JPWO2005053742A1 - Medicament containing antibody composition - Google Patents

Medicament containing antibody composition Download PDF

Info

Publication number
JPWO2005053742A1
JPWO2005053742A1 JP2005516043A JP2005516043A JPWO2005053742A1 JP WO2005053742 A1 JPWO2005053742 A1 JP WO2005053742A1 JP 2005516043 A JP2005516043 A JP 2005516043A JP 2005516043 A JP2005516043 A JP 2005516043A JP WO2005053742 A1 JPWO2005053742 A1 JP WO2005053742A1
Authority
JP
Japan
Prior art keywords
antibody
sugar chain
fucose
acetylglucosamine
antibody composition
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
JP2005516043A
Other languages
Japanese (ja)
Inventor
設楽 研也
研也 設楽
丹羽 倫平
倫平 丹羽
Original Assignee
協和醗酵工業株式会社
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 協和醗酵工業株式会社 filed Critical 協和醗酵工業株式会社
Publication of JPWO2005053742A1 publication Critical patent/JPWO2005053742A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation

Abstract

本発明は抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを組み合わせてなる医薬を提供する。In the present invention, among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50%. Provided is a medicament comprising a combination of the above antibody composition and at least one kind of drug.

Description

本発明は抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを組み合わせてなる医薬に関する。  In the present invention, among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50%. The present invention relates to a pharmaceutical comprising a combination of the above antibody composition and at least one kind of drug.

抗体依存性細胞障害活性(以下、ADCC活性と略記する)は、NK細胞、単球・マクロファージ、顆粒球など、エフェクター細胞と呼ばれる細胞集団に発現するFcγ受容体に、抗体のFc領域が結合することにより起こる。NK細胞は細胞上のFcγ受容体の一種FcγRIIIaを介して強いADCC活性を誘導する。FcγRIIIa遺伝子は158番目のアミノ酸に機能的な遺伝子多型が存在し、158番目のアミノ酸がValine(以下Valと称する)のFcγRIIIaはPhenylalanine(以下Pheと称する)のFcγRIIIaよりも強いADCC活性を誘導する[ジャーナル・オブ・クリニカル・インベスティゲーション(J.Clin.Invest.),100,1059−1070(1997)]。近年Val/Valの遺伝子型を有する患者において、その他の遺伝子型の患者よりもRituxanの臨床効果が有意に高いことが示されており[ブラッド(blood),99,754−758(2002)、アースライティス・アンド・リューマティズム(Arthritis Reum.),48,455−459(2003)]、ヒトIgG1サブクラスの抗体医薬の臨床効果にADCC活性が重要な役割を果たすことが示唆されている。
抗体などの糖タンパク質の糖鎖は、タンパク質部分との結合様式により、アスパラギンと結合する糖鎖(N−グリコシド結合糖鎖)とセリン、スレオニンなどと結合する糖鎖(O−グリコシル結合糖鎖)の2種類に大別される。N−グリコシド結合糖鎖は、以下の構造式(I)に示す基本となる共通のコア構造を有する[生物化学実験法23−糖蛋白質糖鎖研究法(学会出版センター)高橋禮子編(1989年)]。

Figure 2005053742
上記構造式(I)において、GlcNAcはN−アセチルグルコサミン、Manはマンノースを示す。またアスパラギンと結合する糖鎖の末端を還元末端、反対側を非還元末端という。
抗体分子は重鎖と軽鎖が2分子ずつ会合した4量体として構成される。IgG1型ヒト抗体は、重鎖のFc領域中に存在するN末端から297番目のアスパラギンに上記のコア構造を有する糖鎖が結合している。このコア構造にさらに非還元末端側にN−アセチルグルコサミン、ガラクトース、シアル酸、還元末端のN−アセチルグルコサミンの6位にフコースがそれぞれ付加する可能性があるが、通常これらの成分は均一ではなく、抗体を生産する細胞によって変化する[トレンズ・イン・バイオテクノロジー(Trends Biotechnol.),15,26−32(l997)]。ヒトIgG1のADCC活性は、この糖鎖中のガラクトース[ヒューマン・アンティボディズ・アンド・ハイブリドーマズ(Hum Antibodies Hybridomas),5,143−151(1994)、ヒューマン・アンティボディズ・アンド・ハイブリドーマズ(Hum Antibodies Hybridomas),6,82−88(1995)]、N−アセチルグルコサミン[ネイチャー・バイオテクノロジー(Nat.Biotechnol.),17,176−180,(1999)、バイオテクノロジー・アンド・バイオエンジニアリング(Biotechnol.Bioeng.),74,288−294,(2001)]、フコース[ジャーナル・オブ・バイオロジカル・ケミストリー(J.Biol.Chem.),277,26733−26740(2002)、ジャーナル・オブ・バイオロジカル・ケミストリー(J.Biol.Chem.),278,3466−3473(2003)]の含有量に影響を受けることが報告されている。中でも最も影響が大きいのはフコースであり、還元末端のN−アセチルグルコサミンの6位にフコースが付加した糖鎖の割合が少ないヒトIgG1型の抗体はADCC活性を顕著に増強する[ジャーナル・オブ・バイオロジカル・ケミストリー(J.Biol.Chem.),277,26733−26740(2002)、ジャーナル・オブ・バイオロジカル・ケミストリー(J.Biol.Chem.),278,3466−3473(2003)]。しかしこれらの報告はいずれも抗体単独でのADCC活性であり、他の薬剤によってフコースの量が少ないヒトIgG1型の抗体のADCC活性がさらに増強するかどうかは知られていない。
一方ADCC活性はエフェクター細胞を刺激するサイトカインによって上昇することが知られている。例えばインターロイキン(以下IL)−2[キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),46,213(1998)、キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),51,171(2002)、ブラッド(Blood),93,3922(1999)、ジャパニーズ・ジャーナル・オブ・キャンサー・リサーチ(Jpn.J.Cancer Res.),87,497(1996)]、IL−12[ブラッド(Blood),93,3922(1999)、ジャパニーズ・ジャーナル・オブ・キャンサー・リサーチ(Jpn.J.Cancer Res.),87,497(1996)]、IL−15[ブラッド(Blood),93,3922(1999)、ジャーナル・オブ・エクスペリメンタル・メディスン(J.Exp.Med),180,1395(1994)]、インターフェロン(以下IFN)−α[キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),46,213(1998)、キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),51,171(2002)]、IFN−γ[キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),46,213(1998)、キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),51,171(2002)、ジャパニーズ・ジャーナル・オブ・キャンサー・リサーチ(Jpn.J.Cancer Res.),87,497(1996)]、マクロファージ−コロニー刺激因子(以下M−CSF)[キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),46,213(1998)、ブラッド(Blood),93,3922(1999)、ジャパニーズ・ジャーナル・オブ・キャンサー・リサーチ(Jpn.J.Cancer Res.),87,497(1996)、ジャパニーズ・ジャーナル・オブ・キャンサー・リサーチ(Jpn.J.Cancer Res.),81,79(1990)]等、様々なサイトカインをエフェクター細胞にin vitroで加えて刺激することにより、ADCC活性を上昇させることが知られている。
化学療法剤のような低分子の薬剤がin vitroのADCC活性を増強することは知られていないが、化学療法と抗体との併用により優れた治療効果が得られることが知られている。抗HER2/neuヒト化抗体rhuMAb HER2(Herceptin、Roche社)はtaxane系抗癌剤との併用療法により乳癌に対して顕著な効果を示すことが知られている[Clinical Therapeutics,21,309(1999)]。また、抗CD20ヒト型キメラ抗体IDEC−C2B8(Rituxan、IDEC社)は多剤療法との併用療法によりB細胞リンパ腫に対して顕著な効果を示すことが知られている[J.Clin.Oncol.,17,268(1999)]。Antibody-dependent cytotoxicity (hereinafter abbreviated as ADCC activity) is that the Fc region of an antibody binds to Fcγ receptors expressed in cell populations called effector cells such as NK cells, monocytes / macrophages, and granulocytes. It happens. NK cells induce strong ADCC activity through FcγRIIIa, a type of Fcγ receptor on cells. The FcγRIIIa gene has a functional gene polymorphism at the 158th amino acid, and FcγRIIIa whose amino acid at 158 is Valine (hereinafter referred to as Val) induces stronger ADCC activity than that of Phenylalanine (hereinafter referred to as Phe). [Journal of Clinical Investigation (J. Clin. Invest.), 100, 1059-1070 (1997)]. Recently, it has been shown that patients with Val / Val genotypes have significantly higher clinical effects of Rituxan than patients with other genotypes [blood, 99, 754-758 (2002), Earth Lytis and Rheumatism (Arthritis Reum.), 48, 455-459 (2003)], it has been suggested that ADCC activity plays an important role in the clinical effects of antibody drugs of the human IgG1 subclass.
The sugar chains of glycoproteins such as antibodies are linked to asparagine (N-glycoside-linked sugar chains) and sugar chains (O-glycosyl-linked sugar chains) that bind to serine, threonine, etc., depending on the mode of binding to the protein moiety There are two main types. N-glycoside-linked sugar chains have a basic common core structure shown in the following structural formula (I) [Biochemical Experimental Method 23-Glycoprotein Sugar Chain Research Method (Academic Publishing Center) Etsuko Takahashi (1989) ]].
Figure 2005053742
In the structural formula (I), GlcNAc represents N-acetylglucosamine, and Man represents mannose. The end of the sugar chain that binds to asparagine is called the reducing end, and the opposite side is called the non-reducing end.
An antibody molecule is configured as a tetramer in which two heavy chains and two light chains are associated. In the IgG1-type human antibody, a sugar chain having the above core structure is bound to the 297th asparagine from the N-terminus present in the Fc region of the heavy chain. In addition to this core structure, fucose may be added to the 6-position of N-acetylglucosamine, galactose, sialic acid, and N-acetylglucosamine at the reducing end on the non-reducing end side, but these components are usually not uniform. , Depending on the cell producing the antibody [Trends Biotechnol., 15, 26-32 (l997)]. The ADCC activity of human IgG1 is determined by galactose [Human Antibodies Hybridomas, 5, 143-151 (1994), Human Antibodies and Hybridomas ( Hum Antibodies Hybridomas, 6, 82-88 (1995)], N-acetylglucosamine [Nature. Biotechnol., 17, 176-180, (1999), Biotechnology and Bioengineering (Biotechnol). Bioeng.), 74, 288-294, (2001)], fucose [Journal of Biological Chemistry (J. Biol. Ch. em.), 277, 26733-26740 (2002), Journal of Biological Chemistry (J. Biol. Chem.), 278, 3466-3473 (2003)]. ing. Of these, fucose has the greatest effect, and a human IgG1-type antibody with a small proportion of sugar chain with fucose added to the 6-position of N-acetylglucosamine at the reducing end significantly enhances ADCC activity [Journal of Biological chemistry (J. Biol. Chem.), 277, 26733-26740 (2002), Journal of biological chemistry (J. Biol. Chem.), 278, 3466-3473 (2003)]. However, these reports are all ADCC activity of the antibody alone, and it is not known whether the ADCC activity of the human IgG1-type antibody with a small amount of fucose is further enhanced by other drugs.
On the other hand, ADCC activity is known to be increased by cytokines that stimulate effector cells. For example, interleukin (hereinafter IL) -2 [Cancer Immunol. Immunother., 46, 213 (1998), Cancer Immunol. Immunother., 51, 171 (2002). ), Blood, 93, 3922 (1999), Japanese Journal of Cancer Research (Jpn. J. Cancer Res.), 87, 497 (1996)], IL-12 [Blood, 93, 3922 (1999), Japanese Journal of Cancer Research (Jpn. J. Cancer Res.), 87, 497 (1996)], IL-15 [Blo od), 93, 3922 (1999), Journal of Experimental Medicine (J. Exp. Med), 180, 1395 (1994)], interferon (hereinafter, IFN) -α [Cancer Immunology Immunotherapy ( Cancer Immunol. (Immunother.), 46, 213 (1998), Cancer Immunol. Immunotherapy (Cancer Immunol. Immunother.), 51, 171 (2002)], IFN-γ [Cancer Immunol. (Cancer Immunol.). Immunother.), 46, 213 (1998), Cancer Immunol. Immunother., 51, 171 2002), Japanese Journal of Cancer Research (Jpn. J. Cancer Res.), 87, 497 (1996)], macrophage-colony stimulating factor (M-CSF) [Cancer Immunology Immunotherapy (Cancer). Immunol. Immunother.), 46, 213 (1998), Blood, 93, 3922 (1999), Japanese Journal of Cancer Research (Jpn. J. Cancer Res.), 87, 497 (1996). , Japanese journal of Cancer research (Jpn.J.Cancer Res.), 81,79 ( 1990)] , etc., to stimulate added in in vitro a variety of cytokines in effector cells More, it is known to increase the ADCC activity.
Although it is not known that a low molecular weight drug such as a chemotherapeutic agent enhances in vitro ADCC activity, it is known that an excellent therapeutic effect can be obtained by the combined use of chemotherapy and an antibody. The anti-HER2 / neu humanized antibody rhuMAb HER2 (Herceptin, Roche) is known to show a remarkable effect on breast cancer by combination therapy with a taxane anticancer agent [Clinical Therapeutics, 21, 309 (1999)]. . Further, it is known that the anti-CD20 human chimeric antibody IDEC-C2B8 (Rituxan, IDEC) has a remarkable effect on B cell lymphoma by combination therapy with multidrug therapy [J. Clin. Oncol. , 17, 268 (1999)].

本発明の目的は、高い治療効果を有する抗体組成物と少なくとも1種類の薬剤とを用いる医薬を提供することにある。
本発明は、以下の(1)〜(14)に関する。
(1) 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを組み合わせてなる医薬。
(2) 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを併用して投与するための医薬。
(3) 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを同時に又は逐次的に投与するための医薬。
(4) 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物を単独で投与した場合よりも、高い治療効果を示すことを特徴とする、上記(1)〜(3)のいずれか1項に記載の医薬。
(5) 高い治療効果を示すことが、高い抗体依存性細胞障害活性を示すことである、上記(4)記載の医薬。
(6) 抗体組成物が、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチンに耐性を有する細胞から生産される抗体組成物である上記(1)〜(5)のいずれか1項に記載の医薬。
(7) 抗体組成物が、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖である抗体組成物である、上記(1)〜(6)のいずれか1項に記載の医薬。
(8) フコースが結合していない糖鎖が、該フコースの1位がN−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にα結合していない糖鎖である、上記(1)〜(7)のいずれか1項に記載の医薬。
(9) 抗体組成物が、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素のゲノム遺伝子がノックアウトされた細胞から生産された抗体組成物である、上記(7)または(8)記載の医薬。
(10) N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素が、α1,6−フコシルトランスフェラーゼである上記(9)記載の医薬。
(11) 薬剤が、蛋白質、低分子の薬剤および生物学的応答調節剤からなる群から選ばれる物質である上記(1)〜(10)のいずれか1項に記載の医薬。
(12) 蛋白質が、サイトカインまたは抗体である、上記(11)記載の医薬。
(13) サイトカインが、IFN−γ、IL−2およびIL−15から選ばれるサイトカインである上記(12)に記載の医薬。
(14) 医薬が、腫瘍を伴う疾患に対する治療薬である、上記(1)〜(13)のいずれか1項に記載の医薬。
本発明の医薬の形態としては、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを組み合わせてなる医薬、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを併用して投与するための医薬、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを同時に又は逐次的に投与するための医薬があげられる。
ここで、組み合わせてなる医薬とは、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを別々に調製し、これらの薬剤を組み合わせて同時にまたは逐次的に投与する医薬であってもよいし、それぞれの薬剤成分を混合させた合剤であってもよい。それぞれの薬剤成分を混合させた合剤には、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物に少なくとも1種類の薬剤を結合させた融合抗体なども包含する。
本発明において、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合とは、該組成物中に含まれるFc領域に結合する全てのN−グリコシド結合複合型糖鎖の合計数に対して、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の数が占める割合をいう。糖鎖の割合は、糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合していない糖鎖の割合が好ましい。
本発明においで、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖とは、該フコースが、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンにα結合していない糖鎖をいい、具体的には、該フコースの1位がN−グリコシド結合複合型糖鎖のN−アセチルグルコサミンの6位にα結合していない糖鎖があげられる。
本発明における抗体組成物としては、N−グリコシド結合複合型糖鎖をFc領域に有する抗体分子を含有してなる組成物であればいかなるものも包含される。
抗体分子とは、重鎖および軽鎖(以下、それぞれH鎖およびL鎖と表記する)の2種類のポリペプチド鎖がそれぞれ2分子ずつ会合した4量体である。H鎖のN末端側の約4分の1とL鎖のN末端側の約2分の1(それぞれ100余アミノ酸)はV領域と呼ばれ、多様性に富み、抗原との結合に直接関与する。V領域以外の部分の大半はC領域と呼ばれる。抗体分子はC領域の相同性によりIgG、IgM、IgA、IgD、IgEの各クラスに分類される。
またIgGクラスはC領域の相同性により、さらにIgG1〜IgG4のサブクラスに分類される。
H鎖はN末端側よりVH、CH1、CH2、CH3の4つのイムノグロブリンドメインに分かれ、CH1とCH2の間にはヒンジ領域と呼ばれる可動性の高いペプチド領域があり、CH1とCH2とが区切られる。ヒンジ領域以降のCH2とCH3からなる構造単位はFc領域と呼ばれ、N−グリコシド結合型糖鎖が結合している。また、この領域は、Fcレセプター、補体などが結合する領域である(免疫学イラストレイテッド原書第5版、2000年2月10日発行、南江堂版、抗体工学入門、1994年1月25日初版、地人書館)。
抗体などの糖蛋白質の糖鎖は、蛋白質部分との結合様式により、アスパラギンと結合する糖鎖(N−グリコシド結合糖鎖)とセリン、スレオニンなどと結合する糖鎖(O−グリコシル結合糖鎖)の2種類に大別される。N−グリコシド結合糖鎖は、以下の構造式(I)に示す基本となる共通のコア構造を有する[生物化学実験法23−糖蛋白質糖鎖研究法(学会出版センター)高橋禮子編(1989年)]。

Figure 2005053742
上記構造式(I)において、アスパラギンと結合する糖鎖の末端を還元末端、反対側を非還元末端という。
N−グリコシド結合糖鎖としては、上記構造式(I)のコア構造を有するものであればいかなるものでもよいが、コア構造の非還元末端にマンノースのみが結合するハイマンノース型、コア構造の非還元末端側にガラクトース−N−アセチルグルコサミン(以下、Gal−GlcNAcと表記する)の枝を並行して1ないしは複数本有し、更にGal−GlcNAcの非還元末端側にシアル酸、バイセクティングのN−アセチルグルコサミンなどの構造を有するコンプレックス型(複合型)、コア構造の非還元末端側にハイマンノース型とコンプレックス型の両方の枝を持つハイブリッド型などがあげられる。
抗体分子のFc領域には、N−グリコシド結合糖鎖が1カ所ずつ結合する領域を有しているので、抗体1分子あたり2本の糖鎖が結合している。抗体分子に結合するN−グルコシド結合糖鎖としては、前記構造式(I)で示されるコア構造を含むいかなる糖鎖も包含されるので、抗体に結合する2本のN−グルコシド結合糖鎖には多数の糖鎖の組み合わせが存在することになる。
したがって、本発明における抗体組成物は、本発明の効果が得られる範囲であれば、単一の糖鎖構造を有する抗体分子から構成されていてもよいし、複数の異なる糖鎖構造を有する抗体分子から構成されていてもよい。
抗体分子としては、抗体のFc領域を含む分子であればいかなる分子も包含される。具体的には、抗体、抗体の断片、Fc領域を含む融合蛋白質などがあげられる。
抗体としては、動物に抗原を免疫し、免疫動物の脾臓細胞より作製したハイブリドーマ細胞が分泌する抗体、遺伝子組換え技術により作製された抗体、すなわち、抗体遺伝子を挿入した抗体発現ベクターを、宿主細胞へ導入することにより取得された抗体などがあげられる。具体的には、ハイブリドーマが生産する抗体、ヒト化抗体、ヒト抗体などをあげることができる。
ハイブリドーマは、ヒト以外の哺乳動物に抗原を免疫して取得されたB細胞と、マウス、ラット等に由来するミエローマ細胞とを細胞融合させて得られる、所望の抗原特異性を有したモノクローナル抗体を生産する細胞をいう。
ヒト化抗体としては、ヒト型キメラ抗体、ヒト型CDR移植抗体などがあげられる。
ヒト型キメラ抗体は、ヒト以外の動物の抗体H鎖V領域(以下、HVまたはVHとも称す)および抗体L鎖V領域(以下、LVまたはVLとも称す)とヒト抗体のH鎖C領域(以下、CHとも称す)およびヒト抗体のL鎖C領域(以下、CLとも称す)とからなる抗体をいう。ヒト以外の動物としては、マウス、ラット、ハムスター、ラビット等、ハイブリドーマを作製することが可能であれば、いかなるものも用いることができる。
ヒト型キメラ抗体は、モノクローナル抗体を生産するハイブリドーマより、VHおよびVLをコードするcDNAを取得し、ヒト抗体CHおよびヒト抗体CLをコードする遺伝子を有する宿主細胞用発現ベクターにそれぞれ挿入してヒト型キメラ抗体発現ベクターを構築し、宿主細胞へ導入することにより発現させ、製造することができる。
ヒト型キメラ抗体のCHとしては、ヒトイムノグロブリン(以下、hIgと表記する)に属すればいかなるものでもよいが、hIgGクラスのものが好適であり、更にhIgGクラスに属するhIgG1、hIgG2、hIgG3、hIgG4といったサブクラスのいずれも用いることができる。また、ヒト型キメラ抗体のCLとしては、hIgに属すればいかなるものでもよく、κクラスあるいはλクラスのものを用いることができる。
ヒト型CDR移植抗体は、ヒト以外の動物の抗体のVHおよびVLのCDRのアミノ酸配列をヒト抗体のVHおよびVLの適切な位置に移植した抗体をいう。
ヒト型CDR移植抗体は、ヒト以外の動物の抗体のVHおよびVLのCDR配列を任意のヒト抗体のVHおよびVLのCDR配列に移植したV領域をコードするcDNAを構築し、ヒト抗体のCHおよびヒト抗体のCLをコードする遺伝子を有する宿主細胞用発現ベクターにそれぞれ挿入してヒト型CDR移植抗体発現ベクターを構築し、該発現ベクターを宿主細胞へ導入することによりヒト型CDR移植抗体を発現させ、製造することができる。
ヒト型CDR移植抗体のCHとしては、hIgに属すればいかなるものでもよいが、hIgGクラスのものが好適であり、更にhIgGクラスに属するhIgG1、hIgG2、hIgG3、hIgG4といったサブクラスのいずれも用いることができる。また、ヒト型CDR移植抗体のCLとしては、hIgに属すればいかなるものでもよく、κクラスあるいはλクラスのものを用いることができる。
ヒト抗体は、元来、ヒト体内に天然に存在する抗体をいうが、最近の遺伝子工学的、細胞工学的、発生工学的な技術の進歩により作製されたヒト抗体ファージライブラリーならびにヒト抗体トランスジェニック非ヒト動物あるいはヒト抗体トランスジェニック植物から得られる抗体等も含まれる。
ヒト体内に存在する抗体は、例えば、ヒト末梢血リンパ球を単離し、EBウイルス等を感染させ不死化、クローニングすることにより、該抗体を生産するリンパ球を培養でき、培養物中より該抗体を精製することができる。
ヒト抗体ファージライブラリーは、ヒトB細胞から調製した抗体遺伝子をファージ遺伝子に挿入することによりFab、一本鎖抗体等の抗体断片をファージ表面に発現させたライブラリーである。該ライブラリーより、抗原を固定化した基質に対する結合活性を指標として所望の抗原結合活性を有する抗体断片を発現しているファージを回収することができる。該抗体断片は、更に遺伝子工学的手法により、2本の完全なH鎖および2本の完全なL鎖からなるヒト抗体分子へも変換することができる。
ヒト抗体トランスジェニック非ヒト動物は、ヒト抗体遺伝子が細胞内に組込まれた動物をいう。具体的には、マウス胚性幹細胞へヒト抗体遺伝子を導入し、該胚性幹細胞を他のマウスの初期胚へ移植後、発生させることによりヒト抗体トランスジェニック非ヒト動物を作製することができる。また、動物の受精卵にヒト抗体遺伝子を導入し、該受精卵を発生させることにヒト抗体トランスジェニック非ヒト動物を作製することもできる。ヒト抗体トランスジェニック非ヒト動物からのヒト抗体の作製方法は、通常のヒト以外の哺乳動物で行われているハイブリドーマ作製方法によりヒト抗体ハイブリドーマを得、培養することで培養物中にヒト抗体を蓄積させることができる。
トランスジェニック非ヒト動物としては、ウシ、ヒツジ、ヤギ、ブタ、ウマ、マウス、ラット、ニワトリ、サルまたはウサギ等があげられる。
抗体の断片は、上記抗体の少なくともFc領域の一部を含んだ断片をいう。Fc領域とは、抗体のH鎖のC末端側の領域、CH2領域およびCH3領域をいう。少なくともFc領域の一部とは、好ましくはCH2領域を含む断片、より好ましくはCH2領域内に存在する1番目のアスパラギン酸を含む領域をいう。IgGクラスのFc領域は、カバット(Kabat)らのEU Index[シーケンシズ・オブ・プロテインズ・オブ・イムノロジカル・インタレスト(Sequences of Proteins of Immunological Interest),5th Ed.Public Health Service,National Institutes of Health,Bethesda,MD.(1991)]のナンバリングで226番目のCysからC末端、あるいは230番目のProからC末端までをいう。抗体の断片としては、H鎖の単量体、H鎖の2量体などがあげられる。本発明におけるFc領域としては、天然型およびその変異型を包含する。
Fc領域の一部を有する融合蛋白質とは、抗体のFc領域の一部を含んだ抗体あるいは抗体の断片と、酵素、サイトカインなどの蛋白質とを融合させた物質(以下、Fc融合蛋白質と称す)をいう。
本発明の抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上、好ましくは60%以上、より好ましくは70%以上、さらに好ましくは80%以上、特に好ましくは90%以上であり、抗体組成物中に含まれるFc領域に結合する全てのN−グリコシド結合複合型糖鎖が、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖であることが最も好ましい。
以上のように抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が高ければ高いほど、抗体組成物は、高い抗体依存性細胞障害活性を有する。
N−グリコシド結合複合型糖鎖をFc領域に有する抗体分子からなる組成物中に含まれる、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合は、抗体分子からヒドラジン分解や酵素消化などの公知の方法[生物化学実験法23−糖タンパク質糖鎖研究法(学会出版センター)高橋禮子編(1989)]を用い、糖鎖を遊離させ、遊離させた糖鎖を蛍光標識又は同位元素標識し、標識した糖鎖をクロマトグラフィー法にて分離することによって決定することができる。また、遊離させた糖鎖をHPAED−PAD法[ジャーナル・オブ・リキッド・クロマトグラフィー(J.Liq.Chromatogr.),6,1577(1983)]によって分析することによっても決定することができる。
本発明において、高い治療効果としては、例えば細胞障害活性が高いこと等があげられる。
抗体組成物の有する細胞障害活性としては抗体依存性細胞障害活性(以下、ADCCと略記する)、補体依存性細胞障害活性(モノクローナル・アンティボディズ)、抗原に結合することによる抗原発現細胞の増殖抑制活性などがあげられる。増殖抑制活性には標的細胞のアポトーシス誘導や分化誘導を促進するものも包含される[キャンサー・リサーチ(Cancer Research)60,7170(2000)、ネイチャー・メディスン(Nature Medicine),644(1995)、セル・グロース・ディファレンシエーション(Cell Growth Differ.),401(1992)]。
ADCC活性とは、生体内で、腫瘍細胞等の細胞表面抗原などに結合した抗体が、抗体Fc領域とエフェクター細胞表面上に存在するFc受容体との結合を介してエフェクター細胞を活性化し、腫瘍細胞等を障害する活性をいう[モノクローナル・アンティボディズ:プリンシプルズ・アンド・アプリケーションズ(Monoclonal Antibodies:Principles and Applications),Wiley−Liss,Inc.,Capter2.1(1995)]。エフェクター細胞としては、ナチュラルキラー細胞、マクロファージ、単球、樹状細胞、顆粒球等の免疫細胞があげられる。またFc受容体はFcα受容体I、Fcε受容体I、Fcε受容体II、Fcγ受容体I、Fcγ受容体IIa、Fcγ受容体IIb、Fcγ受容体IIc、Fcγ受容体IIIa、Fcγ受容体IIIb、Fc受容体n等のタイプに分かれる。この内Fcγ受容体IIIaは主にナチュラルキラー細胞上に発現し、ADCC活性に重要なFc受容体の一つである[モノクローナル・アンティボディズ:プリンシプルズ・アンド・プラクティス(Monoclonal Antibodies:principles and practice),ThirdEdition,Acad.Press,1996(以下、モノクローナル・アンティボディズと略す)]。
したがって、本発明において、抗体分子のFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物を単独で投与した場合よりも、高い治療効果を示すとは、抗体分子のFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と少なくとも1種類以上の薬剤とを組み合わせることにより、該抗体組成物単独で投与した場合に発揮される治療効果よりも高い治療効果を発揮する医薬をいう。
また、本発明の医薬は、抗体分子のFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%未満である抗体組成物と少なくとも1種類の薬剤とを組み合わせてなる医薬よりも、高い治療効果を示す。
本発明の医薬は、薬剤を単独で使用する場合よりも少ない薬剤の量にすることができ、薬剤を単独で高用量を患者に投与した場合に懸念された副作用を低減させることができる。
N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチンに耐性を有する細胞(以下、α1,6−フコース/レクチン耐性細胞と称す)としては、酵母、動物細胞、昆虫細胞、植物細胞など抗体組成物を製造することができる細胞であって、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチン耐性を有する細胞であればいかなる細胞でもよい。
具体的には、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチンに耐性を有する、ハイブリドーマ細胞、ヒト抗体およびヒト化抗体を製造するための宿主細胞、ヒト抗体を生産するためのトランスジェニック非ヒト動物を製造するための胚性幹細胞および受精卵細胞、ヒト抗体を生産するためのトランスジェニック植物を製造するための植物カルス細胞、ミエローマ細胞、トランスジェニック非ヒト動物由来の細胞などがあげられる。ミエローマ細胞はハイブリドーマ細胞を製造する際に融合細胞として用いることができる。また、トランスジェニック非ヒト動物に抗原を免疫し、該動物の脾臓細胞を取り出し、ハイブリドーマ細胞を製造するために用いることができる。
レクチンに耐性を有する細胞とは、培養培地にレクチンを有効濃度与えて細胞培養を行ったときにも、生育が阻害されない細胞をいう。
本発明において、生育が阻害されないレクチン有効濃度は、細胞株に応じて適宜定めればよいが、通常10μg/ml〜10.0mg/ml、好ましくは0.5〜2.0mg/mlである。親株細胞に変異を導入した場合のレクチンの有効濃度とは、該親株細胞が正常に生育できない濃度以上であり、好ましくは親株細胞が正常に生育できない濃度と同濃度、より好ましくは2〜5倍、さらに好ましくは10倍、最も好ましくは20倍以上の濃度をいう。
親株細胞とは、何らかの処理を施す前の細胞、すなわち本発明で用いるα1,6−フコース/レクチン耐性細胞を選択する工程を行う前の細胞、上述した酵素活性を低下または欠失するために遺伝子工学的な処理を行う前の細胞をいう。
親株細胞としては、特に限定はないが、種々の細胞株の親株細胞の具体例として、以下に示す細胞があげられる。
NS0細胞の親株細胞としては、バイオ/テクノロジー(BIO/TECHNOLOGY),10,169(1992)、バイオテクノロジー・バイオエンジニアリング(Biotechnol.Bioeng.),73,261,(2001)等の文献に記載されているNS0細胞があげられる。また、理化学研究所細胞開発銀行に登録されているNS0細胞株(RCB0213)、あるいはこれら株を生育可能な培地に馴化させた亜株などもあげられる。
SP2/0−Ag14細胞の親株細胞としては、ジャーナル・オブ・イムノロジー(J.Immunol.),126,317(1981)、ネイチャー(Nature),276,269(1978)、ヒューマン・アンチィボディズ・アンド・ハイブリドーマズ(Human Antibodies and Hybridomas),,129(1992)等の文献に記載されているSP2/0−Ag14細胞があげられる。また、ATCCに登録されているSP2/0−Ag14細胞(ATCC CRL−1581)あるいはこれら株を生育可能な培地に馴化させた亜株(ATCC CRL−1581.1)などもあげられる。
チャイニーズハムスター卵巣組織由来CHO細胞の親株細胞としては、Journal of Experimental Medicine,108,945(1958)、Proc.Natl.Acad.Sei.USA,60,1275(1968)、Genetics,55,513(1968)、Chromosoma,41,129(1973)、Methods in Cell Science,18,115(1996)、Radiation Research,148,260(1997)、Proc.Natl.Acad.Sci.USA,77,4216(1980)、Proc.Natl.Acad.Sci.60,1275(1968)、Cell,,121(1975)、Molecular Cell Genetics,Appendix I,II(p883−900)等の文献に記載されているCHO細胞などがあげられる。また、ATCCに登録されているCHO−K1株(ATCC CCL−61)、DUXB11株(ATCCCRL−9096)、Pro−5株(ATCC CRL−1781)や、市販のCHO−S株(Lifetechnologies社 Cat#11619)、あるいはこれら株を生育可能な培地に馴化させた亜株などもあげられる。
ラットミエローマ細胞株YB2/3HL.P2.G11.16Ag.20細胞の親株細胞としては、Y3/Ag1.2.3細胞(ATCC CRL−1631)から樹立された株化細胞が包含される。その具体的な例としては、J.Cell.Biol.,93,576(1982)、Methods Enzymol.,73B,1(1981)等の文献に記載されているYB2/3HL.P2.G11.16Ag.20細胞があげられる。また、ATCCに登録されているYB2/3HL.P2.G11.16Ag.20細胞(ATCC CRL−1662)あるいはこれら株を生育可能な培地に馴化させた亜株などもあげられる。
N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチンとしては、該糖鎖構造を認識できるレクチンであれば、いずれのレクチンも包含される。その具体的な例としては、レンズマメレクチンLCA(Lens Culinaris由来のLentil Agglutinin)、エンドウマメレクチンPSA(Pisum sativum由来のPea Lectin)、ソラマメレクチンVFA(Vicia faba由来のAgglutinin)、ヒイロチャワンタケレクチンAAL(Aleuria aurantia由来のLectin)などがあげられる。
本発明において、α1,6−フコース/レクチン耐性細胞としては、一定の有効濃度のレクチン存在下で生育が阻害されない細胞であれば、いずれでもよいが、例えば、以下にあげる少なくとも1つの蛋白質の活性が親株細胞よりも低下または欠失した細胞などがあげられる。
(a)細胞内糖ヌクレオチドGDP−フコースの合成に関与する酵素蛋白質(以下、「GDP−フコース合成酵素」と表記する);
(b)N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素蛋白質(以下、「α1,6−フコース修飾酵素」と表記する);
(c)細胞内糖ヌクレオチドGDP−フコースのゴルジ体への輸送に関与する蛋白質(以下、「GDP−フコース輸送蛋白質」と表記する)。
GDP−フコース合成酵素としては、細胞内で糖鎖へのフコースの供給源である糖ヌクレオチドGDP−フコースの合成に関与する酵素であればいかなる酵素も包含し、細胞内糖ヌクレオチドGDP−フコースの合成に影響を与える酵素などがあげられる。
細胞内の糖ヌクレオチドGDP−フコースは、de novoの合成経路あるいはSalvage合成経路により供給されている。したがって、これら合成経路に関与する酵素はすべてGDP−フコース合成酵素に包含される。
de novoの合成経路に関与するGDP−フコース合成酵素としては、GDP−mannose 4−dehydratase(GDP−マンノース 4−デヒドラターゼ;以下、GMDと表記する)、GDP−keto−6−deoxymannose 3,5−epimerase,4−reductase(GDP−ケト−デオキシマンノース 3,5−エピメラーゼ,4−リダクターゼ;以下、Fxと表記する)などがあげられる。
Salvage合成経路に関与するGDP−フコース合成酵素としては、GDP−beta−L−fucose pyrophosphorylase(GDP−ベータ−L−フコース−ピロホスフォリラーゼ;以下、GFPPと表記する)、Fucokinase(フコキナーゼ)などがあげられる。GMDとしては、配列番号1、2または3で表されるアミノ酸配列を有するGMDがあげられる。
細胞内糖ヌクレオチドGDP−フコースの合成に影響を与える酵素としては、上述の細胞内の糖ヌクレオチドGDP−フコースの合成経路に関与する酵素の活性に影響を与えたり、該酵素の基質となる物質の構造に影響を与える酵素も包含される。
α1,6−フコース修飾酵素としては、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合する反応に関与する酵素であればいかなる酵素も包含される。N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合する反応に関与する酵素としては、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合する反応に影響を与える酵素であればいかなる酵素も包含される。具体的には、α1,6−フコシルトランスフェラーゼやα−L−フコシダーゼなどがあげられる。α1,6−フコシルトランスフェラーゼとしては、配列番号4または5で表されるアミノ酸配列を有するα1,6−フコシルトランスフェラーゼがあげられる。
また、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合する反応に影響を与える酵素としては、上述のN−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する反応に関与する酵素の活性に影響を与えたり、該酵素の基質となる物質の構造に影響を与える酵素も包含される。
GDP−フコース輸送蛋白質としては、細胞内糖ヌクレオチドGDP−フコースのゴルジ体への輸送に関与する蛋白質であればいかなる蛋白質も包含され、具体的には、GDP−フコーストランスポーターなどがあげられる。
また、細胞内糖ヌクレオチドGDP−フコースをゴルジ体内へ輸送する反応に影響を与える蛋白質もGDP−フコース輸送蛋白質に包含され、具体的には上述の細胞内糖ヌクレオチドGDP−フコースのゴルジ体への輸送に関与する蛋白質の活性に影響を与えたり、発現に影響を与える蛋白質などがあげられる。
本発明の製造方法に用いられる細胞を取得する方法としては、α1,6−フコース/レクチン耐性細胞を選択することができる手法であればいかなる手法でも用いることができる。具体的には、上述した蛋白質の活性を低下または欠失させる手法などがあげられる。上述の蛋白質の活性を低下または欠失させる手法としては、
(a)蛋白質の遺伝子を標的した遺伝子破壊の手法;
(b)蛋白質の遺伝子のドミナントネガティブ体を導入する手法;
(c)蛋白質についての突然変異を導入する手法;
(d)蛋白質の遺伝子の転写または翻訳を抑制する手法;
などがあげられる(WO02/31140、WO03/085107、WO03/085118)。
本発明において、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素のゲノム遺伝子がノックアウトされた細胞としては、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素のゲノム遺伝子の発現または機能を完全に抑制された細胞などがあげられる。したがって、本発明において、ゲノム遺伝子がノックアウトされた細胞としては、以下に述べる手法を用いて、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素のゲノム遺伝子をノックアウトした細胞があげられる。
ゲノム遺伝子がノックアウトされた細胞の具体的な例としては、標的となる遺伝子のすべてまたは一部がゲノムから削除された細胞があげられる。このような細胞を取得する方法としては、目的とするゲノムの改変を行うことができれば、いずれの手法でも用いることができるが、遺伝子工学的な手法が望ましい。その具体的な手法としては、上述した酵素の活性を低下または欠失させる手法があげられる。
また、上述した、N−グリコシド結合糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチンに耐性な細胞株を選択する方法を用いることにより、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素のゲノム遺伝子がノックアウトされた細胞を選択することができる。
本発明における抗体組成物としては、疾患に関連する細胞に発現する抗原、もしくは疾患に関連する細胞の増殖や転移など、病態形成に関わる抗原などに対する抗体組成物などがあげられる。具体的にはGD2、GD3、GM2、HER2、CD20、CD22、CD25、CD33、CD52、MAGE、HM1.24、CCケモカイン受容体4(CCR4)、副甲状腺ホルモン関連蛋白(PTHrP)、塩基性線維芽細胞増殖因子、線維芽細胞増殖因子8、塩基性繊維芽細胞増殖因子受容体、線維芽細胞増殖因子8受容体、上皮細胞成長因子受容体(EGFR)、上皮性細胞接着分子(EpCam)、インスリン様増殖因子、インスリン様増殖因子受容体、PMSA、血管内皮細胞増殖因子、血管内皮細胞増殖因子受容体、呼吸器合胞体ウイルス(RSV)、IL−5受容体α鎖などに対する抗体組成物があげられる。
上記の抗体組成物の具体例としては、抗GD2抗体[アンチ・キャンサー・リサーチ(Anticancer Res.),13,331(1993)]、抗GD3抗体[キャンサー・イムノロジー・イムノセラピー(Cancer Immunol.Immunother.),36,260(1993)、WO01/23432、US6437098]、抗GM2抗体[キャンサー・リサーチ(Cancer Res.),54,1511(1994)、US6423511]、抗HER2抗体[プロシーディングス・オブ・ザ・ナショナル・アカデミー・オブ・サイエンス(Proc.Natl.Acad.Sci.USA),89,4285(1992)]、抗CD20抗体[ブラッド(Blood),83,435(1994)、WO03/55993]、抗CD22抗体[セミナーズ・イン・オンコロジー(Semmin.Oncol.),30,253(2003)]、抗CD25抗体[ヒューマン・アンティボディズ(Human Antibodies),10,127−142(2001)]、抗CD33抗体[ジャーナル・オブ・クリニカル・オンコロジー(J.Clin.Oncol.),19,3244(2001)]、抗CD52抗体[プロシーディングス・オブ・ザ・ナショナル・アカデミー・オブ・サイエンス(Proc.Natl.Acad.Sci.USA),89,4285(1992)]、抗MAGE抗体[ブリティッシュ・ジャーナル・オブ・キャンサー(British J.Cancer),83,493(2000)]、抗HM1.24抗体[モレキュラー・イムノロジー(Molecular Immunol.),36,387(1999)]、抗CCR4抗体[WO01/64754、WO03/18635、WO00/42074、WO99/15666、US6245332]、抗副甲状腺ホルモン関連蛋白(PTHrP)抗体[キャンサー(Cancer),88,2909(2000)]、抗線維芽細胞増殖因子8抗体[プロシーディングス・オブ・ザ・ナショナル・アカデミー・オブ・サイエンス(Proc.Natl.Acad.Sci.USA),86,9911(1989)、WO03/002608]、抗線維芽細胞増殖因子8受容体抗体[ジャーナル・オブ・バイオロジカル・ケミストリー(J.Biol.Chem.),265,16455(1990)]、抗上皮細胞成長因子受容体抗体[キャンサー・リサーチ(Cancer Res.),59,1236(1999)]、抗上皮性細胞接着分子抗体[プロシーディングス・オブ・ザ・ナショナル・アカデミー・オブ・サイエンス(Proc.Natl.Acad.Sci.USA),76,1438(1979)]、抗インスリン様増殖因子抗体[ジャーナル・オブ・ニューロサイエンス・リサーチ(J.Neurosci.Res.),40,647(1995)、WO03/93317]、抗インスリン様増殖因子受容体抗体[ジャーナル・オブ・ニューロサイエンス・リサーチ(J.Neurosci.Res.),40,647(1995)]、抗PMSA抗体[ジャーナル・オブ・ウロロジー(J.Urology),160,2396(1998)]、抗血管内皮細胞増殖因子抗体[キャンサー・リサーチ(Cancer Res.),57,4593(1997)]、抗血管内皮細胞増殖因子受容体抗体[オンコジーン(Oncogene),19,2138(2000)]、抗RSV抗体[アンティバイラル・リサーチ(Antiviral Res.),47,57−77(2000)]、抗IL−5受容体α鎖抗体(US6018032、US6538111)などがあげられる。具体的な抗体名としては、ハーセプチン(Herceptin)、リツキサン(Rituxan)、キャンパス(Campath)、アバスチン(Avastin)、ベクサー(Bexxar)、リンフォサイド(LymphoCide)、マイロターグ(Mylotarg)、パノレックス(Panorex)、ゼバリン(Zevalin)[ネイチャー・レビューズ・キャンサー(Nat.Rev.Cancer),,118(2001)]、ゼナパックス(Zenapax)、シムレクト(Simulect)、レミケード(Remicade)、シナジス(Synagis)、レオプロ(ReoPro)[ヒューマン・アンティボディズ(Human Antibodies),10,127−142(2001)]、エンブレル(Enbrel)[エキスパート・オピニオン・オン・ファーマコセラピー(Expert Opinion on Pharmacotherapy),,1137−1148(2001)]、ゾレア(Zolair)[レスピラトリー・メディスン(Respiratory Medicine),97,123−129(2003)]などがあげられる。
本発明において、ADCC活性は、51Cr遊離法、lactate dehydrogenase(LDH)遊離法、フローサイトメトリー等によるin vitroの測定系、あるいは動物モデルを用いたin vivoの評価系等で測定することができる。
本発明において、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物とともに用いられる薬剤としては、サイトカイン、抗体などの蛋白質、低分子の薬剤、生物学的応答調節物質(以下BRMと称する)などがあげられる。
サイトカインとしては、インターロイキン(IL)類、コロニー刺激因子(CSF)類、インターフェロン(IFN)類、腫瘍壊死因子(TNF)類、ケモカイン類、あるいはこれらの組み合わせがあげられる。具体的には、IFN−α、IFN−β、IFN−γ、TNF−α、TNF−β、IL−1α、IL−1β、IL−2、IL−3、IL−4、IL−6、IL−7、IL−9、IL−12、IL−15、IL−17、IL−18、IL−21、fractalkine、GM−CSF、G−CSF、M−CSF、SCF、あるいはこれらの組み合わせがあげられる。好ましくはIL−2、IL−15、IFN−α、IFN−γがあげられる。
低分子の薬剤としては、アミフォスチン(エチオール)[amifostine(ethyol)]、シスプラチン(cisplatin)、ダカルバジン(DTIC)[dacarbazine(DTIC)]、ダクチノマイシン(dactinomycin)、メクロレタミン(ナイトロジェンマスタード)[mechlorethamine(nitrogenmustard)]、ストレプトゾシン(streptozocin)、シクロフォスファミド(cyclophosphamide)、カルムスチン(BCNU)[carmustine(BCNU)]、ロムスチン(CCNU)[lomustine(CCNU)]、ドキソルビシン(アドリアマイシン)[doxorubicin(adriamycin)]、ドキソルビシンリポ(ドキシル)[doxorubicinlipo(doxil)]、ゲムシタビン(ゲムザール)[gemcitabine(gemzar)]、ダウノルビシン(daunorubicin)、ダウノルビシンリポ(ダウノゾーム)[daunorubicinlipo(daunoxome)]、プロカルバジン(procarbazine)、マイトマイシン(mitomycin)、シタラビン(cytarabine)、エトポシド(etoposide)、メトトレキセート(methotrexate)、5−フルオロウラシル(5−fluorouracil)、ビンブラスチン(vinblastine)、ビンクリスチン(vincristine)、ブレオマイシン(bleomycin)、パクリタキセル(タキソール)[paclitaxel(taxol)]、ドセタキセル(タキソテア)[docetaxel(taxotere)]、アルデスロイキン(aldesleukin)、アスパラギナーゼ(asparaginase)、ブスルファン(busulfan)、カルボプラチン(carboplatin)、クラドリビン(cladribine)、カンプトテシン(camptothecin)、CPT−11、10−ヒドロキシ−7−エチル−カンプトテシン(SN38)[10−hydroxy−7−ethyl−camptothecin(SN38)]、フロクスウリジン(floxuridine)、フルダラビン(fludarabine)、ヒドロキシウレア(hydroxyurea)、イホスファミド(ifosfamide)、イダルビシン(idarubicin)、メスナ(mesna)、イリノテカン(irinotecan)、ミトキサントロン(mitoxantrone)、トポテカン(topotecan)、ロイプロリド(leuprolide)、メゲストロール(megestrol)、メルファラン(melpharan)、メルカプトプリン(mercaptopurine)、プリカマイシン(plicamycin)、ミトタン(mitotane)、ペガスパラガーゼ(pegaspargase)、ペントスタチン(pentostatin)、ピポブロマン(pipobroman)、ストレプトゾシン(streptozocin)、タモキシフェン(tamoxifen)、テニポシド(teniposide)、テストラクトン(testolactone)、チオグアニン(thioguanine)、チオテパ(thiotepa)、ウラシルマスタード(uracil mustard)、ビノレルビン(vinorelbine)、クロラムブシル(chlorambucil)、プレドニゾロン(prednisolone)、ビンデシン(vindesine)、ニムスチン(nimstine)、セムスチン(semustin)、カペシタビン(capecitabine)、トムデックス(tomudex)、アザシチジン(azacytidine)、UFT、オキザロプラチン(oxaloplatin)、ゲフィチニブ(イレッサ)[gefitinib(Iressa)]、イマチニブ(STI571)[imatinib(STI571)]、アムサクリン(amsacrine)、オール−トランスレチノイン酸(all−trans retinoic acid)、サリドマイド(thalidomide)、ベキサロテン(ターグレチン)[bexarotene(targretin)]、デキサメタゾン(dexamethasone)、アナストロゾール(アリミデックス)[anastrozole(Alimidex)]、ロイプリン(leuplin)あるいはこれらの組み合わせがあげられる。好ましくは、ビンクリスチン、シクロフォスファミド、エトポシド、メトトレキセートあるいはこれらの組み合わせがあげられる。
抗体としては、上述した抗体があげられる。
本発明におけるBRMとしては、BCG、嫌気性コリネバクテリア、ムラミルジペプチド、トレハロースジミコール酸等の細菌製剤、ピラン共重合体、MVE、ポリ I:C、ピリミジン、チモシン、チムリン、チモポエシン、ピシバニール、フコイダン、クレスチン等があげられる。
本発明の医薬の効果は、例えばin vitroの細胞障害活性測定系によって調べることができる。in vitroの細胞障害活性測定系の例としては、ADCC活性の測定系があげられる。ADCC活性は、抗体の存在下で、抗原を発現する標的細胞と、ヒトあるいはその他の動物より採取した末梢血単核球、単球、マクロファージ、顆粒球等のエフェクター細胞を接触させ、障害された標的細胞の度合いを検出し、これを定量することにより測定することができる。障害された標的細胞の度合いは、51Cr遊離法、標的細胞の酵素活性を検出する方法、フローサイトメーターによる検出法などによって検出することができる。ADCC活性測定系における本発明の免疫担当細胞を活性化させる物質または抗腫瘍活性を有する物質の効果は、ADCC活性測定系中にこれらの物質を添加するか、あるいは標的細胞、またはエフェクター細胞、またはその両者にあらかじめこれらの物質を一定期間曝露してADCC活性に与える影響を観察することにより、測定することができる。
また本発明の医薬の効果は、動物モデルを用いたin vivo抗腫瘍活性を測定することによっても調べることができる。
動物モデルとしては、ヌードマウス等の免疫不全マウスにヒト癌組織由来の培養細胞株を移植した異種移植モデル、培養マウス癌細胞株を正常な免疫系を有する野生型マウスへ移植した同系移植モデルなどがあげられる。
異種移植モデルはヌードマウス等の免疫不全マウスの皮下、皮内、腹腔内、静脈内等様々な部位にヒト癌細胞株を移植することにより作製することができる。
上記動物モデルを用いて抗体の単独投与、免疫担当細胞を活性化させる物質または抗腫瘍活性を有する物質の単独投与の効果と、本発明の医薬の効果とを比較することにより、本発明の医薬の抗腫瘍効果を評価することができる。
本発明の医薬は、単独で投与することも可能ではあるが、通常は薬理学的に許容される一つあるいはそれ以上の担体と一緒に混合し、製剤学の技術分野においてよく知られる任意の方法により製造した医薬製剤として提供するのが望ましい。
投与経路は、治療に際して最も効果的なものを使用するのが望ましく、経口投与、または口腔内、気道内、直腸内、皮下、筋肉内および静脈内等の非経口投与をあげることができ、蛋白質製剤の場合、望ましくは静脈内投与をあげることができる。
投与形態としては、噴霧剤、カプセル剤、錠剤、顆粒剤、シロップ剤、乳剤、座剤、注射剤、軟膏、テープ剤等があげられる。
経口投与に適当な製剤としては、乳剤、シロップ剤、カプセル剤、錠剤、散剤、顆粒剤等があげられる。
乳剤およびシロップ剤のような液体調製物は、水、ショ糖、ソルビトール、果糖等の糖類、ポリエチレングリコール、プロピレングリコール等のグリコール類、ごま油、オリーブ油、大豆油等の油類、p−ヒドロキシ安息香酸エステル類等の防腐剤、ストロベリーフレーバー、ペパーミント等のフレーバー類等を添加剤として用いて製造できる。
カプセル剤、錠剤、散剤、顆粒剤等は、乳糖、ブドウ糖、ショ糖、マンニトール等の賦形剤、デンプン、アルギン酸ナトリウム等の崩壊剤、ステアリン酸マグネシウム、タルク等の滑沢剤、ポリビニルアルコール、ヒドロキシプロピルセルロース、ゼラチン等の結合剤、脂肪酸エステル等の界面活性剤、グリセリン等の可塑剤等を添加剤として用いて製造できる。
非経口投与に適当な製剤としては、注射剤、座剤、噴霧剤等があげられる。
注射剤は、塩溶液、ブドウ糖溶液、あるいは両者の混合物からなる担体等を用いて調製される。
座剤はカカオ脂、水素化脂肪またはカルボン酸等の担体を用いて調製される。
また、噴霧剤は該医薬そのもの、ないしは受容者の口腔および気道粘膜を刺激せず、かつ該医薬を微細な粒子として分散させ吸収を容易にさせる担体等を用いて調製される。
担体として具体的には乳糖、グリセリン等が例示される。該医薬および用いる担体の性質により、エアロゾル、ドライパウダー等の製剤が可能である。また、これらの非経口剤においても経口剤で添加剤として例示した成分を添加することもできる。
投与量または投与回数は、目的とする治療効果、投与方法、治療期間、年齢、体重等により異なるが、通常成人1回当たり抗体量として0.1〜20mg/kgである。抗体と併用する薬剤は、単独で臨床に用いられる場合の投与量と同用量またはそれより少ない用量である。  An object of the present invention is to provide a medicament using an antibody composition having a high therapeutic effect and at least one kind of drug.
  The present invention relates to the following (1) to (14).
  (1) Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% A pharmaceutical comprising a combination of the antibody composition as described above and at least one drug.
  (2) Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% The pharmaceutical for administering combining the antibody composition which is the above, and at least 1 sort (s) of medicine.
  (3) Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% A medicament for administering the antibody composition as described above and at least one drug simultaneously or sequentially.
  (4) Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% The medicament according to any one of (1) to (3) above, which exhibits a higher therapeutic effect than when the antibody composition as described above is administered alone.
  (5) The medicament according to (4) above, wherein the high therapeutic effect is high antibody-dependent cytotoxic activity.
  (6) An antibody composition is produced from a cell having resistance to a lectin that recognizes a sugar chain structure in which N-acetylglucosamine at the N-glycoside-linked complex type sugar chain reducing terminal at position 6 and position 1 of fucose are α-linked. The medicament according to any one of (1) to (5) above, which is an antibody composition.
  (7) Among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the antibody composition is a sugar chain in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end. The medicament according to any one of (1) to (6) above, which is an antibody composition.
  (8) The sugar chain to which fucose is not bonded is a sugar chain in which the 1-position of the fucose is not α-bonded to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex type sugar chain reducing end ( The medicament according to any one of 1) to (7).
  (9) A cell in which the genomic gene of an enzyme involved in sugar chain modification in which the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing terminal is knocked out The medicament according to (7) or (8) above, which is an antibody composition produced from
  (10) The enzyme involved in sugar chain modification in which the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing terminal is α1,6-fucosyltransferase (9) ) The pharmaceutical described.
  (11) The medicament according to any one of (1) to (10), wherein the drug is a substance selected from the group consisting of a protein, a low-molecular drug, and a biological response modifier.
  (12) The medicament according to (11) above, wherein the protein is a cytokine or an antibody.
  (13) The medicament according to (12), wherein the cytokine is a cytokine selected from IFN-γ, IL-2, and IL-15.
  (14) The medicament according to any one of (1) to (13) above, wherein the medicament is a therapeutic agent for a disease involving a tumor.
  As a pharmaceutical form of the present invention, among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, a sugar in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end A pharmaceutical comprising a combination of an antibody composition having a chain ratio of 50% or more and at least one drug, among all N-glycoside-linked complex sugar chains that bind to the Fc region contained in the antibody composition, Pharmaceutical composition and antibody composition for administration in combination of an antibody composition in which the ratio of sugar chain in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more and at least one kind of drug Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained therein, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more An antibody composition, a medicament for administration of at least one agent at the same time or sequentially and the like.
  Here, the combined medicine refers to the non-fucose bound to the N-acetylglucosamine at the reducing end of the sugar chain among all the N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition. The pharmaceutical composition may be prepared by separately preparing an antibody composition having a sugar chain ratio of 50% or more and at least one drug, and combining these drugs simultaneously or sequentially. It may be a mixture in which drug components are mixed. In the mixture in which each drug component is mixed, fucose binds to N-acetylglucosamine at the reducing end of the sugar chain among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition. Also included are fusion antibodies in which at least one drug is bound to an antibody composition in which the proportion of untreated sugar chains is 50% or more.
  In the present invention, among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the ratio of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end The number of sugar chains in which fucose is not bound to N-acetylglucosamine at the reducing end of the sugar chain with respect to the total number of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the composition Refers to the proportion of The proportion of sugar chains is preferably the proportion of sugar chains in which the 1-position of fucose is not α-bonded to the 6-position of N-acetylglucosamine at the sugar chain reducing end.
  In the present invention, a sugar chain in which fucose is not bound to N-acetylglucosamine at the N-glycoside-linked complex type sugar chain reducing end means that the fucose is N-acetyl at the N-glycoside-linked complex type sugar chain reducing end. This refers to a sugar chain that is not α-bonded to glucosamine, specifically, a sugar chain in which the 1-position of the fucose is not α-bonded to the 6-position of N-acetylglucosamine of the N-glycoside-bonded complex sugar chain. .
  The antibody composition in the present invention includes any composition as long as it contains an antibody molecule having an N-glycoside-linked complex sugar chain in the Fc region.
  The antibody molecule is a tetramer in which two molecules of two kinds of polypeptide chains, a heavy chain and a light chain (hereinafter referred to as H chain and L chain, respectively) are associated with each other. About one-quarter of the N-terminal side of the H chain and about one-half (each 100 amino acids) of the N-terminal side of the L chain are called V regions and are rich in diversity and directly involved in antigen binding To do. Most of the parts other than the V region are called C regions. Antibody molecules are classified into IgG, IgM, IgA, IgD, and IgE classes based on C region homology.
  The IgG class is further classified into IgG1 to IgG4 subclasses based on the homology of the C region.
  The H chain is divided into four immunoglobulin domains, VH, CH1, CH2, and CH3, from the N-terminal side. Between CH1 and CH2, there is a highly flexible peptide region called a hinge region, and CH1 and CH2 are separated. . A structural unit consisting of CH2 and CH3 after the hinge region is called an Fc region, and an N-glycoside-linked sugar chain is bound thereto. In addition, this region is a region to which Fc receptor, complement, and the like are bound (Immunology Illustrated Original 5th Edition, issued February 10, 2000, Nanedo Edition, Introduction to Antibody Engineering, January 25, 1994) First edition, Jinjinshokan).
  The sugar chains of glycoproteins such as antibodies are linked to asparagine (N-glycoside-linked sugar chains) and to sugar chains (O-glycosyl-linked sugar chains) that bind to serine, threonine, etc. There are two main types. N-glycoside-linked sugar chains have a basic common core structure shown in the following structural formula (I) [Biochemical Experimental Method 23-Glycoprotein Sugar Chain Research Method (Academic Publishing Center) Etsuko Takahashi (1989) ]].
Figure 2005053742
  In the structural formula (I), the end of the sugar chain that binds to asparagine is called the reducing end, and the opposite side is called the non-reducing end.
  Any N-glycoside-linked sugar chain may be used as long as it has the core structure of the above structural formula (I), but a high mannose type in which only mannose binds to the non-reducing end of the core structure, non-core structure. One or more branches of galactose-N-acetylglucosamine (hereinafter referred to as Gal-GlcNAc) are provided in parallel on the reducing end side, and sialic acid and bisecting N are provided on the non-reducing end side of Gal-GlcNAc. -Complex type (complex type) having a structure such as acetylglucosamine, and a hybrid type having both a high mannose type and a complex type branch on the non-reducing terminal side of the core structure.
  The Fc region of an antibody molecule has a region where N-glycoside-linked sugar chains are bonded one by one, so that two sugar chains are bound per antibody molecule. The N-glucoside-linked sugar chain that binds to the antibody molecule includes any sugar chain that includes the core structure represented by the structural formula (I). Therefore, the two N-glucoside-linked sugar chains that bind to the antibody There will be many combinations of sugar chains.
  Therefore, the antibody composition in the present invention may be composed of an antibody molecule having a single sugar chain structure or an antibody having a plurality of different sugar chain structures as long as the effects of the present invention are obtained. It may be composed of molecules.
  The antibody molecule includes any molecule that contains the Fc region of an antibody. Specific examples include antibodies, antibody fragments, and fusion proteins containing an Fc region.
  As an antibody, an antibody is immunized with an antigen and secreted by a hybridoma cell prepared from a spleen cell of the immunized animal, an antibody prepared by gene recombination technology, that is, an antibody expression vector into which an antibody gene is inserted, And antibodies obtained by introduction into. Specific examples include antibodies produced by hybridomas, humanized antibodies, human antibodies, and the like.
  A hybridoma is a monoclonal antibody having a desired antigen specificity obtained by fusing a B cell obtained by immunizing a mammal other than a human with a myeloma cell derived from a mouse, a rat or the like. A cell that produces.
  Examples of humanized antibodies include human chimeric antibodies and human CDR-grafted antibodies.
  Human-type chimeric antibodies include non-human animal antibody H chain V regions (hereinafter also referred to as HV or VH) and antibody L chain V regions (hereinafter also referred to as LV or VL) and human antibody H chain C regions (hereinafter referred to as LV or VH). , Also referred to as CH) and an L chain C region of a human antibody (hereinafter also referred to as CL). Any animal other than humans can be used as long as it can produce hybridomas such as mice, rats, hamsters, rabbits and the like.
  Human chimeric antibodies are obtained by obtaining cDNAs encoding VH and VL from hybridomas producing monoclonal antibodies and inserting them into expression vectors for host cells having genes encoding human antibody CH and human antibody CL, respectively. A chimeric antibody expression vector can be constructed and expressed by being introduced into a host cell.
  The CH of the human chimeric antibody may be any as long as it belongs to human immunoglobulin (hereinafter referred to as hIg), but the hIgG class is preferred, and hIgG1, hIgG2, hIgG3, Any of the subclasses such as hIgG4 can be used. The CL of the human chimeric antibody may be any as long as it belongs to hIg, and those of κ class or λ class can be used.
  The human CDR-grafted antibody refers to an antibody obtained by grafting the VH and VL CDR amino acid sequences of non-human animal antibodies to appropriate positions of the human antibody VH and VL.
  The human CDR-grafted antibody constructs a cDNA encoding a V region obtained by grafting the VH and VL CDR sequences of a non-human animal antibody into the VH and VL CDR sequences of any human antibody, A human CDR-grafted antibody expression vector is constructed by inserting each into a host cell expression vector having a gene encoding CL of a human antibody, and the human CDR-grafted antibody is expressed by introducing the expression vector into the host cell. Can be manufactured.
  The CH of the human CDR-grafted antibody may be any as long as it belongs to hIg, but the hIgG class is preferable, and any of the subclasses such as hIgG1, hIgG2, hIgG3, and hIgG4 belonging to the hIgG class may be used. it can. The CL of the human CDR-grafted antibody may be any CL as long as it belongs to hIg, and those of κ class or λ class can be used.
  Human antibodies originally refer to antibodies that exist naturally in the human body, but human antibody phage libraries and human antibody transgenics created by recent advances in genetic engineering, cell engineering, and developmental engineering techniques. Antibodies obtained from non-human animals or human antibody transgenic plants are also included.
  The antibody present in the human body can be cultured, for example, by isolating human peripheral blood lymphocytes, infecting and immortalizing and cloning EB virus and the like, and culturing lymphocytes that produce the antibody. Can be purified.
  The human antibody phage library is a library in which antibody fragments such as Fab and single chain antibody are expressed on the phage surface by inserting antibody genes prepared from human B cells into the phage genes. From the library, phages expressing antibody fragments having a desired antigen-binding activity can be collected using the binding activity to the substrate on which the antigen is immobilized as an index. The antibody fragment can be further converted into a human antibody molecule comprising two complete heavy chains and two complete light chains by genetic engineering techniques.
  A human antibody transgenic non-human animal refers to an animal in which a human antibody gene is incorporated into cells. Specifically, a human antibody transgenic non-human animal can be produced by introducing a human antibody gene into a mouse embryonic stem cell, transplanting the embryonic stem cell to an early embryo of another mouse, and generating it. A human antibody transgenic non-human animal can also be produced by introducing a human antibody gene into a fertilized egg of an animal and generating the fertilized egg. A human antibody is produced from a non-human animal transgenic non-human animal by using a conventional method for producing a hybridoma in a mammal other than a human to obtain a human antibody hybridoma and cultivate it to accumulate the human antibody in the culture. Can be made.
  Examples of transgenic non-human animals include cattle, sheep, goats, pigs, horses, mice, rats, chickens, monkeys, and rabbits.
  An antibody fragment refers to a fragment containing at least a part of the Fc region of the antibody. The Fc region refers to a region on the C-terminal side of the H chain of an antibody, a CH2 region, and a CH3 region. At least a part of the Fc region preferably refers to a fragment containing the CH2 region, more preferably a region containing the first aspartic acid present in the CH2 region. The Fc region of the IgG class is described in Kabat et al. EU Index [Sequences of Proteins of Immunological Interest, 5th  Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)], from the 226th Cys to the C-terminal, or from the 230th Pro to the C-terminal. Examples of antibody fragments include H chain monomers and H chain dimers. The Fc region in the present invention includes natural types and mutants thereof.
  A fusion protein having a part of an Fc region is a substance obtained by fusing an antibody or antibody fragment containing a part of an Fc region of an antibody with a protein such as an enzyme or a cytokine (hereinafter referred to as an Fc fusion protein). Say.
  Of all the N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition of the present invention, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50%. Or more, preferably 60% or more, more preferably 70% or more, still more preferably 80% or more, particularly preferably 90% or more, and all N-glycosidic conjugates that bind to the Fc region contained in the antibody composition Most preferably, the type sugar chain is a sugar chain in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end.
  As described above, among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is high. The higher the antibody composition, the higher the antibody-dependent cytotoxic activity.
  The ratio of the sugar chain in which fucose is not bound to N-acetylglucosamine at the reducing end of the sugar chain contained in the composition consisting of the antibody molecule having an N-glycoside-linked complex type sugar chain in the Fc region is determined from the antibody molecule to the hydrazine Using known methods such as degradation and enzymatic digestion [Biochemical Experimental Method 23-Glycoprotein Glycan Research Method (Academic Publishing Center) Takahashi Etsuko (1989)], sugar chains are released and the released sugar chains are fluorescent. It can be determined by labeling or isotope labeling and separating the labeled sugar chain by a chromatographic method. It can also be determined by analyzing the released sugar chain by the HPAED-PAD method [J. Liq. Chromatogr., 6, 1577 (1983)].
  In the present invention, examples of the high therapeutic effect include high cytotoxic activity.
  The cytotoxic activity of the antibody composition includes antibody-dependent cytotoxic activity (hereinafter abbreviated as ADCC), complement-dependent cytotoxic activity (monoclonal antibodies), and antigen-expressing cells that bind to the antigen. Examples thereof include growth inhibitory activity. Antiproliferative activity includes those that promote apoptosis and differentiation induction of target cells [Cancer Research]60, 7170 (2000), Nature Medicine.1644 (1995), Cell Growth Differ.3, 401 (1992)].
  ADCC activity means that an antibody bound to a cell surface antigen such as a tumor cell in a living body activates an effector cell through binding of an antibody Fc region to an Fc receptor present on the effector cell surface. It refers to the activity of damaging cells and the like [Monoclonal Antibodies: Principles and Applications, Wiley-Liss, Inc. , Capter 2.1 (1995)]. Examples of effector cells include immune cells such as natural killer cells, macrophages, monocytes, dendritic cells, and granulocytes. Fc receptor is Fcα receptor I, Fcε receptor I, Fcε receptor II, Fcγ receptor I, Fcγ receptor IIa, Fcγ receptor IIb, Fcγ receptor IIc, Fcγ receptor IIIa, Fcγ receptor IIIb, It is divided into types such as Fc receptor n. Among them, Fcγ receptor IIIa is mainly expressed on natural killer cells and is one of Fc receptors important for ADCC activity [Monoclonal Antibodies: principals and practices. ), Third Edition, Acad. Press, 1996 (hereinafter abbreviated as Monoclonal Antibodies)].
  Therefore, in the present invention, among all N-glycoside-bonded complex sugar chains that bind to the Fc region of an antibody molecule, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more. N-acetyl at the reducing end of the sugar chain of all N-glycoside-bonded complex sugar chains that bind to the Fc region of the antibody molecule means higher therapeutic effect than when the antibody composition is administered alone. By combining an antibody composition in which the proportion of the sugar chain in which fucose is not bound to glucosamine is 50% or more and at least one kind of drug, the therapeutic effect exhibited when the antibody composition alone is administered. Refers to a drug that exhibits a high therapeutic effect.
  In addition, the pharmaceutical of the present invention has a ratio of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end of all N-glycoside-bonded complex sugar chains that bind to the Fc region of the antibody molecule. % Higher than that of a pharmaceutical comprising a combination of an antibody composition of less than% and at least one drug.
  The medicament of the present invention can reduce the amount of the drug as compared with the case where the drug is used alone, and can reduce the side effects which are a concern when a high dose of the drug alone is administered to a patient.
  A cell having resistance to a lectin recognizing a sugar chain structure in which the 6-position of N-acetylglucosamine at the N-glycoside-binding complex type sugar chain reducing end and the 1-position of fucose are α-linked (hereinafter referred to as α1,6-fucose / lectin resistance) Cell)) is a cell capable of producing an antibody composition such as yeast, animal cell, insect cell, plant cell, etc., which is the 6-position of N-acetylglucosamine at the N-glycoside-linked complex type sugar chain reducing end. Any cell may be used as long as it has lectin resistance and recognizes a sugar chain structure in which the 1-position of fucose is α-linked.
  Specifically, a hybridoma cell, a human antibody, which has resistance to a lectin that recognizes a sugar chain structure in which the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing terminal and the 1-position of fucose are α-linked Host cells for producing humanized antibodies, embryonic stem cells and fertilized egg cells for producing transgenic non-human animals for producing human antibodies, and for producing transgenic plants for producing human antibodies Plant callus cells, myeloma cells, cells derived from transgenic non-human animals and the like can be mentioned. Myeloma cells can be used as fusion cells when producing hybridoma cells. Moreover, it can be used for immunizing a transgenic non-human animal with an antigen, taking out the spleen cells of the animal, and producing hybridoma cells.
  A cell having resistance to lectin refers to a cell whose growth is not inhibited even when cell culture is performed with an effective concentration of lectin applied to a culture medium.
  In the present invention, the effective lectin concentration at which growth is not inhibited may be appropriately determined according to the cell line, but is usually 10 μg / ml to 10.0 mg / ml, preferably 0.5 to 2.0 mg / ml. When the mutation is introduced into the parent cell, the effective concentration of the lectin is not less than the concentration at which the parent cell cannot grow normally, preferably the same concentration as the concentration at which the parent cell cannot grow normally, more preferably 2 to 5 times. More preferably, the concentration is 10 times, most preferably 20 times or more.
  The parent cell is a cell before any treatment, that is, a cell before the step of selecting α1,6-fucose / lectin resistant cells used in the present invention, a gene for reducing or eliminating the above-mentioned enzyme activity. A cell before engineering treatment.
  The parent cell is not particularly limited, but specific examples of the parent cell of various cell lines include the cells shown below.
  NS0 cell parent cell lines include Bio / Technology (BIO / TECHNOLOGY),10169 (1992), Biotechnol. Bioeng.,73, 261, (2001), NS0 cells described in the literature. In addition, NS0 cell line (RCB0213) registered with RIKEN Cell Development Bank, or sub-strains in which these strains are conditioned to a medium capable of growth.
  As the parent cell line of SP2 / 0-Ag14 cells, Journal of Immunology (J. Immunol.),126, 317 (1981), Nature,276269 (1978), Human Antibodies and Hybridomas,3, 129 (1992), and the like, SP2 / 0-Ag14 cells. In addition, SP2 / 0-Ag14 cells (ATCC CRL-1581) registered in the ATCC or sub-strains (ATCC CRL-1581.1) in which these strains are conditioned in a viable medium are also included.
  As a parent cell of CHO cells derived from Chinese hamster ovary tissue, Journal of Experimental Medicine,108945 (1958), Proc. Natl. Acad. Sei. USA,60, 1275 (1968), Genetics,55, 513 (1968), Chromosoma,41129 (1973), Methods in Cell Science,18, 115 (1996), Radiation Research,148, 260 (1997), Proc. Natl. Acad. Sci. USA,774216 (1980), Proc. Natl. Acad. Sci.60, 1275 (1968), Cell,6121 (1975), Molecular Cell Genetics, Appendix I, II (p883-900), and the like. In addition, CHO-K1 strain (ATCC CCL-61), DUXB11 strain (ATCC CRL-9096), Pro-5 strain (ATCC CRL-1781) registered in ATCC, commercially available CHO-S strain (Lifetechnologies Cat #) 11619), or substrains obtained by acclimating these strains to a medium capable of growth.
  Rat myeloma cell line YB2 / 3HL. P2. G11.16 Ag. The parent cell of 20 cells includes a cell line established from Y3 / Ag1.2.3 cells (ATCC CRL-1631). Specific examples thereof include J. Org. Cell. Biol. ,93576 (1982), Methods Enzymol. ,73B, 1 (1981) and the like described in the literature such as YB2 / 3HL. P2. G11.16 Ag. There are 20 cells. In addition, YB2 / 3HL. P2. G11.16 Ag. Examples thereof include 20 cells (ATCC CRL-1662) or sub-strains obtained by acclimating these strains to a medium capable of growth.
  As a lectin that recognizes a sugar chain structure in which the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing terminal and the 1-position of fucose are α-bonded, any lectin that can recognize the sugar chain structure can be used. Lectins are also included. Specific examples thereof include lentil lectin LCA (Lens  CulinarisLentil Agglutinin), pea lectin PSA (Pisum  sativumPea Lectin), broad bean lectin VFA (Vicia  fabaDerived from Agglutinin), Hilochawantake lectin AAL (Aleuria  aurantia(Lectin of origin) and the like.
  In the present invention, the α1,6-fucose / lectin-resistant cell may be any cell as long as growth is not inhibited in the presence of a certain effective concentration of lectin. For example, the activity of at least one protein listed below Are cells that are reduced or deleted from the parent cell line.
  (A) an enzyme protein involved in the synthesis of intracellular sugar nucleotide GDP-fucose (hereinafter referred to as “GDP-fucose synthase”);
  (B) Enzyme protein involved in sugar chain modification in which the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing terminal (hereinafter referred to as “α1,6-fucose modifying enzyme”) );
  (C) Protein involved in transport of intracellular sugar nucleotide GDP-fucose to the Golgi apparatus (hereinafter referred to as “GDP-fucose transport protein”).
  The GDP-fucose synthase includes any enzyme that is involved in the synthesis of the sugar nucleotide GDP-fucose, which is the source of fucose to the sugar chain in the cell, and the synthesis of the intracellular sugar nucleotide GDP-fucose. Enzymes that affect
  The intracellular sugar nucleotide GDP-fucose is supplied by a de novo synthesis route or a salvage synthesis route. Therefore, all enzymes involved in these synthetic pathways are included in the GDP-fucose synthase.
  The GDP-fucose synthase involved in the de novo synthesis pathway includes GDP-mannose 4-dehydratase (GDP-mannose 4-dehydratase; hereinafter referred to as GMD), GDP-keto-6-deoxymannose 3,5-epimerase. , 4-reductase (GDP-keto-deoxymannose 3,5-epimerase, 4-reductase; hereinafter referred to as Fx).
  Examples of the GDP-fucose synthase involved in the Salvage synthesis pathway include GDP-beta-L-fucose pyrophosphorylase (GDP-beta-L-fucose-pyrophosphorylase; hereinafter referred to as GFPP), Fucokinase (fucokinase) and the like. can give. Examples of GMD include GMD having the amino acid sequence represented by SEQ ID NO: 1, 2 or 3.
  Examples of the enzyme that affects the synthesis of intracellular sugar nucleotide GDP-fucose include the substances that affect the activity of the enzyme involved in the above-described intracellular sugar nucleotide GDP-fucose synthesis pathway, and substances that serve as substrates for the enzyme. Enzymes that affect structure are also included.
  The α1,6-fucose modifying enzyme includes any enzyme as long as it is an enzyme involved in a reaction in which the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end and the 1-position of fucose are α-bonded. The As an enzyme involved in the reaction of α-bonding the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end and the 1-position of fucose, N-acetylglucosamine at the N-glycosidically linked sugar chain reducing end Any enzyme can be used as long as it affects the reaction of α-bonding the 6th position of 1 and the 1st position of fucose. Specific examples include α1,6-fucosyltransferase and α-L-fucosidase. Examples of α1,6-fucosyltransferase include α1,6-fucosyltransferase having the amino acid sequence represented by SEQ ID NO: 4 or 5.
  In addition, as an enzyme that affects the reaction in which the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end and the 1-position of fucose are α-bonded, the above-mentioned N-glycoside-linked complex sugar chain reducing end Also included are enzymes that affect the activity of the enzyme involved in the α-bonding of the 1-position of fucose to the 6-position of N-acetylglucosamine and the structure of the substance that is the substrate of the enzyme.
  The GDP-fucose transport protein includes any protein as long as it is a protein involved in the transport of intracellular sugar nucleotide GDP-fucose to the Golgi body, and specifically includes a GDP-fucose transporter.
  In addition, proteins that affect the reaction of transporting intracellular sugar nucleotide GDP-fucose into the Golgi are also included in the GDP-fucose transport protein. Specifically, transport of the above-mentioned intracellular sugar nucleotide GDP-fucose to the Golgi body. Examples include proteins that affect the activity of proteins involved in or affect expression.
  As a method for obtaining cells used in the production method of the present invention, any method can be used as long as it can select α1,6-fucose / lectin resistant cells. Specific examples include a technique for reducing or deleting the activity of the above-described protein. As a technique for reducing or deleting the activity of the above-mentioned protein,
  (A) a gene disruption technique targeting protein genes;
  (B) a technique for introducing a dominant negative form of a protein gene;
  (C) a technique for introducing a mutation in a protein;
  (D) a technique for suppressing transcription or translation of a protein gene;
  (WO02 / 31140, WO03 / 085107, WO03 / 085118).
  In the present invention, N-glycoside-linked complex type sugar chain reducing terminal N-acetylglucosamine 6-position N-acetylglucosamine, the cell where the genomic gene of the enzyme involved in sugar chain modification in which the 1-position of fucose is α-linked is knocked out -Cells in which the expression or function of the genome gene of the enzyme involved in the sugar chain modification in which the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the reducing end of the glycoside-linked complex is completely suppressed. It is done. Therefore, in the present invention, the cell in which the genomic gene is knocked out can be obtained by using the method described below, wherein the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end. Examples include cells in which the genomic gene of an enzyme involved in sugar chain modification is knocked out.
  A specific example of a cell in which a genomic gene has been knocked out is a cell in which all or part of the target gene has been deleted from the genome. As a method for obtaining such cells, any method can be used as long as the target genome can be modified, but a genetic engineering method is desirable. Specific examples of the technique include a technique for reducing or deleting the activity of the enzyme described above.
  Moreover, by using the above-mentioned method of selecting a cell line resistant to a lectin that recognizes a sugar chain structure in which the 6-position of N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain and the 1-position of fucose are α-linked. A cell in which the genomic gene of the enzyme involved in the sugar chain modification in which the 1-position of fucose is α-bonded to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end is knocked out can be selected.
  Examples of the antibody composition in the present invention include an antibody composition against an antigen expressed in a cell associated with a disease or an antigen involved in pathogenesis such as proliferation and metastasis of a cell associated with a disease. Specifically, GD2, GD3, GM2, HER2, CD20, CD22, CD25, CD33, CD52, MAGE, HM1.24, CC chemokine receptor 4 (CCR4), parathyroid hormone related protein (PTHrP), basic fibroblast Cell growth factor, fibroblast growth factor 8, basic fibroblast growth factor receptor, fibroblast growth factor 8 receptor, epidermal growth factor receptor (EGFR), epithelial cell adhesion molecule (EpCam), insulin Antibody composition against insulin-like growth factor, insulin-like growth factor receptor, PMSA, vascular endothelial growth factor, vascular endothelial growth factor receptor, respiratory syncytial virus (RSV), IL-5 receptor α chain, etc. It is done.
  Specific examples of the antibody composition include an anti-GD2 antibody [Anticancer Res.,13331 (1993)], anti-GD3 antibody [Cancer Immunol. Immunother.],36, 260 (1993), WO01 / 23432, US6437098], anti-GM2 antibody [Cancer Res.,54, 1511 (1994), US6423511], anti-HER2 antibody [Procedure of the National Academy of Sciences (Proc. Natl. Acad. Sci. USA),894285 (1992)], anti-CD20 antibody [Blood,83, 435 (1994), WO 03/55993], anti-CD22 antibody [Semimin. Oncol.,30, 253 (2003)], anti-CD25 antibody [Human Antibodies],10127-142 (2001)], anti-CD33 antibody [J. Clin. Oncol.],193244 (2001)], anti-CD52 antibody [Procedure of the National Academy of Sciences (Proc. Natl. Acad. Sci. USA),894285 (1992)], anti-MAGE antibody [British J. Cancer,83493 (2000)], anti-HM1.24 antibody [Molecular Immunol.,36, 387 (1999)], anti-CCR4 antibody [WO01 / 64754, WO03 / 18635, WO00 / 42074, WO99 / 15666, US62453332], anti-parathyroid hormone-related protein (PTHrP) antibody [Cancer,88, 2909 (2000)], anti-fibroblast growth factor 8 antibody [Procedure of the National Academy of Sciences (Proc. Natl. Acad. Sci. USA),86, 9911 (1989), WO 03/002608], anti-fibroblast growth factor 8 receptor antibody [J. Biol. Chem.],265, 16455 (1990)], anti-epidermal growth factor receptor antibody [Cancer Res.,59, 1236 (1999)], anti-epithelial cell adhesion molecule antibody [Procedure of the National Academy of Sciences (Proc. Natl. Acad. Sci. USA),761438 (1979)], anti-insulin-like growth factor antibody [J. Neurosci. Res.,40, 647 (1995), WO 03/93317], anti-insulin-like growth factor receptor antibody [J. Neurosci. Res.],40, 647 (1995)], anti-PMSA antibody [J. Urology, 160, 2396 (1998)], anti-vascular endothelial growth factor antibody [Cancer Research.574593 (1997)], anti-vascular endothelial cell growth factor receptor antibody [Oncogene,19, 2138 (2000)], anti-RSV antibody [Antiviral Research.47, 57-77 (2000)], anti-IL-5 receptor α chain antibody (US6018032, US65538111) and the like. Specific antibody names include Herceptin, Rituxan, Campath, Avastin, Bexar, Lymphoside, Mylotarg, Panorex, Zevalin [Nature. Rev. Cancer,1, 118 (2001)], Zenapax, Simulect, Remicade, Synagis, ReoPro [Human Antibodies,10, 127-142 (2001)], Enbrel [Expert Opinion on Pharmacotherapy,21137-1148 (2001)], Zolair [Respiratory Medicine,97, 123-129 (2003)].
  In the present invention, ADCC activity is51By Cr release method, lactate dehydrogenase (LDH) release method, flow cytometry, etc.in vitroUsing a measurement system or animal modelin vivoIt is possible to measure with an evaluation system.
  In the present invention, of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50 Examples of the drug used together with the antibody composition of at least% include cytokines, proteins such as antibodies, low molecular drugs, biological response modifiers (hereinafter referred to as BRM), and the like.
  Cytokines include interleukins (IL), colony stimulating factor (CSF), interferon (IFN), tumor necrosis factor (TNF), chemokines, or combinations thereof. Specifically, IFN-α, IFN-β, IFN-γ, TNF-α, TNF-β, IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-6, IL -7, IL-9, IL-12, IL-15, IL-17, IL-18, IL-21, fractalkine, GM-CSF, G-CSF, M-CSF, SCF, or combinations thereof . IL-2, IL-15, IFN-α, and IFN-γ are preferable.
  Examples of low molecular weight drugs include amifostine (ethiol) [amifostine (ethyl)], cisplatin (daplatin) (DTIC) [dacarbazine (DTIC)], dactinomycin (dactinomycin), mechloretamine (nitrogen mustard) nitrogenmustard)], streptozocin, cyclophosphamide, carmustine (BCNU) [admuthine (BCNU)], romustine (CCNU) , Doxo Bicine lipo (doxyl) [doxorubicinlipo (doxil)], gemcitabine (gemzar) [gemcitabine (gemzar)], daunorubicin (daunorubicin), daunorubicinpine (daunorubicinpine) (Cytarabine), etoposide, methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, bleomycin (Paclitaxel (taxol)), docetaxel (taxotere), aldesleukin, asparaginase, busulfan, carboplatin, carboplatin, carboplatin, carboplatin camptothecin), CPT-11, 10-hydroxy-7-ethyl-camptothecin (SN38) [10-hydroxy-7-ethyl-camptothecin (SN38)], floxuridine, fludarabine, hydroxyurea (urix) ), Ifosfamide (Ifosfamide), idarubicin, mesna, irinotecan, mitoxantrone, topotecan, leuprolide, megestol, megestol Purine (mercaptopurine), prikamycin, mitotane, pegasparagase, pentostatin, pipobroman, streptozocide (instreptozect) oside), test lactone (testolactone), thioguanine, thiotepa, uracil mustard (uracil mustard), vinorelbine (n), chlorambucil (n) Semestin, capecitabine, tomudex, azacitidine, UFT, oxaloplatin, gefitinib (Iressa) [gefitinib (ST) matinib (STI571)], amsacrine, all-trans retinoic acid, thalidomide, bexarotene (targretin) (ex), dexamethasole (ex) ) [Anatrozole (Alimidex)], leuplin, or a combination thereof. Preferably, vincristine, cyclophosphamide, etoposide, methotrexate, or a combination thereof is used.
  Examples of the antibody include the above-described antibodies.
  BRM in the present invention includes BCG, anaerobic corynebacteria, muramyl dipeptide, trehalose dimycolic acid and other bacterial preparations, pyran copolymer, MVE, poly I: C, pyrimidine, thymosin, thymrin, thymopoesin, pisibanil, fucoidan And krestin.
  The effect of the medicament of the present invention is, for example,in vitroIt can be examined by a cytotoxic activity measuring system.in vitroExamples of the cytotoxic activity measurement system include an ADCC activity measurement system. ADCC activity was impaired by contacting target cells expressing the antigen with effector cells such as peripheral blood mononuclear cells, monocytes, macrophages and granulocytes collected from humans or other animals in the presence of antibodies. It can be measured by detecting the degree of the target cell and quantifying it. The degree of target cells damaged is51It can be detected by a Cr release method, a method for detecting enzyme activity of a target cell, a detection method using a flow cytometer, and the like. The effect of the substance that activates the immunocompetent cell of the present invention or the substance having antitumor activity in the ADCC activity measurement system is determined by adding these substances in the ADCC activity measurement system, or by targeting the target cell, effector cell, or It can be measured by exposing these substances to both of them in advance for a certain period and observing the effect on ADCC activity.
  The effect of the medicament of the present invention was obtained using an animal model.in vivoIt can also be examined by measuring antitumor activity.
  Animal models include xenotransplantation models in which cultured cell lines derived from human cancer tissue are transplanted into immunodeficient mice such as nude mice, syngeneic transplantation models in which cultured mouse cancer cell lines are transplanted into wild-type mice having a normal immune system, etc. Can be given.
  A xenograft model can be prepared by transplanting human cancer cell lines to various sites such as subcutaneous, intradermal, intraperitoneal, intravenous, etc. of immunodeficient mice such as nude mice.
  By comparing the effects of single administration of an antibody, a substance that activates immunocompetent cells or a substance having antitumor activity, and the effect of the medicament of the present invention using the above animal model, the medicament of the present invention The antitumor effect of can be evaluated.
  The medicament of the present invention can be administered alone, but usually mixed together with one or more pharmacologically acceptable carriers, and any well known in the technical field of pharmaceutics It is desirable to provide it as a pharmaceutical preparation produced by the method.
  It is desirable to use the most effective route for treatment, and oral administration or parenteral administration such as buccal, intratracheal, rectal, subcutaneous, intramuscular and intravenous can be used. In the case of a preparation, intravenous administration can be preferably mentioned.
  Examples of the dosage form include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
  Suitable formulations for oral administration include emulsions, syrups, capsules, tablets, powders, granules and the like.
  Liquid preparations such as emulsions and syrups include saccharides such as water, sucrose, sorbitol and fructose, glycols such as polyethylene glycol and propylene glycol, oils such as sesame oil, olive oil and soybean oil, p-hydroxybenzoic acid Preservatives such as esters, and flavors such as strawberry flavor and peppermint can be used as additives.
  Capsules, tablets, powders, granules, etc. are excipients such as lactose, glucose, sucrose, mannitol, disintegrants such as starch and sodium alginate, lubricants such as magnesium stearate and talc, polyvinyl alcohol, hydroxy A binder such as propylcellulose and gelatin, a surfactant such as fatty acid ester, a plasticizer such as glycerin and the like can be used as additives.
  Formulations suitable for parenteral administration include injections, suppositories, sprays and the like.
  The injection is prepared using a carrier made of a salt solution, a glucose solution, or a mixture of both.
  Suppositories are prepared using a carrier such as cacao butter, hydrogenated fat or carboxylic acid.
  The spray is prepared using a carrier that does not irritate the drug itself or the recipient's oral cavity and airway mucosa, and disperses the drug as fine particles to facilitate absorption.
  Specific examples of the carrier include lactose and glycerin. Depending on the properties of the medicine and the carrier used, preparations such as aerosols and dry powders are possible. In these parenteral preparations, the components exemplified as additives for oral preparations can also be added.
  The dose or frequency of administration varies depending on the intended therapeutic effect, administration method, treatment period, age, body weight, etc., but is usually 0.1-20 mg / kg as the amount of antibody per adult. The drug used in combination with the antibody is the same dose or less than the dose when used clinically alone.

第1図は、KM2760−1とKM3060のADCC活性に対する各種サイトカインの増強効果を示す。縦軸は細胞障害活性(%)を表す。■はサイトカイン非添加、□はIL−2添加、斜線はIL−15添加時の細胞障害活性をそれぞれ表す。
第2図は、KM3065とRituxanのADCC活性に対する各種サイトカインの増強効果を示す。縦軸は細胞障害活性(%)を表す。図において■はサイトカイン非添加、□はIL−2添加、斜線はIL−15添加時の細胞障害活性をそれぞれ表す。
第3図は、KM3065とRituxanのADCC活性に対する各種サイトカインの増強効果を示す。縦軸は細胞障害活性(%)を表す。図において■はサイトカイン非添加、□はIFN−α添加、斜線はIFN−γ添加時の細胞障害活性をそれぞれ表す。
以下、本発明を詳細に説明する。本願は、2003年12月4日に出願された日本国特許出願2003−406589号の優先権を主張するものであり、当該特許出願の明細書及び/または図面に記載される内容を包含する。
FIG. 1 shows the enhancement effect of various cytokines on the ADCC activities of KM2760-1 and KM3060. The vertical axis represents cytotoxic activity (%). (2) indicates no cytotoxic activity, □ indicates IL-2 addition, and hatched lines indicate cytotoxic activity when IL-15 is added.
FIG. 2 shows the enhancing effect of various cytokines on ADCC activity of KM 3065 and Rituxan. The vertical axis represents cytotoxic activity (%). In the figure, ■ represents the absence of cytokine, □ represents the addition of IL-2, and the diagonal line represents the cytotoxic activity upon addition of IL-15.
FIG. 3 shows the enhancement effect of various cytokines on ADCC activity of KM 3065 and Rituxan. The vertical axis represents cytotoxic activity (%). In the figure, ■ represents the absence of cytokine, □ represents the addition of IFN-α, and the diagonal line represents the cytotoxic activity when IFN-γ was added.
Hereinafter, the present invention will be described in detail. This application claims the priority of the Japan patent application 2003-406589 for which it applied on December 4, 2003, and includes the content described in the specification and / or drawing of the said patent application.

in vitro細胞障害活性における抗CCR4抗体とサイトカインの併用効果
in vitro細胞障害活性における抗CCR4ヒト型キメラ抗体KM2760−1およびKM3060(WO02/31140)とサイトカインの併用効果を以下の方法により測定した。
KM2760−1はラットミエローマ細胞株YB2/0細胞に抗CCR4キメラ抗体発現株を導入して得られた細胞株KM2760−1#58−35−16より生産された抗CCR4キメラ抗体である。KM2760−1#58−35−16株はレクチン耐性能を有し、KM2760−1の全糖鎖中のフコースが結合しない糖鎖含量は87%である。一方KM3060はCHO/DG44細胞株に抗CCR4キメラ抗体発現株を導入して得られた細胞株5−03より生産された抗CCR4キメラ抗体である。KM3060はレクチン耐性能を有さず、KM3060の全糖鎖中のフコースが結合しない糖鎖含量は8%である。KM2760−1とKM3060は同じアミノ酸配列を有し、抗原結合活性も同等である。
(a)エフェクター細胞溶液の調製
健常人静脈血50mLを採取し、ヘパリンナトリウム(清水製薬社製)0.5mLを加え穏やかに混ぜた。これをMONO−POLY分離溶液(大日本製薬株式会社製)を用いて使用説明書に従い、遠心分離(400g、20分間)して単核球層を分離した。RPMI1640−FCS(5)培地で3回遠心分離して洗浄後、同培地を用いて3×10細胞/mLの濃度で再懸濁し、エフェクター細胞溶液とした。
(b)エフェクター細胞のサイトカインによる刺激
上記(a)で得られたエフェクター細胞溶液を50μLずつ、96穴U底プレート(Falcon社製)に分注した。さらにRPMI1640−FCS(5)で希釈した以下の溶液を50μLずつ添加し、5%COインキュベーター内にて3日間静置した。
(1)RPMI1640−FCS(5)のみ(サイトカイン非添加コントロール)
(2)IL−2(ペプロテック社製)2ng/mL
(3)IL−15(ペプロテック社製)2ng/mL
(c)標的細胞溶液の調製
G418(ナカライテスク社製)を0.5mg/mLで含むRPMI1640−FCS(10)培地(FCSを10%含むRPMI1640培地(GIBCO BRL社製))で培養したCCR4/EL4細胞(CCR4遺伝子を導入したマウス胸腺腫細胞、WO01/64754)を遠心分離操作及び懸濁によりRPMI1640−FCS(5)培地(FCSを5%含むRPMI1640培地(GIBCO BRL社製))で洗浄した後、RPMI1640−FCS(5)培地によって、2×10細胞/mLに調製し、標的細胞溶液とした。
(d)細胞障害活性の測定
(b)で調製した96ウェルU字底プレートの各ウェルに(c)で調製した標的細胞溶液の50μL(1×10細胞/ウェル)を分注した。このときエフェクター細胞と標的細胞の比は15:1となる。更に、KM2760−1、あるいはKM3060を各最終濃度1ng/mLあるいは100ng/mLとなるように加えて全量を200μLとし、37℃で4時間反応させた。反応後、プレートを遠心分離し、上清中の乳酸デヒドロゲナーゼ(LDH)活性を、CytoTox96 Non−Radioactive Cytotoxicity Assay(Promega社製)を用いて、添付の説明書にしたがって吸光度データを取得することで測定した。標的細胞自然遊離の吸光度データは、エフェクター細胞溶液、抗体溶液の代わりに培地のみを用いて、また、エフェクター細胞自然遊離の吸光度データは、標的細胞溶液、抗体溶液の代わりに培地のみを用いて、上記と同様の操作を行うことで取得した。標的細胞全遊離の吸光度データは、抗体溶液、エフェクター細胞溶液の代わりに培地を用い、反応終了45分前に15μLの9%Triton X−100溶液を添加し、上記と同様の操作を行い、上清のLDH活性を測定することにより求めた。ADCC活性は次式により求めた。

Figure 2005053742
第1図に結果を図示した。サイトカイン非添加時のKM2760−1のADCC活性はKM3060のADCC活性よりも高いが、サイトカインの添加によりさらに増強された。この結果は、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物の高いADCC活性は、サイトカインによってさらに上昇することを示す。Combined effect of anti-CCR4 antibody and cytokine on cytotoxicity in vitro
The combined effect of anti-CCR4 human chimeric antibodies KM2760-1 and KM3060 (WO02 / 31140) and cytokines in in vitro cytotoxic activity was measured by the following method.
KM2760-1 is an anti-CCR4 chimeric antibody produced from the cell line KM2760-1 # 58-35-16 obtained by introducing an anti-CCR4 chimeric antibody expression strain into the rat myeloma cell line YB2 / 0 cells. The KM2760-1 # 58-35-16 strain has lectin resistance, and the sugar chain content to which fucose in all the sugar chains of KM2760-1 does not bind is 87%. On the other hand, KM3060 is an anti-CCR4 chimeric antibody produced from the cell line 5-03 obtained by introducing an anti-CCR4 chimeric antibody expression strain into the CHO / DG44 cell line. KM3060 does not have lectin resistance, and the glycan content in which fucose in all glycan chains of KM3060 does not bind is 8%. KM2760-1 and KM3060 have the same amino acid sequence and antigen binding activity is also equivalent.
(A) Preparation of effector cell solution 50 mL of healthy human venous blood was collected and 0.5 mL of heparin sodium (manufactured by Shimizu Pharmaceutical Co., Ltd.) was added and gently mixed. This was centrifuged (400 g, 20 minutes) using a MONO-POLY separation solution (manufactured by Dainippon Pharmaceutical Co., Ltd.) to separate the mononuclear cell layer. After washing by centrifugation 3 times with RPMI1640-FCS (5) medium, the suspension was resuspended at a concentration of 3 × 10 6 cells / mL using the same medium to obtain an effector cell solution.
(B) Stimulation of effector cells by cytokines 50 μL of the effector cell solution obtained in (a) above was dispensed into 96-well U-bottom plates (Falcon). Further, 50 μL of the following solutions diluted with RPMI 1640-FCS (5) were added, and allowed to stand in a 5% CO 2 incubator for 3 days.
(1) RPMI 1640-FCS (5) only (control without cytokine addition)
(2) IL-2 (manufactured by Peprotech) 2 ng / mL
(3) IL-15 (manufactured by Peprotech) 2 ng / mL
(C) Preparation of target cell solution CCR4 / cultured in RPMI 1640-FCS (10) medium (RPMI 1640 medium (GIBCO BRL) containing 10% FCS) containing G418 (Nacalai Tesque) at 0.5 mg / mL EL4 cells (mouse thymoma cells introduced with CCR4 gene, WO01 / 64754) were washed with RPMI1640-FCS (5) medium (RPMI1640 medium (GIBCO BRL) containing 5% FCS) by centrifugation and suspension. Then, it adjusted to 2 * 10 < 5 > cell / mL with RPMI1640-FCS (5) culture medium, and it was set as the target cell solution.
(D) Measurement of cytotoxic activity 50 μL (1 × 10 4 cells / well) of the target cell solution prepared in (c) was dispensed into each well of the 96-well U-bottom plate prepared in (b). At this time, the ratio of effector cells to target cells is 15: 1. Further, KM2760-1 or KM3060 was added so as to have a final concentration of 1 ng / mL or 100 ng / mL to make a total volume of 200 μL, and the mixture was reacted at 37 ° C. for 4 hours. After the reaction, the plate was centrifuged, and the lactate dehydrogenase (LDH) activity in the supernatant was measured using CytoTox96 Non-Radioactive Cytotoxicity Assay (manufactured by Promega) by acquiring absorbance data according to the attached instructions. did. Absorbance data of spontaneous release of target cells uses only medium instead of effector cell solution and antibody solution. Absorbance data of spontaneous release of effector cells uses only medium instead of target cell solution and antibody solution. Obtained by performing the same operation as above. Absorbance data for target cell total release was obtained by using medium instead of antibody solution and effector cell solution, adding 15 μL of 9% Triton X-100 solution 45 minutes before the end of the reaction, and performing the same operation as above. It was determined by measuring the LDH activity of Qing. ADCC activity was determined by the following formula.
Figure 2005053742
The results are shown in FIG. The ADCC activity of KM2760-1 when no cytokine was added was higher than the ADCC activity of KM3060, but was further enhanced by the addition of cytokine. As a result, the ratio of sugar chains in which fucose is not bound to N-acetylglucosamine at the reducing end of the sugar chain out of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition is 50. The high ADCC activity of an antibody composition that is greater than or equal to% indicates that it is further increased by cytokines.

in vitro細胞障害活性における抗CD20抗体とサイトカインの併用効果
in vitro細胞障害活性における抗CD20ヒト型キメラ抗体KM3065(WO03/55993)および市販の抗CD20ヒト型キメラ抗体Rituxan(Genentech社製)とサイトカインの併用効果を実施例1に記載の方法により測定した。
サイトカインはIL−2、IL−15(いずれもPeprotech社製、終濃度1ng/mL)、IFN−α(Peprotech社製、終濃度100ng/mL)、IFN−γ(R&D Systems社製、終濃度100ng/mL)を用いた。KM3065はラットミエローマ細胞株YB2/0細胞に抗CD20キメラ抗体発現株を導入して得られた細胞株KM3065より生産された抗CD20キメラ抗体である。KM3065株はレクチン耐性能を有し、KM3065の全糖鎖中のフコースが結合しない糖鎖含量は96%である。一方Rituxanの全糖鎖中のフコースが結合しない糖鎖含量は6%である。KM3065とRituxanは同じアミノ酸配列を有し、抗原結合活性も同等である。
第2図および第3図に結果を図示した。サイトカイン非添加時のKM3065のADCC活性はRituxanのADCC活性よりも高いが、サイトカインの添加によりさらに増強された。この結果は、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物の高いADCC活性は、サイトカインによってさらに上昇することを示す。
Effect of combined use of anti-CD20 antibody and cytokine on in vitro cytotoxic activity
The combined effect of the anti-CD20 human chimeric antibody KM 3065 (WO03 / 55993) and the commercially available anti-CD20 human chimeric antibody Rituxan (Genentech) and cytokine in the in vitro cytotoxic activity was measured by the method described in Example 1.
Cytokines are IL-2, IL-15 (all manufactured by Peprotech, final concentration 1 ng / mL), IFN-α (manufactured by Peprotech, final concentration 100 ng / mL), IFN-γ (manufactured by R & D Systems, final concentration 100 ng). / ML) was used. KM 3065 is an anti-CD20 chimeric antibody produced from the cell line KM 3065 obtained by introducing an anti-CD20 chimeric antibody expression strain into the rat myeloma cell line YB2 / 0 cells. The KM 3065 strain has lectin resistance, and the sugar chain content to which fucose in all the sugar chains of KM 3065 does not bind is 96%. On the other hand, the sugar chain content in which fucose is not bound in all sugar chains of Rituxan is 6%. KM 3065 and Rituxan have the same amino acid sequence and antigen binding activity is also equivalent.
The results are shown in FIG. 2 and FIG. The ADCC activity of KM 3065 when no cytokine was added was higher than that of Rituxan, but was further enhanced by the addition of cytokine. As a result, the ratio of sugar chains in which fucose is not bound to N-acetylglucosamine at the reducing end of the sugar chain out of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition is 50. The high ADCC activity of an antibody composition that is greater than or equal to% indicates that it is further increased by cytokines.

抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と少なくとも1種類の薬剤とを用いる医薬は、高い治療効果を有することが期待される。  Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more. A medicine using an antibody composition and at least one drug is expected to have a high therapeutic effect.

Claims (14)

抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを組み合わせてなる医薬。Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more. A medicament comprising a combination of an antibody composition and at least one drug. 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを併用して投与するための医薬。Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more. A medicament for administering an antibody composition in combination with at least one drug. 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物と、少なくとも1種類の薬剤とを同時に又は逐次的に投与するための医薬。Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more. A medicament for administering an antibody composition and at least one drug simultaneously or sequentially. 抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖の割合が50%以上である抗体組成物を単独で投与した場合よりも、高い治療効果を示すことを特徴とする、請求項1〜3のいずれか1項に記載の医薬。Of all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition, the proportion of sugar chains in which fucose is not bound to N-acetylglucosamine at the sugar chain reducing end is 50% or more. The medicine according to any one of claims 1 to 3, which exhibits a higher therapeutic effect than when the antibody composition is administered alone. 高い治療効果を示すことが、高い抗体依存性細胞障害活性を示すことである、請求項4記載の医薬。The medicament according to claim 4, wherein the high therapeutic effect is a high antibody-dependent cytotoxic activity. 抗体組成物が、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位とフコースの1位がα結合した糖鎖構造を認識するレクチンに耐性を有する細胞から生産される抗体組成物である請求項1〜5のいずれか1項に記載の医薬。Antibody composition produced from a cell having resistance to a lectin that recognizes a sugar chain structure in which N-acetylglucosamine at the N-glycoside-linked complex type sugar chain reducing terminal at position 6 of N-acetylglucosamine and position 1 of fucose are α-linked The medicine according to any one of claims 1 to 5, which is a product. 抗体組成物が、抗体組成物中に含まれるFc領域に結合する全N−グリコシド結合複合型糖鎖のうち、糖鎖還元末端のN−アセチルグルコサミンにフコースが結合していない糖鎖である抗体組成物である、請求項1〜6のいずれか1項に記載の医薬。An antibody in which the antibody composition is a sugar chain in which fucose is not bound to N-acetylglucosamine at the reducing end of the sugar chain among all N-glycoside-bonded complex sugar chains that bind to the Fc region contained in the antibody composition The pharmaceutical according to any one of claims 1 to 6, which is a composition. フコースが結合していない糖鎖が、該フコースの1位がN−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にα結合していない糖鎖である、請求項1〜7のいずれか1項に記載の医薬。The sugar chain to which fucose is not bonded is a sugar chain in which the 1-position of the fucose is not α-bonded to the 6-position of N-acetylglucosamine at the reducing end of the N-glycoside bond complex type sugar chain. The medicament according to any one of the above. 抗体組成物が、N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素のゲノム遺伝子がノックアウトされた細胞から生産された抗体組成物である、請求項7または8記載の医薬。An antibody composition is produced from a cell in which the genomic gene of an enzyme involved in sugar chain modification in which the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end is knocked out. The medicament according to claim 7 or 8, which is an antibody composition. N−グリコシド結合複合型糖鎖還元末端のN−アセチルグルコサミンの6位にフコースの1位がα結合する糖鎖修飾に関与する酵素が、α1,6−フコシルトランスフェラーゼである請求項9記載の医薬。10. The pharmaceutical according to claim 9, wherein the enzyme involved in the sugar chain modification in which the 1-position of fucose is α-linked to the 6-position of N-acetylglucosamine at the N-glycoside-linked complex sugar chain reducing end is α1,6-fucosyltransferase. . 薬剤が、蛋白質、低分子の薬剤および生物学的応答調節剤からなる群から選ばれる物質である請求項1〜10のいずれか1項に記載の医薬。The medicament according to any one of claims 1 to 10, wherein the drug is a substance selected from the group consisting of a protein, a low-molecular drug, and a biological response modifier. 蛋白質が、サイトカインまたは抗体である、請求項11記載の医薬。The medicament according to claim 11, wherein the protein is a cytokine or an antibody. サイトカインが、IFN−γ、IL−2およびIL−15から選ばれるサイトカインである請求項12に記載の医薬。The medicament according to claim 12, wherein the cytokine is a cytokine selected from IFN-γ, IL-2 and IL-15. 医薬が、腫瘍を伴う疾患に対する治療薬である、請求項1〜13のいずれか1項に記載の医薬。The medicament according to any one of claims 1 to 13, wherein the medicament is a therapeutic agent for a disease involving a tumor.
JP2005516043A 2003-12-04 2004-12-03 Medicament containing antibody composition Pending JPWO2005053742A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2003406589 2003-12-04
JP2003406589 2003-12-04
PCT/JP2004/018441 WO2005053742A1 (en) 2003-12-04 2004-12-03 Medicine containing antibody composition

Publications (1)

Publication Number Publication Date
JPWO2005053742A1 true JPWO2005053742A1 (en) 2007-06-28

Family

ID=34650287

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2005516043A Pending JPWO2005053742A1 (en) 2003-12-04 2004-12-03 Medicament containing antibody composition

Country Status (2)

Country Link
JP (1) JPWO2005053742A1 (en)
WO (1) WO2005053742A1 (en)

Families Citing this family (514)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3528611B2 (en) * 1998-07-16 2004-05-17 Jfeスチール株式会社 Method and apparatus for cutting binding material
SG165344A1 (en) 2004-10-05 2010-10-28 Genentech Inc Method for treating vasculitis
JP2009507040A (en) * 2005-09-02 2009-02-19 グライコフィ, インコーポレイテッド Immunoglobulin containing mainly GLCNACMAN3GLCNAC2 glycoform
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
ES2547689T3 (en) 2005-12-02 2015-10-08 Genentech, Inc. Compositions and methods for the treatment of diseases and disorders associated with cytokine signaling that involve antibodies that bind to IL-22 and IL-22R
EP1973950B1 (en) 2006-01-05 2014-09-17 Genentech, Inc. Anti-ephb4 antibodies and methods using the same
AR059851A1 (en) 2006-03-16 2008-04-30 Genentech Inc ANTIBODIES OF EGFL7 AND METHODS OF USE
CA2652945C (en) 2006-05-30 2015-06-02 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
WO2008063776A2 (en) 2006-10-12 2008-05-29 Genentech, Inc. Antibodies to lymphotoxin-alpha
PT2502938E (en) 2006-10-27 2015-06-05 Genentech Inc Antibodies and immunoconjugates and uses therefor
CA2676766A1 (en) 2007-02-09 2008-08-21 Genentech, Inc. Anti-robo4 antibodies and uses therefor
EP3789400A1 (en) 2007-09-26 2021-03-10 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
LT2514436T (en) 2007-11-07 2018-04-10 Genentech, Inc. Il-22 for use in treating microbial disorders
TWI580694B (en) 2007-11-30 2017-05-01 建南德克公司 Anti-vegf antibodies
KR102100066B1 (en) 2008-10-14 2020-04-10 제넨테크, 인크. Immunoglobulin variants and uses thereof
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
BRPI0918204A2 (en) 2008-12-23 2015-12-08 Genentech Inc igv variant pharmaceutical composition and kit
TW201544123A (en) 2009-03-20 2015-12-01 Genentech Inc Anti-HER antibodies
UA108199C2 (en) 2009-03-25 2015-04-10 ANTIBODY AGAINST α5β1 AND ITS APPLICATION
EP2679600A1 (en) 2009-03-25 2014-01-01 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
TW201106972A (en) 2009-07-27 2011-03-01 Genentech Inc Combination treatments
WO2011014750A1 (en) 2009-07-31 2011-02-03 Genentech, Inc. Inhibition of tumor metastasis using bv8- or g-csf-antagonists
ES2599076T3 (en) 2009-09-02 2017-01-31 Genentech, Inc. Smoothened mutant and methods of use thereof
JP6077745B2 (en) * 2009-09-30 2017-02-08 富士フイルムRiファーマ株式会社 Anti-podoplanin antibody and pharmaceutical composition comprising anti-podoplanin antibody
US8435511B2 (en) 2009-10-22 2013-05-07 Genentech, Inc. Anti-hepsin antibodies and methods using same
WO2011056502A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Bone morphogenetic protein receptor type ii compositions and methods of use
WO2011056497A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor type iib compositions and methods of use
WO2011056494A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor-like kinase-1 antagonist and vegfr3 antagonist combinations
CA2780221A1 (en) 2009-11-04 2011-05-12 Fabrus Llc Methods for affinity maturation-based antibody optimization
KR101968766B1 (en) 2009-11-05 2019-04-12 제넨테크, 인크. Methods and composition for secretion of heterologous polypeptides
EP2509626B1 (en) 2009-12-11 2016-02-10 F.Hoffmann-La Roche Ag Anti-vegf-c antibodies and methods using same
CA2784385A1 (en) 2009-12-23 2011-06-30 Genentech, Inc. Anti-bv8 antibodies and uses thereof
RU2573994C2 (en) 2010-02-10 2016-01-27 Иммьюноджен, Инк Anti-cd20 antibodies and thereof application
US9556249B2 (en) 2010-02-18 2017-01-31 Genentech, Inc. Neuregulin antagonists and use thereof in treating cancer
KR101899835B1 (en) 2010-03-24 2018-09-19 제넨테크, 인크. Anti-lrp6 antibodies
WO2011147834A1 (en) 2010-05-26 2011-12-01 Roche Glycart Ag Antibodies against cd19 and uses thereof
RU2613886C2 (en) 2010-06-03 2017-03-21 Дженентек, Инк. Antibodies and immunoconjugates rendered by immuno-positron emission tomography, methods of application
EP2582729A4 (en) 2010-06-18 2014-05-28 Hoffmann La Roche Anti-axl antibodies and methods of use
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2011161189A1 (en) 2010-06-24 2011-12-29 F. Hoffmann-La Roche Ag Anti-hepsin antibodies and methods of use
EP2591004A1 (en) 2010-07-09 2013-05-15 F.Hoffmann-La Roche Ag Anti-neuropilin antibodies and methods of use
WO2012010582A1 (en) 2010-07-21 2012-01-26 Roche Glycart Ag Anti-cxcr5 antibodies and methods of use
RU2013106216A (en) 2010-08-03 2014-09-10 Ф. Хоффманн-Ля Рош Аг BIOMARKERS OF CHRONIC Lymphocytic Leukemia
KR20130049196A (en) 2010-08-05 2013-05-13 에프. 호프만-라 로슈 아게 Anti-mhc antibody anti-viral cytokine fusion protein
WO2012020038A1 (en) 2010-08-13 2012-02-16 Roche Glycart Ag Anti-tenascin-c a2 antibodies and methods of use
CR20180142A (en) 2010-08-13 2018-04-05 Roche Glycart Ag ANTI-FAP ANTIBODIES AND METHODS OF USE (Divisional Exp: 2013-0038)
CN103080132B (en) 2010-08-25 2016-06-08 弗·哈夫曼-拉罗切有限公司 The antibody of anti-IL-18R1 and purposes thereof
DK2612151T3 (en) 2010-08-31 2017-10-02 Genentech Inc BIOMARKETS AND METHODS OF TREATMENT
DK2625197T3 (en) 2010-10-05 2016-10-03 Genentech Inc Smoothened MUTANT AND METHODS OF USING THE SAME
EP2638070B1 (en) 2010-11-10 2016-10-19 F.Hoffmann-La Roche Ag Methods and compositions for neural disease immunotherapy
AR084342A1 (en) 2010-12-16 2013-05-08 Genentech Inc DIAGNOSIS AND TREATMENTS RELATED TO THE INHIBITION OF TH2
JP6253987B2 (en) 2010-12-20 2017-12-27 ジェネンテック, インコーポレイテッド Anti-mesothelin antibodies and immunoconjugates
US20120195910A1 (en) 2010-12-22 2012-08-02 Genentech, Inc. Anti-pcsk9 antibodies and methods of use
WO2012092539A2 (en) 2010-12-31 2012-07-05 Takeda Pharmaceutical Company Limited Antibodies to dll4 and uses thereof
EP2681240B1 (en) 2011-02-28 2017-08-16 F. Hoffmann-La Roche AG Monovalent antigen binding proteins
BR112013019975A2 (en) 2011-02-28 2017-08-01 Hoffmann La Roche "Antigen binding proteins, pharmaceutical composition, use of an antigen binding protein, method for treating a patient and method for preparing an antigen binding protein, nucleic acid, vector and host cell"
MX342240B (en) 2011-04-07 2016-09-21 Genentech Inc Anti-fgfr4 antibodies and methods of use.
WO2012146630A1 (en) 2011-04-29 2012-11-01 F. Hoffmann-La Roche Ag N-terminal acylated polypeptides, methods for their production and uses thereof
JP5987053B2 (en) 2011-05-12 2016-09-06 ジェネンテック, インコーポレイテッド Multiple reaction monitoring LC-MS / MS method for detecting therapeutic antibodies in animal samples using framework signature peptides
MX370233B (en) 2011-05-16 2019-12-06 Genentech Inc Fgfr1 agonists and methods of use.
BR112013032235A2 (en) 2011-06-15 2016-11-22 Hoffmann La Roche anti-human epo receptor antibodies and methods of use
BR112013029746B1 (en) 2011-06-22 2021-02-02 F. Hoffmann-La Roche Ag method for recombinant production of a complex, complex, pharmaceutical formulation and use of the complex
KR20140045440A (en) 2011-06-30 2014-04-16 제넨테크, 인크. Anti-c-met antibody formulations
KR20140068877A (en) 2011-08-17 2014-06-09 제넨테크, 인크. Inhibition of angiogenesis in refractory tumors
CN103890007A (en) 2011-08-17 2014-06-25 霍夫曼-拉罗奇有限公司 Neuregulin antibodies and uses thereof
RU2014109038A (en) 2011-08-23 2015-09-27 Рош Гликарт Аг ANTIBODIES TO CHONDROITINSULFATE PROTEOGLYCAN MELANOMA
MX2014003094A (en) 2011-09-15 2014-04-25 Genentech Inc Methods of promoting differentiation.
BR112014006419A2 (en) 2011-09-19 2018-08-07 Genentech Inc Methods to Treat a Cancer Patient, Kit and Article
AU2012319150B2 (en) 2011-10-05 2017-08-17 Genentech, Inc. Methods of treating liver conditions using Notch2 antagonists
PT2766393T (en) 2011-10-14 2018-10-15 Hoffmann La Roche Anti-htra1 antibodies and methods of use
RU2014119426A (en) 2011-10-15 2015-11-20 Дженентек, Инк. WAYS OF APPLICATION OF SCD1 ANTAGONISTS
WO2013059531A1 (en) 2011-10-20 2013-04-25 Genentech, Inc. Anti-gcgr antibodies and uses thereof
AR088509A1 (en) 2011-10-28 2014-06-11 Genentech Inc THERAPEUTIC COMBINATIONS AND METHODS TO TREAT MELANOMA
MX2014005885A (en) 2011-11-21 2014-09-04 Genentech Inc Purification of anti-c-met antibodies.
US20140335084A1 (en) 2011-12-06 2014-11-13 Hoffmann-La Roche Inc. Antibody formulation
WO2013092720A1 (en) 2011-12-22 2013-06-27 F. Hoffmann-La Roche Ag Full length antibody display system for eukaryotic cells and its use
SG10201700169PA (en) 2011-12-22 2017-02-27 Hoffmann La Roche Expression vector element combinations, novel production cell generation methods and their use for the recombinant production of polypeptides
SG11201403223PA (en) 2011-12-22 2014-07-30 Hoffmann La Roche Expression vector organization, novel production cell generation methods and their use for the recombinant production of polypeptides
AR089434A1 (en) 2011-12-23 2014-08-20 Genentech Inc PROCEDURE TO PREPARE FORMULATIONS WITH HIGH CONCENTRATION OF PROTEINS
US10774132B2 (en) 2012-01-09 2020-09-15 The Scripps Research Instittue Ultralong complementarity determining regions and uses thereof
JP2015509091A (en) 2012-01-09 2015-03-26 ザ スクリプス リサーチ インスティテュート Humanized antibody
CN104066449B (en) 2012-01-18 2018-04-27 霍夫曼-拉罗奇有限公司 Anti- LRP5 antibody and application method
CA2862424A1 (en) 2012-01-18 2013-07-25 Genentech, Inc. Methods of using fgf19 modulators
TW201334789A (en) 2012-01-31 2013-09-01 Genentech Inc Anti-IgE antibodies and methods using same
RU2014133069A (en) 2012-02-11 2016-04-10 Дженентек, Инк. R-SPONDIN TRANSLOCATIONS AND WAYS WITH THEIR USE
JP6152120B2 (en) 2012-02-15 2017-06-21 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Affinity chromatography based on Fc receptors
RU2014136886A (en) 2012-03-27 2016-05-20 Дженентек, Инк. DIAGNOSTIC AND TREATMENT TYPES RELATED TO HER3 INHIBITORS
AR090549A1 (en) 2012-03-30 2014-11-19 Genentech Inc ANTI-LGR5 AND IMMUNOCATE PLAYERS
BR112014027166A2 (en) 2012-05-01 2017-06-27 Genentech Inc antibody, nucleic acid, host cell, method for producing an antibody, immunoconjugate, pharmaceutical formulation, treatment method, proliferation inhibiting method and detection methods.
WO2013170191A1 (en) 2012-05-11 2013-11-14 Genentech, Inc. Methods of using antagonists of nad biosynthesis from nicotinamide
RU2625771C2 (en) 2012-05-23 2017-07-18 Дженентек, Инк. Therapeutics selection method
MX2014014830A (en) 2012-06-15 2015-05-11 Genentech Inc Anti-pcsk9 antibodies, formulations, dosing, and methods of use.
BR112014030843A2 (en) 2012-07-04 2019-10-15 Hoffmann La Roche anti-theophylline antibody, pharmaceutical formulation and use of the antibody
CA2872192A1 (en) 2012-07-04 2014-01-09 F. Hoffmann-La Roche Ag Anti-biotin antibodies and methods of use
CN107973856B (en) 2012-07-04 2021-11-23 弗·哈夫曼-拉罗切有限公司 Covalently linked antigen-antibody conjugates
CN110042114A (en) 2012-07-05 2019-07-23 弗·哈夫曼-拉罗切有限公司 Expression and excretory system
IN2014DN10652A (en) 2012-07-09 2015-09-11 Genentech Inc
BR112015000441A2 (en) 2012-07-09 2017-12-19 Genentech Inc Immunoconjugates, Pharmaceutical Formulation and Treatment Method and Method for Inhibiting Proliferation of a CD22 Positive Cell
CN104411337A (en) 2012-07-09 2015-03-11 基因泰克公司 Immunoconjugates comprising anti-cd79b antibodies
EA201590173A1 (en) 2012-07-09 2015-09-30 Дженентек, Инк. IMMUNOCONGUTS CONTAINING ANTIBODIES TO CD22
EA201590237A1 (en) * 2012-07-18 2015-05-29 Гликотоп Гмбх NEW METHODS OF ANTIBODY TREATMENT AGAINST HER2 WITH LOW FUZOZYLATION
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
US9725512B2 (en) 2012-11-08 2017-08-08 Hoffmann-La Roche Inc. HER3 antibodies binding to the beta-hairpin of HER3
MA38176A1 (en) 2012-11-13 2017-06-30 Genentech Inc Novel anti-haemagglutinin antibody, useful for the treatment, inhibition or prevention of viral influenza infection
AU2013360775B2 (en) 2012-12-21 2018-04-12 F. Hoffmann-La Roche Ag Disulfide-linked multivalent MHC class I comprising multi-function proteins
US10980804B2 (en) 2013-01-18 2021-04-20 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
WO2014116749A1 (en) 2013-01-23 2014-07-31 Genentech, Inc. Anti-hcv antibodies and methods of using thereof
CN104994879A (en) 2013-02-22 2015-10-21 霍夫曼-拉罗奇有限公司 Methods of treating cancer and preventing drug resistance
RU2015140921A (en) 2013-02-26 2017-04-03 Роше Гликарт Аг ANTIBODIES TO MCSP
JP2016510751A (en) 2013-03-06 2016-04-11 ジェネンテック, インコーポレイテッド Methods of treating and preventing anticancer drug resistance
CA2903480A1 (en) 2013-03-14 2014-09-25 Genentech, Inc. Combinations of a mek inhibitor compound with an her3/egfr inhibitor compound and methods of use
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
BR112015022604A2 (en) 2013-03-14 2017-10-24 Genentech Inc Uses of a Chromatin Modifier Modulator and an Egfr Antagonist
MX2015010777A (en) 2013-03-14 2016-04-25 Genentech Inc Anti-b7-h4 antibodies and immunoconjugates.
SG11201507427QA (en) 2013-03-15 2015-10-29 Genentech Inc Compositions and methods for diagnosis and treatment of hepatic cancers
BR112015023262B8 (en) 2013-03-15 2024-02-06 Ac Immune Sa Isolated antibody, immunoconjugate, pharmaceutical formulation and uses of antibody
KR102158924B1 (en) 2013-03-15 2020-09-22 제넨테크, 인크. Biomarkers and methods of treating pd-1 and pd-l1 related conditions
CN105143265A (en) 2013-03-15 2015-12-09 豪夫迈·罗氏有限公司 Anti-crth2 antibodies and their use
EP2968537A1 (en) 2013-03-15 2016-01-20 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
UA118843C2 (en) 2013-03-15 2019-03-25 Дженентек, Інк. Il-22 polypeptides and il-22 fc fusion proteins and methods of use
UA118028C2 (en) 2013-04-03 2018-11-12 Рош Глікарт Аг Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use
WO2014177459A2 (en) 2013-04-29 2014-11-06 F. Hoffmann-La Roche Ag Fc-receptor binding modified asymmetric antibodies and methods of use
KR102282134B1 (en) 2013-04-29 2021-07-27 에프. 호프만-라 로슈 아게 Human fcrn-binding modified antibodies and methods of use
SG10201810481UA (en) 2013-04-29 2018-12-28 Hoffmann La Roche Fcrn-binding abolished anti-igf-1r antibodies and their use in the treatment of vascular eye diseases
EP2999716A2 (en) 2013-05-20 2016-03-30 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
JP6469094B2 (en) 2013-05-31 2019-02-13 ジェネンテック, インコーポレイテッド Anti-cell wall tycoic acid antibodies and conjugates
MX369758B (en) 2013-05-31 2019-11-20 Genentech Inc Anti-wall teichoic antibodies and conjugates.
WO2015017146A2 (en) 2013-07-18 2015-02-05 Fabrus, Inc. Antibodies with ultralong complementarity determining regions
JP6687520B2 (en) 2013-07-18 2020-04-22 トーラス バイオサイエンシズ リミテッド ライアビリティ カンパニー Humanized antibody with extremely long complementarity determining regions
ME03476B (en) 2013-08-01 2020-01-20 Five Prime Therapeutics Inc Afucosylated anti-fgfr2iiib antibodies
CN105518027A (en) 2013-09-17 2016-04-20 豪夫迈·罗氏有限公司 Methods of using anti-LGR5 antibodies
JP6502931B2 (en) 2013-10-11 2019-04-17 アメリカ合衆国 TEM 8 antibody and use thereof
WO2015052230A1 (en) 2013-10-11 2015-04-16 F. Hoffmann-La Roche Ag Multispecific domain exchanged common variable light chain antibodies
WO2015054670A1 (en) 2013-10-11 2015-04-16 Genentech, Inc. Nsp4 inhibitors and methods of use
KR20160070136A (en) 2013-10-18 2016-06-17 제넨테크, 인크. Anti-rsp02 and/or anti-rsp03 antibodies and their uses
RU2016119425A (en) 2013-10-23 2017-11-28 Дженентек, Инк. METHODS FOR DIAGNOSIS AND TREATMENT OF EOSINOPHILIC DISEASES
EP3071597B1 (en) 2013-11-21 2020-07-29 F.Hoffmann-La Roche Ag Anti-alpha-synuclein antibodies and methods of use
LT3079719T (en) 2013-12-09 2019-12-10 Allakos Inc Anti-siglec-8 antibodies and methods of use thereof
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
MX2016007958A (en) 2013-12-17 2016-08-03 Genentech Inc Anti-cd3 antibodies and methods of use.
CN105899535A (en) 2013-12-17 2016-08-24 豪夫迈·罗氏有限公司 Methods of treating cancer using pd-1 axis binding antagonists and an anti-cd20 antibody
CA2934028A1 (en) 2013-12-17 2015-06-25 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
NZ720515A (en) 2013-12-17 2022-12-23 Genentech Inc Methods of treating cancers using pd-1 axis binding antagonists and taxanes
TWI670283B (en) 2013-12-23 2019-09-01 美商建南德克公司 Antibodies and methods of use
CN105873616B (en) 2014-01-03 2020-06-05 豪夫迈·罗氏有限公司 Covalently linked polypeptide toxin-antibody conjugates
CN105873615B (en) 2014-01-03 2020-12-25 豪夫迈·罗氏有限公司 Covalently linked helicar-anti-helicar antibody conjugates and uses thereof
WO2015101586A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
WO2015103549A1 (en) 2014-01-03 2015-07-09 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Neutralizing antibodies to hiv-1 env and their use
PL3092251T3 (en) 2014-01-06 2021-08-02 F. Hoffmann-La Roche Ag Monovalent blood brain barrier shuttle modules
CA2931986A1 (en) 2014-01-15 2015-07-23 F. Hoffmann-La Roche Ag Fc-region variants with modified fcrn- and maintained protein a-binding properties
BR112016015693A2 (en) 2014-01-24 2017-10-24 Genentech Inc Method To Treat Prostate Cancer And Antibody
DK3102197T3 (en) 2014-02-04 2018-11-19 Genentech Inc Smoothened mutant and methods for its use
PL3102230T3 (en) 2014-02-08 2021-11-15 F.Hoffmann-La Roche Ag Methods of treating alzheimer's disease
AU2015214058B2 (en) 2014-02-08 2020-07-09 Genentech, Inc. Methods of treating Alzheimer's Disease
AR099465A1 (en) 2014-02-12 2016-07-27 Genentech Inc ANTI-JAGGED1 ANTIBODIES AND METHODS OF USE
MX2016010729A (en) 2014-02-21 2016-10-26 Genentech Inc Anti-il-13/il-17 bispecific antibodies and uses thereof.
EP4014995A1 (en) 2014-02-28 2022-06-22 Allakos Inc. Methods and compositions for treating siglec-8 associated diseases
PL3116999T3 (en) 2014-03-14 2021-12-27 F.Hoffmann-La Roche Ag Methods and compositions for secretion of heterologous polypeptides
US20170107294A1 (en) 2014-03-21 2017-04-20 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
MA39776A (en) 2014-03-24 2017-02-01 Hoffmann La Roche Cancer treatment with c-met antagonists and correlation of the latter with hgf expression
RU2016142476A (en) 2014-03-31 2018-05-07 Дженентек, Инк. COMBINED THERAPY, INCLUDING ANTI-ANGIOGENESIS AGENTS AND AGONISTS BINDING OX40
MA51552A (en) 2014-03-31 2021-03-24 Hoffmann La Roche ANTI-OX40 ANTIBODIES AND METHODS OF USE
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
RU2016150370A (en) 2014-05-22 2018-06-26 Дженентек, Инк. Antibodies and immunoconjugates against GPC3
KR20170005016A (en) 2014-05-23 2017-01-11 제넨테크, 인크. Mit biomarkers and methods using the same
JP2017518989A (en) * 2014-05-27 2017-07-13 アカデミア シニカAcademia Sinica Anti-CD20 glycoengineered antibody group and use thereof
CA2949982A1 (en) 2014-06-11 2015-12-17 Genentech, Inc. Anti-lgr5 antibodies and uses thereof
EP3154589A1 (en) 2014-06-13 2017-04-19 Genentech, Inc. Methods of treating and preventing cancer drug resistance
CA2947504A1 (en) 2014-06-26 2015-12-30 F. Hoffmann-La Roche Ag Anti-brdu antibodies and methods of use
JP7032929B2 (en) 2014-07-11 2022-03-09 ヴェンタナ メディカル システムズ, インク. Anti-PD-L1 antibody and its diagnostic use
CN106488775A (en) 2014-07-11 2017-03-08 基因泰克公司 NOTCH approach suppresses
JP7286267B2 (en) 2014-08-28 2023-06-05 バイオアトラ インコーポレイテッド Conditionally active chimeric antigen receptor for modified T cells
TWI751102B (en) 2014-08-28 2022-01-01 美商奇諾治療有限公司 Antibodies and chimeric antigen receptors specific for cd19
TW201625689A (en) 2014-09-12 2016-07-16 建南德克公司 Anti-B7-H4 antibodies and immunoconjugates
JP7085837B2 (en) 2014-09-12 2022-06-17 ジェネンテック, インコーポレイテッド Anti-HER2 antibody and immune complex
PE20170670A1 (en) 2014-09-12 2017-06-06 Genentech Inc ANTI-CLL-1 ANTIBODIES AND IMMUNOCONJUGATES
KR20170055521A (en) 2014-09-17 2017-05-19 제넨테크, 인크. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
LT3262071T (en) 2014-09-23 2020-06-25 F. Hoffmann-La Roche Ag Method of using anti-cd79b immunoconjugates
US9732148B2 (en) 2014-10-16 2017-08-15 Genentech, Inc. Anti-α-synuclein antibodies and methods of use
US10626176B2 (en) 2014-10-31 2020-04-21 Jounce Therapeutics, Inc. Methods of treating conditions with antibodies that bind B7-H4
CN106796235B (en) 2014-11-03 2021-01-29 豪夫迈·罗氏有限公司 Assays for detecting T cell immune subsets and methods of use thereof
CN107109484B (en) 2014-11-03 2021-12-14 豪夫迈·罗氏有限公司 Methods and biomarkers for efficacy prediction and assessment of OX40 agonist treatment
RU2017119185A (en) 2014-11-05 2018-12-05 Дженентек, Инк. ANTIBODIES AGAINST FGFR2 / 3 AND WAYS OF THEIR APPLICATION
JP6576456B2 (en) 2014-11-06 2019-09-18 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Fc region variants with modified FcRn binding properties and protein A binding properties
CN107073126A (en) 2014-11-06 2017-08-18 豪夫迈·罗氏有限公司 Combination treatment comprising OX40 combinations activator and TIGIT inhibitor
WO2016071376A2 (en) 2014-11-06 2016-05-12 F. Hoffmann-La Roche Ag Fc-region variants with modified fcrn-binding and methods of use
EP3218403B1 (en) 2014-11-10 2020-05-13 F.Hoffmann-La Roche Ag Anti-interleukin-33 antibodies and uses thereof
EP3552488A1 (en) 2014-11-10 2019-10-16 F. Hoffmann-La Roche AG Animal model for nephropathy and agents for treating the same
EP3218397B8 (en) 2014-11-14 2021-04-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to ebola virus glycoprotein and their use
BR112017010198A2 (en) 2014-11-17 2017-12-26 Genentech Inc combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
US11008403B2 (en) 2014-11-19 2021-05-18 Genentech, Inc. Anti-transferrin receptor / anti-BACE1 multispecific antibodies and methods of use
CN107108745B (en) 2014-11-19 2021-01-12 基因泰克公司 Antibodies against BACE1 and their use for immunotherapy of neurological diseases
US10508151B2 (en) 2014-11-19 2019-12-17 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
SG11201704056XA (en) 2014-11-20 2017-06-29 Hoffmann La Roche Combination therapy of t cell activating bispecific antigen binding molecules cd3 abd folate receptor 1 (folr1) and pd-1 axis binding antagonists
BR112017011478A2 (en) 2014-12-03 2018-02-27 Genentech, Inc. antibody-antibiotic conjugate compound, antibody-antibiotic conjugate, pharmaceutical composition, methods of treating a bacterial infection and exterminating staph aureus, process for compound preparation, kit and antibiotic-binder intermediate
EP3226911A1 (en) 2014-12-03 2017-10-11 F.Hoffmann-La Roche Ag Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof
PL3227332T3 (en) 2014-12-03 2020-06-15 F. Hoffmann-La Roche Ag Multispecific antibodies
US9975949B2 (en) 2014-12-05 2018-05-22 Genentech, Inc. Anti-CD79b antibodies and methods of use
WO2016094566A2 (en) 2014-12-10 2016-06-16 Genentech, Inc. Blood brain barrier receptor antibodies and methods of use
PT3233921T (en) 2014-12-19 2021-12-09 Chugai Pharmaceutical Co Ltd Anti-c5 antibodies and methods of use
WO2016111947A2 (en) 2015-01-05 2016-07-14 Jounce Therapeutics, Inc. Antibodies that inhibit tim-3:lilrb2 interactions and uses thereof
CN107428833B (en) 2015-01-16 2021-11-02 朱诺治疗学股份有限公司 ROR 1-specific antibodies and chimeric antigen receptors
CN113956354A (en) 2015-01-22 2022-01-21 中外制药株式会社 Combinations and methods of use of two or more anti-C5 antibodies
JP2018512597A (en) 2015-02-04 2018-05-17 ジェネンテック, インコーポレイテッド Mutant smoothened and method of using the same
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
WO2016138160A1 (en) 2015-02-24 2016-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Middle east respiratory syndrome coronavirus immunogens, antibodies, and their use
CN107430117A (en) 2015-03-16 2017-12-01 豪夫迈·罗氏有限公司 Detection and quantitative IL 13 method and the purposes in diagnosing and treating Th2 relevant diseases
WO2016146833A1 (en) 2015-03-19 2016-09-22 F. Hoffmann-La Roche Ag Biomarkers for nad(+)-diphthamide adp ribosyltransferase resistance
WO2016154003A1 (en) 2015-03-20 2016-09-29 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Neutralizing antibodies to gp120 and their use
RU2742241C2 (en) 2015-03-23 2021-02-04 Джоунс Терапьютикс, Инк. Antibodies to icos
WO2016153572A1 (en) 2015-03-25 2016-09-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Bispecific multivalent fusion proteins
EP3277314A4 (en) 2015-04-03 2018-08-29 Eureka Therapeutics, Inc. Constructs targeting afp peptide/mhc complexes and uses thereof
SI3286315T1 (en) 2015-04-24 2021-09-30 F. Hoffmann-La Roche Ag Methods of identifying bacteria comprising binding polypeptides
WO2016179003A1 (en) 2015-05-01 2016-11-10 Genentech, Inc. Masked anti-cd3 antibodies and methods of use
WO2016179194A1 (en) 2015-05-04 2016-11-10 Jounce Therapeutics, Inc. Lilra3 and method of using the same
JP6963508B2 (en) 2015-05-11 2021-11-10 ジェネンテック, インコーポレイテッド Compositions and Methods for Treating Lupus Nephritis
JP7048319B2 (en) 2015-05-12 2022-04-05 ジェネンテック, インコーポレイテッド Treatment and diagnosis methods for cancer
CN107771182A (en) 2015-05-29 2018-03-06 豪夫迈·罗氏有限公司 The anti-Ebola virus glycoproteins antibody of humanization and application method
JP7144935B2 (en) 2015-05-29 2022-09-30 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods for cancer
CN107810012A (en) 2015-06-02 2018-03-16 豪夫迈·罗氏有限公司 Use the composition and method of the anti-Antybody therapy sacred diseases of IL 34
WO2016196975A1 (en) 2015-06-03 2016-12-08 The United States Of America, As Represented By The Secretary Department Of Health & Human Services Neutralizing antibodies to hiv-1 env and their use
US10112990B2 (en) 2015-06-05 2018-10-30 Genentech, Inc. Anti-Tau antibodies and methods of use
MX2017015937A (en) 2015-06-08 2018-12-11 Genentech Inc Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists.
MX2017014740A (en) 2015-06-08 2018-08-15 Genentech Inc Methods of treating cancer using anti-ox40 antibodies.
EP3307780A1 (en) 2015-06-15 2018-04-18 Genentech, Inc. Antibodies and immunoconjugates
JP2018526972A (en) 2015-06-16 2018-09-20 ジェネンテック, インコーポレイテッド Anti-CD3 antibody and method of use
CR20180031A (en) 2015-06-16 2018-05-07 Genentech Inc ANTIBODIES MATURED BY AFFINITY AND HUMANIZED FOR FCRH5 AND METHODS FOR USE
JP6996983B2 (en) 2015-06-16 2022-02-21 ジェネンテック, インコーポレイテッド Anti-CLL-1 antibody and how to use
WO2016205531A2 (en) 2015-06-17 2016-12-22 Genentech, Inc. Anti-her2 antibodies and methods of use
CA2987797A1 (en) 2015-06-17 2016-12-22 Christopher Robert Bebbington Methods and compositions for treating fibrotic diseases
EP3310815A1 (en) 2015-06-17 2018-04-25 F. Hoffmann-La Roche AG Methods of treating locally advanced or metastatic breast cancers using pd-1 axis binding antagonists and taxanes
LT3313879T (en) 2015-06-24 2022-03-25 F. Hoffmann-La Roche Ag Anti-transferrin receptor antibodies with tailored affinity
CA2989936A1 (en) 2015-06-29 2017-01-05 Genentech, Inc. Type ii anti-cd20 antibody for use in organ transplantation
WO2017001350A1 (en) 2015-06-29 2017-01-05 Ventana Medical Systems, Inc. Materials and methods for performing histochemical assays for human pro-epiregulin and amphiregulin
CN105384825B (en) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 A kind of bispecific chimeric antigen receptor and its application based on single domain antibody
EP3932953A1 (en) 2015-08-28 2022-01-05 F. Hoffmann-La Roche AG Anti-hypusine antibodies and uses thereof
JP6904947B2 (en) 2015-09-22 2021-07-21 スプリング バイオサイエンス コーポレーション Anti-OX40 antibody and its diagnostic applications
CN116987187A (en) 2015-09-23 2023-11-03 豪夫迈·罗氏有限公司 Optimized variants of anti-VEGF antibodies
BR112018005931A2 (en) 2015-09-24 2018-10-09 Abvitro Llc hiv antibody compositions and methods of use
MA43345A (en) 2015-10-02 2018-08-08 Hoffmann La Roche PYRROLOBENZODIAZEPINE ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
NZ741067A (en) 2015-10-02 2023-07-28 Hoffmann La Roche Bispecific anti-human cd20/human transferrin receptor antibodies and methods of use
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
EP3150636A1 (en) 2015-10-02 2017-04-05 F. Hoffmann-La Roche AG Tetravalent multispecific antibodies
CA2997809A1 (en) 2015-10-07 2017-04-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Il-7r-alpha specific antibodies for treating acute lymphoblastic leukemia
CA2998208A1 (en) 2015-10-22 2017-04-27 Jounce Therapeutics, Inc. Gene signatures for determining icos expression
US10604577B2 (en) 2015-10-22 2020-03-31 Allakos Inc. Methods and compositions for treating systemic mastocytosis
WO2017072210A1 (en) 2015-10-29 2017-05-04 F. Hoffmann-La Roche Ag Anti-variant fc-region antibodies and methods of use
EP3184547A1 (en) 2015-10-29 2017-06-28 F. Hoffmann-La Roche AG Anti-tpbg antibodies and methods of use
WO2017075173A2 (en) 2015-10-30 2017-05-04 Genentech, Inc. Anti-factor d antibodies and conjugates
MX2018004509A (en) 2015-10-30 2018-08-01 Genentech Inc Anti-htra1 antibodies and methods of use thereof.
WO2017079479A1 (en) 2015-11-03 2017-05-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Neutralizing antibodies to hiv-1 gp41 and their use
WO2017079768A1 (en) 2015-11-08 2017-05-11 Genentech, Inc. Methods of screening for multispecific antibodies
WO2017091580A1 (en) 2015-11-23 2017-06-01 Five Prime Therapeutics, Inc. Predicting response to cancer treatment with fgfr2 inhibitors
CA3005592C (en) 2015-12-18 2024-01-23 Chugai Seiyaku Kabushiki Kaisha Anti-c5 antibodies and methods of use
ES2837428T3 (en) 2016-01-08 2021-06-30 Hoffmann La Roche CEA-Positive Cancer Treatment Procedures Using PD-1 Axis Binding Antagonists and Anti-CEA / Anti-CD3 Bispecific Antibodies
CN114113625A (en) 2016-01-20 2022-03-01 基因泰克公司 High dose treatment for alzheimer's disease
JP2019509721A (en) 2016-02-04 2019-04-11 キュリス,インコーポレイテッド Mutant smoothened and method of using the same
JP6821693B2 (en) 2016-02-29 2021-01-27 ジェネンテック, インコーポレイテッド Treatment and diagnosis for cancer
ES2904286T3 (en) 2016-03-15 2022-04-04 Chugai Pharmaceutical Co Ltd Cancer Treatment Methods Using PD-1 Axis Binding Antagonists and Anti-GPC3 Antibodies
CN108700598A (en) 2016-03-25 2018-10-23 豪夫迈·罗氏有限公司 The drug of the total antibody of multichannel and antibody conjugate quantifies measuring method
US20170319688A1 (en) 2016-04-14 2017-11-09 Genentech, Inc. Anti-rspo3 antibodies and methods of use
WO2017181079A2 (en) 2016-04-15 2017-10-19 Genentech, Inc. Methods for monitoring and treating cancer
KR20190003958A (en) 2016-04-15 2019-01-10 제넨테크, 인크. Treatment and monitoring of cancer
SG11201808994YA (en) 2016-04-15 2018-11-29 Bioatla Llc Anti-axl antibodies, antibody fragments and their immunoconjugates and uses thereof
WO2017192589A1 (en) 2016-05-02 2017-11-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to influenza ha and their use and identification
PE20181890A1 (en) 2016-05-02 2018-12-11 Hoffmann La Roche CONTORSBODY - A MONOCATENARIO DIANA LEAGUE
CN109071640B (en) 2016-05-11 2022-10-18 豪夫迈·罗氏有限公司 Modified anti-tenascin antibodies and methods of use
ES2930255T3 (en) 2016-05-13 2022-12-09 Bioatla Inc Anti-Ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
JP2019522633A (en) 2016-05-20 2019-08-15 ジェネンテック, インコーポレイテッド PROTAC antibody conjugates and methods of use
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
JP7133477B2 (en) 2016-06-24 2022-09-08 ジェネンテック, インコーポレイテッド Anti-polyubiquitin multispecific antibody
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
MX2018015721A (en) 2016-07-29 2019-05-27 Chugai Pharmaceutical Co Ltd Bispecific antibody exhibiting increased alternative fviii-cofactor-function activity.
US20240018268A1 (en) 2016-07-29 2024-01-18 Juno Therapeutics, Inc. Anti-idiotypic antibodies against anti-cd19 antibodies
SG11201801024XA (en) 2016-08-05 2018-05-30 Chugai Pharmaceutical Co Ltd Therapeutic or preventive compositions for il-8-related diseases
CN109476748B (en) 2016-08-08 2023-05-23 豪夫迈·罗氏有限公司 Methods for treatment and diagnosis of cancer
US10870694B2 (en) 2016-09-02 2020-12-22 Dana Farber Cancer Institute, Inc. Composition and methods of treating B cell disorders
WO2018049083A1 (en) 2016-09-07 2018-03-15 The Regents Of The University Of California Antibodies to oxidation-specific epitopes
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
EP3515932B1 (en) 2016-09-19 2023-11-22 F. Hoffmann-La Roche AG Complement factor based affinity chromatography
EP4268845A3 (en) 2016-09-23 2024-02-28 F. Hoffmann-La Roche AG Uses of il-13 antagonists for treating atopic dermatitis
US11072660B2 (en) 2016-10-03 2021-07-27 Juno Therapeutics, Inc. HPV-specific binding molecules
JP7050770B2 (en) 2016-10-05 2022-04-08 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Method for preparing antibody drug conjugate
EP3523451A1 (en) 2016-10-06 2019-08-14 Genentech, Inc. Therapeutic and diagnostic methods for cancer
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
JP2019535250A (en) 2016-10-29 2019-12-12 ジェネンテック, インコーポレイテッド Anti-MIC antibody and method of use
HUE057559T2 (en) 2016-11-02 2022-06-28 Jounce Therapeutics Inc Antibodies to pd-1 and uses thereof
EP3541843A1 (en) 2016-11-15 2019-09-25 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
CN110023334B (en) 2016-11-21 2023-11-14 科雅博有限责任公司 anti-GP 73 antibodies and immunoconjugates
KR20230146126A (en) 2016-12-07 2023-10-18 제넨테크, 인크. Anti-tau antibodies and methods of use
CN117820467A (en) 2016-12-07 2024-04-05 基因泰克公司 anti-TAU antibodies and methods of use
AU2017375946A1 (en) 2016-12-12 2019-06-20 Genentech, Inc. Methods of treating cancer using anti-PD-l1 antibodies and antiandrogens
JOP20190134A1 (en) 2016-12-23 2019-06-02 Potenza Therapeutics Inc Anti-neuropilin antigen-binding proteins and methods of use thereof
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
US11274157B2 (en) 2017-01-12 2022-03-15 Eureka Therapeutics, Inc. Constructs targeting histone H3 peptide/MHC complexes and uses thereof
EP3580235A1 (en) 2017-02-10 2019-12-18 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use
MY197534A (en) 2017-02-10 2023-06-21 Genentech Inc Anti-tryptase antibodies, compositions thereof, and uses thereof
MX2019010295A (en) 2017-03-01 2019-11-21 Genentech Inc Diagnostic and therapeutic methods for cancer.
EP3600442A1 (en) 2017-03-22 2020-02-05 Genentech, Inc. Optimized antibody compositions for treatment of ocular disorders
SG10201911225WA (en) 2017-03-28 2020-01-30 Genentech Inc Methods of treating neurodegenerative diseases
JOP20190203A1 (en) 2017-03-30 2019-09-03 Potenza Therapeutics Inc Anti-tigit antigen-binding proteins and methods of use thereof
CN110621787A (en) 2017-04-14 2019-12-27 豪夫迈·罗氏有限公司 Methods for diagnosis and treatment of cancer
SG11201909048TA (en) 2017-04-21 2019-11-28 Genentech Inc Use of klk5 antagonists for treatment of a disease
KR20200012860A (en) 2017-04-26 2020-02-05 유레카 쎄라퓨틱스, 인코포레이티드 Constructs that specifically recognize glypican 3 and uses thereof
CN110799541A (en) 2017-04-27 2020-02-14 特沙诺有限公司 Antibody agents against lymphocyte activation gene-3 (LAG-3) and uses thereof
CN110831628A (en) 2017-05-05 2020-02-21 爱乐科斯公司 Methods and compositions for treating allergic eye diseases
WO2018209125A1 (en) 2017-05-10 2018-11-15 Fred Hutchinson Cancer Research Center Epstein barr virus antibodies, vaccines, and uses of the same
CN110621336A (en) 2017-05-16 2019-12-27 戊瑞治疗有限公司 Combination of an anti-FGFR 2 antibody with a chemotherapeutic agent in the treatment of cancer
AU2018304458B2 (en) 2017-07-21 2021-12-09 Foundation Medicine, Inc. Therapeutic and diagnostic methods for cancer
ES2963157T3 (en) 2017-07-26 2024-03-25 Forty Seven Inc Anti-SIRP-alpha antibodies and related methods
JP7382922B2 (en) 2017-09-20 2023-11-17 中外製薬株式会社 Dosing regimen for combination therapy using PD-1 system binding antagonists and GPC3 targeting agents
EP3692063A1 (en) 2017-10-03 2020-08-12 Juno Therapeutics, Inc. Hpv-specific binding molecules
CA3078974A1 (en) 2017-10-12 2019-04-18 Immunowake Inc. Vegfr-antibody light chain fusion protein
JP7256197B2 (en) 2017-11-01 2023-04-11 ジュノー セラピューティクス インコーポレイテッド Antibodies and chimeric antigen receptors specific for B-cell maturation antigens
MX2020004567A (en) 2017-11-06 2020-08-13 Genentech Inc Diagnostic and therapeutic methods for cancer.
BR112020010753A2 (en) 2017-12-01 2020-11-24 The Regents Of The University Of California anti-cxcr5 antibodies and compositions and uses thereof
US20210070845A1 (en) 2017-12-15 2021-03-11 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
AU2018389111A1 (en) 2017-12-22 2020-06-18 Jounce Therapeutics, Inc. Antibodies to LILRB2
TW201929907A (en) 2017-12-22 2019-08-01 美商建南德克公司 Use of PILRA binding agents for treatment of a Disease
EP3732202A4 (en) 2017-12-28 2022-06-15 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against tigit
KR20200104886A (en) 2017-12-28 2020-09-04 난징 레전드 바이오테크 씨오., 엘티디. Antibodies and variants against PD-L1
US20210079067A1 (en) 2018-01-02 2021-03-18 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to ebola virus glycoprotein and their use
AU2019205090A1 (en) 2018-01-05 2020-08-06 Ac Immune Sa Misfolded TDP-43 binding molecules
WO2019137541A1 (en) 2018-01-15 2019-07-18 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against pd-1
EP3740505A1 (en) 2018-01-16 2020-11-25 Lakepharma Inc. Bispecific antibody that binds cd3 and another target
BR112020015016A2 (en) 2018-01-26 2020-12-29 Genentech, Inc. PHARMACEUTICAL COMPOSITIONS, METHODS OF TREATMENT OF INFLAMMATORY INTESTINAL DISEASE, INHIBITION OF MICROBIAL INFECTION IN THE INTESTINE AND ACCELERATING OR IMPROVING WOUND HEALING
KR20200115546A (en) 2018-01-26 2020-10-07 제넨테크, 인크. IL-22 Fc fusion protein and method of use
CA3081125A1 (en) 2018-02-01 2019-08-08 Innovent Biologics (Suzhou) Co., Ltd. Fully humanized anti-b cell maturation antigen (bcma) single-chain antibody and use thereof
PE20211116A1 (en) 2018-02-08 2021-06-23 Genentech Inc BISPECIFIC ANTIGEN BINDING MOLECULES AND METHODS OF USE
CA3226165A1 (en) 2018-02-09 2019-08-15 Genentech, Inc. Therapeutic and diagnostic methods for mast cell-mediated inflammatory diseases
EP3752530A1 (en) 2018-02-14 2020-12-23 ABBA Therapeutics AG Anti-human pd-l2 antibodies
RU2020130795A (en) 2018-02-21 2022-03-21 Дзе Юнайтед Стэйтс Оф Америка, Эс Репрезентед Бай Дзе Секретэри, Департмент Оф Хелт Энд Хьюман Сервисиз NEUTRALIZING ANTIBODIES TO ENV HIV-1 AND THEIR USE
AU2019226085A1 (en) 2018-02-21 2020-09-17 Genentech, Inc. Dosing for treatment with IL-22 Fc fusion proteins
US20200399376A1 (en) 2018-02-26 2020-12-24 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
US20200040103A1 (en) 2018-03-14 2020-02-06 Genentech, Inc. Anti-klk5 antibodies and methods of use
US11485782B2 (en) 2018-03-14 2022-11-01 Beijing Xuanyi Pharmasciences Co., Ltd. Anti-claudin 18.2 antibodies
CN116327926A (en) 2018-03-15 2023-06-27 中外制药株式会社 Anti-dengue virus antibodies with cross-reactivity to Zika virus and methods of use
CN111886254B (en) 2018-03-30 2023-12-08 南京传奇生物科技有限公司 Single domain antibodies against LAG-3 and uses thereof
TW202011029A (en) 2018-04-04 2020-03-16 美商建南德克公司 Methods for detecting and quantifying FGF21
SG11202009284TA (en) 2018-04-05 2020-10-29 Juno Therapeutics Inc T cell receptors and engineered cells expressing same
BR112020023118A2 (en) 2018-05-14 2021-04-13 Werewolf Therapeutics, Inc. ACTIVE ACTIVABLE INTERLEUKIN POLYPEPTIDS 12 AND METHODS OF USE OF THESE
KR20210021467A (en) 2018-05-14 2021-02-26 웨어울프 세라퓨틱스, 인크. Activatable interleukin-2 polypeptide and method of use thereof
EP3805400A4 (en) 2018-06-04 2022-06-29 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule showing changed half-life in cytoplasm
BR112020026384A2 (en) 2018-06-23 2021-03-30 Genentech, Inc. METHODS FOR TREATING AN INDIVIDUAL WITH LUNG CANCER AND FOR TREATING AN INDIVIDUAL WITH SMALL CELL LUNG CANCER, KITS, ANTIBODY ANTI-PD-L1 AND COMPOSITION
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
CA3104147A1 (en) 2018-07-18 2020-01-23 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
BR112021000727A2 (en) 2018-07-20 2021-04-13 Surface Oncology, Inc. ANTI-CD112R COMPOSITIONS AND METHODS
JPWO2020027330A1 (en) 2018-08-03 2021-08-19 中外製薬株式会社 Antigen-binding molecule containing two antigen-binding domains linked to each other
EP3608674A1 (en) 2018-08-09 2020-02-12 Regeneron Pharmaceuticals, Inc. Methods for assessing binding affinity of an antibody variant to the neonatal fc receptor
SG10202106830VA (en) 2018-08-10 2021-08-30 Chugai Pharmaceutical Co Ltd Anti-cd137 antigen-binding molecule and utilization thereof
TW202021618A (en) 2018-08-17 2020-06-16 美商23與我有限公司 Anti-il1rap antibodies and methods of use thereof
GB201814281D0 (en) 2018-09-03 2018-10-17 Femtogenix Ltd Cytotoxic agents
US20220098613A1 (en) 2018-09-12 2022-03-31 Fred Hutchinson Cancer Research Center Reducing cd33 expression to selectively protect therapeutic cells
WO2020061060A1 (en) 2018-09-19 2020-03-26 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
EP4249917A3 (en) 2018-09-21 2023-11-08 F. Hoffmann-La Roche AG Diagnostic methods for triple-negative breast cancer
AU2019347751A1 (en) 2018-09-27 2021-04-29 Xilio Development, Inc. Masked cytokine polypeptides
KR20210074286A (en) 2018-10-05 2021-06-21 파이브 프라임 테라퓨틱스, 인크. Anti-FGFR2 antibody formulation
WO2020081493A1 (en) 2018-10-16 2020-04-23 Molecular Templates, Inc. Pd-l1 binding proteins
EP3867646A1 (en) 2018-10-18 2021-08-25 F. Hoffmann-La Roche AG Diagnostic and therapeutic methods for sarcomatoid kidney cancer
EP3880714A4 (en) 2018-11-16 2022-07-20 Memorial Sloan Kettering Cancer Center Antibodies to mucin-16 and methods of use thereof
JP2022511502A (en) 2018-12-05 2022-01-31 ジェネンテック, インコーポレイテッド Diagnostic Methods and Diagnostic Compositions for Cancer Immunotherapy
WO2020117257A1 (en) 2018-12-06 2020-06-11 Genentech, Inc. Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
EP3883609A2 (en) 2018-12-20 2021-09-29 The United States of America, as represented by the Secretary, Department of Health and Human Services Ebola virus glycoprotein-specific monoclonal antibodies and uses thereof
AR117453A1 (en) 2018-12-20 2021-08-04 Genentech Inc CF OF MODIFIED ANTIBODIES AND METHODS TO USE THEM
AR117327A1 (en) 2018-12-20 2021-07-28 23Andme Inc ANTI-CD96 ANTIBODIES AND METHODS OF USE OF THEM
CA3120474A1 (en) 2018-12-21 2020-06-25 23Andme, Inc. Anti-il-36 antibodies and methods of use thereof
MX2021007768A (en) 2018-12-26 2021-08-24 Xilio Dev Inc Anti-ctla4 antibodies and methods of use thereof.
WO2020141145A1 (en) 2018-12-30 2020-07-09 F. Hoffmann-La Roche Ag Anti-rabbit cd19 antibodies and methods of use
TW202043272A (en) 2019-01-14 2020-12-01 美商建南德克公司 Methods of treating cancer with a pd-1 axis binding antagonist and an rna vaccine
CN113329770A (en) 2019-01-24 2021-08-31 中外制药株式会社 Novel cancer antigen and antibody against said antigen
MA54863A (en) 2019-01-29 2021-12-08 Juno Therapeutics Inc TYROSINE KINASE RECEPTOR-LIKE (ROR1) RECEPTOR ORPHAN-1 SPECIFIC CHIMERA ANTIGENIC ANTIBODIES AND RECEPTORS
GB201901197D0 (en) 2019-01-29 2019-03-20 Femtogenix Ltd G-A Crosslinking cytotoxic agents
JP2022521773A (en) 2019-02-27 2022-04-12 ジェネンテック, インコーポレイテッド Dosing for treatment with anti-TIGIT antibody and anti-CD20 antibody or anti-CD38 antibody
KR20210138588A (en) 2019-03-08 2021-11-19 제넨테크, 인크. Methods for detecting and quantifying membrane-associated proteins on extracellular vesicles
TW202100556A (en) 2019-03-14 2021-01-01 美商建南德克公司 Treatment with her2 t cell-dependent bispecific antibodies
US20220143094A1 (en) 2019-04-19 2022-05-12 Chugai Seiyaku Kabushiki Kaisha Chimeric receptor that recognizes engineered site in antibody
WO2020214995A1 (en) 2019-04-19 2020-10-22 Genentech, Inc. Anti-mertk antibodies and their methods of use
WO2020227228A2 (en) 2019-05-03 2020-11-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use
EP3962947A2 (en) 2019-05-03 2022-03-09 F. Hoffmann-La Roche AG Methods of treating cancer with an anti-pd-l1 antibody
AU2020275415A1 (en) 2019-05-14 2021-11-25 Genentech, Inc. Methods of using anti-CD79B immunoconjugates to treat follicular lymphoma
CA3137512A1 (en) 2019-05-14 2020-11-19 Werewolf Therapeutics, Inc. Separation moieties and methods and use thereof
US20230085439A1 (en) 2019-05-21 2023-03-16 University Of Georgia Research Foundation, Inc. Antibodies that bind human metapneumovirus fusion protein and their use
MX2021014274A (en) 2019-05-23 2022-01-06 Ac Immune Sa Anti-tdp-43 binding molecules and uses thereof.
AU2020299382A1 (en) 2019-07-02 2022-01-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind EGFRvIII and their use
JPWO2021010326A1 (en) 2019-07-12 2021-01-21
CN114641490B (en) 2019-08-06 2023-06-06 新旭生技股份有限公司 Antibodies that bind to pathological TAU species and uses thereof
TW202118512A (en) 2019-09-12 2021-05-16 美商建南德克公司 Compositions and methods of treating lupus nephritis
US20220348687A1 (en) 2019-09-20 2022-11-03 Genentech, Inc. Dosing for anti-tryptase antibodies
JP2022548978A (en) 2019-09-27 2022-11-22 ジェネンテック, インコーポレイテッド Dosing for Treatment with Drugs Anti-TIGIT and Anti-PD-L1 Antagonist Antibodies
TW202122114A (en) 2019-10-18 2021-06-16 美商建南德克公司 Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
MX2022005400A (en) 2019-11-06 2022-05-24 Genentech Inc Diagnostic and therapeutic methods for treatment of hematologic cancers.
JP2023504736A (en) 2019-12-06 2023-02-06 ジュノー セラピューティクス インコーポレイテッド Anti-idiotypic antibodies against GPRC5D target binding domains and related compositions and methods
US20230028050A1 (en) 2019-12-06 2023-01-26 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
AU2020403145A1 (en) 2019-12-13 2022-07-07 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
CR20220357A (en) 2019-12-27 2022-08-24 Chugai Pharmaceutical Co Ltd Anti-ctla-4 antibody and use thereof
CN113045655A (en) 2019-12-27 2021-06-29 高诚生物医药(香港)有限公司 anti-OX 40 antibodies and uses thereof
CN110818795B (en) 2020-01-10 2020-04-24 上海复宏汉霖生物技术股份有限公司 anti-TIGIT antibodies and methods of use
WO2022050954A1 (en) 2020-09-04 2022-03-10 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
WO2021194481A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
CN116650628A (en) 2020-01-31 2023-08-29 基因泰克公司 Method for inducing neoepitope specific T cells with PD-1 axis binding antagonists and RNA vaccines
CN115768800A (en) 2020-01-31 2023-03-07 克利夫兰诊所基金会 Anti-mullerian hormone receptor 2 antibodies and methods of use
TW202144395A (en) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 Anti-CD137 antigen-binding molecule for use in cancer treatment
IL295023A (en) 2020-02-14 2022-09-01 Jounce Therapeutics Inc Antibodies and fusion proteins that bind to ccr8 and uses thereof
WO2021168292A1 (en) 2020-02-20 2021-08-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Epstein-barr virus monoclonal antibodies and uses thereof
JP2023516945A (en) 2020-02-28 2023-04-21 上海復宏漢霖生物技術股▲フン▼有限公司 Anti-CD137 constructs and uses thereof
CA3169910A1 (en) 2020-02-28 2021-09-02 Shanghai Henlius Biotech, Inc. Anti-cd137 constructs, multispecific antibody and uses thereof
CR20220461A (en) 2020-03-13 2022-10-21 Genentech Inc Anti-interleukin-33 antibodies and uses thereof
KR20220156575A (en) 2020-03-19 2022-11-25 제넨테크, 인크. Isotype Selective Anti-TGF-beta Antibodies and Methods of Use
CA3169967A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Tie2-binding agents and methods of use
CN115698717A (en) 2020-04-03 2023-02-03 基因泰克公司 Methods of treatment and diagnosis of cancer
WO2021207662A1 (en) 2020-04-10 2021-10-14 Genentech, Inc. Use of il-22fc for the treatment or prevention of pneumonia, acute respiratory distress syndrome, or cytokine release syndrome
WO2021217051A1 (en) 2020-04-24 2021-10-28 Genentech, Inc. Methods of using anti-cd79b immunoconjugates
JP2023523145A (en) 2020-04-27 2023-06-02 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Isotype-independent antibody against lipoprotein (a)
WO2021222167A1 (en) 2020-04-28 2021-11-04 Genentech, Inc. Methods and compositions for non-small cell lung cancer immunotherapy
EP4146283A1 (en) 2020-05-03 2023-03-15 Levena (Suzhou) Biopharma Co., Ltd. Antibody-drug conjugates (adcs) comprising an anti-trop-2 antibody, compositions comprising such adcs, as well as methods of making and using the same
EP4157881A1 (en) 2020-05-27 2023-04-05 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Antibodies specifically recognizing nerve growth factor and uses thereof
EP4157461A1 (en) 2020-05-29 2023-04-05 23Andme, Inc. Anti-cd200r1 antibodies and methods of use thereof
TW202210515A (en) 2020-06-02 2022-03-16 美商當康生物科技有限公司 Anti-cd93 constructs and uses thereof
CN116529260A (en) 2020-06-02 2023-08-01 当康生物技术有限责任公司 anti-CD 93 constructs and uses thereof
CN115698719A (en) 2020-06-12 2023-02-03 基因泰克公司 Methods and compositions for cancer immunotherapy
KR20230025691A (en) 2020-06-16 2023-02-22 제넨테크, 인크. Methods and compositions for treating triple negative breast cancer
WO2021257124A1 (en) 2020-06-18 2021-12-23 Genentech, Inc. Treatment with anti-tigit antibodies and pd-1 axis binding antagonists
JP2023531222A (en) 2020-06-22 2023-07-21 アルミラル・ソシエダッド・アノニマ ANTI-IL-36 ANTIBODY AND METHODS OF USE THEREOF
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds
CN116783215A (en) 2020-07-29 2023-09-19 当康生物技术有限责任公司 anti-CD 93 constructs and uses thereof
JP2023536602A (en) 2020-08-03 2023-08-28 ジェネンテック, インコーポレイテッド Diagnostic and therapeutic methods for lymphoma
US20230324408A1 (en) 2020-08-05 2023-10-12 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
EP4192854A1 (en) 2020-08-07 2023-06-14 Genentech, Inc. Flt3 ligand fusion proteins and methods of use
TW202221026A (en) 2020-08-14 2022-06-01 瑞士商Ac 免疫有限公司 Humanized anti-TDP-43 binding molecules and uses thereof
EP4204448A2 (en) 2020-08-27 2023-07-05 cureab GmbH Anti-golph2 antibodies for macrophage and dendritic cell differentiation
JP2023541627A (en) 2020-09-14 2023-10-03 イシュノス サイエンシズ ソシエテ アノニム Antibodies that bind to IL1RAP and uses thereof
MX2023003685A (en) 2020-09-30 2023-06-02 Dren Bio Inc Anti-cd94 antibodies and methods of use thereof.
CA3193952A1 (en) 2020-10-05 2022-04-14 Bernard Martin Fine Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
EP4232475A1 (en) 2020-10-20 2023-08-30 Kantonsspital St. Gallen Antibodies or antigen-binding fragments specifically binding to gremlin-1 and uses thereof
JP2023545566A (en) 2020-10-20 2023-10-30 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy with PD-1 axis binding antagonist and LRRK2 inhibitor
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
WO2022098638A2 (en) 2020-11-04 2022-05-12 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
IL302217A (en) 2020-11-04 2023-06-01 Genentech Inc Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies and anti-cd79b antibody drug conjugates
WO2022098628A2 (en) 2020-11-04 2022-05-12 Genentech, Inc. Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
TW202237639A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
TW202237638A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
WO2022132904A1 (en) 2020-12-17 2022-06-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies targeting sars-cov-2
WO2022140797A1 (en) 2020-12-23 2022-06-30 Immunowake Inc. Immunocytokines and uses thereof
WO2022162203A1 (en) 2021-01-28 2022-08-04 Vaccinvent Gmbh Method and means for modulating b-cell mediated immune responses
AU2022212599A1 (en) 2021-01-28 2023-08-17 Universität Ulm Method and means for modulating b-cell mediated immune responses
CN117120084A (en) 2021-01-28 2023-11-24 维肯芬特有限责任公司 Methods and means for modulating B cell mediated immune responses
KR20230142790A (en) 2021-02-09 2023-10-11 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Antibodies targeting the coronavirus SPIKE PROTEIN
CA3210753A1 (en) 2021-02-09 2022-08-18 University Of Georgia Research Foundation, Inc. Human monoclonal antibodies against pneumococcal antigens
TW202302626A (en) 2021-02-15 2023-01-16 日商武田藥品工業股份有限公司 Cell therapy compositions and methods for modulating tgf-b signaling
JP2024508488A (en) 2021-03-01 2024-02-27 エクシリオ デベロップメント, インコーポレイテッド Combination of masked CTLA4 and PD1/PD-L1 antibodies to treat cancer
US20220306743A1 (en) 2021-03-01 2022-09-29 Xilio Development, Inc. Combination of ctla4 and pd1/pdl1 antibodies for treating cancer
JP2024509169A (en) 2021-03-03 2024-02-29 ソレント・セラピューティクス・インコーポレイテッド Antibody-drug conjugates including anti-BCMA antibodies
EP4301472A1 (en) 2021-03-05 2024-01-10 Dynamicure Biotechnology LLC Anti-vista constructs and uses thereof
IL305758A (en) 2021-03-10 2023-11-01 Immunowake Inc Immunomodulatory molecules and uses thereof
WO2022198192A1 (en) 2021-03-15 2022-09-22 Genentech, Inc. Compositions and methods of treating lupus nephritis
WO2022197877A1 (en) 2021-03-19 2022-09-22 Genentech, Inc. Methods and compositions for time delayed bio-orthogonal release of cytotoxic agents
EP4314049A1 (en) 2021-03-25 2024-02-07 Dynamicure Biotechnology LLC Anti-igfbp7 constructs and uses thereof
AR125344A1 (en) 2021-04-15 2023-07-05 Chugai Pharmaceutical Co Ltd ANTI-C1S ANTIBODY
CN117321078A (en) 2021-04-30 2023-12-29 豪夫迈·罗氏有限公司 Administration for combination therapy with anti-CD 20/anti-CD 3 bispecific antibody and anti-CD 79B antibody drug conjugates
WO2022241446A1 (en) 2021-05-12 2022-11-17 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
AU2022273303A1 (en) 2021-05-14 2023-11-02 Genentech, Inc. Agonists of trem2
WO2022255440A1 (en) 2021-06-04 2022-12-08 Chugai Seiyaku Kabushiki Kaisha Anti-ddr2 antibodies and uses thereof
KR20240019109A (en) 2021-06-09 2024-02-14 에프. 호프만-라 로슈 아게 Combination of a specific BRAF inhibitor (Paradox Break) and a PD-1 axis binding antagonist for use in the treatment of cancer
WO2022266660A1 (en) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
CN117616123A (en) 2021-06-25 2024-02-27 中外制药株式会社 anti-CTLA-4 antibodies
CA3220353A1 (en) 2021-06-25 2022-12-29 Chugai Seiyaku Kabushiki Kaisha Use of anti-ctla-4 antibody
WO2023283611A1 (en) 2021-07-08 2023-01-12 Staidson Biopharma Inc. Antibodies specifically recognizing tnfr2 and uses thereof
CN115812082A (en) 2021-07-14 2023-03-17 舒泰神(北京)生物制药股份有限公司 Antibody specifically recognizing CD40 and application thereof
WO2023004386A1 (en) 2021-07-22 2023-01-26 Genentech, Inc. Brain targeting compositions and methods of use thereof
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023019239A1 (en) 2021-08-13 2023-02-16 Genentech, Inc. Dosing for anti-tryptase antibodies
WO2023021055A1 (en) 2021-08-19 2023-02-23 F. Hoffmann-La Roche Ag Multivalent anti-variant fc-region antibodies and methods of use
AU2022332303A1 (en) 2021-08-27 2024-02-01 Genentech, Inc. Methods of treating tau pathologies
WO2023034750A1 (en) 2021-08-30 2023-03-09 Genentech, Inc. Anti-polyubiquitin multispecific antibodies
AU2022345251A1 (en) 2021-09-17 2024-03-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Synthetic humanized llama nanobody library and use thereof to identify sars-cov-2 neutralizing antibodies
TW202321308A (en) 2021-09-30 2023-06-01 美商建南德克公司 Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023081818A1 (en) 2021-11-05 2023-05-11 American Diagnostics & Therapy, Llc (Adxrx) Monoclonal antibodies against carcinoembryonic antigens, and their uses
WO2023086807A1 (en) 2021-11-10 2023-05-19 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
WO2023091887A1 (en) 2021-11-16 2023-05-25 Genentech, Inc. Methods and compositions for treating systemic lupus erythematosus (sle) with mosunetuzumab
TW202334202A (en) 2021-11-16 2023-09-01 瑞士商Ac免疫有限公司 Novel molecules for therapy and diagnosis
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023154824A1 (en) 2022-02-10 2023-08-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies that broadly target coronaviruses
TW202342519A (en) 2022-02-16 2023-11-01 瑞士商Ac 免疫有限公司 Humanized anti-tdp-43 binding molecules and uses thereof
TW202342520A (en) 2022-02-18 2023-11-01 美商樂天醫藥生技股份有限公司 Anti-programmed death-ligand 1 (pd-l1) antibody molecules, encoding polynucleotides, and methods of use
WO2023173026A1 (en) 2022-03-10 2023-09-14 Sorrento Therapeutics, Inc. Antibody-drug conjugates and uses thereof
US20230414750A1 (en) 2022-03-23 2023-12-28 Hoffmann-La Roche Inc. Combination treatment of an anti-cd20/anti-cd3 bispecific antibody and chemotherapy
WO2023192827A1 (en) 2022-03-26 2023-10-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Bispecific antibodies to hiv-1 env and their use
WO2023192881A1 (en) 2022-03-28 2023-10-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to hiv-1 env and their use
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023194565A1 (en) 2022-04-08 2023-10-12 Ac Immune Sa Anti-tdp-43 binding molecules
WO2023198727A1 (en) 2022-04-13 2023-10-19 F. Hoffmann-La Roche Ag Pharmaceutical compositions of anti-cd20/anti-cd3 bispecific antibodies and methods of use
WO2023201299A1 (en) 2022-04-13 2023-10-19 Genentech, Inc. Pharmaceutical compositions of therapeutic proteins and methods of use
WO2023203177A1 (en) 2022-04-20 2023-10-26 Kantonsspital St. Gallen Antibodies or antigen-binding fragments pan-specifically binding to gremlin-1 and gremlin-2 and uses thereof
WO2023215737A1 (en) 2022-05-03 2023-11-09 Genentech, Inc. Anti-ly6e antibodies, immunoconjugates, and uses thereof
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023235699A1 (en) 2022-05-31 2023-12-07 Jounce Therapeutics, Inc. Antibodies to lilrb4 and uses thereof
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2023237706A2 (en) 2022-06-08 2023-12-14 Institute For Research In Biomedicine (Irb) Cross-specific antibodies, uses and methods for discovery thereof
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020407A1 (en) 2022-07-19 2024-01-25 Staidson Biopharma Inc. Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024030829A1 (en) 2022-08-01 2024-02-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind to the underside of influenza viral neuraminidase
WO2024028732A1 (en) 2022-08-05 2024-02-08 Janssen Biotech, Inc. Cd98 binding constructs for treating brain tumors
WO2024028731A1 (en) 2022-08-05 2024-02-08 Janssen Biotech, Inc. Transferrin receptor binding proteins for treating brain tumors
WO2024037633A2 (en) 2022-08-19 2024-02-22 Evive Biotechnology (Shanghai) Ltd Formulations comprising g-csf and uses thereof
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024054822A1 (en) 2022-09-07 2024-03-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Engineered sars-cov-2 antibodies with increased neutralization breadth
WO2024054929A1 (en) 2022-09-07 2024-03-14 Dynamicure Biotechnology Llc Anti-vista constructs and uses thereof
WO2024064826A1 (en) 2022-09-22 2024-03-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001064754A1 (en) * 2000-03-03 2001-09-07 Kyowa Hakko Kogyo Co., Ltd. Gene recombinant antibody and its fragment
WO2002031140A1 (en) * 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
WO2002078739A1 (en) * 2001-03-29 2002-10-10 Kyowa Hakko Kogyo Co., Ltd. Drugs containing genetically modified antibody against ganglioside gd3
WO2003035835A2 (en) * 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
WO2003055993A1 (en) * 2001-12-25 2003-07-10 Kyowa Hakko Kogyo Co., Ltd. Composition of antibody specifically binding to cd20
WO2003085119A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003085118A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Process for producing antibody composition
WO2003085102A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cell with depression or deletion of the activity of protein participating in gdp-fucose transport
WO2003085107A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
JP2007533639A (en) * 2003-10-16 2007-11-22 ラボラトワール・フランセ・デュ・フラクシオンマン・エ・デ・ビョテクノロジー Anti-RHESUS-DIgG3 of YB2 / 0 cell line with strong phagocytic activity

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2420835T3 (en) * 1999-04-09 2013-08-27 Kyowa Hakko Kirin Co., Ltd. Procedure to control the activity of immunofunctional molecules
AU7449000A (en) * 1999-09-30 2001-04-30 Kyowa Hakko Kogyo Co. Ltd. Human type complementation-determining domain transplanted antibody against ganglioside gd2 and derivative of this antibody

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001064754A1 (en) * 2000-03-03 2001-09-07 Kyowa Hakko Kogyo Co., Ltd. Gene recombinant antibody and its fragment
WO2002031140A1 (en) * 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
WO2002078739A1 (en) * 2001-03-29 2002-10-10 Kyowa Hakko Kogyo Co., Ltd. Drugs containing genetically modified antibody against ganglioside gd3
WO2003035835A2 (en) * 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
WO2003055993A1 (en) * 2001-12-25 2003-07-10 Kyowa Hakko Kogyo Co., Ltd. Composition of antibody specifically binding to cd20
WO2003085119A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003085118A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Process for producing antibody composition
WO2003085102A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cell with depression or deletion of the activity of protein participating in gdp-fucose transport
WO2003085107A1 (en) * 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
JP2007533639A (en) * 2003-10-16 2007-11-22 ラボラトワール・フランセ・デュ・フラクシオンマン・エ・デ・ビョテクノロジー Anti-RHESUS-DIgG3 of YB2 / 0 cell line with strong phagocytic activity

Also Published As

Publication number Publication date
WO2005053742A1 (en) 2005-06-16

Similar Documents

Publication Publication Date Title
JPWO2005053742A1 (en) Medicament containing antibody composition
US20200017593A1 (en) Method of producing antibodies with n-glycoside-linked sugar chains lacking fucosylation

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20071005

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20101020

A02 Decision of refusal

Free format text: JAPANESE INTERMEDIATE CODE: A02

Effective date: 20110419