TWI535454B - Conjugated factor viii molecules - Google Patents

Conjugated factor viii molecules Download PDF

Info

Publication number
TWI535454B
TWI535454B TW102147349A TW102147349A TWI535454B TW I535454 B TWI535454 B TW I535454B TW 102147349 A TW102147349 A TW 102147349A TW 102147349 A TW102147349 A TW 102147349A TW I535454 B TWI535454 B TW I535454B
Authority
TW
Taiwan
Prior art keywords
molecule
factor viii
domain
fviii
peg
Prior art date
Application number
TW102147349A
Other languages
Chinese (zh)
Other versions
TW201412333A (en
Inventor
夏恩 迪菲斯
Original Assignee
諾沃 諾迪士克公司
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 諾沃 諾迪士克公司 filed Critical 諾沃 諾迪士克公司
Publication of TW201412333A publication Critical patent/TW201412333A/en
Application granted granted Critical
Publication of TWI535454B publication Critical patent/TWI535454B/en

Links

Description

共軛因子VIII分子 Conjugated factor VIII molecule

本發明係關於共軛凝血因子VIII分子。具體而言,本發明係關於具有改良之循環半衰期之共軛因子VIII分子。 The present invention relates to conjugated Factor VIII molecules. In particular, the invention relates to conjugated Factor VIII molecules having an improved circulating half-life.

A型血友病係由凝血因子VIII(FVIII)活性缺陷或功能障礙而引起之遺傳性出血病症。臨床表現不在於一期止血-血凝塊形成正常發生-而在於由於缺乏二期凝血酶形成導致凝塊不穩定。該疾病係藉由靜脈內注射分離自血液或重組產生之凝血因子FVIII來治療。 Hemophilia A is an inherited bleeding disorder caused by deficiency or dysfunction of factor VIII (FVIII) activity. The clinical manifestation is not that the first stage of hemostasis - the formation of blood clots occurs normally - but that the clot is unstable due to the lack of stage II thrombin formation. The disease is treated by intravenous injection of blood or recombinantly produced factor FVIII.

當前治療建議正自傳統的按需治療向預防轉移。內源性FVIII之循環半衰期為12-14小時且因此預防性治療需要一週實施數次以使患者獲得實質上無症狀之生活。靜脈內(IV)投與對於許多人(尤其兒童及未成年人)而言導致明顯不便及/或疼痛。因此,業內需要具有因子VIII活性之新穎因子VIII產物,其較佳在結構上係同質的、較佳係安全的且較佳具有顯著延長之循環半衰期以減少因子VIII之每週投與次數。此外,業內亦需要用於獲得及產生該等分子之相對簡單的方法。 Current treatment recommendations are shifting from traditional on-demand treatment to prevention. The circulating half-life of endogenous FVIII is 12-14 hours and thus prophylactic treatment requires several times a week to allow the patient to obtain a substantially asymptomatic life. Intravenous (IV) administration causes significant inconvenience and/or pain for many people, especially children and minors. Thus, there is a need in the art for a novel Factor VIII product having Factor VIII activity which is preferably structurally homogeneous, preferably safe, and preferably has a significantly extended circulating half-life to reduce the number of weekly doses of Factor VIII. In addition, there is a need in the industry for a relatively simple method for obtaining and producing such molecules.

業內已知對因子VIII實施聚乙二醇化來延長循環半衰期。然而,獲得具有同質結構以及顯著改良之循環半衰期之安全產物仍為障礙。可用於產生共軛因子VIII分子之方法通常極為費力,及/或往往導致產量較低及/或產物結構不同質。WO2008011633中提出使用人工構造之O-連接糖基化位點來獲得具有延長之治療性蛋白質循環半衰期的治療 性蛋白質,然而,其中未揭示共軛因子VIII分子。 It is known in the art to PEGylate Factor VIII to extend the circulating half-life. However, obtaining a safe product with a homogenous structure and a significantly improved cyclic half-life is still an obstacle. Methods that can be used to generate conjugated Factor VIII molecules are generally laborious and/or often result in lower yields and/or product structure identities. WO2008011633 proposes the use of artificially constructed O-linked glycosylation sites to obtain treatments with prolonged therapeutic protein circulating half-life Sexual proteins, however, do not reveal conjugated factor VIII molecules.

在第一態樣中,本發明係關於具有改良之循環半衰期的B結構域截短型因子VIII分子,該分子經由截短之B結構域中之O-連接寡糖與親水性聚合物共價共軛,其中因子VIII激活導致共價共軛之側基移除。 In a first aspect, the invention relates to a B domain truncated Factor VIII molecule having an improved circulating half-life, the molecule covalently covalently with a hydrophilic polymer via an O-linked oligosaccharide in a truncated B domain Conjugation in which Factor VIII activation results in the removal of covalently conjugated side groups.

在其他態樣中,本發明進一步係關於獲得該等分子之方法、該等分子之用途及包含該等分子之醫藥組合物。 In other aspects, the invention is further directed to methods of obtaining such molecules, uses of such molecules, and pharmaceutical compositions comprising such molecules.

因此,本文提供具有改良之循環半衰期之共軛因子VIII分子,其中共軛側基(例如親水性聚合物)在激活後移除。本發明分子較佳具有同質結構-至少對於親水性聚合物在截短之B結構域中的位置而言-且較佳具有有利的安全性質。另外,本文進一步提供獲得該等分子之相對簡單方法。較佳地,本發明之激活之因子VIII分子與內源性激活之因子VIII類似。 Accordingly, provided herein are conjugated Factor VIII molecules having an improved circulating half-life in which a conjugated pendant group (eg, a hydrophilic polymer) is removed upon activation. The molecules of the invention preferably have a homogenous structure - at least for the position of the hydrophilic polymer in the truncated B domain - and preferably have advantageous security properties. In addition, a relatively simple method of obtaining such molecules is further provided herein. Preferably, the activated Factor VIII molecule of the invention is similar to endogenously activated Factor VIII.

在圖中,共軛基團之大小有時稱為「K」,在本文中其與KDa(千道爾頓)具有相同含義。 In the figure, the size of the conjugated group is sometimes referred to as "K", which has the same meaning as KDa (kiloton) herein.

圖1:O-連接寡糖之糖聚乙二醇化過程的示意圖。此圖並不代表達成實例中所獲得產物之可能方式的詳盡無遺列示。 Figure 1: Schematic representation of the PEGylation process of O-linked oligosaccharides. This figure does not represent an exhaustive list of possible ways of achieving the products obtained in the examples.

圖2:反應混合物在Source 15Q上之離子交換層析(A)。所收集溶離份之分子標記(左)的SDS-PAGE(B)。 Figure 2: Ion exchange chromatography (A) of the reaction mixture on Source 15Q. SDS-PAGE (B) of the molecular marker (left) of the collected fractions.

圖3:在superdex 200尺寸排除層析上對封端產物實施純化。 Figure 3: Purification of the capped product on a superdex 200 size exclusion chromatography.

圖4:使用多種aPTT試劑之O-糖聚乙二醇化rFVIII的凝固活性。 (A)顯示凝固活性與產色活性之比。(B)顯示比凝固活性。 Figure 4: Coagulation activity of O-glycol PEGylated rFVIII using various aPTT reagents. (A) shows the ratio of coagulation activity to chromogenic activity. (B) shows specific clotting activity.

圖5:40K-PEG-[O]-N8在FVIII KO小鼠中之活體內效果(阻塞時間)。 Figure 5: In vivo effect (blocking time) of 40K-PEG-[O]-N8 in FVIII KO mice.

圖6:顯示根據本發明產生糖聚乙二醇化因子FVIII所涉及製程步驟之流程圖。 Figure 6: A flow chart showing the process steps involved in the production of the sugar PEGylation factor FVIII in accordance with the present invention.

圖7:實例中產生之本發明之因子VIII分子的示意圖。 Figure 7: Schematic representation of the Factor VIII molecule of the invention produced in the Examples.

圖8:顯示可用於本發明實施例中之代表性具支鏈PEG聚合物,其在本文中稱為「SA-甘油-PEG」。 Figure 8: shows a representative branched PEG polymer useful in the examples of the invention, which is referred to herein as "SA-glycerol-PEG."

定義: definition:

因子VIII分子:FVIII/因子VIII係主要由肝細胞產生之大的複合糖蛋白。FVIII由2351個胺基酸組成,包括信號肽,且藉由同源性界定含有數種不同結構域。存在三種A結構域、一種B結構域、及兩種C結構域。結構域順序可列示為NH2-A1-A2-B-A3-C1-C2-COOH。FVIII以在B-A3邊界分開之兩條鏈形式在血漿中循環。該等鏈藉由二價金屬離子結合連接。A1-A2-B鏈稱為重鏈(HC),而A3-C1-C2稱為輕鏈(LC)。 Factor VIII Molecule: FVIII/Factor VIII is a large complex glycoprotein produced primarily by hepatocytes. FVIII consists of 2351 amino acids, including signal peptides, and is defined by homology to contain several different domains. There are three A domains, one B domain, and two C domains. The domain sequence can be listed as NH2-A1-A2-B-A3-C1-C2-COOH. FVIII circulates in plasma in the form of two strands separated at the B-A3 border. The chains are linked by a divalent metal ion. The A1-A2-B chain is referred to as the heavy chain (HC), and the A3-C1-C2 is referred to as the light chain (LC).

內源性因子VIII分子以具有不同大小B結構域之分子的集合體形式在活體內循環。在活體內很可能發生B結構域之漸進性酶促移除,導致具有不同大小B結構域之分子的集合體。通常認為,位置740處之裂解(藉由其移除B結構域之最後一部分)的發生與凝血酶激活有關。然而,此並不能排除例如位置740處之裂解位點已經損害之因子VIII變體可能具有活性。 The endogenous Factor VIII molecule circulates in vivo in the form of an aggregate of molecules having different size B domains. Progressive enzymatic removal of the B domain is likely to occur in vivo, resulting in aggregates of molecules with different size B domains. It is generally believed that the occurrence of cleavage at position 740 (by which the last portion of the B domain is removed) is associated with thrombin activation. However, this does not exclude that, for example, a Factor VIII variant that has been disrupted by the cleavage site at position 740 may be active.

本文所用之「因子VIII」或「FVIII」係指為內源性凝血途徑之成員且為凝血所必需之人類血漿糖蛋白。「天然FVIII」係顯示於SEQ ID NO.1(胺基酸1-2332)中之全長人類FVIII分子。B結構域跨越SEQ ID NO 1中之胺基酸741-1648。 As used herein, "Factor VIII" or "FVIII" refers to a human plasma glycoprotein that is a member of the endogenous coagulation pathway and is essential for coagulation. "Native FVIII" is a full length human FVIII molecule shown in SEQ ID NO. 1 (amino acid 1-2332). The B domain spans the amino acid 741-1648 in SEQ ID NO 1.

SEQ ID NO 1: SEQ ID NO 1:

本發明之因子VIII分子係B結構域截短型因子FVIII分子,其中剩餘結構域對應於SEQ ID NO.1中之1-740號及1649-2332號胺基酸中所 示之序列。因此,本發明分子係在轉化宿主細胞(較佳來源於哺乳動物)中產生之重組分子。然而,剩餘結構域(即三種A結構域及兩種C結構域)可與SEQ ID NO 1中所示之胺基酸序列(胺基酸1-740及1649-2332)略有不同,例如約1%、2%、3%、4%或5%。具體而言,在剩餘結構域中引入胺基酸修飾(取代、缺失等)來改良(例如)因子VIII與諸如vW因子、LPR、多種受體、其他凝血因子、細胞表面等多種其他部分的結合能力似乎是可行的。此外,本發明之因子VIII分子在例如截短之B結構域及/或該等分子之一或多種其他結構域中包含其他轉譯後修飾似乎亦是可行的。該等其他轉譯後修飾可呈與本發明之因子VIII分子共軛之多種分子形式,例如聚合化合物、肽化合物、脂肪酸衍生化合物等。 The Factor VIII molecule of the invention is a B domain truncated factor FVIII molecule, wherein the remaining domains correspond to amino acids 1-740 and 1649-2332 of SEQ ID NO. The sequence shown. Thus, the molecules of the invention are recombinant molecules produced in a transformed host cell, preferably derived from a mammal. However, the remaining domains (ie, the three A domains and the two C domains) may differ slightly from the amino acid sequences (amino acids 1-740 and 1649-2332) shown in SEQ ID NO: 1, for example, 1%, 2%, 3%, 4% or 5%. Specifically, amino acid modifications (substitutions, deletions, etc.) are introduced in the remaining domains to improve, for example, the binding of Factor VIII to various other components such as vW factors, LPR, multiple receptors, other coagulation factors, cell surfaces, and the like. The ability seems to be feasible. Furthermore, it may also be feasible for the Factor VIII molecules of the invention to include other post-translational modifications in, for example, a truncated B domain and/or one or more other domains of such molecules. Such other post-translational modifications may be in a variety of molecular forms that are conjugated to the Factor VIII molecules of the invention, such as polymeric compounds, peptide compounds, fatty acid-derived compounds, and the like.

本發明之因子VIII分子無論是否在B結構域外經修飾、是否具有其他轉譯後修飾皆具有因子VIII活性,意味著能夠在凝血級聯反應中以與FVIII功能類似或等效之方式起作用、經由與激活血小板上之FIXa相互作用誘導形成FXa、並支持形成血凝塊。該活性可在活體外藉由業內熟知技術進行評價,例如凝塊分析、內源性凝血酶潛能分析等。本發明之因子VIII分子所具有之FVIII活性佔天然人類FVIII之至少約10%、至少20%、至少30%、至少40%、至少50%、至少60%、至少70%、至少80%、至少90%、及100%或甚至100%以上。 The Factor VIII molecule of the present invention has Factor VIII activity whether modified in the B domain or whether it has other post-translational modifications, meaning that it can function in a similar or equivalent manner to FVIII function in the coagulation cascade. Interaction with FIXa on activated platelets induces the formation of FXa and supports the formation of blood clots. This activity can be assessed in vitro by techniques well known in the art, such as clot analysis, endogenous thrombin potential analysis, and the like. The Factor VIII molecule of the invention has at least about 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least FVIII activity of native human FVIII. 90%, and 100% or even 100% or more.

B結構域:因子VIII中之B結構域跨越SEQ ID NO 1中之胺基酸741-1648。B結構域在數個不同位點處裂解,使得循環血漿FVIII分子具有較大異質性。高度糖基化B結構域之確切功能尚未可知。吾人已知該結構域對於凝血級聯反應中之FVIII活性不是必要的。以下事實支持此明顯之功能缺乏:B結構域缺失型/截短型FVIII似乎具有與全長天然FVIII所見相同之活體內特性。儘管如此,有跡象表明B結構域可減少與細胞膜之連接,至少在無血清條件下。 B domain: The B domain in Factor VIII spans the amino acid 741-1648 in SEQ ID NO 1. The B domain is cleaved at several different sites, resulting in greater heterogeneity of circulating plasma FVIII molecules. The exact function of the highly glycosylated B domain is not known. It is known that this domain is not essential for FVIII activity in the coagulation cascade. The following facts support this apparent lack of function: B domain deleted/truncated FVIII appears to have the same in vivo characteristics as seen for full length native FVIII. Despite this, there are indications that the B domain can reduce the attachment to the cell membrane, at least in serum-free conditions.

B結構域截短型/缺失型因子VIII分子:所合成之內源性全長FVIII呈單鏈前體分子形式。在分泌之前,前體裂解成重鏈及輕鏈。 重組B結構域缺失型FVIII可由兩種不同策略產生。分別合成呈兩條不同多肽鏈形式之不含B結構域之重鏈及輕鏈(雙鏈策略)或合成呈單前體多肽鏈形式之B結構域缺失型FVIII,其以與全長FVIII前體相同之方式裂解成重鏈及輕鏈(單鏈策略)。 B-domain truncated/deleted factor VIII molecule: The endogenous full-length FVIII synthesized is in the form of a single-stranded precursor molecule. Prior to secretion, the precursor is cleaved into heavy and light chains. Recombinant B domain deleted FVIII can be produced by two different strategies. Synthesis of a B-domain-free heavy and light chain (double-stranded strategy) in the form of two different polypeptide chains, or a B-domain deleted FVIII in the form of a single precursor polypeptide chain, respectively, with a full-length FVIII precursor In the same way, it is cleaved into heavy and light chains (single chain strategy).

在B結構域缺失型FVIII前體多肽中,重鏈及輕鏈部分通常由連接體分開。為使引入B結構域缺失型FVIII中免疫原性抗原決定基之風險最小化,連接體序列較佳衍生自FVIII B結構域。連接體必須包含將B結構域缺失型FVIII前體多肽分成重鏈及輕鏈之蛋白酶識別位點。在全長FVIII之B結構域中,胺基酸1644-1648構成此識別位點。導致B結構域缺失型FVIII激活時連接體移除之凝血酶位點位於重鏈中。因此,連接體之大小及胺基酸序列不可能影響藉由凝血酶激活之其自剩餘FVIII分子之移除。B結構域缺失有利於產生FVIII。儘管如此,B結構域部分可包括於連接體中而不降低生產率。B結構域對生產率之負性效應並未歸因於具有任何特定大小或序列之B結構域。 In a B domain deleted FVIII precursor polypeptide, the heavy and light chain portions are typically separated by a linker. To minimize the risk of introducing an immunogenic epitope in a B domain deleted FVIII, the linker sequence is preferably derived from the FVIII B domain. The linker must comprise a protease recognition site that separates the B domain deleted FVIII precursor polypeptide into a heavy chain and a light chain. Amino acids 1644-1648 constitute this recognition site in the B domain of full length FVIII. The thrombin site that results in the removal of the linker upon activation of the B domain deleted FVIII is located in the heavy chain. Thus, the size of the linker and the amino acid sequence are unlikely to affect its removal from the remaining FVIII molecules by thrombin activation. Deletion of the B domain facilitates the production of FVIII. Nonetheless, the B domain portion can be included in the connector without reducing productivity. The negative effect of the B domain on productivity is not due to the B domain of any particular size or sequence.

截短之B結構域可含有數個O-糖基化位點。然而,根據一較佳實施例,該分子在截短之B結構域中包含僅一個、或者兩個、三個或四個O-連接寡糖。 The truncated B domain may contain several O-glycosylation sites. However, according to a preferred embodiment, the molecule comprises only one, or two, three or four O-linked oligosaccharides in the truncated B domain.

根據一較佳實施例,截短之B結構域包含僅一個潛在的O-糖基化位點且親水性聚合物與該O-糖基化位點共價共軛。 According to a preferred embodiment, the truncated B domain comprises only one potential O-glycosylation site and the hydrophilic polymer is covalently conjugated to the O-glycosylation site.

本發明B結構域截短型分子中之O-連接寡糖可附接至藉由重組手段及/或藉由截短B結構域暴露「隱藏」O-糖基化位點而人工產生之O-糖基化位點。在兩種情形下,該等分子均可藉由以下來產生:設計B結構域截短型因子VIII胺基酸序列並隨後使該胺基酸序列經受電腦(in silico)分析以預測截短之B結構域中O-糖基化位點之概率。可在適宜 宿主細胞中合成以相對高概率具有該等糖基化位點之分子,然後分析糖基化模式並隨後選拔在截短之B結構域中具有O-連接糖基化之分子。用於產生重組因子VIII蛋白質之適宜宿主細胞較佳來源於哺乳動物以確保分子被糖基化。在本發明實施中,細胞係哺乳動物細胞,更佳為已確立之哺乳動物細胞系,包括但不限於CHO(例如,ATCC CCL 61)、COS-1(例如,ATCC CRL 1650)、幼倉鼠腎(BHK)、及HEK293(例如,ATCC CRL 1573;Graham等人,J.Gen.Virol.36:59-72,1977)細胞系。較佳之BHK細胞系係tk-ts13 BHK細胞系(Waechter及Baserga,Proc.Natl.Acad.Sci.USA 79:1106-1110,1982),下文中稱為BHK 570細胞。BHK 570細胞系可以ATCC登錄號CRL 10314自美國典型培養物保藏中心(American Type Culture Collection),12301Parklawn Dr.,Rockville,MD 20852得到。tk-ts13 BHK細胞系亦可以登錄號CRL 1632自ATCC得到。較佳之CHO細胞系係可以登錄號CCI61自ATCC得到之CHO K1細胞系以及細胞系CHO-DXB11及CHO-DG44。 The O-linked oligosaccharide in the B domain truncated molecule of the invention can be attached to an O artificially produced by recombinant means and/or by exposing a "hidden" O-glycosylation site by truncating the B domain. - glycosylation site. In both cases, the molecules can be produced by designing a B domain truncated Factor VIII amino acid sequence and subsequently subjecting the amino acid sequence to in silico analysis to predict truncation Probability of O-glycosylation sites in the B domain. Can be suitable Molecules having such glycosylation sites are synthesized in host cells with relatively high probability, and then the glycosylation pattern is analyzed and subsequently the molecules having O-linked glycosylation in the truncated B domain are selected. Suitable host cells for the production of recombinant Factor VIII proteins are preferably derived from mammals to ensure that the molecule is glycosylated. In the practice of the invention, cell line mammalian cells, more preferably established mammalian cell lines, including but not limited to CHO (eg, ATCC CCL 61), COS-1 (eg, ATCC CRL 1650), baby hamster kidney (BHK), and HEK293 (eg, ATCC CRL 1573; Graham et al, J. Gen. Virol. 36: 59-72, 1977) cell lines. A preferred BHK cell line is the tk-ts13 BHK cell line (Waechter and Baserga, Proc. Natl. Acad. Sci. USA 79: 1106-1110, 1982), hereinafter referred to as BHK 570 cells. The BHK 570 cell line is available under the ATCC Accession No. CRL 10314 from the American Type Culture Collection, 12301 Parklawn Dr., Rockville, MD 20852. The tk-ts13 BHK cell line is also available from ATCC under accession number CRL 1632. A preferred CHO cell line is the CHO K1 cell line derived from CCC61 from ATCC and the cell lines CHO-DXB11 and CHO-DG44.

其他適宜細胞系包括但不限於大鼠Hep I(大鼠肝細胞瘤;ATCC CRL 1600)、大鼠Hep II(大鼠肝細胞瘤;ATCC CRL 1548)、TCMK(ATCC CCL 139)、人類肺(ATCC HB 8065)、NCTC 1469(ATCC CCL 9.1);DUKX細胞(CHO細胞系)(Urlaub及Chasin,Proc.Natl.Acad.Sci.USA 77:4216-4220,1980)(DUKX細胞亦稱為DXB11細胞)、及DG44(CHO細胞系)(Cell,33:405,1983;及Somatic Cell and Molecular Genetics 12:555,1986)。亦可使用3T3細胞、Namalwa細胞、骨髓瘤及骨髓瘤與其他細胞之融合物。在一些實施例中,細胞可為突變體或重組細胞,例如,與其衍生自之細胞類型相比,表現質量或數量不同之催化蛋白質轉譯後修飾之酶譜系(例如,諸如糖基轉移酶及/或糖苷酶等糖基化酶、或諸如前肽等加工酶)的細胞。DUKX細 胞(CHO細胞系)尤其佳。 Other suitable cell lines include, but are not limited to, rat Hep I (rat hepatoma; ATCC CRL 1600), rat Hep II (rat hepatoma; ATCC CRL 1548), TCMK (ATCC CCL 139), human lung ( ATCC HB 8065), NCTC 1469 (ATCC CCL 9.1); DUKX cells (CHO cell line) (Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77: 4216-4220, 1980) (DUKX cells are also known as DXB11 cells) ), and DG44 (CHO cell line) (Cell, 33: 405, 1983; and Somatic Cell and Molecular Genetics 12: 555, 1986). 3T3 cells, Namalwa cells, myeloma, and fusions of myeloma with other cells can also be used. In some embodiments, the cell can be a mutant or recombinant cell, eg, a zymogram line of a catalytic protein post-translational modification that exhibits a qualitative or quantitative difference in performance compared to the cell type from which it is derived (eg, such as a glycosyltransferase and/or Or a cell such as a glycosylase such as a glycosidase or a processing enzyme such as a propeptide. DUKX fine Cells (CHO cell lines) are particularly preferred.

當前較佳之細胞係HEK293、COS、中國倉鼠卵巢(CHO)細胞、幼倉鼠腎(BHK)及骨髓瘤細胞,中國倉鼠卵巢(CHO)細胞尤佳。 The currently preferred cell lines are HEK293, COS, Chinese hamster ovary (CHO) cells, baby hamster kidney (BHK) and myeloma cells, and Chinese hamster ovary (CHO) cells are particularly preferred.

因此,本發明之發明者顯示可藉由截短B結構域來激活因子VIII B結構域中之「隱藏」O-糖基化位點。儘管不欲受限於任何理論,但此現象可歸因於所改變之截短之B結構域中該分子之三級結構。由此使得「隱藏」O-糖基化位點在截短之B結構域中可以糖基化。此方法之一個優點係提供具有有利安全性質(例如,就變應原性而言)之重組分子。另一優點可為,其代表獲得B結構域中具有O-連接寡糖之B結構域截短變體之較簡單方法,此乃因B結構域中糖基化位點內在充足,先前證實在重組蛋白質中構造人工O-糖基化位點較為困難。 Thus, the inventors of the present invention have shown that a "hidden" O-glycosylation site in the Factor VIII B domain can be activated by truncating the B domain. Although not wishing to be bound by any theory, this phenomenon can be attributed to the tertiary structure of the molecule in the altered truncated B domain. This allows the "hidden" O-glycosylation site to be glycosylated in the truncated B domain. One advantage of this approach is to provide recombinant molecules with advantageous safety properties (e.g., in terms of allergenicity). Another advantage may be that it represents a relatively simple method of obtaining a B domain truncation variant with an O-linked oligosaccharide in the B domain, since the glycosylation site in the B domain is inherently sufficient, previously confirmed in It is difficult to construct artificial O-glycosylation sites in recombinant proteins.

wt FVIII分子中B結構域之長度為約907個胺基酸。本發明分子中截短之B結構域之長度可自約10個胺基酸至約700個胺基酸不等,例如約12-500個胺基酸、12-400個胺基酸、12-300個胺基酸、12-200個胺基酸、15-100個胺基酸、15-75個胺基酸、15-50個胺基酸、15-45個胺基酸、20-45個胺基酸、20-40個胺基酸、或20-30個胺基酸。該截短之B結構域可包含重鏈片段及/或輕鏈片段及/或未在wt FVIII分子中發現之人工引入序列。術語「B結構域截短」及「B結構域缺失」在本文中可互換使用。 The length of the B domain in the wt FVIII molecule is about 907 amino acids. The length of the truncated B domain of the molecules of the invention may vary from about 10 amino acids to about 700 amino acids, such as from about 12 to about 500 amino acids, from 12 to 400 amino acids, 12- 300 amino acids, 12-200 amino acids, 15-100 amino acids, 15-75 amino acids, 15-50 amino acids, 15-45 amino acids, 20-45 Amino acid, 20-40 amino acids, or 20-30 amino acids. The truncated B domain may comprise a heavy chain fragment and/or a light chain fragment and/or an artificially introduced sequence not found in the wt FVIII molecule. The terms "B domain truncation" and "B domain deletion" are used interchangeably herein.

改良之循環半衰期:與野生型因子VIII分子相比,本發明分子具有改良之循環半衰期,較佳延長之循環半衰期。循環半衰期較佳延長至少10%、較佳至少15%、較佳至少20%、較佳至少25%、較佳至少30%、較佳至少35%、較佳至少40%、較佳至少45%、較佳至少50%、較佳至少55%、較佳至少60%、較佳至少65%、較佳至少70%、較佳至少75%、較佳至少80%、較佳至少85%、較佳至少90%、較佳至少95%、較佳至少100%,更佳至少125%、更佳至少150%、更佳至少 175%、更佳至少200%,且最佳至少250%或300%。甚至更佳地,該等分子之循環半衰期相對於野生型FVIII之循環半衰期延長至少400%、500%、600%、或甚至700%。 Improved circulating half-life: The molecules of the invention have an improved circulating half-life, preferably a prolonged circulating half-life, compared to wild-type Factor VIII molecules. Preferably, the cycle half-life is extended by at least 10%, preferably at least 15%, preferably at least 20%, preferably at least 25%, preferably at least 30%, preferably at least 35%, preferably at least 40%, and preferably at least 45%. Preferably at least 50%, preferably at least 55%, preferably at least 60%, preferably at least 65%, preferably at least 70%, preferably at least 75%, preferably at least 80%, preferably at least 85%, more preferably Preferably at least 90%, preferably at least 95%, preferably at least 100%, more preferably at least 125%, more preferably at least 150%, more preferably at least 175%, more preferably at least 200%, and most preferably at least 250% or 300% . Even more preferably, the circulating half-life of the molecules is extended by at least 400%, 500%, 600%, or even 700% relative to the circulating half-life of wild-type FVIII.

親水性聚合物:本發明之修飾基團/親水性聚合物較佳非天然存在。在一個實例中,「非天然存在之修飾基團」係聚合物修飾基團,其中至少一種聚合物部分非天然存在。在另一實例中,非天然存在之修飾基團係經修飾之碳水化合物。選擇修飾基團官能化之位置以不妨礙「修飾糖」以酶促方式添加至多肽上。「修飾糖」亦係指經修飾基團官能化且為天然或修飾酶(例如糖基轉移酶)之受質的任何糖基模擬部分。 Hydrophilic polymer: The modifying group/hydrophilic polymer of the present invention is preferably non-naturally occurring. In one example, a "non-naturally occurring modifying group" is a polymeric modifying group in which at least one polymeric moiety is not naturally occurring. In another example, the non-naturally occurring modifying group is a modified carbohydrate. The position at which the modifying group is functionalized is selected so as not to prevent the "modified sugar" from being added to the polypeptide in an enzymatic manner. "Modified sugar" also refers to any glycosyl analog moiety that is functionalized with a modifying group and is a substrate of a native or modified enzyme (eg, a glycosyltransferase).

添加至多肽之聚合物修飾基團可改變該多肽之特性,例如,其生物可利用性、生物活性或其活體內半衰期。本發明之聚合物實例包括水溶性聚合物,其可為直鏈或具支鏈且可包括一或多種獨立選擇之聚合物部分,例如聚伸烷基二醇及其衍生物。本發明之聚合物修飾基團可包括水溶性聚合物,例如聚乙二醇及其衍生物(PEG、m-PEG)、聚丙二醇及其衍生物(PPG、m-PPG)及諸如此類。 The polymeric modifying group added to the polypeptide can alter the properties of the polypeptide, for example, its bioavailability, biological activity, or its in vivo half-life. Examples of polymers of the present invention include water soluble polymers which may be linear or branched and which may include one or more independently selected polymeric moieties such as polyalkylene glycols and derivatives thereof. The polymer modifying groups of the present invention may include water soluble polymers such as polyethylene glycol and its derivatives (PEG, m-PEG), polypropylene glycol and its derivatives (PPG, m-PPG), and the like.

術語「水溶性」係指在水中具有一定可檢測溶解度之部分。檢測及/或定量水溶解度之方法已為業內所熟知。本發明之實例性水溶性聚合物包括肽、糖類、聚醚、聚胺、聚羧酸及諸如此類。肽可具有混合序列且可由單一胺基酸構成,例如,聚離胺酸。多醣實例係聚唾液酸。聚醚實例係聚乙二醇,例如,m-PEG。聚胺實例為聚乙烯亞胺,且聚丙烯酸係代表性聚羧酸。 The term "water soluble" refers to a moiety that has a certain detectable solubility in water. Methods for detecting and/or quantifying water solubility are well known in the art. Exemplary water soluble polymers of the invention include peptides, saccharides, polyethers, polyamines, polycarboxylic acids, and the like. The peptide may have a mixed sequence and may be composed of a single amino acid, for example, a polylysine. An example of a polysaccharide is polysialic acid. An example of a polyether is polyethylene glycol, for example, m-PEG. An example of a polyamine is polyethyleneimine, and a polyacrylic acid is a representative polycarboxylic acid.

本發明水溶性聚合物之聚合物骨架可為聚乙二醇(即PEG)。與本發明有關之術語PEG包括呈任何形式之聚乙二醇,包括烷氧基PEG、二官能團PEG、多臂PEG、叉狀PEG、具支鏈PEG、有側基的PEG(即具有一或多個側接聚合物骨架之官能團的PEG或有關聚合物)、或其 中具有可降解鍵之PEG。 The polymer backbone of the water soluble polymer of the present invention may be polyethylene glycol (i.e., PEG). The term PEG in connection with the present invention includes polyethylene glycol in any form, including alkoxy PEG, difunctional PEG, multi-arm PEG, forked PEG, branched PEG, pendant PEG (ie having one or a plurality of PEG or related polymers flanked by functional groups of the polymer backbone, or a PEG having a degradable bond.

聚合物骨架可為直鏈或具支鏈。具支鏈聚合物骨架已為業內眾所周知。通常,具支鏈聚合物具有中心分支核心部分及複數個與該中心分支核心連接之聚合物直鏈。PEG常以具支鏈形式使用,其可藉由將氧化乙烯添加至諸如甘油、季戊四醇及山梨醇等多元醇中來製備。中心分支部分亦可衍生自諸如離胺酸或半胱胺酸等數種胺基酸。在一個實例中,具支鏈聚乙二醇可以通式R(-PEG-OH)m表示,其中R表示核心部分,例如甘油或季戊四醇,且m表示臂的個數。多臂PEG分子(例如彼等闡述於美國專利第5,932,462號中者,該專利之全部內容以引用方式併入本文中)亦可用作聚合物骨架。 The polymer backbone can be linear or branched. Branched polymer backbones are well known in the art. Typically, a branched polymer has a central branched core portion and a plurality of polymeric linear chains attached to the central branched core. PEG is often used in a branched form, which can be prepared by adding ethylene oxide to a polyol such as glycerin, pentaerythritol, and sorbitol. The central branch portion can also be derived from several amino acids such as lysine or cysteine. In one example, a branched polyethylene glycol can be represented by the formula R(-PEG-OH)m, wherein R represents a core moiety, such as glycerol or pentaerythritol, and m represents the number of arms. Multi-arm PEG molecules, such as those described in U.S. Pat.

圖8顯示可用於本發明實施例中之代表性具支鏈PEG聚合物,其在本文中稱為「SA-甘油-PEG」。圖8A顯示與聚糖或多肽之胺基酸連接之CMP-SA-甘油-PEG或SA-甘油-PEG的實例性SA-甘油-PEG組份。圖8B顯示透過Gal殘基與聚糖或多肽連接之SA-甘油-PEG部分。圖8C顯示透過Gal-GalNAc殘基與聚糖或多肽連接之SA-甘油-PEG部分。圖8D顯示透過Gal-GalNAc部分與多肽之胺基酸連接之SA-甘油-PEG部分。在多個實施例中,AA係蘇胺酸或絲胺酸。在一實例性實施例中,藉由刪除FVIII多肽之B結構域將AA轉化成O-連接糖基化位點。下文段落中關於聚合物分子量之論述普遍適用於圖8中所顯示之具支鏈PEG。在圖8中,下標「n」表示可提供如段落中所論述之具有期望分子量之直鏈(且因此具支鏈)m-PEG的任何整數。在多個實施例中,「n」經選擇以使直鏈m-PEG部分為約20KDa至約40KDa,例如約20KDa、約30KDa或約40KDa。對應於該等m-PEG分子量之整數對應於約400(例如約455)至約900(例如約910)。因此,「n」經選擇以提供為約40KDa至約80KDa(例如,約40KDa、約50KDa、約60KDa、約70KDa、或約80KDa)之具支鏈PEG。 Figure 8 shows a representative branched PEG polymer that can be used in embodiments of the invention, which is referred to herein as "SA-glycerol-PEG." Figure 8A shows an exemplary SA-glycerol-PEG component of CMP-SA-glycerol-PEG or SA-glycerol-PEG linked to the amino acid of a glycan or polypeptide. Figure 8B shows the SA-glycerol-PEG moiety attached to a glycan or polypeptide via a Gal residue. Figure 8C shows the SA-glycerol-PEG moiety attached to a glycan or polypeptide via a Gal-GalNAc residue. Figure 8D shows the SA-glycerol-PEG moiety attached to the amino acid of the polypeptide through the Gal-GalNAc moiety. In various embodiments, the AA is threonine or serine. In an exemplary embodiment, AA is converted to an O-linked glycosylation site by deletion of the B domain of the FVIII polypeptide. The discussion of polymer molecular weight in the following paragraphs is generally applicable to the branched PEG shown in Figure 8. In Figure 8, the subscript "n" indicates any integer that provides a linear (and therefore branched) m-PEG having the desired molecular weight as discussed in the paragraph. In various embodiments, "n" is selected such that the linear m-PEG moiety is from about 20 KDa to about 40 KDa, such as about 20 KDa, about 30 KDa, or about 40 KDa. An integer corresponding to the molecular weight of the m-PEGs corresponds to from about 400 (e.g., from about 455) to about 900 (e.g., about 910). Thus, "n" is selected to provide a branched PEG of from about 40 KDa to about 80 KDa (eg, about 40 KDa, about 50 KDa, about 60 KDa, about 70 KDa, or about 80 KDa).

許多其他聚合物亦適用於本發明。非肽及水溶性之聚合物骨架尤其可用於本發明。適宜聚合物之實例包括但不限於其他聚伸烷基二醇,例如聚丙二醇(「PPG」)、乙二醇與丙二醇及諸如此類之共聚物、聚(氧乙烯化多元醇)、聚(烯醇)、聚(乙烯吡咯啶酮)、聚(羥基丙基甲基丙烯醯胺)、聚([α]-羥酸)、聚(乙烯醇)、聚磷腈、聚噁唑啉、聚(N-丙烯醯基嗎啉)(例如闡述於美國專利第5,629,384號中者,該專利之全部內容以引用方式併入本文中)、以及共聚物、三元共聚物、及其混合物。 Many other polymers are also suitable for use in the present invention. Non-peptide and water soluble polymer backbones are especially useful in the present invention. Examples of suitable polymers include, but are not limited to, other polyalkylene glycols such as polypropylene glycol ("PPG"), copolymers of ethylene glycol and propylene glycol, and the like, poly(oxyethylated polyols), poly(enols) ), poly(vinylpyrrolidone), poly(hydroxypropylmethacrylamide), poly([α]-hydroxy acid), poly(vinyl alcohol), polyphosphazene, polyoxazoline, poly(N) - propylene decyl morpholine (for example, as described in U.S. Patent No. 5,629,384, the disclosure of which is incorporated herein in its entirety by reference in its entirety in its entirety)

儘管聚合物骨架之各鏈的分子量可能有所變化,但通常介於約100Da至約160,000Da範圍內,例如,約5,000Da至約100,000Da。更具體而言,本發明各共軛親水性聚合物之大小可自約500Da至約80,000Da不等,例如約1000Da至約80,000Da;約2000Da至約70,000Da;約5000至約70,000Da;約5000至約60,000Da;約10,000至約70,000Da;約20,000至約60,000Da;約30,000至約60,000Da;約30,000至約50,000Da;或約30,000至約40,000Da。應瞭解,該等大小代表估計值而非準確量測值。根據一較佳實施例,本發明分子與親水性聚合物異質群體共軛,例如,大小為例如10,000、40,000、或80,000Da +/-約5000、約4000、約3000、約2000、或約1000Da之PEG。 While the molecular weight of each chain of the polymer backbone may vary, it is typically in the range of from about 100 Da to about 160,000 Da, for example, from about 5,000 Da to about 100,000 Da. More specifically, the conjugated hydrophilic polymers of the present invention may range in size from about 500 Da to about 80,000 Da, such as from about 1000 Da to about 80,000 Da; from about 2000 Da to about 70,000 Da; from about 5,000 to about 70,000 Da; 5000 to about 60,000 Da; about 10,000 to about 70,000 Da; about 20,000 to about 60,000 Da; about 30,000 to about 60,000 Da; about 30,000 to about 50,000 Da; or about 30,000 to about 40,000 Da. It should be understood that these dimensions represent estimates rather than accurate measurements. According to a preferred embodiment, the molecules of the invention are conjugated to a heterogeneous population of hydrophilic polymers, for example, having a size of, for example, 10,000, 40,000, or 80,000 Da +/- about 5,000, about 4,000, about 3,000, about 2,000, or about 1000 Da. PEG.

O-連接寡糖:N-聚糖及O-聚糖二者均可藉由產生蛋白質之細胞附接至蛋白質上。當初生蛋白自核糖體易位至內質網時,細胞N-糖基化器件識別並糖基化胺基酸鏈中之N-糖基化信號(N-X-S/T基元)(Kiely等人,1976;Glabe等人,1980)。 O-linked oligosaccharides: Both N-glycans and O-glycans can be attached to proteins by the production of proteins. When the nascent protein translocates from the ribosome to the endoplasmic reticulum, the cellular N-glycosylation device recognizes and phosphorylates the N-glycosylation signal (NXS/T motif) in the amino acid chain (Kiely et al., 1976; Glabe et al., 1980).

同樣,可將O-聚糖附接至胺基酸鏈中之特定O-糖基化位點上,但觸發O-糖基化之基元較N-糖基化信號更為異質,且吾人預測胺基酸序列中之O-糖基化位點的能力尚不充分(Julenius等人,2004)。因 此,人工O-糖基化位點之構造具有一定不確定性。通常假定天然FVIII分子不含有任何O-糖基化位點,且熟習此項技術者因此預期需要構建至少一個人工O-糖基化位點並插入至與實施本發明有關之B結構域中。 Similarly, O-glycans can be attached to specific O-glycosylation sites in the amino acid chain, but the motif that triggers O-glycosylation is more heterogeneous than the N-glycosylation signal, and The ability to predict O-glycosylation sites in amino acid sequences is not sufficient (Julenius et al., 2004). because Thus, the construction of artificial O-glycosylation sites has some uncertainty. It is generally assumed that the native FVIII molecule does not contain any O-glycosylation sites, and those skilled in the art are therefore expected to construct at least one artificial O-glycosylation site and insert into the B domain associated with the practice of the invention.

因此,將截短型因子VIII B結構域中之O-連接寡糖共價連接至天然存在之O-連接糖基化序列或藉由重組技術人工構建之O-連接糖基化序列。 Thus, an O-linked oligosaccharide in the truncated Factor VIII B domain is covalently linked to a naturally occurring O-linked glycosylation sequence or an O-linked glycosylation sequence artificially constructed by recombinant techniques.

根據本發明之一較佳實施例,將O-連接寡糖連接至天然存在之O-連接糖基化序列,此O-連接糖基化序列在野生型因子VIII分子中不經受糖基化,但由於B結構域被截短而變得可經O-糖基化。其實例顯示於實例及SEQ ID NO 2中(截短之B結構域對應於胺基酸742-763)。 以下情形似乎可能:即使B結構域在稍微不同的地方被截短,即截短之B結構域與SEQ ID NO 2相比稍微較短(例如較SEQ ID NO 2短1、2、3、4、或5個胺基酸)或較長(例如1、2、3、4、5、6、7、8、9、10、15、20、25、30、35、40、45、或50個胺基酸),SEQ ID NO 2中之「隱藏」O-糖基化位點亦變為經糖基化。該方法藉由截短B結構域來激活「隱藏」O-糖基化位點而非產生人工O-糖基化位點,其具有產生具有有利安全性質(即變應原性降低等)之分子的優點。因子VIII B結構域中之其他O-糖基化位點同樣可藉由以不同方式截短分子來激活。 According to a preferred embodiment of the invention, the O-linked oligosaccharide is linked to a naturally occurring O-linked glycosylation sequence which is not subjected to glycosylation in the wild-type Factor VIII molecule, However, since the B domain is truncated, it becomes O-glycosylated. Examples thereof are shown in the examples and in SEQ ID NO 2 (the truncated B domain corresponds to the amino acid 742-763). It may seem possible that even if the B domain is truncated at a slightly different place, the truncated B domain is slightly shorter than SEQ ID NO 2 (eg shorter than SEQ ID NO 2 1, 2, 3, 4) Or 5 amino acids) or longer (eg 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50) The amino acid, the "hidden" O-glycosylation site in SEQ ID NO 2, also becomes glycosylated. The method activates a "hidden" O-glycosylation site by truncating the B domain rather than creating an artificial O-glycosylation site, which has the advantage of producing favorable safety properties (ie, reduced allergenicity, etc.) The advantages of molecules. Other O-glycosylation sites in the Factor VIII B domain can also be activated by truncating the molecule in different ways.

O-連接寡糖之糖聚乙二醇化:可改變O-聚糖之生物合成並利用在生物合成之相對早期添加唾液酸殘基來終止。某些唾液酸轉移酶能夠作用於GalNAcα-Ser/Thr、或早期O-聚糖核心亞型(在核心1 GalT作用後)。術語T抗原與Galβ1-3GalNAcα-Ser/Thr二糖之存在有關。該等結構之產生涉及糖基轉移酶之間對相同受質之競爭且因此高爾基體內糖基轉移酶之實際表現程度及亞細胞分佈決定O-聚糖生物合成及多樣 化之結構結果。如圖1中所示,僅Galβ1-3GalNAcα-Ser/Thr二糖適於糖聚乙二醇化。 O-linked oligosaccharide sugar PEGylation: The biosynthesis of O-glycans can be altered and terminated by the addition of sialic acid residues at a relatively early stage of biosynthesis. Certain sialyltransferases are capable of acting on GalNAcα-Ser/Thr, or early O-glycan core subtypes (after core 1 GalT action). The term T antigen is associated with the presence of Galβ1-3GalNAcα-Ser/Thr disaccharide. The production of such structures involves competition between the glycosyltransferases for the same substrate and thus the actual extent of glycosyltransferase activity in the Golgi and the subcellular distribution that determines the structural results of O-glycan biosynthesis and diversification. As shown in Figure 1, only Galβ1-3GalNAcα-Ser/Thr disaccharide is suitable for glycoPEGylation.

然而,通過用唾液酸酶或核心1 GalT或其組合來處理蛋白質可大大增加該結構之可得量。作為糖聚乙二醇化過程之結果,通過α3鍵結至目標蛋白之Galβ1-3GalNAcα-Ser/Thr二糖將唾液酸PEG添加至天然結構中(圖1)。 However, the availability of this structure can be greatly increased by treating the protein with sialidase or core 1 GalT or a combination thereof. As a result of the PEGylation process of the sugar, sialic acid PEG was added to the native structure by the Galβ1-3GalNAcα-Ser/Thr disaccharide bonded to the target protein by α3 (Fig. 1).

亦可將其他親水性聚合物附接至O-連接寡糖上。以酶促方式經由O-聚糖使其他親水性聚合物與FVIII共軛之基本要求係能夠如WO03031464中所揭示經由游離胺基使其偶聯至甘胺醯基-唾液酸衍生物。此可通過彼等熟習此項技術者所習知之多種偶聯化學法來達成。 具活性生物相容聚合物之實例包括聚氧化烯烴,例如但不限於聚乙二醇(PEG)、2-(甲基丙烯醯氧基)乙基磷醯膽鹼(mPC)聚合物(如WO03062290中所述)、右旋糖苷、多聚乙醯神經胺酸或其他基於碳水化合物之聚合物、胺基酸或特定肽序列之聚合物、生物素衍生物、聚乙烯醇(PVA)、聚羧酸酯、聚乙烯吡咯啶酮、聚乙烯-共-馬來酸酐、聚苯乙烯-共-蘋果酸酐、聚噁唑啉、聚丙烯醯基嗎啉、肝素、白蛋白、纖維素、殼聚糖水解產物、諸如羥基乙基澱粉及羥基丙基澱粉等澱粉、糖原、瓊脂糖及其衍生物、瓜爾豆膠、支鏈澱粉、菊糖、黃原膠、角叉菜膠、果膠、海藻酸水解產物、其他生物聚合物及其任何等效物。 Other hydrophilic polymers can also be attached to the O-linked oligosaccharides. The basic requirement for enzymatically conjugate other hydrophilic polymers to FVIII via O-glycans can be coupled to the glycidinyl-sialic acid derivative via a free amine group as disclosed in WO03031464. This can be achieved by a variety of coupling chemistries known to those skilled in the art. Examples of active biocompatible polymers include polyalkylene oxides such as, but not limited to, polyethylene glycol (PEG), 2-(methacryloxy)ethylphosphonium choline (mPC) polymers (eg, WO03062290) Said), dextran, polyacetamide, or other carbohydrate-based polymers, amino acids or polymers of specific peptide sequences, biotin derivatives, polyvinyl alcohol (PVA), polycarboxylates Acid ester, polyvinylpyrrolidone, polyethylene-co-maleic anhydride, polystyrene-co-malic anhydride, polyoxazoline, polypropylene decylmorpholine, heparin, albumin, cellulose, chitosan Hydrolysate, starch such as hydroxyethyl starch and hydroxypropyl starch, glycogen, agarose and its derivatives, guar gum, amylopectin, inulin, xanthan gum, carrageenan, pectin, Alginate hydrolysate, other biopolymers, and any equivalents thereof.

醫藥組合物:醫藥組合物在本文中較佳意欲涵蓋包含本發明因子VIIII分子之適於非經腸投與的組合物,例如即用型無菌水性組合物或可於例如水或水性緩衝液中重構之乾燥無菌組合物。本發明組合物可包含多種醫藥上可接受之賦形劑、穩定劑等。 Pharmaceutical Compositions: Pharmaceutical Compositions It is preferred herein to encompass compositions suitable for parenteral administration comprising a Factor VIIII molecule of the invention, such as a ready-to-use sterile aqueous composition or may be, for example, in water or an aqueous buffer. Reconstituted dry sterile composition. The compositions of the present invention may comprise a variety of pharmaceutically acceptable excipients, stabilizers and the like.

該等組合物中之額外成份可包括潤濕劑、乳化劑、抗氧化劑、增積劑、張力調節劑、螯合劑、金屬離子、油質媒劑、蛋白質(例 如,人類血清白蛋白、明膠或蛋白質)及兩性離子(例如,胺基酸,例如甜菜鹼、牛磺酸、精胺酸、甘胺酸、離胺酸及組胺酸)。當然,該等額外成份不應不利地影響本發明醫藥調配物之總體穩定性。非經腸投與可藉由藉助注射器(視情況鋼筆型注射器)皮下、肌內、腹膜內或靜脈內注射來實施。或者,非經腸投與可藉助輸注幫浦來實施。另一選擇係組合物可為適於呈鼻或肺噴霧形式投與FVIII化合物之溶液或懸浮液。作為又一選擇,含有本發明FVIII化合物之醫藥組合物亦可適於經皮投與(例如藉由無針注射或貼片,視情況為離子導入貼片)、或經黏膜(例如含服)投與。 Additional ingredients in such compositions may include wetting agents, emulsifying agents, antioxidants, accumulating agents, tonicity adjusting agents, chelating agents, metal ions, oily vehicles, proteins (eg, For example, human serum albumin, gelatin or protein) and zwitterions (eg, amino acids such as betaines, taurine, arginine, glycine, lysine, and histidine). Of course, such additional ingredients should not adversely affect the overall stability of the pharmaceutical formulations of the present invention. Parenteral administration can be carried out by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe (optional pen-type syringe). Alternatively, parenteral administration can be carried out by means of an infusion pump. Another selection system composition can be a solution or suspension suitable for administration of the FVIII compound in the form of a nasal or pulmonary spray. As a further alternative, a pharmaceutical composition comprising a compound of the invention FVIII may also be suitable for transdermal administration (for example by needle-free injection or patch, optionally as an iontophoresis patch), or via a mucosa (for example, containing a garment). Cast.

因此,在第一態樣中,本發明係關於具有改良之循環半衰期的B結構域截短型因子VIII分子,該分子經由截短之B結構域中之O-連接寡糖與親水性聚合物共價共軛,其中因子VIII激活(分子激活)導致共價共軛之親水性聚合物移除。 Thus, in a first aspect, the invention relates to a B domain truncated Factor VIII molecule having an improved circulating half-life via an O-linked oligosaccharide and a hydrophilic polymer in a truncated B domain Covalent conjugation in which factor VIII activation (molecular activation) results in the removal of a covalently conjugated hydrophilic polymer.

根據一個實施例,親水性聚合物係PEG。PEG聚合物之大小可自10,000至約160,000Da不等;例如10,000至80,000Da,例如約10,000;15,000;20,000;25,000;30,000;35,000;40,000;45,000;50,000;55,000;60,000;65,000;70,000;75,000;或80,000Da。較佳地,將O-連接寡糖附接至藉由截短B結構域而非藉由插入未在wt FVIII分子中發現之人工O-糖基化位點而產生之O-糖基化位點上。 According to one embodiment, the hydrophilic polymer is PEG. The size of the PEG polymer can vary from 10,000 to about 160,000 Da; for example, 10,000 to 80,000 Da, such as about 10,000; 15,000; 20,000; 25,000; 30,000; 35,000; 40,000; 45,000; 50,000; 55,000; 60,000; 65,000; 70,000; Or 80,000 Da. Preferably, the O-linked oligosaccharide is attached to the O-glycosylation site produced by truncating the B domain rather than by insertion of an artificial O-glycosylation site not found in the wt FVIII molecule. Point.

根據一尤佳實施例,本發明分子包含如SEQ ID NO 2中所示之胺基酸序列。該等分子具有激活之FVIII分子與天然活性FVIII分子相同的特徵。該特徵在安全評價中似乎具有有利特性。 According to a particularly preferred embodiment, the molecule of the invention comprises an amino acid sequence as set forth in SEQ ID NO 2. These molecules have the same characteristics that the activated FVIII molecule is identical to the naturally active FVIII molecule. This feature seems to have advantageous characteristics in the safety evaluation.

本發明亦係關於包含本發明分子之醫藥組合物。 The invention also relates to pharmaceutical compositions comprising the molecules of the invention.

本發明進一步係關於獲得本發明分子之方法,其中該方法包含使B結構域截短型因子VIII分子與諸如PEG基團等親水性聚合物經由 截短之B結構域中之O-連接寡糖共軛。因此,本發明亦係關於藉由該等方法獲得或可藉由該等方法獲得之分子。 The invention further relates to a method of obtaining a molecule of the invention, wherein the method comprises passing a B domain truncated Factor VIII molecule to a hydrophilic polymer such as a PEG group via The O-linked oligosaccharide is conjugated in the truncated B domain. Accordingly, the invention also relates to molecules obtained by or by such methods.

在另一態樣中,本發明係關於治療血友病性疾病之方法,其包含向有需要之患者投與治療有效量之本發明分子。 In another aspect, the invention relates to a method of treating a hemophilia disorder comprising administering to a patient in need thereof a therapeutically effective amount of a molecule of the invention.

本文所用之術語「治療」係指對有需要之任何人類或其他動物個體實施醫學治療。期望該個體已由從業醫師實施身體檢查,該從業醫師給出嘗試性或確定性診斷,該診斷指出使用該特定治療有益於該人類或其他動物個體之健康。該治療之時間選擇及目的可根據個體體質現狀因人而異。因此,該治療可為預防治療、姑息治療、對症治療及/或治癒性治療。 The term "treatment" as used herein refers to the medical treatment of any human or other animal individual in need thereof. It is expected that the individual has performed a physical examination by a medical practitioner who gives a tentative or definitive diagnosis indicating that the use of the particular treatment is beneficial to the health of the human or other animal individual. The timing and purpose of the treatment may vary from person to person depending on the current state of the individual. Thus, the treatment can be prophylactic, palliative, symptomatic, and/or curative.

在再一態樣中,本發明係關於本發明分子作為藥劑之用途以及本發明分子用以製造用於治療血友病之藥劑的用途。 In still another aspect, the invention relates to the use of a molecule of the invention as a medicament and to the use of a molecule of the invention for the manufacture of a medicament for the treatment of hemophilia.

在最後態樣中,本發明係關於構造本發明之B結構域截短型因子VIII分子之方法,該方法包含:(i)截短B結構域並視情況對該截短型因子VIII分子之胺基酸序列實施分析以識別潛在的O-連接糖基化位點;(ii)在適宜宿主細胞中產生該分子;及(iii)選拔在截短之B結構域中具有O-連接聚糖之分子。 In a final aspect, the invention relates to a method of constructing a B domain truncated Factor VIII molecule of the invention, the method comprising: (i) truncating the B domain and optionally the truncated Factor VIII molecule Analysis of the amino acid sequence to identify potential O-linked glycosylation sites; (ii) production of the molecule in a suitable host cell; and (iii) selection of O-linked glycans in the truncated B domain The molecule.

實例Instance 實例1Example 1 產生重組B結構域截短型O-糖基化因子VIII Production of recombinant B domain truncated O-glycosylation factor VIII

在SEQ ID NO 2中給出B結構域缺失型因子VIII分子之胺基酸序列的實例。此多肽亦可稱為「N8」。該分子包含21胺基酸殘基連接體序列(SFSQNSRHPSQNPPVLKRHQR-加以下劃線之S係絲胺酸殘基,其具有在實例2中經聚乙二醇化之O-聚糖)。 An example of an amino acid sequence of a B domain deleted Factor VIII molecule is given in SEQ ID NO 2. This polypeptide may also be referred to as "N8". This molecule contains the 21 amino acid residue linker sequence (SFSQNSRHP S QNPPVLKRHQR - underlined S- line serine residue having the PEGylated O-glycan in Example 2).

本發明之因子VIII分子在實例中可以多種方式提及-但因子VIII分子之所有提及皆係指本發明之因子VIII分子、或另一選擇為在轉化成 本發明因子VIII分子過程中之因子VIII分子。 The Factor VIII molecule of the present invention may be mentioned in various ways in the examples - but all references to the Factor VIII molecule refer to the Factor VIII molecule of the present invention, or alternatively selected to be converted into A Factor VIII molecule in the process of the Factor VIII molecule of the invention.

SEQ ID NO 2: SEQ ID NO 2:

細胞系及培養方法: Cell line and culture method:

使用因子VIII cDNA來構建編碼具有如SEQ ID NO 2中所示胺基酸序列之B結構域缺失型因子VIII的哺乳動物表現質粒。該質粒編碼包含全長人類因子VIII之胺基酸1-740的因子VIII重鏈及包含全長人類因子VIII之胺基酸1649-2332的因子VIII輕鏈。藉由21胺基酸連接體將該等重鏈及輕鏈序列與全長人類因子VIII之胺基酸741-750及1638-1648序列連接。將中國倉鼠卵巢(CHO)細胞用BDD因子VIII編碼質粒轉染並利用二氫葉酸還原酶系統進行選拔,最終產生在無動物組份培養基中培養之純系懸浮液生產細胞。 A mammalian expression plasmid encoding a B domain deleted type Factor VIII having the amino acid sequence as shown in SEQ ID NO 2 was constructed using Factor VIII cDNA. This plasmid encodes a Factor VIII heavy chain comprising amino acid 1-740 of full length human Factor VIII and a Factor VIII light chain comprising amino acid 1649-2332 of full length human Factor VIII. The heavy and light chain sequences are ligated to the amino acid 741-750 and 1638-1648 sequences of full length human Factor VIII by a 21 amino acid linker. Chinese hamster ovary (CHO) cells were transfected with the BDD Factor VIII-encoding plasmid and selected using the dihydrofolate reductase system to finally produce pure suspension production cells cultured in animal-free medium.

此方法之第一步驟係接種細胞小瓶,自工作細胞庫小瓶至化學限定及無動物組份生長培養基中。解凍後,先將細胞在T-燒瓶中進行培育。解凍後一天或兩天,將細胞轉移至搖瓶中,並藉由連續稀釋擴展培養物體積以使細胞密度保持在0.2-3.0×106個細胞/ml之間。下一步驟為將搖瓶培養物轉移至種子生物反應器中。此時再次擴展培養物體積,隨後最終轉移至生產生物反應器中。所有接種物擴展步驟皆使用相同的化學限定及無動物組份培養基。在轉移至生產生物反應器中之後,向培養基中補充能夠增大產物濃度之組份。在生產生物反應器中,以重複分批方法以三天為週期培養細胞。收穫時,將80-90%培養物體積轉移至收穫槽中。隨後用新鮮培養基稀釋剩餘培養液以達到 初始細胞密度,並隨後開始新的生長期。 The first step of the method is to inoculate a cell vial from a working cell bank vial to a chemically defined and animal free growth medium. After thawing, the cells were first incubated in a T-flask. One day or two after thawing, the cells were transferred to shake flasks and expanded by serial dilution to make the volume of the culture cell density was maintained between 0.2-3.0 × 10 6 cells / ml. The next step is to transfer the shake flask culture to the seed bioreactor. At this point the culture volume is expanded again and then finally transferred to the production bioreactor. The same chemically defined and animal-free medium was used for all inoculum expansion steps. After being transferred to the production bioreactor, the medium is supplemented with a component capable of increasing the concentration of the product. In a production bioreactor, cells were cultured in a three-day cycle in a repeated batch process. At harvest, 80-90% of the culture volume was transferred to the harvesting tank. The remaining medium is then diluted with fresh medium to achieve initial cell density and then a new growth phase begins.

藉由離心及過濾來淨化收穫批料並轉移至儲存槽中,隨後開始純化過程。將緩衝液添加至儲存槽中之游離細胞收穫物中以穩定pH值。 The harvest batch is purified by centrifugation and filtration and transferred to a storage tank, after which the purification process begins. The buffer is added to the free cell harvest in the storage tank to stabilize the pH.

到生產運行結束時,收集細胞並冷凍以終了生產細胞庫。測試該細胞庫之黴漿菌、無菌度及病毒污染。 At the end of the production run, the cells are collected and frozen to end the production cell bank. The cell bank was tested for mycoplasma, sterility and virus contamination.

純化:使用包括以下之四步驟純化程序,自細胞培養基分離B結構域缺失型因子VIII:在Capto MMC管柱上之濃縮步驟、免疫吸附層析步驟、陰離子交換層析及最後的凝膠過濾步驟。通常使用以下程序:將11公升無菌過濾培養基抽吸至Capto MMC管柱(GE Healthcare,Sweden)(1.6×12cm)上,該管柱已經過15ml/min流速之緩衝液A(20mM咪唑、10mM CaCl2、50mM NaCl、0.02% Tween 80,pH=7.5)平衡。將管柱用75ml緩衝液A洗滌,隨後用含有1.5M NaCl之75ml緩衝液A洗滌。用20mM咪唑、10mM CaCl2、0.02% Tween 80、2.5M NaCl、8M乙二醇(pH=7.5)以1ml/min流速來溶離蛋白質。收集8ml溶離份並分析其因子VIII活性(CoA-測試)。集中含有因子VIII之溶離份且通常獲得約50ml之集合體積。 Purification: Separation of B domain-deficient Factor VIII from cell culture media using a four-step purification procedure consisting of a concentration step on a Capto MMC column, an immunoadsorption chromatography step, an anion exchange chromatography, and a final gel filtration step . The following procedure is typically used: 11 liters of sterile filter medium is pumped onto a Capto MMC column (GE Healthcare, Sweden) (1.6 x 12 cm) which has been subjected to a flow rate of 15 ml/min of buffer A (20 mM imidazole, 10 mM CaCl). 2 , 50 mM NaCl, 0.02% Tween 80, pH = 7.5) equilibrated. The column was washed with 75 ml of buffer A, followed by washing with 75 ml of buffer A containing 1.5 M NaCl. The protein was eluted with 20 mM imidazole, 10 mM CaCl 2 , 0.02% Tween 80, 2.5 M NaCl, 8 M ethylene glycol (pH = 7.5) at a flow rate of 1 ml/min. 8 ml of the fraction was collected and analyzed for factor VIII activity (CoA-test). Concentrations containing Factor VIII are concentrated and typically achieve an aggregate volume of approximately 50 ml.

已研發出對抗因子VIII之單株抗體(Kjalke Eur J Biochem 234 773)。已由抗原決定基之圖譜分析(結果未顯示)發現,此抗體F25能從胺基酸殘基725至740中識別出重鏈的之遠C-端序列。基本上如製造商所述將此F25抗體以2.4mg/ml凝膠之密度偶聯至NHS激活瓊脂糖4FF(GE Healthcare,Bio-Sciences AB,Uppsala,Sweden)上。將來自先前步驟之集合液用20mM咪唑、10mM CaCl2、0.02% Tween 80(pH=7.3)稀釋10倍並施加至F25瓊脂糖管柱(1.6×9.5cm)上,該管柱已用0.5ml/min流速之20mM咪唑、10mM CaCl2、150mM NaCl、0.02% Tween 80、1M甘油(pH=7.3)平衡。用平衡緩衝液洗滌管柱,直至UV信號穩定,並隨後用20mM咪唑、10mM CaCl2、0.65M NaCl(pH=7.3)洗滌管柱,直至UV信號再次穩定。用20mM咪唑、10mM CaCl2、0.02% Tween 80、2.5M NaCl、50%乙二醇(pH=7.3)以1ml/min流速來溶離因子VIII。收集1ml溶離份並分析其因子VIII活性(CoA-測試)。集中含有因子VIII之溶離份且通常獲得約25ml之集合體積。 A monoclonal antibody against Factor VIII has been developed (Kjalke Eur J Biochem 234 773). A map analysis of the epitope (not shown) has been found to recognize that the antibody F25 recognizes the far C-terminal sequence of the heavy chain from amino acid residues 725 to 740. This F25 antibody was coupled to NHS activated agarose 4FF (GE Healthcare, Bio-Sciences AB, Uppsala, Sweden) at a density of 2.4 mg/ml gel essentially as described by the manufacturer. The pool from the previous step was diluted 10-fold with 20 mM imidazole, 10 mM CaCl 2 , 0.02% Tween 80 (pH = 7.3) and applied to a F25 agarose column (1.6 x 9.5 cm) which had been used with 0.5 ml. The /min flow rate was equilibrated with 20 mM imidazole, 10 mM CaCl 2 , 150 mM NaCl, 0.02% Tween 80, 1 M glycerol (pH = 7.3). The column was washed with equilibration buffer until the UV signal was stable, and then the column was washed with 20 mM imidazole, 10 mM CaCl 2 , 0.65 M NaCl (pH = 7.3) until the UV signal stabilized again. Factor VIII was eluted with 20 mM imidazole, 10 mM CaCl 2 , 0.02% Tween 80, 2.5 M NaCl, 50% ethylene glycol (pH = 7.3) at a flow rate of 1 ml/min. 1 ml of the fraction was collected and analyzed for factor VIII activity (CoA-test). Concentrations containing Factor VIII are concentrated and typically achieve an aggregate volume of about 25 ml.

製備供離子交換步驟用之緩衝液A(20mM咪唑、10mM CaCl2、0.02% Tween 80、1M甘油,pH=7.3)及緩衝液B(20mM咪唑、10mM CaCl2、0.02% Tween 80、1M甘油、1M NaCl,pH=7.3)。用85%緩衝液A/15%緩衝液B以2ml/min流速平衡Macro-Prep 25Q Support管柱(1×10cm)(Bio-Rad Laboratories,Hercules,CA,USA)。將來自先前步驟之集合液用緩衝液A稀釋10倍,並以2ml/min流速抽吸至管柱上。用85%緩衝液A/15%緩衝液B以2ml/min流速洗滌管柱並用自15%緩衝液B至70%緩衝液B之線性梯度,以2ml/min流速經120ml來溶離因子VIII。收集2ml溶離份並分析其因子VIII活性(CoA-測試)。集中含有因子VIII之溶離份且通常獲得約36ml之集合液體積。 Prepare buffer A for the ion exchange step (20 mM imidazole, 10 mM CaCl 2 , 0.02% Tween 80, 1 M glycerol, pH = 7.3) and buffer B (20 mM imidazole, 10 mM CaCl 2 , 0.02% Tween 80, 1 M glycerol, 1 M NaCl, pH = 7.3). The Macro-Prep 25Q Support column (1 x 10 cm) (Bio-Rad Laboratories, Hercules, CA, USA) was equilibrated with 85% buffer A/15% buffer B at a flow rate of 2 ml/min. The pool from the previous step was diluted 10-fold with buffer A and pumped onto the column at a flow rate of 2 ml/min. The column was washed with 85% buffer A/15% buffer B at a flow rate of 2 ml/min and factor VIII was eluted with 120 ml at a flow rate of 2 ml/min using a linear gradient from 15% buffer B to 70% buffer B. 2 ml of the fraction was collected and analyzed for Factor VIII activity (CoA-test). Concentrations containing Factor VIII are concentrated and typically a volume of pool of approximately 36 ml is obtained.

將來自先前步驟之集合液施加至Superdex 200製備級(GE Healthcare,Bio-Sciences AB,Uppsala,Sweden)管柱(2.6×60cm)中,用20mM咪唑、10mM CaCl2、0.02% Tween 80、1M甘油、150mM NaCl(pH=7.3)以1ml/min平衡及溶離該管柱。收集3ml溶離份並分析其因子VIII活性(CoA-測試)。集中含有因子VIII之溶離份且通常獲得約57ml之集合液體積。將含有因子VIII之集合液於-80℃下儲存。 The pool from the previous step was applied to a Superdex 200 preparative (GE Healthcare, Bio-Sciences AB, Uppsala, Sweden) column (2.6 x 60 cm) with 20 mM imidazole, 10 mM CaCl 2 , 0.02% Tween 80, 1 M glycerol. 150 mM NaCl (pH = 7.3) was equilibrated and dissolved at 1 ml/min. 3 ml of the fraction was collected and analyzed for Factor VIII activity (CoA-test). Concentrations containing Factor VIII are concentrated and typically a volume of pool of approximately 57 ml is obtained. The pool containing Factor VIII was stored at -80 °C.

藉由CoA活性及ELISA量測所判斷,使用上述四步驟純化程序獲得約15%之總產率。 The overall yield of about 15% was obtained using the four-step purification procedure described above as judged by CoA activity and ELISA measurements.

用於製造N8之細胞系係重組中國倉鼠卵巢(CHO)細胞系,其經過 由pTSV7表現載體與含有編碼F8-500蛋白質之cDNA的插入物組成之pTSV7中的表現質粒#814 F8-500穩定轉染。「N8」在本文中意欲對應於具有如SEQ ID NO 2中所列示之胺基酸序列的蛋白質。自N-端開始,F8-500蛋白質(N8)由FVIII信號肽(胺基酸-19至-1)、繼之無B結構域之FVIII重鏈(胺基酸1-740)、21胺基酸連接體(SFSQNSRHPSQNPPVLKRHQR)、及FVIII輕鏈(野生型人類FVIII之胺基酸1649-2332)組成。21胺基酸連接體序列係衍生自FVIII B結構域且由全長FVIII之胺基酸741-750及1638-1648組成。 Cell line used to make N8 recombinant Chinese hamster ovary (CHO) cell line The expression plasmid #814 F8-500 in pTSV7 consisting of the pTSV7 expression vector and the insert containing the cDNA encoding the F8-500 protein was stably transfected. "N8" is intended herein to correspond to a protein having an amino acid sequence as set forth in SEQ ID NO 2. Starting from the N-terminus, the F8-500 protein (N8) consists of the FVIII signal peptide (amino acid-19 to -1), followed by the B-free FVIII heavy chain (amino acid 1-740), 21 amino group. The acid linker (SFSQNSRHPSQNPPVLKRHQR) and the FVIII light chain (amino acid 1649-2332 of wild type human FVIII) are composed. The 21 amino acid linker sequence is derived from the FVIII B domain and consists of the amino acids 741-750 and 1638-1648 of full length FVIII.

用pTSV7中之814 F8-500轉染CHO細胞並利用二氫葉酸還原酶系統進行選拔,最終產生在無動物組份培養基中培養之純系懸浮液生產細胞。藉由解凍工作細胞庫小瓶並在轉移至生產生物反應器中之前擴展細胞來開始生產運行。所有接種物擴展步驟皆使用相同的化學限定及無動物組份培養基。在轉移至生產生物反應器中之後,向培養基中補充能夠增大產物濃度之組份。在生產生物反應器中,以重複分批方法以三天為週期培養細胞。收穫時,將80-90%培養物體積轉移至收穫槽中。隨後用新鮮培養基稀釋剩餘培養液以達到初始細胞密度,並隨後開始新的生長期。藉由離心及過濾來淨化收穫批料並將其轉移至儲存槽中,隨後開始純化過程。將緩衝液添加至儲存槽中之游離細胞收穫物中以穩定pH值。 CHO cells were transfected with 814 F8-500 in pTSV7 and selected using a dihydrofolate reductase system to ultimately produce pure suspension production cells cultured in animal-free medium. Production runs are initiated by thawing the working cell bank vials and expanding the cells prior to transfer to the production bioreactor. The same chemically defined and animal-free medium was used for all inoculum expansion steps. After being transferred to the production bioreactor, the medium is supplemented with a component capable of increasing the concentration of the product. In a production bioreactor, cells were cultured in a three-day cycle in a repeated batch process. At harvest, 80-90% of the culture volume was transferred to the harvesting tank. The remaining medium is then diluted with fresh medium to achieve initial cell density and then a new growth phase begins. The harvest batch is purified by centrifugation and filtration and transferred to a storage tank, after which the purification process begins. The buffer is added to the free cell harvest in the storage tank to stabilize the pH.

實例2Example 2 重組B結構域截短型及O-糖基化之因子VIII之聚乙二醇化: PEGylation of recombinant B domain truncated and O-glycosylated factor VIII:

利用以下程序使實例1中獲得之重組因子VIII分子與聚乙二醇(PEG)共軛。 The recombinant Factor VIII molecule obtained in Example 1 was conjugated to polyethylene glycol (PEG) using the following procedure.

為使實例1中獲得之重組因子VIII分子有效地糖聚乙二醇化,大於5mg/ml之FVIII濃度較佳。由於FVIII在此濃度下通常不溶解,故對所選緩衝液組合物實施篩選(一些該等結果參見表1)。 In order to efficiently PEGylate the recombinant Factor VIII molecule obtained in Example 1, a concentration of FVIII greater than 5 mg/ml is preferred. Since FVIII is generally insoluble at this concentration, screening is performed on the selected buffer composition (some of these results are shown in Table 1).

基於該等考慮,吾人發現含有50mM MES、50mM CaCl2、150mM NaCl、20%甘油(pH 6.0)之緩衝液係適宜反應緩衝液。 Based on these considerations, we have found that a buffer containing 50 mM MES, 50 mM CaCl 2 , 150 mM NaCl, 20% glycerol (pH 6.0) is a suitable reaction buffer.

將經如上所述純化之重組FVIII在反應緩衝液中藉由在Poros 50 HQ管柱上利用分步溶離、在Sartorius Vivaspin(PES)過濾器上(以10kDa為截斷值)或在Amicon 10kDa MWCO PES過濾器上實施離子交換來濃縮至6-10mg/mL濃度。藉由將因子VIII(BDD)(最終約4.7mg/mL)與唾液酸酶(產脲節桿菌(A.ureafaciens))(159mU/mL)、CMP-SA-甘油-PEG-40kDa(5mol.eq.)及MBP-ST3Gal1(540mU)混合於反應緩衝液(50mM MES、50mM CaCl2、150mM NaCl、20%甘油、0.5mM抗蛋白酶素,pH 6.0)中來開始對FVIII實施糖聚乙二醇化。在32℃下培育反應混合物,直至總轉化產率為約20-30%。 Recombinant FVIII purified as described above in a reaction buffer by stepwise dissolution on a Poros 50 HQ column, on a Sartorius Vivaspin (PES) filter (with a cutoff value of 10 kDa) or at Amicon 10kDa MWCO PES Ion exchange was performed on the filter to concentrate to a concentration of 6-10 mg/mL. By factor VIII (BDD) (final about 4.7 mg/mL) with sialidase (A. ureafaciens) (159 mU/mL), CMP-SA-glycerol-PEG-40kDa (5 mol. eq) .) and MBP-ST3Gal1 (540 mU) were mixed in reaction buffer (50 mM MES, 50 mM CaCl 2 , 150 mM NaCl, 20% glycerol, 0.5 mM anti-protease, pH 6.0) to initiate sugar pegylation of FVIII. The reaction mixture was incubated at 32 ° C until the total conversion yield was about 20-30%.

培育後,將樣品用緩衝液A(25mM Tris、5mM CaCl2、20mM NaCl、20%甘油,pH 7.5)稀釋並施加至Source 15Q管柱(1cm內徑x 6cm,4.7mL,1mL/min,280nm)上。將結合物質用緩衝液A洗滌並使用分步梯度用緩衝液B(25mM Tris、5mM CaCl2、1M NaCl、20%甘油,pH 7.5)實施溶離。在約25%緩衝液B下自管柱中溶離出經糖聚 乙二醇化之因子VIII-(O)-SA-甘油-PEG-40kDa。圖2顯示反應混合物在Source 15Q上之離子交換層析。 After incubation, the samples were diluted with buffer A (25 mM Tris, 5 mM CaCl 2 , 20 mM NaCl, 20% glycerol, pH 7.5) and applied to a Source 15Q column (1 cm id x 6 cm, 4.7 mL, 1 mL/min, 280 nm) )on. The binding substance was washed with buffer A and eluted with buffer B (25 mM Tris, 5 mM CaCl 2 , 1 M NaCl, 20% glycerol, pH 7.5) using a step gradient. The sugar PEGylated Factor VIII-(O)-SA-glycerol-PEG-40kDa was eluted from the column at about 25% buffer B. Figure 2 shows ion exchange chromatography of the reaction mixture on Source 15Q.

為封端唾液酸酶處理期間暴露於N-聚糖之游離半乳糖部分,將因子VIII-SA-甘油-PEG-40kDa(最終為1.0mg/mL)之集合溶離份與CMP-SA(2,000mol eq)及MBP-SBD-ST3Gal3(400mU/mL)混合於反應緩衝液(50mM MES、20mM CaCl2、150mM NaCl、10mM MnCl2、20%甘油,pH 6.0)中並在32℃下培育11小時。 For the exposure to the free galactose moiety of the N-glycan during the capsidylase treatment, the pooled fraction of Factor VIII-SA-glycerol-PEG-40kDa (final 1.0 mg/mL) was combined with CMP-SA (2,000 mol) Eq) and MBP-SBD-ST3Gal3 (400 mU/mL) were mixed in reaction buffer (50 mM MES, 20 mM CaCl 2 , 150 mM NaCl, 10 mM MnCl 2 , 20% glycerol, pH 6.0) and incubated at 32 ° C for 11 hours.

在Superdex 200管柱(10cm內徑×300mm;280nm)上藉由凝膠過濾將所得封端之經糖聚乙二醇化因子VIII-SA-甘油-PEG-40kDa與CMP-SA及ST3GalIII分開,該管柱已用50mM MES、50mM CaCl2、150mM NaCl、10%甘油(pH 6.0)以0.25mL/min流速平衡。產物因子VIII-SA-甘油-PEG-40kDa在38min時溶離出來。圖3顯示使用Superdex 200尺寸排除層析對封端產物實施純化。收集峰溶離份,等分並隨後實施分析。 The resulting blocked glycated PEGylated Factor VIII-SA-glycerol-PEG-40kDa was separated from CMP-SA and ST3GalIII by gel filtration on a Superdex 200 column (10 cm inner diameter x 300 mm; 280 nm). The column has been equilibrated with 50 mM MES, 50 mM CaCl 2 , 150 mM NaCl, 10% glycerol (pH 6.0) at a flow rate of 0.25 mL/min. The product factor VIII-SA-glycerol-PEG-40kDa eluted at 38 min. Figure 3 shows the purification of the capped product using Superdex 200 size exclusion chromatography. The peak fractions were collected, aliquoted and subsequently analyzed.

封端程序之目的係降低共軛因子VIII分子之活體內清除率。 The purpose of the capping procedure is to reduce the in vivo clearance of the conjugated Factor VIII molecule.

實例3Example 3 在產色FVIII活性分析中O-聚糖聚乙二醇化rFVIII之活性: O-glycan PEGylated rFVIII activity in chromogenic FVIII activity assay:

在產色FVIII分析中使用Coatest SP試劑(Chromogenix)如下對實例2中獲得之O-糖聚乙二醇化rFVIII的活性實施評價:將rFVIII樣品及校準物(來自NIBSC之第7國際FVIII標準物)稀釋於Coatest分析緩衝液(50mM Tris、150mM NaCl、1% BSA,pH 7.3,含有防腐劑)中。將50μl樣品、標準物及緩衝液陰性對照一式兩份添加至96孔微量滴定板(Nunc)中。將來自Coatest SP套組之因子IXa/因子X試劑、磷脂試劑及CaCl2以5:1:3(vol:vol:vol)混合並將75μl此物質添加至孔中。在於室溫下培育15min後,添加50μl因子Xa受質S-2765/凝血酶抑制劑I-2581混合物並在室溫下將反應物培育10min,隨後添加25μl 1M檸檬酸 (pH 3)。在Spectramax微量滴定板讀數器(Molecular Devices)上使用620nm作為參考波長量測415nm處之吸光度。減去所有樣品之陰性對照值並藉由吸光度值與FVIII濃度之圖形之線性回歸繪製校準曲線。比活性係藉由將樣品活性除以蛋白質濃度來計算,該蛋白質濃度係藉由尺寸排除HPLC並積分HPLC層析譜中之輕鏈峰(即不包括PEG部分)來測定。表2中之數據顯示,O-糖聚乙二醇化rFVIII化合物之比產色活性得以保持,意味著聚乙二醇化變體中似乎保留因子VIII活性。 The activity of the O-glycol PEGylated rFVIII obtained in Example 2 was evaluated in the chromogenic FVIII assay using Coatest SP reagent (Chromogenix) as follows: rFVIII sample and calibrator (7th International FVIII standard from NIBSC) Dilute in Coatest assay buffer (50 mM Tris, 150 mM NaCl, 1% BSA, pH 7.3, containing preservative). 50 μl of sample, standards, and buffer negative controls were added in duplicate to 96-well microtiter plates (Nunc). Factor IXa/Factor X reagent, phospholipid reagent and CaCl 2 from the Coatest SP kit were mixed at 5:1:3 (vol:vol:vol) and 75 μl of this material was added to the wells. After incubation for 15 min at room temperature, 50 μl of Factor Xa substrate S-2765/thrombin inhibitor I-2581 mixture was added and the reaction was incubated for 10 min at room temperature followed by the addition of 25 μl of 1 M citric acid (pH 3). The absorbance at 415 nm was measured on a Spectramax microtiter plate reader (Molecular Devices) using 620 nm as the reference wavelength. Negative control values for all samples were subtracted and a calibration curve was drawn by linear regression of the absorbance values and FVIII concentration plots. The specific activity was calculated by dividing the sample activity by the protein concentration, which was determined by size exclusion HPLC and integrating the light chain peaks in the HPLC chromatogram (ie, excluding the PEG moiety). The data in Table 2 shows that the specific chromogenic activity of the O-glycolized pegylated rFVIII compound is maintained, meaning that Factor VIII activity appears to be retained in the PEGylated variant.

圓括號中所注明之數據係單獨測定數值之平均偏差及標準偏差。 The data indicated in parentheses are the average deviation and standard deviation of the values measured separately.

實例4Example 4 在FVIII凝固活性分析中O-聚糖聚乙二醇化rFVIII之活性 Activity of O-glycan PEGylated rFVIII in FVIII coagulation activity assay

在FVIII凝固分析中進一步評價O-糖聚乙二醇化rFVIII之活性。將rFVIII樣品稀釋於HBS/BSA(20mM hepes、150mM NaCl,pH 7.4,含有1% BSA)中至約10U/ml,隨後10倍稀釋於含有VWF(Dade Behring)之FVIII缺乏血漿中。隨後將樣品及校準用血漿標準物(HemosIL校準血漿,來自Instrumentation Laboratory)稀釋於HBS/BSA中以產生四種(樣品)或六種(校準物)不同濃度。在ACL9000儀器(Instrumentation laboratory)上使用單因子程式來量測凝固時間,其中 將樣品/標準物與等體積的含有VWF(Dade Behring)、鈣及aPTT試劑之FVIII缺乏血漿混合,並量測凝固時間。使用以下試劑:Synthasil(HemosIL,Instrumentation Laboratory)、肌動蛋白FS(激活部分促凝血酶原激酶時間(Activated PTT)試劑,Dade Behring)、Stago(STA® PTT-A,Stago)、及dAPPTin(DAPPTIN®TC,Technoclone)。樣品活性係基於凝固時間對校準物濃度之半對數圖來計算。 The activity of O-glycol PEGylated rFVIII was further evaluated in the FVIII coagulation assay. The rFVIII sample was diluted in HBS/BSA (20 mM hepes, 150 mM NaCl, pH 7.4, containing 1% BSA) to about 10 U/ml, followed by 10-fold dilution in FVIII-deficient plasma containing VWF (Dade Behring). Samples and calibration plasma standards (HemosIL calibrated plasma from Instrumentation Laboratory) were then diluted in HBS/BSA to produce four (samples) or six (calibrators) different concentrations. A single factor program is used to measure the clotting time on an ACL 9000 instrument (Instrumentation laboratory), where The sample/standard was mixed with an equal volume of FVIII-deficient plasma containing VWF (Dade Behring), calcium and aPTT reagent, and the clotting time was measured. The following reagents were used: Synthasil (HemosIL, Instrumentation Laboratory), actin FS (Activated PTT reagent, Dade Behring), Stago (STA® PTT-A, Stago), and dAPPTin (DAPPTIN) ® TC, Technoclone). Sample activity is calculated based on a half log plot of clotting time versus calibrant concentration.

端視PEG大小及所用aPTT試劑,O-糖聚乙二醇化rFVIII化合物(分別為對照、10、40及80kDA PEG)之凝固活性(圖4)有不同程度的降低。使用Synthasil或dAPPTin作為aPTT試劑導致凝固活性隨PEG大小逐漸降低。使用Stago's aPTT試劑,對於所評價之所有三種O-糖聚乙二醇化N8化合物均觀察到比凝固活性降低50%。當使用肌動蛋白FS作為aPTT試劑時,比凝固活性保持在10,000IU/mg左右。數據表明,aPTT分析受存在之PEG部分影響,然而,使用選擇之aPTT試劑(例如肌動蛋白FS)O-糖聚乙二醇化時rFVIII之比凝固活性並未減弱。 The coagulation activity (Fig. 4) of the O-sugar PEGylated rFVIII compounds (control, 10, 40 and 80 kDA PEG, respectively) was reduced to varying degrees depending on the size of the PEG and the aPTT reagent used. The use of Synthasil or dAPPTin as an aPTT reagent resulted in a gradual decrease in coagulation activity with PEG size. Using Stago's aPTT reagent, a 50% reduction in coagulation activity was observed for all three O-glycolized PEGylated N8 compounds evaluated. When actin FS was used as the aPTT reagent, the specific clotting activity was maintained at about 10,000 IU/mg. The data indicates that the aPTT assay is affected by the presence of the PEG moiety, however, the specific clotting activity of rFVIII is not attenuated when PEGylated with the selected aPTT reagent (eg, actin FS) O-glycan.

實例5:Example 5: rFVIII之O-連接聚乙二醇化對輔因子活性及FVIII激活速率的影響 Effect of O-linked PEGylation of rFVIII on Cofactor Activity and FVIII Activation Rate

將激活之FVIII納入至FIXa-FVIIIa複合物中會使FIXa催化之FX激活的催化效率提高五個數量級(van Dieijen等人,(1981)J Biol Chem 256:3433)且FIXa-FVIIIa複合物組裝及FX激活動力學之表徵係FVIIIa分子之功能完整性之靈敏量度。藉由在磷脂及凝血酶激活之rFVIII或PEG-rFVIII存在下測定FIXa催化之FX激活的動力學參數來表徵凝血酶激活之rFVIII或PEG-rFVIII的輔因子活性。利用FVIIIa活性分析(FIXa-輔因子活性分析),對固定濃度(0.1nM)之rFVIIIa或FIXa分別實施FIXa及FVIIIa之相互滴定以獲得FIXa對於rFVIIIa之表觀親和常數(K½FIXa)及功能FVIIIa濃度。FX激活之米氏常數(Michaelis constant)(km)及轉換數(kcat)係自針對固定濃度之FIXa-FVIIIa複合物滴定FX來 獲得。 Incorporation of activated FVIII into the FIXa-FVIIIa complex increases the catalytic efficiency of FIXa-catalyzed FX activation by five orders of magnitude (van Dieijen et al., (1981) J Biol Chem 256:3433) and FIXa-FVIIIa complex assembly and Characterization of FX activation kinetics is a sensitive measure of the functional integrity of the FVIIIa molecule. The cofactor activity of thrombin-activated rFVIII or PEG-rFVIII was characterized by measuring the kinetic parameters of FIXa-catalyzed FX activation in the presence of phospholipid and thrombin-activated rFVIII or PEG-rFVIII. Using FVIIIa activity assay (FIXa-cofactor activity assay), FIXa and FVIIIa were titrated to each other at a fixed concentration (0.1 nM) of rFVIIIa or FIXa to obtain the apparent affinity constant (K 1⁄2 FIXa ) and functional FVIIIa concentration of FIXa for rFVIIIa. . The Michaelis constant (k m ) and the number of conversions (k cat ) of FX activation were obtained by titrating FX against a fixed concentration of FIXa-FVIIIa complex.

如下實施FIXa-輔因子活性分析:藉由在37℃下將rFVIII(通常為0.7nM,1U/mL)與5nM人類α-凝血酶一起培育恰好30秒來製備用於每一測試之新鮮的凝血酶激活之rFVIII及PEG-rFVIII變體。隨後,藉由將上文激活反應物二次取樣至FIXa、磷脂囊泡(磷脂TGT,來自Rossix[Mölndal,Sweden])、水蛭素、廣譜蛋白酶抑制劑(Pefabloc)Xa及CaCl2之製備好的混合物中來定量FX激活速率;藉由添加FX並容許在37℃下進行30秒或60秒來開始FX激活。藉由將FX激活反應物稀釋至含有EDTA之冰冷緩衝液中來終止激活。使用FXa特異性產色受質藉由在ELISA讀數器中讀取405nM處之吸光度來定量FXa濃度。利用使用經純化FXa製得之參考曲線將吸光度轉化成FXa濃度。使用自激活之rFVIII或PEG-rFVIII變體組裝之FIXa-rFVIIIa複合物的轉換數將FX激活速率轉化成rFVIIIa濃度。 FIXa-cofactor activity assay was performed as follows: Fresh coagulation for each test was prepared by incubating rFVIII (usually 0.7 nM, 1 U/mL) with 5 nM human alpha-thrombin for exactly 30 seconds at 37 °C. Enzyme activated rFVIII and PEG-rFVIII variants. Subsequently, preparation was carried out by subsampling the above activated reactants to FIXa, phospholipid vesicles (phospholipid TGT, from Rossix [Mölndal, Sweden]), hirudin, broad-spectrum protease inhibitors (Pefabloc) Xa and CaCl 2 . The FX activation rate was quantified in the mixture; FX activation was initiated by adding FX and allowing 30 or 60 seconds at 37 °C. Activation was terminated by diluting the FX activating reaction into ice cold buffer containing EDTA. FXa concentration was quantified using FXa-specific chromogenic receptors by reading the absorbance at 405 nM in an ELISA reader. The absorbance was converted to FXa concentration using a reference curve prepared using purified FXa. The turnover number of the FIXa-rFVIIIa complex assembled using self-activated rFVIII or PEG-rFVIII variants converts the FX activation rate to rFVIIIa concentration.

凝血酶催化之rFVIII激活速率係藉由在含有0.7nM rFVIII或PEG-rFVIII及0.13nM人類α-凝血酶之混合物中定量rFVIIIa之初始(0-3min)形成來量測。FVIIIa之形成與時間成線性關係。將FVIIIa激活速率表示為每分鐘每莫耳最初存在之rFVIII所形成rFVIIIa之莫耳數(v/[rFVIII]0)。 The thrombin-catalyzed rate of rFVIII activation was measured by quantifying the initial (0-3 min) formation of rFVIIIa in a mixture containing 0.7 nM rFVIII or PEG-rFVIII and 0.13 nM human alpha-thrombin. The formation of FVIIIa is linear with time. The FVIIIa activation rate is expressed as the number of moles of rFVIIIa (v/[rFVIII] 0 ) formed per minute of rFVIII originally present per mole.

rFVIII之O-連接糖聚乙二醇化不影響於激活之rFVIII存在下凝血酶催化之rFVIII激活速率或FIXa催化之FX激活的km或kcat(參見表3)。此外,O-連接糖聚乙二醇化不影響rFVIIIa-FIXa相互作用之表觀Kd(K½FIXa)。 O- linked glycosylation of rFVIII PEGylated rFVIII does not affect the presence of thrombin catalyzed activation rate of the catalytic FIXa or FX activation k m or k cat (Table 3) activation of rFVIII. Furthermore, O- linked glycosylation pegylation does not affect the apparent K d (K ½FIXa) Interaction of rFVIIIa-FIXa.

圖4顯示使用多種aPTT試劑之O-糖聚乙二醇化rFVIII的凝固活性。數據以凝固活性與產色活性之比(A)或以比凝固活性(B)顯示。顯示來自三項獨立實驗之值的平均值及標準偏差。 Figure 4 shows the clotting activity of O-glycol PEGylated rFVIII using various aPTT reagents. The data is shown as the ratio of coagulation activity to chromogenic activity (A) or to specific coagulation activity (B). The mean and standard deviation of the values from the three independent experiments are shown.

表3:rFVIII激活速率及藉由FIXa之FX激活的動力學常數 Table 3: Kinetic constants of rFVIII activation rate and FX activation by FIXa

數據係3-6次量測之平均值及標準偏差。 The data is the average and standard deviation of 3-6 measurements.

實例6Example 6 糖聚乙二醇化B結構域缺失型(BDD)-FVIII在FVIII KO小鼠及vWF KO小鼠中之藥物代謝動力學 Pharmacokinetics of glycoPEGylated B domain deleted (BDD)-FVIII in FVIII KO mice and vWF KO mice

在靜脈內投與280IU/kg至FVIII KO小鼠中之後對具有多種PEG大小之糖聚乙二醇化BDD-FVIII的藥物代謝動力學實施研究。 The pharmacokinetics of PEGylated BDD-FVIII with various PEG sizes were performed after intravenous administration of 280 IU/kg to FVIII KO mice.

研究以下化合物:BDD-FVIII、BDD-FVIII-10K PEG(O-聚糖,0129-0000-1005)、BDD-FVIII-40K PEG(O-聚糖,0129-0000-1003)、BDD-FVIII-2x40K PEG(O及N-聚糖0129-0000-1008-1A)、BDD-FVIII-80K PEG(N-聚糖,0129-0000-1012,O-聚糖0129-0000-1009)。 The following compounds were studied: BDD-FVIII, BDD-FVIII-10K PEG (O-glycan, 0129-0000-1005), BDD-FVIII-40K PEG (O-glycan, 0129-0000-1003), BDD-FVIII- 2x40K PEG (O and N-glycan 0129-0000-1008-1A), BDD-FVIII-80K PEG (N-glycan, 0129-0000-1012, O-glycan 0129-0000-1009).

動物研究設計:基於C57B1/6外顯子16基因剔除之背景,在Taconic M&B飼養因子VIII基因剔除(FVIII KO)小鼠。以約1:1之比例使用重量大約為25g及19-26週齡之雄性與雌性小鼠。小鼠未完全回交。在該小鼠品系中未檢測到FVIII。 Animal Research Design: Based on the background of C57B1/6 exon 16 gene knockout, Taconic M&B was reared to Factor VIII gene knockout (FVIII KO) mice. Male and female mice weighing approximately 25 g and 19-26 weeks of age were used at a ratio of approximately 1:1. The mice did not return completely. No FVIII was detected in this mouse strain.

在尾靜脈中給小鼠單次靜脈內注射280IU/kg上文所列示化合物。若小鼠係經靜脈外圍投藥,則用另一隻小鼠替換該小鼠。投藥後,自投藥前直至投藥後64小時使用未經塗敷之玻璃毛細管採集眼血管叢血液樣品。自每隻小鼠取三份樣品,且在每一時間點採集2、3或4份樣品。將血液在檸檬酸鈉(9:1)中穩定化並稀釋於FVIII COA SP緩衝液(1:4)中,隨後在4000g下離心5分鐘。將自經稀釋血液獲得之血漿在乾冰中於保持-80℃下冷凍,隨後藉助FVIII產色活性及/或FVIII 抗原分析實施定量分析。 Mice were given a single intravenous injection of 280 IU/kg of the compounds listed above in the tail vein. If the mouse is administered intravenously, the mouse is replaced with another mouse. After administration, blood samples of the ocular plexus were collected from uncoated glass capillaries from the time of administration until 64 hours after administration. Three samples were taken from each mouse and 2, 3 or 4 samples were taken at each time point. The blood was stabilized in sodium citrate (9:1) and diluted in FVIII COA SP buffer (1:4), followed by centrifugation at 4000 g for 5 minutes. The plasma obtained from the diluted blood is frozen in dry ice at -80 ° C, followed by FVIII chromogenic activity and / or FVIII Antigen analysis was performed quantitatively.

定量血漿分析: Quantitative plasma analysis:

藉由使用來自Coatest SP套組(Chromogenix)之試劑來測定FVIII產色活性。將經稀釋血漿樣品、存於Coatest SP緩衝液中之校準物(ILS校準血漿)、及緩衝液陰性對照(50μl)一式兩份添加至96孔微量滴定板(Nunc)中。將來自Coatest SP套組之因子IXa/因子X試劑、磷脂試劑及CaCl2以5:1:3(vol:vol:vol)混合並將75μl此物質添加至孔中。在於室溫下培育15min後,添加50μl因子Xa受質S-2765/凝血酶抑制劑I-2581混合物並在室溫下將反應物培育10min,隨後添加25μl 2%檸檬酸。在Spectramax微量滴定板讀數器(Molecular Devices)上量測405nm處之吸光度。自藉由校準用國際血漿標準物(ILS)稀釋物製得之校準曲線來計算血漿樣品中之FVIII活性。 FVIII chromogenic activity was determined by using reagents from the Coatest SP kit (Chromogenix). Diluted plasma samples, calibrators (ILS calibrated plasma) stored in Coatest SP buffer, and buffer negative controls (50 [mu]l) were added in duplicate to 96-well microtiter plates (Nunc). Factor IXa/Factor X reagent, phospholipid reagent and CaCl 2 from the Coatest SP kit were mixed at 5:1:3 (vol:vol:vol) and 75 μl of this material was added to the wells. After incubation for 15 min at room temperature, 50 μl of Factor Xa receptor S-2765/thrombin inhibitor I-2581 mixture was added and the reaction was incubated for 10 min at room temperature followed by the addition of 25 μl of 2% citric acid. Absorbance at 405 nm was measured on a Spectramax microtiter plate reader (Molecular Devices). The FVIII activity in the plasma samples was calculated from calibration curves prepared by calibrating dilutions with International Plasma Standards (ILS).

FVIII抗原分析係自Diagnostica Stago(Asserachrom VIII:CAg)購得之ELISA套組,其使用兩種針對人類FVIII之輕鏈的單株抗體。在藉由該套組提供之coatest SP稀釋緩衝液中將校準物(化合物之稀釋物)或血漿樣品稀釋至少50倍,將其施加至預塗覆孔中並按照製造商說明書實施ELISA。用於報告藥物代謝動力學研究之值係基於自化合物本身製得之標準曲線。 The FVIII antigen assay was an ELISA kit purchased from Diagnostica Stago (Asserachrom VIII: CAg) using two monoclonal antibodies directed against the light chain of human FVIII. The calibrant (diluted compound) or plasma sample was diluted at least 50-fold in the coatest SP dilution buffer provided by the kit, applied to the pre-coated wells and the ELISA was performed according to the manufacturer's instructions. The values used to report pharmacokinetic studies are based on standard curves prepared from the compounds themselves.

藥物代謝動力學參數估計: Estimation of pharmacokinetic parameters:

藥物代謝動力學分析係藉由數據之非分域方法(NCA)使用ILS作為校準物(數據基於產色活性)、使用化合物本身作為校準物(數據基於ELISA)來實施。自數據估計以下參數:Cmax(在於第一取樣時間點靜脈內投與後之最大濃度)、Tmax(在於第一時間點靜脈內投與後之最大濃度時間)、AUC0-∞(自0時間至無窮之曲線下面積)、T½(終末半衰期)、CL(清除率)及Vss(穩態分佈容積)。所有計算皆係使用WinNonlin Pro 4.1版本實施。 Pharmacokinetic analysis was performed by non-domain method (NCA) of data using ILS as a calibrator (data based on chromogenic activity), using the compound itself as a calibrator (data based on ELISA). The following parameters were estimated from the data: Cmax (the maximum concentration after intravenous administration at the first sampling time point), Tmax (the maximum concentration time after intravenous administration at the first time point), AUC0-∞ (from time 0 to time) The area under the infinite curve), T1⁄2 (terminal half-life), CL (clearance rate), and Vss (steady-state distribution volume). All calculations were performed using WinNonlin Pro version 4.1.

在靜脈內注射280IU/Kg BDD-EVIII、BDD-FVIII-10KDa PEG、BDD-FVIII-40KDa PEG、BDD-FVIII-2x40KDa PEG及BDD-FVIII-80KDa PEG至FVIII KO小鼠中後,隨PEG大小增加半衰期延長至7.8h(BDD-FVIII)至15-16h範圍內(表4),此對應於2倍增加。類似地,隨PEG大小增加,清除率降低且MRT增加(表4)。 After intravenous injection of 280IU/Kg BDD-EVIII, BDD-FVIII-10KDa PEG, BDD-FVIII-40KDa PEG, BDD-FVIII-2x40KDa PEG and BDD-FVIII-80KDa PEG into FVIII KO mice, the PEG size increases The half-life was extended to 7.8 h (BDD-FVIII) to 15-16 h (Table 4), which corresponds to a 2-fold increase. Similarly, as PEG size increased, clearance decreased and MRT increased (Table 4).

結論: in conclusion:

在靜脈內投與280IU/kg至FVIII KO小鼠中之後,與BDD-FVIII相比,BDD-FVIII之糖聚乙二醇化使T½延長1.3-2.1倍。隨PEG基團大小在10KDa至80KDa PEG範圍內增加,吾人觀察到T½延長。 After intravenous administration of 280 IU/kg to FVIII KO mice, PEGylation of BDD-FVIII sugars increased T1⁄2 by 1.3-2.1 fold compared to BDD-FVIII. As the size of the PEG group increased in the range of 10 KDa to 80 KDa PEG, we observed an increase in T1⁄2.

實例7Example 7 在FeCl3誘導之損傷模型中於A型血友病小鼠中與艾菲特(Advate)相比40K-PEG-[O]-N8之延長止血效果 Prolonged hemostasis of 40K-PEG-[O]-N8 compared with Advate in type A hemophilia mice induced by FeCl 3 induced injury

在FeCl3誘導之損傷模型中於A型血友病(F8-KO)小鼠中研究40K-PEG-[O]-N8相對於重組FVIII(艾菲特)之作用持續時間。 Mice Study 40K-PEG- [O] -N8 phase duration to effect recombination of FVIII (Yifei Te) in the hemophilia A (F8-KO) in the FeCl 3 model of injury induced in it.

將小鼠麻醉並置於加熱墊(37℃)上以保持體溫。暴露頸動脈並藉由超音將量測血液流量之流量探針(0.5 PSB奈米探針)置於動脈周圍。 藉由將短暫浸於10% FeCl3溶液中之濾紙(2×5mm)放置於暴露之頸動脈周圍來誘導損傷(鐵調介之化學氧化)。3min後移除濾紙。隨後將動脈用0.9% NaCl洗滌三次並最後施加消毒用外科潤滑劑(Surgilube)(聲音耦合劑)以排除流量探針中之空氣並確保血液流量之量測最佳化。在移除FeCl3飽和濾紙後記錄血液流量(ml/min)達25min並藉由量測自移除FeCl3飽和濾紙直至血液流量為0ml/min之時間(min)來測定阻塞時間。若在25min後未發生阻塞,則阻塞時間報告為25min,即使在觀察期間未發生阻塞。用艾菲特(280U/kg)、40K-PEG-[O]-N8(280U/kg)、或媒劑對F8-KO小鼠(n=6-10)實施治療。在投藥後5min(急性效應)或24、48、60、及72小時用FeCl3誘導損傷。在移除FeCl3後記錄血液流量(ml/min)達25min,並隨後測定阻塞時間。 Mice were anesthetized and placed on a heating pad (37 ° C) to maintain body temperature. A flow probe (0.5 PSB nano probe) that exposes the carotid artery and measures blood flow by supersonics is placed around the artery. Injury (iron-mediated chemical oxidation) was induced by placing a filter paper (2 x 5 mm) briefly immersed in a 10% FeCl 3 solution around the exposed carotid artery. Remove the filter paper after 3 minutes. The artery is then washed three times with 0.9% NaCl and finally a surgical surgical lubricant (Surgilube) is applied to exclude air from the flow probe and to ensure that the blood flow is optimized. Blood flow (ml / min) 25min and was determined by the amount of time measured from the blocking saturated filter paper to remove FeCl 3 until blood flow was 0ml / min of time (min) after removal of the recording paper saturated FeCl 3. If no blockage occurred after 25 min, the block time was reported to be 25 min, even though no blockage occurred during the observation period. Treatment with F8-KO mice (n=6-10) was performed with Effort (280 U/kg), 40K-PEG-[O]-N8 (280 U/kg), or vehicle. The injury was induced with FeCl 3 5 min (acute effect) or 24, 48, 60, and 72 hours after administration. Blood flow (ml/min) was recorded for 25 min after removal of FeCl 3 and the blocking time was subsequently determined.

在經媒劑治療之F8-KO小鼠中未發生阻塞,而在經40KDa-PEG-[O]-N8及艾菲特治療之所有小鼠中皆於投藥後5min(急性效應)發生阻塞,平均阻塞時間分別為4.3±0.4min及5.2±0.7min。在經40KDa-PEG-[O]-N8治療之F8-KO小鼠中,在投藥後72小時平均阻塞時間延長至13.8±3.4min。相比之下,經艾菲特治療之F8-KO小鼠在24及48小時後分別具有13.0±3.4min及15.9±2.9min之阻塞時間。重要的是,在投與艾菲特60及72小時未觀察到阻塞。在經40KDa-PEG-[O]-N8治療之所有小鼠中,在投藥後24小時觀察到阻塞,而經艾菲特治療之小鼠僅67%發生阻塞。在72小時後,在經40KDa-PEG-[O]-N8治療之小鼠中63%仍然可以看到阻塞,而在投與艾菲特後60及72小時未觀察到阻塞。 No occlusion occurred in the vehicle-treated F8-KO mice, but in all mice treated with 40KDa-PEG-[O]-N8 and Effort, the occlusion occurred 5 minutes after the administration (acute effect). The mean occlusion time was 4.3 ± 0.4 min and 5.2 ± 0.7 min, respectively. In F8-KO mice treated with 40KDa-PEG-[O]-N8, the mean occlusion time was extended to 13.8 ± 3.4 min at 72 hours after administration. In contrast, F8-KO mice treated with Effort had a blocking time of 13.0 ± 3.4 min and 15.9 ± 2.9 min after 24 and 48 hours, respectively. Importantly, no obstruction was observed after 60 and 72 hours of Effort administration. In all mice treated with 40KDa-PEG-[O]-N8, occlusion was observed 24 hours after administration, whereas only 67% of Ephel-treated mice developed occlusion. After 72 hours, occlusion was still visible in 63% of 40KDa-PEG-[O]-N8 treated mice, whereas no obstruction was observed at 60 and 72 hours after administration of Effie.

40KDa-PEG-[O]-N8於F8-KO小鼠中之延長效果。 The prolongation effect of 40KDa-PEG-[O]-N8 in F8-KO mice.

在投藥280IU/kg 40KDa-PEG-[O]-N8、280IU/kg艾菲特、或媒劑後5min(急性效應)、24、48、60、及72小時用FeCl3誘導損傷。在移除FeCl3後記錄血液流量(ml/min)達25min,並隨後測定阻塞時間。在 投藥後60及72小時,在投藥艾菲特之小鼠中未發生阻塞。顯示每一群組6-10隻小鼠之平均值及SEM。利用Kruskal-Wallis檢驗(包括Dunn事後檢驗(Dunn's post test))來比較不同組間之阻塞時間。*:p<0.05;**:p<0.01。 The injury was induced with FeCl 3 at 5 min (acute effect), 24, 48, 60, and 72 hours after administration of 280 IU/kg 40 KDa-PEG-[O]-N8, 280 IU/kg Effort, or vehicle. Blood flow (ml/min) was recorded for 25 min after removal of FeCl 3 and the blocking time was subsequently determined. At 60 and 72 hours after administration, no blockage occurred in the mice administered with Effie. The mean and SEM of 6-10 mice per group are shown. Kruskal-Wallis test (including Dunn's post test) was used to compare the block time between different groups. *: p <0.05; **: p < 0.01.

總之,在FeCl3誘導之損傷模型中於F8-KO小鼠中與艾菲特相比40KDa-PEG-[O]-N8之止血效果顯著延長。 In short, compared to the model of FeCl 3 -induced damage to the F8-KO mice with Yifei Te 40KDa-PEG- [O] -N8 hemostatic effect of significantly extended.

<110> 丹麥商諾沃-諾迪士克公司 <110> Danish company Novo-Nodisk

<120> 共軛因子VIII分子 <120> Conjugated Factor VIII Molecule

<130> 101961-5191WO <130> 101961-5191WO

<140> 098106523 <140> 098106523

<141> 2009-02-27 <141> 2009-02-27

<150> US 61/032,006 <150> US 61/032,006

<151> 2008-02-27 <151> 2008-02-27

<150> US 61/043,354 <150> US 61/043,354

<151> 2008-04-08 <151> 2008-04-08

<150> US 61/058,869 <150> US 61/058,869

<151> 2008-06-04 <151> 2008-06-04

<160> 2 <160> 2

<170> PatentIn vcrsion 3.5 <170> PatentIn vcrsion 3.5

<210> 1 <210> 1

<211> 2332 <211> 2332

<212> PRT <212> PRT

<213> 智人 <213> Homo sapiens

<400> 1 <400> 1

<210> 2 <210> 2

<211> 1445 <211> 1445

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> B-結構域截短型人類因子VIII <223> B-domain truncated human factor VIII

<400> 2 <400> 2

Claims (17)

一種具有改良之循環半衰期的B結構域截短型因子VIII分子,該分子經由該截短之B結構域中之O-連接寡糖與親水性聚合物共價共軛,其中因子VIII之激活導致該共價共軛親水性聚合物之移除。 A B domain truncated Factor VIII molecule having an improved circulating half-life, the molecule being covalently conjugated to a hydrophilic polymer via an O-linked oligosaccharide in the truncated B domain, wherein activation of Factor VIII results in Removal of the covalently conjugated hydrophilic polymer. 如請求項1之分子,其中該因子VIII前體多肽之重鏈及輕鏈部分係藉由連接體分開,其中該連接體之序列係衍生自因子VIII B結構域,且其中該連接體包含蛋白酶識別位點,該蛋白酶可將B結構域缺失型FVIII前體多肽分成重鏈及輕鏈。 The molecule of claim 1, wherein the heavy and light chain portions of the Factor VIII precursor polypeptide are separated by a linker, wherein the sequence of the linker is derived from a Factor VIII B domain, and wherein the linker comprises a protease At the recognition site, the protease can separate the B domain deleted FVIII precursor polypeptide into a heavy chain and a light chain. 如請求項1或2之分子,其中該B結構域的長度為20-30個胺基酸。 A molecule according to claim 1 or 2, wherein the B domain is 20-30 amino acids in length. 如請求項1或2之分子,其中該B結構域的長度為20個胺基酸。 The molecule of claim 1 or 2, wherein the B domain is 20 amino acids in length. 如請求項1或2之分子,其中該親水性聚合物係多醣。 The molecule of claim 1 or 2, wherein the hydrophilic polymer is a polysaccharide. 如請求項1或2之分子,其中該親水性聚合物係PEG。 The molecule of claim 1 or 2, wherein the hydrophilic polymer is PEG. 如請求項6之分子,其中該PEG之大小係約40,000Da。 The molecule of claim 6, wherein the PEG is about 40,000 Da in size. 如請求項1或2之分子,其中該分子包含如SEQ ID NO 2中所示之胺基酸序列。 The molecule of claim 1 or 2, wherein the molecule comprises an amino acid sequence as set forth in SEQ ID NO 2. 如請求項1或2之分子,其中該B結構域較SEQ ID NO 2短一個胺基酸。 A molecule according to claim 1 or 2, wherein the B domain is one amino acid shorter than SEQ ID NO 2. 如請求項1或2之分子,其中該分子係由包含下列步驟之方法製得:a)以編碼包含SEQ ID NO 2的因子VIII分子之載體轉染哺乳動物宿主細胞;b)在適合於宿主細胞中表現該因子VIII分子的條件下培養步驟 a)之宿主細胞;c)自步驟b)之宿主細胞培養物收穫該因子VIII分子;及d)經由O-連接寡糖將該因子VIII分子與親水性聚合物共價共軛。 A molecule according to claim 1 or 2, wherein the molecule is produced by a method comprising the steps of: a) transfecting a mammalian host cell with a vector encoding a Factor VIII molecule comprising SEQ ID NO 2; b) being suitable for the host The culture step under conditions in which the factor VIII molecule is expressed in the cell Host cell of a); c) harvesting the Factor VIII molecule from the host cell culture of step b); and d) covalently conjugated the Factor VIII molecule to the hydrophilic polymer via an O-linked oligosaccharide. 如請求項1或2之分子,其係用作用於治療血友病之藥劑。 A molecule according to claim 1 or 2 for use as an agent for the treatment of hemophilia. 如請求項1或2之分子,其係藉由皮下投與來治療血友病。 A molecule according to claim 1 or 2, which is administered by subcutaneous administration to treat hemophilia. 如請求項1或2之分子,其係藉由靜脈內投與來治療血友病。 The numerator of claim 1 or 2 is for the treatment of hemophilia by intravenous administration. 一種醫藥組合物,其包含如請求項1至10中任一項之分子。 A pharmaceutical composition comprising the molecule of any one of claims 1 to 10. 如請求項14之醫藥組合物,其係用作用於治療血友病之藥劑。 A pharmaceutical composition according to claim 14 which is for use as an agent for the treatment of hemophilia. 如請求項14之醫藥組合物,其係藉由皮下投與來治療血友病。 The pharmaceutical composition of claim 14, which is for treating hemophilia by subcutaneous administration. 如請求項14之醫藥組合物,其係藉由靜脈內投與來治療血友病。 The pharmaceutical composition of claim 14 which is administered by intravenous administration to treat hemophilia.
TW102147349A 2008-02-27 2009-02-27 Conjugated factor viii molecules TWI535454B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US3200608P 2008-02-27 2008-02-27
US4335408P 2008-04-08 2008-04-08
US5886908P 2008-06-04 2008-06-04

Publications (2)

Publication Number Publication Date
TW201412333A TW201412333A (en) 2014-04-01
TWI535454B true TWI535454B (en) 2016-06-01

Family

ID=51842289

Family Applications (1)

Application Number Title Priority Date Filing Date
TW102147349A TWI535454B (en) 2008-02-27 2009-02-27 Conjugated factor viii molecules

Country Status (7)

Country Link
BR (1) BRPI0908485B1 (en)
HU (1) HUS1900042I1 (en)
NL (1) NL301007I2 (en)
NO (1) NO2019046I1 (en)
PT (1) PT2257311E (en)
RU (1) RU2015144646A (en)
TW (1) TWI535454B (en)

Also Published As

Publication number Publication date
NL301007I2 (en) 2020-04-16
HUS1900042I1 (en) 2019-10-28
RU2015144646A (en) 2018-12-28
NO2019046I1 (en) 2019-12-16
BRPI0908485A2 (en) 2015-08-18
PT2257311E (en) 2014-07-28
NL301007I1 (en) 2019-10-02
TW201412333A (en) 2014-04-01
BRPI0908485B1 (en) 2022-02-22

Similar Documents

Publication Publication Date Title
TWI425953B (en) Conjugated factor viii molecules
JP5914363B2 (en) Factor VIII molecule with reduced VWF binding
AU2009239641B2 (en) Factor IX conjugates with extended half-lives
RU2744370C2 (en) Blood coagulation protein conjugates
JP6573989B2 (en) A truncated von Willebrand factor polypeptide for the treatment of hemophilia
KR20100110799A (en) Chemically modified factor ix
TWI535454B (en) Conjugated factor viii molecules
AU2013204960B2 (en) Conjugated factor VII molecules