin, complement C5α′ chain, complement C7, α-1 B glycoprotein, B-2-glycoprotein, vitamin D-binding Protein, Inter-α-trypsin inhibitor heavy chain H2, α-1B-glycoprotein, angiotensinogen precursor, angiotensin-1, angiotensin-2, angiotensin-3, GARP protein, β -2-glycoprotein, Clusterin (Apo J), integrin α-8 precursor glycoprotein, integrin α-8 heavy chain, integrin α-8 light chain, hepatitis C virus particles, elf- 5. Kininogen, HSP33-homologous, lysinyl endopeptidase and protein 32 precursor rich in repeated leucine.

b.抗體b. Antibody

本發明說明所使用的術語「抗體」廣泛地涵蓋了能被岩藻醣基化的完整抗體及其片段。例如,抗體包括完全組裝的免疫球蛋白(例如多株、單株、單特異性、多特異性、嵌合、去免疫、人源化、人類、靈長類化、單鏈、單結構域、合成及重組抗體);具所需活性或功能之完整抗體的部分(例如,抗體的免疫片段,其含有Fab、Fab’、F(ab’)2、Fv、scFv、單域片段);以及含有能被岩藻醣基化之Fc結構域的胜肽和蛋白(例如,Fc-融合蛋白)。 The term "antibody" used in the description of the present invention broadly covers intact antibodies and fragments thereof that can be fucosylated. For example, antibodies include fully assembled immunoglobulins (eg, multiple strains, single strains, monospecific, multispecific, chimeric, deimmunized, humanized, human, primate, single chain, single domain, (Synthetic and recombinant antibodies); parts of intact antibodies with desired activity or function (eg, immunological fragments of antibodies that contain Fab, Fab', F(ab')2, Fv, scFv, single-domain fragments); and contain Peptides and proteins of the Fc domain that can be fucosylated (eg, Fc-fusion proteins).

本發明說明所使用的術語「去岩藻醣基化抗體」係指在相較於在天然條件下所產生的抗體,岩藻醣基化被抑制或顯著降低的條件下所產生的抗體或其片段。以本發明方法所產生的去岩藻醣基化抗體可為完全(100%)去岩藻醣基化,或者可包括岩藻醣基化及去岩藻醣基化的分子的混合。例如,在一些實施例中,由本發明方法所產生之抗體可含有約20%至約100%的去岩藻醣基化分子。在另一些實施例中,由本發明方法所產生之抗體可含有約40%至約100%的去岩藻醣基化分子。在某些實施例中,由本發明方法所產生的抗體可含有約至少20%、30%、40%、50%、60%、70%、80%、90%、91%、92%、93%、94%、95%、96%、97、98%、99%或100%的去岩藻醣基化分子。並非所有的N-醣化抗體或其片段(例如,Fc-融合蛋白)皆需為去岩藻醣基化。 The term "defucosylated antibody" as used in the description of the present invention refers to an antibody produced under conditions in which fucosylation is inhibited or significantly reduced compared to antibodies produced under natural conditions or Fragment. The defucosylated antibody produced by the method of the present invention may be fully (100%) defucosylated, or may include a mixture of fucosylated and defucosylated molecules. For example, in some embodiments, the antibodies produced by the methods of the present invention may contain about 20% to about 100% of afucosylated molecules. In other embodiments, the antibody produced by the method of the present invention may contain about 40% to about 100% of afucosylated molecules. In certain embodiments, the antibodies produced by the methods of the invention may contain about at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93% , 94%, 95%, 96%, 97, 98%, 99% or 100% of fucosylated molecules. Not all N-glycated antibodies or fragments thereof (eg, Fc-fusion proteins) need to be defucosylated.

b.抗體種類b. Antibody type

可使用本發明方法可將任何抗體生產為去岩藻醣基化抗 體。對於可使用本發明方法產生的抗體的種類並無限制。以下為可產生的抗體的非詳盡列表。 Any antibody can be produced as a defucosylated antibody using the method of the present invention. There is no limitation on the types of antibodies that can be produced using the method of the present invention. The following is a non-exhaustive list of antibodies that can be produced.

辨識腫瘤相關抗原的抗體的範例包括抗-GD2抗體、抗-GD3抗體、抗-GM2抗體、抗-HER2抗體、抗-CD52抗體、抗-MAGE抗體、抗-HM124抗體、抗-甲狀旁腺激素相關蛋白(parathyroid hormone-related protein,PTHrP)抗體、抗-鹼性纖維細胞生長因子抗體與抗-FGF8抗體、抗-鹼性纖維細胞生長因子受體抗體與抗-FGFS受體抗體、抗-類胰島素生長因子抗體、抗-類胰島素生長因子受體抗體、抗-PMSA抗體、抗-血管內皮細胞生長因子抗體、抗-血管內皮細胞生長因子受體抗體及其類似物。 Examples of antibodies that recognize tumor-associated antigens include anti-GD2 antibodies, anti-GD3 antibodies, anti-GM2 antibodies, anti-HER2 antibodies, anti-CD52 antibodies, anti-MAGE antibodies, anti-HM124 antibodies, anti-parathyroid glands Hormone-related protein (PTHrP) antibody, anti-basic fibroblast growth factor antibody and anti-FGF8 antibody, anti-basic fibroblast growth factor receptor antibody and anti-FGFS receptor antibody, anti- Insulin-like growth factor antibodies, anti-insulin-like growth factor receptor antibodies, anti-PMSA antibodies, anti-vascular endothelial cell growth factor antibodies, anti-vascular endothelial cell growth factor receptor antibodies and the like.

辨識過敏或發炎相關抗原的抗體的範例包括抗-IL-6抗體、抗-IL-6受體抗體、抗-IL-5抗體、抗-IL-5受體抗體與抗-IL-4抗體、抗-腫瘤壞死因子抗體、抗-腫瘤壞死因子受體抗體、抗-CCR4抗體、抗-趨化因子抗體、抗-趨化因子受體抗體及其類似物。 Examples of antibodies that recognize antigens associated with allergy or inflammation include anti-IL-6 antibodies, anti-IL-6 receptor antibodies, anti-IL-5 antibodies, anti-IL-5 receptor antibodies and anti-IL-4 antibodies, Anti-tumor necrosis factor antibody, anti-tumor necrosis factor receptor antibody, anti-CCR4 antibody, anti-chemokine antibody, anti-chemokine receptor antibody and the like.

辨識循環器官疾病相關抗原的抗體的範例包括抗-GpIIb/IIIa抗體、抗-血小板衍生生長因子抗體、抗-血小板衍生生長因子受體抗體及抗-血液凝固因子抗體及其類似物。 Examples of antibodies that recognize antigens associated with circulating organ diseases include anti-GpIIb/IIIa antibodies, anti-platelet-derived growth factor antibodies, anti-platelet-derived growth factor receptor antibodies, and anti-blood coagulation factor antibodies and the like.

辨識病毒或細菌感染相關抗原的抗體的範例包括抗-gpl 20抗體、抗-CD4抗體、抗-CCR4抗體與抗-維羅毒素抗體及其類似物。 Examples of antibodies that recognize antigens associated with viral or bacterial infections include anti-gpl 20 antibodies, anti-CD4 antibodies, anti-CCR4 antibodies, and anti-verotoxin antibodies and the like.

許多治療性抗體為市售可得的,例如,結合VEGF(如,Bevacizumab(AVASTIN®))、EGFR(如,Cetuximab(ERBITUX®))、HER2(如,Trastuzumab(HERCEPTIN®))與CD20(如,Rituximab(RITUXAN®))的抗體,及結合TNFa(如,Etanecept(ENBREL®),其包括TNF受體的受體 結合域(p75))、CD2(如,Alefacept(AMEVIVE®),其包含LFA-3的CD2-結合區)、或B7(Abatacept(ORENCIA®),其包括CTLA4的B7-結合區)抗體的Fc-融合蛋白。 Many therapeutic antibodies are commercially available, for example, binding VEGF (eg, Bevacizumab (AVASTIN®)), EGFR (eg, Cetuximab (ERBITUX®)), HER2 (eg, Trastuzumab (HERCEPTIN®)) and CD20 (eg , Rituximab (RITUXAN®)), and TNFa (eg, Etanecept (ENBREL®), which includes the receptor binding domain of the TNF receptor (p75)), CD2 (eg, Alefacept (AMEVIVE®), which contains LFA -3 CD2-binding region), or B7 (Abatacept (ORENCIA®), which includes CTLA4's B7-binding region) antibody Fc-fusion protein.

3.製備去岩藻醣基化蛋白的方法3. Method for preparing defucosylated protein

在低岩藻醣基化細胞中產生本發明去岩藻醣基化蛋白,其包括去岩藻醣基化抗體。使用本領域習知技術,例如以編碼蛋白之表現載體轉染低岩藻醣基化細胞,可在低岩藻醣基化細胞中表現去岩藻醣基化蛋白。 Defucosylated proteins of the invention are produced in hypofucosylated cells, which include defucosylated antibodies. Using techniques known in the art, such as transfection of low-fucosylated cells with expression vectors encoding proteins, defucosylated proteins can be expressed in low-fucosylated cells.

可使用本領域習知技術,來製備編碼蛋白的表現載體。例如,將胺基酸序列反轉錄成核酸序列,以構築表現載體,較佳使用對於在其中表現蛋白質之生物體而言最佳化的核酸密碼子。然後,將編碼蛋白的核酸和其任何調控片段組裝並插入至所欲的表現載體中。表現載體可含有額外的核酸序列,例如蛋白標籤、篩選標誌、或控制蛋白表現的調控序列,如同上述含有經修飾的酵素的表現載體。表現載體接著可利用轉染導入宿主細胞。可使用已知本領域習知技術進行轉染。例如,可使用以化學為基礎的方法(如,脂質、磷酸鈣、陽離子聚合物、DEAE-葡聚醣、活化的樹枝形大分子、磁珠等),藉由以儀器為基礎的方法(如,電穿孔、生物噴射技術、顯微注射、雷射/光注射等),或藉由以病毒為基礎的方法進行轉染。之後在適合所選表現系統及宿主的條件下,可在經轉染的細胞中表現蛋白。可接著使用親和性管柱或其它本領域已知技術純化表現的蛋白。 Techniques known in the art can be used to prepare expression vectors encoding proteins. For example, to reverse transcribe an amino acid sequence into a nucleic acid sequence to construct an expression vector, it is preferable to use a nucleic acid codon optimized for the organism in which the protein is expressed. Then, the nucleic acid encoding the protein and any regulatory fragments thereof are assembled and inserted into the desired expression vector. The expression vector may contain additional nucleic acid sequences, such as protein tags, screening markers, or regulatory sequences that control protein expression, as described above for expression vectors containing modified enzymes. The expression vector can then be introduced into the host cell using transfection. Transfection can be performed using techniques known in the art. For example, chemical-based methods (eg, lipids, calcium phosphate, cationic polymers, DEAE-dextran, activated dendrimers, magnetic beads, etc.) can be used, with instrument-based methods (eg , Electroporation, biojet technology, microinjection, laser/light injection, etc.), or transfection by virus-based methods. The protein can then be expressed in the transfected cells under conditions suitable for the chosen expression system and host. The expressed protein can then be purified using affinity columns or other techniques known in the art.

將編碼經修飾的酵素(欲成為一低岩藻醣基化細胞)的核酸及編碼蛋白(欲表現此蛋白)的核酸,以任意的順序轉染至宿主細胞,以產生去岩藻醣基化蛋白。例如,可先將編碼經修飾的酵素(欲成為低岩藻醣基化 細胞)的核酸轉染至宿主細胞中,再以編碼一蛋白(欲表現此蛋白)的核酸轉染。或者,可先將編碼蛋白(欲表現此蛋白)的核酸轉染至宿主細胞中,再以編碼經修飾的酵素(欲成為一低岩藻醣基化細胞)的核酸轉染。在另一變化中,可同時將編碼經修飾的酵素(欲成為一低岩藻醣基化細胞)的核酸與編碼蛋白(欲表現此蛋白)的核酸轉染至宿主細胞中。 Transfect the nucleic acid encoding the modified enzyme (to be a low-fucosylated cell) and the nucleic acid encoding the protein (to express this protein) in any order into the host cell to produce defucosylated protein. For example, a nucleic acid encoding a modified enzyme (to be a hypofucosylated cell) can be transfected into a host cell, and then a nucleic acid encoding a protein (to express this protein) can be transfected. Alternatively, the nucleic acid encoding the protein (to express this protein) can be transfected into the host cell first, and then the nucleic acid encoding the modified enzyme (to be a low-fucosylated cell) can be transfected. In another variation, the nucleic acid encoding the modified enzyme (to be a hypofucosylated cell) and the nucleic acid encoding the protein (to express this protein) can be transfected into the host cell at the same time.

在一特定實施例中,依照下列步驟,藉由先製備低岩藻醣基化細胞,然後將編碼蛋白質的核酸轉染至低岩藻醣基化細胞中,以產生去岩藻醣基化蛋白:a)獲得適合表現蛋白的宿主細胞;b)將編碼經修飾的酵素的核酸轉染至此宿主細胞中;c)篩選及/或分離具低岩藻醣基化的轉染細胞;d)將編碼蛋白的核酸轉染至低岩藻醣基化細胞中;e)篩選及/或分離經以編碼蛋白之核酸轉染的低岩藻醣基化細胞;f)誘導蛋白在低岩藻醣基化細胞中的表現。 In a specific embodiment, according to the following steps, by preparing low-fucosylated cells first, and then transfecting the protein-encoding nucleic acid into the low-fucosylated cells to produce defucosylated proteins : A) obtain a host cell suitable for protein expression; b) transfect the nucleic acid encoding the modified enzyme into this host cell; c) screen and/or isolate transfusified cells with low fucosylation; d) Transfection of protein-encoding nucleic acid into low-fucosylated cells; e) screening and/or isolation of low-fucosylated cells transfected with protein-encoding nucleic acid; f) induction of proteins in low-fucosylated Performance in cells.

在另一實施例中,依照下列步驟,藉由先將編碼蛋白的核酸轉染至宿主細胞中,再以編碼經修飾的酵素的核酸轉染此細胞,以產生去岩藻醣基化蛋白:a)獲得適合表現蛋白的宿主細胞;b)將編碼蛋白的核酸轉染至宿主細胞中;c)篩選及/或分離經以編碼蛋白的核酸轉染的細胞;d)將編碼經修飾的酵素的核酸轉染至步驟(c)的細胞中;e)篩選及/或分離步驟(d)中之具低岩藻醣基化的轉染細胞; f)誘導蛋白在低岩藻醣基化細胞中的表現。 In another embodiment, according to the following steps, by transfecting the nucleic acid encoding the protein into the host cell, and then transfecting the cell with the nucleic acid encoding the modified enzyme, the defucosylated protein is produced: a) obtain a host cell suitable for protein expression; b) transfect the nucleic acid encoding the protein into the host cell; c) screen and/or isolate cells transfected with the nucleic acid encoding the protein; d) encode the modified enzyme Transfected nucleic acid into cells of step (c); e) screening and/or isolation of transfected cells with low fucosylation in step (d); f) induced protein in low fucosylated cells Performance.

在上述實施例的變化中,去岩藻醣基化蛋白可依下述步驟產生:a)獲得表現或過度表現蛋白的宿主細胞;b)將編碼經修飾的酵素的核酸轉染至此細胞中;c)篩選及/或分離具低岩藻醣基化的轉染細胞;d)誘導蛋白在低岩藻醣基化細胞中的表現。 In a variation of the above embodiment, the defucosylated protein can be produced as follows: a) obtaining a host cell that expresses or overexpresses the protein; b) transfects the nucleic acid encoding the modified enzyme into this cell; c) Screen and/or isolate transfected cells with low fucosylation; d) Induce protein performance in low fucosylated cells.

在又一實施例中,如下,藉由同時將編碼經修飾的酵素(欲成為一低岩藻醣基化細胞)的核酸和編碼蛋白(欲表現此蛋白)的核酸轉染至宿主細胞,以產生去岩藻醣基化蛋白:a)獲得適合表現一蛋白的宿主細胞;b)將編碼蛋白的第一核酸及編碼經修飾的酵素的第二核酸轉染至宿主細胞;c)篩選及/或分離表現蛋白且具有低岩藻醣基化的轉染細胞;d)誘導蛋白在低岩藻醣基化細胞中的表現。 In yet another embodiment, as follows, by simultaneously transfecting a nucleic acid encoding a modified enzyme (to be a hypofucosylated cell) and a nucleic acid encoding a protein (to express this protein) into a host cell, to To produce afucosylated protein: a) obtain a host cell suitable for expressing a protein; b) transfect the first nucleic acid encoding the protein and the second nucleic acid encoding the modified enzyme into the host cell; c) screen and/or Or isolate transfected cells that express protein and have low fucosylation; d) Induce protein performance in low fucosylated cells.

使用上述的本發明方法產生去岩藻醣基化蛋白,包括抗體,可以本領域習知方法純化。例如,藉由本發明方法所產生的去岩藻醣基化蛋白,包括抗體,可以生理化學分餾、抗體類特異性親和力、抗原特異性親和力等方式純化。 The afucosylated proteins, including antibodies, produced using the method of the present invention described above can be purified by methods known in the art. For example, the defucosylated proteins, including antibodies, produced by the method of the present invention can be purified by physiochemical fractionation, antibody-specific affinity, and antigen-specific affinity.

4.去岩藻醣基化抗體改善的特性4. Improved properties of defucosylated antibodies

相較於使用標準方式所產生的抗體,以本發明方法生產的去岩藻醣基化抗體具有改善的特性。 Compared to antibodies produced using standard methods, the defucosylated antibodies produced by the method of the present invention have improved properties.

可藉由ELISA、螢光法及其類似方式,來測量去岩藻醣基化抗體的活性。可藉由測量其ADCC和CDC等方式,來評估抗原陽性的培養細胞株的細胞毒性活性。可使用相對接近人類的動物種類的適當模型,來評估抗體在人體內的安全性和治療效果。 The activity of defucosylated antibodies can be measured by ELISA, fluorescence method and the like. The cytotoxic activity of antigen-positive cultured cell lines can be evaluated by measuring their ADCC and CDC. Appropriate models of animal species that are relatively close to humans can be used to assess the safety and therapeutic effects of antibodies in humans.

a.增加的ADCC活性a. Increased ADCC activity

相較於使用標準方式所產生的抗體,本發明的去岩藻醣基化抗體具有增加的ADCC活性。 Compared to antibodies produced using standard methods, the defucosylated antibodies of the present invention have increased ADCC activity.

本發明方法所用之「ADCC活性」係指抗體引起抗體依賴性細胞毒性(ADCC)反應的能力。ADCC為細胞媒介的反應,其中表現FcRs之抗原非特異性細胞毒性細胞(如,自然殺手(NK)細胞、嗜中性細胞與巨噬細胞)辨識結合至目標細胞表面的抗體,接著造成目標細胞的分解(即,殺死)。ADCC中的主要媒介細胞是自然殺手(NK)細胞。NK細胞表現FcγRIII,其中FcγRIIIA為活化受體而FcγRIIIB為抑制受體。單核細胞表現FcγRI、FcγRII與FcγRIII。可使用體外分析法,如實施例3中描述的分析法,來直接評定ADCC活性。 "ADCC activity" used in the method of the present invention refers to the ability of an antibody to cause an antibody-dependent cytotoxicity (ADCC) response. ADCC is a cell-mediated response in which antigen-specific cytotoxic cells that express FcRs (eg, natural killer (NK) cells, neutrophils, and macrophages) recognize antibodies that bind to the surface of target cells, which then cause the target cells Decomposition (ie, kill). The main mediator cells in ADCC are natural killer (NK) cells. NK cells express FcyRIII, where FcyRIIIA is an activated receptor and FcyRIIIB is an inhibitory receptor. Monocytes express FcγRI, FcγRII and FcγRIII. In vitro assays, such as the assay described in Example 3, can be used to directly assess ADCC activity.

可使用體外分析法,來直接評定ADCC活性。在一些實施例中,本發明的去岩藻醣基化抗體的ADCC活性至少是其野生型對照抗體的0.5、1、2、3、5、10、20、50、100倍。 In vitro assays can be used to directly assess ADCC activity. In some embodiments, the ADCC activity of the defucosylated antibody of the present invention is at least 0.5, 1, 2, 3, 5, 10, 20, 50, 100 times its wild-type control antibody.

由於去岩藻醣基化抗體具有增加的ADCC活性,相較於其岩藻醣基化抗體,可以較低的量或濃度,來給予去岩藻醣基化的治療抗體。在一些實施例中,本發明的去岩藻醣基化抗體的濃度可比其岩藻醣基化抗體的濃度低至少2、3、5、10、20、30、50或100倍。在一些實施例中,相 較於其野生型對應物,本發明的去岩藻醣基化抗體可展現更高的最大目標細胞分解(maximal target cell lysis)。例如,本發明的去岩藻醣基化抗體的最大目標細胞分解(maximal targe cell lysis)比其野生型抗體的最大目標細胞分解高10%、15%、20%、25%、30%、40%、50%或更多。 Since the defucosylated antibody has increased ADCC activity, the defucosylated therapeutic antibody can be administered in a lower amount or concentration than its fucosylated antibody. In some embodiments, the concentration of the afucosylated antibody of the invention may be at least 2, 3, 5, 10, 20, 30, 50, or 100 times lower than the concentration of its fucosylated antibody. In some embodiments, the defucosylated antibody of the present invention can exhibit a higher maximum target cell lysis than its wild-type counterpart. For example, the maximum target cell breakdown of the defucosylated antibody of the present invention is 10%, 15%, 20%, 25%, 30%, 40 higher than the maximum target cell breakdown of its wild-type antibody %, 50% or more.

b.增加的CDC活性b. Increased CDC activity

相較於使用標準方法產生的抗體,本發明的去岩藻醣基化抗體具有增加的補體依賴性細胞毒性(CDC)活性。 Compared to antibodies produced using standard methods, the defucosylated antibodies of the invention have increased complement dependent cytotoxicity (CDC) activity.

本發明方法所使用的「CDC活性」係指辨識在目標細胞上結合的抗體,接著使目標細胞分解的補體系統中的一或多個成分的反應。本發明的去岩藻醣基化抗體不會降低或抑制CDC活性,但相反地,其維持類似或高於其岩藻醣基化對應物的CDC活性。 The "CDC activity" used in the method of the present invention refers to the reaction of one or more components in the complement system that recognizes the antibody bound to the target cell and then decomposes the target cell. The defucosylated antibody of the present invention does not reduce or inhibit CDC activity, but on the contrary, it maintains CDC activity similar to or higher than that of its fucosylated counterpart.

本發明更提供具有增強的CDC功能的去岩藻醣基化抗體。在一實施例中,本發明的Fc變異體具有增加的CDC活性。在另一實施例中,所述去岩藻醣基化抗體具有比相對的分子高至少2倍,或至少3倍,或至少5倍,或至少10倍,或至少50倍,或至少100倍的CDC活性。 The present invention further provides defucosylated antibodies with enhanced CDC function. In one embodiment, the Fc variants of the invention have increased CDC activity. In another embodiment, the defucosylated antibody has at least 2-fold, or at least 3-fold, or at least 5-fold, or at least 10-fold, or at least 50-fold, or at least 100-fold higher than the relative molecule CDC activity.

4.使用去岩藻醣基化抗體4. Use defucosylated antibodies

可以靜脈內(intravenously,i.v.)、皮下(subcutaneously,s.c.)、肌內(intra-muscularly,i.m.)、皮內(intradermal,i.d.)、腹膜內(intraperitoneal,i.p.)或經由任何黏膜表面,如口服(orally,p.o.)、舌下(sublingually,s.l.)、臉頰、鼻、直腸、陰道或經由肺部途徑,來給予本發明的去岩藻醣基化抗體。 It can be intravenously (iv), subcutaneously (sc), intra-muscularly (im), intradermal (id), intraperitoneal (ip) or via any mucosal surface, such as oral ( Orally, po), sublingually (sl), cheek, nose, rectum, vagina, or via the pulmonary route to administer the defucosylated antibody of the present invention.

對於治療或預防各種疾病,包括癌症、發炎性疾病、免疫及自體免疫疾病、過敏、循環器官疾病(如,動脈硬化)以及病毒或細菌感染, 去岩藻醣基化抗體是有用。 Defucosylated antibodies are useful for the treatment or prevention of various diseases, including cancer, inflammatory diseases, immune and autoimmune diseases, allergies, circulatory organ diseases (eg, arteriosclerosis), and viral or bacterial infections.

本發明的去岩藻醣基化抗體的劑量會依據給予的對象及特定模式而改變。可根據本發明所屬技術領域中具有通常知識者所熟知的許多因子,包括但不限於,抗體目標、對象的種類以及對象的大小/體重,而改變所需的劑量。劑量可為0.1至100,000μg/kg體重。可單劑或多劑給予去岩藻醣基化抗體。去岩藻醣基化抗體可在24小時期間中給予一次,在24小時期間給予多次,或持續注入。可持續地或以特定時程給予去岩藻醣基化抗體。可由獲自動物模型的劑量-反應曲線推斷有效劑量。 The dosage of the defucosylated antibody of the present invention will vary depending on the subject to be administered and the specific mode. The required dose can be changed according to many factors well known to those of ordinary skill in the technical field to which the invention belongs, including but not limited to, antibody target, type of subject, and size/weight of the subject. The dose may be 0.1 to 100,000 μg/kg body weight. Defucosylated antibodies can be administered in single or multiple doses. The defucosylated antibody can be administered once in a 24-hour period, multiple times during a 24-hour period, or continuous injection. Defucosylated antibodies can be administered continuously or on a specific schedule. The effective dose can be inferred from the dose-response curve obtained from the animal model.

4.特定實施例4. Specific embodiments

本發明的特定實施例包括,但不限於以下: Specific embodiments of the invention include, but are not limited to the following:

(1)一種在宿主細胞中生產去岩藻醣基化蛋白的方法,包括將至少一個編碼岩藻醣基化途徑(fucosylation pathway)之經修飾之酵素的核酸導入至宿主細胞中,以產生去岩藻醣基化蛋白。 (1) A method for producing afucosylated protein in a host cell, comprising introducing at least one nucleic acid encoding a modified enzyme of the fucosylation pathway into the host cell to produce Fucosylated protein.

(2)如(1)之方法,其中此經修飾的酵素源自於GDP-甘露醣4,6-脫水酶(GDP-mannose 4,6-dehydrogenase,GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(GDP-4-keto-6-deoxy-D-mannose epinierase-reductase,FX)、或岩藻醣轉移酶(fucosyltransferase,FUT)。 (2) The method of (1), wherein the modified enzyme is derived from GDP-mannose 4,6-dehydrogenase (GMD), GDP-4-keto-6 -Deoxy-D-mannose isomerase-reductase (GDP-4-keto-6-deoxy-D-mannose epinierase-reductase, FX), or fucosyltransferase (FUT).

(3)如(1)之方法,其中經修飾的酵素源自於岩藻醣轉移酶(FUT)。 (3) The method of (1), wherein the modified enzyme is derived from fucose transferase (FUT).

(4)如(1)之方法,其中經修飾的酵素源自於α-1,6-岩藻醣轉移酶(FUT8)。 (4) The method of (1), wherein the modified enzyme is derived from α-1,6-fucose transferase (FUT8).

(5)如(1)之方法,其中經修飾的酵素於宿主細胞中降低或抑制經修飾之酵素所源自的野生型酵素的活性。 (5) The method of (1), wherein the modified enzyme reduces or inhibits the activity of the wild-type enzyme derived from the modified enzyme in the host cell.

(6)如(1)之方法,其中修飾酵素抑制或降低宿主細胞中的岩藻醣基化。 (6) The method of (1), wherein the modified enzyme inhibits or reduces fucosylation in the host cell.

(7)如(1)之方法,其中於宿主細胞中所產生的去岩藻醣基化蛋白為去岩藻醣基化抗體或其片段。 (7) The method according to (1), wherein the defucosylated protein produced in the host cell is a defucosylated antibody or a fragment thereof.

(8)如(7)之方法,其中去岩藻醣基化抗體或其片段至少90%去岩藻醣基化。 (8) The method of (7), wherein the defucosylated antibody or fragment thereof is at least 90% defucosylated.

(9)如(7)之方法,其中去岩藻醣基化抗體,相較於其岩藻醣基化抗體,具有增加的抗體依賴性細胞毒性(antibody-dependent cellular cytotoxicity,ADCC)活性。 (9) The method of (7), wherein the defucosylated antibody has increased antibody-dependent cellular cytotoxicity (ADCC) activity compared to its fucosylated antibody.

(10)如(7)之方法,其中去岩藻醣基化抗體的補體依賴性細胞毒性(CDC)相較於其岩藻醣基化對應物,未被降低或抑制。 (10) The method of (7), wherein the complement-dependent cytotoxicity (CDC) of the defucosylated antibody is not reduced or inhibited compared to its fucosylated counterpart.

(11)一種在宿主細胞中產生去岩藻醣基化蛋白的方法,包括將編碼岩藻醣基化途徑之經修飾之酵素的第一核酸序列,以及編碼欲產生的蛋白之第二核酸序列轉染至宿主細胞中。 (11) A method for producing afucosylated protein in a host cell, comprising a first nucleic acid sequence encoding a modified enzyme of the fucosylation pathway, and a second nucleic acid sequence encoding the protein to be produced Transfected into host cells.

(12)如(11)之方法,其中宿主細胞同時轉染第一核酸序列與第二核酸序列。 (12) The method of (11), wherein the host cell transfects the first nucleic acid sequence and the second nucleic acid sequence simultaneously.

(13)如(12)之方法,其中所產生的去岩藻醣基化蛋白為抗體和/或其片段。 (13) The method according to (12), wherein the defucosylated protein produced is an antibody and/or a fragment thereof.

(14)如(11)之方法,其中第一核酸編碼源自於GDP-甘露醣4,6-脫水酶(GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(FX)、及/或岩藻醣轉移酶(FUT)的經修飾的酵素。 (14) The method according to (11), wherein the first nucleic acid encodes a GDP-mannose 4,6-dehydratase (GMD), GDP-4-keto-6-deoxy-D-mannose isomerase -Modified enzymes of reductase (FX) and/or fucose transferase (FUT).

(15)如(11)之方法,其中第一核酸編碼源自於α-1,6-岩藻醣轉移酶(FUT8)的經修飾的酵素。 (15) The method according to (11), wherein the first nucleic acid encodes a modified enzyme derived from α-1,6-fucose transferase (FUT8).

(16)如(11)之方法,包括下列步驟:a)將編碼經修飾的酵素的第一核酸轉染至宿主細胞中;b)篩選及分離低岩藻醣基化的轉染細胞;c)將編碼欲產生之蛋白的第二核酸轉染至低岩藻醣基化細胞中; d)在此低岩藻醣基化細胞中表現欲產生之蛋白。 (16) The method according to (11), including the following steps: a) transfecting the first nucleic acid encoding the modified enzyme into the host cell; b) screening and isolating the low-fucosylated transfected cells; c ) Transfect the second nucleic acid encoding the protein to be produced into the hypofucosylated cell; d) express the protein to be produced in the low fucosylated cell.

(17)如(11)之方法,其中所產生的去岩藻醣基化蛋白為抗體及/或其片段。 (17) The method according to (11), wherein the defucosylated protein produced is an antibody and/or a fragment thereof.

(18)如(11)之方法,包括以下步驟:a)將編碼所欲產生的蛋白的第二核酸轉染至宿主細胞;b)篩選及分離轉染第二核酸的細胞;c)將編碼經修飾的酵素之第一核酸轉染至於步驟(b)中的細胞;d)篩選及分離低岩藻醣基化的轉染細胞;e)在此低岩藻醣基化細胞中表現欲產生之蛋白。 (18) The method according to (11), including the following steps: a) transfecting a second nucleic acid encoding the protein to be produced into a host cell; b) screening and isolating cells transfected with the second nucleic acid; c) transcoding The first nucleic acid of the modified enzyme is transfected into the cell in step (b); d) Screening and isolation of transfusified cells with low fucosylation; e) Demonstration of production in this low fucosylated cell Of protein.

(19)如(18)之方法,其中所產生之去岩藻醣基化蛋白為抗體及/或其片段。 (19) The method according to (18), wherein the defucosylated protein produced is an antibody and/or a fragment thereof.

5.額外實施例5. Additional embodiments

本發明的額外實施例包括,但不限於以下: Additional embodiments of the invention include, but are not limited to the following:

(1)一種產生去岩藻醣基化抗體的方法,包括:將編碼至少一經修飾的酵素的核酸導入至宿主細胞中,以在宿主細胞中產生去岩藻醣基化抗體。 (1) A method for producing defucosylated antibodies, comprising: introducing a nucleic acid encoding at least one modified enzyme into a host cell to produce defucosylated antibodies in the host cell.

(2)如(1)之方法,其中經修飾的酵素源自於GDP-甘露醣4,6-脫水酶(GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(FX)、或岩藻醣轉移酶(FUT)。 (2) The method of (1), wherein the modified enzyme is derived from GDP-mannose 4,6-dehydratase (GMD), GDP-4-keto-6-deoxy-D-mannose isomerase -Reductase (FX), or fucose transferase (FUT).

(3)如(1)之方法,其中經修飾的酵素源自於岩藻醣轉移酶(FUT)。 (3) The method of (1), wherein the modified enzyme is derived from fucose transferase (FUT).

(4)如(1)之方法,其中經修飾的酵素源自於α-1,6-岩藻醣轉移酶(FUT8)。 (4) The method of (1), wherein the modified enzyme is derived from α-1,6-fucose transferase (FUT8).

(5)如(1)之方法,其中經修飾的酵素抑制宿主細胞中的野生型岩藻醣基化酵素的活性。 (5) The method of (1), wherein the modified enzyme inhibits the activity of wild-type fucosylated enzyme in the host cell.

(6)如(1)之方法,其中經修飾的酵素抑制及/或降低宿主細胞中抗體的岩藻醣基化。 (6) The method of (1), wherein the modified enzyme inhibits and/or reduces fucosylation of the antibody in the host cell.

(7)如(1)之方法,其中去岩藻醣基化抗體具有增加的ADCC。 (7) The method of (1), wherein the defucosylated antibody has increased ADCC.

(8)如(1)之方法,其中去岩藻醣基化抗體的CDC活性未被降低或抑制。 (8) The method of (1), wherein the CDC activity of the defucosylated antibody is not reduced or inhibited.

(9)一種產生去岩藻醣基化抗體的方法,包括:a)提供宿主細胞,b)將編碼至少一經修飾的酵素的核酸導入至宿主細胞中,以及c)在宿主細胞中產生去岩藻醣基化抗體。 (9) A method for producing defucosylated antibodies, comprising: a) providing a host cell, b) introducing a nucleic acid encoding at least one modified enzyme into the host cell, and c) producing de-rock in the host cell Fucosylated antibody.

(10)如(9)之方法,其中於步驟(a)中,宿主細胞包括至少一編碼抗體的核酸。 (10) The method of (9), wherein in step (a), the host cell includes at least one nucleic acid encoding an antibody.

(11)如(9)之方法,更將編碼抗體的核酸導入至步驟(b)後的宿主細胞中。 (11) As in the method of (9), the nucleic acid encoding the antibody is further introduced into the host cell after step (b).

(12)如(9)之方法,其中經修飾的酵素源自於GDP-甘露醣4,6-脫水酶(GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(FX)、及/或岩藻醣轉移酶(FUT)。 (12) The method of (9), wherein the modified enzyme is derived from GDP-mannose 4,6-dehydratase (GMD), GDP-4-keto-6-deoxy-D-mannose isomerase -Reductase (FX), and/or fucose transferase (FUT).

(13)如(9)之方法,其中經修飾的酵素源自於岩藻醣轉移酶(FUT)。 (13) The method of (9), wherein the modified enzyme is derived from fucose transferase (FUT).

(14)如(9)之方法,其中經修飾的酵素源自於α-1,6-岩藻醣轉移酶(FUT8)。 (14) The method of (9), wherein the modified enzyme is derived from α-1,6-fucose transferase (FUT8).

(15)如(9)之方法,其中經修飾的酵素抑制宿主細胞中岩藻醣基化酵素的活性。 (15) The method of (9), wherein the modified enzyme inhibits the activity of fucosylase in the host cell.

(16)如(9)之方法,其中經修飾的酵素抑制及降低宿主細胞中抗體的醣基化。 (16) The method of (9), wherein the modified enzyme inhibits and reduces the glycosylation of the antibody in the host cell.

(17)如(9)之方法,其中去岩藻醣基化抗體具有增加的ADCC。 (17) The method of (9), wherein the defucosylated antibody has increased ADCC.

(18)如(9)之方法,其中去岩藻醣基化抗體的CDC活性未被降低或抑制。 (18) The method of (9), wherein the CDC activity of the defucosylated antibody is not reduced or inhibited.

(19)一種以(1)或(9)之方法所產生的去岩藻醣基化抗體,其中去岩藻醣基化抗體具有增加的ADCC活性。 (19) A defucosylated antibody produced by the method of (1) or (9), wherein the defucosylated antibody has increased ADCC activity.

(20)如(19)之方法,其中去岩藻醣基化抗體為人類抗體或其片段。 (20) The method according to (19), wherein the defucosylated antibody is a human antibody or a fragment thereof.

(21)如(19)之方法,其中去岩藻醣基化抗體維持原有的CDC活性。 (21) The method according to (19), wherein the defucosylated antibody maintains the original CDC activity.

(22)一種醫藥組成物,包括(19)之去岩藻醣基化抗體及藥學上可接受之載體或賦形劑。 (22) A pharmaceutical composition comprising the defucosylated antibody of (19) and a pharmaceutically acceptable carrier or excipient.

(23)一種無或具低岩藻醣基化的細胞,包括編碼至少一經修飾的酵素的核酸。 (23) A cell with no or low fucosylation, including a nucleic acid encoding at least one modified enzyme.

額外的本發明特定實施例包括,但不限於以下實施例。 Additional specific embodiments of the present invention include, but are not limited to the following embodiments.

【實施例1】[Example 1]

岩藻醣基化途徑中經修飾之酵素的製備及表現此經修飾之酵素的穩定細胞株Preparation of modified enzyme in the fucosylation pathway and stable cell lines expressing the modified enzyme

1.細胞株1. Cell line

市售CHOdhfr(-)細胞株(ATCC CRL-9096),其為缺乏二氫葉酸還原酶活性的CHO細胞突變株,購自於菌種保存及研究中心(Culture Collection and Research Center(CCRC),台灣)。將CHOdhfr(-)細胞株分成3個分開的培養物,且如下所示進行處理:將第一培養物轉染編碼RITUXAN®(Rituximab,一種抗蛋白質CD20的嵌合單株抗體)的表現載體。獲得表現RITUXAN®的穩定細胞株且鑑別為RC79。 Commercially available CHOdhfr(-) cell line (ATCC CRL-9096), which is a mutant strain of CHO cells lacking dihydrofolate reductase activity, purchased from Culture Collection and Research Center (CCRC), Taiwan ). The CHOdhfr (-) cell line was divided into 3 separate cultures and treated as follows: The first culture was transfected with a expression vector encoding RITUXAN® (Rituximab, a chimeric monoclonal antibody against protein CD20). A stable cell line expressing RITUXAN® was obtained and identified as RC79.

將第二培養物轉染編碼HERCEPTIN®(Trastuzumab,一種抗蛋白質HER2的單株抗體)。獲得表現HERCEPTIN®的穩定細胞株且鑑別為HC59。 The second culture was transfected encoding HERCEPTIN® (Trastuzumab, a monoclonal antibody against the protein HER2). A stable cell line expressing HERCEPTIN® was obtained and identified as HC59.

第三培養物為未經處理的細胞,且維持為CHOdhfr(-)細胞 株。 The third culture was untreated cells and was maintained as CHOdhfr(-) cell line.

2.編碼FUT8與GMD之經修飾的酵素之表現載體的構築2. Construction of expression vector encoding modified enzymes of FUT8 and GMD

構築許多編碼經修飾之酵素FUT8與GMD的表現載體。 Constructed many expression vectors encoding modified enzymes FUT8 and GMD.

F83M、F8M1、F8M2、F8M3及F8D1突變型分別表示對α-1,6-岩藻醣轉移酶,野生型FUT8蛋白(GenBank No.NP_058589.2),的不同修飾。表1總結針對各FUT8載體對於野生型核酸序列所做的修飾,及在所表現的酵素中所產生的胺基酸改變。特別是,F83M代表在野生型FUT8蛋白中於R365A、D409A與D453A有3個修飾的突變型。F8M1、F8M2與F8M3分別代表野生型FUT8蛋白中各自在K369E、D409K與S469V有一個修飾的突變型。F8D1代表於野生型FUT8蛋白中,具有於第365至386位之胺基酸殘基刪除的突變型。 The F83M, F8M1, F8M2, F8M3 and F8D1 mutants represent different modifications to α-1,6-fucose transferase and wild-type FUT8 protein (GenBank No. NP_058589.2), respectively. Table 1 summarizes the modifications made to the wild-type nucleic acid sequence for each FUT8 vector and the amino acid changes produced in the enzymes shown. In particular, F83M represents a mutant with three modifications in R365A, D409A and D453A in the wild-type FUT8 protein. F8M1, F8M2, and F8M3 respectively represent mutants with a modification in K369E, D409K, and S469V in the wild-type FUT8 protein. F8D1 represents a mutant with deletion of amino acid residues at positions 365 to 386 in the wild-type FUT8 protein.

表2總結GMD載體對於野生型核酸序列所做的修飾,及在所表現的酵素中所產生的胺基酸改變。特別是,突變型GMD4M表示對GDP-甘露醣4,6-脫水酶,野生型GMD蛋白(GenBank No.NP_001233625.1,的修飾,其在野生型GMD蛋白中有4個突變,在T155A、E157A、Y179A與K183A。 Table 2 summarizes the modifications made by the GMD vector to the wild-type nucleic acid sequence, and the amino acid changes produced in the enzymes represented. In particular, the mutant GMD4M represents a modification of GDP-mannose 4,6-dehydratase, the wild-type GMD protein (GenBank No. NP_001233625.1, which has 4 mutations in the wild-type GMD protein, in T155A, E157A , Y179A and K183A.

所有編碼F83M、F8M1、F8M2、F8M3、F8D1及GMD4M的核酸序列由GeneDireX公司合成,並次選殖至pHD表現載體(pcDNA3.1Hygro,Invitrogen,Carlsbad,CA,cat.no.V870-20,具dhfr基因)的PacI/EcoRv或BamHI/EcoRV位置,以形成pHD/F83M、pHD/F8M1、pHD/F8M2、pHD/F8M3、pHD/F8D1及pHD/GMD4M質體。 All nucleic acid sequences encoding F83M, F8M1, F8M2, F8M3, F8D1, and GMD4M were synthesized by GeneDireX and sub-selected into the pHD expression vector (pcDNA3.1Hygro, Invitrogen, Carlsbad, CA, cat.no.V870-20, with dhfr Gene) PacI/EcoRv or BamHI/EcoRV positions to form pHD/F83M, pHD/F8M1, pHD/F8M2, pHD/F8M3, pHD/F8D1 and pHD/GMD4M plastids.

3.表現經修飾的酵素之穩定重組細胞株的製備3. Preparation of stable recombinant cell lines expressing modified enzymes

將pHD/F83M、pHD/F8M1、pHD/F8M2、pHD/F8M3、 pHD/F8D1及pHD/GMD4M質體藉由電穿孔(PA4000 PULSEAGILE® electroporator,Cyto Pulse Sciences)轉染至不同細胞株中,包括(a)RC79細胞株(表現RITUXAN®的CHO細胞)、(b)HC59細胞株(表現HERCEPTIN®的CHO細胞)、以及(c)CHOdhfr(-)細胞(缺乏二氫葉酸還原酶活性的CHO細胞突變株)。 Transfection of pHD/F83M, pHD/F8M1, pHD/F8M2, pHD/F8M3, pHD/F8D1 and pHD/GMD4M plastids by electroporation (PA4000 PULSEAGILE® electroporator, Cyto Pulse Sciences) into different cell lines, including ( a) RC79 cell line (CHO cell expressing RITUXAN®), (b) HC59 cell line (CHO cell expressing HERCEPTIN®), and (c) CHOdhfr (-) cell (CHO cell lacking dihydrofolate reductase activity mutation) Strain).

a. RC79細胞a. RC79 cells

轉染的RC79細胞株先培養於含0.1至0.25mg/mL潮黴素(Hygromycin)的RC79培養基中(含有0.4μM MTX、0.5mg/mL遺傳黴素(Geneticin)、0.05mg/mL博來黴素(Zeocin)、4mM Glutamax-I和0.01% F-68的EX-CELL®302無血清培養基)中。接著,將轉染細胞培養於含有0.4μM MTX、0.5mg/mL遺傳黴素、0.05mg/mL博來黴素、4mM Glutamax-I,0.01% F-68與0.25mg/mL潮黴素的EX-CELL®302無血清培養基中,並如以下所述,以扁豆凝集素(Lens culinaris agglutinin,LCA)分離,以產生5個細胞庫(cell pool),包括RC79F83M、RC79F8M1、RC79F8M2、RC79F8M3、RC79F8D1與RC79-GMD4M細胞株。 The transfected RC79 cell line was first cultivated in RC79 medium containing 0.1 to 0.25 mg/mL Hygromycin (containing 0.4 μM MTX, 0.5 mg/mL Geneticin, 0.05 mg/mL bleomycin) (Zeocin), 4 mM Glutamax-I and 0.01% F-68 in EX-CELL® 302 serum-free medium). Next, the transfected cells were cultured in EX containing 0.4 μM MTX, 0.5 mg/mL geneticin, 0.05 mg/mL bleomycin, 4 mM Glutamax-I, 0.01% F-68 and 0.25 mg/mL hygromycin -CELL®302 serum-free medium and separated with Lens culinaris agglutinin (LCA) as described below to produce 5 cell pools, including RC79F83M, RC79F8M1, RC79F8M2, RC79F8M3, RC79F8D1 and RC79-GMD4M cell line.

b. HC59細胞b. HC59 cells

轉染的HC59細胞株先培養於含0.1至0.25mg/mL潮黴素的HC59培養基(含有0.8μM MTX、0.5mg/mL遺傳黴素、0.05mg/mL博來黴素與4mM Glutamax-I的EX-CELL® 325 PF CHO培養基)。接著,將轉染的細胞培養於含有0.8μM MTX、0.5mg/mL遺傳黴素、0.05mg/mL博來黴素、4mM Glutamax-I與0.25mg/mL潮黴素的EX-CELL® 325 PF CHO培養基中,並如以下所述,以LCA分離,以產生HC59F83M細胞株的細胞庫。 The transfected HC59 cell line was first cultivated in HC59 medium containing 0.1 to 0.25 mg/mL hygromycin (containing 0.8 μM MTX, 0.5 mg/mL geneticin, 0.05 mg/mL bleomycin and 4 mM Glutamax-I EX-CELL® 325 PF CHO medium). Next, the transfected cells were cultured in EX-CELL® 325 PF containing 0.8 μM MTX, 0.5 mg/mL geneticin, 0.05 mg/mL bleomycin, 4 mM Glutamax-I and 0.25 mg/mL hygromycin In CHO medium and as described below, it was isolated with LCA to produce a cell bank of HC59F83M cell line.

c. CHOdhfrc. CHOdhfr (-)(-) 細胞cell

CHOdhfr(-)細胞先培養於含4mM Glutamax-I與0.1至0.25mg/mL潮黴素的EX-CELL® 325 PF CHO培養基。接著,將轉染的細胞培養於含有4mM Glutamax-I、0.25mg/mL潮黴素與0.01μM MTX的EX-CELL® 325 PF CHO培養基中,以產生C109F83M細胞株的細胞庫。 CHOdhfr (-) cells were first cultured in EX-CELL® 325 PF CHO medium containing 4 mM Glutamax-I and 0.1 to 0.25 mg/mL hygromycin. Next, the transfected cells were cultured in EX-CELL® 325 PF CHO medium containing 4 mM Glutamax-I, 0.25 mg/mL hygromycin and 0.01 μM MTX to generate a cell bank of C109F83M cell line.

4.具低岩藻醣基化細胞的分離4. Isolation of cells with low fucosylation

在此實施例中使用羅丹明(Rhodamine)標記的扁豆凝集素(Lens culinaris agglutinin,LCA)(Vector Laboratories,Cat.RL-1042),來篩選具低岩藻醣基化的細胞。 In this example, Rhodamine-labeled Lens culinaris agglutinin (LCA) (Vector Laboratories, Cat. RL-1042) was used to select cells with low fucosylation.

所有的RC79、HC59及CHO轉染株受到含有潮黴素的初步篩選培養基作為篩選壓力,接著使用可辨識N-連接寡醣之α-1,6-岩藻醣基化三甘露醣核心結構,並使表現此結構的細胞往細胞死亡途徑的LCA,來進行最終篩選。先以1.2 x 105細胞/mL,將RC79、HC59或CHO的轉染株種於含有0.4mg/mL LCA的2.5mL新鮮培養基中,且在第3或4天時計數細胞存活率。將細胞培養於此初步篩選培養基中,直到細胞存活率達到80%。在細胞存活率達到80%後,以1.2 x 105cells/mL,將細胞重新懸浮於含有梯度濃度之LCA的新鮮篩選培養基中。LCA篩選重複數次,直到LCA最終濃度達到0.6至1.2mg/mL。 All RC79, HC59 and CHO transfected strains were subjected to the initial selection medium containing hygromycin as the selection pressure, and then the core structure of α-1,6-fucosylated trimannose, which can identify N-linked oligosaccharides, The cells that express this structure are sent to the LCA of the cell death pathway for final screening. First to 1.2 x 10 5 cells / mL, the RC79, HC59 or CHO transfected lines were seeded in 2.5mL fresh media containing 0.4mg / mL LCA, the survival rate and the cells were counted on day 3 or 4. The cells were cultured in this preliminary screening medium until the cell survival rate reached 80%. After the cell survival rate reached 80%, the cells were resuspended in fresh screening medium containing gradient concentrations of LCA at 1.2 x 10 5 cells/mL. The LCA screening is repeated several times until the final LCA concentration reaches 0.6 to 1.2 mg/mL.

為分析細胞表面上的岩藻醣的量,以LCA標誌細胞,且以流式細胞術進行分析。首先,將細胞種於不含LCA的完全培養基14天,以去除來自篩選試劑LCA的訊號干擾。接著,以1mL冰冷PBS清洗3 x 105細胞2次,且重新懸浮於含1%牛血清白蛋白與5μg/mL LCA的200μl冰PBS中。在 冰上培養30分鐘後,以1mL冰冷PBS清洗細胞2次。將細胞重新懸浮於350μl冷的PBS中,並使用FACScaliburTM流式細胞儀(BD Biosciences,San Jose,CA)進行分析。 To analyze the amount of fucose on the cell surface, cells were labeled with LCA and analyzed by flow cytometry. First, the cells were planted in complete medium without LCA for 14 days to remove signal interference from the screening reagent LCA. Subsequently, 1mL ice-cold PBS at 3 x 10 5 cells were washed twice, and resuspended in 1% bovine serum albumin and 5μg / mL LCA in 200μl of ice-cold PBS. After incubating on ice for 30 minutes, the cells were washed twice with 1 mL of ice-cold PBS. The cells were resuspended in 350 μl cold PBS and analyzed using a FACScalibur flow cytometer (BD Biosciences, San Jose, CA).

接著,以10mL的冰冷PBS清洗1 x 107細胞2次,且重新懸浮於6.5mL含1%牛血清白蛋白與5μg/mL LCA的冰冷PBS中。在冰上培養30分鐘後,以10mL冷的PBS清洗細胞2次。將細胞重新懸浮於1mL含1%熱滅活的胎牛血清(GIBCO,Cat.10091-148)與抗生素-抗黴菌劑(Invitrogen,Cat.15240062)的冰冷PBS中。 Next, 1 x 10 7 cells were washed twice with 10 mL of ice-cold PBS, and resuspended in 6.5 mL of ice-cold PBS containing 1% bovine serum albumin and 5 μg/mL LCA. After incubating on ice for 30 minutes, the cells were washed twice with 10 mL of cold PBS. The cells were resuspended in 1 mL of ice-cold PBS containing 1% heat-inactivated fetal bovine serum (GIBCO, Cat. 10091-148) and antibiotic-antimycotic agent (Invitrogen, Cat. 15240062).

以FACSAriaTM或InfluxTM Cell Sorter(BD Biosciences,San Jose,CA)分析及分類細胞。對於不同的細胞株,需要1-3次的分類,以產生具低岩藻醣基化細胞的同質族群。此外,使用CLONEPIXTM 2系統(MOLECULAR DEVICES®),來分離具低岩藻醣基化的穩定細胞株,並移至96孔盤。在培養約2週後,將細胞移至6孔盤,並再以流式細胞術分析。然後,將具低岩藻醣基化細胞移至用於饋料批次(fed-batch)培養的過濾管,以評估細胞表現和由所得細胞純化之抗體的岩藻醣基化程度。 Cells were analyzed and sorted with FACSAria or Influx Cell Sorter (BD Biosciences, San Jose, CA). For different cell lines, 1-3 classifications are required to produce a homogeneous population with low fucosylated cells. In addition, the CLONEPIX 2 system (MOLECULAR DEVICES®) was used to isolate stable cell lines with low fucosylation and moved to 96-well plates. After culturing for about 2 weeks, the cells were transferred to a 6-well plate and analyzed by flow cytometry. Then, the cells with low fucosylation were moved to a filter tube for fed-batch culture to evaluate the cell performance and the degree of fucosylation of antibodies purified from the resulting cells.

5.表現RITUXAN®之C109F83M細胞株的製備5. Preparation of C109F83M cell line expressing RITUXAN®

在以LCA分離低岩藻醣基化CHOdhfr(-)細胞(C109F83M細胞)後,將編碼RITUXAN®的核酸以電穿孔的方式(PA4000 PULSEAGILE® electroporator,Cyto Pulse Sciences)轉染至細胞中。分離出C109F83M的低岩藻醣單一細胞株,AF97,並以電穿孔的方式轉染編碼RITUXAN®的核酸,以表現RITUXAN®。將轉染株移至含有非選擇性培養基的25T燒瓶燒瓶中,以回復生長。48小時後,將轉染株培養於含有4mM GlutaMAX-I、潮黴素-B、 博來黴素與0.01μM MTX的篩選培養基。使用CLONEPIX TM 2系統挑選出單一細胞,以產生AF97抗-CD20細胞株。 After isolating the low-fucosylated CHOdhfr (-) cells (C109F83M cells) with LCA, the nucleic acid encoding RITUXAN® was transfected into the cells by electroporation (PA4000 PULSEAGILE® electroporator, Cyto Pulse Sciences). A low-fucose single cell line of C109F83M, AF97, was isolated and transfected with nucleic acid encoding RITUXAN® to express RITUXAN® by electroporation. The transfected strain was transferred to a 25T flask flask containing non-selective medium to resume growth. After 48 hours, the transfected strain was cultured in a selection medium containing 4 mM GlutaMAX-I, hygromycin-B, bleomycin and 0.01 μM MTX. Single cells were selected using the CLONEPIX™ 2 system to produce AF97 anti-CD20 cell line.

所獲得的細胞為表現RITUXAN®的低岩藻醣基化CHOdhfr(-)細胞,且在此稱為AF97抗-CD20細胞株。 The obtained cells are low-fucosylated CHOdhfr (-) cells expressing RITUXAN®, and are referred to herein as AF97 anti-CD20 cell lines.

【實施例2】[Example 2] 去岩藻醣基化抗體的表現及分析Performance and analysis of defucosylated antibody 1.抗體的表現與純化1. Antibody performance and purification

將實施例1中所獲得的具低岩藻醣基化活性的細胞以批次或饋料批次培養,來表現抗體。由細胞所純化的抗體進行單醣分析,以對Fc區中的醣鏈進行定量分析。 The cells with low fucosylation activity obtained in Example 1 were cultured in batches or fed batches to express antibodies. The antibody purified from the cells is subjected to monosaccharide analysis to quantitatively analyze the sugar chain in the Fc region.

將重組RC79細胞培養於含有4mM Glutamax與0.01% F-68的EX-CELL® 302無血清培養基中,且維持於37℃、5% CO2的震盪培養箱(Infors Multitron Pro)中。 The recombinant RC79 cells were cultured in EX-CELL® 302 serum-free medium containing 4 mM Glutamax and 0.01% F-68, and maintained in a shaking incubator (Infors Multitron Pro) at 37° C. and 5% CO 2 .

將重組HC59細胞培養於含有0.8μM MTX、0.5mg/mL遺傳黴素、0.05mg/mL博來黴素、4mM Glutamax-I與0.25mg/mL潮黴素的EX-CELL® 325 PF CHO培養基中,且維持於37℃、5% CO2的震盪培養箱(Infors Multitron Pro)中。 Recombinant HC59 cells were cultured in EX-CELL® 325 PF CHO medium containing 0.8 μM MTX, 0.5 mg/mL geneticin, 0.05 mg/mL bleomycin, 4 mM Glutamax-I and 0.25 mg/mL hygromycin And maintained in a shaking incubator (Infors Multitron Pro) at 37°C, 5% CO 2 .

每天常規地監測細胞培養的參數。使用血球計數器(hemocytometer)以錐藍質(trypan blue)排除,來判定細胞密度及存活率。當細胞存活率低於60%時,藉由離心來收集條件培養基並以蛋白A樹脂來純化所表現的抗體。以5倍管柱體積的0.1M Tris,pH 8.3,來平衡蛋白A管柱,接著將樣本添加至管柱中。以0.1M Tris,pH 8.3(2倍管柱體積)及PBS,pH 6.5(10倍管柱體積)清洗掉未結合的蛋白。進一步以0.1M醋酸鈉,pH 6.5(10倍管柱體積)清洗管柱。最後,以0.1M甘胺酸,pH 2.8沖提抗體,並以相同沖提體積的0.1M Tris,pH 8.3進行中和。 Cell culture parameters are routinely monitored daily. A hemocytometer was used to exclude trypan blue to determine cell density and survival rate. When the cell survival rate is less than 60%, the conditioned medium is collected by centrifugation and the expressed antibody is purified with protein A resin. The protein A column was equilibrated with 5 column volumes of 0.1M Tris, pH 8.3, and then the sample was added to the column. Unbound protein was washed away with 0.1M Tris, pH 8.3 (2 column volumes) and PBS, pH 6.5 (10 column volumes). The column was further washed with 0.1 M sodium acetate, pH 6.5 (10 times the column volume). Finally, the antibody was eluted with 0.1 M glycine, pH 2.8, and neutralized with the same elution volume of 0.1 M Tris, pH 8.3.

2.抗體的N-多醣圖譜分析的判定2. Judgment of antibody N-polysaccharide map analysis

以ACQUITY UPLC®系統分析N-多醣圖譜。首先,在37℃下,以含有在0.3mL消化緩衝液(15mM Tris-HCl、pH 7.0)中之3 U PNGase-F,來消化0.3mg抗體樣本18小時。,釋放出的N-多醣藉由使用AMICON®Ultra-0.5mL 30K裝置,於13,000rpm下5分鐘之超過濾,以從抗體分離,然後冷凍乾燥3小時。接著,將乾燥的N-多醣溶於30μL的ddH2O與45μL的2-AB標記試劑(含0.34M氨基苯甲醯胺與1M氰基硼氫化鈉的DMSO-醋酸(7:3 v/v)溶劑)中,且於65℃下培養3小時。以PD MINITRAPTM G10尺寸排除管柱除去過量的2-AB標記試劑。將標記的N-多醣冷凍乾燥隔夜,並重新溶於50μL ddH2O中,以進行UPLC檢測。在60℃下,藉由ACQUITYUPLC®系統和Glycan BEH醯胺管柱,來獲得N-聚醣圖譜。以100mM甲酸銨,pH4.5/乙腈線性梯度,來分離不同形式的N-聚醣。 The N-polysaccharide map was analyzed with the ACQUITY UPLC® system. First, at 37°C, 0.3 mg of antibody sample was digested with 3 U PNGase-F in 0.3 mL of digestion buffer (15 mM Tris-HCl, pH 7.0) for 18 hours. The released N-polysaccharide was separated from the antibody by ultrafiltration using an AMICON® Ultra-0.5mL 30K device at 13,000 rpm for 5 minutes, and then freeze-dried for 3 hours. Next, the dried N-polysaccharide was dissolved in 30 μL of ddH 2 O and 45 μL of 2-AB labeling reagent (DMSO-acetic acid containing 0.34M aminobenzamide and 1M sodium cyanoborohydride (7:3 v/v ) In the solvent) and incubate at 65°C for 3 hours. Remove excess 2-AB labeling reagent in PD MINITRAP G10 size exclusion column. The labeled N-polysaccharide was freeze-dried overnight and redissolved in 50 μL ddH 2 O for UPLC detection. At 60°C, the N-glycan pattern was obtained by ACQUITYUPLC® system and Glycan BEH amide column. Different forms of N-glycans were separated with a linear gradient of 100 mM ammonium formate, pH 4.5/acetonitrile.

流式細胞儀的結果顯示,在所有細胞類型中,過度表現F83M蛋白的細胞表面上,LCA的結合極低。類似地,在過度表現F8M1、F8M2、F8M3、F8D1或GMD4M蛋白的RC79細胞上,未偵測到LCA結合(數據未顯示)。 The results of flow cytometry showed that LCA binding was extremely low on the cell surface overexpressing F83M protein in all cell types. Similarly, LCA binding was not detected on RC79 cells overexpressing F8M1, F8M2, F8M3, F8D1 or GMD4M proteins (data not shown).

表3顯示,具有未修飾的岩藻醣基化途徑之RC79與HC59細胞以及藉由過度表現F83M經修飾的酵素,來修飾其岩藻醣基化途徑的RC79與HC59細胞中所產生之抗體的N-聚醣圖譜。表3中的數據顯示,具有未修 飾岩藻醣基化途徑之細胞中所產生的抗-CD20與抗-ErbB2抗體大部分被重度岩藻醣基化。特別是,在這些細胞中,僅3.67%的抗-CD20與3.64%的抗-ErbB2抗體被去岩藻醣基化。相較之下,過度表現F83M經修飾之酵素的細胞中所產生之抗體具有非常低的岩藻醣基化程度。特別是,在過度表現F83M經修飾之酵素的細胞中,約98.86-98.91%的抗-CD20與約92.12-96.52%的抗-ErbB2抗體被去岩藻醣基化。 Table 3 shows the antibodies produced in RC79 and HC59 cells with unmodified fucosylation pathway and RC79 and HC59 cells whose fucosylation pathway is modified by overexpressing F83M modified enzyme N-glycan map. The data in Table 3 shows that the anti-CD20 and anti-ErbB2 antibodies produced in cells with unmodified fucosylation pathways are mostly heavily fucosylated. In particular, in these cells, only 3.67% of anti-CD20 and 3.64% of anti-ErbB2 antibodies were defucosylated. In contrast, antibodies produced in cells that overexpress F83M modified enzymes have a very low degree of fucosylation. In particular, in cells that overexpress F83M modified enzymes, approximately 98.86-98.91% of anti-CD20 and approximately 92.12-96.52% of anti-ErbB2 antibodies are defucosylated.

此外,表4顯示具有未修飾的岩藻基化途徑之RC79細胞以及藉由過度表現F8M1、F8M2、F8M3、F8D1或GMD4M經修飾的酵素之一,來修飾其岩藻醣基化途徑的RC79細胞中所產生之抗體的N-聚醣圖譜。表4的數據顯示,具有未修飾的岩藻醣基化途徑的RC79細胞中所產生之抗-CD20抗體大部分被重岩藻醣基化。特別是,在這些細胞中,僅3.67%的抗-CD20抗體為去岩藻醣基化。相較之下,過度表現經修飾的酵素之RC79細胞中所產生的抗體具有非常低的岩藻醣基化程度。特別是,由過度表現F8M1、F8M2、F8M3、F8D1或GMD4M經修飾的酵素之細胞所產生之抗-CD20抗體的去岩藻醣基化程度為約92.78%至約97.16%,如表4所示。 In addition, Table 4 shows RC79 cells with unmodified fucosylation pathway and RC79 cells with modified fucosylation pathway by overexpressing one of the modified enzymes of F8M1, F8M2, F8M3, F8D1 or GMD4M N-glycan profile of the antibody produced in The data in Table 4 shows that the anti-CD20 antibody produced in RC79 cells with an unmodified fucosylation pathway is mostly heavily fucosylated. In particular, in these cells, only 3.67% of the anti-CD20 antibody was defucosylated. In contrast, antibodies produced in RC79 cells that over-express modified enzymes have a very low degree of fucosylation. In particular, the degree of defucosylation of anti-CD20 antibodies produced by cells that overexpress F8M1, F8M2, F8M3, F8D1, or GMD4M modified enzymes is about 92.78% to about 97.16%, as shown in Table 4 . .

表4亦顯示由過度表現FUT8經修飾的酵素(F8M1、F8M2,F8M3,F8D1)之一的細胞所產生之抗體的去岩藻醣基化程度為95.70至97.16%,且由過度表現GMD經修飾的酵素(GMD4M)所生產之抗體的去岩藻醣基化程度為92.78%。這些結果證實,在過度表現FUT8經修飾的酵素之細胞中所產生之抗體的去岩醣基化程度高於在過度表現GMD突變型蛋白的細胞中所生產之抗體。 Table 4 also shows that the degree of defucosylation of antibodies produced by cells that overexpress one of FUT8 modified enzymes (F8M1, F8M2, F8M3, F8D1) is 95.70 to 97.16%, and is modified by overexpression of GMD The antibody produced by the enzyme (GMD4M) has a degree of defucosylation of 92.78%. These results confirm that the antibodies produced in cells that over-express FUT8 modified enzymes are more de-glycosylated than those produced in cells that over-express GMD mutant proteins.

表34的結果證實,已經設計來表現抗體的宿主細胞可轉染 表現岩藻醣基化途徑中經修飾的酵素(FUT8或GMD)的載體。此結果也顯示,在這些轉染細胞中所產生的抗體是去岩藻醣基化抗體。 The results in Tables 3 and 4 confirm that host cells that have been designed to express antibodies can be transfected with vectors that express modified enzymes (FUT8 or GMD) in the fucosylation pathway. This result also shows that the antibodies produced in these transfected cells are defucosylated antibodies.

此外,評估AF97細胞株中所產生之抗體的岩藻醣基化程度。表5的結果顯示,過度表現F83M經修飾的酵素之AF97細胞中所產生之抗體具有非常低岩藻醣基化程度。特別是,97.83%的在AF97細胞株中所產生之抗-CD20抗體為去岩藻醣基化。相較之下,市售RITUXAN®(MABTHERA®)具有3.92%的去岩藻醣基化程度。 In addition, the degree of fucosylation of antibodies produced in the AF97 cell line was evaluated. The results in Table 5 show that the antibodies produced in AF97 cells that overexpress F83M modified enzymes have a very low degree of fucosylation. In particular, 97.83% of the anti-CD20 antibodies produced in AF97 cell lines were defucosylated. In comparison, commercially available RITUXAN® (MABTHERA®) has a degree of defucosylation of 3.92%.

表5的結果顯示,可在先轉染編碼經修飾的酵素之核酸,接著第二次轉染編碼抗體之核酸的細胞中,產生去岩藻醣基化抗體。因此,可使用本發明方法,將細胞修飾以產生去岩藻醣基化抗體。 The results in Table 5 show that defucosylated antibodies can be produced in cells that were first transfected with nucleic acids encoding modified enzymes and then transfected with nucleic acids encoding antibodies a second time. Therefore, the method of the present invention can be used to modify cells to produce defucosylated antibodies.

3.重組細胞中FUT8蛋白的表現3. Expression of FUT8 protein in recombinant cells

將RC79細胞與表現FUT8經修飾的酵素(如,F8M1、F8M2、F8M3、或F8D1)的重組細胞的沉澱物分解於含有磷酸酶抑制劑雞尾酒(Sigma-Aldrich,Cat.S8820)的1% Triton X-100中。以DCTM(清潔劑)蛋白質分析(BIO-RAD)試劑,來確認分解細胞的上清液中的蛋白濃度。將含有30μg蛋白的上清液的每個樣品,以12.5%十二烷基磺酸鈉-聚丙烯醯胺膠體電泳(sodium dodecyl sulfate-polyacrylamide gel electrophoresis,SDS-PAGE)來分離並移至硝化纖維素膜上。藉由使用含有120mM NaCl、0.1% gelatin(w/w)與0.1% TWEEN®20(聚乙二醇山梨醣醇酐單月桂酸酯(v/w)之25mM Tris-HCl(pH7.4),來封阻硝化纖維素膜1小時,並在4℃下分別與抗-FUT8抗體(Abcam,Cat.ab204124,1:500)及GAPDH抗體(GeneTex,Cat.GT239,1:10000)分別培養隔夜。膜以25mM Tris-HCl(pH7.4)(含有120mM NaCl, 0.1% gelatin(w/w)and 0.1% TWEEN® 20(v/w))清洗膜3次,5分鐘,接著在室溫下與山羊抗-兔子IgG(Jackson ImmunoResearch,Cat.111-035-144)和山羊抗-小鼠IgG HRP(GeneTex,Cat.GTX213111-01,1:10000)分別反應1小時。額外清洗後,以SIGMAFAST DAB with Metal Enhancer(Sigma,Cat.D0426),來分析膜。 Decompose RC79 cells and recombinant cells expressing FUT8 modified enzymes (eg, F8M1, F8M2, F8M3, or F8D1) into 1% Triton X containing phosphatase inhibitor cocktail (Sigma-Aldrich, Cat. S8820) -100. DC TM (cleaner) protein analysis (BIO-RAD) reagent was used to confirm the protein concentration in the supernatant of decomposed cells. Each sample containing 30 μg of protein supernatant was separated and transferred to nitrocellulose by 12.5% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) On the plain film. By using 25mM Tris-HCl (pH7.4) containing 120mM NaCl, 0.1% gelatin (w/w) and 0.1% TWEEN® 20 (polyethylene glycol sorbitan monolaurate (v/w), The nitrocellulose membrane was blocked for 1 hour and incubated with anti-FUT8 antibody (Abcam, Cat.ab204124, 1:500) and GAPDH antibody (GeneTex, Cat.GT239, 1:10000) at 4°C overnight. The membrane was washed 3 times with 25 mM Tris-HCl (pH 7.4) (containing 120 mM NaCl, 0.1% gelatin (w/w) and 0.1% TWEEN® 20 (v/w)) for 5 minutes, then at room temperature with Goat anti-rabbit IgG (Jackson ImmunoResearch, Cat. 111-035-144) and goat anti-mouse IgG HRP (GeneTex, Cat. GTX213111-01, 1:10000) were reacted for 1 hour respectively. After additional washing, use SIGMAFAST DAB with Metal Enhancer (Sigma, Cat. D0426) to analyze the membrane.

第1圖為西方墨點法,其顯示RC79親源細胞中及表現經修飾的酵素之RC79重組細胞中的FUT8蛋白的表現。重組細胞及RC79親源中FUT8蛋白的表現量類似。這些結果表示,表現經修飾的酵素之RC79細胞中之去岩藻醣基化抗體的產生與FUT8蛋白的表現量無關。這些結果顯示FUT8經修飾的酵素會干擾細胞中的野生型FUT8蛋白,以抑制及/或降低岩藻醣基化途徑,使重組細胞有效率地產生去岩藻醣基化抗體。 FIG 1 is a western blot showing expression of FUT8 protein source parent cells RC79 and RC79 recombinant cell in the performance of the modified enzyme. The expression levels of FUT8 protein in recombinant cells and RC79 parent are similar. These results indicate that the production of defucosylated antibodies in RC79 cells expressing modified enzymes is independent of the amount of FUT8 protein expressed. These results show that FUT8 modified enzymes can interfere with wild-type FUT8 protein in cells to inhibit and/or reduce the fucosylation pathway, allowing recombinant cells to efficiently produce defucosylated antibodies.

相較於其它依靠抑制或向下調控野生型FUT8基因、或使用RNAi,來減少FUT8蛋白表現的方式,使用本發明方法產生去岩藻醣基化抗體的機制為新穎且獨特的。 Compared with other methods that rely on inhibiting or down-regulating the wild-type FUT8 gene, or using RNAi to reduce FUT8 protein expression, the mechanism for producing defucosylated antibodies using the method of the present invention is novel and unique.

4.重組細胞的穩定性4. Stability of recombinant cells

評估表現F83M經修飾的酵素之RC79重組細胞的穩定性。 The stability of RC79 recombinant cells expressing F83M modified enzyme was evaluated.

將RC79重組細胞培養在不含篩選試劑的培養基3個月。每週以流式細胞術分析,來監測細胞的岩藻醣基化,且每月以ACQUITY UPLC® System與Glycan BEH Amide管柱,判定經純化的抗體的N-聚醣的組成,持續3個月,如上所述。LCA非結合性特性在90天的評估期期間被維持,表示在整個研究過程中,岩藻醣基化途徑被抑制及/或降低(第2圖)。 The RC79 recombinant cells were cultured in medium without screening reagents for 3 months. Weekly flow cytometry analysis was used to monitor the fucosylation of the cells, and the ACQUITY UPLC® System and Glycan BEH Amide columns were used monthly to determine the composition of the purified antibody N-glycans for 3 consecutive Month, as described above. The non-binding properties of LCA were maintained during the 90-day evaluation period, indicating that the fucosylation pathway was inhibited and/or reduced throughout the study ( Figure 2 ).

此外,在72天的期間,評估在5個穩定的RC79F83M細胞株 (R4-R8)中所產生之抗-CD20抗體的去岩藻醣基化程度。如表6所示,所有RC79F83M細胞株在72天的研究中,皆產生高度去岩藻醣基化抗體。 In addition, during the 72-day period, the degree of defucosylation of anti-CD20 antibodies produced in 5 stable RC79F83M cell lines (R4-R8) was evaluated. As shown in Table 6 , all RC79F83M cell lines produced highly defucosylated antibodies during the 72-day study.

此研究的這些結果證明,藉由本發明方法製備的表現經修飾的酵素之重組細胞株為穩定的且長時間產生高度去岩藻醣基化的抗體。 These results of this study prove that the recombinant cell line expressing the modified enzyme prepared by the method of the present invention is stable and produces highly defucosylated antibodies for a long time.

【實施例3】[Example 3] 去岩藻醣基化抗體的ADCC活性ADCC activity of defucosylated antibody

為了評估實施例2所獲得之經純化的抗-CD20抗體的體外細胞毒性,依照下述方法測量ADCC活性。 In order to evaluate the in vitro cytotoxicity of the purified anti-CD20 antibody obtained in Example 2, the ADCC activity was measured according to the following method.

1.效應細胞溶液的製備1. Preparation of effector cell solution

將自健康捐贈者的人類週邊血液(100mL)加至含有肝素鈉的VACUTAINER®試管。將全血樣本與RPMI 1640無血清(SF)培養基以1:1稀釋並輕輕混合。使用Ficoll-Paque PLUS,藉由緩慢地將24mL經稀釋的血液添加到Ficoll-Paque,並在25℃下以400 x g離心32分鐘,以分離單核細胞。將血沉棕黃層(buffy coat)充分地分配到兩個含有20mL RPMI 1640培養基的50mL離心管中,然後混合2次。接著,將混合物在25℃下,以1,200rpm離心12分鐘,獲得上清液。將RPMI 1640 SF培養基(13mL)添加至上清液,以重新懸浮PBMC細胞。將細胞在25℃下,以1,200rpm離心12分鐘,獲得上清液。將RPMI培養基(10mL)加至上清液中,以重新懸浮PBMC細胞。將充足體積的PBMC細胞懸浮液添加至75T燒瓶,且每個燒瓶約15mL的細胞,其最終細胞密度為1.5 x 106細胞/mL。將IL-2(2.5μg/mL)以最終濃度為3ng/mL加到所有燒瓶中。將PBMC細胞培養於37℃,5% CO2的培養箱中18小時。收集經IL-2所刺激的PBMC細胞,並在25℃下,以1,200rpm離心12分鐘,然 後丟棄上清液。加入PBS(10mL),並與細胞混合。將細胞在25℃下以1,200rpm離心12分鐘,以去除上清液。將細胞以RPMI AM重新懸浮,且將最終濃度調整為2×107細胞/mL。 Add human peripheral blood (100 mL) from healthy donors to a VACUTAINER® test tube containing heparin sodium. The whole blood sample was diluted 1:1 with RPMI 1640 serum-free (SF) medium and mixed gently. Using Ficoll-Paque PLUS, monocytes were isolated by slowly adding 24 mL of diluted blood to Ficoll-Paque and centrifuging at 400 xg for 32 minutes at 25°C. The buffy coat was fully dispensed into two 50 mL centrifuge tubes containing 20 mL RPMI 1640 medium, and then mixed twice. Next, the mixture was centrifuged at 1,200 rpm for 12 minutes at 25°C to obtain a supernatant. RPMI 1640 SF medium (13 mL) was added to the supernatant to resuspend PBMC cells. The cells were centrifuged at 1,200 rpm for 12 minutes at 25°C to obtain a supernatant. RPMI medium (10 mL) was added to the supernatant to resuspend PBMC cells. A sufficient volume of PBMC cell suspension was added to the 75T flask, and each flask had approximately 15 mL of cells, and its final cell density was 1.5 x 106 cells/mL. IL-2 (2.5 μg/mL) was added to all flasks at a final concentration of 3 ng/mL. PBMC cells were cultured in a 37°C, 5% CO 2 incubator for 18 hours. PBMC cells stimulated with IL-2 were collected and centrifuged at 1,200 rpm for 12 minutes at 25°C, and then the supernatant was discarded. PBS (10 mL) was added and mixed with the cells. The cells were centrifuged at 1,200 rpm for 12 minutes at 25°C to remove the supernatant. The cells were resuspended at RPMI AM, and the final concentration was adjusted to 2×10 7 cells/mL.

2.目標細胞溶液的製備2. Preparation of target cell solution

將來自75T燒瓶的細胞懸浮液以1,000rpm離心5分鐘,去除上清液,然後以10mL 1X PBS清洗。將經清洗的細胞以1,200rpm離心5分鐘,去除上清液。以RPMI分析培養基,將細胞重新懸浮,以製備5 x 105細胞/mL的目標細胞溶液。將目標細胞溶液(40μL的5 x 105細胞/mL)加至V底的96孔細胞培養盤的孔洞中。接著,分別將20μL的經製備的市售RITUXAN®溶液(MABTHERA®)(25-0.0025μg/mL)(陽性對照組)、去岩藻醣基化抗體(R1細胞株)溶液(25-0.0025μg/mL)、或RPMI分析培養基(陰性對照組)加至孔洞中,且與目標細胞溶液混合。將V底的96孔細胞培養盤培養於37℃,5% CO2的培養箱中30至60分鐘。 The cell suspension from the 75T flask was centrifuged at 1,000 rpm for 5 minutes, the supernatant was removed, and then washed with 10 mL of 1X PBS. The washed cells were centrifuged at 1,200 rpm for 5 minutes, and the supernatant was removed. In RPMI assay medium, the cells were resuspended to prepare a target cell solution was 5 x 10 5 cells / mL. The target cell solution (40 L of 5 x 10 5 cells / mL) were added to a V-bottom 96-well culture plate holes cells. Next, 20 μL of the prepared commercially available RITUXAN® solution (MABTHERA®) (25-0.0025 μg/mL) (positive control group) and defucosylated antibody (R1 cell line) solution (25-0.0025 μg) /mL), or RPMI analysis medium (negative control group) was added to the hole, and mixed with the target cell solution. The V-bottom 96-well cell culture plate was cultured in a 37°C, 5% CO 2 incubator for 30 to 60 minutes.

3. ADCC活性分析3. ADCC activity analysis

將效應細胞溶液(40μL的8 x 105效應細胞/孔洞)或40μL的RPMI分析培養基加至盤中,以與目標細胞溶液反應。將盤子以300 x g離心4分鐘。將盤子於37℃,5% CO2反應4小時。在收集上清液前,將CYTOTOX 96®的裂解溶液(10μL)添加至Tmax與BlkV組的盤中,反應1小時。將V底的96孔細胞培養盤以300 x g離心4分鐘,且將50μL的上清液從96孔細胞培養盤移至平底分析盤的孔洞中。 The effector cell solution (40 L of 8 x 10 5 effector cells / hole) or 40 L of RPMI was added to the culture dish analysis, to the reaction solution to a target cell. Centrifuge the plate at 300 xg for 4 minutes. The plate was reacted at 37°C with 5% CO 2 for 4 hours. Before collecting the supernatant, the lysis solution (10 μL) of CYTOTOX 96® was added to the trays of the Tmax and BlkV group, and reacted for 1 hour. The V-bottom 96-well cell culture dish was centrifuged at 300 xg for 4 minutes, and 50 μL of supernatant was transferred from the 96-well cell culture dish to the hole of the flat-bottom analysis dish.

將乳酸脫氫酶(lactate dehydrogenase,LDH)(2μL)加至10mL之LDH陽性對照稀釋劑中,以製備LDH陽性對照溶液。將製備的LDH陽性 對照組溶液(50μL)添加至96孔平底分析盤的孔洞中。 Lactate dehydrogenase (LDH) (2 μL) was added to 10 mL of LDH positive control diluent to prepare LDH positive control solution. The prepared LDH positive control solution (50 [mu]L) was added to the hole of the 96-well flat bottom analysis dish.

將LDH重建基質混合物(50μL)添加至分析盤的每個測試孔洞。將盤子覆蓋,並於室溫下於黑暗中培養30分鐘。將終止溶液(50μL)添加至盤子之每個測試孔洞。在加入終止溶液後,立即記錄波長490nm的吸光值。使用各組(S、PBMC、T、E和Tmax)空白移除吸光值,以下列算式計算ADCC活性。 LDH reconstituted matrix mixture (50 μL) was added to each test hole of the analytical disk. Cover the plate and incubate in the dark at room temperature for 30 minutes. Stop solution (50 μL) was added to each test hole of the plate. Immediately after adding the stop solution, the absorbance at a wavelength of 490 nm was recorded. Using each group (S, PBMC, T, E, and Tmax) blank removal absorbance values, ADCC activity was calculated according to the following formula.

Figure 107130630-A0202-12-0039-1
Figure 107130630-A0202-12-0039-1

其中S為樣本(目標細胞+PBMC+抗-CD20抗體)的LDH釋放的吸光值;PBMC為目標細胞及PBMC的LDH釋放的吸光值;E為PBMC的LDH釋放的吸光值;T為目標細胞的自發性LDH釋放的吸光值;且Tmax為目標細胞的最大LDH釋放的吸光值。 Where S is the absorbance value of the LDH released by the sample (target cell + PBMC + anti-CD20 antibody); PBMC is the absorbance value of the LDH released by the target cell and PBMC; E is the absorbance value of the LDH released by the PBMC; T is the spontaneity of the target cell The absorbance value released by sexual LDH; and Tmax is the absorbance value of the maximum LDH released by the target cell.

相較於市售RITUXAN®(MABTHERA®),去岩藻醣基化抗-CD20抗體(R1細胞株)在來自捐贈者1(第3a圖)及捐贈者2(第3b圖)兩者的PBMC細胞中誘導顯著地更強且更高的ADCC反應。 Compared to the commercially available RITUXAN® (MABTHERA®), the defucosylated anti-CD20 antibody (R1 cell line) was found in PBMC from both donor 1 ( Figure 3a ) and donor 2 ( Figure 3b ) A significantly stronger and higher ADCC response is induced in the cells.

表7所示,來自RC79F83MR1細胞株之去岩藻醣基化抗-CD20抗體的EC50顯著低於市售RITUXAN®,其為岩藻醣基化抗-CD20抗體。特別是,去岩藻醣基化抗-CD20抗體(R1細胞株)在來自捐贈者1和2之PBMC細胞中,分別具有為1.7ng/mL與4.6ng/mL的EC50。相較之下,岩藻醣基化抗-CD20抗體(MABTHERA®)在來自捐贈者1和2之PBMC細胞中分別具有18.2ng/mL與35.0ng/mL的EC50As shown in Table 7 , the EC 50 of the defucosylated anti-CD20 antibody from the RC79F83MR1 cell line was significantly lower than that of the commercially available RITUXAN®, which is a fucosylated anti-CD20 antibody. In particular, the defucosylated anti-CD20 antibody (R1 cell line) had an EC 50 of 1.7 ng/mL and 4.6 ng/mL in PBMC cells from donors 1 and 2, respectively. In contrast, fucosylated anti -CD20 antibody (MABTHERA®) each having EC 18.2ng / mL and 35.0ng / mL in the 50 PBMC cells from donor 1 and 2 of the.

此研究的這些結果證明去岩藻醣基化抗-CD20抗體(R1細胞株)展現的ADCC活性為岩藻醣基化抗-CD20抗體(MABTHERA®)的7.68至10.7倍。 These results of this study demonstrate that the defucosylated anti-CD20 antibody (R1 cell line) exhibited ADCC activity 7.68 to 10.7 times that of the fucosylated anti-CD20 antibody (MABTHERA®).

【實施例4】[Example 4] 去岩藻醣基化抗體的結合親和性Binding affinity of defucosylated antibodies

使用偶聯至具有胺偶聯套組之BIACORE®CM5晶片的抗-組胺酸(抗-His)抗體和BIACORE®X100控制軟體的固定精靈(immobilization wizard),評估岩藻醣基化及去岩藻醣基化抗-CD20抗體對於His標記之FcγRIIIa重組蛋白的結合親和性。 Use an immobilization wizard coupled to the anti-histidine (anti-His) antibody of BIACORE® CM5 chip with amine coupling kit and BIACORE® X100 control software to evaluate fucosylation and de-rocking Binding affinity of fucosylated anti-CD20 antibody to His-tagged FcγRIIIa recombinant protein.

以10μL/分鐘的速率,將His標記之FcγRIIIa重組蛋白(1μg/mL),注入至抗-His抗體固定之CM5晶片上,持續20秒。 At a rate of 10 μL/min, His-labeled FcγRIIIa recombinant protein (1 μg/mL) was injected onto the anti-His antibody-immobilized CM5 wafer for 20 seconds.

將來自細胞株1的去岩藻醣基化抗-CD20抗體(5、10、20、40及80nM)、市售岩藻醣基化抗-CD20抗體RITUXAN®(MABTHERA®)(20、40、80、160及320nM)和市售去岩藻醣基化抗-CD20抗體GAZYVA®(obinutuzumab)(5、10、20、40或80nM)分別以30μL/分鐘的流速注射通過晶片,持續3分鐘。電泳緩衝液以30μL/分鐘的流速流經晶片5分鐘。將甘胺酸、pH 1.5(10mM)以30μL/分鐘的流速注射至晶片60秒。 Defucosylated anti-CD20 antibodies (5, 10, 20, 40, and 80 nM) from cell line 1 and commercially available fucosylated anti-CD20 antibodies RITUXAN® (MABTHERA®) (20, 40, 80, 160, and 320 nM) and commercially available defucosylated anti-CD20 antibody GAZYVA® (obinutuzumab) (5, 10, 20, 40, or 80 nM) were injected through the wafer at a flow rate of 30 μL/min for 3 minutes, respectively. The electrophoresis buffer flows through the wafer at a flow rate of 30 μL/min for 5 minutes. Glycine, pH 1.5 (10 mM) was injected into the wafer at a flow rate of 30 μL/min for 60 seconds.

利用BIACORE® X100評估軟體,來分析各循環的應感圖譜,以獲得平衡解離常數(KD)、結合速率常數(Ka)和解離速率常數(Kd)的值。每個循環的應感圖譜以1:1 Langmuir結合模型適配。若Chi2值低於1/10X Rmax值,則適配模型適當且動力學的結合參數是可信的。 Use BIACORE® X100 evaluation software to analyze the stress map of each cycle to obtain the values of equilibrium dissociation constant (K D ), binding rate constant (Ka) and dissociation rate constant (Kd). The stress map of each cycle is adapted with 1:1 Langmuir combined with the model. If the Chi 2 value is lower than 1/10X Rmax value, then the appropriate adaptation model and kinetic binding parameters are credible.

第4a至4c圖顯示經測試之三種抗體的典型SPR應感圖譜。典型的SPR應感圖譜顯示,此分析所使用的條件(例如,結合時間、解離時間和抗體濃度範圍)是適當的。此外,三種抗體之Chi2值低於1/10X Rmax值,表示1:1 Langmuir模型適合作為所有三種抗體的應感圖譜適配。 Figures 4a to 4c show typical SPR response patterns of the three antibodies tested. A typical SPR response profile shows that the conditions used for this analysis (eg, binding time, dissociation time, and antibody concentration range) are appropriate. In addition, the Chi 2 value of the three antibodies is lower than 1/10X Rmax value, indicating that the 1:1 Langmuir model is suitable for the adaptation of the response map of all three antibodies.

表8所示,去岩藻醣基化抗-CD20抗體(R1細胞株)對FcγRIIIa的結合親和性比MABTHERA®強10倍(R1細胞株的KD=13.0nM,MABTHERA®=151.5nM)。此外,去岩藻醣基化抗-CD20抗體(R1細胞株)對FcγRIIIa的結合親和性比GAZYVA®強3倍(R1細胞株的KD=13.0nM,GAZYVA®=39.9nM)。 As shown in Table 8, to fucosylated anti -CD20 antibody (R1 cell line) binding affinity for the FcγRIIIa 10 times stronger than MABTHERA® (K D R1 cell line = 13.0nM, MABTHERA® = 151.5nM) . In addition, the defucosylated anti-CD20 antibody (R1 cell line) has a binding affinity to FcγRIIIa that is three times stronger than GAZYVA® (K D = 13.0 nM for R1 cell line, GAZYVA® = 39.9 nM).

此研究的這些結果證實,根據本發明製備的去岩藻醣基化抗-CD20抗體(R1細胞株)具有比市售岩藻醣基化抗-CD20抗體RITUXAN®(MABTHERA®)及市售去岩藻醣基化抗-CD20抗體(GAZYVA®)更強的FcγRIIIa結合親和性。 These results of this study confirm that the afucosylated anti-CD20 antibody (R1 cell line) prepared according to the present invention has a higher price than the commercially available fucosylated anti-CD20 antibody RITUXAN® (MABTHERA®) and the commercially available Fucosylated anti-CD20 antibody (GAZYVA®) has stronger FcγRIIIa binding affinity.

【實施例5】[Example 5] 去岩藻醣基化抗體的CDC活性CDC activity of defucosylated antibody

評估以本發明方法所產生之去岩藻醣基化抗體的CDC活性。 The CDC activity of the defucosylated antibody produced by the method of the present invention is evaluated.

以RPMI培養基培養Daudi細胞,且當細胞密度達到1×106細胞/mL時,進行繼代培養(繼代培養密度:2-3 x 105細胞/mL)。收集Daudi細胞,並以300rpm離心5分鐘。以RPMI培養基重新懸浮細胞,以製備濃度為1 x 105細胞/mL的細胞懸浮液。在重新懸浮後,將100μL的細胞懸浮液或100μL的RPMI培養基種至白色96孔盤的孔洞中。 Daudi cells were cultured in RPMI medium, and when the cell density reached 1×10 6 cells/mL, subculture was carried out (subculture density: 2-3×10 5 cells/mL). Daudi cells were collected and centrifuged at 300 rpm for 5 minutes. Cells were resuspended in RPMI medium to prepare a concentration of 1 x 10 5 cells / mL cell suspension. After resuspension, seed 100 μL of cell suspension or 100 μL of RPMI medium into the holes of a white 96-well dish.

以120μg/mL至0.234μg/mL的濃度,將市售可得RITUXAN®(MABTHERA®)與去岩藻醣基化抗-CD20抗體(R1細胞株)製備於生理食鹽水中。接著,將濃度為120μg/mL至0.234μg/mL之25μL的RITUXAN®(MABTHERA®)或去岩藻醣基化抗-CD20抗體(R1細胞株)添加至含有Daudi細胞或RPMI培養基的白色96孔盤的孔洞中。將CELLTITER-GLO®試劑(20μL)加至每個孔洞中,接著混合。將盤置於微量盤震盪器上,以750rpm震盪2分鐘,接著在黑暗中於室溫培養10分鐘。藉由插有高靈敏度螢光盒的多模式讀取器,來偵測發光強度(積分時間:1秒),以計算抗-CD20抗體的EC50值及抗體的相關CDC活性。 The commercially available RITUXAN® (MABTHERA®) and defucosylated anti-CD20 antibody (R1 cell line) were prepared in physiological saline at a concentration of 120 μg/mL to 0.234 μg/mL. Next, 25 μL of RITUXAN® (MABTHERA®) or defucosylated anti-CD20 antibody (R1 cell line) at a concentration of 120 μg/mL to 0.234 μg/mL was added to white 96 wells containing Daudi cells or RPMI medium Holes in the plate. Add CELLTITER-GLO® reagent (20 μL) to each well and then mix. The plate was placed on a microplate shaker and shaken at 750 rpm for 2 minutes, and then incubated in the dark at room temperature for 10 minutes. A multi-mode reader with a high-sensitivity fluorescent box was inserted to detect the luminous intensity (integration time: 1 second) to calculate the EC 50 value of the anti-CD20 antibody and the relevant CDC activity of the antibody.

第5圖顯示去岩藻醣基化抗-CD20抗體(R1細胞株)與RITUXAN®之CDC活性的比較。去岩藻醣基化抗-CD20抗體(R1細胞株)的EC50值為0.682μg/mL,其高於RITUXAN®的EC50值(EC50=0.582μg/mL)。 Figure 5 shows a comparison of CDC activity of defucosylated anti-CD20 antibody (R1 cell line) and RITUXAN®. The EC 50 value of the defucosylated anti-CD20 antibody (R1 cell line) was 0.682 μg/mL, which was higher than the EC 50 value of RITUXAN® (EC 50 = 0.582 μg/mL).

GAZYVA®是一種市售去岩藻醣基化抗-CD20抗體,已顯示能誘導ADCC活性,但抑制CDC活性(E.Mössener et al.(2010);C.Ferrara et al.(2011))。從其它研究獲得的結果顯示為了達到有效率的癌症治療,應該提高GAZYVA®的量。相較之下,本實施例及實施例5的結果證明,藉由本發明方法產生的去岩藻醣基化抗-CD20抗體誘導ADCC活性,並維持類似於其岩藻醣基化抗體的CDC活性。因此,比起GAZYVA®,本發明的去岩藻醣基化抗-CD20抗體表現更佳。 GAZYVA® is a commercially available defucosylated anti-CD20 antibody that has been shown to induce ADCC activity but inhibit CDC activity (E. Mössener et al. (2010); C. Ferrara et al. (2011)). The results obtained from other studies indicate that in order to achieve efficient cancer treatment, the amount of GAZYVA® should be increased. In contrast, the results of this example and Example 5 demonstrate that the defucosylated anti-CD20 antibody produced by the method of the present invention induces ADCC activity and maintains CDC activity similar to its fucosylated antibody . Therefore, the defucosylated anti-CD20 antibody of the present invention performs better than GAZYVA®.

【實施例6】[Example 6] 去岩藻醣基化抗體在動物模型中的功效證據Evidence of the efficacy of defucosylated antibodies in animal models

在此實施例中,使用B細胞淋巴瘤皮下異種移植模型,來證 明本發明的去岩藻醣基化抗體的抗腫瘤功效。SU-DHL-4為B細胞淋巴瘤細胞株,其在細胞膜上表現大量的CD20,且可成長並皮下形成固態腫瘤。因此,開發SCID/Beige小鼠中的異種移植模型,以比較去岩藻醣基化抗體(R1細胞株)與市售可得RITUXAN®(MABTHERA®)的抗腫瘤功效。 In this example, a B-cell lymphoma subcutaneous xenograft model was used to demonstrate the anti-tumor efficacy of the defucosylated antibody of the present invention. SU-DHL-4 is a B-cell lymphoma cell line that displays a large amount of CD20 on the cell membrane and can grow and form solid tumors under the skin. Therefore, a xenograft model in SCID/Beige mice was developed to compare the antitumor efficacy of the defucosylated antibody (R1 cell line) with the commercially available RITUXAN® (MABTHERA®).

以RPMI培養基(CM),將SU-DHL-4細胞培養於燒瓶中。當細胞濃度達0.8-1.0 x 106細胞/mL時,收集細胞懸浮液並以300g離心5分鐘,以去除上清液。以含有一些條件培養基的新培養基(新鮮CM:條件CM=9:1),將細胞重新懸浮。細胞以1:2至1:10的比例(接種的細胞數量:總獲得細胞數量)繼代培養,且細胞濃度為至少1 x 105細胞/mL。將培養盤培養於37℃。SU-DHL-4細胞培養於5個150T的燒瓶。當細胞濃度到達0.8至1.0 x 106細胞/mL時,將細胞懸浮液收集至50mL試管,之後以1,200rpm離心5分鐘,以移除上清液。使用無血清RPMI培養基,將細胞濃度調整至1 x 108細胞/mL。在冰上,使用具18G針頭之預冷針筒,於50mL試管中混合細胞懸浮液與等體積的MATRIGEL®。最終細胞濃度為5 x 107細胞/mL。使用具23G *1”針頭的預冷1mL針筒,將5 x 107細胞/mL的Matrigel-SU-DHL-4細胞混合物(100uL)皮下注射至每隻小鼠(SCID/Beige小鼠)背部區域的右側。總接種細胞數量為5 x 106細胞。每3或4天使用卡尺測量每隻小鼠的腫瘤體積,且以等式:V=0.5 x ab2計算,其中a與b分別代表腫瘤的長度及寬度。 SU-DHL-4 cells were cultured in flasks with RPMI medium (CM). When the cell concentration of 0.8-1.0 x 10 6 cells / mL, the cell suspension was collected and centrifuged at 300g for 5 minutes to remove the supernatant. Resuspend the cells in a new medium containing some conditioned medium (fresh CM: conditioned CM=9:1). Cells at ratio of 1: 2 to 1:10 (number of cells seeded: obtaining the total number of cells) subcultured and the cells at a concentration of at least 1 x 10 5 cells / mL. The culture plate was cultured at 37°C. SU-DHL-4 cells were cultured in 5 150T flasks. When the cell concentration reached 0.8 to 1.0 x 10 6 cells/mL, the cell suspension was collected into a 50 mL test tube, and then centrifuged at 1,200 rpm for 5 minutes to remove the supernatant. Using serum-free RPMI medium, adjust the cell concentration to 1 x 10 8 cells/mL. On ice, use a pre-cooled syringe with an 18G needle and mix the cell suspension with an equal volume of MATRIGEL® in a 50 mL test tube. The final cell concentration is 5 x 107 cells/mL. Using a pre-chilled 1 mL syringe with a 23G *1” needle, 5 x 10 7 cells/mL Matrigel-SU-DHL-4 cell mixture (100 uL) was injected subcutaneously into the back of each mouse (SCID/Beige mice) The right side of the area. The total number of inoculated cells is 5 x 10 6 cells. The tumor volume of each mouse is measured using calipers every 3 or 4 days, and is calculated by the equation: V=0.5 x ab 2 , where a and b represent respectively The length and width of the tumor.

當腫瘤體積達約200mm3(198.25±55.53mm3)時,其在接種腫瘤後約20天發生,將小鼠分成三組,每組5隻,之後以食鹽水(載體),市售RITUXAN®(MABTHERA)或無岩藻醣基化抗-CD20抗體(R1細胞株)治療。小鼠每週注射0.2mL的0.1mg/mL抗體,共3週。每週兩次,以電子秤和 數位卡尺測量所有小鼠的體重與腫瘤大小。於治療期結束時,將小鼠犧牲,取出腫瘤組織,並秤重。接著,使用10%福爾馬林緩衝液,於室溫下固定腫瘤組織,供後續試驗。 When the tumor volume reached about 200 mm 3 (198.25±55.53 mm 3 ), it occurred about 20 days after tumor inoculation, and the mice were divided into three groups of 5 mice each, followed by saline (vehicle), commercially available RITUXAN® (MABTHERA) or afucosylated anti-CD20 antibody (R1 cell line) treatment. Mice were injected with 0.2 mL of 0.1 mg/mL antibody weekly for 3 weeks. Twice a week, the weight and tumor size of all mice were measured with electronic scales and digital calipers. At the end of the treatment period, the mice were sacrificed, tumor tissue was removed, and weighed. Then, 10% formalin buffer was used to fix the tumor tissue at room temperature for subsequent experiments.

第6圖所示,去岩藻醣基化抗-CD20抗體(R1細胞株)顯示比RITUXAN®顯著更強的抗-腫瘤功效。在載體組與去岩藻醣基化抗-CD20抗體(R1細胞株)治療組之間,腫瘤體積具有統計上的顯著差異(P<0.001,以學生t檢定)。相對地,相較於僅載體組,RITUXAN®在腫瘤體積上不具有統計上顯著差異。在1mg/kg劑量下,去岩藻醣基化抗-CD20抗體(R1細胞株)比起RITUXAN®更有效地抑制腫瘤生長(R1細胞株=468±148mm3;RITUXAN®=1407±241mm3),如表9中所示。去岩藻醣基化抗-CD20抗體(R1細胞株)與RITUXAN®組之間在腫瘤體積上具有統計上顯著差異(P<0.001)。 As shown in Figure 6 , the defucosylated anti-CD20 antibody (R1 cell line) showed significantly stronger anti-tumor efficacy than RITUXAN®. There was a statistically significant difference in tumor volume between the vehicle group and the defucosylated anti-CD20 antibody (R1 cell line) treatment group (P<0.001, tested by Student's t). In contrast, RITUXAN® did not have statistically significant differences in tumor volume compared to the vehicle-only group. At a dose of 1 mg/kg, defucosylated anti-CD20 antibody (R1 cell line) inhibited tumor growth more effectively than RITUXAN® (R1 cell line=468±148mm 3 ; RITUXAN®=1407±241mm 3 ) , As shown in Table 9 . There was a statistically significant difference in tumor volume between the defucosylated anti-CD20 antibody (R1 cell line) and the RITUXAN® group (P<0.001).

去岩藻醣基化抗-CD20抗體(R1細胞株)治療組的腫瘤重量顯著小於僅載體組(P<0.001)的腫瘤重量,如第7圖中所示。然而,在相同劑量下,相較於載體組,RITUXAN®在腫瘤重量上並未顯示統計上顯著差異。因此,相較於RITUXAN®組,在腫瘤重量方面,去岩藻醣基化抗-CD20抗體(R1細胞株)顯著降低(R1細胞株=0.27±0.15g;RITUXAN®=0.62±0.09g,P<0.01),如表9所示。去岩藻醣基化抗-CD20抗體(R1細胞株)比起RITUXAN®更有效地抑制腫瘤生長,其亦可對應於腫瘤體積的結果。 The tumor weight of the defucosylated anti-CD20 antibody (R1 cell line) treatment group was significantly smaller than that of the vehicle-only group (P<0.001), as shown in Figure 7 . However, at the same dose, RITUXAN® did not show a statistically significant difference in tumor weight compared to the vehicle group. Therefore, compared with the RITUXAN® group, in terms of tumor weight, the defucosylated anti-CD20 antibody (R1 cell line) was significantly reduced (R1 cell line=0.27±0.15g; RITUXAN®=0.62±0.09g, P <0.01), as shown in Table 9 . Defucosylated anti-CD20 antibody (R1 cell line) inhibits tumor growth more effectively than RITUXAN®, which can also correspond to the results of tumor volume.

在治療期間,所有組的小鼠體重逐漸增加,如第8圖所示。 During the treatment period, the weight of mice in all groups gradually increased, as shown in Figure 8 .

這些研究的結果顯示去岩藻醣基化抗-CD20抗體(R1細胞株)是安全的。 The results of these studies indicate that the defucosylated anti-CD20 antibody (R1 cell line) is safe.

Figure 107130630-A0202-12-0045-2
Figure 107130630-A0202-12-0045-2

Figure 107130630-A0202-12-0046-3
Figure 107130630-A0202-12-0046-3

Figure 107130630-A0202-12-0047-4
Figure 107130630-A0202-12-0047-4

Figure 107130630-A0202-12-0048-5
Figure 107130630-A0202-12-0048-5

Figure 107130630-A0202-12-0049-6
Figure 107130630-A0202-12-0049-6

Figure 107130630-A0202-12-0050-7
Figure 107130630-A0202-12-0050-7

Figure 107130630-A0202-12-0050-8
Figure 107130630-A0202-12-0050-8

Figure 107130630-A0202-12-0051-9
Figure 107130630-A0202-12-0051-9

Figure 107130630-A0202-12-0051-10
Figure 107130630-A0202-12-0051-10

<110> 聯亞藥業聯合生物製藥彭文君 陳惠蓉 <110> Lianya Pharmaceutical United Biopharma Peng Wenjun Chen Huirong

<120> 去岩藻醣基化抗體的製造方法 <120> Method for producing defucosylated antibody

<130> 2018.XX.XX <130> 2018.XX.XX

<160> 17 <160> 17

<170> PatentIn version 3.5 <170> PatentIn version 3.5

<210> 1 <210> 1

<211> 1728 <211> 1728

<212> DNA <212> DNA

<213> 小鼠 <213> mouse

<400> 1

Figure 107130630-A0202-12-0052-11
Figure 107130630-A0202-12-0053-12
<400> 1
Figure 107130630-A0202-12-0052-11
Figure 107130630-A0202-12-0053-12

<210> 2 <210> 2

<211> 575 <211> 575

<212> PRT <212> PRT

<213> 人類 <213> Human

<400> 2

Figure 107130630-A0202-12-0053-13
Figure 107130630-A0202-12-0054-14
Figure 107130630-A0202-12-0055-15
Figure 107130630-A0202-12-0056-16
Figure 107130630-A0202-12-0057-17
<400> 2
Figure 107130630-A0202-12-0053-13
Figure 107130630-A0202-12-0054-14
Figure 107130630-A0202-12-0055-15
Figure 107130630-A0202-12-0056-16
Figure 107130630-A0202-12-0057-17

<210> 3 <210> 3

<211> 1728 <211> 1728

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F83M經修飾之酵素的核酸序列,野生型FUT8基因第1093-1095、1225-1227與1357-1359位置被改變 <223> Nucleic acid sequence of F83M modified enzyme, positions 1093-1095, 1225-1227 and 1357-1359 of wild-type FUT8 gene were changed

<400> 3

Figure 107130630-A0202-12-0057-18
Figure 107130630-A0202-12-0058-19
<400> 3
Figure 107130630-A0202-12-0057-18
Figure 107130630-A0202-12-0058-19

<210> 4 <210> 4

<211> 575 <211> 575

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F83M經修飾之酵素的胺基酸序列,野生型FUT8蛋白第365、409與453位置的殘基被改變 <223> Amino acid sequence of F83M modified enzyme, residues at positions 365, 409 and 453 of wild-type FUT8 protein were changed

<400> 4

Figure 107130630-A0202-12-0059-20
Figure 107130630-A0202-12-0060-21
Figure 107130630-A0202-12-0061-22
Figure 107130630-A0202-12-0062-23
<400> 4
Figure 107130630-A0202-12-0059-20
Figure 107130630-A0202-12-0060-21
Figure 107130630-A0202-12-0061-22
Figure 107130630-A0202-12-0062-23

<210> 5 <210> 5

<211> 1728 <211> 1728

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8M1經修飾之酵素的核酸序列,野生型FUT8基因第1105-1107位置被改變 <223> Nucleic acid sequence of F8M1 modified enzyme, position 1105-1107 of wild-type FUT8 gene was changed

<400> 5

Figure 107130630-A0202-12-0062-24
Figure 107130630-A0202-12-0063-25
<400> 5
Figure 107130630-A0202-12-0062-24
Figure 107130630-A0202-12-0063-25

<210> 6 <210> 6

<211> 575 <211> 575

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8M1經修飾之酵素的胺基酸序列,野生型FUT8蛋白第369位置的殘基被改變 <223> The amino acid sequence of the modified enzyme of F8M1, the residue at position 369 of the wild-type FUT8 protein was changed

<400> 6

Figure 107130630-A0202-12-0064-26
Figure 107130630-A0202-12-0065-27
Figure 107130630-A0202-12-0066-28
Figure 107130630-A0202-12-0067-29
<400> 6
Figure 107130630-A0202-12-0064-26
Figure 107130630-A0202-12-0065-27
Figure 107130630-A0202-12-0066-28
Figure 107130630-A0202-12-0067-29

<210> 7 <210> 7

<211> 1728 <211> 1728

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8M2經修飾之酵素的核酸序列,野生型FUT8基因第1225-1227位置被改變 <223> Nucleic acid sequence of F8M2 modified enzyme, position 1225-1227 of wild type FUT8 gene was changed

<400> 7

Figure 107130630-A0202-12-0067-30
Figure 107130630-A0202-12-0068-31
<400> 7
Figure 107130630-A0202-12-0067-30
Figure 107130630-A0202-12-0068-31

<210> 8 <210> 8

<211> 575 <211> 575

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8M2經修飾之酵素的胺基酸序列,野生型FUT8蛋白第409位置的殘基被改變 <223> The amino acid sequence of the modified enzyme of F8M2, the residue at position 409 of the wild-type FUT8 protein was changed

<400> 8

Figure 107130630-A0202-12-0069-32
Figure 107130630-A0202-12-0070-33
Figure 107130630-A0202-12-0071-34
Figure 107130630-A0202-12-0072-35
<400> 8
Figure 107130630-A0202-12-0069-32
Figure 107130630-A0202-12-0070-33
Figure 107130630-A0202-12-0071-34
Figure 107130630-A0202-12-0072-35

<210> 9 <210> 9

<211> 1728 <211> 1728

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8M3經修飾之酵素的核酸序列,野生型FUT8基因第1405-1407位置被改變 <223> Nucleic acid sequence of F8M3 modified enzyme, position 1405-1407 of wild-type FUT8 gene was changed

<400> 9

Figure 107130630-A0202-12-0072-36
Figure 107130630-A0202-12-0073-37
<400> 9
Figure 107130630-A0202-12-0072-36
Figure 107130630-A0202-12-0073-37

<210> 10 <210> 10

<211> 575 <211> 575

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8M3經修飾之酵素的胺基酸序列,野生型FUT8蛋白第469位置的殘基被改變 <223> The amino acid sequence of the modified enzyme of F8M3, the residue at position 469 of the wild-type FUT8 protein was changed

<400> 10

Figure 107130630-A0202-12-0074-38
Figure 107130630-A0202-12-0075-39
Figure 107130630-A0202-12-0076-40
Figure 107130630-A0202-12-0077-41
<400> 10
Figure 107130630-A0202-12-0074-38
Figure 107130630-A0202-12-0075-39
Figure 107130630-A0202-12-0076-40
Figure 107130630-A0202-12-0077-41

<210> 11 <210> 11

<211> 1665 <211> 1665

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8D1經修飾之酵素的核酸序列,野生型FUT8基因第1087-1149位置被刪除 <223> Nucleic acid sequence of F8D1 modified enzyme, position 1087-1149 of wild-type FUT8 gene was deleted

<400> 11

Figure 107130630-A0202-12-0077-42
Figure 107130630-A0202-12-0078-43
<400> 11
Figure 107130630-A0202-12-0077-42
Figure 107130630-A0202-12-0078-43

<210> 12 <210> 12

<211> 548 <211> 548

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8D1經修飾之酵素的胺基酸序列,野生型FUT8蛋白第365-386位置的殘基被刪除 <223> Amino acid sequence of F8D1 modified enzyme, residues at positions 365-386 of wild-type FUT8 protein are deleted

<400> 12

Figure 107130630-A0202-12-0079-44
Figure 107130630-A0202-12-0080-45
Figure 107130630-A0202-12-0081-46
Figure 107130630-A0202-12-0082-47
<400> 12
Figure 107130630-A0202-12-0079-44
Figure 107130630-A0202-12-0080-45
Figure 107130630-A0202-12-0081-46
Figure 107130630-A0202-12-0082-47

<210> 13 <210> 13

<211> 1119 <211> 1119

<212> DNA <212> DNA

<213> 小鼠 <213> mouse

<400> 13

Figure 107130630-A0202-12-0082-48
Figure 107130630-A0202-12-0083-49
<400> 13
Figure 107130630-A0202-12-0082-48
Figure 107130630-A0202-12-0083-49

<210> 14 <210> 14

<211> 372 <211> 372

<212> PRT <212> PRT

<213> 小鼠 <213> mouse

<400> 14

Figure 107130630-A0202-12-0083-50
Figure 107130630-A0202-12-0084-51
Figure 107130630-A0202-12-0085-52
<400> 14
Figure 107130630-A0202-12-0083-50
Figure 107130630-A0202-12-0084-51
Figure 107130630-A0202-12-0085-52

<210> 15 <210> 15

<211> 1119 <211> 1119

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> GMD4M經修飾之酵素的核酸序列,野生型GMD基因第463-465、469-471、535-537與547-549位置被改變 <223> Nucleic acid sequence of GMD4M modified enzyme, position 463-465, 469-471, 535-537 and 547-549 of wild-type GMD gene were changed

<400> 15

Figure 107130630-A0202-12-0086-53
<400> 15
Figure 107130630-A0202-12-0086-53

<210> 16 <210> 16

<211> 372 <211> 372

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> GMD4M經修飾之酵素的胺基酸序列,野生型GMD基因第155、157、179與183位置的殘基被改變 <223> Amino acid sequence of GMD4M modified enzyme, residues at positions 155, 157, 179 and 183 of wild-type GMD gene were changed

<400> 16

Figure 107130630-A0202-12-0087-54
Figure 107130630-A0202-12-0088-55
Figure 107130630-A0202-12-0089-56
<400> 16
Figure 107130630-A0202-12-0087-54
Figure 107130630-A0202-12-0088-55
Figure 107130630-A0202-12-0089-56

<210> 17 <210> 17

<211> 63 <211> 63

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> F8D1突變基因中被刪除的核酸序列 <223> Deleted nucleic acid sequence in F8D1 mutant gene

<400> 17

Figure 107130630-A0202-12-0089-57
<400> 17
Figure 107130630-A0202-12-0089-57

Claims (19)

一種在一宿主細胞中產生一去岩藻醣基化蛋白的方法,包括將至少一編碼岩藻醣基化途徑(fucosylation pathway)之經修飾之酵素的核酸導入至該宿主細胞中,以產生該去岩藻醣基化蛋白。 A method for producing a defucosylated protein in a host cell, comprising introducing at least one nucleic acid encoding a modified enzyme of the fucosylation pathway into the host cell to produce the Defucosylated protein. 如申請專利範圍第1項所述之方法,其中該經修飾之酵素源自於GDP-甘露醣4,6-脫水酶(GDP-mannose 4,6-dehydratase,GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(GDP-4-keto-6-deoxy-D-mannose epinierase-reductase,FX)、或岩藻醣轉移酶(fucosyltransferase,FUT)。 The method as described in item 1 of the patent application scope, wherein the modified enzyme is derived from GDP-mannose 4,6-dehydratase (GDP-mannose 4,6-dehydratase, GMD), GDP-4-keto -6-deoxy-D-mannose isomerase-reductase (GDP-4-keto-6-deoxy-D-mannose epinierase-reductase, FX), or fucosyltransferase (FUT). 如申請專利範圍第1項所述之方法,其中該經修飾之酵素源自於岩藻醣轉移酶(FUT)。 The method as described in item 1 of the patent application scope, wherein the modified enzyme is derived from fucose transferase (FUT). 如申請專利範圍第1項所述之方法,其中該經修飾之酵素源自於α-1,6-岩藻醣轉移酶(FUT8)。 The method as described in item 1 of the patent application scope, wherein the modified enzyme is derived from α-1,6-fucose transferase (FUT8). 如申請專利範圍第1項所述之方法,其中該經修飾之酵素於該宿主細胞中降低或抑制該經修飾之酵素所源自的野生型酵素的活性。 The method as described in item 1 of the patent application scope, wherein the modified enzyme reduces or inhibits the activity of the wild-type enzyme from which the modified enzyme is derived in the host cell. 如申請專利範圍第1項所述之方法,其中該經修飾之酵素抑制或降低該宿主細胞中的岩藻醣基化。 The method as described in item 1 of the patent application scope, wherein the modified enzyme inhibits or reduces fucosylation in the host cell. 如申請專利範圍第1項所述之方法,其中該宿主細胞中所產生的該去岩藻醣基化蛋白為一去岩藻醣基化抗體或其片段。 The method according to item 1 of the patent application scope, wherein the defucosylated protein produced in the host cell is a defucosylated antibody or fragment thereof. 如申請專利範圍第7項所述之方法,其中該去岩藻醣基化抗體或其片段至少90%去岩藻醣基化。 The method according to item 7 of the patent application scope, wherein the defucosylated antibody or fragment thereof is at least 90% defucosylated. 如申請專利範圍第7項所述之方法,其中該去岩藻醣基化抗體,相較於其岩藻醣基化抗體,具有增加的抗體依賴性細胞毒性(antibody-dependent cellular cytotoxicity,ADCC)活性。 The method as described in item 7 of the patent application range, wherein the defucosylated antibody has increased antibody-dependent cellular cytotoxicity (ADCC) compared to its fucosylated antibody active. 如申請專利範圍第7項所述之方法,其中該去岩藻醣基化抗體的補體依賴性細胞毒性(complement dependent cytotoxicity,CDC),相較於其岩藻醣基化對應物,未被降低抑制。 The method as described in item 7 of the patent application scope, wherein the complement dependent cytotoxicity (CDC) of the defucosylated antibody is not reduced compared to its fucosylated counterpart inhibition. 一種在一宿主細胞中產生一去岩藻醣基化蛋白的方法,包括將一編碼岩藻醣基化途徑之一經修飾之酵素的第一核酸序列,以及一編碼欲產生的蛋白之第二核酸序列轉染至該宿主細胞中。 A method for producing a defucosylated protein in a host cell, comprising a first nucleic acid sequence encoding a modified enzyme of one of the fucosylation pathways, and a second nucleic acid encoding a protein to be produced The sequence is transfected into the host cell. 如申請專利範圍第11項所述之方法,其中該宿主細胞同時轉染該第一核酸序列與該第二核酸序列。 The method according to item 11 of the patent application scope, wherein the host cell transfects the first nucleic acid sequence and the second nucleic acid sequence simultaneously. 如申請專利範圍第12項所述之方法,其中所產生的該去岩藻醣基化蛋白為一抗體或其片段。 The method as described in item 12 of the patent application scope, wherein the defucosylated protein produced is an antibody or a fragment thereof. 如申請專利範圍第11項所述之方法,其中該第一核酸編碼源自於GDP-甘露醣4,6-脫水酶(GMD)、GDP-4-酮基-6-脫氧-D-甘露醣異構酶-還原酶(FX)、及/或岩藻醣轉移酶(FUT)的一經修飾之酵素。 The method as described in item 11 of the patent application scope, wherein the first nucleic acid encoding is derived from GDP-mannose 4,6-dehydratase (GMD), GDP-4-keto-6-deoxy-D-mannose A modified enzyme of isomerase-reductase (FX) and/or fucose transferase (FUT). 如申請專利範圍第11項所述之方法,其中該第一核酸編碼源自於α-1,6-岩藻醣轉移酶(FUT8)的一經修飾之酵素。 The method according to item 11 of the patent application scope, wherein the first nucleic acid encodes a modified enzyme derived from α-1,6-fucose transferase (FUT8). 如申請專利範圍第11項所述之方法,包括下列步驟:a)將編碼該經修飾之酵素的該第一核酸轉染至該宿主細胞;b)篩選及分離低岩藻醣基化的轉染細胞;c)將編碼該欲產生之蛋白的該第二核酸轉染至該低岩藻醣基化細胞;d)在該低岩藻醣基化細胞中表現該欲產生之蛋白。 The method as described in item 11 of the patent application scope includes the following steps: a) transfecting the first nucleic acid encoding the modified enzyme into the host cell; b) screening and isolating the low-fucosylated transfection Staining the cell; c) transfecting the second nucleic acid encoding the protein to be produced into the low-fucosylated cell; d) expressing the protein to be produced in the low-fucosylated cell. 如申請專利範圍第16項所述之方法,其中所產生的該去岩藻醣基化蛋白為一抗體及/或其片段。 The method according to item 16 of the patent application scope, wherein the defucosylated protein produced is an antibody and/or a fragment thereof. 如申請專利範圍第11項所述之方法,包括以下步驟:a)將編碼該所欲產生之蛋白的該第二核酸轉染至該宿主細胞;b)篩選及分離經轉染該第二核酸的細胞;c)將編碼該經修飾之酵素之該第一核酸轉染至於步驟(b)中的該細胞;d)篩選及分離低岩藻醣基化的轉染細胞;e)在該低岩藻醣基化細胞中表現該欲產生之蛋白。 The method as described in item 11 of the patent application scope includes the following steps: a) transfecting the second nucleic acid encoding the desired protein into the host cell; b) screening and isolating the transfected second nucleic acid Cells; c) transfect the first nucleic acid encoding the modified enzyme to the cells in step (b); d) select and isolate low-fucosylated transfected cells; e) at the low The fucosylated cells express the protein to be produced. 如申請專利範圍第18項所述之方法,其中該所產生之去岩藻醣基化蛋白為一抗體及/或其片段。 The method as described in item 18 of the patent application scope, wherein the produced defucosylated protein is an antibody and/or a fragment thereof.
TW107130630A 2018-08-31 2018-08-31 Manufacture method of afucosylated antibodies TWI748124B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW107130630A TWI748124B (en) 2018-08-31 2018-08-31 Manufacture method of afucosylated antibodies

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW107130630A TWI748124B (en) 2018-08-31 2018-08-31 Manufacture method of afucosylated antibodies

Publications (2)

Publication Number Publication Date
TW202010844A true TW202010844A (en) 2020-03-16
TWI748124B TWI748124B (en) 2021-12-01

Family

ID=70766697

Family Applications (1)

Application Number Title Priority Date Filing Date
TW107130630A TWI748124B (en) 2018-08-31 2018-08-31 Manufacture method of afucosylated antibodies

Country Status (1)

Country Link
TW (1) TWI748124B (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2409712A1 (en) * 2010-07-19 2012-01-25 International-Drug-Development-Biotech Anti-CD19 antibody having ADCC and CDC functions and improved glycosylation profile

Also Published As

Publication number Publication date
TWI748124B (en) 2021-12-01

Similar Documents

Publication Publication Date Title
US9751942B2 (en) Anti-LAMP5 antibody and utilization thereof
EP2595662B1 (en) Anti-cd19 antibody having adcc and cdc functions and improved glycosylation profile
EP2596024B1 (en) Anti-cd19 antibody having adcc function with improved glycosylation profile
US10308970B2 (en) Methods for controlling fucosylation levels in proteins
JP2009114201A (en) Antibody glycosylation variant having increased antibody-dependent cellular cytotoxicity
KR20090077911A (en) Use of human cells of myeloid leukaemia origin for expression of antibodies
AU2018438767B9 (en) Afucosylated antibodies and manufacture thereof
Wang et al. The interplay of protein engineering and glycoengineering to fine‐tune antibody glycosylation and its impact on effector functions
KR102651432B1 (en) Afucosylated antibodies and methods for their preparation
TWI748124B (en) Manufacture method of afucosylated antibodies
TWI745615B (en) Afucosylated antibodies and cell lines expressing the afucosylated antibodies