WO2016163433A1 - Composition comprising anti-fgfr2 antibody and another agent - Google Patents

Composition comprising anti-fgfr2 antibody and another agent Download PDF

Info

Publication number
WO2016163433A1
WO2016163433A1 PCT/JP2016/061333 JP2016061333W WO2016163433A1 WO 2016163433 A1 WO2016163433 A1 WO 2016163433A1 JP 2016061333 W JP2016061333 W JP 2016061333W WO 2016163433 A1 WO2016163433 A1 WO 2016163433A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
combination
composition according
contained
amino acid
Prior art date
Application number
PCT/JP2016/061333
Other languages
French (fr)
Japanese (ja)
Inventor
能伸 塩瀬
さおり 石田
Original Assignee
第一三共株式会社
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 第一三共株式会社 filed Critical 第一三共株式会社
Priority to JP2017511041A priority Critical patent/JP6887944B2/en
Publication of WO2016163433A1 publication Critical patent/WO2016163433A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Inorganic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A combination comprising an antibody capable of binding to human fibroblast growth factor receptor 2 (hFGFR2) and an agent that is different from the antibody.

Description

抗FGFR2抗体と他剤を含む組成物Composition comprising anti-FGFR2 antibody and other agent
 本発明は、新規な抗体もしくはその機能断片またはその修飾体および他の有効成分の組合せ、該抗体もしくはその機能断片またはその修飾体および他の有効成分を含有する医薬組成物等に関する。 The present invention relates to a novel antibody or a functional fragment thereof or a combination thereof and a combination of other active ingredients, and a pharmaceutical composition containing the antibody or a functional fragment thereof or a modification thereof and other active ingredients.
 癌の治療では、手術による癌部分の除去、抗癌剤や放射線により癌細胞を殺すことで治癒や延命効果が期待される。しかしながら、多くの癌に対しては単独の抗癌剤治療だけでは十分な効果が得られないため、様々な薬剤との併用により治療が試みられている。抗癌剤にはドキソルビシン、シスプラチン、タキソール、あるいはカンプトテシンなどの化学療法剤や、クリゾチニブ、ダサチニブ、ラパチニブなどの分子標的薬、ベバシズマブ、セツキシマブ、トラスツズマブなどの癌治療抗体、アナストロゾール、エキセメスタン、タモキシフェンなどのホルモン剤などがあり、癌の種類や進行度、治療状況により、使われる抗癌剤やその併用療法も決定される。細胞の増殖、生存を司るレセプターに対し、細胞外領域に作用し、直接増殖シグナルを遮断したり、ADCCを介して抗腫瘍活性を示す癌治療抗体と、同じレセプターに対する薬剤や、異なるレセプターを阻害する薬剤、あるいは化学療法剤等を併用することで抗癌作用を強める方策は以前より行われてきた。 In the treatment of cancer, healing and life-prolonging effects are expected by removing cancerous parts by surgery and killing cancer cells with anticancer drugs and radiation. However, since a sufficient effect cannot be obtained only with a single anticancer drug treatment for many cancers, treatments are being attempted in combination with various drugs. Anticancer agents include chemotherapeutic agents such as doxorubicin, cisplatin, taxol, or camptothecin; molecular targeting drugs such as crizotinib, dasatinib, and lapatinib; cancer therapeutic antibodies such as bevacizumab, cetuximab, and trastuzumab; hormones such as anastrozole, exemestane, and tamoxifen There are drugs, etc., and the anticancer drug used and its combination therapy are also determined depending on the type and progression of cancer and the treatment status. Receptors that control cell growth and survival act on the extracellular domain and directly block growth signals or inhibit anti-tumor activity through ADCC, drugs against the same receptor, or inhibit different receptors Measures to strengthen the anti-cancer effect by using a combination drug or a chemotherapeutic agent have been used for some time.
 Laptinib (Her2 TKI)とHerceptin (抗Her2抗体)の併用は、乳癌治療において併用効果が認められ、lapatinibの作用によりHer2が細胞表面に蓄積することで、Her2の効果を強めている可能性が示唆されている(非特許文献1)。また、ErbBファミリー全体を阻害するafatinibと、抗EGFRモノクローナル抗体セツキシマブのEGFRの二重阻害においても強い抗癌作用を示すことが明らかとなっている(非特許文献2)。このように、受容体型tyrosine kinaseに対し、それを認識する抗体と、そのtyrosin kinase活性を阻害する低分子薬剤との併用は、強い増殖阻害を引き起こし、有用な治療法に繋がる可能性がある。他にも、大腸癌治療において、化学療法剤であるカンプトテシンとEGFR阻害剤であるセツキシマブとの併用やアバスチンとの併用は、標準治療として確立されており、大腸癌治療に大きなメリットを示している。 The combined use of Laptinib (Her2 TKI) and Herceptin (anti-Her2 antibody) has been shown to have a combined effect in breast cancer treatment, suggesting that Her2 may accumulate on the cell surface due to the action of lapatinib, thereby enhancing the effect of Her2. (Non-Patent Document 1). In addition, it has been clarified that a strong anticancer activity is exhibited even in the double inhibition of EGFR of afatinib that inhibits the whole ErbB family and the anti-EGFR monoclonal antibody cetuximab (Non-patent Document 2). Thus, the combined use of an antibody recognizing the receptor-type tyrosine kinase and a low-molecular-weight drug that inhibits the tyrosin kinase activity may cause strong growth inhibition and lead to a useful therapeutic method. In addition, the combination of chemotherapeutic camptothecin with EGFR inhibitor cetuximab and Avastin has been established as a standard treatment for the treatment of colorectal cancer, and has shown great benefits for colorectal cancer treatment. .
 しかしながら、抗FGFR2抗体と併用することにより、抗癌作用が増強される薬剤についてはほとんど知られていない(特許文献1及び2)。 However, little is known about drugs whose anticancer effect is enhanced by using in combination with anti-FGFR2 antibodies (Patent Documents 1 and 2).
米国特許出願公開US2014/0322220A1号US patent application publication US2014 / 0322220A1 米国特許出願公開US2015/0050273A1号US Patent Application Publication No. US2015 / 0050273A1
 本発明の一つの課題は抗FGFR2抗体および他剤の組合せ、ならびに、該抗体および他剤を含有する医薬組成物等を提供することである。 One object of the present invention is to provide a combination of an anti-FGFR2 antibody and another agent, and a pharmaceutical composition containing the antibody and the other agent.
 また、本発明の他の一つの課題は、癌の治療又は予防に使用される抗FGFR2抗体および他剤を組合せ、ならびに、癌の治療又は予防に使用される、該抗体および他剤を含有する医薬組成物を提供することにある。 Another subject of the present invention is a combination of an anti-FGFR2 antibody and other agents used for the treatment or prevention of cancer, and the antibody and other agents used for the treatment or prevention of cancer. It is to provide a pharmaceutical composition.
 さらに、本発明の他の一つの課題は、抗FGFR2抗体と他剤とを組み合わせて投与することにより、または、該組成物を投与することにより、癌を治療又は予防する方法を提供することにある。 Furthermore, another object of the present invention is to provide a method for treating or preventing cancer by administering an anti-FGFR2 antibody and another agent in combination, or by administering the composition. is there.
 発明者らは上記課題を解決するために検討を行い、抗癌活性を有する抗FGFR2抗体に様々な抗癌剤(例えば、ドキソルビシン、シスプラチン、タキソール、ドセタキセル、フルオロウラシルあるいはカンプトテシンなどの化学療法剤や、クリゾチニブ、ダサチニブ、ラパチニブなどの分子標的薬、ベバシズマブ、セツキシマブ、トラスツズマブ、ラムシルマブなどの癌治療抗体、アナストロゾール、エキセメスタン、タモキシフェンなどのホルモン剤など)を組み合わせて使用することにより、優れた抗腫瘍効果が得られることを見出し、本発明を完成させた。 The inventors have studied to solve the above-mentioned problems, and various anticancer agents (for example, doxorubicin, cisplatin, taxol, docetaxel, fluorouracil or camptothecin, chemotherapeutic agents such as crizotinib, anti-FGFR2 antibody having anticancer activity, Combined with molecular targeted drugs such as dasatinib and lapatinib, cancer therapeutic antibodies such as bevacizumab, cetuximab, trastuzumab, and ramcilmab, and hormonal drugs such as anastrozole, exemestane, and tamoxifen) The present invention has been completed.
 本発明は、
(1)下記(i)乃至(ix)の特徴を有するモノクローナル抗体もしくはその機能断片、および、トポイソメラーゼ阻害剤、微小管阻害剤、プラチナ製剤、抗VEGFR2抗体及び5-フルオロウラシル系抗がん剤よりなる群から選択される一つ又は二つ以上の他剤の組合せ、又は、該抗体もしくはその機能断片、および、トポイソメラーゼ阻害剤、微小管阻害剤、プラチナ製剤、抗VEGFR2抗体及び5-フルオロウラシル系抗がん剤よりなる群から選択される一つ又は二つ以上の他剤を含む医薬組成物:
(i)ヒト2型線維芽細胞増殖因子受容体(hFGFR2)IIIb及びIIIcに特異的に結合する;
(ii)hFGFR2の免疫グロブリン様ドメイン2又は免疫グロブリン様ドメイン3に結合する;
(iii)ヒト1型線維芽細胞増殖因子受容体(hFGFR1)、ヒト3型線維芽細胞増殖因子受容体(hFGFR3)及びヒト4型線維芽細胞増殖因子受容体(hFGFR4)には特異的に結合しない;
(iv)抗体依存性細胞傷害活性を有する;
(v)抗体依存性細胞媒介食活性を有する;
(vi)hFGFR2に対する中和活性を有する;
(vii)in vivo抗腫瘍活性を有する;
(viii)hFGFR2へのFGFの結合を阻害する;
(ix)キメラ抗体、ヒト化抗体又はヒト抗体である、
(2)
 該抗体が配列表の配列番号1(図1)に示されるアミノ酸配列からなるCDRL1、配列表の配列番号2(図2)に示されるアミノ酸配列からなるCDRL2及び配列表の配列番号3(図3)に示されるアミノ酸配列からなるCDRL3を含むことからなる軽鎖、並びに、配列表の配列番号4(図4)に示されるアミノ酸配列からなるCDRH1、配列表の配列番号5(図5)に示されるアミノ酸配列からなるCDRH2及び配列表の配列番号6(図6)に示されるアミノ酸配列もしくは該アミノ酸配列のアミノ末端から数えて1つ目又は2つ目のアミノ酸が他のアミノ酸に置換されてなるアミノ酸配列からなるCDRH3を含むことからなる重鎖からなる、(1)に記載の組合せ又は医薬組成物、
(3)
 該抗体が下記(i)乃至(vi)から選択される、(1)又は(2)に記載の組合せ又は医薬組成物:
(i)配列番号は8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号18(図12)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H19/L1);
(ii)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号16(図11)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H12/L1);
(iii)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号12(図9)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H8/L1);
(iv)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号20(図16)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H9/L1);
(v)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号14(図10)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H11/L1);
(vi)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号10(図8)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H5/L1)、
(4)
 該抗体が(3)の(i)乃至(vi)のいずれか一つに記載の抗体の重鎖および軽鎖のアミノ酸配列とそれぞれ95%以上同一なアミノ酸配列を含む重鎖および軽鎖を含んでなり、且つヒトFGFR2に結合する、(1)に記載の組合せ又は医薬組成物、
(5)
 該抗体もしくはその機能断片が、(2)又は(3)の(i)乃至(vi)のいずれか一つに記載の抗体が認識する抗原上の部位に結合する、(1)に記載の組合せ又は医薬組成物、
The present invention
(1) A monoclonal antibody or a functional fragment thereof having the following characteristics (i) to (ix), and a topoisomerase inhibitor, a microtubule inhibitor, a platinum preparation, an anti-VEGFR2 antibody and a 5-fluorouracil anticancer agent. A combination of one or two or more other agents selected from the group, or the antibody or a functional fragment thereof, and a topoisomerase inhibitor, a microtubule inhibitor, a platinum preparation, an anti-VEGFR2 antibody and a 5-fluorouracil anti-antibody A pharmaceutical composition comprising one or more other agents selected from the group consisting of cancer agents:
(I) specifically binds to human type 2 fibroblast growth factor receptor (hFGFR2) IIIb and IIIc;
(Ii) binds to immunoglobulin-like domain 2 or immunoglobulin-like domain 3 of hFGFR2;
(Iii) Specific binding to human type 1 fibroblast growth factor receptor (hFGFR1), human type 3 fibroblast growth factor receptor (hFGFR3) and human type 4 fibroblast growth factor receptor (hFGFR4) do not do;
(Iv) has antibody-dependent cytotoxic activity;
(V) has antibody-dependent cell-mediated phagocytic activity;
(Vi) has neutralizing activity against hFGFR2;
(Vii) has in vivo anti-tumor activity;
(Viii) inhibits the binding of FGF to hFGFR2;
(Ix) a chimeric antibody, a humanized antibody or a human antibody,
(2)
The antibody has CDRL1 consisting of the amino acid sequence shown in SEQ ID NO: 1 (FIG. 1), CDRL2 consisting of the amino acid sequence shown in SEQ ID NO: 2 (FIG. 2), and SEQ ID NO: 3 (FIG. 3). ), A light chain comprising CDRL3 consisting of the amino acid sequence shown in FIG. 4, a CDRH1 consisting of the amino acid sequence shown in SEQ ID NO: 4 (FIG. 4) in the sequence listing, and SEQ ID NO: 5 (FIG. 5) in the sequence listing. CDRH2 comprising the amino acid sequence and the amino acid sequence shown in SEQ ID NO: 6 (FIG. 6) of the sequence listing or the first or second amino acid counted from the amino terminus of the amino acid sequence are substituted with other amino acids. The combination or pharmaceutical composition according to (1), comprising a heavy chain comprising CDRH3 comprising an amino acid sequence,
(3)
The combination or pharmaceutical composition according to (1) or (2), wherein the antibody is selected from the following (i) to (vi):
(I) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 18 (FIG. 12) A humanized antibody comprising a chain (hFR2-14_H19 / L1);
(Ii) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 16 (FIG. 11) A humanized antibody comprising (hFR2-14_H12 / L1);
(Iii) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 12 (FIG. 9) A humanized antibody comprising (hFR2-14_H8 / L1);
(Iv) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 20 (FIG. 16) A humanized antibody comprising (hFR2-14_H9 / L1);
(V) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 14 (FIG. 10) A humanized antibody comprising (hFR2-14_H11 / L1);
(Vi) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 10 (FIG. 8) A humanized antibody (hFR2-14_H5 / L1),
(4)
The antibody comprises a heavy chain and a light chain each comprising 95% or more amino acid sequence identical to the amino acid sequence of the heavy chain and the light chain of the antibody according to any one of (i) to (vi) And the combination or pharmaceutical composition according to (1), which binds to human FGFR2;
(5)
The combination according to (1), wherein the antibody or a functional fragment thereof binds to a site on the antigen recognized by the antibody according to any one of (2) or (3) (i) to (vi) Or a pharmaceutical composition,
(6)
 該抗体もしくはその機能断片が、(2)又は(3)の(i)乃至(vi)のいずれか一つに記載の抗体と、hFGFR2への結合において競合する、(1)に記載の組合せ又は医薬組成物、
(7)
 下記の工程(i)及び(ii)を含むことからなる方法により得られる抗体もしくはその機能断片および他剤の組合せ又は該抗体もしくはその機能断片および他剤を含む医薬組成物:
(i)下記(ア)又は(イ)に記載の細胞を培養する工程;
(ア)下記(a)乃至(c)のいずれか一つに記載のヌクレオチドが挿入された組換えベクター又は該ヌクレオチドが導入された組換え細胞:
(a)(1)乃至(6)のいずれか一つに記載の抗体の重鎖または軽鎖の一部または全部のアミノ酸配列をコードする塩基配列を含んでなるヌクレオチド;
(b)(1)乃至(6)のいずれか一つに記載の抗体の重鎖または軽鎖の一部または全部のアミノ酸配列をコードする塩基配列を含む塩基配列からなるヌクレオチド;
(c)(1)乃至(6)のいずれか一つに記載の抗体の重鎖または軽鎖の一部または全部のアミノ酸配列をコードする塩基配列からなるヌクレオチド;
(イ)(1)乃至(6)のいずれか一つに記載の抗体又はその機能断片を産生する細胞、
および
(ii)前記工程(i)で得られた培養物から(1)乃至(6)のいずれか一つに記載の抗体又はその機能断片を回収する工程、
(8)
 該抗体の重鎖または軽鎖のアミノ末端またはカルボキシル末端において1乃至5個のアミノ酸が欠失してなる、(1)乃至(7)のいずれか一つに記載の組合せ又は医薬組成物、
(9)
 (1)乃至(8)のいずれか一つに記載の抗体もしくはその機能断片の糖鎖修飾体および他剤の組合せ、又は、該修飾体および他剤を含む医薬組成物、
(10)
 癌の治療もしくは予防に使用される、(1)乃至(9)のいずれか一つに記載の組合せ又は医薬組成物、
(6)
The combination according to (1), wherein the antibody or a functional fragment thereof competes with the antibody according to any one of (i) to (vi) of (2) or (3) in binding to hFGFR2, or Pharmaceutical composition,
(7)
A combination of an antibody or a functional fragment thereof obtained by a method comprising the following steps (i) and (ii) and another agent, or a pharmaceutical composition comprising the antibody or a functional fragment thereof and the other agent:
(I) a step of culturing the cell according to (a) or (b) below;
(A) A recombinant vector into which the nucleotide according to any one of the following (a) to (c) is inserted or a recombinant cell into which the nucleotide has been introduced:
(A) a nucleotide comprising a base sequence encoding part or all of the amino acid sequence of the heavy chain or light chain of the antibody according to any one of (1) to (6);
(B) a nucleotide consisting of a base sequence comprising a base sequence encoding part or all of the amino acid sequence of the heavy chain or light chain of the antibody according to any one of (1) to (6);
(C) a nucleotide comprising a base sequence encoding part or all of the amino acid sequence of the heavy chain or light chain of the antibody according to any one of (1) to (6);
(A) a cell producing the antibody or the functional fragment thereof according to any one of (1) to (6),
And (ii) recovering the antibody or functional fragment thereof according to any one of (1) to (6) from the culture obtained in the step (i),
(8)
The combination or pharmaceutical composition according to any one of (1) to (7), wherein 1 to 5 amino acids are deleted at the amino terminus or carboxyl terminus of the heavy or light chain of the antibody,
(9)
(1) to (8) a combination of a modified sugar chain of the antibody or a functional fragment thereof according to any one of (1) to (8) and another agent, or a pharmaceutical composition comprising the modified agent and the other agent;
(10)
The combination or pharmaceutical composition according to any one of (1) to (9), which is used for treatment or prevention of cancer,
(11)
 癌がhFGFR2陽性である、(10)に記載の組合せ又は医薬組成物、
(12)
 該抗体もしくはその機能断片がさらなる化合物とコンジュゲートされた、(1)乃至(11)のいずれか一つに記載の組合せ又は医薬組成物、
(13)
 他剤がトポイソメラーゼ阻害剤である、(1)乃至(12)のいずれか一つに記載の組合せ又は医薬組成物、
(14)
 トポイソメラーゼ阻害剤と併用するための、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物、
(15)
 トポイソメラーゼ阻害剤を含み、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物、
(11)
The combination or pharmaceutical composition according to (10), wherein the cancer is hFGFR2 positive,
(12)
The combination or pharmaceutical composition according to any one of (1) to (11), wherein the antibody or functional fragment thereof is conjugated with a further compound;
(13)
The combination or pharmaceutical composition according to any one of (1) to (12), wherein the other agent is a topoisomerase inhibitor;
(14)
An antibody or a functional fragment thereof included in the combination or composition according to any one of (1) to (8) or a combination or composition according to (9) for use in combination with a topoisomerase inhibitor A pharmaceutical composition comprising the modified form,
(15)
An antibody or functional fragment thereof, or a modified form contained in the combination or composition according to (9), which comprises a topoisomerase inhibitor and is contained in the combination or composition according to any one of (1) to (8) A pharmaceutical composition that increases the action of the antibody, the functional fragment, or the modified product,
(16)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含み、トポイソメラーゼ阻害剤と併用することにより、該トポイソメラーゼ阻害剤の作用を上昇させる医薬組成物、
(17)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が組み合わせて投与されることを特徴とする医薬組成物、
(18)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、(17)に記載の医薬組成物、
(19)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が、単一の製剤に有効成分として含有されることを特徴とする、(17)に記載の医薬組成物、
(20)
(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が組み合わせて投与されることを特徴とする癌の治療方法、
(16)
An antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9), and a topoisomerase inhibitor; A pharmaceutical composition that increases the action of the topoisomerase inhibitor when used in combination;
(17)
An antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9), and a topoisomerase inhibitor A pharmaceutical composition characterized by being administered in combination;
(18)
An antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9), and a topoisomerase inhibitor The pharmaceutical composition according to (17), which is contained as an active ingredient in different preparations and administered at the same time or at different times,
(19)
An antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9), and a topoisomerase inhibitor The pharmaceutical composition according to (17), which is contained as an active ingredient in a single preparation,
(20)
An antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9), and a topoisomerase inhibitor A method for treating cancer, characterized by being administered in combination;
(21)
 トポイソメラーゼ阻害剤がイリノテカン、トポテカン及びエトポシドよりなる群から選択される一つ又は二つ以上である、(13)乃至(19)のいずれか一つに記載の医薬組成物又は(20)に記載の治療方法、
(22)
 他剤が微小管阻害剤である、(1)乃至(12)のいずれか一つに記載の組合せ又は医薬組成物、
(23)
 微小管阻害剤と併用するための、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物、
(24)
 微小管阻害剤を含み、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物、
(25)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含み、微小管阻害剤と併用することにより、該微小管阻害剤の作用を上昇させる医薬組成物、
(21)
The pharmaceutical composition according to any one of (13) to (19), wherein the topoisomerase inhibitor is one or more selected from the group consisting of irinotecan, topotecan and etoposide, or (20) Method of treatment,
(22)
The combination or pharmaceutical composition according to any one of (1) to (12), wherein the other agent is a microtubule inhibitor,
(23)
In combination with the microtubule inhibitor, the antibody or the functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the combination or composition according to (9) A pharmaceutical composition comprising a modified product,
(24)
An antibody or a functional fragment thereof, which is included in the combination or composition according to any one of (1) to (8), or a modified form which is included in the combination or composition according to (9), comprising a microtubule inhibitor A pharmaceutical composition that increases the action of the antibody, the functional fragment or the modified product,
(25)
A microtubule inhibitor comprising an antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9) A pharmaceutical composition that increases the action of the microtubule inhibitor when used in combination with
(26)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が組み合わせて投与されることを特徴とする医薬組成物、
(27)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、(26)に記載の医薬組成物、
(28)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が、単一の製剤に有効成分として含有されることを特徴とする、(26)に記載の医薬組成物、
(29) 
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が組み合わせて投与されることを特徴とする癌の治療方法、
(30)
 微小管阻害剤がパクリタキセル、ドセタキセル、ビンクリスチン、ビンブラスチン、エリブリン、ビンデシン及びビノレルビンよりなる群から選択される一つ又は二つ以上である、(22)乃至(28)のいずれか一つに記載の医薬組成物又は(29)に記載の治療方法、
(26)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and a microtubule inhibitor Are administered in combination, a pharmaceutical composition,
(27)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and a microtubule inhibitor Are contained in different preparations as active ingredients, and are administered at the same time or at different times, the pharmaceutical composition according to (26),
(28)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and a microtubule inhibitor Is contained in a single preparation as an active ingredient, The pharmaceutical composition according to (26),
(29)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and a microtubule inhibitor A method of treating cancer, characterized by being administered in combination,
(30)
The medicament according to any one of (22) to (28), wherein the microtubule inhibitor is one or more selected from the group consisting of paclitaxel, docetaxel, vincristine, vinblastine, eribulin, vindesine and vinorelbine. A composition or a treatment method according to (29),
(31)
 他剤がプラチナ製剤である、(1)乃至(12)のいずれか一つに記載の組合せ又は医薬組成物、
(32)
 プラチナ製剤と併用するための、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物、
(33)
 プラチナ製剤を含み、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物、
(34)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含み、プラチナ製剤と併用することにより、該プラチナ製剤の作用を上昇させる医薬組成物、
(35)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が組み合わせて投与されることを特徴とする医薬組成物、
(31)
The combination or pharmaceutical composition according to any one of (1) to (12), wherein the other agent is a platinum preparation,
(32)
The antibody or functional fragment thereof contained in the combination or composition described in any one of (1) to (8) or the modification contained in the combination or composition described in (9) for use in combination with a platinum preparation A pharmaceutical composition comprising the body,
(33)
In combination with an antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9) A pharmaceutical composition that increases the action of the antibody, the functional fragment or the modified product,
(34)
(1) The antibody or functional fragment thereof contained in the combination or composition according to any one of (8) or a functional fragment thereof, or the modified product contained in the combination or composition according to (9), and used in combination with a platinum preparation A pharmaceutical composition for increasing the action of the platinum preparation,
(35)
A combination of the antibody or the functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified product contained in the combination or composition according to (9), and a platinum preparation A pharmaceutical composition characterized by being administered,
(36)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、(35)に記載の医薬組成物、
(37)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が、単一の製剤に有効成分として含有されることを特徴とする、(35)に記載の医薬組成物、
(38)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が組み合わせて投与されることを特徴とする癌の治療方法
(39)
 プラチナ製剤がシスプラチン、オキサリプラチン、カルボプラチン及びネダプラチンよりなる群から選択される一つ又は二つ以上である、(31)乃至(37)のいずれか一つに記載の医薬組成物又は(38)に記載の治療方法、
(40)
 他剤が抗VEGFR2抗体である、(1)乃至(12)のいずれか一つに記載の組合せ又は医薬組成物、
(36)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and a platinum preparation, The pharmaceutical composition according to (35), which is contained as an active ingredient in different preparations and administered at the same time or at different times,
(37)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and a platinum preparation, The pharmaceutical composition according to (35), which is contained as an active ingredient in a single preparation,
(38)
A combination of the antibody or the functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified product contained in the combination or composition according to (9), and a platinum preparation (39) A method of treating cancer characterized by
The pharmaceutical composition according to any one of (31) to (37), wherein the platinum preparation is one or more selected from the group consisting of cisplatin, oxaliplatin, carboplatin and nedaplatin or (38) The treatment method described,
(40)
The combination or pharmaceutical composition according to any one of (1) to (12), wherein the other agent is an anti-VEGFR2 antibody,
(41)
 抗VEGFR2抗体と併用するための、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物、
(42)
 抗VEGFR2抗体を含み、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物、
(43)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含み、抗VEGFR2抗体と併用することにより、該抗VEGFR2抗体の作用を上昇させる医薬組成物、
(44)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が組み合わせて投与されることを特徴とする医薬組成物、
(45)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、(44)に記載の医薬組成物、
(41)
An antibody or a functional fragment thereof included in the combination or composition according to any one of (1) to (8) for use in combination with an anti-VEGFR2 antibody, or a combination or composition described in (9) A pharmaceutical composition comprising the modified form,
(42)
An anti-VEGFR2 antibody, an antibody or a functional fragment thereof included in the combination or composition according to any one of (1) to (8), or a modified form included in the combination or composition according to (9); A pharmaceutical composition that increases the action of the antibody, the functional fragment, or the modified product,
(43)
An antibody or a functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or a modified form contained in the combination or composition according to (9), and an anti-VEGFR2 antibody; A pharmaceutical composition that increases the action of the anti-VEGFR2 antibody when used in combination;
(44)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and an anti-VEGFR2 antibody A pharmaceutical composition characterized by being administered in combination;
(45)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and an anti-VEGFR2 antibody The pharmaceutical composition according to (44), which is contained as an active ingredient in different preparations and administered at the same time or at different times,
(46)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が、単一の製剤に有効成分として含有されることを特徴とする、(44)に記載の医薬組成物、
(47) 
(1)乃至(8)のいずれか一つに記載の組合せ又は医薬組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が組み合わせて投与されることを特徴とする癌の治療方法、
(48)
 抗VEGFR2抗体がラムシルマブである、(40)乃至(46)のいずれか一つに記載の医薬組成物又は(47)に記載の治療方法、
(49)
 他剤が5-フルオロウラシル系抗がん剤である、(1)乃至(12)のいずれか一つに記載の組合せ又は医薬組成物、
(50)
 5-フルオロウラシル系抗がん剤と併用するための、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物、
(46)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and an anti-VEGFR2 antibody The pharmaceutical composition according to (44), which is contained as an active ingredient in a single preparation,
(47)
The antibody or functional fragment thereof contained in the combination or pharmaceutical composition according to any one of (1) to (8) or the modified form contained in the combination or composition according to (9), and an anti-VEGFR2 antibody A method of treating cancer, characterized by being administered in combination,
(48)
The pharmaceutical composition according to any one of (40) to (46) or the treatment method according to (47), wherein the anti-VEGFR2 antibody is ramcilmab,
(49)
The combination or pharmaceutical composition according to any one of (1) to (12), wherein the other agent is a 5-fluorouracil anticancer agent,
(50)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) for use in combination with a 5-fluorouracil anticancer agent, or the combination according to (9) or A pharmaceutical composition comprising a modification contained in the composition;
(51)
 5-フルオロウラシル系抗がん剤を含み、(1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物、
(52)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体を含み、5-フルオロウラシル系抗がん剤と併用することにより、該5-フルオロウラシル系抗がん剤の作用を上昇させる医薬組成物、
(53)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が組み合わせて投与されることを特徴とする医薬組成物、
(54)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、(53)に記載の医薬組成物、
(55)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が、単一の製剤に有効成分として含有されることを特徴とする、(53)に記載の医薬組成物、
(51)
An antibody or a functional fragment thereof, or a combination or composition according to (9), which comprises a 5-fluorouracil anticancer agent and is contained in the combination or composition according to any one of (1) to (8) A pharmaceutical composition for increasing the action of the antibody, the functional fragment or the modified product by using in combination with the modified product,
(52)
A 5-fluorouracil system comprising an antibody or a functional fragment thereof included in the combination or composition according to any one of (1) to (8) or a modified substance included in the combination or composition according to (9) A pharmaceutical composition that increases the action of the 5-fluorouracil anticancer agent by using in combination with an anticancer agent;
(53)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and the 5-fluorouracil system A pharmaceutical composition, wherein anticancer agents are administered in combination;
(54)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and the 5-fluorouracil system The pharmaceutical composition according to (53), wherein the anticancer agent is contained as an active ingredient in different preparations and administered at the same time or at different times,
(55)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and the 5-fluorouracil system The pharmaceutical composition according to (53), wherein the anticancer agent is contained as an active ingredient in a single preparation,
(56)
 (1)乃至(8)のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は(9)に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が組み合わせて投与されることを特徴とする癌の治療方法、及び、
(57)
 5-フルオロウラシル系抗がん剤がフルオロウラシル、テガフール、テガフール・ギメラシル・オテラシルカリウム、テガフール・ウラシル、カペシタビン、カルモフール、ドキシフルリジンである、(49)乃至(55)のいずれか一つに記載の医薬組成物又は(56)に記載の治療方法、等に関する。
(56)
The antibody or functional fragment thereof contained in the combination or composition according to any one of (1) to (8) or the modified substance contained in the combination or composition according to (9), and the 5-fluorouracil system A method for treating cancer, comprising administering a combination of anticancer agents; and
(57)
The pharmaceutical composition according to any one of (49) to (55), wherein the 5-fluorouracil anticancer agent is fluorouracil, tegafur, tegafur, gimeracil, oteracil potassium, tegafur uracil, capecitabine, carmofur, or doxyfluridine. Or a treatment method according to (56).
 本発明の提供する抗体および他剤の組合せを用いることにより各種癌の治療または予防が可能となる。 It is possible to treat or prevent various cancers by using the combination of the antibody and other agents provided by the present invention.
ヒト化FR2-14抗体軽鎖(hFR2-14_L1)のCDR1のアミノ酸配列(配列表の配列番号1)CDR1 amino acid sequence of humanized FR2-14 antibody light chain (hFR2-14_L1) (SEQ ID NO: 1 in the sequence listing) ヒト化FR2-14抗体軽鎖(hFR2-14_L1)のCDR2のアミノ酸配列(配列表の配列番号2)Amino acid sequence of CDR2 of humanized FR2-14 antibody light chain (hFR2-14_L1) (SEQ ID NO: 2 in the sequence listing) ヒト化FR2-14抗体軽鎖(hFR2-14_L1)のCDR3のアミノ酸配列(配列表の配列番号3)CDR3 amino acid sequence of humanized FR2-14 antibody light chain (hFR2-14_L1) (SEQ ID NO: 3 in the Sequence Listing) ヒト化FR2-14抗体重鎖(hFR2-14_H19)のCDR1のアミノ酸配列(配列表の配列番号4)CDR1 amino acid sequence of humanized FR2-14 antibody heavy chain (hFR2-14_H19) (SEQ ID NO: 4 in the sequence listing) ヒト化FR2-14抗体重鎖(hFR2-14_H19)のCDR2のアミノ酸配列(配列表の配列番号5)CDR2 amino acid sequence of humanized FR2-14 antibody heavy chain (hFR2-14_H19) (SEQ ID NO: 5 in the sequence listing) ヒト化FR2-14抗体重鎖(hFR2-14_H19)のCDR3のアミノ酸配列(配列表の配列番号6)CDR3 amino acid sequence of humanized FR2-14 antibody heavy chain (hFR2-14_H19) (SEQ ID NO: 6 in the sequence listing) hFR2-14_L1のアミノ酸配列(配列表の配列番号8)。うちアミノ酸番号1乃至20はシグナル配列であり、通常大部分の成熟hFR2-14_L1のアミノ酸配列には含まれない。可変領域はアミノ酸番号21乃至130、定常領域はアミノ酸番号131乃至235。Amino acid sequence of hFR2-14_L1 (SEQ ID NO: 8 in the sequence listing). Among them, amino acid numbers 1 to 20 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_L1. The variable region is amino acid numbers 21 to 130, and the constant region is amino acid numbers 131 to 235. hFR2-14_H5のアミノ酸配列(配列表の配列番号10)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H5のアミノ酸配列には含まれない。可変領域はアミノ酸番号20乃至137、定常領域はアミノ酸番号138乃至467。Amino acid sequence of hFR2-14_H5 (SEQ ID NO: 10 in the sequence listing). Among them, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H5. The variable region is amino acid numbers 20 to 137, and the constant region is amino acid numbers 138 to 467. hFR2-14_H8のアミノ酸配列(配列表の配列番号12)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H8のアミノ酸配列には含まれない。可変領域はアミノ酸番号20乃至137、定常領域はアミノ酸番号138乃至467。Amino acid sequence of hFR2-14_H8 (SEQ ID NO: 12 in the sequence listing). Of these, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H8. The variable region is amino acid numbers 20 to 137, and the constant region is amino acid numbers 138 to 467. hFR2-14_H11のアミノ酸配列(配列表の配列番号14)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H11のアミノ酸配列には含まれない。可変領域はアミノ酸番号20乃至137、定常領域はアミノ酸番号138乃至467。Amino acid sequence of hFR2-14_H11 (SEQ ID NO: 14 in the sequence listing). Of these, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H11. The variable region is amino acid numbers 20 to 137, and the constant region is amino acid numbers 138 to 467. hFR2-14_H12のアミノ酸配列(配列表の配列番号16)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H12のアミノ酸配列には含まれない。可変領域はアミノ酸番号20乃至137、定常領域はアミノ酸番号138乃至467。The amino acid sequence of hFR2-14_H12 (SEQ ID NO: 16 in the sequence listing). Among them, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H12. The variable region is amino acid numbers 20 to 137, and the constant region is amino acid numbers 138 to 467. hFR2-14_H19のアミノ酸配列(配列表の配列番号18)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H19のアミノ酸配列には含まれない。可変領域はアミノ酸番号20乃至137、定常領域はアミノ酸番号138乃至467。Amino acid sequence of hFR2-14_H19 (SEQ ID NO: 18 in the sequence listing). Of these, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequence of most mature hFR2-14_H19. The variable region is amino acid numbers 20 to 137, and the constant region is amino acid numbers 138 to 467. ヒト胃癌細胞株SNU-16移植ヌードマウスにおける、hFR2-14_H19/L1単独投与、CPT-11単独投与およびhFR2-14_H19/L1とCPT-11の併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 10mg/kg、シンボル三角はCPT-11 40mg/kg、シンボル四角はhFR2-14_H19/L1 10mg/kg+CPT-11 40mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, CPT-11 single administration, and hFR2-14_H19 / L1 and CPT-11 combined administration in the human gastric cancer cell line SNU-16 transplanted nude mouse. No symbol indicates no treatment control group, symbol circle indicates hFR2-14_H19 / L1 10 mg / kg, symbol triangle indicates CPT-11 40 mg / kg, and symbol square indicates hFR2-14_H19 / L1 10 mg / kg + CPT-11 40 mg / kg. ヒト胃癌細胞株SNU-16移植NOD-scidマウスにおける、hFR2-14_H19/L1単独投与、Paclitaxel単独投与またはhFR2-14_H19/L1とPaclitaxelの併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 10mg/kg、シンボル三角はPaclitaxel 15mg/kg、シンボル四角はhFR2-14_H19/L1 10mg/kg+Pacltaxel 15mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, Paclitaxel single administration, or combined administration of hFR2-14_H19 / L1 and Paclitaxel in human gastric cancer cell line SNU-16 transplanted NOD-scid mouse. No symbol indicates an untreated control group, symbol circle indicates hFR2-14_H19 / L1 10 mg / kg, symbol triangle indicates Paclitaxel 15 mg / kg, symbol square indicates hFR2-14_H19 / L1 10 mg / kg + Pacltaxel 15 mg / kg. ヒト肺癌細胞株KNS-62移植NOD-scidマウスにおける、hFR2-14_H19/L1単独投与、Cisplatin単独投与またはhFR2-14_H19/L1とCisplatinの併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 10mg/kg、シンボル三角はCisplatin 5mg/kg、シンボル四角はhFR2-14_H19/L1 10mg/kg+Cisplatin 5mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, Cisplatin single administration, or combined administration of hFR2-14_H19 / L1 and Cisplatin in the human lung cancer cell line KNS-62 transplanted NOD-scid mouse. No symbol indicates an untreated control group, symbol circle indicates hFR2-14_H19 / L1 10 mg / kg, symbol triangle indicates Ciplatin 5 mg / kg, symbol square indicates hFR2-14_H19 / L1 10 mg / kg + Cisplatin 5 mg / kg. ヒト化FR2-14重鎖(hFR2-14_H9)のアミノ酸配列(配列番号20)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H 9のアミノ酸配列には含まれない。可変領域はアミノ酸番号20乃至137、定常領域はアミノ酸番号138乃至467。Amino acid sequence of humanized FR2-14 heavy chain (hFR2-14_H9) (SEQ ID NO: 20). Among them, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequence of most mature hFR2-14_H9. The variable region is amino acid numbers 20 to 137, and the constant region is amino acid numbers 138 to 467. ヒト胃癌細胞株SNU-16移植ヌードマウスにおける、hFR2-14_H19/L1単独投与、Cisplatin単独投与またはhFR2-14_H19/L1とCisplatinの併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 1mg/kg、シンボル三角はCisplatin 5mg/kg、シンボル四角はhFR2-14_H19/L1 1mg/kg+Cisplatin 5mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, Cisplatin single administration, or combined administration of hFR2-14_H19 / L1 and Cisplatin in a human gastric cancer cell line SNU-16 transplanted nude mouse. No symbol indicates an untreated control group, symbol circle indicates hFR2-14_H19 / L1 1 mg / kg, symbol triangle indicates Ciplatin 5 mg / kg, symbol square indicates hFR2-14_H19 / L1 1 mg / kg + Cisplatin 5 mg / kg. ヒト胃癌細胞株SNU-16移植ヌードマウスにおける、hFR2-14_H19/L1単独投与、Docetaxel単独投与またはhFR2-14_H19/L1とDocetaxelの併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 1mg/kg、シンボル三角はDocetaxel 1mg/kg、シンボル四角はhFR2-14_H19/L1 1mg/kg+Docetaxel 1mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, Docetaxel single administration, or combined administration of hFR2-14_H19 / L1 and Docetaxel in the human gastric cancer cell line SNU-16 transplanted nude mouse. No symbol indicates an untreated control group, symbol circles indicate hFR2-14_H19 / L1 1 mg / kg, symbol triangles indicate Docetaxel 1 mg / kg, and symbol squares indicate hFR2-14_H19 / L1 1 mg / kg + Doctaxel 1 mg / kg. ヒト胃癌細胞株SNU-16移植ヌードマウスにおける、hFR2-14_H19/L1単独投与、抗VEGFR2抗体単独投与またはhFR2-14_H19/L1と抗VEGFR2抗体の併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 1mg/kg、シンボル三角は抗VEGFR2抗体 20mg/kg、シンボル四角はhFR2-14_H19/L1 1mg/kg+抗VEGFR2抗体 20mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, anti-VEGFR2 antibody single administration, or combined administration of hFR2-14_H19 / L1 and an anti-VEGFR2 antibody in the human gastric cancer cell line SNU-16 transplanted nude mouse. No symbol indicates an untreated control group, symbol circle indicates hFR2-14_H19 / L1 1 mg / kg, symbol triangle indicates anti-VEGFR2 antibody 20 mg / kg, and symbol square indicates hFR2-14_H19 / L1 1 mg / kg + anti-VEGFR2 antibody 20 mg / kg. ヒト胃癌細胞株SNU-16移植ヌードマウスにおける、hFR2-14_H19/L1単独投与、5-FU単独投与またはhFR2-14_H19/L1と5-FUの併用投与でのin vivo抗腫瘍効果を示した図。シンボル無しは無処置対照群、シンボル丸はhFR2-14_H19/L1 1mg/kg、シンボル三角は5-FU 50mg/kg、シンボル四角はhFR2-14_H19/L1 1mg/kg+5-FU 50mg/kgを示す。The figure which showed the in vivo anti-tumor effect by hFR2-14_H19 / L1 single administration, 5-FU single administration, or hFR2-14_H19 / L1 and 5-FU combined administration in the human stomach cancer cell line SNU-16 transplanted nude mouse. No symbol indicates no treatment control group, symbol circle indicates hFR2-14_H19 / L1 1 mg / kg, symbol triangle indicates 5-FU 50 mg / kg, symbol square indicates hFR2-14_H19 / L1 1 mg / kg + 5-FU 50 mg / kg.
 1.定義
 本発明において、「遺伝子」とは、蛋白質のアミノ酸をコードする塩基配列が含まれるヌクレオチドまたはその相補鎖を意味し、例えば、蛋白質のアミノ酸をコードする塩基配列が含まれるヌクレオチドまたはその相補鎖であるポリヌクレオチド、オリゴヌクレオチド、DNA、mRNA、cDNA、cRNA等は「遺伝子」の意味に含まれる。かかる遺伝子は一本鎖、二本鎖又は三本鎖以上のヌクレオチドであり、DNA鎖とRNA鎖の会合体、一本のヌクレオチド鎖上にリボヌクレオチド(RNA)とデオキシリボヌクレオチド(DNA)が混在するもの及びそのようなヌクレオチド鎖を含む二本鎖又は三本鎖以上のヌクレオチドも「遺伝子」の意味に含まれる。本発明の「FGFR2遺伝子」としては、例えば、FGFR2蛋白質のアミノ酸配列をコードする塩基配列が含まれるDNA、mRNA、cDNA、cRNA等をあげることができる。
1. Definitions In the present invention, “gene” means a nucleotide containing a nucleotide sequence encoding a protein amino acid or a complementary strand thereof, for example, a nucleotide containing a nucleotide sequence encoding a protein amino acid or a complementary strand thereof. Certain polynucleotides, oligonucleotides, DNA, mRNA, cDNA, cRNA and the like are included in the meaning of “gene”. Such a gene is a single-stranded, double-stranded, or triple-stranded nucleotide, and an assembly of a DNA strand and an RNA strand, and ribonucleotide (RNA) and deoxyribonucleotide (DNA) are mixed on one nucleotide strand. Also included within the meaning of “gene” are double-stranded or triple-stranded nucleotides comprising such nucleotide chains. Examples of the “FGFR2 gene” of the present invention include DNA, mRNA, cDNA, cRNA and the like containing a base sequence encoding the amino acid sequence of the FGFR2 protein.
 本発明において、「ヌクレオチド」、「核酸」および「核酸分子」は同義であり、例えば、DNA、RNA、プローブ、オリゴヌクレオチド、ポリヌクレオチド、プライマー等も「ヌクレオチド」の意味に含まれる。かかるヌクレオチドは一本鎖、二本鎖又は三本以上の鎖からなるヌクレオチドであり、DNA鎖とRNA鎖の会合体、一本のヌクレオチド鎖上にリボヌクレオチド(RNA)とデオキシリボヌクレオチド(DNA)が混在するもの及びそのようなヌクレオチド鎖を含む二本鎖又は三本以上の鎖の会合体も「ヌクレオチド」の意味に含まれる。 In the present invention, “nucleotide”, “nucleic acid” and “nucleic acid molecule” are synonymous, and for example, DNA, RNA, probe, oligonucleotide, polynucleotide, primer and the like are also included in the meaning of “nucleotide”. Such nucleotide is a nucleotide composed of a single strand, a double strand, or three or more strands, and an assembly of a DNA strand and an RNA strand, and ribonucleotide (RNA) and deoxyribonucleotide (DNA) on a single nucleotide strand. Also included within the meaning of “nucleotide” are those that are intermingled and aggregates of two or more strands containing such nucleotide strands.
 本発明において、「ポリペプチド」、「ペプチド」および「蛋白質」は同義である。 In the present invention, “polypeptide”, “peptide” and “protein” are synonymous.
 本発明において、「抗原」を「免疫原」の意味に用いることがある。 In the present invention, “antigen” is sometimes used to mean “immunogen”.
 本発明において、「細胞」には、動物個体に由来する各種細胞、継代培養細胞、初代培養細胞、細胞株、組換え細胞及び微生物等も含まれる。 In the present invention, “cell” includes various cells derived from individual animals, subculture cells, primary culture cells, cell lines, recombinant cells, microorganisms, and the like.
 本発明においては、FGFR2、FGFR2IIIb、FGFR2IIIc、FGFR3、FGFRs等を認識する抗体を、それぞれ「抗FGFR2抗体」、「抗FGFR2IIIb抗体」、「抗FGFR2IIIc抗体」、「抗FGFR3抗体」及び「抗FGFRs抗体」等と表記することがある。かかる抗体には、キメラ化抗体、ヒト化抗体、ヒト抗体等が含まれる。 In the present invention, antibodies that recognize FGFR2, FGFR2IIIb, FGFR2IIIc, FGFR3, FGFRs, etc. are designated as “anti-FGFR2 antibody”, “anti-FGFR2IIIb antibody”, “anti-FGFR2IIIc antibody”, “anti-FGFR3 antibody” and “anti-FGFRs antibody”, respectively. Or the like. Such antibodies include chimerized antibodies, humanized antibodies, human antibodies and the like.
 本発明における「抗体の機能断片」とは、元の抗体が奏する機能の少なくとも一部を奏する抗体断片を意味する。「抗体の機能断片」としては、例えば、Fab、F(ab’)2、scFv、Fab’、一本鎖免疫グロブリン等をあげることができるが、それらに限定されるものではない。かかる抗体の機能断片は、抗体蛋白質の全長分子をパパイン、ペプシン等の酵素で処理することによって得られたものに加え、組換え遺伝子を用いて適当な宿主細胞において産生された組換え蛋白質であってもよい。 In the present invention, the “functional fragment of an antibody” means an antibody fragment that exhibits at least a part of the function exhibited by the original antibody. Examples of the “functional fragment of an antibody” include, but are not limited to, Fab, F (ab ′) 2, scFv, Fab ′, single chain immunoglobulin and the like. Such functional fragments of antibodies are recombinant proteins produced in appropriate host cells using recombinant genes in addition to those obtained by treating full-length antibody protein molecules with enzymes such as papain and pepsin. May be.
 本発明において、抗体が結合する「部位」、すなわち抗体が認識する「部位」とは、抗体が結合又は認識する抗原上の部分ペプチド又は部分高次構造を意味する。本発明においては、かかる部位のことをエピトープ、抗体の結合部位とも呼ぶ。本発明の抗FGFR2抗体が結合又は認識するFGFR2蛋白質上の部位としては、FGFR2蛋白質上の部分ペプチド又は部分高次構造等を例示することができる。 In the present invention, the “site” to which the antibody binds, that is, the “site” recognized by the antibody means a partial peptide or a partial higher order structure on the antigen to which the antibody binds or recognizes. In the present invention, such a site is also referred to as an epitope or an antibody binding site. Examples of the site on the FGFR2 protein to which the anti-FGFR2 antibody of the present invention binds or recognizes include a partial peptide or a partial higher order structure on the FGFR2 protein.
 抗体分子の重鎖及び軽鎖にはそれぞれ3箇所の相補性決定領域(CDR:Complemetarity determining region)があることが知られている。相補性決定領域は、超可変領域(hypervariable domain)とも呼ばれ、抗体の重鎖及び軽鎖の可変領域内にあって、一次構造の変異性が特に高い部位であり、重鎖及び軽鎖のポリペプチド鎖の一次構造上において、通常、それぞれ3ヶ所に分離している。本発明においては、抗体の相補性決定領域について、重鎖の相補性決定領域を重鎖アミノ酸配列のアミノ末端側からCDRH1、CDRH2、CDRH3と表記し、軽鎖の相補性決定領域を軽鎖アミノ酸配列のアミノ末端側からCDRL1、CDRL2、CDRL3と表記する。これらの部位は立体構造の上で相互に近接し、結合する抗原に対する特異性を決定している。 It is known that there are three complementarity determining regions (CDRs) in each of the heavy and light chains of an antibody molecule. The complementarity-determining region is also called a hypervariable domain, and is located in the variable region of the heavy and light chains of the antibody and has a particularly high primary structure variability. In the primary structure of the polypeptide chain, it is usually separated at three points. In the present invention, for the complementarity determining region of an antibody, the complementarity determining region of the heavy chain is denoted as CDRH1, CDRH2, CDRH3 from the amino terminal side of the heavy chain amino acid sequence, and the complementarity determining region of the light chain is defined as the light chain amino acid. CDRL1, CDRL2, and CDRL3 are represented from the amino terminal side of the sequence. These sites are close to each other on the three-dimensional structure and determine the specificity for the antigen to be bound.
 本発明において、「抗体変異体」とは、元の抗体が有するアミノ酸配列においてアミノ酸が置換、欠失、付加及び/又は挿入(以下、「変異」と総称する)してなるアミノ酸配列を有し、且つ本発明のFGFR2蛋白質に結合するポリペプチドを意味する。かかる抗体変異体における変異アミノ酸の数は、1乃至2、3、4、5、6、7、8、9、10、12、15、20、25、30、40又は50個である。かかる抗体変異体も本発明の「抗体」に包含される。  In the present invention, the “antibody variant” has an amino acid sequence in which amino acids are substituted, deleted, added and / or inserted (hereinafter collectively referred to as “mutation”) in the amino acid sequence of the original antibody. And a polypeptide that binds to the FGFR2 protein of the present invention. The number of mutated amino acids in such antibody variants is 1 to 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, 40 or 50. Such antibody variants are also included in the “antibody” of the present invention. *
 本発明において、「1乃至数個」における「数個」とは、3乃至10個を指す。 In the present invention, “several” in “1 to several” refers to 3 to 10.
 本発明の抗体が奏する活性・性質としては、例えば、生物的活性、理化学的性質等を挙げることができ、具体的には、各種生物活性、抗原やエピトープに対する結合活性、製造や保存時における安定性、熱安定性等をあげることができる。 Examples of the activity / property exhibited by the antibody of the present invention include biological activity, physicochemical properties, and the like. Specifically, various biological activities, binding activity to antigens and epitopes, stability during production and storage Property, heat stability, etc. can be raised.
 本発明において、「ストリンジェントな条件下でハイブリダイズする」とは、5×SSCを含む溶液中で65℃にてハイブリダイゼーションを行い、ついで2×SSC-0.1%SDSを含む水溶液中で65℃にて20分間、0.5×SSC-0.1%SDSを含む水溶液中で65℃にて20分間、ならびに、0.2×SSC-0.1%SDSを含む水溶液中で65℃にて20分間、それぞれ洗浄する条件又はそれと同等の条件でハイブリダイズすることを意味する。SSCとは150mM NaCl-15mMクエン酸ナトリウムの水溶液であり、n×SSCはn倍濃度のSSCを意味する。 In the present invention, “hybridize under stringent conditions” means hybridization at 65 ° C. in a solution containing 5 × SSC, and then in an aqueous solution containing 2 × SSC-0.1% SDS. 20 minutes at 65 ° C. in an aqueous solution containing 0.5 × SSC-0.1% SDS for 20 minutes at 65 ° C. and 65 ° C. in an aqueous solution containing 0.2 × SSC-0.1% SDS Means to hybridize under the conditions of washing for 20 minutes or under equivalent conditions. SSC is an aqueous solution of 150 mM NaCl-15 mM sodium citrate, and nx SSC means an n-fold concentration of SSC.
 本発明において「細胞傷害」とは、何らかの形で、細胞に病理的な変化をもたらすことを指し、直接的な外傷にとどまらず、DNAの切断や塩基の二量体の形成、染色体の切断、細胞分裂装置の損傷、各種酵素活性の低下などあらゆる細胞の構造や機能上の損傷を意味する。本発明において「細胞傷害活性」とは上記細胞傷害を引き起こすことを意味する。 In the present invention, “cytotoxicity” refers to causing a pathological change in a cell in some form, and is not limited to direct trauma, but also includes DNA breakage, base dimer formation, chromosome breakage, It means any structural or functional damage of cells, such as damage to cell division apparatus or reduction of various enzyme activities. In the present invention, “cytotoxic activity” means causing the above cytotoxicity.
 本発明において「抗体依存性細胞傷害活性」とは、「antibody dependent cellular cutotoxicity(ADCC)活性」を指し、NK細胞が抗体を介して腫瘍細胞等の標的細胞を傷害する作用活性を意味する。 In the present invention, “antibody-dependent cytotoxic activity” refers to “antibody dependent cellular toxicity (ADCC) activity” and means an activity in which NK cells damage target cells such as tumor cells via antibodies.
 本発明において「抗体依存性細胞媒介食活性」とは「andibody dependent cell phagocytosis(ADCP)活性」を指し、単球やマクロファージ細胞が抗体を介して腫瘍細胞等の標的細胞を貪食する作用活性を意味する。「抗体依存性貪食活性」ともいう。 In the present invention, “antibody-dependent cell-mediated phagocytic activity” refers to “andibody dependent cell phagocytosis (ADCP) activity”, which means that monocyte or macrophage cells phagocytose target cells such as tumor cells via antibodies. To do. Also called “antibody-dependent phagocytic activity”.
 本発明において「補体依存性細胞傷害作用活性」とは、「complement dependent cytotoxicity(CDC)活性」を指し、補体が抗体を介して腫瘍細胞等の標的細胞を傷害する作用活性を意味する。 In the present invention, “complement-dependent cytotoxic activity” refers to “complement dependent cytotoxicity (CDC) activity” and means an activity that complements a target cell such as a tumor cell via an antibody.
 本発明において「癌」と「腫瘍」は同じ意味で用いられる。 In the present invention, “cancer” and “tumor” are used interchangeably.
 本発明において「他の有効成分」又は「他の薬剤」とは、抗FGFR2抗体と組み合わせて使われるか、または本発明の医薬組成物に抗FGFR2抗体とともに含まれる疾患の治療又は予防活性を有する成分又は薬剤である。好適な態様において、かかる組合せまたは医薬組成物は癌の治療又は予防に有効であり、より好適な態様において、医薬組成物は癌の治療又は予防において相乗効果を示す。 In the present invention, “other active ingredient” or “other drug” is used in combination with an anti-FGFR2 antibody, or has a therapeutic or prophylactic activity for a disease included in the pharmaceutical composition of the present invention together with an anti-FGFR2 antibody. An ingredient or drug. In preferred embodiments, such a combination or pharmaceutical composition is effective in treating or preventing cancer, and in more preferred embodiments, the pharmaceutical composition exhibits a synergistic effect in treating or preventing cancer.
 本発明において「相乗効果」とは、複数の有効成分を組み合わせて使用した場合または複数の有効成分を1つの医薬組成物に含有せしめた場合、それぞれを有効成分が示す抗癌活性を加算したより強い抗癌活性を示すことをいう。例えば、有効成分AおよびBの奏する抗癌活性がそれぞれaおよびbであって、AとBを組み合わせて使用するかまたはAとBを1つの医薬組成物に含有せしめて使用した場合に、その抗癌活性がaとbを加算したより大きければ、該組合せまたは医薬組成物は相乗効果を奏するとみなすことができる。別の例として、有効成分AおよびBの奏する抗癌活性がそれぞれaおよびゼロ(0:すなわちBが抗癌活性を奏さない濃度)であって、AとBを組み合わせて使用しするかまたはAとBを1つの医薬組成物に含有せしめた場合、その抗癌活性がaより大きければ、該組合せまたは医薬組成物は相乗効果を奏するとみなすことができる。また別の例として、有効成分AおよびBの奏する抗癌活性がいずれもゼロ(0:すなわちAとBがいずれも抗癌活性を奏さない濃度)であって、AとBを組み合わせて使用しするかまたはAとBを1つの医薬組成物に含有せしめた場合、その抗癌活性がゼロ(0)より大きければ、該組合せまたは医薬組成物は相乗効果を奏するとみなすことができる。 In the present invention, “synergistic effect” means that when a plurality of active ingredients are used in combination or when a plurality of active ingredients are contained in one pharmaceutical composition, the anticancer activity indicated by each active ingredient is added to each. It refers to showing strong anticancer activity. For example, when the anticancer activities exhibited by the active ingredients A and B are a and b, respectively, and A and B are used in combination or A and B are contained in one pharmaceutical composition, If the anticancer activity is greater than the sum of a and b, the combination or pharmaceutical composition can be considered to have a synergistic effect. As another example, the anticancer activities exhibited by the active ingredients A and B are a and zero (0: that is, B is a concentration at which B does not exhibit anticancer activity), respectively, and A and B are used in combination or A When B and B are contained in one pharmaceutical composition, if the anticancer activity is greater than a, the combination or the pharmaceutical composition can be regarded as having a synergistic effect. As another example, the anticancer activity exhibited by active ingredients A and B is both zero (0: that is, the concentration at which neither A nor B exhibits anticancer activity), and A and B are used in combination. Or if A and B are included in one pharmaceutical composition, the combination or pharmaceutical composition can be considered to have a synergistic effect if its anti-cancer activity is greater than zero (0).
 2.抗原蛋白質
(2-1)特性
 FGFRsは線維芽細胞増殖因子(FGF)に結合する受容体蛋白質である。本発明において、FGFRsは脊椎動物由来、好適には哺乳動物由来であり、より好適にはヒト由来である。ヒトFGF及びFGFRsは、22のFGF(FGF1乃至14、および、FGF16乃至23)、ならびにチロシンキナーゼドメインを有する4つのFGFR(FGFR1乃至4)にそれぞれ分類される。FGFRsは、2つあるいは3つの免疫グロブリン様ドメイン(IgD1乃至3)から成るリガンド結合部位を含む細胞外領域、1回膜貫通領域、およびチロシンキナーゼドメインを含む細胞内領域から構成されている。このうち、FGFR1、FGFR2およびFGFR3には、IIIbおよびIIIcと呼ばれる各2つのスプライシングバリアントが存在する。これらのアイソフォームはIgD3の後半において約50のアミノ酸配列が異なっており、異なる組織分布およびリガンド特異性を示す。FGFRsは次のような活性を有する:(1)FGFに結合する;(2)かかる結合によりFGFRsは二量体化する;(3)かかる二量体化によりFGFRsの特定のチロシン残基がリン酸化される;(4)かかるリン酸化によりFGFR基質2α(FRS2α)等のアダプター蛋白質のリクルートが促進される;(5)該FGFRsを発現している細胞又は組織にFGF刺激によるシグナルを伝達するか又はシグナル伝達を活性化する。
本発明においてFGFR2蛋白質は以下の性質を有する。
(i)FGFに結合する。
FGFR2IIIb蛋白質は、通常、FGF1、FGF3、FGF7(KGF)、FGF10,FGF22及びFGF23からなる群より選択される1又は2以上に結合するが、他のFGFに結合してもよく、変異型の場合は前記群に含まれるFGFであっても結合しないことがある。
2. Antigen protein (2-1) characteristics FGFRs are receptor proteins that bind to fibroblast growth factor (FGF). In the present invention, FGFRs are derived from vertebrates, preferably from mammals, and more preferably from humans. Human FGFs and FGFRs are classified into 22 FGFs (FGF1 to 14 and FGF16 to 23), and 4 FGFRs having a tyrosine kinase domain (FGFR1 to 4), respectively. FGFRs are composed of an extracellular region containing a ligand binding site consisting of two or three immunoglobulin-like domains (IgD1 to 3), a single transmembrane region, and an intracellular region containing a tyrosine kinase domain. Among these, FGFR1, FGFR2 and FGFR3 have two splicing variants called IIIb and IIIc, respectively. These isoforms differ by about 50 amino acid sequences in the second half of IgD3 and show different tissue distribution and ligand specificity. FGFRs have the following activities: (1) bind to FGF; (2) FGFRs dimerize by such binding; (3) such dimerization causes specific tyrosine residues of FGFRs to be phosphorylated. (4) The phosphorylation promotes recruitment of adapter proteins such as FGFR substrate 2α (FRS2α); (5) Does FGF-stimulated signals or signals transmit to cells or tissues expressing the FGFRs? Or activate signaling.
In the present invention, the FGFR2 protein has the following properties.
(I) binds to FGF.
The FGFR2IIIb protein normally binds to one or more selected from the group consisting of FGF1, FGF3, FGF7 (KGF), FGF10, FGF22, and FGF23, but may bind to other FGFs, and in the case of a mutant type May not bind even to FGF included in the group.
 FGFR2IIIc蛋白質は、通常、FGF1、FGF2、FGF4、FGF6、FGF9、FGF17、FGF18、FGF21、FGF23からなる群より選択される1又は2以上に結合する。他のFGFに結合してもよく、変異型の場合は前記群に含まれるFGFであっても結合しないことがある。
(ii)FGFR2発現細胞又は組織内にFGF刺激により生じるシグナルを伝達する。
The FGFR2IIIc protein normally binds to one or more selected from the group consisting of FGF1, FGF2, FGF4, FGF6, FGF9, FGF17, FGF18, FGF21, and FGF23. It may bind to other FGF, and in the case of a mutant type, it may not bind even if it is an FGF included in the group.
(Ii) Transmit signals generated by FGF stimulation into FGFR2-expressing cells or tissues.
 FGF刺激により生じるシグナル伝達としては、特に限定されるものではないが、例えば、FGFR2自己リン酸化、FGFR基質のリクルート及びその促進、それらを介したMAPK,PI3K、Akt、細胞外シグナル制御キナーゼ(ERK)等のシグナル経路の活性化等をあげることができる。FGFR基質としては、FGFR基質2α(FRS2α)等を例示することができる。 Signal transduction caused by FGF stimulation is not particularly limited. For example, FGFR2 autophosphorylation, FGFR substrate recruitment and promotion thereof, MAPK, PI3K, Akt, extracellular signal-regulated kinase (ERK) via them. ) And the like. Examples of the FGFR substrate include FGFR substrate 2α (FRS2α).
 かかるシグナル伝達の活性化及びその抑制を評価するための試験方法は、特に限定されものではなく、公知の方法から任意に選択することができるが、例えば、ERKシグナル伝達を評価する系として、後述のElk1ルシフェラーゼレポーターアッセイをあげることができる。
(iii)本発明においてFGFR2IIIb蛋白質は、次の(a)乃至(d)のいずれか一つに記載のアミノ酸配列(以下、「FGFR2IIIbアミノ酸配列」という)を含んでなるか、FGFR2IIIbアミノ酸配列を含むアミノ酸配列からなるか、またはFGFR2IIIbアミノ酸配列からなる:
(a)NP_075259のアミノ酸配列;
(b)NP_075259のアミノ酸配列と80%、82%、84%、86%、88%、90%、92%、94%、96%、98%又は99%以上の配列同一性を示し且つFGF結合活性を有するポリペプチドのアミノ酸配列;
(c)NP_075259のアミノ酸配列において1乃至50,40、35、30、25、20、15、12、10、8、6、5、4、3もしくは2個、または1個のアミノ酸が置換、欠失、付加または挿入してなり且つFGF結合活性を有するポリペプチドのアミノ酸配列;及び、
(d)NP_075259のアミノ酸配列をコードする塩基配列に相補的な塩基配列を有するヌクレオチドとストリンジェントな条件下でハイブリダイズするヌクレオチドの有する塩基配列によりコードされ、且つ、FGF結合活性を有するポリペプチドのアミノ酸配列。
The test method for evaluating the activation of such signal transduction and its suppression is not particularly limited and can be arbitrarily selected from known methods. For example, a system for evaluating ERK signal transduction is described later. Elk1 luciferase reporter assay.
(Iii) In the present invention, the FGFR2IIIb protein comprises the amino acid sequence described in any one of the following (a) to (d) (hereinafter referred to as “FGFR2IIIb amino acid sequence”) or comprises the FGFR2IIIb amino acid sequence: Consisting of the amino acid sequence or consisting of the FGFR2IIIb amino acid sequence:
(A) the amino acid sequence of NP — 075259;
(B) 80%, 82%, 84%, 86%, 88%, 90%, 92%, 94%, 96%, 98% or 99% sequence identity with NP — 075259 amino acid sequence and FGF binding An amino acid sequence of a polypeptide having activity;
(C) In the amino acid sequence of NP — 075259, 1 to 50, 40, 35, 30, 25, 20, 15, 12, 10, 8, 6, 5, 4, 3 or 2, or 1 amino acid is substituted or missing An amino acid sequence of a polypeptide that is deleted, added or inserted and has FGF binding activity; and
(D) A polypeptide encoded by a nucleotide sequence of a nucleotide that hybridizes under stringent conditions with a nucleotide having a nucleotide sequence complementary to the nucleotide sequence encoding the amino acid sequence of NP — 075259 and having an FGF binding activity Amino acid sequence.
 (b)乃至(d)記載のいずれか一つに記載のポリペプチドは、FGF結合活性に加え、FGFR2の有する他の活性を有していてもよい。 The polypeptide described in any one of (b) to (d) may have other activities possessed by FGFR2 in addition to the FGF binding activity.
 本発明においてFGFR2IIIc蛋白質は、次の(a)乃至(d)のいずれか一つに記載のアミノ酸配列(以下、「FGFR2IIIcアミノ酸配列」という)を含んでなるか、FGFR2IIIcアミノ酸配列を含むアミノ酸配列からなるか、またはFGFR2IIIcアミノ酸配列からなる:
(a)NP_000132のアミノ酸配列;
(b)NP_000132のアミノ酸配列と80%以上、82%以上、84%以上、86%以上、88%以上、90%以上、92%以上、94%以上、96%以上、98%以上又は99%以上の配列同一性を示し且つFGF結合活性を有するポリペプチドのアミノ酸配列;
(c)NP_000132のアミノ酸配列において1乃至50、45、40、35、30、25、20、15、12、10、8、6、5、4、3もしくは2個、または1個のアミノ酸が置換、欠失、付加または挿入してなり且つFGF結合活性を有するポリペプチドのアミノ酸配列;及び、
(d)NP_000132のアミノ酸配列をコードする塩基配列に相補的な塩基配列を有するヌクレオチドとストリンジェントな条件下でハイブリダイズするヌクレオチドの有する塩基配列によりコードされ、且つ、FGF結合活性を有するポリペプチドのアミノ酸配列。
In the present invention, the FGFR2IIIc protein comprises the amino acid sequence described in any one of the following (a) to (d) (hereinafter referred to as “FGFR2IIIc amino acid sequence”) or is derived from an amino acid sequence comprising the FGFR2IIIc amino acid sequence. Or consists of the FGFR2IIIc amino acid sequence:
(A) the amino acid sequence of NP_000132;
(B) NP — 000132 amino acid sequence and 80% or higher, 82% or higher, 84% or higher, 86% or higher, 88% or higher, 90% or higher, 92% or higher, 94% or higher, 96% or higher, 98% or higher, or 99% An amino acid sequence of a polypeptide having the above sequence identity and having FGF binding activity;
(C) In the amino acid sequence of NP — 000132, 1 to 50, 45, 40, 35, 30, 25, 20, 15, 12, 10, 8, 6, 5, 4, 3 or 2 or 1 amino acid is substituted. An amino acid sequence of a polypeptide that is deleted, added or inserted and has FGF binding activity; and
(D) A polypeptide encoded by a nucleotide sequence of a nucleotide that hybridizes under stringent conditions with a nucleotide having a nucleotide sequence complementary to the nucleotide sequence encoding the amino acid sequence of NP_000132 and having an FGF binding activity Amino acid sequence.
 (b)乃至(d)記載のいずれか一つに記載のポリペプチドは、FGF結合活性に加え、FGFR2の有する他の活性を有していてもよい。 The polypeptide described in any one of (b) to (d) may have other activities possessed by FGFR2 in addition to the FGF binding activity.
 本発明のFGFR2蛋白質は天然型(非組換型)及び組換え型のいずれであってもよい。また、担体やタグなど他のペプチドや蛋白質との融合物もFGFR2蛋白質の意味に含まれる。さらに、PEG等のポリマー付加を含む化学修飾、及び/又は、糖鎖修飾を含む生物学的修飾がなされたものもFGFR2蛋白質の意味に含まれる。また、FGFR2蛋白質断片も本発明のFGFR2蛋白質の意味に含まれる。FGFR2蛋白質断片のうち上記(i)及び/又は(ii)に記載の性質を具備したものをFGFR2蛋白質機能断片と呼ぶ。
(2-2)抗原遺伝子
 本発明においてFGFR2IIIb遺伝子は、FGFR2IIIbアミノ酸配列をコードする塩基配列(以下、「FGFR2遺伝子配列」という)を含んでなるか、FGFR2遺伝子配列を含む塩基配列からなるか、またはFGFR2遺伝子配列からなる。 本発明においてFGFR2IIIc遺伝子は、FGFR2IIIcアミノ酸配列をコードする塩基配列(以下、「FGFR2遺伝子配列」という)を含んでなるか、FGFR2遺伝子配列を含む塩基配列からなるか、またはFGFR2遺伝子配列からなる。
(2-3)抗原蛋白質の調製
 本発明のFGFR2蛋白質は、動物組織(体液を含む)、該組織由来の細胞もしくは該細胞培養物からの精製及び単離、遺伝子組換え、インビトロ翻訳、化学合成等により調製することができる。
The FGFR2 protein of the present invention may be either natural (non-recombinant) or recombinant. In addition, fusion with other peptides and proteins such as carriers and tags are also included in the meaning of the FGFR2 protein. Furthermore, the chemical modification including addition of a polymer such as PEG and / or biological modification including sugar chain modification is also included in the meaning of the FGFR2 protein. FGFR2 protein fragments are also included in the meaning of the FGFR2 protein of the present invention. Among the FGFR2 protein fragments, those having the properties described in (i) and / or (ii) above are referred to as FGFR2 protein functional fragments.
(2-2) Antigen gene In the present invention, the FGFR2IIIb gene comprises a base sequence encoding the FGFR2IIIb amino acid sequence (hereinafter referred to as "FGFR2 gene sequence"), consists of a base sequence containing the FGFR2 gene sequence, or It consists of the FGFR2 gene sequence. In the present invention, the FGFR2IIIc gene comprises a base sequence encoding the FGFR2IIIc amino acid sequence (hereinafter referred to as “FGFR2 gene sequence”), consists of a base sequence containing the FGFR2 gene sequence, or consists of an FGFR2 gene sequence.
(2-3) Preparation of antigen protein The FGFR2 protein of the present invention is purified and isolated from animal tissues (including body fluids), cells derived from the tissues or cell cultures, genetic recombination, in vitro translation, chemical synthesis. Etc. can be prepared.
 3.抗体
(3-1)抗体の分類
 本発明の抗体は、モノクローナル抗体およびポリクローナル抗体のいずれであってもよい。本発明のモノクローナル抗体としては、非ヒト動物由来の抗体(非ヒト動物抗体)、ヒト由来の抗体(ヒト抗体)、キメラ化抗体(「キメラ抗体」ともいう)、ヒト化抗体等をあげることができる。
3. Antibody (3-1) Antibody Classification The antibody of the present invention may be either a monoclonal antibody or a polyclonal antibody. Examples of the monoclonal antibody of the present invention include non-human animal-derived antibodies (non-human animal antibodies), human-derived antibodies (human antibodies), chimerized antibodies (also referred to as “chimeric antibodies”), humanized antibodies, and the like. it can.
 非ヒト動物抗体としては、哺乳類、鳥類等の脊椎動物に由来する抗体等をあげることができる。哺乳類由来の抗体としては、マウス抗体、ラット抗体等げっ歯類由来の抗体等をあげることができる。鳥類由来の抗体としては、ニワトリ抗体等をあげることができる。抗ヒトFGFR2ラットモノクローナル抗体としては、FR2-10、FR2-13、FR2-14(WO2013/154206)等をあげることができる。 Examples of non-human animal antibodies include antibodies derived from vertebrates such as mammals and birds. Examples of the antibody derived from mammals include antibodies derived from rodents such as mouse antibodies and rat antibodies. Examples of avian-derived antibodies include chicken antibodies. Examples of the anti-human FGFR2 rat monoclonal antibody include FR2-10, FR2-13, FR2-14 (WO2013 / 154206) and the like.
 キメラ化抗体としては、非ヒト動物抗体由来の可変領域とヒト抗体(ヒト免疫グロブリン)定常領域とを結合してなる抗体等をあげることができるが、それに限定されるものではない。非ヒト動物抗体由来の可変領域とヒト抗体定常領域が結合してなるキメラ化抗体としては、前述のラットモノクローナル抗体FR2-10、FR2-13又はFR2-14由来の重鎖及び軽鎖可変領域、ならびに、ヒト重鎖及び軽鎖定常領域を有する、cFR2-10、cFR2-13、cFR2-14(WO2013/154206)等をそれぞれあげることができる。 Examples of the chimerized antibody include, but are not limited to, an antibody obtained by binding a variable region derived from a non-human animal antibody and a human antibody (human immunoglobulin) constant region. Chimeric antibodies formed by combining a variable region derived from a non-human animal antibody and a human antibody constant region include the heavy and light chain variable regions derived from the aforementioned rat monoclonal antibody FR2-10, FR2-13 or FR2-14, In addition, cFR2-10, cFR2-13, cFR2-14 (WO2013 / 154206) having human heavy chain and light chain constant regions can be exemplified.
 ヒト化抗体としては、非ヒト動物抗体の可変領域中のCDRをヒト抗体(ヒト免疫グロブリンの可変領域)に移植したもの、CDRに加え非ヒト動物抗体のフレームワーク領域の配列も一部ヒト抗体に移植したもの、それらのいずれかの非ヒト動物抗体由来の1つ又は2つ以上のアミノ酸をヒト型のアミノ酸で置換したもの等をあげることができるが、それらに限定されるものではない。非ヒト動物抗体の可変領域中のCDRとしては、前述のFR2-10、FR2-13又はFR2-14由来の重鎖可変領域中のCDRH1乃至CDRH3および軽鎖可変領域中のCDRL1乃至CDRL3、それらのCDRのアミノ酸配列において1又は2個のアミノ酸が他のアミノ酸に置換されたもの等を例示することができる。 As humanized antibodies, CDRs in the variable region of a non-human animal antibody are transplanted to a human antibody (variable region of human immunoglobulin), and in addition to the CDR, the sequence of the framework region of the non-human animal antibody is also partially human antibody However, the present invention is not limited to these, and those obtained by substituting one or more amino acids derived from any of these non-human animal antibodies with human amino acids. CDRs in the variable region of the non-human animal antibody include CDRH1 to CDRH3 in the heavy chain variable region derived from FR2-10, FR2-13, or FR2-14 described above, and CDRL1 to CDRRL3 in the light chain variable region, Examples include those in which one or two amino acids are substituted with other amino acids in the CDR amino acid sequence.
 ヒト抗体としては、本発明の抗原を認識する抗体であれば特に限定されるものではないが、本発明の抗体のCDRを有する抗体と同一の部位に結合するヒト抗体、前述のFR2-10,FR2-13又はFR2-14とFGFR2上で同一の部位に結合するヒト抗体等を例示することができる。 The human antibody is not particularly limited as long as it is an antibody that recognizes the antigen of the present invention, but a human antibody that binds to the same site as the antibody having the CDR of the antibody of the present invention, the aforementioned FR2-10, Examples include human antibodies that bind to the same site on FR2-13 or FR2-14 and FGFR2.
 本発明における抗体は、FGFR2に結合する活性を有していれば、複数の異なる抗体に由来する部分から構成される抗体であってもよく、例えば、複数の異なる抗体間で重鎖及び/又は軽鎖を交換したもの、重鎖及び/又は軽鎖の全長を交換したもの、可変領域のみ又は定常領域のみを交換したもの、CDRの全部又は一部のみを交換したもの等をあげることができる。キメラ化抗体の重鎖可変領域と軽鎖可変領域は、異なる本発明の抗体に由来してもよい。ヒト化抗体の重鎖及び軽鎖の可変領域中のCDRH1乃至CDRH3並びにCDRL1乃至CDRL3は、2種又はそれ以上の本発明の抗体に由来してもよい。ヒト抗体の重鎖及び軽鎖の可変領域中のCDRH1乃至CDRH3並びにCDRL1乃至CDRL3は、2種又はそれ以上の本発明の抗体が有するCDRの組合せであってもよい。そのような複数の異なる抗体に由来する部分から構成される抗体は、(3-3)乃至(3-6)記載の1つ又は2つ以上の活性を有していてもよい。 The antibody in the present invention may be an antibody composed of portions derived from a plurality of different antibodies as long as it has an activity of binding to FGFR2, for example, a heavy chain and / or between a plurality of different antibodies. Examples include those in which the light chain has been exchanged, those in which the full length of the heavy chain and / or light chain has been exchanged, those in which only the variable region or only the constant region has been exchanged, and those in which all or part of the CDR has been exchanged. . The heavy chain variable region and the light chain variable region of the chimerized antibody may be derived from different antibodies of the present invention. CDRH1 to CDRH3 and CDRL1 to CDRL3 in the variable regions of the heavy and light chains of the humanized antibody may be derived from two or more antibodies of the invention. CDRH1 to CDRH3 and CDRL1 to CDRL3 in the variable region of the heavy chain and light chain of a human antibody may be a combination of CDRs of two or more antibodies of the present invention. Such an antibody composed of a portion derived from a plurality of different antibodies may have one or more activities described in (3-3) to (3-6).
 本発明のモノクローナル抗体のアイソタイプは特に限定されるものではないが、例えば、IgG1、IgG2、IgG3、IgG4等のIgG、IgM、IgA1、IgA2等のIgA、IgD、IgE等をあげることができ、好適にはIgGおよびIgMを挙げることができる。
(3-2)抗体の結合特異性
 本発明の抗体はFGFR2蛋白質を認識する。言い換えれば、本発明の抗体はFGFR2蛋白質に結合する。かかる抗体は「抗FGFR2抗体」と表記される。また、本発明の好適な抗体はFGFR2蛋白質を特異的に認識する。言い換えれば、本発明の好適な抗体はFGFR2蛋白質に特異的に結合する。さらに、本発明の、より好適な抗体はFGFR2IIIb蛋白質及び/又はFGFR2IIIc蛋白質に特異的に結合し、より一層好適な抗体はFGFR2IIIb蛋白質及び/又はFGFR2IIIc蛋白質の有する免疫グロブリン様ドメイン(以下、「Ig様ドメイン」という)に特異的に結合する。かかるIg様ドメインとしては、Ig様ドメイン2、Ig様ドメイン3等を例示することができる。
The isotype of the monoclonal antibody of the present invention is not particularly limited, and examples thereof include IgG such as IgG1, IgG2, IgG3, and IgG4, IgA such as IgM, IgA1, and IgA2, IgD, IgE, and the like. Can include IgG and IgM.
(3-2) Binding specificity of antibody The antibody of the present invention recognizes the FGFR2 protein. In other words, the antibody of the present invention binds to the FGFR2 protein. Such antibodies are designated as “anti-FGFR2 antibodies”. The preferred antibody of the present invention specifically recognizes the FGFR2 protein. In other words, preferred antibodies of the present invention specifically bind to the FGFR2 protein. Furthermore, the more preferable antibody of the present invention specifically binds to the FGFR2IIIb protein and / or FGFR2IIIc protein, and the more preferable antibody is an immunoglobulin-like domain (hereinafter referred to as “Ig-like”) possessed by the FGFR2IIIb protein and / or FGFR2IIIc protein. Domain)). Examples of such Ig-like domains include Ig-like domain 2, Ig-like domain 3, and the like.
 本発明において「特異的な認識」、すなわち「特異的な結合」とは、非特異的な吸着ではない結合を意味する。結合が特異的であるか否かの判定基準としては、例えば、解離定数(Dissociation Consitant:以下、「KDという」)をあげることができる。本発明の好適な抗体のFGFR2蛋白質に対するKD値は1×10-5M以下、5×10-6M以下、2×10-6M以下または1×10-6M以下、より好適には5×10-7M以下、2×10-7M以下または1×10-7M以下、より一層好適には5×10-8M以下、2×10-8M以下または1×10-8M以下、さらにより一層好適には5×10-9M以下、2×10-9M以下または1×10-9M以下、最適には5×10-10M以下、2×10-10M以下または1×10-10M以下である。 In the present invention, “specific recognition”, that is, “specific binding” means binding that is not non-specific adsorption. As a criterion for determining whether or not the binding is specific, for example, a dissociation constant (hereinafter referred to as “KD”) can be given. The KD value for the FGFR2 protein of a suitable antibody of the present invention is 1 × 10 −5 M or less, 5 × 10 −6 M or less, 2 × 10 −6 M or less, or 1 × 10 −6 M or less, more preferably 5 × 10 −7 M or less, 2 × 10 −7 M or less or 1 × 10 −7 M or less, and more preferably 5 × 10 −8 M or less, 2 × 10 −8 M or less, or 1 × 10 −8 M Below, even more preferably 5 × 10 −9 M or less, 2 × 10 −9 M or less, or 1 × 10 −9 M or less, optimally 5 × 10 −10 M or less, 2 × 10 −10 M or less Or 1 × 10 −10 M or less.
 本発明における抗原と抗体の結合は、ELISA法、RIA法、Surface Plasmon Resonance解析法等により測定または判定することができる。
(3-3)抗体の抗腫瘍活性
 本発明の抗体は、抗腫瘍活性を有し、好適にはイン・ビボ(in vivo)で抗腫瘍活性を有する。本発明において、抗腫瘍活性と抗癌活性は同義である。
The binding between the antigen and the antibody in the present invention can be measured or determined by ELISA method, RIA method, Surface Plasmon Resonance analysis method or the like.
(3-3) Antitumor activity of antibody The antibody of the present invention has antitumor activity, and preferably has antitumor activity in vivo. In the present invention, antitumor activity and anticancer activity are synonymous.
 本発明において、抗腫瘍活性とは、腫瘍組織及び/又は腫瘍細胞の増殖、悪性化、浸潤、転移、腫瘍サイズ増大又は重量増加等を抑制する活性を意味する。 In the present invention, the antitumor activity means an activity of suppressing the growth, malignancy, invasion, metastasis, tumor size increase or weight increase of tumor tissue and / or tumor cells.
 抗腫瘍活性は、定法に従って評価することができる。イン・ビボ抗腫瘍活性は、例えば、ヒト癌組織又は癌細胞を移植した非ヒト動物モデル(ゼノグラフト)を用いることにより、ヒト腫瘍に対する効果を評価することができる。ゼノグラフトに用いる非ヒト動物としては、ヌードマウス等のマウスやラット等をあげることができる。 Antitumor activity can be evaluated according to a standard method. The in vivo antitumor activity can be evaluated for its effect on human tumors by using, for example, a non-human animal model (xenograft) transplanted with human cancer tissue or cancer cells. Examples of non-human animals used for xenograft include mice such as nude mice and rats.
 また、抗腫瘍活性は、癌細胞の細胞増殖に対する抑制または阻害活性としても評価することができる。
(3-4)抗体の細胞傷害活性
 本発明の抗FGFR2抗体は、抗体依存性細胞傷害(ADCC)活性及び/又は補体依存性細胞傷害(CDC)活性及び/又は抗体依存性細胞媒介食活性(ADCP)活性を有していてもよい。本発明の好適な抗体はADCC活性を有し、より好適な抗体は、FGFR2発現細胞に対してADCC活性を有する。ADCC活性、CDC活性およびADCP活性は公知の方法により測定することができる。
(3-5)シグナル伝達に対する抗体の作用
本発明の抗FGFR2抗体の有する生物学的活性及び性質は、FGFR2を介したFGFシグナルを通じて評価することもできる。FGFR2を介したFGF刺激によるシグナル伝達としては、特に限定されるものではないが、例えば、FGFR2自己リン酸化、FGFR基質のリクルート及びその促進、それらを介したMAPK,PI3K、Akt、細胞外シグナル制御キナーゼ(ERK)等のシグナル経路の活性化等をあげることができる。
The antitumor activity can also be evaluated as a suppressive or inhibitory activity against cell proliferation of cancer cells.
(3-4) Cytotoxic activity of antibody The anti-FGFR2 antibody of the present invention has antibody-dependent cytotoxicity (ADCC) activity and / or complement-dependent cytotoxicity (CDC) activity and / or antibody-dependent cell-mediated phagocytic activity. It may have (ADCP) activity. A preferred antibody of the present invention has ADCC activity, and a more preferred antibody has ADCC activity against FGFR2-expressing cells. ADCC activity, CDC activity and ADCP activity can be measured by known methods.
(3-5) Effect of antibody on signal transduction The biological activity and properties of the anti-FGFR2 antibody of the present invention can also be evaluated through FGFR2-mediated FGF signals. Signal transduction by FGF stimulation via FGFR2 is not particularly limited, but includes, for example, FGFR2 autophosphorylation, FGFR substrate recruitment and its promotion, MAPK, PI3K, Akt, extracellular signal control through them Examples include activation of signal pathways such as kinase (ERK).
 また、本発明の好適な抗体はFGFR2に対する中和活性を有し、より好適にはFGFR2IIIb及び/又はFGFR2IIIcに対する中和活性を有する。中和活性とは、FGFR2のリガンドによるFGFR2活性化を阻害又は抑制する活性を意味する。例えば、FGF依存性のFGFR2介在性シグナル、シグナル伝達等を阻害する抗体は、かかる中和活性を有すると判定することができる。
(3-6)抗体の受容体-リガンド間の結合を阻害する活性
本発明の好適な抗体はFGFR2とそのリガンドの結合を阻害し、より好適にはFGFR2IIIb及び/又はFGFR2IIIcとFGFの結合を阻害する。かかる受容体-リガンド間の結合阻害は、競合的阻害および非競合的阻害のいずれであってもよい。FGFR2IIIbおよびFGFR2IIIcに対するリガンドとしては、FGF1、FGF3、FGF7、FGF10、FGF22およびFGF23、ならびに、FGF1、FGF2、FGF4、FGF6、FGF9、FGF17、FGF18、FGF21およびFGF23を、それぞれ例示することができる。
(3-7)モノクローナル抗体
 本発明はモノクローナル抗体を提供する。モノクローナル抗体には、ラット抗体、マウス抗体、ウサギ抗体、ニワトリ抗体、魚類抗体等の非ヒト動物由来のモノクローナル抗体、キメラ抗体、ヒト化抗体、ヒト抗体、それらの機能断片、それらの修飾体等が含まれる。このうち、ラットモノクローナル抗体の例としては、FR2-14抗体(WO2013/154206)等をあげることができる。
The preferred antibody of the present invention has neutralizing activity against FGFR2, more preferably neutralizing activity against FGFR2IIIb and / or FGFR2IIIc. The neutralizing activity means an activity that inhibits or suppresses FGFR2 activation by the FGFR2 ligand. For example, an antibody that inhibits FGF-dependent FGFR2-mediated signals, signal transduction and the like can be determined to have such neutralizing activity.
(3-6) Activity of Inhibiting Receptor-Ligand Binding of Antibody A preferred antibody of the present invention inhibits the binding of FGFR2 and its ligand, and more preferably inhibits the binding of FGFR2IIIb and / or FGFR2IIIc to FGF. To do. Such receptor-ligand binding inhibition may be either competitive inhibition or non-competitive inhibition. Examples of ligands for FGFR2IIIb and FGFR2IIIc include FGF1, FGF3, FGF7, FGF10, FGF22 and FGF23, and FGF1, FGF2, FGF4, FGF6, FGF9, FGF17, FGF18, FGF21 and FGF23, respectively.
(3-7) Monoclonal antibody The present invention provides a monoclonal antibody. Monoclonal antibodies include rat antibodies, mouse antibodies, rabbit antibodies, chicken antibodies, monoclonal antibodies derived from non-human animals such as fish antibodies, chimeric antibodies, humanized antibodies, human antibodies, functional fragments thereof, modified versions thereof, etc. included. Of these, examples of rat monoclonal antibodies include the FR2-14 antibody (WO2013 / 154206).
 FR2-14は抗ヒトFGFR2ラットモノクローナル抗体である(WO2013/154206)。 FR2-14 is an anti-human FGFR2 rat monoclonal antibody (WO2013 / 154206).
 本発明の抗体変異体には、好適には、蛋白質の分解もしくは酸化に対する感受性の低下、生物活性の改善、抗原結合能の改善、又は理化学的性質もしくは機能的性質の付与等がなされている。そのような抗体変異体の例として、抗体の有するアミノ酸配列において保存的アミノ酸置換されてなるアミノ酸配列を有する抗体をあげることができる。保存的アミノ酸置換とは、アミノ酸側鎖に関連のあるアミノ酸グループ内で生じる置換である(WO2013/154206)。 The antibody variants of the present invention preferably have reduced sensitivity to protein degradation or oxidation, improved biological activity, improved antigen binding ability, or imparted physicochemical or functional properties. As an example of such an antibody variant, an antibody having an amino acid sequence obtained by conservative amino acid substitution in the amino acid sequence of the antibody can be mentioned. A conservative amino acid substitution is a substitution that occurs within an amino acid group associated with an amino acid side chain (WO2013 / 154206).
 蛋白質中に含まれるアスパラギン酸は、そのC末側に連結するアミノ酸が小さい側鎖を有する場合は異性化によりイソアスパラギン酸に変換され易く、一方で、アスパラギンの場合は脱アミド化によりアスパラギン酸に変換され易く、さらに異性化によりイソアスパラギン酸に変換される可能性がある。そのような異性化や脱アミド化が進めば、蛋白質の安定性に影響が生じ得る。そこで、かかる異性化や脱アミド化を回避するために、蛋白質中のアスパラギン酸やアスパラギンあるいはそれらに隣接するアミノ酸等を他のアミノ酸で置換することができる。かかるアミノ酸置換がなされた抗体変異体は、元の抗体が有する抗原結合活性を保持していることが好ましい。 Aspartic acid contained in a protein is easily converted to isoaspartic acid by isomerization when the amino acid linked to the C-terminal side has a small side chain, whereas in the case of asparagine, it is converted to aspartic acid by deamidation. It is easily converted and may be further converted to isoaspartic acid by isomerization. As such isomerization and deamidation proceed, protein stability may be affected. Therefore, in order to avoid such isomerization and deamidation, aspartic acid, asparagine or amino acids adjacent to them in the protein can be substituted with other amino acids. It is preferable that the antibody variant in which such amino acid substitution is made retains the antigen-binding activity of the original antibody.
 本発明の抗体のアミノ酸配列において保守的アミノ酸置換がなされたアミノ酸配列を有する抗体変異体、ならびに、FR2-14由来のCDRH1乃至CDRH3及びCDRL1乃至CDRL3のいずれかのアミノ酸配列において保守的アミノ酸変異がなされたアミノ酸配列を有する該CDRを含むマウス抗体、ラット抗体、キメラ化抗体、ヒト化抗体、ヒト抗体等も本発明に包含される。 An antibody variant having an amino acid sequence in which a conservative amino acid substitution has been made in the amino acid sequence of the antibody of the present invention, and a conservative amino acid mutation in any of the amino acid sequences of CDRH1 to CDRH3 and CDRL1 to CDRL3 derived from FR2-14 Also included in the present invention are mouse antibodies, rat antibodies, chimerized antibodies, humanized antibodies, human antibodies and the like containing the CDRs having the amino acid sequence.
 FR2-14由来のCDRH1乃至CDRH3及びCDRL1乃至CDRL3のいずれか1つ又は2つ以上のアミノ酸配列において1乃至数個、好適には1乃至3個、より好適には1又は2個、最適には1個のアミノ酸が他のアミノ酸に置換されてなるアミノ酸配列を有するCDRH1乃至CDRH3及びCDRL1乃至CDRL3を含む抗体の変異体であって、且つヒトFGFR2に結合するものは、本発明の抗体の変異体に包含される。 One to several, preferably 1 to 3, more preferably 1 or 2, and most preferably, in any one or two or more amino acid sequences of CDRH1 to CDRH3 and CDRL1 to CDRL3 derived from FR2-14 A variant of an antibody comprising CDRH1 to CDRH3 and CDRL1 to CDRL3 having an amino acid sequence in which one amino acid is substituted with another amino acid and binding to human FGFR2 is a variant of the antibody of the present invention. Is included.
 FR2-14の好適な変異体としては、例えば、CDRH3のアミノ酸配列において1又は2個のアミノ酸、好適にはアミノ酸末端から数えて1つ目又は2つ目のアミノ酸が他のアミノ酸で置換されてなるアミノ酸配列を有するCDRH3を含むキメラ抗体、ヒト化抗体、ヒト抗体等をあげることができる。 
FR2-14重鎖CDRH3のアミノ酸が置換された好適な変異体としては、例えば、(i)1番目のアミノ酸であるアスパラギン酸がグルタミン酸で置換された変異体(配列番号10並びに図8:118番目のアミノ酸がGlu)、(ii)2番目のアミノ酸であるグリシンがチロシン、アラニン、トリプトファン、バリン、アルギニン、アスパラギン、メチオニン、ロイシン、リジン、イソロイシン、ヒスチジン、フェニルアラニン、グルタミンまたはグルタミン酸で、好適にはフェニルアラニン、ヒスチジン、リジン又はロイシン(配列番号12、14、16、18、20並びに図9乃至12及び16:119番目のアミノ酸がPhe、Lys、His又はLeu)で、より好適にはロイシンで置換された変異体(配列番号16又は18並びに図11又は12:119番目のアミノ酸がLeu)、(iii)8番目のアミノ酸であるスレオニンがアラニンで置換されたヒト化抗体、キメラ抗体、ヒト抗体、その機能断片等を例示することができる。具体的には、配列表の配列番号8、10、12、14、16、18及び20に示されるアミノ酸配列のアミノ酸番号118-126からなるCDRH3が挙げられる。
As a suitable variant of FR2-14, for example, in the amino acid sequence of CDRH3, 1 or 2 amino acids, preferably the 1st or 2nd amino acid counted from the amino acid end is substituted with another amino acid. A chimeric antibody, a humanized antibody, a human antibody, etc. containing CDRH3 having the amino acid sequence
Examples of suitable mutants in which the amino acid of FR2-14 heavy chain CDRH3 is substituted include, for example, (i) a mutant in which aspartic acid, which is the first amino acid, is substituted with glutamic acid (SEQ ID NO: 10 and FIG. 8: 118th position). (Ii) the second amino acid glycine is tyrosine, alanine, tryptophan, valine, arginine, asparagine, methionine, leucine, lysine, isoleucine, histidine, phenylalanine, glutamine or glutamic acid, preferably phenylalanine Histidine, lysine or leucine (SEQ ID NOs: 12, 14, 16, 18, 20 and FIGS. 9-12 and 16: 119th amino acid is Phe, Lys, His or Leu), more preferably leucine. Variant (SEQ ID NO: 16 or 1 11 or 12: Illustrated are humanized antibodies, chimeric antibodies, human antibodies, functional fragments thereof and the like in which the 119th amino acid is Leu) and (iii) the 8th amino acid threonine is substituted with alanine. . Specifically, CDRH3 consisting of amino acid numbers 118-126 of the amino acid sequence shown in SEQ ID NOs: 8, 10, 12, 14, 16, 18, and 20 in the sequence listing can be mentioned.
 また、複数の抗体に由来するCDRH1乃至CDRH3およびCDRL1乃至CDRL3を有する抗体も、抗体変異体に含まれる。そのような変異体の例として、CDRH3のみある抗体に由来し、CDRH1及びCDRH2ならびにCDRL1乃至CDRL3は別の抗体に由来する抗体変異体をあげることができる。 Further, antibodies having CDRH1 to CDRH3 and CDRL1 to CDRL3 derived from a plurality of antibodies are also included in the antibody variants. Examples of such a variant include antibody variants derived from an antibody having only CDRH3, and CDRH1 and CDRH2 and CDRL1 to CDRL3 are derived from other antibodies.
 本発明においては抗体変異体も「抗体」に包含される。 In the present invention, antibody variants are also encompassed by “antibodies”.
 本発明の抗体の定常領域としては、とくに限定されるものではないが、ヒトの疾患を治療または予防するための本発明の抗体としては、好適にはヒト抗体のものが使用される。ヒト抗体の重鎖定常領域としては、例えば、Cγ1、Cγ2、Cγ3、Cγ4、Cμ、Cδ、Cα1、Cα2、Cε等をあげることができる。ヒト抗体の軽鎖定常領域としては、例えば、Cκ、Cλ等をあげることができる。
(3-8)抗体の機能断片
 本発明は一つの態様として、本発明の抗FGFR2抗体の機能断片を提供する。抗体の機能断片とは、該抗体の有する機能の少なくとも一部を保持する断片を意味する。かかる抗体の機能としては、一般的には、抗原結合活性、抗原の活性を調節する活性、抗体依存性細胞障害(ADCC)活性及び抗体依存性細胞媒介食(ADCP)活性等を挙げることができる。本発明の抗FGFR2抗体の機能としては、例えば、FGFR2蛋白質結合活性、ADCC活性、ADCP活性、FGFR2に対する中和活性、イン・ビボ抗腫瘍活性、FGFR2とそのリガンドの結合を阻害する活性等をあげることができる。
The constant region of the antibody of the present invention is not particularly limited, but a human antibody is preferably used as the antibody of the present invention for treating or preventing a human disease. Examples of the heavy chain constant region of a human antibody include Cγ1, Cγ2, Cγ3, Cγ4, Cμ, Cδ, Cα1, Cα2, and Cε. Examples of the light chain constant region of a human antibody include Cκ and Cλ.
(3-8) Functional Fragment of Antibody As an aspect, the present invention provides a functional fragment of the anti-FGFR2 antibody of the present invention. The functional fragment of an antibody means a fragment that retains at least a part of the function of the antibody. Such antibody functions generally include antigen binding activity, activity regulating antigen activity, antibody-dependent cytotoxicity (ADCC) activity, antibody-dependent cell-mediated food (ADCP) activity, and the like. . Examples of the function of the anti-FGFR2 antibody of the present invention include FGFR2 protein binding activity, ADCC activity, ADCP activity, neutralizing activity against FGFR2, in vivo antitumor activity, activity of inhibiting the binding of FGFR2 and its ligand, and the like. be able to.
 抗体の機能断片としては、該抗体の有する活性の少なくとも一部を保持している該抗体の断片であれば特に限定されないが、例えば、Fab、F(ab’)2、Fv、重鎖及び軽鎖のFvを適当なリンカーで連結させたシングルチェインFv(scFv)、diabody(diabodies)、線状抗体、及び抗体断片より形成された多特異性抗体、F(ab’)2を還元条件下で処理した抗体の可変領域の一価の断片であるFab’等をあげることができるが、それらに限定されるものではない。リンカー部分を保有するscFvのように、本発明の抗体の断片以外の部分を含む分子も、本発明の抗体の機能断片の意味に包含される。 The functional fragment of the antibody is not particularly limited as long as it is a fragment of the antibody that retains at least a part of the activity of the antibody. For example, Fab, F (ab ′) 2, Fv, heavy chain and light chain A single chain Fv (scFv), a diabody (diabodies), a linear antibody, and a multispecific antibody formed from an antibody fragment, in which Fv of the chain is linked with an appropriate linker, F (ab ′) 2 under reducing conditions Fab ′, which is a monovalent fragment of the variable region of the treated antibody, can be mentioned, but is not limited thereto. Molecules containing portions other than the fragments of the antibody of the present invention, such as scFv having a linker moiety, are also encompassed within the meaning of the functional fragment of the antibody of the present invention.
 抗体蛋白質のアミノ末端及び/又はカルボキシ末端のアミノ酸が1乃至数個又はそれ以上を欠失してなり、且つ該抗体の有する機能の少なくとも一部を保持している分子も、抗体の機能断片の意味に包含される。例えば、哺乳類培養細胞で生産される抗体の重鎖のカルボキシル末端のリジン残基が欠失することが知られており(Journal of Chromatography A,705:129-134(1995))、また、同じく重鎖カルボキシル末端のグリシン、リジンの2アミノ酸残基が欠失し、新たにカルボキシル末端に位置するプロリン残基がアミド化されることが知られている(Analytical Biochemistry,360:75-83(2007))。しかし、これらの重鎖配列の欠失及び修飾は、抗体の抗原結合能及びエフェクター機能(補体の活性化や抗体依存性細胞障害作用など)には影響を及ぼさない。そのような抗体の機能断片の修飾体も、本発明の抗体もしくはその機能断片、又はその修飾体(後述)に包含される。 A molecule in which one or more amino acid and / or carboxy terminal amino acids of an antibody protein are deleted and which retains at least a part of the functions of the antibody is also a functional fragment of an antibody. Included in the meaning. For example, it is known that the lysine residue at the carboxyl terminus of the heavy chain of an antibody produced in cultured mammalian cells is deleted (Journal of Chromatography A, 705: 129-134 (1995)), and also heavy. It is known that two amino acid residues of glycine and lysine at the chain carboxyl terminus are deleted and a proline residue newly located at the carboxyl terminus is amidated (Analytical Biochemistry, 360: 75-83 (2007)). ). However, deletion and modification of these heavy chain sequences do not affect the antigen-binding ability and effector function (such as complement activation and antibody-dependent cytotoxicity) of the antibody. Such modified antibody functional fragments are also encompassed by the antibody of the present invention or functional fragments thereof, or modified products thereof (described later).
 本発明の抗体又はその機能断片は、少なくとも2種類の異なる抗原に対して特異性を有する多特異性抗体であってもよい。多特異性抗体は、2種類の異なる抗原に結合する二重特異性抗体(bispecific antibody)に限定されず、3種以上の異なる抗原に対して特異性を有する抗体も本発明の「多特異性抗体」の意味に包含される。 The antibody or functional fragment thereof of the present invention may be a multispecific antibody having specificity for at least two different antigens. The multispecific antibody is not limited to a bispecific antibody that binds to two different antigens, and an antibody having specificity for three or more different antigens is also the “multispecific antibody of the present invention. It is encompassed within the meaning of “antibody”.
 本発明の多特異性抗体は、全長抗体又はその機能断片であってもよい(例えば、F(ab’)2二重特異性抗体)。 本発明の抗体の一つの態様は、一本鎖抗体(以下、「scFv」という)である。また、2つ以上のscFvをポリペプチドリンカーで結合させて作製されるBiscFvやMultiscFv、一本鎖イムノグロブリン、単一ドメイン抗体、ナノボディ等も、本発明における抗体の機能性断片の意味に包含される。
(3-9)ヒト化抗体およびヒト抗体
 本発明はその一つの態様において、ヒト化抗体又はその機能断片を提供する。
The multispecific antibody of the present invention may be a full-length antibody or a functional fragment thereof (eg, F (ab ′) 2 bispecific antibody). One embodiment of the antibody of the present invention is a single chain antibody (hereinafter referred to as “scFv”). In addition, BiscFv, MultiscFv, single chain immunoglobulin, single domain antibody, Nanobody, etc. prepared by linking two or more scFvs with a polypeptide linker are also included in the meaning of the functional fragment of the antibody in the present invention. .
(3-9) Humanized antibody and human antibody In one embodiment, the present invention provides a humanized antibody or a functional fragment thereof.
 本発明の抗FGFR2ヒト化抗体又はその機能断片は、抗腫瘍活性を有し、好適にはイン・ビボ(in vivo)で抗腫瘍活性を有する。また、かかるヒト化抗体又はその機能断片は、好適にはFGFR2IIIb蛋白質及び/又はFGFR2IIIc蛋白質に特異的に結合し、より好適にはそれらの蛋白質の有するIg様ドメインに結合する。さらに、かかるヒト化抗体又はその機能断片は、好適にはADCC活性及び/又はADCP活性を有する。また、本発明のヒト化抗体又はその機能断片はFGFR2に対する中和活性を有し、好適にはFGFR2IIIb及び/又はFGFR2IIIcに対する中和活性を有し、より好適にはFGFR2IIIb及びFGFR2IIIcに対する中和活性を有する。さらに、本発明のヒト化抗体又はその機能断片は、好適にはFGFR2とそのリガンドの結合を阻害する。 The anti-FGFR2 humanized antibody of the present invention or a functional fragment thereof has antitumor activity, and preferably has antitumor activity in vivo. Moreover, such a humanized antibody or a functional fragment thereof preferably binds specifically to the FGFR2IIIb protein and / or FGFR2IIIc protein, and more preferably binds to an Ig-like domain of those proteins. Furthermore, such a humanized antibody or functional fragment thereof preferably has ADCC activity and / or ADCP activity. Further, the humanized antibody of the present invention or a functional fragment thereof has neutralizing activity against FGFR2, preferably has neutralizing activity against FGFR2IIIb and / or FGFR2IIIc, and more preferably has neutralizing activity against FGFR2IIIb and FGFR2IIIc. Have. Furthermore, the humanized antibody or functional fragment thereof of the present invention preferably inhibits the binding of FGFR2 and its ligand.
 本発明の好適なヒト化抗体の例としては、配列表の配列番号1(図2)に示されるアミノ酸配列からなるCDRL1、配列表の配列番号2(図2)に示されるアミノ酸配列からなるCDRL2及び配列表の配列番号3(図3)に示されるアミノ酸配列からなるCDRL3を含む可変領域を有する軽鎖、並びに、配列表の配列番号4(図4)に示されるアミノ酸配列からなるCDRH1、配列表の配列番号5(図5)に示されるアミノ酸配列からなるCDRH2及び配列表の配列番号6(図6)に示されるアミノ酸配列からなるCDRH3(但し、アミノ末端から数えて1つ目又は2つ目のアミノ酸は前述の通り他のアミノ酸で置換されていてもよい)を含む可変領域を有する重鎖からなり、本発明のFGFR2蛋白質を認識するヒト化抗体もしくは該抗体のFGFR2蛋白質結合活性を保持している該抗体の断片、またはその変異体等をあげることができる。 Examples of suitable humanized antibodies of the present invention include CDRL1 consisting of the amino acid sequence shown in SEQ ID NO: 1 (FIG. 2) of the sequence listing, and CDRL2 consisting of the amino acid sequence shown in SEQ ID NO: 2 (FIG. 2) of the sequence listing. And a light chain having a variable region comprising CDRL3 consisting of the amino acid sequence shown in SEQ ID NO: 3 (FIG. 3) in the sequence listing, and CDRH1 consisting of the amino acid sequence shown in SEQ ID NO: 4 (FIG. 4) in the sequence listing, CDRH2 consisting of the amino acid sequence shown in SEQ ID NO: 5 (FIG. 5) in the sequence table and CDRH3 consisting of the amino acid sequence shown in SEQ ID NO: 6 (FIG. 6) in the sequence table (however, the first or two counted from the amino terminus) A humanized antibody that recognizes the FGFR2 protein of the present invention, comprising a heavy chain having a variable region containing the amino acid of the eye (which may be substituted with another amino acid as described above) Properly can be mentioned fragment of said antibody that retain the FGFR2 protein binding activity of the antibody or variant thereof or the like.
 本発明のより好適なヒト化抗体はヒト化FR2-14抗体およびその変異体であり、以下にその例としてhFR2-14_H1/L1乃至hFR2-14_H19/L1(WO2013/154206)を挙げることができるが、それらに限定されるものではなく、例えば、hFR2-14_H1/L1乃至hFR2-14_H19/L1のいずれか一つのヒト化抗体の重鎖可変領域を含む重鎖、ならびに、hFR2-14_H1/L1乃至hFR2-14_H19/L1のいずれか一つのヒト化抗体の軽鎖可変領域を含む軽鎖、を含んでなる抗体も、本発明のより好適なヒト化抗体に含まれる。 More preferred humanized antibodies of the present invention are humanized FR2-14 antibodies and variants thereof, and examples thereof include hFR2-14_H1 / L1 to hFR2-14_H19 / L1 (WO2013 / 154206). For example, but not limited to, a heavy chain comprising the heavy chain variable region of any one of hFR2-14_H1 / L1 to hFR2-14_H19 / L1, and hFR2-14_H1 / L1 to hFR2 An antibody comprising a light chain comprising the light chain variable region of any one of −14_H19 / L1 is also included in the more preferred humanized antibody of the present invention.
 hFR2-14_H19/L1はWO2013/154206の実施例9において得られた、糖鎖修飾が調節されたヒト化抗体である。当該抗体の軽鎖の塩基配列は配列番号7のヌクレオチド番号61乃至705を、アミノ酸配列は配列番号8(図7)のアミノ酸番号21乃至235を、重鎖の塩基配列は配列番号17のヌクレオチド番号58乃至1401を、アミノ酸配列は配列番号18(図12)のアミノ酸番号20乃至467を、それぞれ含んでいる。 HFR2-14_H19 / L1 is a humanized antibody with modified sugar chain modification obtained in Example 9 of WO2013 / 154206. The light chain base sequence of the antibody is nucleotide numbers 61 to 705 of SEQ ID NO: 7, the amino acid sequence is amino acid numbers 21 to 235 of SEQ ID NO: 8 (FIG. 7), and the heavy chain base sequence is the nucleotide number of SEQ ID NO: 17. 58 to 1401, and the amino acid sequence includes amino acid numbers 20 to 467 of SEQ ID NO: 18 (FIG. 12), respectively.
 hFR2-14_H12/L1の、軽鎖の塩基配列は配列番号7のヌクレオチド番号61乃至705を、アミノ酸配列は配列番号8(図7)のアミノ酸番号21乃至235を、重鎖の塩基配列は配列番号15のヌクレオチド番号58乃至1401を、アミノ酸配列は配列番号16(図11)のアミノ酸番号20乃至467を、それぞれ含んでいる。 The light chain base sequence of hFR2-14_H12 / L1 is nucleotide numbers 61 to 705 of SEQ ID NO: 7, the amino acid sequence is amino acid numbers 21 to 235 of SEQ ID NO: 8 (FIG. 7), and the heavy chain base sequence is SEQ ID NO: 15 nucleotide numbers 58 to 1401, and the amino acid sequence includes amino acid numbers 20 to 467 of SEQ ID NO: 16 (FIG. 11), respectively.
 hFR2-14_H8/L1の、軽鎖の塩基配列は配列番号7のヌクレオチド番号61乃至705を、アミノ酸配列は配列番号8(図7)のアミノ酸番号21乃至235を、重鎖の塩基配列は配列番号11のヌクレオチド番号58乃至1401を、アミノ酸配列は配列番号12(図9)のアミノ酸番号20乃至467を、それぞれ含んでいる。 The light chain base sequence of hFR2-14_H8 / L1 is nucleotide numbers 61 to 705 of SEQ ID NO: 7, the amino acid sequence is amino acid numbers 21 to 235 of SEQ ID NO: 8 (FIG. 7), and the heavy chain base sequence is SEQ ID NO: 11 nucleotide numbers 58 to 1401, and the amino acid sequence includes amino acid numbers 20 to 467 of SEQ ID NO: 12 (FIG. 9), respectively.
 hFR2-14_H9/L1の、軽鎖の塩基配列は配列番号7のヌクレオチド番号61乃至705を、アミノ酸配列は配列番号8(図7)のアミノ酸番号21乃至235を、重鎖の塩基配列は配列番号19のヌクレオチド番号58乃至1401を、アミノ酸配列は配列番号20(図16)のアミノ酸番号20乃至467を、それぞれ含んでいる。 The light chain base sequence of hFR2-14_H9 / L1 is nucleotide numbers 61 to 705 of SEQ ID NO: 7, the amino acid sequence is amino acid numbers 21 to 235 of SEQ ID NO: 8 (FIG. 7), and the heavy chain base sequence is SEQ ID NO: 19 nucleotide numbers 58 to 1401, and the amino acid sequence includes amino acid numbers 20 to 467 of SEQ ID NO: 20 (FIG. 16), respectively.
 hFR2-14_H11/L1抗体の、軽鎖の塩基配列は配列番号7のヌクレオチド番号61乃至705を、アミノ酸配列は配列番号8(図7)のアミノ酸番号21乃至235を、重鎖の塩基配列は配列番号13のヌクレオチド番号58乃至1401を、アミノ酸配列は配列番号14(図10)のアミノ酸番号20乃至467を、それぞれ含んでいる。 The light chain base sequence of the hFR2-14_H11 / L1 antibody is nucleotide numbers 61 to 705 of SEQ ID NO: 7, the amino acid sequence is amino acid numbers 21 to 235 of SEQ ID NO: 8 (FIG. 7), and the heavy chain base sequence is the sequence The nucleotide number 58 to 1401 of No. 13 and the amino acid sequence include amino acid numbers 20 to 467 of SEQ ID No. 14 (FIG. 10), respectively.
 hFR2-14_H5/L1の、軽鎖の塩基配列は配列番号7のヌクレオチド番号61乃至705を、アミノ酸配列は配列番号8(図7)のアミノ酸番号21乃至235を、重鎖の塩基配列は配列番号9のヌクレオチド番号58乃至1401を、アミノ酸配列は配列番号10(図8)のアミノ酸番号20乃至467を、それぞれ含んでいる。 The light chain base sequence of hFR2-14_H5 / L1 is nucleotide numbers 61 to 705 of SEQ ID NO: 7, the amino acid sequence is amino acid numbers 21 to 235 of SEQ ID NO: 8 (FIG. 7), and the heavy chain base sequence is SEQ ID NO: 9 nucleotide numbers 58 to 1401, and the amino acid sequence includes amino acid numbers 20 to 467 of SEQ ID NO: 10 (FIG. 8), respectively.
 これらのヒト化抗体はTm値が高く立体構造安定性に優れ、FGFR2に対する高い結合活性、優れた熱安定性、FGFR2リガンド依存性のFGFR2シグナル中和活性、ADCC活性、ADCP活性、FGFR2リガンドとFGFR2の結合を阻害する活性、in vivo抗腫瘍活性等を有しており、過酷な条件に曝しても高い抗原結合活性を維持した。 These humanized antibodies have high Tm values, excellent conformational stability, high binding activity to FGFR2, excellent thermal stability, FGFR2 ligand-dependent FGFR2 signal neutralization activity, ADCC activity, ADCP activity, FGFR2 ligand and FGFR2 It has an activity to inhibit the binding of, an in vivo antitumor activity and the like, and maintains a high antigen binding activity even when exposed to harsh conditions.
 本発明のより好適なヒト化抗体はhFR2-14_H12/L1およびhFR2-14_H19/L1である。 More preferred humanized antibodies of the present invention are hFR2-14_H12 / L1 and hFR2-14_H19 / L1.
 本発明のヒト化hFR2-14_H1/L1乃至hFR2-14_H19/L1抗体のいずれか一つに記載の抗体の重鎖または軽鎖のアミノ酸配列と90%以上、92%以上、94%以上、96%以上、98%以上又は99%以上の同一なアミノ酸配列を含む重鎖または軽鎖を含み、且つFGFR2に結合する抗体又はその機能断片も、本発明に包含される。かかる配列同一性は、好適には94%以上、より好適には96%以上、より一層好適には98%以上、最適には99%以上である。また、それらの抗体は、好適には(3-3)乃至(3-6)記載の活性を一つ又はそれ以上有している。 The amino acid sequence of the heavy or light chain of the antibody according to any one of the humanized hFR2-14_H1 / L1 to hFR2-14_H19 / L1 antibodies of the present invention and 90% or more, 92% or more, 94% or more, 96% As described above, an antibody or a functional fragment thereof comprising a heavy chain or a light chain comprising 98% or more or 99% or more of the same amino acid sequence and binding to FGFR2 is also included in the present invention. Such sequence identity is preferably 94% or more, more preferably 96% or more, even more preferably 98% or more, and optimally 99% or more. These antibodies preferably have one or more of the activities described in (3-3) to (3-6).
 二種類のアミノ酸配列間の同一性あるいは相同性は、Blast algorithm version 2.2.2(Altschul, Stephen F.,Thomas L.Madden,Alejandro A.Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J.Lipman(1997),「Gapped BLAST and PSI-BLAST:a new generation of protein database search programs」,Nucleic Acids Res.25:3389-3402)のデフォルトパラメーターを使用することによって決定することができる。Blast algorithmは、例えば、インターネットでhttp://blast.ncbi.nlm.nih.gov/にアクセスすることによっても使用することができる。 The identity or homology between the two types of amino acid sequences is Blast algorithm version 2.2.2 (Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, ZhengWebbhand J. Lipman (1997), “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res. 25: 3389-3402). Blast algorithm can also be used by accessing http://blast.ncbi.nlm.nih.gov/ on the Internet, for example.
 本発明のヒト化hFR2-14_H1/L1乃至hFR2-14_H19/L1抗体のいずれか一つに記載の抗体の重鎖または軽鎖のアミノ酸配列において1乃至10、8、6、5、4、3、もしくは2個、または1個のアミノ酸が置換、欠失、付加または挿入してなるアミノ酸配列を含む重鎖または軽鎖を含み、且つFGFR2に結合する抗体又はその機能断片も、本発明に包含される。かかるアミノ酸変異は好適には置換であり、変異されるアミノ酸の個数は好適には1乃至5個、より好適には1乃至4個、より一層好適には1乃至3個、さらにより一層好適には1乃至2個、最適には1個である。また、それらの抗体は、好適には(3-3)乃至(3-6)記載の活性を一つ又はそれ以上有している。 1 to 10, 8, 6, 5, 4, 3, in the amino acid sequence of the heavy chain or light chain of the antibody according to any one of the humanized hFR2-14_H1 / L1 to hFR2-14_H19 / L1 antibodies of the present invention, Alternatively, an antibody or functional fragment thereof comprising a heavy or light chain comprising an amino acid sequence in which two or one amino acid is substituted, deleted, added or inserted, and binds to FGFR2, is also encompassed in the present invention. The Such amino acid mutations are preferably substitutions, and the number of amino acids to be mutated is preferably 1 to 5, more preferably 1 to 4, even more preferably 1 to 3, and even more preferably. Is one to two, and most preferably one. These antibodies preferably have one or more of the activities described in (3-3) to (3-6).
 ヒト化hFR2-14_H5/L1、hFR2-14_H8/L1、hFR2-14_H9/L1、hFR2-14_H11/L1、hFR2-14_H12/L1、hFR2-14_H19/L1抗体のいずれか一つに記載の抗体の重鎖または軽鎖のアミノ酸配列をコードする塩基配列に相補的な塩基配列を有するヌクレオチドとストリンジェントな条件下でハイブリダイズするヌクレオチドの有する塩基配列によりコードされるアミノ酸配列を含む重鎖または軽鎖を含み、且つFGFR2に結合する抗体又はその機能断片も、本発明に包含される。それらの抗体は、好適には(3-3)乃至(3-6)記載の活性を一つ又はそれ以上有している。 The heavy chain of the antibody according to any one of humanized hFR2-14_H5 / L1, hFR2-14_H8 / L1, hFR2-14_H9 / L1, hFR2-14_H11 / L1, hFR2-14_H12 / L1, and hFR2-14_H19 / L1 antibodies Or a heavy chain or a light chain comprising an amino acid sequence encoded by a nucleotide sequence having a nucleotide that hybridizes under stringent conditions with a nucleotide having a nucleotide sequence complementary to the nucleotide sequence encoding the amino acid sequence of the light chain In addition, an antibody or a functional fragment thereof that binds to FGFR2 is also encompassed in the present invention. These antibodies preferably have one or more of the activities described in (3-3) to (3-6).
 本発明は別の一つの態様において、ヒト抗体又はその機能断片を提供する。本発明のヒト抗体は、FGFR2に結合するヒト由来の抗体であれば特に限定されるものではないが、好適には(3-3)乃至(3-6)記載の活性を一つ又はそれ以上有している。
(3-10)エピトープに結合する抗体
 本発明の提供する抗体と「同一の部位に結合する抗体」も本発明の抗体に含まれる。ある抗体と「同一の部位に結合する抗体」とは、該抗体が認識する抗原分子上の部位に結合する他の抗体を意味する。第一抗体が結合する抗原分子上の部分ペプチド又は部分立体構造に第二抗体が結合すれば、第一抗体と第二抗体は同一の部位に結合すると判定することができる。また、第一抗体の抗原に対する結合に対して第二抗体が競合する、すなわち、第二抗体が第一抗体と抗原の結合を妨げることを確認することによって、具体的な結合部位のペプチド配列又は立体構造が決定されていなくても、第一抗体と第二抗体が同一の部位に結合すると判定することができる。さらに、第一抗体と第二抗体が同一の部位に結合し、且つ第一抗体が抗腫瘍活性など本発明の抗体の一つの態様に特徴的な効果を有する場合、第二抗体も同様の活性を有する蓋然性は極めて高い。従って、第一の抗FGFR2抗体の結合する部位に第二の抗FGFR2抗体が結合すれば、第一抗体と第二抗体がFGFR2蛋白質上の同一の部位に結合すると判定することができる。また、第一の抗FGFR2抗体のFGFR2蛋白質への結合に対して第二の抗FGFR2抗体が競合することを確認できれば、第一抗体と第二抗体がFGFR2蛋白質上の同一の部位に結合する抗体と判定することができる。
In another embodiment, the present invention provides a human antibody or a functional fragment thereof. The human antibody of the present invention is not particularly limited as long as it is a human-derived antibody that binds to FGFR2, but preferably has one or more of the activities described in (3-3) to (3-6). Have.
(3-10) Antibody that binds to epitope “An antibody that binds to the same site” as the antibody provided by the present invention is also included in the antibody of the present invention. An “antibody that binds to the same site” as an antibody means another antibody that binds to a site on an antigen molecule that the antibody recognizes. If the second antibody binds to the partial peptide or partial conformation on the antigen molecule to which the first antibody binds, it can be determined that the first antibody and the second antibody bind to the same site. In addition, by confirming that the second antibody competes for the binding of the first antibody to the antigen, that is, the second antibody prevents the binding between the first antibody and the antigen, the peptide sequence of the specific binding site or Even if the three-dimensional structure is not determined, it can be determined that the first antibody and the second antibody bind to the same site. Furthermore, when the first antibody and the second antibody bind to the same site, and the first antibody has a characteristic effect on one embodiment of the antibody of the present invention such as antitumor activity, the second antibody has the same activity. The probability of having is very high. Therefore, if the second anti-FGFR2 antibody binds to the site to which the first anti-FGFR2 antibody binds, it can be determined that the first antibody and the second antibody bind to the same site on the FGFR2 protein. Further, if it can be confirmed that the second anti-FGFR2 antibody competes with the binding of the first anti-FGFR2 antibody to the FGFR2 protein, an antibody that binds the first antibody and the second antibody to the same site on the FGFR2 protein. Can be determined.
 本発明のヒト化抗体FR2-10、FR2-13又はFR2-14が認識するFGFR2蛋白質上の部位に結合する抗体も本発明に含まれる。  
(3-11)抗体の修飾体
 本発明は、抗体又はその機能断片の修飾体を提供する。本発明の抗体又はその機能断片の修飾体とは、本発明の抗体又はその機能断片に化学的又は生物学的な修飾が施されてなるものを意味する。化学的な修飾体には、アミノ酸骨格への化学部分の結合、N-結合またはO-結合炭水化物鎖の化学修飾体等が含まれる。生物学的な修飾体には、翻訳後修飾(例えば、N-結合またはO-結合への糖鎖付加、N末またはC末のプロセッシング、脱アミド化、アスパラギン酸の異性化、メチオニンの酸化)されたもの、原核生物宿主細胞を用いて発現させることによりN末にメチオニン残基が付加したもの等が含まれる。また、本発明の抗体または抗原の検出または単離を可能にするために標識されたもの、例えば、酵素標識体、蛍光標識体、アフィニティ標識体もかかる修飾物の意味に含まれる。このような本発明の抗体又はその機能断片の修飾物は、元の本発明の抗体又はその機能断片の安定性および血中滞留性の改善、抗原性の低減、かかる抗体又は抗原の検出又は単離等に有用である。
Antibodies that bind to a site on the FGFR2 protein recognized by the humanized antibody FR2-10, FR2-13 or FR2-14 of the present invention are also included in the present invention.
(3-11) Modified Antibody The present invention provides a modified antibody or a functional fragment thereof. The modified form of the antibody of the present invention or a functional fragment thereof means a product obtained by chemically or biologically modifying the antibody of the present invention or a functional fragment thereof. Chemical modifications include chemical modifications to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and the like. Biological modifications include post-translational modifications (eg, glycosylation to N- or O-links, N- or C-terminal processing, deamidation, aspartic acid isomerization, methionine oxidation) And those in which a methionine residue is added to the N-terminal by expression using a prokaryotic host cell. In addition, those modified to allow detection or isolation of the antibody or antigen of the present invention, for example, an enzyme label, a fluorescent label, and an affinity label are also included in the meaning of such a modified product. Such a modified product of the antibody of the present invention or a functional fragment thereof can be used to improve the stability and blood retention of the original antibody of the present invention or a functional fragment thereof, to reduce antigenicity, to detect or simply detect such an antibody or antigen. Useful for separation.
 化学的修飾体に含まれる化学部分としては、ポリエチレングリコール等の水溶性ポリマーを例示することができる。  Examples of the chemical moiety contained in the chemically modified product include water-soluble polymers such as polyethylene glycol. *
 生物学的な修飾体としては、酵素処理や細胞処理等により修飾が施されたもの、遺伝子組換えによりタグ等他のペプチドが付加された融合体、ならびに内因性又は外来性の糖鎖修飾酵素を発現する細胞を宿主として調製されたもの等をあげることができる。 Biologically modified substances include those modified by enzyme treatment or cell treatment, fusions to which other peptides such as tags have been added by genetic recombination, and endogenous or exogenous sugar chain-modifying enzymes. And the like, which are prepared using a cell expressing the protein as a host.
 本発明の抗体又はその機能断片に結合している糖鎖修飾を調節すること(グリコシル化、脱フコース化等)によって、抗体依存性細胞傷害活性を増強することが可能である。抗体の糖鎖修飾の調節技術としては、例えば、WO99/54342、WO00/61739、WO02/31140等に記載された方法が知られているが、これらに限定されるものではない。本発明の抗体の修飾体には当該糖鎖修飾が調節された抗体も含まれる。 It is possible to enhance antibody-dependent cytotoxic activity by regulating the modification of the sugar chain bound to the antibody of the present invention or a functional fragment thereof (glycosylation, defucose, etc.). For example, methods described in WO99 / 54342, WO00 / 61739, WO02 / 31140 and the like are known as techniques for regulating antibody sugar chain modification, but are not limited thereto. The modified form of the antibody of the present invention includes an antibody in which the sugar chain modification is regulated.
 かかる修飾は、抗体またはその機能断片における任意の位置に、または所望の位置において施されてもよく、1つ又は2つ以上の位置に同一又は2種以上の異なる修飾がなされてもよい。 Such modification may be performed at any position in the antibody or a functional fragment thereof or at a desired position, and one or two or more positions may be subjected to the same or two or more different modifications.
 本発明において「抗体断片の修飾体」は「抗体の修飾体の断片」をもその意味に含むものである。
本発明においては抗体の修飾体、その機能断片の修飾体を、単に「抗体」、「抗体の機能断片」と呼ぶことがある。
hFR2-14_H19/L1は糖鎖修飾が調節された、すなわち脱フコース化されたヒト化抗体であり(WO2013/154206の実施例9)、本発明の抗体、本発明の抗体の修飾体に包含される。
In the present invention, “modified antibody fragment” includes “modified antibody fragment” in its meaning.
In the present invention, a modified antibody and a modified functional fragment thereof are sometimes simply referred to as “antibody” or “functional fragment of an antibody”.
hFR2-14_H19 / L1 is a humanized antibody in which glycosylation is regulated, that is, defucose (Example 9 of WO2013 / 154206), and is included in the antibody of the present invention and the modified antibody of the present invention. The
 4.抗体の製造方法
(4-1)ハイブリドーマを用いる方法
 本発明の一つの態様として、抗FGFR2抗体は、例えば、コーラーとミルスタインの方法(Kohler and Milstein,Nature (1975)256,p.495-497、Kennet,R.ed.,Monoclonal Antibodies,p.365-367,Plenum Press,N.Y.(1980))に従って取得することができる(WO2013/154206)。
(4-2)細胞免疫法
 天然型のFGFR2蛋白質を発現する細胞、組換え型FGFR2蛋白質またはその断片を発現する細胞等を免疫原として使用することにより、前記のハイブリドーマ法により抗FGFR2抗体を調製することができる(WO2013/154206)。
(4-3)遺伝子組換え
 本発明の抗体は、その重鎖アミノ酸配列をコードする塩基配列が含まれるヌクレオチド(重鎖ヌクレオチド)及びその軽鎖アミノ酸配列をコードする塩基配列が含まれるヌクレオチド(軽鎖ヌクレオチド)、又は、かかる重鎖ヌクレオチドが挿入されたベクター及び軽鎖ヌクレオチドが挿入されたベクターを宿主細胞に導入し、該細胞を培養した後その培養物からかかる抗体を回収することにより調製することができ、そのようにして得られた抗体も本発明に含まれる(WO2013/154206)。
(4-4)DNA免疫法
 本発明の抗FGFR2抗体は、DNA免疫法を使用して得ることもできる。抗原発現プラスミドをマウスやラットなどの動物個体に遺伝子導入し、抗原を個体内で発現させることによって、抗原に対する免疫を誘導する。遺伝子導入の手法には、直接プラスミドを筋肉に注射する方法や、リポソームやポリエチレンイミンなどの導入試薬を静脈注射する方法、ウイルスベクターを用いる手法、プラスミドを付着させた金粒子をGene Gunにより射ち込む手法、急速に大量のプラスミド溶液を静脈注射するHydrodynamic法などが存在する。
(4-5)ヒト化抗体のデザイン法および調製法
 ヒト化抗体としては、非ヒト動物抗体のCDRのみがヒト由来の抗体に組込まれた抗体(Nature(1986)321,p.522-525参照)、CDR移植法によりCDRの配列に加え一部のフレームワークのアミノ酸残基もヒト抗体に移植した抗体(WO90/07861号、US6972323号公報参照)、それらのいずれかにおける非ヒト動物抗体の1つまたは2つ以上のアミノ酸がヒト型のアミノ酸で置換されてなる抗体等を挙げることができるが、それらに限定されるものではない。
(4-6)ヒト抗体の調製法
 本発明の抗体としては、さらに、ヒト抗体を挙げることができる。抗FGFR2ヒト抗体とは、ヒト由来の抗体のアミノ酸配列からなる抗FGFR2抗体を意味する。抗FGFR2ヒト抗体は、ヒト抗体の重鎖と軽鎖の遺伝子を含むヒトゲノムDNA断片を有するヒト抗体産生マウスを用いた方法、ヒト抗体ライブラリーより選別したファージディスプレイ由来のヒト抗体を取得する方法等の公知の方法により取得することができる(WO2013/154206)。
(4-7)抗体の機能断片の調製法
 一本鎖抗体を作成する方法は公知である。scFvにおいて、重鎖可変領域と軽鎖可変領域は、コンジュゲートを作らないようなリンカー、好ましくはポリペプチドリンカーを介して連結される。scFvにおける重鎖可変領域及び軽鎖可変領域は、同一の抗体に由来してもよく、別々の抗体に由来してもよい。 本発明の抗体は、多量化して抗原に対する親和性を高めたものであってもよい。多量化する抗体としては、1種類の抗体であっても、同一の抗原の複数のエピトープを認識する複数の抗体であってもよい。
4). Antibody Production Method (4-1) Method Using Hybridoma As one embodiment of the present invention, an anti-FGFR2 antibody is prepared, for example, by the method of Kohler and Milstein (Kohler and Milstein, Nature (1975) 256, p. 495-497. , Kennet, R. ed., Monoclonal Antibodies, p. 365-367, Plenum Press, NY (1980)) (WO2013 / 154206).
(4-2) Cell immunization method An anti-FGFR2 antibody is prepared by the hybridoma method described above using cells expressing a natural FGFR2 protein, cells expressing a recombinant FGFR2 protein or a fragment thereof as an immunogen. (WO2013 / 154206).
(4-3) Genetic Recombination The antibody of the present invention comprises a nucleotide containing a base sequence encoding its heavy chain amino acid sequence (heavy chain nucleotide) and a nucleotide containing a base sequence encoding its light chain amino acid sequence (light chain). A chain nucleotide), or a vector into which such a heavy chain nucleotide is inserted and a vector into which a light chain nucleotide is inserted into a host cell, culturing the cell, and then recovering the antibody from the culture. The antibody thus obtained is also included in the present invention (WO2013 / 154206).
(4-4) DNA immunization The anti-FGFR2 antibody of the present invention can also be obtained using DNA immunization. An antigen expression plasmid is introduced into an animal individual such as a mouse or a rat, and the antigen is expressed in the individual to induce immunity to the antigen. Gene transfer methods include direct injection of plasmids into muscles, intravenous injection of introduction reagents such as liposomes and polyethyleneimine, methods using viral vectors, and gold particles with plasmids attached to them by Gene Gun. There are techniques, such as the Hydrodynamic method in which a large amount of plasmid solution is intravenously injected.
(4-5) Design method and preparation method of humanized antibody As a humanized antibody, an antibody in which only a CDR of a non-human animal antibody is incorporated into a human-derived antibody (Nature (1986) 321; see pages 522-525). ), Antibodies obtained by grafting some framework amino acid residues in addition to CDR sequences to human antibodies by the CDR grafting method (see WO90 / 07861, US6972323), 1 of non-human animal antibodies in any of them Examples thereof include, but are not limited to, antibodies in which one or two or more amino acids are substituted with human amino acids.
(4-6) Preparation Method of Human Antibody The antibody of the present invention can further include a human antibody. The anti-FGFR2 human antibody means an anti-FGFR2 antibody consisting of the amino acid sequence of a human-derived antibody. The anti-FGFR2 human antibody is a method using a human antibody-producing mouse having a human genomic DNA fragment containing the heavy and light chain genes of a human antibody, a method of obtaining a human antibody derived from a phage display selected from a human antibody library, etc. (WO2013 / 154206).
(4-7) Preparation Method of Functional Fragment of Antibody A method for producing a single chain antibody is known. In scFv, the heavy chain variable region and the light chain variable region are linked via a linker that does not form a conjugate, preferably a polypeptide linker. The heavy chain variable region and the light chain variable region in scFv may be derived from the same antibody or different antibodies. The antibody of the present invention may be an antibody having increased affinity for an antigen by multimerization. The antibody that multiplies may be one type of antibody or a plurality of antibodies that recognize multiple epitopes of the same antigen.
 本発明の抗体は、アミノ酸配列が異なる複数種類の抗FGFR2抗体の混合物、すなわちポリクローナル抗体であってもよい。 The antibody of the present invention may be a mixture of a plurality of types of anti-FGFR2 antibodies having different amino acid sequences, that is, a polyclonal antibody.
 抗体の修飾物として、ポリエチレングリコール(PEG)等の各種分子と結合した抗体を使用することもできる。 As the modified antibody, an antibody conjugated with various molecules such as polyethylene glycol (PEG) can also be used.
 本発明の抗体は、更にこれらの抗体と他の薬剤がコンジュゲートを形成しているもの(Immunoconjugate)でもよい。このような抗体の例としては、該抗体が放射性物質や薬理作用を有する化合物と結合している物を挙げることができる(WO2013/154206)。
(4-8)抗体の精製
 得られた抗体は、均一にまで精製することができる。抗体の分離、精製は通常の蛋白質で使用されている分離、精製方法を使用すればよい。
The antibody of the present invention may be one in which these antibody and another drug form a conjugate (Immunoconjugate). Examples of such antibodies include those in which the antibody is bound to a radioactive substance or a compound having a pharmacological action (WO2013 / 154206).
(4-8) Purification of antibody The obtained antibody can be purified to homogeneity. Separation and purification of antibodies may be carried out using separation and purification methods used for ordinary proteins.
 例えばクロマトグラフィーカラム、フィルター、限外濾過、塩析、透析、調製用ポリアクリルアミドゲル電気泳動、等電点電気泳動等を適宜選択、組み合わせれば、抗体を分離、精製することができるが、これらに限定されるものではない。
(4-9)組換抗体
 本発明は、本発明の抗体もしくはその機能断片またはその修飾体をコードするヌクレオチド(抗体遺伝子)又はベクターが導入された細胞を培養し、その培養物から抗体もしくはその機能断片またはその修飾体を回収する工程、を含む、抗体もしくはその機能断片またはその修飾体の製造方法により得られた抗体もしくはその機能断片またはその修飾体も、本発明に含まれる。
For example, antibodies can be separated and purified by appropriately selecting and combining chromatography columns, filters, ultrafiltration, salting out, dialysis, preparative polyacrylamide gel electrophoresis, isoelectric focusing, etc. It is not limited to.
(4-9) Recombinant antibody The present invention comprises culturing a cell into which a nucleotide (antibody gene) or vector encoding the antibody of the present invention or a functional fragment thereof or a modified form thereof or a vector has been introduced. An antibody or a functional fragment thereof or a modified product thereof obtained by a method for producing an antibody or a functional fragment thereof or a modified product thereof, comprising the step of recovering the functional fragment or a modified product thereof is also included in the present invention.
 5.医薬組成物
 本発明は抗FGFR2抗体もしくはその機能断片またはその修飾体を含む医薬組成物を提供する。
5. Pharmaceutical Composition The present invention provides a pharmaceutical composition comprising an anti-FGFR2 antibody or a functional fragment thereof or a modified form thereof.
 本発明の医薬組成物は、FGFR2もしくはそのリガンドの過剰発現またはFGFR2の変異もしくは遺伝子増幅によるFGFR2シグナル異常又は亢進、または、FGFR2のアイソフォームのスイッチングにより惹起されるか又は増悪化される各種疾患(以下、「FGFR2に関わる疾患」という)、とりわけ各種癌の治療又は予防に有用である。 The pharmaceutical composition of the present invention can be used for various diseases (such as FGFR2 or its ligand overexpression, FGFR2 mutation or gene amplification caused by FGFR2 signal abnormality or enhancement, or FGFR2 isoform switching). Hereinafter, it is useful for the treatment or prevention of "a disease related to FGFR2"), particularly various cancers.
 かかる治療又は予防の対象となる癌の惹起又は増悪化の原因としては、FGFR2遺伝子のイントロン内の一塩基置換(SNP)、FGFR2の高発現、FGFR2を恒常的に活性化するミスセンス変異、FGFR2遺伝子の増幅又は過剰発現、FGFR2IIIbからFGFR2IIIcへのスイッチング等を例示することができる。 Causes of the induction or exacerbation of cancer that is the subject of treatment or prevention include single base substitution (SNP) in the intron of the FGFR2 gene, high expression of FGFR2, a missense mutation that constantly activates FGFR2, and the FGFR2 gene Amplification or overexpression of FGFR2IIIb to FGFR2IIIc can be exemplified.
 かかる癌種としては、例えば、乳癌、子宮内膜癌、卵巣癌、非小細胞肺癌などの肺癌、胃癌、前立腺癌、腎癌、肝臓癌、膵臓癌、大腸癌、食道癌、膀胱癌、子宮頚癌、血液癌、リンパ腫、脳腫瘍、胆管癌、悪性黒色腫等をあげることができ、好適にはFGFR2蛋白質を発現しているそれらの癌をあげることができる。 Examples of such cancer types include lung cancer such as breast cancer, endometrial cancer, ovarian cancer, non-small cell lung cancer, gastric cancer, prostate cancer, kidney cancer, liver cancer, pancreatic cancer, colon cancer, esophageal cancer, bladder cancer, uterus. Cervical cancer, blood cancer, lymphoma, brain tumor, bile duct cancer, malignant melanoma and the like can be mentioned, and those cancers expressing FGFR2 protein can be mentioned preferably.
 本発明において、疾患の治療または予防には、かかる疾患、好適にはFGFR2蛋白質を発現している個体におけるかかる疾患の発症の予防、増悪化又は進行の抑制又は阻害、かかる疾患に罹患した個体が呈する一つ又は二つ以上の症状の軽減、増悪化若しくは進行の抑制または寛解、二次性疾患の治療又は予防等が含まれるが、それらに限定されるものではない。 In the present invention, for the treatment or prevention of a disease, prevention of such disease, preferably the onset of the disease in an individual expressing FGFR2 protein, suppression or inhibition of progression or progression, Examples include, but are not limited to, alleviation of one or more symptoms present, suppression or remission of progression or progression, treatment or prevention of secondary diseases, and the like.
 本発明の医薬組成物には、治療または予防に有効な量の抗FGFR2抗体又は該抗体の機能断片と薬学上許容される希釈剤、担体、可溶化剤、乳化剤、保存剤及び/又は補助剤を含有せしめることができる。 The pharmaceutical composition of the present invention comprises a therapeutically or prophylactically effective amount of an anti-FGFR2 antibody or a functional fragment of the antibody and a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and / or adjuvant. Can be contained.
 「治療または予防に有効な量」とは、特定の疾患、投与形態および投与径路につき治療又は予防効果を奏する量を意味し、「薬理学的に有効な量」と同義である。 “Therapeutically or prophylactically effective amount” means an amount that exhibits a therapeutic or prophylactic effect for a specific disease, administration form, and administration route, and is synonymous with “pharmacologically effective amount”.
 本発明の医薬組成物には、pH、浸透圧、粘度、透明度、色、等張性、無菌性、該組成物又はそれに含まれる抗体の安定性、溶解性、徐放性、吸収性、浸透性、剤型、強度、性状、形状等を変化させたり、維持したり、保持したりするための物質(以下、「製剤用の物質」という)を含有せしめることができる。製剤用の物質としては、薬理学的に許容される物質であれば特に限定されるものではない。例えば、非毒性又は低毒性であることは、製剤用の物質が好適に具備する性質である。 The pharmaceutical composition of the present invention includes pH, osmotic pressure, viscosity, transparency, color, isotonicity, sterility, stability of the composition or antibody contained therein, solubility, sustained release, absorbability, penetration. Substances for changing, maintaining, and maintaining properties, dosage forms, strength, properties, shapes, etc. (hereinafter referred to as “substances for formulation”) can be included. The substance for the preparation is not particularly limited as long as it is a pharmacologically acceptable substance. For example, non-toxicity or low toxicity is a property that a drug substance preferably has.
 製剤用の物質として、例えば、以下のものをあげることができるが、これらに限定されるものではない; グリシン、アラニン、グルタミン、アスパラギン、ヒスチジン、アルギニン又はリジン等のアミノ酸類、抗菌剤、アスコルビン酸、硫酸ナトリウム又は亜硫酸水素ナトリウム等の抗酸化剤、リン酸、クエン酸、ホウ酸バッファー、炭酸水素ナトリウム、トリス-塩酸(Tris-Hcl)溶液等の緩衝剤、マンニトールやグリシン等の充填剤、エチレンジアミン四酢酸(EDTA)等のキレート剤、カフェイン、ポリビニルピロリジン、β-シクロデキストリンやヒドロキシプロピル-β-シクロデキストリン等の錯化剤、グルコース、マンノース又はデキストリン等の増量剤、単糖類、二糖類やグルコース、マンノースやデキストリン等の他の炭水化物、着色剤、香味剤、希釈剤、乳化剤やポリビニルピロリジン等の親水ポリマー、低分子量ポリペプチド、塩形成対イオン、塩化ベンズアルコニウム、安息香酸、サリチル酸、チメロサール、フェネチルアルコール、メチルパラベン、プロピルパラベン、クロレキシジン、ソルビン酸又は過酸化水素等の防腐剤、グリセリン、プロピレングリコール又はポリエチレングリコール等の溶媒、マンニトール又はソルビトール等の糖アルコール、懸濁剤、PEG、ソルビタンエステル、ポリソルビテート20やポリソルビテート80等ポリソルビテート、トリトン(triton)、トロメタミン(tromethamine)、レシチン又はコレステロール等の界面活性剤、スクロースやソルビトール等の安定化増強剤、塩化ナトリウム、塩化カリウム、マンニトールやソルビトール等の弾性増強剤、輸送剤、希釈剤、賦形剤、及び/又は薬学上の補助剤。 Examples of the substance for the preparation include, but are not limited to, the following: amino acids such as glycine, alanine, glutamine, asparagine, histidine, arginine or lysine, antibacterial agent, ascorbic acid Antioxidants such as sodium sulfate or sodium bisulfite, phosphoric acid, citric acid, boric acid buffer, sodium hydrogen carbonate, buffer such as tris-hydrochloric acid (Tris-Hcl) solution, filler such as mannitol and glycine, ethylenediamine Chelating agents such as tetraacetic acid (EDTA), caffeine, polyvinylpyrrolidine, complexing agents such as β-cyclodextrin and hydroxypropyl-β-cyclodextrin, bulking agents such as glucose, mannose or dextrin, monosaccharides, disaccharides, Glucose, mannose and dextri Other carbohydrates such as glycine, coloring agents, flavoring agents, diluents, hydrophilic polymers such as emulsifiers and polyvinylpyrrolidine, low molecular weight polypeptides, salt-forming counterions, benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, Preservatives such as methylparaben, propylparaben, chlorexidine, sorbic acid or hydrogen peroxide, solvents such as glycerin, propylene glycol or polyethylene glycol, sugar alcohols such as mannitol or sorbitol, suspending agents, PEG, sorbitan esters, polysorbate 20 And polysorbites such as 80 and polysorbites, surfactants such as triton, tromethamine, lecithin and cholesterol, stabilization enhancers such as sucrose and sorbitol, sodium chloride Potassium, potassium chloride, mannitol and elasticity enhancers such as sorbitol, transport agent, diluent, excipient, and / or pharmaceutically adjuvants.
 これらの製剤用の物質の添加量は、抗FGFR2抗体もしくはその機能断片またはその修飾体の重量に対して0.001乃至1000倍、好適には0.01乃至100倍、より好適には0.1乃至10倍である。 The amount of these substances for preparations added is 0.001 to 1000 times, preferably 0.01 to 100 times, more preferably 0.001 times the weight of the anti-FGFR2 antibody or functional fragment thereof or a modified product thereof. 1 to 10 times.
 抗FGFR2抗体もしくはその機能断片またはその修飾体をリポソーム中に含有せしめたイムノリポソーム、抗体とリポソームとが結合してなる抗体修飾体(米国特許第6214388号等)を含有する医薬組成物も、本発明の医薬組成物に含まれる。 An immunoliposome containing an anti-FGFR2 antibody or a functional fragment thereof or a modified product thereof in a liposome, or a pharmaceutical composition containing an antibody modified product (US Pat. No. 6,214,388, etc.) formed by binding an antibody and a liposome is also present in the present invention. Included in the pharmaceutical composition of the invention.
 賦形剤や担体は、通常液体又は固体であり、注射用の水、生理食塩水、人工脳脊髄液、その他の、経口投与又は非経口投与用の製剤に用いられる物質であれば特に限定されない。生理食塩水としては、中性のもの、血清アルブミンを含むもの等をあげることができる。 The excipient and carrier are usually liquid or solid, and are not particularly limited as long as they are water used for injection, physiological saline, artificial cerebrospinal fluid, and other substances used for oral or parenteral administration. . Examples of the physiological saline include neutral ones and those containing serum albumin.
 緩衝剤としては、医薬組成物の最終pHが7.0乃至8.5になるように調整されたTrisバッファー、同じく4.0乃至5.5になるように調整された酢酸バッファー、同じく5.0乃至8.0になるように調整されたクエン酸バッファー、同じく5.0乃至8.0になるように調整されたヒスチジンバッファー等を例示することができる。 Examples of the buffer include Tris buffer adjusted so that the final pH of the pharmaceutical composition is 7.0 to 8.5, acetate buffer adjusted so as to be 4.0 to 5.5, and 5. Examples thereof include a citrate buffer adjusted to 0 to 8.0, a histidine buffer adjusted to 5.0 to 8.0, and the like.
 本発明の医薬組成物は、固体、液体、懸濁液等である。凍結乾燥製剤をあげることができる。凍結乾燥製剤を成型するには、スクロース等の賦形剤を用いることができる。 The pharmaceutical composition of the present invention is a solid, liquid, suspension or the like. Freeze-dried preparations can be mentioned. An excipient such as sucrose can be used to mold the lyophilized preparation.
 本発明の医薬組成物の投与径路としては、経腸投与、局所投与及び非経口投与のいずれでもよく、例えば、静脈内投与、動脈内投与、筋肉内投与、皮内投与、皮下投与、腹腔内投与、経皮投与、骨内投与、関節内投与等をあげることができる。 The administration route of the pharmaceutical composition of the present invention may be enteral administration, topical administration or parenteral administration, for example, intravenous administration, intraarterial administration, intramuscular administration, intradermal administration, subcutaneous administration, intraperitoneal administration Administration, transdermal administration, intraosseous administration, intraarticular administration and the like can be mentioned.
 かかる医薬組成物の組成は、投与方法、抗体のFGFR2蛋白質結合親和性等に応じて決定することができる。本発明の抗FGFR2抗体もしくはその機能断片またはその修飾体のFGFR2蛋白質に対する親和性が高いほど(KD値が低いほど)、少ない投与量でその薬効を発揮し得る。 The composition of such a pharmaceutical composition can be determined according to the administration method, the FGFR2 protein binding affinity of the antibody, and the like. The higher the affinity of the anti-FGFR2 antibody of the present invention or a functional fragment thereof or a modified product thereof for the FGFR2 protein (the lower the KD value), the more effective the drug can be exerted.
 本発明の抗FGFR2抗体の投与量は、薬理学的に有効な量であれば限定されず、個体の種、疾患の種類、症状、性別、年齢、持病、該抗体のFGFR2蛋白質結合親和性又はその生物活性、その他の要素に応じて適宜決定することができるが、通常、0.01乃至1000mg/kg、好適には0.1乃至100mg/kgを、1乃至180日間に1回、又は1日2回若しくは3回以上投与することができる。 The dose of the anti-FGFR2 antibody of the present invention is not limited as long as it is a pharmacologically effective amount, and the species of the individual, the type of disease, symptoms, sex, age, chronicity, FGFR2 protein binding affinity of the antibody or Although it can be appropriately determined depending on its biological activity and other factors, it is usually 0.01 to 1000 mg / kg, preferably 0.1 to 100 mg / kg once every 1 to 180 days, or 1 It can be administered twice or more times a day.
 医薬組成物の形態としては、注射剤(凍結乾燥製剤、点滴剤を含む)、坐剤、経鼻型吸収製剤、経皮型吸収製剤、舌下剤、カプセル、錠剤、軟膏剤、顆粒剤、エアーゾル剤、丸剤、散剤、懸濁剤、乳剤、点眼剤、生体埋め込み型製剤等を例示することができる。 The form of the pharmaceutical composition includes injections (including lyophilized preparations and infusions), suppositories, nasal absorption preparations, transdermal absorption preparations, sublingual preparations, capsules, tablets, ointments, granules, aerosols. Examples thereof include pills, pills, powders, suspensions, emulsions, eye drops, and implantable preparations.
 抗FGFR2抗体もしくはその機能断片またはその修飾体(以下「抗FGFR2抗体等」という)は、他剤と組み合わせて用いることができる。抗FGFR2等、またはそれを有効成分として含む医薬組成物は、他剤、すなわち抗FGFR2抗体等以外の化合物を有効成分としての医薬組成物と同時にあるいは個別に投与することができる。例えば、他剤を投与した後に抗FGFR2抗体等を有効成分として含む医薬組成物を投与するか、抗FGFR2抗体等を有効成分として含む医薬組成物を投与した後に、他剤を投与するか、または、抗FGFR2抗体等を有効成分として含む医薬組成物と他剤とを同時に投与してもよい。単一の医薬組成物中に抗FGFR2抗体等および他剤の両方の有効成分が含まれる場合と、両有効成分が複数の医薬組成物に分かれて含まれる場合のいずれも、本発明においては「抗FGFR2抗体等および他剤を含む医薬組成物」という。本発明において、かかる「医薬組成物」は「抗FGFR2抗体等と他剤を組み合わせて投与される医薬組成物」と同義である。 The anti-FGFR2 antibody or a functional fragment thereof or a modified form thereof (hereinafter referred to as “anti-FGFR2 antibody etc.”) can be used in combination with other agents. Anti-FGFR2 or the like, or a pharmaceutical composition containing it as an active ingredient, can be administered simultaneously with or separately from other agents, that is, compounds other than anti-FGFR2 antibody or the like as the active ingredient. For example, after administering another agent, a pharmaceutical composition containing an anti-FGFR2 antibody or the like as an active ingredient is administered, or after administering a pharmaceutical composition containing an anti-FGFR2 antibody or the like as an active ingredient, or the other agent is administered, or A pharmaceutical composition containing an anti-FGFR2 antibody or the like as an active ingredient and another agent may be administered simultaneously. In the present invention, both the case where the active ingredients of both the anti-FGFR2 antibody and the like and other agents are contained in a single pharmaceutical composition, and the case where both active ingredients are separately contained in a plurality of pharmaceutical compositions are used in the present invention. It is referred to as “pharmaceutical composition containing anti-FGFR2 antibody and the like and other agents”. In the present invention, such a “pharmaceutical composition” is synonymous with “a pharmaceutical composition administered in combination with an anti-FGFR2 antibody or the like and another agent”.
 本発明において、抗FGFR2抗体等と他剤を「組み合わせて投与される」とは、ある一定期間において、被投与対象の体内に、抗FGFR2抗体等と他剤が取り込まれることを意味する。抗FGFR2抗体等と他剤が単一製剤中に含まれた製剤が投与されてもよく、またそれぞれが別々に製剤化され、別々に投与されても良い。別々に製剤化される場合、その投与の時期は特に限定されず、同時に投与されてもよく、時間を置いて異なる時間に、又は、異なる日に、投与されても良い。抗FGFR2抗体等と他剤が、それぞれ異なる時間又は日に投与される場合、その投与の順番は特に限定されない。通常、それぞれの製剤は、それぞれの投与方法に従って投与されるため、それらの投与は、同一回数となる場合もあり、異なる回数となる場合もある。また、それぞれが別々に製剤化される場合、各製剤の投与方法(投与経路)は同じであってもよく、異なる投与方法(投与経路)で投与されてもよい。また、抗FGFR2抗体等と他剤が同時に体内に存在する必要は無く、ある一定期間(例えば一ヶ月間、好ましくは1週間、さらに好ましくは数日間、さらにより好ましくは1日間)の間に体内に取り込まれていればよく、いずれかの投与時にもう一方の有効成分が体内から消失していてもよい。 In the present invention, “administered in combination” with an anti-FGFR2 antibody or the like and another agent means that the anti-FGFR2 antibody or the like and the other agent are taken into the body of the administration target for a certain period. A preparation containing an anti-FGFR2 antibody or the like and another agent in a single preparation may be administered, or each may be formulated separately and administered separately. When formulated separately, the timing of administration is not particularly limited, and may be administered at the same time, and may be administered at different times or on different days. When the anti-FGFR2 antibody and other agents are administered at different times or days, the order of administration is not particularly limited. Usually, since each formulation is administered according to each administration method, the administration may be the same number of times or may be different times. Moreover, when each is formulated separately, the administration method (administration route) of each formulation may be the same, and may be administered by different administration methods (administration route). Further, the anti-FGFR2 antibody and the like and the other agent need not be present in the body at the same time, and may be present in the body during a certain period (for example, one month, preferably one week, more preferably several days, even more preferably one day). The other active ingredient may disappear from the body at any time of administration.
 「抗FGFR2抗体等と他剤を組み合わせて投与される医薬組成物」の投与形態としては、例えば、1)抗FGFR2抗体等と他剤を含む単一の製剤の投与、2)抗FGFR2抗体等と他剤とを別々に製剤化して得られる2種の製剤の同一投与経路での同時投与、3)抗FGFR2抗体等と他剤を別々に製剤化して得られる2種の製剤の同一投与経路での時間差をおいての投与、4)抗FGFR2抗体等と他剤を別々に製剤化して得られる2種の製剤の異なる投与経路での同時投与、5)抗FGFR2抗体等と他剤とを別々に製剤化して得られる2種の製剤の異なる投与経路での時間差をおいての投与などを挙げることができる。「抗FGFR2抗体等と他剤を組み合わせて投与される医薬組成物」の投与量、投与間隔、投与形態、製剤等は、抗FGFR2抗体を含有する医薬組成物のそれらに準ずるが、それらに限定されるものではない。 Examples of the administration form of “pharmaceutical composition administered in combination with anti-FGFR2 antibody etc.” and other agents include, for example, 1) administration of a single preparation containing anti-FGFR2 antibody etc. and other agents, 2) anti-FGFR2 antibody etc. 3) Simultaneous administration of the two preparations obtained by separately formulating the other agent with the same administration route, 3) Same administration route of the two preparations obtained by separately formulating the anti-FGFR2 antibody and the other agent 4) Simultaneous administration of two preparations obtained by separately formulating anti-FGFR2 antibody and other agents in different administration routes, 5) Anti-FGFR2 antibody and other agents For example, administration of two kinds of preparations obtained by separate preparations at different time intervals in different administration routes can be mentioned. The dosage, administration interval, dosage form, formulation, etc. of the “pharmaceutical composition administered in combination with an anti-FGFR2 antibody and the like” are based on those of the pharmaceutical composition containing the anti-FGFR2 antibody, but are not limited thereto. Is not to be done.
 かかる医薬組成物は、それが2種の異なる製剤にされた場合には、それらを含むキットであってもよい。 Such pharmaceutical compositions may be kits containing them when they are made into two different formulations.
 本発明において、抗FGFR2抗体等および他剤の「組合せ」とは、抗FGFR2抗体等および他剤を「組み合わせて投与される」ことを意味する。   In the present invention, the “combination” of anti-FGFR2 antibody and the like and other agents means that “anti-FGFR2 antibody and the like and other agents are“ administered in combination ”. *
 本発明において、「他剤」は抗癌剤であれは特に限定されるものではなく、好適には化学療法剤、分子標的薬、癌治療抗体、ホルモン剤であり、より好適にはトポイソメラーゼ阻害剤、微小管阻害剤、プラチナ製剤、抗VEGFR2抗体、代謝拮抗剤である。トポイソメラーゼ阻害剤の好適な例としては、イリノテカン、トポテカン、エトポシド等を挙げることができる。微小管阻害剤の好適な例としては、パクリタキセル、ドセタキセル、ビンクリスチン、ビンブラスチン、ビンデシン、エリブリン、ビノレルビン、パクリタクセル内包ミセルNK105等を挙げることができる。プラチナ製剤の好適な例としては、オキサルプラチン、カルボプラチン、シスプラチン、ネダプラチン、シスプラチン内包ミセルNC-6004等を挙げることができる。抗VEGFR2抗体の好適な例としては、ラムシルマブ等を挙げることができる。代謝拮抗剤としては、5-フルオロウラシル系抗がん剤(例えば、5-FU又は生体内において5-FUに代謝され抗腫瘍効果を発揮する化合物等)が好ましく、5-フルオロウラシル系抗がん剤の好適な例としては、フルオロウラシル(5-FU)、テガフール、テガフール・ギメラシル・オテラシルカリウム(S-1又はTS-1)、テガフール・ウラシル(UFT)、カペシタビン、カルモフール、ドキシフルリジン等を挙げることができる。ただし、本発明の組合せ又は医薬組成物に含まれる「他剤」はこれらに限定されない。 In the present invention, the “other agent” is not particularly limited as long as it is an anticancer agent, and is preferably a chemotherapeutic agent, a molecular target agent, a cancer therapeutic antibody, or a hormonal agent, more preferably a topoisomerase inhibitor, Tube inhibitor, platinum preparation, anti-VEGFR2 antibody, antimetabolite. Preferable examples of the topoisomerase inhibitor include irinotecan, topotecan, etoposide and the like. Preferable examples of the microtubule inhibitor include paclitaxel, docetaxel, vincristine, vinblastine, vindesine, eribulin, vinorelbine, paclitaxel-containing micelle NK105, and the like. Preferable examples of the platinum preparation include oxalplatin, carboplatin, cisplatin, nedaplatin, cisplatin-containing micelle NC-6004 and the like. Preferable examples of the anti-VEGFR2 antibody include ramucirumab. The antimetabolite is preferably a 5-fluorouracil anticancer agent (for example, 5-FU or a compound that is metabolized to 5-FU in vivo and exhibits an antitumor effect), and is a 5-fluorouracil anticancer agent. Preferred examples of such include fluorouracil (5-FU), tegafur, tegafur gimeracil oteracil potassium (S-1 or TS-1), tegafur uracil (UFT), capecitabine, carmofur, doxyfluridine and the like. it can. However, the “other agent” contained in the combination or pharmaceutical composition of the present invention is not limited thereto.
 本発明の組合せ又は医薬組成物には、さらに他の医薬を使用することができる。さらなる他の医薬としては、ワクチン、放射線療法(放射線療法剤)など各種治療法等をあげることができる。 In the combination or pharmaceutical composition of the present invention, other pharmaceuticals can be used. Still other medicaments include various therapeutic methods such as vaccines and radiation therapy (radiotherapy agents).
 本発明は癌などFGFRに関わる疾患の治療方法または予防方法、該疾患の治療用または予防用医薬組成物を調製するための本発明の抗体の使用、該疾患の治療または予防のための本発明の抗体の使用をも提供する。本発明の抗体を含む治療用または予防用キットも本発明に含まれる。 The present invention relates to a method for treating or preventing a disease associated with FGFR such as cancer, use of the antibody of the present invention for preparing a pharmaceutical composition for treating or preventing the disease, and the present invention for treating or preventing the disease. Also provided is the use of antibodies. A therapeutic or prophylactic kit containing the antibody of the present invention is also included in the present invention.
 以下、実施例において本発明を更に詳細に説明するが、本発明はこれらに限定されない。 Hereinafter, the present invention will be described in more detail with reference to Examples, but the present invention is not limited thereto.
 なお、下記実施例に関する各操作は特に明示がない限り、当業者が使用する実験書に記載の方法により行うか、または、市販の試薬やキットを用いる場合には市販品の指示書に従って行った。 In addition, unless otherwise specified, each operation relating to the following examples was performed by the method described in the experiment document used by those skilled in the art, or when using a commercially available reagent or kit, it was performed according to the instructions for the commercially available product. .
 実施例1. FGFR2発現胃癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)とCPT-11とのin vivo併用効果
 ヒト胃癌細胞株SNU―16(ATCCより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて5×107cells/mLに調整し、細胞懸濁液をヌードマウス(CAnN.Cg-Foxnlnu/CrlCrlj、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(各群n=5)、hFR2-14_H19/L1(WO2013/154206)は10mg/kgにて腹腔内に投与し、CPT-11は40mg/kgにてマウスに尾静脈内投与した。hFR2-14_H19/L1は移植10、17、24日後に投与し、CPT-11は移植10日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
Example 1. In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and CPT-11 against FGFR2-expressing gastric cancer cell line Human gastric cancer cell line SNU-16 (purchased from ATCC) PBS and Matrigel (purchased from Japan BD) The cell suspension was adjusted to 5 × 10 7 cells / mL with a 1: 1 mixture, and the cell suspension was nude mice (CAnN.Cg-Foxnl nu / CrlCrlj, female, 5-6 weeks old, purchased from Charles River, Japan) 0.1 mL was transplanted subcutaneously into the axilla. Grouping was performed using the tumor volume value (each group n = 5), hFR2-14_H19 / L1 (WO2013 / 154206) was intraperitoneally administered at 10 mg / kg, and CPT-11 was administered at 40 mg / kg in mice. Administered into the tail vein. hFR2-14_H19 / L1 was administered 10, 17, and 24 days after transplantation, and CPT-11 was administered 10 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
 腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植36日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 36 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
 hFR2-14_H19/L1単独投与、CPT-11単独投与、およびhFR2-14_H19/L1とCPT-11併用投与の結果を図13に示した。数値は平均値±標準誤差(SE)を示す。移植36日後におけるTGIは、hFR2-14_H19/L1単独投与群で24%、CPT-11単独投与群で41%であったのに対し、hFR2-14_H19/L1とCPT-11併用投与群では80%であり、併用により抗腫瘍効果が増強されることが示された。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
The results of hFR2-14_H19 / L1 single administration, CPT-11 single administration, and hFR2-14_H19 / L1 and CPT-11 combined administration are shown in FIG. A numerical value shows an average value +/- standard error (SE). TGI at 36 days after transplantation was 24% in the hFR2-14_H19 / L1 single administration group and 41% in the CPT-11 single administration group, compared to 80% in the hFR2-14_H19 / L1 and CPT-11 combination administration group It was shown that the antitumor effect was enhanced by the combined use.
 実施例2. FGFR2発現胃癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)とPaclitaxelとのin vivo併用効果
 ヒト胃癌細胞株SNU―16(ATCCより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて5×107cells/mLに調整し、細胞懸濁液をNOD-scidマウス(NOD.CB17-Prkdcscid/J、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(Paclitaxel単独投与群はn=4、他の群はn=5)、hFR2-14_H19/L1は10mg/kgにて腹腔内に投与し、Paclitaxelは15mg/kgにてマウスに尾静脈内投与した。hFR2-14_H19/L1は移植10、17、24日後に投与し、Paclitaxelは移植10日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植39日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Example 2 In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and Paclitaxel against FGFR2-expressing gastric cancer cell line Human gastric cancer cell line SNU-16 (purchased from ATCC) PBS and Matrigel (purchased from Japan BD) 1: The cell suspension was adjusted to 5 × 10 7 cells / mL with 1 mixture, and the cell suspension was NOD-scid mice (NOD.CB17-Prkdc scid / J, female, 5-6 weeks old, purchased from Charles River, Japan) 0.1 mL was transplanted subcutaneously into the axilla. The tumor volume was used for grouping (n = 4 in the Paclitaxel single administration group, n = 5 in the other groups), hFR2-14_H19 / L1 was intraperitoneally administered at 10 mg / kg, and Paclitaxel was 15 mg / kg. Mice were administered intravenously into the tail vein. hFR2-14_H19 / L1 was administered 10, 17, and 24 days after transplantation, and Paclitaxel was administered 10 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 39 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
hFR2-14_H19/L1単独投与、Paclitaxel単独投与、およびhFR2-14_H19/L1とPaclitaxel併用投与の結果を図14に示した。数値は平均値±標準誤差(SE)を示す。移植39日後におけるTGIは、hFR2-14_H19/L1単独投与群で60%、Paclitaxel単独投与群で53%であったのに対し、hFR2-14_H19/L1とPaclitaxel併用投与群では、98%であった。先行技術の抗FGFR2抗体では、Paclitaxelとの併用により、抗腫瘍効果が増強することが示されているが、腫瘍が縮退するには到らなかった(US2015/0050273 A1)。一方、本発明におけるCPT-11あるいはPaclitaxelとの併用においては、併用効果により大幅な抗腫瘍活性の増強と、腫瘍縮退が認められ、とりわけPaclitaxelとの併用では、39日において5例中3例に腫瘍の完全消失が認められた。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
The results of hFR2-14_H19 / L1 single administration, Paclitaxel single administration, and hFR2-14_H19 / L1 and Paclitaxel combined administration are shown in FIG. A numerical value shows an average value +/- standard error (SE). TGI 39 days after transplantation was 60% in the hFR2-14_H19 / L1 single administration group and 53% in the Paclitaxel single administration group, compared with 98% in the hFR2-14_H19 / L1 and Paclitaxel combination administration group. . The anti-FGFR2 antibody of the prior art has been shown to enhance the antitumor effect when used in combination with Paclitaxel, but the tumor has not reached degeneracy (US2015 / 0050273 A1). On the other hand, in the combined use with CPT-11 or Paclitaxel in the present invention, significant enhancement of antitumor activity and tumor degeneration were observed due to the combined use effect. In particular, in the combined use with Paclitaxel, 3 out of 5 cases in 39 days Complete disappearance of the tumor was observed.
 実施例3. FGFR2発現肺癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)とCisplatinとのin vivo併用効果
 ヒト肺癌細胞株KNS―62(財団法人ヒューマンサイエンス振興財団、ヒューマンサイエンス研究資源バンクより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて1.4×107cells/mLに調整し、細胞懸濁液をNOD-scidマウス(NOD.CB17-Prkdcscid/J、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(各群n=5)、hFR2-14_H19/L1は10mg/kgにて腹腔内に投与し、Cisplatinは5mg/kgにてマウスに尾静脈内投与した。hFR2-14_H19/L1は移植7、14、21日後に投与し、Cisplatinは移植7、21日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
Example 3 FIG. In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and cisplatin against FGFR2-expressing lung cancer cell line Human lung cancer cell line KNS-62 (Purchased from Human Science Promotion Foundation, Human Science Research Resource Bank) PBS and Matrigel (purchased from Japan BD) were adjusted to 1.4 × 10 7 cells / mL with a 1: 1 mixture, and the cell suspension was treated with NOD-scid mice (NOD.CB17-Prkdc scid / J, female, 0.1 mL was transplanted subcutaneously into the axilla of 5-6 weeks old (purchased from Charles River, Japan). The tumor volume value was used for grouping (each group n = 5), hFR2-14_H19 / L1 was administered intraperitoneally at 10 mg / kg, and Cisplatin was administered into mice via the tail vein at 5 mg / kg. hFR2-14_H19 / L1 was administered 7, 14, 21 days after transplantation, and Cisplatin was administered 7, 21 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
 腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植36日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 36 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
hFR2-14_H19/L1単独投与、Cisplatin単独投与、およびhFR2-14_H19/L1とCisplatin併用投与の結果を図15に示した。数値は平均値±標準誤差(SE)を示す。移植36日後におけるTGIは、hFR2-14_H19/L1単独投与群で16%、Cisplatin単独投与群で27%であったのに対し、hFR2-14_H19/L1とCisplatin併用投与群では、52%であった。無処置対照群(Untreated control)とhFR2-14_H19/L1とCisplatin併用投与群との間に、有意差(t検定、p<0.05)が認められ、併用効果が示された。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
FIG. 15 shows the results of hFR2-14_H19 / L1 single administration, Cisplatin single administration, and hFR2-14_H19 / L1 and Cisplatin combination administration. A numerical value shows an average value +/- standard error (SE). TGI at 36 days after transplantation was 16% in the hFR2-14_H19 / L1 single administration group and 27% in the Ciplatin single administration group, compared with 52% in the hFR2-14_H19 / L1 and Cisplatin combination administration group. . A significant difference (t test, p <0.05) was observed between the untreated control group and the hFR2-14_H19 / L1 and Cisplatin combination administration group, indicating a combined effect.
 実施例4. FGFR2発現胃癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)とCisplatinとのin vivo併用効果
 ヒト胃癌細胞株SNU-16(ATCCより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて5×10cells/mLに調整し、細胞懸濁液をヌードマウス(CAnN.Cg-Foxn1[nu]/CrlCrlj[Foxn1nu/Foxn1nu]、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(各群n=8)、hFR2-14_H19/L1は1mg/kgにて、Cisplatinは5mg/kgにてマウスにそれぞれ尾静脈内投与した。hFR2-14_H19/L1は移植6、13、20日後に投与し、Cisplatinは移植6日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植27日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Example 4 In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and Cisplatin against FGFR2-expressing gastric cancer cell line Human gastric cancer cell line SNU-16 (purchased from ATCC) PBS and Matrigel (purchased from Japan BD) 1: The cell suspension was adjusted to 5 × 10 7 cells / mL with a mixed solution of 1 and nude cells (CAnN.Cg-Foxn1 [nu] / CrlCrlj [Foxn1nu / Foxn1nu], female, 5-6 weeks old, Charles, Japan) (Purchased from River) 0.1 mL was transplanted subcutaneously into the axilla. The mice were divided into groups using the tumor volume values (n = 8 for each group), and hFR2-14_H19 / L1 was administered into the tail vein at 1 mg / kg and Cisplatin at 5 mg / kg. hFR2-14_H19 / L1 was administered 6, 13, and 20 days after transplantation, and Cisplatin was administered 6 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 27 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
hFR2-14_H19/L1単独投与、Cisplatin単独投与、およびhFR2-14_H19/L1とCisplatin併用投与の結果を図17に示した。数値は平均値±標準誤差(SE)を示す。移植27日後におけるTGIは、hFR2-14_H19/L1単独投与群で68%、Cisplatin単独投与群で23%であったのに対し、hFR2-14_H19/L1とCisplatin併用投与群では、87%であった。Cisplatin単独投与群と、hFR2-14_H19/L1とCisplatin併用投与群との間に、有意差(Dunnett型多重比較検定、p<0.0001)が認められ、併用効果が示された。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
The results of hFR2-14_H19 / L1 single administration, Cisplatin single administration, and hFR2-14_H19 / L1 and Cisplatin combination administration are shown in FIG. A numerical value shows an average value +/- standard error (SE). TGI 27 days after transplantation was 68% in the hFR2-14_H19 / L1 single administration group and 23% in the Ciplatin single administration group, compared with 87% in the hFR2-14_H19 / L1 and Cisplatin combination administration group. . A significant difference (Dunnett multiple comparison test, p <0.0001) was observed between the Cisplatin single administration group and the hFR2-14_H19 / L1 and Cisplatin combination administration group, indicating a combined effect.
 実施例5. FGFR2発現胃癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)とDocetaxelとのin vivo併用効果
 ヒト胃癌細胞株SNU-16(ATCCより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて5×10cells/mLに調整し、細胞懸濁液をヌードマウス(CAnN.Cg-Foxn1[nu]/CrlCrlj[Foxn1nu/Foxn1nu]、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(各群n=8)、hFR2-14_H19/L1は1mg/kgにて、Docetaxelは1mg/kgにてマウスにそれぞれ尾静脈内投与した。hFR2-14_H19/L1は移植6、13、20日後に投与し、Docetaxelは移植6日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植27日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Embodiment 5 FIG. In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and Docetaxel against FGFR2-expressing gastric cancer cell line Human gastric cancer cell line SNU-16 (purchased from ATCC) PBS and Matrigel (purchased from Japan BD) 1: The cell suspension was adjusted to 5 × 10 7 cells / mL with a mixed solution of 1 and nude cells (CAnN.Cg-Foxn1 [nu] / CrlCrlj [Foxn1nu / Foxn1nu], female, 5-6 weeks old, Charles, Japan) (Purchased from River) 0.1 mL was transplanted subcutaneously into the axilla. The mice were divided into groups using the tumor volume values (n = 8 for each group), and hFR2-14_H19 / L1 was administered to the mice at 1 mg / kg and Docetaxel was administered to the mice at 1 mg / kg via the tail vein. hFR2-14_H19 / L1 was administered 6, 13, and 20 days after transplantation, and Docetaxel was administered 6 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 27 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
hFR2-14_H19/L1単独投与、Docetaxel単独投与、およびhFR2-14_H19/L1とDocetaxel併用投与の結果を図18に示した。数値は平均値±標準誤差(SE)を示す。移植27日後におけるTGIは、hFR2-14_H19/L1単独投与群で68%、Docetaxel単独投与群で16%であったのに対し、hFR2-14_H19/L1とDocetaxel併用投与群では、73%であった。Docetaxel単独投与群と、hFR2-14_H19/L1とDocetaxel併用投与群との間に、有意差(Dunnett型多重比較検定、p<0.005)が認められ、併用効果が示された。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
The results of hFR2-14_H19 / L1 single administration, Docetaxel single administration, and hFR2-14_H19 / L1 and Docetaxel combined administration are shown in FIG. A numerical value shows an average value +/- standard error (SE). TGI 27 days after transplantation was 68% in the hFR2-14_H19 / L1 single administration group and 16% in the Docetaxel single administration group, compared with 73% in the hFR2-14_H19 / L1 and Docetaxel combination administration group. . A significant difference (Dunnett multiple comparison test, p <0.005) was observed between the Docetaxel single administration group and the hFR2-14_H19 / L1 and Docetaxel combination administration group, indicating a combined effect.
 実施例6. FGFR2発現胃癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)と抗VEGFR2抗体とのin vivo併用効果
 ヒト胃癌細胞株SNU-16(ATCCより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて5×10cells/mLに調整し、細胞懸濁液をヌードマウス(CAnN.Cg-Foxn1[nu]/CrlCrlj[Foxn1nu/Foxn1nu]、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(各群n=8)、hFR2-14_H19/L1は1mg/kgにて、抗マウスVEGFR2抗体DC101(Bio-X-cellより購入)は20mg/kgにてマウスにそれぞれ尾静脈内投与した。hFR2-14_H19/L1は移植6、13、20日後に投与し、抗VEGFR2抗体は移植6、9、13、16、20日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植27日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Example 6 In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and anti-VEGFR2 antibody against FGFR2-expressing gastric cancer cell line Human gastric cancer cell line SNU-16 (purchased from ATCC) PBS and Matrigel (purchased from Japan BD) The cell suspension was adjusted to 5 × 10 7 cells / mL with a 1: 1 mixture, and the cell suspension was treated with nude mice (CAnN.Cg-Foxn1 [nu] / CrlCrlj [Foxn1nu / Foxn1nu], female, 5-6 weeks old, 0.1 mL was transplanted subcutaneously into the axilla region (purchased from Japan Charles River). Grouping was performed using tumor volume values (n = 8 for each group), hFR2-14_H19 / L1 at 1 mg / kg, and anti-mouse VEGFR2 antibody DC101 (purchased from Bio-X-cell) at 20 mg / kg. Each mouse was administered via the tail vein. hFR2-14_H19 / L1 was administered 6, 13, and 20 days after transplantation, and anti-VEGFR2 antibody was administered 6, 9, 13, 16, and 20 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 27 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
hFR2-14_H19/L1単独投与、抗VEGFR2抗体単独投与、およびhFR2-14_H19/L1と抗VEGFR2抗体併用投与の結果を図19に示した。数値は平均値±標準誤差(SE)を示す。移植27日後におけるTGIは、hFR2-14_H19/L1単独投与群で68%、抗VEGFR2抗体単独投与群で28%であったのに対し、hFR2-14_H19/L1と抗VEGFR2抗体併用投与群では、91%であった。抗VEGFR2抗体単独投与群と、hFR2-14_H19/L1と抗VEGFR2抗体併用投与群との間に、有意差(Dunnett型多重比較検定、p<0.0001)が認められ、併用効果が示された。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
The results of hFR2-14_H19 / L1 single administration, anti-VEGFR2 antibody single administration, and hFR2-14_H19 / L1 and anti-VEGFR2 antibody combination administration are shown in FIG. A numerical value shows an average value +/- standard error (SE). TGI 27 days after transplantation was 68% in the hFR2-14_H19 / L1 single administration group and 28% in the anti-VEGFR2 antibody single administration group, whereas it was 91 in the hFR2-14_H19 / L1 and anti-VEGFR2 antibody combination administration group. %Met. There was a significant difference (Dunnett multiple comparison test, p <0.0001) between the anti-VEGFR2 antibody alone administration group and the hFR2-14_H19 / L1 and anti-VEGFR2 antibody combination administration group, indicating the combined effect .
 実施例7. FGFR2発現胃癌細胞株に対するヒト化抗FGFR2抗体(hFR2-14_H19/L1)と5-FUとのin vivo併用効果
 ヒト胃癌細胞株SNU-16(ATCCより購入)をPBSとマトリゲル(日本BDより購入)1:1の混液にて5×10cells/mLに調整し、細胞懸濁液をヌードマウス(CAnN.Cg-Foxn1[nu]/CrlCrlj[Foxn1nu/Foxn1nu]、雌性、5~6週齢、日本チャールス・リバーより購入)の腋窩部皮下に0.1mL移植した。腫瘍体積値を用いて群分けを行い(各群n=8)、hFR2-14_H19/L1は1mg/kgにて、5-FUは50mg/kgにてマウスにそれぞれ尾静脈内投与した。hFR2-14_H19/L1は移植7、14日後に投与し、5-FUは移植7、10、14、17日後に投与した。経時的に腫瘍の長径(mm)および短径(mm)を電子デジタルノギス(株式会社ミツトヨ製)で計測し、以下に示す計算式により腫瘍体積を算出した。
腫瘍体積(mm)=1/2×[腫瘍長径]×[腫瘍短径]×[腫瘍短径]
抗腫瘍効果は、移植21日後に以下に示す計算式により腫瘍増殖抑制率(TGI%)で評価した。
Example 7 In vivo combined effect of humanized anti-FGFR2 antibody (hFR2-14_H19 / L1) and 5-FU against FGFR2-expressing gastric cancer cell line Human gastric cancer cell line SNU-16 (purchased from ATCC) PBS and Matrigel (purchased from Japan BD) The cell suspension was adjusted to 5 × 10 7 cells / mL with a 1: 1 mixture, and the cell suspension was treated with nude mice (CAnN.Cg-Foxn1 [nu] / CrlCrlj [Foxn1nu / Foxn1nu], female, 5-6 weeks old, 0.1 mL was transplanted subcutaneously into the axilla region (purchased from Japan Charles River). The mice were divided into groups using the tumor volume values (n = 8 for each group), and hFR2-14_H19 / L1 was administered into the tail vein at 1 mg / kg and 5-FU at 50 mg / kg. hFR2-14_H19 / L1 was administered 7, 14 days after transplantation, and 5-FU was administered 7, 10, 14, 17 days after transplantation. The major axis (mm) and minor axis (mm) of the tumor were measured over time with an electronic digital caliper (manufactured by Mitutoyo Corporation), and the tumor volume was calculated using the following formula.
Tumor volume (mm 3 ) = 1/2 × [tumor major axis] × [tumor minor axis] × [tumor minor axis]
The antitumor effect was evaluated by the tumor growth inhibition rate (TGI%) by the following formula 21 days after transplantation.
   TGI (%) = (1-A/B)×100  
   A : 化合物投与群の判定日の平均腫瘍体積 
   B : 無処置対照群の判定日の平均腫瘍体積 
hFR2-14_H19/L1単独投与、5-FU単独投与、およびhFR2-14_H19/L1と5-FU併用投与の結果を図20に示した。数値は平均値±標準誤差(SE)を示す。移植21日後におけるTGIは、hFR2-14_H19/L1単独投与群で49%、5-FU単独投与群で32%であったのに対し、hFR2-14_H19/L1と5-FU併用投与群では、79%であった。5-FU単独投与群と、hFR2-14_H19/L1と5-FU併用投与群との間に、有意差(Dunnett型多重比較検定、p<0.0001)が認められ、併用効果が示された。
TGI (%) = (1-A / B) x 100
A: Average tumor volume on the date of determination in the compound administration group
B: Average tumor volume on the date of determination in the untreated control group
FIG. 20 shows the results of hFR2-14_H19 / L1 single administration, 5-FU single administration, and hFR2-14_H19 / L1 and 5-FU combined administration. A numerical value shows an average value +/- standard error (SE). TGI 21 days after transplantation was 49% in the hFR2-14_H19 / L1 single administration group and 32% in the 5-FU single administration group, whereas it was 79% in the hFR2-14_H19 / L1 and 5-FU combined administration group. %Met. A significant difference (Dunnett-type multiple comparison test, p <0.0001) was observed between the 5-FU single administration group and the hFR2-14_H19 / L1 and 5-FU combination administration group, indicating a combination effect. .
 本発明の提供する抗体と他剤の組合せにより各種癌の治療または予防が可能となる。 The combination of the antibody and other agents provided by the present invention makes it possible to treat or prevent various cancers.
配列番号1:ヒト化FR2-14軽鎖(hFR2-14_L1)のCDR1のアミノ酸配列(図1)
配列番号2:ヒト化FR2-14軽鎖(hFR2-14_L1)のCDR2のアミノ酸配列(図2)
配列番号3:ヒト化FR2-14軽鎖(hFR2-14_L1)のCDR3のアミノ酸配列(図3)
配列番号4:ヒト化FR2-14重鎖(hFR2-14_H19)のCDR1のアミノ酸配列(図4)
配列番号5:ヒト化FR2-14重鎖(hFR2-14_H19)のCDR2のアミノ酸配列(図5)
配列番号6:ヒト化FR2-14重鎖(hFR2-14_H19)のCDR3のアミノ酸配列(図6)
配列番号7:ヒト化FR2-14軽鎖(hFR2-14_L1)の塩基配列。うちヌクレオチド番号1乃至60はシグナル配列をコードする配列である。
配列番号8:ヒト化FR2-14軽鎖(hFR2-14_L1)のアミノ酸配列(図7)。うちアミノ酸番号1乃至20はシグナル配列であり、通常大部分の成熟hFR2-14_L1のアミノ酸配列には含まれない。
配列番号9:ヒト化FR2-14重鎖(hFR2-14_H5)の塩基配列。うちヌクレオチド番号1乃至57はシグナル配列をコードする配列である。
配列番号10:ヒト化FR2-14重鎖(hFR2-14_H5)のアミノ酸配列(図8)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H5のアミノ酸配列には含まれない。
配列番号11:ヒト化FR2-14重鎖(hFR2-14_H8)の塩基配列。うちヌクレオチド番号1乃至57はシグナル配列をコードする配列である。
配列番号12:ヒト化FR2-14重鎖(hFR2-14_H8)のアミノ酸配列(図9)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H8のアミノ酸配列には含まれない。
配列番号13:ヒト化FR2-14重鎖(hFR2-14_H11)の塩基配列。うちヌクレオチド番号1乃至57はシグナル配列をコードする配列である。
配列番号14:ヒト化FR2-14重鎖(hFR2-14_H11)のアミノ酸配列(10)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H11のアミノ酸配列には含まれない。
配列番号15:ヒト化FR2-14重鎖(hFR2-14_H12)の塩基配列。うちヌクレオチド番号1乃至57はシグナル配列をコードする配列である。
配列番号16:ヒト化FR2-14重鎖(hFR2-14_H12)のアミノ酸配列(図11)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H12のアミノ酸配列には含まれない。
配列番号17:ヒト化FR2-14重鎖(hFR2-14_H19)の塩基配列。うちヌクレオチド番号1乃至57はシグナル配列をコードする配列である。
配列番号18:ヒト化FR2-14重鎖(hFR2-14_H19)のアミノ酸配列(図12)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H19のアミノ酸配列には含まれない。
配列番号19:ヒト化FR2-14重鎖(hFR2-14_H9)の塩基配列。うちヌクレオチド番号1乃至57はシグナル配列をコードする配列である。
配列番号20:ヒト化FR2-14重鎖(hFR2-14_H9)のアミノ酸配列(図16)。うちアミノ酸番号1乃至19はシグナル配列であり、通常大部分の成熟hFR2-14_H9のアミノ酸配列には含まれない。
SEQ ID NO: 1: Amino acid sequence of CDR1 of humanized FR2-14 light chain (hFR2-14_L1) (FIG. 1)
SEQ ID NO: 2: amino acid sequence of CDR2 of humanized FR2-14 light chain (hFR2-14_L1) (FIG. 2)
SEQ ID NO: 3: Amino acid sequence of CDR3 of humanized FR2-14 light chain (hFR2-14_L1) (FIG. 3)
SEQ ID NO: 4: Amino acid sequence of CDR1 of humanized FR2-14 heavy chain (hFR2-14_H19) (FIG. 4)
SEQ ID NO: 5: Amino acid sequence of CDR2 of humanized FR2-14 heavy chain (hFR2-14_H19) (FIG. 5)
SEQ ID NO: 6: amino acid sequence of CDR3 of humanized FR2-14 heavy chain (hFR2-14_H19) (FIG. 6)
SEQ ID NO: 7: nucleotide sequence of humanized FR2-14 light chain (hFR2-14_L1) Among these, nucleotide numbers 1 to 60 are sequences encoding a signal sequence.
SEQ ID NO: 8: Amino acid sequence of humanized FR2-14 light chain (hFR2-14_L1) (FIG. 7). Among them, amino acid numbers 1 to 20 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_L1.
SEQ ID NO: 9: nucleotide sequence of humanized FR2-14 heavy chain (hFR2-14_H5) Of these, nucleotide numbers 1 to 57 are sequences encoding a signal sequence.
SEQ ID NO: 10: Amino acid sequence of humanized FR2-14 heavy chain (hFR2-14_H5) (FIG. 8). Among them, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H5.
SEQ ID NO: 11: nucleotide sequence of humanized FR2-14 heavy chain (hFR2-14_H8) Of these, nucleotide numbers 1 to 57 are sequences encoding a signal sequence.
SEQ ID NO: 12: amino acid sequence of humanized FR2-14 heavy chain (hFR2-14_H8) (FIG. 9). Of these, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H8.
SEQ ID NO: 13: nucleotide sequence of humanized FR2-14 heavy chain (hFR2-14_H11) Of these, nucleotide numbers 1 to 57 are sequences encoding a signal sequence.
SEQ ID NO: 14 amino acid sequence (10) of humanized FR2-14 heavy chain (hFR2-14_H11). Of these, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H11.
SEQ ID NO: 15: nucleotide sequence of humanized FR2-14 heavy chain (hFR2-14_H12) Of these, nucleotide numbers 1 to 57 are sequences encoding a signal sequence.
SEQ ID NO: 16 amino acid sequence of humanized FR2-14 heavy chain (hFR2-14_H12) (FIG. 11). Among them, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H12.
SEQ ID NO: 17: nucleotide sequence of humanized FR2-14 heavy chain (hFR2-14_H19) Of these, nucleotide numbers 1 to 57 are sequences encoding a signal sequence.
SEQ ID NO: 18: amino acid sequence of humanized FR2-14 heavy chain (hFR2-14_H19) (FIG. 12). Of these, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequence of most mature hFR2-14_H19.
SEQ ID NO: 19: base sequence of humanized FR2-14 heavy chain (hFR2-14_H9) Of these, nucleotide numbers 1 to 57 are sequences encoding a signal sequence.
SEQ ID NO: 20: amino acid sequence of humanized FR2-14 heavy chain (hFR2-14_H9) (FIG. 16). Among them, amino acid numbers 1 to 19 are signal sequences, and are usually not included in the amino acid sequences of most mature hFR2-14_H9.

Claims (57)

  1. 下記(i)乃至(ix)の特徴を有するモノクローナル抗体もしくはその機能断片、および、トポイソメラーゼ阻害剤、微小管阻害剤、プラチナ製剤、抗VEGFR2抗体及び5-フルオロウラシル系抗がん剤よりなる群から選択される一つ又は二つ以上の他剤の組合せ、又は、該抗体もしくはその機能断片、および、トポイソメラーゼ阻害剤、微小管阻害剤、プラチナ製剤、抗VEGFR2抗体及び5-フルオロウラシル系抗がん剤よりなる群から選択される一つ又は二つ以上の他剤を含む医薬組成物:
    (i)ヒト2型線維芽細胞増殖因子受容体(hFGFR2)IIIb及びIIIcに特異的に結合する;
    (ii)hFGFR2の免疫グロブリン様ドメイン2又は免疫グロブリン様ドメイン3に結合する;
    (iii)ヒト1型線維芽細胞増殖因子受容体(hFGFR1)、ヒト3型線維芽細胞増殖因子受容体(hFGFR3)及びヒト4型線維芽細胞増殖因子受容体(hFGFR4)には特異的に結合しない;
    (iv)抗体依存性細胞傷害活性を有する;
    (v)抗体依存性細胞媒介食活性を有する;
    (vi)hFGFR2に対する中和活性を有する;
    (vii)in vivo抗腫瘍活性を有する;
    (viii)hFGFR2へのFGFの結合を阻害する;
    (ix)キメラ抗体、ヒト化抗体又はヒト抗体である。
    Selected from the group consisting of monoclonal antibodies or functional fragments thereof having the following characteristics (i) to (ix), and topoisomerase inhibitors, microtubule inhibitors, platinum preparations, anti-VEGFR2 antibodies and 5-fluorouracil anticancer agents. A combination of one or two or more other agents, or the antibody or a functional fragment thereof, and a topoisomerase inhibitor, microtubule inhibitor, platinum preparation, anti-VEGFR2 antibody and 5-fluorouracil anticancer agent A pharmaceutical composition comprising one or more other agents selected from the group consisting of:
    (I) specifically binds to human type 2 fibroblast growth factor receptor (hFGFR2) IIIb and IIIc;
    (Ii) binds to immunoglobulin-like domain 2 or immunoglobulin-like domain 3 of hFGFR2;
    (Iii) Specific binding to human type 1 fibroblast growth factor receptor (hFGFR1), human type 3 fibroblast growth factor receptor (hFGFR3) and human type 4 fibroblast growth factor receptor (hFGFR4) do not do;
    (Iv) has antibody-dependent cytotoxic activity;
    (V) has antibody-dependent cell-mediated phagocytic activity;
    (Vi) has neutralizing activity against hFGFR2;
    (Vii) has in vivo anti-tumor activity;
    (Viii) inhibits the binding of FGF to hFGFR2;
    (Ix) A chimeric antibody, humanized antibody or human antibody.
  2.  該抗体が配列表の配列番号1(図1)に示されるアミノ酸配列からなるCDRL1、配列表の配列番号2(図2)に示されるアミノ酸配列からなるCDRL2及び配列表の配列番号3(図3)に示されるアミノ酸配列からなるCDRL3を含むことからなる軽鎖、並びに、配列表の配列番号4(図4)に示されるアミノ酸配列からなるCDRH1、配列表の配列番号5(図5)に示されるアミノ酸配列からなるCDRH2及び配列表の配列番号6(図6)に示されるアミノ酸配列もしくは該アミノ酸配列のアミノ末端から数えて1つ目又は2つ目のアミノ酸が他のアミノ酸に置換されてなるアミノ酸配列からなるCDRH3を含むことからなる重鎖からなる、請求項1に記載の組合せ又は医薬組成物。 The antibody has CDRL1 consisting of the amino acid sequence shown in SEQ ID NO: 1 (FIG. 1), CDRL2 consisting of the amino acid sequence shown in SEQ ID NO: 2 (FIG. 2), and SEQ ID NO: 3 (FIG. 3). ), A light chain comprising CDRL3 consisting of the amino acid sequence shown in FIG. 4 and a CDRH1 consisting of the amino acid sequence shown in SEQ ID NO: 4 (FIG. 4) in the sequence listing, and SEQ ID NO: 5 (FIG. 5) in the sequence listing. CDRH2 comprising the amino acid sequence and the amino acid sequence shown in SEQ ID NO: 6 (FIG. 6) of the sequence listing or the first or second amino acid counted from the amino terminus of the amino acid sequence are substituted with other amino acids. The combination or pharmaceutical composition according to claim 1, comprising a heavy chain comprising CDRH3 comprising an amino acid sequence.
  3.  該抗体が下記(i)乃至(vi)から選択される、請求項1又は2に記載の組合せ又は医薬組成物:
    (i)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号は18(図12)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H19/L1);
    (ii)配列番号は8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号16(図11)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H12/L1);
    (iii)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号12(図9)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H8/L1);
    (iv)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号20(図16)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H9/L1);
    (v)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号14(図10)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H11/L1);
    (vi)配列番号8(図7)で示されるアミノ酸配列のアミノ酸番号21乃至235を含む軽鎖、および、配列番号10(図8)で示されるアミノ酸配列のアミノ酸番号20乃至467を含む重鎖、を含んでなるヒト化抗体(hFR2-14_H5/L1)。
    The combination or pharmaceutical composition according to claim 1 or 2, wherein the antibody is selected from the following (i) to (vi):
    (I) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7), and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 18 (FIG. 12). A humanized antibody comprising a chain (hFR2-14_H19 / L1);
    (Ii) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 16 (FIG. 11) A humanized antibody comprising a chain (hFR2-14_H12 / L1);
    (Iii) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 12 (FIG. 9) A humanized antibody comprising (hFR2-14_H8 / L1);
    (Iv) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 20 (FIG. 16) A humanized antibody comprising (hFR2-14_H9 / L1);
    (V) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 14 (FIG. 10) A humanized antibody comprising (hFR2-14_H11 / L1);
    (Vi) a light chain comprising amino acid numbers 21 to 235 of the amino acid sequence represented by SEQ ID NO: 8 (FIG. 7) and a heavy chain comprising amino acid numbers 20 to 467 of the amino acid sequence represented by SEQ ID NO: 10 (FIG. 8) A humanized antibody (hFR2-14_H5 / L1).
  4.  該抗体が請求項3の(i)乃至(vi)のいずれか一つに記載の抗体の重鎖および軽鎖のアミノ酸配列とそれぞれ95%以上同一なアミノ酸配列を含む重鎖および軽鎖を含んでなり、且つヒトFGFR2に結合する、請求項1に記載の組合せ又は医薬組成物。 The antibody comprises a heavy chain and a light chain each comprising 95% or more amino acid sequence identical to the amino acid sequence of the heavy chain and the light chain of the antibody according to any one of (i) to (vi) of claim 3 The combination or pharmaceutical composition according to claim 1, which binds to human FGFR2.
  5.  該抗体もしくはその機能断片が、請求項2又は3の(i)乃至(vi)のいずれか一つに記載の抗体が認識する抗原上の部位に結合する、請求項1に記載の組合せ又は医薬組成物。 The combination or pharmaceutical according to claim 1, wherein the antibody or a functional fragment thereof binds to a site on an antigen recognized by the antibody according to any one of (i) to (vi) of claim 2 or 3. Composition.
  6.  該抗体もしくはその機能断片が、請求項2又は3の(i)乃至(vi)のいずれか一つに記載の抗体と、hFGFR2への結合において競合する、請求項1に記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to claim 1, wherein the antibody or a functional fragment thereof competes with the antibody according to any one of (i) to (vi) of claim 2 or 3 in binding to hFGFR2. object.
  7.  下記の工程(i)及び(ii)を含むことからなる方法により得られる抗体もしくはその機能断片および他剤の組合せ又は該抗体もしくはその機能断片および他剤を含む医薬組成物:
    (i)下記(ア)又は(イ)に記載の細胞を培養する工程;
    (ア)下記(a)乃至(c)のいずれか一つに記載のヌクレオチドが挿入された組換えベクター又は該ヌクレオチドが導入された組換え細胞:
    (a)請求項1乃至6のいずれか一つに記載の抗体の重鎖または軽鎖の一部または全部のアミノ酸配列をコードする塩基配列を含んでなるヌクレオチド;
    (b)請求項1乃至6のいずれか一つに記載の抗体の重鎖または軽鎖の一部または全部のアミノ酸配列をコードする塩基配列を含む塩基配列からなるヌクレオチド;
    (c)請求項1乃至6のいずれか一つに記載の抗体の重鎖または軽鎖の一部または全部のアミノ酸配列をコードする塩基配列からなるヌクレオチド;
    (イ)請求項1乃至6のいずれか一つに記載の抗体又はその機能断片を産生する細胞、
    および
    (ii)前記工程(i)で得られた培養物から請求項1乃至6のいずれか一つに記載の抗体又はその機能断片を回収する工程。
    A combination of an antibody or a functional fragment thereof obtained by a method comprising the following steps (i) and (ii) and another agent, or a pharmaceutical composition comprising the antibody or a functional fragment thereof and the other agent:
    (I) a step of culturing the cell according to (a) or (b) below;
    (A) A recombinant vector into which the nucleotide according to any one of the following (a) to (c) is inserted or a recombinant cell into which the nucleotide has been introduced:
    (A) a nucleotide comprising a base sequence encoding part or all of the amino acid sequence of the heavy chain or light chain of the antibody according to any one of claims 1 to 6;
    (B) a nucleotide consisting of a base sequence comprising a base sequence encoding part or all of the amino acid sequence of the heavy chain or light chain of the antibody according to any one of claims 1 to 6;
    (C) a nucleotide consisting of a base sequence encoding part or all of the amino acid sequence of the heavy chain or light chain of the antibody according to any one of claims 1 to 6;
    (A) a cell producing the antibody or functional fragment thereof according to any one of claims 1 to 6,
    And (ii) recovering the antibody or functional fragment thereof according to any one of claims 1 to 6 from the culture obtained in the step (i).
  8.  該抗体の重鎖または軽鎖のアミノ末端またはカルボキシル末端において1乃至5個のアミノ酸が欠失してなる、請求項1乃至7のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 7, wherein 1 to 5 amino acids are deleted from the amino terminus or carboxyl terminus of the heavy or light chain of the antibody.
  9.  請求項1乃至8のいずれか一つに記載の抗体もしくはその機能断片の糖鎖修飾体および他剤の組合せ又は該修飾体および他剤を含む医薬組成物。 A combination of a modified sugar chain of the antibody or functional fragment thereof according to any one of claims 1 to 8 and another agent, or a pharmaceutical composition comprising the modified agent and the other agent.
  10.  癌の治療もしくは予防に使用される、請求項1乃至9のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 9, which is used for treatment or prevention of cancer.
  11.  癌がhFGFR2陽性である、請求項10に記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to claim 10, wherein the cancer is hFGFR2 positive.
  12.  該抗体もしくはその機能断片がさらなる化合物とコンジュゲートされた、請求項1乃至11のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 11, wherein the antibody or functional fragment thereof is conjugated with a further compound.
  13.  他剤がトポイソメラーゼ阻害剤である、請求項1乃至12のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 12, wherein the other agent is a topoisomerase inhibitor.
  14.  トポイソメラーゼ阻害剤と併用するための、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物。 The antibody or functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or the modified substance contained in the combination or composition according to claim 9, for use in combination with a topoisomerase inhibitor. A pharmaceutical composition comprising
  15.  トポイソメラーゼ阻害剤を含み、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物。 It contains a topoisomerase inhibitor and is used in combination with an antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified form contained in the combination or composition according to claim 9. The pharmaceutical composition which raises the effect | action of this antibody, this functional fragment, or this modification.
  16.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含み、トポイソメラーゼ阻害剤と併用することにより、該トポイソメラーゼ阻害剤の作用を上昇させる医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified form contained in the combination or composition according to claim 9, and used in combination with a topoisomerase inhibitor. The pharmaceutical composition which raises the effect | action of this topoisomerase inhibitor by this.
  17.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が組み合わせて投与されることを特徴とする医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a topoisomerase inhibitor. A pharmaceutical composition to be administered.
  18.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、請求項17に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a topoisomerase inhibitor, 18. A pharmaceutical composition according to claim 17, characterized in that it is contained as an active ingredient in another formulation and administered at the same time or at different times.
  19.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が、単一の製剤に有効成分として含有されることを特徴とする、請求項17に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance and a topoisomerase inhibitor contained in the combination or composition according to claim 9 The pharmaceutical composition according to claim 17, which is contained as an active ingredient in one preparation.
  20.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、トポイソメラーゼ阻害剤が組み合わせて投与されることを特徴とする癌の治療方法。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a topoisomerase inhibitor. A method for treating cancer, characterized by being administered.
  21.  トポイソメラーゼ阻害剤がイリノテカン、トポテカン及びエトポシドよりなる群から選択される一つ又は二つ以上である、請求項13乃至19のいずれか一つに記載の医薬組成物又は請求項20に記載の治療方法。 The pharmaceutical composition according to any one of claims 13 to 19 or the treatment method according to claim 20, wherein the topoisomerase inhibitor is one or more selected from the group consisting of irinotecan, topotecan and etoposide. .
  22.  他剤が微小管阻害剤である、請求項1乃至12のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 12, wherein the other agent is a microtubule inhibitor.
  23.  微小管阻害剤と併用するための、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物。 The antibody or functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8 or the modification contained in the combination or composition according to claim 9 for use in combination with a microtubule inhibitor A pharmaceutical composition comprising a body.
  24.  微小管阻害剤を含み、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物。 A microtubule inhibitor, which is used in combination with an antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified form contained in the combination or composition according to claim 9. By doing so, the pharmaceutical composition which raises the effect | action of this antibody, this functional fragment, or this modified body.
  25.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含み、微小管阻害剤と併用することにより、該微小管阻害剤の作用を上昇させる医薬組成物。 Use in combination with a microtubule inhibitor, comprising the antibody or functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or the modified form contained in the combination or composition according to claim 9. To increase the action of the microtubule inhibitor.
  26.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が組み合わせて投与されることを特徴とする医薬組成物。 A combination of an antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a microtubule inhibitor. The pharmaceutical composition characterized by being administered.
  27.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、請求項26に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a microtubule inhibitor, 27. The pharmaceutical composition according to claim 26, which is contained as an active ingredient in different preparations and administered at the same time or at different times.
  28.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が、単一の製剤に有効成分として含有されることを特徴とする、請求項26に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a microtubule inhibitor, 27. The pharmaceutical composition according to claim 26, which is contained as an active ingredient in a single preparation.
  29.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、微小管阻害剤が組み合わせて投与されることを特徴とする癌の治療方法。 A combination of an antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a microtubule inhibitor. A method for treating cancer, characterized by being administered.
  30.  微小管阻害剤がパクリタキセル、ドセタキセル、ビンクリスチン、ビンブラスチン、エリブリン、ビンデシン及びビノレルビンよりなる群から選択される一つ又は二つ以上である、請求項22乃至28のいずれか一つに記載の医薬組成物又は請求項29に記載の治療方法。 The pharmaceutical composition according to any one of claims 22 to 28, wherein the microtubule inhibitor is one or more selected from the group consisting of paclitaxel, docetaxel, vincristine, vinblastine, eribulin, vindesine and vinorelbine. Or the treatment method of Claim 29.
  31.  他剤がプラチナ製剤である、請求項1乃至12のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 12, wherein the other agent is a platinum preparation.
  32.  プラチナ製剤と併用するための、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8 or a modified form contained in the combination or composition according to claim 9 for use in combination with a platinum preparation. A pharmaceutical composition comprising.
  33.  プラチナ製剤を含み、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物。 A platinum preparation is used, and is used in combination with an antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified form contained in the combination or composition according to claim 9. To increase the action of the antibody, the functional fragment or the modified product.
  34.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含み、プラチナ製剤と併用することにより、該プラチナ製剤の作用を上昇させる医薬組成物。 The antibody or functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or the functional fragment thereof, or the modified substance contained in the combination or composition according to claim 9, and used in combination with a platinum preparation To increase the action of the platinum preparation.
  35.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が組み合わせて投与されることを特徴とする医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8 or a functional fragment thereof, or a modification contained in the combination or composition according to claim 9, and a platinum preparation are administered in combination. The pharmaceutical composition characterized by the above-mentioned.
  36.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、請求項35に記載の医薬組成物。 The antibody or the functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or the modified substance contained in the combination or composition according to claim 9, and the platinum preparation, 36. The pharmaceutical composition according to claim 35, which is contained as an active ingredient in the preparation and administered at the same time or at different times.
  37.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が、単一の製剤に有効成分として含有されることを特徴とする、請求項35に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a platinum preparation, 36. The pharmaceutical composition according to claim 35, which is contained as an active ingredient in the preparation.
  38.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、プラチナ製剤が組み合わせて投与されることを特徴とする癌の治療方法。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8 or a functional fragment thereof, or a modification contained in the combination or composition according to claim 9, and a platinum preparation are administered in combination. A method for treating cancer, comprising:
  39.  プラチナ製剤がシスプラチン、オキサリプラチン、カルボプラチン及びネダプラチンよりなる群から選択される一つ又は二つ以上である、請求項31乃至37のいずれか一つに記載の医薬組成物又は請求項38に記載の治療方法。 The pharmaceutical composition according to any one of claims 31 to 37 or the claim 38, wherein the platinum preparation is one or more selected from the group consisting of cisplatin, oxaliplatin, carboplatin and nedaplatin. Method of treatment.
  40.  他剤が抗VEGFR2抗体である、請求項1乃至12のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 12, wherein the other agent is an anti-VEGFR2 antibody.
  41.  抗VEGFR2抗体と併用するための、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物。 The antibody or the functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8 or the modified substance contained in the combination or composition according to claim 9 for use in combination with an anti-VEGFR2 antibody A pharmaceutical composition comprising
  42.  抗VEGFR2抗体を含み、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物。 An anti-VEGFR2 antibody is included and used in combination with an antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modification contained in the combination or composition according to claim 9. The pharmaceutical composition which raises the effect | action of this antibody, this functional fragment, or this modification.
  43.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含み、抗VEGFR2抗体と併用することにより、該抗VEGFR2抗体の作用を上昇させる医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and used in combination with an anti-VEGFR2 antibody Thereby increasing the action of the anti-VEGFR2 antibody.
  44.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が組み合わせて投与されることを特徴とする医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and an anti-VEGFR2 antibody in combination. A pharmaceutical composition to be administered.
  45.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、請求項44に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and an anti-VEGFR2 antibody, 45. Pharmaceutical composition according to claim 44, characterized in that it is contained as an active ingredient in another formulation and administered at the same time or at different times.
  46.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が、単一の製剤に有効成分として含有されることを特徴とする、請求項44に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9 and an anti-VEGFR2 antibody are The pharmaceutical composition according to claim 44, which is contained as an active ingredient in one preparation.
  47.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、抗VEGFR2抗体が組み合わせて投与されることを特徴とする癌の治療方法。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and an anti-VEGFR2 antibody in combination. A method for treating cancer, characterized by being administered.
  48.  抗VEGFR2抗体がラムシルマブである、請求項40乃至46のいずれか一つに記載の医薬組成物又は請求項47に記載の治療方法。 48. The pharmaceutical composition according to any one of claims 40 to 46 or the treatment method according to claim 47, wherein the anti-VEGFR2 antibody is ramcilmab.
  49.  他剤が5-フルオロウラシル系抗がん剤である、請求項1乃至12のいずれか一つに記載の組合せ又は医薬組成物。 The combination or pharmaceutical composition according to any one of claims 1 to 12, wherein the other agent is a 5-fluorouracil anticancer agent.
  50.  5-フルオロウラシル系抗がん剤と併用するための、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含む医薬組成物。 The antibody or functional fragment thereof, or the combination or composition according to claim 9, which is included in the combination or composition according to any one of claims 1 to 8, for use in combination with a 5-fluorouracil anticancer agent. A pharmaceutical composition comprising the modified product.
  51.  5-フルオロウラシル系抗がん剤を含み、請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体と併用することにより、該抗体、該機能断片又は該修飾体の作用を上昇させる医薬組成物。 An antibody or a functional fragment thereof, or a combination or composition according to claim 9, comprising a 5-fluorouracil anticancer agent and contained in the combination or composition according to any one of claims 1 to 8. The pharmaceutical composition which raises the effect | action of this antibody, this functional fragment, or this modified body by using together with a modified body.
  52.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体を含み、5-フルオロウラシル系抗がん剤と併用することにより、該5-フルオロウラシル系抗がん剤の作用を上昇させる医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, wherein the 5-fluorouracil anti-antibodies are A pharmaceutical composition that increases the action of the 5-fluorouracil-based anticancer agent when used in combination with a cancer drug.
  53.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が組み合わせて投与されることを特徴とする医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a 5-fluorouracil anti-antibody. A pharmaceutical composition comprising a combination of cancer drugs.
  54.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が、それぞれ別の製剤に有効成分として含有され、同時にまたは異なる時間に投与されることを特徴とする、請求項53に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a 5-fluorouracil anti-antibody. 54. The pharmaceutical composition according to claim 53, wherein the cancer agent is contained as an active ingredient in different preparations, and is administered at the same time or at different times.
  55.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が、単一の製剤に有効成分として含有されることを特徴とする、請求項53に記載の医薬組成物。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a 5-fluorouracil anti-antibody. The pharmaceutical composition according to claim 53, wherein the cancer drug is contained as an active ingredient in a single preparation.
  56.  請求項1乃至8のいずれか一つに記載の組合せ又は組成物に含まれる抗体もしくはその機能断片又は請求項9に記載の組合せ又は組成物に含まれる修飾体、および、5-フルオロウラシル系抗がん剤が組み合わせて投与されることを特徴とする癌の治療方法。 An antibody or a functional fragment thereof contained in the combination or composition according to any one of claims 1 to 8, or a modified substance contained in the combination or composition according to claim 9, and a 5-fluorouracil anti-antibody. A method for treating cancer, comprising administering a cancer drug in combination.
  57.  5-フルオロウラシル系抗がん剤がフルオロウラシル、テガフール、テガフール・ギメラシル・オテラシルカリウム、テガフール・ウラシル、カペシタビン、カルモフール、ドキシフルリジンである、請求項49乃至55のいずれか一つに記載の医薬組成物又は請求項56に記載の治療方法。 The pharmaceutical composition according to any one of claims 49 to 55, wherein the 5-fluorouracil anticancer agent is fluorouracil, tegafur, tegafur, gimeracil, oteracil potassium, tegafur uracil, capecitabine, carmofur, doxyfluridine 57. A treatment method according to claim 56.
PCT/JP2016/061333 2015-04-08 2016-04-07 Composition comprising anti-fgfr2 antibody and another agent WO2016163433A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2017511041A JP6887944B2 (en) 2015-04-08 2016-04-07 Composition containing anti-FGFR2 antibody and other agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2015079468 2015-04-08
JP2015-079468 2015-04-08

Publications (1)

Publication Number Publication Date
WO2016163433A1 true WO2016163433A1 (en) 2016-10-13

Family

ID=57072444

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2016/061333 WO2016163433A1 (en) 2015-04-08 2016-04-07 Composition comprising anti-fgfr2 antibody and another agent

Country Status (3)

Country Link
JP (1) JP6887944B2 (en)
TW (1) TW201642904A (en)
WO (1) WO2016163433A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023164662A1 (en) * 2022-02-25 2023-08-31 Amgen Inc. Treatment of squamous non small cell lung cancer

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012508184A (en) * 2008-11-07 2012-04-05 ギャラクシー バイオテック, エルエルシー Monoclonal antibody against fibroblast growth factor receptor 2
JP2013529076A (en) * 2010-05-11 2013-07-18 アベオ ファーマシューティカルズ, インコーポレイテッド Anti-FGFR2 antibody
WO2013154206A1 (en) * 2012-04-09 2013-10-17 第一三共株式会社 Anti-fgfr2 antibody
JP2015504434A (en) * 2011-11-23 2015-02-12 バイエル・インテレクチュアル・プロパティ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツングBayer Intellectual Property GmbH Anti-FGFR2 antibody and use thereof
WO2015053407A1 (en) * 2013-10-08 2015-04-16 第一三共株式会社 Combination of anti-fgfr2 antibody and other agent

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012508184A (en) * 2008-11-07 2012-04-05 ギャラクシー バイオテック, エルエルシー Monoclonal antibody against fibroblast growth factor receptor 2
JP2013529076A (en) * 2010-05-11 2013-07-18 アベオ ファーマシューティカルズ, インコーポレイテッド Anti-FGFR2 antibody
JP2015504434A (en) * 2011-11-23 2015-02-12 バイエル・インテレクチュアル・プロパティ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツングBayer Intellectual Property GmbH Anti-FGFR2 antibody and use thereof
WO2013154206A1 (en) * 2012-04-09 2013-10-17 第一三共株式会社 Anti-fgfr2 antibody
WO2015053407A1 (en) * 2013-10-08 2015-04-16 第一三共株式会社 Combination of anti-fgfr2 antibody and other agent

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CLARKE J.M. ET AL.: "Targeted inhibition of VEGF Receptor-2:An update on Ramucirumab", EXPERT OPIN.BIOL.THER., vol. 13, no. 8, 2013, pages 1187 - 1196, XP055320040 *
DAMIA G. ET AL.: "Striking activity of E-3810 combined with paclitaxel and 5FU in the triple negative breast cancer model MDA-MB-231.", CANCER RESEARCH, vol. 71, no. 8, 2011 *
IVAN M. ET AL.: "Blockade of FGF signaling: therapeutic promise for ovarian cancer.", CANCER BIOLOGY & THERAPY, vol. 10, no. 5, 2010, pages 505 - 508, XP055320037 *
MATSUDA Y. ET AL.: "Fibroblast growth factor receptor-2 IIIc as a novel molecular target in colorectal cancer.", CURRENT COLORECTAL CANCER REPORTS, vol. 10, no. 1, 2014, pages 20 - 26, XP055202148 *
ZHANG J. ET AL.: "Activity of AZD4547, a potent and selective FGF-receptor inhibitor, alone and in combination with chemotherapy in standard xenograft and human primary explant models of FGF-receptor 2 amplified gastric cancer.", CANCER RESEARCH, vol. 72, no. 8, 2012 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023164662A1 (en) * 2022-02-25 2023-08-31 Amgen Inc. Treatment of squamous non small cell lung cancer

Also Published As

Publication number Publication date
TW201642904A (en) 2016-12-16
JP6887944B2 (en) 2021-06-16
JPWO2016163433A1 (en) 2018-02-22

Similar Documents

Publication Publication Date Title
JP7118117B2 (en) Selective method for producing antibody-drug conjugates
EP2844675B1 (en) Humanized pan-her antibody compositions
JP6124377B2 (en) New anti-DR5 antibody
JP6967853B2 (en) Antibodies that bind to ErbB-2 and ErbB-3
EP2635604B1 (en) Pan-her antibody composition
JP6039428B2 (en) Anti-HER2 antibodies and compositions
CA2813796C (en) Erbb3 binding antibody
Ko et al. Combination of novel HER2-targeting antibody 1E11 with trastuzumab shows synergistic antitumor activity in HER2-positive gastric cancer
JP6509735B2 (en) Combination of anti-FGFR2 antibody and other agents
WO2021115303A1 (en) Anti-claudin18.2 monoclonal antibody, preparation method therefor and use thereof
KR20220152318A (en) Medicines for the treatment and/or prevention of cancer
CA3058341A1 (en) Antibodies for the treatment of erbb-2/erbb-3 positive tumors
CN114181310B (en) anti-TIGIT antibody, and pharmaceutical composition and use thereof
JP6887944B2 (en) Composition containing anti-FGFR2 antibody and other agents
CN114616245A (en) anti-CD 38 antibody and application thereof
CN111032081A (en) ErbB-2 and ErbB-3 targeting agents and bispecific antibodies
JP2016182135A (en) Anti-her2 antibodies and compositions
CA3202006A1 (en) Treatment of cancers with an antibody that binds lgr5 and egfr
JP2024515879A (en) Anti-SIGLEC Compositions and Uses Thereof
JP2023523006A (en) Cancer treatment with antibodies that bind to LGR5 and EGFR
JP2024516230A (en) Therapeutic and diagnostic methods and compositions for cancer
JP2020507338A (en) Monoclonal and humanized antibodies to cancer glycopeptides

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16776599

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017511041

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16776599

Country of ref document: EP

Kind code of ref document: A1