JPH05213775A - Bfa antibody - Google Patents

Bfa antibody

Info

Publication number
JPH05213775A
JPH05213775A JP1996892A JP1996892A JPH05213775A JP H05213775 A JPH05213775 A JP H05213775A JP 1996892 A JP1996892 A JP 1996892A JP 1996892 A JP1996892 A JP 1996892A JP H05213775 A JPH05213775 A JP H05213775A
Authority
JP
Japan
Prior art keywords
antibody
bfa
human
cells
erbb
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
JP1996892A
Other languages
Japanese (ja)
Inventor
Yoshihiro Sugiyama
孔宏 杉山
Yoshinori Kashiwabara
美紀 柏原
Masafumi Shibamori
雅文 柴森
Kyohei Deguchi
恭平 出口
Kenichi Imagawa
健一 今川
Mikio Kikuchi
幹雄 菊地
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Otsuka Pharmaceutical Co Ltd
Original Assignee
Otsuka Pharmaceutical Co Ltd
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 Otsuka Pharmaceutical Co Ltd filed Critical Otsuka Pharmaceutical Co Ltd
Priority to JP1996892A priority Critical patent/JPH05213775A/en
Publication of JPH05213775A publication Critical patent/JPH05213775A/en
Pending legal-status Critical Current

Links

Landscapes

  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

PURPOSE:To obtain a BFA antibody effective for treating cancers, especially malignant tumors such as human mammary cancer. CONSTITUTION:A complex antibody wherein one antibody constituting the complex antibody is human c-erbB-2 specific antibody and the other is human lymphocyte antibody. Especially the complex antibody wherein an antibody recognizing surface antigen of target cell is anti-human c-erbB-2 antibody and an antibody which is bonded to an effector cell and transmits a signal is an anti-human lymphocyte antibody. A therapeutic agent for cancer comprising the complex antibody as an essential component.

Description

【発明の詳細な説明】Detailed Description of the Invention

【0001】[0001]

【産業上の利用分野】本発明は癌治療に有効な複合抗
体、より詳しくはヒト乳癌等の悪性腫瘍治療に有効な複
合抗体及び癌治療剤に関する。
TECHNICAL FIELD The present invention relates to a composite antibody effective for treating cancer, and more particularly to a composite antibody effective for treating malignant tumors such as human breast cancer and a therapeutic agent for cancer.

【0002】[0002]

【従来の技術】従来、癌免疫療法においてモノクローナ
ル抗体は抗体依存性細胞性細胞傷害作用(ADCC;an
tibody-dependent celluar cytotoxicity)[Martin,J.
S.,et al., Blood, 73, 1431-1439 (1989)] と或るいは
補体依存性溶菌作用(complement-dependent cytolysis)
[Irie,R.F.,et al., Proc.Natl.Acad.U.S.A., 83, 8649
-8698 (1986)] に基づく受動免疫により広く臨床試験が
なされた。しかしながら、癌治療のために必要な効能の
ある細胞傷害性免疫反応を誘導するモノクローナル抗体
は例外的であった。之等の臨床結果はモノクローナル抗
体による癌治療の研究の限界を示すものであった。
2. Description of the Related Art Conventionally, in cancer immunotherapy, a monoclonal antibody has an antibody-dependent cellular cytotoxicity (ADCC;
tibody-dependent celluar cytotoxicity) [Martin, J.
S., et al., Blood, 73 , 1431-1439 (1989)] or complement-dependent cytolysis.
[Irie, RF, et al., Proc.Natl.Acad.USA, 83 , 8649
-8698 (1986)] has been widely clinically tested by passive immunization. However, the monoclonal antibody that induces the efficacious cytotoxic immune response required for cancer treatment was exceptional. Their clinical results have shown the limit of research on cancer treatment with monoclonal antibodies.

【0003】上記モノクロナール抗体による問題点を克
服するための有力な方法として、異なった二種類の抗体
分子を解離させ、再結合してハイブリッド抗体を得る試
みがなされ、これは古くはウサギポリクローナル抗体を
用いて行なわれている[Nisonoff,A. and M.M.Rivers, A
rch.biochem.Biophys., 93, 460 (1961)] が、実際にこ
れが広く知られたのはモノクロナール抗体の作製技術が
開発されてからである。[Koehler,G. and C.Milstein,
Nature, 256, 495 (1975)]。
As a promising method for overcoming the problems caused by the above-mentioned monoclonal antibody, an attempt was made to dissociate two different kinds of antibody molecules and re-bond them to obtain a hybrid antibody, which was a rabbit polyclonal antibody in the old days. [Nisonoff, A. and MMRivers, A
rch.biochem.Biophys., 93 , 460 (1961)], but it was not widely known until the technology for producing monoclonal antibodies was developed. [Koehler, G. and C. Milstein,
Nature, 256, 495 (1975)].

【0004】これら二種類のモノクロナール抗体のそれ
ぞれの半分ずつを化学的に結合させてハイブリッド抗体
(モノマータイプ)を作製する方法は数多く報告され、
之等は一般にF(ab′)2 分子を材料として作製され
ている。例えばブレンナン(Brennan)らはDTT(ジチ
オスレイトール)の還元剤を用いて該F(ab′)2
子を還元処理し、一方のFab′のSH基を5,5′−
ジチオビス(2−ニトロ安息香酸)(DTNB)で保護
してFab′−SNBとし、他方のFab′−SHと混
合することにより、高収率でモノマータイプのハイブリ
ッド抗体を作製することに成功している[Brennan,M.,
P.F.Davison and H.Pavlus, Science, 229, 81-83 (198
5) ; Nitta,T., et al., Eur.J.Immunol., 19, 1437-14
41 (1989)参照]。
Many methods have been reported for producing a hybrid antibody (monomer type) by chemically binding each half of these two types of monoclonal antibodies.
In general, they are produced using F (ab ') 2 molecules as a material. For example, Brennan et al. Reduced the F (ab ') 2 molecule with a reducing agent of DTT (dithiothreitol) to reduce the SH group of one Fab' to 5,5'-.
Fab'-SNB was protected with dithiobis (2-nitrobenzoic acid) (DTNB) and mixed with the other Fab'-SH to successfully prepare a monomer-type hybrid antibody in high yield. [Brennan, M.,
PFDavison and H. Pavlus, Science, 229, 81-83 (198
5); Nitta, T., Et al., Eur.J.Immunol., 19 , 1437-14
41 (1989)].

【0005】また別の方法としては、異なった二種類の
抗体分子を架橋剤SPDP[N−スクシンイミジル−3
−(1−ビリジルジチオ)プロピオネート]により連結
したダイマータイプのハイブリッド抗体を作製する方法
[Staerz,U.D., et al., Nature, 314, 628-631 (1985)]
や、二種類の抗体F(ab′)2 フラグメントを同様に
して架橋剤SPDPを用いて結合させたダイマータイプ
のハイブリッド抗体を作製する方法[Nitta,T., et al.,
Eur.J.Immunol.,19, 1437-1441 (1989)] 等がある。
Another method is to use two different antibody molecules as a cross-linking agent SPDP [N-succinimidyl-3].
-(1-Vyridyldithio) propionate] to produce a dimer-type hybrid antibody
[Staerz, UD, et al., Nature, 314, 628-631 (1985)]
Alternatively, a method for producing a dimer type hybrid antibody in which two types of antibody F (ab ′) 2 fragments are similarly bound using a crosslinking agent SPDP [Nitta, T., et al.,
Eur.J.Immunol., 19 , 1437-1441 (1989)].

【0006】ハイブリッド抗体を作製するもう一つの手
段としては、細胞融合法を利用する方法がある。例え
ば、ある抗原に対する抗体生産ハイブリドーマを、別の
抗原で免疫した動物の脾細胞と融合させて、ハイブリッ
ド抗体産生ハイブリドーマを得、該ハイブリドーマの培
養によって目的とするハイブリッド抗体を得る方法[Mil
stein,C. and A.C.Cuello, Nature, 305, 537 (1983)]
や、異なった抗体産生ハイブリドーマを相互に細胞融合
させてハイブリッド抗体産生ハイブリドーマを得、これ
より所望の抗体を得る方法[Staerz,U.D. and M.J.Beva
n, Proc.Natl.Acad.Sci, U.S.A., 83, 1453 (1986) ; L
anzavecchia,A. and D.Scheidegger, Eur.J.Immunol.,1
7, 105 (1987)] 等がある。
[0006] As another means for producing a hybrid antibody, there is a method utilizing a cell fusion method. For example, a method in which an antibody-producing hybridoma for one antigen is fused with splenocytes of an animal immunized with another antigen to obtain a hybrid antibody-producing hybridoma, and the hybrid antibody of interest is obtained by culturing the hybridoma [Mil
stein, C. and ACCuello, Nature, 305 , 537 (1983)]
Alternatively, a method of obtaining hybrid antibody-producing hybridomas by fusing different antibody-producing hybridomas with each other and obtaining a desired antibody therefrom [Staerz, UD and MJBeva
n, Proc.Natl.Acad.Sci, USA, 83, 1453 (1986); L
anzavecchia, A. and D.Scheidegger, Eur.J.Immunol., 1
7 , 105 (1987)] etc.

【0007】更に、別の型のハイブリッド抗体として
は、抗原結合活性を有する可変部(V)領域をマウスハ
イブリドーマ由来で、免疫活性を有する定常部(C)領
域をヒト由来のものにしたものが知られている。該抗体
の製造方法は遺伝子組み換え技術を用いて試みられた1
984年のモリソン(Morrison) の他、様々な特異性を
もつこの種ハイブリッド抗体の作製方法が例示されてい
る[Morrison SL et al.,Proc.Natl.Acad.Sci.U.S.A.,8
1, 6851 (1984) : Sharon J. et al., Nature, 309,364
(1984) : Neuberger,M.S., et al., Nature, 312, 604
(1984) : Boulianne,G.L., et al., Nature, 312, 634
(1984)等]。更に、新しい型のハイブリッド抗体とし
ては、V領域の相補性決定領域(CDR)のみをマウス
由来としたもの(Reshaped 抗体)をも例示できる[Jone
s,P.T., et al., Nature, 321, 552 (1986) : Riechman
n L., et al., Nature, 332, 323 (1988) ]。
Further, as another type of hybrid antibody, a variable region (V) region having an antigen-binding activity is derived from a mouse hybridoma, and a constant region (C) region having an immunological activity is derived from a human. Are known. The method for producing the antibody has been attempted using gene recombination technology.
In addition to Morrison of 984, a method for producing this kind of hybrid antibody having various specificities is exemplified [Morrison SL et al., Proc. Natl. Acad. Sci. USA, 8
1 , 6851 (1984): Sharon J. et al., Nature, 309, 364
(1984): Neuberger, MS, et al., Nature, 312 , 604
(1984): Boulianne, GL, et al., Nature, 312 , 634.
(1984) etc.]. Further, as a new type of hybrid antibody, a mouse derived only from the complementarity determining region (CDR) of V region (Reshaped antibody) can be exemplified [Jone
s, PT, et al., Nature, 321 , 552 (1986): Riechman
n L., et al., Nature, 332 , 323 (1988)].

【0008】上記ハイブリッド抗体の応用は以下に述べ
る癌治療の他に、イムノアッセイ系への利用[Suresh,M.
R., et al., Proc.Natl.Acad.Sci. U.S.A., 83, 7989-7
993(1986)] 、免疫組織化学での利用[Milsten,C. and
A.C.Cuello, Nature, 305, 537 (1983)]等が報告されて
いる。また、上記モノクロナール抗体による癌治療より
一歩進歩した形でかかる異なった二種類の抗体のハイブ
リッド抗体[複合抗体(以下、BFA;Bifunctional an
tibodyと略称する)の一方の抗体が癌細胞と会合した抗
原を認識し、該BFAの他方の抗体がTリンパ球のT細
胞抗原を認識するBFAを作製することができる。該B
FAの構造に基ずき、標的とする癌細胞に結合すること
ができるBFAが標的癌細胞に対し細胞傷害活性を持つ
ことができる。この機序により癌治療への期待がなされ
た。その後、インビトロにおいて、BFAの活性がいく
つかの腫瘍細胞で研究された[Mezzanica,D., et al., I
nt.J.Cancer,41, 609-615 (1988) ; Mansfield,P.F., e
t al., Cancer Immunol.Immunother, 33, 247-254 (199
1) ; Oshimi,K., et al., Blood,77, 1044-1049 (1991)
; Nitta,T., et al., E.J.Immunol.,19, 1437-1441 (1
989)] 。
The above-mentioned hybrid antibody is applied to immunoassay systems in addition to cancer treatment described below [Suresh, M.
R., et al., Proc.Natl.Acad.Sci. USA, 83 , 7989-7
993 (1986)], use in immunohistochemistry [Milsten, C. and
ACCuello, Nature, 305, 537 (1983)] has been reported. In addition, a hybrid antibody [complex antibody (hereinafter, referred to as BFA; Bifunctional an
BFA in which one antibody (abbreviated as tibody) recognizes an antigen associated with cancer cells and the other antibody recognizes the T cell antigen of T lymphocytes. The B
Based on the structure of FA, BFA capable of binding to a target cancer cell can have cytotoxic activity against the target cancer cell. This mechanism has led to expectations for cancer treatment. Subsequently, the activity of BFA was studied in vitro in some tumor cells [Mezzanica, D., et al., I.
nt.J.Cancer, 41 , 609-615 (1988); Mansfield, PF, e
t al., Cancer Immunol. Immunon, 33 , 247-254 (199
1); Oshimi, K., et al., Blood, 77 , 1044-1049 (1991)
Nitta, T., et al., EJImmunol., 19 , 1437-1441 (1
989)].

【0009】更に神経膠腫、卵巣癌、肺癌に対してもB
FAの臨床研究が着手された[Nitta,T., et al., Lance
t, 335, 368-371 (1990) ; de Leij,L., et al., Fonda
tionNationale de Transfusion Sanguine, Les Ulis Fr
ance, 249-253 (1990)]。
[0009] Furthermore, B can be used for glioma, ovarian cancer, and lung cancer.
Clinical study of FA undertaken [Nitta, T., et al., Lance
t, 335 , 368-371 (1990); de Leij, L., et al., Fonda
tionNationale de Transfusion Sanguine, Les Ulis Fr
ance, 249-253 (1990)].

【0010】一方、近年、細胞生物学の著しい進歩によ
り細胞における増殖因子レセプターと発癌との係わりが
注目されてきた。数ある癌遺伝子産物のうちの一つとし
て特に受容体型チロシンキナーゼ(チロシン残基特異的
蛋白質リン酸化酵素)のうち、上皮細胞成長因子(epide
rmal growth factor; EGF)と該EGFの受容体(レ
セプター)と酷似する蛋白質をコードする関連癌遺伝子
として見い出されたc−erbB−2遺伝子[Yamamoto,
T., et al., Nature, 319, 230-234 (1986)]の癌遺伝子
産物であるp185遺伝子は、種々のヒト悪性腫瘍にお
いて該遺伝子の増幅と癌遺伝子産物の高発現が認められ
ており、特に乳癌、胃癌、肺癌、膵臓癌中に高頻度のc
−erbB−2遺伝子の増幅が認められている。その腺
癌、特に乳癌については、ヒト乳癌と乳癌由来の培養細
胞にc−erbB−2遺伝子の増幅が認められ[King,C.
R., et al., Science, 229, 974-976 (1985) ; Yamamot
o,T., et al., Nature, 319, 230-234 (1986)]、該c−
erbB−2遺伝子の増幅の程度が、乳癌の予後と強い
相関を示すことが既に見出されている[Slamon,D.J., et
al., Science, 235, 177-182 (1987)] 。しかしなが
ら、該遺伝子産物の発現は正常成人組織には、極まれに
しか認められない[Natali,P.G., et al., Int.J.Cance
r, 45, 457-461 (1990)] 。
On the other hand, in recent years, due to remarkable progress in cell biology, attention has been paid to the relationship between growth factor receptors and carcinogenesis in cells. Among the numerous oncogene products, among the receptor tyrosine kinases (tyrosine residue-specific protein kinases), epidermal growth factor (epide
rmal growth factor (EGF) and a c-erbB-2 gene found as a related oncogene encoding a protein that closely resembles the receptor of EGF [Yamamoto,
T., et al., Nature, 319, 230-234 (1986)], the oncogene product p185 gene, has been observed to be amplified and highly expressed oncogene product in various human malignancies. , Especially in breast cancer, gastric cancer, lung cancer, pancreatic cancer
-Amplification of the erbB-2 gene has been observed. Regarding the adenocarcinoma, especially breast cancer, amplification of the c-erbB-2 gene was observed in human breast cancer and cultured cells derived from breast cancer [King, C.
R., et al., Science, 229 , 974-976 (1985); Yamamot
o, T., et al., Nature, 319, 230-234 (1986)], c-
It has already been found that the degree of amplification of the erbB-2 gene has a strong correlation with the prognosis of breast cancer [Slamon, DJ, et.
al., Science, 235 , 177-182 (1987)]. However, expression of the gene product is rarely observed in normal adult tissues [Natali, PG, et al., Int. J. Cancer.
r, 45 , 457-461 (1990)].

【0011】上記c−erbB−2蛋白質に対する抗体
やモノクロナール抗体も既に開発され、病理材料のみな
らず、手術材料を直ちに免疫染色法によって検査する方
法も開発されつつあり[Masuko,T., et al., Jpn.J.Canc
er Res.,80, 10-14 (1989)]; Yamada,Y., et al., Jpn.
J.Cancer Res., 80, 1192-1198 (1989)] 、c−erb
B−2遺伝子産物を認識する抗体としても、例えばc−
erbB−2遺伝子のC末端領域を認識するポリクロー
ナル抗体、pAb1(T4881)[トリトンバイオサ
イエンス社製(Triton Bioscience Inc.;Alameda,CA)]や
キナーゼドメインを認識するポリクローナル抗体Ab−
1[オンコジーンサイエンス(OncogeneScience Inc.;Ma
nhasset,NY)] や、c−erbB−2の細胞外ドメイン
を認識するモノクローナル抗体、SV2−61γ[株式
会社ニチレン(特開平2−150293号公報参照)]
等が知られ、本発明者らも先に腺癌、特に乳癌の診断剤
及び治療剤を提供する目的でヒト乳癌細胞株SK−BR
−3(ATCC寄託番号;ATCC HTB30)の培養上清で免
疫した哺乳動物の免疫細胞と哺乳動物の骨髄細胞との融
合により形成されたハイブリドーマにより産生され、c
−erbB−2関連蛋白質に特異的に反応する抗c−e
rbB−2モノクローナル抗体、GFD−OA−p18
5−1を作製した[Ouzge,Alper, et al., Cell Growth
& Differentation, 1, 591-599 (1990)]。上記GFD−
OA−p185−1抗体は、c−erbB−2遺伝子を
発現するヒト癌細胞株SK−BR−3及び、A−549
(ヒト肺癌細胞株;ATCC CCL185 )のインビトロでの増
殖を有意に抑制した(特願平3−229835号)。上
記各抗体は、c−erbB−2蛋白質とそれぞれ反応
し、c−erbB−2遺伝子産物の発現と関連する疾
患、特に腺癌、中でも乳癌等の診断に有用であるが(特
開平3−191865号公報参照)、これらの抗体は上
記悪性腫瘍の治療剤としては、前記したようにモノクロ
ーナル抗体であり、必ずしも良好な治療効果が予想され
ない。本発明者らはより一般的な上皮癌に対するBFA
による免疫治療法を開発するために、c−erbB−2
遺伝子産物に関連するものを標的とするBFAを開発
し、インビトロにおけるBFAの抗腫瘍活性を評価し、
ここに本発明を完成するに至った。
Antibodies against the c-erbB-2 protein and monoclonal antibodies have already been developed, and not only pathological materials but also methods for immediately examining surgical materials by immunostaining are being developed [Masuko, T., et. al., Jpn.J.Canc
er Res., 80 , 10-14 (1989)]; Yamada, Y., et al., Jpn.
J. Cancer Res., 80 , 1192-1198 (1989)], c-erb
As an antibody that recognizes the B-2 gene product, for example, c-
A polyclonal antibody that recognizes the C-terminal region of the erbB-2 gene, pAb1 (T4881) [Triton Bioscience Inc. (Alameda, CA)], or a polyclonal antibody Ab- that recognizes the kinase domain.
1 [Oncogene Science Inc.; Ma
nhasset, NY)], or a monoclonal antibody that recognizes the extracellular domain of c-erbB-2, SV2-61γ [Nichiren Co., Ltd. (see Japanese Patent Laid-Open No. 2-150293)].
And the like, and the present inventors have previously provided a human breast cancer cell line SK-BR for the purpose of providing a diagnostic agent and therapeutic agent for adenocarcinoma, particularly breast cancer.
-3 (ATCC deposit number; ATCC HTB30) produced by a hybridoma formed by fusion of immune cells of a mammal immunized with the culture supernatant of ATCC HTB30 and bone marrow cells of the mammal, c
-An anti-ce specifically reacting with erbB-2-related protein
rbB-2 monoclonal antibody, GFD-OA-p18
5-1 was prepared [Ouzge, Alper, et al., Cell Growth
& Differentation, 1, 591-599 (1990)]. GFD-
The OA-p185-1 antibody is a human cancer cell line SK-BR-3 expressing the c-erbB-2 gene and A-549.
In vitro proliferation of (human lung cancer cell line; ATCC CCL185) was significantly suppressed (Japanese Patent Application No. 3-229835). Each of the above antibodies reacts with the c-erbB-2 protein, and is useful for diagnosing diseases associated with the expression of the c-erbB-2 gene product, particularly adenocarcinoma, especially breast cancer (Japanese Patent Laid-Open No. 3-191865). As described above, these antibodies are monoclonal antibodies as therapeutic agents for the above-mentioned malignant tumors, and a good therapeutic effect is not necessarily expected. We have BFA for more common epithelial cancers
C-erbB-2 for developing an immunotherapy method by
Develop BFA targeting those related to the gene product and evaluate the anti-tumor activity of BFA in vitro,
The present invention has been completed here.

【0012】尚、c−erbB−2に関連するBFAつ
いては、今だ報告はなく、本発明によりBFAを構成す
る一方の抗体が抗ヒトc−erbB−2抗体からなる複
合抗体を提供することが初めて可能となった。
It should be noted that there has been no report on BFA related to c-erbB-2, and the present invention provides a composite antibody in which one antibody constituting BFA is an anti-human c-erbB-2 antibody. It became possible for the first time.

【0013】[0013]

【発明が解決しようとする問題点】本発明はc−erb
B−2遺伝子を発現する細胞、特に乳癌、肺癌、胃癌、
膵臓癌等の悪性腫瘍細胞等の標的細胞と、細胞傷害性T
リンパ球細胞(Cytotoxic TLymphocytes) 等のエフェク
ター細胞とを特異的に結合させることができ、しかもこ
れによって上記エフェクター細胞の有する上記標的細胞
に対する細胞傷害活性を増強させ得る新しいBFA(複
合抗体)、及びこれを利用した悪性腫瘍等の臨床治療剤
を提供することを目的とするものである。
DISCLOSURE OF THE INVENTION Problems to be Solved by the Invention
Cells expressing the B-2 gene, especially breast cancer, lung cancer, gastric cancer,
Target cells such as malignant tumor cells such as pancreatic cancer and cytotoxic T
A new BFA (complex antibody) capable of specifically binding to effector cells such as lymphocyte cells (Cytotoxic TLymphocytes), and further enhancing the cytotoxic activity of the effector cells against the target cells, and The purpose of the present invention is to provide a clinical therapeutic agent for malignant tumors and the like.

【0014】[0014]

【問題を解決するための手段】本発明によれば、BFA
(複合抗体)を構成する一方の抗体がヒトc−erbB
−2抗体であり、他方がヒトリンパ球抗体であることを
特徴とするBFA、殊に標的細胞の表面抗原を認識する
抗体が抗ヒトc−erbB−2抗体であって、エフェク
ター細胞に結合してシグナル伝達を行なう抗体が抗ヒト
リンパ球抗体である上記BFA、抗ヒトc−erbB−
2抗体がGDF−OA−p185−1である上記BF
A、及び上記抗ヒトリンパ球抗体が抗CD3抗体であ
る、ヒトc−erbB−2関連蛋白に特異的に反応する
上記BFAが提供される。また、本発明によれば、上記
BFAを必須成分とすることを特徴とする癌治療剤が提
供される。
According to the present invention, a BFA
One antibody constituting (complex antibody) is human c-erbB
-2 antibody, and the other is a human lymphocyte antibody, BFA, in particular, the antibody that recognizes the surface antigen of the target cell is an anti-human c-erbB-2 antibody, which binds to effector cells. The above BFA and anti-human c-erbB- in which the antibody that carries out signal transduction is an anti-human lymphocyte antibody.
2 The above BF in which the antibody is GDF-OA-p185-1
A, and the above BFA that specifically reacts with the human c-erbB-2 related protein, wherein the anti-human lymphocyte antibody is an anti-CD3 antibody. Further, according to the present invention, there is provided a cancer therapeutic agent comprising the above BFA as an essential component.

【0015】本発明抗体は、抗原(標的細胞及びエフェ
クター細胞)に対する特異的結合力が強く、またその利
用によって、CTL等のエフェクター細胞の有する標的
細胞に対する細胞傷害活性を活性化乃至増強させ得る特
徴を有しているが、該エフェクター細胞が正常細胞にま
で攻撃するような活性化は行なうことなく、更にFcレ
セプターを介したADCC等による悪影響の可能性をも
回避されている。従って本発明抗体は、殊に悪性腫瘍等
の臨床治療に有効である。
The antibody of the present invention has a strong specific binding ability to antigens (target cells and effector cells), and its use can activate or enhance the cytotoxic activity of effector cells such as CTL against target cells. However, it does not activate the effector cells to attack normal cells, and further avoids the possibility of adverse effects due to ADCC or the like via Fc receptors. Therefore, the antibody of the present invention is particularly effective for clinical treatment of malignant tumors and the like.

【0016】以下、本発明抗体、殊に一方の抗体が抗ヒ
トc−erbB−2抗体のFab′部分であり、他方の
抗体が抗ヒトリンパ球抗体のFab′部分であり、之等
が結合した本発明抗体の作製法につき詳述する。
Hereinafter, the antibodies of the present invention, particularly one of the antibodies is the Fab 'portion of the anti-human c-erbB-2 antibody, and the other antibody is the Fab' portion of the anti-human lymphocyte antibody. The method for producing the antibody of the present invention will be described in detail.

【0017】本発明抗体の作製のための材料とする抗
体、即ちモノクローナル抗体としては、例えば抗c−e
rbB−2抗体等の抗体乃至抗腫瘍抗体、及び抗CD3
抗体等の抗ヒトリンパ球抗体をそれぞれ利用できる。上
記抗c−erbB−2抗体に属する抗c−erbB−2
抗体は癌細胞表面に発現しているc−erbB−2遺伝
子産物を認識するものであり、これにはGFD−OA−
p185−1[Alper,O.,et al., Cell Growth & Differ
entiation, 1, 591-599 (1990)]、遺伝子組換え技術を
用いてヒトc−erbB−2遺伝子を適当な発現ベクタ
ーに組込んで得られる組換えDNAを適当な動物細胞に
導入し、その動物細胞を形質転換し、ヒトc−erbB
−2遺伝子を発現している細胞を選択し、その細胞表面
に発現している細胞をc−erbB−2モノクローナル
抗体作製のための免疫抗原として作製したSV2−61
抗体やSV2−61γ抗体(特開平2−150293号
公報)、TAb251抗体、TAb255抗体、TAb
256抗体、TAb258抗体、TAb259抗体(P
CT公開特許W091−02062号公報)、9G6抗
体[Van de Vijver M.J., et al., New Eng.J.Med., 31
9, 1239 (1988)]等が含まれる。
The antibody used as a material for producing the antibody of the present invention, that is, a monoclonal antibody is, for example, anti-ce
Antibodies such as rbB-2 antibody or antitumor antibody, and anti-CD3
Anti-human lymphocyte antibodies such as antibodies can be used. Anti-c-erbB-2 belonging to the above anti-c-erbB-2 antibody
The antibody recognizes the c-erbB-2 gene product expressed on the surface of cancer cells, which includes GFD-OA-
p185-1 [Alper, O., et al., Cell Growth & Differ
entiation, 1, 591-599 (1990)], a recombinant DNA obtained by incorporating the human c-erbB-2 gene into an appropriate expression vector using a gene recombination technique is introduced into an appropriate animal cell, and Transforming animal cells into human c-erbB
SV2-61 in which cells expressing the C-2 gene were selected, and the cells expressing on the cell surface were prepared as an immunizing antigen for preparing a c-erbB-2 monoclonal antibody
Antibody, SV2-61γ antibody (JP-A-2-150293), TAb251 antibody, TAb255 antibody, TAb
256 antibody, TAb258 antibody, TAb259 antibody (P
CT published patent W091-202062), 9G6 antibody [Van de Vijver MJ, et al., New Eng. J. Med., 31 .
9 , 1239 (1988)] etc. are included.

【0018】更に上記抗ヒトリンパ球抗体に属する抗C
D3抗体の具体例としては、UCHT1抗体[抗CD3
ε抗体:マウスIgG1 :Beverley,P.C.L. and Callar
d,R.E., Eur.J.Immunol.,11, 329-334 (1981): インペ
リアル・キャンサー・リサーチファンデーション(Imper
ial Cancer Reseach Foundation,UK)]、OKT3抗体
[抗CD3抗体:マウスIgG2a:Kung,P.C., et al.,
Science, 206, 347-349 (1979)] 等を例示できる。該U
CHT1抗体はTリンパ球の細胞表面に発現したヒトC
D3ε抗原を認識する抗体であることが知られている。
Further, anti-C belonging to the above-mentioned anti-human lymphocyte antibody
Specific examples of the D3 antibody include UCHT1 antibody [anti-CD3
ε antibody: mouse IgG 1 : Beverley, PCL and Callar
d, RE, Eur.J.Immunol., 11 , 329-334 (1981): Imperial Cancer Research Foundation (Imper
ial Cancer Reseach Foundation, UK)], OKT3 antibody [anti-CD3 antibody: mouse IgG 2a : Kung, PC, et al.,
Science, 206 , 347-349 (1979)] and the like. The U
CHT1 antibody is human C expressed on the cell surface of T lymphocytes.
It is known to be an antibody that recognizes the D3ε antigen.

【0019】之等各抗体はそれぞれのモノクローナル抗
体産生ハイブリドーマを新たに作製するか又は既知のモ
ノクローナル抗体産生ハイブリドーマを利用して、公知
の方法[例えばKoehler,G. and C.Milstein, Nature, 2
56, 495 (1975)等参照]に従い製造することができる。
その一般的方法としては、まずモノクローナル抗体産生
ハイブリドーマを適当な培養用培地、例えば10%FC
S(牛胎児血清)を含むRPMI−1640培地(日水
製薬社製)等を用いて培養する。この培養ハイブリドー
マをPBS(リン酸緩衝食塩水)又はFCSを含まない
RPMI−1640培地に懸濁させ、該懸濁液を、予め
プリスタン(2,6,10,14−テトラメチルペンタ
デカン)を腹腔内投与して飼育されたBALB/cヌー
ドマウスに腹腔内投与する。上記投与の約7〜14日後
にマウス腹部が膨れてきた時点で、該マウスの腹腔より
腹水を抜き、次いで該腹水を遠心分離して細胞を除き腹
水を得る。この腹水から一般的方法、例えばプロティン
A[Forsgren,A. and J.Sjoequist, J.Immmunol.,97, 82
2 (1966)] を用いたアフィニティークロマトグラフィー
等を行なうことにより、目的のモノクローナル抗体を採
取、精製することができる。上記精製は、より詳しくは
1ml当りの腹水に2〜4mlの結合バッファー(1.5M
グリシン、3M NaCl、pH8.9)を加えた混合
液を、例えばプロティンAセファロイン(チッソ株式会
社製)カラムにアプライし、抗体をカラムに吸着させた
後、0.1Mクエン酸ナトリウム緩衝液(pH3.0〜
5.0)で溶出させることにより実施できる。
For each antibody, a monoclonal antibody-producing hybridoma is newly prepared, or a known monoclonal antibody-producing hybridoma is used, and a known method [eg, Koehler, G. and C. Milstein, Nature, 2
56 , 495 (1975), etc.].
As a general method, first, a hybridoma producing a monoclonal antibody is first treated with an appropriate culture medium, for example, 10% FC.
Culture is performed using RPMI-1640 medium (Nissui Pharmaceutical Co., Ltd.) containing S (fetal bovine serum). This cultured hybridoma was suspended in PBS (phosphate buffered saline) or RPMI-1640 medium containing no FCS, and the suspension was preliminarily injected with pristane (2,6,10,14-tetramethylpentadecane) intraperitoneally. It is intraperitoneally administered to the administered BALB / c nude mouse. Approximately 7 to 14 days after the above administration, when the abdomen of the mouse swells, ascites is drained from the abdominal cavity of the mouse, and then the ascites is centrifuged to remove cells to obtain ascites. From this ascites, a general method such as Protein A [Forsgren, A. and J. Sjoequist, J. Immunol., 97 , 82
2 (1966)], the target monoclonal antibody can be collected and purified by performing affinity chromatography or the like. More specifically, the above purification is carried out by adding 2 to 4 ml of binding buffer (1.5 M to 1 ml of ascites fluid).
Glycine, 3M NaCl, pH 8.9) was added to the mixture, for example, Protein A Cephaloin (manufactured by Chisso Corporation) column, and the antibody was adsorbed to the column, followed by 0.1M sodium citrate buffer ( pH 3.0 ~
It can be carried out by eluting with 5.0).

【0020】上記の如くして得られるモノクロナール抗
体からのF(ab′)2 の調製は、既に報告されている
方法[例えばParham,P., J.Immunol., 131, 2895-2902
(1983)]に準じて、ペプシン消化させることにより行な
うことができる。この調製の具体的一例としては、以下
の方法を例示することができる。即ち、まず5〜10mg
の抗体を含む0.1Mクエン酸ナトリウム溶液(pH
4.1〜4.5)に、抗体の1/10〜1/100倍重
量のペプシン(シグマ社製)を加えて混合し、37℃の
恒温槽で1〜8時間消化反応を行なう。該反応液に1M
トリス溶液を加えてpHを8.0として反応を停止さ
せ、20mMトリス塩酸、2mM EDTA及び150
mM NaClからなる溶液(pH8.0:以後「TE
S緩衝液」という)で平衡化したTSK−SW−G30
00(トーソー社製)カラムを用いてゲル濾過を行な
い、最初のピークをプールする。次に、この反応液に2
〜4倍容量の結合バッファーを加えてプロティンAカラ
ムにアプライして素通り画分を回収し、ペプシン未消化
の完全な抗体分子をカラムに吸着させて除去する。かく
して得られた素通り画分を、分子量10000の膜を用
いて眼外濾過を行なうことにより、濃縮F(ab′)2
画分が得られる。抗体により収量は異なるが、通常10
mgの抗体から約4〜6mg程度のF(ab′)2 が得られ
る。
The preparation of F (ab ') 2 from the monoclonal antibody obtained as described above can be carried out by a method already reported [eg Parham, P., J. Immunol., 131 , 2895-2902].
(1983)], followed by digestion with pepsin. The following method can be illustrated as a specific example of this preparation. That is, 5-10 mg
0.1M sodium citrate solution (pH)
4.1 to 4.5), 1/10 to 1/100 times the weight of the antibody by weight of pepsin (manufactured by Sigma) is added and mixed, and the digestion reaction is performed in a 37 ° C. thermostat for 1 to 8 hours. 1M in the reaction solution
The reaction was stopped by adding Tris solution to pH 8.0 and adding 20 mM Tris hydrochloric acid, 2 mM EDTA and 150 mM.
Solution consisting of mM NaCl (pH 8.0:
"S buffer") equilibrated with TSK-SW-G30
Gel filtration is performed using a 00 (Tosoh) column, and the first peak is pooled. Then add 2 to this reaction mixture.
Approximately 4 times the volume of binding buffer is added and applied to a Protein A column to collect a flow-through fraction, and complete antibody molecules undigested with pepsin are adsorbed to the column and removed. The flow-through fraction thus obtained is subjected to extraocular filtration using a membrane having a molecular weight of 10,000 to obtain concentrated F (ab ') 2
Fractions are obtained. Yield varies depending on the antibody, but usually 10
About 4 to 6 mg of F (ab ′) 2 is obtained from mg of antibody.

【0021】上記の如くして得られるF(ab′)2
ラグメントを用いて本発明抗体、即ち上記標的細胞に対
する抗体のFab′[モノマー]とエフェクター細胞に
対する抗体のFab′[モノマー]とが結合したBFA
は、例えば新田らの方法[Nitta,T., et al., Eur.J.Im
munol., 19, 1437-1441 (1989)] に従って作製すること
ができる。
The F (ab ') 2 fragment obtained as described above is used to bind the antibody of the present invention, that is, Fab' [monomer] of the antibody to the target cells and Fab '[monomer] of the antibody to effector cells. BFA
For example, the method of Nitta et al. [Nitta, T., et al., Eur.J.Im
munol., 19 , 1437-1441 (1989)].

【0022】上記方法は、まず標的細胞の表面抗原を認
識する抗体、例えば抗c−erbB−2抗体のF(a
b′)2 フラグメントを含むTES緩衝液に、最終濃度
が1〜2mMとなるようにDTT(和光純薬社製)水溶
液を加えて、pH7.5で室温にて30分間反応を行な
った後、最終濃度が5mMとなるようにDTNB(和光
純薬社製)水溶液を加えて、更に室温で30分間反応を
行なわせ、この反応液をTES緩衝液で平衡化したセフ
ァデックスG25カラム(ファルマシァ社製)を用いて
ゲル濾過を行なうことにより実施され、かくして標的細
胞に対する抗体Fab′−SNBを収得できる。別に、
エフェクター細胞に結合し且つシグナル伝達を行なう抗
体、例えば抗CD3抗体のF(ab′)2 フラグメント
を含むTES緩衝液に、最終濃度が1〜2mMとなるよ
うにDTT水溶液を加えて、pH7.5で室温にて30
分間反応を行ない、得られる反応液をTES緩衝液で平
衡化したセファデッスクG25カラムを用いてゲル濾過
を行なうことにより、エフェクター細胞に対する抗体F
ab′−SHを収得できる。上記で得られるFab′−
SHを含むTES緩衝液に、先に得られた標的細胞を認
識する抗体のFab′−SNBをほぼ1:1のモル比で
加えて、限外濾過膜で濃縮した後、室温で4時間程度反
応させることにより、標的細胞に対する抗体のFab′
[モノマー]とエフェクター細胞に対する抗体Fab′
[モノマー]とが結合した所望のBFAを収得できる。
In the above method, first, an antibody recognizing the surface antigen of the target cell, for example, F (a) of anti-c-erbB-2 antibody is used.
b ′) To a TES buffer containing 2 fragments, an aqueous solution of DTT (manufactured by Wako Pure Chemical Industries, Ltd.) was added so that the final concentration was 1-2 mM, and the reaction was carried out at room temperature for 30 minutes at pH 7.5. An aqueous solution of DTNB (manufactured by Wako Pure Chemical Industries, Ltd.) was added so that the final concentration was 5 mM, and the reaction was further performed at room temperature for 30 minutes, and the reaction solution was equilibrated with a TES buffer solution (Sephadex G25 column, manufactured by Pharmacia). The antibody Fab'-SNB against the target cells can be obtained by carrying out gel filtration using the above method. Apart from
To a TES buffer containing an antibody that binds to effector cells and performs signal transduction, for example, an F (ab ′) 2 fragment of anti-CD3 antibody, a DTT aqueous solution is added so that the final concentration is 1-2 mM, and the pH is 7.5. At room temperature at 30
The reaction was carried out for a minute, and the resulting reaction solution was subjected to gel filtration using a Sephadex G25 column equilibrated with TES buffer to give antibody F against effector cells.
Ab'-SH can be obtained. Fab′-obtained above
Fab'-SNB, an antibody that recognizes the target cells obtained above, was added to a TES buffer containing SH at a molar ratio of about 1: 1 and concentrated with an ultrafiltration membrane, followed by about 4 hours at room temperature. By reacting, Fab ′ of the antibody against the target cell
[Monomer] and antibody Fab ′ against effector cells
A desired BFA bound with [monomer] can be obtained.

【0023】上記においては、また抗体Fab′−SN
Bの作製をエフェクター細胞に対する抗体につき行な
い、また抗体Fab′−SHの作製を標的細胞に対する
抗体につき行なうこともでき、これによっても所望のB
FAを収得することができる。
In the above, also the antibody Fab'-SN
It is also possible to make B for antibodies to effector cells, and to make antibody Fab'-SH for antibodies to target cells, which also results in desired B
FA can be acquired.

【0024】かくして得られたBFAをTSK−ゲルG
3000SW(トーソー社製)を使用するHPLC(高
速液体クロマトグラフィー)に付すことにより、未反応
のFab′−チオール誘導体(分子量約55kd)を分
離して、分子量約110kdを持つBFAを精製するこ
とができる。
The BFA thus obtained was mixed with TSK-Gel G.
By subjecting it to HPLC (high performance liquid chromatography) using 3000 SW (manufactured by Tosoh Corporation), the unreacted Fab′-thiol derivative (molecular weight: about 55 kd) can be separated, and BFA having a molecular weight of about 110 kd can be purified. it can.

【0025】非還元条件下で、0.1%SDS(ドデシ
ル硫酸ナトリウム:シグマ社製)の存在下にて、7.5
%PAGE(ポリアクリルアミドゲル電気泳動)分析を
行なうことにより、上記で精製されたBFAが予想され
た分子量を持っていることの確認を行なうことができ
る。
7.5, under non-reducing conditions, in the presence of 0.1% SDS (sodium dodecyl sulfate: Sigma).
By performing a% PAGE (polyacrylamide gel electrophoresis) analysis, it can be confirmed that the BFA purified above has the expected molecular weight.

【0026】また、上記方法に従って標的細胞に対する
抗体のF(ab′)n[ポリマー]とエフェクター細胞
に対する抗体F(ab′)n[ポリマー]とが結合した
BFAを作製することが可能である。該F(ab′)n
[ポリマー]とF(ab′)n[ポリマー]とが結合し
たBFAを作製する方法については、本発明者らが先に
出願した複合抗体の製造法に記載されている(特願平3
−229835号参照)。該方法に従って標的細胞に対
する腕を複数とする(F(ab′)n[ポリマー]利
用)とすることによって、得られるBFAは、その標的
細胞に対する結合性がより高められ、これによって本発
明所望の治療効果が一層増強され得ると考えられる。
Further, it is possible to prepare a BFA in which the antibody F (ab ') n [polymer] for the target cells and the antibody F (ab') n [polymer] for the effector cells are bound according to the above method. The F (ab ') n
A method for producing a BFA in which [polymer] and F (ab ′) n [polymer] are bound is described in the method for producing a composite antibody previously filed by the present inventors (Japanese Patent Application No. Hei.
-229835). By using a plurality of arms for a target cell (using F (ab ′) n [polymer]) according to the method, the BFA obtained has a higher binding property to the target cell, which is desirable for the present invention. It is considered that the therapeutic effect can be further enhanced.

【0027】上記如くして得られる本発明のBFAの利
用によれば、CTL等のエフェクター細胞の標的細胞に
対する傷害活性を高めることができる。例えば、抗c−
erbB−2抗体と抗CD3抗体とを用いて作製した本
発明BFAを用いれば、c−erbB−2遺伝子産物を
細胞表面に発現している癌細胞を効果的に傷害乃至壊死
させることができる。之等の事実は、後記する試験例に
示すように、既に報告された試験方法[Staerz,U.D. J.
W.Yewdell and M.J.Bevan, Eur.J.Immunol., 17, 571-5
74 (1987) ; Nitta,T., et al., Eur.J.Immunol.,19, 1
437-1441 (1989)] に従う試験により確認できる。
By using the BFA of the present invention obtained as described above, the toxic activity of effector cells such as CTL against target cells can be enhanced. For example, anti-c-
By using the BFA of the present invention prepared by using the erbB-2 antibody and the anti-CD3 antibody, cancer cells expressing the c-erbB-2 gene product on the cell surface can be effectively injured or necrotic. These facts are based on the already reported test method [Staerz, UDJ, as shown in the test example below.
W. Yewdell and MJ Bevan, Eur. J. Immunol., 17 , 571-5
74 (1987); Nitta, T., Et al., Eur.J.Immunol., 19 , 1
437-1441 (1989)].

【0028】尚、上記においては、エフェクター細胞と
して組織適合性抗原型の一致したリンパ球を用いねばな
らないという拘束性もなく、例えばクローン化したCT
L等を用いることができ、また健常人の末梢血リンパ球
(PBL:peripheral bloodlymphocyle) を用いてもよ
く、更にクーロン化CTL乃至PBLをインターロイキ
ンー2等の存在下で3日以上培養してLAK(lymphokin
e activated killer cells) を誘導して用いてもよい。
健常人の末梢血リンパ球の調製は、比重分離法に従って
以下の如くして行なうことができる。即ち、まずヘパリ
ン採血した健常人の血液10mlに2〜3倍重量のPBS
緩衝液を加えて混合する。別に、この希釈血液の半量の
比重分離液(例えばフィコールパック:ファルマシア社
製を用いることができる)を加えた遠沈管に希釈血液の
全量を重層した後、400×g、室温で30〜40分間
密度匂配遠心分離を行ない、遠心後に遠沈管の血漿と比
重分離液との間にできる白濁層を吸い取り、10%FC
S含有RPMI−1640培地により洗浄する。かくし
て末梢血リンパ球1〜2×107 個を得ることができ
る。またLAKは得られたPBLを106 個/mlとなる
ように10%FCS含有RPMI−1640培地に懸濁
し、培地1ml当り10〜1000単位のインターロイキ
ンー2を加えて培養することにより誘導することができ
る。
In the above, there is no restriction that lymphocytes having a histocompatibility-matching antigen type must be used as effector cells. For example, cloned CT
L, etc. may be used, or peripheral blood lymphocytes (PBL) of a healthy person may be used, and further, clonal CTL or PBL is cultured for 3 days or more in the presence of interleukin-2. LAK (lymphokin
It may be used by inducing e activated killer cells).
Preparation of peripheral blood lymphocytes of a healthy person can be performed as follows according to the specific gravity separation method. That is, 10 ml of blood of a healthy person who had heparinized blood was first added to 2-3 times the weight of PBS.
Add buffer and mix. Separately, after overlaying the whole volume of the diluted blood on a centrifuge tube to which a specific gravity separation liquid (for example, Ficoll Pack: manufactured by Pharmacia Co., Ltd.) of the diluted blood is added, 400 × g, at room temperature for 30 to 40 minutes. Density gradient centrifugation is performed, and after centrifugation, the white turbid layer formed between the plasma in the centrifuge tube and the specific gravity separation liquid is absorbed, and 10% FC
Wash with S-containing RPMI-1640 medium. Thus, 1 to 2 × 10 7 peripheral blood lymphocytes can be obtained. LAK is induced by suspending the obtained PBLs in RPMI-1640 medium containing 10% FCS at 10 6 cells / ml and culturing by adding 10 to 1000 units of interleukin-2 per 1 ml of the medium. be able to.

【0029】また放射性同位元素51Crを含むクロム酸
ナトリウム(Na2 51CrO4 )による標的細胞の標識
は、まず標的細胞のペレット(1〜2×106 細胞)
に、3.7MBq/mlの上記クロム酸ナトリウム(NE
N社製、第一化学製品社販売)を加え、37℃、5%C
2 存在下で60〜90分間培養を行ない、次いで培養
後に10%FCS含有RPMI−1640培地を用いて
十分に洗浄し、過剰の未反応のクロミウム(51Cr)を
除くことにより行なうことができる。かくしてクロミウ
ム(51Cr)標識標的細胞を収得できる。
The labeling of the target cells with sodium chromate (Na 2 51 CrO 4 ) containing the radioisotope 51 Cr is carried out by first pelleting the target cells (1-2 × 10 6 cells).
In addition, 3.7 MBq / ml of the above sodium chromate (NE
N, manufactured by Daiichi Pure Chemicals Co., Ltd.) and added at 37 ° C, 5% C
It can be carried out by culturing in the presence of O 2 for 60 to 90 minutes, followed by thorough washing with 10% FCS-containing RPMI-1640 medium to remove excess unreacted chromium ( 51 Cr). .. Thus, target cells containing chromium ( 51 Cr) can be obtained.

【0030】上記クロミウム(51Cr)標識標的細胞、
エフェクター細胞及びハイブリッド抗体を用いた細胞傷
害試験は、より詳しくは、まず適当な濃度(通常0.0
1〜3μg/ml)に希釈したBFAの100μlとエフ
ェクター細胞106 /100μlとを混合し、室温で3
0分間反応させ、反応液を10%FCS含有RPMI−
1640を用いて洗浄し、抗体の結合したエフェクター
細胞を沈殿として回収する(この操作で未反応のBFA
を除くことができる)。次に、各濃度の抗体が結合した
エフェクター細胞のそれぞれ150μl(エフェクター
細胞数:2×105 )と、標的細胞の50μl(細胞
数:104 )とを、それぞれ96穴のU底培養プレート
(コーニング社製)の各ウェルに添加し、37℃、5%
CO2 存在下で培養して、標的細胞の特異傷害反応を行
なう。反応後に各ウェルの放射能活性をカウントする。
BFAを介して、エフェクター抗体と標的細胞とを結合
させた時に起こる標的細胞の特異的傷害の程度(%)
は、次の計算式(1)に従って求めることができる。
Target cells labeled with chromium ( 51 Cr),
More specifically, the cytotoxicity test using the effector cells and the hybrid antibody should be conducted at an appropriate concentration (usually 0.0
1~3μg / ml) and 100 [mu] l effector cells 10 6/100 [mu] l of diluted BFA were mixed, at room temperature for 3
The reaction was allowed to proceed for 0 minutes, and the reaction solution was added with RPMI containing 10% FCS
Wash with 1640 and collect the antibody-bound effector cells as a precipitate (unreacted BFA by this procedure).
Can be excluded). Next, 150 μl of each effector cell (the number of effector cells: 2 × 10 5 ) to which the antibody of each concentration was bound and 50 μl of the target cell (the number of cells: 10 4 ) were respectively added to a 96-well U-bottom culture plate ( Corning), at 37 ℃, 5%
The cells are cultured in the presence of CO 2 to carry out a specific injury reaction of target cells. After the reaction, the radioactivity of each well is counted.
Degree (%) of specific damage of target cells that occurs when effector antibody and target cells are bound via BFA
Can be obtained according to the following calculation formula (1).

【0031】 特異的傷害%=[(a−b)/(c−d)]×100 (1) [aはBFAを介したエフェクター細胞による標的細胞
の特異的51Cr放出を示す実測値を、bは標的細胞のみ
を培養した時のウェル中の放射能活性(自然放出)を、
cは1%NP−40(シグマ社製)溶液を用いて標的細
胞を溶解させた時の反応液中の放射活性(最大放出)を
それぞれ示す。] 上記細胞傷害試験によれば、本発明BFA、特に抗c−
erbB−2抗体のFab′と抗CD3抗体のFab′
とを用いて作製した本発明BFAは、100ng/mlの用
量で約25%の特異的標的細胞傷害が観察され、このこ
とから高い標的細胞傷害能を有することが確認された
(後記する図1−A参照)。これに対して、抗c−er
bB−2抗体のF(ab′)2 フラグメントと抗CD3
抗体のF(ab′)2 フラグメントとを1:1の比率で
単に混合した混合液では、1000ng/mlまで濃度を上
げても有意な細胞傷害性は認められなかった(後記する
図1−B参照)。また、各親のF(ab′)2 フラグメ
ントも1000ng/mlまで濃度を上げても有意な細胞傷
害性は認められなかった。
Specific injury% = [(a−b) / (c−d)] × 100 (1) [a is a measured value showing specific 51 Cr release of target cells by BFA-mediated effector cells, b shows the radioactivity (spontaneous release) in the well when only the target cells were cultured,
c indicates the radioactivity (maximum release) in the reaction solution when the target cells were lysed using a 1% NP-40 (manufactured by Sigma) solution. ] According to the above cytotoxicity test, the BFA of the present invention, particularly anti-c-
Fab ′ of erbB-2 antibody and Fab ′ of anti-CD3 antibody
The BFA of the present invention produced by using and was observed to have a specific target cell cytotoxicity of about 25% at a dose of 100 ng / ml, which confirmed that it has a high target cell cytotoxicity (Fig. 1 described later). -See A). On the other hand, anti-c-er
F (ab ') 2 fragment of bB-2 antibody and anti-CD3
No significant cytotoxicity was observed even when the concentration was raised to 1000 ng / ml in the mixed solution in which the antibody F (ab ') 2 fragment was simply mixed at a ratio of 1: 1 (Fig. 1-B described later). reference). Also, the F (ab ') 2 fragment of each parent did not show significant cytotoxicity even when the concentration was increased to 1000 ng / ml.

【0032】このように、標的細胞に対する抗体のFa
b′フラグメントとエフェクター細胞に対する抗体のF
ab′フラグメントとが結合した本発明のBFAは、こ
れが10ng/mlという極微量から標的細胞の特異的細胞
傷害活性を示す(後記する図1−A参照)ことから、c
−erbB−2遺伝子産物を細胞表面に発現している癌
細胞による悪性腫瘍の治療剤、特に乳癌治療剤として有
用である。
Thus, the Fa of the antibody against the target cell is
b'fragment and F of antibody to effector cells
The BFA of the present invention bound to the ab ′ fragment shows a specific cytotoxic activity of target cells from a very small amount of 10 ng / ml (see FIG. 1-A described later).
-It is useful as a therapeutic agent for malignant tumors caused by cancer cells expressing the erbB-2 gene product on the cell surface, particularly as a therapeutic agent for breast cancer.

【0033】また本発明BFAは、その標的細胞に対す
る高い結合力維持性を応用して、上記治療剤の他にも、
例えば従来のモノクロナール抗体に代わって、各種の診
断薬として利用できる。
In addition to the above-mentioned therapeutic agents, the BFA of the present invention applies the ability to maintain high binding power to target cells to
For example, it can be used as various diagnostic agents in place of conventional monoclonal antibodies.

【0034】以上のように、本発明抗体は悪性腫瘍治療
剤として有効であり、従って本発明にかかる悪性腫瘍治
療剤をも提供するものである。この本発明悪性腫瘍治療
剤は、上記BFAをその必須成分として含有することを
基本として、他は通常の製剤技術乃至この種BFAを用
いる免疫療法等で慣用されている技術手段に従い調製す
ることができる。即ち、該治療剤は通常本発明抗体と共
に適当な医薬製剤担体を配合して製剤組成物の形態に調
製される。用いられる担体としては使用形態に応じた製
剤の調製に通常慣用される各種のもの、例えば充填剤、
増量剤、結合剤、表面活性剤、緩衝液、安定化剤等の賦
形剤乃至希釈剤のいずれでもよく、調製される製剤形態
は、これが本発明治療剤成分を効果的に含有する状態で
あればよく、例えば錠剤、粉末剤等の固剤であってもよ
いが、通常液剤、懸濁剤、乳剤等の注射剤形態とするの
がよい。またこれは使用前に適当な担体の添加により液
状となし得る乾燥品の形態とすることもでき、いずれの
形態も常法に従い調製できる。また各形態の製剤はその
形態に応じて適当な投与経路、例えば注射剤形態の製剤
では静脈内、筋肉内、皮下、皮内、腹腔内投与され、固
型形態の製剤は経口乃至経腸投与される。勿論、予め体
外でリンパ球等に本発明抗体を反応させてから、それを
静脈内、腹腔内、感染病巣等に投与することも有用な方
法である。
As described above, the antibody of the present invention is effective as a therapeutic agent for malignant tumor, and therefore also provides a therapeutic agent for malignant tumor according to the present invention. This therapeutic agent for malignant tumors of the present invention is based on the fact that it contains the above-mentioned BFA as an essential component thereof, and can be prepared according to other conventional formulation techniques or technical means commonly used in immunotherapy using this type of BFA. it can. That is, the therapeutic agent is usually prepared in the form of a pharmaceutical composition by mixing a suitable pharmaceutical carrier with the antibody of the present invention. As the carrier to be used, various kinds of carriers usually used for the preparation of a formulation depending on the use form, for example, a filler,
Any of excipients or diluents such as a bulking agent, a binder, a surface active agent, a buffer solution, a stabilizer, etc. may be used, and the preparation form to be prepared is such that it effectively contains the therapeutic agent component of the present invention. It may be a solid preparation such as tablets and powders, but it is usually in the form of injections such as solutions, suspensions and emulsions. In addition, this may be in the form of a dried product which can be made into a liquid form by adding an appropriate carrier before use, and any form can be prepared by a conventional method. In addition, the preparations of each form are administered by an appropriate administration route depending on the form, for example, intravenous, intramuscular, subcutaneous, intradermal, and intraperitoneal injections are given for preparations in the form of injections, and oral or enteral administrations for preparations in the solid form To be done. Of course, it is also a useful method to react the antibody of the present invention with lymphocytes or the like in vitro in advance and then administer the antibody to intravenous, intraperitoneal, infectious lesion or the like.

【0035】本発明治療剤の投与量は該製剤の投与方
法、投与形態、使用目的、適用患者等に応じて適宜決定
され一定ではないが、一般には本発明抗体の量が約0.
00001〜80重量%程度含有されるものとするのが
よく、この製剤は一日成人一人当り約0.01μg〜1
0mg程度の範囲で適用されるのが好ましい。かくして本
発明治療剤の投与によれば、これを投与された患者の体
内においてリンパ球の細胞傷害性が増強され、かくして
所望の治療効果が奏される。
The dose of the therapeutic agent of the present invention is appropriately determined depending on the administration method, dosage form, purpose of use, patient to be applied, etc. of the preparation and is not constant, but generally the amount of the antibody of the present invention is about 0.
It is preferable to contain about 0,0001 to 80% by weight, and this preparation contains about 0.01 µg to 1 per adult per day.
It is preferably applied in the range of about 0 mg. Thus, the administration of the therapeutic agent of the present invention enhances the cytotoxicity of lymphocytes in the body of the patient to whom the therapeutic agent is administered, thus exerting the desired therapeutic effect.

【0036】また、本発明治療剤を用いた免疫療法の
内、養子免疫療法、即ち一度患者生体より採取したリン
パ球を何等かの手段で活性化した後、再度患者生体内に
戻して治療を行なう方法は、例えば次の如くして実施で
きる。即ち、患者の末梢血約100mlからリンパ球を分
離し、IL−2約100U/mlを添加した無血清培地で
約1週間程度培養し、得られるLAK細胞(リンホカイ
ン活性化キラー細胞)約1×108 個を本発明のBFA
約100〜1000μg、好ましくは約100μg前後
と共に患者体内に注入することにより実施され、この処
置は患者の病状、年齢等に応じて通常週に数回に分けて
行なうことができる。
In the immunotherapy using the therapeutic agent of the present invention, adoptive immunotherapy, that is, lymphocytes once collected from the patient's body are activated by some means and then returned to the patient's body for treatment. The method to be performed can be implemented as follows, for example. That is, lymphocytes were separated from about 100 ml of peripheral blood of a patient and cultured in a serum-free medium supplemented with about 100 U / ml of IL-2 for about 1 week to obtain about 1 × of LAK cells (lymphokine-activated killer cells) obtained. 10 8 BFA of the present invention
It is carried out by injecting into the patient's body together with about 100 to 1000 μg, preferably about 100 μg, and this treatment can be usually divided into several times a week depending on the medical condition, age and the like of the patient.

【0037】[0037]

【発明の効果】本発明抗体は、抗原(標的細胞及びエフ
ェクター細胞)に対する特異的結合力が強く、その利用
によりCTL等のエフェクター細胞の有する標的細胞に
対する細胞傷害活性を活性化乃至増強させ得る特徴を有
している。
INDUSTRIAL APPLICABILITY The antibody of the present invention has a strong specific binding power to antigens (target cells and effector cells), and its use can activate or enhance the cytotoxic activity of effector cells such as CTL against target cells. have.

【0038】[0038]

【実施例】以下、本発明BFA及びこれを用いた悪性腫
瘍治療剤につきより詳細に説明するため、実施例を挙げ
る本発明はこれに限定されない。
EXAMPLES The BFA of the present invention and a therapeutic agent for malignant tumor using the same will be described in more detail below, but the present invention is not limited thereto.

【0039】[0039]

【実施例1】BFAの製造 1) UCHT1腹水の調製 UCHT1細胞[抗CD3ε抗体:マウスIgG1 :Bev
erley,P.C.L. and Callard,R.E., Eur.J.Immunol.,11,
329-334 (1981): インペリアル・キャンサー・リサーチ
ファンデーション(Imperial Cancer Reseach Foundatio
n,UK) より入手]を10%FCS(フロー社製)を含む
RPMI−1640培地(日水製薬社製)を用いて、3
7℃、5%CO2 存在下で培養してハイブリドーマを得
た。
Example 1 Production of BFA 1) Preparation of UCHT1 ascites UCHT1 cells [anti-CD3ε antibody: mouse IgG 1 : Bev
erley, PCL and Callard, RE, Eur.J.Immunol., 11 ,
329-334 (1981): Imperial Cancer Research Foundation
n, UK)] using RPMI-1640 medium (manufactured by Nissui Pharmaceutical Co., Ltd.) containing 10% FCS (manufactured by Flow Co.).
Hybridomas were obtained by culturing at 7 ° C. in the presence of 5% CO 2 .

【0040】別にBALB/c(日本チャールズリバー
社製)の腹腔内に、一匹当り0.5mlのプリスタン
(2,6,10,14−テトラメチルペンタデカン、和
光純薬社製)を投与して1〜2週間飼育を行なった。
Separately, 0.5 ml of pristane (2,6,10,14-tetramethylpentadecane, manufactured by Wako Pure Chemical Industries, Ltd.) was administered intraperitoneally to BALB / c (produced by Charles River Japan). The animals were raised for 1-2 weeks.

【0041】次に先に得られたUCHT1細胞を、PB
Sに懸濁し、その1×107 細胞/0.5mlずつを各
マウス腹腔内に投与した。上記投与の2〜3週間目にマ
ウスの腹腔が膨れてきた時点で、マウスの腹腔より腹水
を抜き、該腹水を日立遠心機05PR−22(日立製作
所社製)を用いて10000rpm、30分間、4℃で
遠心分離し、UCHT1抗体を含む腹水上清を得た。
Next, the UCHT1 cells obtained above were treated with PB
The cells were suspended in S, and 1 × 10 7 cells / 0.5 ml was intraperitoneally administered to each mouse. At the time when the abdominal cavity of the mouse was swollen 2 to 3 weeks after the above administration, ascites was drained from the abdominal cavity of the mouse, and the ascites was 10,000 rpm using Hitachi Centrifuge 05PR-22 (manufactured by Hitachi, Ltd.) for 30 minutes, After centrifugation at 4 ° C., ascites supernatant containing UCHT1 antibody was obtained.

【0042】2) UCHT1−F(ab′)2 フラグ
メントの調製 上記1)で得られたUCHT1腹水上清に2倍容量の
1.5Mグリシン及び3M NaClからなる緩衝液
(pH8.9)を加えて混合し、混合液を、固定化プロ
テインA(Immobilized-Protein A (Repligen 社))カ
ラムにアプライし、UCHT1抗体をカラムに吸着させ
た。充分に1.5Mグリシン及び3M NaClからな
る緩衝液(pH8.9)で洗浄した後、吸着しているU
CHT1抗体を、0.1Mクエン酸ナトリウム緩衝液
(pH3.0〜5.0)で溶出させて、IgG溶液を得
た。
2) Preparation of UCHT1-F (ab ') 2 fragment To the UCHT1 ascites supernatant obtained in 1) above was added a buffer (pH 8.9) consisting of 2 volumes of 1.5 M glycine and 3 M NaCl. And mixed, and the mixture was applied to an immobilized protein A (Immobilized-Protein A (Repligen)) column to adsorb the UCHT1 antibody to the column. After sufficiently washing with a buffer solution (pH 8.9) consisting of 1.5 M glycine and 3 M NaCl, the adsorbed U
The CHT1 antibody was eluted with 0.1 M sodium citrate buffer (pH 3.0 to 5.0) to obtain an IgG solution.

【0043】上記で得られたIgG溶液2ml(IgG
10mg)を、0.1Mクエン酸ナトリウム緩衝液(p
H4.1)に対して4℃で一晩透析した。透析液を回収
後、これに抗体の1/10〜1/25量(w/w)のペ
プシン(シグマ社製) を加えて混合し、37℃の恒温槽
で6〜8時間消化反応を行なった。反応後、反応液に1
Mトリス溶液1/2容量を加えて反応を停止させ、15
0Mm NaCl含有20mMリン酸緩衝液(pH7.
0)で平衡化したTSK−G3000SW(トーソー社
製)カラム(21.5mmID×60cm)を用いてゲ
ル濾過を行ない、F(ab′)2 画分をプールした。
2 ml of the IgG solution obtained above (IgG
10 mg) in 0.1 M sodium citrate buffer (p
It was dialyzed against H4.1) overnight at 4 ° C. After collecting the dialysate, 1/10 to 1/25 amount (w / w) of the antibody, pepsin (manufactured by Sigma), was added and mixed, and the digestion reaction was performed in a thermostat at 37 ° C for 6 to 8 hours. It was After the reaction, add 1 to the reaction mixture.
Stop the reaction by adding 1/2 volume of M Tris solution,
20 mM phosphate buffer containing 0 Mm NaCl (pH 7.
Gel filtration was performed using a TSK-G3000SW (manufactured by Tosoh Corporation) column (21.5 mm ID × 60 cm) equilibrated with 0) to pool F (ab ′) 2 fractions.

【0044】次に、この分取した画分に1.5Mグリシ
ン及び3M NaClからなる緩衝液(pH8.9)を
加え、その混合液をプロティンAカラムにアプライして
素通り画分を回収し、カラムにペプシン未消化のUHC
L1抗体を吸着させて除去した。かくして得られた素通
りの画部を、分子量10000の膜を用いて限外濾過
(アミコン社製、ダイアフローメンブレンYM10、4
3mm膜を使用)を行ない、バッファーを20mMトリ
ス塩酸、2mM EDTA及び150mM NaClか
らなる溶液(pH8.0:以後「TES緩衝液」とい
う)に交換して、UCHT1のF(ab′)2 フラグメ
ントの4mgを得た。
Next, a buffer solution (pH 8.9) consisting of 1.5 M glycine and 3 M NaCl was added to the collected fractions, and the mixture was applied to a protein A column to collect a flow-through fraction, UHC undigested with pepsin on the column
The L1 antibody was adsorbed and removed. The thus-obtained flow-through section was subjected to ultrafiltration using a membrane having a molecular weight of 10,000 (Amicon, Diaflow membrane YM10, 4).
3 mm membrane was used) and the buffer was exchanged with a solution consisting of 20 mM Tris-hydrochloric acid, 2 mM EDTA and 150 mM NaCl (pH 8.0: hereinafter referred to as “TES buffer”), and the F (ab ′) 2 fragment of UCHT1 was replaced. 4 mg was obtained.

【0045】3) GFD−OA−p185−1腹水の
調製 まずヒトc−erbB−2遺伝子産物を認識する抗体と
してのGFD−OA−p185−1抗体を以下のように
調製した。即ち、GFD−OA−p185−1抗体を産
生するハイブリドーマGFD−OA−p185−1(微
工研菌寄第12206号)を上記1)と同様にして培養
した後、該細胞をマウスの腹腔内に投与し、該腹水より
GFD−OA−p185−1抗体を含む腹水上清を得
た。
3) Preparation of GFD-OA-p185-1 ascites First, the GFD-OA-p185-1 antibody as an antibody recognizing the human c-erbB-2 gene product was prepared as follows. That is, the hybridoma GFD-OA-p185-1 (GFD-OA-p185-1 antibody-producing hybridoma GFD-OA-p185-1) was cultured in the same manner as in 1) above, and then the cells were intraperitoneally injected into mice. The ascites supernatant containing the GFD-OA-p185-1 antibody was obtained from the ascites.

【0046】4) GFD−OA−p185−1−F
(ab′)2 フラグメントの調製 上記2)と同様にして3)で得られたGFD−OA−p
185−1腹水上清に2倍容量の1.5Mグリシン及び
3M NaClからなる緩衝液(pH8.9)を加えた
混合液を、プロティンAカラムにアプライし、GFD−
OA−p185−1抗体をカラム吸着させた。同バッフ
ァーで洗浄した後、吸着しているGFD−OA−p18
5−1抗体を、0.1Mクエン酸ナトリウム緩衝液(p
H3.0〜5.0)で溶出させて、IgGを含む溶液を
得た。
4) GFD-OA-p185-1-F
Preparation of (ab ') 2 fragment GFD-OA-p obtained in 3) in the same manner as in 2) above
The 185-1 ascites supernatant was mixed with 2 volumes of a buffer (pH 8.9) consisting of 1.5 M glycine and 3 M NaCl, and the mixture was applied to a protein A column to give GFD-
The OA-p185-1 antibody was adsorbed on the column. After washing with the same buffer, adsorbed GFD-OA-p18
5-1 antibody was added to 0.1 M sodium citrate buffer (p
H3.0-5.0) was eluted to obtain a solution containing IgG.

【0047】上記で得られたIgGを含む溶液2ml
(IgG10mg)を、0.1Mクエン酸ナトリウム緩
衝液(pH4.1)に対して4℃で一晩透析した。透析
液を回収後、これに抗体の1/10〜1/25量(w/
w)のペプシンを加えて混合し、37℃の恒温槽で4〜
8時間消化反応を行なった。反応後、反応液に1Mトリ
ス溶液1/2容量を加えて反応を停止させ、150Mm
NaCl含有20mMリン酸緩衝液(pH7.0)で
平衡化したTSK−G3000SWカラムを用いてゲル
濾過を行ない、F(ab′)2 画分をプールした。
2 ml of the solution containing IgG obtained above
(IgG 10 mg) was dialyzed against 0.1 M sodium citrate buffer (pH 4.1) at 4 ° C. overnight. After collecting the dialysate, 1/10 to 1/25 the amount of the antibody (w /
w) pepsin was added and mixed, and the mixture was mixed in a thermostat at 37 ° C for 4 to
The digestion reaction was performed for 8 hours. After the reaction, 1/2 volume of 1 M Tris solution was added to the reaction solution to stop the reaction, and 150 Mm
Gel filtration was performed using a TSK-G3000SW column equilibrated with 20 mM phosphate buffer (pH 7.0) containing NaCl, and F (ab ′) 2 fractions were pooled.

【0048】次に、この画分に1.5Mグリシン及び3
M NaClからなる緩衝液(pH8.9)を加え、そ
の混合液をプロティンAカラムにアプライして素通り画
分を回収し、カラムにペプシン未消化のGFD−OA−
p185−1抗体を吸着させて除去した。かくして得ら
れた素通りの画部を、分子量10000の膜を用いて限
外濾過を行ない、バッファーをTES緩衝液に交換し
て、GFD−OA−p185−1のF(ab′)2 フラ
グメント5mgを得た。
Next, 1.5 M glycine and 3 were added to this fraction.
A buffer solution (pH 8.9) consisting of M NaCl was added, the mixture solution was applied to a protein A column to collect a flow-through fraction, and pepsin-undigested GFD-OA- was applied to the column.
The p185-1 antibody was adsorbed and removed. The flow-through fraction thus obtained was subjected to ultrafiltration using a membrane having a molecular weight of 10,000, and the buffer was exchanged with a TES buffer solution to obtain 5 mg of F (ab ') 2 fragment of GFD-OA-p185-1. Obtained.

【0049】5) GFD−OA−p185−1Fa
b′とUCHT1−Fab′とのBFAの作製 上記4)で調製したGFD−OA−p185−1抗体の
F(ab′)2 フラグメント3mgを含む1.0mlT
ES緩衝液に、20μlの100mMジチオスレイトー
ル(DTT;和光純薬社製)水溶液を加えて、pH7.
5、室温にて30分間反応を行なった後、引き続き10
mM DTNB水溶液(和光純薬社製)1〜2倍量を加
えて、更に室温で30分間反応を行なわせた。この反応
液を窒素ガスで置換したTES緩衝液で平衡化したセフ
ァデックスG25カラムを用いてゲル濾過し、最初のピ
ークをプールして、GFD−OA−p185−1−Fa
b′の混合ニトロフェニルジスルフィド誘導体(GFD
−OA−p185−1−Fab′−SNB)を収得し
た。
5) GFD-OA-p185-1Fa
Preparation of BFA of b ′ and UCHT1-Fab ′ 1.0 ml T containing 3 mg of F (ab ′) 2 fragment of GFD-OA-p185-1 antibody prepared in 4) above
20 μl of 100 mM dithiothreitol (DTT; manufactured by Wako Pure Chemical Industries) aqueous solution was added to the ES buffer to adjust the pH to 7.
5. After reacting for 30 minutes at room temperature, continue to 10
A 1-2 times amount of mM DTNB aqueous solution (manufactured by Wako Pure Chemical Industries, Ltd.) was added, and the reaction was further performed at room temperature for 30 minutes. The reaction solution was subjected to gel filtration using a Sephadex G25 column equilibrated with a TES buffer solution substituted with nitrogen gas, and the first peak was pooled to give GFD-OA-p185-1-Fa.
b'mixed nitrophenyl disulfide derivative (GFD
-OA-p185-1-Fab'-SNB) was obtained.

【0050】別に、上記2)で調製したUCHT1抗体
のF(ab′)2 フラグメント3mgを含む1.0ml
TES緩衝液に、20μlのDTT水溶液を加えて、p
H7.5で室温にて30分間反応を行なった後、得られ
た反応液を窒素ガスで置換したTES緩衝液で平衡化し
たセファデックスG25カラムを用いてゲル濾過し、最
初のピークをプールして、UCHT1−Fab′−SH
を収得した。
Separately, 1.0 ml containing 3 mg of F (ab ') 2 fragment of UCHT1 antibody prepared in 2) above.
To the TES buffer solution, add 20 μl of DTT aqueous solution and p
After reacting for 30 minutes at room temperature under H7.5, the obtained reaction solution was subjected to gel filtration using a Sephadex G25 column equilibrated with a TES buffer solution substituted with nitrogen gas, and the first peak was pooled. UCHT1-Fab'-SH
Was obtained.

【0051】上記UCHT1−Fab′−SHに、先に
調製したGFD−OA−p185−1−Fab′−SN
Bを加え、限外濾過膜(YM10)で濃縮後、室温で4
時間反応させ、以後、4℃で反応を継続させることによ
って、GFD−OA−p185−1−Fab′とUCT
H1−Fab′のモノマー同士が結合したBFAを得
た。
GFD-OA-p185-1-Fab'-SN prepared above was added to the UCHT1-Fab'-SH.
After adding B and concentrating with an ultrafiltration membrane (YM10), at room temperature, 4
After reacting for a period of time and then continuing the reaction at 4 ° C., GFD-OA-p185-1-Fab ′ and UCT were obtained.
BFA in which monomers of H1-Fab ′ were bound to each other was obtained.

【0052】上記で得られたBFAを含むTES緩衝液
をTSKゲルG3000SW(トーソー社製)を使用し
たHPLC(高速液体クロマトグラフィー)又はイオン
交換クロマトグラフィーやハイドロキシアパタイトカラ
ムクロマトグラフィーに付すことにより未反応のFa
b′−チオール誘導体(分子量約55kd)から目的の
BFAを分離して精製した。
The TES buffer solution containing BFA obtained above was subjected to HPLC (high performance liquid chromatography) using TSK gel G3000SW (manufactured by Tosoh Corp.), ion exchange chromatography or hydroxyapatite column chromatography to unreact. Fa
The target BFA was separated and purified from the b'-thiol derivative (molecular weight about 55 kd).

【0053】精製したBFAを非還元条件下で、0.1
%SDS(ドデシル硫酸ナトリウム、シグマ社製)存在
下に、7.5%PAGE分析を行ない、精製したBFA
の分子量を確認した。
The purified BFA was added to 0.1% under non-reducing conditions.
Purified BFA by performing 7.5% PAGE analysis in the presence of% SDS (sodium dodecyl sulfate, manufactured by Sigma).
The molecular weight of was confirmed.

【0054】上記SDS−PAGEの結果、分子量約1
10kdの位置にバンドを確認し、このことからBFA
は予想した分子量約110kdをもつことが確認され
た。
As a result of the above SDS-PAGE, the molecular weight was about 1
The band was confirmed at the position of 10 kd, and from this, the BFA
Was confirmed to have the expected molecular weight of about 110 kd.

【0055】[0055]

【実施例2】実施例1で得られた本発明BFAとGFD
−OA−p185−1−F(ab′)2 フラグメント及
びUCHT1−F(ab′)2 フラグメントとの免疫学
的反応性を、7つのヒト癌細胞株と健常ヒトボランティ
アから調製した末梢血リンパ球(PBL)を使用して以
下の通り試験した。
Example 2 BFA and GFD of the present invention obtained in Example 1
-OA-p185-1-F (ab ') 2 fragment and UCHT1-F (ab') 2 fragment were immunoreactive with peripheral blood lymphocytes prepared from seven human cancer cell lines and healthy human volunteers. Tested as follows using (PBL).

【0056】この試験には、以下に示す7つのヒト癌細
胞株(いずれもアメリカン・タイプ・カルチャー・コレ
クション、ATCC;Rockville, Md. U.S.A. より入
手)を用いた。
In this test, the following seven human cancer cell lines (all were obtained from American Type Culture Collection, ATCC; Rockville, Md. USA) were used.

【0057】ZR−75−1(ヒト乳癌細胞株:ATC
C CRL1500) SK−BR−3(ヒト乳腺癌細胞株:ATCC HTB
30) MCF−7(ヒト乳腺癌細胞株:ATCC HTB2
2) PANC−1(ヒト膵臓、上皮腺癌細胞株:ATCC
CRL1469) MIAPaCa−2(ヒト膵臓癌細胞株:ATCC C
RL1420) ASPC−1(ヒト転移性膵臓癌細胞株:ATCC C
RL1682) BxPC−3(原発性ヒト膵臓癌細胞株:ATCC C
RL1687) 上記7つのヒト癌細胞株と健常ボランティアのPBL
を、それぞれ0.1%BSA及び0.1%NaN3 を含
むPBS緩衝液で洗浄した後、同PBS緩衝液中に10
μgの各抗体を加え、氷上にて30分間反応させた。反
応後、細胞を2回洗浄し、次いでFITC−複合抗マウ
スIgG(Tago Inc., U.S.A. 社製)を加え、更に氷上
にて30分間反応させた。反応後、2回洗浄し、次いで
各抗体の反応活性をオルト・スペクトラムIII (Ortho
Diagnostic Inc., U.S.A. 社製)を使用したフロー・サ
イトメトリーによって測定した。尚、陽性細胞は対数表
示蛍光強度を所有しているものとして定量した。
ZR-75-1 (human breast cancer cell line: ATC
C CRL 1500) SK-BR-3 (human breast adenocarcinoma cell line: ATCC HTB
30) MCF-7 (human breast adenocarcinoma cell line: ATCC HTB2
2) PANC-1 (human pancreas, epithelial adenocarcinoma cell line: ATCC
CRL1469) MIAPaCa-2 (human pancreatic cancer cell line: ATCC C
RL1420) ASPC-1 (human metastatic pancreatic cancer cell line: ATCC C
RL1682) BxPC-3 (primary human pancreatic cancer cell line: ATCC C
RL1687) The above 7 human cancer cell lines and PBL of healthy volunteers
Were washed with PBS buffer containing 0.1% BSA and 0.1% NaN 3 , respectively, and then washed with 10% PBS buffer.
μg of each antibody was added and reacted on ice for 30 minutes. After the reaction, the cells were washed twice, FITC-conjugated anti-mouse IgG (Tago Inc., USA) was added, and the mixture was further reacted on ice for 30 minutes. After the reaction, the plate was washed twice, and then the reaction activity of each antibody was measured by Ortho Spectrum III (Ortho Spectrum III).
It was measured by flow cytometry using Diagnostic Inc., USA. Positive cells were quantified as possessing logarithmic display fluorescence intensity.

【0058】その結果を第1表に示す。The results are shown in Table 1.

【0059】[0059]

【表1】 [Table 1]

【0060】該表中、数字は各抗体と反応した多彩防の
パーセント(%)を示す。またコントロール(対照)は
第2抗体のみを表わし、NTは未試験を示す。
In the table, the numbers indicate the percentage (%) of multicolor defense reacted with each antibody. The control represents only the second antibody, and NT represents untested.

【0061】該表より、抗c−erbB−2遺伝子産物
F(ab′)2 フラグメントは、2つの乳癌細胞株ZR
−75−1及びSK−BR−3に対して強く反応した。
之等の2つの癌細胞株は、豊富にc−erbB−2mR
NAを発現することが知られている癌細胞株である[Kr
aus,M.H., et al., EMBO J., 6, 605-610 (1987)]。ま
たこのF(ab′)2 フラグメントは、癌細胞株MCF
−7、PANC−1、MIAPaCa−2及びASPC
−1と弱い反応性(前の2つの癌細胞株に比較して)を
示し、癌細胞株BxPC−3に対してはほとんど反応性
を示さなかった。更に、PBLとは反応しなかった。
From the table, it can be seen that the anti-c-erbB-2 gene product F (ab ') 2 fragment was associated with two breast cancer cell lines ZR.
Reacted strongly with -75-1 and SK-BR-3.
These two cancer cell lines are rich in c-erbB-2mR
A cancer cell line known to express NA [Kr
aus, MH, et al., EMBO J., 6 , 605-610 (1987)]. In addition, this F (ab ') 2 fragment was used as a cancer cell line MCF.
-7, PANC-1, MIAPaCa-2 and ASPC
It showed a weak reactivity with -1 (compared to the previous two cancer cell lines) and little reactivity with the cancer cell line BxPC-3. Furthermore, it did not react with PBL.

【0062】抗CD3εF(ab′)2 フラグメント
は、PBLにのみ反応し、癌細胞株とは反応しなかっ
た。本発明BFAは、2つのヒト乳癌細胞株ZR−75
−1及びSK−BR−3とPBLに対して強く反応し
た。また該BFAの反応活性は、他のヒト癌細胞株に対
しては上記抗c−erbB−2遺伝子産物F(ab′)
2 フセグメントと同様であった。
The anti-CD3εF (ab ') 2 fragment reacted only with PBL and not with the cancer cell line. The BFA of the present invention comprises two human breast cancer cell lines ZR-75.
-1 and SK-BR-3 reacted strongly with PBL. Further, the reaction activity of the BFA is the above anti-c-erbB-2 gene product F (ab ') against other human cancer cell lines.
It was similar to the 2 segment.

【0063】之等の試験結果から、本発明のBFAは期
待された免疫学的特徴を保有するものであることが明ら
かとなった。
From the above test results, it was revealed that the BFA of the present invention possesses the expected immunological characteristics.

【0064】[0064]

【実施例3】実施例1で得られた本発明BFAの細胞傷
害活性測定を、エフェクター細胞としてPBLを、標的
細胞としてZR−75−1及びSK−BR−3をそれぞ
れ使用して、BFAを介して之等各エフェクター細胞と
標的細胞とが結合した時に起こる標的細胞の特異的な溶
解を指標として、以下の方法に従い実施した。
Example 3 The cytotoxic activity of the BFA of the present invention obtained in Example 1 was measured using PBL as effector cells and ZR-75-1 and SK-BR-3 as target cells, respectively. Using the specific lysis of the target cells, which occurs when the respective effector cells and the target cells are bound to each other, as an index, the following procedure was performed.

【0065】 標的細胞の51Crによる標識 10%FCSを含むRPMI−1640培地を用いて、
ZR−75−1細胞及びSK−BR−3細胞を、37℃
で5%CO2 存在下で培養した。次に、培養上清を除去
し、PBS(-) 25mlで洗浄後、それぞれの培養細胞
に0.05%トリプシン−0.05%EDTA−PBS
溶液2.5mlを加え、37℃で1分間インキュベート
して、細胞を剥がし、10%FCS含有RPMI−16
40培地20mlに懸濁させて中和し、1000rp
m、8分間室温で2回同培地で遠心洗浄した。
Labeling of target cells with 51 Cr Using RPMI-1640 medium containing 10% FCS,
ZR-75-1 cells and SK-BR-3 cells were incubated at 37 ° C.
Were cultured in the presence of 5% CO 2 . Then, the culture supernatant was removed, and after washing with 25 ml of PBS (-) , 0.05% trypsin-0.05% EDTA-PBS was added to each cultured cell.
Add 2.5 ml of the solution and incubate at 37 ° C. for 1 minute to detach the cells and remove RPMI-16 containing 10% FCS.
Suspend in 20 ml of 40 medium to neutralize, 1000 rp
Centrifugation with the same medium was performed twice at room temperature for 8 minutes.

【0066】次いで、得られた各細胞の沈殿(5×10
6 細胞)に3.7MBqのクロム酸ナトリウム(Na 2
51CrO4 :NEN社製)を加え、37℃で5%CO2
存在下で60分間培養を行ない、培養液を10%FCS
含有RPMI−1640培地を用いて2回洗浄して過剰
のクロム酸ナトリウムを除去し、血球計算盤で細胞数を
数えた後、培養液の濃度が2×105 細胞/mlとなる
ように10%FCS含有RPMI−1640培地にて希
釈した。
Then, the resulting pellets of each cell (5 × 10 5
6 cells) with 3.7 MBq of sodium chromate (Na 2
51 CrO 4 : NEN) was added and 5% CO 2 was added at 37 ° C.
Incubate for 60 minutes in the presence of 10% FCS
Excess sodium chromate was removed by washing twice with the RPMI-1640 medium containing the cells, and after counting the number of cells with a hemocytometer, the concentration of the culture solution was adjusted to 2 × 10 5 cells / ml. It was diluted with RPMI-1640 medium containing% FCS.

【0067】 エフェクター細胞(PBL)の調製 ヘパリン採血した健常人血液50mlにPBS緩衝液5
0mlを加えて混合し、別に50mlの遠心管4本に比
重分離液(リンポサイトセパレーションメディウム:フ
ロー(Flow Laboratorues) 社製)を15mlずつ加えて
おき、先の血液25mlずつを之等各管に重層した。そ
の後、日立遠心機O5PR−22を用いて1500rp
m、30分間、室温にて密度勾配遠心分離を行なった。
Preparation of effector cells (PBL) PBS buffer 5 was added to 50 ml of heparin-collected blood of a healthy person.
Add 0 ml and mix. Separately, add 15 ml of specific gravity separation liquid (Lymphosite Separation Medium: Flow (Flow Laboratorues)) to 4 50 ml centrifuge tubes, and add 25 ml of the above blood to each tube. Overlaid. After that, 1500 rpm using Hitachi Centrifuge O5PR-22
Density gradient centrifugation was performed at room temperature for 30 minutes.

【0068】遠心後に4本の遠心管中の血漿と比重分離
液との間にできる白濁層をパスツールピペットを用いて
吸い取り、これを新しい50mlの遠心管に移し、25
mlの10%FCS含有RPMI−1640培地を加え
て混合し、1000rpm、5分間遠心してPBLを沈
殿として回収した。10%FCS含有RPMI−164
0培地による洗浄操作を3回繰り返した後、得られた沈
殿を10%FCS含有RPMI−1640培地に懸濁さ
せた。
After centrifugation, the white turbid layer formed between the plasma and the specific gravity separated liquid in the four centrifuge tubes was sucked up using a Pasteur pipette, and this was transferred to a new 50 ml centrifuge tube.
ml of RPMI-1640 medium containing 10% FCS was added and mixed, and centrifuged at 1000 rpm for 5 minutes to recover PBL as a precipitate. RPMI-164 containing 10% FCS
After the washing operation with 0 medium was repeated 3 times, the obtained precipitate was suspended in 10% FCS-containing RPMI-1640 medium.

【0069】 各抗体の調製 実施例1で得られた本発明のGFD−OA−p185−
1−Fab′−UCHT1−Fab′結合型BFA、こ
のBFA作製の出発材料として用いたGFD−OA−p
185−1−F(ab′)2 フラグメントとUCHT1
−F(ab′)2 フラグメントとの1:1混合物、GF
D−OA−p185−1−F(ab′)2 フラグメント
及びUCHT1−F(ab′)2 フラグメントのそれぞ
れを、10%FCS含有RPMI−1640培地を用い
て、それぞれをマイレックスGVフィルター0.22μ
M(日本ミリポアー社製)を用いて無菌濾過した後、2
ng/ml、20ng/ml、200ng/ml及び2
000ng/mlの各濃度に希釈調製した。
Preparation of each antibody GFD-OA-p185-of the present invention obtained in Example 1
1-Fab'-UCHT1-Fab 'linked BFA, GFD-OA-p used as a starting material for the preparation of this BFA
185-1-F (ab ') 2 fragment and UCHT1
1: 1 mixture with -F (ab ') 2 fragment, GF
Each of the D-OA-p185-1-F (ab ′) 2 fragment and the UCHT1-F (ab ′) 2 fragment was subjected to RPMI-1640 medium containing 10% FCS, and each of them was subjected to Mylex GV filter 0.22 μ.
After aseptic filtration using M (manufactured by Japan Millipore), 2
ng / ml, 20 ng / ml, 200 ng / ml and 2
Dilution was prepared to each concentration of 000 ng / ml.

【0070】 細胞傷害活性試験 上記で調製した各種濃度の抗体100μl及びエフェ
クター細胞50μl(細胞数=5×104 個、Effecto
r;E)と、標的細胞の50μl(1×104 個、Taget;T
)とを、それぞれ96穴U底培養プレート(ファルコ
ン(Falcon)社製)の各ウェルに添加(Effector/Taget
=E/T=5)し、37℃、5%CO2 存在下で4時間
培養して、標的細胞の特異的傷害反応を行なわせた。次
に、培養上清をスーパーネータント・コレクション・シ
ステム(大日本製薬社製)で採取し、測定チューブに入
れ、該チューブ内に含まれる放射能活性をカウントし
た。また、上記と同様な条件でそれぞれの抗体を、エフ
ェクター細胞と標的細胞の比率(E/T)を0、10及
び20に代えて、反応させた。
Cytotoxic activity test 100 μl of various concentrations of antibody prepared above and 50 μl of effector cells (cell number = 5 × 10 4 , Effecto)
r; E) and 50 μl of target cells (1 × 10 4 cells, Taget; T)
) Is added to each well of a 96-well U-bottom culture plate (Falcon) (Effector / Taget).
= E / T = 5), and the cells were cultured at 37 ° C. in the presence of 5% CO 2 for 4 hours to cause a specific damaging reaction of target cells. Next, the culture supernatant was collected by a Supernatant Collection System (manufactured by Dainippon Pharmaceutical Co., Ltd.), put in a measurement tube, and the radioactivity contained in the tube was counted. Further, each antibody was reacted under the same conditions as described above, except that the ratio (E / T) of effector cells to target cells was changed to 0, 10 and 20.

【0071】各抗体を介してエフェクター細胞と標的細
胞とを結合させた時に起こる標的細胞の特異的傷害活性
の程度(%)を、前記計算式(1)に従って求めた。
The degree (%) of the specific cytotoxic activity of the target cells, which occurs when the effector cells and the target cells are bound via each antibody, was determined according to the above calculation formula (1).

【0072】得られた結果を図1及び図2に示す。The obtained results are shown in FIGS. 1 and 2.

【0073】図1はZR−75−1細胞に対して本発明
BFAを用いた結果を示しており、図2はZR−75−
1細胞に対してGFD−OA−p185−1−F(a
b′)2 フラグメントとUCHT1−F(ab′)2
ラグメントとの1:1混合物を用いた場合の結果を示し
ている。
FIG. 1 shows the results of using the BFA of the present invention on ZR-75-1 cells, and FIG. 2 shows ZR-75-1.
GFD-OA-p185-1-F (a
The results are shown using a 1: 1 mixture of the b ') 2 fragment and the UCHT1-F (ab') 2 fragment.

【0074】各図において横軸は用いた抗体の絶対量
(ng/ml)を、縦軸は標的細胞の特異的傷害%(%
Specific Cytotoxicity )を示し、図中(1)はE/T
=0の場合を、(2)はE/T=5の場合を、(3)は
E/T=10の場合を、また(4)はE/T=20の場
合を、それぞれ示す。
In each figure, the horizontal axis represents the absolute amount of antibody used (ng / ml), and the vertical axis represents the specific injury% (%) of target cells.
Specific Cytotoxicity), where (1) is E / T
= 0, (2) shows E / T = 5, (3) shows E / T = 10, and (4) shows E / T = 20.

【0075】上記図1より、PBLと結合した本発明の
BFAはZR−75−1細胞のケースにおいて、有意な
細胞傷害活性を保有することが確認された。この細胞傷
害活性の発現に必要なBFAの濃度は、10ng/ml
以上であり、またE/T比は5以上であった。また、本
発明BFAは100ng/mlの用量で且つE/T比が
20で最大約25%の特異的標的細胞傷害が観察でき、
このことから高い標的細胞傷害活性を有することが確認
された。これに対して、GFD−OA−p185−1−
F(ab′)2 フラグメントとUCHT1−F(a
b′)2 フラグメントとの1:1混合物を用いた場合
は、1000ng/mlまで濃度を上げても有意な細胞
傷害活性は認められなかった(図2参照)。
From the above FIG. 1, it was confirmed that the BFA of the present invention bound to PBL possesses significant cytotoxic activity in the case of ZR-75-1 cells. The concentration of BFA required for expression of this cytotoxic activity is 10 ng / ml.
Above, the E / T ratio was 5 or more. In addition, the BFA of the present invention can observe specific target cell damage of up to about 25% at a dose of 100 ng / ml and an E / T ratio of 20,
From this, it was confirmed to have a high target cytotoxic activity. On the other hand, GFD-OA-p185-1-
F (ab ') 2 fragment and UCHT1-F (a
When a 1: 1 mixture with b ′) 2 fragment was used, no significant cytotoxic activity was observed even when the concentration was increased to 1000 ng / ml (see FIG. 2).

【0076】更に、GFD−OA−p185−1−F
(ab′)2 フラグメント及びUCHT1−F(a
b′)2 フラグメントのそれぞれを用いて同一試験を行
なった結果、之等の各フラグメントは1000ng/m
lまで濃度を上げても有意な細胞傷害活性は認められな
かった。
Furthermore, GFD-OA-p185-1-F
(Ab ') 2 fragment and UCHT1-F (a
b ') The same test was carried out using each of the 2 fragments, and as a result, each of these fragments was 1000 ng / m 2.
No significant cytotoxic activity was observed even when the concentration was increased to 1.

【0077】以上の結果より、本発明BFAは10ng
/mlという極微小量から標的細胞の特異的細胞傷害活
性を示し、このことからc−erbB−2遺伝子産物を
細胞表面に発現している癌細胞よりなる悪性腫瘍の治療
剤、特に乳癌治療剤として有用であることが分かる。
From the above results, the BFA of the present invention is 10 ng
A therapeutic agent for malignant tumors, particularly a breast cancer therapeutic agent, which is composed of cancer cells expressing a c-erbB-2 gene product on the cell surface, which exhibits a specific cytotoxic activity of target cells from an extremely small amount of 1 / ml. It turns out to be useful as

【0078】また本発明BFAの標的細胞に対する高い
結合力維持性を応用すれば、本発明所望の上記治療剤の
他にも、例えば従来のモノクロール抗体に代わって、各
種の診断薬としての利用も期待できる。
In addition to the above therapeutic agents desired in the present invention, for example, the BFA of the present invention can be used as various diagnostic agents in place of conventional monoclonal antibodies in addition to the above therapeutic agents desired in the present invention. Can be expected.

【図面の簡単な説明】[Brief description of drawings]

【図1】本発明BFAの細胞傷害活性を調べたグラフで
ある。
FIG. 1 is a graph showing the cytotoxic activity of the BFA of the present invention.

【図2】図1と対比して対照とする抗体混合物の細胞傷
害活性を調べたグラフである。
FIG. 2 is a graph showing the cytotoxic activity of the antibody mixture used as a control in comparison with FIG. 1.

───────────────────────────────────────────────────── フロントページの続き (72)発明者 出口 恭平 徳島県徳島市末広4丁目1−9−703号 (72)発明者 今川 健一 徳島県板野郡北島町新喜来字北ハリ1−83 (72)発明者 菊地 幹雄 神奈川県鎌倉市今泉台4−29−14 ─────────────────────────────────────────────────── ─── Continuation of front page (72) Inventor Kyohei Exit 1-9-703 Suehiro, Tokushima City, Tokushima Prefecture (72) Inventor Kenichi Imagawa 1-83 Kitahari, Kitakijima, Itano-gun, Tokushima Prefecture (72) ) Inventor Mikio Kikuchi 4-29-14 Imaizumidai, Kamakura City, Kanagawa Prefecture

Claims (6)

【特許請求の範囲】[Claims] 【請求項1】 複合抗体を構成する一方の抗体がヒトc
−erbB−2特異性抗体であり、他方がヒトリンパ球
抗体であることを特徴とする複合抗体。
1. One of the antibodies constituting the composite antibody is human c
-A composite antibody characterized in that it is an erbB-2 specific antibody and the other is a human lymphocyte antibody.
【請求項2】 標的細胞の表面抗原を認識する抗体が抗
ヒトc−erbB−2抗体であって、エフェクター細胞
に結合してシグナル伝達を行う抗体が抗ヒトリンパ球抗
体である請求項1に記載の複合抗体。
2. The antibody recognizing the surface antigen of a target cell is an anti-human c-erbB-2 antibody, and the antibody that binds to effector cells to perform signal transduction is an anti-human lymphocyte antibody. Complex antibody.
【請求項3】 抗ヒトc−erbB−2抗体がGFD−
OA−p185−1である請求項1又は2に記載の複合
抗体。
3. The anti-human c-erbB-2 antibody is GFD-
The composite antibody according to claim 1 or 2, which is OA-p185-1.
【請求項4】 抗ヒトリンパ球抗体が抗CD3抗体であ
る請求項1又は2に記載の複合抗体。
4. The composite antibody according to claim 1, wherein the anti-human lymphocyte antibody is an anti-CD3 antibody.
【請求項5】 ヒトc−erbB−2関連蛋白に特異的
に反応する請求項1又は2に記載の複合抗体。
5. The composite antibody according to claim 1 or 2, which specifically reacts with a human c-erbB-2 related protein.
【請求項6】 請求項3に記載の複合抗体を必須成分と
することを特徴とする癌治療剤。
6. A therapeutic agent for cancer, which comprises the complex antibody according to claim 3 as an essential component.
JP1996892A 1992-02-05 1992-02-05 Bfa antibody Pending JPH05213775A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP1996892A JPH05213775A (en) 1992-02-05 1992-02-05 Bfa antibody

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP1996892A JPH05213775A (en) 1992-02-05 1992-02-05 Bfa antibody

Publications (1)

Publication Number Publication Date
JPH05213775A true JPH05213775A (en) 1993-08-24

Family

ID=12013993

Family Applications (1)

Application Number Title Priority Date Filing Date
JP1996892A Pending JPH05213775A (en) 1992-02-05 1992-02-05 Bfa antibody

Country Status (1)

Country Link
JP (1) JPH05213775A (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6627196B1 (en) 1999-08-27 2003-09-30 Genentech, Inc. Dosages for treatment with anti-ErbB2 antibodies
US7041292B1 (en) 1999-06-25 2006-05-09 Genentech, Inc. Treating prostate cancer with anti-ErbB2 antibodies
US7371376B1 (en) 1996-10-18 2008-05-13 Genentech, Inc. Anti-ErbB2 antibodies
EP2289944A2 (en) 2003-10-10 2011-03-02 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
EP2311945A1 (en) 2003-10-14 2011-04-20 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies substituting for functional proteins
US9334331B2 (en) 2010-11-17 2016-05-10 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US10759870B2 (en) 2017-09-29 2020-09-01 Chugai Seiyaku Kabushiki Kaisha Multispecific antigen-binding molecules having blood coagulation factor VIII (FVIII) cofactor function-substituting activity and pharmaceutical formulations containing such a molecule as an active ingredient
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11124576B2 (en) 2013-09-27 2021-09-21 Chungai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
US11150254B2 (en) 2014-09-26 2021-10-19 Chugai Seiyaku Kabushiki Kaisha Method for measuring reactivity of FVIII
US11214623B2 (en) 2014-09-26 2022-01-04 Chugai Seiyaku Kabushiki Kaisha Antibody capable of neutralizing substance having activity alternative to function of coagulation factor VIII (FVIII)
US11352438B2 (en) 2016-09-06 2022-06-07 Chugai Seiyaku Kabushiki Kaisha Methods of using a bispecific antibody that recognizes coagulation factor IX and/or activated coagulation factor IX and coagulation factor X and/or activated coagulation factor X
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05170667A (en) * 1991-12-26 1993-07-09 Kyowa Hakko Kogyo Co Ltd Bi-specific antibody

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05170667A (en) * 1991-12-26 1993-07-09 Kyowa Hakko Kogyo Co Ltd Bi-specific antibody

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7371376B1 (en) 1996-10-18 2008-05-13 Genentech, Inc. Anti-ErbB2 antibodies
US7041292B1 (en) 1999-06-25 2006-05-09 Genentech, Inc. Treating prostate cancer with anti-ErbB2 antibodies
US6627196B1 (en) 1999-08-27 2003-09-30 Genentech, Inc. Dosages for treatment with anti-ErbB2 antibodies
US7371379B2 (en) 1999-08-27 2008-05-13 Genentech, Inc. Dosages for treatment with anti-ErbB2 antibodies
US10280228B2 (en) 1999-08-27 2019-05-07 Genentech, Inc. Treatment with anti-ErbB2 antibodies
US10160811B2 (en) 1999-08-27 2018-12-25 Genentech, Inc. Treatment with anti-ErbB2 antibodies
EP3085783A1 (en) 2003-10-10 2016-10-26 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
US8062635B2 (en) 2003-10-10 2011-11-22 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
EP2289944A2 (en) 2003-10-10 2011-03-02 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
EP2311945A1 (en) 2003-10-14 2011-04-20 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies substituting for functional proteins
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US11168344B2 (en) 2005-03-31 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US10934344B2 (en) 2006-03-31 2021-03-02 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9334331B2 (en) 2010-11-17 2016-05-10 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies
US10450381B2 (en) 2010-11-17 2019-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of treatment that include the administration of bispecific antibodies
US11124576B2 (en) 2013-09-27 2021-09-21 Chungai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US11150254B2 (en) 2014-09-26 2021-10-19 Chugai Seiyaku Kabushiki Kaisha Method for measuring reactivity of FVIII
US11214623B2 (en) 2014-09-26 2022-01-04 Chugai Seiyaku Kabushiki Kaisha Antibody capable of neutralizing substance having activity alternative to function of coagulation factor VIII (FVIII)
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11352438B2 (en) 2016-09-06 2022-06-07 Chugai Seiyaku Kabushiki Kaisha Methods of using a bispecific antibody that recognizes coagulation factor IX and/or activated coagulation factor IX and coagulation factor X and/or activated coagulation factor X
US10759870B2 (en) 2017-09-29 2020-09-01 Chugai Seiyaku Kabushiki Kaisha Multispecific antigen-binding molecules having blood coagulation factor VIII (FVIII) cofactor function-substituting activity and pharmaceutical formulations containing such a molecule as an active ingredient

Similar Documents

Publication Publication Date Title
JPH05213775A (en) Bfa antibody
JP3213314B2 (en) Monoclonal antibodies against stem cell factor receptor
EP3858856A1 (en) Anti-b7-h3 monoclonal antibody and use thereof in cell therapy
CA2129183C (en) Bispecific trigger molecules recognizing lymphocyte antigen cd2 and tumor antigens
JP3541296B2 (en) Monoclonal antibodies that specifically bind to tumor vascular endothelial cells and their use
AU2002240719B2 (en) Antibodies against cancer
CN110869389A (en) anti-ROR 1 antibodies and methods of making and using the same
JP2979318B2 (en) Immunotherapy of tumor with monoclonal antibody against 17-1A antigen
AU2002240719A1 (en) Antibodies against cancer
CN112830969B (en) Monoclonal antibody specifically binding to human Claudin18.2, and medicine and kit containing monoclonal antibody
CN113354739B (en) Chimeric antigen receptor for targeted expression of Claudin18.2 cell and application thereof
CN110563843A (en) Monoclonal antibody targeting human VISTA protein
CN116323676A (en) Bispecific antibodies against Claudin18.2 and CD3 and uses thereof
CN114591434B (en) anti-Siglec 15 antibody and preparation method and application thereof
WO2022194201A1 (en) Cldn18.2-targeting antibody or antigen binding fragment thereof and use thereof
JP2023525778A (en) Anti-BCMA antibody and chimeric antigen receptor
CN105385694B (en) Anti-human CD98 monoclonal antibody 98-3H3 light and heavy chain variable region gene and its application
EP1528936B1 (en) Combination of tumor-associated surface protein antigens and tumor-associated sugars in the treatment and diagnosis of cancer
WO2021244553A1 (en) Tetravalent bispecific antibody against pd-1 and egfr
US7189824B2 (en) Tumor vessel endothelial cell-binding monoclonal antibodies
JPH05304987A (en) Human type monoclonal antibody and gene coding the same, hybridoma and antitumor agent
CN110563848A (en) bispecific antibody and application thereof
WO2023143535A1 (en) Antibody targeting il-18bp and use thereof
CN117430711A (en) Bispecific fusion protein for treating liver cell type liver cancer and application thereof
CA2116753C (en) Monoclonal antibodies recognizing the epidermal growth factor-receptor, cells and methods for producing them and compositions containing them