TWI748124B - Manufacture method of afucosylated antibodies - Google Patents

Manufacture method of afucosylated antibodies Download PDF

Info

Publication number
TWI748124B
TWI748124B TW107130630A TW107130630A TWI748124B TW I748124 B TWI748124 B TW I748124B TW 107130630 A TW107130630 A TW 107130630A TW 107130630 A TW107130630 A TW 107130630A TW I748124 B TWI748124 B TW I748124B
Authority
TW
Taiwan
Prior art keywords
antibody
cells
protein
cell
nucleic acid
Prior art date
Application number
TW107130630A
Other languages
Chinese (zh)
Other versions
TW202010844A (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

Landscapes

  • Preparation Of Compounds By Using Micro-Organisms (AREA)

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 functional 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 functions of humans, including catalysis, signal transduction, message transmission between cells, and molecular recognition and binding. Many glycoproteins have been used for therapeutic purposes, and in the past two decades, the recombinant form of naturally occurring secreted glycoproteins has been the main product of the biotechnology industry. Examples include erythropoietin (EPO), therapeutic monoclonal antibody (therapeutic monoclonal antibody, therapeutic mAb), tissue plasminogen activator (tPA), interferon-β (interferon-β) , IFN-β), granulocyte-macrophage colony stimulating factor (granulocyte-macrophage colony stimulating factor, GM-CSF) and human chorionic gonadotrophin (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 in the diagnosis, prevention and treatment of various human diseases due to their long half-life in the 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 the effector function of IgG antibodies, and IgG1 subtypes have been reported. The class of antibodies has 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 antibody requires antibody Fc region and effector cells, such as killer cells, natural killer cells, activated macrophages or similar cells and antibody receptors 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 antibody hinge region and C region (hereinafter referred to as "Cγ2 domain"), and sugar chains 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。 Reducing or inhibiting the fucosylation of the N-polysaccharide of the antibody or Fc fusion protein can enhance ADCC activity. ADCC generally participates in the activation of natural killer (NK) cells and relies on the recognition of antibody-coated cells by Fc receptors on the surface of NK cells. The binding of the Fc region to the Fc receptor on NK cells will be affected by the glycosylation status of the Fc region. In addition, the type of N-polysaccharide in the Fc region also affects ADCC activity. Therefore, for an antibody composition or an Fc fusion protein composition, the relative amount of the defucosylated N-polysaccharide can be increased to increase the binding affinity to FcγRIII, or the ADCC activity of the composition.

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 all been shown to be very important to how 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 patterns achieved in specific host organisms, including the introduction or overexpression of certain enzymes involved in 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-terminal-linked oligosaccharides are G2 oligosaccharides. G2 means that it has two terminal Gals and does not have the biantannary structure of NeuAcs. WO99/22764 mentions substantially no antibody composition of glycoproteins with N-terminally-linked G1, G0 or G-1 oligosaccharides in the CH2 domain. G1 refers to a bifurcation structure with one Gal and no NeuAcs, G0 refers to a bifurcation structure without terminal NeuAcs or Gals, and G-1 refers to one GlcNAc less than 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 reported that, compared with 73% of antibodies expressed by NSO (mouse myeloma) cells, 47% of anti-hIL-5R antibodies expressed by YB2/0 (rat myeloma) cells had α1 -6 sugar chain linked by fucose. 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 RNAi can be used to inhibit the function of α1,6-fucosyltransferase to control the modification of fucose bound to sugar chains. A method of using cells to produce an antibody composition, which includes the use of anti-lectin cells, the lectin recognizes the sugar chain, and the first position of fucose is combined through the α-bond in the sugar chain linked by the compound 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 effector function of human IgG1 subclass antibodies, and by changing the structure of the sugar chain, it may be possible to prepare antibodies with stronger effector functions. However, the structure of sugar chains is diverse and complex, and solving the physiological role of sugar chains is still insufficient and expensive. Therefore, there is a need for a method of producing afucosylated antibodies.

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 the defucosylated protein produced by the method of the present invention and the cell that produces the defucosylated protein. Compared with naturally occurring fucosylated antibodies, the defucosylated antibodies of the present invention have increased antibody-dependent cellular cytotoxicity (ADCC) activity.

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 present 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 the fucosylation of the antibody in the host cell. The modified enzyme can be derived from an enzyme in the fucosylation pathway. In certain 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 a specific embodiment, the modified enzyme may be derived from α-1,6-fucose transferase (FUT8). The modified enzyme can inhibit the function of the enzyme naturally present in the host cell's fucosylation pathway, thereby inhibiting the fucosylation of the antibody 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, the method for producing afucosylated protein (including afucosylated antibody) includes (a) providing a host cell, (b) encoding a modified fucosylation pathway The nucleic acid of the enzyme is introduced into the host cell, and (c) the afucosylated 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 present invention relates to defucosylated proteins produced by the method of the present invention, including defucosylated antibodies. Compared with 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 present invention also relates to cells that produce 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 embodiments of the present invention are provided in the following examples, 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. In RC79 recombinant cells expressing mutant FUT8 enzyme, the expression level of FUT8 protein is similar to or the same as the expression level of FUT8 protein in 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 peak of the long dashed line represents the F83M-expressing RC79 recombinant cells stained with rhodamine-LCA. The gray-filled peaks represent F83M-expressing RC79 recombinant cells (negative control group) that have not been stained with rhodamine-LCA. The peak of the short dashed line represents RC79 cells stained with rhodamine-LCA (parental cells that do not express 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 the defucosylated anti-CD20 monoclonal antibody (R1 cell line) is 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. The His-tag FcγRIIIa (1μg/mL) and 5-80nM defucosylated anti-CD20 monoclonal antibody (Figure 4a) , 20-320nM RITUXAN® (Figure 4b) or 5-80nM GAZYVA® (Figure 4a) Figure 4c) , flow through the anti-His antibody-immobilized CM5 chip in sequence at a flow rate of 30 μL/min. Compared with 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 equivalent 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 the 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 RITUXAN® and the vector 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。圖中的數據點表示體重的平均值±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 figure represent the mean ± SD of body weight (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 afucosylated antibodies with improved activity. The present invention also relates to the antibody produced by the method of the invention and the cell producing the afucosylated antibody. Compared with naturally occurring fucosylated antibodies, the defucosylated antibodies of the present invention have increased antibody-dependent cellular 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 implementations are provided 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 belongs can understand that the modifications or changes of the embodiments clearly described in the description of the present invention will 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 terms used in the description are only used to describe specific embodiments and do not limit the present invention. The headings used below are only for the purpose of organizing the chapters and should not be seen as limiting the subject 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 mentioned in the description of the present invention can be fully incorporated as a whole (including parts), 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 specified, all technical and scientific terms used in the description of the present invention have the same meaning as those commonly understood by those with ordinary knowledge in the technical field to which the present invention belongs. Unless the context indicates otherwise, the singular "one" and "the" in the description of the invention include the plural meaning. Similarly, unless the context dictates 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 understood that all the amino acid sizes, all molecular weights or molecular mass values of the multipeptides are approximate values and are for description only. Although methods and materials similar or identical to those described in the description of 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 the event of a conflict, it will be subject to the restrictions of this release (including the explanation of terms). In addition, the materials, methods, and examples are merely illustrative in nature, and are not intended to limit the present invention.

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

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 present 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 aglycosylated antibody, including host cells derived from yeast, insects, amphibians, fish, reptiles, birds, mammals or humans, or fusion tumors. The host cell may be an unmodified cell or cell line, or a genetically modified cell line (for example, to promote the production of biological products). In some embodiments, the host cell is a cell strain that has been modified to allow growth under desired conditions, such as in a 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 generally 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 in the description of this invention include 293T (embryo 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 (fetal 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 (Ovary), OVCAR- 8 (ovary), P388 (leukemia), P388/ADR (leukemia), PC-3 (prostate), PERC6® (El-deformed 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 (ovary), 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 in the description of the present invention include monkey kidney (CVI-76) transfected with SV40 (COS-7), African green monkey kidney (VERO-76), green A cell line 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. Modification of 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 afucosylated antibody of the present invention can be produced in a host cell, and the fucosylation pathway in the host cell has been altered in a way 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 naturally occurring or wild-type enzymes in the fucosylation pathway, which is modified or destroyed in a way that changes or destroys 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, covalent linkage chemistry or protein moiety, introducing amino acid substitutions, insertions and/or deletions, and/or Any combination of it. 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 non-natural amino acids. Non-naturally occurring amino acids include, but are not limited to, ε-N lysine, ß-alanine, ornithine, leucine, norvaline, hydroxyproline, thyroxine, and γ-amine Butyric acid, homoserine, citrulline, aminobenzoic acid, 6-aminohexanoic acid (Aca; 6-Aminohexanoic acid), hydroxyproline, thiol propionic acid (MPA), 3-nitro-tyrosine Amino acid, pyroglutamate and the like. Naturally occurring amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, and isoleucine , Leucine, Lysine, Methionine, Phenylalanine, 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 can be derived from any naturally occurring enzyme in the fucosylation pathway. For example, the modified enzyme can 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), galactoside 2-α-L-fucose transferase 2 (FUT2), galactoside 3 ( 4)-L-Fucosyltransferase (FUT3), α(1,3)Fucosyltransferase, bone marrow-specific (FUT4), α-(1,3)-Fucosyltransferase (FUT5), α-(1,3)-Fucosyltransferase (FUT6), α-(1,3)-Fucosyltransferase (FUT7), α-(1,6)-Fucosyltransferase (FUT8) , Α-(1,3)-Fucosyltransferase (FUT9), Protein O-Fucosyltransferase 1 (POFUT1), Protein O-Fucosyltransferase 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 afucosylated 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 way 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 fucosylation pathway of the host cell is changed by introducing a nucleic acid encoding a modified enzyme in the fucosylation pathway into the host cell. For example, the nucleic acid molecule encoding the modified enzyme is inserted into the expression vector and transfected into the host cell. The nucleic acid molecule encoding the modified enzyme can be transiently introduced into the host cell, or stably fused into the genome of the host cell. Standard recombinant DNA methods can be used to produce 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 exhibit 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, nucleic acids may contain protein tags, screening markers, or regulatory sequences that control protein expression in host cells, 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 well known. Those with ordinary knowledge in the technical field to which the present invention pertains can understand that the selection of expression vectors, including the selection of regulatory fragments, can be based on several factors, including the selection of the host cell to be transformed, the expression level of the required protein, etc. Exemplary regulatory sequences for mammalian host cell expression include viral fragments that direct high protein expression in mammalian cells, for example, derived from cytomegalovirus (CMV) (for example, CMV promoter and/or enhancer Simian virus 40 (Simian Virus, SV40) (for example, SV40 promoter and/or enhancer), adenovirus (for example, adenovirus major late promoter (AdMLP)) and polyoma virus The 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 can be 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 present invention relates to host cells expressing modified enzymes in the fucosylation pathway. The performance of the modified enzyme in the host cell can interfere with the activity of the wild-type enzyme, resulting in inhibition or reduction of the fucosylation pathway. Therefore, the protein (eg, antibody) produced in the host cell expressing the modified enzyme is defucosylated (afucosylated).

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。 As used in the present invention, the phrase "hypofucosylated cell" or "hypofucosylated host cell" refers to that because the cell expresses a modified enzyme in the fucosylation pathway, fucose Cells in which the basic 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 expression vectors containing nucleic acid sequences encoding modified enzymes in the fucosylation pathway into host cells. Transfection can be performed using techniques known in the art. For example, chemical-based methods (such as lipids, calcium phosphate, cationic polymers, DEAE-dextran, activated dendrimers, magnetic beads, etc.) can be used, and instrument-based methods (such as , Electroporation, biojet technology, microinjection, laser/photoinjection, 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 selection markers present on the expression vector can be used to screen and isolate transfected cells from untransfected cells. In addition, various techniques can be used to screen and isolate transfected cells with the fucosylation pathway inhibited or reduced from the cells with the normal fucosylation pathway. For example, antibodies, lectins, metabolic markers, or chemoenzyme strategies can be used to determine fucosylation. In addition, by exposing the transfected cells to lentil lectin (Lens culinaris agglutinin, LCA, Vector laboratories L-1040), cells with inhibited or reduced fucosylation pathways can be screened out. LCA recognizes the α-1,6-fucosylated trimannose core structure of the N-terminally connected oligosaccharide, and allows the cells expressing this structure to enter the cell death pathway. Therefore, cells that survive exposure to LCA have an inhibited or reduced fucosylation pathway and are considered as hypofucosylated 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 present invention relates to a method for producing defucosylated protein. In some embodiments, the afucosylated protein is an afucosylated 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), ceruloplasmin , Α-2-macroglobulin, α-2-HS-glycoprotein, α-fetoprotein, haptoglobin, fibrinogen gamma chain precursor, immunoglobulin (including IgG, IgA, IgM, IgD, IgE And similar), APO-D, kininogen, 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 anaphylactoxin, 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 anaphylactoxin, complement C5 α'chain, complement C7, α-1 B glycoprotein, B-2-glycoprotein, vitamin D-binding Protein, Inter-α-trypsin inhibitor heavy chain H2, α-1B-glycoprotein, precursor of angiotensin, angiotensin-1, angiotensin-2, angiotensin-3, GARP protein, β -2-glycoprotein, Clusterin (Apo J), integrin alpha-8 precursor glycoprotein, integrin alpha-8 heavy chain, integrin alpha-8 light chain, hepatitis C virus particles, elf- 5. Kininogen, HSP33-homologous, lysine endopeptidase and protein 32 precursors rich in repeat 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 (e.g., multiple strains, single strains, monospecific, multispecific, chimeric, deimmunized, humanized, human, primatized, single chain, single domain, Synthetic and recombinant antibodies); part of a complete antibody with the desired activity or function (for example, an immune fragment of an antibody containing Fab, Fab', F(ab')2, Fv, scFv, single domain fragments); and Peptides and proteins of the Fc domain that can be fucosylated (for example, 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" used in the description of the present invention refers to an antibody produced under conditions where fucosylation is inhibited or significantly reduced compared to an antibody produced under natural conditions. Fragment. The defucosylated antibody produced by the method of the present invention may be completely (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% afucosylated molecules. In other embodiments, the antibodies produced by the methods of the invention may contain about 40% to about 100% afucosylated molecules. In certain embodiments, the antibody produced by the method of the present 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% defucosylated molecules. Not all N-glycosylated antibodies or fragments thereof (for example, Fc-fusion proteins) need to be afucosylated.

b.抗體種類b. Types of antibodies

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 an afucosylated antibody using the method of the present invention. There are no restrictions 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 antibody, anti-GD3 antibody, anti-GM2 antibody, anti-HER2 antibody, anti-CD52 antibody, anti-MAGE antibody, anti-HM124 antibody, anti-parathyroid gland Hormone-related protein (parathyroid 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 their analogs.

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 related to 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 circulatory 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 their analogs.

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 (e.g., Bevacizumab (AVASTIN®)), EGFR (e.g., Cetuximab (ERBITUX®)), HER2 (e.g., Trastuzumab (HERCEPTIN®)) and CD20 (e.g., , Rituximab (RITUXAN®)), and binding TNFa (e.g., Etanecept (ENBREL®), which includes the receptor binding domain (p75) of the TNF receptor), CD2 (e.g., Alefacept (AMEVIVE®), which contains LFA -3 CD2-binding region), or B7 (Abatacept (ORENCIA®), which includes the B7-binding region of CTLA4) 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。 The afucosylated protein of the present invention is produced in hypofucosylated cells, which includes afucosylated antibody. Using techniques known in the art, such as transfecting hypofucosylated cells with a protein-encoding expression vector, the defucosylated protein can be expressed in the hypofucosylated 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 protein-encoding expression vectors. For example, the amino acid sequence is reverse transcribed into a nucleic acid sequence to construct an expression vector, preferably using nucleic acid codons 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, just like the above-mentioned 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 (such as lipids, calcium phosphate, cationic polymers, DEAE-dextran, activated dendrimers, magnetic beads, etc.) can be used, and instrument-based methods (such as , Electroporation, biojet technology, microinjection, laser/photoinjection, etc.), or transfection by virus-based methods. Then, under conditions suitable for the selected expression system and host, the protein can be expressed in the transfected cells. An affinity column or other techniques known in the art can then be used to purify the expressed protein.

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 become a hypofucosylated cell) and the nucleic acid encoding the protein (to express the protein) into the host cell in any order to produce defucosylation protein. For example, a nucleic acid encoding a modified enzyme (to be a hypofucosylated cell) can be transfected into a host cell first, and then transfected with a nucleic acid encoding a protein (the protein is to be expressed). Alternatively, the nucleic acid encoding the protein (to express the protein) can be transfected into the host cell first, and then transfected with the nucleic acid encoding the modified enzyme (to be a hypofucosylated cell). In another variation, the nucleic acid encoding the modified enzyme (to be a hypofucosylated cell) and the nucleic acid encoding the protein (to express the 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, a defucosylated protein is produced by first preparing hypofucosylated cells, and then transfecting nucleic acid encoding the protein into the hypofucosylated cells : A) Obtain a host cell suitable for expressing the protein; b) Transfect the nucleic acid encoding the modified enzyme into the host cell; c) Screen and/or isolate transfected cells with hypofucosylation; d) Add The nucleic acid encoding the protein is transfected into low-fucosylated cells; e) screening and/or isolating the low-fucosylated cells transfected with the nucleic acid encoding the protein; f) inducing the protein in low-fucosylated cells Performance in chemical 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, a nucleic acid encoding a protein is first transfected into a host cell, and then the cell is transfected with a nucleic acid encoding a modified enzyme to produce afucosylated protein: a) Obtain a host cell suitable for expressing the protein; 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 a modified enzyme Transfect the nucleic acid of step (c) into the cells in step (c); e) screening and/or isolating the transfected cells with hypofucosylation in step (d); f) inducing the protein in the hypofucosylated cells In the 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 afucosylated protein can be produced according to the following steps: a) obtaining a host cell expressing or overexpressing the protein; b) transfecting the nucleic acid encoding the modified enzyme into the cell; c) Screening and/or isolating transfected cells with hypofucosylation; d) Inducing protein expression in hypofucosylation 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 another embodiment, as follows, by simultaneously transfecting the nucleic acid encoding the modified enzyme (to be a hypofucosylated cell) and the nucleic acid encoding the protein (to express the protein) to the host cell, 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) screening and/ Or isolate the transfected cells that express the protein and have hypofucosylation; d) Induce the expression of the protein in the hypofucosylation 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 production of afucosylated proteins, including antibodies, using the methods of the present invention described above, can be purified by methods known in the art. For example, the afucosylated protein produced by the method of the present invention, including antibodies, can be purified by methods such as physiological and chemical 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 afucosylated 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 and similar methods. The cytotoxic activity of antigen-positive cultured cell lines can be evaluated by measuring ADCC and CDC. Appropriate models of animal species that are relatively close to humans can be used to evaluate 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 with antibodies produced using standard methods, the afucosylated 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。 The "ADCC activity" used in the method of the present invention refers to the ability of an antibody to cause an antibody-dependent cellular cytotoxicity (ADCC) response. ADCC is a cell-mediated reaction in which non-specific cytotoxic cells expressing FcRs (eg, natural killer (NK) cells, neutrophils, and macrophages) recognize antibodies bound to the surface of target cells, and then create target cells Decomposition (ie, kill). The main vector cells in ADCC are natural killer (NK) cells. NK cells express FcγRIII, in which FcγRIIIA is an activating receptor and FcγRIIIB is an inhibitory receptor. Monocytes express FcyRI, FcyRII and FcyRIII. In vitro assays, such as those 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 afucosylated antibody of the present invention is at least 0.5, 1, 2, 3, 5, 10, 20, 50, 100 times that of 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 defucosylated antibodies have increased ADCC activity, compared with their fucosylated antibodies, defucosylated therapeutic antibodies can be administered in a lower amount or concentration. In some embodiments, the concentration of the afucosylated antibody of the present invention may be at least 2, 3, 5, 10, 20, 30, 50, or 100 times lower than the concentration of the fucosylated antibody. In some embodiments, compared to its wild-type counterpart, the afucosylated antibody of the present invention can exhibit higher maximal target cell lysis. For example, the maximum target cell lysis of the defucosylated antibody of the present invention is 10%, 15%, 20%, 25%, 30%, 40% higher than the maximum target cell lysis of the 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 with antibodies produced using standard methods, the afucosylated antibodies of the present 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 afucosylated 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 afucosylated antibodies with enhanced CDC function. In one embodiment, the Fc variant of the present invention has increased CDC activity. In another embodiment, the afucosylated antibody has at least 2 times, or at least 3 times, or at least 5 times, or at least 10 times, or at least 50 times, or at least 100 times higher than the corresponding 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 (intradermal, id), intraperitoneal (intraperitoneal, ip) or via any mucosal surface, such as oral ( Orally, po), sublingually (sl), cheek, nose, rectum, vagina or via pulmonary route, the defucosylated antibody of the present invention is administered.

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。 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, defucosylated antibodies are useful.

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 afucosylated antibody of the present invention will vary depending on the subject and specific mode of administration. The required dosage can be changed according to many factors well known to those with ordinary knowledge in the technical field of the present invention, including but not limited to, the antibody target, the type of the subject, and the size/weight of the subject. The dose can be 0.1 to 100,000 μg/kg body weight. The afucosylated antibody can be administered in a single dose or in multiple doses. Defucosylated antibodies can be administered once during a 24-hour period, multiple times during a 24-hour period, or continuous infusion. The afucosylated antibody 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 embodiment

本發明的特定實施例包括,但不限於以下: Specific embodiments of the present 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 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 defucosylation 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 (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 fucosyltransferase (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 from which the modified enzyme is derived 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 afucosylated protein produced in the host cell is an afucosylated 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 according to (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 according to (7), wherein the defucosylated antibody has an increased antibody-dependent cellular cytotoxicity (ADCC) activity compared to the 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 according to (7), wherein the complement-dependent cytotoxicity (CDC) of the afucosylated 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 combining 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 Transfection 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 is simultaneously transfected with the first nucleic acid sequence and the second nucleic acid sequence.

(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 afucosylated 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 of (11), wherein the first nucleic acid code is derived from 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 of (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) Transfect the first nucleic acid encoding the modified enzyme into host cells; b) Screen and isolate transfected cells with hypofucosylation; 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 hypofucosylated 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 afucosylated 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) Transfect the second nucleic acid encoding the protein to be produced into the host cell; b) Screen and isolate the cells transfected with the second nucleic acid; c) Transform the encoding The first nucleic acid of the modified enzyme is transfected into the cells in step (b); d) the transfected cells with hypofucosylation are screened and isolated; e) the hypofucosylated cells are expressed to be produced的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 afucosylated protein produced is an antibody and/or a fragment thereof.

5.額外實施例5. Additional Examples

本發明的額外實施例包括,但不限於以下: Additional embodiments of the present 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 afucosylated antibody, comprising: introducing a nucleic acid encoding at least one modified enzyme into a host cell to produce the afucosylated antibody 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 fucosyltransferase (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 according to (1), wherein the modified enzyme inhibits the activity of the wild-type fucosylation 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 afucosylated 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 afucosylated 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 of producing a defucosylated antibody, comprising: a) providing a host cell, b) introducing a nucleic acid encoding at least one modified enzyme into the host cell, and c) producing a defucosylated antibody in the host cell Alcosylated antibodies.

(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 according to (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) 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 fucosyltransferase (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 fucosylation enzyme 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 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 afucosylated 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 afucosylated 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 afucosylated 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 of (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 without or with hypofucosylation, comprising 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 enzymes in the fucosylation pathway and stable cell lines expressing the modified enzymes

1.細胞株1. Cell lines

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 CHO cell mutant strain lacking dihydrofolate reductase activity, purchased from Culture Collection and Research Center (CCRC), Taiwan ). The CHOdhfr (-) cell line was divided into 3 separate cultures and processed 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 to encode HERCEPTIN® (Trastuzumab, a monoclonal antibody against the protein HER2). Obtained a stable cell line expressing HERCEPTIN® and identified it 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 is untreated cells and maintained as a CHOdhfr(-) cell strain.

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 for modified enzymes encoding FUT8 and GMD

構築許多編碼經修飾之酵素FUT8與GMD的表現載體。 Construct 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 respectively represent different modifications to α-1,6-fucose transferase, wild-type FUT8 protein (GenBank No.NP_058589.2). 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 expressed enzymes. 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 wild-type FUT8 protein with a modified mutant type in K369E, D409K and S469V respectively. F8D1 represents the mutant type in the wild-type FUT8 protein with amino acid residues at positions 365 to 386 deleted.

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 to the wild-type nucleic acid sequence by the GMD vector and the amino acid changes produced in the expressed enzymes. In particular, mutant GMD4M represents a modification of GDP-mannose 4,6-dehydratase, wild-type GMD protein (GenBank No.NP_001233625.1, which has 4 mutations in 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 then cloned into the pHD expression vector (pcDNA3.1Hygro, Invitrogen, Carlsbad, CA, cat.no.V870-20, with dhfr Gene) Pad/EcoRv or BamHI/EcoRV position 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)。 The pHD/F83M, pHD/F8M1, pHD/F8M2, pHD/F8M3, pHD/F8D1 and pHD/GMD4M plastids were transfected into different cell lines by electroporation (PA4000 PULSEAGILE® electroporator, Cyto Pulse Sciences), including ( a) RC79 cell line (CHO cells expressing RITUXAN®), (b) HC59 cell line (CHO cells expressing HERCEPTIN®), and (c) CHOdhfr (-) cells (CHO cell mutations lacking dihydrofolate reductase activity) 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 cultured in RC79 medium containing 0.1 to 0.25mg/mL Hygromycin (containing 0.4μM MTX, 0.5mg/mL Geneticin (Geneticin), 0.05mg/mL bleomycin) (Zeocin), 4mM Glutamax-I and 0.01% F-68 EX-CELL®302 serum-free medium). Then, the transfected cells were cultured in EX containing 0.4μM MTX, 0.5mg/mL geneticin, 0.05mg/mL bleomycin, 4mM Glutamax-I, 0.01% F-68 and 0.25mg/mL hygromycin -CELL®302 serum-free medium, and as described below, separated with Lens culinaris agglutinin (LCA) to generate 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 cultured 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 4mM Glutamax-I) EX-CELL® 325 PF CHO medium). Then, the transfected cells were cultured on EX-CELL® 325 PF containing 0.8μM MTX, 0.5mg/mL geneticin, 0.05mg/mL bleomycin, 4mM Glutamax-I and 0.25mg/mL hygromycin In CHO medium, and as described below, separated by LCA to generate a cell bank of the 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 are first cultured in EX-CELL® 325 PF CHO medium containing 4mM Glutamax-I and 0.1 to 0.25mg/mL hygromycin. Next, the transfected cells were cultured in EX-CELL® 325 PF CHO medium containing 4mM Glutamax-I, 0.25mg/mL hygromycin and 0.01μM MTX to generate a cell bank of the C109F83M cell line.

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

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 screen cells with hypofucosylation.

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 a preliminary selection medium containing hygromycin as selection pressure, and then used to identify the core structure of α-1,6-fucosylated trimannose, which can identify N-linked oligosaccharides. And let the cells expressing this structure go to the LCA of the cell death pathway for final screening. The transfected strains of RC79, HC59 or CHO were planted in 2.5 mL fresh medium containing 0.4 mg/mL LCA at 1.2 x 10 5 cells/mL, and the cell survival rate was counted on the 3rd or 4th day. The cells are cultured in this preliminary screening medium until the cell survival rate reaches 80%. After the cell survival rate reached 80%, the cells were resuspended in a fresh selection medium containing a gradient concentration of LCA at 1.2 x 10 5 cells/mL. The LCA screening is repeated several times until the final concentration of LCA 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, the cells were labeled with LCA and analyzed by flow cytometry. First, the cells were planted in a 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。 Then, wash the 1 x 10 7 cells twice with 10 mL of ice-cold PBS, and resuspend them 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。 FACSAria TM or Influx TM Cell Sorter (BD Biosciences, San Jose, CA) was used to analyze and classify cells. For different cell lines, 1-3 classifications are required to generate a homogeneous population of hypofucosylated cells. In addition, the CLONEPIX TM 2 system (MOLECULAR DEVICES®) was used to isolate stable cell lines with low fucosylation and transferred to a 96-well plate. After culturing for about 2 weeks, the cells were transferred to a 6-well plate and analyzed by flow cytometry. Then, the hypofucosylated cells were moved to a filter tube used for fed-batch culture to evaluate the cell performance and the degree of fucosylation of the antibody purified from the obtained 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 hypofucosylated 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 single low-fucose cell line of C109F83M, AF97, was isolated and transfected with nucleic acid encoding RITUXAN® by electroporation to express RITUXAN®. The transfected strain was transferred to a 25T flask containing non-selective medium to restore 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. The CLONEPIX TM 2 system was used to select single cells to generate AF97 anti-CD20 cell lines.

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 hypofucosylated 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 antibodies 1.抗體的表現與純化1. Expression and purification of antibodies

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 feed batches to express antibodies. The antibody purified from the cells is subjected to monosaccharide analysis to quantitatively analyze the sugar chains 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。 Recombinant RC79 cells were cultured in EX-CELL® 302 serum-free medium containing 4mM Glutamax and 0.01% F-68, and maintained at 37℃, 5% CO 2 in a shaking incubator (Infors Multitron Pro).

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 0.8μM MTX, 0.5mg/mL geneticin, 0.05mg/mL bleomycin, 4mM Glutamax-I and 0.25mg/mL hygromycin , And maintained in a shaking incubator (Infors Multitron Pro) at 37°C and 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。 The parameters of cell culture are routinely monitored every day. Use a hemocytometer to exclude trypan blue to determine the 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. Equilibrate the protein A column with 5 column volumes of 0.1M Tris, pH 8.3, and then add the sample to the column. Wash the unbound protein with 0.1M Tris, pH 8.3 (2 times the column volume) and PBS, pH 6.5 (10 times the column volume). The column was further cleaned with 0.1M sodium acetate, pH 6.5 (10 times the column volume). Finally, the antibody was extracted with 0.1M glycine, pH 2.8, and neutralized with the same extraction volume of 0.1M Tris, pH 8.3.

2.抗體的N-多醣圖譜分析的判定2. Determination of antibody N-polysaccharide profile 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。 Analyze the N-polysaccharide profile with ACQUITY UPLC® system. First, digest 0.3 mg of antibody samples with 3 U PNGase-F in 0.3 mL digestion buffer (15 mM Tris-HCl, pH 7.0) at 37°C for 18 hours. The released N-polysaccharides were separated from the antibody by ultrafiltration using an AMICON®Ultra-0.5mL 30K device at 13,000rpm 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 (containing 0.34M aminobenzamide and 1M sodium cyanoborohydride in DMSO-acetic acid (7:3 v/v). ) Solvent) and incubate at 65°C for 3 hours. The PD MINITRAP TM G10 size exclusion column is used to remove excess 2-AB labeling reagent. The labeled N-polysaccharide was freeze-dried overnight and re-dissolved in 50 μL ddH 2 O for UPLC detection. At 60°C, the N-glycan profile was obtained by ACQUITYUPLC® system and Glycan BEH Amide column. A linear gradient of 100 mM ammonium formate, pH 4.5/acetonitrile was used to separate different forms of N-glycans.

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 in all cell types, the binding of LCA was extremely low on the cell surface that overexpressed F83M protein. Similarly, on RC79 cells overexpressing F8M1, F8M2, F8M3, F8D1, or GMD4M protein, no LCA binding was detected (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 results of antibodies produced in RC79 and HC59 cells with unmodified fucosylation pathways and RC79 and HC59 cells with modified fucosylation pathways by overexpression of F83M modified enzymes N-glycan profile. The data in Table 3 shows that most of the anti-CD20 and anti-ErbB2 antibodies produced in cells with unmodified fucosylation pathways are heavily fucosylated. In particular, in these cells, only 3.67% of the anti-CD20 and 3.64% of the anti-ErbB2 antibodies were defucosylated. In contrast, antibodies produced in cells overexpressing the F83M modified enzyme have a very low degree of fucosylation. In particular, in cells overexpressing F83M modified enzymes, about 98.86-98.91% of anti-CD20 and about 92.12-96.52% of anti-ErbB2 antibodies were 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 pathways and RC79 cells with fucosylation pathways modified by overexpression of one of F8M1, F8M2, F8M3, F8D1 or GMD4M modified enzymes The N-glycan profile of the antibody produced in. The data in Table 4 shows that most of the anti-CD20 antibodies produced in RC79 cells with unmodified fucosylation pathway are heavily fucosylated. In particular, in these cells, only 3.67% of the anti-CD20 antibodies were afucosylated. In contrast, antibodies produced in RC79 cells that overexpress the modified enzyme have a very low degree of fucosylation. In particular, the degree of defucosylation of anti-CD20 antibodies produced by cells overexpressing F8M1, F8M2, F8M3, F8D1, or GMD4M modified enzymes ranged from 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 the FUT8 modified enzymes (F8M1, F8M2, F8M3, F8D1) is 95.70 to 97.16%, and is modified by overexpression of GMD The degree of defucosylation of the antibody produced by the enzyme (GMD4M) is 92.78%. These results confirm that the degree of deglycosylation of antibodies produced in cells overexpressing FUT8 modified enzymes is higher than that of antibodies produced in cells overexpressing 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 expressing modified enzymes (FUT8 or GMD) in the fucosylation pathway. This result also shows that the antibodies produced in these transfected cells are afucosylated 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 the antibodies produced in the AF97 cell line was evaluated. The results in Table 5 show that the antibodies produced in AF97 cells that overexpress the F83M modified enzyme have a very low degree of fucosylation. In particular, 97.83% of the anti-CD20 antibodies produced in the AF97 cell line are defucosylated. In contrast, the 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 the afucosylated antibody can be produced in cells that are first transfected with the nucleic acid encoding the modified enzyme and then transfected with the nucleic acid encoding the antibody for the second time. Therefore, the methods of the present invention can be used to modify cells to produce afucosylated antibodies.

3.重組細胞中FUT8蛋白的表現3. The performance 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。 The precipitate of RC79 cells and recombinant cells expressing FUT8 modified enzymes (eg, F8M1, F8M2, F8M3, or F8D1) was decomposed in 1% Triton X containing a phosphatase inhibitor cocktail (Sigma-Aldrich, Cat.S8820) -100 in. The DC TM (cleaning agent) protein analysis (BIO-RAD) reagent was used to confirm the protein concentration in the supernatant of the decomposed cells. Each sample of the supernatant containing 30μg protein was separated by 12.5% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to nitrocellulose On the 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), To block the nitrocellulose membrane for 1 hour, and separately incubate 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 with 25mM Tris-HCl (pH7.4) (containing 120mM NaCl, 0.1% gelatin(w/w) and 0.1% TWEEN® 20(v/w)) for 3 times for 5 minutes, and 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. After additional washing, SIGMAFAST DAB with Metal Enhancer (Sigma, Cat.D0426), to analyze the film.

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 of FUT8 protein in recombinant cells and RC79 parent is similar. These results indicate that the production of afucosylated antibody in RC79 cells expressing the modified enzyme has nothing to do with the expression level of FUT8 protein. These results show that FUT8 modified enzymes can interfere with the wild-type FUT8 protein in cells to inhibit and/or reduce the fucosylation pathway, so that recombinant cells can 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 the expression of the FUT8 protein, the mechanism of using the method of the present invention to produce afucosylated antibodies 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。 To evaluate the stability of RC79 recombinant cells expressing F83M modified enzyme.

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 a medium without selection reagents for 3 months. Weekly flow cytometry analysis is used to monitor the fucosylation of the cells, and the ACQUITY UPLC® System and Glycan BEH Amide column are used to determine the composition of the N-glycan of the purified antibody every month, lasting 3 Month, as mentioned 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 72 days, 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 afucosylated 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。 The 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 afucosylated 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 antibodies

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, 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 a healthy donor 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, 24mL of diluted blood is slowly added to Ficoll-Paque and centrifuged at 400 xg for 32 minutes at 25°C to separate monocytes. The buffy coat was fully distributed into two 50 mL centrifuge tubes containing 20 mL of 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 the 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 the PBMC cells. A sufficient volume of PBMC cell suspension was added to the 75T flask, and about 15 mL of cells per flask, the 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. The PBMC cells were cultured in a 37°C, 5% CO 2 incubator for 18 hours. PBMC cells stimulated by IL-2 were collected and centrifuged at 1,200 rpm for 12 minutes at 25°C, and then the supernatant was discarded. Add PBS (10 mL) and mix with the cells. The cells were centrifuged at 1,200 rpm at 25°C for 12 minutes to remove the supernatant. The cells were resuspended in 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. The medium was analyzed with RPMI, and the cells were resuspended to prepare a target cell solution of 5 x 10 5 cells/mL. Add the target cell solution (40 μL of 5 x 10 5 cells/mL) to the holes of the 96-well cell culture plate at the bottom of the V. Next, add 20 μL of the prepared commercially available RITUXAN® solution (MABTHERA®) (25-0.0025μg/mL) (positive control group) and defucosylated antibody (R1 cell line) solution (25-0.0025μg). /mL), or RPMI analysis medium (negative control group) is added to the hole and mixed with the target cell solution. Incubate the V-bottom 96-well cell culture dish 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, 5% CO 2 for 4 hours. Before collecting the supernatant, add the lysis solution (10 μL) of CYTOTOX 96® to the dish of the Tmax and BlkV groups, and react for 1 hour. Centrifuge the V-bottom 96-well cell culture dish at 300 xg for 4 minutes, and transfer 50 μL of the supernatant 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。 Add lactate dehydrogenase (LDH) (2 μL) to 10 mL of LDH positive control diluent to prepare LDH positive control solution. The prepared LDH positive control solution (50 µL) was added to the holes of the 96-well flat-bottomed analysis disc.

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。 Add the LDH reconstitution matrix mixture (50 μL) to each test hole of the analysis disc. Cover the dish and incubate in the dark at room temperature for 30 minutes. Add stop solution (50 μL) to each test hole of the plate. Immediately after adding the stop solution, the absorbance at 490 nm was recorded. Using each group (S, PBMC, T, E, and Tmax) blank removal absorbance value, ADCC activity was calculated with 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 released by the LDH of the sample (target cell + PBMC + anti-CD20 antibody); PBMC is the absorbance value released by the LDH of the target cell and PBMC; E is the absorbance value released by the LDH of the PBMC; T is the spontaneous emission value of the target cell The absorbance value of sexual LDH release; and Tmax is the absorbance value of the maximum LDH release of 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 with the commercially available RITUXAN® (MABTHERA®), the defucosylated anti-CD20 antibody (R1 cell line) is in PBMC from both donor 1 (Figure 3a ) and donor 2 ( Figure 3b) A significantly stronger and higher ADCC response is induced in the cell.

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, to RC79F83MR1 fucose from cell lines of antibody-based anti -CD20 EC 50 significantly lower than commercially available RITUXAN®, which is a fucosylated anti -CD20 antibody. In particular, the defucosylated anti-CD20 antibody (R1 cell line) has 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 ADCC activity of the defucosylated anti-CD20 antibody (R1 cell line) is 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 afucosylated antibody

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 anti-histidine (anti-His) antibody coupled to BIACORE®CM5 chip with amine coupling kit and immobilization wizard of BIACORE®X100 control software to evaluate fucosylation and de-rocking The binding affinity of the alcosylated anti-CD20 antibody to the 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, the His-labeled FcγRIIIa recombinant protein (1 μg/mL) was injected onto the anti-His antibody-immobilized CM5 chip 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。 The defucosylated anti-CD20 antibody (5, 10, 20, 40, and 80 nM) from cell line 1, and the commercially available fucosylated anti-CD20 antibody RITUXAN® (MABTHERA®) (20, 40, 80, 160 and 320 nM) and the commercially available defucosylated anti-CD20 antibody GAZYVA® (obinutuzumab) (5, 10, 20, 40 or 80 nM) were injected through the chip at a flow rate of 30 μL/min for 3 minutes. The running buffer was flowed through the wafer for 5 minutes at a flow rate of 30 μL/min. Glycine, pH 1.5 (10 mM) was injected into the wafer for 60 seconds at a flow rate of 30 μL/min.

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 response pattern of each cycle to obtain the equilibrium dissociation constant (K D ), the association rate constant (Ka) and the dissociation rate constant (Kd) values. The response map of each cycle is adapted with a 1:1 Langmuir combined model. If the Chi 2 value is lower than 1/10X Rmax, the fitting model is appropriate and the 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 profiles of the three antibodies tested. A typical SPR response profile shows that the conditions used in this analysis (for example, binding time, dissociation time, and antibody concentration range) are appropriate. In addition, the Chi 2 values of the three antibodies are lower than the 1/10X Rmax value, indicating that the 1:1 Langmuir model is suitable for adaptation to the response maps 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 , the binding affinity of defucosylated anti-CD20 antibody (R1 cell line) to FcγRIIIa is 10 times stronger than MABTHERA® (K D of R1 cell line = 13.0 nM, MABTHERA® = 151.5 nM) . In addition, the binding affinity of defucosylated anti-CD20 antibody (R1 cell line) to FcγRIIIa is three times stronger than that of GAZYVA® (K D of R1 cell line = 13.0 nM, 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 confirmed that the defucosylated anti-CD20 antibody (R1 cell strain) prepared according to the present invention has a higher performance than the commercially available fucosylated anti-CD20 antibody RITUXAN® (MABTHERA®) and the commercially available defucosylated anti-CD20 antibody. 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 antibodies

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 afucosylated antibody produced by the method of the present invention was 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 performed (subculture density: 2-3×10 5 cells/mL). Daudi cells were collected and centrifuged at 300 rpm for 5 minutes. Resuspend the cells in RPMI medium to prepare a cell suspension with a concentration of 1 x 10 5 cells/mL. After resuspension, 100 μL of cell suspension or 100 μL of RPMI medium was seeded into the holes of the white 96-well plate.

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 the defucosylated anti-CD20 antibody (R1 cell line) were prepared in physiological saline at a concentration of 120μg/mL to 0.234μg/mL. Next, add 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 to 96 white wells containing Daudi cells or RPMI medium In the hole of the disk. Add CELLTITER-GLO® reagent (20μL) to each hole and mix. Place the disk on a micro disk shaker, shake at 750 rpm for 2 minutes, and then incubate in the dark at room temperature for 10 minutes. Detect the luminous intensity (integration time: 1 second) by inserting a multi-mode reader with a high-sensitivity fluorescent box 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 the CDC activity comparison of defucosylated anti-CD20 antibody (R1 cell line) and RITUXAN®. The EC 50 value of the defucosylated anti-CD20 antibody (R1 cell line) is 0.682 μg/mL, which is 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, which 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 show that in order to achieve effective cancer treatment, the amount of GAZYVA® should be increased. In contrast, the results of this example and example 5 prove 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 for 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 subcutaneous xenograft model of B-cell lymphoma 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 expresses 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 anti-tumor efficacy of afucosylated antibody (R1 cell line) and 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。 The 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. The cells were resuspended in a new medium containing some conditioned medium (fresh CM: conditioned CM=9:1). The cells are subcultured at a ratio of 1:2 to 1:10 (number of cells seeded: total number of cells obtained), and the cell concentration is at least 1 x 10 5 cells/mL. The culture plate was incubated at 37°C. SU-DHL-4 cells were cultured in five 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 108 cells/mL. On ice, use a pre-cooled syringe with an 18G needle to mix the cell suspension with an equal volume of MATRIGEL® in a 50 mL test tube. The final cell concentration is 5 x 10 7 cells/mL. Using a pre-cooled 1mL syringe with a 23G *1" needle, inject 5 x 10 7 cells/mL Matrigel-SU-DHL-4 cell mixture (100uL) subcutaneously into the back of each mouse (SCID/Beige mouse) The right side of the area. The total number of inoculated cells is 5 x 10 6 cells. Use a caliper to measure the tumor volume of each mouse every 3 or 4 days, and calculate with 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 200mm 3 (198.25±55.53mm 3 ), it occurred about 20 days after tumor inoculation. The mice were divided into three groups, 5 mice in each group, and then 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 every week for 3 weeks. Twice a week, the body 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, the tumor tissues were taken out, 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, using Student t test). In contrast, compared to the vehicle-only group, RITUXAN® has no statistically significant difference in tumor volume. At a dose of 1mg/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 result 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 the 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 show 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> Union Asia Pharmaceuticals United Biopharmaceuticals Peng Wenjun Chen Huirong

<120> 去岩藻醣基化抗體的製造方法 <120> Manufacturing method of 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> The nucleic acid sequence of F83M modified enzyme, the positions 1093-1095, 1225-1227 and 1357-1359 of the wild-type FUT8 gene have been 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> The amino acid sequence of F83M modified enzyme, the residues at positions 365, 409 and 453 of the wild-type FUT8 protein are 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 has been 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 F8M1 modified enzyme, the residue at position 369 of the wild-type FUT8 protein is 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 has been 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 F8M2 modified enzyme, the residue at position 409 of wild-type FUT8 protein is 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, wild-type FUT8 gene position 1405-1407 is 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 F8M3 modified enzyme, the residue at position 469 of the wild-type FUT8 protein is 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> The amino acid sequence of F8D1 modified enzyme, the residues at positions 365-386 of the 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, positions 463-465, 469-471, 535-537 and 547-549 of the wild-type GMD gene have been 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> The amino acid sequence of GMD4M modified enzyme, the residues at positions 155, 157, 179 and 183 of the wild-type GMD gene are 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> The deleted nucleic acid sequence in the F8D1 mutant gene

<400> 17

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

Claims (16)

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,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)或岩藻醣轉移酶(fucosyltransferase,FUT),編碼該經修飾之酵素的核酸係擇自於由SEQ ID Nos:3、5、7、9、11或15組成之群組,該經修飾之酵素於該宿主細胞中可降低或抑制該經修飾之酵素所源自的野生型酵素的活性。 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, wherein the modified enzyme is derived from GDP-mannose 4,6-dehydratase (GDP-mannose 4,6-dehydratase, GMD) or fucosyltransferase (FUT) , The nucleic acid encoding the modified enzyme is selected from the group consisting of SEQ ID Nos: 3, 5, 7, 9, 11, or 15, and the modified enzyme can reduce or inhibit the modified enzyme in the host cell The activity of the wild-type enzyme from which the modified enzyme is derived. 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 described in item 1 of the scope of patent application, wherein the modified enzyme is derived from 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α-1,6-岩藻醣轉移酶(FUT8)。 The method described in item 1 of the scope of the patent application, 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抑制或降低該宿主細胞中的岩藻醣基化。 The method according to claim 1, 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 claim 1, wherein the afucosylated protein produced in the host cell is an afucosylated antibody or a fragment thereof. 如申請專利範圍第5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 claim 5, wherein the defucosylated antibody or fragment thereof is at least 90% defucosylated. 如申請專利範圍第5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 according to claim 5, 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5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 described in item 5 of the scope of patent application, wherein the afucosylated antibody is complement dependent Complement dependent cytotoxicity (CDC), compared to its fucosylated counterpart, is not reduced and inhibited. 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,其中該第一核酸編碼源自於GDP-甘露醣4,6-脫水酶(GMD)及/或岩藻醣轉移酶(FUT)的一經修飾之酵素,且該第一核酸序列係擇自於由SEQ ID Nos:3、5、7、9、11及/或15組成之群組,該經修飾之酵素於該宿主細胞中可降低或抑制該經修飾之酵素所源自的野生型酵素的活性。 A method for producing a defucosylated protein in a host cell, comprising combining a first nucleic acid sequence encoding a modified enzyme of a fucosylation pathway, and a second nucleic acid encoding the protein to be produced The sequence is transfected into the host cell, wherein the first nucleic acid encodes a modified enzyme derived from GDP-mannose 4,6-dehydratase (GMD) and/or fucose transferase (FUT), and the The first nucleic acid sequence is selected from the group consisting of SEQ ID Nos: 3, 5, 7, 9, 11 and/or 15. The modified enzyme can reduce or inhibit the modified enzyme in the host cell The activity of the wild-type enzyme from which it is derived. 如申請專利範圍第9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 claim 9, wherein the host cell is simultaneously transfected with the first nucleic acid sequence and the second nucleic acid sequence. 如申請專利範圍第10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 according to claim 10, wherein the afucosylated protein produced is an antibody or a fragment thereof. 如申請專利範圍第9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 claim 9, 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9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 described in item 9 of the scope of the patent application includes the following steps: a) transfecting the first nucleic acid encoding the modified enzyme into the host cell; b) screening and isolating hypofucosylated transfectants Transfecting the cell; c) transfecting the second nucleic acid encoding the protein to be produced into the hypofucosylated cell; d) expressing the protein to be produced in the hypofucosylated cell. 如申請專利範圍第13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 13 of the scope of the patent application, wherein the afucosylated protein produced is an antibody and/or a fragment thereof. 如申請專利範圍第9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 described in item 9 of the scope of patent application includes the following steps: a) transfecting the second nucleic acid encoding the protein to be produced into the host cell; b) screening and isolating the transfected second nucleic acid C) transfecting the first nucleic acid encoding the modified enzyme into the cell in step (b); d) screening and isolating transfected cells with low fucosylation; e) in the low The fucosylated cell expresses the protein to be produced. 如申請專利範圍第15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 according to claim 15, wherein the afucosylated protein produced 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 TW202010844A (en) 2020-03-16
TWI748124B true 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)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130224190A1 (en) * 2010-07-19 2013-08-29 International - Drug - Development - Biotech Anti-cd19 antibody having adcc and cdc functions and improved glycosylation profile

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130224190A1 (en) * 2010-07-19 2013-08-29 International - Drug - Development - Biotech Anti-cd19 antibody having adcc and cdc functions and improved glycosylation profile

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Malphettes L et al., "Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies", Biotechnology and Bioengineering, vol.106, no.5, p.774-783, 2010/08/01
Malphettes L et al., "Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies", Biotechnology and Bioengineering, vol.106, no.5, p.774-783, 2010/08/01; *
Ng BG et al., "Biallelic Mutations in FUT8 Cause a Congenital Disorder of Glycosylation with Defective Fucosylation", The American Journal of Human Genetics, vol.102, no.1, p.188-195, 2018/01/04 *
Yamane-Ohnuki N et al., "Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity",Biotechnology and Bioengineering, vol.87, no.5, p.614-622, 2004/09/05
Yamane-Ohnuki N et al., "Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity",Biotechnology and Bioengineering, vol.87, no.5, p.614-622, 2004/09/05; *

Also Published As

Publication number Publication date
TW202010844A (en) 2020-03-16

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
US9120856B2 (en) Anti-CD19 antibody having ADCC function with improved glycosylation profile
JP2009114201A (en) Antibody glycosylation variant having increased antibody-dependent cellular cytotoxicity
AU2018438767B9 (en) Afucosylated antibodies and manufacture thereof
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