TW201019961A - Combination therapy - Google Patents

Combination therapy Download PDF

Info

Publication number
TW201019961A
TW201019961A TW098134878A TW98134878A TW201019961A TW 201019961 A TW201019961 A TW 201019961A TW 098134878 A TW098134878 A TW 098134878A TW 98134878 A TW98134878 A TW 98134878A TW 201019961 A TW201019961 A TW 201019961A
Authority
TW
Taiwan
Prior art keywords
amine
ethynylphenyl
phenyl
antibody
ethoxy
Prior art date
Application number
TW098134878A
Other languages
Chinese (zh)
Inventor
Ellen Filvaroff
Mark Merchant
Original Assignee
Genentech Inc
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 Genentech Inc filed Critical Genentech Inc
Publication of TW201019961A publication Critical patent/TW201019961A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Abstract

The present invention relates generally to the fields of molecular biology and growth factor regulation. More specifically, the invention relates to combination therapies for the treatment of pathological conditions, such as cancer.

Description

201019961 六、發明說明: 【發明所屬之技術領域】 本發明大體而言係關於分子生物學及生長因子調控領 域。更具體言之,本發明係關於治療諸如癌症之病理性病 狀之組合療法。 * 本申請案根據35 USC 119(e)主張2008年10月17曰申請之 美國臨時專利申請案第61/106,513號之優先權,該申請案 之内容係以引用之方式併入本文中。 ❹ 【先前技術】 HGF為由間充質獲得之多效性因子,其對若干不同細胞 類型具有細胞分裂(mitogenic)、細胞移動(motogenic)及形 態發生(morphogenic)活性。HGF效應係經由特異性酷胺酸 激酶c-met介導,且時常在多種腫瘤中觀察到異常HGF及c- met表現。參見例如Maulik等人,Cytokine & Growth Factor201019961 VI. Description of the Invention: TECHNICAL FIELD OF THE INVENTION The present invention relates generally to the field of molecular biology and growth factor regulation. More specifically, the present invention relates to a combination therapy for treating a pathological condition such as cancer. * This application claims priority to U.S. Provisional Patent Application Serial No. 61/106,513, filed on Jan. 27, 2008, the entire disclosure of which is hereby incorporated by reference. ❹ [Prior Art] HGF is a pleiotropic factor obtained from mesenchyme, which has mitogenic, motogenic, and morphogenic activities against several different cell types. The HGF effector is mediated via the specific valine kinase c-met, and abnormal HGF and c-met expression is often observed in a variety of tumors. See, for example, Maulik et al., Cytokine & Growth Factor

Reviews (2002), 13 :41-59 ; Danilkovitch-Miagkova及 Zbar, J. Clin. Invest. (2002),109(7):863-867。腫瘤進展及轉移 ❹. 涉及HGF/c-Met信號傳導路徑之調控。參見例如Trusolino 及 Comoglio,Nature Rev. (2002), 2:289-300 ° HGF結合Met受體酪胺酸激酶(RTK)之細胞外域且調控各 種生物過程,諸如細胞分散、增殖及存活。HGF-Met信號 傳導對正常胚胎發育(尤其肌肉祖細胞遷移及肝及神經系 統發育)而言必不可少(Bladt等人,Nature (1995),376, 768-771 ; Hamanoue等人,Faseb J (2000),14,399-406 ; Maina等人,Cell (1996),87,531-542 ; Schmidt等人, 143939.doc 201019961Reviews (2002), 13:41-59; Danilkovitch-Miagkova and Zbar, J. Clin. Invest. (2002), 109(7): 863-867. Tumor progression and metastasis ❹. Involves the regulation of the HGF/c-Met signaling pathway. See, for example, Trusolino and Comoglio, Nature Rev. (2002), 2: 289-300 ° HGF binds to the extracellular domain of Met receptor tyrosine kinase (RTK) and regulates various biological processes such as cell dispersion, proliferation and survival. HGF-Met signaling is essential for normal embryonic development (especially muscle progenitor migration and liver and nervous system development) (Bladt et al, Nature (1995), 376, 768-771; Hamanoue et al., Faseb J ( 2000), 14, 399-406; Maina et al., Cell (1996), 87, 531-542; Schmidt et al., 143939.doc 201019961

Nature (1995),373, 699-702 ; Uehara等人,Nature (1995), 373, 702-705)。Met及HGF基因剔除小鼠之發育表型極類 似,表明HGF為Met受體之同源配位體(Schmidt等人, 1995,同上;Uehara等人,1995,同上)。HGF-Met亦在肝 再生、血管生成及傷口癒合中發揮作用(Bussolino等人,J Cell Biol (1992),119, 629-641 ; Matsumoto 及 Nakamura, Exs (1993),65,225-249 ; Nusrat等人,J Clin Invest (1994) 93, 2056-2065)。前驅體Met受體經歷蛋白水解分裂並形成 經由雙硫鍵連接之細胞外α亞單位及跨膜β亞單位(Tempest 等人,Br J Cancer (1988),58,3-7)。β亞單位含有細胞質 激酶域且在C端具有多受質對接位點(multi-substrate docking site),銜接蛋白(adapter protein)在該位點處結合 且開始信號傳導(Bardelli 等人,Oncogene (1997),15, 3103-3111 ; Nguyen等人,J Biol Chem (1997), 272, 20811-20819 ; Pelicci等人,Oncogene (1995),10, 1631-1638 ; Ponzetto等人,Cell (1994),77,261-271 ; Weidner等人, Nature (1996), 384,173-176)。HGF結合後,Met之活化分 別經由Gabl及Grb2/Sos介導之PI3激酶及Ras/MAPK活化引 起酪胺酸磷酸化及下游信號傳導,從而驅動細胞運動及增殖 (Furge等人,Oncogene (2000), 19, 5582-5589 ; Hartmann 等人,J Biol Chem (1994),269,21936-21939 ; Ponzetto 等 人,J Biol Chem (1996),271,14119-14123 ; Royal及 Park, J Biol Chem (1995), 270, 27780-27787) 〇 已顯示Met在致癌物處理之骨肉瘤細胞株中轉型(Cooper 143939.doc 201019961 等人,Nature (1984),311,29-33 ; Park等人,Cell (1986), 45,895-904)。在多種人類癌症中觀察到Met過度表現或基 因擴增。舉例而言,Met蛋白在結腸直腸癌中過度表現至 少5倍,且據報導在肝轉移中經基因擴增(Di Renzo等人, Clin Cancer Res (1995), 1,147-154 ; Liu等人,Oncogene (1992),7,181-185)。亦報導Met蛋白在口腔鱗狀細胞癌、 肝細胞癌、腎細胞癌、乳房癌及肺癌中過度表現(Jin等 人,Cancer (1997),79,749-760 ; Morello 等人,J Cell Physiol (2001), 189,285-290 ; Natali 等人,Int J Cancer (1996),69,212-217 ; Olivero等人,Br J Cancer (1996), 74, 1862-1868 ; Suzuki等人,Br J Cancer (1996),74,1862-1868)。另外,已在肝細胞癌、胃癌及結腸直腸癌中觀察 到 mRNA過度表現(Boix等人,Hepatology (1994),19, 88-91 ; Kuniyasu等人,Int J Cancer (1993), 55, 72-75 ; Liu等 人,Oncogene (1992),7, 181-185)。 已在腎乳頭狀癌中發現Met之激酶域發生若干引起組成 性受體活化之突變(Olivero等人,Int J Cancer (1999),82, 640-643 ; Schmidt 等人,Nat Genet (1997), 16, 68-73 ; Schmidt等人,Oncogene (1999), 18,2343-2350)。此等活 化突變使得組成性Met酪胺酸磷酸化且引起MAPK活化、 病灶形成(focus formation)及腫瘤形成(Jeffers等人,Proc Natl Acad Sci U S A (1997),94, 11445-11450)。另外,此 等突變增強細胞運動及侵襲(Giordano等人,Faseb J (2000),14, 399-406 ; Lorenzato等人,Cancer Res (2002), 143939.doc 201019961 62,7025-703 0)。轉型細胞中之HGF依賴性Met活化介導運 動、分散及遷移增強,最終導致侵襲性腫瘤生長及轉移 (Jeffers 等人,Mol Cell Biol (1996), 16,1115-1125 ; Meiners等人,Oncogene (1998),16,9-20)。 已顯示Met與驅動受體活化、轉型及侵襲之其他蛋白質 相互作用。在贅生性細胞中,據報導Met與細胞外基質 (ECM)組件(諸如層黏連蛋白)之受體α6β4整合素相互作 用,促進HGF依賴性侵襲性生長(Trusolino等人,Cell (2001) ,107,643-654)。另外,已顯示Met之細胞外域與信 號蛋白(semaphorin)家族之成員叢蛋白Bl(plexin B1)相互 作用且增強侵襲性生長(Giordano等人,Nat Cell Biol (2002) , 4, 720-724)。此外,亦報導腫瘤形成及轉移中涉及 之CD44v6與Met及HGF形成複合物且引起Met受體活化 (Orian-Rousseau等人,Genes Dev (2002), 16,3074-3086)。Nature (1995), 373, 699-702; Uehara et al, Nature (1995), 373, 702-705). The developmental phenotype of Met and HGF knockout mice is very similar, indicating that HGF is a cognate ligand for the Met receptor (Schmidt et al, 1995, supra; Uehara et al, 1995, supra). HGF-Met also plays a role in liver regeneration, angiogenesis and wound healing (Bussolino et al, J Cell Biol (1992), 119, 629-641; Matsumoto and Nakamura, Exs (1993), 65, 225-249; Nusrat Et al, J Clin Invest (1994) 93, 2056-2065). The precursor Met receptor undergoes proteolytic cleavage and forms an extracellular alpha subunit and a transmembrane beta subunit linked by a disulfide bond (Tempest et al, Br J Cancer (1988), 58, 3-7). The beta subunit contains a cytoplasmic kinase domain and has a multi-substrate docking site at the C-terminus, where the adaptor protein binds and initiates signaling (Bardelli et al., Oncogene (1997) ), 15, 3103-3111; Nguyen et al, J Biol Chem (1997), 272, 20811-20819; Pelicci et al, Oncogene (1995), 10, 1631-1638; Ponzetto et al, Cell (1994), 77 , 261-271; Weidner et al, Nature (1996), 384, 173-176). After HGF binding, activation of Met causes tyrosine phosphorylation and downstream signaling via Gabl and Grb2/Sos-mediated activation of PI3 kinase and Ras/MAPK, respectively, thereby driving cell movement and proliferation (Furge et al., Oncogene (2000) , 19, 5582-5589; Hartmann et al, J Biol Chem (1994), 269, 21936-21939; Ponzetto et al, J Biol Chem (1996), 271, 14119-14123; Royal and Park, J Biol Chem (1995) ), 270, 27780-27787) 〇 has been shown to transform Met in carcinogen-treated osteosarcoma cell lines (Cooper 143939.doc 201019961 et al, Nature (1984), 311, 29-33; Park et al, Cell (1986) ), 45, 895-904). Met overexpression or gene amplification was observed in a variety of human cancers. For example, Met proteins are at least 5-fold overexpressed in colorectal cancer and have been reported to be genetically amplified in liver metastases (Di Renzo et al, Clin Cancer Res (1995), 1, 147-154; Liu et al. , Oncogene (1992), 7, 181-185). Met protein has also been reported to be overexpressed in oral squamous cell carcinoma, hepatocellular carcinoma, renal cell carcinoma, breast cancer, and lung cancer (Jin et al, Cancer (1997), 79, 749-760; Morello et al, J Cell Physiol ( 2001), 189, 285-290; Natali et al, Int J Cancer (1996), 69, 212-217; Olivero et al, Br J Cancer (1996), 74, 1862-1868; Suzuki et al, Br J Cancer (1996), 74, 1862-1868). In addition, excessive mRNA expression has been observed in hepatocellular carcinoma, gastric cancer, and colorectal cancer (Boix et al, Hepatology (1994), 19, 88-91; Kuniyasu et al, Int J Cancer (1993), 55, 72- 75; Liu et al., Oncogene (1992), 7, 181-185). Several mutations that cause constitutive receptor activation have been found in the kinase domain of Met in renal papillary carcinoma (Olivero et al, Int J Cancer (1999), 82, 640-643; Schmidt et al, Nat Genet (1997), 16, 68-73; Schmidt et al., Oncogene (1999), 18, 2343-2350). Such viable mutations phosphorylate constitutive Met tyrosine and cause MAPK activation, focus formation, and tumor formation (Jeffers et al, Proc Natl Acad Sci U S A (1997), 94, 11445-11450). In addition, such mutations enhance cell movement and invasion (Giordano et al, Faseb J (2000), 14, 399-406; Lorenzato et al, Cancer Res (2002), 143939. doc 201019961 62, 7025-703 0). HGF-dependent Met activation in transformed cells mediates increased movement, dispersion, and migration, ultimately leading to invasive tumor growth and metastasis (Jeffers et al, Mol Cell Biol (1996), 16, 1115-1125; Meiners et al, Oncogene ( 1998), 16, 9-20). Met has been shown to interact with other proteins that drive receptor activation, transformation, and invasion. In neoplastic cells, it has been reported that Met interacts with the receptor α6β4 integrin of the extracellular matrix (ECM) component (such as laminin) to promote HGF-dependent invasive growth (Trusolino et al., Cell (2001), 107, 643-654). In addition, the extracellular domain of Met has been shown to interact with plexin B1, a member of the semaphorin family, and enhance invasive growth (Giordano et al, Nat Cell Biol (2002), 4, 720-724). In addition, it has also been reported that CD44v6 involved in tumor formation and metastasis forms a complex with Met and HGF and causes Met receptor activation (Orian-Rousseau et al., Genes Dev (2002), 16, 3074-3086).

Met為受體酪胺酸激酶(RTK)(包括Ron及Sea)之子族之成 員(Maulik等人,Cytokine Growth Factor Rev (2002), 13, 41-59)。對Met之細胞外域結構之預測表明其與信號蛋白 及叢蛋白共享同源性。Met之N端含有具有約500個胺基酸 之Sema域,該域在所有信號蛋白及叢蛋白中均為保守的。 信號蛋白及叢蛋白屬於首次關於在神經發育中之作用進行 描述的分泌型及膜結合型蛋白質之大家族(Van Vactor及 Lorenz,Curr Bio (1999),19,R201-204)。然而,新近已使 信號蛋白過度表現與腫瘤侵襲及轉移相關聯。叢蛋白、信 號蛋白及整合素中可見之富集半胱胺酸之PSI域(亦稱為 143939.doc 201019961Met is a member of the subfamily of receptor tyrosine kinase (RTK), including Ron and Sea (Maulik et al., Cytokine Growth Factor Rev (2002), 13, 41-59). Prediction of the extracellular domain structure of Met indicates that it shares homology with signaling proteins and plexifins. The N-terminus of Met contains a Sema domain of about 500 amino acids, which is conserved across all signaling proteins and plexifins. Signal proteins and plexins are a large family of secreted and membrane-bound proteins that were first described for their role in neurodevelopment (Van Vactor and Lorenz, Curr Bio (1999), 19, R201-204). However, recent overexpression of signaling proteins has been associated with tumor invasion and metastasis. The PSI domain of cysteine enriched in plexin, signal protein and integrin (also known as 143939.doc 201019961)

Met 相關序列域(Met Related Sequence domain))鄰接於 Sema域,其後繼之以四個IPT重複單元,該等重複單元為 叢蛋白及轉錄因子中可見之免疫球蛋白樣區域。最近研究 表明Met Sema域足以滿足HGF及肝素結合(Gherardi等人, Proc Natl Acad Sci U S A (2003),100(21):12039-44)。 如上文所說明,Met受艎酪胺酸激酶由其同源配位體 HGF活化,且受體峨酸化活化MAPK、PI-3激酶及PLC-γ之 下游路徑(L. Trusolino及 Ρ· M. Comoglio,Nat Rev Cancer 2, 289 (2002) ; C. Birchmeier等人,Nat Rev Mol Cell Biol 4, 915 (2003))。激酶域内之Y1234/Y1235之磷酸化對Met激酶 活化至關重要,而多受質對接位點中之Y1349及Y1356對 於src同源區-2(SH2)結合、磷酸酪胺酸結合(PTB)及Met結 合域(MBD)蛋白(C. Ponzetto 等人,Cell 77, 261 (1994) ; K· Μ· Weidner等人,Nature 384,173 (1996) ; G. Pelicci等人, Oncogene 10, 16;31 (1995))介導下游信號傳導路徑之活化較 為重要。已充分表徵另一近膜填酸化位點(juxtamembrane phosphorylation site) Y1003與Cbl E3連接酶之胳胺酸激酶 結合(TKB)域的結合(P. Peschard 等人,Mol Cell 8, 995 (2001) ; P. Peschard, N. Ishiyama, T. Lin, S. Lipkowitz, M. Park, J Biol Chem 279,29565 (2004))。據報導 Cbl 結合驅 動内吞蛋白(endophilin)介導之受體内飲作用、泛素化及後 續受體降解(A. Petrem等人,Nature 416,187 (2002))。此 受體下調機制先前已在亦具有類似Cbl結合位點之EGFR家 族中進行描述(K. Shtiegman, Y. Yarden,Semin Cancer Biol 143939.doc 201019961 13,29 (2003) ; M. D. Marmor, Y. Yarden, Oncogene 23, 2057 (2004) ; P. Peschard, M. Park, Cancer Cell 3,519 (2003))。已報導多種腫瘤中存在Met及HGF之調控異常。 在數種癌症中觀察到配位體驅動之Met活化。在肺癌、乳 房癌及多發性骨髓瘤中觀察到升高之血清及瘤内HGF(J. M. Siegfried等人,Ann Thorac Surg 66,1915 (1998) ; Ρ· C. Ma等人,Anticancer Res 23,49 (2003) ; B. E. Elliott等 人,Can J Physiol Pharmacol 80, 91 (2002) ; C. Seidel 等 人,Med Oncol 15,145 (1998))。已在諸如結腸直腸癌、 肺癌、胃癌及腎癌之各種癌症中報導Met及/或HGF之過度 表現、Met擴增或突變,且認為其驅動非配位體依賴性受 體活化(C. Birchmeier等人,Nat Rev Mol Cell Biol 4,915 (2003) ; G. Maulik等人,Cytokine Growth Factor Rev 13, 41 (2002))。另外,肝小鼠模型中Met之誘導性過度表現會 引發肝細胞癌,表明受體過度表現驅動非配位體依賴性腫 瘤形成(R· Wang等人,J Cell Biol 153, 1023 (2001))。關於 家族性及偶發性腎乳頭狀癌(RPC)患者,報導暗示癌症中 涉及Met之最強有力的證據。Met激酶域中引起受體組成性 活化之突變在RPC中經鑑別為生殖系及體細胞突變(L. Schmidt等人,Nat Genet 16,68 (1997))。在轉瘦基因小鼠 模型中引入此等突變會引起腫瘤形成及轉移。(M. Jeffers 等人,Proc Natl Acad Sci U S A 94, 11445 (1997))。 表皮生長因子受體(EGFR)家族包含四種參與諸如分化及 增殖之細胞反應的密切相關之受體(HER1/EGFR、HER2、 143939.doc 201019961 HER3及HER4)。EGFR激酶或其配位體TGF-α之過度表現 時常與許多癌症相關,該等癌症包括乳房癌、肺癌、結腸 直腸癌、卵巢癌、腎細胞癌、膀胱癌、頭頸部癌、神經膠 母細胞瘤及星形細胞瘤,且咸信該過度表現促進此等腫瘤 之惡性生長。亦發現EGFR基因(EGFRvIII)中之特異性缺失 突變增加細胞腫瘤形成性。EGFR刺激之信號傳導路徑之 活化促進多個潛在促進癌症之過程(例如增殖、血管生 成、細胞運動及侵襲)、減少細胞凋亡及誘導抗藥性。增 加之HER1/EGFR表現時常與晚期疾病、轉移及不良預後相 關。舉例而言,在NSCLC及胃癌中,顯示增加之HER1/EGFR 表現與高轉移率、不良腫瘤分化及增加之腫瘤增殖相關。 在NSCLC及神經膠母細胞瘤中觀察到活化受體之内在蛋 白酪胺酸激酶活性及/或增加下游信號傳導之突變°然 而,突變作為主要機制在賦予EGF受體抑制劑(例如埃羅替 尼(erlotinib)(TARCEVA®)或吉非替尼(gefitinib))敏感性中 之作用引起爭議。已報導全長EGF受體之突變形式預測對 EGF受體酪胺酸激酶抑制劑吉非替尼之反應性(Paez,J. G. 等人,(2004) Science 304:1497-1500; Lynch,T. J·等人, (2004) Ν· Engl. J. Med. 3 50:2129-2139)。細胞培養研究已 顯示表現EGF受體之該等突變形式之細胞株(亦即H3255)對 EGF受體酪胺酸激酶抑制劑吉非替尼所致之生長抑制更敏 感,且需要更高濃度的吉非替尼來抑制表現野生型EGF受 體之腫瘤細胞株》此等觀察結果表明,雖然EGF受體之特 異性突變形式可能反映較高的對EGF受體抑制劑之敏感 143939.doc 201019961 性,但不鑑別完全無反應表型。 使用直接抑制EGFR之激酶活性之化合物及藉由阻斷 EGFR活化來降低EGFR激酶活性之抗體作為抗腫瘤劑的研 發為熱門研究領域(de Bono J.S.及Rowinsky,E.K. (2002) Trends in Mol. Medicine 8:S19-S26 ; Dancey,J.及The Met Related Sequence domain is adjacent to the Sema domain, which is followed by four IPT repeat units, which are immunoglobulin-like regions visible in the plexus and transcription factors. Recent studies have shown that the Met Sema domain is sufficient for HGF and heparin binding (Gherardi et al, Proc Natl Acad Sci U S A (2003), 100(21): 12039-44). As explained above, Met-induced tyrosine kinase is activated by its cognate ligand HGF, and receptor citrate activates the downstream pathways of MAPK, PI-3 kinase and PLC-γ (L. Trusolino and Ρ·M. Comoglio, Nat Rev Cancer 2, 289 (2002); C. Birchmeier et al, Nat Rev Mol Cell Biol 4, 915 (2003)). Phosphorylation of Y1234/Y1235 in the kinase domain is critical for Met kinase activation, whereas Y1349 and Y1356 in the multi-substrate docking site for src homology-2 (SH2) binding, phosphotyrosine binding (PTB) and Met binding domain (MBD) protein (C. Ponzetto et al, Cell 77, 261 (1994); K. Μ Weidner et al, Nature 384, 173 (1996); G. Pelicci et al, Oncogene 10, 16; (1995)) It is important to mediate the activation of downstream signaling pathways. The binding of another juxtamembrane phosphorylation site, Y1003, to the lysine kinase binding (TKB) domain of Cbl E3 ligase has been well characterized (P. Peschard et al, Mol Cell 8, 995 (2001); P. Peschard, N. Ishiyama, T. Lin, S. Lipkowitz, M. Park, J Biol Chem 279, 29565 (2004)). It has been reported that Cbl binds to endophilin-mediated receptor endocytosis, ubiquitination, and subsequent receptor degradation (A. Petrem et al, Nature 416, 187 (2002)). This receptor down-regulation mechanism has previously been described in the EGFR family, which also has a Cbl binding site (K. Shtiegman, Y. Yarden, Semin Cancer Biol 143939. doc 201019961 13, 29 (2003); MD Marmor, Y. Yarden , Oncogene 23, 2057 (2004); P. Peschard, M. Park, Cancer Cell 3, 519 (2003)). Regulatory abnormalities in Met and HGF have been reported in various tumors. Ligand-driven Met activation was observed in several cancers. Elevated serum and intratumoral HGF are observed in lung cancer, breast cancer, and multiple myeloma (JM Siegfried et al, Ann Thorac Surg 66, 1915 (1998); Ρ·C. Ma et al, Anticancer Res 23, 49 (2003); BE Elliott et al, Can J Physiol Pharmacol 80, 91 (2002); C. Seidel et al, Med Oncol 15, 145 (1998)). Overexpression of Met and/or HGF, Met amplification or mutation has been reported in various cancers such as colorectal cancer, lung cancer, gastric cancer and renal cancer, and is believed to drive non-ligand-dependent receptor activation (C. Birchmeier Et al., Nat Rev Mol Cell Biol 4, 915 (2003); G. Maulik et al., Cytokine Growth Factor Rev 13, 41 (2002)). In addition, inducible overexpression of Met in a mouse model of liver triggers hepatocellular carcinoma, suggesting that receptor overexpression drives non-ligand-dependent tumor formation (R· Wang et al, J Cell Biol 153, 1023 (2001)) . For patients with familial and sporadic renal papillary carcinoma (RPC), the report suggests the strongest evidence for Met in cancer. Mutations that cause constitutive activation of the receptor in the Met kinase domain have been identified in the RPC as germline and somatic mutations (L. Schmidt et al, Nat Genet 16, 68 (1997)). Introduction of such mutations in a mouse model of transgenic mice causes tumor formation and metastasis. (M. Jeffers et al., Proc Natl Acad Sci U S A 94, 11445 (1997)). The epidermal growth factor receptor (EGFR) family contains four closely related receptors involved in cellular responses such as differentiation and proliferation (HER1/EGFR, HER2, 143939.doc 201019961 HER3 and HER4). Overexpression of EGFR kinase or its ligand TGF-α is often associated with many cancers, including breast cancer, lung cancer, colorectal cancer, ovarian cancer, renal cell carcinoma, bladder cancer, head and neck cancer, and glioblastoma. Tumors and astrocytoma, and the excessive expression of this tumor promotes the malignant growth of these tumors. It has also been found that specific deletion mutations in the EGFR gene (EGFRvIII) increase cell tumor formation. Activation of the EGFR-stimulated signaling pathway promotes multiple processes that potentially promote cancer (e.g., proliferation, angiogenesis, cell motility, and invasion), reduces apoptosis, and induces resistance. Increased HER1/EGFR performance is often associated with advanced disease, metastasis, and poor prognosis. For example, in NSCLC and gastric cancer, increased HER1/EGFR performance is associated with high metastatic rate, poor tumor differentiation, and increased tumor proliferation. Mutations in the intrinsic protein tyrosine kinase activity of activated receptors and/or increased downstream signaling are observed in NSCLC and glioblastoma. However, mutations serve as the primary mechanism in conferring EGF receptor inhibitors (eg, erlotidine). The role of erlotinib (TARCEVA®) or gefitinib (gefitinib) is controversial. Mutant forms of the full-length EGF receptor have been reported to predict reactivity to the EGF receptor tyrosine kinase inhibitor gefitinib (Paez, JG et al, (2004) Science 304: 1497-1500; Lynch, T. J. Et al., (2004) Ν Engl. J. Med. 3 50:2129-2139). Cell culture studies have shown that cell lines exhibiting these mutated forms of EGF receptor (ie, H3255) are more sensitive to growth inhibition by the EGF receptor tyrosine kinase inhibitor gefitinib and require higher concentrations. Gefitinib inhibits tumor cell lines expressing wild-type EGF receptors. These observations suggest that although specific mutations in the EGF receptor may reflect higher sensitivity to EGF receptor inhibitors 143939.doc 201019961 , but does not identify a completely unresponsive phenotype. The use of compounds that directly inhibit the kinase activity of EGFR and antibodies that reduce EGFR kinase activity by blocking EGFR activation has been a hot research area as an anti-tumor agent (de Bono JS and Rowinsky, EK (2002) Trends in Mol. Medicine 8 :S19-S26 ; Dancey, J. and

Sausville, E.A. (2003) Nature Rev. Drug Discovery 2:92-313)。多項研究表明、揭示或提出,一些EGFR激酶抑制 劑在與某些其他抗癌或化學治療劑或治療組合使用時可改 良腫瘤細胞殺死或腫瘤形成扼殺(例如Herbst, R.S·等人, (2001) Expert Opin. Biol. Ther· 1:719-732 ; Solomon,B.等 人,(2003) Int. J. Radiat. Oncol. Biol. Phys· 55:713-723 ; Krishnan,S.等人,(2003) Frontiers in Bioscience 8,el-13 ; Grunwald, V. 及 Hidalgo, M. (2003) J. Nat· Cancer Inst. 95:851-867 ; Seymour L. (2003) Current Opin. Investig. Drugs 4(6):658-666 ; Khalil,Μ·Υ.等人,(2003) Expert Rev. Anticancer Ther.3:367-380 ; Bulgaru,A.M·等人’ (2003) Expert Rev. Anticancer Ther.3:269-279 ; Dancey, J. &Sausville,E.A.(2003)NatureRev.DrugDiscovery2:92-313 ; Ciardiello,F.等人,(2000) Clin. Cancer Res· 6:2053-2063 ;及專利公開案第US 2003/0157104號)。 埃羅替尼(例如鹽酸埃羅替尼,亦稱為TARCEVA®或 OSI-774)為EGFR激酶之可經口利用之抑制劑。在活體 外,埃羅替尼在若干人類腫瘤細胞株(包括結腸直腸癌及 乳房癌)中顯示具有針對EGFR激酶之實質抑制活性(Moyer 143939.doc -10- 201019961 J.D·等人,(1997) Cancer Res. 57:4838),且臨床前評估顯 示針對若干表現EGFR之人類腫瘤異種移植物之活性 (Pollack, V.A.等人,(1999) J. Pharmacol. Exp. Ther· 291:739)。在臨床試驗中,埃羅替尼在若干適應症中顯示 活性,該等適應症包括頭頸部癌(Soulieres,D.等人, (2004) J. Clin. Oncol. 22:77)、NSCLC (Perez-Soler R等 人,(2001) Proc· Am· Soc. Clin· Oncol. 20:310a,摘要 1235)、CRC (Oza,M.等人,(2003) Proc· Am. Soc. Clin. Oncol. 22:196a,摘要785)及MBC (Winer, E.等人,(2002) Breast Cancer Res. Treat. 76:5115a,摘要445 ; Jones, R.J. 等人,(2003) Proc. Am. Soc. Clin. Oncol. 22:45a,摘要 180)。在III期試驗中,埃羅替尼單一療法在晚期難治癒性 NSCLC患者中顯著延長存活,延遲疾病進展且延遲肺癌相 關症狀之惡化(Shepherd, F·等人 ’(2004) J. Clin. Oncology, 22:14S (7月15日增刊),摘要7022)。在2004年11月,美國 食品與藥物管理局(FDA)批准TARCEVA®在至少一種先前 化學療法方案失敗後用於治療局部晚期或轉移性非小細胞 肺癌(NSCLC)患者。 血管系統之發育為許多生理及病理過程之基本要求。諸 如胚胎及腫瘤之生長活躍之組織需要足夠的血液供應。該 等組織藉由產生促血管生成(pro-angiogenic)因子(其經由 稱為血管生成(angiogenesis)之過程促進新血管形成)來滿 足此需求。血管形成為複雜但有序之生物事件,其包括全 部或多個以下步驟:a)内皮細胞(EC)自現有EC增殖或自祖 143939.doc • 11 · 201019961 細胞(progenitor cell)分化;b) EC遷移且聚結形成條索狀 (cord-like)結構;c)隨後血管索進行管生成(tubulogenesis) 以形成具有中央腔之金管;d)現有血管索或血管發芽形成 二級血管;e)原有血管叢進行進一步重塑及再成型;及f) 募集内皮外(peri-endothelial)細胞來包袠内皮管,從而向 血管提供維持及調節功能;該等細胞包括小毛細管之外被 細胞(pericyte)、較大血管之平滑肌細胞及心臟中之心肌細 胞。Hanahan,D·Seience 277:48-50 (1997);Hogan,B.L· 及 Kolodziej, P. A. Nature Reviews Genetics· 3:513-23 (2002) ; Lubarsky,B.及 Krasnow,M. A. Cell· 112:19-28 (2003) 。 現已充分確定多種病症之發病機制均涉及血管生成。此 等病症包括實體腫瘤及轉移;動脈粥樣硬化;晶狀體後纖 維組織增生;血管瘤;慢性炎症;眼内新生血管疾病,諸 如增殖性視網膜病(例如糖尿病性視網膜病)、年齡相關之 黃斑變性(AMD)、新生血管性青光眼;移植角膜組織及其 他組織之免疫排斥;類風濕性關節炎;及牛皮癣。 Folkman等人,J. Biol. Chem·,267:10931-10934 (1992); Klagsbrun等人,Annu. Rev. Physiol. 53:217-239 (1991); 及 Garner A., 「Vascular diseases」,Pathobiology of Ocular Disease. A Dynamic Approach, Garner A.,Klintworth GK編, 第 2版(Marcel Dekker,NY, 1994),第 1625-1710頁。 在腫瘤生長之情況下,血管生成似乎對於增生轉變成腫 瘤形成及向腫瘤生長及轉移提供營養至關重要。Folkman 143939.doc -12- 201019961 等人,Nature 339:58 (1989)。新血管生成使得腫瘤細胞與 正常細胞相比獲取生長優勢及增殖自發性(pr〇liferative autonomy)。腫瘤通常以單個異常細胞開始,該異常細胞 可能因與可用毛細管床之距離而僅增殖至數立方毫米之尺 寸’且腫瘤可能長時間保持「休眠」而不進一步生長及擴 散(dissemination)。一些腫瘤細胞隨後轉變為血管生成表 型而活化内皮細胞,内皮細胞增殖且成熟為新的毛細血 管。此等新形成之血管不僅能使原發性腫瘤持續生長,而 且能使轉移性腫瘤細胞擴散且再移生(rec〇1〇nizati〇n)。相 應地,已在乳房癌以及數種其他腫瘤中觀察到腫瘤切片中 之微血管密度與患者存活之間存在相關性。冒以如打等 人,N· Engl. J. Med 324:1-6 (1991) ; Horak等人,Lancet 340:1120-1124 (1992); Macchiarini等人,1^11。61 340:145- 146 (1992)。雖然控制血管生成轉變之確切機制尚未完全 瞭解,但咸信腫瘤塊之新金管生成係由大量企管生成刺激 φ 劑及抑制劑之淨平衡所致(Folkman, 1995, Nat. Med. 1(1):27-31)〇 血管發育之過程受到嚴格調控。迄今為止,已顯示大量 分子(主要為周圍細胞所產生之分泌因子)調控EC分化、增 殖、遷移及聚結成為條索狀結構。舉例而言,已鑑別血管 内皮生長因子(VEGF)為刺激血管生成及誘導血管滲透性所 涉及之關鍵因子。Ferrara等人,Endocr. Rev· 18:4_25 (1997)。發現即使單一 VEGF等位基因喪失亦會導致胚胎死 亡,指出此因子在血管系統發育及分化中發揮不可替代之 143939.doc -13- 201019961 作用。此外,已顯示VEGF為與腫瘤及眼内病症相關之新 血管生成的關鍵介趙。Ferrara等人,Endocr. Rev.同上。 大多數所檢查之人類腫瘤過度表現VEGF mRNA。Berkman 等人,J. Clin. Invest. 91:153-159 (1993) ; Brown等人, Human Pathol. 26:86-91 (1995) ; Brown等人,Cancer Res. 53:4727-4735 (1993) ; Mattern等人,' Brit. J. Cancer 73:931-934 (1996) ; Dvorak 等人,Am. J. Pathol. 146:1029-1039 (1995)。 抗VEGF中和抗體抑制裸小鼠中多種人類腫瘤細胞株之 生長(Kim 等人,Nature 362:841-844 (1993) ; Warren 等 人,J. Clin. Invest. 95:1789-1797 (1995) ; BorgstrSm 等 人,Cancer Res· 56:4032-4039 (1996) ; Melnyk 等人, Cancer Res. 56:921-924 (1996))’且亦抑制缺血性視網膜病 症模型中之眼内血管生成。Adamis等人,Arch. Ophthalmol. 114:66-71 (1996)。因此,抗VEGF單株抗體或VEGF作用之 其他抑制劑為治療腫瘤及各種眼内新生血管病症之有前景 之候選物。此等抗體描述於例如1 998年1月14日公開之EP 817,648 及 1998 年 10 月 15 日公開之 WO 98/45331 及 WO 98/45332中。 癌症為人類健康最致命的威脅之一。僅在美國,每年有 近13 0萬新患者罹患癌症,且癌症為繼心血管疾病之後的 第二大死亡原因’約佔死亡原因的1/4。大部分此等死亡 由實體腫瘤造成。雖然某些癌症之醫學治療已取得顯著進 步,但所有癌症之總五年存活率在過去20年裏僅提高了約 143939.doc •14· 201019961 1 〇%。即使癌症之治療取得顯著進步,仍需尋求改良療 法。 本文中所引用之所有參考文獻(包括專利申請案及公開 案)均以全文引用之方式併入本文中。 【發明内容】 本發明提供治療諸如癌症之病理性病狀之組合療法,其 中c-met结抗劑與VEGF括抗劑組合,或其中c-met拮抗劑與 VEGF拮抗劑及EGFR拮抗劑組合,從而提供顯著抗腫瘤活 性。 在一態樣中,本發明提供治療個體癌症之方法,該等方 法包含向個體投與治療有效量之c-met拮抗劑及VEGF拮抗 劑。 在另一態樣中,本發明提供治療個體癌症之方法,該等 方法包含向個體投與治療有效量之c-met拮抗劑、VEGF拮 抗劑及EGFR拮抗劑。 在某些實施例中,VEGF拮抗劑為干擾VEGF與細胞受體 結合之化合物。該等阻斷VEGF之拮抗劑之實例包括(但不 限於)可溶性VEGF受體、對VEGF具有特異性之適體或肽 體、及抗VEGF抗體。在一實施例中,抗VEGF抗體為貝伐 單抗(bevacizumab) 〇 在某些實施例中,當組合使用時,投與在約0.05 mg/kg 至約1 5 mg/kg範圍内之貝伐單抗。在一實施例中,可向個 體投與一或多個以下劑量:約0.5 mg/kg、1 ·0 mg/kg、2.0 mg/kg、3.0 mg/kg ' 4.0 mg/kg、5.0 mg/kg、6.0 mg/kg、 143939.doc -15- 201019961 7.0 mg/kg、7.5 mg/kg、8.0 mg/kg、9.0 mg/kg、10 mg/kg 或15 mg/kg(或其任何組合)。該等劑量可間歇地投與,例 如每天、每三天、每週或每兩至三週投與一次。 c-met拮抗劑之實例包括(但不限於)可溶性c-met受體、 可溶性HGF變異體、對c-met或HGF具有特異性之適體或肽 體' c-met小分子、抗c-met抗體及抗HGF抗體。在一些實 施例中,c-met拮抗劑為抗c_met抗體。 在一些實施例中,抗c-met抗體為包含Fc區之單臂抗體 (one-armed antibody)(亦即重鍵可變域及輕鍵可變域形成 單一抗原結合臂),其中Fc區包含第一及第二Fc多肽,其 中該第一及第二Fc多肽呈複合物形式存在且形成與包含該 抗原結合臂之Fab分子相比增加該抗體片段之穩定性的Fc 區。Sausville, E.A. (2003) Nature Rev. Drug Discovery 2: 92-313). A number of studies have shown, revealed or suggested that some EGFR kinase inhibitors can improve tumor cell killing or tumor formation killing when used in combination with certain other anti-cancer or chemotherapeutic agents or treatments (eg Herbst, RS et al., 2001). ) Expert Opin. Biol. Ther· 1:719-732 ; Solomon, B. et al., (2003) Int. J. Radiat. Oncol. Biol. Phys· 55:713-723 ; Krishnan, S. et al. 2003) Frontiers in Bioscience 8, el-13; Grunwald, V. and Hidalgo, M. (2003) J. Nat· Cancer Inst. 95:851-867; Seymour L. (2003) Current Opin. Investig. Drugs 4( 6): 658-666; Khalil, Μ·Υ. et al., (2003) Expert Rev. Anticancer Ther. 3:367-380; Bulgaru, AM· et al. (2003) Expert Rev. Anticancer Ther. 3:269 -279; Dancey, J. & Sausville, EA (2003) NatureRev. DrugDiscovery 2: 92-313; Ciardiello, F. et al., (2000) Clin. Cancer Res. 6: 2053-2063; and Patent Publication No. US 2003/0157104). Erlotinib (such as erlotinib hydrochloride, also known as TARCEVA® or OSI-774) is an orally administrable inhibitor of EGFR kinase. In vitro, erlotinib has been shown to have substantial inhibitory activity against EGFR kinase in several human tumor cell lines, including colorectal cancer and breast cancer (Moyer 143939.doc -10- 201019961 JD· et al., (1997) Cancer Res. 57:4838), and preclinical evaluation showed activity against several human tumor xenografts expressing EGFR (Pollack, VA et al, (1999) J. Pharmacol. Exp. Ther. 291:739). In clinical trials, erlotinib has been shown to be active in several indications, including head and neck cancer (Soulieres, D. et al., (2004) J. Clin. Oncol. 22:77), NSCLC (Perez). - Soler R et al. (2001) Proc· Am. Soc. Clin Oncol. 20:310a, Abstract 1235), CRC (Oza, M. et al., (2003) Proc. Am. Soc. Clin. Oncol. 22 : 196a, Abstract 785) and MBC (Winer, E. et al., (2002) Breast Cancer Res. Treat. 76: 5115a, Abstract 445; Jones, RJ et al., (2003) Proc. Am. Soc. Clin. Oncol 22:45a, abstract 180). In a phase III trial, erlotinib monotherapy significantly prolonged survival in patients with advanced refractory NSCLC, delaying disease progression and delaying the progression of lung cancer-related symptoms (Shepherd, F. et al. (2004) J. Clin. Oncology , 22:14S (Supplementary Supplement, July 15), Abstract 7022). In November 2004, the US Food and Drug Administration (FDA) approved TARCEVA® for the treatment of patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) after failure of at least one prior chemotherapy regimen. The development of the vascular system is a fundamental requirement for many physiological and pathological processes. Organizations such as embryos and tumors that are actively growing need adequate blood supply. These tissues meet this need by producing pro-angiogenic factors that promote neovascularization via a process called angiogenesis. Angiogenesis is a complex but ordered biological event that includes all or more of the following steps: a) endothelial cell (EC) proliferation from existing EC or differentiation from progenitor cells; 143. EC migrates and coalesces to form a cord-like structure; c) subsequent vascular cord tubulogenesis to form a gold tube with a central lumen; d) existing vascular cord or blood vessel sprouts to form a secondary blood vessel; e) The original vascular plexus is further remodeled and reshaped; and f) the peri-endothelial cells are recruited to encapsulate the endothelial tube, thereby providing maintenance and regulation to the blood vessels; the cells include cells outside the small capillaries ( Pericyte), smooth muscle cells of larger blood vessels and cardiomyocytes in the heart. Hanahan, D. Seience 277:48-50 (1997); Hogan, BL· and Kolodziej, PA Nature Reviews Genetics·3:513-23 (2002); Lubarsky, B. and Krasnow, MA Cell·112:19-28 (2003). It has been well established that the pathogenesis of a variety of conditions involves angiogenesis. These conditions include solid tumors and metastases; atherosclerosis; post-lens fibrous tissue hyperplasia; hemangioma; chronic inflammation; intraocular neovascular diseases such as proliferative retinopathy (eg, diabetic retinopathy), age-related macular degeneration (AMD), neovascular glaucoma; immune rejection of transplanted corneal tissue and other tissues; rheumatoid arthritis; and psoriasis. Folkman et al, J. Biol. Chem., 267: 10931-10934 (1992); Klagsbrun et al, Annu. Rev. Physiol. 53:217-239 (1991); and Garner A., "Vascular diseases", Pathobiology A Dynamic Approach, Garner A., Klintworth GK, 2nd ed. (Marcel Dekker, NY, 1994), pp. 1625-1710. In the case of tumor growth, angiogenesis appears to be essential for the conversion of hyperplasia into tumor formation and nutrition for tumor growth and metastasis. Folkman 143939.doc -12- 201019961 et al, Nature 339:58 (1989). Neovascularization allows tumor cells to acquire growth advantage and pr〇liferative autonomy compared to normal cells. Tumors usually begin with a single abnormal cell that may only proliferate to a size of a few cubic millimeters due to the distance from the available capillary bed' and the tumor may remain "sleeping" for a long time without further growth and dissemination. Some tumor cells then transform into an angiogenic phenotype that activates endothelial cells, which proliferate and mature into new capillary blood vessels. These newly formed blood vessels not only enable the primary tumor to grow continuously, but also allow the metastatic tumor cells to spread and re-contribute (rec〇1〇nizati〇n). Accordingly, there has been a correlation between microvessel density in tumor sections and patient survival observed in breast cancer as well as in several other tumors. Take the example of N. Engl. J. Med 324:1-6 (1991); Horak et al., Lancet 340:1120-1124 (1992); Macchiarini et al., 1^11.61 340:145- 146 (1992). Although the exact mechanism controlling angiogenic transformation is not fully understood, the new gold tube production of the Xianxin tumor block is caused by a large number of chemistries generating a net balance of stimulating agents and inhibitors (Folkman, 1995, Nat. Med. 1(1) :27-31) The process of vascular development is strictly regulated. To date, a large number of molecules (mainly secreted factors produced by surrounding cells) have been shown to regulate EC differentiation, proliferation, migration and coalescence into a cord-like structure. For example, vascular endothelial growth factor (VEGF) has been identified as a key factor involved in stimulating angiogenesis and inducing vascular permeability. Ferrara et al., Endocr. Rev. 18:4_25 (1997). It was found that even a single VEGF allele loss can lead to embryonic death, indicating that this factor plays an irreplaceable role in vascular system development and differentiation. 143939.doc -13- 201019961 Role. In addition, VEGF has been shown to be a key mediator of neovascularization associated with tumors and intraocular disorders. Ferrara et al., Endocr. Rev. Ibid. Most human tumors examined overexpress VEGF mRNA. Berkman et al, J. Clin. Invest. 91: 153-159 (1993); Brown et al, Human Pathol. 26: 86-91 (1995); Brown et al, Cancer Res. 53: 4727-4735 (1993) Mattern et al, 'Brit. J. Cancer 73: 931-934 (1996); Dvorak et al, Am. J. Pathol. 146: 1029-1039 (1995). Anti-VEGF neutralizing antibodies inhibit the growth of various human tumor cell lines in nude mice (Kim et al, Nature 362: 841-844 (1993); Warren et al, J. Clin. Invest. 95: 1789-1797 (1995) BorgstrSm et al, Cancer Res. 56:4032-4039 (1996); Melnyk et al, Cancer Res. 56:921-924 (1996)) and also inhibits intraocular angiogenesis in a model of ischemic retinopathy. Adamis et al., Arch. Ophthalmol. 114: 66-71 (1996). Thus, anti-VEGF monoclonal antibodies or other inhibitors of VEGF action are promising candidates for the treatment of tumors and various intraocular neovascular disorders. Such antibodies are described in, for example, EP 817,648, published Jan. 14, 998, and WO 98/45331 and WO 98/45332, issued Oct. 15, 1998. Cancer is one of the most deadly threats to human health. In the United States alone, nearly 1.3 million new patients suffer from cancer each year, and cancer is the second leading cause of death after cardiovascular disease, which accounts for about a quarter of the cause of death. Most of these deaths are caused by solid tumors. Although the medical treatment of certain cancers has made significant progress, the total five-year survival rate of all cancers has only increased by about 143939.doc •14·201019961 1 〇% over the past 20 years. Even if the treatment of cancer has made significant progress, it is still necessary to seek improved treatment. All references (including patent applications and publications) cited herein are hereby incorporated by reference in their entirety. SUMMARY OF THE INVENTION The present invention provides a combination therapy for treating a pathological condition such as cancer, wherein a c-met antagonist is combined with a VEGF antagonist, or wherein a c-met antagonist is combined with a VEGF antagonist and an EGFR antagonist, thereby Provides significant anti-tumor activity. In one aspect, the invention provides a method of treating cancer in a subject, the method comprising administering to the individual a therapeutically effective amount of a c-met antagonist and a VEGF antagonist. In another aspect, the invention provides a method of treating cancer in a subject, the method comprising administering to the individual a therapeutically effective amount of a c-met antagonist, a VEGF antagonist, and an EGFR antagonist. In certain embodiments, the VEGF antagonist is a compound that interferes with the binding of VEGF to a cellular receptor. Examples of such antagonists that block VEGF include, but are not limited to, soluble VEGF receptors, aptamers or peptides specific for VEGF, and anti-VEGF antibodies. In one embodiment, the anti-VEGF antibody is bevacizumab. In certain embodiments, when used in combination, administration of bevacizated in the range of from about 0.05 mg/kg to about 15 mg/kg Monoclonal antibody. In one embodiment, one or more of the following doses can be administered to an individual: about 0.5 mg/kg, 1 · 0 mg/kg, 2.0 mg/kg, 3.0 mg/kg '4.0 mg/kg, 5.0 mg/kg , 6.0 mg/kg, 143939.doc -15- 201019961 7.0 mg/kg, 7.5 mg/kg, 8.0 mg/kg, 9.0 mg/kg, 10 mg/kg or 15 mg/kg (or any combination thereof). The doses can be administered intermittently, for example once a day, every three days, every week or every two to three weeks. Examples of c-met antagonists include, but are not limited to, soluble c-met receptors, soluble HGF variants, aptamers or peptibodies that are specific for c-met or HGF, c-met small molecules, anti-c- Met antibody and anti-HGF antibody. In some embodiments, the c-met antagonist is an anti-c-met antibody. In some embodiments, the anti-c-met antibody is a one-armed antibody comprising an Fc region (ie, a heavy-bond variable domain and a light-bond variable domain form a single antigen-binding arm), wherein the Fc region comprises The first and second Fc polypeptides, wherein the first and second Fc polypeptides are present as a complex and form an Fc region that increases the stability of the antibody fragment compared to a Fab molecule comprising the antigen binding arm.

在一些實施例中,抗c-met抗體包含(a)第一多肽,其包 含具有如下序列之重鏈可變域:EVQLVESGGGLVQPGGS LRLSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNS DTRFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYC ATYRSYVTPLDYWGQGTLVTVSS (SEQ ID NO: 10) > CHI 序列及第一Fc多肽;(b)第二多肽,其包含具有如下序列之 輕鏈可變域:DIQMTQSPSSLSASVGDRVTITCKSSQSLLYT SSQKNYLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSG SGTDFTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIK R(SEQ ID NO: 11)及CL1序列;及(c)包含第二Fc多肽之第 三多肽,其中重鏈可變域及輕鏈可變域呈複合物形式存在 143939.doc •16· 201019961 且形成單一抗原結合臂,其中第一及第二Fc多肽呈複合物 形式存在且形成與包含該抗原結合臂之Fab分子相比增加 該抗體片段之穩定性的Fc區。在一些實施例中,第一多肽 包含圖1中描繪之Fc序列(SEQ ID NO: 12),且第二多肽包 含圖2中描繪之Fc序列(SEQ ID NO: 13)。在一些實施例 中,第一多肽包含圖2中描繪之Fc序列(SEQ ID NO: 13), 且第二多肽包含圖1中描繪之Fc序列(SEQ ID NO: 12)。In some embodiments, the anti-c-met antibody comprises (a) a first polypeptide comprising a heavy chain variable domain comprising: EVQLVESGGGLVQPGGS LRLSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNS DTRFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYC ATYRSYVTPLDYWGQGTLVTVSS (SEQ ID NO: 10) > CHI sequence and first Fc polypeptide; (b) a second polypeptide comprising a light chain variable domain having the sequence: DIQMTQSPSSLSASVGDRVTITCKSSQSLLYT SSQKNYLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSG SGTDFTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIK R (SEQ ID NO: 11) and CL1 sequence; and (c) third comprising the second Fc polypeptide a polypeptide, wherein the heavy chain variable domain and the light chain variable domain are present in a complex form 143939.doc •16·201019961 and form a single antigen binding arm, wherein the first and second Fc polypeptides are present in a complex form and form An Fab molecule comprising the antigen binding arm is compared to an Fc region that increases the stability of the antibody fragment. In some embodiments, the first polypeptide comprises the Fc sequence depicted in Figure 1 (SEQ ID NO: 12) and the second polypeptide comprises the Fc sequence depicted in Figure 2 (SEQ ID NO: 13). In some embodiments, the first polypeptide comprises the Fc sequence depicted in Figure 2 (SEQ ID NO: 13) and the second polypeptide comprises the Fc sequence depicted in Figure 1 (SEQ ID NO: 12).

在一些實施例中,抗c-met抗體包含(a)包含重鏈可變域 之第一多肽,該多肽包含序列EVQLVESGGGLVQPGGSLR LSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNSDT RFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYCAT YRSYVTPLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPE VTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVSLSCAVKGFYPSD IAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 14) ; (b)包含輕鏈可變域之第二多肽,該多肽包含序列 DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKNYLAW YQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGTDFTLTIS SLQPEDFATYYCQQYYAYPWTFGQGTKVEIKRTVAAPSVFI 143939.doc -17· 201019961 FPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV THQGLSSPVTKSFNRGEC (SEQ ID NO: 15);及包含FC序 列之第三多肽,該多肽包含序列CPPCPAPELLGGPSVFLFP PKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK VSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQV SLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 13),其中重鏈可變域及輕鏈可變域 呈複合物形式存在且形成單一抗原結合臂,其中第一及第 二Fc多肽呈複合物形式存在且形成與包含該抗原結合臂之 Fab分子相比增加該抗體片段之穩定性的Fc區。 在一實施例中,抗c-met抗體包含重鏈可變域,該重鏈 可變域包含圖1中描繪之CDR1-HC、CDR2-HC及CDR3-HC 序列(SEQ ID NO: 4、5及/或9)中之一或多者。在一些實施 例中,該抗體包含輕鏈可變域,該輕鏈可變域包含圖1中 描繪之 CDR1-LC、CDR2-LC及 CDR3-LC序列(SEQ ID NO: 1、2及/或3)中之一或多者。在一些實施例中,重鏈可變域 包含圖1中描繪之FR1-HC、FR2-HC、FR3-HC及FR4-HC序 列(SEQ ID NO: 21-24)。在一些實施例中,輕鏈可變域包 含圖1中描繪之FR1-LC、FR2-LC、FR3-LC及FR4-LC序列 (SEQ ID NO: 16-19)。 適用於本發明方法中之其他抗c-met抗體描述於本文中 143939.doc • 18 - 201019961 且在此項技術中已知。 在一態樣中’抗c-met抗體包含至少一個促進抗體片段 内Fc序列之異二聚、而最小化同二聚之特徵。該(等)特徵 改良免疫球蛋白群體之產率及/或純度及/或均質性。在一 個實施例中,該抗體包含構成如WO 2005/063816中所述之 「杵(knobs)」及「臼(holes)」之Fc突變。舉例而言,臼突 變在Fc多肽中可為T366A、L368A及/或Y407V之一或多 者,空穴(cavity)突變可為T366W。 在某些實施例中,投與劑量為每三週約15 mg/kg之杬c_ met抗體。 在某些實施例中,EGFR拮抗劑為埃羅替尼。在某些實 施例中,在三週週期之每天投與劑量15〇 mg之埃羅替尼。 在某些實施例中,在三週週期之每天投與劑量1〇〇 mg之埃 羅替尼。在某些實施例中,在三週週期之每天投與劑量5〇 mg之埃羅替尼。 可使用本發明方法來影響任何適合病理狀態。舉例而 e,可使用本發明方法治療不同癌症,實體腫瘤與液體腫 瘤以及似軟組織腫瘤。可用本發明治療改善之癌症之非限 制性實例包括乳癌、結腸直腸癌、直腸癌、非小細胞肺 癌、非霍奇金氏淋巴瘤(n〇n_H〇dgkins 、 腎細胞癌、前列腺癌、肝癌(諸如肝細胞癌)、胰腺癌、軟 、卫織肉瘤、卡波西氏肉瘤(kap〇si,s…⑶脱)、類癌瘤 (carcinoid Carcinoma)、頭頸癌、黑素瘤、卵巢癌、胃癌、 間皮瘤及多發性骨髓瘤。在某些態樣中,癌症為轉移性 143939.doc •19- 201019961 的。在其他態樣中,癌症為非轉移性的。 在一些實施例中,抗c_met抗體(諸如MetMAb)及抗VEGF 抗體(諸如貝伐單抗)用於諸如非小細胞肺癌之癌症之組合 療法中。在另一實施例中,抗det抗體(諸如MetMAb)、 抗VEGF抗體(諸如貝伐單抗)及egFr抑制劑(諸如埃羅替 - 尼)用於諸如非小細胞肺癌之癌症之組合療法中。 視欲治療之特定癌症適應症而定,本發明之組合療法可 與其他治療劑(諸如化學治療劑)或其他療法(諸如放射療法 或手術)組合。許多已知的化學治療劑可用於本發明之組 ❹ 合療法中。在某些實施例中,本發明之組合療法可與一種 以上化學治療劑組合。在某些實施例中,使用治療特定適 應症之標準化學治療劑。在另一實施例中,用於組合中之 各治療劑之劑量或頻率等於或小於相應藥劑在無其他藥劑 下使用時之劑量或頻率。 【實施方式】 L定義 除非另外指示,否則如本文所使用之術語「肝細胞生長 〇 因子」或「HGF」係指能夠在允許活化HGF/c—met信號傳 導路徑之條件下發生該過程的任何原生或變異(原生或合 成)HGF多狀。術語「野生型HGF」泛指包含天然存在之 ·. HGF蛋白之胺基酸序列的多肽。術語「野生型序列」 - 泛扣天然存在之HGF中可見之胺基酸序列。(>111以為11(317之 已知受體,經由該受體在生物學上實現HGF細胞内信號傳 導。 143939.doc •20· 201019961 如本文所使用之術語「HGF變異體」係指在原生HGF序 列中包括一或多個胺基酸突變之HGF多肽。該一或多個胺 基酸突變視情況包括胺基酸取代。 「天然序列」多肽包含與源於自然界之多肽具有相同胺 基酸序列的多肽。因此,天然序列多肽可具有任何哺乳動 物之天然存在之多肽的胺基酸序列。該天然序列多肽可自 自然界分離,或可利用重組或合成方法產生。術語「天然 序列」多肽特定涵蓋多肽之天然存在之截斷或分泌形式 (例如細胞外域序列)、多肽之天然存在變異體形式(或者例 如剪接形式)及天然存在之等位基因變異體。 多肽「變異體」意謂與天然序列多肽具有至少約80%之 胺基酸序列一致性之生物活性多肽。該等變異體包括例如 在多肽之N端或C端添加或缺失一或多個胺基酸殘基之多 肽。變異體通常應與天然序列多肽具有至少約80%之胺基 酸序列一致性,更佳至少約90%之胺基酸序列一致性,且 甚至更佳至少約95%之胺基酸序列一致性。 「EGFR」意謂受體酪胺酸激酶多肽表皮生長因子受 體’其係描述於Ullrich等人,Nature (1984) 309:418425 中’或者稱為Her-1及c-erbB基因產物;以及其變異體,諸 如EGFRvIII。EGFR之變異體亦包括缺失、取代及插入性 變異體’例如 Lynch等人(New England Journal of Medicine 2004,350:2129)、Paez等人(Science 2004,304:1497)、Pao 等人(PNAS 2004,101:13306)中描述之變異體。 「生物樣品」(可互換稱為r樣品」或「組織或細胞樣 143939.doc •21 - 201019961 品」)涵蓋多種自個艘獲得之樣品類型,且可用於診斷或 監測檢定。料義涵蓋生物來源之血液及其他液體樣品, 實體組織樣品(諸如生檢試樣)或由其獲得之組織培養物或 細胞’及其子代。該定義亦包括已在取得後以任何方式加 以操縱之樣品’操縱方式諸如用試劑處理、溶解、或富集 某些組分(諸如蛋白質或聚核苷酸)或出於切片之目的包埋 於半固體或固體基質中。術語「生物樣品」涵蓋臨床樣 品’且亦包括培養物中之細胞、細胞上清液、細胞溶解產 物 β血漿、生物體液及組織樣品。生物樣品之來源 可為實體組織,來自新鮮、冷;東及/或保藏之器官或組織 樣品或生物檢體或抽出物;血液或任何血液組分;體液, 諸如腦脊髓液、羊水、腹膜液或間質液;個體妊娠期或發 育中任何時間之細胞。在一些實施例中,生物樣品係獲自 原發性或轉移性腫瘤。生物樣品可含有在自然界中非天然 與組織互混之化合物,諸如防腐劑、抗凝血劑、緩衝劑、 固定劑、營養素、抗生素或其類似物。 「抗c-met抗體」為以足夠的親和力及特異性結合 之抗體。所選擇之抗體通常應對c_met具肴足夠強的結合 親和力’舉例而§,抗體結合人類“met之Kd值可在1 〇〇 nM-1 pM之間。抗體親和力可利用以下方法測定,例如基 於表面電漿共振之檢定(諸如PCT申請公開案第w〇 2005/012359號中所述之BlAcore檢定)、酶聯免疫吸附檢定 (ELISA)及競爭檢定(例如RIA)。在某些實施例中,可使用 抗c-met抗體作為乾向及干擾涉及c-met活性之疾病或病狀 143939.doc -22- 201019961 之治療劑。另外,可使抗體經受其他生物活性檢定,例如 以評估其作為治療劑之效用。該等檢定在此項技術中為已 知的且視靶抗原及抗體之預定用途而定。 「c-met拮抗劑」(可互換稱為「c-met抑制劑」)為干擾c-met活化或功能之藥劑。c-met抑制劑之實例包括c-met抗 體、HGF抗體、小分子c-met拮抗劑、c-met酪胺酸激酶抑 制劑、反義及抑制性RNA(例如shRNA)分子(參見例如WO 2004/87207)。c-met抑制劑較佳為結合c-met之抗體或小分 子。在一特定實施例中,c-met抑制劑對c-met之結合親和 力(解離常數)為約1,〇〇〇 nM或更低。在另一實施例中,c-met抑制劑對c-met之結合親和力為約100 nM或更低。在另 一實施例中,c-met抑制劑對c-met之結合親和力為約50 nM或更低。在一特定實施例中,c-met抑制劑與c-met共價 結合。在一特定實施例中,c-met抑制劑抑制c-met信號傳 導之IC50為1,000 nM或更低。在另一實施例中,c-met抑制 劑抑制c-met信號傳導之IC50為500 nM或更低。在另一實 施例中,c-met抑制劑抑制c-met信號傳導之1C50為50 nM 或更低。在某些實施例中,c-met拮抗劑降低或抑制c-met 之表現含量或生物活性達至少10%、20%、30%、40%、 50%、60%、70%、80%、90%或更高。 「c-met活化」係指活化或填酸化c-met受體。c-met活化 一般引起信號轉導(例如由c-met或受質多肽中之c-met受體 磷酸化酪胺酸殘基之細胞内激酶域引起)。c-met活化可由 結合相關c-met受體之c-met配位體(HGF)介導。HGF與c- 143939.doc •23- 201019961 met之結合可活化c-met之激酶域,從而引起c-met中酷胺酸 殘基之磷酸化及/或其他受質多肽中酪胺酸殘基之磷酸 化。 術語「VEGF」或「VEGF-A」用於指如Leung等人, Science, 246:1306 (1989)及 Houck 等人,Mol. Endocrin., 5:1806 (1991)描述之165-胺基酸人類血管内皮細胞生長因 子及相關12卜胺基酸、189-胺基酸及206-胺基酸人類血管 内皮細胞生長因子,以及其天然存在之等位基因形式及已 加工形式。VEGF-A為包括 VEGF-B、VEGF-C、VEGF-D、 VEGF-E、VEGF-F及P1GF之基因家族之一部分。VEGF-A 主要結合兩種高親和力受體酪胺酸激酶,即VEGFR-l(Flt-1)及VEGFR-2(Flk-l/KDR),後者為VEGF-A之血管内皮細 胞的細胞分裂信號之主要傳遞質。另外,神經纖毛蛋白-1 (neuropilin-1)已經鑑別為肝素結合性VEGF-A同功異型物 之受體且可在血管發育中發揮作用。術語「VEGF」或 「VEGF-A」亦指來自諸如小鼠、大鼠或靈長類動物之非 人類物種之VEGF。來自特定物種之VEGF有時係用諸如 hVEGF(指人類VEGF)或mVEGF(指鼠類VEGF)之術語指 示。術語「VEGF」亦用於指包含165-胺基酸人類血管内 皮細胞生長因子之胺基酸8至109或1至109之多肽的截斷形 式或片段。在本申請案中,對VEGF之任何此類形式之提 及可例如用「VEGF(8-109)」、「VEGF(1-109)」或 「VEGF165」加以鑑別。「截斷」之原生VEGF之胺基酸位 置係如原生VEGF序列中所指示進行編號。舉例而言,截 143939.doc -24- 201019961 斷之原生VEGF中之胺基酸位置17(甲硫胺酸)在原生VEGF 中亦為位置17(甲硫胺酸)。截斷之原生VEGF對KDR及Flt-1受體之結合親和力與原生VEGF相當。 如本文所使用之術語「VEGF變異體」係指在原生VEGF 序列中包括一或多個胺基酸突變之VEGF多肽。該一或多 個胺基酸突變視情況包括胺基酸取代。應注意,出於簡記 本文所述之VEGF變異體之代號的目的,編號係指沿推定 原生VEGF之胺基酸序列之胺基酸殘基位置(提供於Leung 等人,同上及Houck等人,同上中)。 「VEGF生物活性」包括與任何VEGF受體結合’或任何 VEGF信號傳導活性,諸如調控正常與異常血管生成 (angiogenesis)及血管再生(vasculogenesis)(Ferrar a 及 Davis-Smyth (1997) Endocrine Rev. 18:4-25 ; Ferrara (1999) J. Mo/. Mei 77:527-543);促進胚胎血管再生及血管生成 (Carmeliet 等人,(1996) iVaiwre 380:435-439 ; Ferrara 等 人,(1996) 380:439-442);及調節雌性生殖道中之 週期性血管增生、及骨生長及軟骨形成(Ferrara等人, (1998) MeA 4:336-340 ; Gerber等人,(1999) iVaiwreIn some embodiments, the anti-c-met antibody comprises (a) a first polypeptide comprising a heavy chain variable domain, the polypeptide comprising the sequence EVQLVESGGGLVQPGGSLR LSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNSDT RFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYCAT YRSYVTPLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPE VTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVSLSCAVKGFYPSD IAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 14) ; (b) a second polypeptide comprising a light chain variable domain of the polypeptide comprising the sequence DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKNYLAW YQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGTDFTLTIS SLQPEDFATYYCQQYYAYPWTFGQGTKVEIKRTVAAPSVFI 143939.doc -17 · 201019961 FPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV THQGLSSPVTKSFNRGEC (SEQ ID NO: 15); and the third polypeptide comprising a FC sequence , the polypeptide comprises the sequence CPPCPAPELLGGPSVFLFP P KPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK VSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQV SLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 13), wherein the heavy chain variable domain and a light chain variable domain form and form a single antigen binding arm is a complex form, wherein the first and second Fc polypeptides form complexes in the form of There is and forms an Fc region that increases the stability of the antibody fragment compared to a Fab molecule comprising the antigen binding arm. In one embodiment, the anti-c-met antibody comprises a heavy chain variable domain comprising the CDR1-HC, CDR2-HC and CDR3-HC sequences depicted in Figure 1 (SEQ ID NO: 4, 5 And/or 9) one or more. In some embodiments, the antibody comprises a light chain variable domain comprising the CDR1-LC, CDR2-LC and CDR3-LC sequences depicted in FIG. 1 (SEQ ID NO: 1, 2, and/or One or more of 3). In some embodiments, the heavy chain variable domain comprises the FR1-HC, FR2-HC, FR3-HC, and FR4-HC sequences depicted in Figure 1 (SEQ ID NO: 21-24). In some embodiments, the light chain variable domain comprises the FR1-LC, FR2-LC, FR3-LC, and FR4-LC sequences depicted in Figure 1 (SEQ ID NOS: 16-19). Other anti-c-met antibodies suitable for use in the methods of the invention are described herein in 143939.doc • 18 - 201019961 and are known in the art. In one aspect, the anti-c-met antibody comprises at least one feature that promotes heterodimerization of the Fc sequence within the antibody fragment while minimizing homodimerization. This (etc.) feature improves the yield and/or purity and/or homogeneity of the immunoglobulin population. In one embodiment, the antibody comprises an Fc mutation that constitutes "knobs" and "holes" as described in WO 2005/063816. For example, the mutation can be one or more of T366A, L368A and/or Y407V in the Fc polypeptide, and the cavity mutation can be T366W. In certain embodiments, the administered dose is about 15 mg/kg of 杬c_ met antibody every three weeks. In certain embodiments, the EGFR antagonist is erlotinib. In certain embodiments, a dose of 15 mg of erlotinib is administered daily for a three week period. In certain embodiments, a dose of 1 mg of erlotinib is administered daily for a three week period. In certain embodiments, a dose of 5 mg of erlotinib is administered daily for a three week period. The methods of the invention can be used to affect any suitable pathological condition. By way of example, the methods of the invention can be used to treat different cancers, solid tumors and fluid tumors, as well as soft tissue tumors. Non-limiting examples of cancers that can be treated with the present invention include breast cancer, colorectal cancer, rectal cancer, non-small cell lung cancer, non-Hodgkin's lymphoma (n〇n_H〇dgkins, renal cell carcinoma, prostate cancer, liver cancer ( Such as hepatocellular carcinoma), pancreatic cancer, soft, atherosclerosis, Kaposi's sarcoma (kap〇si, s... (3) off), carcinoid carcinoma, head and neck cancer, melanoma, ovarian cancer, stomach cancer , mesothelioma, and multiple myeloma. In some aspects, the cancer is metastatic 143939.doc • 19-201019961. In other aspects, the cancer is non-metastatic. In some embodiments, the resistance A c_met antibody (such as MetMAb) and an anti-VEGF antibody (such as bevacizumab) are used in combination therapy such as cancer of non-small cell lung cancer. In another embodiment, an anti-det antibody (such as MetMAb), an anti-VEGF antibody ( Such as bevacizumab) and egFr inhibitors (such as erlotinib) are used in combination therapies such as cancer of non-small cell lung cancer. Depending on the particular cancer indication to be treated, the combination therapy of the present invention may Other therapeutic agents A combination such as a chemotherapeutic agent or other therapy such as radiation therapy or surgery. Many known chemotherapeutic agents are useful in the group therapy of the present invention. In certain embodiments, the combination therapy of the present invention can be combined with a Combination of the above chemotherapeutic agents. In certain embodiments, a standard chemotherapeutic agent for treating a particular indication is used. In another embodiment, the dosage or frequency of each therapeutic agent used in the combination is equal to or less than the corresponding agent at Dosage or frequency when used under other agents. [Embodiment] L Definitions The term "hepatocyte growth factor" or "HGF" as used herein, unless otherwise indicated, refers to the ability to activate HGF/c-met signals. Any native or variant (native or synthetic) HGF polymorphism of the process under the conditions of the conduction pathway. The term "wild-type HGF" refers broadly to a polypeptide comprising the amino acid sequence of a naturally occurring HGF protein. The term "wild type" Sequence" - the amino acid sequence visible in naturally occurring HGF. (>111 is 11 (the known receptor for 317, biologically via this receptor) Now HGF intracellular signaling. 143939.doc • 20· 201019961 The term “HGF variant” as used herein refers to an HGF polypeptide comprising one or more amino acid mutations in the native HGF sequence. Amino acid mutagenesis includes, as appropriate, amino acid substitution. A "native sequence" polypeptide comprises a polypeptide having the same amino acid sequence as a polypeptide derived from nature. Thus, a native sequence polypeptide can have an amine of any mammalian naturally occurring polypeptide. The native sequence polypeptide may be isolated from nature or may be produced by recombinant or synthetic methods. The term "native sequence" polypeptide specifically encompasses the naturally occurring truncated or secreted form of the polypeptide (eg, extracellular domain sequences), the natural presence of the polypeptide Variant forms (or, for example, spliced forms) and naturally occurring allelic variants. A polypeptide "variant" means a biologically active polypeptide having at least about 80% amino acid sequence identity to a native sequence polypeptide. Such variants include, for example, polypeptides having one or more amino acid residues added or deleted at the N-terminus or C-terminus of the polypeptide. The variant will typically have at least about 80% amino acid sequence identity to the native sequence polypeptide, more preferably at least about 90% amino acid sequence identity, and even more preferably at least about 95% amino acid sequence identity. . "EGFR" means the receptor tyrosine kinase polypeptide epidermal growth factor receptor' is described in Ullrich et al, Nature (1984) 309:418425 'or as the Her-1 and c-erbB gene products; and Variants, such as EGFRvIII. Variants of EGFR also include deletions, substitutions, and insertional variants such as Lynch et al. (New England Journal of Medicine 2004, 350: 2129), Paez et al. (Science 2004, 304: 1497), Pao et al. (PNAS 2004). , 101:13306) variants described. “Biological samples” (interchangeable as r samples) or “tissue or cell-like 143939.doc •21 - 201019961 products” cover a wide range of sample types obtained from individual vessels and can be used for diagnostic or monitoring tests. It is intended to encompass blood and other liquid samples of biological origin, solid tissue samples (such as biopsy samples) or tissue cultures or cells obtained therefrom and their progeny. This definition also includes samples that have been manipulated in any manner after they have been obtained, such as manipulations, dissolution, or enrichment of certain components (such as proteins or polynucleotides) or entrapment for slicing purposes. In a semi-solid or solid matrix. The term "biological sample" encompasses clinical samples' and also includes cells in culture, cell supernatants, cell lysate beta plasma, biological fluids, and tissue samples. The source of the biological sample may be a solid tissue from fresh, cold; eastern and / or preserved organ or tissue samples or biological specimens or extracts; blood or any blood components; body fluids, such as cerebrospinal fluid, amniotic fluid, peritoneal fluid Or interstitial fluid; cells of an individual during pregnancy or at any time during development. In some embodiments, the biological sample is obtained from a primary or metastatic tumor. The biological sample may contain compounds which are not naturally intermixed with the tissue in nature, such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics or the like. An "anti-c-met antibody" is an antibody that binds with sufficient affinity and specificity. The selected antibody should generally be sufficient for c_met to have a strong binding affinity'. For example, §, antibody binding to human "met Kd value can be between 1 〇〇 nM-1 pM. Antibody affinity can be determined by the following method, for example based on surface A plasma resonance assay (such as the BlAcore assay described in PCT Application Publication No. WO 2005/012359), an enzyme-linked immunosorbent assay (ELISA), and a competition assay (eg, RIA). In some embodiments, An anti-c-met antibody is used as a therapeutic agent for dryness and interference with diseases or conditions involving c-met activity 143939. doc -22-201019961. Additionally, antibodies can be subjected to other biological activity assays, for example to assess their use as therapeutic agents The utility of these assays is known in the art and depends on the intended use of the target antigen and antibody. "c-met antagonists" (interchangeably referred to as "c-met inhibitors") are interference c -met activation or functional agent. Examples of c-met inhibitors include c-met antibodies, HGF antibodies, small molecule c-met antagonists, c-met tyrosine kinase inhibitors, antisense and inhibitory RNA (eg, shRNA) molecules (see, eg, WO 2004). /87207). The c-met inhibitor is preferably an antibody or a small molecule that binds to c-met. In a particular embodiment, the binding affinity (dissociation constant) of the c-met inhibitor to c-met is about 1, 〇〇〇 nM or lower. In another embodiment, the binding affinity of the c-met inhibitor to c-met is about 100 nM or less. In another embodiment, the binding affinity of the c-met inhibitor to c-met is about 50 nM or less. In a specific embodiment, the c-met inhibitor is covalently bound to c-met. In a specific embodiment, the c-met inhibitor inhibits c-met signaling with an IC50 of 1,000 nM or less. In another embodiment, the c-met inhibitor inhibits c-met signaling with an IC50 of 500 nM or less. In another embodiment, the c-met inhibitor inhibits c-met signaling with a 1 C50 of 50 nM or less. In certain embodiments, the c-met antagonist reduces or inhibits the expression level or biological activity of c-met by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or higher. "C-met activation" refers to the activation or filling of the c-met receptor. C-met activation generally results in signal transduction (e. g., caused by c-met or the intracellular kinase domain of the c-met receptor phosphorylating tyrosine residues in the polypeptide). C-met activation can be mediated by a c-met ligand (HGF) that binds to the relevant c-met receptor. Binding of HGF to c-143939.doc •23-201019961 met activates the kinase domain of c-met, resulting in phosphorylation of valine residues in c-met and/or tyrosine residues in other receptor polypeptides Phosphorylation. The term "VEGF" or "VEGF-A" is used to mean 165-amino acid human as described by Leung et al, Science, 246: 1306 (1989) and Houck et al, Mol. Endocrin., 5: 1806 (1991). Vascular endothelial growth factor and related 12 amino acid, 189-amino acid and 206-amino acid human vascular endothelial growth factor, as well as naturally occurring allelic forms and processed forms thereof. VEGF-A is part of a gene family including VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF-F and P1GF. VEGF-A mainly binds to two high-affinity receptor tyrosine kinases, namely VEGFR-1 (Flt-1) and VEGFR-2 (Flk-1/KDR), which is the cell division signal of vascular endothelial cells of VEGF-A. The main pass quality. In addition, neuropilin-1 has been identified as a receptor for heparin-binding VEGF-A isoforms and may play a role in vascular development. The term "VEGF" or "VEGF-A" also refers to VEGF from non-human species such as mice, rats or primates. VEGF from a particular species is sometimes indicated by terms such as hVEGF (referred to as human VEGF) or mVEGF (referred to as murine VEGF). The term "VEGF" is also used to refer to a truncated form or fragment of a polypeptide comprising amino acid 8 to 109 or 1 to 109 of the 165-amino acid human endothelium cell growth factor. In the present application, the reference to any such form of VEGF can be identified, for example, by "VEGF (8-109)", "VEGF (1-109)" or "VEGF165". The amino acid position of the native VEGF that is "truncated" is numbered as indicated in the native VEGF sequence. For example, the amino acid position 17 (methionine) in the native VEGF of 143939.doc -24-201019961 is also position 17 (methionine) in native VEGF. The binding affinity of the truncated native VEGF to KDR and Flt-1 receptors is comparable to native VEGF. The term "VEGF variant" as used herein refers to a VEGF polypeptide comprising one or more amino acid mutations in the native VEGF sequence. The one or more amino acid mutations optionally include amino acid substitutions. It should be noted that for the purpose of abbreviating the designation of the VEGF variant described herein, the numbering refers to the position of the amino acid residue along the putative amino acid sequence of the native VEGF (provided in Leung et al., supra and Houck et al. Ibid.) "VEGF biological activity" includes binding to any VEGF receptor' or any VEGF signaling activity, such as regulation of normal and abnormal angiogenesis and vasculogenesis (Ferrar a and Davis-Smyth (1997) Endocrine Rev. 18 : 4-25; Ferrara (1999) J. Mo/. Mei 77: 527-543); promotes embryonic angiogenesis and angiogenesis (Carmeliet et al., (1996) iVaiwre 380: 435-439; Ferrara et al., 1996 380: 439-442); and regulation of periodic vascular proliferation, bone growth and cartilage formation in the female reproductive tract (Ferrara et al., (1998) MeA 4:336-340; Gerber et al., (1999) iVaiwre

Mei 5:623-628)。作為多效性生長因子,VEGF除作為血管 生成及血管再生中之血管生成因子外,亦在其他生理過程 中展現多種生物效應,該等生理過程諸如内皮細胞存活、 血管滲透及無管擴張、單核細胞趨化性及妈流入(Ferrara 及 Davis-Smyth (1997),同上;及Cebe-Suarez等人,Ce//. Mo/, h/e &ζ·. 63:601-615 (2006))。此外,最近研究已報導 143939.doc -25- 201019961 VEGF對少數非内皮細胞類型(諸如視網膜色素上皮細胞、 胰管細胞及神經勒細胞(Schwann cell))具有細胞分裂效 應。Guerrin等人,(1995) 乂 Ce// Ρ;2>;5ζ·ο/. 164:385-394 ; Oberg-Welsh^ A J (1997) Mol. Cell. Endocrinol. 126:125-132 ; Sondell^-A * (1999) Neurosci. 19:573 1-5740 o 「血管生成抑制劑」或「抗血管生成劑」係指直接或間 接抑制血管生成、血管再生或不期望的血管滲透之小分子 量物質、聚核普酸、多狀、分離蛋白、重組蛋白、抗體或 其結合物或融合蛋白。應瞭解,抗血管生成劑包括結合及 阻斷血管生成因子或其受體之血管生成活性之藥劑。舉例 而言,抗血管生成劑為如以上所定義之血管生成劑之抗體 或其他抬抗劑,例如VEGF-A或VEGF-A受體(例如KDR受 體或Flt-Ι受體)之抗體、抗PDGFR抑制劑(諸如 GLEEVEC®(甲磺酸伊馬替尼(Imatinib Mesylate)))。抗血 管生成劑亦包括原生血管生成抑制劑’例如血管抑制素、 内皮抑制素等。參見例如Klagsbrun及D'Amore, P/jyno/.,53:217-39 (1991) ; Streit 及 Detmar, 22:3172-3179 (2003)(例如,表3列舉惡性黑素瘤之抗血管 生成療法);Ferrara及 Alitalo, iVaiwre Me山ϋβ 5:1359-1364 (1999) ; Tonini等人,22:6549-6556 (2003)(例 如,表2列舉已知的抗血管生成因子);及Sato. /«ί· «/. C/ζ'π. 8:200-206 (2003)(例如,表1列舉臨床試驗中使用 之抗jk管生成劑)。 「VEGF拮抗劑」係指能夠中和、阻斷、抑制、消除、 143939.doc -26- 201019961 降低或干擾VEGF活性(包括其與一或多種VEGF受體之結 合)之分子(肽基或非肽基)。在某些實施例中,VEGF拮抗 劑降低或抑制VEGF之表現含量或生物活性達至少10%、 20%、30%、40%、50%、60%、70%、80%、90%或更高。 在一實施例中,受VEGF拮抗劑抑制之VEGF為VEGF(8-109)、VEGF(1-109)或VEGF165。適用於本發明方法中之 VEGF拮抗劑包括特異性結合VEGF之肽基或非肽基化合 物,諸如抗VEGF抗體及其抗原結合片段、特異性結合 VEGF之多肽或其片段,及特異性結合VEGF從而隔絕其與 一或多種受體(例如可溶性VEGF受體蛋白或其VEGF結合 片段,或嵌合VEGF受體蛋白)結合的受體分子及衍生物; 與編碼VEGF多肽之核酸分子之至少一片段互補的反義核 鹼基(nucleobase)寡聚物;與編碼VEGF多肽之核酸分子之 至少一片段互補的小RNA ;靶向VEGF之核酶;VEGF之肽 體;及VEGF適體。 「抗VEGF抗體」為以足夠的親和力及特異性結合VEGF 之抗體。所選擇之抗鱧通常應對VEGF具有足夠強的結合 親和力,例如抗體結合hVEGF之Kd值可在100 nM-1 pM之 間。抗體親和力可利用以下方法測定,例如基於表面電漿 共振之檢定(諸如PCT申請公開案第WO 2005/012359號中 所述之BIAcore檢定)、酶聯免疫吸附檢定(ELISA)及競爭 檢定(例如RIA)。在某些實施例中,可使用本發明之抗 VEGF抗體作為靶向及干擾涉及VEGF活性之疾病或病狀之 治療劑。另外,可使抗體經受其他生物活性檢定,例如以 143939.doc -27- 201019961 評估其作為治療劑之效用。該等檢定在此項技術中為已知 的且視把抗原及抗體之預定用途而定。實例包括HUVEC 抑制檢定(如以下實例中所述);腫瘤細胞生長抑制檢定(例 如WO 89/06692中所述);抗體依賴性細胞之細胞毒性 (ADCC)及補體介導之細胞毒性(CDC)檢定(美國專利 5,5〇0,362);及激動活性或造血作用檢定(參見w〇 95/27062”抗VEGF抗體通常應不結合其他VEGF同系物 (諸如VEGF-B或VEGF-C),亦不結合其他生長因子(諸如 P1GF、PDGF 或 bFGF)。 在某些實施例中,抗VEGF抗體包括與利用融合瘤ATCC HB 10709產生之單株抗VEGF抗體A4.6.1結合相同抗原決 定基之單株抗體;根據Presta等人,及以57:4593-4599 (1997)產生之重組人類化抗VEGF單株抗體。在一實 施例中,抗VEGF抗體為「貝伐單抗(BV)」,亦稱為 「rhuMAb VEGF」或「AVASTIN®」。其包含突變人類IgGl 構架區及來自阻斷人類VEGF與其受體結合之鼠類抗 hVEGF單株抗體A.4.6.1之抗原結合互補決定區。貝伐單抗 之約93%的胺基酸序列(包括大部分構架區)係衍生自人類 IgGl,且約7%之序列係衍生自鼠類抗體A4.6.1。貝伐單抗 之分子質量為約149,000道爾頓,且其經糖基化。貝伐單 抗已獲FDA批准與化學療法方案組合用於治療轉移性結腸 直腸癌(CRC)及非小細胞肺癌(NSCLC)。Hurwitz等人,N. Engl. J. Med. 350:2335-42 (2004) ; Sandler# A » N. Engl. J· Med. 3 55:2542-50 (2006)。目前,貝伐單抗正處於許多 143939.doc -28 · 201019961 正在進行之用於治療各種癌症適應症之臨床試驗的研究 中。Kerbel,*/. C/z«. Owco/· 19:45S-5 IS (2001) ; De Vore等 A 5 Proc. Am. Soc. Clin. Oncol. 19:485a. (2000) ; Hurwitz 等人,C//«· Cawcer 6:66-69 (2006) ; Johnson等 人,Proc. Jm. C7i«. 20:315a (2001) ; Kabbinavar 等人,·/. Once?/· 21:60-65 (2003) ; Miller等人,Mei 5: 623-628). As a pleiotropic growth factor, VEGF, in addition to its angiogenic factors in angiogenesis and angiogenesis, also exhibits a variety of biological effects in other physiological processes such as endothelial cell survival, vascular infiltration, and ductless expansion. Nuclear cell chemotaxis and maternal inflow (Ferrara and Davis-Smyth (1997), supra; and Cebe-Suarez et al., Ce//. Mo/, h/e & ζ·. 63:601-615 (2006) ). In addition, recent studies have reported that 143939.doc -25-201019961 VEGF has a cell division effect on a few non-endothelial cell types such as retinal pigment epithelial cells, pancreatic duct cells, and Schwann cells. Guerrin et al., (1995) 乂Ce// Ρ;2>;5ζ·ο/. 164:385-394; Oberg-Welsh^ AJ (1997) Mol. Cell. Endocrinol. 126:125-132 ; Sondell^- A * (1999) Neurosci. 19:573 1-5740 o "Angiogenesis inhibitor" or "anti-angiogenic agent" refers to a small molecular weight substance that directly or indirectly inhibits angiogenesis, angiogenesis or undesired vascular penetration. Nucleic acid, polymorphism, isolated protein, recombinant protein, antibody or combination thereof or fusion protein. It will be appreciated that an anti-angiogenic agent includes an agent that binds to and blocks the angiogenic activity of an angiogenic factor or its receptor. For example, an anti-angiogenic agent is an antibody or other antagonist of an angiogenic agent as defined above, such as an antibody to a VEGF-A or VEGF-A receptor (eg, a KDR receptor or a Flt-Ι receptor), Anti-PDGFR inhibitor (such as GLEEVEC® (Imatinib Mesylate)). Anti-angiogenic agents also include pro-angiogenic inhibitors such as angiostatin, endostatin and the like. See, for example, Klagsbrun and D'Amore, P/jyno/., 53:217-39 (1991); Streit and Detmar, 22:3172-3179 (2003) (eg, Table 3 lists anti-angiogenic therapies for malignant melanoma) ); Ferrara and Alitalo, iVaiwre Me Hawthorn 5: 1359-1364 (1999); Tonini et al, 22: 6549-6556 (2003) (for example, Table 2 lists known anti-angiogenic factors); and Sato. «ί· «/. C/ζ'π. 8:200-206 (2003) (for example, Table 1 lists anti-jk tube generators used in clinical trials). "VEGF antagonist" means a molecule that is capable of neutralizing, blocking, inhibiting, eliminating, 143939.doc -26-201019961 reducing or interfering with VEGF activity, including its binding to one or more VEGF receptors (peptidyl or non-peptide) Peptidyl). In certain embodiments, the VEGF antagonist reduces or inhibits the expression level or biological activity of VEGF by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more high. In one embodiment, the VEGF that is inhibited by the VEGF antagonist is VEGF (8-109), VEGF (1-109), or VEGF165. VEGF antagonists suitable for use in the methods of the invention include peptidyl or non-peptidyl compounds that specifically bind to VEGF, such as anti-VEGF antibodies and antigen-binding fragments thereof, polypeptides that specifically bind VEGF or fragments thereof, and specifically bind to VEGF Isolates a receptor molecule and derivative thereof that binds to one or more receptors (eg, a soluble VEGF receptor protein or a VEGF-binding fragment thereof, or a chimeric VEGF receptor protein); is complementary to at least a fragment of a nucleic acid molecule encoding a VEGF polypeptide An antisense nucleobase oligomer; a small RNA complementary to at least a fragment of a nucleic acid molecule encoding a VEGF polypeptide; a ribozyme that targets VEGF; a peptibody of VEGF; and a VEGF aptamer. An "anti-VEGF antibody" is an antibody that binds VEGF with sufficient affinity and specificity. The selected anti-sputum generally has a sufficiently strong binding affinity for VEGF, e.g., the antibody binding to hVEGF may have a Kd value between 100 nM and 1 pM. Antibody affinity can be determined by methods such as surface plasma resonance based assays (such as the BIAcore assay described in PCT Application Publication No. WO 2005/012359), enzyme-linked immunosorbent assay (ELISA), and competition assays (eg, RIA). ). In certain embodiments, an anti-VEGF antibody of the invention can be used as a therapeutic agent that targets and interferes with a disease or condition involving VEGF activity. Alternatively, the antibody can be subjected to other biological activity assays, e.g., as 143939.doc -27-201019961 for its utility as a therapeutic. Such assays are known in the art and depend on the intended use of the antigen and antibody. Examples include HUVEC inhibition assays (as described in the Examples below); tumor cell growth inhibition assays (eg, as described in WO 89/06692); antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CDC) Assay (US Patent 5,5,0,362); and agonistic activity or hematopoietic assay (see w〇95/27062) anti-VEGF antibodies should generally not bind to other VEGF homologs (such as VEGF-B or VEGF-C), nor Binding to other growth factors (such as P1GF, PDGF or bFGF). In certain embodiments, the anti-VEGF antibody comprises a monoclonal antibody that binds to the same epitope as a monoclonal anti-VEGF antibody A4.6.1 produced using the fusionoma ATCC HB 10709. According to Presta et al., and recombinant humanized anti-VEGF monoclonal antibody produced at 57:4593-4599 (1997). In one embodiment, the anti-VEGF antibody is "bevacizumab (BV)", also known as "rhuMAb VEGF" or "AVASTIN®", which comprises a mutant human IgG1 framework region and an antigen binding complementarity determining region from a murine anti-hVEGF monoclonal antibody A.4.6.1 that blocks binding of human VEGF to its receptor. Approximately 93% of the amino acid sequence resistant to the package The majority of the framework regions are derived from human IgG1, and about 7% of the sequence is derived from murine antibody A4.6.1. The molecular mass of bevacizumab is about 149,000 Daltons, and it is glycosylated. Valamycin has been approved by the FDA in combination with a chemotherapy regimen for the treatment of metastatic colorectal cancer (CRC) and non-small cell lung cancer (NSCLC). Hurwitz et al., N. Engl. J. Med. 350:2335-42 ( 2004); Sandler# A » N. Engl. J. Med. 3 55:2542-50 (2006). Currently, bevacizumab is in the process of many 143939.doc -28 · 201019961 ongoing treatment for various cancer adaptations In the study of clinical trials, Kerbel, */. C/z«. Owco/· 19:45S-5 IS (2001); De Vore et al. A 5 Proc. Am. Soc. Clin. Oncol. 19:485a. (2000); Hurwitz et al., C//«· Cawcer 6:66-69 (2006); Johnson et al., Proc. Jm. C7i«. 20:315a (2001); Kabbinavar et al., /. Once? /· 21:60-65 (2003); Miller et al.

Ca«.及ej. 7>eai. 94 :增子ij 1:S6 (2005) o 貝伐單抗及其他人類化抗VEGF抗體進一步描述於2005 ® 年2月26日頒予之美國專利第6,884,879號中。其他抗體包 括如PCT公開案第WO 2005/012359號、PCT公開案第WO 2005/044853號及美國專利申請案60/991,302中所述之G6或 Β20系列抗體(例如G6-31、Β20-4.1),此等專利申請案之内 容明確地以引用之方式併入本文中。關於其他抗體,參見美 國專利第7,060,269號、第6,582,959號、第6,703,020號、第 6,054,297號;WO 98/45332 ; WO 96/30046 ; WO 94/10202 ; ΕΡ 0666868Β1 ;美國專利申請公開案第2006009360號、第 9 20050186208號、第 20030206899號、第 20030190317號、 第 20030203409號及第 20050112126 號;及 Popkov 等人, Journal of Immunological Methods 288:149-164 (2004)° 其 他抗體包括結合人類VEGF上包含殘基F17、M18、D19、 Y21、Y25、Q89、191、K101、E103 及 C104 或者包含殘基 F17、Y21、Q22、Y25、D63、183 及 Q89之功能抗原決定 基的抗體。. 根據本發明之「G6系列抗體」為源自根據PCT公開案第 143939.doc -29- 201019961 WO 2005/012359號之圖 7、24-26及 34-35 中任一者之 G6抗 體或G6衍生抗體的序列之抗VEGF抗體,該公開案之全部 揭示内容明確地以引用之方式併入本文中。亦參見PCT公 開案第WO 2005/044853號,其全部揭示内容明確地以引用 之方式併入本文中。在一實施例中,G6系列抗體結合人類 VEGF上包含殘基 F17、Y21、Q22、Y25、D63、183 及 Q89 之功能抗原決定基。 根據本發明之「B20系列抗鱧」為源自根據PCT公開案 第WO 2005/012359號之圖27-29中任一者之B20抗體或B20 衍生抗體的序列之抗VEGF抗體,該公開案之全部揭示内 容明確地以引用之方式併入本文中。亦參見PCT公開案第 WO 2005/044853號及美國專利申請案60/991,302,此等專 利申請案之内容明確地以引用之方式併入本文中。在一實 施例中,B20系列抗體結合人類VEGF上包含殘基F17、 M18、D19、Y21、Y25、Q89、191、K101、E103 及 C104 之功能抗原決定基。 根據本發明之「功能抗原決定基」係指抗原中在能量方 面對抗體結合有貢獻之胺基酸殘基。抗原中任一個在能量 方面有貢獻之殘基的突變(例如由於丙胺酸或同系物突變 而使野生型VEGF突變)均將破壞抗體之結合,以致抗體之 相對親和力比(突變型VEGF的IC50/野生型VEGF的IC50)將 大於5(參見WO 2005/012359之實例2)。在一實施例中,相 對親和力比係由溶液結合噬菌體呈現ELISA來測定。簡言 之,在4°C下用於PBS中之濃度為2 pg/ml之待測試抗體的 143939.doc -30· 201019961Ca«. and ej. 7>eai. 94: Zengzi ij 1:S6 (2005) o Bevacizumab and other humanized anti-VEGF antibodies are further described in US Patent No. 6,884,879, issued February 26, 2005 No. Other antibodies include G6 or Β20 series antibodies (e.g., G6-31, Β20-4.1) as described in PCT Publication No. WO 2005/012359, PCT Publication No. WO 2005/044853, and U.S. Patent Application Serial No. 60/991,302. The contents of these patent applications are expressly incorporated herein by reference. For other antibodies, see U.S. Patent Nos. 7,060,269, 6,582,959, 6, 703, 020, 6, 054, 297, WO 98/45332, WO 96/30046, WO 94/10202, ΕΡ 0666868Β1, and U.S. Patent Application Publication No. 2006009360, No. 9 20050186208, No. 20030206899, No. 20030190317, No. 20030203409, and No. 20050112126; and Popkov et al, Journal of Immunological Methods 288: 149-164 (2004) ° Other antibodies including binding to human VEGF containing residues F17 M18, D19, Y21, Y25, Q89, 191, K101, E103 and C104 or an antibody comprising functional epitopes of residues F17, Y21, Q22, Y25, D63, 183 and Q89. The "G6 series antibody" according to the present invention is a G6 antibody or G6 derived from any one of Figures 7, 24-26 and 34-35 according to PCT Publication No. 143939.doc -29-201019961 WO 2005/012359. Anti-VEGF antibodies that are derived from the sequence of the antibody, the entire disclosure of which is expressly incorporated herein by reference. See also PCT Publication No. WO 2005/044853, the entire disclosure of which is expressly incorporated herein by reference. In one embodiment, the G6 series antibody binds to a functional epitope of residues F17, Y21, Q22, Y25, D63, 183 and Q89 on human VEGF. The "B20 series anti-caries" according to the present invention is an anti-VEGF antibody derived from the sequence of the B20 antibody or the B20-derived antibody according to any one of Figures 27-29 of PCT Publication No. WO 2005/012359, the disclosure of which is The entire disclosure is expressly incorporated herein by reference. See also PCT Publication No. WO 2005/044853 and U.S. Patent Application Serial No. 60/991, the disclosure of each of which is expressly incorporated by reference. In one embodiment, the B20 series of antibodies bind to functional determinants of residues F17, M18, D19, Y21, Y25, Q89, 191, K101, E103 and C104 on human VEGF. The "functional epitope" according to the present invention means an amino acid residue in an antigen which contributes to antibody binding in energy. Mutations in residues that are energy-dependent in any of the antigens (eg, mutations in wild-type VEGF due to mutations in alanine or homologs) will disrupt antibody binding, resulting in a relative affinity ratio of antibodies (IC50/ of mutant VEGF) The wild type VEGF will have an IC50) greater than 5 (see Example 2 of WO 2005/012359). In one embodiment, the relative affinity ratio is determined by the solution-bound phage displaying an ELISA. Briefly, the antibody to be tested at a concentration of 2 pg/ml in PBS at 4 ° C 143939.doc -30· 201019961

Fab形式塗布96孔Maxisorp免疫板(NUNC)隔夜,且在室溫 下用 PBS、0.5〇/〇 BSA及 0·05ο/〇 Tween20(PBT)阻斷 2小時。 首先,在室溫下在經Fab塗布的板上培育噬菌體呈現 hVEGF丙胺酸點突變體(殘基8-109形式)或野生型 hVEGF(8-109)於PBT中之連續稀釋液15分鐘,且用PBS、 0.05% Tween20(PBST)洗滌板。使用按1:5000稀釋於PBT中 之抗M13單株抗體辣根過氧化酶(Amer sham Pharmacia)結 合物來偵測結合之噬菌體,用3,3',5,5’-四甲基聯苯胺 (TMB,Kirkegaard & Perry Labs, Gaithersburg, MD)受質顯 色約5分鐘,用1.0 M H3P〇4中止且用分光光度法在45 0 nm 下讀數。IC50值之比(IC50,ala/IC50,wt)表示結合親和力減 小之倍數(相對結合親和力)。 「EGFR拮抗劑」(可互換稱為「EGFR抑制劑」)為干擾 c-met活化或功能之藥劑。EGFR抑制劑之實例包括EGFR抗 體、EGFR配位體抗體、小分子EGFR拮抗劑、EGFR酪胺 酸激酶抑制劑、反義及抑制性RNA(例如shRNA)分子(參見 例如WO 2004/87207)。EGFR抑制劑較佳為結合EGFR之抗 體或小分子。在一些實施例中,EGFR抑制劑為靶向EGFR 之藥物(EGFR-targeted drug)。在一特定實施例中,EGFR 抑制劑對EGFR之結合親和力(解離常數)為約1,000 nM或更 低。在另一實施例中,EGFR抑制劑對EGFR之結合親和力 為約100 nM或更低。在另一實施例中,EGFR抑制劑對 EGFR之結合親和力為約50 nM或更低。在一特定實施例 中,EGFR抑制劑與EGFR共價結合。在一特定實施例中, 143939.doc -31- 201019961 EGFR抑制劑抑制EGFR信號傳導之IC50為1,000 nM或更 低。在另一實施例中,EGFR抑制劑抑制EGFR信號傳導之 IC50為500 nM或更低。在另一實施例中,EGFR抑制劑抑 制EGFR信號傳導之IC50為50 nM或更低。在某些實施例 中,EGFR拮抗劑降低或抑制EGFR之表現含量或生物活性 達至少 10%、20%、30%、40%、50%、60%、70%、80%、 90%或更高。 「EGFR活化」係指EGFR之活化或磷酸化。EGFR活化 一般引起信號轉導(例如由EGFR或受質多肽中之EGFR受 體磷酸化酪胺酸殘基之細胞内激酶域引起)。EGFR活化可 藉由EGFR配位體與包含EGFR的EGFR二聚體之結合來介 導。EGFR配位體與EGFR二聚體之結合可活化二聚體中一 或多個EGFR之激酶域,從而引起一或多個EGFR中酪胺酸 殘基之磷酸化及/或其他受質多肽中酪胺酸殘基之磷酸 化。 如本文所使用之術語「靶向EGFR之藥物」係指結合 EGFR且抑制EGFR活化之治療劑。該等藥劑之實例包括結 合EGFR之抗體及小分子。結合EGFR之抗體之實例包括 MAb 579(ATCC CRL HB 8506)、MAb 455(ATCC CRL HB8507)、MAb 225(ATCC CRL 8508)、MAb 528(ATCC CRL 8509)(參見Mendelsohn等人之美國專利第4,943,533號)及其 變異鱧,諸如嵌合225(C225或西妥昔單抗(Cetuximab); ERBUTIX®)及再成型人類 225(H225)(參見 WO 96/40210, Imclone Systems Inc.); IMC-11F8,一 種把向 EGFR之完全 143939.doc -32- 201019961 人類抗體(Imcl〇ne);結合π型突變EGFR之抗體(美國專利 第5,212,290號);如美國專利第5,891,996號所述之結合 EGFR之人類化及嵌合抗體;及結合EGFR之人類抗體,諸 如 ABX-EGF(參見 WO 98/50433,Abgenix) ; EMD 55900 (Stragliotto等人,*/. Cawcer 32A:636-640 (1996)); EMD7200(馬妥珠單抗(matuzumab)),一種針對EGFR之人 類化EGFR抗體,其與EGF及TGF-α競爭結合EGFR ;及 mAb 806 或人類化 mAb 806(Johns 等人,*/· ·δζ·ο/· C/iewi· 279(29):30375-30384 (2004))。抗 EGFR 抗體可與細胞毒性 劑結合,從而產生免疫結合物(參見例如EP659,439A2, Merck Patent GmbH)。結合EGFR之小分子之實例包括 ZD1839 或吉非替尼(IRESSA ; Astra Zeneca) ; CP-358774 或埃羅替尼(TARCEVATM ; Genentech/OSI);及 AG1478、 AG1571(SU 5271 ; Sugen) ; EMD-7200。 片語「基因擴增」係指在特定細胞或細胞株中形成基因 或基因片段之多個複本之方法。複製區(一段擴增之DNA) 時常稱為「擴增子(amplicon)」》所產生之信使RNA (mRNA)之量(亦即基因表現含量)通常亦與所表現之特定基 因產生之複本數目成比例增加。 「酪胺酸激酶抑制劑」為在一定程度上抑制酪胺酸激酶 (諸如c-met受體)之酪胺酸激酶活性之分子。 「顯示c-met及/或EGFR表現、擴增或活化j之癌症或生 物樣品為在診斷測試中表現(包括過度表現)c_met及/或 EGFR、具有擴增之c-met及/或EGFR基因及/或展示c-met及/ 143939.doc • 33· 201019961 或EGFR之活化或磷酸化的樣品。 「不顯示c-met及/或EGFR表現、擴增或活化」之癌症或 生物樣品為在診斷測試中不表現(包括過度表現)C-met及/ 或EGFR、不具有擴增之c_inet;及/或EGFR基因及/或不展示 c-met及/或EGFR之活化或磷酸化的樣品。 「顯示c-met及/或EGFR活化」之癌症或生物樣品為在診 斷測試中展示c-met及/或EGFR之活化或填酸化之樣品。該 活化可直接(例如藉由利用ELISA量測c-met及/或EGFR碌酸 化)或間接確定。 「不顯示c-met及/或EGFR活化」之癌症或生物樣品為在 診斷測試中不展示c-met & /或EGFR之活化或填酸化之樣 品。該活化可直接(例如藉由利用ELISA量測c-met及/或 EGFR鱗酸化)或間接確定。 「不顯示c-met及/或EGFR擴增」之癌症或生物樣品為在 診斷測試中不具有擴增之c-met及/或EGFR基因之樣品。 「顯示c-met及/或EGFR擴增」之癌症或生物樣品為在診 斷測試中具有擴增之c-met及/或EGFR基因之樣品。 本文中之「磷酸基-ELIS A檢定」為在酶聯免疫吸附檢定 (ELISA)中使用通常為抗體之試劑來偵測璘酸化c-met& /或 EGFR、受質或下游信號傳導分子,從而評估一或多種c_ met及/或EGFR之磷酸化的檢定。較佳使用偵測磷酸化c-met及/或EGFR之抗體。該檢定可對較佳來自新鮮或冷凍 生物樣品之細胞溶解產物執行。 「c-met及/或EGFR過度表現或擴增」之癌細胞為與相同 143939.doc -34- 201019961 組織類型之非癌性細胞相比,c-met及/或EGFR蛋白或基因 之含量顯著較高的細胞。該過度表現可由基因擴增或增加 之轉錄或轉譯引起。c-met及/或EGFR過度表現或擴增可在 診斷或預後檢定中藉由評估細胞表面上存在之c-met及/或 EGFR蛋白之含量增加(例如經由免疫組織化學檢定;IHC) 來確定。或者或另外,可例如經由發光原位雜交(fluorescent in situ hybridization)(FISH ;參見 1998 年 10 月公開之 WO 98/45479)、南方墨點法(southern blotting)或聚合酶鏈反應 (PCR)技術(諸如定量即時PCR(qRT-PCR))來量測細胞中c-met及/或EGFR編碼核酸之含量。除上述檢定以外,熟練 人士亦可利用各種活體内檢定。舉例而言,可將患者體内 之細胞曝露於視情況標記可偵測標記(例如放射性同位素) 之抗體,且可藉由例如外部掃描放射性或藉由分析自先前 曝露於抗體之患者獲取之生檢來評估抗體與患者細胞的結 合。 「不過度表現或擴增c-met及/或EGFR」之癌細胞為與相 同組織類型之非癌性細胞相比,c-met及/或EGFR蛋白或基 因之含量不高於正常含量的細胞。 如本文所使用之術語「突變」意謂特定蛋白質或核酸 (基因、RNA)之胺基酸或核酸序列分別相對於野生型蛋白 質或核酸存在差異。突變蛋白或核酸可自基因之一個等位 基因(雜合)或兩個等位基因(純合)表現或可見於該一或兩 個等位基因上,且可為體細胞系或生殖細胞系。在本發明 中,突變一般為體細胞突變。突變包括序列重排,諸如插 143939.doc -35- 201019961 入、缺失及點突變(包括單一核苦酸/胺基酸多形現象)。 「抑制」為與參考值相比降低或減小活性、功能及/或 量。 蛋白質「表現」係指基因中編碼之資訊轉變為信使 RNA(mRNA),隨後轉變為蛋白質。 本文中,「表現」相關蛋白質(諸如HER受體或HER配位 體)之樣品或細胞為確定存在編碼蛋白質之mRNA或蛋白質 (包括其片段)之樣品或細胞。 「「免疫結合物」(可互換稱為「抗體_藥物結合物」或 「ADC」)意謂結合一或多種細胞毒性劑之抗體,該一或 多種細胞毒性劑諸如化學治療劑、藥物、生長抑制劑、毒 素(例如蛋白質毒素、細菌、真菌、植物或動物來源之酶 促活性毒素或其片段)或放射性同位素(亦即放射結合物)。 如本文所使用之片語「抗原結合臂」係指本發明之抗體 片段中能夠特異性結合相關靶分子之組成部分。抗原結合 臂一般且較佳為免疫球蛋白多肽序列(例如免疫球蛋白輕 鏈及重鏈之CDR及/或可變域序列)之複合物。 如本文所使用之片語「N端截斷重鏈」係指包含全長免 疫球蛋白重鏈之部分而非全部的多肽,其中缺失的部分為 通常位於重鏈端區域之部分。缺失之部分可包括(但不 限於)可變域、CH1、及鉸鏈序列之一部分或全部。一般而 言,若不存在野生型鉸鏈序列,則N端截斷重鏈之剩餘恆 定域應包含能夠鍵聯另一 Fc序列之組件(亦即如本文所述 之「第一」Fc多肽卜舉例而言,該組件可為能夠形成雙 143939.doc -36· 201019961 硫鍵之經修飾殘基或添加之半胱胺酸殘基。 如本文所使用之術語「Fc區」泛指包含免疫球蛋白重鏈 之C端多肽序列之二聚體複合物,其中c端多肽序列為可 藉由木瓜蛋白酶消化完整抗體而獲得之序列。Fc區可包含 原生或變異Fc序列。雖然免疫球蛋白重鏈之pc序列之邊界 可變化,但人類IgG重鏈Fc序列通常係界定為自約位置 Cys226處之胺基酸殘基或自约位置pr〇230伸展至Fc序列之 羧基端。免疫球蛋白之F c序列一般包含兩個恆定域,即 CH2域及CH3域,且視情況包含CH4域。本文中,「Fc多 肽」意謂構成Fc區之一種多肽。Fc多狀可自諸如IgG 1、 IgG2、IgG3 或 IgG4 亞型、IgA、IgE、IgD 或 IgM 之任何合 適的免疫球蛋白獲得。在一些實施例中,Fc多肽包含野生 型鉸鏈序列之一部分或全部(一般在N端)。在一些實施例 中,Fc多肽不包含功能型或野生型鉸鏈序列。 術語「Fc受體」及「FcR」用於描述結合抗體之Fc區的 受體。舉例而言’ FcR可為天然序列人類FcR。一般而 言,FcR為結合IgG抗體(γ受體)且包括Fc(RI、Fc(RII及 Fc(RIII亞類之受體(包括此等受體之等位基因變異體及另 外剪接形式)的FcR。Fc(RII受體包括Fc(RIIA(「活化受 體」)及Fc(RIIB(「抑制受體」),Fc(RIIA及Fc(RIIB具有類 似胺基酸序列’主要不同在於其細胞質域。其他同型之免 疫球蛋白亦可由某些FcR結合(參見例如Jane way等人, Immuno Biology · the immune system in health and disease, (Elsevier Science Ltd·,NY)(第 4版,1999))。活化受體 Fc 143939.doc 37 · 201019961 (RIIA在其細胞質域中含有基於免疫受體酪胺酸之活化基 元(immunoreceptor tyrosine-based activation motif , ITAM”抑制受體Fc(RIIB在其細胞質域中含有基於免疫受體 酷胺酸之抑制基元(immunoreceptor tyrosine-based inhibition motif» ITIM)(.^ idL Daeron, Annu. Rev. Immunol. 15:203- 234 (1997)中)。FcR 綜述於 Ravetch 及 Kinet, .*The Fab format was coated with 96-well Maxisorp immunoplate (NUNC) overnight and blocked with PBS, 0.5 〇/〇 BSA and 0·05ο/〇 Tween 20 (PBT) for 2 hours at room temperature. First, phage were incubated on Fab-coated plates at room temperature to present serial dilutions of hVEGF alanine point mutant (residue 8-109 format) or wild-type hVEGF (8-109) in PBT for 15 minutes, and The plates were washed with PBS, 0.05% Tween 20 (PBST). The anti-M13 monoclonal antibody horseradish peroxidase (Amer sham Pharmacia) conjugate diluted 1:5000 in PBT was used to detect bound phage using 3,3',5,5'-tetramethylbenzidine (TMB, Kirkegaard & Perry Labs, Gaithersburg, MD) was subjected to color development for about 5 minutes, stopped with 1.0 M H3P〇4 and read spectrophotometrically at 45 0 nm. The ratio of IC50 values (IC50, ala/IC50, wt) represents a fold reduction in binding affinity (relative binding affinity). "EGFR antagonists" (interchangeably referred to as "EGFR inhibitors") are agents that interfere with c-met activation or function. Examples of EGFR inhibitors include EGFR antibodies, EGFR ligand antibodies, small molecule EGFR antagonists, EGFR tyrosine kinase inhibitors, antisense and inhibitory RNA (e.g., shRNA) molecules (see, e.g., WO 2004/87207). The EGFR inhibitor is preferably an antibody or small molecule that binds to EGFR. In some embodiments, the EGFR inhibitor is an EGFR-targeted drug. In a specific embodiment, the binding affinity (dissociation constant) of the EGFR inhibitor to EGFR is about 1,000 nM or less. In another embodiment, the binding affinity of the EGFR inhibitor to EGFR is about 100 nM or less. In another embodiment, the binding affinity of the EGFR inhibitor to EGFR is about 50 nM or less. In a specific embodiment, the EGFR inhibitor is covalently bound to EGFR. In a specific embodiment, 143939.doc -31-201019961 EGFR inhibitors inhibit EGFR signaling with an IC50 of 1,000 nM or less. In another embodiment, the EGFR inhibitor inhibits EGFR signaling with an IC50 of 500 nM or less. In another embodiment, the EGFR inhibitor inhibits EGFR signaling with an IC50 of 50 nM or less. In certain embodiments, the EGFR antagonist reduces or inhibits the expression level or biological activity of EGFR by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more high. "EGFR activation" refers to activation or phosphorylation of EGFR. EGFR activation generally results in signal transduction (e. g., caused by the intracellular kinase domain of EGFR receptor phosphorylated tyrosine residues in EGFR or a polypeptide of interest). EGFR activation can be mediated by binding of an EGFR ligand to an EGFR dimer comprising EGFR. Binding of an EGFR ligand to an EGFR dimer activates one or more kinase domains of a EGFR in a dimer, thereby causing phosphorylation of tyrosine residues in one or more EGFR and/or other receptor polypeptides Phosphorylation of tyrosine residues. The term "drug targeted to EGFR" as used herein refers to a therapeutic agent that binds to EGFR and inhibits EGFR activation. Examples of such agents include antibodies and small molecules that bind EGFR. Examples of antibodies that bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see US Patent No. 4,943,533 to Mendelsohn et al.) And its variants, such as chimeric 225 (C225 or Cetuximab; ERBUTIX®) and reshaped human 225 (H225) (see WO 96/40210, Imclone Systems Inc.); IMC-11F8, A fully 143939.doc-32-201019961 human antibody (Imcl〇ne) to EGFR; an antibody that binds to a π-type mutant EGFR (U.S. Patent No. 5,212,290); as described in U.S. Patent No. 5,891,996, incorporated herein by reference. Humanized and chimeric antibodies; and human antibodies that bind to EGFR, such as ABX-EGF (see WO 98/50433, Abgenix); EMD 55900 (Stragliotto et al, */. Cawcer 32A: 636-640 (1996)); EMD7200 (Matuzumab), a humanized EGFR antibody against EGFR that competes with EGF and TGF-α for binding to EGFR; and mAb 806 or humanized mAb 806 (Johns et al., */··δζ· ο/· C/iewi· 279(29): 30375-30384 (2004)). The anti-EGFR antibody can be combined with a cytotoxic agent to produce an immunoconjugate (see, e.g., EP 659, 439 A2, Merck Patent GmbH). Examples of small molecules that bind to EGFR include ZD1839 or gefitinib (IRESSA; Astra Zeneca); CP-358774 or erlotinib (TARCEVATM; Genentech/OSI); and AG1478, AG1571 (SU 5271; Sugen); EMD- 7200. The phrase "gene amplification" refers to a method of forming multiple copies of a gene or gene fragment in a particular cell or cell line. The amount of messenger RNA (mRNA) produced by the replication region (a piece of amplified DNA), often referred to as the "amplicon" (ie, the amount of gene expression), is usually also the number of copies produced by the particular gene being expressed. Proportionate increase. A "tyrosine kinase inhibitor" is a molecule that inhibits the tyrosine kinase activity of a tyrosine kinase (such as a c-met receptor) to some extent. "A cancer or biological sample showing c-met and/or EGFR expression, amplification or activation is a manifestation (including overexpression) of c_met and/or EGFR in a diagnostic test, with an amplified c-met and/or EGFR gene And/or show c-met and / 143939.doc • 33· 201019961 or a sample of activation or phosphorylation of EGFR. “A cancer or biological sample that does not show c-met and/or EGFR expression, amplification or activation” is Samples that do not exhibit (including overexpression) C-met and/or EGFR, have no amplified c_inet; and/or EGFR genes and/or do not display activation or phosphorylation of c-met and/or EGFR. A cancer or biological sample that shows "c-met and/or EGFR activation" is a sample that exhibits activation or acidification of c-met and/or EGFR in a diagnostic test. This activation can be determined directly (e.g., by measuring c-met and/or EGFR acidification using ELISA) or indirectly. A cancer or biological sample that does not display c-met and/or EGFR activation is a sample that does not exhibit activation or acidification of c-met & / / EGFR in diagnostic tests. This activation can be determined directly (e.g., by measuring c-met and/or EGFR squaring using ELISA) or indirectly. A cancer or biological sample that does not display c-met and/or EGFR amplification is a sample that does not have an amplified c-met and/or EGFR gene in a diagnostic test. A cancer or biological sample showing "c-met and/or EGFR amplification" is a sample having an amplified c-met and/or EGFR gene in a diagnostic test. As used herein, "phosphoryl-ELIS A assay" is the use of a reagent, typically an antibody, in an enzyme-linked immunosorbent assay (ELISA) to detect c-met & / or EGFR, a receptor or a downstream signaling molecule, thereby A assay to assess phosphorylation of one or more c-mets and/or EGFR. Preferably, antibodies that detect phosphorylated c-met and/or EGFR are used. This assay can be performed on cell lysates preferably from fresh or frozen biological samples. The cancer cells of "c-met and/or EGFR overexpressed or expanded" have significant levels of c-met and/or EGFR protein or gene compared to non-cancerous cells of the same 143939.doc -34-201019961 tissue type. Higher cells. This overexpression can be caused by gene amplification or increased transcription or translation. Overexpression or amplification of c-met and/or EGFR can be determined in a diagnostic or prognostic assay by assessing the increase in the amount of c-met and/or EGFR protein present on the cell surface (eg, via immunohistochemical assay; IHC) . Alternatively or additionally, for example, via fluorescence in situ hybridization (FISH; see WO 98/45479, published October 1998), southern blotting or polymerase chain reaction (PCR) techniques. (such as quantitative real-time PCR (qRT-PCR)) to measure the amount of c-met and/or EGFR-encoding nucleic acid in a cell. In addition to the above tests, skilled individuals can also use a variety of in vivo tests. For example, cells in a patient can be exposed to antibodies that optionally detect detectable labels (eg, radioisotopes) and can be obtained by, for example, external scanning of radioactivity or by analysis of patients previously exposed to antibodies. The assay assesses the binding of the antibody to the patient's cells. A cancer cell that does not overexpress or amplify c-met and/or EGFR is a cell with a c-met and/or EGFR protein or gene content that is not higher than normal cells compared to a non-cancerous cell of the same tissue type. . The term "mutation" as used herein means that the amino acid or nucleic acid sequence of a particular protein or nucleic acid (gene, RNA) differs from the wild type protein or nucleic acid, respectively. A mutein or nucleic acid can be expressed from or can be found on one or both alleles of a gene (homozygous) or both alleles, and can be a somatic cell line or a germ cell line . In the present invention, the mutation is generally a somatic mutation. Mutations include sequence rearrangements, such as insertions, deletions, and point mutations (including single nucleotides/amino acid polymorphisms). "Inhibition" is the reduction or decrease in activity, function and/or amount compared to a reference value. Protein "performance" refers to the conversion of information encoded in a gene into messenger RNA (mRNA), which is then converted to protein. As used herein, a sample or cell that "expresses" a related protein (such as a HER receptor or a HER ligand) is a sample or cell that determines the presence of mRNA or protein (including fragments thereof) encoding the protein. "Immunoconjugate" (interchangeably referred to as "antibody-drug conjugate" or "ADC") means an antibody that binds to one or more cytotoxic agents, such as chemotherapeutics, drugs, growth. Inhibitors, toxins (eg, protein toxins, bacterial, fungal, enzymatically active toxins or fragments thereof of plant or animal origin) or radioisotopes (ie, radioconjugates). The phrase "antigen-binding arm" as used herein refers to a component of an antibody fragment of the present invention which is capable of specifically binding to a related target molecule. The antigen binding arm is generally and preferably a complex of immunoglobulin polypeptide sequences (e.g., CDRs and/or variable domain sequences of immunoglobulin light and heavy chains). As used herein, the phrase "N-terminal truncated heavy chain" refers to a polypeptide comprising a portion, but not all, of the full-length immunoglobulin heavy chain, wherein the deleted portion is the portion of the heavy chain end region. The missing portion may include, but is not limited to, a variable domain, a CH1, and a portion or all of the hinge sequence. In general, if no wild-type hinge sequence is present, the remaining constant domain of the N-terminal truncated heavy chain should comprise a component capable of binding another Fc sequence (ie, a "first" Fc polypeptide as described herein). The component may be a modified residue capable of forming a double 143939.doc -36·201019961 sulfur bond or an added cysteine residue. The term "Fc region" as used herein generally refers to a heavy immunoglobulin-containing molecule. A dimeric complex of a C-terminal polypeptide sequence of a chain, wherein the c-terminal polypeptide sequence is a sequence obtainable by papain digestion of an intact antibody. The Fc region may comprise a native or variant Fc sequence. Although the immunoglobulin heavy chain is pc The boundaries of the sequences may vary, but the human IgG heavy chain Fc sequence is generally defined as an amino acid residue at position Cys226 or from the approximate position pr〇230 to the carboxy terminus of the Fc sequence. Generally, two constant domains, namely a CH2 domain and a CH3 domain, and optionally a CH4 domain are included. Herein, "Fc polypeptide" means a polypeptide constituting an Fc region. The Fc polymorphism can be derived from, for example, IgG 1, IgG2, IgG3 or IgG4 subtype, IgA, IgE Any suitable immunoglobulin of IgD or IgM is obtained. In some embodiments, the Fc polypeptide comprises part or all of a wild-type hinge sequence (generally at the N-terminus). In some embodiments, the Fc polypeptide does not comprise a functional or wild type Hinge sequences. The terms "Fc receptor" and "FcR" are used to describe a receptor that binds to the Fc region of an antibody. For example, 'FcR can be a native sequence human FcR. In general, FcR is a binding IgG antibody (gamma receptor) And includes Fc (RI, Fc (RII and Fc) receptors of the RIII subclass (including allelic variants of these receptors and additional spliced forms) FcR. Fc (RII receptors include Fc (RIIA ( "Activated receptors" and Fc (RIIB ("inhibiting receptors"), Fc (RIIA and Fc (RIIB has a similar amino acid sequence' mainly differs in its cytoplasmic domain. Other isotypes of immunoglobulins may also be caused by certain FcRs Binding (see, for example, Jane Way et al, Immuno Biology. The immune system in health and disease, (Elsevier Science Ltd., NY) (4th ed., 1999)). Activated receptor Fc 143939.doc 37 · 201019961 (RIIA Immunoreceptor based on its cytoplasmic domain The immunoreceptor tyrosine-based activation motif (ITAM) inhibits receptor Fc (RIIB contains immunoreceptor tyrosine-based inhibition motifs (ITIM) in its cytoplasmic domain). (.^ idL Daeron, Annu. Rev. Immunol. 15:203-234 (1997)). FcR is reviewed in Ravetch and Kinet, .*

Immunol 9:457-92 (1991) ; Capel^A. » Immunomethods 4:25-34 (1994);及 de Haas等人,J· Lab. Clin. Med. 126:330-41 (1995)中。本文中之術語「FcR」涵蓋其他FcR,包括有待 © 於將來鑑別之FcR。該術語亦包括新生兒受艘(neonatal receptor) FcRn,該受體負責將母體IgG轉移至胎兒(Guyer 等人,《/. 117:587 (19*76);及 Kim 等人,X 24:249 (1994)) 0 如本文所使用之「鉸鏈區」、「鉸鏈序列」及其變化形式 包括此項技術中已知之含義,該含義說明於例如Jane way 等人,Immuno Biology · the immune system in health and disease, (Elsevier Science Ltd.,NY)(第 4版,1999) ; Bloom 等人,Protein Science (1997),6:407-415 ; Humphreys 等 人,J. Immunol· Methods (1997),209:193-202 中。 如本文所使用之「激動劑抗體」為模擬相關多肽(例如 HGF)之至少一種功能活性之抗體。 在整個本說明書及申請專利範圍中,免疫球蛋白重鏈中 殘基之編號均為Kabat等人,Sequences of Proteins of Immunological Interest,第 5 版,Public Health Service, 143939.doc -38- 201019961Immunol 9:457-92 (1991); Capel^A. » Immunomethods 4:25-34 (1994); and de Haas et al, J. Lab. Clin. Med. 126:330-41 (1995). The term "FcR" as used herein encompasses other FcRs, including FcR to be identified in the future. The term also includes neonatal receptor FcRn, which is responsible for the transfer of maternal IgG to the fetus (Guyer et al., /. 117:587 (19*76); and Kim et al., X 24:249). (1994)) 0 "Hinge region", "hinge sequence" and variations thereof as used herein includes meanings known in the art, which are illustrated, for example, by Jane Way et al., Immuno Biology · the immune system in health And disease, (Elsevier Science Ltd., NY) (4th ed., 1999); Bloom et al, Protein Science (1997), 6: 407-415; Humphreys et al, J. Immunol. Methods (1997), 209: 193-202. An "agonist antibody" as used herein is an antibody that mimics at least one functional activity of a related polypeptide (e.g., HGF). Throughout this specification and the scope of the patent application, the numbering of residues in the immunoglobulin heavy chain is Kabat et al., Sequences of Proteins of Immunological Interest, 5th Edition, Public Health Service, 143939.doc -38-201019961

National Institutes of Health,Bethesda,Md. (1991)中之Εϋ 索引編號,該文獻明確地以引用之方式併入本文中。 「Kabat中之EU索引」係指人類][gGi EU抗體之殘基編 號。 術語「抗體」以最廣泛意義使用且特別涵蓋單株抗體 ' (包括全長單株抗體)、多株抗體、多特異性抗體(例如雙特 異性抗體)、單價抗體、多價抗體及抗體片段,只要其展 現期望的生物活性即可。 ® 「抗體片段」僅包含完整抗體之一部分,其中該部分較 佳保留當存在於完整抗體中時通常與彼部分相關的至少一 個、較佳大部分或所有功能。在一實施例中,抗體片段包 含完整抗體之抗原結合位點,從而保留結合抗原之能力。 在另一實施例中,抗體片段(例如包含Fc區之抗體片段)保 留當存在於完整抗體中時通常與^區相關之至少一個生物 功能,諸如FcRn結合、抗體半衰期調節、ADC(:功能及補 ❿ 體結合。在一實施例中,抗體片段為活體内半衰期實質上 與完整抗體類似之單價抗體。舉例而言,該抗體片段可包 含一個與能夠賦予片段活體内穩定性之^序列連接之抗原 結合臂。在一實施例中’本發明抗體為如W〇 2〇〇5/〇63816 中所述之單臂抗體。在_實施例中,單臂抗體包含構成如National Institutes of Health, Bethesda, Md. (1991), index number, which is expressly incorporated herein by reference. "EU index in Kabat" refers to the number of residues in human [gGi EU antibodies. The term "antibody" is used in its broadest sense and specifically covers monoclonal antibodies ' (including full-length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (eg, bispecific antibodies), monovalent antibodies, multivalent antibodies, and antibody fragments. As long as it exhibits the desired biological activity. The "antibody fragment" contains only a portion of the intact antibody, wherein the portion preferably retains at least one, preferably most or all of the functions normally associated with the portion when present in the intact antibody. In one embodiment, the antibody fragment comprises the antigen binding site of the intact antibody, thereby retaining the ability to bind antigen. In another embodiment, an antibody fragment (eg, an antibody fragment comprising an Fc region) retains at least one biological function normally associated with a region when present in an intact antibody, such as FcRn binding, antibody half-life regulation, ADC (: function and In one embodiment, the antibody fragment is a monovalent antibody having a half-life in vivo substantially similar to an intact antibody. For example, the antibody fragment may comprise a sequence that is capable of conferring in vivo stability to the fragment. The antigen binding arm. In one embodiment, the antibody of the invention is a one-armed antibody as described in W〇2〇〇5/〇63816. In the embodiment, the one-armed antibody comprises a composition such as

W〇 2005/063816中所述之「杵」及「白」之Fc突變。舉例 而s,臼犬變在Fc多肽中可為T366A、L368A&/或Y4〇7V 之一或多者,空穴突變可為T366W。 「阻斷」抗體或抗體「拮抗劑」為抑制或減小所結合抗 143939.doc -39- 201019961 原之生物活性的抗體。較佳阻斷抗體或拮抗劑抗體完全抑 制抗原之生物活性。 抗體之「功能性抗原結合位點」為能夠結合無抗原之位 點。雖然抗原結合位點之抗原結合親和力不必與衍生抗原 結合位點之親本抗體一樣強,但結合抗原之能力必須可使 用多種已知用於評估抗體與抗原的結合之方法中的任—方 法來量測。此外,本文之多價抗體之各抗原結合位點的抗 原結合親和力無需在數量上相同。對於本文之多聚抗體, 功能性抗原結合位點之數目可使用如美國專利申請公開案 第20050186208號之實例2中所述之超速離心分析來評估。 根據該分析方法,組合乾抗原與多聚抗體之不同比率,且 假定不同數目之功能結合位點計算複合物之平均分子量。 比較此等理論值與所獲得之實際實驗值以評估功能結合位 點之數目。 「物種依賴性抗體」為對來自第一哺乳動物物種之抗原 之結合親和力強於對來自第二哺乳動物物種之抗原同系物 的結合親和力之抗體。雖然物種依賴性抗體通常「特異性 結合」一種人類抗原(亦即結合親和力(κ〇值不超過約丄X 1〇·7 Μ,較佳不超過約lxl0_8 M,且最佳不超過約^1〇 9 M),但對來自第二非人類哺乳動物物種之抗原同系物的結 合親和力比對該人類抗原之結合親和力弱至少約5〇倍或 至少約500倍,或至少約1000倍。物種依賴性抗體可為如 上文所定義之各種類型之抗體中的任一者。在一實施例 中’物種依賴性抗體為人類化或人類抗體。 143939.doc •40- 201019961 如本文所使用之「抗體突變體」或「抗體變異體」係指 物種依賴性抗體之胺基酸序列變異體’其中物種依賴性抗 體之一或多個胺基酸殘基已經修飾。該等突變體必然與物 種依賴性抗體具有小於100%之序列一致性或相似性。在 一實施例中,抗體突變體將具有與物種依賴性抗體之重鍵 或輕鏈可變域之胺基酸序列具有至少75%、更佳至少 80%、更佳至少85%、更佳至少90%,且最佳至少95%之胺 基酸序列一致性或相似性的胺基酸序列。對於該序列之— 致性或相似性在本文中係定義為在序列比對及必要時引人 間隙以達成最大序列一致性百分比之後,候選序列中與物 種依賴性抗體殘基一致(亦即相同殘基)或類似(亦即來自基 於共同側鏈特性之同一組的胺基酸殘基,參見下文)之胺 基酸殘基的百分比。在可變域外部的抗體序列中進行N 端、C端或内部延伸、缺失或插入均不應視為影響序列一 致性或相似性。 「嵌合VEGF受體蛋白」為具有源自至少兩種不同蛋白 質之胺基酸序列之VEGF受體分子,其中至少一種蛋白質 為VEGF受體蛋白。在某些實施例中,嵌合VEGF受體蛋白 能夠結合VEGF且抑制其生物活性。 除非另外指示,否則表述「多價抗體」在整個說明書中 用於表示包含三個或三個以上抗原結合位點之抗體。多價 抗體較佳經工程改造而具有三個或三個以上抗原結合位 點’且一般不為天然序列IgM或IgA抗體。 「Fv」片段為含有完整抗原識別及結合位點之抗體片 143939.doc -41 · 201019961 段。該區域係由緊密締合之一個重鏈可變域與一個輕鏈可 變域的二聚體組成,該締合在性質上可為共價的,例如在 scFv中。在該組態中,各可變域之三個CDR相互作用以界 定Vh-Vl二聚體表面上之抗原結合位點。總體而言,六個 CDR或其子集對抗體賦予抗原結合特異性。然而,即使單 個可變域(或僅包含三個抗原特異性CDR的一半Fv)亦具有 識別且結合抗原之能力,但其親和力通常低於整個結合位 點。 如本文所使用之「抗體可變域」係指抗體分子之輕鏈及 重鏈之部分,包括互補決定區(CDR ;亦即CDR1、CDR2及 CDR3)及構架區(FR)之胺基酸序列。VH係指重鏈之可變 域。VL係指輕鏈之可變域。根據本發明中所使用之方法, 可根據 Kabat(Sequences of Proteins of Immunological Interest(National Institutes of Health, Bethesda, Md., 1987 及1991))來定義指定給CDR及FR之胺基酸位置。抗體或抗 原結合片段之胺基酸編號亦係根據Kabat之編號。 如本文所使用之術語「互補決定區」(CDR ;亦即 CDR1、CDR2及CDR3)係指抗體可變域中對抗原結合而言 必需存在之胺基酸殘基。各可變域通常具有三個CDR區, 經鑑別為CDR1、CDR2及CDR3。各互補決定區可包含來 自如Kabat所定義之「互補決定區」的胺基酸殘基(亦即輕 鏈可變域中約殘基24-34 (L1)、50-56 (L2)及89-97 (L3), 及重鏈可變域中 31-35 (HI)、50-65 (H2)及 95-102 (H3); Kabat 等人,Sequences of Proteins of Immunological 143939.doc -42· 201019961 TVziereW,第 5版,Public Health Service, National Institutes of Health,Bethesda,MD. (1991))及/或來自「高變環」之 殘基(亦即輕鏈可變域中約殘基26-32 (L1)、50-52 (L2)及 91-96 (L3),及重鏈可變域中 26-32 (HI)、53-55 (H2)及 96-101 (H3) ; Chothia 及 Lesk,·/. Mo/· 5/〇/. 196:901-917 (1987))。在一些情況下,互補決定區可包括來自根據 Kabat所定義之CDR區與高變環之胺基酸。舉例而言,抗 體4D5之重鏈之CDRH1包括胺基酸26至35。 「構架區」(下文稱為FR)為除CDR殘基以外的可變域殘 基。各可變域通常具有四個FR,經鑑別為FR1、FR2、FR3 及FR4。若根據Kabat定義CDR,則輕鏈FR殘基位於約殘基 1-23 (LCFR1)、35-49 (LCFR2)、57-88 (LCFR3)及 98-107 (LCFR4)處,且重鏈FR殘基位於重鏈殘基中約殘基1-30 (HCFR1)、36-49 (HCFR2)、66-94 (HCFR3)及 103-113 (HCFR4)處。若CDR包含來自高變環之胺基酸殘基,則輕 鏈FR殘基位於輕鏈中約殘基1-25 (LCFR1)、33-49 (LCFR2)、53-90 (LCFR3)及 97-107 (LCFR4)處,且重鏈 FR 殘基位於重鏈殘基中約殘基1-25 (HCFR.1)、33-52 (HCFR2)、56-95 (HCFR3)及 102-113 (HCFR4)處。在一些 情況下,當CDR包含來自如Kabat所定義之CDR與高變環 之胺基酸時,FR殘基應作相應調整。舉例而言,當 CDRH1包括胺基酸H26-H35時,重鏈FR1殘基位於位置1-25處,且FR2殘基位於么置36-49處。 「Fab」片段含有輕鏈可變域及恆定域,及重鏈可變域 143939.doc -43- 201019961 及第一恆定域(CH1)。F(ab')2抗體片段包含一對Fab片段, 該對Fab片段通常藉由其間之鉸鏈半胱胺酸共價連接於接 近其羧基端處。此項技術亦已知抗體片段之其他化學偶 合。 「單鏈Fv」或「scFv」抗體片段包含抗體之VH及VL域, 其中此等域存在於單個多肽鏈中。Fv多肽一般在VH與VL 域之間另外包含多肽連接子,該連接子使得scFv能夠形成抗 原結合所期望的結構。關於scFv之综述,參見Pluckthun, The Pharmacology of Monoclonal Antibodies,第]Λ3 卷, Rosenburg及Moore編,Springer-Verlag, New York,第 269-315 頁(1994)。 術語「雙功能抗體」係指具有兩個抗原結合位點的小抗 體片段,該等片段包含與同一多肽鏈(VH及VL)中之輕鏈可 變域(vL)連接的重鏈可變域(VH)。藉由使用由於過短而無 法在同一鏈上的兩個域之間配對的連接子,迫使該等域與 另一鏈上之互補域配對且產生兩個抗原結合位點。雙功能 抗體更詳細描述於例如EP 404,097 ; WO 93/11161 ;及 .Hollinger等人,Proc. iVai/· Sci. USA, 90 6444-6448 (1993)中。 表述「線性抗體」係指如Zapata等人,/Voieiw 8(10):1057-1062 (1995)中所述之抗體。簡言之,該等抗體 包含一對串聯Fd區段(VH-CH1-VH-CH1),其與互補輕鏈多 肽一起形成一對抗原結合區。線性抗體可具有雙特異性或 單特異性。 143939.doc -44- 201019961 修飾語「單株」表示由實質上均質抗體群體獲得之抗體 特徵,且不應理解為需要藉由任何特定方法產生抗體。舉 例而言,欲根據本發明使用之單株抗體可利用多種技術製 備,該等技術包括例如融合瘤方法(例如,Kohler及 Milstein, Nature, 256:495-97 (1975) ; Hongo 等人,The Fc mutations of "杵" and "白" described in W〇 2005/063816. For example, the canine can be one or more of T366A, L368A&/ or Y4〇7V in the Fc polypeptide, and the hole mutation can be T366W. A "blocking" antibody or antibody "antagonist" is an antibody that inhibits or reduces the biological activity of the original antibody against 143939.doc-39-201019961. Preferably, the blocking antibody or antagonist antibody completely inhibits the biological activity of the antigen. The "functional antigen binding site" of an antibody is a site capable of binding to an antigen-free site. Although the antigen binding affinity of the antigen binding site does not have to be as strong as the parent antibody from which the antigen binding site is derived, the ability to bind the antigen must be able to use any of the methods known for assessing the binding of the antibody to the antigen. Measure. Furthermore, the antigen binding affinities of the various antigen binding sites of the multivalent antibodies herein need not be identical in number. For the multimeric antibodies herein, the number of functional antigen binding sites can be assessed using ultracentrifugation analysis as described in Example 2 of U.S. Patent Application Publication No. 20050186208. According to this analytical method, different ratios of dry antigen to multimeric antibody are combined, and assuming a different number of functional binding sites to calculate the average molecular weight of the complex. These theoretical values are compared to the actual experimental values obtained to assess the number of functional binding sites. A "species-dependent antibody" is an antibody that binds to an antigen from a first mammalian species more strongly than to an antigenic homologue from a second mammalian species. Although species-dependent antibodies typically "specifically bind" to a human antigen (ie, binding affinity (a value of κ〇 does not exceed about 〇X 1〇·7 Μ, preferably does not exceed about lxl0_8 M, and optimally does not exceed about ^1) 〇 9 M), but the binding affinity for an antigen homolog from a second non-human mammalian species is at least about 5 fold or at least about 500 fold, or at least about 1000 fold weaker than the binding affinity for the human antigen. A sex antibody can be any of the various types of antibodies as defined above. In one embodiment, the 'species-dependent antibody is a humanized or human antibody. 143939.doc • 40- 201019961 "Antibody" as used herein "Mutant" or "antibody variant" refers to an amino acid sequence variant of a species-dependent antibody in which one or more amino acid residues of a species-dependent antibody have been modified. These mutants are necessarily species dependent The antibody has a sequence identity or similarity of less than 100%. In one embodiment, the antibody mutant will have at least 75% of the amino acid sequence of the heavy or light chain variable domain of the species dependent antibody. More preferably an amino acid sequence of at least 80%, more preferably at least 85%, more preferably at least 90%, and most preferably at least 95% amino acid sequence identity or similarity. For the sequence or similarity In this context, it is defined that after sequence alignment and, if necessary, introducing a gap to achieve a maximum sequence identity percentage, the candidate sequence is identical (ie the same residue) or similar (ie from a The percentage of amino acid residues of the same group of amino acid residues of the common side chain character, see below). N-terminal, C-terminal or internal extension, deletion or insertion in the antibody sequence outside the variable domain It should be considered to affect sequence identity or similarity. A "chimeric VEGF receptor protein" is a VEGF receptor molecule having an amino acid sequence derived from at least two different proteins, at least one of which is a VEGF receptor protein. In certain embodiments, a chimeric VEGF receptor protein is capable of binding to VEGF and inhibiting its biological activity. Unless otherwise indicated, the expression "multivalent antibody" is used throughout the specification to indicate that it comprises three or more An antibody to the original binding site. The multivalent antibody is preferably engineered to have three or more antigen binding sites' and is generally not a native sequence IgM or IgA antibody. The "Fv" fragment is a complete antigen recognition and binding. Site antibody fragment 143939.doc -41 · 201019961. This region consists of a tightly associated heavy chain variable domain and a light chain variable domain dimer, which can be a total of Valuable, for example in scFv. In this configuration, the three CDRs of each variable domain interact to define an antigen binding site on the surface of the Vh-V1 dimer. Overall, six CDRs or their sub- The set confers antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three antigen-specific CDRs) has the ability to recognize and bind antigen, but its affinity is generally lower than the entire binding site. As used herein, "antibody variable domain" refers to a portion of the light and heavy chains of an antibody molecule, including the complementarity determining regions (CDRs; ie, CDR1, CDR2 and CDR3) and the amino acid sequence of the framework region (FR). . VH refers to the variable domain of the heavy chain. VL refers to the variable domain of the light chain. According to the method used in the present invention, the position of the amino acid assigned to the CDR and FR can be defined according to Kabat (Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991)). The amino acid number of the antibody or antigen-binding fragment is also numbered according to Kabat. The term "complementarity determining region" (CDR; i.e., CDR1, CDR2, and CDR3) as used herein refers to an amino acid residue that is necessary for antigen binding in an antibody variable domain. Each variable domain typically has three CDR regions identified as CDR1, CDR2 and CDR3. Each of the complementarity determining regions may comprise an amino acid residue from a "complementarity determining region" as defined by Kabat (i.e., about residues 24-34 (L1), 50-56 (L2) and 89 in the light chain variable domain. -97 (L3), and heavy chain variable domains 31-35 (HI), 50-65 (H2), and 95-102 (H3); Kabat et al., Sequences of Proteins of Immunological 143939.doc -42· 201019961 TVziereW, 5th Edition, Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) and/or residues from the "hypervariable loop" (ie, about 26-32 residues in the light chain variable domain) (L1), 50-52 (L2) and 91-96 (L3), and heavy chain variable domains 26-32 (HI), 53-55 (H2) and 96-101 (H3); Chothia and Lesk, ·/. Mo/· 5/〇/. 196:901-917 (1987)). In some cases, the complementarity determining region can include an amino acid from a CDR region and a hypervariable loop as defined by Kabat. For example, the CDRH1 of the heavy chain of antibody 4D5 includes amino acids 26 to 35. A "framework region" (hereinafter referred to as FR) is a variable domain residue other than a CDR residue. Each variable domain typically has four FRs identified as FR1, FR2, FR3, and FR4. If the CDRs are defined according to Kabat, the light chain FR residues are located at about residues 1-23 (LCFR1), 35-49 (LCFR2), 57-88 (LCFR3), and 98-107 (LCFR4), and the heavy chain FR remains The base is located at about residues 1-30 (HCFR1), 36-49 (HCFR2), 66-94 (HCFR3), and 103-113 (HCFR4) in the heavy chain residue. If the CDR comprises an amino acid residue from a hypervariable loop, the light chain FR residue is located in the light chain at residues 1-25 (LCFR1), 33-49 (LCFR2), 53-90 (LCFR3) and 97- 107 (LCFR4), and the heavy chain FR residue is located in the heavy chain residue about residues 1-25 (HCFR.1), 33-52 (HCFR2), 56-95 (HCFR3), and 102-113 (HCFR4) At the office. In some cases, when the CDRs comprise an amino acid from a CDR and a hypervariable ring as defined by Kabat, the FR residue should be adjusted accordingly. For example, when CDRH1 comprises the amino acid H26-H35, the heavy chain FR1 residue is at positions 1-25 and the FR2 residue is at position 36-49. The "Fab" fragment contains a light chain variable domain and a constant domain, and a heavy chain variable domain 143939.doc -43 - 201019961 and a first constant domain (CH1). The F(ab')2 antibody fragment comprises a pair of Fab fragments which are typically covalently linked to their carboxy terminus by a hinged cysteine in between. Other chemical couplings of antibody fragments are also known in the art. A "single-chain Fv" or "scFv" antibody fragment comprises the VH and VL domains of an antibody, wherein such domains are present in a single polypeptide chain. Fv polypeptides typically additionally comprise a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding. For a review of scFv, see Pluckthun, The Pharmacology of Monoclonal Antibodies, vol. 3, ed. Rosenburg and Moore, Springer-Verlag, New York, pp. 269-315 (1994). The term "bifunctional antibody" refers to a small antibody fragment having two antigen binding sites comprising a heavy chain variable domain linked to a light chain variable domain (vL) in the same polypeptide chain (VH and VL). (VH). By using a linker that is too short to be paired between two domains on the same strand, the domains are forced to pair with complementary domains on the other strand and create two antigen-binding sites. Bifunctional antibodies are described in more detail in, for example, EP 404,097; WO 93/11161; and Hollinger et al, Proc. iVai/. Sci. USA, 90 6444-6448 (1993). The expression "linear antibody" refers to an antibody as described in Zapata et al., /Voieiw 8(10): 1057-1062 (1995). Briefly, the antibodies comprise a pair of tandem Fd segments (VH-CH1-VH-CH1) which together with the complementary light chain polypeptide form a pair of antigen binding regions. Linear antibodies can have bispecific or monospecificity. 143939.doc -44- 201019961 The modifier "single plant" refers to the characteristics of an antibody obtained from a substantially homogeneous population of antibodies and is not to be construed as requiring production of the antibody by any particular method. For example, monoclonal antibodies to be used in accordance with the present invention can be prepared using a variety of techniques including, for example, fusion knob methods (e.g., Kohler and Milstein, Nature, 256:495-97 (1975); Hongo et al.

Hybridoma, 14 (3):253-260 (1995) ; Harlow 等人,Hybridoma, 14 (3): 253-260 (1995); Harlow et al.

Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press,第 2 版,1988) ; Hammerling 等人, Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N_Y.,1981))、重組DNA方法(參見例如美國專利 第4,816,567號)、噬菌體呈現技術(參見例如Clackson等 人,352:624-628 (1991) ; Marks等人,·/· Μο/. Βίο/· 222:581-597 (1992) ; Sidhu等人,《/· Mo/·扪〇/· 338(2):299-310 (2004) ; Lee 等人,《/. Mo/·价〇/. 340(5):1073-1093 (2004) ; Fellouse, Proc. Natl. Acad. Sci. USA 101(34):12467-12472 (2004);及 Lee 等人,《/./所//21^〇/.^/6从〇心 284(1-2):119-132 (2004)),及在動物中產生具有部分或所有編碼 人類免疫球蛋白序列之人類免疫球蛋白基因座或基因的人 類或人類樣抗體之技術(參見例如WO 1998/24893 ; WO 1996/34096 ; WO 1996/33735 ; WO 1991/10741 ; Jakobovits 等人,户roc· ΛΓαί/. 90:2551 (1993);Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd edition, 1988); Hammerling et al, Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N_Y., 1981)), recombinant DNA methods (see for example U.S. Patent No. 4,816,567, phage display technology (see, e.g., Clackson et al, 352: 624-628 (1991); Marks et al., / / Μ ο ο ο ο ο ο ο ο ο ο ο ο ο ο ο ο Person, "/· Mo/·扪〇/· 338(2): 299-310 (2004); Lee et al., "/. Mo/. Price 〇/. 340(5): 1073-1093 (2004); Fellouse, Proc. Natl. Acad. Sci. USA 101(34): 12467-12472 (2004); and Lee et al., /////21^〇/.^/6 from 〇心284(1- 2): 119-132 (2004)), and techniques for producing human or human-like antibodies having partial or all human immunoglobulin loci or genes encoding human immunoglobulin sequences in animals (see, for example, WO 1998/24893 WO 1996/34096; WO 1996/33735; WO 1991/10741 ; Jakobovits et al., household roc· ΛΓαί/. 90:2551 (1993);

Jakobovits^ Λ. ? Nature 362:255-258 (1993) ; Bruggemann 等人,Fear ζ·« 7:33 (1993);美國專利第 5,545,807 號;第 5,545,806號;第 5,569,825號;第 5,625,126號;第 143939.doc -45- 201019961 5,633,425號;及第 5,661,016號;Marks等人,价-o/rec/mo/o幻; 10:779-783 (1992) ; Lonberg 等人,368:856-859 (1994) ; Morrison,iVaiMre 368:812-813 (1994) ; Fishwild等 人,Nature Biotechnol. 14:845-851 (1996) ; Neuberger, Nature Biotechnol. 14:826 (1996);及 Lonberg 及 Huszar, 13:65-93 (1995))。 本文中之單株抗體特別包括「嵌合」抗體,其中重鏈及/ 或輕鏈之一部分與自特定物種衍生或屬於特定抗體類別或 子類之抗體中的相應序列一致或同源,而鏈之其餘部分與 自另一物種衍生或屬於另一抗體類別或子類之抗體中的相 應序列一致或同源;以及該等抗體之片段,只要其展現期 望的生物活性即可(參見例如美國專利第4,816,567號;及 Morrison 等人,Ρτ-oc. Natl. Acad. Sci. USA 81:6851-6855 (1984))。嵌合抗體包括PRIMATIZED®抗體,其中抗體之 抗原結合區係衍生自藉由例如用相關抗原使獼猴(macaque monkey)免疫而產生之抗體。 非人類(例如鼠類)抗體之「人類化」形式為含有自非人 類免疫球蛋白衍生之最小序列的嵌合抗體。人類化抗體主 要為人類免疫球蛋白(接受者抗體(recipient antibody))’其 中來自接受者之高變區(hypervariable region)的殘基經來 自諸如小鼠、大鼠、兔或非人類靈長類動物之非人類物種 (供給者抗體(donor antibody))之高變區的具有期望特異 性、親和力及能力之殘基置換。在一些情況下,人類免疫 球蛋白之Fv構架區(FR)殘基經相應非人類殘基置換。此 143939.doc -46- 201019961 外,人類化抗體可包含接受者抗體或供給者抗體中未見之 殘基。進行此等修飾以進一步改進抗體效能。一般而言, 人類化抗體應包含實質上全部的至少一個,通常兩個可變 域’其中高變環全部或實質上全部對應於非人類免疫球蛋 白之高變環’且FR區全部或實質上全部為人類免疫球蛋白 序列之FR區。人類化抗體視情況亦將包含免疫球蛋白悝定 區(Fc)(通常為人類免疫球蛋白恆定區)之至少一部分。關 於更多詳情,參見 Jones等人,321:522-525 (1986); Riechmann等人,Waiwre 332:323-329 (1988) ; &Presta,Cwr. (9/7. «Sirwci. 2:593-596 (1992) ° 「人類抗體」為具有對應於由人類產生之抗體的胺基酸 序列之胺基酸序列及/或已使用任何如本文所揭示之製備 人類抗體之技術所製備的抗體。此人類抗體之定義特別排 除包含非人類抗原結合殘基之人類化抗體。人類抗體可使 用此項技術中已知之各種技術產生。在一實施例中,人類 抗體係選自嗟菌體文庫’其中該嗤菌體文庫表現人類抗體 (Vaughan^ A » Nature Biotechnology 14:309-314 (1996);Jakobovits^ Λ. ? Nature 362:255-258 (1993); Bruggemann et al., Fear ζ·« 7:33 (1993); US Patent No. 5,545,807; 5,545,806; 5,569,825; 5,625,126; .doc -45-201019961 5,633,425; and 5,661,016; Marks et al., price -o/rec/mo/o illusion; 10:779-783 (1992); Lonberg et al., 368:856-859 ( 1994); Morrison, iVaiMre 368:812-813 (1994); Fishwild et al, Nature Biotechnol. 14:845-851 (1996); Neuberger, Nature Biotechnol. 14:826 (1996); and Lonberg and Huszar, 13: 65-93 (1995)). The monoclonal antibodies herein specifically include "chimeric" antibodies in which one of the heavy and/or light chains is identical or homologous to the corresponding sequence in an antibody derived from a particular species or belonging to a particular antibody class or subclass, and the chain The remainder is identical or homologous to the corresponding sequence in an antibody derived from another species or belonging to another antibody class or subclass; and fragments of such antibodies as long as they exhibit the desired biological activity (see, for example, US patents) No. 4,816,567; and Morrison et al., Ρτ-oc. Natl. Acad. Sci. USA 81:6851-6855 (1984)). Chimeric antibodies include PRIMATIZED® antibodies wherein the antigen binding region of the antibody is derived from an antibody produced by, for example, immunizing a macaque monkey with a related antigen. The "humanized" form of a non-human (e.g., murine) antibody is a chimeric antibody containing minimal sequence derived from a non-human immunoglobulin. Humanized antibodies are primarily human immunoglobulins (recipient antibodies) where residues from the recipient's hypervariable region are derived from, for example, mice, rats, rabbits or non-human primates. Residue replacement of the hypervariable region of an animal's non-human species (donor antibody) with the desired specificity, affinity, and ability. In some instances, the Fv framework region (FR) residues of human immunoglobulin are replaced by corresponding non-human residues. In addition to 143939.doc -46-201019961, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further improve antibody potency. In general, a humanized antibody should comprise substantially all of at least one, usually two variable domains 'where the hypervariable loops wholly or substantially all correspond to the hypervariable loop of the non-human immunoglobulin' and the FR regions are all or substantially All of them are FR regions of human immunoglobulin sequences. The humanized antibody will also optionally comprise at least a portion of an immunoglobulin binding region (Fc), typically a human immunoglobulin constant region. For more details, see Jones et al., 321:522-525 (1986); Riechmann et al., Waiwre 332:323-329 (1988); &Presta, Cwr. (9/7. «Sirwci. 2:593 -596 (1992) ° "Human antibody" is an amino acid sequence having an amino acid sequence corresponding to an antibody produced by a human and/or an antibody which has been prepared using any of the techniques for preparing a human antibody as disclosed herein. The definition of such human antibodies specifically excludes humanized antibodies comprising non-human antigen-binding residues. Human antibodies can be produced using various techniques known in the art. In one embodiment, the human anti-system is selected from the group of bacteria. The bacillus library exhibits human antibodies (Vaughan^ A » Nature Biotechnology 14:309-314 (1996);

Sheets等人,iVa". 心/. 95:6157-6162 (1998);Sheets et al., iVa". Heart/. 95:6157-6162 (1998);

Hoogenboom 及 Winter, «/. Mol. Biol., 227:381 (1991); Marks等人,《/· M〇/· 5ζ·ο/·,222:581 (1991))。人類抗體亦可 藉由向例如内源免疫球蛋白基因已部分或完全失活之小鼠 之轉殖基因動物中引入人類免疫球蛋白基因座而製備。激 發後’觀察到人類抗體產生,其在包括基因重排、裝配及 抗體譜系之各方面均與人類中所見非常類似。該方法描述 143939.doc •47- 201019961 於例如美國專利第5,545,807號、第5,545,806號、第 5,569,825號、第 5,625,126號、第 5,633,425號、第 5,661,016 號及以下科技出版物中:Marks等人,5io/TecA«o/6>幻; 10:779-783 (1992) ; Lonberg 等人,Nature 368:856-859 (1994) ; Morrison,368:812-13 (1994) ; Fishwild等 * 尺,Nature Biotechnology 14:845-51 (1996) » Neuberger, : JVaiwre 14:826 (1996) ; Lonberg 及 Huszar, 13:65-93 (1995)。或者,人類抗體可 經由產生針對靶抗原之抗體之人類B淋巴細胞的永生化來 ® 製備(該等B淋巴細胞可自個體回收或可能已經活體外免 疫)。參見例如 Cole 等人 ’Monoclonal Antibodies and Cancer Therapy, Alan R. Liss,第 77 頁(1985) ; Boerner 等 尺,J. Immunol·,147 (1):86-95 (1991);及美國專利第 5,750,373號 ° 「裸抗體」為未結合異源分子(諸如細胞毒性部分或放 射性標記)之抗體。 「親和力成熟」抗體為在一或多個CDR中具有一或多種 變化之抗體,與不具有該(等)變化之親本抗體相比,該 (等)變化可改良抗體對抗原之親和力。較佳之親和力成熟 抗體將對靶抗原具有奈莫耳(nanomolar)或甚至皮莫耳 (picomolar)親和力。親和力成熟抗體係利用此項技術中已 知之程序產生。Marks等人 ’ Bio/Technology 10:779-783 (1992)描述藉由VH及VL域改組(shuffling)達成之親和力成 熟。CDR及/或構架殘基之隨機突變誘發描述於Barbas等 143939.doc • 48- 201019961 A » Proc Nat. Acad. Sci, USA 91:3809-3813 (1994) ; Schier 等人,Gene 169:147-155 (1995) ; Yelton等人,《/· /wwwwo/· 155:1994-2004 (1995) ; Jackson等人,·/. 154(7):3310-9 (1995);及 Hawkins等人,乂 Mi>/· 5zo/· 226:889-896 (1992) 中o 具有指定抗體之「生物特徵」之抗體為具有該抗體之一 或多個區別於結合相同抗原之其他抗體的生物特徵之抗 體。 為篩選結合與相關抗體結合之抗原上之抗原決定基的抗 體,可執行常規交叉阻斷檢定,諸如Antibodies,A Laboratory Manual, Cold Spring Harbor Laboratory, Harlow 及David Lane編(1988)中所述之檢定。Hoogenboom and Winter, «/. Mol. Biol., 227:381 (1991); Marks et al., "/· M〇/· 5ζ·ο/·, 222:581 (1991)). Human antibodies can also be prepared by introducing a human immunoglobulin locus into a transgenic animal, such as a mouse in which the endogenous immunoglobulin gene has been partially or completely inactivated. Human antibody production was observed after stimulation, which is very similar to that seen in humans in all aspects including gene rearrangement, assembly, and antibody lineage. The method is described in, for example, U.S. Patent Nos. 5,545,807, 5,545,806, 5,569,825, 5,625,126, 5,633,425, 5,661,016 and below: Marks et al. 5io/TecA«o/6>Magic; 10:779-783 (1992); Lonberg et al, Nature 368:856-859 (1994); Morrison, 368:812-13 (1994); Fishwild et al. Biotechnology 14: 845-51 (1996) » Neuberger, JVaiwre 14:826 (1996); Lonberg and Huszar, 13:65-93 (1995). Alternatively, human antibodies can be prepared by immortalization of human B lymphocytes that produce antibodies against the target antigen (the B lymphocytes can be recovered from the individual or may have been immunized in vitro). See, for example, Cole et al. 'Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, page 77 (1985); Boerner et al, J. Immunol, 147 (1): 86-95 (1991); and U.S. Patent 5,750,373 No. "Naked antibody" is an antibody that does not bind to a heterologous molecule, such as a cytotoxic moiety or a radioactive label. "Affinity maturation" antibodies are antibodies that have one or more changes in one or more CDRs that alter the affinity of the antibody for the antigen as compared to a parent antibody that does not have this (equal) change. Preferred affinity matured antibodies will have nanomolar or even picomolar affinity for the target antigen. Affinity mature resistance systems are generated using procedures known in the art. Marks et al., Bio/Technology 10:779-783 (1992) describe affinity maturity achieved by VH and VL domain shuffling. Random mutation induction of CDR and/or framework residues is described in Barbas et al. 143939.doc • 48-201019961 A » Proc Nat. Acad. Sci, USA 91:3809-3813 (1994); Schier et al., Gene 169:147- 155 (1995); Yelton et al., ///wwwwo/. 155:1994-2004 (1995); Jackson et al., /. 154(7):3310-9 (1995); and Hawkins et al. Mi>/· 5zo/· 226:889-896 (1992) The antibody having the "biological characteristics" of the specified antibody is an antibody having one or more of the antibodies which are different from the biological characteristics of other antibodies that bind to the same antigen. To screen for antibodies that bind to an epitope on an antigen that binds to the relevant antibody, routine cross-blocking assays can be performed, such as those described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Harlow, and David Lane (1988). .

為增加含有本發明之胺基酸序列的抗體或多肽之半衰 期,可如(例如)美國專利5,739,277中所述,使救助受體 (salvage receptor)結合抗原決定基與抗體(尤其抗體片段) 連接。舉例而言,編碼救助受體結合抗原決定基之核酸分 子可與編碼本發明多肽序列之核酸同框連接,以致經工程 改造之核酸分子所表現之融合蛋白包含救助受體結合抗原 決定基及本發明之多肽序列》如本文所使用之術語「救助 受體結合抗原決定基」係指IgG分子(例如IgG1、IgG2、 IgG3或IgG4)之Fc區中負責增加IgG分子之活體内血清半衰 期之抗原決定基(例如Ghetie等人,Rev. Immunol. 18:739-766 (2000),表1)。以下文獻中亦描述Fe區中具有 取代且血清半衰期增加之抗體:WO 00/42072 ; WO 143939.doc • 49- 201019961 02/060919 ; Shields 等人,J. Biol. Chem. 276:6591-6604 (2001) Hinton, J. Biol. Chem. 279:6213-6216 (2004)。在 另一實施例中,亦可例如藉由連接其他多肽序列來增加血 清半衰期。舉例而言,適用於本發明方法之抗體或其他多 肽可連接血清白蛋白或血清白蛋白中結合FcRn受體之部分 或血清白蛋白結合肽,以致血清白蛋白結合該抗體或多 肽,該等多肽序列例如揭示於WO 01/45746中。在一較佳 實施例中,待連接之血清白蛋白肽包含胺基酸序列 DICLPRWGCLW(SEQ ID NO: 32)。在另一實施例中,Fab 之半衰期係利用此等方法增加。關於血清白蛋白結合肽序 歹1J ,亦參見Dennis等人,J. Biol. Chem. 277:35035-35043 (2002) 。 「經分離」多肽或「經分離」抗體為已經鑑別且自自然 環境之組分中分離及/或回收之抗體。其自然環境之污染 組分為會干擾多肽或抗體之診斷或治療用途的物質,且可 包括酶、激素及其他蛋白或非蛋白溶質。在較佳實施例 中,多肽或抗體將純化至:(1)如勞立法(Lowry method)所 測定,多肽或抗體佔95重量%以上且最佳99重量%以上; (2)藉由使用旋杯式測序儀足以獲得N端或内部胺基酸序列 之至少15個殘基的程度;或(3)由在還原或非還原條件下使 用考馬斯藍(Coomassie blue)或較佳使用銀染色(silver stain)之SDS-PAGE鎩定為均質(homogeneity)。由於多肽自 然環境之至少一種組分將不存在,故經分離多肽或抗體包 括原位(in situ)處於重組細胞内之多肽或抗體。然而,通 143939.doc -50- 201019961 常將藉由至少一個純化步驟來製備經分離多肽或抗體。 「片段」意謂多肽或核酸分子之一部分,其較佳含有參 考核酸分子或多肽之全長之至少10%、20%、30%、40%、 50%、60%、70%、80%、90〇/〇、95°/〇或更長。片段可含有 10、20、30、40、50、60、70、80、90 或 1〇〇、200、 300、400、500、600個或更多核苷酸,或 1〇、2〇、3〇、 40 、 50 、 60 、 70 、 80 、 90 、 1〇〇 、 120 、 140 、 160 、 180 、 190、200個或更多胺基酸。 治療」係指治療性處理與預防措施。需要治療者包括 已患良性、癌前期或非轉移性腫瘤者,以及需預防癌症發 生或復發者。 術語「治療有效量」係指治療劑治療或預防哺乳動物之 疾病或病症之量。在癌症情況下,治療劑之治療有效量可 減少癌細胞數目;減小原發性腫瘤大小;抑制(亦即在一 疋程度上減緩且較佳終止)癌細胞浸潤至周邊器官中;抑 φ 制(亦即在一定程度上減緩且較佳終止)腫瘤轉移;在一定 程度上抑制腫瘤生長;及/或在一定程度上減輕一或多種 與病症相關之症狀。就藥物可阻止生長及/或殺死現有癌 細胞而言,其可抑制細胞生長及/或具有細胞毒性。對於 癌症療法,活體内功效可例如藉由評估存活持續時間、疾 病進展時間(TTP)、反應率(RR)、反應持續時間及/或生活 品質來量測。 術語「癌症」及「癌性」仙或描述哺㈣物中通常以 不受調控之細胞生長為特徵之生理狀況。該定義中包括良 I43939.doc •51- 201019961 ^•生癌症及惡性癌症^ Γ早期癌症」或Γ早期腫瘤」意謂不 具侵襲性或轉移性之癌症,或分類為0期、I期或η期癌症 之癌症°癌症之實例包括(但不限於)癌瘤、淋巴瘤、胚細 胞瘤(包括神經管胚細胞瘤及視網膜胚細胞瘤)、肉瘤(包括 脂肪肉瘤及滑膜細胞肉瘤)、神經内分泌腫瘤(包括類癌腫 瘤、胃泌素瘤及胰島細胞癌)、間皮瘤、神經鞘瘤(包括聽 神紐瘤)、脊膜瘤、腺癌、黑素瘤及白血病或淋巴惡性疾 病。該等癌症之更特定實例包括鱗狀細胞癌(例如上皮鱗 狀細胞癌)、肺癌(包括小細胞肺癌(SCLC)、非小細胞肺癌 (NSCLC)、肺腺癌及肺鱗狀細胞癌)、腹膜癌、肝細胞癌、 月癌(包括胃腸癌)、胰腺癌、神經膠母細胞瘤、子宮頸 癌、卵巢癌、肝癌〇iver cancer)、膀胱癌、肝細胞瘤 (hepatoma)、乳房癌(包括轉移性乳房癌)、結腸癌、直腸 癌、結腸直腸癌、子宮内膜或子宮癌、唾液腺癌、腎癌、前 列腺癌、陰門癌、曱狀腺癌、肝癌㈣心⑽in_)、肛 門癌㉟癌、睪丸癌、食道癌、膽道腫瘤以及頭頸部 癌0 術語「癌前期(pre_cancer〇us)」係指通常在癌症之前或 發展成癌症之病狀或生長。「癌前期」生長將具有以異常 細胞週期調控、增殖或分化為特徵之細胞,其可用細胞週 期調控、細胞增殖或分化之指標確定。 厂 發育 厂 發育不良」意謂組織、器官赤 益S或細胞之任何異常生長或 。發育不良較佳為高等級或癌前期。 轉移」 意謂癌症自其原發位 點擴散至身體之其他位 143939.doc -52· 201019961 置。癌細胞可脫離原發性腫瘤,滲透至淋巴及血管中,經 由血流循環且在身體其他部位之正常組織中的遠位病灶中 生長(轉移)。轉移可為局部的或遠位的。轉移為連續過 程,按序發生腫瘤細胞脫離原發性腫瘤,經由血流行進, 且停留在某一遠處位點。在該新位點處,細胞建立血液供 應且會生長形成威脅生命的物質。 腫瘤細胞内之刺激性與抑制性分子路徑調控該行為且 遠處位點中腫瘤細胞與宿主細胞之間的相互作用亦很明 顯。 「非轉移性」意謂癌症為良性的,或停留在原發性位點 處且未滲透至淋巴或血管系統中或除原發性位點以外之組 織中。非轉移性癌症一般為屬於0期、J期或π期癌症及有 時III期癌症之任何癌症。 「原發性腫瘤」或「原發性癌症」意謂最初癌症且不意 謂位於個體體内之另一組織、器官或位置中之轉移性病 變。 「良性腫瘤」或「良性癌症」意謂仍位於起源位點處且 不具有浸潤、侵襲或轉移至遠處位點之能力的腫瘤。 「腫瘤負擔(tumor burden)」意謂體内癌細胞之數目、 腫瘤大小或癌症之量。腫瘤負擔亦稱為腫瘤負荷(加以的 load)。 「腫瘤數目」意謂腫瘤之數目。 「個體」意謂哺乳動物,包括(但不限於)人類或非人類 哺乳動物,諸如牛、馬、犬、羊或猶^個體較佳為人類。 143939.doc -53- 201019961 術語「抗癌療法」係指適用於治療癌症之療法。抗癌治 療劑之實例包括(但不限於)例如化學治療劑、生長抑制 劑、細胞毒性劑、放射療法中所用之藥劑、抗血管生成 劑、細胞凋亡劑、抗微管蛋白劑及治療癌症之其他藥劑、 抗CD20抗體、血小板衍生之生長因子抑制劑(例如 Gleevec™(甲磺酸伊馬替尼))、COX-2抑制劑(例如塞内昔 布(celecoxib))、干擾素、細胞激素、與一或多種以下標乾 ErbB2、ErbB3、ErbB4、PDGFR-β、BlyS、APRIL、 BCMA或VEGF受體、TRAIL/Apo2結合之拮抗劑(例如中和 抗體),及其他生物活性劑及有機化學劑等。本發明中亦 包括其組合。 如本文所使用之術語「細胞毒性劑」係指一種抑制或阻 止細胞功能及/或引起細胞破壞的物質。該術語意欲包括 放射性同位素(例如,I131、I125、Y9G及Re186)、化學治療劑 及毒素(諸如細菌、真菌、植物或動物來源之酶促活性毒 素或其片段)。 「化學治療劑」為適用於治療癌症之化合物。化學治療 劑之實例包括適用於治療癌症之化合物。化學治療劑之實 例包括:烷化劑,諸如塞替派(thiotepa)及CYTOXAN®環 磷醯胺;烷基磺酸鹽類,諸如白消安(busulfan)、英丙舒 凡(improsulfan)及0底泊舒凡(piposulfan);氣丙。定(aziridine) 類,諸如苯0坐多巴(benzodopa)、卡巴酿i (carboquone)、米 特多巴(meturedopa)及尤利多巴(uredopa);伸乙基亞胺 (ethylenimine)類及甲基密胺(methylamelamine)類,包括六 143939.doc -54- 201019961To increase the half-life of an antibody or polypeptide comprising an amino acid sequence of the invention, a salvage receptor binding epitope can be linked to an antibody (especially an antibody fragment) as described in, for example, U.S. Patent 5,739,277. For example, a nucleic acid molecule encoding a rescue receptor binding epitope can be ligated in-frame with a nucleic acid encoding a polypeptide sequence of the invention such that the fusion protein represented by the engineered nucleic acid molecule comprises a rescue receptor binding epitope and Polypeptide Sequence of the Invention As used herein, the term "responsible receptor binding epitope" refers to an antigenic epitope in the Fc region of an IgG molecule (eg, IgG1, IgG2, IgG3, or IgG4) that is responsible for increasing the in vivo serum half-life of an IgG molecule. Base (e.g., Ghetie et al, Rev. Immunol. 18:739-766 (2000), Table 1). Antibodies having substitutions and increased serum half-life in the Fe region are also described in the following documents: WO 00/42072; WO 143939.doc • 49-201019961 02/060919; Shields et al., J. Biol. Chem. 276:6591-6604 ( 2001) Hinton, J. Biol. Chem. 279: 6213-6216 (2004). In another embodiment, the serum half-life can also be increased, e.g., by ligation of other polypeptide sequences. For example, an antibody or other polypeptide suitable for use in the methods of the invention may be linked to a portion of a serum albumin or serum albumin that binds to an FcRn receptor or a serum albumin binding peptide such that serum albumin binds to the antibody or polypeptide, such polypeptides Sequences are for example disclosed in WO 01/45746. In a preferred embodiment, the serum albumin peptide to be linked comprises the amino acid sequence DICLPRWGCLW (SEQ ID NO: 32). In another embodiment, the half-life of the Fab is increased using such methods. For serum albumin binding peptide sequence 歹1J, see also Dennis et al, J. Biol. Chem. 277:35035-35043 (2002). An "isolated" polypeptide or an "isolated" antibody is one which has been identified and isolated and/or recovered from components of the natural environment. Contaminants in their natural environment are substances that interfere with the diagnostic or therapeutic use of the polypeptide or antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. In a preferred embodiment, the polypeptide or antibody will be purified to: (1) the polypeptide or antibody comprises 95% by weight or more and preferably 99% by weight or more as determined by the Lowry method; (2) by using a spin The cup sequencer is sufficient to obtain at least 15 residues of the N-terminal or internal amino acid sequence; or (3) by Coomassie blue or preferably silver staining under reducing or non-reducing conditions The SDS-PAGE of (silver stain) is determined to be homogeneity. Since at least one component of the polypeptide natural environment will not be present, the isolated polypeptide or antibody comprises a polypeptide or antibody that is in situ in a recombinant cell. However, 143939.doc -50-201019961 will often be prepared by at least one purification step to prepare an isolated polypeptide or antibody. "Fragment" means a portion of a polypeptide or nucleic acid molecule that preferably contains at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90 of the total length of the reference nucleic acid molecule or polypeptide. 〇/〇, 95°/〇 or longer. Fragments may contain 10, 20, 30, 40, 50, 60, 70, 80, 90 or 1 〇〇, 200, 300, 400, 500, 600 or more nucleotides, or 1 〇, 2 〇, 3 〇, 40, 50, 60, 70, 80, 90, 1 〇〇, 120, 140, 160, 180, 190, 200 or more amino acids. Treatment refers to therapeutic treatment and preventive measures. Those in need of treatment include those who have developed benign, precancerous or non-metastatic tumors, as well as those who need to prevent cancer from developing or recurring. The term "therapeutically effective amount" refers to an amount of a therapeutic agent that treats or prevents a disease or condition in a mammal. In the case of cancer, a therapeutically effective amount of the therapeutic agent can reduce the number of cancer cells; reduce the size of the primary tumor; inhibit (i.e., slow down and better terminate) cancer cells infiltrate into peripheral organs; (i.e., to some extent slow and preferably terminate) tumor metastasis; to some extent inhibit tumor growth; and/or to some extent alleviate one or more symptoms associated with the condition. It inhibits cell growth and/or is cytotoxic insofar as the drug prevents growth and/or kills existing cancer cells. For cancer therapy, in vivo efficacy can be measured, for example, by assessing duration of survival, time to disease progression (TTP), response rate (RR), duration of response, and/or quality of life. The terms "cancer" and "cancerous" or describe the physiological condition in which the growth of cells is usually characterized by unregulated cell growth. This definition includes good I43939.doc •51- 201019961 ^•born cancer and malignant cancer^ Γearly cancer” or early stage tumor means “non-invasive or metastatic cancer, or classified as stage 0, stage I or η Cancer of cancer cancer Examples include, but are not limited to, carcinoma, lymphoma, blastoma (including chorioblastoma and retinoblastoma), sarcoma (including liposarcoma and synovial sarcoma), nerve Endocrine tumors (including carcinoid tumors, gastrinoma and islet cell carcinoma), mesothelioma, schwannomas (including hearing neoplasms), meningioma, adenocarcinoma, melanoma, and leukemia or lymphoid malignancies. More specific examples of such cancers include squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer (including small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), lung adenocarcinoma, and lung squamous cell carcinoma), Peritoneal cancer, hepatocellular carcinoma, lunar cancer (including gastrointestinal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer (hepatoma) Including metastatic breast cancer), colon cancer, rectal cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, genital cancer, squamous cell carcinoma, liver cancer (4) heart (10) in_), anal cancer 35 Cancer, testicular cancer, esophageal cancer, biliary tract cancer, and head and neck cancer. The term "pre-cancer" refers to the condition or growth that usually precedes cancer or develops into cancer. "Precancerous" growth will have cells characterized by abnormal cell cycle regulation, proliferation or differentiation, which can be determined by indicators of cell cycle regulation, cell proliferation or differentiation. Plant development plant dysplasia means any abnormal growth of tissue, organ, or cell. Dysplasia is preferably high grade or precancerous. “Transfer” means that the cancer spreads from its original site to the rest of the body. 143939.doc -52· 201019961. Cancer cells can detach from the primary tumor, penetrate into the lymph and blood vessels, and grow (metastasize) through the bloodstream and in distant lesions in normal tissues in other parts of the body. The transfer can be local or distant. The metastasis is a continuous process, in which the tumor cells are detached from the primary tumor, and the blood is prevalent, and stays at a distant site. At this new site, the cells establish blood supply and grow to form life-threatening substances. The stimulatory and inhibitory molecular pathways within tumor cells regulate this behavior and the interaction between tumor cells and host cells in distant sites is also evident. "Non-metastatic" means that the cancer is benign, or stays at the primary site and does not penetrate into the lymphatic or vascular system or in tissues other than the primary site. Non-metastatic cancers are generally any cancer that is stage 0, J or π cancer and sometimes stage III cancer. "Primary tumor" or "primary cancer" means an initial cancer and does not mean a metastatic disease in another tissue, organ or location within the individual. A "benign tumor" or "benign cancer" means a tumor that is still at the site of origin and does not have the ability to infiltrate, invade, or metastasize to a distant site. "Tumor burden" means the number of cancer cells in the body, the size of the tumor, or the amount of cancer. The tumor burden is also called the tumor load (load). "Number of tumors" means the number of tumors. "Individual" means a mammal, including but not limited to a human or non-human mammal, such as a cow, horse, dog, sheep, or an individual, preferably a human. 143939.doc -53- 201019961 The term "anticancer therapy" refers to a therapy that is suitable for the treatment of cancer. Examples of anti-cancer therapeutics include, but are not limited to, for example, chemotherapeutic agents, growth inhibitors, cytotoxic agents, agents used in radiation therapy, anti-angiogenic agents, apoptotic agents, anti-tubulin agents, and cancer treatments. Other agents, anti-CD20 antibodies, platelet-derived growth factor inhibitors (eg GleevecTM (imatinib mesylate)), COX-2 inhibitors (eg celecoxib), interferons, cytokines An antagonist (eg, a neutralizing antibody) that binds to one or more of the following standard ErbB2, ErbB3, ErbB4, PDGFR-β, BlyS, APRIL, BCMA or VEGF receptors, TRAIL/Apo2, and other bioactive agents and organic chemistry Agents, etc. Combinations of the invention are also included in the invention. The term "cytotoxic agent" as used herein refers to a substance that inhibits or blocks cellular function and/or causes cell destruction. The term is intended to include radioisotopes (e.g., I131, I125, Y9G, and Re186), chemotherapeutic agents, and toxins (such as enzymatically active toxins of bacterial, fungal, plant or animal origin or fragments thereof). A "chemotherapeutic agent" is a compound suitable for treating cancer. Examples of chemotherapeutic agents include compounds suitable for the treatment of cancer. Examples of chemotherapeutic agents include: alkylating agents such as thiotepa and CYTOXAN® cyclophosphamide; alkyl sulfonates such as busulfan, improsulfan and Piposulfan; gas C. Aziridine, such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimine and methyl Methylamine (methylamelamine), including six 143939.doc -54- 201019961

曱密胺(altretamine)、三伸乙基密胺(triethylenemelamine)、 三伸乙基鱗醯胺(trietylenephosphoramide)、三伸乙基硫代 鱗醯胺(triethiylenethiophosphoramide)及三經曱基密胺 (trimethylolomelamine);多聚乙醯(acetogenin)類(尤其布 拉他辛(bullatacin)及布拉他辛酮(bullatacinone));喜樹驗 (camptothecin)(包括合成類似物拓朴替康(topotecan));苔 蘚蟲素(bryostatin);卡利斯達汀(callystatin); CC-1065(包 括其阿多來新(adozelesin)、卡折來新(carzelesin)及比折來 新(bizelesin)合成類似物);念珠藻環肽(cryptophycin)類 (尤其念珠藻環肽1及念珠藻環肽8);海兔毒素(dolastatin); 多卡米辛(duocarmycin)(包括合成類似物KW-2189及CB1-TM1);艾權素(eleutherobin);盤克斯達汀(pancratistatin); 沙考的汀(sarcodictyin);海錦抑素(spongistatin);氮芬 (nitrogen mustard)類,諸如苯丁酸氮芥、萘氮芥 (chlornaphazine)、氯填醯胺(cholophosphamide)、雌莫司 ί丁 (estramustine)、異環鱗酿胺(ifosfamide)、氮芬(mechlorethamine)、 鹽酸氮芥氧化物(mechlorethamine oxide hydrochloride) ' 美法命、新恩比興(novembichin)、膽固醇苯乙酸氮芬 (phenesterine)、潑尼莫司 ί丁(prednimustine)、氣乙環破醯 胺(trofosfamide)、展嘴咬氮芥(uracil mustard);亞石肖基脲 (nitrosurea)類,諸如卡莫司汀(carmustine)、氣腺黴素 (chlorozotocin)、福莫司汀(fotemustine)、洛莫司汀(lomustine)、 尼莫司汀(nimustine)及拉甯司汀(ranimnustine);抗生素, 諸如稀二炔抗生素(例如卡奇徽素(calicheamicin),尤其卡 143939.doc -55- 201019961 奇黴素γΐΐ及卡奇徽素ωΐΐ(參見例如Angew,C/^ew/wi/·五汰 五《茗/·,33:183-186 (1994));達米辛(dynemicin),包括達米 辛A ;雙膦酸鹽(bisphosphonate)類,諸如氣屈膦酸鹽 (clodronate);艾斯帕米辛(esperamicin);以及新製癌菌素 (neocarzinostatin)發色團及相關色蛋白烯二炔抗生素發色 團)、阿克那黴素(aclacinomysin)、放線菌素(actinomycin)、 奥斯拉米辛(authramycin)、偶氮絲胺酸(azaserine)、博來 黴素(bleomycins)、放線菌素C(cactinomycin)、卡拉比辛 (carabicin)、洋紅黴素(carminomycin)、嗜癌菌素(carzinophilin)、 色黴素(chromomycini)、更生黴素(dactinomycin)、道諾黴 素(daunorubicin)、地托比星(detorubicin) 、 6-重氮基-5-側 氧基-L-正白胺酸、ADRIAMYCIN®阿黴素(doxorubicin)(包 括嗎啉基-阿黴素、氰基嗎啉基-阿黴素、2-吡咯啉基-阿黴 素及脫氧阿黴素(deoxydoxombicin))、表柔比星(epirubicin)、 依索比星(esorubicin)、黃膽素(idarubicin)、麻西羅黴素 (marcellomycin)、絲裂黴素(諸如絲裂黴素C(mitomycin (1:))、黴盼酸(1]1)^〇卩1^11〇1^^(^(1)、諾加黴素(11〇§&1&1]1>^11)'撖 欖黴素(olivomycin)、培洛徽素(peplomycin)、泊非黴素 (potfiromycin)、嗓吟黴素(puromycin)、奎那黴素(quelamycin)、 羅多比星(rodorubicin)、鏈黑菌素(streptonigrin)、鏈佐星 (streptozocin)、殺結核菌素(tubercidin)、烏苯美司(ubenimex)、 淨司他丁(zinostatin)、左柔比星(zorubicin);抗代謝物, 諸如曱胺喋呤及5-氟尿嘧啶(5-FU);葉酸類似物,諸如迪 諾特寧(denopterin)、甲胺嗓呤、蝶羅呤(pteropterin)、三 143939.doc -56- 201019961 甲曲沙(trimetrexate);嗓呤類似物,諸如敦達拉濱 (fludarabine)、6-魏基嗓吟、嗟 π米嗓吟(thiamiprine)、硫鳥 嗓呤(thioguanine) ; °密咬類似物,諸如安西他濱Alkretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide, and trimethylolomelamine ; acetogenin (especially bullatacin and bullatacinone); camptothecin (including synthetic analogue topotecan); moss Bryostatin; callistatin; CC-1065 (including its adozelesin, carzelesin, and bizelesin synthetic analogues); rosary Algae cryptophycin (especially Candida cyclic peptide 1 and Nostoccal cyclic peptide 8); dolastatin; doocarmycin (including synthetic analogues KW-2189 and CB1-TM1); Eleutherobin; pancratistatin; sarcodictyin; spongistatin; nitrogen mustard, such as chlorambucil, naphthyl mustard (chlornaphazine), chlorolamine (cholophosphamide), Estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride 'methodamine, new novembichin, cholesterol phenylacetic acid Phenosterine, prednimustine, trofosfamide, uracil mustard, and nitrosurea, such as carmustine Carmustine), chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics, such as diacetylene antibiotics (eg calicheamicin, especially card 143939.doc -55- 201019961 gibberellin gamma 卡 and kawachi ω ΐΐ (see eg Angew, C/^ew/wi/·五五五《茗/·, 33: 183-186 (1994)); dynemicin, including damicin A; bisphosphonate, such as clodronate; esperamicin ; and neocarzinostatin chromophores and related colored eggs Olefin diacetyl antibiotic chromophore), aclacinomysin, actinomycin, authramycin, azaserine, bleomycins ), actinomycin C (cactinomycin), carabincin, carminomycin, carzinophilin, chromomycini, dactinomycin, daunorubicin (daunorubicin), detorubicin, 6-diazo-5-oxo-L-positral acid, ADRIAMYCIN® doxorubicin (including morpholinyl-doxorubicin, cyanide) Benzolinyl-doxorubicin, 2-pyrroline-doxorubicin and deoxydoxombicin), epirubicin, esorubicin, idarubicin , marcellomycin (marcellomycin), mitomycin (such as mitomycin C (mitomycin (1:)), mycophenolic acid (1] 1) ^ 〇卩 1 ^ 11 〇 1 ^ ^ (^ ( 1), nogamycin (11〇§ &1&1]1>^11) 'olivomycin, peplomycin, potfiromycin, 嗓吟Puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex ), ketostatin, zorubicin; antimetabolites such as amidoxime and 5-fluorouracil (5-FU); folic acid analogues such as denoptinin, Methotrexate, pteropterin, 143939.doc -56- 201019961 Trimetrexate; 嗓呤 analogs, such as fludarabine, 6-weig 嗓吟, 嗟π Thiamiprine, thioguanine; ° bite analogs, such as amphetamine

(ancitabine)、阿紮胞苦(azacitidine)、6·氮尿普、卡莫氟 (carmofur)、阿糖胞普(cytarabine)、雙脫氧尿普 (dideoxyuridine)、脫氧氟尿普(doxifluridine)、依諾他濱 (enocitabine)、氟尿苦(floxuridine);雄激素,諸如卡普睪 嗣(calusterone)、丙酸屈他雄鲷(dromostanolone propionate)、環 硫雄醇(epitiostanol)、美雄烧(mepitiostane)、睪内醋 (testolactone);抗腎上腺素(anti-adrenal)類,諸如胺魯米 特(aminoglutethimide)、米托坦(mitotane)、曲洛司坦 (trilostane);葉酸補充劑,諸如夫羅林酸(frolinic acid); 醋葡搭内醋(aceglatone);醒構醯胺糖普(aldophosphamide glycoside);胺基乙醯丙酸(胺基levulinic acid);伊利盧拉 (eniluracil);安^丫嗖(amsacrine);倍思塔布(bestrabucil); 比生群(bisantrene);埃達曲克(edatraxate);得弗伐胺 (defofamine);秋水仙胺(demecolcine);地0丫酿(diaziquone); 艾弗利散(elfornithine);依利醋銨(elliptinium acetate);埃 坡徽素(epothilone);依託格魯(etoglucid);墙酸鎵;經基 腺;香兹多酷體(lentinan);羅尼達寧(lonidainine);美登 素類(maytansinoids),諸如美登素(maytansine)及安絲菌素 (ansamitocin);米托胍膝(mitoguazone);米托蒽醒 (mitoxantrone);莫比達摩(mopidanmol);碗拉維林 (nitraerine);喷司他丁(pentostatin);凡那明(phenamet); 143939.doc -57- 201019961 0比柔比星(pirarubicin);洛索蒽酿(losoxantrone);足葉草 酸(podophyllinic acid) ; 2-乙基醢肼(2-ethylhydrazide);丙 卡巴肼(procarbazine) ; PSK® 多酷複合物(JHS Natural Products, Eugene, OR);雷佐生(razoxane);根瘤菌素 (rhizoxin);西佐喃(sizofiran);鍺螺胺(spirogermanium); 細交鏈孢菌酮酸(tenuazonic acid);三亞胺酿(triaziquone); 2,2’,2M-三氯三乙基胺;單端孢黴烯族毒素(trichothecene) 類(尤其T-2毒素、弗納庫林A(verracurin A)、桿孢菌素 A(roridin A)及胺癸叮(anguidine));胺基曱酸 Θ旨(urethan); 長春地辛(vindesine);達卡巴唤(dacarbazine);甘露莫司 汀(mannomustine);二漠甘露醇(mitobronitol);二溴衛矛 醇(mitolactol) ; 0辰泊溴烧(pipobroman);曱托辛(gacytosine); 阿拉伯糖(「Ara-C」);環磷醯胺;塞替派;紫杉烷類 (taxoid),例如 TAXOL® 太平洋紫杉醇(paclitaxel,Bristol-Myers Squibb Oncology, Princeton, N.J.) 、 ABRAXANETM 不含十六醇聚氧乙稀醚(Cremophor-free)的太平洋紫杉醇之 白蛋白工程改造奈米粒子調配物(American Pharmaceutical Partners, Schaumberg, Illinois)及 TAXOTERE® 多西他賽 (docetaxel 5 Rhone- Poulenc Rorer, Antony, France);克羅 南布(chloranbucil) ; GEMZAR®吉西他濱(gemcitabine) ; 6-硫基鳥嘌呤;巯基嘌呤;曱胺喋呤;鉑類似物’諸如順鉑 (cisplatin)及卡波銘(carboplatin);長春驗;銘(platinum); 依託泊苷(etoposide)(VP-16);異環磷醯胺;米托蒽醌;長 春新鹼;NAVELBINE®長春瑞賓(vinorelbine);諾凡特龍 143939.doc -58- 201019961(ancitabine), azacitidine, hexazone, carmofur, cytarabine, dideoxyuridine, doxifluridine, Enocitabine, floxuridine; androgens, such as caluterone, dromostanolone propionate, epitiostolol, mepitiostane , testolactone; anti-adrenal, such as aminoglutethimide, mitotane, trilostane; folic acid supplements, such as furolin Folinic acid; aceglatone; awake aldophosphamide glycoside; alanine levulinic acid; eniluracil; (amsacrine); bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; Elfornithine; elliptinium acetate ; epothilone; etoglucid; gallium wall; transbasin; lentinan; lonidainine; maytansinoids, such as Maytansine and ansamitocin; mitoguazone; mitoxantrone; mopidanmol; bowl of nitraerine; pentastatin (pentostatin); phennamet; 143939.doc -57- 201019961 0 pirarubicin; losoxantrone; podophyllinic acid; 2-ethyl hydrazine ( 2-ethylhydrazide); procarbazine; PSK® complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; Syrrogermanium; tenuazonic acid; triaziquone; 2,2',2M-trichlorotriethylamine; trichothecene (trichothecene) In particular, T-2 toxin, verracurin A, roridin A, and anguidine; (urethan); vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; 0 pospobroman ); gacytosine; arabinose ("Ara-C"); cyclophosphamide; thiotepa; taxoid, such as TAXOL® paclitaxel (Bristol-Myers Squibb Oncology, Princeton, NJ), ABRAXANETM Crepes-free, pacliphor-free, paclitaxel-based albumin engineered nanoparticle formulation (American Pharmaceutical Partners, Schaumberg, Illinois) and TAXOTERE® docetaxel (docetaxel 5 Rhone- Poulenc Rorer, Antony, France); chloranbucil; GEMZAR® gemcitabine; 6-thioguanine; guanidinium; amidoxime; platinum analogues such as cisplatin (cisplatin) and carboplatin; vintin; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® vinorelbine Bin (vinorelbi Ne); Novartron 143939.doc -58- 201019961

(novantrone);替尼泊苷(teniposide);依達曲沙(edatrexate); 道諾徽素(daunomycin);胺基嗓吟(aminopterin);希羅達 (xeloda); 伊班膦酸鹽(ibandronate); 依立替康 (irinotecan)(開普拓(Camptosar),CPT-11),包括依立替康 與5-FU及曱醢四氫葉酸(leucovorin)之治療方案;拓撲異構 酶抑制劑RFS 2000 ;二氟甲基鳥胺酸(DMFO);類視色素 (retinoid)類,諸如視黃酸;卡培他濱(capecitabine);康柏 斯達汀(combretastatin);甲醯四氫葉酸(LV);奥赛力鉑 (oxaliplatin),包括奥賽力鉑治療方案(卩01^0又);?〖(:-α、Raf、H-Ras、EGFR(例如埃羅替尼(TarcevaTM))及 VEGF-A之抑制劑,其減少細胞增殖;及上述任一者之醫 藥學上可接受之鹽、酸或衍生物。 該定義中亦包括用於調控或抑制激素對腫瘤之作用的抗 激素劑,諸如抗雌激素及選擇性雌激素受體調節劑 (SERM),包括例如他莫昔芬(tamoxifen)(包括NOLVADEX® 他莫昔芬)、雷諾昔盼(raloxifene)、曲洛昔芬(droloxifene)、 4-經基他莫昔芬、曲沃昔芬(trioxifene)、雷諾昔盼 (keoxifene)、LY117018、奥那司酮(onapristone)及 FARES TON· 托瑞米芬(FARESTON‘toremifene);抑制芳香酶之芳香酶 抑制劑,其調控腎上腺中之雌激素產生,諸如4(5)-咪唑、 胺魯米特、MEGASE® 乙酸曱地孕 _ (megestrol acetate)、 AROMASIN® 依西美坦(exemestane)、弗米斯坦(formestanie)、 法屈 °坐(fadrozole)、RIVISOR® 伏羅峻(vorozole)、FEMARA® 來曲唾(letrozole)及 ARIMIDEX® 安美達鍵(anastrozole); 143939.doc -59- 201019961 及抗雄激素,諸如敗他胺(flutamide)、尼魯胺(nilutamide)、 比卡魯胺(bicalutamide)、亮丙立德(leuprolide)及戈舍瑞林 (goserelin);以及曲沙他濱(troxacitabine)(—種 1,3-二氧戊 環核苷胞嘧啶類似物);反義寡核苷酸,尤其抑制異常細 胞增殖所涉及之信號傳導路徑中之基因表現的反義募核苷 酸,諸如PKC-ct,Raf及H-Ras ;核糖核酸酶,諸如VEGF表 現抑制劑(例如ANGIOZYME®核糖核酸酶)及HER2表現抑 制劑;疫苗,諸如基因療法疫苗,例如ALLOVECTIN®疫 苗、LEUVECTIN® 疫苗及 VAXID® 疫苗;PROLEUKIN® rIL-2 ; LURTOTECAN®拓撲異構酶 1抑制劑;ABARELIX® rmRH ;長春瑞賓及艾斯帕米辛(參見美國專利第4,675,187 號);及上述任一者之醫藥學上可接受之鹽、酸或衍生 物。 如本申請案中所使用之術語「前藥」係指醫藥學活性物 質之前驅物或衍生物形式,與母體藥物相比,其對腫瘤細 胞的細胞毒性較小,且能夠酶促活化或轉化為更具活性之 親本形式。參見例如 Wilman, 「Prodrugs in Cancer Chemotherapy」 Biochemical Society Transactions, ,第 375-382 頁,615th Meeting Belfast(1986);及 Stella 等人, 「Prodrugs: A Chemical Approach to Targeted Drug Delivery」,Direcied Drwg Borchardt 等人(編), 第247-267頁,Humana Press (1985)。本發明之前藥包括 (但不限於)含有磷酸鹽之前藥、含有硫代磷酸鹽之前藥、 含有硫酸鹽之前藥、含有肽之前藥、經D·胺基酸修飾之前 143939.doc -60- 201019961 藥、糖基化前藥、含β-内醯胺之前藥、含有視情況經取代 之苯氧基乙醯胺的前藥或含視情況經取代之苯乙醯胺的前 藥、5-氟胞嘧啶及其他5-氟尿苷前藥,其可轉化為更具活 性之無細胞毒性藥物。可衍生為用於本發明之前藥形式之 細胞毒性藥物之實例包括(但不限於)上述化學治療劑。(novantrone); teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate ); irinotecan (Camptosar, CPT-11), including irinotecan and 5-FU and leucovorin; topoisomerase inhibitor RFS 2000 Difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; combretastatin; formazan tetrahydrofolate (LV) Oxaliplatin, including Osili Platinum treatment plan (卩01^0 again); (: -α, Raf, H-Ras, EGFR (eg, erlotinib (TarcevaTM)) and an inhibitor of VEGF-A, which reduces cell proliferation; and a pharmaceutically acceptable salt of any of the above, Acids or derivatives. Also included in the definition are anti-hormonal agents for regulating or inhibiting the effects of hormones on tumors, such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (tamoxifen). (including NOLVADEX® tamoxifen), raloxifene, droloxifene, 4-pyroxyzin, trioxifene, keoxifene, LY117018, onapristone and FARES TON· toremifene (FARESTON 'toremifene); an aromatase inhibitor that inhibits aromatase, which regulates estrogen production in the adrenal gland, such as 4(5)-imidazole, amine Lumet, MEGASE® megestrol acetate, AROMASIN® exemestane, formestanie, fadrozole, RIVISOR® vorozole, FEMARA® Letrozole and ARIMIDEX® (A) Nastrozole); 143939.doc -59- 201019961 and antiandrogens, such as flutamide, nilutamide, bicalutamide, leuprolide and goserelin (goserelin); and troxacitabine (a 1,3-dioxolan nucleoside cytosine analog); antisense oligonucleotides, particularly in the signal transduction pathway involved in the inhibition of abnormal cell proliferation Antisense nucleotides expressed by genes such as PKC-ct, Raf and H-Ras; ribonucleases such as VEGF expression inhibitors (eg ANGIOZYME® ribonuclease) and HER2 expression inhibitors; vaccines, such as gene therapy Vaccines such as ALLOVECTIN® vaccine, LEUVECTIN® vaccine and VAXID® vaccine; PROLEUKIN® rIL-2; LURTOTECAN® topoisomerase 1 inhibitor; ABARELIX® rmRH; vinorelbine and espartame (see US Patent 4,675,187) And a pharmaceutically acceptable salt, acid or derivative of any of the above. The term "prodrug" as used in this application refers to a precursor or derivative form of a pharmaceutically active substance, and Maternal medicine It is small compared to the cytotoxicity of tumor cells, and is capable of being enzymatically activated or converted into the more active parent form of the present. See, for example, Wilman, "Prodrugs in Cancer Chemotherapy" Biochemical Society Transactions, , pp. 375-382, 615th Meeting Belfast (1986); and Stella et al., "Prodrugs: A Chemical Approach to Targeted Drug Delivery", Direcied Drwg Borchardt et al. (eds.), pp. 247-267, Humana Press (1985). Prodrugs of the present invention include, but are not limited to, pre-phosphate containing drugs, prodrugs containing thiophosphates, prodrugs containing sulfates, prodrugs containing peptides, modified with D-amino acids 143939.doc -60-201019961 Drug, glycosylation prodrug, prodrug containing β-namidoxime, prodrug containing optionally substituted phenoxyacetamide or prodrug containing phenethylamine optionally substituted, 5-fluoro Cytosine and other 5-fluorouridine prodrugs that can be converted to more active, non-cytotoxic drugs. Examples of cytotoxic drugs which may be derivatized for use in the prodrug form of the present invention include, but are not limited to, the above chemotherapeutic agents.

「放射療法」意謂使用定向γ射線或β射線來引發足夠的 細胞損害以致限制其正常作用之能力或完全毀壞細胞。應 瞭解,此項技術中已知許多確定治療劑量及持續時間之方 法。典型治療呈一次投藥形式給予,且典型劑量範圍為每 天10至200個單位(戈瑞(Gray))。 「降低或抑制」意謂能夠引起20%、30%、40%、50%、 60%、70%、75%、80%、85%、90%、95% 或更高之總體 降低。降低或抑制可指所治療病症之症狀、轉移之存在或 大小、原發性腫瘤之大小,或血管生成病症中血管之大小 或數目。 治療劑 本發明係關於c-met拮抗劑及VEGF拮抗劑以組合療法治 療諸如腫瘤之病理性病狀的用途。在另一態樣中,本發明 係關於c-met拮抗劑、VEGF拮抗劑及EGFR拮抗劑以組合 療法治療個體中諸如腫瘤之病理性病狀的用途。 c-met拮抗劑 適用於本發明方法之c-met拮抗劑包括特異性結合c-met 之多肽、抗c-met抗體、c-met小分子、特異性結合c-met之 受體分子及衍生物、及融合蛋白。c-met拮抗劑亦包括c- 143939.doc •61 - 201019961 met多肽、針對c-met及HGF之RNA適體及肽體之拮抗變異 體。作為適用於本發明方法之c-met拮抗劑,亦包括抗HGF 抗體、抗HGF多肽、特異性結合HGF之c-met受體分子及衍 生物。此等拮抗劑中每一者之實例描述如下。 適用於本發明方法之抗c-met抗體包括以足夠親和力及 特異性結合c-met且可降低或抑制c-met活性的任何抗體。 所選抗體通常應對c-met具有足夠強的結合親和力,例如 抗體結合人類c-met之Kd值可在100 nM-1 pM之間。抗體親 和力可利用以下方法測定,例如基於表面電漿共振之檢定 (諸如PCT申請公開案第WO 2〇05/012359號中所述之 BIAcore檢定)、酶聯免疫吸附檢定(ELISA)及競爭檢定(例 如RIA)。較佳可使用本發明之抗c-met抗體作為靶向及干 擾涉及c-met/HGF活性之疾病或病狀之治療劑。另外,可 使抗體經受其他生物活性檢定,例如以評估其作為治療劑 之效用。該等檢定在此項技術中為已知的且視靶抗原及抗 體之預定用途而定。"Radiotherapy" means the use of directional gamma rays or beta rays to induce sufficient cellular damage to limit its ability to function normally or to completely destroy cells. It should be understood that a number of methods are known in the art for determining the therapeutic dose and duration. Typical treatments are administered in a single administration with a typical dose ranging from 10 to 200 units per day (Gray). "Reducing or inhibiting" means causing an overall decrease of 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% or higher. Reduced or inhibited may refer to the symptoms of the condition being treated, the presence or size of metastases, the size of the primary tumor, or the size or number of blood vessels in the angiogenic condition. Therapeutic Agents The present invention relates to the use of a combination therapy with a c-met antagonist and a VEGF antagonist for the treatment of a pathological condition such as a tumor. In another aspect, the invention relates to the use of a combination therapy to treat a pathological condition, such as a tumor, in a subject in combination with a c-met antagonist, a VEGF antagonist, and an EGFR antagonist. C-met antagonists C-met antagonists suitable for use in the methods of the invention include polypeptides that specifically bind c-met, anti-c-met antibodies, c-met small molecules, receptor molecules that specifically bind c-met, and derivatives And fusion proteins. C-met antagonists also include c-143939.doc • 61 - 201019961 met polypeptide, antagonistic variants of RNA aptamers and peptibodies against c-met and HGF. As c-met antagonists suitable for use in the methods of the invention, anti-HGF antibodies, anti-HGF polypeptides, c-met receptor molecules and derivatives which specifically bind to HGF are also included. Examples of each of these antagonists are described below. Anti-c-met antibodies suitable for use in the methods of the invention include any antibody which binds c-met with sufficient affinity and specificity and which reduces or inhibits c-met activity. The selected antibody should generally have sufficient binding affinity for c-met, for example, the antibody binding to human c-met may have a Kd value between 100 nM and 1 pM. Antibody affinities can be determined by methods such as surface plasmon resonance based assays (such as the BIAcore assay described in PCT Application Publication No. WO 2 〇 05/012359), enzyme-linked immunosorbent assay (ELISA), and competition assays ( For example, RIA). Preferably, the anti-c-met antibody of the present invention can be used as a therapeutic agent for targeting and interfering with diseases or conditions involving c-met/HGF activity. Alternatively, the antibody can be subjected to other biological activity assays, e.g., to assess its utility as a therapeutic. Such assays are known in the art and depend on the intended use of the target antigen and antibody.

抗c-met抗體(可呈單臂抗體形式提供)在此項技術中為已 知的。參見例如Martens, T等人,(2006) Clin Cancer Res 12(20 Pt 1):6144 ; US 6,468,529 ; WO 2006/015371 ; WO 2007/063816 ; US7,408,043 ; WO 2009/007427 ; WO 2005/016382 ; WO 2007/126799。在一些實施例中,抗c-met抗體為MetMAb。MetMAb之序列顯示於圖9及10中。 MetMAb(亦稱為 OA5D5v2)亦描述於 WO 2006/015371 及 Jin 等人,Cancer Res (2008) 68:4360 中。 143939.doc -62- 201019961 在一些實施例中,抗c-met抗體包含(a)第一多肽,其包 含具有如下序列之重鏈可變域:EVQLVESGGGLVQPGGSL RLSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNSD TRFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYCA TYRSYVTPLDYWGQGTLVTVSS(SEQ ID NO: 10)、CH1 序 列及第一 Fc多肽;(b)第二多肽,其包含具有如下序列之輕 鏈可變域:DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTS SQKNYLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSGS GTDFTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIKR (SEQ ID NO: 11)及CL1序列;及(c)包含第二Fc多肽之第三 多肽,其中重鏈可變域及輕鏈可變域呈複合物形式存在且 形成單一抗原結合臂,其中第一及第二Fc多肽呈複合物形 式存在且形成與包含該抗原結合臂之Fab分子相比增加該 抗體片段之穩定性的Fc區。在一些實施例中,第一多肽包 含圖1中描繪之Fc序列(SEQ ID NO: 12),且第二多肽包含 圖2中描繪之Fc序列(SEQ ID NO: 13)。在一些實施例中, 第一多肽包含圖2中描繪之Fc序列(SEQ ID NO: 13),且第 二多肽包含圖1中描繪之Fc序列(SEQ ID NO: 12)。Anti-c-met antibodies, which may be provided in the form of a one-armed antibody, are known in the art. See, for example, Martens, T et al, (2006) Clin Cancer Res 12 (20 Pt 1): 6144; US 6,468,529; WO 2006/015371; WO 2007/063816; US 7,408,043; WO 2009/007427; WO 2005/016382; WO 2007/126799. In some embodiments, the anti-c-met antibody is MetMAb. The sequence of MetMAb is shown in Figures 9 and 10. MetMAb (also known as OA5D5v2) is also described in WO 2006/015371 and Jin et al, Cancer Res (2008) 68:4360. 143939.doc -62-201019961 In some embodiments, the anti-c-met antibody comprises (a) a first polypeptide comprising a heavy chain variable domain comprising: EVQLVESGGGLVQPGGSL RLSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNSD TRFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYCA TYRSYVTPLDYWGQGTLVTVSS (SEQ ID NO: 10) a CH1 sequence and a first Fc polypeptide; (b) a second polypeptide comprising a light chain variable domain having the sequence: DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTS SQKNYLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSGS GTDFTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIKR (SEQ ID NO: 11) and CL1 sequence; and (c) comprising a second a third polypeptide of an Fc polypeptide, wherein the heavy chain variable domain and the light chain variable domain are present in a complex form and form a single antigen binding arm, wherein the first and second Fc polypeptides are present in a complex form and are formed and included The Fab molecule of the antigen binding arm is an Fc region that increases the stability of the antibody fragment. In some embodiments, the first polypeptide comprises the Fc sequence depicted in Figure 1 (SEQ ID NO: 12) and the second polypeptide comprises the Fc sequence depicted in Figure 2 (SEQ ID NO: 13). In some embodiments, the first polypeptide comprises the Fc sequence depicted in Figure 2 (SEQ ID NO: 13) and the second polypeptide comprises the Fc sequence depicted in Figure 1 (SEQ ID NO: 12).

在一些實施例中,抗c-met抗體包含(a)包含重鏈可變域 之第一多肽,該多肽包含序列EVQLVESGGGLVQPGGSLR LSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNSDT RFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYCAT YRSYVTPLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS 143939.doc •63- 201019961 SGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPE VTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVSLSCAVKGFYPSD : IAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR : WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 14) ; (b)包含輕鏈可變域之第二多肽,該多肽包含序列 DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKNYLAW ❿ YQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGTDFTLTIS SLQPEDFATYYCQQYYAYPWTFGQGTKVEIKRTVAAPSVFI FPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV THQGLSSPVTKSFNRGEC (SEQ ID NO: 15);及包含 FC 序 列之第三多肽,該多肽包含序列CPPCPAPELLGGPSVFLFP PKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK VSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQV SLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 13),其中重鏈可變域及輕鏈可變域 呈複合物形式存在且形成單一抗原結合臂,其中第一及第 二Fc多肽呈複合物形式存在且形成與包含該抗原結合臂之 Fab分子相比增加該抗體片段之穩定性的Fc區。 143939.doc -64 - 201019961 抗c-met抗體(可呈單臂抗體形式提供)在此項技術中為已 知的(參見例如 Martens, T等人 ’(2006) Clin Cancer Res 12(20 Pt 1):6144 ; US 6,468,529 ; WO 2006/015371 ; WO 2007/063 816)。在一實施例中,抗c-met抗體包含重鏈可變 域,該重鏈可變域包含圖1中描繪之CDR1-HC、CDR2-HC 及CDR3-HC序列(SEQ ID NO: 4、5及/或9)中之一或多者。 在一些實施例中,該抗體包含輕鏈可變域,該輕鏈可變域 包含圖1中描繪之CDR1-LC、CDR2-LC及CDR3-LC序列 (SEQ ID NO: 1、2及/或3)中之一或多者。在一些實施例 中,重鏈可變域包含圖1中描繪之FR1-HC、FR2-HC、 FR3-HC 及 FR4-HC 序列(SEQ ID NO: 21-24)。在一些實施例 中,輕鏈可變域包含圖1中描繪之FR1-LC、FR2-LC、FR3-LC及 FR4-LC序列(SEQ ID NO: 16-19)。 在其他實施例中,該抗體包含寄存於美國菌種保存中心 (American Type Culture Collection)中之寄存編號為 ATCC HB-11894(融合瘤 1A3.3.13)或 HB-11895(融合瘤 5D5.11.6) 之融合瘤細胞株所產生的單株抗體之一或多個CDR序列。 在一態樣中,抗c-met抗體包含: (a)至少一個、兩個、三個、四個或五個選自由以下組 成之群的高變區(CDR)序列: ⑴ 包含序列A1-A17之CDR-L1,其中A1-A17為 KSSQSLLYTSSQKNYLA (SEQ ID NO: 1), (ii) 包含序列B1-B7之CDR-L2,其中B1-B7為 WASTRES (SEQ ID NO: 2), 143939.doc -65- 201019961 (iii) 包含序列C1-C9之CDR-L3,其中C1-C9為 QQYYAYPWT (SEQ ID NO: 3) > (iv) 包含序列D1-D10之CDR-H1,其中D1-D10為 GYTFTSYWLH (SEQ ID NO: 4), (v) 包含序列E1-E18之CDR-H2,其中E1-E18為 GMIDPSNSDTRFNPNFKD (SEQ ID NO: 5),及 (vi) 包含序列F1-F11之CDR-H3,其中F1-F11為 XYGSYVSPLDY (SEQ ID NO: 6)且X不為 R ; 及(b)至少一個變異CDR,其中該變異CDR序列包含SEQ ID NO: 1、2、3、4、5或6中描緣之序列之至少一個殘基 的修飾。在一實施例中,本發明抗體之CDR-L1包含序列 SEQ ID NO: 1。在一實施例中,本發明抗體之CDR-L2包 含序列SEQ ID NO: 2。在一實施例中,本發明抗體之CDR-L3包含序列SEQ ID NO: 3。在一實施例中,本發明抗體之 CDR-H1包含序列SEQ ID NO: 4。在一實施例中,本發明 抗體之CDR-H2包含序列SEQ ID NO: 5。在一實施例中, 本發明抗體之CDR-H3包含序列SEQ ID NO: 6。在一實施 例中,CDR-H3 包含 TYGSYVSPLDY (SEQ ID NO: 7)。在 一實施例中,CDR-H3 包含 SYGSYVSPLDY (SEQ ID NO: 8)。在一實施例中,包含此等序列(如本文所述組合;)之本 發明抗體為人類化或人類抗體。 在一態樣中,本發明提供包含1、2、3、4、5或6個CDR 之抗體,其中各CDR包含選自由SEQ ID NO: 1、2、3、 143939.doc •66- 201019961 4、5、6、7及8組成之群之序列或由該序列組成或基本上 由該序列組成,且其中SEQ ID NO: 1對應於CDR-L1 ’ SEQ ID NO: 2 對應於 CDR-L2,SEQ ID NO: 3 對應於 CDR-L3 ’ SEQ ID NO: 4 對應於 CDR-H1,SEQ ID NO: 5對應於 CDR-H2,且SEQ ID NO: 6、7或8對應於CDR-H3。在一實施例 中,本發明抗體包含 CDR-L1、CDR-L2、CDR-L3、CDR-Hl、CDR-H2 及 CDR-H3,其中各依序包含 SEQ ID NO: 1、 2、3、4、5及7。在一實施例中,本發明抗體包含CDR-❹ LI、CDR-L2、CDR-L3、CDR-H1、CDR-H2 及 CDR-H3, 其中各依序包含SEQ ID NO: 1、2、3、4、5及8。 本發明抗體中之變異CDR可具有CDR中一或多個殘基之 修飾。在一實施例中,CDR-L2變異體在呈任何組合之以 下位置處包含1-5(1、2、3、4或5)個取代:B1(M或L)、 B2(P、T、G 或 S)、B3(N、G、R 或 T)、B4(I、N 或 F)、 B5(P、I、L 或 G)、B6(A、D、T或 V)及 B7(R、I、Μ或 G)。 在一實施例中,CDR-H1變異體在呈任何組合之以下位置 ❿ 處包含 1-5(1、2、3、4或 5)個取代:D3(N、Ρ、L、S、A、 I)、D5(I、S 或 Y)、D6(G、D、T、K、R)、D7(F、Η、R、 . S、T或V)及D9(M或V)。在一實施例中’ CDR-H2變異體在 呈任何組合之以下位置處包含1-4(1、2、3或4)個取代: E7(Y)、E9(I)、E10(I)、E14(T或 Q)、E15(D、K、S、T或 V)、E16(L)、E17(E、Η、N或 D)及 E18(Y、E 或 H)。在一實 施例中,CDR-H3變異體在呈任何組合之以下位置處包含 1-5(1、2、3、4 或 5)個取代:F1(T、S)、F3(R、S、Η、 143939.doc •67- 201019961 Τ、A、Κ)、F4(G)、F6(R、F、M、T、E、K、A、L、 W)、F7(L、I、T、R、K、V)、F8(S、A)、F10(Y、N)及 F11(Q、S、H、F)。各位置之後的圓括號中之字母指示說 明性取代(亦即置換)胺基酸;熟習此項技術者將顯而易 見,可按常規使用此項技術中已知及/或本文描述之技術 評估在本文所述之情況下其他胺基酸作為取代胺基酸之適 用性。在一實施例中,CDR-L1包含序列SEQ ID NO: 1。 在一實施例中,變異CDR-H3中之F1為T。在一實施例中, 變異CDR-H3中之F1為S。在一實施例中,變異CDR-H3中 之F3為R。在一實施例中,變異CDR-H3中之F3為S。在一 實施例中,變異CDR-H3中之F7為T。在一實施例中,本發 明抗體包含變異CDR-H3,其中F1為T或S,F3為R或S,且 F7 為 T。 在一實施例中’本發明抗體包含變異CDR-H3,其中F1 為T,F3為R,且F7為T。在一實施例中,本發明抗體包含 變異CDR-H3,其中F1為S。在一實施例中,本發明抗體包 含變異CDR-H3 ’其中F1為T,且F3為R。在一實施例中, 本發明抗體包含變異CDR-H3,其中F1為S,F3為RXF7為 T。在一實施例中,本發明抗體包含變異cdr_H3,其中F1 為T,F3為S,F7為T,且F8為S。在一實施例中,本發明 抗體包含變異CDR-H3,其中F1為T,F3為S,F7為T,且 F8為A。在一些實施例中,該變異CDR-H3抗體進一步包含 CDR-L1、CDR-L2、CDR-L3、CDR-H1 及 CDR-H2,其中各 依序包含SEQ ID NO: i、2、3、4及5中描繪之序列。在一 143939.doc • 68- 201019961 些實施例中,此等抗體進一步包含人類子群III重鏈構架共 同序列。在此等抗體之一實施例中,構架共同序列在位置 71、73及/或78處包含取代。在此等抗體之一些實施例 中,位置71為A,73為T及/或78為A。在此等抗體之一實 施例中,此等抗體進一步包含人類κΐ輕鏈構架共同序列》 在一實施例中,本發明抗體包含變異CDR-L2,其中Β6 為V。在一些實施例中,該變異CDR-L2抗體進一步包含 CDR-L1、CDR-L3、CDR-H1、CDR-H2 及 CDR-H3,其中 各依序包含SEQ ID NO: 1、3、4、5及6中描繪之序列。在 一些實施例中,該變異CDR-L2抗體進一步包含CDR-L1、 CDR-L3、CDR-H1、CDR-H2 及 CDR-H3,其中各依序包含 SEQ ID NO: 1、3、4、5及7中描繪之序列。在一些實施例 中,該變異CDR-L2抗體進一步包含CDR-L1、CDR-L3、 CDR-H1、CDR-H2 及 CDR-H3,其中各依序包含 SEQ ID NO: 1、3、4、5及8中描繪之序列。在一些實施例中,此 等抗體進一步包含人類子群III重鏈構架共同序列。在此等 抗體之一實施例中,構架共同序列在位置71、73及/或78 處包含取代。在此等抗體之一些實施例中,位置71為A, 73為T及/或78為A。在此等抗體之一實施例中,此等抗體 進一步包含人類κΐ輕鍵構架共同序列。 在一實施例中,本發明抗體包含變異CDR-H2,其中Ε14 為Τ’ Ε15為Κ’且Ε17為Ε。在一實施例中,本發明抗體包 含變異CDR-H2,其中Ε17為Ε »在一些實施例中,該變異 CDR-H3 抗體進一步包含 CDR-L1、CDR-L2、CDR-L3、 143939.doc -69- 201019961 CDR-H1 及 CDR-H3,其中各依序包含 SEQ ID NO: 1、2、 3、4及6中描繪之序列。在一些實施例中,該變異CDR-H2 抗體進一步包含 CDR-L1、CDR-L2、CDR-L3、CDR-H1 及 CDR-H3,其中各依序包含SEQ ID NO: 1、2、3、4及7中 描繪之序列。在一些實施例中,該變異CDR-H2抗體進一 步包含 CDR-L1、CDR-L2、CDR-L3、CDR-H1 及 CDR-H3, 其中各依序包含SEQ ID NO: 1、2、3、4及8中描繪之序 列。在一些實施例中,此等抗體進一步包含人類子群III重 鏈構架共同序列。在此等抗體之一實施例中,構架共同序 列在位置71、73及/或78處包含取代。在此等抗體之一些 實施例中,位置71為A,73為T及/或78為A。在此等抗體 之一實施例中,此等抗體進一步包含人類κΐ輕鏈構架共同 序列。 在其他實施例中,本發明之c-met抗體特異性結合c-met Sema域或其變異體之至少一部分。在一實例中,本發明之 拮抗劑抗體特異性結合至少一個選自由以下組成之群之序 列:LDAQT (SEQ ID NO: 25)(例如 c_met之殘基 269-273)、 LTEKRKKRS (SEQ ID NO: 26)(例如 c_met 之殘基 3GO-308) 、 KPDSAEPM (SEQ ID NO: 27)( 例如 c-met之殘基 350-357)及NVRCLQHF (SEQ ID NO: 28)(例如 c-met之殘基 381-388)。在一實施例中,本發明之拮抗劑抗體特異性結合由 至少一個選自由以下組成之群之序列的一部分或全部所形 成之構形抗原決定基:LDAQT (SEQ ID NO: 25)(例如c-met之殘基269-273)、LTEKRKKRS (SEQ ID NO: 26)(例如 143939.doc •70· 201019961 c_met之殘基 300-308)、KPDSAEPM (SEQ ID NO: 27)(例如 c-met之殘基 350-357)及NVRCLQHF (SEQ ID NO: 28)(例如 c-met之殘基381-388)。在一實施例中,本發明之拮抗劑抗 體特異性結合與以下序列具有至少50%、60%、70%、 80%、90%、95%、98%序列一致性或相似性之胺基酸序 列:LDAQT (SEQ ID NO: 25)、LTEKRKKRS (SEQ ID NO: 26)、KPDSAEPM (SEQ ID NO: 27)及/或NVRCLQHF (SEQ ID NO: 28)。 在一態樣中,抗c-met抗體包含至少一個促進抗體片段 内Fc序列之異二聚、而最小化同二聚之特徵。該(等)特徵 會改良免疫球蛋白群體之產率及/或純度及/或均質性。在 一實施例中,該抗體包含構成如以下文獻中所述之「杵」 及「白」之Fc突變:WO 2005/063816 ; Ridgeway,J等人, Prot Eng (1996) 9:617-21 ; Zhu Z等人,Prot Sci (1997) 6:781-8。舉例而言,臼突變在Fc多肽中可為T366A、 L368A及/或Y407V之一或多者,空穴突變可為T366W。 抗HGF抗體在此項技術中為熟知的。參見例如Kim KJ等 人,Clin Cancer Res. (2006) 12(4):1292-8 ; WO 2007/115049 ; WO 2009/002521 ; WO 2007/143098 ; WO 2007/017107 ; WO 2005/017107 ; L2G7 ; AMG-102。 本發明方法中可使用特異性結合HGF之c-met受體分子 或其片段來例如結合且隔絕HGF蛋白’從而阻止其傳導信 號。c-met受體分子或其HGF結合片段較佳為可溶形式。在 一些實施例中,該受體之可溶形式係藉由結合HGF而對c- 143939.doc -71- 201019961 met蛋白之生物活性具有抑制效應,從而阻止其結合乾細 胞表面上所存在之其天然受體。亦包括c-met受體融合蛋 白,其實例描述如下。 本發明之可溶性c-met受體蛋白或敌合c-met受體蛋白包 括不經由跨膜域固定於細胞表面上之c-met受體蛋白。因 而,c-met受體之可溶形式(包括嵌合受體蛋白)雖然能夠結 合HGF且使之失活,但不包含跨膜域且因此一般不與表現 該分子之細胞之細胞膜聯結。參見例如Kong-Beltran,Μ等 人,Cancer Cell (2004) 6(1): 75-84。 本發明方法中可使用HGF分子或其片段,該等HGF分子 或其片段特異性結合c-met且阻斷或降低c-met之活化,從 而阻止其傳導信號。 適體為形成特異性結合靶分子(諸如HGF多肽)之三級結 構之核酸分子。適體之產生及治療用途在此項技術中非常 明確。參見例如美國專利第5,475,096號。HGF適體為聚乙 二醇化修飾之募核苷酸,其採用使其能夠結合細胞外HGF 之三維構形。關於適體之其他資訊可見於美國專利申請公 開案第20060148748號中。 肽體為與編碼免疫球蛋白分子之片段或一部分之胺基酸 序列連接的肽序列。多肽可衍生自由任何關於特異性結合 之方法(包括但不限於噬菌體呈現技術)所選擇之隨機序 列。在一較佳實施例中,所選多肽可與編碼免疫球蛋白之 Fc部分之胺基酸序列連接。特異性結合且拮抗HGF或c-met 之肽體亦適用於本發明方法。 143939.doc -72- 201019961In some embodiments, the anti-c-met antibody comprises (a) a first polypeptide comprising a heavy chain variable domain, the polypeptide comprising the sequence EVQLVESGGGLVQPGGSLR LSCAASGYTFTSYWLHWVRQAPGKGLEWVGMIDPSNSDT RFNPNFKDRFTISADTSKNTAYLQMNSLRAEDTAVYYCAT YRSYVTPLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTS GGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS 143939.doc • 63- 201019961 SGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPE VTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVSLSCAVKGFYPSD: IAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR : WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 14); (b) a second polypeptide comprising a light chain variable domain, the polypeptide comprising the sequence DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKNYLAW ❿ YQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGTDFTLTIS SLQPEDFATYYCQQYYAYPWTFGQGTKVEIKRTVAAPSVFI FPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV THQGLSSPVTKSFNRGEC (SEQ ID NO: 15); and FC comprises a first sequence of a tripeptide comprising the sequence CPPCPAPELL GGPSVFLFP PKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCK VSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQV SLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 13), wherein the heavy chain variable domain and a light chain variable domain as a form of a complex and form a single antigen binding arm, wherein the first and second Fc polypeptides form complexes The form exists and forms an Fc region that increases the stability of the antibody fragment compared to a Fab molecule comprising the antigen binding arm. 143939.doc -64 - 201019961 Anti-c-met antibodies (available in the form of a one-armed antibody) are known in the art (see, for example, Martens, T et al. (2006) Clin Cancer Res 12 (20 Pt 1 ): 6144; US 6,468,529; WO 2006/015371; WO 2007/063 816). In one embodiment, the anti-c-met antibody comprises a heavy chain variable domain comprising the CDR1-HC, CDR2-HC and CDR3-HC sequences depicted in Figure 1 (SEQ ID NO: 4, 5) And/or 9) one or more. In some embodiments, the antibody comprises a light chain variable domain comprising the CDR1-LC, CDR2-LC and CDR3-LC sequences depicted in FIG. 1 (SEQ ID NO: 1, 2, and/or One or more of 3). In some embodiments, the heavy chain variable domain comprises the FR1-HC, FR2-HC, FR3-HC, and FR4-HC sequences depicted in Figure 1 (SEQ ID NO: 21-24). In some embodiments, the light chain variable domain comprises the FR1-LC, FR2-LC, FR3-LC, and FR4-LC sequences depicted in Figure 1 (SEQ ID NOS: 16-19). In other embodiments, the antibody comprises the accession number ATCC HB-11894 (fusion tumor 1A3.3.13) or HB-11895 (fusion tumor 5D5.11.6) deposited in the American Type Culture Collection. One or more CDR sequences of a monoclonal antibody produced by the fusion of the tumor cell line. In one aspect, the anti-c-met antibody comprises: (a) at least one, two, three, four or five hypervariable region (CDR) sequences selected from the group consisting of: (1) comprising sequence A1- CDR-L1 of A17, wherein A1-A17 is KSSQSLLYTSSQKNYLA (SEQ ID NO: 1), (ii) comprises CDR-L2 of sequence B1-B7, wherein B1-B7 is WASTRES (SEQ ID NO: 2), 143939.doc -65- 201019961 (iii) CDR-L3 comprising the sequence C1-C9, wherein C1-C9 is QQYYAYPWT (SEQ ID NO: 3) > (iv) CDR-H1 comprising the sequence D1-D10, wherein D1-D10 are GYTFTSYWLH (SEQ ID NO: 4), (v) comprises CDR-H2 of sequence E1-E18, wherein E1-E18 is GMIDPSNSDTRFNPNFKD (SEQ ID NO: 5), and (vi) CDR-H3 comprising sequence F1-F11, Wherein F1-F11 is XYGSYVSPLDY (SEQ ID NO: 6) and X is not R; and (b) at least one variant CDR, wherein the variant CDR sequence comprises SEQ ID NO: 1, 2, 3, 4, 5 or 6. Modification of at least one residue of the sequence of the strand. In one embodiment, the CDR-L1 of an antibody of the invention comprises the sequence SEQ ID NO: 1. In one embodiment, the CDR-L2 of an antibody of the invention comprises the sequence SEQ ID NO: 2. In one embodiment, the CDR-L3 of the antibody of the invention comprises the sequence of SEQ ID NO: 3. In one embodiment, the CDR-H1 of an antibody of the invention comprises the sequence of SEQ ID NO: 4. In one embodiment, the CDR-H2 of the antibody of the invention comprises the sequence of SEQ ID NO: 5. In one embodiment, the CDR-H3 of the antibody of the invention comprises the sequence of SEQ ID NO: 6. In one embodiment, CDR-H3 comprises TYGSYVSPLDY (SEQ ID NO: 7). In one embodiment, CDR-H3 comprises SYGSYVSPLDY (SEQ ID NO: 8). In one embodiment, an antibody of the invention comprising such sequences (as described herein;) is a humanized or human antibody. In one aspect, the invention provides an antibody comprising 1, 2, 3, 4, 5 or 6 CDRs, wherein each CDR comprises a SEQ ID NO: 1, 2, 3, 143939. doc • 66-201019961 4 a sequence consisting of, consisting of, or consisting essentially of the sequence of 5, 6, 7, and 8 and wherein SEQ ID NO: 1 corresponds to CDR-L1 'SEQ ID NO: 2 corresponds to CDR-L2, SEQ ID NO: 3 corresponds to CDR-L3 'SEQ ID NO: 4 corresponds to CDR-H1, SEQ ID NO: 5 corresponds to CDR-H2, and SEQ ID NO: 6, 7 or 8 corresponds to CDR-H3. In one embodiment, an antibody of the invention comprises CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 2, 3, 4 , 5 and 7. In one embodiment, the antibody of the invention comprises CDR-❹ LI, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 2, 3, 4, 5 and 8. A variant CDR in an antibody of the invention may have a modification of one or more residues in the CDR. In one embodiment, the CDR-L2 variant comprises 1-5 (1, 2, 3, 4 or 5) substitutions at any of the following positions: B1 (M or L), B2 (P, T, G or S), B3 (N, G, R or T), B4 (I, N or F), B5 (P, I, L or G), B6 (A, D, T or V) and B7 (R) , I, Μ or G). In one embodiment, the CDR-H1 variant comprises 1-5 (1, 2, 3, 4 or 5) substitutions at any position below any combination: D3 (N, Ρ, L, S, A, I), D5 (I, S or Y), D6 (G, D, T, K, R), D7 (F, Η, R, . S, T or V) and D9 (M or V). In one embodiment, the 'CDR-H2 variant comprises 1-4 (1, 2, 3 or 4) substitutions at any of the following positions: E7 (Y), E9 (I), E10 (I), E14 (T or Q), E15 (D, K, S, T or V), E16 (L), E17 (E, Η, N or D) and E18 (Y, E or H). In one embodiment, the CDR-H3 variant comprises 1-5 (1, 2, 3, 4 or 5) substitutions at any of the following positions: F1 (T, S), F3 (R, S, Η, 143939.doc •67- 201019961 Τ, A, Κ), F4(G), F6 (R, F, M, T, E, K, A, L, W), F7 (L, I, T, R, K, V), F8 (S, A), F10 (Y, N), and F11 (Q, S, H, F). The letters in parentheses after each position indicate an illustrative substitution (i.e., replacement) of the amino acid; it will be apparent to those skilled in the art that it can be routinely evaluated using techniques known in the art and/or described herein. The suitability of other amino acids as substituted amino acids in the case described. In one embodiment, CDR-L1 comprises the sequence SEQ ID NO: 1. In one embodiment, F1 in the variant CDR-H3 is T. In one embodiment, F1 in the variant CDR-H3 is S. In one embodiment, F3 in the variant CDR-H3 is R. In one embodiment, F3 in the variant CDR-H3 is S. In one embodiment, F7 in the variant CDR-H3 is T. In one embodiment, an antibody of the invention comprises a variant CDR-H3, wherein F1 is T or S, F3 is R or S, and F7 is T. In one embodiment, an antibody of the invention comprises a variant CDR-H3, wherein F1 is T, F3 is R, and F7 is T. In one embodiment, an antibody of the invention comprises a variant CDR-H3, wherein F1 is S. In one embodiment, an antibody of the invention comprises a variant CDR-H3' wherein F1 is T and F3 is R. In one embodiment, an antibody of the invention comprises a variant CDR-H3, wherein F1 is S and F3 is RXF7 is T. In one embodiment, an antibody of the invention comprises a variant cdr_H3, wherein F1 is T, F3 is S, F7 is T, and F8 is S. In one embodiment, an antibody of the invention comprises a variant CDR-H3, wherein F1 is T, F3 is S, F7 is T, and F8 is A. In some embodiments, the variant CDR-H3 antibody further comprises CDR-L1, CDR-L2, CDR-L3, CDR-H1 and CDR-H2, wherein each sequence comprises SEQ ID NO: i, 2, 3, 4 And the sequence depicted in 5. In a 143939.doc • 68-201019961 embodiment, the antibodies further comprise a human subgroup III heavy chain framework consensus sequence. In one embodiment of such antibodies, the framework consensus sequence comprises a substitution at positions 71, 73 and/or 78. In some embodiments of such antibodies, position 71 is A, 73 is T and/or 78 is A. In one embodiment of such antibodies, the antibodies further comprise a human kappa light chain framework consensus sequence. In one embodiment, the antibody of the invention comprises a variant CDR-L2, wherein Β6 is V. In some embodiments, the variant CDR-L2 antibody further comprises CDR-L1, CDR-L3, CDR-H1, CDR-H2 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 3, 4, 5 And the sequence depicted in 6. In some embodiments, the variant CDR-L2 antibody further comprises CDR-L1, CDR-L3, CDR-H1, CDR-H2 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 3, 4, 5 And the sequence depicted in 7. In some embodiments, the variant CDR-L2 antibody further comprises CDR-L1, CDR-L3, CDR-H1, CDR-H2 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 3, 4, 5 And the sequence depicted in 8. In some embodiments, the antibodies further comprise a human subgroup III heavy chain framework consensus sequence. In one embodiment of such antibodies, the framework consensus sequence comprises a substitution at positions 71, 73 and/or 78. In some embodiments of such antibodies, position 71 is A, 73 is T and/or 78 is A. In one embodiment of such antibodies, the antibodies further comprise a human kappa light linkage framework consensus sequence. In one embodiment, an antibody of the invention comprises a variant CDR-H2, wherein Ε14 is Τ' Ε15 is Κ' and Ε17 is Ε. In one embodiment, an antibody of the invention comprises a variant CDR-H2, wherein Ε17 is Ε» In some embodiments, the variant CDR-H3 antibody further comprises CDR-L1, CDR-L2, CDR-L3, 143939.doc - 69-201019961 CDR-H1 and CDR-H3, each of which comprises the sequence depicted in SEQ ID NO: 1, 2, 3, 4 and 6. In some embodiments, the variant CDR-H2 antibody further comprises CDR-L1, CDR-L2, CDR-L3, CDR-H1 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 2, 3, 4 And the sequence depicted in 7. In some embodiments, the variant CDR-H2 antibody further comprises CDR-L1, CDR-L2, CDR-L3, CDR-H1 and CDR-H3, wherein each sequence comprises SEQ ID NO: 1, 2, 3, 4 And the sequence depicted in 8. In some embodiments, the antibodies further comprise a human subgroup III heavy chain framework consensus sequence. In one embodiment of such antibodies, the framework common sequence comprises a substitution at positions 71, 73 and/or 78. In some embodiments of such antibodies, position 71 is A, 73 is T and/or 78 is A. In one embodiment of such antibodies, the antibodies further comprise a human kappa light chain framework consensus sequence. In other embodiments, a c-met antibody of the invention specifically binds at least a portion of a c-met Sema domain or a variant thereof. In one example, an antagonist antibody of the invention specifically binds to at least one sequence selected from the group consisting of LDAQT (SEQ ID NO: 25) (eg, residue 269-273 of c_met), LTEKRKKRS (SEQ ID NO: 26) (eg residue 3GO-308 of c_met), KPDSAEPM (SEQ ID NO: 27) (eg residues 350-357 of c-met) and NVRCLQHF (SEQ ID NO: 28) (eg residues of c-met) 381-388). In one embodiment, an antagonist antibody of the invention specifically binds to a conformational epitope formed by at least one or a portion of a sequence selected from the group consisting of LDAQT (SEQ ID NO: 25) (eg, c -met residue 269-273), LTEKRKKRS (SEQ ID NO: 26) (eg, 143939.doc • 70. 201019961 c_met residue 300-308), KPDSAEPM (SEQ ID NO: 27) (eg c-met Residues 350-357) and NVRCLQHF (SEQ ID NO: 28) (e.g., residues 381-388 of c-met). In one embodiment, an antagonist antibody of the invention specifically binds to an amino acid having at least 50%, 60%, 70%, 80%, 90%, 95%, 98% sequence identity or similarity to the following sequences Sequence: LDAQT (SEQ ID NO: 25), LTEKRKKRS (SEQ ID NO: 26), KPDSAEPM (SEQ ID NO: 27) and/or NVRCLQHF (SEQ ID NO: 28). In one aspect, the anti-c-met antibody comprises at least one feature that promotes heterodimerization of the Fc sequence within the antibody fragment while minimizing homodimerization. This (etc.) feature improves the yield and/or purity and/or homogeneity of the immunoglobulin population. In one embodiment, the antibody comprises an Fc mutation that constitutes "杵" and "white" as described in the following literature: WO 2005/063816; Ridgeway, J et al, Prot Eng (1996) 9: 617-21; Zhu Z et al., Prot Sci (1997) 6:781-8. For example, the 臼 mutation can be one or more of T366A, L368A, and/or Y407V in the Fc polypeptide, and the hole mutation can be T366W. Anti-HGF antibodies are well known in the art. See, for example, Kim KJ et al, Clin Cancer Res. (2006) 12(4): 1292-8; WO 2007/115049; WO 2009/002521; WO 2007/143098; WO 2007/017107; WO 2005/017107; L2G7; AMG-102. A c-met receptor molecule or a fragment thereof that specifically binds to HGF can be used in the method of the present invention, for example, to bind and isolate the HGF protein' to prevent its conduction signal. The c-met receptor molecule or its HGF binding fragment is preferably in a soluble form. In some embodiments, the soluble form of the receptor has an inhibitory effect on the biological activity of the c-143939.doc-71-201019961 met protein by binding to HGF, thereby preventing its binding to its natural presence on the surface of the stem cell. Receptor. Also included are c-met receptor fusion proteins, examples of which are described below. The soluble c-met receptor protein or the entrapped c-met receptor protein of the present invention includes a c-met receptor protein that is not immobilized on the cell surface via a transmembrane domain. Thus, the soluble form of the c-met receptor (including chimeric receptor proteins), although capable of binding to HGF and inactivating it, does not comprise a transmembrane domain and is therefore generally not associated with cell membranes of cells expressing the molecule. See, for example, Kong-Beltran, et al., Cancer Cell (2004) 6(1): 75-84. HGF molecules or fragments thereof can be used in the methods of the invention, and such HGF molecules or fragments thereof specifically bind to c-met and block or reduce the activation of c-met, thereby preventing its conduction signal. An aptamer is a nucleic acid molecule that forms a tertiary structure that specifically binds to a target molecule, such as an HGF polypeptide. The production of aptamers and their therapeutic use are well established in this technology. See, for example, U.S. Patent No. 5,475,096. The HGF aptamer is a poly(ethylene glycol) modified nucleotide that adopts a three-dimensional configuration that enables it to bind to extracellular HGF. Further information on aptamers can be found in U.S. Patent Application Publication No. 20060148748. A peptidic body is a peptide sequence linked to an amino acid sequence encoding a fragment or a portion of an immunoglobulin molecule. The polypeptide may be derived from any random sequence selected for any method of specific binding, including but not limited to phage display technology. In a preferred embodiment, the selected polypeptide can be linked to an amino acid sequence encoding the Fc portion of an immunoglobulin. Peptides that specifically bind to and antagonize HGF or c-met are also suitable for use in the methods of the invention. 143939.doc -72- 201019961

c-met拮抗劑包括小分子,諸如US 5,792,783、US 5,834,504、US 5,880,141、US 6,297,238、US 6,599,902、 US 6,790,852 > US 2003/0125370 ' US 2004/0242603、US 2004/0198750、US 2004/0110758、US 2005/0009845、US 2005/0009840 、 US 2005/0245547 、 US 2005/0148574 、 US 2005/0101650 、 US 2005/0075340 、 US 2006/0009453 、 US 2006/0009493、WO 98/007695 ' WO 2003/000660 ' WO 2003/087026、WO 2003/097641、WO 2004/076412、WO 2005/004808、WO 2005/121125、WO 2005/030140、WO 2005/070891、WO 2005/080393、WO 2006/014325、WO 2006/021886 ' WO 2006/021 881、WO 2007/103308 中描述 之化合物。PHA-665752為c-met催化活性以及多種腫瘤細 胞之細胞生長、細胞運動、侵襲及形態之小分子ATP競爭 性活性位點抑制劑(Ma等人,(2005) Clin. Cancer Res· 11:2312-2319 ; Christensen 等人,(2003) Cancer Res· 63:7345-7355) ° EGFR拮抗劑 EGFR拮抗劑包括抗體,諸如稱為尼妥珠單抗 (nimotuzumab)(YM Biosciences)之人類化單株抗體;完全 人類 ABX-EGF(帕尼單抗(panitumumab),Abgenix Inc·); 以及稱為 Ε1·1、E2.4、Ε2·5、E6.2、E6.4、E2.ll、Ε6·3及 E7.6.3且描述於US 6,235,883 中之完全人類抗體;MDX-447(Medarex Inc·)。帕妥珠單抗(2C4)為人類化抗體,其直 接結合HER2,但干擾HER2-EGFR二聚,從而抑制EGFR傳 143939.doc -73- 201019961 導信號。結合EGFR之抗體之其他實例包括MAb 579(ATCC CRL HB 8506)、MAb 455(ATCC CRL HB8507)、MAb 225(ATCC CRL 8508)、MAb 528(ATCC CRL 8509)(參見 Mendelsohn等人之美國專利第4,943,533號)及其變異體, 諸如嵌合225(C225或西妥昔單抗;ERBUTIX®)及再成型人 類 225(H225)(參見 WO 96/40210, Imclone Systems Inc.); IMC-11F8,一種靶向EGFR之完全人類抗體(Imclone);結 合11型突變£〇?11之抗體(美國專利第5,212,290號);如美國 專利第5,891,996號所述之結合EGFR之人類化及嵌合抗 體;及結合EGFR之人類抗體,諸如ABX-EGF(參見WO 98/50433,Abgenix) ; EMD 55900(Stragliotto等人,£狀*/· 32A:636-640 (1996)) ; EMD7200(馬妥珠單抗),一 種針對EGFR之人類化EGFR抗體,其與EGF及TGF-α競爭 結合EGFR ;及mAb 806或人類化mAb 806(Johns等人,X 价〇/· 279(29):30375-30384 (2004))。抗 EGFR 抗體可 與細胞毒性劑結合,從而產生免疫結合物(參見例如 EP659,439A2,Merck Patent GmbH) 〇 適用於本發明方法之抗EGFR抗體包括以足夠親和力及 特異性結合EGFR且可降低或抑制EGFR活性之任何抗體。 所選抗體通常應對EGFR具有足夠強的結合親和力’例如 抗體結合人類EGFR之Kd值可在1〇〇 nM-1 pM之間。抗體親 和力可利用以下方法測定,例如基於表面電漿共振之檢定 (諸如PCT申請公開案第WO 2005/012359號中所述之 BIAcore檢定)、酶聯免疫吸附檢定(ELISA)及競爭檢定(例 143939.doc • 74- 201019961 如RIA)。較佳可使用本發明之抗c-met抗體作為靶向及干 擾涉及EGFR/EGFR配位體活性之疾病或病狀之治療劑。另 外,可使抗體經受其他生物活性檢定,例如以評估其作為 治療劑之效用。該等檢定在此項技術中為已知的且視靶抗 原及抗體之預定用途而定。 雙特異性抗體為對至少兩種不同抗原決定基具有結合特 異性之抗體》例示性雙特異性抗體可結合EGFR及c-met。 在另一實例中,例示性雙特異性抗體可結合同一蛋白(例 如c-met蛋白)之兩種不同抗原決定基。或者,c-met或 EGFR臂可與結合白血球上之觸發分子之臂組合以便在表 現c-met或EGFR之細胞中聚集細胞防禦機制,該觸發分子 諸如T細胞受體分子(例如CD2或CD3)或IgG之Fc受體 (FcyR)(諸如 FcyRI (CD64)、FcyRII (CD32)及 FcyRIII (CD1 6))。雙特異性抗體亦可用於將細胞毒性劑定位於表 現EGFR或c-met之細胞中。此等抗體具有EGFR或c-met結 合臂及結合細胞毒性劑(例如沙泊寧(saporin)、抗干擾素-α、長春花屬生物驗(vinca alkaloid)、蓖麻毒素A鏈、甲胺 喋呤或放射性同位素半抗原)之臂。雙特異性抗體可製備 為全長抗體或抗體片段(例如F(ab')2雙特異性抗體)。 EGFR拮抗劑亦包括小分子,諸如:US5616582 ' US5457105 、 US5475001 、 US5654307 、 US5679683 、 US6084095 、 US6265410 、 US6455534 、 US6521620 、 US6596726 、 US6713484 、 US5770599 、 US6140332 、 US5866572 、 US6399602 ' US6344459 ' US6602863 、 143939.doc -75- 201019961 US6391874、WO 9814451、WO 9850038、WO 9909016、 WO 9924037、WO 9935146、WO 0132651、US6344455、 US5760041、US6002008、US5747498 中描述之化合物。特 定小分子EGFR拮抗劑包括OSI-774 (CP-358774,埃羅替 尼,OSI Pharmaceuticals) ; PD 183805 (CI 1033,N-[4-[(3 -氣-4-氟苯基)胺基]-7-[3-(4-嗎咐基)丙氧基]-6-啥σ坐琳 基]-2-丙烯醯胺二鹽酸鹽,Pfizer Inc.) ; Iressa®(ZD 1839, 吉非替尼,AstraZeneca) ; ZM 105180 ((6-胺基-4-(3 -曱基 苯基-胺基)-喹唑啉,Zeneca) ; BIBX-1382 (N8-(3-氯-4-氟-苯基)-N2-(l -甲基-哌啶-4-基)-嘧啶并[5,4-d]嘧啶-2,8-二 胺,Boehringer Ingelheim) ; PKI-166 ((R)-4-[4-[(l-苯基乙 基)胺基]-1Η-π比洛并[2,3-d]嘴咬-6-基]-苯酌·);(R)-6-(4-經 基苯基)-4-[(1-苯基乙基)胺基]-7H-°比略并[2,3-d]°^咬); CL-387785 (N-[4-[(3- >臭苯基)胺基]-6-啥峻琳基]-2-丁快酿 胺);EKB-569 (N-[4-[(3-氣_4_氟苯基)胺基]-3-氰基-7-乙氧 基-6-哇琳基]-4-(二甲基胺基)-2- 丁稀酿胺);拉帕替尼 (lapatinib)(Tykerb, GlaxoSmithKline) ; ZD6474 (Zactima, AstraZeneca) ; CUDC-lOl(Curis);卡奈替尼(canertinib) (CI-1033) ; AEE788 (6·[4-[(4-乙基-1-哌嗪基)曱基]苯基]-1^-[(11^)-1-苯基乙基]-711-11比0各并[2,3-(1]嘴咬-4-胺,1^〇 2003013541,Novartis)及ΡΚΙ166 (4-[4-[[(lR)-l-苯基乙基] 胺基]-7H-吡咯并[2,3-d]嘧啶-6-基]-笨酚,WO 9702266, Novartis) °C-met antagonists include small molecules such as US 5,792,783, US 5,834,504, US 5,880,141, US 6,297,238, US 6,599,902, US 6,790,852 > US 2003/0125370 ' US 2004/0242603, US 2004/0198750, US 2004/0110758, US 2005/0009845, US 2005/0009840, US 2005/0245547, US 2005/0148574, US 2005/0101650, US 2005/0075340, US 2006/0009453, US 2006/0009493, WO 98/007695 'WO 2003/000660 ' WO 2003/087026, WO 2003/097641, WO 2004/076412, WO 2005/004808, WO 2005/121125, WO 2005/030140, WO 2005/070891, WO 2005/080393, WO 2006/014325, WO 2006/021886 ' WO Compounds described in 2006/021 881, WO 2007/103308. PHA-665752 is a small molecule ATP competitive active site inhibitor of c-met catalytic activity and cell growth, cell movement, invasion and morphology of various tumor cells (Ma et al., (2005) Clin. Cancer Res 11:2312 -2319; Christensen et al, (2003) Cancer Res· 63:7345-7355) ° EGFR antagonist EGFR antagonists include antibodies, such as humanized monoclonal antibodies called nimotuzumab (YM Biosciences) Complete human ABX-EGF (panitumumab, Abgenix Inc.); and known as Ε1·1, E2.4, Ε2·5, E6.2, E6.4, E2.ll, Ε6·3 And fully human antibody of E7.6.3 and described in US 6,235,883; MDX-447 (Medarex Inc.). Pertuzumab (2C4) is a humanized antibody that binds directly to HER2 but interferes with the dimerization of HER2-EGFR, thereby inhibiting EGFR signaling 143939.doc-73-201019961. Other examples of antibodies that bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see U.S. Patent No. 4,943,533 to Mendelsohn et al. And its variants, such as chimeric 225 (C225 or cetuximab; ERBUTIX®) and reshaped human 225 (H225) (see WO 96/40210, Imclone Systems Inc.); IMC-11F8, a target A fully human antibody to EGFR (Imclone); an antibody that binds to a type 11 mutation of 〇11 (U.S. Patent No. 5,212,290); a humanized and chimeric antibody that binds to EGFR as described in U.S. Patent No. 5,891,996; Human antibodies that bind to EGFR, such as ABX-EGF (see WO 98/50433, Abgenix); EMD 55900 (Stragliotto et al., £*/·32A: 636-640 (1996)); EMD7200 (martuzumab) , a humanized EGFR antibody against EGFR that competes with EGF and TGF-α for binding to EGFR; and mAb 806 or humanized mAb 806 (Johns et al., X-valent ·/· 279(29): 30375-30384 (2004) ). The anti-EGFR antibody can bind to a cytotoxic agent to produce an immunoconjugate (see, for example, EP 659, 439 A2, Merck Patent GmbH). Anti-EGFR antibodies suitable for use in the methods of the invention include binding to EGFR with sufficient affinity and specificity and can reduce or inhibit Any antibody that is active in EGFR. The selected antibody will generally have a sufficiently strong binding affinity for EGFR', e.g., the antibody binding to human EGFR may have a Kd value between 1 〇〇 nM-1 pM. Antibody affinity can be determined by methods such as surface plasma resonance based assays (such as the BIAcore assay described in PCT Application Publication No. WO 2005/012359), enzyme-linked immunosorbent assay (ELISA), and competition assays (example 143939). .doc • 74- 201019961 as RIA). Preferably, the anti-c-met antibody of the present invention can be used as a therapeutic agent for targeting and interfering with diseases or conditions involving EGFR/EGFR ligand activity. In addition, the antibody can be subjected to other biological activity assays, e.g., to assess its utility as a therapeutic agent. Such assays are known in the art and depend on the intended use of the target antigen and antibody. Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplary bispecific antibodies can bind to EGFR and c-met. In another example, an exemplary bispecific antibody can bind to two different epitopes of the same protein (e. g., c-met protein). Alternatively, the c-met or EGFR arm can be combined with an arm that binds to a trigger molecule on a white blood cell to aggregate a cellular defense mechanism in a cell that exhibits c-met or EGFR, such as a T cell receptor molecule (eg, CD2 or CD3). Or Fc receptors of IgG (FcyR) (such as FcyRI (CD64), FcyRII (CD32) and FcyRIII (CD1 6)). Bispecific antibodies can also be used to localize cytotoxic agents in cells that exhibit EGFR or c-met. These antibodies have EGFR or c-met binding arms and binding cytotoxic agents (eg saporin, anti-interferon-α, vinca alkaloid, ricin A chain, methotrexate) The arm of a sputum or radioisotope hapten. Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g., F(ab')2 bispecific antibodies). EGFR antagonists also include small molecules such as: US5616582 'US5457105, US5475001, US5654307, US5679683, US6084095, US6265410, US6455534, US6521620, US6596726, US6713484, US5770599, US6140332, US5866572, US6399602 'US6344459 ' US6602863, 143939.doc-75- The compounds described in 201019961 US Pat. No. 6,391, 874, WO 9814451, WO 9850038, WO 9909016, WO 9924037, WO 9935146, WO 0132651, US6344455, US5760041, US6002008, US5747498. Specific small molecule EGFR antagonists include OSI-774 (CP-358774, erlotinib, OSI Pharmaceuticals); PD 183805 (CI 1033, N-[4-[(3-fluoro-4-fluorophenyl)amino) -7-[3-(4-Mercapto)propoxy]-6-啥σ 琳 ]]-2-propenylamine dihydrochloride, Pfizer Inc.; Iressa® (ZD 1839, Ji Fei Azin, AstraZeneca); ZM 105180 ((6-Amino-4-(3-mercaptophenyl-amino)-quinazoline, Zeneca); BIBX-1382 (N8-(3-chloro-4-fluoro) -phenyl)-N2-(l-methyl-piperidin-4-yl)-pyrimido[5,4-d]pyrimidine-2,8-diamine, Boehringer Ingelheim); PKI-166 ((R) -4-[4-[(l-phenylethyl)amino]-1Η-πpiro[2,3-d] mouth bite-6-yl]-benzene discretion·);(R)-6 -(4-Phenylphenyl)-4-[(1-phenylethyl)amino]-7H-° ratio slightly [2,3-d]°^ bite); CL-387785 (N-[ 4-[(3- >odor phenyl)amino]-6-啥 琳 基 ]]-2-butyl fast-acting amine); EKB-569 (N-[4-[(3- gas_4_fluorine) Phenyl)amino]-3-cyano-7-ethoxy-6-wowenyl]-4-(dimethylamino)-2-butanthene); lapatinib (Tykerb, GlaxoSmithKline) ; ZD6474 (Zactima, AstraZeneca) ; CUDC-lOl (Curis); Carnamate (canertinib) (CI-1033) ; AEE788 (6·[4-[(4-ethyl-1-piperazinyl)indolyl]phenyl]-1^-[(11^)-1-phenyl) Base]-711-11 than 0 each [2,3-(1] mouth bite-4-amine, 1^〇2003013541, Novartis) and ΡΚΙ166 (4-[4-[[(lR)-l-phenyl) Ethyl]amino]-7H-pyrrolo[2,3-d]pyrimidin-6-yl]-indolol, WO 9702266, Novartis) °

在一特定實施例中,EGFR拮抗劑具有根據US 143939.doc -76· 201019961In a specific embodiment, the EGFR antagonist has according to US 143939.doc -76· 201019961

5,75 7,498(以引用之方式併入本文中)之通式I5,75 7,498 (incorporated herein by reference)

其中: m為1、2或3 ; 鹵基、羥基、羥 三氟甲基及-(Cr S 或 NH ; 各R1獨立地選自由以下組成之群:氫 胺基、羧基、硝基、胍基、脲基、氰基 C4伸烧基)-W-(苯基),其中w為單鍵、〇 或各R1獨立地選自R9及經氰基取代2Ci_C4烷基,其中 R9係選自由以下組成之群:R5、_〇R6、_NR6R6、_c(〇)r7 、-NHOR5、-0C(0)R6、氰基、Aj_YR5 ; r、Ci C4^ 基,R獨立地為氫或R5; “為“、_〇r6或-Nr6r6 ; a係選 自N-哌啶基、N-嗎啉基、N_吡咯啶基、4_R6_哌嗪_丨_基、 咪唑-1-f、4-»比啶_小基、_(CiC4伸烷基)(c〇2H)、苯 氧基笨基、苯基硫基、CVC4烯基及-(CVC4伸烷 基)C(0)NR R,且Y為s、8〇或8〇2;其中r5、_〇尺6及 魏R6中6之燒基部分視情況經⑴個齒基取代基取代,且 R 〇R及NR R中之垸基部分視情況經1或2個R9基團取 視情況選用之取代基之烧基部分視情況經 子"Γ ’限制條件為兩個雜原子不連接於同-碳原 143939.doc -77· 201019961 或各R1獨立地選自-NHS02R5、鄰苯二醯亞胺基_(Cl_c4)-烧基磺酿基胺基、苯甲醯胺基、苯磺醯基胺基、3_苯基膝 基、2-側氧基吡咯啶基、2,5_二側氧基吡咯啶_丨_基及 R1G-(c2-c4)-烷醯基胺基,其中Ri〇選自鹵基、_or6、c2_e4 烷醯氧基、-C(〇)R7及_NR6R6 ;且其中該等基團_ nhso2r5、鄰苯二醯亞胺基_(Cl_c4)_烷基磺醯基胺基、苯 甲酿胺基、苯磺醯基胺基、3-苯基脲基、2-侧氧基吡咯咬_ 卜基、2,5-二側氧基吡咯啶_丨_基及Ri〇_(C2_C4)_烷醯基胺基 視情況經1或2個獨立地選自鹵基、Ci-C4烷基、氰基、甲 烷磺醯基及Ci-C*烷氧基之取代基取代; 或兩個R1基團連同其所連接之碳一起形成5_8員環,該 環包括1或2個選自〇、s及N之雜原子; R2為氫或CrC6院基,該CrC6烧基視情況經1至3個獨立 地選自i基、CVC4烷氧基、_NR6R6及-so2r5之取代基取 代; η為1或2 ’且各R3獨立地選自氫、鹵基、經基、c〗_c6燒 基、烷氧基,其中該等R3基團之烷基部分 視情況經1至3個獨立地選自鹵基、烷氧基、_NR6R6 及-SO2R之取代基取代;且 R4為疊氮基或-(乙炔基)-Rn ’其中R11為氫或Cl-C6烷 基,該C「C6烷基視情況經羥基、_〇R6或_NR6R6取代。 在一特定實施例中,EGFR拮抗劑為選自由以下組成之 群之式I化合物: (6,7-二甲氧基喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6,7-二 143939.doc -78 · 201019961 甲氧基喹唑啉-4-基)-[3-(3'-羥基丙炔-1-基)笨基]-胺;[3_ (2-(胺基曱基)_乙炔基)苯基]_(6,7_二甲氧基嗜。坐琳_4-基)_ 胺;(3-乙炔基苯基)_(6-墙基噎唾淋-4-基)-胺;(6,7-二甲氧 基喹唑啉-4-基)-(4-乙炔基苯基)-胺;(6,7-二甲氧基喹唑 琳-4-基)-(3-乙快基-2-曱基苯基)-胺;(6-胺基啥β坐琳_4-基)-(3-乙炔基苯基)-胺;(3-乙炔基苯基)-(6-甲烷磺醯基胺 基喹唑啉-4-基)-胺;(3-乙炔基笨基)-(6,7-亞甲基二氧基喧 »坐-4-基)-胺;(6,7-二曱氧基喹唑琳-4-基)-(3-乙炔基-6-甲基苯基)-胺;(3-乙炔基苯基)-(7-硝基啥坐琳_4_基)_胺; (3-乙炔基苯基H6-(f-曱笨磺醯基胺基)喹唑啉_4-基]-胺; (3-乙炔基苯基)-{6-[2'-鄰苯二酿亞胺基-乙_1’_基-績酿基胺 基]啥β坐琳_4-基}-胺,(3-乙快基苯基)-(6-脈基喧β坐嘴_4_ 基)-胺;(7-胺基喹唑啉-4-基)-(3·乙炔基苯基)_胺;(3_乙炔 基苯基)-(7-甲氧基喹唑啉-4-基)-胺;(6-甲氧羰基喹唑啉_ 4-基)-(3-乙炔基苯基)-胺;(7-曱氧羰基喹唑啉_4_基)_(3_乙 快基苯基)-胺;[6,7-雙(2-曱氧基乙氧基)啥唾琳·4_基]_(3-乙炔基苯基)-胺;(3-疊氮基苯基)-(6,7-二甲氧基喹唑啉_4_ 基)-胺,(3 -疊氣基氣本基)-(6,7 -二甲乳基喧吐琳-4-基)_ 胺;(4-疊氮基苯基Η6,7-二曱氧基喹唑啉-4·基)-胺;(3-乙 炔基苯基)-(6-甲烷磺醯基-喹唑啉-4-基)·胺;(6-乙烷硫基-喧β坐琳-4-基)-(3-乙炔基苯基)-胺;(6,7-二甲氧基-喧β坐琳_ 4-基)-(3-乙炔基-4-氟-苯基.)-胺;(6,7-二曱氧基-喹唑啉_4_ 基)-[3-(丙炔-1,-基)_苯基]-胺;[6,7-雙-(2-甲氧基-乙氧基)-啥°坐琳-4 -基]-(5 -乙快基_2 -甲基-笨基)·胺;[6,7 -雙- (2 -曱氧 143939.doc • 79- 201019961 基-乙氧基)-喹唑啉-4-基]-(3-乙炔基-4-氟-苯基)-胺;[6,7-雙-(2-氣-乙氧基)·喹唑啉_4_基]_(3_乙炔基-苯基)_胺;[6_ (2-氣-乙氧基)-7-(2-甲氧基-乙氧基)-喹唑啉_4-基]-(3-乙炔 基-苯基)-胺;[6,7-雙-(2-乙醯氧基-乙氧基)_喹唑啉_4_基]-(3-乙炔基-苯基)-胺;2-[4-(3-乙炔基-苯基胺基)-7-(2-羥基-乙氧基)-喹唾啉-6-基氧基]-乙醇;[6-(2-乙醯氧基-乙氧 基)-7-(2-曱氧基-乙氧基)·喹唑啉_4_基]_(3_乙炔基-苯基)_ 胺’[7-(2-氣-乙氧基)_6-(2-甲氧基-乙氧基)-喧!1坐琳_4-基]_ (3-乙炔基-苯基)_胺;[7_(2_乙醯氧基-乙氧基)_6_(2_曱氧 基-乙氧基)-喹唑啉-4-基]-(3_乙炔基·苯基)-胺;2-[4-(3·乙 炔基-苯基胺基)-6-(2-羥基-乙氧基)-喹唑啉-7-基氧基]-乙 醇;2-[4-(3-乙炔基-苯基胺基甲氧基_乙氧基)_喹唑 啉-6-基氧基]-乙醇;2-[4-(3-乙炔基-苯基胺基)-6-(2-曱氧 基-乙氧基)-喹唑啉-7-基氧基]-乙醇;[6-(2-乙醯氧基-乙氧 基)-7-(2-曱氧基-乙氧基)_喹唑啉·4_基]_(3_乙炔基-苯基)_ 胺;(3-乙炔基-苯基曱氧基_乙氧基)_7-[2-(4-甲基-0辰嗪-1-基)-乙氧基]-喹唑啉_4_基卜胺;(3-乙炔基-苯基 [7-(2-甲礼基_乙氧基)_6-(2-嗎琳-4-基)-乙氧基)-嗜嗤琳_4_ 基卜胺:^^-二乙氧基喹唑啉-丨-基卜^-乙炔基苯基卜胺; (6,7-二丁氧基喹唑啉·丨-基^、乙块基苯基)_胺;(6,7•二異 丙氧基喹唑啉-1·基)-(3-乙炔基苯基)-胺;(6,7-二乙氧基啥 唑啉-1-基)-(3-乙炔基-2·曱基-苯基)-胺;[6,7-雙-(2-曱氧 基-乙氧基)-喹唑啉-1-基]-(3-乙炔基-2-曱基-苯基)-胺;(3_ 乙炔基苯基Η6-(2-羥基-乙氧基)-7-(2-甲氧基-乙氧基)·喹 143939.doc •80- 201019961 °坐琳-1-基]-胺;[6,7-雙-(2-經基-乙氧基)-啥》坐琳-i_基]_(3_ 乙炔基苯基)-胺;2-[4-(3-乙炔基-苯基胺基)-6-(2-甲氧基_ 乙氧基)-喧°坐琳-7-基氧基]_乙醇;(6,7-二丙氧基-喧唾琳_ 4-基)-(3-乙快基-苯基)-胺;(6,7-二乙氧基-啥β坐琳_4_基)_ (3-乙炔基-5-氟·苯基)-胺;(6,7-二乙氧基喹唑啉_4_基)_(3_ 乙炔基-4-氟-苯基)-胺;(6,7-二乙氧基-喧°坐琳-4-基)-(5-乙 快基-2-甲基-苯基)-胺;(6,7-二乙氧基-喧°坐琳-4-基)-(3 -乙 炔基-4-甲基-苯基)-胺;(6-胺基甲基-7-甲氧基-嗟η坐嘛_4_ 基)-(3-乙炔基·苯基)-胺;(6-胺基曱基-7-甲氧基-喹唑嘴_4-基)-(3 -乙快基苯基)-胺;(6-胺基幾基甲基-7·甲氧基_喧η坐 琳-4-基)-(3 -乙快基苯基)-胺;(6-胺基幾基乙基-7-甲氧基_ 喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6-胺基羰基甲基_7-乙 氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6-胺基羰基乙基_ 7-乙氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6-胺基幾基 甲基-7-異丙氧基-啥唾琳-4-基)-(3-乙炔基苯基)·胺;(6-胺 基羰基甲基-7-丙氧基-喹唑淋-4-基)-(3-乙炔基苯基)_胺; (6-胺基羰基甲基-7-曱氧基-喹唑琳-4-基)-(3 -乙炔基苯基)_ 胺,(6-胺基幾基乙基-7-異丙氧基-啥吐琳-4-基)-(3-乙炔基 苯基)-胺;及(6-胺基叛基乙基-7-丙氧基-喧β坐琳_4·基)_(3_ 乙炔基苯基)-胺;(6,7-二乙氧基啥唾琳_1_基)_(3·乙快基苯 基)-胺;(3-乙炔基苯基)-[6-(2-羥基-乙氧基)-7-(2-曱氧基-乙氧基)-喹唑啉-1-基]-胺;[6,7-雙-(2-羥基-乙氧基)_喹唑 琳-1-基]-(3-乙炔基苯基)-胺;[6,7-雙-(2-甲氧基-乙氧基)_ 喹唑啉-1-基]-(3-乙炔基苯基)-胺;(6,7-二甲氧基喹唑啉_ι_ 143939.doc • 81 · 201019961 基M3-乙快基苯基)-胺·’(3-乙快基笨基)_(6_甲烷磺醯基胺 基-喧唾琳-1-基)-胺;及(6-胺基-嗜唾琳_1_基)_(3乙快美苯 基)-胺。 在一特定實施例中’式I之EGFR拮抗劑為^乂^乙炔基苯 基)-6,7-雙(2-曱氧基乙氧基)·4-喧啥琳胺。在一特定實施例 中,EGFR拮抗劑Ν-(3-乙炔基苯基)-6,7-雙(2_曱氧基乙氧 基喹唑啉胺係呈鹽酸鹽形式。在另—特定實施例中, EGFR拮抗劑Ν-(3-乙炔基苯基)-6,7-雙(2-甲氧基乙氧基)_4_ 喧°坐琳胺係呈實質上均勻之結晶多晶型物形式(在W〇 01/34,574中描述為多晶型物Β),該形式展現X光粉末繞射 圖之特徵峰以2Θ。表示在約6.26、12.48、13.39、16.96、 20.20、21.10、22.98、24.46、25.14 及 26.91 處。Ν-(3-乙快 基苯基)-6,7-雙(2-曱氧基乙氧基)-4-啥峻琳胺之該多晶型物 形式係稱為Tarceva™以及OSI-774、CP-358774及埃羅替 尼。 式I化合物、其醫藥學上可接受之鹽及前藥(下文稱為活 性化合物)可由已知適用於製備化學相關化合物之任何方 法製備。活性化合物一般可如US 5,747,498中所揭示之一 般流程I所示使用適當經取代之胺由適當經取代之喹唑啉 製備: 143939.doc -82· 201019961Wherein: m is 1, 2 or 3; halo, hydroxy, hydroxytrifluoromethyl and -(Cr S or NH; each R1 is independently selected from the group consisting of: aminino, carboxyl, nitro, fluorenyl , ureido, cyano C4 alkyl) -W-(phenyl), wherein w is a single bond, hydrazine or each R1 is independently selected from R9 and cyano substituted 2Ci_C4 alkyl, wherein R9 is selected from the group consisting of Group: R5, _〇R6, _NR6R6, _c(〇)r7, -NHOR5, -0C(0)R6, cyano, Aj_YR5; r, Ci C4^, R is independently hydrogen or R5; , _〇r6 or -Nr6r6; a is selected from the group consisting of N-piperidinyl, N-morpholinyl, N-pyrrolidinyl, 4_R6_piperazine-丨-yl, imidazole-1-f, 4-»pyridinium _ small group, _(CiC4 alkylene) (c〇2H), phenoxyphenyl, phenylthio, CVC4 alkenyl and -(CVC4 alkyl) C(0)NR R, and Y is s , 8〇 or 8〇2; wherein the calcined moiety of 6 in r5, _〇6 and Wei R6 is substituted by (1) a dentate substituent, and the thiol moiety of R 〇R and NR R is optionally 1 or 2 R9 groups are taken as appropriate. The alkyl group of the substituent is selected as the case where the two heteroatoms are not connected to the same carbon source. 43939.doc -77· 201019961 or each R1 is independently selected from -NHS02R5, phthalimido-(Cl_c4)-alkylsulfonylamino, benzhydrylamine, phenylsulfonylamino, a 3-phenylphenyl group, a 2-sided oxypyrrolidinyl group, a 2,5-di-oxypyrrolidinyl group, and an R1G-(c2-c4)-alkylmercaptoamine group, wherein Ri is selected from the group consisting of Halogen, _or6, c2_e4 alkoxy, -C(〇)R7 and _NR6R6; and wherein the groups _nhso2r5, phthalic acid iodide-(Cl_c4)-alkylsulfonylamino group, Benzoylamino, phenylsulfonylamino, 3-phenylureido, 2-sided oxypyrrole, 2,5-di-oxypyrrolidine, and Ri〇_( The C2_C4)-alkylamino group is optionally substituted by 1 or 2 substituents independently selected from halo, Ci-C4 alkyl, cyano, methanesulfonyl and Ci-C* alkoxy; or The R1 group together with the carbon to which it is attached forms a 5-8 membered ring comprising 1 or 2 heteroatoms selected from the group consisting of ruthenium, s and N; R2 is hydrogen or a CrC6 base, the CrC6 alkyl group being subjected to 1 Substituting to three substituents independently selected from the group consisting of i group, CVC4 alkoxy group, _NR6R6 and -so2r5; η is 1 or 2' and each R3 is independently selected from hydrogen and halogen a radical, a radical, a c-alkyl group, an alkoxy group, wherein the alkyl moiety of the R3 group is optionally one to three substituents independently selected from the group consisting of halo, alkoxy, _NR6R6 and -SO2R Substituting; and R4 is azido or -(ethynyl)-Rn ' wherein R11 is hydrogen or Cl-C6 alkyl, and the C"C6 alkyl group is optionally substituted with hydroxy, _R6 or _NR6R6. In a specific embodiment, the EGFR antagonist is a compound of formula I selected from the group consisting of: (6,7-dimethoxyquinazolin-4-yl)-(3-ethynylphenyl)-amine (6,7-two 143939.doc -78 · 201019961 methoxyquinazolin-4-yl)-[3-(3'-hydroxypropyn-1-yl)phenyl]-amine; [3_ ( 2-(Aminoguanidino)-ethynyl)phenyl]-(6,7-dimethoxy-isocyanin- 4-yl)-amine; (3-ethynylphenyl)-(6-wall (6,7-dimethoxyquinazolin-4-yl)-(4-ethynylphenyl)-amine; (6,7-dimethoxy Quinazolin-4-yl)-(3-ethylidyl-2-mercaptophenyl)-amine; (6-amino 啥β坐琳-4-yl)-(3-ethynylphenyl)- Amine; (3-ethynylphenyl)-(6-methanesulfonylaminoquinazolin-4-yl)-amine; (3-ethynylphenyl)-(6,7-methylenedioxy (喧, -4-yl)-amine; (6,7-dimethoxy quinazolin-4-yl)-(3-ethynyl-6-methylphenyl)-amine; (3-acetylene Phenyl)-(7-nitroindole _4_yl)-amine; (3-ethynylphenyl H6-(f-indolesulfonylamino)quinazoline-4-yl]- Amine; (3-ethynylphenyl)-{6-[2'-ortho Di-n-imino-ethyl-7'-yl-y-glycosylamino] 啥β sitin _4-yl}-amine, (3-ethyl-bromophenyl)-(6-fluorenyl 喧β sitting mouth _4_yl)-amine; (7-aminoquinazolin-4-yl)-(3.ethynylphenyl)-amine; (3-ethynylphenyl)-(7-methoxyquinazoline) 4-yl)-amine; (6-methoxycarbonylquinazolin-4-yl)-(3-ethynylphenyl)-amine; (7-fluorenyloxyquinazoline-4-yl)-( 3_B-phenylphenyl)-amine; [6,7-bis(2-decyloxyethoxy) 啥 琳 · 4 4 4 4 ; ; ; ; 3- 3- Azidophenyl)-(6,7-dimethoxyquinazolin-4-yl)-amine, (3-isole-based gas-based)-(6,7-dimethyllacyl oxime- 4-yl)-amine; (4-azidophenylphosphonium 6,7-dimethoxyoxyquinazolin-4-yl)-amine; (3-ethynylphenyl)-(6-methanesulfonyl) -quinazolin-4-yl)amine; (6-ethanethio-indolyl-4-yl)-(3-ethynylphenyl)-amine; (6,7-dimethoxy -喧β坐琳_ 4-yl)-(3-ethynyl-4-fluoro-phenyl.)-amine; (6,7-dimethoxy-quinazoline-4-yl)-[3-( Propyne-1,-yl)-phenyl]-amine; [6,7-bis-(2-methoxy-ethoxy)-啥°琳琳-4-yl]-(5-ethylhexyl-2-methyl-phenyl)amine; [6,7-bis-(2-oxo 143939.doc • 79- 201019961 base-ethoxyl )-quinazolin-4-yl]-(3-ethynyl-4-fluoro-phenyl)-amine; [6,7-bis-(2-a-ethoxy)quinazoline_4_ [7-(3-ethane-ethoxy)-7-(2-methoxy-ethoxy)-quinazoline-4-yl]- (3-ethynyl-phenyl)-amine; [6,7-bis-(2-acetoxy-ethoxy)-quinazoline-4-yl]-(3-ethynyl-phenyl) -amine; 2-[4-(3-ethynyl-phenylamino)-7-(2-hydroxy-ethoxy)-quinoxalin-6-yloxy]-ethanol; [6-(2 -Ethyloxy-ethoxy)-7-(2-decyloxy-ethoxy)quinazoline-4-yl]-(3-ethynyl-phenyl)-amine '[7-( 2-gas-ethoxy)_6-(2-methoxy-ethoxy)-oxime! 1 sits _4-yl]-(3-ethynyl-phenyl)-amine; [7_(2_ Ethyloxy-ethoxy)-6-(2-methoxy-ethoxy)-quinazolin-4-yl]-(3-ethynylphenyl)-amine; 2-[4-(3 Ethyl-phenylamino)-6-(2-hydroxy-ethoxy)-quinazolin-7-yloxy]-ethanol; 2-[4-(3-ethynyl-phenylamino) Methoxy-ethoxy) quinazoline -6-yloxy]-ethanol; 2-[4-(3-ethynyl-phenylamino)-6-(2-decyloxy-ethoxy)-quinazolin-7-yloxy ]-ethanol; [6-(2-acetoxy-ethoxy)-7-(2-decyloxy-ethoxy)-quinazoline·4-yl]-(3-ethynyl-benzene ()) amine; (3-ethynyl-phenyl fluorenyloxy-ethoxy)_7-[2-(4-methyl-Octosin-1-yl)-ethoxy]-quinazoline _ 4_Ketamine; (3-ethynyl-phenyl[7-(2-methylsulfanyl-ethoxy)-6-(2-morphin-4-yl)-ethoxy)-isophilic _ 4_ kibamine: ^^-diethoxyquinazoline-丨-kib^-ethynylphenyl-amine; (6,7-dibutoxyquinazoline·丨-yl^, B block Phenyl)-amine; (6,7•diisopropoxyquinazolin-1·yl)-(3-ethynylphenyl)-amine; (6,7-diethoxyoxazoline-1 -yl)-(3-ethynyl-2-indenyl-phenyl)-amine; [6,7-bis-(2-decyloxy-ethoxy)-quinazolin-1-yl]-( 3-ethynyl-2-indenyl-phenyl)-amine; (3_ethynylphenylphosphonium 6-(2-hydroxy-ethoxy)-7-(2-methoxy-ethoxy)·quino 143939 .doc •80- 201019961 ° sitin-1-yl]-amine; [6,7-bis-(2-trans-ethoxy-)-啥" sit Lin-i_基]_( 3-_ ethynylphenyl)-amine; 2-[4-(3-ethynyl-phenylamino)-6-(2-methoxy-ethoxy)-喧°坐琳-7-yloxy ]_Ethanol; (6,7-dipropoxy-hydrazinyl-4-yl)-(3-ethyl-p-phenyl)-amine; (6,7-diethoxy-啥β sitting _4_基)_(3-ethynyl-5-fluoro-phenyl)-amine; (6,7-diethoxyquinazolin-4-yl)-(3_ethynyl-4-fluoro-benzene (6,7-diethoxy-oxime-cylin-4-yl)-(5-ethylidyl-2-methyl-phenyl)-amine; (6,7-diethyl) Oxy-喧°坐琳-4-yl)-(3-ethynyl-4-methyl-phenyl)-amine; (6-aminomethyl-7-methoxy-嗟η sitting _4_ (6-Aminoalkynyl-7-methoxy-quinazolyl-4-yl)-(3-ethoxyphenyl)-amine; (6-Aminomethyl-7-methoxy-Nytyl-4-yl)-(3-ethoxyphenyl)-amine; (6-Aminoethylethyl-7- Methoxy-quinazolin-4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylmethyl-7-ethoxy-quinazolin-4-yl)-(3 -ethynylphenyl)-amine; (6-aminocarbonylethyl-7-ethoxy-quinazolin-4-yl)-(3-ethynylphenyl) -amine; (6-aminomethylmethyl-7-isopropoxy-indolyl-4-yl)-(3-ethynylphenyl)amine; (6-aminocarbonylmethyl-7) -propoxy-quinazolin-4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylmethyl-7-decyloxy-quinazoline-4-yl)-( 3-(ethynylphenyl)-amine, (6-aminoethylethyl-7-isopropoxy-oxiolin-4-yl)-(3-ethynylphenyl)-amine; -Amino-t- ethylethyl-7-propoxy-??-?? _4·yl)_(3_ethynylphenyl)-amine; (6,7-diethoxy 啥 琳 _1 _1 _ base )-(3·B-ylphenyl)-amine; (3-ethynylphenyl)-[6-(2-hydroxy-ethoxy)-7-(2-decyloxy-ethoxy)- Quinazolin-1-yl]-amine; [6,7-bis-(2-hydroxy-ethoxy)-quinazoline-1-yl]-(3-ethynylphenyl)-amine; [6 ,7-bis-(2-methoxy-ethoxy)-quinazolin-1-yl]-(3-ethynylphenyl)-amine; (6,7-dimethoxyquinazoline _ Ip_ 143939.doc • 81 · 201019961 base M3-ethyl-bromophenyl)-amine·'(3-ethyl-based phenyl)-(6-methanesulfonylamino-hydrazin-1-yl)- Amine; and (6-amino-saltyl-l-yl)-(3B-b-phenylene)-amineIn a particular embodiment, the EGFR antagonist of Formula I is ethoxylated phenyl)-6,7-bis(2-decyloxyethoxy) 4-indolylamine. In a specific embodiment, the EGFR antagonist Ν-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy quinazolinamine is in the form of the hydrochloride salt. In the examples, the EGFR antagonist Ν-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)_4_ 坐 ° sitalamine is a substantially homogeneous crystalline polymorph Form (described as polymorph 〇 in W〇01/34,574), which exhibits a characteristic peak of the X-ray powder diffraction pattern of 2 Θ, expressed at about 6.26, 12.48, 13.39, 16.96, 20.20, 21.10, 22.98, 24.46, 25.14 and 26.91. The polymorphic form of Ν-(3-ethyl-bromophenyl)-6,7-bis(2-decyloxyethoxy)-4-indenylamine is called TarcevaTM and OSI-774, CP-358774 and erlotinib. The compounds of formula I, their pharmaceutically acceptable salts and prodrugs (hereinafter referred to as active compounds) may be any of those known to be suitable for the preparation of chemically related compounds. Process Preparation. The active compounds can generally be prepared from the appropriately substituted quinazolines using the appropriate substituted amines as shown in General Scheme I, as disclosed in US 5,747,498: 143939.doc -82· 201019961

如流程I中所示,使適當4-取代之喹唑啉2(其中X為合適 的可置換離去基,諸如鹵基、芳氧基、烷基亞磺醯基、烧 基磺醯基(諸如三氟甲烷磺醯氧基)、芳基亞磺醯基、芳基 石黃醯基、石夕烧氧基、氰基、π比唾并、三嗤并或四β坐并), 143939.doc -83- 201019961 較佳4-氣喹唑啉與適當胺或胺鹽酸鹽4或5(其中R4係如上所 述且Y為Βι·、I或三氟甲烧_績醯氧基)在溶劑(諸如(ci·:) 醇、二甲基甲醯胺(DMF)、N_甲基料咬_2,、氯仿、^ 猜、四氫吱味(THF)、M-二喔垸"比錢其他非質子性容 劑)中反應。反應可在驗存在下實現,該驗較佳為驗金屬 或驗土金屬碳酸鹽或氫氧化物,或三級胺驗,諸如対、 2,6-二甲基吡啶、三甲基吡啶、N_甲基·嗎啉、三乙胺、扣 二曱基胺基-吡啶或N,N_二甲基苯胺。此等鹼在下文中稱 為合適的鹼。維持反應混合物在約周圍溫度至約溶劑回流 溫度、較佳約35°C至約回流溫度之溫度下,直至實質上不 可偵測到剩餘4-齒基喹唑啉,通常歷時約2至約24小時。 反應較佳在惰性氛圍(諸如乾燥氮氣)下執行。 一般以化學計量方式混合反應物。當使用胺4或5之鹽 (通常鹽酸鹽)之化合物使用胺鹼時,較佳使用過量胺鹼, 一般1當量過量之胺鹼。(或者,若不使用胺鹼,則可使用 過量之胺4或5)。 對於使用位阻胺4(諸如2·烷基-3_乙炔基苯胺)或極具反 應性之4-_基喹唑啉之化合物,較佳使用第三丁醇或極性 非質子性溶劑(諸如DMF或N_甲基吡咯啶酮)作為溶劑。 或者,使4-取代喹唑啉2(其中X為羥基或側氧基(且2•氮 經氫化))與四氣化碳及視情況支撐於惰性聚合物上的視情 況經取代之三芳基膦(例如聚合物支撐之三苯基膦, Alddch目錄號36,645-5,其為每公克樹脂含有3毫莫耳磷 之2%與二乙烯苯交聯的聚苯乙烯)在溶劑(諸如四氣化碳、 143939.doc • 84 · 201019961 氣仿、二氣乙烷、四氫呋喃、乙腈或其他非質子性溶劑或 其在〇物)中反應。維持反應混合物在約周圍溫度至回流 溫度、較佳約35。(:至回流溫度之温度下2至24小時❹使該 .混合物直接或在藉由例如真空蒸發而移除溶劑且添加合適 的替代溶劑(諸如(CVC6)醇、DMF、N-甲基吡咯啶酮、 吡啶或1,4-二噁烷)後與適當的胺或胺鹽酸鹽4或5反應。隨 後,維持反應混合物在約周圍溫度至溶劑回流溫度、較佳 約35°C至約回流溫度之溫度下,直至實質上完成產物形 成,通常歷時約2至約24小時。反應較佳在惰性氛圍(諸如 乾燥氮氣)下進行。 當使用化合物4(其中γ為Br、][或三氟曱烷磺醯氧基)作 為起始物質與喹唑啉2反應時,形成式3化合物(其中Ri、 R2、R3及Y係如上所述)。藉由使化合物3與合適的鈀試劑 (諸如肆(三苯基膦)鈀或雙(三苯基膦)二氣化鈀)在合適之路 易斯酸(Lewis acid)(諸如氣化亞銅)及合適之炔烴(諸如三 φ 甲基矽烷基乙炔、炔丙醇或3-(N,N-二甲基胺基)_丙炔)存 在下在溶劑(諸如二乙胺或三乙胺)中反應,使化合物3轉化 為式1化合物(其中R4為Rn乙炔基且Rn係如以上所定義)。 可藉由用重氮化劑(諸如酸及亞硝酸鹽(例如乙酸及 NaN〇2))處理化合物3(其中γ為NH2),接著用疊氮化物(諸 •如NaNs)處理所得產物,使化合物3轉化為化合物其中r4 為疊氮化物)。 為製備R1為胺基或羥胺基之式I化合物,使…為硝美之 相應式I化合物還原。 143939.doc -85 - 201019961 還原宜利用許多已知用於該等轉化之程序中之任一者來 進订。可例如藉由在反應惰性溶劑巾在合適之金屬催化劑 (一諸如把、10或鎳)存在下氫㈣基化合物來進行還原。另 種合適還原劑為例如活化金屬,諸如活化鐵(藉由用稀 酸溶液(諸如鹽酸)洗務鐵粉而產生)。因此,可例如藉由將 硝基化合物及活化金屬與濃鹽酸於溶劑(例如水與醇之混 合物:該醇例如甲醇或乙醇)中之混合物加熱至例如5(rc 至15(TC範圍内之溫度(宜加熱至…或約贼成行還原。 另一種合適類別之還原劑為鹼金屬連二亞硫酸鹽(諸如連 二亞硫酸鈉),其可在(Cl_C4)烷酸、(Ci_C6)烷醇、水或其 混合物中使用。 為製備R2或R3合併有-級或二級胺基部分(除意欲與啥 唑啉反應之胺基外)之式丨化合物,較佳在上述反應之前保 護該游離胺基,接著在與4_(取代)喹唑啉2進行上述反應之 後脫除保護基。 可使用數種熟知之氮保護基。該等基團包括(CiC6)烷氧 基羰基、視情況經取代之苯甲氧羰基、芳氧基羰基、三苯 曱基、乙烯氧基羰基、〇-硝基苯基磺醯基、二苯膦基 (diphenylphosphinyl)、對曱苯磺醯基及苯甲基。氮保護基 之加入可在約01至約50t:之溫度(較佳約周圍溫度),在三 級胺鹼(諸如三乙胺、二異丙基乙胺或吡啶,較佳三乙胺) 存在或不存在下,在氣化烴溶劑(諸如二氣甲烷或丨,2_二氣 乙烷)或醚溶劑(諸如乙二酵二甲醚、二乙二醇二曱醚或 THF)中進行。或者,且使用肖頓_鮑曼(sch〇tten_Baumann) 143939.doc -86- 201019961 條件連接保護基。 在化合物2與5之上述偶合反應之後,可利用熟習此項技 術者已知之去保護方法移除保護基,諸如對於經第三丁氧 基幾基保護之產物在二氣甲烷中用三氟乙酸處理。As shown in Scheme I, a suitable 4-substituted quinazoline 2 (wherein X is a suitable dissociable leaving group such as halo, aryloxy, alkylsulfinyl, alkylsulfonyl) Such as trifluoromethanesulfonyloxy), arylsulfinyl, aryl fluorenyl, oxime, cyano, π-salt, triterpene or tetra-β, and 143939.doc-83 - 201019961 Preferably 4-oxoquinazoline and the appropriate amine or amine hydrochloride 4 or 5 (wherein R4 is as described above and Y is Βι, I or trifluoromethane) in a solvent (such as (ci·:) alcohol, dimethylformamide (DMF), N_methyl bite_2, chloroform, ^ guess, tetrahydroanthracene (THF), M-dioxime " Reaction in an aprotic solvent). The reaction can be carried out in the presence of a test, preferably a metal or soil test metal carbonate or hydroxide, or a tertiary amine test such as ruthenium, 2,6-lutidine, trimethylpyridine, N _Methyl morpholine, triethylamine, decylamino-pyridine or N,N-dimethylaniline. These bases are hereinafter referred to as suitable bases. Maintaining the reaction mixture at a temperature from about ambient temperature to about the reflux temperature of the solvent, preferably from about 35 ° C to about reflux temperature, until substantially no residual 4-dentate quinazoline is detected, typically from about 2 to about 24 hour. The reaction is preferably carried out under an inert atmosphere such as dry nitrogen. The reactants are typically mixed stoichiometrically. When an amine base is used as the compound using the salt of the amine 4 or 5 (usually the hydrochloride salt), it is preferred to use an excess of the amine base, usually 1 equivalent of an excess of the amine base. (Alternatively, if an amine base is not used, an excess of amine 4 or 5 can be used). For compounds using a hindered amine 4 (such as 2·alkyl-3_ethynylaniline) or a highly reactive 4-aminoquinazoline, it is preferred to use a third butanol or a polar aprotic solvent (such as DMF or N-methylpyrrolidone) as a solvent. Alternatively, a 4-substituted quinazoline 2 (wherein X is a hydroxy or pendant oxy group (and 2 • nitrogen is hydrogenated)) and a tetra-carbonated carbon and, optionally, a substituted triaryl group supported on an inert polymer Phosphine (eg, polymer supported triphenylphosphine, Alddch Cat. No. 36, 645-5, which contains 2 millimoles of phosphorus per 2 grams of phosphorus crosslinked with divinylbenzene) in a solvent such as four gases Carbon, 143939.doc • 84 · 201019961 Gas, di-ethane, tetrahydrofuran, acetonitrile or other aprotic solvent or its reaction in the sputum. The reaction mixture is maintained at about ambient temperature to reflux temperature, preferably about 35. (: 2 to 24 hours at the temperature of the reflux temperature, the mixture is removed directly or by evaporation, for example, by vacuum and a suitable replacement solvent (such as (CVC6) alcohol, DMF, N-methylpyrrolidine) is added. The ketone, pyridine or 1,4-dioxane is then reacted with the appropriate amine or amine hydrochloride 4 or 5. Subsequently, the reaction mixture is maintained at about ambient temperature to the reflux temperature of the solvent, preferably from about 35 ° C to about reflux. The temperature is formed until the product formation is substantially complete, usually for about 2 to about 24 hours. The reaction is preferably carried out under an inert atmosphere such as dry nitrogen. When compound 4 is used (wherein γ is Br,] [or trifluoro When decanesulfonyloxy) is reacted as a starting material with quinazoline 2, a compound of formula 3 (wherein Ri, R2, R3 and Y are as described above) is formed by compound 3 with a suitable palladium reagent (such as Ruthenium (triphenylphosphine) palladium or bis(triphenylphosphine) dipalladium palladium) in a suitable Lewis acid (such as vaporized cuprous) and a suitable alkyne (such as tri-φ methyl decyl) In the presence of acetylene, propargyl alcohol or 3-(N,N-dimethylamino)-propyne Reacting in a reagent such as diethylamine or triethylamine to convert compound 3 to a compound of formula 1 wherein R4 is Rn ethynyl and Rn is as defined above. It may be by using a diazotizing agent such as an acid and Nitrite (eg, acetic acid and NaN〇2)) treats compound 3 (where γ is NH2), followed by treatment of the resulting product with an azide (such as NaNs) to convert compound 3 to a compound where r4 is an azide) . To prepare a compound of formula I wherein R1 is an amine or hydroxylamine group, the corresponding compound of formula I is reduced. 143939.doc -85 - 201019961 The restoration should be made using any of a number of procedures known for such conversion. The reduction can be carried out, for example, by reacting an inert solvent bath with a hydrogen (tetra) based compound in the presence of a suitable metal catalyst such as p, 10 or nickel. Another suitable reducing agent is, for example, an activated metal such as activated iron (produced by washing iron powder with a dilute acid solution such as hydrochloric acid). Thus, for example, a mixture of a nitro compound and an activated metal with concentrated hydrochloric acid in a solvent such as a mixture of water and an alcohol: the alcohol such as methanol or ethanol is heated to, for example, 5 (rc to 15 (temperature in the range of TC) (It should be heated to ... or reduced by thief. Another suitable class of reducing agent is alkali metal dithionite (such as sodium dithionite), which can be in (Cl_C4) alkanoic acid, (Ci_C6) alkanol, water or For use in the preparation of a mixture of R2 or R3 having a - or a secondary amine moiety (other than an amine group intended to react with an oxazoline), preferably protecting the free amine group prior to the above reaction, The protecting group is then removed after the above reaction with 4_(substituted)quinazoline 2. Several well-known nitrogen protecting groups may be used. These groups include (CiC6) alkoxycarbonyl, optionally substituted benzoyl Oxycarbonyl, aryloxycarbonyl, triphenylsulfonyl, ethyleneoxycarbonyl, fluorenyl-nitrophenylsulfonyl, diphenylphosphinyl, p-toluenesulfonyl and benzyl. Nitrogen protecting group The addition can be at a temperature of from about 01 to about 50 t: Between ambient temperature, in the presence or absence of a tertiary amine such as triethylamine, diisopropylethylamine or pyridine, preferably triethylamine, in a gasified hydrocarbon solvent (such as di-methane or hydrazine) , 2_di-ethane) or ether solvent (such as ethylenediamine dimethyl ether, diethylene glycol dioxime ether or THF). Or, and use Schoonton_Baumann 143939.doc -86-201019961 Conditional attachment of protecting groups. After the above coupling reactions of compounds 2 and 5, the protecting groups can be removed by methods known to those skilled in the art, such as for products protected by a third butoxy group. Treatment with trifluoroacetic acid in di-methane.

關於保護基及其用途之描述,參見T. w. Greene&p G M. Wuts,「Protective Groups in Organic Synthesis」,第 2 版 ’ John Wiley & Sons, New York,1991。 為製備R或R2為羥基之式〗化合物,較佳裂解Rl*R2為 (C1-C4)烷氧基之式丨化合物。 該裂解反應宜利用許多已知用於此轉化之程序之任一者 進行。〇-脫烷基化可採用在15(rc至175。0用熔融鹽酸吡啶 (20-30當量)處理經保護之式〗衍生物。或者,裂解反應可 以例如用鹼金屬(Cl_C4)烷基硫化物(諸如乙硫醇鈉)處理經 保護之喹唑啉衍生物或用鹼金屬二芳基磷化物(諸如二苯 基磷化鋰)處理來進行。裂解反應亦 宜用三鹵化哪或三鹵 φ 化鋁(諸如三溴化硼)處理經保護之喹唑啉衍生物來進行。 該等反應較佳在反應惰性溶劑存在下在適合溫度進行。 R1或R2為(Cl-C:4)烷基亞磺醯基或(a·。)烷基磺醯基之式 I化合物較佳藉由氧化…或尺2為(Ci_C4)烷基硫基之式〗化合 物製備。硫基氧化成亞磺酿基及/或磺醯基之適合氧化劑 在此項技術中已知,例如過氧化氫、過酸(諸如3_氣過氧苯 甲酸或過氧乙酸)、鹼金屬過氧硫酸鹽(諸如過氧單硫酸 鉀)、二氧化鉻或氣態氧在鉑存在下。一般在儘可能溫和 之條件下使用化學計算量之氧化劑進行氣化以減小過度氧 143939.doc -87- 201019961 化之風險及對其他官能基之破壞。反應一般在適合溶劑 (諸如二氣甲烷、氣仿、丙酮、四氫呋喃或第三丁基曱基 鍵)中且在約-25C至50C之溫度(較佳在或近周圍溫度,亦 即在15°C至35°C範圍内)進行。當期望具有亞磺醯基之化 合物時’應使用較溫和氧化劑(諸如偏過碘酸鈉或偏過蛾 酸鉀)’宜在極性溶劑(諸如乙酸或乙醇)中。含有(Ci_c4)烧 基磺醯基之式I化合物可藉由氧化相應(Ci_c4)烷基亞磺醯 基化合物以及相應(CrCU)烧基硫基化合物而獲得。 R為視情況經取代之(C2-C4)烧醯基胺基、腺基、3_苯基 脲基、苯▼醯胺基或磺醯胺基之式j化合物可藉由醯化或 磺醯化R1為胺基之相應化合物來製備。合適之醯化劑為此 項技術中已知用於醯化胺基形成醢胺基之任何試劑,在合 適鹼存在下例如醯基齒,例如(C2_C4)烷醯氣或(C2_C4)烷醯 溴、或苯甲醯氯或苯甲酿溴;烷酸酐或混合酸酐,例如乙 酸酐或由烷酸與(C〗-C4)烷氧基羰基鹵(例如(C1_C4)烷氧基 羰基氣)反應形成之混合酸酐。在製備…為脲基或3·苯基脲 基之式I化合物時,合適的醯化劑為例如氰酸鹽(例如鹼金 屬氰酸鹽’諸如氰酸鈉)或異氰酸酯(諸如異氰酸苯酯p N_ 磺醯化可用合適的磺醯基齒或磺醯基酸酐在三級胺鹼存在 下進行。醯化或磺醯化一般在反應惰性溶劑中且在約_30<>c 至120C範圍内之溫度下(宜在周圍溫度或約周圍溫度下)進 行。 R為(C1-C4)烷氧基或經取代之((^_C4)烷氧基,或R丨為 (CrC4)烷基胺基或經取代之單_N_(C】_CJ烷基胺基或二_ 143939.doc • 88 - 201019961 NA-CCi-C4)烷基胺基之式I化合物係藉由較佳在合適鹼存 在下分別烷基化R1為羥基或胺基之相應化合物來製備。在 合適鹼存在下’在反應惰性溶劑中且在約1〇t>c至l4〇〇c範 圍内之溫度下(宜在周圍溫度或約周圍溫度下),合適之烷 基化劑包括烷基_或經取代之烷基画,例如視情況經取代 之(CVC4)烷基氣、((:丨-(:4)烷基溴或(Cl_c4)烷基碘。 為製備R1為經胺基取代、經氧基取代或經氰基取代之 (Ci-CO烧基取代基的式I化合物’較佳在合適驗存在下使 R1為具有可經胺基、烷氧基或氰基置換之基團的(Ci_c4)烷 基取代基之相應化合物與適當胺'醇或氰化物反應。反應 較佳在反應惰性溶劑或稀釋劑中且在約1〇〇C至l〇(rc範圍 内之溫度下(較佳在周圍溫度或約周圍溫度下)進行。 R為羧基取代基或包括羧基之取代基的式j化合物係藉 由水解R1為(Ci-C4)烷氧基羰基取代基或包括(Cl_c4)烷氧基 幾基之取代基之相應化合物來製備。水解宜例如在鹼性條 件下(例如在驗金屬氫氧化物存在下)進行。 R1為胺基、(CVC4)烷基胺基、二[(CVCJ烷基]胺基、"比 咯啶-1-基、N-哌啶基、N-嗎啉基、哌嗪-1-基、‘(CVCO 烧基略嗪-1-基或(Ci-Cd烷硫基之式I化合物可藉由在合適 驗存在下使R1為胺或硫醇可置換基團之相應化合物與適當 的胺或硫醇反應來製備。反應較佳在反應惰性溶劑或稀釋 劑中且在約10°C至180T:範圍内(宜在100°c至150。(:範圍内) 之溫度下進行。 R1為2·側氧基吼咯啶_丨_基或2_側氧基哌啶_丨_基之式 143939.doc -89 - 201019961 合物係藉由在合適驗存在下環化Rl為函基_(cvc4)院酿基 胺基之相應化合物來製備。反應較佳在反應惰性溶劑或稀 釋劑中且在約KTC至⑽。C範圍内之溫度下(宜在周圍溫度 或約周圍溫度下)進行。 在製備R1為胺甲酿基、經取代之胺甲醯基、炫醯氧基或 銓取代之烷醯氧基之式合物時,宜胺甲醯化或醯化汉丨 為羥基之相應化合物。 +此項技術中已知之用於醯化羥基芳基部分形成烷醯氧基 芳基之合適醯化劑通常在合適鹼存在下包括例如(c2_c4)烷 醯基鹵、(C^-C:4)烷醯基酸酐及如上所述之混合酸酐,且可 使用其合適的經取代衍生物。或者,可藉助於縮合劑(諸 如碳化一亞胺)使(C2-C4)燒酸或其適當經取代之衍生物與 R為羥基之式I化合物偶合。在製備Ri為胺甲醯基或經取 代之胺甲醯基之式I化合物時,通常在合適的鹼存在下, 合適之胺曱醯化劑為例如氰酸鹽或烷基異氰酸酯或芳基異 氰酸酯。或者,諸如R1為羥基之式[化合物的氯甲酸酯或 羰基咪唑衍生物之合適中間物可例如藉由用光氣(或光氣 4效物)或幾基二味嗤處理該衍生物而產生。隨後,可使 所得中間物與適當胺或經取代之胺反應產生期望的胺曱醯 基衍生物。 R1為胺基羰基或經取代之胺基羰基之化合物可藉由 胺解R1為羧基之合適中間物來製備。 R1為羧基之式I化合物之活化及偶合可利用熟習此項技 術者已知之多種方法進行。合適方法包括活化羧基為具有 143939.doc -90· 201019961 適當的與期望胺偶合之反應性之酸鹵化物、疊氮化物、對 稱或混合酸酐或活性酯。該等類型之中間物的實例及其製 備及用於與胺偶合之用途可廣泛見於文獻中;例如M. Bodansky 及 A. Bodansky, 「The Practice of Peptide Synthesis」,Springer-Verlag, New York, 1984 ° 戶斤得式 I化 合物可利用標準方法分離及純化,該等方法諸如溶劑移除 及再結晶或層析。 所述反應流程I之起始物質(例如胺、喹唑啉及胺保護基) 可容易地獲得,或可由熟習此項技術者使用習知之有機合 成方法容易地合成。舉例而言,2,3-二氫-1,4-苯并噁嗪衍 生物之製備係描述於R. C. ElderHeld, W· H. Todd, S_ Gerber,第 12章,「Heterocyclic Compounds」,第 6卷,R. C. Elderfield編,John Wiley and Sons,Inc., Ν·Υ·, 1957 中0 R. C. Elderfield 及 E. E. Harris 在 Elderfield, 「Heterocyclic Compounds」一書之第6卷第13章中描述經取代之2,3-二氫 苯并噻嗪基化合物。 在另一特定實施例中,EGFR拮抗劑具有如US 5,457,105 (以引用之方式併入本文中)中所述之通式II,For a description of the protecting groups and their uses, see T. w. Greene & p G M. Wuts, "Protective Groups in Organic Synthesis", 2nd edition 'John Wiley & Sons, New York, 1991. For the preparation of a compound of the formula wherein R or R2 is hydroxy, it is preferred to cleave a compound of the formula wherein R1*R2 is a (C1-C4) alkoxy group. The cleavage reaction is preferably carried out using any of a number of procedures known for use in this transformation. The oxime-dealkylation can be carried out by treating the protected formula with 15% rc to 17.5 with molten pyridine hydrochloride (20-30 equivalents). Alternatively, the cleavage reaction can be vulcanized, for example, with an alkali metal (Cl_C4) alkyl group. Treatment of the protected quinazoline derivative or treatment with an alkali metal diaryl phosphide such as lithium diphenylphosphinate is carried out by treatment with a substance such as sodium ethoxide. The cleavage reaction is also preferably trihalogenated or trihalide. Treatment of the protected quinazoline derivative with φ aluminum (such as boron tribromide) is preferably carried out at a suitable temperature in the presence of a reaction inert solvent. R1 or R2 is (Cl-C: 4) alkane The compound of the formula I of the sulfinyl group or the (a.)alkylsulfonyl group is preferably prepared by oxidizing a compound of the formula (I) or a caliper of the formula (Ci_C4)alkylthio. Suitable oxidizing agents for the group and/or sulfonyl group are known in the art, such as hydrogen peroxide, peracids (such as 3-hydroxy peroxybenzoic acid or peracetic acid), alkali metal peroxysulfates (such as peroxygen). Potassium monosulfate), chromium dioxide or gaseous oxygen in the presence of platinum, generally under mildest conditions Gasification with a stoichiometric amount of oxidant to reduce the risk of excess oxygen and damage to other functional groups. The reaction is generally in a suitable solvent (such as di-methane, gas, acetone, tetrahydrofuran or In the third butyl fluorenyl bond) and at a temperature of about -25 C to 50 C (preferably at or near ambient temperature, i.e., in the range of 15 ° C to 35 ° C). When it is desired to have a sulfinyl group In the case of a compound, a milder oxidizing agent (such as sodium metaperiodate or potassium molybdate) should be used. It is preferably in a polar solvent such as acetic acid or ethanol. The compound of formula I containing (Ci_c4)alkylsulfonyl can be borrowed. Obtained by oxidizing the corresponding (Ci_c4) alkylsulfinyl compound and the corresponding (CrCU) alkylthio compound. R is optionally substituted (C2-C4) decylamino group, gland base, 3-benzene The compound of formula j wherein the ureido group, the phenyl oxime group or the sulfonamide group can be prepared by deuteration or sulfonation of the corresponding compound wherein R1 is an amine group. Suitable oximation agents are known in the art. Any reagent which forms a guanamine group on a guanidinium group, in the presence of a suitable base, for example Base teeth, such as (C2_C4) alkane or (C2_C4) alkane bromine, or benzamidine chloride or benzyl bromine; alkanoic anhydride or mixed anhydride, such as acetic anhydride or from alkanoic acid and (C-C4) alkane A mixed acid anhydride formed by the reaction of an oxycarbonyl halide (for example, a (C1_C4) alkoxycarbonyl gas). When preparing a compound of the formula I which is a ureido group or a 3-phenylureido group, a suitable oximation agent is, for example, a cyanate salt. (for example an alkali metal cyanate such as sodium cyanate) or an isocyanate (such as phenyl isocyanate p N_sulfonation can be carried out in the presence of a tertiary amine base using a suitable sulfonyl or sulfonyl anhydride. Alternatively, the sulfonation is carried out in a reaction inert solvent and at a temperature in the range of from about -30 <>> to 120 C (preferably at ambient temperature or about ambient temperature). R is a (C1-C4) alkoxy group or a substituted ((^_C4) alkoxy group, or R is a (CrC4) alkylamino group or a substituted mono-N_(C)_CJ alkylamino group or _ 143939.doc • 88 - 201019961 NA-CCi-C4) Alkylamino compounds of formula I are prepared by alkylating R1 to a corresponding compound of a hydroxy or amine group, preferably in the presence of a suitable base. In the presence of a suitable base 'in a reaction inert solvent and at a temperature in the range of from about 1 Torr to about 4 〇〇c (preferably at ambient temperature or about ambient temperature), suitable alkylating agents include alkyl groups _ Or substituted alkyl, such as optionally substituted (CVC4) alkyl gas, ((: 丨-(:4) alkyl bromide or (Cl_c4) alkyl iodide. For the preparation of R1 is substituted by an amine group, The compound of formula I which is substituted by an oxy group or substituted with a cyano group (Ci-COalkyl substituent) preferably has R1 as a group which may be substituted by an amine group, an alkoxy group or a cyano group in the presence of a suitable test. The corresponding compound of the (Ci_c4)alkyl substituent is reacted with an appropriate amine 'alcohol or cyanide. The reaction is preferably carried out in a reaction inert solvent or diluent at a temperature in the range of from about 1 ° C to about 1 Torr ( Preferably, the compound of formula j wherein R is a carboxy substituent or a substituent comprising a carboxy group is hydrolyzed by R1 to a (Ci-C4) alkoxycarbonyl substituent or includes (Cl_c4). The corresponding compound of the substituent of the alkoxy group is prepared. The hydrolysis is preferably carried out, for example, under basic conditions (for example, in the presence of a metal hydroxide). R1 is an amine group, (CVC4) alkylamino group, two [ (CVCJ alkyl)amino, "pyrrolidin-1-yl, N-piperidinyl, N-morpholinyl, piperazin-1-yl, '(CVCO pyrrolizin-1-yl or The Ci-Cd alkylthio group of the formula I can be prepared by reacting a corresponding compound wherein R1 is an amine or a thiol replaceable group with a suitable amine or thiol in the presence of a suitable test. The reaction is preferably carried out in a reaction inert solvent. Or in a diluent and in the range of about 10 ° C to 180 T: (preferably in the range of 100 ° C to 150 ° (in range). R1 is 2 · side oxy-pyrrolidine _ 丨 _ group or 2 _ Side Oxypiperidine 丨 基 基 143 143939.doc -89 - 201019961 The compound is prepared by cyclizing Rl to the corresponding compound of the functional group _(cvc4) in the presence of a suitable test. Preferably, the reaction is carried out in a reaction inert solvent or diluent at a temperature in the range of from about KTC to (10) C (preferably at ambient temperature or about ambient temperature). In the preparation of R1 is an amine methyl group, substituted In the case of a formula of an amine methyl sulfhydryl group, a fluorenyloxy group or a hydrazine substituted alkoxy group, it is preferred to catalyze or oxime the corresponding compound of the hydroxy group. Suitable oximation agents for the hydroxyaryl moiety to form an alkoxy aryl group typically include, for example, (c2_c4)alkyl fluorenyl halide, (C^-C:4) alkanoic acid anhydride, and mixtures as described above, in the presence of a suitable base An acid anhydride, and a suitable substituted derivative thereof can be used. Alternatively, the (C2-C4) succinic acid or a suitably substituted derivative thereof can be coupled with a compound of formula I wherein R is hydroxy by means of a condensing agent such as a carbamide. In the preparation of a compound of formula I wherein Ri is an aminomethyl sulfhydryl group or a substituted amine mercapto group, a suitable amine oximation agent is, for example, a cyanate or an alkyl isocyanate or an aryl isocyanate, usually in the presence of a suitable base. . Alternatively, a suitable intermediate such as a chloroformate or a carbonylimidazole derivative of the compound of the formula [wherein R1 is hydroxy" may be treated, for example, by treatment with phosgene (or phosgene 4) or a few bases of miso. produce. The resulting intermediate can then be reacted with the appropriate amine or substituted amine to yield the desired amine mercapto derivative. A compound wherein R1 is an aminocarbonyl group or a substituted aminocarbonyl group can be prepared by subjecting R1 to a suitable intermediate of a carboxyl group. Activation and coupling of a compound of formula I wherein R1 is a carboxy group can be carried out by a variety of methods known to those skilled in the art. Suitable methods include activating the carboxyl group as an acid halide, azide, symmetrical or mixed anhydride or active ester having the appropriate reactivity with the desired amine coupling of 143939.doc-90.201019961. Examples of such intermediates and their preparation and their use for coupling with amines are widely found in the literature; for example, M. Bodansky and A. Bodansky, "The Practice of Peptide Synthesis", Springer-Verlag, New York, 1984 The compound of formula I can be isolated and purified by standard methods such as solvent removal and recrystallization or chromatography. The starting materials of the reaction scheme I (e.g., amines, quinazolines, and amine protecting groups) are readily available or can be readily synthesized by those skilled in the art using conventional organic methods of synthesis. For example, the preparation of 2,3-dihydro-1,4-benzoxazine derivatives is described in RC ElderHeld, W. H. Todd, S_ Gerber, Chapter 12, "Heterocyclic Compounds", Volume 6 , ed. Elderfield, John Wiley and Sons, Inc., Ν·Υ·, 1957, 0 RC Elderfield and EE Harris, in Elderfield, “Heterocyclic Compounds”, Volume 6, Chapter 13, Chapter 2, 3 a -dihydrobenzothiazinyl compound. In another specific embodiment, the EGFR antagonist has the formula II as described in US 5,457,105 (incorporated herein by reference)

II 143939.doc -91 - 201019961 其中: m為1、2或3 ;且 各R1獨立地為6-羥基、7-羥基、胺基、羧基、胺甲醯 基、脲基、(1-4C)烷氧基羰基、N-(1-4C)烷基胺曱醯基、 Ν,Ν·二[(1-4C)烷基]胺曱醯基、羥胺基、(卜4C)烷氧基胺 基、〇4C)烷醯氧基胺基、三氟甲氧基、〇4C)烷基、6-(1-4C)烷氧基、7-(l-4C)烷氧基、(1-3C)伸烷基二氧基、 (1-4C)烷基胺基、二[(1-4C)烷基]胺基、°比咯啶-1-基、N-哌啶基、N-嗎啉基、哌嗪-1-基、4-(l-4C)烷基哌嗪-1-基、 (1-4C)烷硫基、(1-4C)烷基亞磧醢基、(1_4C)烷基磺醯基、 溴甲基、二溴曱基、羥基-(1-4C)烷基、(2-4C)烧醯氧基- (1-4C)烷基、(1-4C)烷氧基-(1-4C)烷基、羧基-U-4C)烧 基、(1-4C)烷氧基羰基-(1-4C)烷基、胺曱醯基-(卜40)烧 基、N-(1-4C)烷基胺曱醯基-(1-4C)烷基、N,N-二[(1-4C)烧 基]胺甲醯基-(1-4C)烷基、胺基-U-4C)烷基、(1-4C)烧基 胺基_(1_4C)烷基、二[(1-4C)烧基]胺基·(卜4C)烷基、N_〇底 啶基_(1·4〇烷基、N-嗎啉基-(1-4C)烷基、哌嗪-1-基-(卜 4C)炫基、4-(1-40)院基°底嗓-1-基-U-4C)院基、經基_(2-4C)烷氧基-(1-4C)烷基、(1-4C)烷氧基-(2-4C)烧氧基-(卜 4C)烷基、羥基-(2-4C)烷基胺基-(卜4C)炫•基、(1-4C)燒氣 基-(2-4C)烷基胺基-(1-4C)烷基、(1-4C)院硫基-(1-4C)炫 基、羥基-(2-4C)烷硫基_(1_4C)烷基、(1-4C)烷氧基-(2-4C) 烷硫基-(1-4C)烷基、苯氧基-(1-4C)烷基、苯胺基-(i-4C) 烷基、苯硫基-0-4C)烷基、氰基-(1-4C)烷基、_基_(2_ 143939.doc • 92· 201019961 4C)烷氧基、羥基-(2-4C)烷氧基、(2-4C)烷醯氧基-(2-4C) 烷氧基、(1-4C)烷氧基-(2-4C)烷氧基、羧基-(1-4C)烷氧 基、(1-4C)烷氧基羰基-(1-4C)烷氧基、胺曱醯基-(1-4C)烷 氧基、N-(1-4C)烷基胺甲醯基-(1-4C)烷氧基、Ν,Ν-二[(Ια)烷基] 胺 曱醯基-(1-4C)烷氧基 、胺基-(2-4C)烷 氧基、 (1-4C)烷基胺基-(2-4C)烷氧基、二[(1-4C)烷基]胺基-(2-4C)烷氧基、(2-4C)烷醯氧基、羥基-(2-4C)烷醯氧基、(1-4C)烷氧基-(2-4C)烷醯氧基、苯基-(1-4C)烷氧基、苯氧基-(2-4C)烷氧基、苯胺基-(2-4C)烷氧基、苯硫基-(2-4C)烷氧 基、Ν-哌啶基-(2-4C)烷氧基、Ν-嗎啉基-(2-4C)烷氧基、 哌嗪-1-基-(2-4C)烷氧基、4-(l-4C)烷基哌嗪-1-基-(2-4C) 烷氧基、鹵基-(2-4C)烷基胺基、羥基-(2-4C)烷基胺基、 (2-4C)烷醯氧基-(2-4C)烷基胺基、(1-4C)烷氧基-(2-4C)烷 基胺基、羧基-(1-4C)烷基胺基、(1-4C)烷氧基羰基-(1-4C) 烷基胺基、胺曱醯基-(1-4C)烷基胺基、N-(1-4C)烷基胺甲 醯基-(1-4C)烷基胺基、N,N-二[(1-4C)烷基]胺曱醯基-(1-4C)烷基胺基、胺基-(2-4C)烷基胺基、(1-4C)烷基胺基-(2-4C)烷基胺基、二[(1-4C)烷基]胺基-(2-4C)烷基胺基、苯 基-(1-4C)烷基胺基、苯氧基-(2-4C)烷基胺基、苯胺基-(2-4C)烷基胺基、苯硫基-(2-4C)烷基胺基、(2-4C)烷醯基胺 基、(1-4C)烷氧基羰基胺基、(1-4C)烷基磺醯基胺基、苯 甲酿胺基、苯確醯胺基(benzenesulphonamido)、3-苯基脲 基、2 -側氧基η比洛咬-1 -基、2,5 -二側氧基°比洛咬-1 -基、函 基-(2-4C)烷醯基胺基、羥基-(2-4C)烷醯基胺基、(1-4C)烷 143939.doc -93- 201019961 氧基-(2-4C)烷醯基胺基、羧基-(2-4C)烷醯基胺基、(1-4C) 烷氧基羰基-(2-4C)烷醯基胺基、胺甲醯基-(2-4C)烷醯基胺 基、N-(1-4C)烷基胺甲醯基-(2-4C)烷醯基胺基、N,N-二 [(1-4C)烷基]胺甲醯基-(2-4C)烷醯基胺基、胺基-(2-4C)烷 醯基胺基、(1-4C)烷基胺基-(2-4C)烷醯基胺基或二[(卜4C) 烷基]胺基-(2-4C)烷醯基胺基,且其中該苯甲醯胺基或苯 磺醯胺基取代基或在R1取代基中之任何苯胺基、苯氧基或 苯基可視情況具有一或兩個鹵基、(1-4C)烷基或(1-4C)烷 氧基取代基; η為1或2 ;且 各R2獨立地為氫、羥基、卤基、三氟甲基、胺基、硝 基、氰基、(1-4C)烷基、(1-4C)烷氧基、(1-4C)烷基胺基、 二[(1-4C)烷基]胺基、(1-4C)烷硫基、(1-4C)烷基亞磺醯基 或(1-4C)烷基磺醯基;或其醫藥學上可接受之鹽;例外為 排除4-(4’-羥基苯胺基)-6-甲氧基喹唑啉、4-(4,-羥基苯胺 基)-6,7-亞甲基二氧基喹唑啉、6-胺基-4-(4'-胺基苯胺基) 喹唑啉、4-苯胺基-6-甲基喹唑啉或其鹽酸鹽及4-苯胺基-6,7-二曱氧基喹唑啉或其鹽酸鹽。 在一特定實施例中,EGFR拮抗劑為選自由以下組成之 群之式II化合物:4-(3·-氯-4'-氟苯胺基)_6,7-二曱氧基喹唑 啉;4-(3’,4·-二氣苯胺基)-6,7-二甲氧基喹唑啉;6,7_二甲 氧基-4-(3'-硝基苯胺基)-喹唑啉;6,7-二乙氧基-4-(3,-甲基 苯胺基)-喹唑啉;6-曱氧基-4-(3’-甲基苯胺基)-喹唑啉;4-(3,-氯苯胺基)-6-曱氧基喹唑啉;6,7-伸乙基二氧基-4-(3,- 143939.doc 94 201019961 甲基苯胺基)-喧唾琳;&险i 7 听,6,胺基-7-甲氧基-4·(3,-甲基苯胺 基)-喹唑啉;4-(3’-甲基笨胺基)_6_脲基喹唑啉;6_(2_甲氧 基乙氧基曱基)_4·(3,·甲基苯胺基)_啥唾i ; 6,7-二-(2-甲氧 基乙氧基)-4-(3,-曱基笨胺基)_㈣嘛;6二甲基胺基_4· (31-甲基苯胺基)喹唑啉;6_苯甲醯胺基_4_(3,_甲基苯胺基) 喹唑啉,6,7-二甲氧基_4_(3,_三氟曱基苯胺基)_喹唑啉;6_ 羥基-7-甲氧基-4-(3,-曱基苯胺基)_喹唑啉;7_羥基_6_甲氧 基-4-(3’-甲基笨胺基喹唑啉;7_胺基_4_(3,_甲基苯胺基)_ 喹唑啉;6_胺基_4_(3,_甲基苯胺基)喹唑啉;卜胺基_4_(3,_ 氯苯胺基)-喹唑啉;6-乙醯胺基_4-(3'-甲基苯胺基)_喹唑 啉;6-(2-甲氧基乙基胺基)_4_(3,_曱基苯胺基)_喹唑啉;7_ (2-甲氧基乙醯胺基)_4·(3Ι_曱基苯胺基)_喹唑啉;7_(2-羥基 乙氧基)-6 -甲氧基-4-(3甲基苯胺基)·啥唾琳;7_(2_甲氧基 乙氧基)-6-甲氧基-4-(3·-甲基苯胺基)_啥。坐琳;6_胺基_4· (3’-甲基苯胺基)-喧π坐琳。 式II之噎唾啉衍生物或其醫藥學上可接受之鹽可利用已 知適用於製備化學相關化合物之任何方法來製備。_種合 適方法例如為US 4,322,420中所使用之方法。必需之起始 物質可自市面上購得,或利用標準有機化學程序獲得。 (a)宜在合適鹼存在下使ζ為可置換基團之喹唑啉⑴與 苯胺(ii)反應。 143939.doc -95· 201019961II 143939.doc -91 - 201019961 wherein: m is 1, 2 or 3; and each R1 is independently 6-hydroxy, 7-hydroxy, amine, carboxyl, aminemethanyl, ureido, (1-4C) Alkoxycarbonyl, N-(1-4C)alkylamine fluorenyl, hydrazine, hydrazine bis[(1-4C)alkyl]amine fluorenyl, hydroxylamine, (4C) alkoxyamino , 4C) alkanomethoxyamino group, trifluoromethoxy group, 〇4C) alkyl group, 6-(1-4C) alkoxy group, 7-(l-4C) alkoxy group, (1-3C) Alkyldioxy, (1-4C)alkylamino, bis[(1-4C)alkyl]amino, pyrrolidin-1-yl, N-piperidinyl, N-morpholinyl , piperazin-1-yl, 4-(l-4C)alkylpiperazin-1-yl, (1-4C)alkylthio, (1-4C)alkylphosphonium, (1-4C)alkyl Sulfonyl, bromomethyl, dibromoindolyl, hydroxy-(1-4C)alkyl, (2-4C) decyloxy-(1-4C)alkyl, (1-4C)alkoxy- (1-4C)alkyl, carboxy-U-4C)alkyl, (1-4C)alkoxycarbonyl-(1-4C)alkyl, aminyl-(b40)alkyl, N-( 1-4C)alkylamine fluorenyl-(1-4C)alkyl, N,N-bis[(1-4C)alkyl]aminocarboxamido-(1-4C)alkyl, amine-U -4C)alkyl, (1-4C)alkylamino-(1_4C)alkyl, bis[(1-4C)alkyl] Base (Bu 4C)alkyl, N_decridinyl-(1,4-tetraalkyl, N-morpholinyl-(1-4C)alkyl, piperazin-1-yl-(b 4C) Base, 4-(1-40) yard base, bottom 嗓-1-yl-U-4C), ketone-(2-4C)alkoxy-(1-4C)alkyl, (1-4C Alkoxy-(2-4C)alkoxy-(Bu 4C)alkyl, hydroxy-(2-4C)alkylamino-(Bu 4C)Hyun, (1-4C) aerated base - (2-4C)alkylamino-(1-4C)alkyl, (1-4C)-indolyl-(1-4C) leukoyl, hydroxy-(2-4C)alkylthio-(1_4C)alkane , (1-4C) alkoxy-(2-4C)alkylthio-(1-4C)alkyl, phenoxy-(1-4C)alkyl, anilino-(i-4C)alkyl , phenylthio-0-4C)alkyl, cyano-(1-4C)alkyl, _yl_(2_ 143939.doc • 92· 201019961 4C) alkoxy, hydroxy-(2-4C) alkoxy , (2-4C) alkanomethoxy-(2-4C) alkoxy, (1-4C) alkoxy-(2-4C)alkoxy, carboxy-(1-4C)alkoxy, (1-4C) alkoxycarbonyl-(1-4C)alkoxy, amidino-(1-4C)alkoxy, N-(1-4C)alkylaminemethanyl-(1- 4C) alkoxy, anthracene, fluorenyl-bis[(Ια)alkyl]aminoindolyl-(1-4C)alkoxy, amino-(2-4C)alkoxy, (1-4C)alkane Amino-(2-4C)alkoxy, bis[(1-4C)alkane Amino-(2-4C)alkoxy, (2-4C)alkoxy, hydroxy-(2-4C)alkyloxy, (1-4C)alkoxy-(2-4C) Alkyloxy, phenyl-(1-4C)alkoxy, phenoxy-(2-4C)alkoxy, anilino-(2-4C)alkoxy, phenylthio-(2-4C Alkoxy, hydrazine-piperidinyl-(2-4C)alkoxy, indole-morpholinyl-(2-4C)alkoxy, piperazin-1-yl-(2-4C)alkoxy 4-(l-4C)alkylpiperazin-1-yl-(2-4C)alkoxy, halo-(2-4C)alkylamino, hydroxy-(2-4C)alkylamino (2-4C) alkanomethoxy-(2-4C)alkylamino, (1-4C) alkoxy-(2-4C)alkylamino, carboxy-(1-4C)alkylamine , (1-4C) alkoxycarbonyl-(1-4C)alkylamino, amidino-(1-4C)alkylamino, N-(1-4C)alkylaminecarbamyl -(1-4C)alkylamino, N,N-bis[(1-4C)alkyl]aminoindolyl-(1-4C)alkylamino, amino-(2-4C)alkyl Amino, (1-4C)alkylamino-(2-4C)alkylamino, bis[(1-4C)alkyl]amino-(2-4C)alkylamino, phenyl-( 1-4C) alkylamino group, phenoxy-(2-4C)alkylamino group, anilino-(2-4C)alkylamino group, phenylthio-(2-4C)alkylamino group, (2-4C) alkanoylamino, (1-4C) alkoxy Carbonylamino, (1-4C)alkylsulfonylamino, benzoylamino, benzenesulphonamido, 3-phenylureido, 2-oxooxy η -yl, 2,5-di-tertiary oxyl-l-l-l-yl, aryl-(2-4C)alkylhydrazino, hydroxy-(2-4C)alkylamino, (1- 4C) alkane 143939.doc -93- 201019961 oxy-(2-4C)alkylmercaptoamine, carboxy-(2-4C)alkylalkylamino, (1-4C) alkoxycarbonyl-(2- 4C) alkyl mercaptoamine, amine mercapto-(2-4C)alkylhydrazino, N-(1-4C)alkylamine, mercapto-(2-4C)alkyldecylamine, N , N-bis[(1-4C)alkyl]aminecarbamyl-(2-4C)alkylhydrazino, amino-(2-4C)alkylhydrazino, (1-4C)alkyl Amino-(2-4C)alkylhydrazine-amine or bis[(bu-4C)alkyl]amino-(2-4C)alkylmercaptoamine, and wherein the benzhydrylamine or benzenesulfonamide The substituent or any of the anilino, phenoxy or phenyl groups in the R1 substituent may optionally have one or two halo, (1-4C)alkyl or (1-4C) alkoxy substituents; Is 1 or 2; and each R2 is independently hydrogen, hydroxy, halo, trifluoromethyl, amine, nitro, cyano, (1-4C) , (1-4C) alkoxy, (1-4C)alkylamino, bis[(1-4C)alkyl]amino, (1-4C)alkylthio, (1-4C)alkyl Sulfosyl or (1-4C)alkylsulfonyl; or a pharmaceutically acceptable salt thereof; the exception is the exclusion of 4-(4'-hydroxyanilino)-6-methoxyquinazoline, 4 -(4,-hydroxyanilino)-6,7-methylenedioxyquinazoline, 6-amino-4-(4'-aminoanilino)quinazoline, 4-anilino-6 -methylquinazoline or its hydrochloride and 4-anilino-6,7-dimethoxyoxyquinazoline or its hydrochloride. In a specific embodiment, the EGFR antagonist is a compound of formula II selected from the group consisting of 4-(3·-chloro-4'-fluoroanilino)-6,7-dimethoxy quinazoline; -(3',4·-dianiline)-6,7-dimethoxyquinazoline; 6,7-dimethoxy-4-(3'-nitroanilino)-quinazoline ;6,7-diethoxy-4-(3,-methylanilino)-quinazoline; 6-decyloxy-4-(3'-methylanilino)-quinazoline; 4- (3,-chloroanilino)-6-methoxy quinazoline; 6,7-extended ethyldioxy-4-(3,- 143939.doc 94 201019961 methylanilino)-喧 琳 琳; & Insurance i 7 Listen, 6, Amino-7-methoxy-4·(3,-Methylanilino)-quinazoline; 4-(3'-Methylaminoamino)-6-ureido Quinazoline; 6-(2-methoxyethoxymethyl)_4·(3,·methylanilino)_啥sai; 6,7-di-(2-methoxyethoxy)- 4-(3,-decylamino)-(iv); 6-dimethylamino _4·(31-methylanilino)quinazoline; 6-benzamide _4_(3,_A Benzylamino) quinazoline, 6,7-dimethoxy_4_(3,-trifluoromethylanilino)-quinazoline; 6-hydroxy-7-methoxy-4-(3,-曱Alkylamino) _ quinazoline; 7-hydroxy-6-methoxy-4-(3'-methyl amidoquinazoline; 7-amino-4-(3,-methylanilino)-quinazoline; 6_Amino_4_(3,-methylanilino)quinazoline; amidino_4_(3,-chloroanilino)-quinazoline; 6-acetamido_4-(3'- Methylanilino)-quinazoline; 6-(2-methoxyethylamino)_4_(3,-mercaptoanilino)-quinazoline; 7-(2-methoxyethenylamino) _4·(3Ι_mercaptoanilide)_quinazoline; 7-(2-hydroxyethoxy)-6-methoxy-4-(3methylanilino)·啥啥琳; 7_(2_甲Oxyethoxyethoxy)-6-methoxy-4-(3·-methylanilino)-啥.Shen Lin; 6_Amino_4·(3'-methylanilino)-喧π The quinone derivative of the formula II or a pharmaceutically acceptable salt thereof can be prepared by any method known to be suitable for the preparation of a chemically related compound. A suitable method is, for example, the method used in US 4,322,420. Starting materials are commercially available or can be obtained using standard organic chemistry procedures. (a) The quinazoline (1) which is a displaceable group is preferably reacted with aniline (ii) in the presence of a suitable base. c -95· 201019961

合適之可置換基團z為例如南基、烷氧基、芳氧基或磺 酿氧基’例如氣基、溴基、甲氣 ^ 戌丞T軋基、苯氧基、甲烷磺醯氧 基或f苯-對磺醯氧基。 合適之鹼為例如有機胺鹼, 例妝瞰諸如吡啶、2,6-二甲基吡 啶、三甲基吡啶、4-二甲基胺基咻a & ❹ 签妝丞比啶、二乙胺、嗎啉、Ν- 甲基嗎啉或二氮雜雙環[54〇]十一 」丁 烯,或例如鹼金屬或 驗土金屬碳酸鹽或氫氧化物, ^ 例如碳酸鈉、碳酸鉀、碳酸 鈣、氫氧化鈉或氫氧化鉀。 反應較佳在合適之惰性溶劑或稀釋劑存在下進行,該合 適之惰性溶劑或稀釋劑為例如燒醇或醋,諸如甲醇、乙 醇、異丙醇或乙酸乙醋;南化溶劑,諸如二氣甲烧、氣仿 或四氣化碳;醚,諸如四氫〇夫 風天淹或1,4-二噁烷;芳族溶 ❿ 劑’諸如甲苯;或偶極非質早、、交 F負于冷劑,诸如Ν,Ν_二甲基甲醯 胺、Ν,Ν· 一甲基乙酿胺、Ν-甲Λ· ηι4· Μ 甲基比嘻咬_2_網或二甲亞 石風。反應宜在例如1 〇 至1 $ 〇。广益® 士,& 主範圍内(較佳20〇c至肋艽範 圍内)之溫度下進行。 式II之啥唾琳衍生物可利用 〜用5亥方法呈游離鹼形式獲得, 或者其可呈與式H-Z(其中7 i亡,^ 、 、有上文所定義之含義)之酸形 成之鹽形式獲得。當期望自鹽蒋 曰盟獲得游離鹼時,可使用習知 程序用如上文所定義之合適鹼處理鹽。 143939.doc •96· 201019961 (b)為製備R1或R2為羥基之式Π化合物,裂解…或…為 U-^c)烷氧基之式π之喹唑啉衍生物。 裂解反應宜利用許多已知用於此類轉化之程序中之任一 、 者來進行。可例如藉由用鹼金屬(1_4C)烷基硫化物(諸如乙 硫醇鈉)處理喹唑啉衍生物,或例如用鹼金屬二芳基磷化 • 物(諸如二苯基磷化鋰)處理來進行反應。或者,裂解反應 宜例如藉由用三豳化硼或三豳化鋁(諸如三溴化硼)處理喹 唑啉衍生物來進行。該等反應較佳在如上文所定義之合適 惰性溶劑或稀釋劑存在下且在合適溫度下進行。 (c )為製備R或R為(1 · 4 C )烧基亞續酿基或(1 _ 4 c)烧基 磺醯基之式II化合物,氧化Ri*R2為(1_4C)烷硫基之式 喹唑啉衍生物。 合適之氧化劑為例如此項技術中已知之用於氧化硫基形 成亞磺醯基及/或磺醯基之任何試劑,例如過氧化氫、過 酸(諸如3·氣過氧苯甲酸或過氧乙酸)、鹼金屬過氧硫酸鹽 φ (諸如過氧單硫酸鉀)、三氧化鉻或在鉑存在下之氣態氧。 一般在儘可能溫和之條件下使用所需化學計量之量的氧化 劑進行氧化以減小過度氧化之風險及對其他官能基之破 壞。反應一般在合適溶劑或稀釋劑(諸如二氯甲烷、氣 仿、丙嗣、四負1吱嗔或第三丁基曱基醚)中且在例如_25它 至50C之溫度下(宜在周圍溫度或約周圍溫度下,亦即在 15C至35C範圍内)進行。當需要具有亞磺醯基之化合物 時,亦宜在極性溶劑(諸如乙酸或乙醇)中使用較溫和氧化 劑(例如偏過碘酸鈉或偏過碘酸鉀)。應瞭解,當需要含有 143939.doc -97· 201019961 (1-4C)烷基磺醯基之式π化合物時,可藉由氧化相應(1_4C) 烧基亞續醯基化合物以及相應(1-4C)烷硫基化合物而獲得 該式II化合物。 (d) 為製備R1為胺基之式II化合物,還原Ri為硝基之式】 之喹唑琳衍生物。 還原宜利用許多已知用於此類轉化之程序中之任一者來 進行。可例如藉由在如上文所定義之惰性溶劑或稀釋劑中 在合適金屬催化劑(諸如鈀或鉑)存在下氫化硝基化合物溶 液來進行還原。另一種合適還原劑為例如活化金屬,諸如 活化鐵(藉由用稀酸溶液(諸如鹽酸)洗滌鐵粉而產生)。因 此,可例如藉由將硝基化合物及活化金屬於合適溶劑或稀 釋劑(諸如水與醇之混合物,該醇例如曱醇或乙醇)中之混 合物加熱至例如50。(:至150°C範圍内之溫度(宜加熱至7(rc 或約70°C)進行還原。 (e) 為製備R1為(2-4C)烧醯基胺基或經取代之(2_4〇院 醯基胺基、脲基、3-苯基脲基或苯甲醯胺基,或R2為乙醯 胺基或苯甲醯胺基之式^化合物,醢化以或尺2為胺基之式 II之喹唑啉衍生物。 合適之醯化劑為例如此項技術中已知之用於醯化胺基形 成醯胺基之任何試劑,例如宜在如上文所定義之合適鹼存 在下之酿基鹵’例如(2_4C)烷醯基氣或(2_4C)烷醯基溴、 或苯曱醯氣或苯甲醯溴;在如上文所定義之合適鹼存在下 的烷酸酐或混合酸酐’例如(2_4C)烷酸酐(諸如乙酸酐)或 由烷酸與(1-4C)烷氧基羰基鹵(例如(1_4C)烷氧基羰基氣)反 143939.doc •98- 201019961 應形成之混合酸酐。在製備R1為脲基或3-苯基脲基之式II 化合物時,合適之醯化劑為例如氰酸鹽(例如驗金屬氰酸 鹽’諸如氰酸鈉)或例如異氰酸酯(諸如異氰酸苯酯)。醯化 一般在如上文所定義之合適惰性溶劑或稀釋劑中且在例 如-3〇°C至120°C範圍内之溫度下(宜在周圍溫度或約周圍溫 度下)進行。 (0為製備R1為(1-4C)烷氧基或經取代之(1-4C)烷氧 基’或R1為(1-4C)烷基胺基或經取代之(1-4C)烷基胺基之 式II化合物,較佳在如上文所定義之合適鹼存在下烷基化 Rl酌情為羥基或胺基的式II之喹唑啉衍生物。 在如上文所定義之合適鹼存在下,在如上文所定義之合 適惰性溶劑或稀釋劑中且在例如1(TC至140°C範圍内之溫 度下(宜在周圍溫度或約周圍溫度下),合適之烷基化劑為 例如此項技術中已知之用於烷基化羥基形成烷氧基或經取 代之院氧基或用於烷基化胺基形成烷基胺基或經取代之烷 基胺基的任何試劑,例如烷基i或經取代之烷基_,例如 (1-4C)院基氣、(1_4C)烷基溴或(14C)烷基碘或經取代之 (1-4C)院基氣、經取代之(丨_4(:)烷基溴或經取代之(14(:)烷 基蛾。 (g) 為製備R1為羧基取代基或包括羧基之取代基的式π 化合物’水解W為〇_4C)烷氧基羰基取代基或包括(1-4C) 烧氧基羰基之取代基的式Η之喹唑啉衍生物。 水解宜例如在驗性條件下進行。 (h) 為製備R1為經胺基取代、經氧基取代、經硫基取代 143939.doc •99· 201019961 或經氰基取代之(1-4C)烷基取代基的式II化合物,較佳在 如上文所定義之合適鹼存在下使R1為具有如上文所定義之 可置換基團之(1-4C)烷基取代基的式II之喹唑啉衍生物與 適當胺、醇、硫醇或氰化物反應。 反應較佳在如上文所定義之合適惰性溶劑或稀釋劑中且 在例如10°C至loot範圍内之溫度下(宜在周圍溫度或約周 圍溫度下)進行。 當需要式II之喹唑啉衍生物之醫藥學上可接受之鹽時, 可例如藉由使用習知程序使該化合物與例如合適酸反應而 獲得該鹽。 在一特定實施例中,EGFR括抗劑為如US 5,770,599(以 引用之方式併入本文中)中所揭示之式II,化合物,Suitable displaceable groups z are, for example, a southern group, an alkoxy group, an aryloxy group or a sulfonyloxy group such as a gas group, a bromo group, a methyl group, a phenoxy group, a phenoxy group, a methanesulfonyloxy group. Or f benzene-p-sulfonyloxy. Suitable bases are, for example, organic amine bases, such as pyridine, 2,6-lutidine, trimethylpyridine, 4-dimethylamino 咻a & 签 丞 丞 、, diethylamine , morpholine, Ν-methylmorpholine or diazabicyclo[54〇]undecene butene, or for example an alkali metal or soil test metal carbonate or hydroxide, ^ such as sodium carbonate, potassium carbonate, calcium carbonate , sodium hydroxide or potassium hydroxide. The reaction is preferably carried out in the presence of a suitable inert solvent or diluent such as an alcohol or vinegar such as methanol, ethanol, isopropanol or ethyl acetate; a solvent such as a gas. Aromatic, gas-like or tetra-vaporized carbon; ethers, such as tetrahydrofurfury or 1,4-dioxane; aromatic solvents such as toluene; or dipolar non-premature, F-negative In the cold agent, such as hydrazine, hydrazine dimethyl dimethyl carbamide, hydrazine, hydrazine, methyl ketone, Ν-methyl Λ η 4 4 4 4 甲基 甲基 甲基 甲基 _2 _2 _2 _2 _2 _2 _2 _2 _2 _2 . The reaction is preferably, for example, from 1 〇 to 1 $ 〇. It is carried out at the temperature of the main range (preferably 20〇c to the cost of the ribs). The salicin derivative of formula II can be obtained as a free base by using the method of 5 hai, or it can be formed as a salt with an acid of the formula HZ (wherein 7 i, ^, , as defined above) Form obtained. When it is desired to obtain the free base from the salt, the salt can be treated with a suitable base as defined above using conventional procedures. 143939.doc •96· 201019961 (b) is a quinazoline derivative of the formula π which is a ruthenium compound in which R1 or R2 is a hydroxyl group, which is cleaved or otherwise is a U-^c) alkoxy group. The cleavage reaction is preferably carried out using any of a number of procedures known for such transformation. The quinazoline derivative can be treated, for example, by treatment with an alkali metal (1_4C) alkyl sulfide such as sodium ethanethiolate or, for example, with an alkali metal diaryl phosphating agent such as lithium diphenylphosphide. To react. Alternatively, the cleavage reaction is preferably carried out, for example, by treating the quinazoline derivative with boron trioxide or aluminum trichloride (such as boron tribromide). Preferably, the reactions are carried out in the presence of a suitable inert solvent or diluent as defined above and at a suitable temperature. (c) is a compound of formula II wherein R or R is (1·4 C)alkyl or a (1 _ 4 c)alkylsulfonyl group, and the Ri*R2 is oxidized to (1_4C)alkylthio a quinazoline derivative. Suitable oxidizing agents are, for example, any of the agents known in the art for oxidizing sulfur groups to form sulfinyl groups and/or sulfonyl groups, such as hydrogen peroxide, peracids such as 3·gas peroxybenzoic acid or peroxygen Acetic acid), alkali metal peroxosulfate φ (such as potassium peroxymonosulfate), chromium trioxide or gaseous oxygen in the presence of platinum. Oxidation is generally carried out using the stoichiometric amount of oxidizing agent as mildly as possible to reduce the risk of excessive oxidation and damage to other functional groups. The reaction is generally carried out in a suitable solvent or diluent such as dichloromethane, gas, propionium, tetra-n- or tri-decyl ether and at a temperature of, for example, _25 to 50 C (preferably around) The temperature is about or about ambient temperature, that is, in the range of 15C to 35C. When a compound having a sulfinyl group is desired, it is also preferred to use a milder oxidizing agent (e.g., sodium metaperiodate or potassium metaperiodate) in a polar solvent such as acetic acid or ethanol. It should be understood that when a compound of the formula π containing 143939.doc -97· 201019961 (1-4C)alkylsulfonyl group is required, the corresponding (1_4C) alkyl group can be oxidized and the corresponding (1-4C) An alkylthio compound to obtain the compound of the formula II. (d) A quinazoline derivative of the formula wherein R is a nitro group for the preparation of a compound of formula II wherein R1 is an amine group. The reduction should be carried out using any of a number of procedures known for such transformations. The reduction can be carried out, for example, by hydrogenating a solution of the nitro compound in the presence of a suitable metal catalyst such as palladium or platinum in an inert solvent or diluent as defined above. Another suitable reducing agent is, for example, an activated metal such as activated iron (produced by washing iron powder with a dilute acid solution such as hydrochloric acid). Thus, for example, a mixture of the nitro compound and the activating metal in a suitable solvent or diluent such as a mixture of water and an alcohol, such as decyl alcohol or ethanol, can be heated to, for example, 50. (: to a temperature in the range of 150 ° C (preferably heated to 7 (rc or about 70 ° C) for reduction. (e) For the preparation of R1 is (2-4C) decylamino group or substituted (2_4 〇 a compound of the formula hydrazino, ureido, 3-phenylureido or benzylideneamine, or a compound wherein R2 is an acetamino group or a benzylamino group, which is an amine group or a ruler 2 A quinazoline derivative of the formula II. Suitable oximation agents are, for example, any of the agents known in the art for the oximation of an amine group to form a guanamine group, for example, in the presence of a suitable base as defined above. Alkyl halides such as (2_4C) alkane sulfhydryl or (2_4C) alkanoyl bromide, or phenylhydrazine or benzamidine bromide; alkanoic anhydride or mixed anhydride in the presence of a suitable base as defined above, for example ( 2_4C) an alkanoic anhydride (such as acetic anhydride) or a mixed acid anhydride formed by an alkanoic acid and a (1-4C) alkoxycarbonyl halide (for example, (1-4C) alkoxycarbonyl gas) 143939.doc • 98-201019961. When preparing a compound of formula II wherein R1 is a ureido group or a 3-phenylureido group, suitable oximation agents are, for example, cyanates (for example metal citrates such as sodium cyanate) or for example isocyanic acid An ester (such as phenyl isocyanate). The oximation is generally in a suitable inert solvent or diluent as defined above and at a temperature in the range of, for example, -3 ° C to 120 ° C (preferably at ambient temperature or about (at ambient temperature) (0 is the preparation of R1 is (1-4C) alkoxy or substituted (1-4C) alkoxy' or R1 is (1-4C) alkylamino group or substituted ( 1-4C) Alkylamino group of a compound of formula II, preferably alkylated R1, in the presence of a suitable base as defined above, as defined above as a hydroxy or a quinazoline derivative of formula II. In the presence of a suitable base, in a suitable inert solvent or diluent as defined above and at, for example, 1 (TC to 140 ° C temperature (preferably at ambient temperature or about ambient temperature), a suitable alkyl group The agent is, for example, any of those known in the art for alkylating a hydroxyl group to form an alkoxy group or a substituted alkoxy group or for alkylating an amine group to form an alkylamino group or a substituted alkylamine group. a reagent such as an alkyl i or a substituted alkyl group, for example, (1-4C), a base gas, a (1 to 4C) alkyl bromide or a (14C) alkyl iodide or a substituted (1- 4C) House-based gas, substituted (丨_4(:) alkyl bromide or substituted (14(:)) alkyl moth. (g) Formula for preparing R1 as a carboxyl substituent or a substituent including a carboxyl group The π compound 'hydrolyzes W is a 〇-4C) alkoxycarbonyl substituent or a quinazoline derivative of the formula 包括 including a substituent of the (1-4C) alkoxycarbonyl group. The hydrolysis is preferably carried out, for example, under the conditions of the test. (h) for the preparation of a compound of formula II wherein R1 is substituted by an amine group, substituted by an oxy group, substituted by a thio group, 143939.doc • 99· 201019961 or substituted by a cyano group (1-4C) alkyl group, preferably A quinazoline derivative of formula II wherein R1 is a (1-4C)alkyl substituent having a replaceable group as defined above in the presence of a suitable base as defined above, together with an appropriate amine, alcohol, thiol Or cyanide reaction. The reaction is preferably carried out in a suitable inert solvent or diluent as defined above and at a temperature in the range of, for example, 10 ° C to the loot (preferably at ambient temperature or about ambient temperature). When a pharmaceutically acceptable salt of a quinazoline derivative of the formula II is desired, the salt can be obtained, for example, by reacting the compound with, for example, a suitable acid using a conventional procedure. In a particular embodiment, the EGFR antagonist is a compound of formula II, as disclosed in US 5,770,599, which is incorporated herein by reference.

其中: η為1、2或3 ; 各R2獨立地為鹵基或三氟1甲基; R3為(1-4C)烷氧基;且 R1為二[(1-4C)烷基]胺基-(2-4C)烷氧基、吡咯啶-卜基-(2-4C)烧氧基、Ν-旅唆基-(2-4C)院氧基、嗎淋基-(2-4C) 炫*氧基、派唤-l -基-(2-4C)院氧基、4-(l-4C)炫》基°底11 桊-1_ 143939.doc -100- 201019961 基-(2-4C)烷氧基、咪唑-丨-基-口·^)烷氧基、二[(1_4C)烧 氧基-(2-4C)烷基]胺基_(2-4C)烷氧基、义噻嗎啉基_(2_4C) 烷氧基、1-側氧基-N-嗟嗎啉基-(2-4C)烷氧基或二側氧 基-N-嘆嗎琳基-(2-4C)烧氧基,且其中任何以上所提及之 包含不連接於N或〇原子之CH2(亞甲基)的Ri取代基均視情 況在該CH2基團上具有羥基取代基; 或其醫藥學上可接受之鹽。 在一特定實施例中,EGFR拮抗劑為選自由以下組成之 群之式ΙΓ化合物:4-(3,-氣-4,-氟苯胺基)-7-曱氧基_6_(2-吡 咯啶-1-基乙氧基)-喹唑啉;4-(3,-氣-4,-氟苯胺基)_7-曱氧 基-6-(2-N-嗎啉基乙氧基喹唑啉;4_(3,_氯_4,·氟苯胺基 6-(3-二乙基胺基丙氧基)_7_甲氧基喹唑啉;4_(3,氣·4,氟 苯胺基)-7-甲氧基-6-(3-吡嘻啶-1-基丙氧基)_喹唑琳;4_ (3’-氣-4'-氟苯胺基)_6-(3-二甲基胺基丙氧基)_7_甲氧基喹 吐啉;4-(3’,4'-二氟苯胺基)_7_甲氧基-6-(3-Ν-嗎啉基丙氧 基)-喧°坐啉;4-(3’-氣-4,-氟苯胺基)-7-曱氧基-6-(3-Ν-哌啶 基丙氧基)-喹唑啉;4-(3,-氣-4,-氟苯胺基)-7-曱氧基-6-(3-Ν-嗎啉基丙氧基)-喹唑啉;4_(31_氣_41_氟苯胺基)_6_(2_二 曱基胺基乙氧基)-7·甲氧基喹唑啉;4-(2,,4'-二氟苯胺基)-6-(3·二甲基胺基丙氧基)_7_曱氧基喹唑啉;4_(2,,41_二氟苯 胺基)-7-甲氧基-6-(3-Ν-嗎啉基丙氧基)-喹唑琳;4-(3,-氣-4·-氟苯胺基)-6-(2-咪唑-1-基乙氧基)_7_曱氧基喹唑啉;4-(3·-氣-41-氟苯胺基)-6-(3-咪唑-1-基丙氧基)-7-甲氧基喹唑 啉;4-(3'-氣-41-氟苯胺基)-6-(2·二曱基胺基乙氧基)-7-甲氧 143939.doc -101- 201019961 基喹唑啉,4-(2',4’-二氟苯胺基)-6-(3-二曱基胺基丙氧基)_ 7-甲氧基喹唑啉;4_(2,,4,_二氟苯胺基)_7_甲氧基_6_(3_n_ 嗎啉基丙氧基)-喹唑啉;4_(31_氣_4,_氟苯胺基)6(2_咪唑_ 1-基乙氧基)-7-甲氧基喹唑啉;及4_(3,_氣_4’_氟苯胺基)_6_ (3-咪唑-1·基丙氧基)_7_甲氧基喹唑啉。 在一特定實施例中,EGFR拮抗劑為式化合物,亦即 4_(3H氣苯胺基)·7_甲氧基_6_(3_Ν_嗎琳基丙氧基卜嗜 唑啉,或者稱為ZD 1839、吉非替尼及Iressa®。 式II之喹唑啉衍生物或其醫藥學上可接受之鹽可 知適用於製備化學相關化合物之任何方法來製備。合適方 法包括例如us 5616582、us 558〇87〇、us 5475〇〇1及仍 5569658中說明之方法。除非另外說t _n R2 ^ R1具有上文llUn,之㈣琳衍生物所定義之任何含義。 必需之起始物質可自市面上購得,或利用標準有機化學程 序獲得。 ⑷宜在合適驗存在下使2為可置換基團之㈣琳(Hi)與 苯胺(iv)反應。Wherein: η is 1, 2 or 3; each R2 is independently halo or trifluoromethyl; R3 is (1-4C) alkoxy; and R1 is bis[(1-4C)alkyl]amino -(2-4C) alkoxy, pyrrolidin-buki-(2-4C) alkoxy, oxime-branches-(2-4C) alkoxy, morphine-(2-4C) *oxy, 派-l-yl-(2-4C) alkoxy, 4-(l-4C) 炫" base 11 桊-1_ 143939.doc -100- 201019961 base-(2-4C) Alkoxy, imidazolium-hydrazino-yl-hydroxyl, bis[(1_4C)alkoxy-(2-4C)alkyl]amino-(2-4C)alkoxy, thiophene Phytyl-(2_4C) alkoxy, 1-o-oxy-N-indolyl-(2-4C)alkoxy or di-oxy-N-singly-based-(2-4C) An oxy group, and any of the above-mentioned Ri substituents comprising a CH2 (methylene group) not bonded to a N or a ruthenium atom, optionally having a hydroxy substituent on the CH2 group; or pharmaceutically acceptable Accept the salt. In a specific embodiment, the EGFR antagonist is a hydrazine compound selected from the group consisting of 4-(3,-gas-4,-fluoroanilino)-7-decyloxy-6-(2-pyrrolidine) -1-ylethoxy)-quinazoline; 4-(3,-gas-4,-fluoroanilino)-7-methoxy-6-(2-N-morpholinylethoxyquinazoline ; 4_(3,_Chloro-4, fluoroanilino 6-(3-diethylaminopropoxy)_7-methoxyquinazoline; 4_(3, gas·4, fluoroanilino)- 7-Methoxy-6-(3-pyridin-1-ylpropoxy)-quinazoline; 4_(3'-gas-4'-fluoroanilino)_6-(3-dimethylamine Propyloxy)-7-methoxyquinoxaline; 4-(3',4'-difluoroanilino)-7-methoxy-6-(3-indole-morpholinopropoxy)-indole °Situline; 4-(3'-gas-4,-fluoroanilino)-7-decyloxy-6-(3-indole-piperidinylpropoxy)-quinazoline; 4-(3, - gas-4,-fluoroanilino)-7-decyloxy-6-(3-indole-morpholinopropoxy)-quinazoline; 4_(31_gas_41_fluoroanilino)_6_( 2-didecylaminoethoxy)-7-methoxyquinazoline; 4-(2,,4'-difluoroanilino)-6-(3.dimethylaminopropylpropoxy) _7_曱oxyquinazoline; 4_(2,,41-difluoroanilinyl -7-methoxy-6-(3-indolyl-morpholinylpropoxy)-quinazoline; 4-(3,-aero-4-fluoroanilino)-6-(2-imidazole-1 -ylethoxy)-7-methoxy quinazoline; 4-(3·-gas-41-fluoroanilino)-6-(3-imidazol-1-ylpropoxy)-7-methoxy Quinazoline; 4-(3'-gas-41-fluoroanilino)-6-(2·didecylaminoethoxy)-7-methoxy 143939.doc -101- 201019961 quinazoline, 4-(2',4'-difluoroanilino)-6-(3-didecylaminopropoxy)-7-methoxyquinazoline; 4_(2,4,-difluoroaniline _7_methoxy_6_(3_n_morpholinylpropoxy)-quinazoline; 4_(31_gas_4,-fluoroanilino)6(2-imidazolium-1-ylethoxy)- 7-methoxyquinazoline; and 4_(3,_qi_4'-fluoroanilino)_6_(3-imidazolium-1-ylpropoxy)_7-methoxyquinazoline. In a specific implementation In the case, the EGFR antagonist is a compound of the formula, that is, 4_(3H-anilino)·7_methoxy_6_(3_Ν_morphine-propoxy-oxazoline, or ZD 1839, Gefitse And Iressa®. The quinazoline derivative of formula II or a pharmaceutically acceptable salt thereof is known to be suitable for use in any method for preparing chemically related compounds. Suitable methods include, for example, the methods described in us 5616582, us 558〇87〇, us 5475〇〇1 and still 5569658. Unless otherwise stated, t _n R2 ^ R1 has any meaning as defined by llUn above. The necessary starting materials are commercially available or obtained using standard organic chemical procedures. (4) It is preferred to react (4) with a cyclizable group (Hi) with an aniline (iv) in the presence of a suitable test.

合適之可置換基圏Z為例如南基、烧氧基、芳氧基或續 醯氧基’例如氣基、溴基、甲氧基、笨氧基、甲_氧 基或甲苯-4-磺醯氧基。 143939.doc •102· 201019961Suitable substituted bases Z are, for example, a southern group, an alkoxy group, an aryloxy group or a hydrazine group such as a gas group, a bromo group, a methoxy group, a phenoxy group, a methyl group or a toluene-4-sulfonate. Alkoxy. 143939.doc •102· 201019961

合適之驗為例如有機胺鹼,諸如…2各二甲基吼 咬、三甲基咕咬、I二甲基胺基。比嘴、三乙胺、嗎琳、N 甲基嗎啉或二氮雜雙環[5.4.0]十— ''' 7_浠,或例如驗金屬或 鹼土金屬碳酸鹽或氫氧化物,例如 J如碳酸鈉、碳酸鉀、碳酸 約、氫氧化鈉或氫氧化卸。或者人 *有,合適鹼為例如鹼金屬或 鹼土金屬胺化物,例如胺化鈉或售 义雙(二甲基矽烷基)胺化 納0Suitable tests are, for example, organic amine bases such as ... 2 each of dimethyl hydrazine, trimethyl hydrazine, and I dimethylamino. Than mouth, triethylamine, morphine, N-methylmorpholine or diazabicyclo [5.4.0] tens - ''' 7_浠, or for example metal or alkaline earth metal carbonate or hydroxide, such as J Such as sodium carbonate, potassium carbonate, carbonic acid, sodium hydroxide or hydroxide. Or human *, a suitable base is, for example, an alkali metal or alkaline earth metal amine, such as sodium amination or a bis(dimethylalkylalkyl) aminated nano

反應較佳在合適之惰性溶劑或稀釋劑存在下進行,該合 適之惰性溶劑或稀釋劑為例如垸醇或酯,諸如甲醇、乙 醇、異丙醇或乙酸乙醋;自化溶劑,諸如二氯甲院、氣仿 或四氯化碳;醚,諸如四氫呋喃或154_二噁烷;芳族溶 劑,諸如曱苯;或偶極非質子溶劑,諸如N,N_二甲基甲醯 胺、N,N-二甲基乙醯胺、N_曱基吡咯啶_2_酮或二甲亞 砜。反應宜在例如1〇。〇至150。〇;範圍内(較佳2(^至8〇1範 圍内)之溫度下進行。 式ΙΓ之喹唑啉衍生物可利用該方法呈游離鹼形式獲得, 或者其可呈與式H-Z(其中Z具有上文所定義之含義)之酸形 成之鹽形式獲得。當期望自鹽獲得游離鹼時,可使用習知 程序用如上文所定義之合適驗處理鹽。 (b)為製備R1為經胺基取代之(2-4C)烷氧基之式ΙΓ化合 物,宜在如上文所定義之合適鹼存在下烷基化尺1為羥基之 式ΙΓ之喹唑啉衍生物。 在如上文所疋義之合適驗存在下’在如上文所定義之合 適惰性溶劑或稀釋劑中且在例如lot:至140°C範圍内之溫 143939.doc -103· 201019961 度下(宜在80。(:下或約80°C下)’合適之炫•基化劑為例如此 項技術中已知之用於烷基化羥基形成經胺基取代之烷氧基 的任何試劑,例如經胺基取代之烷基鹵’例如經胺基取代 之(2-4C)烷基氣、經胺基取代之(2-4C)烷基溴或經胺基取 代之(2-4C)烷基碘。 (c) 為製備R1為經胺基取代之(2-4C)烷氧基之式II’化合 物’宜在如上文所定義之合適鹼存在下使R1為羥基-(2-4C) 烧氧基之式ΙΓ化合物或其反應性衍生物與適當胺反應。 R為經基-(2-4C)燒氧基之式ΙΓ化合物之合適反應性衍生 物為例如鹵基-(2-4C)烷氧基或磺酿氧基-(2-4C)烷氧基,諸 如溴-(2-4C)烷氧基或曱烷磺醯氧基_(2_4C)烷氧基。 反應較佳在如上文所定義之合適惰性溶劑或稀釋劑存在 下且在例如10°C至150。(:範圍内之溫度下(宜在5(rc下或約 50C下)進行。 (d) 為製備R1為羥基-胺基_(2_4C)烷氧基之式…化合物, 使R1為2,3-環氧基丙氧基或3,4_環氧基丁氡基之式π,化合 物與適當胺反應。 反應較佳在如上文所疋義之合適惰性溶劑或稀釋劑存在 下且在例如H)m5(rc範圍内之溫度下(宜在听下或約 70°C下)進行。 當需要式生物的醫藥學上可接受之鹽(例 如式Π,之啥嗤琳衍生物之單酸或二酸加成鹽)時,可例如 藉由使用習知程序使該化合物與例如合適酸反應而獲得該 143939.doc 201019961 在一特定實施例中,EGFR拮抗劑為如wo 993 5146(以引 用之方式併入本文中)中所揭示之式ΙΠ化合物或其鹽或溶 劑合物,The reaction is preferably carried out in the presence of a suitable inert solvent or diluent such as a sterol or an ester such as methanol, ethanol, isopropanol or ethyl acetate; a solvent such as dichloro A hospital, gas-like or carbon tetrachloride; an ether such as tetrahydrofuran or 154-dioxane; an aromatic solvent such as toluene; or a dipolar aprotic solvent such as N,N-dimethylformamide, N , N-dimethylacetamide, N_decylpyrrolidine-2-one or dimethyl sulfoxide. The reaction is preferably, for example, 1 Torr. 〇 to 150. 〇; in the range (preferably in the range of 2 (^ to 8〇1). The quinazoline derivative of the formula can be obtained as a free base by this method, or it can be represented by the formula HZ (wherein Z) The salt form of the acid formed having the meaning defined above is obtained. When it is desired to obtain the free base from the salt, a suitable procedure can be used to prepare the salt as defined above using a conventional procedure. (b) Preparation of R1 as an amine a quinazoline derivative of the formula 2-4, wherein the quinazoline derivative of the formula 1 is a hydroxy group, in the presence of a suitable base as defined above. Where appropriate, in the presence of a suitable inert solvent or diluent as defined above and at a temperature in the range of, for example, lot: to 140 ° C, 143939.doc -103.201019961 degrees (preferably at 80. (: below or about At 80 ° C, 'suitable singly-based agents are, for example, any of the reagents known in the art for alkylating a hydroxyl group to form an amino-substituted alkoxy group, such as an amine-substituted alkyl halide. For example, an amine group substituted (2-4C) alkyl gas, an amine substituted (2-4C) alkyl bromide or an amine Substituted (2-4C) alkyl iodide. (c) For the preparation of a compound of formula II' wherein R1 is an amino substituted (2-4C) alkoxy group, it is preferred to have a suitable base as defined above. R1 is a hydroxy-(2-4C) alkoxy compound of the formula or a reactive derivative thereof, which is reacted with a suitable amine. R is a suitable reactive derivative of a hydrazine compound of the group -(2-4C) alkoxy group. For example, halo-(2-4C)alkoxy or sulfo-oxy-(2-4C)alkoxy, such as bromo-(2-4C)alkoxy or decanesulfonyloxy_(2_4C) Alkoxy. The reaction is preferably carried out in the presence of a suitable inert solvent or diluent as defined above and at a temperature in the range of, for example, 10 ° C to 150 (suitably at 5 (rc or about 50 C) (d) For the preparation of a compound of the formula: wherein R1 is hydroxy-amino-(2_4C)alkoxy, R1 is 2,3-epoxypropoxy or 3,4-epoxybutenyl, π, The compound is reacted with a suitable amine. The reaction is preferably carried out in the presence of a suitable inert solvent or diluent as defined above and at a temperature in the range of, for example, H)m5 (preferably at or about 70 ° C). When the pharmaceutically acceptable salt of the desired organism is (e.g., a mono- or diacid addition salt of a hydrazine derivative), which can be obtained, for example, by reacting the compound with, for example, a suitable acid using conventional procedures to obtain the 143939.doc 201019961 in a particular implementation In an embodiment, the EGFR antagonist is a hydrazine compound, or a salt or solvate thereof, as disclosed in WO 993 5146, which is incorporated herein by reference.

/U HN/U HN

III X為N或CH ; Y為CR1且v為N ; 或Y為N且V為CR1 ; 或Y為CR1且V為CR2 ; 或Y為CR2且V為CR1 ;III X is N or CH; Y is CR1 and v is N; or Y is N and V is CR1; or Y is CR1 and V is CR2; or Y is CR2 and V is CR1;

Rl表示基團CH3S02CH2CH2NHCH2-Ar-,其中Ar係選自 苯基、吱喃、噻吩、吡咯及噻唑’其各可視情況經一或兩 個齒基、Cw烷基或Cw烷氧基取代; R2係選自包含以下之群:氫、鹵基、羥基、Cl.4烷基、 烷氧基、Cl·4烷基胺墓及二[Ci 4烷基]胺基; U表示苯基、吨啶基、3H_咪唑基、吲哚基、異吲哚 基、朵琳基、異吲哚啉基、1H_吲唑基、2,3_二氫_1H_, 唑基、1H-苯并咪唑基、2,3-二氳-1H-苯并咪唑基或1H-苯 并二唾基’其係經R3基團取代且視情況經至少一個獨立地 選擇之R4基團取代; 143939.doc •105- 201019961 R3係選自包含以下之群:苯甲基、鹵基苯甲基、二鹵基 苯曱基及三齒基笨甲基、苯曱醯基、0比啶基曱基、《比啶基 甲氧基、苯氧基、笨甲氧基、齒基苯甲氧基、二鹵基苯甲 氧基及三鹵基苯曱氧基及苯磺醯基;或尺3表示三鹵基曱基 苯甲基或三鹵基甲基苯甲氧基·, 或R3表示下式基團R1 represents a group CH3S02CH2CH2NHCH2-Ar-, wherein the Ar is selected from the group consisting of phenyl, decyl, thiophene, pyrrole and thiazole, each of which may be optionally substituted by one or two dentate groups, Cw alkyl or Cw alkoxy; R2 Selected from the group consisting of hydrogen, halo, hydroxy, Cl.4 alkyl, alkoxy, Cl.4 alkylamine tomb and bis[Ci 4 alkyl]amine; U represents phenyl, oxaridinyl , 3H-imidazolyl, fluorenyl, isodecyl, dolantyl, isoindolinyl, 1H-carbazolyl, 2,3-dihydro-1H_, oxazolyl, 1H-benzimidazolyl, 2,3-dioxin-1H-benzimidazolyl or 1H-benzodisalyl' is substituted by an R3 group and optionally substituted with at least one independently selected R4 group; 143939.doc •105- 201019961 R3 is selected from the group consisting of benzyl, halobenzyl, dihalobenzoyl and tridentylmethyl, benzoinyl, 0-pyridylthio, and pyridyl a methoxy group, a phenoxy group, a phenoxy group, a benzoyl methoxy group, a dihalo benzyloxy group, a trihalophenyl fluorenyloxy group, and a benzene sulfonyl group; or a calilem 3 represents a trihalo fluorenyl group Benzyl or trihalomethylbenzyloxy·, or R3 A group of formula

其中各R5獨立地選自鹵素、C1-4烷基及C1-4烷氧基;且 η為0至3 ;且 各R4獨立地為羥基、鹵素、Cl-4烷基、c2-4烯基、C2.4炔 基、c】·4烧氧基、胺基、Cl_4烷基胺基、二[Cl4烷基]胺 基、Cw烧硫基、Cl 4烷基亞磺醯基、Cl_4烷基磺酿基、Cl 4 烧基幾基、叛基、胺甲醯基、Cl-4烷氧基羰基、Cl_4烷醯基 胺基、N-%-4烷基)胺曱醯基、n,N-二(Cw烷基)胺曱醯 基、氰基、硝基及三氟甲基。 在一特定實施例中,式ni2EGFR拮抗劑排除:(1_苯曱 基1H-吲唾-5-基)-(6-(5-((2-甲烧續醯基·乙基胺基)_甲基)_ 呋喃-2-基)-吡啶并[3,4_d]嘧啶_4_基_胺;(4_苯曱氧基笨 基)-(6-(5-((2-曱烷磺醯基-乙基胺基)_甲基)_呋喃_2_基)-。比 咬并[3,4-d]喷咬_4-基-胺;(1-苯甲基-1H-吲。坐_5_基)-(6-(5· ((2-曱烷磺醯基_乙基胺基)_甲基)_呋喃_2基)喹唑啉心基- 143939.doc •106· 201019961 胺(1苯甲基Η-吲唑-5-基)-(7-(5-((2-甲烷磺醯基·乙基 胺基)-甲基)-呋喃_2_基)_喹唑啉_4_基_胺;及(1苯甲基_ιη_ 吲唑基)-(6-(5-((2-甲烷磺醯基-乙基胺基)_甲基)·卜曱基_ 吡咯基)-喹唑啉-4-基-胺。 在一特定實施例中,式III之EGFR拮抗劑係選自由以下 組成之群:4·(4_氟苯甲氧基)_苯基Η6-(5-((2·甲烧績醯基_ 胺基)曱基)_〇夫喃-2-基)-η比咬并[3,4-d]唆咬_4_基)_胺; ((氟*苯甲氧基)-本基)-(6-(5-((2-甲烧績醯基-乙基胺基) 甲基)呋喃-2-基)-D比啶并[3,4_d]嘧啶-4-基)-胺;(4-苯磺醯 基·苯基)-(6-(5-((2-曱烷磺醯基·乙基胺基)_甲基)_呋喃_2_ 基比啶并[3,4-d]嘧啶-4-基)·胺;(4_苯甲氧基-苯基)_(6_ (3-((2-甲烷磺酿基_乙基胺基)_甲基)苯基)比啶并[34d] 嘧啶-4-基)·胺;(4_苯曱氧基_苯基)_(6_(5·((2甲烷磺醯基· 乙基胺基)-曱基)-»夫喃_2·基)喹唑琳_4_基)_胺;(4-(3-氟苯 甲氧基-笨基)-(6-(4-((2-甲烷磺醯基-乙基胺基)_甲基)_呋 喃-2-基)-η比啶并[34_d]嘧啶_4基)胺;(4苯曱氧基苯基)_ (6-(2-((2-甲烷磺醯基乙基胺基)_甲基噻唑_4_基)喹唑啉_ 4-基)-胺;N-{4-[(3-氟笨甲基)氧基]苯基}_6_[5·({[2_(曱烷 石黃酿基)乙基]胺基}甲基)_2_呋喃基]_4_喹唑啉胺;Ν-{4-[(3-氟苯甲基)氧基]-3-甲氧基苯基}_6_[5·({[2_(甲烷磺醯 基)乙基]胺基}甲基)-2-呋喃基]_4_喹唑啉胺;Ν-[4-(苯曱氧 基)苯基]-7-甲氧基-6-[5-({[2-(甲烷磺醯基)乙基]胺基}甲 基)·2·呋喃基]-4-喹唑啉胺;Ν_[4_(苯甲氧基)苯基]_6_[4_ ({[2-(甲烷磺醯基)乙基]胺基}曱基)_2_呋喃基]_4_喹唑啉 143939.doc -107- 201019961 胺;Ν-{4-[(3·氟苯甲基)氧基]-3·甲氧基苯基}-6-[2-({[2-(曱烷磺醯基)乙基]胺基}曱基)-l,3-噻唑-4-基]-4-喹唑啉 胺;N-{4-[(3-溴笨曱基)氧基]苯基}_6-[2-({|;2•(曱烷磺醯 基)乙基]胺基}甲基)-1,3-噻峻-4-基]-4-喹唑淋胺;N-{4-[(3-氟苯曱基)氧基]苯基)_6-[2-({[2-(甲烷磺醯基)乙基]胺 基}罗基)1,3-噻唑-4-基]-4-喹唑啉胺;N-[4-(苯甲氧基)-3-氟苯基]-6-[2-({[2-(甲烷磺醯基)乙基]胺基)曱基)_1,3_噻唑_ 4-基]-4-喹唑啉胺;N-(l-苯甲基-1H-吲唑-5-基)-7-曱氧基-6-[5-({[2-(甲烷磺醯基)乙基]胺基)曱基)_2_呋喃基]_4_喹唑 啉胺·’ 6-[5-({[2-(曱烷磺醯基)乙基]胺基;)甲基)_2_呋喃基]_ N-(4-{[3-(三氟甲基)苯甲基]氧基)苯基)_4_喹唑啉胺;N_ {3-氟-4-[(3-氟苯甲基)氧基]苯基}_6_[5_({[2_(曱烷磺醯基) 乙基]胺基)甲基)-2-呋喃基]-4-喹唾琳胺;N-{4-[(3-溴苯甲 基)氧基]苯基)-6-[5-({[2-(曱烷磺醯基)乙基;]胺基)曱基)_2_ 咬喃基]-4-喹唑啉胺;N-[4-(苯甲氧基)苯基]-6_[3-({[2-(甲 院項醯基)乙基]胺基}曱基)_2_呋喃基]_4_喹唑琳胺;n-[1-(3-氟苯甲基)·ιη-吲唑-5-基]-6-[2-({[2-(曱烷磺酿基)乙基] 胺基}曱基)-1,3-噻唑-4-基]-4-喹唑啉胺;6-[5-({[2-(曱统 磺醯基)乙基]胺基)甲基)_2_呋喃基μΝ_[4 (苯磺醯基)苯基]_ 4-喹唑啉胺;6-[2-({[2-(甲烷磺醯基)乙基]胺基)曱基)_ι,3_ 噻唑-4-基]·Ν-[4-(笨磺醯基)苯基]_4_喹唑啉胺; (曱院磺醯基)乙基]胺基}甲基Μ,、噻唑·4_基]_Ν_(Μ[3· (二氟曱基)苯甲基]氧基)苯基)_4_喹唑啉胺;Ν_{ 3 -氟-4-[(3-氟苯甲基)氧基]苯基)·6_[2_({[2(甲烷磺醯基)乙基]胺 143939.doc -108- 201019961 基}甲基)-l,3-噻唑-4-基]-4-喹唑啉胺;N-(l-苯甲基-1H-吲 唑-5-基)-6-[2-({[2-(甲烷磺醯基;)乙基]胺基)甲基)_13_噻 唑-4-基]-4-喹唑啉胺;!^_(3_氟_4_苯甲氧基苯基 (甲烧續酿基)乙基]胺基)甲基)_〗,3_嗟唾·4_基]_4_喹唑琳 胺;N-(3-氯-4-笨甲氧基苯基;(甲烷磺醯基)乙 基]胺基)甲基)-1,3-噻唑-4-基]-4-喹唑啉胺;N-{3-氣-4-[(3-氟苯甲基)氧基]苯基}-6-[5-({[2-(甲烷磺醯基)乙基]胺基)甲 基)-2-呋喃基]-4-喹唑啉胺;6-[5-({[2-(曱烷磺醯基)乙基] 胺基)甲基)-2-咬淹基]-7-甲氧基-N-(4-苯磺醯基)苯基-4-喹 唾琳胺;N-[4-(苯甲氧基)苯基]_7_氟_6_[5_({[2_(甲烷磺醯 基)乙基]胺基)甲基)-2-呋喃基]-4-喹唑啉胺;N-(l-苯甲基-1H-吲唾-5-基)-7-氟-6-[5-({[2-(甲烷磺醢基)乙基]胺基}甲 基)-2-呋喃基]-4-喹峻嘛胺;N-[4-(苯續醯基)苯基]-7-氟-6-[5-({[2-(甲烷磺醯基)乙基]胺基}甲基)_2_呋喃基]_4_喹唑啉 胺;N-(3-三氟甲基-4-苯甲氧基苯基)-6-[5-({[2_(曱烷磺醯 基)乙基]胺基)甲基)-4-吱味基]-4-啥唾嘴胺;及其鹽及溶 劑合物。 在一特定實施例中,EGFR拮抗劑為N-[3-氣-4-[(3-氟苯 基)曱氧基]苯基]-6-[5-[[[2-(曱基磺醢基)乙基]胺基]甲基]-2-呋喃基]-4-喹唑啉胺二甲苯磺酸鹽(拉帕替尼)。 在一特定實施例中,EGFR拮抗劑為如WO 0132651(以引 用之方式併入本文中)中所揭示之式IV化合物, 143939.doc -109· 201019961Wherein each R5 is independently selected from the group consisting of halogen, C1-4 alkyl and C1-4 alkoxy; and η is 0 to 3; and each R4 is independently hydroxy, halogen, Cl-4 alkyl, c2-4 alkenyl , C2.4 alkynyl, c 4·alkoxy, amine, Cl 4 alkylamino, bis[Cl 4 alkyl]amine, Cw thiol, Cl 4 alkylsulfinyl, Cl 4 alkyl Sulfonic acid group, Cl 4 alkyl group, ruthenium, amine carbaryl group, Cl-4 alkoxycarbonyl group, Cl 4 alkylalkylamino group, N-%-4 alkyl group, amine fluorenyl group, n, N - bis(Cw alkyl)amine fluorenyl, cyano, nitro and trifluoromethyl. In a specific embodiment, the antagonist of the formula ni2 EGFR is excluded: (1-benzoinyl 1H-indolyl-5-yl)-(6-(5-((2-methylpyrene)ethylamino) _Methyl)-furan-2-yl)-pyrido[3,4-d]pyrimidin-4-yl-amine; (4-benzoyloxyphenyl)-(6-(5-((2-decane)) Sulfhydryl-ethylamino)-methyl)-furan-2-yl)-. than biting and [3,4-d] nits 4-yl-amine; (1-phenylmethyl-1H-吲.Sit_5_yl)-(6-(5·((2-decanesulfonyl)ethyl)ethyl)-furan-2-ylquinazoline core - 143939.doc • 106· 201019961 Amine (1 benzyl hydrazino-indazol-5-yl)-(7-(5-((2-methanesulfonyl)ethylamino)-methyl)-furan-2-yl) _ quinazoline _4_ yl-amine; and (1 benzyl-_ιη_carbazolyl)-(6-(5-((2-methanesulfonyl-ethylamino)-methyl)) _ Pyrrolyl)-quinazolin-4-yl-amine. In a particular embodiment, the EGFR antagonist of Formula III is selected from the group consisting of: 4·(4-fluorobenzyloxy)-phenyl Η6-(5-((2·甲烧烧基基基基基基基)_〇夫喃-2-yl)-η ratio bite [3,4-d] bite _4_ base)_ Amine; ((fluoro*benzyloxy)-benzyl)-(6 -(5-((2-methylpropioninyl-ethylamino)methyl)furan-2-yl)-D-pyrido[3,4-d]pyrimidin-4-yl)-amine; (4- Phenylsulfonyl-phenyl)-(6-(5-((2-decanesulfonyl)ethylamino)-methyl)-furan-2_pyridinium[3,4-d]pyrimidine -4-yl)-amine; (4-phenyloxy-phenyl)-(6-(3-((2-methanesulfonyl-ethylamino))methyl)phenyl)pyridinium[ 34d] pyrimidin-4-yl)-amine; (4-benzoyloxy-phenyl)-(6_(5·((2 methanesulfonyl)ethylamino)-indenyl)-» (2) quinazoline _4_yl)-amine; (4-(3-fluorobenzyloxy-phenyl)-(6-(4-((2-methanesulfonyl-ethylamino)) ) _methyl)-furan-2-yl)-n-pyrido[34_d]pyrimidin-4-yl)amine; (4-benzoyloxyphenyl)_(6-(2-((2-methanesulfonyl)) Ethylethylamino)-methylthiazole-4-yl)quinazoline-4-yl)-amine; N-{4-[(3-fluoromethyl)oxy]phenyl}_6_[5· ({[2_(decane fluorescein) ethyl]amino}methyl)_2_furanyl]_4_quinazolinamine; Ν-{4-[(3-fluorobenzyl)oxy] -3-methoxyphenyl}_6_[5·({[2_(methanesulfonyl)ethyl]amino}methyl)-2-furanyl]_4_ quin啉-[4-(phenylhydroxy)phenyl]-7-methoxy-6-[5-({[2-(methanesulfonyl)ethyl)amino}methyl)· 2·furanyl]-4-quinazolinamine; Ν_[4_(benzyloxy)phenyl]_6_[4_({[2-(methanesulfonyl)ethyl]amino}indenyl)_2_ Furanyl]_4_quinazoline 143939.doc -107- 201019961 Amine; Ν-{4-[(3·fluorobenzyl)oxy]-3.methoxyphenyl}-6-[2-( {[2-(decanesulfonyl)ethyl]amino}indolyl)-l,3-thiazol-4-yl]-4-quinazolinamine; N-{4-[(3-bromo) Mercapto)oxy]phenyl}_6-[2-({|;2•(decanesulfonyl)ethyl]amino}methyl)-1,3-thiasen-4-yl]-4 - quinazoline; N-{4-[(3-fluorophenylindenyl)oxy]phenyl)_6-[2-({[2-(methanesulfonyl)ethyl)amino}}} 1,3-thiazol-4-yl]-4-quinazolinamine; N-[4-(benzyloxy)-3-fluorophenyl]-6-[2-({[2-(methane) Sulfhydryl)ethyl]amino)indenyl)_1,3-thiazole-4-yl]-4-quinazolinamine; N-(l-benzyl-1H-carbazol-5-yl)- 7-decyloxy-6-[5-({[2-(methanesulfonyl)ethyl)amino)indolyl)_2_furanyl]_4_quinazolinamine·' 6-[5-( {[2-(decanesulfonyl)ethyl]amine;) Base)_2_furanyl]_N-(4-{[3-(trifluoromethyl)benzyl]oxy)phenyl)-4-quinazolinamine; N_ {3-fluoro-4-[( 3-fluorobenzyl)oxy]phenyl}_6_[5_({[2_(decanesulfonyl)ethyl]amino)methyl)-2-furanyl]-4-quinalin; N-{4-[(3-Bromobenzyl)oxy]phenyl)-6-[5-({[2-(decanesulfonyl)ethyl;]amino)indolyl)_2_ bite喃 ]]-4-quinazolinamine; N-[4-(benzyloxy)phenyl]-6_[3-({[2-(A))]]]}}}} _2_furanyl]_4_quinazoline; n-[1-(3-fluorobenzyl)·ιη-carbazol-5-yl]-6-[2-({[2-(decane) Sulfonic acid)ethyl]amino}indenyl)-1,3-thiazol-4-yl]-4-quinazolinamine; 6-[5-({[2-( sulfonyl)) Amino]methyl)methyl)_2_furanylΝ[[(phenylsulfonyl)phenyl]-4-quinazolinamine; 6-[2-({[2-(methanesulfonyl))ethyl) Amino) sulfhydryl)ethyl]amino]amino]amino]amino]amino]amino]amino]amino]amino]amino]amino]amino]amino]amino]amino] }methylhydrazine, thiazole·4_yl]_Ν_(Μ[3·(difluoroindolyl)benzyl)oxy)phenyl)_4_quinazolinamine;Ν_{ 3 -fluoro-4-[ (3- Benzyl)oxy]phenyl)·6_[2_({[2(methanesulfonyl)ethyl)amine 143939.doc -108- 201019961 yl}methyl)-l,3-thiazol-4-yl ]-4-quinazolinamine; N-(l-benzyl-1H-indazol-5-yl)-6-[2-({[2-(methanesulfonyl)))]] )methyl)_13_thiazol-4-yl]-4-quinazolinamine; ^_(3_Fluoro-4_benzyloxyphenyl (methyl sulphonate) ethyl]amino)methyl)_, 3_嗟Sept. 4_yl]_4_ quinazoline; N-(3-chloro-4-phenoxyphenyl; (methanesulfonyl)ethyl]amino)methyl)-1,3-thiazol-4-yl]-4-quinazolinamine; N-{3-Ga-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulfonyl)ethyl)amino)methyl) -2-furyl]-4-quinazolinamine; 6-[5-({[2-(decanesulfonyl)ethyl]amino)methyl)-2-)) Methoxy-N-(4-phenylsulfonyl)phenyl-4-quinoxalin; N-[4-(benzyloxy)phenyl]_7_fluoro_6_[5_({[2_( Methanesulfonyl)ethyl]amino)methyl)-2-furyl]-4-quinazolinamine; N-(l-benzyl-1H-indole-5-yl)-7-fluoro -6-[5-({[2-(methanesulfonyl)ethyl)amino}methyl)-2-furanyl]-4- quinacamine; N-[4-(phenyl hydrazino) Phenyl]-7-fluoro-6-[5-({[2-(methanesulfonyl)ethyl)amino}methyl)_2-furanyl]_4_quinazolinamine; N-(3) -trifluoromethyl-4-benzyloxyphenyl)-6-[5-({[2_(decanesulfonyl)ethyl]amino)methyl)-4-oxime]-4 - 啥 啥 胺 amine; and its salts and solvents Thereof. In a specific embodiment, the EGFR antagonist is N-[3- gas-4-[(3-fluorophenyl)decyloxy]phenyl]-6-[5-[[[2-(indenyl) Indenyl)ethyl]amino]methyl]-2-furanyl]-4-quinazolinamine xylene sulfonate (lapatinib). In a particular embodiment, the EGFR antagonist is a compound of formula IV as disclosed in WO 0132651 (incorporated herein by reference), 143939.doc -109.

NN

IV 其中: m為1至3之整數;IV where: m is an integer from 1 to 3;

R1表示鹵基或C〗.3烷基; X1表示-0-; R2係選自以下三類基團中之—類: 1) Cw烧基R3(其中R3為哌 或兩個 W羥基烷基及C〗·4烷氧基之 心基,其可具有 選自羥基、鹵基、C〗-4烷基、c 取代基); 2) C2·5稀基R3(其中R3係如本文中所定義); 3) CM炔基R3(其中R3係如本文中所定義广 且其中任何烧基、烯基或块基均可具有一或多個選自經❹ 基、齒基及胺基之取代基;或其鹽。 在一特定實施例中,EGFR拮抗劑係選自由以下組成之 群:4-(4-氣-2-氟苯胺基)-6-甲氧基_7_(i•曱基0辰咬_4_基曱 氧基)喹唑啉;4-(2-氟-4-甲基苯胺基)_6_甲氧基_7_(i_曱基 哌啶-4-基甲氧基)喹唑啉;4-(4-溴_2-氟苯胺基)-6-曱氡基-7-(1-曱基哌啶-4-基曱氧基)喹唑啉;4-(4-氣-2,6-二氟苯胺 基)-6 -曱氧基- 7-(1-甲基略咬-4-基甲氡基)啥。坐淋;4-(4-漠- 143939.doc -110- 201019961 2,6-二氟苯胺基)-6-甲氧基-7-(1-曱基哌啶-4-基曱氧基)喹 〇坐嚇·,4-(4-乳-2 -氣苯胺基)-6 -曱氧基- 7- (旅咬-4-基甲氧基) 喧ί坐琳,4-(2 -氣-4-甲基苯胺基)-6-甲氧基- 7- (旅唆-4-基曱 氧基)啥β坐淋,4-(4 -》臭-2 -氣苯胺基)-6-曱氧基- 7- (派咬-4-基 甲氧基)喹唑啉;4-(4-氣-2,6-二氟苯胺基)-6-甲氧基-7-(哌 啶-4-基曱氧基)喹唑啉;4-(4-溴-2,6-二氟苯胺基)-6-曱氧 基-7-(哌啶-4-基甲氧基)喹唑啉;及其醫藥學上可接受之鹽 及溶劑合物。 在一特定實施例中,EGFR拮抗劑為4-(4-溴-2-氟苯胺 基)-6-甲氧基-7-(1-曱基哌啶-4-基曱氧基)喹唑啉(Zactima) 及其鹽。 VEGF拮抗劑 VEGF拮抗劑係指能夠結合VEGF、降低VEGF表現含量 或中和、阻斷、抑制、消除、降低或干擾VEGF活性(包括 VEGF與一或多種VEGF受體之結合及VEGF介導之血管生 成及内皮細胞存活或增殖)之分子。適用於本發明方法之 VEGF拮抗劑包括特異性結合VEGF之多肽、抗VEGF抗體 及其抗原結合片段、特異性結合VEGF從而隔絕其與一或 多種受體之結合的受體分子及衍生物、融合蛋白(例如 VEGF-Trap (Regeneron))及 VEGFm-白樹素(Peregrine)。 VEGF拮抗劑亦包括VEGF多肽、針對VEGF之RNA適體及 肽體的拮抗變異體。此等物質各自之實例描述如下。 適用於本發明方法之抗VEGF抗體包括以足夠親和力及 特異性結合VEGF且可降低或抑制VEGF之生物活性之任何 143939.doc -111 - 201019961 抗體或其抗原結合片段。抗VEGF抗體通常應不結合其他 VEGF同系物(諸如VEGF-B或VEGF-C),亦不結合其他生長 因子(諸如P1GF、PDGF或bFGF)。該等抗VEGF抗體之實例 包括(但不限於)本文「定義」中所提供之抗VEGF抗體。 兩種充分表徵之VEGF受體為VEGFR1(亦稱為Flt-Ι)及 VEGFR2(對於鼠類同系物,亦稱為KDR及FLK-1)。雖然各 VEGF家族成員之各受體之特異性不同,但VEGF-A結合 Flt-1與KDR。全長Flt-1受體包括具有7個Ig域之細胞外 域、跨膜域及具有酷胺酸激酶活性之細胞内域。細胞外域 參與VEGF之結合,而細胞内域參與信號轉導。 本發明方法中可使用特異性結合VEGF之VEGF受體分子 或其片段來結合且隔絕VEGF蛋白,從而阻止其傳導信 號。在某些實施例中,VEGF受體分子或其VEGF結合片段 為可溶形式,諸如sFlt-1。該受體之可溶形式係藉由結合 VEGF而對VEGF蛋白之生物活性具有抑制效應,從而阻止 其結合把細胞表面上所存在之其天然受體。亦包括VEGF 受體融合蛋白,其實例描述如下。 嵌合VEGF受體蛋白為具有源自至少兩種不同蛋白質之 胺基酸序列的受體分子,其中至少一種蛋白質為能夠結合 VEGF且抑制其生物活性之VEGF受體蛋白(例如fit-1或KDR 受體)。在某些實施例中,本發明之嵌合VEGF受體蛋白係 由僅源自兩種不同的VEGF受體分子之胺基酸序列組成; 然而’包含fit-1及/或KDR受體之細胞外配位艎結合區之 1、2、3、4、5、6或所有7個Ig樣域的胺基酸序列可與其 143939.doc •112- 201019961 他無關蛋白質之胺基酸序列(例如免疫球蛋白序列)連接。 與Ig樣域組合之其他胺基酸序列對一般熟習此項技術者而 言將顯而易見。嵌合VEGF受體蛋白之實例包括可溶性Flt-1/Fc、KDR/Fc或 Flt-1/KDR/Fc(亦稱為 VEGF Trap)。(參見 例如PCT申請公開案第WO 97/44453號)。 本發明之可溶性VEGF受體蛋白或嵌合VEGF受體蛋白包 括不經由跨膜域固定於細胞表面上之VEGF受體蛋白。因 而,VEGF受體之可溶形式(包括嵌合受體蛋白)雖然能夠結 合VEGF且使之失活,但不包含跨膜域且因此一般不與表 現該分子之細胞之細胞膜聯結。 適體為形成特異性結合靶分子(諸如VEGF多肽)之三級 結構之核酸分子。適體之產生及治療用途在此項技術中非 常明確。參見例如美國專利第5,475,096號。VEGF適體為 聚乙二醇化修飾之寡核苷酸,其採用使其能夠結合細胞外 VEGF之三維構形。靶向VEGF以治療年齡相關黃斑變性之 治療上有效適體之一實例為略加他尼(pegaptanib) (Macugen™,OSI)。關於適體之其他資訊可見於美國專利 申請公開案第20060148748號中。 肽體為與編碼免疫球蛋白分子之片段或一部分之胺基酸 序列連接的肽序列。多肽可衍生自由任何關於特異性結合 之方法(包括但不限於噬菌體呈現技術)所選擇之隨機序 列。在一實施例中,所選多肽可與編碼免疫球蛋白之Fc部 分之胺基酸序列連接。特異性結合且拮抗VEGF之肽體亦 適用於本發明方法。 143939.doc • 113- 201019961 组合療法 本發明係關於組合使用卜爪^拮抗劑及VEGF拮抗劑,且 在一些態樣中’係關於組合使用c_met拮抗劑、VEGF拮抗 劑及EGFR拮抗劑作為意欲由此等治療劑之組合活性提供 有益效應之特定治療方案的一部分^組合之有益效應包括 (但不限於)由治療劑之組合所產生之藥物動力學或藥效學 共同作用。本發明尤其適用於治療處於各病期之各種類型 之癌症。 術語癌症包括增殖病症之集合,包括(但不限於)癌前期 生長、良性腫瘤及惡性腫瘤。良性腫瘤保持定位於起源位 點且不具有浸潤、侵襲或轉移至遠處位點之能力。惡性腫 瘤將侵襲且損傷其周圍之其他組織。其亦能夠脫離最初位 點且通常經由血流或經由淋巴結所處之淋巴系統擴散至身 體之其他部分(轉移)。原發性腫瘤係根據產生該等腫瘤之 纪織類型分類;轉移性腫瘤係根據產生癌細胞之組織類型 分類。惡性腫瘤之細胞隨時間變得愈加異常且看來更不像 正常細胞。癌細胞之此外觀變化係稱為腫瘤等級,且癌細 胞係描述為充分分化(低級)、中度分化、不良分化或未分 化(高級)。充分分化細胞看起來相當正常且類似於產生其 之正常細胞《未分化細胞為已變得異常以致不再可能確定 細胞之起源的細胞。 癌症分期系統描述癌症在解剖學上已擴散多遠且試圖將 具有類似預後及治療之患者歸為同一病期組中。可進行數 種測試來幫助癌症分期’包括生檢及某些成像測試 143939.doc •114- 201019961 (imaging test)(諸如胸部x光攝影(chest x-ray)、乳房x光攝 影(mammogram)、骨掃描、CT掃描及MRI掃描)。亦使用 血液測試及臨床評估來評估患者之總體健康狀況且偵測癌 症是否已擴散至某些器官。 為對癌症進行分期,美國癌症聯合委員會(American Joint Committee on Cancer)首先使用TNM分類系統以字母 分類排列癌症(尤其實體腫瘤)。對癌症指定字母T(腫瘤大 小)、Ν(可觸知結節)及/或Μ(轉移)。ΤΙ、Τ2、Τ3及Τ4描述 遞增大小之原發性病變;NO、Nl、Ν2、Ν3指示進行性發 展之結節涉入(node involvement);且M0及Ml反映無遠位 轉移存在或存在遠位轉移。 在亦稱為總病期分組(Overall Stage Grouping)或羅馬數 字分期(Roman Numeral Staging)之第二分期方法中,综合 原發性病變之大小及結節擴散及遠位轉移之存在,將癌症 分為0至IV期。在此系統中,將病例分組為由羅馬數字I至 IV表示之四期,或分類為「復發性」。對於一些癌症,0期 係稱為「原位」或「Tis」,諸如乳房癌之乳腺管原位癌或 小葉原位癌。高級腺瘤亦可分類為0期。I期癌症一般為通 常可治癒之小的局部癌症,而IV期通常表示不能施行手術 之癌症或轉移性癌症。II期及III期癌症通常為局部晚期及/ 或展現局部淋巴結涉入。較高級數字一般指示較廣泛疾 病,包括較大腫瘤大小及/或癌症擴散至附近淋巴結及/或 鄰近原發性腫瘤之器官。雖然此等病期係經精確定義,但 各類癌症之定義不相同且為熟習此項技術者所知。 H3939.doc -115- 201019961 許多癌症登記處(諸如美國國家癌症研究所之監視、流 行病學及最終結果程式(Surveillance, Epidemiology, and End Results Program,SEER))使用總結分期(summary staging)。此系統用於所有類型之癌症。其將癌症病例分 組成5個主要類別: 及位為僅存在於開始之細胞層中之早期癌症。 々新允為侷限於開始器官,而無擴散跡象之癌症。 厘域#為已擴散超出最初(原發)位點至附近淋巴結或器 官及組織之癌症。 磋位為已自原發位點擴散至遠位器官或遠位淋巴結之癌 症。 未如读用於描述無足夠資訊來指示病期之病例。 另外,癌症通常在原發性腫瘤移除後數月或數年復發。 在所有可見腫瘤均已去除後復發之癌症係稱為復發性疾 病。在原發性腫瘤區域内復發之疾病為局部復發性,而轉 移復發之疾病係稱為遠位復發。 腫瘤可為實體腫瘤或非實體或軟組織腫瘤。軟組織腫瘤 之實例包括白血病(例如慢性骨腾性白血病、急性骨趙性 白血病、成年急性淋巴母細胞白血病、急性骨髓性白血 病、成熟B細胞急性淋巴母細胞白血病、慢性淋巴球性白 血病、前淋巴球性白血病或毛細胞白血病)或淋巴瘤(例如 非霍奇金氏淋巴瘤(non-Hodgkin’s lymphoma)、皮膚T細胞 淋巴瘤或霍奇金氏病(Hodgkin’s disease))。實趙趙瘤包括 除血液、骨锻或淋巴系統以外之身體組織之任何癌症。實 143939.doc -116· 201019961R1 represents a halo group or a C.3 alkyl group; X1 represents -0-; R2 is selected from the following three groups: 1) Cw alkyl R3 (wherein R3 is a pipe or two W-hydroxyalkyl groups) And a core group of C alkoxy group, which may have a group selected from the group consisting of a hydroxyl group, a halogen group, a C alkyl group, a c substituent; 2) a C 2 ·5 rare group R 3 (wherein R 3 is as herein described) 3) CM alkynyl R3 (wherein R3 is as defined herein and any of the alkyl, alkenyl or block groups may have one or more substituents selected from the group consisting of fluorenyl, dentate and amine groups Or a salt thereof. In a particular embodiment, the EGFR antagonist is selected from the group consisting of 4-(4-Gas-2-fluoroanilino)-6-methoxy-7-(i• fluorenyl) 0辰咬_4_yloxy)quinazoline; 4-(2-fluoro-4-methylanilino)_6_methoxy_7_(i-mercaptopiperidin-4-ylmethoxy a quinazoline; 4-(4-bromo-2-fluoroanilino)-6-mercapto-7-(1-indolylpiperidin-4-ylindoleoxy)quinazoline; 4-(4) - gas-2,6-difluoroanilino)-6-decyloxy-7-(1-methyl-singe-4-ylmethylindenyl) hydrazine. sitting-in; 4-(4- desert- 143939. Doc -110- 201019961 2,6-Difluoroanilino)-6-methoxy-7-(1-mercaptopiperidone 4--4-mercaptooxy)quinoquinone, 4-(4-milo-2-oxoanilino)-6-indolyloxy-7-(Big -4-ylmethoxy) 喧ί sitting Lin, 4-(2- gas-4-methylanilino)-6-methoxy-7-((唆-4-基曱ethoxy)啥β坐淋,4-(4 -》臭-2 - aza-anilino)-6-decyloxy-7-(pyrylene-4-ylmethoxy)quinazoline; 4-(4-gas-2,6-difluoroanilino)-6-methoxy -7-(piperidin-4-ylmethoxy)quinazoline; 4-(4-bromo-2,6-difluoroanilino)-6-decyloxy-7-(piperidin-4- Methoxy)quinazoline; and pharmaceutically acceptable salts and solvates thereof. In a particular embodiment, the EGFR antagonist is 4-(4-bromo-2-fluoroanilino)-6- Methoxy-7-(1-mercaptopiperidin-4-ylindoleoxy)quinazoline (Zactima) and its salts. VEGF antagonist VEGF antagonist refers to the ability to bind VEGF, reduce VEGF expression content or neutralize a molecule that blocks, inhibits, eliminates, reduces or interferes with VEGF activity, including binding of VEGF to one or more VEGF receptors, and VEGF-mediated angiogenesis and endothelial cell survival or proliferation. VEGF antagonism suitable for use in the methods of the invention Agent includes specific binding to VEGF Peptides, anti-VEGF antibodies and antigen-binding fragments thereof, receptor molecules and derivatives which specifically bind to VEGF to isolate their binding to one or more receptors, fusion proteins (such as VEGF-Trap (Regeneron)) and VEGFm-white ( Peregrine). VEGF antagonists also include VEGF polypeptides, antagonists of RNA aptamers against VEGF, and antagonists of peptibodies. Examples of each of these materials are described below. Anti-VEGF antibodies suitable for use in the methods of the invention include any of the 143939.doc-111-201019961 antibodies or antigen-binding fragments thereof that bind to VEGF with sufficient affinity and specificity and which reduce or inhibit the biological activity of VEGF. Anti-VEGF antibodies should generally not bind to other VEGF homologs (such as VEGF-B or VEGF-C) or other growth factors (such as P1GF, PDGF or bFGF). Examples of such anti-VEGF antibodies include, but are not limited to, the anti-VEGF antibodies provided in the "Definitions" herein. Two well characterized VEGF receptors are VEGFR1 (also known as Flt-Ι) and VEGFR2 (for murine homologs, also known as KDR and FLK-1). Although the specificity of each receptor of each VEGF family member is different, VEGF-A binds to Flt-1 and KDR. The full-length Flt-1 receptor includes an extracellular domain having seven Ig domains, a transmembrane domain, and an intracellular domain having tyrosine kinase activity. The extracellular domain is involved in the binding of VEGF, while the intracellular domain is involved in signal transduction. A VEGF receptor molecule or a fragment thereof that specifically binds to VEGF can be used in the method of the invention to bind and sequester the VEGF protein, thereby preventing its conduction signal. In certain embodiments, the VEGF receptor molecule or VEGF binding fragment thereof is in a soluble form, such as sFlt-1. The soluble form of the receptor has an inhibitory effect on the biological activity of the VEGF protein by binding to VEGF, thereby preventing its binding to its natural receptor present on the cell surface. VEGF receptor fusion proteins are also included, examples of which are described below. A chimeric VEGF receptor protein is a receptor molecule having an amino acid sequence derived from at least two different proteins, wherein at least one of the proteins is a VEGF receptor protein capable of binding to VEGF and inhibiting its biological activity (eg, fit-1 or KDR) Receptor). In certain embodiments, a chimeric VEGF receptor protein of the invention consists of an amino acid sequence derived only from two different VEGF receptor molecules; however, a cell comprising a fit-1 and/or KDR receptor The amino acid sequence of 1, 2, 3, 4, 5, 6, or all 7 Ig-like domains of the outer coordination 艎 binding region may be associated with its amino acid sequence (eg, immunization) with its 143939.doc •112-201019961 The globin sequence) is linked. Other amino acid sequences in combination with an Ig-like domain will be apparent to those of ordinary skill in the art. Examples of chimeric VEGF receptor proteins include soluble Flt-1/Fc, KDR/Fc or Flt-1/KDR/Fc (also known as VEGF Trap). (See, for example, PCT Application Publication No. WO 97/44453). The soluble VEGF receptor protein or chimeric VEGF receptor protein of the invention comprises a VEGF receptor protein that is not immobilized on the cell surface via a transmembrane domain. Thus, soluble forms of the VEGF receptor (including chimeric receptor proteins), while capable of binding to and inactivating VEGF, do not comprise a transmembrane domain and are therefore generally not associated with cell membranes of cells expressing the molecule. An aptamer is a nucleic acid molecule that forms a tertiary structure that specifically binds to a target molecule, such as a VEGF polypeptide. The production and therapeutic use of aptamers is well established in the art. See, for example, U.S. Patent No. 5,475,096. The VEGF aptamer is a PEGylated oligonucleotide that is configured to bind to the three-dimensional configuration of extracellular VEGF. An example of a therapeutically effective aptamer that targets VEGF to treat age-related macular degeneration is pegaptanib (MacugenTM, OSI). Further information on aptamers can be found in U.S. Patent Application Publication No. 20060148748. A peptidic body is a peptide sequence linked to an amino acid sequence encoding a fragment or a portion of an immunoglobulin molecule. The polypeptide may be derived from any random sequence selected for any method of specific binding, including but not limited to phage display technology. In one embodiment, the selected polypeptide can be linked to an amino acid sequence encoding the Fc portion of an immunoglobulin. Peptides that specifically bind to and antagonize VEGF are also suitable for use in the methods of the invention. 143939.doc • 113-201019961 Combination Therapy The present invention relates to the use of a combination of a paw antagonist and a VEGF antagonist, and in some aspects 'for the combined use of a c-met antagonist, a VEGF antagonist and an EGFR antagonist as intended The beneficial effects of a combination of specific therapeutic regimens that provide a beneficial effect of such therapeutic agents include, but are not limited to, pharmacokinetic or pharmacodynamic effects resulting from a combination of therapeutic agents. The invention is particularly useful for treating various types of cancer at various stages of the disease. The term cancer includes a collection of proliferative disorders including, but not limited to, precancerous growth, benign tumors, and malignant tumors. Benign tumors remain localized at the site of origin and do not have the ability to infiltrate, invade, or metastasize to distant sites. A malignant tumor will invade and damage other tissues around it. It can also be detached from the original site and typically spread to other parts of the body (metastasis) via the bloodstream or via the lymphatic system in which the lymph nodes are located. Primary tumors are classified according to the type of tissue from which the tumors are produced; metastatic tumors are classified according to the type of tissue from which the cancer cells are produced. Cells of malignant tumors become more abnormal over time and appear to be less like normal cells. This change in appearance of cancer cells is called tumor grade, and the cancer cell line is described as sufficiently differentiated (lower), moderately differentiated, poorly differentiated, or undifferentiated (advanced). Fully differentiated cells appear to be quite normal and similar to the normal cells from which they are produced. "Undifferentiated cells are cells that have become abnormal so that it is no longer possible to determine the origin of the cell. The cancer staging system describes how far the cancer has been anatomically spread and attempts to classify patients with similar prognosis and treatment into the same disease group. Several tests can be performed to help with cancer staging' including biopsy and some imaging tests 143939.doc •114- 201019961 (imaging test) (such as chest x-ray, mammogram, Bone scan, CT scan and MRI scan). Blood tests and clinical assessments are also used to assess the overall health of the patient and to detect if the cancer has spread to certain organs. To stage cancer, the American Joint Committee on Cancer first uses a TNM classification system to sort cancers (especially solid tumors) in alphabetical order. Specify the letter T (tumor size), sputum (tactile nodules), and/or sputum (metastasis) for cancer. ΤΙ, Τ2, Τ3, and Τ4 describe incremental lesions of primary lesions; NO, Nl, Ν2, Ν3 indicate progressive involvement of node involvement; and M0 and Ml reflect the absence of distant metastasis or the presence of distant sites Transfer. In the second staging method, also known as Overall Stage Grouping or Roman Numeral Staging, the cancer is divided into the size of the primary lesion and the presence of nodular spread and distant metastasis. 0 to IV. In this system, cases are grouped into four phases, represented by Roman numerals I through IV, or classified as "recurrent." For some cancers, stage 0 is called "in situ" or "Tis", such as breast ductal carcinoma in situ or lobular carcinoma in situ. Advanced adenomas can also be classified as stage 0. Stage I cancers are generally small, localized cancers that are usually curable, while stage IV usually indicates cancer or metastatic cancer that cannot be operated on. Stage II and III cancers are usually locally advanced and/or exhibit local lymph node involvement. Higher grades generally indicate a broader range of diseases, including larger tumor sizes and/or cancer spreading to nearby lymph nodes and/or organs adjacent to the primary tumor. Although these stages are precisely defined, the definitions of each type of cancer are not the same and are known to those skilled in the art. H3939.doc -115- 201019961 Many cancer registries (such as the National Cancer Institute's Surveillance, Epidemiology, and End Results Program (SEER)) use summary staging. This system is used for all types of cancer. It divides cancer cases into five main categories: and is an early cancer that exists only in the initial cell layer. Xin Xin is a cancer that is limited to the beginning of the organ without signs of spread.厘域# is a cancer that has spread beyond the original (primary) site to nearby lymph nodes or organs and tissues. The study is a cancer that has spread from the primary site to the distal or distant lymph nodes. Not as read to describe cases where there is not enough information to indicate the stage of the disease. In addition, cancer usually recurs months or years after the primary tumor is removed. A cancer that recurs after all visible tumors have been removed is called a recurrent disease. The disease that recurs in the primary tumor area is locally recurrent, and the disease that metastasizes and relapses is called distant recurrence. The tumor can be a solid tumor or a non-solid or soft tissue tumor. Examples of soft tissue tumors include leukemia (eg, chronic osteogenic leukemia, acute osteogenic leukemia, adult acute lymphoblastic leukemia, acute myeloid leukemia, mature B cell acute lymphoblastic leukemia, chronic lymphocytic leukemia, anterior lymphocytes) Leukemia or hairy cell leukemia) or lymphoma (eg, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, or Hodgkin's disease). Shi Zhao Zhao Tu includes any cancer of body tissue other than blood, bone forging or lymphatic system. Real 143939.doc -116· 201019961

體腫瘤可進一步分為上皮細胞起源之實體腫瘤及非上皮細 胞起源之實體肢瘤。上皮細胞實體腰瘤之實例包括胃腸道 腫瘤、結腸腫瘤、乳房腫瘤、前列腺腫瘤、肺腫瘤、腎腫 瘤、肝Μ瘤、胰腫瘤、卵巢腫瘤、頭頸部腫瘤、口腔腫 瘤、胃腫瘤、十一指腸肢瘤、小腸腫瘤、大腸腫瘤、肛門 腫瘤、膽囊腫瘤、唇腫瘤、鼻咽腫瘤、皮膚腫瘤、子宮腫 瘤、雄性生殖器腫瘤、泌尿器腫瘤、膀胱腫瘤及皮膚腫 瘤。非上皮起源之實體腫瘤包括肉瘤、腦腫瘤及骨腫瘤。 除本文描述之治療方案外’亦可使患者經受手術移除癌 細胞及/或放射療法。 孜興弟二(第 可與之組合其他治療方案 三、第四等)化學治療劑,其中該第二化學治療劑為另 種不同的抗代謝物化學治療劑,或不為抗代謝物之化學治 療劑舉例而s ’第二化學治療劑可為紫杉烷(諸如紫杉 德(加。㈣或太平洋紫杉醇或多烯紫杉醇)、抗代謝物藥 物(諸如吉西他濱或5.氟尿㈣)、卡培他濱❹基化學治 =劑(諸如卡翻、順麵或奥赛力鈷)、蒽環黴素(諸如阿黴 括脂質體阿黴素)、拓朴替康、培美㈣' 長春花 驗(諸如長春瑞賓)及取286。可投與不同化學治 療劑之混合液(cocktail)」。 以;:抗=體及咖拮抗劑組合之其他治療劑包括 物例: 針對腫瘤相關抗原之抗體’·抗激素化合 Γ如ft雌激素化合物(諸如他莫昔芬)或芳香酶抑制 保心樂(阻止或降低與療法相關之任何心肌功能: 143939.doc -117- 201019961 礙);細胞激素;抗血管生成劑(尤其由Genentech以商標 AVASTIN J出售之貝伐單抗);路胺酸激酶抑制劑,諸如舒 尼替尼(sunutinib)(SUTENT)及索拉非尼(sorafenib) ; COX 抑制劑(例如COX-1或COX-2抑制劑);非類固醇消炎藥 物,塞來昔布(CELEBREX7);法呢基轉移酶抑制劑(例 如,替皮法尼/可獲自 Johnson and Johnson之ZARNESTRA7 R115777 或可獲自 Schering-Plough 之洛那法尼 SCH66336 (Lonafarnib SCH66336)) ; mTOR抑制劑,諸如RAD001 及 特癌適(temsirolimus);結合癌胚蛋白質CA 125之抗體,諸 如奥格伐單抗(Oregovomab)(MoAb B43.13) ; HER2 疫苗(諸 如來自 Pharmexia之 HER2 AutoVac疫苗,或來自 Dendreon 之APC8024蛋白質疫苗,或來自GSK/Corixa之HER2肽疫 苗);另一 HER靶向療法(例如曲妥珠單抗、西妥昔單抗、 ABX-EGF、EMD7200、吉非替尼、埃羅替尼、帕尼單抗、 CP724714、CI1033、GW572016、IMC-11F8、TAK165 等); Raf及/或ras抑制劑(參見例如WO 2003/86467);阿黴素鹽 酸鹽脂質體注射液(DOXIL®);拓撲異構酶I抑制劑,諸如 拓朴替康;紫杉烷;HER2及EGFR雙重酪胺酸激酶抑制 劑,諸如拉帕替尼 /GW572016 ; TLK286(TELCYTA®); EMD-7200 ;治療噁心之藥物,諸如血清素拮抗劑、類固 醇或苯并二氮呼;預防或治療皮瘆之藥物或標準痤瘡療 法,包括局部或口服抗生素;治療或預防腹瀉之藥物; 降低體溫之藥物,諸如乙醯胺苯酚、苯海拉明 (diphenhydramine)或唤咬(meperidine);造血生長因子等。 143939.doc -118· 201019961 任何上述共投與藥劑之合適劑量均為目前所使用之劑量, 且歸因於該藥劑與抗e_met抗體及egfr拮抗劑之組合作用 (協同作用),該等劑量可降低,或例如治療醫師所確定, 該等劑量可增加。 . 當抑制劑為抗體時,所投與之抗體較佳為裸抗體。然 而,所投與之抑制劑可與細胞毒性劑結合。所結合之抑制 劑及/或其所結合之抗原較佳經細胞内化,從而使得結合 物殺死其所結合之癌細胞之治療功效增加^在一較佳實施 例中,細胞毒性劑靶向或干擾癌細胞中之核酸。該等細胞 毒性劑之實例包括美登素類、卡奇黴素、核糖核酸酶及 DNA核酸内切酶。 在一些實施例中,例如在療法之前及/或療法期間及/或 療法之後,使本文之患者經受診斷測試。一般而言,若進 行#斷測試,則可自需要療法之患者獲得樣品。當個體患 有癌症時,樣品可為腫瘤樣品或其他生物樣品,諸如生物 φ 體液,包括(但不限於)血液、尿、唾液、腹水或衍生物(諸 如血清及血漿)及其類似物。 在一些實施例中’個體之癌症表現c-met及/或EGFR。測 • 疋c-met或EOFR表現之方法在此項技術中為已知的 且本文 描述某些方法。 在一些實施例中’個體之血清表現高含量之IL8。在一 些實施例中’個體之血清表現超過約15〇 pg/mliIL8,或 在一些實施例中,超過約5〇 pg/mli IL8。在一些實施例 中’個體之jk清表現超過約1〇 pg/m卜2〇 pg/m卜3〇 pg/ml 143939.doc -119· 201019961 或更高之IL8。測定IL8血清濃度之方法在此項技術中為已 知的,且本發明實例中描述一種方法。 在一些實施例中,個體之血清表現高含量之HGF。在一 些實施例中,個體之血清表現超過約5,000 pg/ml、10,000 pg/ml或 50,000 pg/ml之HGF ° 在一些實施例中,例如來自經c-met括抗劑治療(且在一 些實施例中,進一步經VEGF拮抗劑治療)之患者之腫瘤或 血清的樣品中mRNA或蛋白質表現之降低預後例如對治療 之反應或c-met拮抗劑之活性,且在一些實施例中,預後c-met拮抗劑及VEGF拮抗劑之活性。在一些實施例中,數種 血管生成因子(諸如介白素8(IL8)、血管内皮細胞生長因子 A(VEGFA)、EPH受體A2(EphA2)、血管生成素樣蛋白 4(Angptl4)及Ephrin B2(EFNB2))之表現降低預後例如對治 療之反應或c-met拮抗劑之活性(且在一些實施例中,預後 VEGF拮抗劑之活性)。表現之降低可相對於未經處理之樣 品或參考標準值或相對於經c-met拮抗劑治療(或經c-met拮 抗劑及VEGF拮抗劑治療)之前患者之表現含量來確定。 在一些實施例中,例如來自患者腫瘤或血清之樣品中 HGF或IL8表現之降低預後例如對治療之反應或c-met拮抗 劑(且在一些實施例中,VEGF拮抗劑)之活性。在一實施例 中,血清中IL8表現之超過50%之降低或超過70%之降低 (例如相對於治療之前患者之IL8表現含量)指示對治療之反 應。表現之降低可相對於未經處理之樣品或參考標準值或 相對於經c-met拮抗劑治療(或經c-met拮抗劑及VEGF拮抗 143939.doc -120- 201019961 劑治療)之前患者之表現含量來確定。 在一些實施例中,例如來自經c-met拮抗劑治療(且在一 些實施例中,進一步經VEGF拮抗劑治療)之患者之腫瘤或 血清的樣品中mRNA或蛋白質表現之增加預後例如對治療 之反應或c-met拮抗劑(且在一些實施例中,VEGF拮抗劑) 之活性。表現之降低可相對於未經處理之樣品或參考標準 值或相對於經c-met拮抗劑治療(或經c-met拮抗劑及VEGF 拮抗劑治療)之前患者之表現含量來確定。 在一些實施例中,FDG-PET成像預後例如對治療之反應 或c-met拮抗劑之活性(且在一些實施例中,VEGF拮抗劑之 活性)。 因此,本發明亦提供預後方法。因此,所揭示之方法可 提供獲得適用於評估病症未來病程之資料及資訊(包括選 擇治療患者之適當療法)之簡便、高效且潛在具有成本效 益的方法。 在另一態樣中,本發明提供評估患有或疑似患有癌症之 患者的方法,該方法包含:基於比較患者之生物樣品中血 管内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血 管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現 與對照樣品中血管内皮細胞生長因子A(VEGFA)、EPH受 體A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現來預測患者之癌症預後;其中患者生物 樣品相對於對照樣品之血管内皮細胞生長因子A(VEGFA)、 EPH受體A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或 143939.doc •121 · 201019961Body tumors can be further divided into solid tumors of epithelial origin and solid limb tumors of non-epithelial origin. Examples of epithelial solid lumbar tumors include gastrointestinal tumors, colon tumors, breast tumors, prostate tumors, lung tumors, kidney tumors, hepatic tumors, pancreatic tumors, ovarian tumors, head and neck tumors, oral tumors, stomach tumors, and eleven fingers. Intestinal limb tumor, small intestine tumor, large intestine tumor, anal tumor, gallbladder tumor, lip tumor, nasopharyngeal tumor, skin tumor, uterine tumor, male genital tumor, urinary tumor, bladder tumor and skin tumor. Solid tumors of non-epithelial origin include sarcomas, brain tumors, and bone tumors. In addition to the treatment regimens described herein, patients can also undergo surgical removal of cancer cells and/or radiation therapy. A chemotherapeutic agent, which may be combined with other therapeutic regimens, third, fourth, etc., wherein the second chemotherapeutic agent is a different anti-metabolite chemotherapeutic agent, or a chemical that is not an antimetabolite The therapeutic agent is exemplified by s 'the second chemotherapeutic agent may be a taxane (such as yew (plus (4) or paclitaxel or docetaxel), an antimetabolite drug (such as gemcitabine or 5. fluorourethane (four)), card Peitabin thiol chemotherapy = agent (such as card turn, cistern or Osereco cobalt), anthracycline (such as amylin liposomal doxorubicin), topotecan, pemei (four) 'Changchun flower test (such as vinorelbine) and 286. can be administered a mixture of different chemotherapeutic agents. </ RTI>;: anti-body and coffee antagonist combination of other therapeutic agents including examples: antibodies against tumor-associated antigens '·Anti-hormone compounds such as ft estrogen compounds (such as tamoxifen) or aromatase inhibits the heart (to prevent or reduce any myocardial function associated with therapy: 143939.doc -117- 201019961); cytokines; Anti-angiogenic agents (especially by Genen Tech is sold as bevacizumab under the trademark AVASTIN J; glutamate kinase inhibitors such as sunutinib (SUTENT) and sorafenib; COX inhibitors (eg COX-1 or COX) -2 inhibitor); non-steroidal anti-inflammatory drug, celecoxib (CELEBREX7); farnesyltransferase inhibitor (eg, tipifani/ZARNESTRA7 R115777 available from Johnson and Johnson or available from Schering-Plough Lonfarnib SCH66336); mTOR inhibitors such as RAD001 and temsirolimus; antibodies that bind to carcinoembryonic protein CA 125, such as Oregovomab (MoAb B43.13) HER2 vaccine (such as the HER2 AutoVac vaccine from Pharmexia, or the APC8024 protein vaccine from Dendreon, or the HER2 peptide vaccine from GSK/Corixa); another HER targeted therapy (eg trastuzumab, cetuximab) , ABX-EGF, EMD7200, gefitinib, erlotinib, panitumumab, CP724714, CI1033, GW572016, IMC-11F8, TAK165, etc.); Raf and/or ras inhibitors (see eg WO 2003/86467 ); doxorubicin hydrochloride Liposomal injection (DOXIL®); topoisomerase I inhibitors such as topotecan; taxane; HER2 and EGFR dual tyrosine kinase inhibitors such as lapatinib/GW572016; TLK286 (TELCYTA® EMD-7200; a drug for treating nausea, such as a serotonin antagonist, a steroid or a benzodiazepine; a drug for preventing or treating dermatitis or a standard acne treatment, including topical or oral antibiotics; a drug for treating or preventing diarrhea; A drug that lowers body temperature, such as acetaminophen phenol, diphenhydramine or meperidine; hematopoietic growth factor. 143939.doc -118· 201019961 The appropriate dose of any of the above co-administered agents is the currently used dose, and due to the combination of the agent with anti-e_met antibody and egfr antagonist (synergy), the dose can be The dosage may be decreased, or as determined by the treating physician, for example. When the inhibitor is an antibody, the antibody administered is preferably a naked antibody. However, the inhibitor administered can be combined with a cytotoxic agent. The combined inhibitor and/or the antigen to which it binds is preferably intracellularly internalized such that the therapeutic effect of the conjugate to kill the cancer cells to which it binds is increased. In a preferred embodiment, the cytotoxic agent is targeted. Or interfere with nucleic acids in cancer cells. Examples of such cytotoxic agents include maytansinoids, calicheamicin, ribonucleases, and DNA endonucleases. In some embodiments, the patient herein is subjected to a diagnostic test, such as prior to and/or during therapy and/or after therapy. In general, if a # break test is performed, the sample can be obtained from a patient in need of therapy. When the individual has cancer, the sample can be a tumor sample or other biological sample, such as a biological fluid, including but not limited to blood, urine, saliva, ascites or derivatives (such as serum and plasma) and the like. In some embodiments, the cancer of an individual exhibits c-met and/or EGFR. Methods for measuring 疋c-met or EOFR performance are known in the art and some methods are described herein. In some embodiments, the serum of the individual exhibits a high level of IL8. In some embodiments, the serum of an individual exhibits more than about 15 〇 pg/mliIL8, or in some embodiments, more than about 5 〇 pg/mli IL8. In some embodiments, the individual's jk clear performance exceeds about 1 〇 pg/m b 2 〇 pg/m b 3 〇 pg/ml 143939.doc - 119 · 201019961 or higher IL8. Methods for determining the serum concentration of IL8 are known in the art, and a method is described in the examples of the present invention. In some embodiments, the individual's serum exhibits a high level of HGF. In some embodiments, the individual's serum exhibits an HGF of more than about 5,000 pg/ml, 10,000 pg/ml, or 50,000 pg/ml. In some embodiments, for example, from a c-metase treatment (and in some implementations) In the case of a tumor or serum sample of a patient further treated with a VEGF antagonist, the mRNA or protein exhibits a reduced prognosis such as a response to treatment or a c-met antagonist activity, and in some embodiments, a prognosis c- Activity of met antagonists and VEGF antagonists. In some embodiments, several angiogenic factors (such as interleukin 8 (IL8), vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and Ephrin The performance of B2 (EFNB2)) reduces the prognosis, for example, the response to treatment or the activity of a c-met antagonist (and in some embodiments, the activity of a prognostic VEGF antagonist). The decrease in performance can be determined relative to the untreated sample or reference standard value or relative to the level of performance of the patient prior to treatment with c-met antagonist (or treatment with c-met antagonist and VEGF antagonist). In some embodiments, for example, HGF or IL8 in a sample from a patient's tumor or serum exhibits a reduced prognosis such as a response to treatment or a c-met antagonist (and in some embodiments, a VEGF antagonist). In one embodiment, a decrease of more than 50% of the performance of IL8 in serum or a decrease of more than 70% (e.g., relative to the level of IL8 performance of the patient prior to treatment) is indicative of a response to treatment. Performance may be reduced relative to untreated sample or reference standard values or relative to patients treated with c-met antagonist (or treated with c-met antagonist and VEGF antagonist 143939.doc-120-201019961) The content is determined. In some embodiments, for example, an increase in mRNA or protein expression in a sample of a tumor or serum from a patient treated with a c-met antagonist (and in some embodiments, further treated with a VEGF antagonist), eg, for treatment The activity of the reaction or c-met antagonist (and in some embodiments, a VEGF antagonist). The decrease in performance can be determined relative to the untreated sample or reference standard value or relative to the performance level of the patient prior to treatment with c-met antagonist (or treatment with c-met antagonist and VEGF antagonist). In some embodiments, the FDG-PET imaging prognosis is, for example, a response to treatment or an activity of a c-met antagonist (and in some embodiments, the activity of a VEGF antagonist). Accordingly, the present invention also provides a prognostic method. Thus, the disclosed method can provide a simple, efficient, and potentially cost-effective method of obtaining information and information suitable for assessing the future course of a disease, including the selection of an appropriate therapy for treating the patient. In another aspect, the invention provides a method of assessing a patient having or suspected of having cancer, the method comprising: based on comparing a patient's biological sample with vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2) , angiopoietin-like protein 4 (Angptl4) and / or Ephrin B2 (EFNB2) and vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 in control samples Angptl4) and/or Ephrin B2 (EFNB2) is used to predict the prognosis of a patient's cancer; wherein the patient's biological sample is vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like relative to the control sample. Protein 4 (Angptl4) and / or 143939.doc •121 · 201019961

Ephrin B2(EFNB2)表現預後患者之癌症。在一些實施例 中,該方法進一步包含(a)自患者獲得生物樣品(例如在治 療之前及/或治療期間);及(b)偵測生物樣品中血管内皮細 胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血管生成素 樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現。在一些 實施例中,患者生物樣品相對於對照樣品之血管内皮細胞 生長因子A(VEGFA)、EPH受體A2(EphA2)、血管生成素樣 蛋白4(Angptl4)及/或Ephrin B2(EFNB2)表現增加預後患者 之癌症。在一些實施例中,患者生物樣品相對於對照樣品 之血管内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、 血管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)表現 降低預後患者之癌症。 在另一態樣中,本發明提供評估經歷癌症治療之患者的 方法,該方法包含:基於比較患者之生物樣品(例如企清) 中血管内皮細胞生長因子A(VEGFA)、EPH受體 A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現與在治療之前獲取之患者生物樣品中血 管内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血 管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現 來預測患者之癌症預後,其中經歷治療之患者的血清中金 管内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血 管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現 相對於治療前樣品中之表現降低預後患者之癌症。 在一些實施例中,預後癌症包含提供以下任一或多者之 143939.doc -122- 201019961 預示或預測(預後):對治療(例如用c-met拮抗劑(諸如抗c-met抗體)或c_met拮抗劑及VEGF拮抗劑治療)之反應、c-met结抗劑(諸如抗e-met抗體)或c-met括抗劑及VEGF拮抗 劑之活性、對治療(例如用c-met拮抗劑或c-met拮抗劑及 VEGF拮抗劑治療)之反應、治療(例如用c_met拮抗劑或c_ ·- met拮抗劑及VEGF拮抗劑治療)之活性、易患或經診斷患 有癌症之患者之存活持續時間、無復發存活之持續時間、 易患或經診斷患有癌症之患者無進展存活之持續時間、易 ® 患或經診斷患有癌症之患者群組之反應率、易患或經診斷 患有癌症之患者或患者群組之反應持續時間、及/或易患 或經診斷患有癌症之患者之轉移可能性。在一些實施例 中,預示或預測存活持續時間增加。在一些實施例中,預 示或預測存活持續時間減少。在一些實施例中,預示或預 測無復發存活持續時間增加。在一些實施例中,預示或預 測無復發存活持續時間減少,在一些實施例中,預示或預 籲 測反應率增加在一些實施例中,預示或預測反應率降 低。在一些實施例中’預測或預示反應持續時間增加。在 一些實施例中,預測或預示反應持續時間減少。在一些實 .施例中,預測或預示轉移可能性增加。在一些實施例中, 預測或預示轉移可能性降低。 在另μ樣中本發明提供為患有或疑似患有癌症之患 者選擇治療之方法,兮耸古、土 h人·,、* β寻方去包含.(a)基於比較患者之生 物樣〇〇中血s内皮細胞生長因子A 受體 (EphA2)血管生成素樣蛋白4(Angptl4)及/或Ephrin 143939.doc -123- 201019961 B2(EFNB2)之表現與對照樣品中血管内皮細胞生長因子 A(VEGFA)、EPH受體A2(EphA2)、血管生成素樣蛋白 4(Angptl4)及/或Ephrin B2(EFNB2)之表現來預測患者之癌 症預後,其中患者生物樣品相對於對照樣品之血管内皮細 胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血管生成素 樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)表現預後患者之 癌症,及(b)在步驟(a)之後,為患者選擇癌症治療,其中 治療之選擇係基於步驟(a)中確定之患者預後。在一些實施 例中,該等方法進一步包含(c)獲得患者生物樣品;(d)偵 測生物樣品中血管内皮細胞生長因子A(VEGFA)、EPH受 體A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現,其中患者生物樣品中血管内皮細胞生 長因子A(VEGFA)、EPH受體A2(EphA2)、血管生成素樣蛋 白4(Angptl4)及/或Ephrin B2(EFNB2)之表現預後癌症。在 一些實施例中,患者生物樣品相對於對照樣品之血管内皮 細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血管生成 素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)表現增加預後 患者之癌症。在一些實施例中,患者生物樣品相對於對照 樣品之血管内皮細胞生長因子A(VEGFA)、EPH受體 A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)表現降低預後患者之癌症。 在另一態樣中,本發明提供為經歷癌症治療之患者選擇 治療之方法,該等方法包含:(a)基於比較患者生物樣品 (例如血清)中血管内皮細胞生長因子A(VEGFA)、EPH受體 143939.doc -124- 201019961 A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現與治療之前獲取之患者生物樣品中血管 内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血管 生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現來 預測患者之癌症預後,其中經歷治療之患者的血清中血管 内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血管 生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現相 對於治療前樣品中之表現預後患者之癌症;及(b)在步驟 (a)之後,為患者選擇癌症治療,其中治療之選擇係基於步 驟(a)中確定之患者預後。在一些實施例中,該等方法進一 步包含(c)獲得患者生物樣品;(d)偵測生物樣品中血管内 皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血管生 成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)之表現,其 中患者生物樣品中血管内皮細胞生長因子A(VEGFA)、 EPH受體A2(EphA2)、血管生成素樣蛋白4(Angptl4)及/或 Ephrin B2(EFNB2)之表現預後癌症。在一些實施例中,患 者生物樣品相對於對照樣品之血管内皮細胞生長因子 A(VEGFA)、EPH受體A2(EphA2)、血管生成素樣蛋白 4(Angptl4)及/或Ephrin B2(EFNB2)表現增加預後患者之癌 症。在一些實施例中,患者生物樣品相對於對照樣品之血 管内皮細胞生長因子A(VEGFA)、EPH受體A2(EphA2)、血 管生成素樣蛋白4(Angptl4)及/或Ephrin B2(EFNB2)表現降 低預後患者之癌症。 本文中之生物樣品可為固定樣品(例如福馬林固定之石 143939.doc -125- 201019961 蠟包埋(FFPE)樣品)或冷凍樣品。 測定mRNA或蛋白質表現之多種方法包括(但不限於)確 定基因表現譜、聚合酶鏈反應(PCR)(包括定量即時 PCR(qRT-PCR))、微陣列分析、基因表現連續分析(serial analysis of gene expression,SAGE)、MassARRAY、藉由 大規模平行信號測序(Massively Parallel Signature Sequencing,MPSS)進行基因表現分析、蛋白質組研究、 免疫組織化學(IHC)等。較佳定量mRNA。此類mRNA分析 較佳使用聚合酶鏈反應(PCR)技術或藉由微陣列分析進 行。當採用PCR時,PCR之較佳形式為定量即時PCR(qRT-PCR)。在一實施例中,若一或多個以上所說明之基因之表 現例如與相同腫瘤類型之其他樣品相比位於中值或中值以 上,則視為陽性表現。中值表現含量之確定可基本上與基 因表現之量測同時進行,或可能先前已經確定。 多篇公開期刊文章(例如:Godfrey等人,J. Molec. Diagnostics 2:84-91 (2000) ; Specht等人,Am. J. Pathol. 158:419-29 (2001))中給出使用固定的石蠟包埋組織作為 RNA來源來確定基因表現譜之代表性方案之步驟,包括 mRNA分離、純化、引子擴展及擴增。簡言之,代表性方 法以切割石蠟包埋之腫瘤組織樣品之約10微克厚的切片開 始。隨後提取RNA,且移除蛋白質及DNA。分析RNA濃度 後,必要時可包括RNA修復及/或擴增步驟,且使用基因 特異性啟動子逆轉錄RNA,接著進行PCR。最後,分析資 料以基於所檢查之腫瘤樣品中所確定之特徵基因表現模式 143939.doc -126- 201019961 來確定可用於患者之最佳治療選項。 基因或蛋白質表現之偵測可直接或間接確定。 可確定癌症中c-met及/或EGFR之表現或擴增(直接或間 接)。此可利用各種診斷/預後檢定。在一實施例中,可利 用IHC分析c-met及/或EGFR之過度表現。可使來自腫瘤生 檢之石蠟包埋的組織切片經受IHC檢定,且匹配如下之c-met及/或EGFR蛋白質染色強度標準: 分數〇 :未觀察到染色或在少於10%之腫瘤細胞中觀察 到膜染色。 分數1+ :在超過10%之腫瘤細胞中偵測到模糊/勉強可覺 察之膜染色。細胞上僅有部分細胞膜染色。 分數2+ :在超過10%之腫瘤細胞中觀察到弱至中等完全 膜染色。 分數3+ :在超過10%之腫瘤細胞中觀察到中等至強完全 膜染色。 在一些實施例中,c-met及/或EGFR過度表現之評定分數 為0或1 +之腫瘤可表徵為不過度表現c-met及/或EGFR,而 分數為2 +或3 +之腫瘤可表徵為過度表現c-met及/或EGFR。 在一些實施例中,過度表現c-met及/或EGFR之腫瘤可根 據對應於每個細胞所表現的c-met及/或EGFR分子之複本數 目的免疫組織化學分數來分級,且可以生物化學方式進行 測定: 0=0-10,000個複本/細胞, 1+=至少約200,000個複本/細胞, 143939.doc -127- 201019961 2+=至少約500,000個複本/細胞, 3+=至少約2,000,000個複本/細胞。 或者或另外,可對福馬林固定之石蠟包埋踵瘤組,織進行 FISH檢定以確定腫瘤中c-met及/或EGFR擴增之程度(若存 在)。 ·- c-met或EGFR活化可直接(例如藉由碌酸基-ELISA測試 ·. 或偵測磷酸化受體之其他方式)或間接(例如藉由债測活化 之下游信號傳導路徑組件、偵測受體二聚體(例如同二聚 體、異二聚體)、偵測基因表現概況及其類似方式)測定。 φ 類似地,c-met或EGFR組成性活化或非配位體依賴性 EGFR或c-met之存在可直接或間接(例如藉由偵測與組成性 活性相關之受體突變、藉由偵測與組成性活性相關之受體 擴增及其類似方式)偵測。 偵測核酸突變之方法在此項技術中為熟知的。雖然不必 需,但時常擴增樣品中之靶核酸以提供期望量之物質來確 定是否存在突變。擴增技術在此項技術中為熟知的。舉例 而S,擴增產物可能涵蓋或可能不涵蓋編碼相關蛋白質之 〇 所有核酸序列,只要擴增產物包含疑似具有突變之特定胺 基酸/核酸序列位置即可。 在實例中,冑變之存在可藉由使來自樣S之核酸與能 夠與編碼突變核酸之核酸特異性雜交且侦測該雜交的核酸·. 探針接觸來確定。在一實施例中,探針係例如以放射性同 位素(H、p、p等)、發光劑(若丹明、螢光等)或顯色劑 作可偵測物標記。在—些實施例中,探針為反義募聚物, 143939.doc -128- 201019961 例如PNA、N-嗎啉基-胺基磷酸酯、LNA或2,-烷氧基烷氧 基°探針可具有約8個核苷酸至約100個核苷酸,或約1〇至 約75個核苷酸,或約15至約5〇個核苷酸,或約2〇至約3〇個 核普酸。在另一態樣中,本發明之核酸探針提供於鑑別樣 品中之c-met突變之套組中’該套組包含與編碼c_met之核 酸中之突變位點或在該突變位點附近特異性雜交的募核苦 酸。該套組可進一步包含基於使用該套組進行雜交測試之 結果’用c-met拮抗劑治療患含有c-met突變之肢瘤之患者 的說明書。 亦可藉由比較擴增核酸之電泳遷移率與編碼野生型心 met之相應核酸之電泳遷移率來偵測突變。遷移率之差異 指示擴增核酸序列中突變之存在。電泳遷移率可利用任何 適當的分子分離技術(例如在聚丙烯醯膠凝膠上)測定。 亦可使用酶促突變偵測(EMD)來分析核酸,從而偵測突 變(Del Tito專人,Clinical Chemistry 44:731-739, 1998)。 EMD使用噬菌體解離酶丁4核酸内切酶VII,其沿雙鍵DNA 掃描直至其偵測到由核酸變異(諸如點突變、插入及缺失) 導致之鹼基對錯配所引起之結構變形並使其裂解。利用例 如凝膠電泳偵測到兩個由解離酶裂解所形成之短片段指示 存在突變。EMD方法之優點在於由單一方案來鑑別直接自 擴增反應檢定之點突變、缺失及插入,從而消除樣品純化 之必要性,縮短雜交時間及增加信雜比。可檢定含有最多 20倍過量之正常核酸與最多4 kb大小之片段的混合樣品。 然而,EMD掃描無法鑑別突變陽性樣品中發生之特定鹼基 143939.doc -129· 201019961 變化,因此必要時時常需要其他測序程序來鑑別特定突 變。如美國專利第5,869,245號中所表明,可與解離酶T4核 酸内切酶VII類似地使用CEL I酶。 另一簡單的偵測突變之套組為與血色病(Haemochromatosis) StripAssay™ (Viennalabs » http://www.bamburghmarrsh.com/pdf74220.pdf) 類似之反向雜交測試條,其用於债測引起企色病之HF E、 TFR2及FPN1基因中之多個突變。此類檢定係基於PCR擴 增後之序列特異性雜交。對於單一突變檢定,可應用基於 微板之偵測系統,而對於多突變檢定,可使用測試條作為 「巨陣列(macro-array)」。套組可包括用於樣品製備、擴增 及突變偵測之即用型試劑。複合擴增方案提供簡便性且允 許測試極有限體積之樣品。使用直接StripAssay格式,無 需昂貴設備即可在不足5小時内完成20個或20個以上突變 之測試。自樣品分離DNA,且一般在單一(「複合」)擴增 反應中活體外擴增靶核酸(例如利用PCR)且標記生物素。 擴增產物隨後與固定在固體支撐物(諸如測試條)上之募核 苷酸探針(野生型及突變型特異性)選擇性雜交,其中探針 呈平行線或條帶形式固定於固體支撐物上。使用抗生蛋白 鏈菌素-鹼性磷酸酯酶及有色受質來偵測結合之生物素標 記擴增子。此類檢定可偵測本發明之所有突變或其任何子 集。關於特定突變型探針條帶,可能為以下三種信號傳導 圖案之一:⑴僅野生型探針之條帶,指示正常核酸序列, (ii)野生型與突變型探針之條帶,指示雜合基因型,及(iii) 僅突變型探針之條帶,指示純合突變基因型。因此,在一 143939.doc -130- 201019961 態樣中,本發明提供偵測本發明突變之方法,該方法包含 自樣品分離及/或擴增靶(:_〇1以核酸序列,以致擴增產物包 含配位體;使擴增產物與探針接觸,該探針包含配位體之 可偵測結合搭配物且能夠與本發明突變特異性雜交;及隨 後偵測該探針與該擴增產物之雜交。在一實施例中,配位 體為生物素’且結合搭配物包含抗生物素蛋白或抗生蛋白 鏈菌素。在一實施例中,結合搭配物包含可用有色受質偵 測之抗生蛋白鏈菌素_鹼性。在一實施例中,探針固定在 例如測試條上,其中與不同突變互補之探針彼此分開。或 者,用放射性同位素標記擴增核酸,在此情況下,探針不 需包含可偵測標記。 野生型基因之變異涵蓋所有突變形式,諸如插入、逆 位、缺失及/或點突變》在一實施例中,突變為體細胞突 變。體細胞突變為僅發生在某些組織(例如腫瘤組織)中之 突變且不在生殖系中遺傳。生殖系突變可見於任何身體組 織中。 包含靶核酸之樣品可由此項技術熟知且適合腫瘤之特定 類型及位置之方法獲得。冑常使用組織生檢來獲得腫瘤組 織之代表性碎片。或者,可呈已知或認為含有相關腫瘤細 胞之組織/流體形式間接獲得腫瘤細胞。舉例而言,可利 用切除術、支氣管鏡檢查、細針抽吸(flne needle aspimi〇n)、 支氣管刷取(bronchial brushing)或自唾液、胸膜液或血液 中獲得肺癌病變之樣品。可自腫瘤或其他身體樣品(諸如 尿、唾液或血清)偵測突變基因或基因產物。以上所討論 143939.doc -131 - 201019961 之偵測腫瘤樣品中之突變靶基因或基因產物的相同技術可 用於其他身體樣品。癌細胞自腫瘤脫落且出現在該等身體 樣品中。藉由篩選該等身體樣品,可實現諸如癌症之疾病 之簡單早期診斷。另外,藉由測試該等身體樣品之突變靶 基因或基因產物可更容易地監測療法之進程。 此項技術中已知富集腫瘤細胞之組織製劑之方法。舉例 而S ’可自石蠟或冷凍切片分離組織。亦可利用流式細胞 術或雷射捕捉顯微解剖(laser capture microdissection)來分 離癌細胞與正常細胞。此項技術中熟知此等及其他分離腫 瘤細胞與正常細胞之技術。若腫瘤組織受到正常細胞高度 污染,則雖然已知最小化污染及/或偽陽性/陰性結果之技 術(其中一些描述在下文中)’但突變偵測仍可能更困難。 舉例而言’亦可評估樣品中已知與相關腫瘤細胞(但非相 應正常細胞)相關之生物指標(包括突變)的存在,或反之亦 然。 靶核酸中點突變之偵測可使用此項技術熟知之技術藉由 分子選殖靶核酸且測定核酸序列來實現。或者,可使用擴 增技術(諸如聚合酶鏈反應(PCR))直接由來自腫瘤組織之 基因組DNA製劑擴增靶核酸序列。隨後可測定擴增序列之 核酸序列,且自其中鑑別突變。擴增技術在此項技術中為 熟知的’例如Saiki等人,Science 239:487,1988;美國專 利第4,683,203號及第4,683,195號中所述之聚合酶鍵反應。 應注意,適當引子之設計及選擇為此項技術中充分確立 之技術。 143939.doc -132- 201019961 亦可使用此項技術中已知之連接酶鏈反應來擴增靶核酸 序列。參見例如Wu等人,Genomics,第4卷,第560-569頁 (1989)。另外,亦可使用稱為等位基因特異性PCR之技 術。參見例如 Ruano 及 Kidd, Nucleic Acids Research,第 17 卷,第8392頁,1989。根據此技術,使用在3'端與特定靶 核酸突變雜交之引子。若不存在特定突變,則無法觀察到 擴增產物。如歐洲專利申請公開案第0332435號及Newton 等人,Nucleic Acids Research,第 17卷,第 7 頁,1989 中 所揭示,亦可使用擴增阻礙突變系統(Amplification Refractory Mutation System,ARMS)。亦可藉由選殖、測 序及擴增來偵測基因之插入及缺失。另外,可使用該基因 或周圍標記基因之限制片段長度多態性(restriction fragment length polymorphism,RFLP)探針來評分多態片 段中之等位基因變異、或插入。亦可使用單鏈構形多態性 (single stranded conformation polymorphism,SSCP)分析 來偵測等位基因之鹼基變化變異體。參見例如Orita等人, Proc. Natl. Acad· Sci. USA 第 86 卷,第 2766-2770 頁, 1989 ;及Genomics,第 5卷,第 874-879 頁,1989。亦可使 用此項技術中已知之其他偵測插入及缺失之技術。 亦可基於基因之野生型表現產物之變異來偵測野生型基 因之變異。該等表現產物包括mRNA與蛋白質產物。可藉 由擴增且測序mRNA或經由分子選殖由mRNA產生之cDNA 來偵測點突變。可使用此項技術中熟知之DNA測序技術來 測定選殖cDNA之序列。亦可經由聚合酶鏈反應(PCR)測定 143939.doc •133- 201019961 cDNA之序列。 錯配為非100%互補之雜交核酸雙鏈體。總互補性缺乏 可歸因於缺失、插入、逆位、取代或移碼突變。可使用錯 配偵測來偵測靶核酸中之點突變。雖然此等技術之敏感性 可能小於測序技術,但其較容易對大量組織樣品進行。錯 配裂解技術之實例為核糖核酸酶(RNase)保護法,該方法 詳細描述於 Winter 等人,Proc. Natl. Acad. Sci. USA,第 82 卷,第 7575 頁,1985 ;及Meyers等人,Science,第 230卷, 第1242頁,1985中。舉例而言,本發明方法可包括使用與 人類野生型靶核酸互補之經標記核糖核酸探針 (riboprobe)。使源自組織樣品之核糖核酸探針及乾核酸黏 接(雜交)在一起,接著用能夠偵測雙鏈體RNA結構中之某 些錯配之核糖核酸酶A消化。若用核糖核酸酶A偵測錯 配,則其在錯配位點處裂解。因此,當在電泳凝膠基質上 分離黏接之RNA製劑時,若已偵測到錯配且經核糖核酸酶 A裂解,則將可見RNA產物小於核糖核酸探針之全長雙鏈 體RNA、及mRNA或DNA。核糖核酸探針不需為靶核酸 mRN A或基因的全長,而可為靶核酸之一部分,只要其涵 蓋疑經突變之位置即可。若核糖核酸探針僅包含靶核酸 mRNA或基因之區段,則必要時可能期望使用若干此等探 針來篩選整個靶核酸序列之錯配。 可以類似方式使用DNA探針來偵測錯配,例如經由酶促 或化學裂解。參見例如Cotton等人,Proc. Natl. Acad. Sci· USA,第 85 卷,4397,1988 ;及 Shenk 等人,Proc. Natl. 143939.doc •134- 201019961Ephrin B2 (EFNB2) represents cancer in prognosis patients. In some embodiments, the method further comprises (a) obtaining a biological sample from the patient (eg, prior to treatment and/or during treatment); and (b) detecting vascular endothelial growth factor A (VEGFA), EPH in the biological sample Expression of receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2). In some embodiments, the patient biological sample exhibits vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and/or Ephrin B2 (EFNB2) relative to the control sample. Increase the cancer of patients with prognosis. In some embodiments, the patient biological sample exhibits vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and/or Ephrin B2 (EFNB2) relative to the control sample. Reduce the cancer of patients with prognosis. In another aspect, the invention provides a method of assessing a patient undergoing cancer therapy, the method comprising: based on comparing a patient's biological sample (eg, Qiqing) with vascular endothelial growth factor A (VEGFA), EPH receptor A2 ( EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2), and vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), in patient biological samples obtained prior to treatment, The expression of angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) is used to predict the prognosis of cancer in patients. The serum of patients undergoing treatment includes vascular endothelial growth factor A (VEGFA) and EPH receptor A2 (EphA2). , the performance of angiopoietin-like protein 4 (Angptl4) and / or Ephrin B2 (EFNB2) relative to the performance of the pre-treatment sample to reduce cancer in prognosis patients. In some embodiments, the prognostic cancer comprises 143939.doc-122-201019961 predicting or predicting (prognosis) any one or more of the following: for treatment (eg, with a c-met antagonist (such as an anti-c-met antibody) or Reaction of c_met antagonist and VEGF antagonist therapy, activity of c-met antagonist (such as anti-e-met antibody) or c-met antagonist and VEGF antagonist, for treatment (eg with c-met antagonist Or the response of a c-met antagonist and a VEGF antagonist treatment, treatment (eg, treatment with a c-met antagonist or c_·-met antagonist and a VEGF antagonist), survival of a patient susceptible to or diagnosed with cancer Duration, duration of recurrence-free survival, duration of progression-free survival in patients who are predisposed to or diagnosed with cancer, response rate in patients with or susceptible to cancer, predisposition, or diagnosis The duration of response to a patient or group of patients with cancer, and/or the likelihood of metastasis in a patient susceptible to or diagnosed with cancer. In some embodiments, the predicted or predicted survival duration is increased. In some embodiments, the predicted or predicted survival duration is reduced. In some embodiments, an indication of an increase in the duration of recurrence-free survival is predicted or predicted. In some embodiments, the predictive or predictive reduction in the duration of recurrence-free survival, in some embodiments, the predictive or pre-announced increase in response rate, in some embodiments, predicts or predicts a decrease in response rate. In some embodiments, the predicted or predicted duration of the reaction is increased. In some embodiments, the predicted or predicted duration of the reaction is reduced. In some embodiments, the prediction or predictive increase in transfer probability is increased. In some embodiments, the prediction or predictive transfer likelihood is reduced. In another example, the present invention provides a method for selecting a treatment for a patient suffering from or suspected of having cancer, such as squatting, earthy, and sputum. (a) based on comparing biological samples of patients. Expression of vascular endothelial growth factor A receptor (EphA2) angiopoietin-like protein 4 (Angptl4) and/or Ephrin 143939.doc-123-201019961 B2 (EFNB2) and vascular endothelial growth factor A in control samples VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) are used to predict the prognosis of a patient's cancer, wherein the patient's biological sample grows relative to the control sample. Factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) exhibit cancer in prognosis patients, and (b) after step (a), for patients Cancer therapy is selected wherein the choice of treatment is based on the prognosis of the patient as determined in step (a). In some embodiments, the methods further comprise (c) obtaining a patient biological sample; (d) detecting vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein in the biological sample 4 (Angptl4) and/or Ephrin B2 (EFNB2), in which vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) is a prognostic cancer. In some embodiments, the patient biological sample exhibits vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and/or Ephrin B2 (EFNB2) relative to the control sample. Increase the cancer of patients with prognosis. In some embodiments, the patient biological sample exhibits vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and/or Ephrin B2 (EFNB2) relative to the control sample. Reduce the cancer of patients with prognosis. In another aspect, the invention provides methods of selecting a treatment for a patient undergoing cancer treatment, the methods comprising: (a) comparing vascular endothelial growth factor A (VEGFA), EPH in a patient biological sample (eg, serum) Receptor 143939.doc -124- 201019961 A2 (EphA2), Angiopoietin-like Protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) and vascular endothelial growth factor A (VEGFA) in patient biological samples obtained prior to treatment , the expression of EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and / or Ephrin B2 (EFNB2) to predict the prognosis of patients with cancer, wherein the vascular endothelial growth factor A in the serum of patients undergoing treatment (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) are expressed relative to cancer in patients with prognosis in pre-treatment samples; and (b) in steps (a) Thereafter, the patient is selected for cancer treatment, wherein the choice of treatment is based on the patient's prognosis as determined in step (a). In some embodiments, the methods further comprise (c) obtaining a patient biological sample; (d) detecting vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein in the biological sample 4 (Angptl4) and/or Ephrin B2 (EFNB2), in which the patient's biological sample is vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and/or Ephrin B2 (EFNB2) is a prognostic cancer. In some embodiments, the patient biological sample exhibits vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and/or Ephrin B2 (EFNB2) relative to the control sample. Increase the cancer of patients with prognosis. In some embodiments, the patient biological sample exhibits vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4), and/or Ephrin B2 (EFNB2) relative to the control sample. Reduce the cancer of patients with prognosis. The biological sample herein may be a fixed sample (e.g., a formalin fixed stone 143939.doc-125-201019961 wax-embedded (FFPE) sample) or a frozen sample. Various methods for determining mRNA or protein expression include, but are not limited to, determining gene expression profiles, polymerase chain reaction (PCR) (including quantitative real-time PCR (qRT-PCR)), microarray analysis, and serial analysis of Gene expression, SAGE), MassARRAY, gene expression analysis, proteomic research, immunohistochemistry (IHC), etc. by Massively Parallel Signature Sequencing (MPSS). Preferably, the mRNA is quantified. Such mRNA analysis is preferably carried out using polymerase chain reaction (PCR) techniques or by microarray analysis. When PCR is employed, the preferred form of PCR is quantitative real-time PCR (qRT-PCR). In one embodiment, a performance of one or more of the indicated genes is considered positive if, for example, is above or below the median of other samples of the same tumor type. The determination of the median performance level can be performed substantially simultaneously with the measurement of the gene performance, or may have been previously determined. A number of published journal articles (eg, Godfrey et al., J. Molec. Diagnostics 2: 84-91 (2000); Specht et al., Am. J. Pathol. 158: 419-29 (2001)) The paraffin-embedded tissue serves as a source of RNA to determine the steps of a representative protocol for gene expression profiling, including mRNA isolation, purification, primer extension, and amplification. Briefly, a representative method begins with the cutting of about 10 micrograms of a slice of a paraffin-embedded tumor tissue sample. The RNA is then extracted and the protein and DNA are removed. After analyzing the RNA concentration, an RNA repair and/or amplification step may be included as necessary, and the RNA is reverse transcribed using a gene-specific promoter, followed by PCR. Finally, the data was analyzed to determine the optimal treatment options available to the patient based on the characteristic gene expression pattern identified in the tumor sample examined 143939.doc-126-201019961. Detection of gene or protein expression can be determined directly or indirectly. The expression or amplification (direct or indirect) of c-met and/or EGFR in cancer can be determined. This can take advantage of various diagnostic/prognostic assays. In one embodiment, IHC can be used to analyze excessive performance of c-met and/or EGFR. Paraffin-embedded tissue sections from tumor biopsies can be subjected to IHC assays and matched to the following c-met and/or EGFR protein staining intensity criteria: Fraction 〇: No staining or less than 10% of tumor cells were observed Membrane staining was observed. Score 1+: Blurring/reluctantly detectable membrane staining was detected in more than 10% of tumor cells. Only part of the cell membrane was stained on the cells. Fraction 2+: Weak to moderate complete membrane staining was observed in more than 10% of tumor cells. Score 3+: Moderate to strong complete membrane staining was observed in more than 10% of tumor cells. In some embodiments, a tumor with a score of 0 or 1 + with an over-expression of c-met and/or EGFR may be characterized as not overexpressing c-met and/or EGFR, and a tumor having a score of 2 + or 3 + may Characterized by overexpression of c-met and/or EGFR. In some embodiments, tumors that overexpress c-met and/or EGFR can be ranked according to an immunohistochemical fraction corresponding to the number of copies of c-met and/or EGFR molecules exhibited by each cell, and can be biochemically Ways to determine: 0 = 0 - 10,000 replicates / cell, 1 + = at least about 200,000 copies / cell, 143939.doc -127 - 201019961 2+ = at least about 500,000 copies / cell, 3 + = at least about 2,000,000 Replica / cell. Alternatively or additionally, a fumarin-fixed paraffin-embedded tumor group can be subjected to FISH assay to determine the extent of c-met and/or EGFR expansion in the tumor, if any. ·- c-met or EGFR activation can be directly (for example by sulphate-ELISA test or other means of detecting phosphorylated receptors) or indirectly (eg downstream signal transduction pathway components activated by debt testing, Detective Determination of receptor dimers (eg, homodimers, heterodimers, detection of gene expression profiles, and the like). Similarly, the presence of c-met or EGFR constitutively activated or non-ligand-dependent EGFR or c-met can be directly or indirectly (eg, by detecting receptor mutations associated with constitutive activity, by detecting Receptor amplification associated with constitutive activity and similar methods) detection. Methods for detecting nucleic acid mutations are well known in the art. Although not required, the target nucleic acid in the sample is often amplified to provide the desired amount of material to determine if a mutation is present. Amplification techniques are well known in the art. For example, S, the amplification product may or may not encompass all nucleic acid sequences encoding the relevant protein, as long as the amplification product contains a specific amino acid/nucleic acid sequence position suspected of having a mutation. In an example, the presence of a mutated can be determined by contacting a nucleic acid from S-like S with a nucleic acid that is capable of specifically hybridizing to a nucleic acid encoding a mutated nucleic acid and detecting the hybridization. In one embodiment, the probe is labeled, for example, with a radioisotope (H, p, p, etc.), a luminescent agent (rhodamine, fluorescent, etc.) or a color developing agent. In some embodiments, the probe is an antisense polymer, 143939.doc -128-201019961 such as PNA, N-morpholinyl-amino phosphate, LNA or 2,-alkoxy alkoxy The needle can have from about 8 nucleotides to about 100 nucleotides, or from about 1 to about 75 nucleotides, or from about 15 to about 5 nucleotides, or from about 2 to about 3 inches. Nuclear acid. In another aspect, the nucleic acid probe of the present invention is provided in a kit for identifying a c-met mutation in a sample. 'The kit comprises or is specific to a mutation site in a nucleic acid encoding c_met Sexual hybridization raises bitter acid. The kit may further comprise instructions for treating a patient suffering from a limb tumor containing a c-met mutation with a c-met antagonist based on the results of the hybridization test using the kit. Mutations can also be detected by comparing the electrophoretic mobility of the amplified nucleic acid to the electrophoretic mobility of the corresponding nucleic acid encoding the wild type cardiac met. The difference in mobility indicates the presence of a mutation in the amplified nucleic acid sequence. Electrophoretic mobility can be measured using any suitable molecular separation technique (e.g., on a silicone gel gel). Enzyme-induced mutation detection (EMD) can also be used to detect nucleic acids to detect mutations (Del Tito, Clinical Chemistry 44: 731-739, 1998). EMD uses the phage resolving enzyme D4 endonuclease VII, which scans along double-stranded DNA until it detects structural deformation caused by base pair mismatches caused by nucleic acid variations (such as point mutations, insertions and deletions) and It cleaves. The use of, for example, gel electrophoresis to detect two short fragments formed by cleavage of the dissociation enzyme indicates the presence of a mutation. The advantage of the EMD method is that a single protocol is used to identify point mutations, deletions, and insertions directly from the amplification reaction assay, thereby eliminating the need for sample purification, shortening the hybridization time, and increasing the signal-to-noise ratio. A mixed sample containing up to a 20-fold excess of normal nucleic acid and a fragment of up to 4 kb in size can be assayed. However, EMD scans are unable to identify specific base 143939.doc -129· 201019961 changes that occur in mutation-positive samples, so other sequencing procedures are often needed to identify specific mutations when necessary. The CEL I enzyme can be used similarly to the dissociation enzyme T4 endonuclease VII as indicated in U.S. Patent No. 5,869,245. Another simple cluster for detecting mutations is a reverse hybridization test strip similar to Haemochromatosis StripAssayTM (Viennalabs » http://www.bamburghmarrsh.com/pdf74220.pdf), which is used for debt testing. Multiple mutations in the HF E, TFR2 and FPN1 genes of the disease. Such assays are based on sequence-specific hybridization following PCR amplification. For single mutation assays, a microplate-based detection system can be applied, and for multiple mutation assays, a test strip can be used as a "macro-array." Kits can include ready-to-use reagents for sample preparation, amplification, and mutation detection. The complex amplification protocol provides simplicity and allows for the testing of very limited volumes of samples. Using the direct StripAssay format, 20 or more mutations can be tested in less than 5 hours without the need for expensive equipment. DNA is isolated from the sample, and the target nucleic acid is typically amplified in vitro (e.g., using PCR) and labeled with biotin in a single ("complex") amplification reaction. The amplified product is then selectively hybridized to a nucleotide probe (wild type and mutant specific) immobilized on a solid support such as a test strip, wherein the probe is immobilized in solid parallel or strip form on the solid support On the object. Biotin-labeled amplicons were detected using streptavidin-alkaline phosphatase and colored receptors. Such assays can detect all mutations or any subset thereof of the invention. Regarding a particular mutant probe band, it may be one of the following three signaling patterns: (1) a band of wild-type probes only, indicating a normal nucleic acid sequence, (ii) a band of wild-type and mutant probes, indicating miscellaneous A genomic form, and (iii) a band of only mutant probes indicating a homozygous mutant genotype. Thus, in a 143939.doc-130-201019961 aspect, the invention provides a method of detecting a mutation of the invention, the method comprising: isolating and/or amplifying a target from a sample (: 〇1 in a nucleic acid sequence such that amplification The product comprises a ligand; the amplification product is contacted with a probe comprising a detectable binding partner of the ligand and capable of specifically hybridizing to the mutation of the invention; and subsequently detecting the probe and the amplification Hybridization of the product. In one embodiment, the ligand is biotin' and the binding partner comprises avidin or streptavidin. In one embodiment, the binding partner comprises a detectable color acceptor Streptavidin_basic. In one embodiment, the probe is immobilized, for example, on a test strip, wherein probes complementary to different mutations are separated from each other. Alternatively, the nucleic acid is amplified by radioisotope labeling, in which case The probe need not contain a detectable marker. The variation of the wild-type gene covers all mutant forms, such as insertions, inversions, deletions, and/or point mutations. In one embodiment, the mutation is a somatic mutation. Somatic mutation Mutations that occur only in certain tissues (eg, tumor tissue) and are not inherited in the germline. Germline mutations can be found in any body tissue. Samples containing target nucleic acids are well known in the art and are suitable for the particular type and location of the tumor. Methods obtained. Tissue biopsy is often used to obtain representative fragments of tumor tissue. Alternatively, tumor cells can be obtained indirectly from tissue/fluid forms known or believed to contain relevant tumor cells. For example, resection, bronchi can be used Mirror, fine needle aspiration (flne needle aspimi〇n), bronchial brushing or a sample of lung cancer lesions obtained from saliva, pleural fluid or blood. Can be from a tumor or other body sample (such as urine, saliva or Serum) detection of a mutated gene or gene product. The same technique for detecting a mutant target gene or gene product in a tumor sample as discussed above in 143939.doc-131 - 201019961 can be applied to other body samples. In such body samples, diseases such as cancer can be achieved by screening such body samples Simple early diagnosis. In addition, the progress of the therapy can be more easily monitored by testing mutant target genes or gene products of such body samples. Methods for enriching tissue preparations of tumor cells are known in the art. Tissues are isolated from paraffin or cryosections. Flow cytometry or laser capture microdissection can also be used to isolate cancer cells from normal cells. These and other isolated tumor cells and normal cells are well known in the art. Techniques. If tumor tissue is highly contaminated by normal cells, although techniques for minimizing contamination and/or false positive/negative results are known (some of which are described below), mutation detection may still be more difficult. For example, the presence of biological indicators (including mutations) known to be associated with associated tumor cells (but not corresponding normal cells) in the sample may also be assessed, or vice versa. Detection of point mutations in the target nucleic acid can be accomplished by molecularly cloning the target nucleic acid and determining the nucleic acid sequence using techniques well known in the art. Alternatively, the target nucleic acid sequence can be directly amplified from a genomic DNA preparation from tumor tissue using an amplification technique such as polymerase chain reaction (PCR). The nucleic acid sequence of the amplified sequence can then be determined and the mutation identified therefrom. Amplification techniques are well known in the art, such as the polymerase linkage reactions described in Saiki et al., Science 239: 487, 1988; U.S. Patent Nos. 4,683,203 and 4,683,195. It should be noted that the design and selection of appropriate primers is a well-established technique in this technology. 143939.doc -132- 201019961 A ligase chain reaction known in the art can also be used to amplify a target nucleic acid sequence. See, for example, Wu et al., Genomics, Vol. 4, pp. 560-569 (1989). Alternatively, a technique called allele-specific PCR can be used. See, for example, Ruano and Kidd, Nucleic Acids Research, Vol. 17, p. 8392, 1989. According to this technique, a primer that hybridizes to a specific target nucleic acid mutation at the 3' end is used. If no specific mutation is present, the amplification product cannot be observed. Amplification Refractory Mutation System (ARMS) can also be used as disclosed in European Patent Application Publication No. 0332435 and Newton et al., Nucleic Acids Research, Vol. 17, p. 7, 1989. Gene insertions and deletions can also be detected by colonization, sequencing and amplification. Alternatively, a restriction fragment length polymorphism (RFLP) probe of the gene or surrounding marker gene can be used to score allelic variation, or insertion, in a polymorphic fragment. Single stranded conformation polymorphism (SSCP) analysis can also be used to detect base variation variants of alleles. See, for example, Orita et al, Proc. Natl. Acad. Sci. USA, Vol. 86, pp. 2766-2770, 1989; and Genomics, Vol. 5, pp. 874-879, 1989. Other techniques for detecting insertions and deletions known in the art can also be used. Mutations in the wild-type gene can also be detected based on variations in the wild-type performance product of the gene. Such performance products include mRNA and protein products. Point mutations can be detected by amplification and sequencing of mRNA or by cDNA-derived cDNA produced from mRNA. The sequence of the cloned cDNA can be determined using DNA sequencing techniques well known in the art. The sequence of the 143939.doc • 133-201019961 cDNA can also be determined by polymerase chain reaction (PCR). Mismatches are non-100% complementary hybrid nucleic acid duplexes. Lack of total complementarity can be attributed to deletions, insertions, inversions, substitutions or frameshift mutations. Mismatch detection can be used to detect point mutations in target nucleic acids. While these techniques may be less sensitive than sequencing techniques, they are easier to perform on large numbers of tissue samples. An example of a mismatch cleavage technique is the RNase protection method, which is described in detail in Winter et al, Proc. Natl. Acad. Sci. USA, Vol. 82, p. 7575, 1985; and Meyers et al. Science, Vol. 230, p. 1242, 1985. For example, the methods of the invention can include the use of a labeled riboprobe that is complementary to a human wild-type target nucleic acid. The ribonucleic acid probe derived from the tissue sample and the dry nucleic acid are ligated (hybridized) together, followed by digestion with ribonuclease A which is capable of detecting some mismatch in the duplex RNA structure. If RNase A detects a mismatch, it cleaves at the mismatch site. Therefore, when the immobilized RNA preparation is separated on the electrophoresis gel matrix, if a mismatch has been detected and cleavage by ribonuclease A, the visible RNA product is smaller than the full-length duplex RNA of the riboprobe, and mRNA or DNA. The riboprobe is not required to be the full length of the target nucleic acid mRN A or the gene, but may be a part of the target nucleic acid as long as it covers the position of the suspected mutation. If the riboprobe contains only the target nucleic acid mRNA or a segment of the gene, it may be desirable to use several such probes to screen for mismatches throughout the target nucleic acid sequence, if desired. DNA probes can be used in a similar manner to detect mismatches, such as via enzymatic or chemical cleavage. See, for example, Cotton et al., Proc. Natl. Acad. Sci. USA, Vol. 85, 4397, 1988; and Shenk et al., Proc. Natl. 143939.doc • 134-201019961

Acad. Sci. USA,第72卷,第989卷,1975。或者,可利用 錯配雙鏈體相對於匹配雙鏈體之電泳遷移率變化來偵測錯 配。參見例如 Cariello, Human Genetics,第 42卷,第 726 頁,1988。使用核糖核酸探針或DNA探針時,可在雜交之 前擴增可含突變之靶核酸mRNA或DNA。亦可使用南方雜 ' 交來偵測靶核酸DNA之變化,尤其當變化為總重排(gr〇ss rearrangement)(諸如缺失及插入)時。 ❹ 亦可使用等位基因特異性探針來篩選已擴增之靶核酸 DNA序列。此等探針為核酸寡聚物’各含有具有已知突變 之靶核酸基因區域。舉例而言,一種寡聚物可為約3〇個核 苷酸長,對應於靶基因序列之一部分。藉由使用該等等位 基因特異性探針之組,可篩選靶核酸擴增產物來鑑別靶基 因中先别已鑑別之突變的存在。可例如在对綸過濾器 (nylon filter)上進行等位基因特異性探針與擴增靶核酸序 列之雜交》在嚴格雜交條件下與特定探針之雜交指示腫瘤 φ 組織中存在與等位基因特異性探針中相同之突變。 亦可藉由篩選相應野生型蛋白質之變異來偵測野生型靶 基因之變異。舉例而言,可使用與靶基因產物具有免疫反 • 應性之單株抗體來篩選組織,例如已知與基因產物(蛋白 • 質)之特定突變位置結合之抗體。舉例而言,所使用之抗 體可為結合缺失外顯子(例如外顯子14)或結合包含靶蛋白 之缺失部分之構形抗原決定基的抗體。同源抗原之缺乏將 指示突變。亦可使用對突變等位基因之產物具有特異性之 抗體來㈣突變基因產物。可自嗤菌趙呈現文庫鑑別抗 143939.doc •135- 201019961 體。該等免疫學檢定可以此項技術中已知之任何便利格式 進行jtb等格式包括西方墨點法、免疫組織化學檢定及 ELISA檢定。可使用㈣制變異蛋白質之方法來摘測野 生型乾基因之變異。 引子對適用於使用核酸擴增技術(諸如聚合酶鏈反應)來 測定靶核酸之核苷酸序列。單鏈DNA引子對可與靶核酸序 列内部或靶核酸序列周圍之序列黏接以引發靶序列擴增。 亦可使用等位基因特異性引子。該等引子僅與特定突變把 序列黏接,且因此在突變靶序列作為模板下將僅擴增產 物。為促進擴增序列之後續選殖,引子可具有附加於其末 端之限制酶位點序列。此項技術中熟知該等酶及位點。引 子本身可使用此項技術中熟知之技術合成。引子一般可使 用可自市面上購得之募核苷酸合成器產生。特定引子之設 計充分處於此項技術之技能内。 核酸探針適用於若干目的。其可用於與基因組dna南方 雜交及用於核糖核酸酶保護法中以偵測以上已討論之點突 變。可使用探針偵測靶核酸擴增產物。其亦可用於使用其 他技術來偵測野生型基因或mRNA之錯配。可使用酶(例如 S1核酸酶)、化學品(例如羥胺或四氧化餓及哌啶)或與完全 匹配之雜交體相比錯配雜交體之電泳遷移率變化來偵測錯 配此項技術中已知此等技術。參見Novack等人,Proc.Acad. Sci. USA, Vol. 72, vol. 989, 1975. Alternatively, mismatches can be detected by detecting the difference in electrophoretic mobility of the mismatched duplex relative to the matched duplex. See, for example, Cariello, Human Genetics, Vol. 42, p. 726, 1988. When a riboprobe or DNA probe is used, the target nucleic acid mRNA or DNA which can be mutated can be amplified prior to hybridization. Southern hybrids can also be used to detect changes in target nucleic acid DNA, especially when the change is gr〇ss rearrangement (such as deletions and insertions).等 Allele-specific probes can also be used to screen amplified target nucleic acid DNA sequences. These probes are nucleic acid oligomers each containing a target nucleic acid gene region having a known mutation. For example, an oligomer can be about 3 nucleotides long, corresponding to a portion of a target gene sequence. By using a set of such allele-specific probes, the target nucleic acid amplification product can be screened to identify the presence of a previously identified mutation in the target gene. For example, hybridization of an allele-specific probe to an amplified target nucleic acid sequence on a nylon filter will hybridize to a specific probe under stringent hybridization conditions to indicate the presence and allele in the tumor φ tissue. The same mutation in the specific probe. Variations in wild-type target genes can also be detected by screening for variations in the corresponding wild-type proteins. For example, monoclonal antibodies that are immunoreactive with a target gene product can be used to screen for tissue, such as antibodies known to bind to specific mutational positions of a gene product (protein). For example, the antibody used can be an antibody that binds to a deletion exon (e.g., exon 14) or binds to a conformational epitope comprising a deletion portion of the target protein. A lack of a homologous antigen will indicate a mutation. An antibody specific for the product of the mutant allele can also be used to (4) mutate the gene product. It can be used to identify the anti-bacteria Zhao Zhao 143939.doc • 135- 201019961 body. Such immunological assays can be performed in any convenient format known in the art, including jtb format, Western blotting, immunohistochemistry, and ELISA assays. The variation of the wild type dry gene can be extracted by the method of (4) variant protein. A primer pair is suitable for use in nucleic acid amplification techniques, such as polymerase chain reaction, to determine the nucleotide sequence of a target nucleic acid. A single-stranded DNA primer pair can be affixed to a sequence within the target nucleic acid sequence or around the target nucleic acid sequence to initiate amplification of the target sequence. Allele-specific primers can also be used. These primers only bind the sequence to a particular mutation, and thus will only amplify the product under the mutated target sequence as a template. To facilitate subsequent selection of the amplified sequence, the primer may have a restriction enzyme site sequence appended to its terminus. Such enzymes and sites are well known in the art. The primers themselves can be synthesized using techniques well known in the art. Primers are typically produced using commercially available nucleotide synthesizers. The design of a particular primer is well within the skill of this technology. Nucleic acid probes are suitable for several purposes. It can be used to hybridize to the southern part of the genomic DNA and to be used in the ribonuclease protection method to detect the point mutations discussed above. A probe can be used to detect a target nucleic acid amplification product. It can also be used to detect mismatches in wild-type genes or mRNA using other techniques. The mismatch can be detected using an enzyme (eg, S1 nuclease), a chemical (eg, hydroxylamine or tetrazolium and piperidine) or a mismatched hybrid electrophoretic shift change compared to a perfectly matched hybrid. These techniques are known. See Novack et al., Proc.

Natl. Acad. Sci. USA,第 83卷,第 586 頁,1986。探針一 般與激酶域外部之序列互補。可使用整個核酸探針組來構 成偵測靶核酸中之突變之套組。該套組允許與相關靶序列 I43939.doc •136· 201019961 之大區域雜交。探針可彼此重疊或鄰接。 若使用核糖核酸探針來偵測mRNA之錯配,則其一般與 靶基因之mRNA互補。因為RNA探針與有義鏈互補,所以 其並不編碼相應基因產物,因此其為反義探針。核糖核酸 探針一般將用放射性、比色或螢光物質標記,此可由此項 技術中已知之任何方法實現。若使用核糖核酸探針來偵測 DNA之錯配,則其可具有極性有義或反義。類似地,亦可 使用DNA探針來偵測錯配。 在一些情況下,癌症過度表現或不過度表現c-met受體 及/或EGFR。受體過度表現可在診斷或預後檢定中藉由評 估細胞表面上所存在之受體蛋白的含量增加來確定(例如 經由免疫組織化學檢定;IHC)。或者或另外,可例如經由 螢光原位雜交(FISH ;參見1998年10月公開之WO 98/45479)、 南方墨點法(southern blotting)或聚合酶鏈反應(PCR)技術 (諸如即時定量PCR(RT-PCR))來量測細胞中編碼受體之核 酸之含量。除上述檢定外,熟練人員亦可利用各種活體内 檢定。舉例而言,可使患者體内之細胞曝露於視情況標記 可偵測標記(例如放射性同位素)之抗體,且可藉由例如外 部掃描放射性或分析自先前曝露於抗體之患者獲取之生檢 來評估抗體與患者細胞的結合。 化學治療劑 本發明之組合療法可進一步包含一或多種化學治療劑。 組合投藥包括使用分開的調配物或單個醫藥調配物進行共 投藥或並行投藥,及以任一順序進行連續投藥,其中較佳 143939.doc -137- 201019961 存在-段時期’此時兩種(或所有)活性劑同時發揮其生物 活性。 若投與化學治療劑,則投與劑量通常為其已知劑量,或 由於藥物之組合作用或可能因投與抗代謝物化學治療劑所 致之負面㈣㈣視情科低。料化學治㈣之製備及 給藥時程可根據製造商之說明書或如熟練從業者憑 確定加以使用。 上文已揭示可組合之各種化學治療劑。在—些實施例 中,待組合之化學治療劑係選自由以下組成之群:紫朴 類(〜包括多騎㈣及太平洋㈣醇)、長春花類(諸如長^ 瑞賓或長春鹼)、鉑化合物(諸如卡鉑或順鉑)、芳香酶抑制 劑(諸如來曲丨、安美達疑或依西美坦)、抗雌激素(例如氣 維司群(fulvestrant)或他莫昔芬)、依託泊苷、塞替派環 磷醯胺、甲胺喋呤、脂質體阿黴素、聚乙二醇化脂質體阿 黴素、卡培他濱、吉西他濱、COX_2抑制劑(例如塞内昔 布)或蛋白體抑制劑(例如PS342)。 調配物、劑量及投藥 本發明中所使用之治療劑應以與優良醫學規範一致之方 式調配、定劑量及投藥。本文所考慮之因素包括所治療之 特定病症、所治療之特定個體、個別患者之臨床病狀、病 症之病因、藥劑傳遞位點、投藥方法、投藥時程、 組合 藥劑之藥物-藥物相互作用及醫師所知之其他因素。 治療調配物係使用此項技術中已知之標準方法, 稽田 合具有期望純度之活性成分與視情況選用之生理學 143939.doc -138- 201019961 受之載劑、賦形劑或穩定劑而製備(Remington's Pharmaceutical Sciences (第 20版),A. Gennaro 編,2000, Lippincott,Williams &amp; Wilkins, Philadelphia, PA)。可接受 之載劑包括生理鹽水或緩衝液,諸如磷酸鹽、檸檬酸鹽及 其他有機酸;抗氧化劑,包括抗壞血酸;低分子量(小於 約10個殘基)多肽;蛋白質,諸如血清白蛋白、明膠或免 疫球蛋白;親水性聚合物,諸如聚乙烯吡咯啶酮;胺基 酸,諸如甘胺酸、麩醯胺酸、天冬醯胺、精胺酸或離胺 酸;單醣、雙醣及其他碳水化合物,包括葡萄糖、甘露糖 或糊精;螯合劑,諸如EDTA ;糖醇,諸如甘露糖醇或山 梨糖醇;成鹽相對離子,諸如鈉;及/或非離子型界面活 調配物視情況但較佳地含有醫藥學上可接受之鹽,較佳 含有氯化鈉,且較佳為約生理學濃度。本發明之調配物視 情況可含有醫藥學上可接受之防腐劑。在一些實施例中, 防腐劑之濃度範圍為通常以v/v計0.1%至2.0%。合適之防 腐劑包括醫藥技術中所知之防腐劑。苯甲基醇、苯酚、間 曱酚、對羥基苯甲酸甲酯及對羥基苯曱酸丙酯為較佳防腐 劑。本發明調配物視情況可包括濃度為0.005%至0.02%之 醫藥學上可接受之界面活性劑。 根據所治療之特定適應症之需要,本文調配物亦可含有 一種以上活性化合物,較佳為具有彼此不產生不利影響之 互補活性的化合物。該等分子合適地以有效用於預定目的 之量組合存在。 143939.doc -139- 201019961 亦可在膠體藥物傳遞系統(例如脂質體、白蛋白微球 體、微乳液、奈米粒子及奈米膠囊)或巨乳液中將活性成 分裹入例如利用凝聚技術或界面聚合所製備之微膠囊(分 別例如為羥基甲基纖維素或明膠微膠囊及聚(甲基丙烯酸甲 Μ微膠囊)中。該等技術揭示於Remingt〇n,s pharmaceutical Sciences,同上中。 可製備持續釋放製劑。持續釋放製劑之適合實例包括含 有抗體之固體疏水性聚合物半透基質,該等基質呈成形物 品形式.,例如膜或微膠囊。持續釋放基質之實例包括聚 酯;水凝膠(例如聚(2-羥基乙基-甲基丙烯酸酯)或聚(乙烯 醇));聚乳酸交酯(美國專利第3,773,919號);L-麩胺酸與 γ-乙基-L-麩胺酸酯之共聚物;不可降解之乙烯乙酸乙烯 S曰’可降解之乳酸-乙醇酸共聚物,諸如lupron DEPOT (包含乳酸-乙醇酸共聚物及柳培林(〗eUpr〇】ide acetate)之可注射微球體);及聚羥基丁酸。雖然 諸如乙烯乙酸乙烯酯及乳酸—乙醇酸之聚合物能夠釋放分 子超過100天,但某些水凝膠釋放蛋白質較短時期。當囊 封抗體長時間保留在鳢内時,由於其在37。(:下曝露於水分 可能變性或聚集,導致生物活性損失及可能改變免疫原 性。視所涉及之機制而定,可設計合理策略來實現穩定 化°舉例而言,若發現聚集機制為經由硫基-二硫化物互 換形成分子間S-S鍵,則可藉由修改硫氫基殘基、自酸性 溶液凍乾、控制水分含量、使用適當添加劑及開發特定的 聚合物基質組合物來實現穩定化。 143939.doc 201019961 本發明治療劑係根據已知方法向人類患者投與,該等方 法諸如以推注(bolus)形式經靜脈内投與,或藉由連續輸注一 ί又時間,藉由肌肉内、腹膜内、腦脊聽内(intracer〇br〇Spinai)、 皮下、關節内、滑膜内、鞘内、口腔、局部或吸入途徑。 在VEGF结抗劑之情況下,若廣泛副作用或毒性與VEGF拮 抗作用相關’則尤其期望局部投藥。治療應用亦可使用離 趙策略。離體策略包括用編碼卜爪以或EGFR或VEGF拮抗 劑之聚核苷酸轉染或轉導自個體獲得之細胞。隨後使轉染 或轉導之細胞返回個體。該等細胞可為多種類型中之任一 種’包括(但不限於)造血細胞(例如骨髓細胞、巨噬細胞、 單核細胞、樹突狀細胞、T細胞或B細胞)、纖維母細胞、 上皮細胞、内皮細胞、角質細胞或肌細胞。 舉例而言,若c-met或EGFR或VEGf拮抗劑為抗體,則 該抗體利用任何適合方式投與,該等方式包括非經腸、皮 下、腹膜内、肺内及鼻内投藥,若需要局部免疫抑制治 療’則包括病變内投藥》非經腸輸注包括肌肉内、靜脈 内、動脈内、腹膜内或皮下投藥。另外,適當地藉由脈衝 輸注來投與抗體,尤其遞減劑量之抗體。較佳藉由注射, 最佳藉由靜脈内或皮下注射來給藥,部分取決於投藥為短 暫或長期。 在另一實例中,當病症或腫瘤位置允許時,藉由例如直 接注射局部投與c_met或EGFR4VEGF拮抗劑化合物,且 注射可週期性重複。c_met或EGFR或VEGF拮抗劑亦可全 身性傳遞至個體或直接傳遞至腫瘤細胞,例如傳遞至腫瘤 143939.doc -141 - 201019961 或在手術切除腫瘤後傳遞至腫瘤床,以預防或減少局部復 發或轉移。 當抑制劑為抗體時,所投與之抗體較佳為裸抗體。然 而’所投與之抑制劑可與細胞毒性劑結合^所結合之抑制 劑及/或其所結合之抗原較佳經細胞内化,從而使得結合 、 物殺死其所結合之癌細胞之治療功效增加。在一較佳實施 ·: 例中’細胞毒性劑乾向或干擾癌細胞中之核酸。該等細胞 毒性劑之實例包括美登素類、卡奇黴素、核糖核酸酶及 DNA核酸内切酶。 ❿ 任何本文所列之治療劑之合適劑量均為目前所使用之劑 量且在治療醫師認為時可降低或增加。 在一些實施例中’投與足以獲得15微克/毫升或15微克/ 毫升以上之血清合濃度(serum trough concentration)之量的 抗c-met抗體。在一些實施例中,投與劑量為每三週約i 5 mg/kg或更向之抗omet抗體。在一些實施例中,投與劑量 為母三週約15-20mg/kg之抗c-met抗體。 在一些實施例中’在三週時期内投與總劑量為約丨5 © mg/kg或更高之抗c-met抗體。 在一實施例中,EGFR拮抗劑為埃羅替尼❶可在三週週 期之每一天均投與劑量為150 mg之埃羅替尼。在一些實施 · 例中,投與劑量為1〇〇 mg之埃羅替尼。在一些實施例中’ -· 投與劑量為50 mg之埃羅替尼。預期如埃羅替尼之標籤所 指示來減少埃羅替尼之劑量。 在某些實施例中,當組合使用時,投與在約〇 〇5 mg/kg 143939.doc -142· 201019961 至約15 mg/kg範圍内之貝 s*猫&amp;七夕加 仪皁抗。在一實施例中,可向個 體投與一或多個以下劑量: 巧 〇.5 mg/kg、1.0 mg/kg、2 〇 mg/kg、3.0 mg/kg、4·〇 . ,Λ &quot; g/kg、5_G mg/kg、6.0 mg/kg、 7.0 mg/kg、7.5 mg/kg、8 ft n 8 g 8·〇 mg/kg' 9.0 mg/kg、w 或15 mg/kg(或其任何組合 ) 該專劑$可間歇地投與,例 如每天、每三天、每週或 及每兩至二週投與一次。在另一實 施例中,當組合使用時,备 ^ 母隔一週經靜脈内向個體投與1〇Natl. Acad. Sci. USA, Vol. 83, p. 586, 1986. The probe is generally complementary to a sequence external to the kinase domain. The entire set of nucleic acid probes can be used to construct a set that detects mutations in the target nucleic acid. This set allows for hybridization to a large region of the relevant target sequence I43939.doc • 136·201019961. The probes can overlap or abut each other. If a ribonucleic acid probe is used to detect a mismatch in an mRNA, it is generally complementary to the mRNA of the target gene. Because an RNA probe is complementary to a sense strand, it does not encode the corresponding gene product and is therefore an antisense probe. The riboprobe probe will typically be labeled with a radioactive, colorimetric or fluorescent material, which can be accomplished by any method known in the art. If a riboprobe is used to detect a mismatch in DNA, it can be polar or antisense. Similarly, DNA probes can also be used to detect mismatches. In some cases, cancer overexpresses or does not overexpress c-met receptors and/or EGFR. Overexpression of the receptor can be determined in a diagnostic or prognostic assay by assessing the increase in the amount of receptor protein present on the cell surface (e.g., via immunohistochemical assay; IHC). Alternatively or additionally, for example, via fluorescence in situ hybridization (FISH; see WO 98/45479, published October 1998), southern blotting or polymerase chain reaction (PCR) techniques (such as real-time quantitative PCR) (RT-PCR)) to measure the amount of nucleic acid encoding a receptor in a cell. In addition to the above tests, skilled personnel can also use a variety of in vivo tests. For example, cells in a patient can be exposed to antibodies that detect a detectable label (eg, a radioisotope) as appropriate, and can be obtained by, for example, external scanning radioactivity or analysis of a biopsy obtained from a patient previously exposed to the antibody. The binding of the antibody to the patient's cells is assessed. Chemotherapeutic Agents The combination therapies of the invention may further comprise one or more chemotherapeutic agents. Combination administration includes co-administration or concurrent administration using separate formulations or individual pharmaceutical formulations, and continuous administration in either order, with preferably 143939.doc -137-201019961 present-stage period 'at this time two (or All) active agents simultaneously exert their biological activity. If a chemotherapeutic agent is administered, the administered dose will usually be its known dose, or may be due to a combination of drugs or may be negative due to administration of an antimetabolite chemotherapeutic agent. The preparation of the chemical treatment (4) and the time course of administration can be used according to the manufacturer's instructions or as determined by a skilled practitioner. Various chemotherapeutic agents that can be combined have been disclosed above. In some embodiments, the chemotherapeutic agent to be combined is selected from the group consisting of: purple (including multi-riding (four) and Pacific (tetra) alcohol), periwinkle (such as long rubin or vinblastine), Platinum compounds (such as carboplatin or cisplatin), aromatase inhibitors (such as Letrozin, Ammet or Exemestane), antiestrogens (such as fulvestrant or tamoxifen), Etoposide, thiotecycline, methotrexate, liposomal doxorubicin, pegylated liposomal doxorubicin, capecitabine, gemcitabine, COX 2 inhibitor (eg necenab) Or a proteosome inhibitor (eg PS342). Formulations, Dosage, and Administration The therapeutic agents used in the present invention should be formulated, dosed, and administered in a manner consistent with good medical practice. Factors considered herein include the particular condition being treated, the particular individual being treated, the clinical condition of the individual patient, the cause of the condition, the site of the agent, the method of administration, the time course of administration, the drug-drug interaction of the combination agent, and Other factors known to the physician. Therapeutic formulations are prepared using standard methods known in the art, and the active ingredients of the desired purity and the physiologically selected 143939.doc-138-201019961 carrier, excipient or stabilizer are used (Remington's) Pharmaceutical Sciences (20th Edition), edited by A. Gennaro, 2000, Lippincott, Williams &amp; Wilkins, Philadelphia, PA). Acceptable carriers include physiological saline or buffers such as phosphates, citrates and other organic acids; antioxidants, including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins such as serum albumin, gelatin Or immunoglobulin; a hydrophilic polymer such as polyvinylpyrrolidone; an amino acid such as glycine, glutamic acid, aspartame, arginine or lysine; monosaccharide, disaccharide and Other carbohydrates, including glucose, mannose or dextrin; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming ions, such as sodium; and/or non-ionic interfacial ligands Preferably, however, it preferably contains a pharmaceutically acceptable salt, preferably sodium chloride, and preferably about physiological concentration. The formulations of the present invention may optionally contain a pharmaceutically acceptable preservative. In some embodiments, the concentration of the preservative ranges from 0.1% to 2.0%, typically in v/v. Suitable preservatives include those known in the pharmaceutical arts. Phenylmethyl alcohol, phenol, m-nonylphenol, methyl p-hydroxybenzoate and propyl p-hydroxybenzoate are preferred preservatives. The formulations of the present invention may optionally comprise from 5% to 0.02% by weight of a pharmaceutically acceptable surfactant. The formulations herein may also contain more than one active compound, preferably a compound having complementary activities that do not adversely affect each other, depending on the particular indication being treated. The molecules are suitably present in combination in an amount effective for the intended purpose. 143939.doc -139- 201019961 It is also possible to entrap active ingredients in colloidal drug delivery systems (eg liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or macroemulsions, for example using agglomeration techniques or interfaces. The microcapsules prepared by polymerization (for example, hydroxymethylcellulose or gelatin microcapsules and poly(methyl sulfonium methacrylate microcapsules, respectively) are polymerized. These techniques are disclosed in Remingt〇n, s pharmaceutical Sciences, supra. Sustained release formulations. Suitable examples of sustained release formulations include solid hydrophobic polymeric semipermeable matrices containing antibodies, which are in the form of shaped articles, such as films or microcapsules. Examples of sustained release matrices include polyesters; hydrogels (eg poly(2-hydroxyethyl-methacrylate) or poly(vinyl alcohol)); polylactide (U.S. Patent No. 3,773,919); L-glutamic acid and γ-ethyl-L-glutamine Copolymer of acid ester; non-degradable ethylene vinyl acetate S曰' degradable lactic acid-glycolic acid copolymer, such as lupron DEPOT (including lactic acid-glycolic acid copolymer and Liu Pei Lin (〗 〖eUpr Injectable microspheres; and polyhydroxybutyrate. While polymers such as ethylene vinyl acetate and lactic acid-glycolic acid are capable of releasing molecules for more than 100 days, certain hydrogels release proteins for a shorter period of time. When the encapsulated antibody remains in the sputum for a long time, it may be degraded or aggregated due to its exposure to water, resulting in loss of biological activity and possibly altering immunogenicity. Depending on the mechanism involved, it can be designed reasonably. Strategy to achieve stabilization. For example, if the aggregation mechanism is found to form intermolecular SS bonds via thio-disulfide interchange, the sulphur residue can be modified, lyophilized from an acidic solution, and moisture content controlled. Stabilization is achieved using appropriate additives and development of specific polymer matrix compositions. 143939.doc 201019961 Therapeutic agents of the invention are administered to human patients according to known methods, such as intravenously in the form of bolus Inject, or by continuous infusion for a while, by intramuscular, intraperitoneal, intracranial (intracer〇br〇Spinai), subcutaneous, intra-articular, synovial Intrathecal, oral, topical or inhalation routes. In the case of VEGF antagonists, if extensive side effects or toxicity are associated with VEGF antagonism, then topical administration is particularly desirable. Therapeutic applications may also use the off-campus strategy. The cells obtained from the individual are transfected or transduced with a polynucleotide encoding a paw or an EGFR or VEGF antagonist. The transfected or transduced cells are then returned to the individual. The cells can be any of a variety of types. 'Includes, but is not limited to, hematopoietic cells (eg, bone marrow cells, macrophages, monocytes, dendritic cells, T cells, or B cells), fibroblasts, epithelial cells, endothelial cells, keratinocytes, or muscle cells. For example, if the c-met or EGFR or VEGf antagonist is an antibody, the antibody is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal administration, if desired. Immunosuppressive therapy 'includes intralesional administration'. Parenteral infusion includes intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration. In addition, antibodies are administered by pulse infusion, particularly by decreasing doses of the antibody. Preferably, it is administered by injection, preferably by intravenous or subcutaneous injection, depending in part on whether the administration is short or long term. In another example, a c_met or EGFR4 VEGF antagonist compound is administered topically by, for example, direct injection, when the condition or tumor location permits, and the injection can be repeated periodically. C_met or EGFR or VEGF antagonists can also be delivered systemically to the individual or directly to tumor cells, for example to tumor 143939.doc-141 - 201019961 or to the tumor bed after surgical removal of the tumor to prevent or reduce local recurrence or Transfer. When the inhibitor is an antibody, the antibody administered is preferably a naked antibody. However, the inhibitor to which the inhibitor is administered can bind to the cytotoxic agent and/or the antigen to which it binds is preferably intracellularly internalized, thereby allowing the binding to kill the cancer cells to which it binds. Increased efficacy. In a preferred embodiment, the cytotoxic agent is dry or interferes with nucleic acids in cancer cells. Examples of such cytotoxic agents include maytansinoids, calicheamicin, ribonucleases, and DNA endonucleases.合适 Any suitable dosage of the therapeutic agents listed herein is the amount currently employed and may be reduced or increased as the treating physician believes. In some embodiments, an anti-c-met antibody is administered in an amount sufficient to obtain a serum trough concentration of 15 μg/ml or 15 μg/ml or more. In some embodiments, the administered dose is about 5 mg/kg or more anti-omet antibody every three weeks. In some embodiments, the anti-c-met antibody is administered at a dose of about 15-20 mg/kg for three weeks. In some embodiments, an anti-c-met antibody is administered at a total dose of about ©5 © mg/kg or higher over a three week period. In one embodiment, the EGFR antagonist is erlotinib and a dose of 150 mg of erlotinib can be administered on each of the three week weeks. In some embodiments, an erlotinib dose of 1 mg is administered. In some embodiments, a dose of 50 mg of erlotinib is administered. It is expected that the dose of erlotinib will be reduced as indicated by the label of erlotinib. In certain embodiments, when used in combination, the shellfish is administered at a dose of about 5 mg/kg 143939.doc -142.201019961 to about 15 mg/kg. In one embodiment, one or more of the following doses can be administered to an individual: 巧〇.5 mg/kg, 1.0 mg/kg, 2 〇mg/kg, 3.0 mg/kg, 4·〇., Λ &quot; g/kg, 5_G mg/kg, 6.0 mg/kg, 7.0 mg/kg, 7.5 mg/kg, 8 ft n 8 g 8·〇mg/kg' 9.0 mg/kg, w or 15 mg/kg (or Any combination) The special dose $ can be administered intermittently, for example once a day, every three days, every week or every two to two weeks. In another embodiment, when used in combination, the parent is administered intravenously to the individual one week apart.

mg/kg之貝伐單抗,戋善= 、 母一週經靜脈内向個體投與 mg/kg之貝伐單抗。 、 治療劑之組合投與通常在規定時間内(通常視所選擇之 組合而定,在數分鐘、數小時、數天或數週内)進行。組 合療法意欲包括以連續方式投與此等治療劑(亦即各治療 劑在不同時間投與)以及以實質上同時方式投與此等治療 劑或至少兩種治療劑。 治療劑可經由相同途徑或不同途徑投與。舉例而言,組 合形式之VEGF、EGFR或c-met拮抗劑可藉由靜脈内注射 而投與’而組合形式之蛋白激酶抑制劑可經口投與。或 者,舉例而言,視特定治療劑而定,該兩種治療劑均可經 口投與’或該兩種治療劑均可藉由靜脈内注射而投與。投 與治療劑之順序亦視特定藥劑而變化。 本申請案預期利用基因療法來投與VEGF、c-met及/或 EGFR拮抗劑。關於使用基因療法來產生細胞内抗體,參 見例如1996年3月14日公開之WO 96/07321。 主要有兩種使核酸(視情況含於載體中)進入患者細胞中 143939.doc •143- 201019961 之方法;活體内及離體。對於活體内傳遞,通常在需要抗 體之位點處直接注射核酸於患者中。對於離體處理,移出 患者細胞,將核酸引入此等分離細胞中,且將經修飾細胞 直接投與患者,或例如囊封於將植入患者中之多孔膜内 (參見例如美國專利第4,892,538號及第5 283 187號)。存在 多種可用於將核酸引入活細胞中之技術。視活體外轉移核 酸至培養細胞内,或活體内轉移核酸至預定宿主之細胞内 而定,該等技術可不同。適於活體外轉移核酸至哺乳動物 細胞内之技術包括使用脂質體、電穿孔、顯微注射、細胞 融合、DEAE-葡聚糖、磷酸鈣沈澱法等。離體傳遞基因之 常用載體為逆轉錄病毒。 目前較佳之活體内核酸轉移技術包括用病毒載體(諸如 腺病毒、單純性疱療I型病毒或腺相關病毒)轉染及基於脂 質之系統(脂質介導之基因轉移之適用脂質為例如 DOTMA、DOPE及DC-Chol)。在一些情形下,可能需要向 核酸源提供靶向靶細胞之.藥劑,諸如對細胞表面膜蛋白或 乾細胞具有特異性之抗體、靶細胞上之受體之配位體等。 當使用脂質體時,可使用結合與内飲作用相關之細胞表面 膜蛋白之蛋白質來靶向及/或促進攝取(例如)對特定細胞類 型具有向性之衣殼蛋白或其片段、在循環中經歷内化之蛋 白質之抗體、及把向細胞内定位且增強細胞内半衰期之蛋 白質。受體介導之内飲作用之技術描述於例如Wu等人,j 如〇/· 262:4429-4432 (1987);及 Wagner等人,Pr〇cMB/kg of bevacizumab, 戋善=, and parental administration of mg/kg of bevacizumab to the individual intravenously. The combination of therapeutic agents is usually administered within a specified period of time (generally within minutes, hours, days or weeks depending on the combination selected). Combination therapies are intended to include administering such therapeutic agents in a continuous manner (i.e., each therapeutic agent is administered at different times) and administering the therapeutic agents or at least two therapeutic agents in a substantially simultaneous manner. The therapeutic agent can be administered via the same route or by different routes. For example, a combined form of a VEGF, EGFR or c-met antagonist can be administered by intravenous injection&apos; and a combined form of a protein kinase inhibitor can be administered orally. Alternatively, for example, depending on the particular therapeutic agent, both therapeutic agents can be administered orally or both therapeutic agents can be administered by intravenous injection. The order in which the therapeutic agent is administered will also vary depending on the particular agent. This application contemplates the use of gene therapy to administer VEGF, c-met and/or EGFR antagonists. For the use of gene therapy to produce intracellular antibodies, see, for example, WO 96/07321, published March 14, 1996. There are two main methods for introducing nucleic acids (as appropriate in a vector) into a patient's cells. 143939.doc • 143-201019961; in vivo and ex vivo. For in vivo delivery, the nucleic acid is typically injected directly into the patient at the site where the antibody is desired. For ex vivo treatment, the patient cells are removed, nucleic acids are introduced into the isolated cells, and the modified cells are administered directly to the patient, or, for example, encapsulated in a porous membrane to be implanted in a patient (see, e.g., U.S. Patent No. 4,892,538 And No. 5 283 187). There are a variety of techniques available for introducing nucleic acids into living cells. Such techniques may vary depending on whether the nucleic acid is transferred in vitro to the cultured cells, or the nucleic acid is transferred in vivo to the cells of the intended host. Techniques suitable for the in vitro transfer of nucleic acids into mammalian cells include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, calcium phosphate precipitation, and the like. A commonly used vector for in vitro delivery of genes is a retrovirus. Currently preferred in vivo nucleic acid transfer techniques include transfection with viral vectors (such as adenovirus, simple blister type I virus or adeno-associated virus) and lipid-based systems (lipid-mediated gene transfer for lipids such as DOTMA, DOPE and DC-Chol). In some cases, it may be desirable to provide a nucleic acid source with an agent that targets a target cell, such as an antibody specific for a cell surface membrane protein or stem cell, a ligand for a receptor on a target cell, and the like. When a liposome is used, a protein that binds to a cell surface membrane protein associated with endocytosis can be used to target and/or facilitate uptake, for example, a capsid protein or fragment thereof that is tropic for a particular cell type, in circulation. An antibody that undergoes internalization of the protein, and a protein that localizes within the cell and enhances intracellular half-life. Techniques for receptor-mediated endocytosis are described, for example, in Wu et al., j. 〇/. 262:4429-4432 (1987); and Wagner et al., Pr.

Jcac/· «Sci. 87:3410-3414 (1990)中。關於目前已 143939.doc 201019961 知之基因標記及基因療法方案之综述,參見Anderson等 人,Sdewce 256:808-813 (1992)。亦參見 WO 93/25673 及 其中引用之參考文獻。 以下為本發明方法及組合物之實例。應瞭解,鑒於以上 所提供之一般描述,可實施各種其他實施例。 實例 材料及方法 細胞株:自物理與化學研究所(Institute of Physical and Chemical Research)(Ibaraki,Japan)之RIKEN生物資源中心 (RIKEN BioResource Center)獲得KP4細胞株。自美國菌種 保存中心(American Type Culture Collection)(Manassas, VA)獲得NCI-H596細胞株。 NCI-H441-shMet-3.ll 及 EBC-l-shMet-3.15 及 4.5 先前已 描述於共同擁有之同在申請中之美國專利公開案第 2009/0226443號中。簡言之,如下製備逆轉錄病毒構築 體:將編碼針對c-met 之 shRNA 序列(5,-GATCCCCGAACAG AATCACTGACATATTCAAGAGATATGTCAGTGATTCTGTT CTTTTTTGGAAA-3' (SEQ ID NO: 29)(shMet 3)及 5'GATCCCCGAAACTGTATGCTGGATGATTCAAGAGATC ATCCAGCATACAGTTTCTTTTTTGGAAA (SEQ ID NO: 30) (shMet 4))之寡核苷酸選殖於HI啟動子下游之pShuttle-Hl 載體的Bglll/Hindlll位點中(David Davis,GNE)。粗體文字 表示乾雜交序列。使用Clonase II酶(Invitrogen)用逆轉錄 病毒 pHUSH-GW載體(Gray D 等人,BMC Biotechnology. 143939.doc -145- 201019961 2007; 7:61)重組此等構築體,從而產生shRNA表現受誘導 性啟動子控制之構築體。用四環素類似物多西環素 (doxycycline)進行處理引起shRNA表現。含有針對GFP之 shRNA之shGFP2對照逆轉錄病毒構築體(Hoeflich等人, Cancer Res. (2006) 66(2):999-1006)由 David Davis, Genentech, Inc.提供。shGFP2含有以下寡核苷酸:Jcac/· «Sci. 87:3410-3414 (1990). For a review of currently known 143939.doc 201019961 known gene signatures and gene therapy protocols, see Anderson et al, Sdewce 256:808-813 (1992). See also WO 93/25673 and references cited therein. The following are examples of the methods and compositions of the present invention. It will be appreciated that in view of the general description provided above, various other embodiments can be implemented. EXAMPLES Materials and Methods Cell lines: KP4 cell lines were obtained from the RIKEN BioResource Center of the Institute of Physical and Chemical Research (Ibaraki, Japan). The NCI-H596 cell line was obtained from the American Type Culture Collection (Manassas, VA). NCI-H441-shMet-3.ll and EBC-l-shMet-3.15 and 4.5 are previously described in commonly-owned co-pending U.S. Patent Publication No. 2009/0226443. Briefly, retroviral constructs were prepared as follows: shRNA sequences encoding c-met (5,-GATCCCCGAACAG AATCACTGACATATTCAAGAGATATGTCAGTGATTCTGTT CTTTTTTGGAAA-3' (SEQ ID NO: 29) (shMet 3) and 5'GATCCCCGAAACTGTATGCTGGATGATTCAAGAGATC ATCCAGCATACAGTTTCTTTTTTGGAAA (SEQ ID NO: 30) (shMet 4)) The oligonucleotide was cloned in the Bglll/Hindlll site of the pShuttle-Hl vector downstream of the HI promoter (David Davis, GNE). Bold letters indicate dry hybridization sequences. Recombination of these constructs with the retroviral pHUSH-GW vector (Gray D et al, BMC Biotechnology. 143939. doc-145-201019961 2007; 7:61) using the Clonase II enzyme (Invitrogen) to generate shRNA expression inducibility The construct of the promoter control. Treatment with the tetracycline analog doxycycline caused shRNA expression. A shGFP2 control retroviral construct containing shRNA directed against GFP (Hoeflich et al, Cancer Res. (2006) 66(2): 999-1006) was supplied by David Davis, Genentech, Inc. shGFP2 contains the following oligonucleotides:

(EGFP) shRNA (有義)5'-GATCCCCAGATCCGCCACAACATCGATTCAA GAGATCGATGTTGTGGCGGATCTTGTTTTTTGGAAA-3 (SEQ ID NO: 31)。使經逆轉錄病毒構築體穩定轉導之純系 在適當培養基+/- 1 pg/ml多西環素(Clontech)中生長以誘導 shRNA表現,且使用抗c-met C-12抗體(Santa Cruz Biotech) 經由西方墨點法篩選c-met敲除。使用抗填酸基-c-met Y1003(Biosource)及抗磷酸基-c-met Y1234/1234(Cell Signaling)抗體建立鱗酸基-c-met(亦稱為pmet或p-pc-met) 墨點。作為對照組,使用抗肌動蛋白1-19抗體(Santa Cruz Biotech)建立肌動蛋白墨點。EBC純系3.1 5及EBC純系4.12 顯示c-met表現及鱗酸基-c-met含量急劇降低,H441純系 3.11及H441純系3.1顯示c-met表現及磷酸基-c-met表現中 度降低,而EBC純系4.5顯示c-met及雄酸基-c-met表現較少 降低。細胞株EBC純系4.5、EBC純系4.12含有構築體 shMet4,且細胞株H441純系3.1、H441純系3.11及EBC純 系3.15含有構築體811]^613。 西方墨點法:為拆分蛋白質,使全部細胞溶解產物在4- 143939.doc -146- 201019961 12% Tris-甘胺酸凝膠(Invitrogen)上跑勝。將凝膠·轉移至 0.45 m确酸纖維素膜(Invitrogen)上。在室溫下,在奥德賽 (Odyssey)阻斷緩衝液(LiCor Biosciences)中阻斷膜30分 鐘,隨後在4°c下在於阻斷緩衝液加0.1 % Tween 20中稀釋 之初級抗體中培育隔夜。用TBST洗滌膜,隨後在室溫下 與於阻斷緩衝液加0.1% Tween 20及0.01% SDS中之根據適 當初級抗體之山羊抗小鼠IR800(Rockland)、山羊抗兔 Alexa Fluor 680(Molecular Probes)及驢抗山羊 Alexa Fluor 680(Molecular Probes)—起培育2小時。藉由使用奥德赛系 統(Odyssey System)掃描來偵測抗體》 細胞株溶解產物:(活體外)用冷TBS清洗細胞。在冰 上,在溶解緩衝液(TBS + 1% NP40+無EDTA的完全最低蛋 白酶抑制劑(Complete Mini-EDTA-Free protease inhibitor) (Roche)+lx磷酸酯酶抑制劑混合液2(Sigma))中溶解細胞10 分鐘(在板上)。在4°C下在l〇〇〇〇xg下離心溶解產物10分 鐘,且轉移溶解產物至新管中。使用BCA檢定(Pierce)定 量溶解產物。 踵瘤溶解產物··用手持式均質機在溶解缓衝液(PBS+1% Triton X-100+無EDTA的完全最低蛋白酶抑制劑(R〇che)+3 X 磷酸醋酶抑制劑混合液2(sigina))中使腫瘤均質化。在冰上 在不定時渦旋下培育溶解產物1小時。在4°c下在l0000xg 下離心溶解產物10分鐘,且轉移溶解產物至新管中。使用 BCA檢定(Pierce)定量溶解產物。 改良性培養基:在細胞處理後,移除培養基,且在l〇〇〇〇xg 143939.doc -147· 201019961 下離心ίο分鐘。隨後轉移改良性培養基至新管中。 微陣列處理:將EBC-l shMet4-12及shGFP-2細胞接種於 10 cm板中(每種條件3份)。10%無Tet之FBS RPMI 1640且培 育隔夜。用新鮮培養基+/- 100 ng/ml多西環素(doxycylin)(dox) 更換培養基。培育板24小時或48小時。經dox處理24小時 後,用100 ng/ml HGF處理經HGF處理之細胞2小時。隨後 使細胞胰蛋白酶化且離心。冷凍離心塊且儲存在-80°C 下。使用Qiagen RNeasy套組,如製造商之說明書所述自 細胞提取RNA。 IL8檢定•·在PBS(pH 7.4)中稀釋抗人類IL8 (R&amp;D Systems,Minneapolis,MN,USA)至 5 pg/mL,且塗布於 Maxisorp 384孔平底板(Nunc,Neptune, NJ)上(每孔 25 μΐ), 在4°C下培育隔夜(16-18小時)。在洗滌緩衝液(PBS,pH 7.4,0.05% Tween 20)中洗滌板3次,且以每孔50 μΐ向各孔 中添加阻斷緩衝液(PBS,pH 7.4,0.5% BSA)。阻斷板1-3 小時;該培育及所有後續培育均在室溫下在回轉式震盪器 上進行。阻斷步驟期間,在樣品稀釋劑(PBS/0.5% BSA/0.05% T-20/5 mM EDTA/0.25% CHAPS/0.2% BGG/10 ppM Proclin)中製備樣品及標準物。洗滌經阻斷之板三次 (如上所述),且向ELISA板中添加樣品及標準物(每孔25 μΐ)。培育1-2小時後,洗滌ELISA板三次(如上所述),且藉 由添加在檢定緩衝液(PBS/0.5% BSA/0.05% Tween 20,pH 7.4)中稀釋至50 ng/mL之經生物素標記之抗人類IL8抗體 (R&amp;D Systems,. Minneapolis, MN, USA)(每孔 25 μΐ)來摘測 143939.doc -148- 201019961(EGFP) shRNA (sense) 5'-GATCCCCAGATCCGCCACAACATCGATTCAA GAGATCGATGTTGTGGCGGATCTTGTTTTTGGGAAA-3 (SEQ ID NO: 31). The pure line stably transduced by the retroviral construct was grown in appropriate medium +/- 1 pg/ml doxycycline (Clontech) to induce shRNA expression, and anti-c-met C-12 antibody (Santa Cruz Biotech) was used. Screening for c-met knockouts by Western blotting. Establishment of luciferyl-c-met (also known as pmet or p-pc-met) ink using anti-acid-based-c-met Y1003 (Biosource) and anti-phospho-c-met Y1234/1234 (Cell Signaling) antibodies point. As a control group, an actin ink dot was established using an anti-actin 1-19 antibody (Santa Cruz Biotech). EBC pure line 3.15 and EBC pure line 4.12 showed a sharp decrease in c-met performance and selenate-c-met content. H441 pure line 3.11 and H441 pure line 3.1 showed a moderate decrease in c-met performance and phosphate-c-met performance. EBC pure line 4.5 showed less reduction in c-met and androstyl-c-met performance. The cell line EBC pure line 4.5 and the EBC pure line 4.12 contained the construct shMet4, and the cell line H441 pure line 3.1, H441 pure line 3.11 and EBC pure line 3.15 contained the construct 811]^613. Western blotting method: To separate proteins, all cell lysates were run over a 4-143939.doc-146-201019961 12% Tris-Glyceric acid gel (Invitrogen). The gel was transferred to a 0.45 m cellulose acetate membrane (Invitrogen). The membrane was blocked in Odyssey blocking buffer (LiCor Biosciences) for 30 minutes at room temperature, followed by incubation in primary antibody diluted in blocking buffer plus 0.1% Tween 20 at 4 °C Overnight. The membrane was washed with TBST, followed by goat anti-mouse IR800 (Rockland), goat anti-rabbit Alexa Fluor 680 (Molecular Probes) according to the appropriate primary antibody in blocking buffer plus 0.1% Tween 20 and 0.01% SDS at room temperature. And cockroach anti-goat Alexa Fluor 680 (Molecular Probes) - incubated for 2 hours. The antibody was lysed by using the Odyssey System scan: (in vitro) the cells were washed with cold TBS. On ice, in lysis buffer (TBS + 1% NP40 + Complete Mini-EDTA-Free protease inhibitor (Roche) + lx phosphatase inhibitor cocktail 2 (Sigma)) Dissolve the cells for 10 minutes (on the plate). The lysate was centrifuged at 10 ° C for 10 minutes at 4 ° C and the lysate was transferred to a new tube. The lysate was quantified using a BCA assay (Pierce). Tumor lysate··Use a hand-held homogenizer in lysis buffer (PBS+1% Triton X-100+ EDTA-free complete minimum protease inhibitor (R〇che) + 3 X phosphatase inhibitor mixture 2 ( The tumor is homogenized in sigina)). The lysate was incubated on ice for 1 hour under occasional vortexing. The lysate was centrifuged at 10000 xg for 10 minutes at 4 ° C and the lysate was transferred to a new tube. The lysate was quantified using a BCA assay (Pierce). Modified medium: After the cells were treated, the medium was removed and centrifuged for 1 minute at l〇〇〇〇xg 143939.doc -147·201019961. The modified medium is then transferred to a new tube. Microarray treatment: EBC-l shMet4-12 and shGFP-2 cells were seeded in 10 cm plates (3 parts each). 10% without Tet FBS RPMI 1640 and incubated overnight. The medium was replaced with fresh medium +/- 100 ng/ml doxycylin (dox). The plates were incubated for 24 hours or 48 hours. After 24 hours of dox treatment, HGF-treated cells were treated with 100 ng/ml HGF for 2 hours. The cells were then trypsinized and centrifuged. The pellet was frozen and stored at -80 °C. RNA was extracted from the cells using the Qiagen RNeasy kit as described in the manufacturer's instructions. IL8 assay • Dilute anti-human IL8 (R&amp;D Systems, Minneapolis, MN, USA) to 5 pg/mL in PBS (pH 7.4) and plate on Maxisorp 384 well flat bottom plate (Nunc, Neptune, NJ) 25 μΐ per well), incubated overnight (16-18 hours) at 4 °C. Plates were washed 3 times in wash buffer (PBS, pH 7.4, 0.05% Tween 20) and blocking buffer (PBS, pH 7.4, 0.5% BSA) was added to each well at 50 μM per well. The plates were blocked for 1-3 hours; the incubation and all subsequent incubations were performed at room temperature on a rotary shaker. Samples and standards were prepared in sample diluent (PBS/0.5% BSA/0.05% T-20/5 mM EDTA/0.25% CHAPS/0.2% BGG/10 ppM Proclin) during the blocking step. The blocked plates were washed three times (as described above) and samples and standards (25 μM per well) were added to the ELISA plate. After 1-2 hours of incubation, the ELISA plate was washed three times (as described above) and diluted by adding the assay buffer (PBS/0.5% BSA/0.05% Tween 20, pH 7.4) to 50 ng/mL. Labeled anti-human IL8 antibody (R&amp;D Systems,. Minneapolis, MN, USA) (25 μM per well) for 143939.doc -148- 201019961

與板結合之人類IL8。培育板1-2小時且如上所述進行洗 滌。隨後向ELISA板中添加按1:40,000稀釋之與HRP結合之 抗生蛋白鏈菌素(Amersham BioSciences, Piscataway, NJ, US A)(每孔25 μΐ),且培育板0.5小時。最後洗滌後,以每 孔 25 μΐ 向 ELISA 板中添加 ΤΜΒ 受質(Kirkegaard&amp;Perry Laboratories, Gaithersburg, MD)。約 5分鐘後,藉由每孔添 加25 μΐ 1.0 Μ磷酸終止顯色。使用SpectraMax 250微量滴 定板讀取器(Molecular Devices, Sunnyvale, CA)測定 450 nm及650 nm下之吸光度。 人類 VEGF165-206 ELISA(偵測 VEGF165):在 4°C 下, 用0.4 pg/ml於50 mM碳酸鹽緩衝液(pH 9.6)中之3.5?8抗體 以每孔 100 μΐ 塗布 MaxiSorp 96孔微孔板(Nunc, Roskilde, Denmark)隔夜,且用於PBS中之0.05%聚山梨醇S旨20(pH 7.4)洗滌。在室溫下,用於PBS中之0.5%牛血清白蛋白、 10 ppm Proclin 300(Supelco,Bellefonte,PA)阻斷板(每孔 150 μΐ) 1小時,且洗滌。以每孔100 μΐ添加於PBS中之 0.5%牛血清白蛋白、0.05%聚山梨醇酯20、10 ppm Proclin 300、5mMEDTA及0.35NNaCl(pH6.3)(檢定緩衝液)中 之 VEGF標準物(1.56-200 pg/ml VEGF165 或 0.041-5.2 pM VEGF,兩倍連續稀釋;Genentech)及樣品(最小1:10稀釋 度)《在室溫下培育板2小時且洗滌。藉由添加於檢定緩衝 液中之經生物素標記之A4.6.1來偵測與板結合之VEGF。 在室溫下培育2小時之後,洗滌板,且添加於檢定緩衝液 中之抗生蛋白鍵菌素-HRP(GE Healthcare, Piscataway, 143939.doc •149· 201019961 NJ),培育30分鐘。洗滌板,接著添加受質3,3',5,5’-四曱基 聯苯胺(Kirkegaard &amp; Perry Laboratories, Gaithersburg, MD)。藉由添加1 M磷酸終止反應,且在450 nm下讀取吸 光度。使用四參數回歸曲線擬合程式(Genentech)分析滴定 曲線。使用屬於標準曲線範圍内之數據點計算樣品中之 VEGF濃度。 VEGF110-206 ELISA(偵測總 VEGF):在 4°C 下,用 0.5 pg/ml於50 mM碳酸鹽緩衝液(pH 9.6)中之抗體2E3以每孔 100 μΐ 塗布 MaxiSorp 96 孔微孔板(Nunc,Roskilde, Denmark)隔夜,且用PBS中之0.05%聚山梨醇酯20(pH 7.4) 洗滌。在室溫下,用於PBS中之0.5%牛血清白蛋白、10 ppm Proclin 300(Supelco, Bellefonte,PA)阻斷板(每孔 150 μ1)1小時,且洗滌。以每孔100 μΐ向板中添加於PBS中之 0.5%牛血清白蛋白、0.05%聚山梨醇酯20、10 ppm Proclin 300、5 mM EDTA、0.25% CHAPS、0.2% 牛 γ-球蛋白 (Sigma, St. Louis,MO)及 0.35 N NaCl (pH 7.4)(檢定緩衝 液)中之 VEGF標準物(1.56-200 pg/ml VEGF1654 0.0409_ 5.24 pM VEGF,兩倍連續稀釋;Genentech)及樣品(最小 1:10稀釋度)。在室溫下培育板2小時且洗滌。藉由添加於 樣品緩衝液中之經生物素標記之Α4·6·1(每孔100 μΐ)來偵測 與板結合之VEGF。在室溫下培育1小時後,洗滌板,且添 加抗生蛋白鍵菌素-HRP(GE Healthcare, Piscataway,NJ), 培育30分鐘(每孔100 μΐ)。洗滌板,接著向板中添加生物 素基酷胺鹽(ELAST ELISA擴增系統,Perkin Elmer Life 143939.doc -150· 201019961Human IL8 combined with a plate. The plates were incubated for 1-2 hours and washed as described above. HRP-bound streptavidin (Amersham BioSciences, Piscataway, NJ, US A) (25 μM per well) diluted 1:40,000 was then added to the ELISA plate and the plates were incubated for 0.5 hours. After the final wash, ΤΜΒ substrate (Kirkegaard &amp; Perry Laboratories, Gaithersburg, MD) was added to the ELISA plate at 25 μM per well. After about 5 minutes, color development was stopped by adding 25 μΐ 1.0 Μ phosphoric acid per well. Absorbance at 450 nm and 650 nm was measured using a SpectraMax 250 microtiter plate reader (Molecular Devices, Sunnyvale, CA). Human VEGF165-206 ELISA (detection of VEGF165): MaxiSorp 96-well microwells were coated with 0.4 μg/ml of 3.5?8 antibody in 50 mM carbonate buffer (pH 9.6) at 100 μΐ per well at 4 °C. Plates (Nunc, Roskilde, Denmark) were overnight and used for 0.05% polysorbate S 20 (pH 7.4) wash in PBS. The plates were incubated with 0.5% bovine serum albumin, 10 ppm Proclin 300 (Supelco, Bellefonte, PA) in PBS for 1 hour at room temperature and washed. VEGF standard in 0.5% bovine serum albumin, 0.05% polysorbate 20, 10 ppm Proclin 300, 5 mM EDTA and 0.35 NNaCl (pH 6.3) (assay buffer) added to PBS in 100 μM per well ( 1.56-200 pg/ml VEGF165 or 0.041-5.2 pM VEGF, two-fold serial dilution; Genentech) and sample (minimum 1:10 dilution) "The plates were incubated for 2 hours at room temperature and washed. Plate-bound VEGF was detected by biotinylated A4.6.1 added to assay buffer. After incubation for 2 hours at room temperature, the plates were washed and added to the assay buffer for streptavidin-HRP (GE Healthcare, Piscataway, 143939.doc • 149. 201019961 NJ) and incubated for 30 minutes. The plate was washed, followed by the addition of the 3,3',5,5'-tetradecylbenzidine (Kirkegaard &amp; Perry Laboratories, Gaithersburg, MD). The reaction was stopped by the addition of 1 M phosphoric acid and the absorbance was read at 450 nm. The titration curve was analyzed using a four parameter regression curve fitting program (Genentech). The VEGF concentration in the sample is calculated using data points that fall within the range of the standard curve. VEGF110-206 ELISA (Total VEGF Detection): MaxiSorp 96-well microplates were coated with 0.5 μg/ml of antibody 2E3 in 50 mM carbonate buffer (pH 9.6) at 100 μΐ per well at 4 °C ( Nunc, Roskilde, Denmark) was washed overnight with 0.05% polysorbate 20 (pH 7.4) in PBS. 0.5% bovine serum albumin, 10 ppm Proclin 300 (Supelco, Bellefonte, PA) blocking plates (150 μl per well) in PBS for 1 hour at room temperature and washed. 0.5% bovine serum albumin, 0.05% polysorbate 20, 10 ppm Proclin 300, 5 mM EDTA, 0.25% CHAPS, 0.2% bovine gamma globulin (Sigma) added to PBS in 100 μM per well. , St. Louis, MO) and 0.35 N NaCl (pH 7.4) (assay buffer) VEGF standard (1.56-200 pg/ml VEGF1654 0.0409_ 5.24 pM VEGF, two-fold serial dilution; Genentech) and sample (minimum 1:10 dilution). The plates were incubated for 2 hours at room temperature and washed. The plate-bound VEGF was detected by biotinylated Α4·6·1 (100 μM per well) added to the sample buffer. After incubating for 1 hour at room temperature, the plates were washed, and anti-protein-HRP (GE Healthcare, Piscataway, NJ) was added and incubated for 30 minutes (100 μM per well). The plate was washed and then biotinylated amide salt was added to the plate (ELAST ELISA Amplification System, Perkin Elmer Life 143939.doc -150· 201019961)

Sciences Inc.,ΜΑ)(每孔100 μΐ)。培育15分鐘後,洗務 板,且添加於樣品緩衝液中之抗生蛋白鏈菌素-HRP(每孔 100 μΐ)。培育30分鐘後,洗滌板,且添加受質3,3·,5,5·-四 甲基聯苯胺(Kirkegaard &amp; Perry Laboratories)(每孔 100 μΐ)。藉由添加1 Μ磷酸(每孔100 μΐ)終止反應。在450 nm下 讀取吸光度。使用四參數回歸曲線擬合程式(Genentech)分 析滴定曲線。使用屬於標準曲線範圍内之數據點計算樣品 中之VEGF濃度。 小鼠VEGF ELISA(偵測總VEGF):在4°C下,用0.5 pg/ml於50 mM碳酸鹽緩衝液(pH 9.6)中之山羊抗小鼠 VEGF(R&amp;D, Minneapolis, MN)以每孔 100 μΐ塗布 MaxiSorp 96孔微孔板(Nunc, Roskilde, Denmark)隔夜,且用於PBS中 之0.05%聚山梨醇酯20(pH 7.4)洗滌。在室溫下,用於PBS 中之 0.5% 牛血清白蛋白、10 ppm Proclin 300(Supelco, Bellefonte,PA)阻斷板(每孔150 μ1)1小時,且洗滌。以每 孔100 μΐ添加於PBS中之0.5%牛血清白蛋白、0.05%聚山梨 醇酯 20、10 ppm Proclin 300、5 mM EDTA、0.25% CHAPS、0.2% 牛 γ-球蛋白(Sigma,St. Louis, MO)及 0_35 N NaCl (pH 7.4)(樣品緩衝液)中之小鼠VEGF標準物(1.0-128 pg/mlVEGF164或 0.026-3.4 pMVEGF,兩倍連續稀釋; R&amp;D Systems, McKinley Place,NE)及樣品(最小稀釋度 1:10)。培育2小時後,藉由添加於樣品緩衝液中之經生物 素標記之山羊抗小鼠VEGF抗體來偵測與板結合之VEGF。 在室溫下培育1小時之後,洗滌板,且添加於PBS中之 143939.doc • 151 · 201019961 0.5 %牛血清白蛋白、0.05%聚山裂醇醋20、10 ppm Pro cl in 300 (pH 7.4)中之抗生蛋白鏈菌素-HRP(GE Healthcare, Piscataway,NJ)。培育45分鐘後,洗滌板,且添加受質 3,3’,5,5'-四甲基聯苯胺(Kirkegaard &amp; Perry Laboratories, Gaithersburg, MD)(每孔100 μΐ)。藉由添加1 Μ磷酸(每孔100 μΐ)終止反應,且在450 nm下讀取吸光度。使用四參數回歸 曲線擬合程式(Genentech)分析滴定曲線。使用屬於標準曲線 範圍内之數據點計算樣品中之VEGF濃度。 活體内異種移植腫瘤研究 你試#於··由Genentech, Inc.之抗體工程改造部門 (Antibody Engineering Department)呈澄清液體形式提供結 合小鼠與人類VEGF之抗VEGF抗體(B20-4.1及B20-4.1.1), 且在1 xPBS中稀釋。亦由Genentech, Inc.之抗體工程改造 部門呈澄清液體形式提供抗c-met單價單株抗體MetMAb (rhuOA5D5v2 ; WO 2007/063816),且在 MetMAb 緩衝液(10 mM組胺酸琥ίό酸鹽(histidine succinate)、4%海藻糖二水 合物、0.02%聚山梨醇酯20,pH 5.7)中稀釋。對照抗體包 括小鼠IgG2a同型10D9-1E11-1F12(抗豬草)抗體及人類 IgGl同型hu5B6(抗gD)抗體,該兩者均呈澄清液體形式自 Genentech,Inc.之抗體工程改造部門獲得,且在lxPBS中 稀釋。由OSI Pharmaceuticals向Genentech之調配物組提供 埃羅替尼(TarcevaTM),且與足量媒劑(甲基纖維素吐溫 (methylcellulose tween,MCT))— 起稱出。在5%蔗糖水中 新鮮製備0.5或1 mg/mL之多西環素(Dox),且每3天定期更 143939.doc -152· 201019961 換。在Dox研究中,給予對照動物5%蔗糖水,該5%蔗糖水 每3天更換一次。材料儲存於設定為維持4°C至8°C範圍内 之溫度之冰箱中。 #禮自 Charles River Laboratories (CRL)獲得 6至 8週大 的裸小鼠(nu/nu),且在進行研究之前,在Genentech之飼 養所(vivarium)馴化至少一週。在Van Andel研究所產生4至 6週大的hu-HGF-Tg-C3H-SCID小鼠,在該研究所中維持該 等小鼠且利用該品系進行所有研究。動物圈養在具有供應 高效率顆粒空氣(HEPA)的過濾器之房間申之通風籠系統 中。研究僅使用看起來健康且無明顯異常之動物。 研究設計 研究1 :在KP4胰腺異種移植腫瘤模型中用MetMAb舆抗 VEGF抗體之組合處理:在由RPMI 1640培養基 (In vitro gen)、2 mM L-麵醯胺酸及10%胎牛血清組成之生 長培養基中培養KP4細胞。為製備供接種於小鼠中之細 胞,使細胞胰蛋白酶化,用10毫升無菌lx磷酸鹽緩衝生理 鹽水(PBS)洗滌。利用錐蟲藍拒染(tryptan blue exclusion) 計數細胞之子集,且將剩餘細胞再懸浮於100 μΐ無菌1 X PBS中至濃度為每毫升5χ107個細胞。用5χ106個ΚΡ4細胞在 右下肩胛區中皮下接種小鼠。監測腫瘤直至其平均體積達 到 230 mm3。 將小鼠隨機分成7組,每組10隻小鼠,且開始處理(概述 於表1中)。組1中之小鼠用100 μι MetMAb缓衝液(IP,一 次)及抗豬草對照抗體(5 mg/kg,IP,每週兩次歷時3週)處 143939.doc -153· 201019961 理。組2中之小鼠用B20-4.1 (5 mg/kg,IP,每週兩次歷時3 週)處理。組3中之小鼠用次治療劑量(sub-therapeutic dose) 之MetMAb (2.5 mg/kg,IP,一次)處理。組4中之小鼠用21 天提供約50%腫瘤抑制之劑量之MetMAb (ED5〇: 7.5 mg/kg,IP,一次)處理。組5中之小鼠用高劑量之MetMAb ' (30 mg/kg,ip,一次)處理。給予組6中之小鼠次治療劑量 ? 之 MetMAb 加抗 VEGF 抗體(2.5 mg/kg MetMAb,IP,一 次,加5 mg/kg B20-4.1,IP,每週兩次歷時3週)。給予組7 中之小鼠ED50之MetMAb加抗VEGF抗體(7.5 mg/kg Φ MetMAb,IP,一次,加5 mg/kg B20-4.1,IP ’ 每週兩次 歷時3週)。每週兩次量測腫瘤體積,且監測動物25天。 表1 KP4胰腺異種移植腫瘤模型中MetMAb與抗VEGF抗體之組合 組 數目/ 性別 測試材料 途 徑 劑量頻率 劑量 (mg/kg) 劑量濃度 (mg/ml) 劑量體 積(μ〇 1 10/F MetMAb緩衝 液;對照抗體 IP 一次; 每週兩次x3週 0 0 100 2 10/F 抗VEGF抗體 (B20-4.1) IP 每週兩次(MetMAb 緩衝液一次)X3週 5 1.25 100 3 10/F MetMAb IP 一次(對照抗體每 週兩次) 2.5 0.625 100 4 10/F MetMAb IP 一次(對照抗體每 週兩次x3週) 7.5 1.875 100 5 10/F MetMAb IP 一次(對照抗體每 週兩次x3週) 30 7.5 100 6 10/F MetMAb+抗 VEGF抗體 (B20-4.1) IP 一次(MetMAb); 每週兩次CB20-4.1) 2.5 (MetMAb); 5 (B20-4.1) 0.625 (MetMAb); 1·25(Β2(Μ.1) 100 7 10/F MetMAb+抗 VEGF抗體 fB20-4.1) IP 一次(MetMAb); 每週兩次(B20-4.1) 2.5 (MetMAb); 5(B20-4.1) 0.625 (MetMAb); 1.25 (B20-4.1) 100 143939.doc -154- 201019961 研究2 :在NCI-H441 NSCLC異種移植腫瘤模型中用c-met敲除舆抗VEGF抗髏之組合處理:在由RPMI 1640培養 基(Invitrogen)、2 mM L-糙醯胺酸及10%胎牛血清組成之 生長培養基中培養NCI-H44l-shMet-3.11細胞。為製備供接 種於小鼠中之細胞,使細胞胰蛋白酶化,用10毫升無菌1X 磷酸鹽緩衝生理鹽水(PBS)洗滌。利用錐蟲藍拒染計數細 胞之子集,且將剩餘細胞再懸浮於100 μΐ無菌1XPBS中至 濃度為每毫升5χ107個細胞。用5xl06個NCI-H441-shMet-4.11細胞在右下肩胛區中皮下接種小鼠。監測腫瘤直至其 平均體積達到200 mm3。 將小鼠隨機分成4組,每組10隻小鼠,且開始處理(概述 於表2中)。組1中之小鼠用抗豬草對照抗體(5 mg/kg,IP, 每週兩次歷時4週)處理,且給予5%蔗糖飲用水。組2中之 小执用抗豬草對照抗體(5 mg/kg,IP,每週兩次歷時4週) 處理’且給予於5%蔗糖飲用水中之多西環素(丨mg/mL)。 組3中之小鼠用抗VEGF抗體(B20-4.1.1,5 mg/kg,IP,每 週兩次歷時4週)處理,且給予5%蔗糖飲用水。組4中之小 鼠用抗VEGF抗體(B20-4.1.1 ’ 5 mg/kg,IP,每週兩次歷時 4週)處理,且給予於5%蔗糖飲用水中之多西環素(1 mg/mLh繼續給予抗體四週,此時雖然維持對動物進行多 西.環素處理,但終止抗體給藥。監測腫瘤體積直至第57 天0 143939.doc -155- 201019961 表2 NCI-441 NSCLC異種移植腫瘤模型te_meti|j^|^VEGF抗體之組合 組 1 數目/ 性別 10/F 測試材料 蔗糖;抗豬 草對照抗體 途徑 飲用水; IP 劑量頻率 任意取食(整個 過程);每週兩 次x4週 劑量 (mg/kg) 5(抗豬草) 劑量濃度 (mg/ml) 1.25(抗豬草) 劑量體 積(μΐ) 100 2 10/F 多西環素; 對照抗體 飲用水; IP 任意取食(整個 過程);每週兩 次x4週 5(抗豬草) 1.25(抗豬 草);1 (Dox) 100 3 10/F 蔗糖;抗 VEGF(B20-4.1.1) 飲用水; IP 任意取食(整個 過程);每週兩 次x4週 5(抗 VEGF) 1.25(抗 VEGF) 100 4 10/F 多西環素; 抗 VEGF (B20-4.1.1) 飲用水; IP 任意取食(整個 過程);每週兩 次X4週 5(抗 VEGF) 1.25(抗 VEGF); 1 (Dox) 100 研究3及4 :在EBC-l-shMet NSCLC異種移植模型中用c-met敲除舆抗VEGF抗艎之组合處理:在由RPMI 1640培養 基(Invitrogen)、2 mM L-麩酿胺酸及1 〇%胎牛血清組成之 生長培養基中培養£801-311^461;-3.15及£801-3111^161;-4.5細 胞株。為製備供接種於小鼠中之細胞,使細胞胰蛋白酶 化,用10毫升無菌lx磷酸鹽緩衝生理鹽水(PBS)洗滌。利 用錐蟲藍拒染計數細胞之子集’且將剩餘細胞再懸浮於 100 μΐ無菌lxPBS中至濃度為每毫升5xl07個細胞。用5xl06 個 EBC-l-shMet-3.1 5及 EBC-l-shMet-4.5 細胞在右下肩胛區 中皮下接種小鼠。監測腫瘤直至其平均體積達到300 mm3 ° 對於 EBC-l-shMet-3.15研究與 EBC-l-shMet-4.5研究’將 小鼠隨機分成2組,每組10隻小鼠’總計4組’且開始處理 143939.doc -156- 201019961 (概述於表3中)。組1中之小鼠具有EBC-l-shMet-3.15腫 瘤,且給予5%蔗糖飲用水。組2中之小鼠具有EBC-1-shMet-3.15腫瘤,且給予於5%蔗糖飲用水中之多西環素(1 mg/mL)。組3中之小鼠具有EBC-l-shMet-4.5腫瘤,且給予 5%蔗糖飲用水。組4中之小鼠具有EBC-l-shMet-4.5腫瘤, 且給予於5¾蔗糖飲用水中之多西環素(丨mg/mL)。監測腫 瘤體積歷時21天。Sciences Inc., ΜΑ) (100 μM per well). After 15 minutes of incubation, the plate was washed and the streptavidin-HRP (100 μM per well) was added to the sample buffer. After incubation for 30 minutes, the plates were washed and the substrate was added with 3,3,5,5-tetramethylbenzidine (Kirkegaard &amp; Perry Laboratories) (100 μM per well). The reaction was stopped by the addition of 1 guanidine phosphate (100 μM per well). Read the absorbance at 450 nm. The titration curve was analyzed using a four parameter regression curve fitting program (Genentech). The VEGF concentration in the sample is calculated using data points that fall within the range of the standard curve. Mouse VEGF ELISA (Total VEGF Detection): goat anti-mouse VEGF (R&amp;D, Minneapolis, MN) at 0.5 pg/ml in 50 mM carbonate buffer (pH 9.6) at 4 °C MaxiSorp 96-well microplates (Nunc, Roskilde, Denmark) were coated overnight at 100 μM per well and washed with 0.05% polysorbate 20 (pH 7.4) in PBS. 0.5% bovine serum albumin, 10 ppm Proclin 300 (Supelco, Bellefonte, PA) blocking plates (150 μl per well) in PBS for 1 hour at room temperature and washed. 0.5% bovine serum albumin, 0.05% polysorbate 20, 10 ppm Proclin 300, 5 mM EDTA, 0.25% CHAPS, 0.2% bovine gamma globulin (Sigma, St.) added to PBS at 100 μM per well. Louis, MO) and mouse VEGF standard in 0_35 N NaCl (pH 7.4) (sample buffer) (1.0-128 pg/ml VEGF164 or 0.026-3.4 pMVEGF, two-fold serial dilution; R&amp;D Systems, McKinley Place, NE) and sample (minimum dilution 1:10). After 2 hours of incubation, the plate-bound VEGF was detected by biotinylated goat anti-mouse VEGF antibody added to the sample buffer. After incubation for 1 hour at room temperature, the plates were washed and added to PBS 143939.doc • 151 · 201019961 0.5% bovine serum albumin, 0.05% polysorbate 20, 10 ppm Pro cl in 300 (pH 7.4 Streptavidin-HRP (GE Healthcare, Piscataway, NJ). After 45 minutes of incubation, the plates were washed and the substrate was added with 3,3',5,5'-tetramethylbenzidine (Kirkegaard &amp; Perry Laboratories, Gaithersburg, MD) (100 μM per well). The reaction was stopped by the addition of 1 guanidine phosphate (100 μM per well) and the absorbance was read at 450 nm. The titration curve was analyzed using a four parameter regression curve fitting program (Genentech). The VEGF concentration in the sample is calculated using data points that fall within the range of the standard curve. In vivo xenograft tumor research you test #于·· Provided anti-VEGF antibody binding to mouse and human VEGF in a clear liquid form by the Antibody Engineering Department of Genentech, Inc. (B20-4.1 and B20-4.1 .1), and diluted in 1 x PBS. The anti-c-met monovalent monoclonal antibody MetMAb (rhuOA5D5v2; WO 2007/063816) was also provided in a clear liquid form by the antibody engineering department of Genentech, Inc., and in MetMAb buffer (10 mM histidine sulphate ( Diluted in histidine succinate), 4% trehalose dihydrate, 0.02% polysorbate 20, pH 5.7). Control antibodies include the mouse IgG2a isotype 10D9-1E11-1F12 (anti-porcine) antibody and the human IgG1 homotype hu5B6 (anti-gD) antibody, both obtained in clear liquid form from the Antibody Engineering Division of Genentech, Inc., and Dilute in lxPBS. Ertotinib (TarcevaTM) was supplied by OSI Pharmaceuticals to the Genentech formulation group and was found to be a sufficient amount of vehicle (methylcellulose tween (MCT)). 0.5 or 1 mg/mL of doxycycline (Dox) was freshly prepared in 5% sucrose water and changed regularly every 3 days 143939.doc -152· 201019961. In the Dox study, control animals were given 5% sucrose water, which was changed every 3 days. The material is stored in a refrigerator set to maintain a temperature in the range of 4 ° C to 8 ° C. #礼自 Charles River Laboratories (CRL) obtained nude mice (nu/nu) 6 to 8 weeks old and domesticated at the Genentech's feeding center (vivarium) for at least one week prior to the study. Four to six week old hu-HGF-Tg-C3H-SCID mice were generated at the Van Andel Institute, and the mice were maintained in the study and all studies were performed using this line. Animals are housed in a ventilated cage system in a room with a filter that supplies high efficiency particulate air (HEPA). The study used only animals that appeared healthy and had no significant abnormalities. Study Design Study 1: Treatment with a combination of MetMAb舆 anti-VEGF antibody in a KP4 pancreatic xenograft tumor model: consisting of RPMI 1640 medium (In vitro gen), 2 mM L-face valeric acid and 10% fetal bovine serum KP4 cells were cultured in growth medium. To prepare cells for inoculation into mice, the cells were trypsinized and washed with 10 ml of sterile lx phosphate buffered saline (PBS). A subset of cells was counted using tryptan blue exclusion and the remaining cells were resuspended in 100 μL sterile 1 X PBS to a concentration of 5χ107 cells per ml. Mice were inoculated subcutaneously in the lower right scapular region with 5χ106 ΚΡ4 cells. Tumors were monitored until their average volume reached 230 mm3. Mice were randomized into 7 groups of 10 mice each and treatment was started (summarized in Table 1). The mice in Group 1 were treated with 100 μM MetMAb buffer (IP, once) and anti-porcine control antibody (5 mg/kg, IP, twice a week for 3 weeks) at 143939.doc -153· 201019961. Mice in Group 2 were treated with B20-4.1 (5 mg/kg, IP, twice a week for 3 weeks). Mice in group 3 were treated with a sub-therapeutic dose of MetMAb (2.5 mg/kg, IP, once). Mice in group 4 were treated with MetMAb (ED5: 7.5 mg/kg, IP, once) at a dose of about 50% tumor inhibition for 21 days. Mice in group 5 were treated with high doses of MetMAb' (30 mg/kg, ip, once). What is the sub-therapeutic dose of the mice in group 6? MetMAb plus anti-VEGF antibody (2.5 mg/kg MetMAb, IP, once, plus 5 mg/kg B20-4.1, IP, twice a week for 3 weeks). MetMAb of mouse ED50 in group 7 was administered anti-VEGF antibody (7.5 mg/kg Φ MetMAb, IP, once, plus 5 mg/kg B20-4.1, IP' twice a week for 3 weeks). Tumor volume was measured twice a week and animals were monitored for 25 days. Table 1 Number of combinations of MetMAb and anti-VEGF antibodies in KP4 pancreatic xenograft tumor model / Sex test material route Dose frequency dose (mg/kg) Dose concentration (mg/ml) Dose volume (μ〇1 10/F MetMAb buffer Control antibody IP once; twice weekly x3 weeks 0 0 100 2 10/F anti-VEGF antibody (B20-4.1) IP twice a week (MetMAb buffer once) X3 weeks 5 1.25 100 3 10/F MetMAb IP once (Control antibody twice a week) 2.5 0.625 100 4 10/F MetMAb IP once (control antibody twice weekly x3 weeks) 7.5 1.875 100 5 10/F MetMAb IP once (control antibody twice a week x 3 weeks) 30 7.5 100 6 10/F MetMAb+anti-VEGF antibody (B20-4.1) IP once (MetMAb); twice weekly CB20-4.1) 2.5 (MetMAb); 5 (B20-4.1) 0.625 (MetMAb); 1·25 (Β2( Μ.1) 100 7 10/F MetMAb+anti-VEGF antibody fB20-4.1) IP once (MetMAb); twice a week (B20-4.1) 2.5 (MetMAb); 5 (B20-4.1) 0.625 (MetMAb); 1.25 ( B20-4.1) 100 143939.doc -154- 201019961 Study 2: Combination treatment with c-met knockout anti-VEGF anti-sputum in NCI-H441 NSCLC xenograft tumor model: in RPMI 1640 culture Yl growth medium (Invitrogen), 2 mM L- roughness acid amide, and 10% fetal bovine serum in the culture composition of the NCI-H44l-shMet-3.11 cells. To prepare cells for inoculation in mice, the cells were trypsinized and washed with 10 ml of sterile 1X phosphate buffered saline (PBS). A subset of the cells was counted using trypan blue exclusion and the remaining cells were resuspended in 100 μL sterile 1X PBS to a concentration of 5χ107 cells per ml. Mice were inoculated subcutaneously in the lower right scapular region with 5 x 106 NCI-H441-shMet-4.11 cells. Tumors were monitored until their average volume reached 200 mm3. Mice were randomized into 4 groups of 10 mice each and treatment was started (summarized in Table 2). Mice in group 1 were treated with anti-porcine control antibody (5 mg/kg, IP, twice a week for 4 weeks) and given 5% sucrose drinking water. The small anti- ragweed control antibody (5 mg/kg, IP, twice a week for 4 weeks) in group 2 was treated with 'doxcycline (丨mg/mL) in 5% sucrose drinking water. . Mice in Group 3 were treated with anti-VEGF antibody (B20-4.1.1, 5 mg/kg, IP, twice a week for 4 weeks) and given 5% sucrose drinking water. The mice in Group 4 were treated with anti-VEGF antibody (B20-4.1.1 '5 mg/kg, IP, twice a week for 4 weeks) and given doxycycline in 5% sucrose drinking water (1 The antibody was continued to be administered for four weeks at mg/mLh, at which time the administration of docecycline was maintained, but antibody administration was stopped. Tumor volume was monitored until day 57. 0 143939.doc -155-201019961 Table 2 NCI-441 NSCLC xenogene Transplanted tumor model te_meti|j^|^ VEGF antibody combination group 1 number / sex 10/F test material sucrose; anti-porcine control antibody pathway drinking water; IP dose frequency arbitrary feeding (whole process); twice weekly x4 Weekly dose (mg/kg) 5 (anti-porcion) Dose concentration (mg/ml) 1.25 (anti-porcelain) Dose volume (μΐ) 100 2 10/F doxycycline; Control antibody drinking water; IP Any feeding (whole process); twice weekly x4 weeks 5 (anti-porcelain) 1.25 (anti-porcelain); 1 (Dox) 100 3 10/F sucrose; anti-VEGF (B20-4.1.1) drinking water; IP arbitrary Food (whole process); twice weekly x4 weeks 5 (anti-VEGF) 1.25 (anti-VEGF) 100 4 10/F doxycycline; anti-VEGF (B20-4.1.1) drinking water; IP Arbitrary feeding (whole process); twice weekly X4 weeks 5 (anti-VEGF) 1.25 (anti-VEGF); 1 (Dox) 100 studies 3 and 4: c-met in the EBC-l-shMet NSCLC xenograft model Knockout combination of anti-VEGF anti-sputum treatment: cultured in growth medium consisting of RPMI 1640 medium (Invitrogen), 2 mM L-branched tyrosine and 1% fetal bovine serum: £801-311^461;-3.15 And £801-3111^161;-4.5 cell strain. To prepare cells for inoculation in mice, the cells were trypsinized and washed with 10 ml of sterile lx phosphate buffered saline (PBS). Dye a subset of counted cells' and resuspend the remaining cells in 100 μl sterile lxPBS to a concentration of 5 x 107 cells per ml. Use 5xl06 EBC-l-shMet-3.1 5 and EBC-l-shMet-4.5 cells in the lower right Mice were subcutaneously inoculated in the scapular region. Tumors were monitored until their average volume reached 300 mm3 °. For EBC-l-shMet-3.15 study and EBC-l-shMet-4.5 study, mice were randomly divided into 2 groups, each group consisting of 10 small The rats 'total 4 groups' and began processing 143939.doc -156-201019961 (summarized in Table 3). The mice in Group 1 had an EBC-l-shMet-3.15 tumor and were given 5% sucrose drinking water. The mice in Group 2 had EBC-1-shMet-3.15 tumors and were administered to doxycycline (1 mg/mL) in 5% sucrose drinking water. The mice in Group 3 had EBC-l-shMet-4.5 tumors and were given 5% sucrose drinking water. The mice in Group 4 had EBC-l-shMet-4.5 tumors and were administered to doxycycline (丨mg/mL) in 53⁄4 sucrose drinking water. The tumor volume was monitored for 21 days.

表3 NCI-441 NSCLC異種移植腫瘤模型中㈣过敲除與抗呢征抗體之組合 組 1 細胞株Table 3 NCI-441 NSCLC xenograft tumor model (4) Combination of knockout and anti-sense antibody group 1 cell line

數目/測試材 性別 辛斗 10/F 途徑 劑量頻率 飲用水任意取食(整 個過程) 10/F多西環飲用水任意取食(整 素 個過程) 3 EBC-l-shMet-4.5 10/F蔗糖飲用水任意取食(整 個過程) 4 EBC-l-shMet·4.5 10/F多西環飲用水任意取食(整 __個過程) 2 EBC-1-shMet- 3.15 EBC-1-shMet- 3.15 蔗糖 劑量 劑量濃度劑量體積 (mg/kg) (mg/ml) (μ〇Number / test material sex 10/F route dose frequency drinking water free feeding (whole process) 10/F Dorset drinking water free feeding (whole process) 3 EBC-l-shMet-4.5 10/F Sucrose drinking water can be eaten freely (the whole process) 4 EBC-l-shMet·4.5 10/F Dorsetian drinking water can be fed ad libitum (complete __ process) 2 EBC-1-shMet- 3.15 EBC-1-shMet- 3.15 sucrose dose dose concentration dose volume (mg/kg) (mg/ml) (μ〇

用對小鼠及人類VEGF(B20-4.1)或單獨人類VEGF(貝伐 單抗)具有選擇性之抗VEGF抗體進一步測試EBC-l-shMet-4.5 NSCLC異種移 植腫瘤 ,以測試人類 或小鼠VEGF對驅動 腫瘤生長之相對貢獻。雖然如上所述接種小鼠,但改為將 小鼠隨機分成3組,每組10隻小鼠,且開始處理(概述於表 4中)。 143939.doc •157- 201019961 表4 小鼠或人類VEGF對EBC-l-shMet-4.5 NSCLC異種移植腫瘤生長 之貢獻的表徵 組 數目/ 性別 測試材料 途徑 劑董頻率 劑量 (mg/kg) 劑量濃度 (mg/ml) 劑量體積 (μΐ) 1 10/F 抗gD對照抗體 (hu5B6) IP 每週兩次x3週 5(抗 gD) 】·25(抗 gD) 100 2 10/F 抗 VEGF (B20-4.1) IP 每週兩次x3週 5(抗小鼠及 人類VEGF) 1.25(抗小鼠及 人類 100 3 10/F 抗 VEGF (貝伐單抗) IP 每週兩次χ3週 5(抗人類 VEGF) 1.25(抗人類 VEGF) 100 研究5 :在EBC-l-shMet-4.5 NSCLC異種移植腫瘤模型 中用c-met敲除、EGFR拮抗劑埃羅替尼及抗VEGF抗逋之 組合處理•·如上所述接種EBC-l-shMet-4.5細胞,將小鼠 隨機分成8組,每組10隻小鼠,且開始處理(概述於表5 中)。初步耐受性研究指示,多西環素與埃羅替尼之組合 可使體重明顯減輕。因此,用飲用水使多西環素之濃度 降低至0.5 mg/mL。此多西環素濃度足以活體内誘導針 對c-met之shRNA。組1中之小鼠(對照組)用抗豬草對照抗 體(5 mg/kg,IP,每週兩次歷時4週)加MCT(l〇〇 μι,PO, QD,χ28天)處理,且給予5%蔗糖飲用水。組2中之小鼠用 對照抗體(5 mg/kg,IP,每週兩次歷時4週)加MCT(100 μί,PO,QD,x28天)處理,且給予於5%蔗糖飲用水中之多 西環素(0.5 mg/mL)。組3中之小鼠用抗VEGF抗體(5 mg/kg, IP,每週兩次歷時4週)加MCT(100 pL,P〇,QD,x28天)處 理,且給予5%蔗糖飲用水。組4中之小鼠用抗豬草對照抗 體(5 mg/kg,IP,每週兩次歷時4週)加埃羅替尼(100 143939.doc -158- 201019961 mg/kg,PO,QD,x28天)處理,且給予5%蔗糖飲用水。組 5中之小鼠用抗VEGF抗體(5 mg/kg,IP ’每週兩次歷時4 週)加MCT(100 μί,P〇,QD,x28天)處理,且給予於5% 蔗糖飲用水中之多西環素(〇·5 mg/mL)。組6中之小鼠用抗 豬草對照抗體(5 mg/kg ’ IP,每週兩次歷時4週)加埃羅替 尼(100 mg/kg,PO,QD,χ28天)處理,且給予於5%嚴糖 飲用水中之多西環素(〇·5 mg/mL)。組7中之小鼠用抗VEGF 抗體(5 mg/kg,IP,每週兩次歷時4週)加埃羅替尼(100 mg/kg,PO,QD,χ28天)處理,且給予5%嚴糖飲用水。 且,組8中之小鼠用抗VEGF抗體(5 mg/kg,IP,每週兩次 歷時4週)加埃羅替尼(100 mg/kg,PO,QD,χ28天)以及於 5%蔗糖飲用水中之多西環素(0.5 mg/mL)處理。 表5 EBC-l-shMet-4.5 NSCLC異種移植腫瘤模型中之c-met敲除 埃羅替尼及抗VEGF抗體之組合 測試材料 途徑 劍量頻率 劑量 (mg/kg) 劑量濃度 (mg/ml) 劑量體積 (μΐ) 抗豬草對照抗體; MCT ;蔗糖 IP ; P0 ; 飲用水 每週兩次x4 週;QD ; 任意取食 5(抗豬草) 1.25(抗 gD) 100 抗豬草抗體; MCT ;多西環素 IP ; PO ; 飲用水 每遇兩次X4 週;QD ; 任意取食 5(抗豬草) 1.25(抗 gD) ; 0.5 (Dox) 100 抗VEGF抗體(B20-4.1.1) ; MCT ; 蔗糖 IP ; PO ; 飲用水 每週兩次X4 週;QD ; 任意取食 5(Β20-4.Π) 1·25(抗 gD) 100 抗豬草抗體;埃羅 替尼;蔗糖 IP ; PO ; 飲用水 每遇兩次X4 遇;QD ; 任意取食 5(抗豬草); 10〇(埃羅替尼) 1.25(抗 gD) ; 25(埃 羅替尼) 100 數目/ ~ 組性別 143939.doc • 159- 201019961 5 6 7 8 10/F 抗VEGF抗體(B20-4.1_1) ; MCT ;多 西環素 IP ; PO ; 飲用水 每週兩次x4 週;QD ; 任意取食 5(B20-4.1.1) 1.25(抗 gD) ; 0.5 (Dox) 100 10/F 抗豬草抗體;埃羅 替尼;多西環素 IP ; PO ; 飲用水 每週兩次χ4 週;QD ; 任意取食 5 (B20-4.1.1); 1〇〇(埃羅替尼) 1.25(抗 gD) ; 25(埃 羅替尼); 0.5 (Dox) 100 10/F 抗VEGF抗體(B20-4.1.1);埃羅替 尼;蔗糖 IP ; PO ; 飲用水 每週兩次χ4 週;QD ; 任意取食 5(B20-4.1.1); 100(埃羅替尼) 1.25(抗 gD) ; 25(埃 羅替尼) 100 10/F 抗VEGF抗體(B20-4.1.1);埃羅替 尼;多西環素 IP ; PO ; 飲用水 每週兩次X4 週;QD ; 任意取食 5(B20-4.1.1); 100(埃羅替尼) 1.25(抗 g〇) ; 25(埃 羅替尼); 0.5 (Dox) 100 研究6 :在NCI-H596 NSCLC異種移植腫瘤模型中用c-met敲除、埃羅替尼及抗VEGF抗餿之組合處理:在由 RPMI 1640 培養基(Invitrogen)、2 mM L-麵醯胺酸及 10% 胎 牛血清組成之生長培養基中培養NCI-H596細胞。為製備供 接種於小鼠中之細胞,使細胞胰蛋白酶化,用1 〇毫升無菌 lx磷酸鹽緩衝生理鹽水(PBS)洗滌。利用錐蟲藍拒染計數 細胞之子集,且將剩餘細胞再懸浮於100 μΐ無菌1 XPBS中 至濃度為每毫升5x106個細胞。使用人類HGF轉殖基因 C3H-SCID小鼠(hu-HGF-Tg-SCID)來模擬旁分泌人類HGF 刺激(Zhang YW 等人,£111^11〇6§1*〇\^1;11〇£&gt;1111111&amp;11&lt;:-11161;-expressing xenografts in a new strain of immunocompromised mice transgenic for human hepatocyte growth factor/scatter factor. Oncogene 24:101-6, 2005)。因為小鼠 HGF 為人類 HGF之不良配位體,故產生此等小鼠,且因而可活體内增 強對HGF起反應之腫瘤細胞株之生長。因為NCI-H596細胞 不能在正常裸或SCID小鼠中形成腫瘤,但能夠在hu-HGF- 143939.doc -160- 201019961EBC-l-shMet-4.5 NSCLC xenograft tumors were further tested with anti-VEGF antibodies selective for mouse and human VEGF (B20-4.1) or human VEGF (bevacizumab) alone to test human or mouse VEGF Relative contribution to driving tumor growth. Although mice were inoculated as described above, mice were instead randomly divided into 3 groups of 10 mice each and treatment was started (summarized in Table 4). 143939.doc •157- 201019961 Table 4 Characterization of the contribution of mouse or human VEGF to EBC-l-shMet-4.5 NSCLC xenograft tumor growth Group number / sex test material route agent Dong frequency dose (mg/kg) dose concentration ( Mg/ml) Dosage volume (μΐ) 1 10/F Anti-gD control antibody (hu5B6) IP twice weekly x3 weeks 5 (anti-gD) 】·25 (anti-gD) 100 2 10/F anti-VEGF (B20-4.1 IP twice weekly x3 weeks 5 (anti-mouse and human VEGF) 1.25 (anti-mouse and human 100 3 10/F anti-VEGF (bevacizumab) IP twice weekly for 3 weeks 5 (anti-human VEGF) 1.25 (anti-human VEGF) 100 Study 5: Combination of c-met knockout, EGFR antagonist erlotinib and anti-VEGF anti-spasm in EBC-l-shMet-4.5 NSCLC xenograft tumor model •· The EBC-l-shMet-4.5 cells were inoculated, and the mice were randomly divided into 8 groups of 10 mice each and started treatment (summarized in Table 5). Preliminary tolerance study indicated that doxycycline and angstrom The combination of rotinib can significantly reduce body weight. Therefore, the concentration of doxycycline is reduced to 0.5 mg/mL with drinking water. This concentration of doxycycline is sufficient for induction in vivo. -met shRNA. The mice in group 1 (control group) were treated with anti-porcine control antibody (5 mg/kg, IP, twice a week for 4 weeks) plus MCT (l〇〇μι, PO, QD, χ28) Day) treated with 5% sucrose drinking water. Group 2 mice were treated with control antibody (5 mg/kg, IP, twice a week for 4 weeks) plus MCT (100 μί, PO, QD, x28 days) Treated and given doxycycline (0.5 mg/mL) in 5% sucrose drinking water. The mice in group 3 were treated with anti-VEGF antibody (5 mg/kg, IP twice a week for 4 weeks) MCT (100 pL, P〇, QD, x28 days) was treated with 5% sucrose drinking water. Group 4 mice were treated with anti-porcine control antibody (5 mg/kg, IP, twice a week for 4 weeks) Treated with erlotinib (100 143939.doc -158-201019961 mg/kg, PO, QD, x28 days) and given 5% sucrose drinking water. Group 5 mice were treated with anti-VEGF antibody (5 mg/ Kg, IP 'twice a week for 4 weeks) plus MCT (100 μί, P〇, QD, x28 days) and given doxycycline (〇·5 mg/mL) in 5% sucrose drinking water The mice in group 6 were treated with anti-porcine control antibody (5 mg/kg 'IP, twice a week) 4 weeks) Jiaai Luo erlotinib (100 mg / kg, PO, QD, χ28 day) treated, and administered in a 5% sugar strict drinking water as much as doxycycline (square · 5 mg / mL). Group 7 mice were treated with anti-VEGF antibody (5 mg/kg, IP, twice a week for 4 weeks) plus erlotinib (100 mg/kg, PO, QD, χ28 days) and given 5% Strict sugar drinking water. Furthermore, mice in group 8 were treated with anti-VEGF antibody (5 mg/kg, IP twice a week for 4 weeks) plus erlotinib (100 mg/kg, PO, QD, χ28 days) and at 5% Treatment with doxycycline (0.5 mg/mL) in sucrose drinking water. Table 5 Combination of c-met knockout erlotinib and anti-VEGF antibody in EBC-l-shMet-4.5 NSCLC xenograft tumor model Test material route Sword frequency dose (mg/kg) Dose concentration (mg/ml) Dosage volume (μΐ) against ragweed control antibody; MCT; sucrose IP; P0; drinking water twice a week for x4 weeks; QD; arbitrary feeding 5 (anti-porcine) 1.25 (anti-gD) 100 anti-porcino antibody; ; doxycycline IP; PO; drinking water every two X4 weeks; QD; arbitrary feeding 5 (anti-porcine) 1.25 (anti-gD); 0.5 (Dox) 100 anti-VEGF antibody (B20-4.1.1) MCT; sucrose IP; PO; drinking water twice a week X4 weeks; QD; arbitrary feeding 5 (Β20-4.Π) 1·25 (anti-gD) 100 anti-porcino antibody; erlotinib; ; PO; drinking water every two X4 encounters; QD; arbitrary feeding 5 (anti-grass); 10 〇 (erlotinib) 1.25 (anti-gD); 25 (erlotinib) 100 number / ~ group Gender 143939.doc • 159- 201019961 5 6 7 8 10/F anti-VEGF antibody (B20-4.1_1); MCT; doxycycline IP; PO; drinking water twice a week x4 weeks; QD; (B20-4.1.1) 1.25 (anti-gD); 0.5 (Dox) 10 0 10/F anti-porcino antibody; erlotinib; doxycycline IP; PO; drinking water twice a week for 4 weeks; QD; any feeding 5 (B20-4.1.1); Rotinib) 1.25 (anti-gD); 25 (erlotinib); 0.5 (Dox) 100 10/F anti-VEGF antibody (B20-4.1.1); erlotinib; sucrose IP; PO; Weeks twice χ 4 weeks; QD; arbitrarily feeding 5 (B20-4.1.1); 100 (erlotinib) 1.25 (anti-gD); 25 (erlotinib) 100 10/F anti-VEGF antibody (B20- 4.1.1); erlotinib; doxycycline IP; PO; drinking water twice a week X4 weeks; QD; arbitrary feeding 5 (B20-4.1.1); 100 (erlotinib) 1.25 ( Anti-g〇); 25 (Errotinib); 0.5 (Dox) 100 Study 6: Combination of c-met knockout, erlotinib and anti-VEGF antispasmodic in NCI-H596 NSCLC xenograft tumor model : NCI-H596 cells were cultured in growth medium consisting of RPMI 1640 medium (Invitrogen), 2 mM L-face valine and 10% fetal bovine serum. To prepare cells for inoculation into mice, the cells were trypsinized and washed with 1 ml of sterile lx phosphate buffered saline (PBS). A subset of cells were counted using trypan blue exclusion and the remaining cells were resuspended in 100 μl sterile 1 X PBS to a concentration of 5 x 106 cells per ml. Human HGF transgenic C3H-SCID mice (hu-HGF-Tg-SCID) were used to mimic paracrine human HGF stimulation (Zhang YW et al., £111^11〇6§1*〇\^1;11〇£ &gt;1111111&amp;11&lt;:-11161;-expressing xenografts in a new strain of immunocompromised mice transgenic for human hepatocyte growth factor/scatter factor. Oncogene 24:101-6, 2005). Since mouse HGF is a poor ligand for human HGF, these mice are produced, and thus the growth of tumor cell lines that respond to HGF can be enhanced in vivo. Because NCI-H596 cells cannot form tumors in normal nude or SCID mice, but can be in hu-HGF- 143939.doc -160- 201019961

Tg-SCID小鼠中形成腫瘤,故此模型表示HGF旁分泌驅動 之腫瘤模型。對動物右側剃毛後,用0.5xl06個NCI-H596 細胞在右下肩胛區中皮下接種NCI-H596細胞。監測腫瘤直 至其平均體積達到100 mm3。Tumors are formed in Tg-SCID mice, so this model represents a tumor model driven by HGF paracrine. After shaving the right side of the animals, NCI-H596 cells were inoculated subcutaneously in the lower right scapular region with 0.5 x 106 NCI-H596 cells. The tumors were monitored until their average volume reached 100 mm3.

將小鼠隨機分成9組,每組10隻小鼠,且開始處理(概述 於表6中)。組1 (未處理組)中之小鼠含有帶腫瘤小鼠,雖然 未處理該等小鼠,但與經處理組並行監測其腫瘤。組2(對 照組)中之小鼠用抗豬草對照抗體(5 mg/kg,IP,每週兩次 歷時 12週)加 MCT(100 μι,PO,QD,x84 天)及 MetMAb緩 衝液(100 μΐ^,IP,每三週一次χ4)處理。組3中之小鼠用對 照抗體(5 mg/kg,IP,每週兩次歷時12週)加MCT(100 μι,PO,QD,x84 天)及 MetMAb(30 mg/kg,IP,每三週一 次X4)處理。組4中之小鼠用抗VEGF抗體(B20-4.1,5 mg/kg,IP,每週兩次歷時 12 週)加 MCT(100 pL,PO, QD,x84天)及MetMAb緩衝液(100 pL,IP,每三週一次χ4) 處理。組5中之小鼠用抗豬草對照抗體(5 mg/kg,IP,每週 兩次歷時12週)加埃羅替尼(100 mg/kg,PO,QD,x84天) 及MetMAb緩衝液(100 pL,IP,每三週一次x4)處理。組6 中之小鼠用抗VEGF抗體(B20-4.1,5 mg/kg,IP,每週兩 次歷時 12 週)加 MCT(100 μί,PO,QD,x84 天)及 MetMAb(30 mg/kg,IP,每三週一次χ4)處理。組7中之小 鼠用抗豬草對照抗體(5 mg/kg,IP,每週兩次歷時12週)加 埃羅替尼(100 mg/kg,PO,QD,x84 天)及 MetMAb(30 mg/kg,IP,每三週一次χ4)處理。組8中之小鼠用抗VEGF 143939.doc •161- 201019961 抗體(B20-4.1,5 mg/kg,IP,每週兩次歷時12週)加埃羅 替尼(100 mg/kg,PO,QD,χ84 天)及 MetMAb緩衝液(100 μΐ^ ’ IP,每三週一次x4)處理。組9中之小鼠用抗VEGF抗 體(B20-4.1,5 mg/kg,IP,每週兩次歷時12週)加埃羅替 尼(100 mg/kg,PO,QD,χ84 天)及 MetMAb(30 mg/kg, IP,每三週一次x4)處理。 表6 在hu-HGF-Tg-C3H-SCID小鼠中生長之NCI-H596 NSCLC異種移植 腫瘤模型中MetMAb、埃羅替尼及抗VEGF抗體的組合 數目/ 測試材料 劑量濃度 劑量體積 組 性別 途徑 劑量頻率 劑量(mg/kg) (mg/ml) (μΐ) 1 10/F 未處理 - - _ - - 2 10/F 抗豬草抗體; IP ; 每週兩次χ12 5(抗豬草) 1·25(抗豬草) 100 MCT ; MetMAb PO ; 週;QDx84 緩衝液 IP 天;Q3Wx4 3 10/F 抗豬草抗體; IP ; 每週兩次xl2 5(抗豬草); 1.25(抗豬草); 100 MCT ; MetMAb PO ; IP 週;QDx84 天;Q3Wx4 30 (MetMAb) 7.5 (MetMAb) 4 10/F 抗VEGF抗體 IP ; 每週兩次xl2 5(抗VEGF) 1.25(抗 VEGF) 100 (B20-4.1); PO ; 週;QDx84 MCT ; MetMAb IP 天;Q3Wx4 緩衝液 5 10/F 抗豬草抗體;埃 IP ; 每週兩次xl2 5(抗豬草); 1.25(抗豬草); 100 羅替尼; PO ; 週;QDx84 100(埃羅替尼) 25(埃羅替尼) MetMAb緩衝液 IP 天;Q3Wx4 6 10/F 抗VEGF抗體 IP ; 每週兩次X12 5(抗VEGF); 1.25(抗 100 (B20-4.1); PO ; 週;QDx84 30 (MetMAb) VEGF) ; 7.5 MCT ; MetMAb IP 天;Q3Wx4 (MetMAb) 7 10/F 抗豬草抗體;埃 IP ; 每週兩次xl2 5(抗豬草); 1.25(抗豬草); 100 羅替尼; PO ; 週;QDx84 1〇〇(埃羅替 25(埃羅替尼); MetMAb IP 天;Q3Wx4 尼;);30 (MetMAb) 7.5 (MetMAb) 8 10/F 抗VEGF抗體 IP ; 每遇兩次xl2 5(抗VEGF); 1.25(抗 100 (B20-4.1);埃羅 PO ; 遇;QDx84 1〇〇(埃羅替尼)VEGF) ; 25(埃 替尼;MetMAb IP 天;Q3Wx4 羅替尼) 緩衝液 9 10/F 抗VEGF抗體 IP ; 每週兩次X12 5(抗VEGF); 1.25(抗 100 (B2(M.l);埃羅 PO ; 週;QDx84 1〇〇(埃羅替 VEGF) ; 25(埃 替尼;AetMAb IP 天;Q3Wx4 尼);30 羅替尼);7.5 (MetMAb) (MetMAb) -162- 143939.doc 201019961 對於此處所報導之所有研究,若腫瘤達到2000 mm3以上 或腫瘤顯示壞死病變跡象,則取消對該等動物之研究。若 任何既定組中超過50%之動物的研究已取消,則停止對該 組之處理且取消對所有動物之研究。對小鼠之所有研究及 處理均遵循實驗動物管理與使用委員會(Institutional Animal Care and Use Committee,IACUC)指南。 廑痨及浚# f琢··使用UltraCal-IV測徑規(型號54-10-111,Fred V· Fowler Company, Inc. ; Newton, ΜΑ)量測踵 瘤體積之兩個尺寸(長度及寬度)。由Excel vl 1.2 (Microsoft Corporation ; Redmond, WA)使用下式計算腫瘤體積: 腫瘤體積(mm3)=(長度x寬度2)x0.5 必我 f 齊分术··使用 KaleidaGraph 3.6(Synergy Software ; Reading, PA)作腫瘤抑制曲線圖。如下計算生長抑制百分 比(%Inh): %Ihn=100x(l-[腫瘤大小(經處理)/腫瘤大小(媒劑)]) 腫瘤發病率係由研究結束時各組中可量測腫瘤之數目來 確定。部分退化(PR)既定為研究期間任一天,大於50%但 小於100%之起始腫瘤體積之腫瘤退化。完全退化(CR)既 定為研究期間任一天,1〇〇%之最初起始腫瘤體積之腫瘤 退化。 使用 JMP軟體 5· 1.2版(SAS Institute ; Cary,NC)來計算平 均腫瘤體積及平均值之標準誤差(SEM)。亦使用JMP軟體 5.1.2版進行資料分析且使用司徒頓t檢驗(Student's t-test) 或杜氏t檢驗(Dunnett'st-test)產生p值。 143939.doc • 163· 201019961 結果 使用圖1中描繪之細胞株,藉由活體外或活體内用hgf 處理(hu-HGF-Tg-SCID)且藉由抑制c-met(使用抗c-met抗體 MetMAb或針對c-met之shRNA)來研究c-met功能。此等細 胞株為配位體誘導(NCI-H596及KP4)或非配位體依賴性 (NCI-H441及EBC-l)c-met活性之代表模型,基於c-met活 化模式加以選擇且包括旁分泌型(NSCLC細胞株NCI-H596)、自分泌型(胰腺細胞株KP4)、c-met過度表現型 (NSCLC細胞株NCI-H441)及局部擴增且過度表現c-met型 (NSCLC細胞株EBC-1)。c-met蛋白之相對表現顯示於圖1B 中。此等結果表明配位體反應性細胞株KP4及NCI H596表 現之c-met之含量遠低於非配位體依賴性活化之細胞株。 EBC-1細胞株表現最高含量之c-met。KP4與NCI-H596為配 位體反應性(圖1C),且此反應受MetMAb之處理抑制。KP4 為自分泌型細胞株,且顯示在無其他HGF存在下經 MetMAb 處理後,p-c-met 減少。為 NCI-H441(圖 1D)及 EBC-1(圖1E)產生表現多西環素可誘導之乾向c-met mRN A之 shRNA的穩定細胞株。雖然在經多西環素處理後4、5或7 天,&gt;1(^1-11441-311^461細胞株3.1及3.11顯示〇-11161;及?-〇11^1 顯著減少,但在對照NCI-H441-shGFPl細胞株中未觀察到 磷酸基-c-met表現之變化(圖1D)。在經多西環素處理後1或 2天,EBC-l-shMet-3.5及4.12細胞株顯示c-met表現之完全 阻斷,而在該相同時間範圍内,EBC-l-shMet-4.5僅顯示c-met表現之部分阻斷(圖1E)。用多西環素處理對照EBC-1- 143939.doc 201019961 shGFP2對c-met表現無影響。 研究NSCLC細胞株中活化c-met或破壞活性c-met信號傳 導之效應。首先,用HGF處理配位體依賴性細胞株(諸如 NCI-H596及HOP92)6小時,且由微陣列分析基因表現。用 HGF處理配位體依賴性細胞株會引起數種血管生成因子 : (包括介白素8(IL8)、血管内皮細胞生長因子A(VEGFA)、 EPH受體A2(EphA2)、血管生成素樣蛋白4(Angptl4)及 Ephrin B2(EFNB2))之基因表現的顯著上調(圖2A)。其次, ® 在多西環素存在或不存在下處理非配位體依賴性NSCLC細 胞株 EBC-l-shMet-3.15 及 EBC-l-shMet-4.12 以減少 c-met含 量,從而評估視活性c-met信號傳導而定之基因表現。觀 察到數種血管生成因子(包括介白素8(IL8)、血管内皮細胞 生長因子A(VEGFA)、EPH受體A2(EphA2)、血管生成素樣 蛋白4(Angptl4)及Ephrin B2(EFNB2))之表現降低,表明在 非配位體依賴性EBC-1細胞中,此等因子之表現視c-met信 泰 號傳導而定(圖2B)。此等結果指示活化c-met會活化一組血 管生成基因。c-met活化可藉由正調促血管生成分子而間 接促進血管生成。 . 因為在配位體依賴性與非配位體依賴性NSCLC細胞株 中,c-met活化均引起VEGF及IL8表現上調,故在細胞培 養物以及異種移植腫瘤中研究VEGF及IL8蛋白表現含量。 用多西環素處理EBC-l-shMet-3.15 及 EBC-l-shMet-4_12 細 胞株以誘導c-met敲除,從而在處理後1天與2天時引起分 泌型VEGF蛋白含量顯著減少,而用多西環素處理對照 143939.doc -165- 201019961 EBC-l-shGFP2細胞株對VEGF含量無影響(圖3A)。在由 EBC-l-shMet-3.15、EBC-l-shMet-4.12 及 EBC-l-shGFP2 細 胞株形成之異種移植腫瘤中,IL8蛋白含量觀察到類似效 應(圖3D)。用HGF處理配位體依賴性細胞株NCI-H596亦引 起VEGF(圖3B)及IL8(圖3E)蛋白含量顯著減少。此外,在 培養物中,c-met敲除會引起用多西環素處理之NCI-H441- shMet-3.ll 及 NCI-H441-shMet-3.1 細胞株(但無 NCI-H441- shGFPl細胞株)之VEGF蛋白含量顯著減少,然而該效應僅 在用多西環素處理4天後才明顯(圖3C)。因為在騰腺(KP4) 細胞中HGF處理不會增加VEGF或IL8含量,故該效應可能 為NSCLC細胞株所特有。此等結果表明,在NSCLC細胞株 中VEGF及IL8表現為Met活化之常見結果且在NSCLC細胞 株中c-met抑制會降低VEGF與IL8之表現。 為測試c-met與VEGF抑制劑之組合是否產生較佳抗腫瘤 活性’使用HGF/Met自分泌型KP4胰腺腫瘤模型來評估c_ met括抗劑抗體MetMAb與抗VEGF抗體之纟且合處理。基於 劑量範圍研究選擇MetMAb之劑量,該等研究基於每三週 給藥一次,定義2.5 mg/kg為次治療劑量,7 5 mg/kg為最 低有效劑量(MED)(對應於50%有效劑量(ΕΕ&gt;5〇)),且3〇 mg/kg為最佳有效劑量。以推薦劑量及時程(5 mg/kg,每 週兩次)利用結合小鼠與人類VEGF之抗VEGF抗體(B20-4.1),且單獨投與抗VEGF抗體會部分抑制κρ4異種移植腫 瘤生長(約22%)(圖4)。單獨投與,當以3〇 mg/kg 投與時,顯示良好的踵瘤抑制(約65%);當以7·5 111§/1^投 143939.doc -166- 201019961 與時,部分抑制腫瘤生長(約24%);且當以2.5 mg/kg投與 時,不抑制腫瘤生長。Mice were randomized into 9 groups of 10 mice each and treatment was started (summarized in Table 6). Mice in group 1 (untreated group) contained tumor-bearing mice, and although the mice were not treated, tumors were monitored in parallel with the treated group. Mice in group 2 (control group) were treated with anti-porcine control antibody (5 mg/kg, IP twice a week for 12 weeks) plus MCT (100 μιη, PO, QD, x84 days) and MetMAb buffer ( 100 μΐ^, IP, once every three weeks χ 4) treatment. Group 3 mice were treated with control antibody (5 mg/kg, IP, twice a week for 12 weeks) plus MCT (100 μιη, PO, QD, x84 days) and MetMAb (30 mg/kg, IP, every three) Once a week X4) processing. Group 4 mice were treated with anti-VEGF antibody (B20-4.1, 5 mg/kg, IP, twice a week for 12 weeks) plus MCT (100 pL, PO, QD, x84 days) and MetMAb buffer (100 pL) , IP, once every three weeks χ 4) processing. Group 5 mice were treated with anti-porcine control antibody (5 mg/kg, IP twice a week for 12 weeks) plus erlotinib (100 mg/kg, PO, QD, x84 days) and MetMAb buffer (100 pL, IP, x4 every three weeks). Group 6 mice were treated with anti-VEGF antibody (B20-4.1, 5 mg/kg, IP, twice a week for 12 weeks) plus MCT (100 μί, PO, QD, x84 days) and MetMAb (30 mg/kg) , IP, once every three weeks χ 4) processing. Group 7 mice were treated with ragweed control antibody (5 mg/kg, IP twice a week for 12 weeks) plus erlotinib (100 mg/kg, PO, QD, x84 days) and MetMAb (30 Mg/kg, IP, once every three weeks χ 4) treatment. Group 8 mice were treated with anti-VEGF 143939.doc •161- 201019961 antibody (B20-4.1, 5 mg/kg, IP, twice a week for 12 weeks) plus erlotinib (100 mg/kg, PO, QD, χ84 days) and MetMAb buffer (100 μΐ^ 'IP, x4 every three weeks). Group 9 mice were treated with anti-VEGF antibody (B20-4.1, 5 mg/kg, IP, twice a week for 12 weeks) plus erlotinib (100 mg/kg, PO, QD, χ84 days) and MetMAb (30 mg/kg, IP, x4 every three weeks). Table 6 Number of combinations of MetMAb, erlotinib and anti-VEGF antibodies in NCI-H596 NSCLC xenograft tumor models grown in hu-HGF-Tg-C3H-SCID mice / test material dose concentration dose volume group sex route dose Frequency dose (mg/kg) (mg/ml) (μΐ) 1 10/F untreated - - _ - - 2 10/F anti-porcino antibody; IP; twice a week χ 12 5 (anti-porcine) 1· 25 (anti-porcelain) 100 MCT; MetMAb PO; week; QDx84 buffer IP days; Q3Wx4 3 10/F anti-porcino antibody; IP; twice weekly xl2 5 (anti-porcelain); 1.25 (anti-porcelain) 100 MCT; MetMAb PO; IP week; QDx84 days; Q3Wx4 30 (MetMAb) 7.5 (MetMAb) 4 10/F anti-VEGF antibody IP; twice weekly xl2 5 (anti-VEGF) 1.25 (anti-VEGF) 100 (B20- 4.1); PO; week; QDx84 MCT; MetMAb IP day; Q3Wx4 buffer 5 10/F anti-porcino antibody; Ai IP; twice weekly xl2 5 (anti-porcelain); 1.25 (anti-porcelain); TIN; PO; week; QDx84 100 (erlotinib) 25 (erlotinib) MetMAb buffer IP days; Q3Wx4 6 10/F anti-VEGF antibody IP; Twice X12 5 (anti-VEGF); 1.25 (anti-100 (B20-4.1); PO; week; QDx84 30 (MetMAb) VEGF); 7.5 MCT; MetMAb IP day; Q3Wx4 (MetMAb) 7 10/F anti-porcine antibody埃IP; twice a week xl2 5 (anti-porcelain); 1.25 (anti-porcelain); 100 rotinib; PO; week; QDx84 1〇〇 (erlotidine 25 (erlotinib); MetMAb IP Day; Q3Wx4 Ni;); 30 (MetMAb) 7.5 (MetMAb) 8 10/F anti-VEGF antibody IP; every two times xl2 5 (anti-VEGF); 1.25 (anti-100 (B20-4.1); Ero PO; ; QDx84 1 〇〇 (erlotinib) VEGF); 25 (Etinib; MetMAb IP days; Q3Wx4 rotinib) Buffer 9 10/F anti-VEGF antibody IP; twice weekly X12 5 (anti-VEGF) 1.25 (anti-100 (B2 (Ml); Ero PO; week; QDx84 1 〇〇 (Errot VEGF); 25 (Etinib; AetMAb IP days; Q3Wx4 Ni); 30 rotinib); 7.5 ( MetMAb) (MetMAb) -162- 143939.doc 201019961 For all studies reported here, if the tumor reached 2000 mm3 or more or the tumor showed signs of necrotic lesions, the study of the animals was cancelled. If more than 50% of the animals in any given group have been canceled, the treatment of the group is stopped and the study of all animals is cancelled. All studies and treatments in mice follow the guidelines of the Institutional Animal Care and Use Committee (IACUC).廑痨和浚# f琢··Measure the two dimensions (length and width) of the tumor volume using the UltraCal-IV caliper (model 54-10-111, Fred V· Fowler Company, Inc.; Newton, ΜΑ) ). Tumor volume was calculated by Excel vl 1.2 (Microsoft Corporation; Redmond, WA) using the following formula: Tumor volume (mm3) = (length x width 2) x 0.5 must be f-segmentation · use KaleidaGraph 3.6 (Synergy Software; Reading , PA) for tumor inhibition curve. The percentage of growth inhibition (%Inh) was calculated as follows: %Ihn=100x (l-[tumor size (treated)/tumor size (vehicle))) The incidence of tumors was the number of tumors that could be measured in each group at the end of the study. to make sure. Partial degeneration (PR) is defined as tumor regression of more than 50% but less than 100% of the initial tumor volume on any day during the study. Complete degeneration (CR) is defined as the tumor regression of 1% of the initial starting tumor volume on any day during the study period. JMP software version 5.1.2 (SAS Institute; Cary, NC) was used to calculate the mean tumor volume and the standard error of the mean (SEM). Data analysis was also performed using JMP software version 5.1.2 and p values were generated using Student's t-test or Dunnett's test. 143939.doc • 163· 201019961 Results using the cell line depicted in Figure 1, by treatment with hgf (hu-HGF-Tg-SCID) in vitro or in vivo and by inhibition of c-met (using anti-c-met antibody MetMAb or shRNA for c-met) to study c-met function. These cell lines are representative models of ligand-induced (NCI-H596 and KP4) or non-ligand-dependent (NCI-H441 and EBC-1) c-met activities, selected based on the c-met activation pattern and included Paracrine (NSCLC cell line NCI-H596), autocrine (pancreatic cell line KP4), c-met overexpression (NSCLC cell line NCI-H441) and locally amplified and overexpressed c-met type (NSCLC cells) Strain EBC-1). The relative performance of the c-met protein is shown in Figure 1B. These results indicate that the ligand-reactive cell lines KP4 and NCI H596 exhibit a much lower level of c-met than non-ligand-dependently activated cell lines. The EBC-1 cell line exhibited the highest level of c-met. KP4 and NCI-H596 are ligand-reactive (Fig. 1C), and this reaction is inhibited by treatment with MetMAb. KP4 is an autocrine cell line and shows a decrease in p-c-met after treatment with MetMAb in the absence of other HGF. A stable cell line expressing dasicyclin-inducible shRNA to c-met mRN A was generated for NCI-H441 (Fig. 1D) and EBC-1 (Fig. 1E). Although 4, 5 or 7 days after treatment with doxycycline, &gt;1 (^1-11441-311^461 cell lines 3.1 and 3.11 showed a significant decrease in 〇-11161; and ?-〇11^1, but in No change in phospho-c-met expression was observed in the control NCI-H441-shGFP1 cell line (Fig. 1D). EBC-l-shMet-3.5 and 4.12 cell lines were treated 1 or 2 days after doxycycline treatment. A complete blockade of c-met performance was shown, and within this same time frame, EBC-l-shMet-4.5 only showed partial blockade of c-met performance (Fig. 1E). Treatment of control EBC-1 with doxycycline - 143939.doc 201019961 shGFP2 has no effect on c-met performance. To study the effect of activating c-met or disrupting active c-met signaling in NSCLC cell lines. First, treatment of ligand-dependent cell lines (such as NCI-) with HGF. H596 and HOP92) were expressed for 6 hours and analyzed by microarray analysis. Treatment of ligand-dependent cell lines with HGF caused several angiogenic factors: (including interleukin 8 (IL8), vascular endothelial growth factor A ( The gene expression of VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and Ephrin B2 (EFNB2) was significantly upregulated (Fig. 2A). Secondly, ® was more Treatment of non-ligand-dependent NSCLC cell lines EBC-l-shMet-3.15 and EBC-l-shMet-4.12 in the presence or absence of cyclin to reduce c-met content, thereby assessing visually active c-met signaling Gene expression. Several angiogenic factors (including interleukin 8 (IL8), vascular endothelial growth factor A (VEGFA), EPH receptor A2 (EphA2), angiopoietin-like protein 4 (Angptl4) and Ephrin B2 were observed. The decrease in performance (EFNB2)) indicates that in non-ligand-dependent EBC-1 cells, the expression of these factors is dependent on c-metxin transmission (Fig. 2B). These results indicate activation of c-met It activates a group of angiogenic genes. c-met activation can indirectly promote angiogenesis by positively regulating angiogenic molecules. Because of c-met activation in ligand-dependent and non-ligand-dependent NSCLC cell lines. Both of them caused up-regulation of VEGF and IL8, so the expression levels of VEGF and IL8 protein were studied in cell culture and xenograft tumors. EBC-l-shMet-3.15 and EBC-l-shMet-4_12 cell lines were treated with doxycycline. Inducing c-met knockdown, resulting in secreted VEG at 1 and 2 days after treatment The F protein content was significantly reduced, while the control 143939.doc-165-201019961 EBC-l-shGFP2 cell line treated with doxycycline had no effect on VEGF content (Fig. 3A). A similar effect was observed in IL8 protein content in xenograft tumors formed from EBC-l-shMet-3.15, EBC-l-shMet-4.12 and EBC-l-shGFP2 cell lines (Fig. 3D). Treatment of the ligand-dependent cell line NCI-H596 with HGF also caused a significant decrease in protein content of VEGF (Fig. 3B) and IL8 (Fig. 3E). In addition, in culture, c-met knockout caused NCI-H441-shMet-3.ll and NCI-H441-shMet-3.1 cell lines treated with doxycycline (but no NCI-H441-shGFP1 cell line) The VEGF protein content was significantly reduced, however this effect was only apparent after 4 days of treatment with doxycycline (Fig. 3C). Since HGF treatment does not increase VEGF or IL8 levels in adeno-gland (KP4) cells, this effect may be unique to NSCLC cell lines. These results indicate that VEGF and IL8 are common results of Met activation in NSCLC cell lines and that c-met inhibition in NSCLC cell lines reduces the expression of VEGF and IL8. To test whether the combination of c-met and VEGF inhibitor produced better anti-tumor activity, the HGF/Met autocrine KP4 pancreatic tumor model was used to evaluate the treatment of the c_ met antagonist antibody MetMAb and the anti-VEGF antibody. The dose of MetMAb was selected based on the dose range study, which was based on administration once every three weeks, defining a therapeutic dose of 2.5 mg/kg and a minimum effective dose (MED) of 75 mg/kg (corresponding to a 50% effective dose (ΕΕ&gt ; 5 〇)), and 3 〇 mg / kg is the best effective dose. Anti-VEGF antibody (B20-4.1) binding to mouse and human VEGF was used at the recommended dose and time course (5 mg/kg twice a week), and administration of anti-VEGF antibody alone partially inhibited κρ4 xenograft tumor growth (about 22%) (Figure 4). When administered alone, when administered at 3〇mg/kg, it showed good tumor suppression (about 65%); when it was 7.5939.doc -166-201019961 with 7·5 111§/1^, partial inhibition Tumor growth (about 24%); and when administered at 2.5 mg/kg, did not inhibit tumor growth.

MetMAb與抗VEGF抗體之組合之功效超過疊加功效(對 次治療劑量之MetMAb或MED之MetMAb而言,分別為約 81 %及約83%)(圖4)。與單獨用MetMAb或抗VEGF處理之組 相比,在組合組内觀察到部分反應(PR)及完全反應(CR)之 數目增加。具體言之,抗VEGF抗體加次治療劑量之 MetMAb(2.5 mg/kg)產生3個PR及1個CR,而單獨各藥劑無 PR或CR。抗VEGF抗體加7.5 mg/kg之MetMAb產生5個PR 及2個CR,而單獨7.5 mg/kg之MetMAb產生5個PR及1個 CR。 此等結果指示次治療劑量或最低有效劑量之MetMAb增 強抗VEGF抗體之抗腫瘤效應,從而產生超過單獨處理之 腫瘤抑制。在人類胰腺腫瘤模型中,VEGF及c-met抑制劑 可顯著改良彼此之活性。 利用NCI-H441-shMet-3.ll NSCLC異種移植模型進一步 研究非配位體(HGF)依賴性細胞株中c-met與VEGF抑制劑 之組合的效應。經由飲用水給予小鼠多西環素,藉此阻斷 活體内c-met表現。Dox處理會誘導針對c-met之shRNA表 現,從而降低c-met之表現。單獨用Dox處理會部分抑制腫 瘤生長(圖5)。單獨用抗VEGF抗體處理動物亦會部分抑制 NCI-H441-shMet-3.ll腫瘤生長(圖5),表明此等腫瘤之生 長及存活部分視VEGF而定。c-met敲除與VEGF抗體處理 之組合顯著改良抗腫瘤活性,從而使得腫瘤停滯(圖5)。與 143939.doc -167· 201019961 單獨用Dox或抗VEGF處理之組相比,在組合處理組中觀察 到PR及CR數目增加。具趙言之,觀察到Dox加抗VEGF抗 體處理組具有1個PR及2個CR,而對照物處理組、D〇x處理 組或抗VEGF抗體處理組無PR及具有1個CR。此等結果表 明,在人類NSCLC腫瘤模型中,VEGF及c-met抑制劑可顯 著改良彼此之活性。 為進一步研究c-met敲除對腫瘤生長之影響,產生EBC-1-shMet NSCLC細胞株(圖 1)。一些純系如同EBC-1-shMet-3.15—樣完全敲除c-met,而其他純系如同EBC-l-shMet-4.5—樣僅部分敲除c-met(圖1;亦參見共同擁有之同在申 請中之美國專利公開案第2009/0226443號)。當此等細胞株 在裸小鼠中生長為異種移植腫瘤時,用多西環素處理小鼠 (經由飲用水)引起反映c-met敲除含量之腫瘤抑制,其中在 EBC-l-shMet-3.15純系中c-met之完全敲除引起腫瘤退化, 而在EBC-lshMet-4.5純系中c-met之部分敲除引起腫瘤生長 延遲(圖6A)。此等結果表明EBC-1細胞株之腫瘤生長及存 活高度視c-met表現而定。EBC-l-shMet-4.5純系為研究其 他治療劑與shRNA介導之c-met敲除組合之活性的理想模 型。 腫瘤血管生成可受由腫瘤或在腫瘤異種移植之情況下由 宿主提供之VEGF影響。EBC-1-shMet細胞株以c-met依賴 性方式表現VEGF(圖2及3)。為確定c-met依賴性VEGF產生 是否為腫瘤進展所需,用單獨結合人類VEGF之抗VEGF抗 體(貝伐單抗,AVASTIN®)或結合人類及小鼠VEGF之抗 143939.doc -168- 201019961 VEGF 抗體(B20-4.1)處理帶 EBC-l-shMet-3.15腫瘤小鼠。 在EBC-l-shMet-3.15 NSCLC異種移植物中,人類抗 VEGF抗艘 AVASTIN®(Genentech,Inc.,South San Francisco, CA)及人類與小鼠可交叉反應性抗體B20-4.1對VEGF之抑 制具有同等功效,指示功能性VEGF在EBC-1模型中為腫瘤 細胞衍生的(圖6B)。此外’ c-met抑制活性之機制不僅僅限 於c-met對VEGF表現之誘導,因為在EBC-l-shMet-3.15異 種移植腫瘤模型中敲除c-met會引起腫瘤退化(圖6A),而抗 VEGF抗體處理會引起腫瘤停滯(圖6B)。 測試c-met敲除與用埃羅替尼及抗VEGF抗體處理組合之 效應,並與用兩兩組合(double combination)處理及單獨用 各藥劑處理作比較。先前研究已顯示當。&quot;^“抑制劑與 EGFR抑制劑組合時,c-met抑制劑顯示顯著增強之活性(同 在申請中之共同擁有之美國專利公開案第2009/0226443號 及第 2009/0226455號)。 c-met敲除、埃羅替尼及抗VEGF抗體之組合產生即時且 持續之腫瘤反應(-92。/。;圖7) ’在此實驗中所測試之任何 方案中均為最大功效。與單獨任一治.療方案相比’ c-met 敲除與抗VEGF抗體處理或埃羅替尼處理之組合亦引起抗 腫瘤活性顯著增加(分別為_77%及-82% ;圖7)。 用適當對照物(蔗糖、MCT及抗猪草抗體;空心圓)處理 帶EBC-l-shMet-4.5腫瘤小鼠不會產生顯著抑制。同樣’ 單獨埃羅替尼對踵瘤生長顯示最小影響(正三角形)(_9〇/〇 ; 圖7)。與以上所示之研究一致,c_met敲除(實心正方形)及 143939.doc -169- 201019961 用抗VEGF抗體處理(倒三角形)會引起腫瘤生長延遲或停滯 (分別為-59%及-70% ;圖7)。相反’埃羅替尼與抗VEGF抗 體之組合與單獨抗VEGF抗體所觀察到之功效相比未顯示 改良(-61% ;圖7)。 c-met敲除與抗VEGF抗體處理之組合亦引起部分反應 (PR)及完全反應(CR)之數目增加:組合處理產生4個PR及1 個CR,而單獨c-met敲除產生2個PR且無CR,且單獨用抗 VEGF抗體處理產生1個PR且無CR 0埃羅替尼加c-met敲除 及抗VEGF抗體方案使反應數目增加至7個PR。 測試用抗c-met抗體MetMAb、埃羅替尼及抗VEGF抗體 處理之效應,並與單獨用各藥劑處理或用兩兩組合處理作 比較。在hu-HGF-Tg-SCID小鼠中產生NSCLC NCI-H596異 種移植物,且用MetMAb、抗VEGF抗體(B20-4· 1)及/或埃 羅替尼處理。The efficacy of the combination of MetMAb and anti-VEGF antibody exceeded the additive efficacy (about 81% and about 83%, respectively, for the sub-therapeutic dose of MetMAb or MED MetMAb) (Figure 4). An increase in the number of partial (PR) and complete (CR) reactions was observed in the combination group compared to the group treated with MetMAb or anti-VEGF alone. Specifically, the anti-VEGF antibody plus a therapeutic dose of MetMAb (2.5 mg/kg) produced 3 PRs and 1 CR, whereas each agent alone did not have PR or CR. Anti-VEGF antibody plus 7.5 mg/kg of MetMAb produced 5 PR and 2 CR, while 7.5 mg/kg of MetMAb alone produced 5 PR and 1 CR. These results indicate that the sub-therapeutic dose or the lowest effective dose of MetMAb enhances the anti-tumor effect of the anti-VEGF antibody, resulting in tumor suppression over treatment alone. In human pancreatic tumor models, VEGF and c-met inhibitors significantly improve each other's activity. The effect of the combination of c-met and VEGF inhibitors in non-ligand (HGF)-dependent cell lines was further investigated using the NCI-H441-shMet-3.ll NSCLC xenograft model. The mice were given doxycycline via drinking water, thereby blocking c-met performance in vivo. Dox treatment induces shRNA expression against c-met, thereby reducing the performance of c-met. Treatment with Dox alone partially inhibited tumor growth (Figure 5). Treatment of animals with anti-VEGF antibodies alone also partially inhibited NCI-H441-shMet-3.ll tumor growth (Fig. 5), indicating that the growth and survival of these tumors depend on VEGF. The combination of c-met knockdown and VEGF antibody treatment significantly improved anti-tumor activity, resulting in tumor stagnation (Figure 5). An increase in the number of PR and CR was observed in the combination treatment group compared to the group treated with Dox or anti-VEGF alone at 143939.doc-167. 201019961. With Zhao Yanzhi, it was observed that the Dox plus anti-VEGF antibody treatment group had 1 PR and 2 CR, while the control treatment group, D〇x treatment group or anti-VEGF antibody treatment group had no PR and had 1 CR. These results indicate that VEGF and c-met inhibitors significantly improve each other's activity in a human NSCLC tumor model. To further investigate the effect of c-met knockdown on tumor growth, EBC-1-shMet NSCLC cell lines were generated (Fig. 1). Some pure lines are completely knocked out of c-met like EBC-1-shMet-3.15, while other pure lines are only partially knocked out of c-met like EBC-l-shMet-4.5 (Fig. 1; see also co-owned U.S. Patent Publication No. 2009/0226443, filed. When these cell lines were grown as xenograft tumors in nude mice, treatment of mice with doxycycline (via drinking water) caused tumor suppression reflecting c-met knockout levels, in EBC-l-shMet- 3.15 Complete knockout of c-met in the pure line caused tumor regression, whereas partial knockdown of c-met in the EBC-lshMet-4.5 pure line caused a delay in tumor growth (Fig. 6A). These results indicate that tumor growth and survival of EBC-1 cell lines are highly dependent on c-met performance. EBC-l-shMet-4.5 pure is the ideal model for studying the activity of other therapeutic agents in combination with shRNA-mediated c-met knockout. Tumor angiogenesis can be affected by VEGF provided by the tumor or by the host in the case of tumor xenografts. The EBC-1-shMet cell line expresses VEGF in a c-met-dependent manner (Figures 2 and 3). To determine whether c-met-dependent VEGF production is required for tumor progression, anti-VEGF antibodies (bevacizumab, AVASTIN®) that bind to human VEGF alone or bind human and mouse VEGF to 143939.doc -168- 201019961 VEGF antibody (B20-4.1) was treated with EBC-l-shMet-3.15 tumor mice. Inhibition of VEGF by human anti-VEGF against AVASTIN® (Genentech, Inc., South San Francisco, CA) and human and mouse cross-reactive antibody B20-4.1 in EBC-l-shMet-3.15 NSCLC xenografts Equivalent, indicating that functional VEGF is tumor cell-derived in the EBC-1 model (Fig. 6B). Furthermore, the mechanism of 'c-met inhibitory activity is not limited to the induction of VEGF expression by c-met, as knocking out c-met in the EBC-l-shMet-3.15 xenograft tumor model causes tumor regression (Fig. 6A). Anti-VEGF antibody treatment caused tumor stagnation (Fig. 6B). The effect of c-met knockdown combined with treatment with erlotinib and anti-VEGF antibody was tested and compared to treatment with a double combination and treatment with each agent alone. Previous studies have shown that. &quot;&quot;&quot;&quot;&quot;&quot;&quot;&quot;&quot;&quot;&quot;&quot;&lt;RTIID=0.0&gt;&gt;&gt; The combination of -met knockout, erlotinib and anti-VEGF antibodies produced an immediate and sustained tumor response (-92%); Figure 7) 'Maximum efficacy in any of the protocols tested in this experiment. The combination of either c-met knockout with anti-VEGF antibody treatment or erlotinib treatment also resulted in a significant increase in anti-tumor activity (_77% and -82%, respectively; Figure 7). Appropriate controls (sucrose, MCT and anti-porcine antibody; open circles) treated mice with EBC-l-shMet-4.5 tumors did not produce significant inhibition. Similarly, erlotinib alone showed minimal effect on tumor growth (positive Triangle) (_9〇/〇; Figure 7). Consistent with the studies shown above, c_met knockout (closed squares) and 143939.doc -169-201019961 treatment with anti-VEGF antibodies (inverted triangles) can cause delayed tumor growth or Stagnant (-59% and -70%, respectively; Figure 7). In contrast, 'erlotinib The combination with the anti-VEGF antibody showed no improvement compared to the efficacy observed with the anti-VEGF antibody alone (-61%; Figure 7). The combination of c-met knockdown and anti-VEGF antibody treatment also caused partial reaction (PR) and Increase in the number of complete responses (CR): combination treatment yielded 4 PRs and 1 CR, whereas c-met knockout alone produced 2 PRs and no CR, and treatment with anti-VEGF antibodies alone yielded 1 PR with no CR 0 The erlotinib plus c-met knockdown and anti-VEGF antibody protocol increased the number of reactions to 7 PR. The effect of treatment with anti-c-met antibody MetMAb, erlotinib and anti-VEGF antibody was tested and used separately Drug treatment or comparison with two-two combination treatment. NSCLC NCI-H596 xenografts were produced in hu-HGF-Tg-SCID mice, and MetMAb, anti-VEGF antibody (B20-4·1) and/or Ero were used. Treatment with nyani.

MetMAb、埃羅替尼及抗VEGF抗體之組合產生所觀察到 之最即時且顯著之腫瘤生長抑制(在第25天時為約95% ;圖 9)。此組中觀察到即時腫瘤體積退化,對應於此組中之9 個PR及1個CR。比較而言,MetMAb加抗VEGF抗體處理組 具有2個PR且無CR。 與未處理小鼠相比,在對照物處理小鼠(MetMAb緩衝 液、MCT及抗豬草抗體)中未觀察到腫瘤生長抑制(圖8)。 與先前研究一致,埃羅替尼(1〇〇 mg/kg,整個過程中每天 均投與)對NCI-H596腫瘤生長無影響,而MetMAb(30 mg/kg,每3週一次)引起部分腫瘤抑制(在第25天時約59%) 143939.doc • 170- 201019961 且埃羅替尼與MetMAb之組合產生強於疊加效應之效應(在 第25天時約89% ;圖8)。 抗VEGF抗體(B20-4.1 ’ 5 mg/kg,每週兩次;倒三角形) 單獨使用時會引起顯著的腫瘤生長抑制(在第25天時約 67。/。),然而當加上MetMAb時’存在顯著疊加效應(在第乃 天時約90% ;圖8) »此效應為c-met抑制所特有,因為當抗 VEGF抗體與埃羅替尼組合時,未觀察到疊加效應(在第25 天時約71% ;圖8)。在隨後時間點,MetMAb加埃羅替尼之 兩兩組合之反應曲線與MetMAb加抗VEGF抗體之反應曲線 之間的差異增加,指示當呈兩兩組合形式使用時’對C_ met與VEGF信號傳導之抑制提供最顯著且持久的抗腫瘤7舌 性。 此等資料表明c-met拮抗劑、VEGF拮抗劑及EGFR枯抗 劑之組合、及c-met拮抗劑及VEGF拮抗劑之組合各顯著抑 制非小細胞肺癌中之腫瘤生長。 雖然為達到清楚理解之目的,已藉由說明及實例之方式 較詳細地描述上述發明,但該等描述及實例不應理解為限 制本發明之範疇。 【圖式簡單說明】 圖1 :選用於異種移植研究之細胞株之综述° 細胞株 係基於c-met活化模式進行選擇且包括旁分泌型(NSCLC^® 胞株NCH-H596)、自分泌型(胰腺細胞株KP4)、c_met過度 表現型(NSCLC細胞株NCI-H441)及局部擴增且過度表現C-met型(NSCLC細胞株EBC-1)。(B-E)西方墨點法’如下顯 143939.doc -171 - 201019961 示填酸基-c-met及c-met蛋白含量:(B) 4種細胞株各在基本 條件下,(C) KP4及H596細胞經HGF(100 ng/ml)及/或單價 抗c-met單株抗體MetMAb(200 nM)處理後,(D)表現Dox可 誘導之針對 c-met 之 shRNA(shMet 3-1 及 shMet 3-11)或Dox 可誘導之針對GFP之shRNA(shGFP 1)的NCI-H441細胞之穩 定純系經Dox處理後,及(E)表現dox可誘導之針對c-met之 shRNA(shMet 3-15、shMet 4-12、shMet 4-5)或 dox 可誘導 之針對GFP之shRNA(shGFP2)的EBC-1細胞之穩定純系經 Dox處理後。Dox處理之EBC-1 shMet純系3-15及4-12完全 阻斷c-met表現,而EBC-1 shMet純系4-5部分阻斷c-met表 現。nd=未處理;Dox=多西環素;NSCLC=非小細胞肺 癌; 圖2 : c-met調控之血管生成因子之mRNA含量。(A)用 HGF處理配位體依賴性細胞株後,與血管生成分子表現相 關之c-met活性。(B)在含EBC-I Met shRNA之細胞中,Dox 處理後24及48小時用HGF處理會恢復基因表現。HGF =肝 細胞生長因子,Dox=多西環素,RMA=穩健多晶片平均值 (robust multi-chip average),IL8 =介白素 8,VEGF A=血管 内皮生長因子A,EPHA2=EPH受體A2,ANGPTL4=血管生 成素樣蛋白 4,EFNB2=ephrin B2 ; 圖3 : c-met調控之VEGF及IL8蛋白含量。(A、C)(A) EBC-1、(C) NCI-H441細胞中VEGF蛋白含量之調節。(D) 使用Dox可誘導之針對c-met之shRNA敲除c-met後’ EBC-1 腫瘤中之IL8蛋白含量。(B、E)用HGF處理NCI-H596細胞 143939.doc -172- 201019961 後,(B) VEGF及(E) IL8蛋白含量之調節。Dox=多西環 素,VEGF=血管内皮生長因子; 圖4:在胰腺腫瘤中,用抗c-met抗體MetMAb處理會增 強抗VEGF抗體功效。按指示處理帶皮下KP4胰腺異種移植 腫瘤之裸小鼠。對照物處理動物顯示不對處理起反應。 MetMAb以最佳劑量(30 mg/kg,IP,一次)、最低有效劑量 (7.5 mg/kg,10,一次)及次治療劑量(2.5 mg/kg,一次)給 予。識別人類與小鼠VEGF之抗VEGF抗體(B20-4.1)以最佳 劑量(5 mg/kg,IP,每週兩次)單獨給予,或與MetMAb(2.5 mg/kg,一次)或最低有效劑量之MetMAb(7.5 kg/kg,一次) 組合給予。作各組(n=10)平均腫瘤體積之曲線圖。在第25 天時計算腫瘤抑制(TI=100x(l-(TV*341/TC* ***)))。出於此 實驗之目的,PR及CR分別既定為腫瘤體積降低超過50%或 腫瘤體積降低達100%。*PS〇.05 ; PR=部分反應,CR=完全 反應,VEGF =血管内皮生長因子,IP=腹膜内; 圖 5 :在NCI-H441-shMet-3.ll NSCLC經瘤中,c-met抑 制會增強抗VEGF抗體之抗腫瘤效應。如下處理帶皮下 NCI-H441 shMet 3-11 NSCLC異種移植腫瘤之裸小鼠:用 單獨Dox(l mg/ml,於飲用水中之5%蔗糖中)處理以活體内 誘導針對c-met之shRNA ;用對照物(於飲用水中之5%蔗糖) 處理;或Dox處理與抗VEGF(B20-4.1)抗體處理組合。作各 組(n=10)平均腫瘤體積之曲線圖。Dox=多西環素, NSCLC=非小細胞肺癌,VEGF=血管内皮生長因子,Ab= 抗體; 143939.doc -173· 201019961 圖6 :表徵EBC-1-shMet模型中之c-met及VEGF抑制。按 指示處理帶皮下 EBC-l-shMet-3.15 或 EBC-l-shMet-4.5 NSCLC 腫瘤之裸小鼠。(A)帶 EBC-l-shMet-3.15 及 EBC-1-shMet-4.5腫瘤之小鼠係經Dox(l mg/ml,於飲用水中之5% 蔗糖中)或蔗糖(於飲用水中之5%蔗糖)處理。(B)帶EBC-1-shmet-3.15腫瘤之動物係經對照抗體(hu5B6)、抗人類 VEGF抗體(貝伐單抗)或抗人類/小鼠VEGF抗體(B20-4.1)(兩者均為5 mg/kg,IP,每週兩次)處理。作各組 (n=10)平均腫瘤體積之曲線圖。IP=腹膜内,Dox=多西環 素,NSCLC=非小細胞肺癌,VEGF=血管内皮生長因子; 圖7 :在EBC-l-shMet-4.5 NSCLC異種移植模型中,c-met、EGFR及VEGF抑制之組合顯著抑制腫瘤生長。帶皮 下EBC-l-shMet-4.5 NSCLC腫瘤之裸小鼠係經蔗糖(於飲用 水中之5%蔗糖)或Dox(0.5 mg/ml,於飲用水中之5%蔗糖 中)處理以誘導針對c-met之shRNA之表現。埃羅替尼(100 mg/kg,PO,每天)及/或抗VEGF抗體(B20-4.1.1,5 mg/kg, IP,每週兩次)係單獨或呈兩兩組合(Dox+B20-4.1.1 ; Dox+ 埃羅替尼;B20-4.1.1+埃羅替尼)或三個組合(Dox+B20-4.1 +埃羅替尼)形成給予。作各組(n=10)平均腫瘤體積之曲 線圖。在第27天時計算腫瘤抑制(ΤΙ=100χ(1-(ΤνΑ817Τ(:*^)))。 出於此實驗之目的,PR及CR分別既定為腫瘤體積降低超 過50%或腫瘤體積降低達100%。PR=部分反應’ CR=完全 反應,KD=敲除,Dox=多西環素,EGFR=表皮生長因子受 體,NSCLC =非小細胞肺癌,VEGF =血管内皮生長因子’ 143939.doc •174· 201019961 IP=腹膜内,PO=經口; 圖8 :在NCI-H596 NSCLC動物模型中,用抗c-met抗體 MetMAb、埃羅替尼及抗VEGF抗體之組合處理會顯著抑制 腫瘤生長。(A)帶皮下腫瘤之人類HGF-轉殖基因SCID(hu-‘ HGF-Tg-SCID)小鼠係未經處理,經對照物處理,經抗c- : met抗體 MetMAb(3 0 mg/kg,IP,每三週一次)、EGFR枯抗 劑埃羅替尼(100 mg/kg,PO,每天)或抗VEGF抗體(B20-4.1,5 mg/kg,IP,每週兩次)單獨處理,或經其兩兩組合 ® (MetMAb+B20-4.1 ; MetMAb+埃羅替尼;B20-4.1 +埃羅替 尼)形式處理或經其三個組合形式處理。(B) MetMAb+埃羅 替尼、MetMAb+抗VEGF抗體、及MetMAb+埃羅替尼+抗 VEGF抗體組之抗腫瘤效應之曲線圖突出組差異之量值。 作各組(n=10)平均腫瘤體積之曲線圖。在第25天時計算腫 瘤抑制(TI=100x(l-(TV*3te/TC#s* 组)))。PR及CR分別既定 為腫瘤體積降低超過50%或腫瘤體積降低達100%。PR=部分 反應,CR=完全反應,EGFR=表皮生長因子受體,NSCLC= 9 非小細胞肺癌,VEGF=血管内皮生長因子,IP=腹膜内, PO=經口 〇 *P&lt;0.02,**P=0.005 ; • 圖 9 :描繪MetMAb(OA5D5v2)之構架(FR)、CDR、第一 恆定域(CL或CH1)及Fc區(Fc)之胺基酸序列。所描繪之Fc 序列包含如WO 2005/063816中所述之「臼」(空穴)突變 T366S、L368A及Y407V ;及 圖10 :描繪Fc多肽之序列,該序列包含如WO 2005/063816 中所述之「杵」(突出)突變T366W。在一實施例中’包含 143939.doc •175· 201019961 此序列之Fc多肽與包含圖9之Fc序列的Fc多肽形成複合 物,從而產生Fc區。 143939.doc 176- 201019961 序列表 &lt;110&gt;美商建南德克公司 &lt;120&gt;組合療法 &lt;130&gt;P4277R1 &lt;140&gt;098134878 &lt;141&gt;2009-10-14 &lt;150&gt;US 61/106,513 &lt;151&gt; 2008-10-17 &lt;160&gt;33 &lt;210&gt;1 &lt;211&gt; 17 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;1The combination of MetMAb, erlotinib and anti-VEGF antibodies produced the most immediate and significant tumor growth inhibition observed (about 95% at day 25; Figure 9). Immediate tumor volume regression was observed in this group, corresponding to 9 PRs and 1 CR in this group. In comparison, the MetMAb plus anti-VEGF antibody treated group had 2 PRs and no CR. No tumor growth inhibition was observed in control-treated mice (MetMAb buffer, MCT, and anti-porcine antibody) compared to untreated mice (Fig. 8). Consistent with previous studies, erlotinib (1 〇〇 mg/kg, administered daily throughout the course) had no effect on NCI-H596 tumor growth, whereas MetMAb (30 mg/kg once every 3 weeks) caused partial tumors Inhibition (approximately 59% at day 25) 143939.doc • 170-201019961 and the combination of erlotinib and MetMAb produced an effect that was stronger than the additive effect (about 89% at day 25; Figure 8). Anti-VEGF antibody (B20-4.1 '5 mg/kg twice weekly; inverted triangle) causes significant tumor growth inhibition when used alone (about 67% at day 25), however when MetMAb is added 'There is a significant additive effect (about 90% in Daysday; Figure 8) » This effect is unique for c-met inhibition, because when the anti-VEGF antibody is combined with erlotinib, no superposition effect is observed (in the first About 71% at 25 days; Figure 8). At the subsequent time point, the difference between the response curve of the MetMAb plus erlotinib combination and the response curve of the MetMAb plus anti-VEGF antibody increased, indicating that C-met and VEGF signaling were used when used in a two-two combination. Inhibition provides the most significant and long lasting anti-tumor 7 tongue. These data indicate that the combination of a c-met antagonist, a VEGF antagonist and an EGFR antagonist, and a combination of a c-met antagonist and a VEGF antagonist each significantly inhibited tumor growth in non-small cell lung cancer. The above description of the invention has been described in detail by way of illustration and example, and the description and examples should not be construed as limiting the scope of the invention. [Simplified illustration] Figure 1: Summary of cell lines selected for xenograft studies ° Cell lines are selected based on c-met activation pattern and include paracrine (NSCLC^® cell line NCH-H596), autocrine (pancreatic cell line KP4), c_met overexpression (NSCLC cell line NCI-H441), and locally amplified and overexpressed C-met type (NSCLC cell line EBC-1). (BE) Western ink dot method 'Shows as follows 143939.doc -171 - 201019961 shows acid-based-c-met and c-met protein content: (B) 4 cell lines under basic conditions, (C) KP4 and H596 cells were treated with HGF (100 ng/ml) and/or monovalent anti-c-met monoclonal antibody MetMAb (200 nM), and (D) showed Dox-inducible shRNA against c-met (shMet 3-1 and shMet) 3-11) or Dox-inducible shRNA (shGFP 1) for NCI-H441 cells, stable pure after Dox treatment, and (E) dox-induced shRNA for c-met (shMet 3-15) The stable pure EBC-1 cells of shMet 4-12, shMet 4-5) or dox-inducible shRNA (shGFP2) against GFP were treated with Dox. Dox-treated EBC-1 shMet pure lines 3-15 and 4-12 completely blocked c-met performance, while EBC-1 shMet pure line 4-5 partially blocked c-met expression. Nd = untreated; Dox = doxycycline; NSCLC = non-small cell lung cancer; Figure 2: mRNA content of c-met regulated angiogenic factors. (A) c-met activity associated with angiogenic molecules after treatment of a ligand-dependent cell line with HGF. (B) Treatment with HGF at 24 and 48 hours after Dox treatment restored gene expression in cells containing EBC-I Met shRNA. HGF = hepatocyte growth factor, Dox = doxycycline, RMA = robust multi-chip average, IL8 = interleukin 8, VEGF A = vascular endothelial growth factor A, EPHA2 = EPH receptor A2, ANGPTL4 = angiopoietin-like protein 4, EFNB2 = ephrin B2; Figure 3: c-met regulated VEGF and IL8 protein content. (A, C) (A) EBC-1, (C) Regulation of VEGF protein content in NCI-H441 cells. (D) IL-8 protein content in EBC-1 tumors after knockdown of c-met by c-met shRNA against Dox inducible. (B, E) Treatment of NCI-H596 cells with HGF 143939.doc -172- 201019961, (B) Regulation of VEGF and (E) IL8 protein content. Dox = doxycycline, VEGF = vascular endothelial growth factor; Figure 4: Treatment with anti-c-met antibody MetMAb enhances the efficacy of anti-VEGF antibodies in pancreatic tumors. Nude mice bearing subcutaneous KP4 pancreatic xenograft tumors were treated as indicated. Control treated animals showed no response to treatment. MetMAb was administered at the optimal dose (30 mg/kg, IP, once), the lowest effective dose (7.5 mg/kg, 10, once) and the sub-therapeutic dose (2.5 mg/kg, once). Human and mouse VEGF anti-VEGF antibody (B20-4.1) was administered at the optimal dose (5 mg/kg, IP twice a week), or with MetMAb (2.5 mg/kg once) or the lowest effective dose MetMAb (7.5 kg/kg, once) was administered in combination. A graph of the mean tumor volume for each group (n=10). Tumor suppression was calculated at day 25 (TI = 100x (l-(TV*341/TC* ***))). For the purposes of this experiment, PR and CR were each determined to reduce tumor volume by more than 50% or tumor volume by 100%. *PS〇.05; PR=partial response, CR=complete response, VEGF=vascular endothelial growth factor, IP=intraperitoneal; Figure 5: c-met inhibition in NCI-H441-shMet-3.ll NSCLC transmembrane It will enhance the anti-tumor effect of anti-VEGF antibodies. Nude mice with subcutaneous NCI-H441 shMet 3-11 NSCLC xenograft tumors were treated as follows: treatment with c-met shRNA in vivo using treatment with Dox alone (1 mg/ml in 5% sucrose in drinking water) Treatment with a control (5% sucrose in drinking water); or Dox treatment in combination with anti-VEGF (B20-4.1) antibody treatment. A graph of the mean tumor volume of each group (n=10). Dox = doxycycline, NSCLC = non-small cell lung cancer, VEGF = vascular endothelial growth factor, Ab = antibody; 143939.doc -173· 201019961 Figure 6: Characterization of c-met and VEGF inhibition in the EBC-1-shMet model . Nude mice bearing subcutaneous EBC-l-shMet-3.15 or EBC-l-shMet-4.5 NSCLC tumors were treated as indicated. (A) Mice with EBC-l-shMet-3.15 and EBC-1-shMet-4.5 tumors were treated with Dox (1 mg/ml in 5% sucrose in drinking water) or sucrose (in drinking water) 5% sucrose) treatment. (B) Animals with EBC-1-shmet-3.15 tumors were treated with control antibody (hu5B6), anti-human VEGF antibody (bevacizumab) or anti-human/mouse VEGF antibody (B20-4.1) (both are 5 mg/kg, IP, twice a week). A graph of the mean tumor volume for each group (n=10). IP = intraperitoneal, Dox = doxycycline, NSCLC = non-small cell lung cancer, VEGF = vascular endothelial growth factor; Figure 7: c-met, EGFR and VEGF in the EBC-l-shMet-4.5 NSCLC xenograft model The combination of inhibition significantly inhibits tumor growth. Nude mice bearing subcutaneous EBC-l-shMet-4.5 NSCLC tumors were treated with sucrose (5% sucrose in drinking water) or Dox (0.5 mg/ml in 5% sucrose in drinking water) to induce The performance of c-met shRNA. Erlotinib (100 mg/kg, PO, daily) and/or anti-VEGF antibody (B20-4.1.1, 5 mg/kg, IP, twice a week) are either alone or in combination (Dox+B20 -4.1.1; Dox+ erlotinib; B20-4.1.1+ erlotinib) or three combinations (Dox+B20-4.1 + erlotinib) form the administration. A graph of the mean tumor volume of each group (n = 10) was made. Tumor inhibition was calculated on day 27 (ΤΙ=100χ(1-(ΤνΑ817Τ(:*^))). For the purposes of this experiment, PR and CR were determined to reduce tumor volume by more than 50% or tumor volume by 100, respectively. %.PR=Partial response 'CR=complete response, KD=knockout, Dox=doxycycline, EGFR=EGFR, NSCLC=non-small cell lung cancer, VEGF=vascular endothelial growth factor' 143939.doc • 174·201019961 IP=intraperitoneal, PO=oral; FIG. 8: In the NCI-H596 NSCLC animal model, treatment with a combination of anti-c-met antibody MetMAb, erlotinib and anti-VEGF antibody significantly inhibited tumor growth. (A) Human HGF-transgenic gene SCID (hu-' HGF-Tg-SCID) mice with subcutaneous tumors were untreated, treated with control, anti-c- : met antibody MetMAb (30 mg/kg) , IP, every three weeks), EGFR inhibitor erlotinib (100 mg/kg, PO, daily) or anti-VEGF antibody (B20-4.1, 5 mg/kg, IP, twice a week) Or treated by its two-in-one combination (MetMAb+B20-4.1; MetMAb+ erlotinib; B20-4.1 + erlotinib) or in combination of three (B) Me The anti-tumor effect of tMAb + erlotinib, MetMAb + anti-VEGF antibody, and MetMAb + erlotinib + anti-VEGF antibody group highlights the magnitude of the difference. The graph of the mean tumor volume of each group (n = 10) Tumor suppression was calculated on day 25 (TI = 100x (l-(TV*3te/TC#s**))). PR and CR were determined to reduce tumor volume by more than 50% or tumor volume by 100%, respectively. PR = partial response, CR = complete response, EGFR = epidermal growth factor receptor, NSCLC = 9 non-small cell lung cancer, VEGF = vascular endothelial growth factor, IP = intraperitoneal, PO = oral 〇 * P &lt; 0.02, ** P = 0.005; • Figure 9: depicts the amino acid sequence of the framework (FR), CDRs, first constant domain (CL or CH1) and Fc region (Fc) of MetMAb (OA5D5v2). The depicted Fc sequence comprises as WO "臼" (hole) mutations T366S, L368A and Y407V as described in 2005/063816; and Figure 10: depicts the sequence of an Fc polypeptide comprising a "杵" (prominent) mutation as described in WO 2005/063816 T366W. In an embodiment 'comprising 143939.doc • 175. 201019961 Fc polypeptide of this sequence forms complex with Fc polypeptide comprising the Fc sequence of Figure 9 Thereof, thereby generating an Fc region. 143939.doc 176- 201019961 Sequence Listing &lt;110&gt;US-based Nandek Corporation &lt;120&gt; Combination Therapy&lt;130&gt;P4277R1 &lt;140&gt;098134878 &lt;141&gt;2009-10-14 &lt;150&gt;US 61 /106,513 &lt;151&gt; 2008-10-17 &lt;160&gt;33 &lt;210&gt;1 &lt;211&gt; 17 &lt;212&gt;PRT &lt;213&gt;Artificial sequence&lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;;1

Lys Ser Ser Gin Ser Leu Leu Tyr Thr Ser Ser Gin Lys Asn Tyr 1 5 10 15Lys Ser Ser Gin Ser Leu Leu Tyr Thr Ser Ser Gin Lys Asn Tyr 1 5 10 15

Leu Ala &lt;210&gt;2 &lt;211&gt;7 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;2Leu Ala &lt;210&gt;2 &lt;211&gt;7 &lt;212&gt; PRT &lt;213&gt; artificial sequence &lt;220&gt;&lt;223&gt; synthetic sequence &lt;400&gt;2

Trp Ala Ser Thr Arg Glu Ser &lt;210&gt;3 &lt;211&gt;9 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;3Trp Ala Ser Thr Arg Glu Ser &lt;210&gt;3 &lt;211&gt;9 &lt;212&gt;PRT &lt;213&gt;Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;3

Gin Gin Tyr Tyr Ala Tyr Pro Trp Thr 5 &lt;210&gt;4 &lt;211&gt;10 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;40Q&gt;4Gin Gin Tyr Tyr Ala Tyr Pro Trp Thr 5 &lt;210&gt;4 &lt;211&gt;10 &lt;212&gt; PRT &lt;213&gt;Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;40Q&gt;4

Gly Tyr Thr Phe Thr Ser Tyr Trp Leu His 5 10 &lt;210&gt;5 &lt;211&gt; 18 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;5Gly Tyr Thr Phe Thr Ser Tyr Trp Leu His 5 10 &lt;210&gt;5 &lt;211&gt; 18 &lt;212&gt;PRT &lt;213&gt; Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;

Gly Met lie Asp Pro Ser Asn Ser Asp Thr Arg Phe Asn Pro Asn 143939.doc 201019961 1 5 10 15Gly Met lie Asp Pro Ser Asn Ser Asp Thr Arg Phe Asn Pro Asn 143939.doc 201019961 1 5 10 15

Phe Lys Asp &lt;210&gt;6 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt;合成序列 &lt;220&gt; &lt;223&gt;X為除R以外之任何胺基酸 &lt;400&gt;6Phe Lys Asp &lt;210&gt;6 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt; Synthetic sequence &lt;220&gt;&lt;223&gt; X is any amino acid other than R &lt;400&gt;

Xaa Tyr Gly Ser Tyr Val Ser Pro Leu Asp Tyr 5 10 &lt;210&gt;7 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;7Xaa Tyr Gly Ser Tyr Val Ser Pro Leu Asp Tyr 5 10 &lt;210&gt;7 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt;Artificial Sequence &lt;220&gt;&lt;223&gt; Synthesis Sequence &lt;400&gt;

Thr Tyr Gly Ser Tyr Val Ser Pro Leu Asp Tyr 5 10 &lt;210&gt;8 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;8Thr Tyr Gly Ser Tyr Val Ser Pro Leu Asp Tyr 5 10 &lt;210&gt;8 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt;Artificial Sequence &lt;220&gt;&lt;223&gt; Synthesis Sequence &lt;400&gt;8

Ser Tyr Gly Ser Tyr Val Ser Pro Leu Asp Tyr 5 10 &lt;210&gt;9 &lt;211&gt; 12 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;9Ser Tyr Gly Ser Tyr Val Ser Pro Leu Asp Tyr 5 10 &lt;210&gt;9 &lt;211&gt; 12 &lt;212&gt;PRT &lt;213&gt;Artificial Sequence &lt;220&gt;&lt;223&gt; Synthesis Sequence &lt;400&gt;9

Ala Thr Tyr Arg Ser Tyr Val Thr Pro Leu Asp Tyr 5 10 &lt;210&gt; 10 &lt;211&gt;119 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;10 GIu Val Gin Leu Val Glu Ser Gly Gly Gly Leu Val Gin Ρω Gly 1 5 10 15Ala Thr Tyr Arg Ser Tyr Val Thr Pro Leu Asp Tyr 5 10 &lt;210&gt; 10 &lt;211&gt;119 &lt;212&gt; PRT &lt;213&gt;Artificial Sequence&lt;220&gt;&lt;223&gt; Synthesis Sequence &lt;400&gt; GIu Val Gin Leu Val Glu Ser Gly Gly Gly Leu Val Gin Ρω Gly 1 5 10 15

Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Tyr Thr Phe Thr 20 25 30Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Tyr Thr Phe Thr 20 25 30

Ser Tyr Trp Leu His Trp Val Axg Gin Ala Pro Gly Lys Gly Leu 35 40 45Ser Tyr Trp Leu His Trp Val Axg Gin Ala Pro Gly Lys Gly Leu 35 40 45

Glu Trp Val Gly Met lie Asp Pro Ser Asn Ser Asp Thr Arg Phe 50 55 60Glu Trp Val Gly Met lie Asp Pro Ser Asn Ser Asp Thr Arg Phe 50 55 60

Asn Pro Asn Phe Lys Asp Arg Phe Thr lie Ser Ala Asp Thr Ser •2· 143939.doc 201019961 65 70 75Asn Pro Asn Phe Lys Asp Arg Phe Thr lie Ser Ala Asp Thr Ser •2· 143939.doc 201019961 65 70 75

Lys Asn Thr Ala Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp 80 85 90Lys Asn Thr Ala Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp 80 85 90

Thr AJa Val Tyr Tyr Cys Ala Thr Tyr Arg Ser Tyr Val Thr Pro 95 100 105Thr AJa Val Tyr Tyr Cys Ala Thr Tyr Arg Ser Tyr Val Thr Pro 95 100 105

Leu Asp Tyr Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser 110 115 &lt;210&gt;11 &lt;211&gt;114Leu Asp Tyr Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser 110 115 &lt;210&gt;11 &lt;211&gt;114

&lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;11&lt;212&gt;PRT &lt;213&gt; Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;

Asp He Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 5 10 15Asp He Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 5 10 15

Gly Asp Arg Val Thr lie Thr Cys Lys Ser Ser Gin Ser Leu Leu 20 25 30Gly Asp Arg Val Thr lie Thr Cys Lys Ser Ser Gin Ser Leu Leu 20 25 30

Tyr Thr Ser Ser Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys 35 40 45Tyr Thr Ser Ser Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys 35 40 45

Pro Gly Lys Ala Pro Lys Leu Leu lie Tyr Trp Ala Ser Thr Arg 50 55 60Pro Gly Lys Ala Pro Lys Leu Leu lie Tyr Trp Ala Ser Thr Arg 50 55 60

Glu Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr 65 70 75Glu Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr 65 70 75

Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro Glu Asp Phe Ala 80 85 90Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro Glu Asp Phe Ala 80 85 90

Thr Tyr Tyr Cys Gin Gin Tyr Tyr Ala Tyr Pro Trp Thr Phe Gly 95 100 105Thr Tyr Tyr Cys Gin Gin Tyr Tyr Ala Tyr Pro Trp Thr Phe Gly 95 100 105

Gin Gly Thr Lys Val Glu lie Lys Arg 110 &lt;210&gt; 12 &lt;211&gt;222 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列Gin Gly Thr Lys Val Glu lie Lys Arg 110 &lt;210&gt; 12 &lt;211&gt;222 &lt;212&gt; PRT &lt;213&gt; Artificial Sequence &lt;220&gt;&lt;223&gt; Synthesis Sequence

&lt;400&gt;12 Cys Pro I&lt;400&gt;12 Cys Pro I

Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val - 10 15Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val - 10 15

Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg 20 25 30Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg 20 25 30

Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp 35 40 45Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp 35 40 45

Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His 50 55 60Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His 50 55 60

Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn Ser Thr Tyr 65 70 75Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn Ser Thr Tyr 65 70 75

Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp Leu Asn 80 85 90Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp Leu Asn 80 85 90

Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala 95 100 105Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala 95 100 105

Pro lie Glu Lys Thr lie Scr Lys Ala Lys Gly Gin Pro Arg Glu 143939.doc 115 201019961 110Pro lie Glu Lys Thr lie Scr Lys Ala Lys Gly Gin Pro Arg Glu 143939.doc 115 201019961 110

Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys 125 130 135Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys 125 130 135

Asn Gin Val Ser Leu Ser Cys Ala Val Lys Gly Phe Tyr Pro Ser 140 145 150Asn Gin Val Ser Leu Ser Cys Ala Val Lys Gly Phe Tyr Pro Ser 140 145 150

Asp lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn 155 160 165Asp lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn 155 160 165

Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe 170 175 180Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe 170 175 180

Leu Val Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gin Gin Gly 185 190 195Leu Val Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gin Gin Gly 185 190 195

Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His 2(X) 205 210Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His 2(X) 205 210

Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys 215 220 &lt;210&gt; 13 &lt;211&gt;222 人工序列 &lt;212&gt; PRT &lt;213&gt; &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;13 Cys Pro 】Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys 215 220 &lt;210&gt; 13 &lt;211&gt;222 Artificial Sequence &lt;212&gt; PRT &lt;213&gt;&lt;220&gt;&lt;223&gt; Synthesis Sequence &lt;400&gt; Cys Pro 】

Pro Cys Pro Ak Pro Glu Leu Leu Gly Gly Pro Ser Val 一 10 15Pro Cys Pro Ak Pro Glu Leu Leu Gly Gly Pro Ser Val a 10 15

Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg 20 25 30Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg 20 25 30

Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp 35 40 45Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp 35 40 45

Pro Glu Val Lys Phe Asn Tip Tyr Val Asp Gly Val Glu Val His 50 55 60Pro Glu Val Lys Phe Asn Tip Tyr Val Asp Gly Val Glu Val His 50 55 60

Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn Ser Thr Tyr 65 70 75Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn Ser Thr Tyr 65 70 75

Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp Leu Asn 80 85 90Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp Leu Asn 80 85 90

Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala 95 100 105Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala 95 100 105

Pro lie Glu Lys Thr lie Ser Lys Ala Lys Gly Gin Pro Arg Glu 110 115 120Pro lie Glu Lys Thr lie Ser Lys Ala Lys Gly Gin Pro Arg Glu 110 115 120

Pro Gin Va3 Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys 125 130 135Pro Gin Va3 Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys 125 130 135

Asn Gin Val Ser Leu Trp Cys Leu Val Lys Gly Phe Tyr Pro Ser 140 145 150Asn Gin Val Ser Leu Trp Cys Leu Val Lys Gly Phe Tyr Pro Ser 140 145 150

Asp lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn 155 160 165Asp lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn 155 160 165

Tyr Lys Thr Hir Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe 170 175 180Tyr Lys Thr Hir Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe 170 175 180

Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Tip Gin Gin Gly 185 190 195Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Tip Gin Gin Gly 185 190 195

Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His 200 205 210Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His 200 205 210

Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys -4- 143939.doc 201019961 215 220 &lt;210&gt; 14 &lt;211&gt;449Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys -4- 143939.doc 201019961 215 220 &lt;210&gt; 14 &lt;211&gt;449

&lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;14&lt;212&gt; PRT &lt;213&gt; Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;14

Glu Val Gin Leu Val Glu Set Gly Gly Gly Leu Val Gin Pro Gly 1 5 10 15Glu Val Gin Leu Val Glu Set Gly Gly Gly Leu Val Gin Pro Gly 1 5 10 15

Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Tyr Thr Phe Thr 20 25 30Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Tyr Thr Phe Thr 20 25 30

Ser Tyr Trp Leu His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu 35 40 45Ser Tyr Trp Leu His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu 35 40 45

Glu Trp Val Gly Met lie Asp Pro Ser Asn Ser Asp Thr Arg Phe 50 55 60Glu Trp Val Gly Met lie Asp Pro Ser Asn Ser Asp Thr Arg Phe 50 55 60

Asn Pro Asn Phe Lys Asp Arg Phe Thr lie Ser Ala Asp Thr Ser 65 70 75Asn Pro Asn Phe Lys Asp Arg Phe Thr lie Ser Ala Asp Thr Ser 65 70 75

Lys Asn Thr Ala Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp 80 85 90Lys Asn Thr Ala Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp 80 85 90

Thr Ala Val Tyr Tyr Cys Ala Thr Tyr Arg Ser Tyt Val Thr Pro 95 100 105Thr Ala Val Tyr Tyr Cys Ala Thr Tyr Arg Ser Tyt Val Thr Pro 95 100 105

Leu Asp Tyr Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser Ala 110 115 120Leu Asp Tyr Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser Ala 110 115 120

Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Set Lys 125 130 135Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Set Lys 125 130 135

Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val Lys Asp 140 145 150Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val Lys Asp 140 145 150

Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu 155 160 165Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu 155 160 165

Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gin Ser Ser Gly 170 175 180Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gin Ser Ser Gly 170 175 180

Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu 185 190 195Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu 185 190 195

Gly Thr Gin Thr Tyr lie Cys Asn Val Asn His Lys Pro Ser Asn 200 205 210Gly Thr Gin Thr Tyr lie Cys Asn Val Asn His Lys Pro Ser Asn 200 205 210

Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr 215 220 225Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr 215 220 225

His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro 230 235 240His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro 230 235 240

Ser Val Phe Leu Pbe Pro Pro Lys Pro Lys Asp Thr Leu Met lie 245 250 255Ser Val Phe Leu Pbe Pro Pro Lys Pro Lys Asp Thr Leu Met lie 245 250 255

Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His 260 265 270Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His 260 265 270

Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu 275 280 285Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu 275 280 285

Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn Ser 290 295 300Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn Ser 290 295 300

Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp 305 310 315Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp 305 310 315

Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu 143939.doc 201019961 320 325 330Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu 143939.doc 201019961 320 325 330

Pro Ala Pro lie Glu Lys Tb: lie Ser Lys Ala Lys Gly Gin Pro 335 340 345Pro Ala Pro lie Glu Lys Tb: lie Ser Lys Ala Lys Gly Gin Pro 335 340 345

Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met 350 355 360Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met 350 355 360

Thr Lys Asn Gin Val Ser Leu Ser Cys Ala Val Lys Gly Phe Tyr 365 370 375Thr Lys Asn Gin Val Ser Leu Ser Cys Ala Val Lys Gly Phe Tyr 365 370 375

Pro Ser Asp lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu 380 385 390Pro Ser Asp lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu 380 385 390

Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser 395 400 405Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser 395 400 405

Phe Phe Leu Val Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gin 410 415 420Phe Phe Leu Val Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gin 410 415 420

Gin Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His 425 430 435Gin Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His 425 430 435

Asn His Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys 440 445 &lt;210&gt;15 &lt;211&gt;220Asn His Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys 440 445 &lt;210&gt;15 &lt;211&gt;220

&lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400〉15&lt;212&gt;PRT &lt;213&gt; artificial sequence &lt;220&gt;&lt;223&gt; synthetic sequence &lt;400>15

Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 5 10 15Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 5 10 15

Gly Asp Arg Val Thr lie Thr Cys Lys Ser Ser Gin Ser Leu Leu 20 25 30Gly Asp Arg Val Thr lie Thr Cys Lys Ser Ser Gin Ser Leu Leu 20 25 30

Tyr Thr Ser Ser Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys 35 40 45Tyr Thr Ser Ser Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Lys 35 40 45

Pro Gly Lys Ala Pro Lys Leu Leu lie Tyr Trp Ala Ser Thr Arg 50 55 60Pro Gly Lys Ala Pro Lys Leu Leu lie Tyr Trp Ala Ser Thr Arg 50 55 60

Glu Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr 65 70 75Glu Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr 65 70 75

Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro Glu Asp Phe Ala 80 85 90Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro Glu Asp Phe Ala 80 85 90

Thr Tyr Tyr Cys Gin Gin Tyr Tyr Ala Tyr Pro Trp Thr Phe Gly 95 100 105Thr Tyr Tyr Cys Gin Gin Tyr Tyr Ala Tyr Pro Trp Thr Phe Gly 95 100 105

Gin Gly Thr Lys Val Glu He Lys Arg Thr Val Ala Ala Pro Ser 110 115 120Gin Gly Thr Lys Val Glu He Lys Arg Thr Val Ala Ala Pro Ser 110 115 120

Val Phe lie Phe Pro Pro Ser Asp Glu Gin Leu Lys Ser Gly Thr 125 130 135Val Phe lie Phe Pro Pro Ser Asp Glu Gin Leu Lys Ser Gly Thr 125 130 135

Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala 140 145 150Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala 140 145 150

Lys Val Gin Trp Lys Vai Asp Asn Ala Leu Gin Ser Gly Asn Ser 155 160 165Lys Val Gin Trp Lys Vai Asp Asn Ala Leu Gin Ser Gly Asn Ser 155 160 165

Gin Glu Ser Val Thr Glu Gin Asp Ser Lys Asp Scr Thr Tyr Ser 170 175 180Gin Glu Ser Val Thr Glu Gin Asp Ser Lys Asp Scr Thr Tyr Ser 170 175 180

Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His 185 190 195Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His 185 190 195

Lys Val Tyr Ala Cys Glu Val Thr His Gin Gly Leu Ser Ser Pro -6- 143939.doc 201019961 200 205 210Lys Val Tyr Ala Cys Glu Val Thr His Gin Gly Leu Ser Ser Pro -6- 143939.doc 201019961 200 205 210

Val Thr Lys Ser Phe Asn Arg Gly Glu Cys 215 220 &lt;210&gt; 16 &lt;211&gt; 23 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列. &lt;400&gt;16Val Thr Lys Ser Phe Asn Arg Gly Glu Cys 215 220 &lt;210&gt; 16 &lt;211&gt; 23 &lt;212&gt; PRT &lt;213&gt; artificial sequence &lt;220&gt;&lt;223&gt; synthetic sequence. &lt;400&gt;

Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 5 10 15Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 5 10 15

Gly Asp Arg Val Thr He Thr Cys 20 &lt;210&gt; 17 &lt;211&gt;15 &lt;212&gt; PRT &lt;213&gt;人工序列Gly Asp Arg Val Thr He Thr Cys 20 &lt;210&gt; 17 &lt;211&gt;15 &lt;212&gt; PRT &lt;213&gt; Artificial sequence

&lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;17 Trp Tyr Gin Gin Lys&lt;220&gt;&lt;223&gt;Synthesis sequence &lt;400&gt;17 Trp Tyr Gin Gin Lys

Pro Gly Lys Ala Pro Lys Leu Leu lie Tyr 10 15 &lt;210&gt; 18 &lt;211&gt;32 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;4O0&gt; 18Pro Gly Lys Ala Pro Lys Leu Leu lie Tyr 10 15 &lt;210&gt; 18 &lt;211&gt;32 &lt;212&gt; PRT &lt;213&gt;Artificial sequence &lt;220&gt;&lt;223&gt;Synthesis sequence &lt;4O0&gt;

Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe 1 5 10 15Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe 1 5 10 15

Thr Leu Thr He Ser Ser Leu Gin Pro Glu Asp Phe Ala Thr Tyr 20 25 30Thr Leu Thr He Ser Ser Leu Gin Pro Glu Asp Phe Ala Thr Tyr 20 25 30

Tyr Cys &lt;210&gt; 19 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;19Tyr Cys &lt;210&gt; 19 &lt;211&gt;11 &lt;212&gt; PRT &lt;213&gt; artificial sequence &lt;220&gt;&lt;223&gt; synthetic sequence &lt;400&gt;

Phe Gly Gin Gly Thr Lys Val Glu lie Lys Arg 5 10 &lt;210&gt; 20 &lt;211&gt; 106 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;20Phe Gly Gin Gly Thr Lys Val Glu lie Lys Arg 5 10 &lt;210&gt; 20 &lt;211&gt; 106 &lt;212&gt;PRT &lt;213&gt;Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;

Thr Val Ala Ala Pro Ser Va! Phe lie Pbe Pro Pro Ser Asp Glu 1 5 10 15Thr Val Ala Ala Pro Ser Va! Phe lie Pbe Pro Pro Ser Asp Glu 1 5 10 15

Gin Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn 143939.doc 201019961 20 25 30Gin Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn 143939.doc 201019961 20 25 30

Phe Tyr Pro Arg Glu Ala Lys Val Gin Trp Lys Val Asp Asn Ala 35 40 45Phe Tyr Pro Arg Glu Ala Lys Val Gin Trp Lys Val Asp Asn Ala 35 40 45

Leu Gin Ser Gly Asn Set Gin Glu Ser Val Thr Glu Gin Asp Ser 50 55 60Leu Gin Ser Gly Asn Set Gin Glu Ser Val Thr Glu Gin Asp Ser 50 55 60

Lys Asp Ser Thr Tyr Ser Leu Ser Ser Thr Leu Tlir Leu Ser Lys 65 70 75Lys Asp Ser Thr Tyr Ser Leu Ser Ser Thr Leu Tlir Leu Ser Lys 65 70 75

Ala Asp Tyr Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His 80 85 90Ala Asp Tyr Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His 80 85 90

Gin Gly Leu Ser Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu 95 100 105 &lt;210&gt;21 &lt;211&gt;25 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;21Gin Gly Leu Ser Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu 95 100 105 &lt;210&gt;21 &lt;211&gt;25 &lt;212&gt;PRT &lt;213&gt;Artificial Sequence&lt;220&gt;&lt;223&gt;SynthesisSequence&lt;;400&gt;21

Glu Val Gin Leu Val Glu Ser Gly Gly Gly Leu Val Gin Pro Gly 1 5 10 15Glu Val Gin Leu Val Glu Ser Gly Gly Gly Leu Val Gin Pro Gly 1 5 10 15

Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser 20 25 &lt;210&gt; 22 &lt;211&gt; 13 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;22Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser 20 25 &lt;210&gt; 22 &lt;211&gt; 13 &lt;212&gt;PRT &lt;213&gt;Artificial sequence &lt;220&gt;&lt;223&gt; Synthesis sequence &lt;400&gt;22

Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val 5 10 &lt;210&gt; 23 &lt;211&gt;30 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;23Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val 5 10 &lt;210&gt; 23 &lt;211&gt;30 &lt;212&gt;PRT &lt;213&gt;Artificial Sequence&lt;220&gt;&lt;223&gt;SynthesisSequence&lt;400&gt; twenty three

Arg Phe Thr lie Ser Ala Asp Thr Ser Lys Asn Thr Ala Tyr Leu 1 5 10 15Arg Phe Thr lie Ser Ala Asp Thr Ser Lys Asn Thr Ala Tyr Leu 1 5 10 15

Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 20 25 30 &lt;210&gt;24 &lt;211&gt;11 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;24Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 20 25 30 &lt;210&gt;24 &lt;211&gt;11 &lt;212&gt;PRT &lt;213&gt;Artificial Sequence&lt;220&gt;&lt;223&gt;SynthesisSequence&lt;;400&gt;24

Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser 5 10 &lt;210&gt;25 143939.doc 201019961 &lt;211&gt;5 &lt;212&gt; PRT &lt;213&gt;智人 &lt;400&gt;25Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser 5 10 &lt;210&gt;25 143939.doc 201019961 &lt;211&gt;5 &lt;212&gt; PRT &lt;213&gt; Homo sapiens &lt;400&gt;25

Leu Asp Ala Gin Thr &lt;210&gt;26 &lt;211&gt;9 &lt;212&gt;PRT &lt;213&gt;智人 &lt;400&gt;26Leu Asp Ala Gin Thr &lt;210&gt;26 &lt;211&gt;9 &lt;212&gt;PRT &lt;213&gt; Homo sapiens &lt;400&gt;26

Leu Thr Glu Lys Arg Lys Lys Arg Ser et MIGlu Λα Al ser T 人asp5 278PR智27ΓΟ 0&gt;1&gt;2&gt;3&gt; &lt;21&lt;21&lt;21&lt;21 &lt;210&gt;28 &lt;211&gt;8 &lt;212&gt;PRT &lt;213&gt;智人 &lt;400&gt; 28Leu Thr Glu Lys Arg Lys Lys Arg Ser et MIGlu Λα Al ser T human asp5 278PR 智27ΓΟ 0&gt;1&gt;2&gt;3&gt;&lt;21&lt;21&lt;21&lt;21&lt;210&gt;28&lt;211&gt;8&lt;212&gt;PRT&lt;213&gt; Homo sapiens &lt;400&gt; 28

Asn Val Arg Cys Leu Gin His Phe &lt;210&gt;29 &lt;211&gt;65 &lt;212&gt; DNA &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;29 gatccccgaa cagaatcact gacatattca agagatatgt cagtgattct 50 gttctttttt ggaaa65 &lt;210&gt;30 &lt;211&gt;65 &lt;212&gt;DNA &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt; 30 gatccccgaa actgtatgct ggatgattca agagatcatc cagcatacag 50 tttctttttt ggaaa65 &lt;210&gt;31 &lt;211&gt;67 &lt;212&gt; DNA &lt;213&gt;人工序列 &lt;220&gt; &lt;223&gt;合成序列 &lt;400&gt;31 gatccccaga tccgccacaa catcgattca agagatcgat gttgtggcgg 50 atcttgtttt ttggaaa67 &lt;210&gt;32 -9- 143939.doc 201019961 &lt;212&gt;PRT &lt;213&gt;人工序列 &lt;220&gt;&lt;223&gt;合成序列 &lt;400&gt;32 Asp lie Cys Leu Pro Arg Trp Gly Cys Leu Trp 5 10 b&gt; Λ &gt; lx «1 lx ^¾^ &lt; A1,Asn Val Arg Cys Leu Gin His Phe &lt;210&gt;29 &lt;211&gt;65 &lt;212&gt; DNA &lt;213&gt;Artificial sequence&lt;220&gt;&lt;223&gt;Synthesissequence&lt;400&gt;29 gatccccgaa cagaatcact gacatattca agagatatgt cagtgattct 50 Gttctttttt ggaaa65 &lt;210&gt;30 &lt;211&gt;65 &lt;212&gt;DNA &lt;213&gt;Artificial sequence&lt;220&gt;&lt;223&gt;Synthesissequence&lt;400&gt; 30 gatccccgaa actgtatgct ggatgattca agagatcatc cagcatacag 50 tttctttttt ggaaa65 &lt;210&gt; 31 &lt;211&gt;67 &lt;212&gt; DNA &lt;213&gt; artificial sequence &lt;220&gt;&lt;223&gt; synthetic sequence &lt;400&gt;31 gatccccaga tccgccacaa catcgattca agagatcgat gttgtggcgg 50 atcttgtttt ttggaaa67 &lt;210&gt;32 -9- 143939. Doc 201019961 &lt;212&gt;PRT &lt;213&gt;Artificial sequence&lt;220&gt;&lt;223&gt;Synthesis sequence&lt;400&gt;32 Asp lie Cys Leu Pro Arg Trp Gly Cys Leu Trp 5 10 b&gt; Λ &gt; lx «1 lx ^3⁄4^ &lt; A1,

Glyuu n As 5&gt;10&gt;iRl&gt;33serserTyrThrLeuGlyTl:Hi serLysAlaserserserPLV sersuvalGlysersersSPTOsAS proLeserGlnserLVrcy AlacyslLeupronHissse uuGlyTrpvalvalASDOLVs ^5¾¾¾¾¾^ pheAlavalprovall I valAlaThrphevalcyssval sers ΐ sThrMserw sSLyloo proGlyproHisserTyrPLyM.;:lar 143939.doc 10-Glyuu n As 5 &gt; 10 &gt; iRl &gt; 33serserTyrThrLeuGlyTl: Hi serLysAlaserserserPLV sersuvalGlysersersSPTOsAS proLeserGlnserLVrcy AlacyslLeupronHissse uuGlyTrpvalvalASDOLVs ^ 5¾¾¾¾¾ ^ pheAlavalprovall I valAlaThrphevalcyssval sers ΐ sThrMserw sSLyloo proGlyproHisserTyrPLyM;:. Lar 143939.doc 10-

Claims (1)

201019961 七、申請專利範圍: 1. 一種c-met拮抗劑與VEGF拮抗劑組合之用途,其用於製 造供治療個體癌症之藥物。 2. 如請求項1之用途,其中該c-met拮抗劑為抗體。 : 3.如請求項2之用途,其中該抗體為單價抗體。 ί 4.如請求項3之用途,其中該抗體為單價且包含Fc區,其 中該Fc區包含第一多肽及第二多肽,其中該第一多肽包 含SEQ ID NO: 12中所述之Fc序列及該第二多肽包含SEQ ❹ ID NO: 13中所述之序列。 5.如請求項4之用途,其中該抗體包含(a)第一多肽,其包 含具有如下序列之重鏈可變域: EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYWLHW VRQAPGKGLEWVGMIDPSNSDTRFNPNFKDRFTISADTS KNTAYLQMNSLRAEDTAVYYCATYRSYVTPLDYWGQGT LVTVSS (SEQ ID NO: 10)、CH1序列及第一 Fc多肽;(b) 第二多肽,其包含具有如下序列之輕鏈可變域: • DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKN YLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGTD FTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIKR (SEQ ID NO: 11)及CL1序列;及(c)包含第二Fc多肽之第 •三多肽,其中該重鏈可變域及該輕鏈可變域呈複合物形 式存在且形成單一抗原結合臂,其中該第一 Fc多肽及該 第二Fc多肽呈複合物形式存在且形成Fc區,與包含該抗 原結合臂之Fab分子相比,其增加該抗體片段之穩定 143939.doc 201019961 性。 6. 如請求項5之用途,其中該第一多肽包含SEQ ID NO: 12 中所述之Fc序列,該第二多肽包含SEQ ID NO: 13中所述 之Fc序列。 7. 如請求項5之用途,其中該第一多肽包含SEQ ID NO: 13 中所述之Fc序列,該第二多肽包含SEQ ID NO: 12中所述 之Fc序列。 8. 如請求項4之用途,其中該抗體為MetMAb。 9. 如請求項1至8中任一項之用途,其中該VEGF拮抗劑為 能特異性結合VEGF之適體(aptamer)。 10. 如請求項1至8中任一項之用途,其中該VEGF拮抗劑為 可溶性VEGF受體蛋白,或其VEGF結合片段,或嵌合 VEGF受體蛋白。 11. 如請求項10之用途,其中該嵌合VEGF受體蛋白至少包 含Flt-Ι或KDR之細胞外域2。 12. 如請求項1至8中任一項之用途,其中該VEGF拮抗劑為 抗VEGF抗體° 13. 如請求項12之用途,其中該抗VEGF抗體為單株抗體。 14. 如請求項13之用途,其中該單株抗體為嵌合抗體、完全 人類抗體或人類化抗體。 15. 如請求項14之用途,其中該抗VEGF抗體為貝伐單抗 (bevacizumab)、G6系列抗體、B20系列抗體或其VEGF結 合片段。 16. 如請求項15之用途,其中該抗VEGF抗體為貝伐單抗。 143939.doc 201019961 17. 如請求項1至8中任一項之用途,其中該藥物進—步包含 EGFR拮抗劑或與EGFR拮抗劑組合使用。 18. 如請求項17之用途,其中該eGFR拮抗劑具有通式工:201019961 VII. Scope of Application: 1. A use of a combination of a c-met antagonist and a VEGF antagonist for the manufacture of a medicament for treating cancer in an individual. 2. The use of claim 1, wherein the c-met antagonist is an antibody. 3. The use of claim 2, wherein the antibody is a monovalent antibody. 4. The use of claim 3, wherein the antibody is monovalent and comprises an Fc region, wherein the Fc region comprises a first polypeptide and a second polypeptide, wherein the first polypeptide comprises the SEQ ID NO: 12 The Fc sequence and the second polypeptide comprise the sequence set forth in SEQ ID NO: 13. 5. The use of claim 4, wherein the antibody comprises (a) a first polypeptide comprising a heavy chain variable domain having the sequence: EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYWLHW VRQAPGKGLEWVGMIDPSNSDTRFNPNFKDRFTISADTS KNTAYLQMNSLRAEDTAVYYCATYRSYVTPLDYWGQGT LVTVSS (SEQ ID NO: 10), CH1 sequence and first Fc polypeptide; (b) a second polypeptide comprising a light chain variable domain having the sequence: • DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKN YLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGTD FTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIKR (SEQ ID NO: 11) and CL1 sequence; and (c) including the second Fc polypeptide a tripeptide, wherein the heavy chain variable domain and the light chain variable domain are present in a complex form and form a single antigen binding arm, wherein the first Fc polypeptide and the second Fc polypeptide are present in a complex form and form an Fc The region, which increases the stability of the antibody fragment compared to the Fab molecule comprising the antigen binding arm, 143939.doc 201019961. 6. The use of claim 5, wherein the first polypeptide comprises the Fc sequence set forth in SEQ ID NO: 12, and the second polypeptide comprises the Fc sequence set forth in SEQ ID NO: 13. 7. The use of claim 5, wherein the first polypeptide comprises the Fc sequence set forth in SEQ ID NO: 13, and the second polypeptide comprises the Fc sequence set forth in SEQ ID NO: 12. 8. The use of claim 4, wherein the antibody is MetMAb. The use according to any one of claims 1 to 8, wherein the VEGF antagonist is an aptamer capable of specifically binding to VEGF. The use according to any one of claims 1 to 8, wherein the VEGF antagonist is a soluble VEGF receptor protein, or a VEGF-binding fragment thereof, or a chimeric VEGF receptor protein. 11. The use of claim 10, wherein the chimeric VEGF receptor protein comprises at least the extracellular domain 2 of Flt-Ι or KDR. 12. The use of any one of claims 1 to 8, wherein the VEGF antagonist is an anti-VEGF antibody. 13. The use of claim 12, wherein the anti-VEGF antibody is a monoclonal antibody. 14. The use of claim 13, wherein the monoclonal antibody is a chimeric antibody, a fully human antibody or a humanized antibody. 15. The use of claim 14, wherein the anti-VEGF antibody is bevacizumab, a G6 series antibody, a B20 series antibody or a VEGF binding fragment thereof. 16. The use of claim 15, wherein the anti-VEGF antibody is bevacizumab. The use of any one of claims 1 to 8, wherein the medicament further comprises or is used in combination with an EGFR antagonist. 18. The use of claim 17, wherein the eGFR antagonist has a general formula: m為1、2或3 ; 各R1獨立地選自由以下組成之群:氫、鹵基、羥基、 羥胺基、叛基、硝基、胍基、脲基、氰基、三氟甲基及 -(CVC4伸烷基)_w-(苯基),其中W為單鍵、〇、s或NH; 或各R1獨立地選自R9及經氰基取代之匚丨-匚4烷基,其中 R9係選自由以下組成之群:R5、-OR6、-NR6R6、-C(〇)R7 、-NHOR5、-〇C(0)R6、氰基、A及·YR5 ; R5 為 CVC4燒 基;R6獨立地為氫或R5 ; R7為R5、-〇R6或_NR6R6 ; A係 選自 N-β底咬基(piperidino)、N-嗎1#基(morpholino)、N-吡咯啶基(pyrrolidino)、4-R6-哌嗪-1-基、咪唑-1-基、4_ 比啶酮-1-基、-(CVC4伸烷基)(C02H)、苯氧基、苯基、 苯基硫基、C2-C4烯基及-(CVC4伸烷基)c(o)nr6r6 ;及 Y為S、SO或S〇2;其中R5、_〇尺6及_NR6R6中之烷基部分 視情況經1至3個画基取代基取代,且R5、-OR6及-NR6R6 中之烧基部分視情況經1或2個R9基團取代,且其中該等 143939.doc 201019961 視it況取代基之烷基部分視情況經鹵基或R9取代,限制 條件為兩個雜原子不連接於同一碳原子; 或各R〗獨立地選自_NHS〇2R5、鄰苯二醯亞胺基 (phthalimid0)-(ci-C4)-烷基磺醯基胺基、苯甲醯胺基、 苯磺醯基胺基、3·苯基脲基、2_側氧基(〇χ〇)„比咯啶 基、2,5-二側氧基吡咯啶_卜基及Rn)_(C2_C4)_烷醯基胺 基,其中R10選自由基、_OR6、C2_C4烷醯氧基、_c(〇)r7 及-nr6r6;且其中該等Rl基團_nhs〇2rS、鄰苯二醯亞胺 基-(Ci-C4)烷基磺醯基胺基、苯甲醯胺基、苯磺醯基胺 基、3-苯基腺基、2·側氧基吡u各啶基、2,5-二側氧基 °比洛咬-1-基及R1G_(C2-C4)-烧醯基胺基視情況經1或2個獨 立地選自鹵基、C丨-C4烷基、氰基、甲烷磺醯基及匚广。 烷氧基之取代基取代; 或兩個R1基團連同其所連接之碳一起形成5_8員環,該 環包括1或2個選自〇、s及N之雜原子; R為氫或C〗-C6烧基’該C〗-C6烧基視情況經1至3個獨 立地選自_基、C!-C4烷氧基、-NR6R6及-S02R5之取代基 取代; η為1或2’且各R3獨立地選自氫、鹵基、經基、c丨_c6 烧基、-NR6R6及CVC4烷氧基’其中該等R3基圈之烷基部 分視情況經1至3個獨立地選自鹵基、(^-(24烷氧基、 -NR6R6及-S02R之取代基取代;及 R4為疊氮基或-(乙炔基)-Rn,其中R11為氫或Cl-C6烷 基’該C「C6烷基視情況經羥基、-OR6或-NR6R6取代。 143939.doc 201019961 19·如請求項17之用途,其中該EGFR拮抗劑為選自由以下 組成之群之式I化合物: (6,7-二甲氧基喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6,7-二甲氧基喹唑啉-4-基羥基丙炔-1-基)笨基]-胺; [3-(2’_(胺基曱基)·乙炔基)苯基μ(6,7_二曱氧基喹唑啉_4_ : 基)-胺;(3-乙炔基苯基)-(6-硝基喹唑啉-4-基)-胺;(6,7- 二曱氧基喹唑啉-4-基)-(4-乙炔基苯基)-胺;(6,7-二曱氧 基喹唑啉-4-基)-(3-乙炔基-2-甲基苯基)-胺;(6-胺基喹唑 m 啉-4-基)-(3-乙炔基苯基)-胺;(3-乙炔基苯基)-(6-曱烷磺 醯基胺基喹唑啉-4-基)-胺;(3-乙炔基苯基)-(6,7-亞甲基 二氧基喹唑啉-4-基)-胺;(6,7-二甲氧基喹唑啉-4-基)-(3-乙炔基-6-甲基苯基)-胺;(3-乙炔基苯基)_(7_硝基喹唑 啉-4-基)-胺;(3-乙炔基苯基)-[6-(4'-甲苯磺醯基胺基)喹 。坐基]-胺;(3_乙炔基苯基)_{6_[2,_鄰苯二醯亞胺基· 乙-Γ-基-績醯基胺基]喹唑啉_4_基}_胺;(3-乙炔基苯基)_ φ (6·胍基喹唑啉-4-基)-胺;(7-胺基喹唑啉-4-基)-(3-乙炔 基苯基)-胺;(3-乙炔基苯基)-(7-甲氧基喹唑啉-4-基)-胺;(6-曱氧羰基喹唑琳_4-基)-(3-乙炔基苯基)-胺;(7-曱 . 氧羰基喹嗤啉-4-基)-(3-乙炔基苯基)-胺;[6,7-雙(2-曱氧 基乙氧基)喹唑啉-4-基]-(3-乙炔基笨基)-胺;(3-疊氮基 苯基)_(6,7-二甲氧基喹唑啉-4-基)-胺;(3-疊氮基_5-氣苯 基Η6,7-二甲氧基喹唑啉-4-基)-胺;(4-疊氮基苯基)-(6,7_二甲氧基喹唑啉-4-基)-胺;(3-乙炔基苯基)-(6·甲烷 磺酿基-啥《坐啉基)-胺;(6·乙烷硫基-喹唑啉-4-基)-(3- 143939.doc 201019961 乙炔基苯基)-胺;(6,7-二曱氧基-啥》坐你-4-基)-(3-乙快 基-4-氟-苯基)·胺;(6,7_二曱氧基-喹唑啉_4_基)-[3·(丙 炔-Γ-基)-苯基]-胺;[6,7_雙_(2-甲氧基-乙氧基)-喹唑啉_ 4·基]-(5-乙炔基-2-甲基-苯基)-胺·,[6,7-雙-(2-曱氧基-乙 氧基)-喹唑啉-4-基]-(3-乙炔基-4-氟-苯基)-胺;[6,7-雙-(2-氯-乙氧基)·喹唑啉_4·基]_(3_乙炔基-苯基)-胺;[6-(2-氣-乙氧基)-7-(2-曱氧基_乙氧基)-喧吐淋-4 -基]-(3 -乙快 基-苯基)-胺;[6,7-雙-(2-乙醯氧基·乙氧基)-喹唑啉-4-基M3-乙炔基-苯基)_胺;2-[4-(3-乙炔基-苯基胺基)_7_ (2-羥基-乙氧基)-喹唑啉_6_基氧基]·乙醇;[6_(2_乙醯氧 基-乙氧基)-7-(2-曱氧基-乙氧基)-嗜唾琳_4_基]-(3 -乙炔 基-苯基)-胺;[7-(2-氣-乙氧基)-6-(2-甲氧基-乙氧基)_喹 唑啉-4-基]-(3-乙炔基-苯基)·胺;[7-(2-乙醯氧基-乙氧 基)-6-(2-甲氧基-乙氧基)-喹嗤琳_4_基]_(3_乙炔基_笨基)_ 胺;2-[4-(3-乙炔基-苯基胺基)_6_(2_經基-乙氧基)_啥。坐 琳-7-基氧基]-乙醇;2-[4-(3-乙炔基-苯基胺基)_7_(2_曱 氧基-乙氧基)-喹唑啉-6-基氧基]-乙醇;2_[4-(3-乙炔基-苯基胺基)-6-(2-甲氧基-乙氧基)_喹唑琳_7_基氧基]-乙 醇;[6·(2-乙醯氧基-乙氧基)-7-(2-曱氧基·乙氧基)_喹唑 啉-4-基]-(3-乙炔基-苯基)-胺;(3-乙炔基_苯基)_《6_(2_甲 氧基-乙氧基)-7-[2-(4-曱基-n底嗓_丨_基)_乙氧基]喹吐琳_ 4-基}-胺;(3-乙快基-苯基)-[7·(2·甲氧基_乙氧基)_6_(2· 嗎琳-4-基)-乙氧基)-啥唑琳-4-基]-胺;(6,7_二乙氧基啥 0坐琳-1-基)-(3-乙炔基苯基)-胺;(6,7-二丁氧基喧。坐琳_ι· 143939.doc -6- 201019961 基)-(3-乙炔基苯基)-胺;(6,7-二異丙氧基喹唑啉-丨·基)_ (3 -乙块基苯基)-胺,(6,7-· —乙氧基啥嗤琳-1-基)-(3 -乙快 基-2-甲基-苯基)-胺;[6,7-雙-(2-曱氧基-乙氧基)-喹唑啉_ 1-基]-(3-乙炔基-2-甲基-苯基)-胺;(3-乙炔基苯基)_[6_ (2-羥基-乙氧基)-7-(2-甲氧基·乙氧基)-喹唑啉_1_基]_ 胺;[6,7-雙-(2-羥基-乙氧基)-喹唑啉-1-基]-(3-乙炔基苯 基)-胺;2-[4-(3-乙炔基-苯基胺基)-6-(2-甲氧基-乙氧基)_ 喹唑啉_7-基氧基]-乙醇;(6,7-二丙氧基-喹唑啉·4_基;)_ (3-乙炔基-苯基)-胺;(6,7-二乙氧基-喹唑啉-4-基)-(3-乙 炔基-5-氟-苯基)-胺;(6,7-二乙氧基-喹唑啉-4-基)-(3-乙 快基-4-氟-苯基)-胺;(6,7-二乙乳基-喧唾琳-4-基)-(5-乙 快基-2-甲基-苯基)-胺;(6,7-二乙氧基-啥唾琳-4-基)-(3-乙炔基-4-甲基-苯基)-胺;(6-胺基甲基-7-甲氧基-喹唑 嚇^-4-基)-(3 -乙快基-苯基)-胺;(6-胺基甲基-7-甲氧基-喧 唑啉-4·基)-(3-乙炔基苯基)-胺;(6-胺基羰基曱基_7_曱氧 基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6-胺基羰基乙基_ 7-甲氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6·胺基幾基 甲基-7-乙氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6_胺 基幾基乙基-7-乙氧基-喹α坐琳-4-基)-(3-乙炔基苯基)_ 胺;(6-胺基羰基曱基-7-異丙氧基-喹唑啉-4-基)_(3•乙块 基苯基)-胺;(6-胺基幾基甲基-7-丙氧基-喧1&gt;坐琳_4-基)_ (3-乙炔基苯基)-胺;(6-胺基羰基甲基-7-甲氧基-喹唑琳_ 4-基)-(3-乙炔基苯基)-胺;(6-胺基羰基乙基-7-異丙氧基_ 喹唾啉-4-基)-(3-乙炔基苯基)-胺;及(6-胺基羰基乙基_7_ 143939.doc 201019961 丙氧基喹唑啉-4·基)-(3-乙炔基苯基)-胺;(6,7·二乙氧基 喹唑啉-1-基)-(3-乙炔基苯基)-胺;(3-乙炔基苯基)-[6-(2-羥基-乙氧基)-7-(2-甲氧基-乙氧基)-喹唑啉-1-基]-胺; [6,7-雙-(2·羥基-乙氧基)_喹唑啉-1-基]-(3-乙炔基苯基)-胺;[6,7-雙-(2-曱氧基-乙氧基)-喹唑啉-1-基]-(3-乙炔基 苯基)-胺;(6,7-二曱氧基喹唑啉-1-基)-(3-乙炔基苯基)-胺;(3-乙炔基苯基)-(6-曱烷磺醯基胺基-喹唑啉-1-基)-胺;及(6-胺基-喹唑啉_ι_基)_(3_乙炔基苯基胺。 20. 21. 22. 23. 24. 25. 如請求項17之用途,其中該式I之EGFR拮抗劑為Ν-(3-乙 炔基苯基)-6,7-雙(2-甲氧基乙氧基)-4-喹唑啉胺。 如請求項17之用途’其中該EGFR拮抗劑Ν·(3·乙炔基苯 基)-6,7-雙(2-甲氧基乙氧基)_‘喹唑蛛胺呈鹽酸鹽形式。 如請求項17之用途,其中該EGFR拮抗劑^^(弘乙炔基苯 基)-6,7-雙(2_甲氧基乙氧基)_4_啥„坐淋胺係呈實質上均勻 之結晶多晶型物形式,該形式展現χ光粉末繞射圖之特 徵峰以 2Θ。表示在約 6.26、12.48、13.39、16.96、20.20、 21’1〇、22.98、24.46、25.14及26.91。 如請求項17之用途,其中該£(31?尺拮抗劑為4(3|氣氟 苯胺基)~7-曱氧基-6-(3-N-嗎啉基丙氧基)喹唑啉。 月求項17之用途,其中該EgfR拮抗劑為N_[3_氣_4_ 氟笨基)曱氧基]苯基]_6-[5-[[[2-(甲基磺酿基)乙基]胺 基]甲基]-2-呋喃基]喹唑啉胺。 ^求項17之㈣,其中―㈣拮抗劑林(4溴_2氟 本胺基)-6-甲氧基_7♦甲基娘咬基甲氧基)啥咬啉。 143939.doc 201019961 26. 如請求項17之用途,其中該抗cmet抗體為MetMAb,該 EGFR拮抗劑為N-(3-乙炔基苯基)-6,7-雙(2-甲氧基乙氧 基)_4_喹唑啉胺,該VEGF拮抗劑為貝伐單抗。 27. 如請求項1至8中任一項之用途,其中該癌症係選自由以 下組成之群:乳癌、結腸直腸癌、直腸癌、非小細胞肺 癌、非霍奇金氏淋巴瘤(non-Hodgkins lymphoma)、腎細 胞癌、前列腺癌、肝癌、胰腺癌、軟組織肉瘤、卡波西氏 肉瘤(kaposi's sarcoma)、類癌瘤(carcinoid carcinoma)、 頭頸癌、胃癌、黑素瘤、卵巢癌、間皮瘤及多發性骨髓 瘤。 28. 如請求項27之用途,其中該癌症為非小細胞肺癌。 29. 如請求項1至8中任一項之用途,其中該藥物進一步包含 化學治療劑或與化學治療劑組合使用。 3 0. —種治療個體癌症之醫藥組合物,其包含治療有效量之 c-met拮抗劑及VEGF拮抗劑。 31. 如請求項30之醫藥組合物,其中該c-met拮抗劑為抗體。 32. 如請求項3 1之醫藥組合物,其中該抗體為單價抗體。 33. 如請求項32之醫藥組合物,其中該抗體為單價且包含Fc 區,其中該Fc區包含第一多肽及第二多肽,其中該第一 多肽包含圖9中所述之Fc序列(SEQ ID NO: 12)且該第二 多肽包含圖10中所述之序列(SEQ ID NO: 13)。 34. 如請求項33之醫藥組合物,其中該抗體包含(a)第一多 肽,其包含具有如下序列之重鏈可變域: EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYWLHW 143939.doc 201019961 VRQAPGKGLEWVGMIDPSNSDTRFNPNFKDRFTISADTS KNTAYLQMNSLRAEDTAVYYCATYRSYVTPLDYWGQGT LVTVSS (SEQ ID NO: 10)、CHI序列及第一 Fc多肽;(b) 第二多肽,其包含具有如下序列之輕鏈可變域: DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKN YLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGT DFTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIKR (SEQ ID NO: 11)及CL1序列;及(c)包含第二Fc多肽之第 三多肽,其中該重鏈可變域及該輕鏈可變域呈複合物形 式存在且形成單一抗原結合臂,其中該第一Fc多肽及該 第二Fc多肽呈複合物形式存在且形成Fe區,與包含該抗 原結合臂之Fab分子相比,其增加該抗體片段之穩定 性。 35.如請求項34之醫藥組合物,其中該第一多肽包含SEQ ID NO: 12中所述之Fc序列,且該第二多肽包含SEQ ID NO: 13中所述之Fc序列。 3 6.如請求項34之醫藥組合物,其中該第一多肽包含SEQ ID NO: 13中所述之Fc序列,且該第二多肽包含SEQ ID NO: 12中所述之F c序列。 37·如請求項33之醫藥組合物,其中該抗體為MetMAb。 38. 如請求項30至37中任一項之醫藥組合物,其中該VEGF 拮抗劑為能特異性結合VEGF之適體。 39. 如請求項30至37中任一項之醫藥組合物,其中該VEGF 拮抗劑為可溶性VEGF受體蛋白,或其VEGF結合片段, 143939.doc -10- 201019961 或嵌合VEGF受體蛋白。 40. 如請求項39之醫藥組合物,其中該嵌合VEGF受體蛋白 至少包含Fit-1或KDR之細胞外域2。 41. 如請求項30至37中任一項之醫藥組合物,其中該VEGF 拮抗劑為抗VEGF抗體。 42. 如請求項41之醫藥組合物,其中該抗VEGF抗體為單株 抗體。 43. 如請求項42之醫藥組合物,其中該單株抗體為嵌合抗 體、完全人類抗體或人類化抗體。 44. 如請求項43之醫藥組合物,其中該抗VEGF抗體為貝伐 單抗、G6系列抗體、B20系列抗體或其VEGF結合片段。 45. 如請求項44之醫藥組合物,其中該抗VEGF抗體為貝伐 單抗。 46. 如請求項30至37中任一項之醫藥組合物,其進一步包含 EGFR拮抗劑。 47. 如請求項46之醫藥組合物,其中該EGFR拮抗劑具有通 式I :m is 1, 2 or 3; each R1 is independently selected from the group consisting of hydrogen, halo, hydroxy, hydroxyamino, thiol, nitro, decyl, ureido, cyano, trifluoromethyl and (CVC4 alkylene)_w-(phenyl), wherein W is a single bond, hydrazine, s or NH; or each R1 is independently selected from R9 and cyano substituted 匚丨-匚4 alkyl, wherein R9 is The following components are selected: R5, -OR6, -NR6R6, -C(〇)R7, -NHOR5, -〇C(0)R6, cyano, A and ·YR5; R5 is CVC4 alkyl; R6 independently Is hydrogen or R5; R7 is R5, -〇R6 or _NR6R6; A is selected from N-β piperidino, N-?1# morpholino, N-pyrrolidino, 4 -R6-piperazin-1-yl, imidazol-1-yl, 4_bistidin-1-yl, -(CVC4alkylene) (C02H), phenoxy, phenyl, phenylthio, C2- C4 alkenyl and -(CVC4alkylene)c(o)nr6r6; and Y is S, SO or S〇2; wherein the alkyl moiety of R5, _〇6 and _NR6R6 is optionally 1 to 3 Substituted by a substituent, and the alkyl moiety in R5, -OR6 and -NR6R6 is optionally substituted with 1 or 2 R9 groups, and wherein the 143939.doc 201019961 The alkyl moiety is optionally substituted with a halo group or R9, with the proviso that the two heteroatoms are not attached to the same carbon atom; or each R is independently selected from _NHS〇2R5, phthalimid0- (ci-C4)-alkylsulfonylamino, benzammonium, benzenesulfonylamino, triphenylphenylurea, 2-tertoxy (fluorene) pipyridyl, 2,5-di-side oxypyrrolidinyl and Rn)-(C2_C4)-alkylalkylamino, wherein R10 is selected as a free radical, _OR6, C2_C4 alkyloxy, _c(〇)r7 and -nr6r6; And wherein the R1 groups _nhs〇2rS, phthalimido-(Ci-C4)alkylsulfonylamino, benzammonium, benzenesulfonylamino, 3-phenyl Glandyl, 2, oxypyrazine, 2,5-dioxyl, butyl-1-yl and R1G_(C2-C4)-calcium-based amines, 1 or 2 Independently selected from halo, C丨-C4 alkyl, cyano, methanesulfonyl and oxime. Substituent substitution of alkoxy; or two R1 groups together with the carbon to which they are attached form a 5-8 ring , the ring comprises 1 or 2 heteroatoms selected from the group consisting of hydrazine, s and N; R is hydrogen or C--C6 alkyl group - the C--C6 alkyl group is optionally 1 3 substituents independently selected from _ group, C!-C4 alkoxy, -NR6R6 and -S02R5; η is 1 or 2' and each R3 is independently selected from hydrogen, halo, thio, c丨_c6 alkyl, -NR6R6 and CVC4 alkoxy' wherein the alkyl portion of the R3 ring is optionally independently selected from halo, (^-(24 alkoxy, -NR6R6 and -) Substituent for S02R; and R4 is azido or -(ethynyl)-Rn, wherein R11 is hydrogen or Cl-C6 alkyl'. The C"C6 alkyl group is optionally substituted with hydroxy, -OR6 or -NR6R6. The use of claim 17, wherein the EGFR antagonist is a compound of formula I selected from the group consisting of: (6,7-dimethoxyquinazolin-4-yl)-(3) -ethynylphenyl)-amine; (6,7-dimethoxyquinazolin-4-ylhydroxypropyn-1-yl)phenyl]-amine; [3-(2'-(amino hydrazine) ()ethynyl)phenyl μ(6,7-dioxaoxyquinazoline-4_:yl)-amine; (3-ethynylphenyl)-(6-nitroquinazolin-4-yl) )-amine; (6,7-dimethoxy quinazolin-4-yl)-(4-ethynylphenyl)-amine; (6,7-dimethoxy quinazolin-4-yl) -(3-ethynyl-2-methylphenyl)-amine; (6-aminoquinazolyl-4-phenyl)-(3-ethynylphenyl)-amine; (3-ethynylphenyl) )-(6-nonanesulfonylaminoquinazolin-4-yl)-amine; (3-ethynylphenyl)-(6,7-methylenedioxyquinazolin-4-yl )-amine; (6,7-dimethoxyquinazolin-4-yl)-(3-ethynyl-6-methylphenyl)-amine; (3-ethynylphenyl)_(7_ Nitroquinazolin-4-yl)-amine; (3-ethynylphenyl)-[6-(4'-toluenesulfonylamino)quina. (3_ethynylphenyl)_{6_[2,_o-phenylenedimino]ethylidene-yl-ylideneamino]quinazoline_4_yl} Amine; (3-ethynylphenyl)_ φ (6. decyl quinazolin-4-yl)-amine; (7-aminoquinazolin-4-yl)-(3-ethynylphenyl) -amine; (3-ethynylphenyl)-(7-methoxyquinazolin-4-yl)-amine; (6-indoleoxycarbonyl quinazoline-4-yl)-(3-ethynylbenzene (7-曱. oxycarbonylquinoxalin-4-yl)-(3-ethynylphenyl)-amine; [6,7-bis(2-decyloxyethoxy) quinazoline 4-(4-ethynylphenyl)-amine; (3-azidophenyl)-(6,7-dimethoxyquinazolin-4-yl)-amine; -azido-5-gas phenyl hydrazine 6,7-dimethoxyquinazolin-4-yl)-amine; (4-azidophenyl)-(6,7-dimethoxyquinazoline (4-ethynylphenyl)-(6.methanesulfonic acid-oxime "sopolinyl"-amine; (6. ethanethio-quinazolin-4-yl )-(3- 143939.doc 201019961 ethynylphenyl)-amine; (6,7-dimethoxy-anthracene) sit--4-yl)-(3-ethyl-furyl-4-fluoro-phenyl )·amine; (6,7-dimethoxy-quinazoline _4_yl)-[ 3 ((propyne-fluorenyl)-phenyl]-amine; [6,7-bis-(2-methoxy-ethoxy)-quinazoline-4-yl]-(5-ethynyl) -2-methyl-phenyl)-amine·,[6,7-bis-(2-decyloxy-ethoxy)-quinazolin-4-yl]-(3-ethynyl-4-fluoro -phenyl)-amine; [6,7-bis-(2-chloro-ethoxy)quinazoline-4(yl)-(3-ethynyl-phenyl)-amine; [6-(2 - gas-ethoxy)-7-(2-decyloxy-ethoxy)-oxime-4-yl]-(3-ethylidyl-phenyl)-amine; [6,7-double -(2-ethoxycarbonyl.ethoxy)-quinazolin-4-yl M3-ethynyl-phenyl)-amine; 2-[4-(3-ethynyl-phenylamino)_7_ ( 2-hydroxy-ethoxy)-quinazoline-6-yloxy]·ethanol; [6_(2_acetoxy-ethoxy)-7-(2-decyloxy-ethoxy) - (7-(2-carboyl-phenyl)-amine; [7-(2-methoxy-ethoxy)-6-(2-methoxy-ethoxy)-quina Oxazolin-4-yl]-(3-ethynyl-phenyl)amine; [7-(2-acetoxy-ethoxy)-6-(2-methoxy-ethoxy)- Quinoline _4_yl]_(3_ethynyl-styl)_amine; 2-[4-(3-ethynyl-phenylamino)_6_(2_carbyl-ethoxy)-啥.琳琳-7-yloxy]-ethanol; 2-[4-(3-ethynyl-phenylamino)-7-(2-methoxy-ethoxy)-quinazolin-6-yloxy ]-ethanol; 2_[4-(3-ethynyl-phenylamino)-6-(2-methoxy-ethoxy)-quinazoline-7-yloxy]-ethanol; [6· (2-acetoxy-ethoxy)-7-(2-decyloxyethoxy)-quinazolin-4-yl]-(3-ethynyl-phenyl)-amine; (3 -ethynyl-phenyl)_6_(2-methoxy-ethoxy)-7-[2-(4-indolyl-n-indolyl-indoleyl)-ethoxy]quinoline_ 4-yl}-amine; (3-ethylfastyl-phenyl)-[7.(2.methoxy-ethoxy)_6_(2·hryn-4-yl)-ethoxy)-oxime (6,7-diethoxyanthracene-1-yl)-(3-ethynylphenyl)-amine; (6,7-dibutoxyanthracene坐琳_ι· 143939.doc -6- 201019961 base)-(3-ethynylphenyl)-amine; (6,7-diisopropoxy quinazoline-anthraceyl)_ (3-B Block phenyl)-amine, (6,7--ethoxyphenyl-1-yl)-(3-ethylidyl-2-methyl-phenyl)-amine; [6,7- Bis-(2-decyloxy-ethoxy)-quinazoline-1-yl]-(3-ethynyl-2-methyl-phenyl)-amine; (3-ethynylbenzene) (6)-[6-(2-hydroxy-ethoxy)-7-(2-methoxyethoxy)-quinazolin-1-yl]-amine; [6,7-bis-(2- Hydroxy-ethoxy)-quinazolin-1-yl]-(3-ethynylphenyl)-amine; 2-[4-(3-ethynyl-phenylamino)-6-(2-A Oxy-ethoxy)-quinazoline-7-yloxy]-ethanol; (6,7-dipropoxy-quinazoline-4-yl;)_(3-ethynyl-phenyl) -amine; (6,7-diethoxy-quinazolin-4-yl)-(3-ethynyl-5-fluoro-phenyl)-amine; (6,7-diethoxy-quinazoline Benzyl-4-yl)-(3-ethylidyl-4-fluoro-phenyl)-amine; (6,7-diethyllacyl-indenyl-4-yl)-(5-ethyl-based) 2-methyl-phenyl)-amine; (6,7-diethoxy-indolyl-4-yl)-(3-ethynyl-4-methyl-phenyl)-amine; (6- Aminomethyl-7-methoxy-quinazoline-4-yl)-(3-ethylidyl-phenyl)-amine; (6-aminomethyl-7-methoxy-carbazole (6-aminocarbonylindenyl-7-nonyloxy-quinazolin-4-yl)-(3-ethynylphenyl) -amine; (6-aminocarbonylethyl-7-methoxy-quinazolin-4-yl)-(3-ethynylphenyl)-amine; (6. Aminomethyl-7-- Ethoxy- Quinazolin-4-yl)-(3-ethynylphenyl)-amine; (6-aminoethylethyl-7-ethoxy-quino-azidin-4-yl)-(3-acetylene (6-aminocarbonylcarbonylindol-7-isopropoxy-quinazolin-4-yl)-(3•ethylphenylphenyl)-amine; (6-amino group Methyl-7-propoxy-oxime 1&gt;sitin_4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylmethyl-7-methoxy-quinazoline _ 4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylethyl-7-isopropoxy-quinoxalin-4-yl)-(3-ethynylphenyl) -amine; and (6-aminocarbonylethyl_7_143939.doc 201019961 propoxyquinazolin-4-yl)-(3-ethynylphenyl)-amine; (6,7·diethoxy Quinazolin-1-yl)-(3-ethynylphenyl)-amine; (3-ethynylphenyl)-[6-(2-hydroxy-ethoxy)-7-(2-methoxy -ethoxy)-quinazolin-1-yl]-amine; [6,7-bis-(2.hydroxy-ethoxy)-quinazolin-1-yl]-(3-ethynylphenyl -Amine; [6,7-bis-(2-decyloxy-ethoxy)-quinazolin-1-yl]-(3-ethynylphenyl)-amine; (6,7-dioxin) Oxyquinazolin-1-yl)-(3-ethynylphenyl)-amine; (3-acetylene Phenyl)-(6-nonanesulfonylamino-quinazolin-1-yl)-amine; and (6-amino-quinazoline-yl)-(3-ethynylphenyl) amine. 20. 21. 22. 23. 24. 25. The use of claim 17, wherein the EGFR antagonist of formula I is Ν-(3-ethynylphenyl)-6,7-bis(2-methoxy Ethoxy)-4-quinazolinamine. The use of claim 17 wherein the EGFR antagonist Ν·(3·ethynylphenyl)-6,7-bis(2-methoxyethoxy)_ quinazoline is in the form of the hydrochloride salt. The use of claim 17, wherein the EGFR antagonist ^ (i-ethynylphenyl)-6,7-bis(2-methoxyethoxy)_4_啥 坐 坐 呈 is substantially uniform A crystalline polymorph form that exhibits a characteristic peak of the diffraction pattern of the calendering powder at 2 Θ, expressed at about 6.26, 12.48, 13.39, 16.96, 20.20, 21'1〇, 22.98, 24.46, 25.14, and 26.91. The use of Item 17, wherein the £ (31 Å antagonist is 4 (3 | fluoroanilino) ~ 7-decyloxy-6-(3-N-morpholinylpropoxy) quinazoline. The use of claim 17, wherein the EgfR antagonist is N_[3_gas_4_fluorophenyl)nonyloxy]phenyl]_6-[5-[[[2-(methylsulfanyl)ethyl]] Amino]methyl]-2-furanyl]quinazolinamine. ^ Item 17 (IV), wherein - (iv) antagonist forest (4 bromo-2-fluorobenzamide)-6-methoxy _7♦ A The use of the anti-cmet antibody is MetMAb and the EGFR antagonist is N-(3-ethynylphenyl)-6. 143939.doc 201019961 26. The use of claim 17, wherein the anti-cmet antibody is MetMAb , 7-bis(2-methoxyethoxy)-4-quinazolinamine, the VEGF antagonist is bevacizumab. 27. As claimed in claim 1. The use according to any one of the preceding claims, wherein the cancer is selected from the group consisting of breast cancer, colorectal cancer, rectal cancer, non-small cell lung cancer, non-Hodgkins lymphoma, kidney cells Cancer, prostate cancer, liver cancer, pancreatic cancer, soft tissue sarcoma, kaposi's sarcoma, carcinoid carcinoma, head and neck cancer, stomach cancer, melanoma, ovarian cancer, mesothelioma and multiple bone marrow 28. The use of claim 27, wherein the cancer is non-small cell lung cancer. The use of any one of claims 1 to 8, wherein the medicament further comprises or is used in combination with a chemotherapeutic agent A pharmaceutical composition for treating cancer in a subject, comprising a therapeutically effective amount of a c-met antagonist and a VEGF antagonist. The pharmaceutical composition according to claim 30, wherein the c-met antagonist is an antibody 32. The pharmaceutical composition of claim 31, wherein the antibody is a monovalent antibody. The pharmaceutical composition of claim 32, wherein the antibody is monovalent and comprises an Fc region, wherein the Fc region comprises a first polypeptide A second polypeptide, wherein the first polypeptide comprises the Fc sequence set forth in Figure 9 (SEQ ID NO: 12) and the second polypeptide comprises the sequence set forth in Figure 10 (SEQ ID NO: 13). The pharmaceutical composition of claim 33, wherein the antibody comprises (a) a first polypeptide comprising a heavy chain variable domain having the sequence: EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYWLHW 143939.doc 201019961 VRQAPGKGLEWVGMIDPSNSDTRFNPNFKDRFTISADTS KNTAYLQMNSLRAEDTAVYYCATYRSYVTPLDYWGQGT LVTVSS (SEQ ID NO: 10), a CHI sequence and a first Fc polypeptide; (b) a second polypeptide comprising a light chain variable domain having the sequence: DIQMTQSPSSLSASVGDRVTITCKSSQSLLYTSSQKN YLAWYQQKPGKAPKLLIYWASTRESGVPSRFSGSGSGT DFTLTISSLQPEDFATYYCQQYYAYPWTFGQGTKVEIKR (SEQ ID NO: 11) and CL1 sequence; and (c) comprising a second Fc a third polypeptide of the polypeptide, wherein the heavy chain variable domain and the light chain variable domain are present in a complex form and form a single antigen binding arm, wherein the first Fc polypeptide and the second Fc polypeptide are present in a complex form And forming an Fe region that increases the resistance compared to the Fab molecule comprising the antigen binding arm Stability fragments. 35. The pharmaceutical composition of claim 34, wherein the first polypeptide comprises the Fc sequence set forth in SEQ ID NO: 12, and the second polypeptide comprises the Fc sequence set forth in SEQ ID NO: 13. The pharmaceutical composition of claim 34, wherein the first polypeptide comprises the Fc sequence set forth in SEQ ID NO: 13, and the second polypeptide comprises the F c sequence set forth in SEQ ID NO: . 37. The pharmaceutical composition of claim 33, wherein the antibody is MetMAb. The pharmaceutical composition according to any one of claims 30 to 37, wherein the VEGF antagonist is an aptamer capable of specifically binding to VEGF. The pharmaceutical composition according to any one of claims 30 to 37, wherein the VEGF antagonist is a soluble VEGF receptor protein, or a VEGF-binding fragment thereof, 143939.doc-10-201019961 or a chimeric VEGF receptor protein. 40. The pharmaceutical composition of claim 39, wherein the chimeric VEGF receptor protein comprises at least the extracellular domain 2 of Fit-1 or KDR. The pharmaceutical composition according to any one of claims 30 to 37, wherein the VEGF antagonist is an anti-VEGF antibody. 42. The pharmaceutical composition of claim 41, wherein the anti-VEGF antibody is a monoclonal antibody. 43. The pharmaceutical composition of claim 42, wherein the monoclonal antibody is a chimeric antibody, a fully human antibody, or a humanized antibody. 44. The pharmaceutical composition of claim 43, wherein the anti-VEGF antibody is bevacizumab, a G6-series antibody, a B20-series antibody or a VEGF-binding fragment thereof. 45. The pharmaceutical composition of claim 44, wherein the anti-VEGF antibody is bevacizumab. The pharmaceutical composition according to any one of claims 30 to 37, which further comprises an EGFR antagonist. 47. The pharmaceutical composition of claim 46, wherein the EGFR antagonist has the general formula I: 其中: m為1、2或3 ; 143939.doc 201019961 各^獨立地選自由以下組成之群:氫、鹵基、μ基、 經胺基、縣、墙基、胍基、腺基、氰基、三氟甲基及 -(cvca烧基)-W_(苯基),其中w為單鍵、〇、s或即; 或各R1獨立地選自r9及經氰基取代2Ci_C4烷基,其中 R9係選自由以下組成之群:r5、_〇r6、_nr6r6、_c(〇)r7 、-NHOR、-〇c(〇)r6、氰基、A及 _yr5 ; R、c _C4烧 基;R6獨立地為氫或R5 ; R7sR5、_〇r4_nr6r6 ; A係 選自N-哌啶基、N-嗎啉基、N-吡咯啶基、4_r6_哌嗪 基、咪唑-1-基、4-吡啶酮·〗_基、_(Ci_C4伸烷 基)(C〇2H)、苯氧基、苯基、苯基硫基、C2-C4烯基及 •(CVC:4 伸烧基)c(〇)NR6R6 ;及 γ為 S、8〇或 s〇2 :其中 R、-OR6及-NR6R6中之烷基部分視情況經1至3個卣基取 代基取代,且R5、_〇R6及-Nr6r6中之烷基部分視情況經 1或2個R9基團取代,且其中該等視情況取代基之烷基部 分視情況經鹵基或R9取代’限制條件為兩個雜原子不連 接於同一碳原子; 或各R1獨立地選自-NHS02R5、鄰苯二醯亞胺基_(Cl_ C4)-烷基磺醯基胺基、苯甲醯胺基、苯磺醯基胺基、3_ 苯基脲基、2-側氧基吡咯啶-1-基、2,5-二側氧基吡咯啶_ 1-基及R10-(C2-C4)-烷醯基胺基,其中R10選自齒基、_〇r6 、C2-C4烷醯氧基、_(:(0)117及_NR6R6 ;且其中該等R1基 團-NHSC^R5、鄰苯二醯亞胺基_(Cl_C4)烷基磺醯基胺 基、苯甲酿胺基、苯磺醯基胺基、3-苯基脲基、2-側氧 基吡咯啶-1-基、2,5-二側氧基吡咯啶-1-基及R1Q-(C2-C4)- 143939.doc • 12- 201019961 貌醯基胺基視情況經1或2個獨立地選自鹵基、Ci-C^烷 基、氰基、曱烷磺醯基及(^-(:4垸氧基之取代基取代; 或兩個R1基團連同其所連接之碳一起形成5_8員環,該 環包括1或2個選自〇、s及N之雜原子; R2為氫或CrC6烷基,該CrCe烷基視情況經1至3個獨 立地選自齒基、C「C4烷氧基、-NR6R6及-S02R5之取代基 取代; π為1或2,且各R3獨立地選自氫、鹵基、羥基、Cl-c6 烧基、烷氧基,其中該等r3基團之烷基部 分視情況經1至3個獨立地選自鹵基、CrCU烷氧基、-NR6R6及-S02R之取代基取代;及 R4為疊氮基或-(乙炔基)-rh,其中R&quot;為氫或Cl-c6烷 基’該CVC6烷基視情況經羥基、_〇R6或_NR6R6取代。 48.如請求項46之醫藥組合物,其中該EGFR拮抗劑為選自 由以下組成之群之式I化合物: (6,7-二甲氧基喹唑啉_4_基)-(3-乙炔基苯基)_胺;(6,7-二甲氧基喹唑啉-4-基)-[3-(3,-羥基丙炔-1-基)苯基]-胺; [3-(2'_(胺基甲基)-乙炔基)苯基卜(6 7_二甲氧基喹唑啉_4_ 基)-胺;(3-乙炔基苯基)-(6-硝基喹唑啉_4_基)_胺;(6,7-二曱氧基喹唑啉-4-基H4-乙炔基苯基)_胺;(6,7·二甲氧 基喹唑啉-4-基)-(3-乙炔基·2-甲基苯基)·胺;(6-胺基喹唑 琳-4-基)-(3-乙炔基苯基)-胺;(3_乙炔基苯基)_(6_甲烷磺 醯基胺基喹唑啉-4-基)-胺;(3-乙快基苯基)-(6,7-亞甲基 二氧基喹唑啉-4-基)·胺;(6,7_二曱氧基喹唑啉_4_基 143939.doc •13· 201019961 乙炔基-6-甲基苯基胺;(3_乙炔基苯基)_(7_硝基喹唑 啉-4-基)-胺;(3-乙炔基苯基)-[6-(4'-甲苯磺醯基胺基)喹 唑啉-4-基]-胺;(3_乙炔基苯基)_{6-[2,-鄰苯二醯亞胺基_ 乙-Γ-基-磺醯基胺基]喹唑啉-4-基}•胺;(3-乙炔基苯基)-(6-胍基喹唑啉-4-基)-胺;(7-胺基喹唑啉·4-基)-(3-乙炔 基苯基)-胺;(3-乙炔基苯基)-(7-曱氧基喹唑啉-4_基)-胺;(6-曱氧羰基喹唑啉-4-基)-(3-乙炔基苯基)-胺;(7-曱 氧羰基喹唑啉-4-基)-(3-乙炔基苯基)-胺;[6,7-雙(2-曱氧 基乙氧基)喹唑啉-4-基]-(3-乙炔基苯基)-胺;(3-疊氮基 孀 苯基)-(6,7-二甲氧基喹唑啉_4_基胺;(3_疊氮基·5_氣苯 基Η6,7-二曱氧基喹唑啉-4-基)-胺;(4·疊氮基苯基)_ (6,7·二甲氧基喹唑啉_4_基)_胺;(3_乙炔基笨基)_(6_曱烷 項醯基-喹唾啉_4_基)_胺;(6_乙烷硫基·喹唑啉_4_基)_(3_ 乙炔基苯基)-胺;(6,7_二甲氧基-喹唑啉_4-基)_(3_乙炔 基-4-氟-苯基)_胺;(6,7_二曱氧基-喹唑啉_4_基)_[3_(丙 快-1’-基)-苯基]-胺;[6,7_雙_(2_甲氧基—乙氧基)_喹唑啉_ 4-基]-(5-乙炔基_2_曱基-苯基)_胺;[6,7_雙_(2_曱氧基-乙 G 氧基)-喧唾啉-4-基]-(3-乙炔基-4-氟-苯基)-胺;[6,7·雙-(2-氣-乙氧基)·喹唑嘴_4基]_(3_乙炔基_笨基)胺;[6_(2_ 氣-乙氧基)-7-(2-甲氧基-乙氧基)_喹唑啉-4-基]-(3-乙炔 基-笨基)-胺;[6,7-雙-(2-乙醯氧基-乙氧基)-喹唑啉-4-基]乙炔基-苯基)-胺;2-[4-(3-乙炔基-苯基胺基)-7-(2_羥基-乙氧基)-喹唑啉-6-基氧基]-乙醇;[6-(2-乙醯氧 基-乙氧基)-7-(2-甲氧基·乙氧基)_啥唑淋-4-基]-(3-乙炔 143939.doc •14- 201019961 基-苯基)-胺;[7-(2-氣-乙氧基)-6-(2-甲氧基-乙氧基)-喹 唾啉-4-基]-(3-乙炔基-苯基)-胺;[7-(2-乙醯氧基-乙氧 基)-6-(2-甲氧基-乙氧基)-喧唑琳_4·基]-(3-乙炔基-苯基)-胺,2-[4-(3-乙快基-本基胺基)-6-(2-經基-乙氧基)-嗜β坐 琳-7-基氧基]-乙醇;2-[4-(3-乙炔基-苯基胺基)-7-(2-曱 氧基-乙氧基)-喹唑啉-6-基氧基]-乙醇;2-[4-(3-乙炔基· 苯基胺基)-6-(2-曱氧基-乙氧基)-啥嗤琳_7_基氧基]-乙 醇》[6-(2 -乙酿氧基-乙氧基)-7-(2-甲氧基-乙氧基)_啥β坐 啉-4-基]-(3-乙炔基·苯基)-胺;(3-乙炔基-苯基χ6_(2_甲 氧基-乙氧基)-7-[2-(4-曱基-旅°秦-1-基)-乙氧基]_啥唾琳_ 4-基卜胺;(3_乙炔基-苯基)·[7-(2-甲氧基-乙氧基)_6_(2_ 嗎啉-4-基)-乙氧基)-喹唑啉-4-基]-胺;(6,7-二乙氧基嗜 唑啉-1-基)-(3-乙炔基苯基)-胺;(6,7-二丁氧基喹唑琳 基)-(3-乙炔基苯基)-胺;(6,7-二異丙氧基喹唑啉-^基)· (3-乙块基苯基)-胺;(6,7-二乙氧基啥唾琳-ΐ_基)_(3_乙炔 基-2-甲基-苯基)-胺;[6,7-雙-(2-甲氧基-乙氧基)_喹唑琳_ 1-基]-(3-乙炔基-2-甲基-苯基)-胺;(3-乙块基苯基) (2 -經基-乙氧基)-7-(2 -甲氧基-乙氧基)-噎唾琳_ι_基]_ 胺,[6,7-雙-(2-經基-乙氧基)-啥吐啭·_1-基]_(3_乙炔基笨 基)-胺;2-[4-(3 -乙快基-苯基胺基)-6-(2-甲氧基_乙氧美) 啥β坐琳-7-基氧基]-乙醇;(6,7 -二丙氧基-啥唾琳基) (3-乙快基-苯基)-胺;(6,7_二乙氧基-啥〇坐啦_4_基)_(3_乙 炔基-5-氟-苯基)-胺;(6,7-二乙氧基-喧唾琳基)_(3_乙 炔基-4-氣-苯基)-胺;(6,7-二乙氧基-喧唾啦_4_基)_(5_乙 143939.doc -15- 201019961 炔基-2-甲基-苯基)-胺;(6,7-二乙氧基-喹唑啉-4-基)-(3-乙炔基-4-甲基-苯基)-胺;(6-胺基甲基-7-甲氧基·喹唑 啉-4-基)-(3-乙炔基-苯基)-胺;(6-胺基甲基-7-曱氧基-喹 β坐琳-4-基)-(3 -乙快基苯基)-胺;(6 -胺基幾基曱基-7-曱氧 基-啥唾淋_4-基)-(3-乙快基苯基)-胺;(6-胺基幾基乙基_ 7-曱氧基-喧°坐琳-4-基)-(3-乙炔基苯基)-胺;(6-胺基幾基 曱基-7-乙氧基·喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6-胺 基羰基乙基-7-乙氧基-喹唑啉·4-基)-(3-乙炔基苯基)-胺;(6-胺基羰基曱基-7·異丙氧基-喹唑啉-4-基)-(3-乙炔 基苯基)-胺;(6_胺基羰基甲基-7-丙氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6-胺基羰基甲基-7-甲氧基-喹唑啉-4-基)-(3·乙炔基苯基)-胺;(6-胺基羰基乙基-7-異丙氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;及(6-胺基羰基乙基-7-丙氧基-喹唑啉-4-基)-(3-乙炔基苯基)-胺;(6,7-二乙氧基 喹唑啉-1-基)-(3-乙炔基苯基)-胺;(3-乙炔基苯基)-[6-(2-羥基-乙氧基)-7-(2-甲氧基-乙氧基)-喹唑啉-1-基]-胺; [6,7-雙-(2-羥基·乙氧基)-喹唑啉-1-基]-(3-乙炔基苯基)-胺;[6,7-雙-(2-曱氧基-乙氧基)-喹唑啉-1-基]_(3_乙炔基 苯基)-胺;(6,7-二甲氧基喹唑啉-1-基)-(3-乙炔基苯基)-胺;(3-乙炔基苯基)·(6·曱烷磺醯基胺基-喹唑啉-1-基)-胺;及(6-胺基-喹唑啉-1-基)-(3-乙炔基苯基)-胺。 49.如請求項46之醫藥組合物,其中該式I之EGFR拮抗劑為 Ν-(3·乙炔基苯基)-6,7-雙(2-甲氧基乙氧基)-4-喹唑啉 胺。 143939.doc -16· 201019961 50. 如請求項46之醫藥組合物,其中該EGFR拮抗劑N-(3-乙 炔基苯基)-6,7-雙(2-曱氧基乙氧基)-4-喹唑啉胺呈鹽酸鹽 形式。 51. 如請求項46之醫藥組合物,其中該EGFR拮抗劑N-(3-乙 ’炔基苯基)-6,7-雙(2-曱氧基乙氧基)-4-喹唑啉胺係呈實質 4 上均勻之結晶多晶型物形式,該形式展現X光粉末繞射 圖之特徵峰以2Θ。表示在約6.26、12.48、13.39、16.96、 20.20、21.10、22.98、24.46、25.14及 26.91。 52. 如請求項46之醫藥組合物,其中該EGFR拮抗劑為4-(3,- 氣-4'-氟苯胺基)-7-甲氧基-6-(3-N-嗎啉基丙氧基)啥唑 淋。 53. 如請求項46之醫藥組合物,其中該EGFR拮抗劑為N-[3-氣-4-[(3·氟苯基)曱氧基]苯基]_6_[5_[[[2_(甲基磺醯基)乙 基]胺基]甲基]-2-吱喃基]-4-喹《坐琳胺。 54. 如請求項46之醫藥組合物,其中該EGFR拮抗劑為4-(4- ^ 溴_2_氟苯胺基)_6_甲氧基-7-(1-曱基裉咬-4-基甲氧基)啥 吐琳。 55. 如請求項46之醫藥組合物,其中該抗cmet抗體為 ; MetMAb,該EGFR拮抗劑為N-(3-乙炔基笨基)_6,7_雙(2- 曱氧基乙氧基)-4-喹唑啉胺,該^^(^拮抗劑為貝伐單 抗。 56. 如請求項30至37中任一項之醫藥組合物,其中該癌症係 選自由以下組成之群:乳癌、結腸直腸癌、直腸癌、非 小細胞肺癌、非霍奇金氏淋巴瘤、腎細胞癌、前列腺 143939.doc 17 201019961 癌、肝癌、胰腺癌、軟組織肉瘤、卡波西氏肉瘤、類癌 瘤、頭頸癌、胃癌、黑素瘤、卵巢癌、間皮瘤及多發性 骨髓瘤。 5 7.如請求項56之醫藥組合物,其中該癌症為非小細胞肺 癌。 58.如請求項30至37中任一項之醫藥組合物,其進一步包含 化學治療劑。 143939.doc 18-Wherein: m is 1, 2 or 3; 143939.doc 201019961 Each is independently selected from the group consisting of hydrogen, halo, mu, amine, county, wall, sulfhydryl, gland, cyano , trifluoromethyl and -(cvcaalkyl)-W_(phenyl), wherein w is a single bond, hydrazine, s or ie; or each R1 is independently selected from r9 and substituted by a cyano group 2Ci_C4 alkyl, wherein R9 It is selected from the group consisting of r5, _〇r6, _nr6r6, _c(〇)r7, -NHOR, -〇c(〇)r6, cyano, A and _yr5; R, c _C4 alkyl; R6 independent The ground is hydrogen or R5; R7sR5, _〇r4_nr6r6; A is selected from N-piperidinyl, N-morpholinyl, N-pyrrolidinyl, 4_r6-piperazinyl, imidazol-1-yl, 4-pyridone ·〗 _ base, _ (Ci_C4 alkylene) (C〇2H), phenoxy, phenyl, phenylthio, C2-C4 alkenyl and • (CVC: 4 extended alkyl) c (〇) NR6R6 And γ are S, 8〇 or s〇2: wherein the alkyl moiety of R, -OR6 and -NR6R6 is optionally substituted with 1 to 3 mercapto substituents, and R5, _〇R6 and -Nr6r6 The alkyl moiety is optionally substituted with 1 or 2 R9 groups, and wherein the alkyl moiety of the substituent is optionally halogenated Or R9 is substituted by the 'restriction condition that two heteroatoms are not attached to the same carbon atom; or each R1 is independently selected from -NHS02R5, phthalic acid imine-(Cl_C4)-alkylsulfonylamino group, benzene Formamyl, phenylsulfonylamino, 3-phenylurea, 2-oxopyrrolidin-1-yl, 2,5-di-oxypyrrolidin-1-yl and R10-(C2- a C4)-alkylalkylamino group, wherein R10 is selected from the group consisting of dentate, _〇r6, C2-C4 alkoxy, _(:(0)117 and _NR6R6; and wherein the R1 group -NHSC^R5 , phthalimide imino-(Cl_C4)alkylsulfonylamino, benzylamino, phenylsulfonylamino, 3-phenylureido, 2-oxopyrrolidine-1- Base, 2,5-di-oxypyrrolidin-1-yl and R1Q-(C2-C4)- 143939.doc • 12-201019961 The mercaptoamine group is optionally selected from halo by 1 or 2 , a Ci_C alkyl group, a cyano group, a decanesulfonyl group, and a substituent of (^-(: 4 methoxy); or two R1 groups together with the carbon to which they are attached form a 5-8 ring, which The ring includes 1 or 2 heteroatoms selected from the group consisting of hydrazine, s and N; R 2 is hydrogen or CrC6 alkyl, and the CrCe alkyl group is optionally selected from the group consisting of dentate groups, C "C4" Substituted by a substituent of oxy, -NR6R6 and -S02R5; π is 1 or 2, and each R3 is independently selected from the group consisting of hydrogen, halo, hydroxy, Cl-c6 alkyl, alkoxy, wherein the r3 group The alkyl moiety is optionally substituted with from 1 to 3 substituents independently selected from halo, CrCU alkoxy, -NR6R6 and -S02R; and R4 is azido or -(ethynyl)-rh, wherein R&quot; Is a hydrogen or Cl-c6 alkyl group. The CVC6 alkyl group is optionally substituted with a hydroxy group, 〇R6 or _NR6R6. The pharmaceutical composition of claim 46, wherein the EGFR antagonist is a compound of formula I selected from the group consisting of: (6,7-dimethoxyquinazolin-4-yl)-(3-acetylene (phenyl))-amine; (6,7-dimethoxyquinazolin-4-yl)-[3-(3,-hydroxypropyn-1-yl)phenyl]-amine; [3-( 2'-(Aminomethyl)-ethynyl)phenyl (6 7-dimethoxyquinazolin-4-yl)-amine; (3-ethynylphenyl)-(6-nitroquinazoline) (6,7-dimethoxy quinazolin-4-yl H4-ethynylphenyl)-amine; (6,7-dimethoxyquinazolin-4- (6-aminoacetylquin-2-yl)-(3-ethynylphenyl)-amine; (3-ethynylbenzene) (6)-(6-methanesulfonylaminoquinazolin-4-yl)-amine; (3-ethyl-bromophenyl)-(6,7-methylenedioxyquinazolin-4- (6,7-dimethoxy quinazoline _4_yl 143939.doc •13· 201019961 ethynyl-6-methylphenylamine; (3_ethynylphenyl)_(7 _Nitroquinazolin-4-yl)-amine; (3-ethynylphenyl)-[6-(4'-toluenesulfonylamino)quinazolin-4-yl]-amine; (3 _acetylene Phenyl)_{6-[2,-o-phenylenediminoimido-ethylidene-yl-sulfonylamino]quinazolin-4-yl}•amine; (3-ethynylphenyl) -(6-nonylquinazolin-4-yl)-amine; (7-aminoquinazoline-4-yl)-(3-ethynylphenyl)-amine; (3-ethynylphenyl) -(7-oxime quinazolin-4-yl)-amine; (6-fluorenyloxyquinazolin-4-yl)-(3-ethynylphenyl)-amine; (7-oxiranylcarbonyl) Quinazolin-4-yl)-(3-ethynylphenyl)-amine; [6,7-bis(2-decyloxyethoxy)quinazolin-4-yl]-(3-ethynyl Phenyl)-amine; (3-azidosulfonylphenyl)-(6,7-dimethoxyquinazolin-4-ylamine; (3_azido-5-phenylphenylphosphonium 6,7) -dimethoxyquinazolin-4-yl)-amine; (4. azidophenyl)-(6,7-dimethoxyquinazolin-4-yl)-amine; (3_acetylene (6-decane fluorenyl-quinoxaline _4_yl)-amine; (6-ethanethio-quinazoline-4-yl)-(3_ethynylphenyl)- Amine; (6,7-dimethoxy-quinazoline-4-yl)-(3-ethynyl-4-fluoro-phenyl)-amine; (6,7-dimethoxy-quinazoline _4_基)_[3_(C-fast-1'-yl)-phenyl]-amine; [6,7_double_(2_methoxy -Ethoxy)-quinazoline-4-yl]-(5-ethynyl-2-indoleyl-phenyl)-amine; [6,7-bis-(2-methoxy-g-oxygen) ))-喧 喧 啉 -4- -4-yl]-(3-ethynyl-4-fluoro-phenyl)-amine; [6,7·bis-(2-gas-ethoxy)·quinazolium _4 Alkyl]-(3-ethynyl-phenyl)amine; [6_(2_g-ethoxy)-7-(2-methoxy-ethoxy)-quinazolin-4-yl]-(3 -ethynyl-styl)-amine; [6,7-bis-(2-acetoxy-ethoxy)-quinazolin-4-yl]ethynyl-phenyl)-amine; 2-[ 4-(3-ethynyl-phenylamino)-7-(2-hydroxy-ethoxy)-quinazolin-6-yloxy]-ethanol; [6-(2-ethyloxy)- Ethoxy)-7-(2-methoxyethoxymethyl)-oxazole-4-yl]-(3-acetylene 143939.doc •14-201019961 phenyl-phenyl)-amine;[7- (2-Gas-ethoxy)-6-(2-methoxy-ethoxy)-quinoxalin-4-yl]-(3-ethynyl-phenyl)-amine; [7-(2 -ethoxycarbonyl-ethoxy)-6-(2-methoxy-ethoxy)-oxazolidine-4-yl]-(3-ethynyl-phenyl)-amine, 2-[4 -(3-ethyl-fast-n-ylamino)-6-(2-carbo-ethoxy)-ββ-lin-7-yloxy]-ethanol; 2-[4-(3-acetylene) Phenyl-phenylamino)-7-(2-decyloxy -ethoxy)-quinazolin-6-yloxy]-ethanol; 2-[4-(3-ethynyl-phenylamino)-6-(2-decyloxy-ethoxy)-啥嗤琳_7_基基]-ethanol][6-(2-ethoxy-ethoxy-ethoxy)-7-(2-methoxy-ethoxy)-啥β-s- s- -4- (3-(3-ethynyl-phenyl)-amine; (3-ethynyl-phenylindole 6_(2-methoxy-ethoxy)-7-[2-(4-mercapto-L. -1-yl)-ethoxy]-啥啥琳_ 4-ylbumin; (3_ethynyl-phenyl)·[7-(2-methoxy-ethoxy)_6_(2_morpholine 4-yl)-ethoxy)-quinazolin-4-yl]-amine; (6,7-diethoxyoxazolin-1-yl)-(3-ethynylphenyl)-amine (6,7-dibutoxyquinazolinyl)-(3-ethynylphenyl)-amine; (6,7-diisopropoxyquinazoline-yl)·(3-ethyl (phenyl))-amine; (6,7-diethoxyhydrazinyl-indoleyl)-(3-ethynyl-2-methyl-phenyl)-amine; [6,7-bis-( 2-methoxy-ethoxy)-quinazoline-1-yl]-(3-ethynyl-2-methyl-phenyl)-amine; (3-ethylphenylphenyl) (2-an -ethoxy)-7-(2-methoxy-ethoxy)-噎 琳 _ ι ι ι ι ι ι ι 胺 胺 胺 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 6 Spit ·_1-yl]_(3_ethynylphenyl)-amine; 2-[4-(3-ethylidyl-phenylamino)-6-(2-methoxy-ethoxyxo) 啥β (7,7-dipropoxy-hydrazinyl) (3-ethyl-p-phenyl)-amine; (6,7-diethoxy- Squatting _4_yl)_(3_ethynyl-5-fluoro-phenyl)-amine; (6,7-diethoxy-hydrazinyl)_(3_ethynyl-4- Gas-phenyl)-amine; (6,7-diethoxy-hydrazino- 4_yl)-(5_B 143939.doc -15- 201019961 alkynyl-2-methyl-phenyl)- Amine; (6,7-diethoxy-quinazolin-4-yl)-(3-ethynyl-4-methyl-phenyl)-amine; (6-aminomethyl-7-methoxy (quinoquinazolin-4-yl)-(3-ethynyl-phenyl)-amine; (6-aminomethyl-7-decyloxy-quino-[beta]-yl-4-yl)-(3- Ethyl phenyl)-amine; (6-amino benzyl-7-methoxy-indolyl-4-yl)-(3-ethyl-bromophenyl)-amine; (6-amine (6-Amino-indenyl-7-ethoxyquine) (oxazolin-4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylethyl-7-ethoxy-quinazoline 4-(4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylindol-7-isopropoxy-quinazolin-4-yl)-(3-ethynylphenyl) -amine; (6-aminocarbonylmethyl-7-propoxy-quinazolin-4-yl)-(3-ethynylphenyl)-amine; (6-aminocarbonylmethyl-7-- Oxy-quinazolin-4-yl)-(3.ethynylphenyl)-amine; (6-aminocarbonylethyl-7-isopropoxy-quinazolin-4-yl)-(3 -ethynylphenyl)-amine; and (6-aminocarbonylethyl-7-propoxy-quinazolin-4-yl)-(3-ethynylphenyl)-amine; (6,7- Diethoxyquinazolin-1-yl)-(3-ethynylphenyl)-amine; (3-ethynylphenyl)-[6-(2-hydroxy-ethoxy)-7-(2 -methoxy-ethoxy)-quinazolin-1-yl]-amine; [6,7-bis-(2-hydroxyethyloxy)-quinazolin-1-yl]-(3- Ethynylphenyl)-amine; [6,7-bis-(2-decyloxy-ethoxy)-quinazolin-1-yl]-(3-ethynylphenyl)-amine; (6, 7-Dimethoxyquinazolin-1-yl)-(3-ethynylphenyl)-amine; (3-ethynylphenyl)·(6·decanesulfonylamino-quinazoline- 1-yl)-amine; and (6-amino-quinazolin-1-yl)-(3-ethynylphenyl)-amine49. The pharmaceutical composition of claim 46, wherein the EGFR antagonist of formula I is Ν-(3. ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinoline Oxazoline amine. The pharmaceutical composition of claim 46, wherein the EGFR antagonist N-(3-ethynylphenyl)-6,7-bis(2-decyloxyethoxy)- 4-quinazolinamine is in the form of the hydrochloride salt. 51. The pharmaceutical composition of claim 46, wherein the EGFR antagonist N-(3-ethyl 'alkynylphenyl)-6,7-bis(2-decyloxyethoxy)-4-quinazoline The amine is in the form of a substantially uniform crystalline polymorph which exhibits a characteristic peak of the X-ray powder diffraction pattern of 2 Å. Expressed at approximately 6.26, 12.48, 13.39, 16.96, 20.20, 21.10, 22.98, 24.46, 25.14 and 26.91. 52. The pharmaceutical composition of claim 46, wherein the EGFR antagonist is 4-(3,-gas-4'-fluoroanilino)-7-methoxy-6-(3-N-morpholinylpropionate Oxy) oxazole. 53. The pharmaceutical composition of claim 46, wherein the EGFR antagonist is N-[3- gas-4-[(3.fluorophenyl)decyloxy]phenyl]_6_[5_[[[2_(A Alkylsulfonyl)ethyl]amino]methyl]-2-indolyl]-4-quino 54. The pharmaceutical composition of claim 46, wherein the EGFR antagonist is 4-(4-^bromo-2-fluoroanilino)-6-methoxy-7-(1-indenyl-4-yl-4-yl Methoxy) 啥吐琳. 55. The pharmaceutical composition of claim 46, wherein the anti-cmet antibody is; MetMAb, the EGFR antagonist is N-(3-ethynylphenyl)-6,7-bis(2-decyloxyethoxy) -4- quinazolinamine, the antagonist is a bevacizumab. The pharmaceutical composition according to any one of claims 30 to 37, wherein the cancer is selected from the group consisting of breast cancer , colorectal cancer, rectal cancer, non-small cell lung cancer, non-Hodgkin's lymphoma, renal cell carcinoma, prostate 143939.doc 17 201019961 cancer, liver cancer, pancreatic cancer, soft tissue sarcoma, Kaposi's sarcoma, carcinoid tumor , head and neck cancer, gastric cancer, melanoma, ovarian cancer, mesothelioma, and multiple myeloma. The pharmaceutical composition of claim 56, wherein the cancer is non-small cell lung cancer. The pharmaceutical composition according to any one of 37, which further comprises a chemotherapeutic agent. 143939.doc 18-
TW098134878A 2008-10-17 2009-10-14 Combination therapy TW201019961A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10651308P 2008-10-17 2008-10-17

Publications (1)

Publication Number Publication Date
TW201019961A true TW201019961A (en) 2010-06-01

Family

ID=41667148

Family Applications (1)

Application Number Title Priority Date Filing Date
TW098134878A TW201019961A (en) 2008-10-17 2009-10-14 Combination therapy

Country Status (3)

Country Link
AR (1) AR073852A1 (en)
TW (1) TW201019961A (en)
WO (1) WO2010045344A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2793545A1 (en) * 2010-05-14 2011-11-17 Genentech, Inc. Treatment methods
EP2611928B1 (en) 2010-09-03 2016-04-27 Academia Sinica Anti-c-met antibody and methods of use thereof
WO2012042421A1 (en) * 2010-09-29 2012-04-05 Pfizer Inc. Method of treating abnormal cell growth
WO2012136685A1 (en) 2011-04-04 2012-10-11 Pieris Ag Methods and compositions for anti-vegf and anti-c-met therapy
GB201113754D0 (en) 2011-08-09 2011-09-21 Glaxo Group Ltd Composition
RU2014114617A (en) * 2011-09-19 2015-10-27 Дженентек, Инк. COMBINED TREATMENTS CONTAINING C-MET ANTAGONISTS AND B-RAF ANTAGONISTS
EP2708556B1 (en) * 2012-09-12 2018-11-07 Samsung Electronics Co., Ltd Pharmaceutical composition for the use in a combination therapy for prevention or treatment of c-met or angiogenesis factor induced diseases
WO2014046198A1 (en) * 2012-09-19 2014-03-27 シスメックス株式会社 Method for obtaining information about hepatocellular carcinoma, and marker and kit for obtaining information about hepatocellular carcinoma
KR102049990B1 (en) * 2013-03-28 2019-12-03 삼성전자주식회사 Fusion protein comprising anti-c-Met antibody and VEGF binding fragment
MX2016012285A (en) * 2014-03-24 2017-01-23 Genentech Inc Cancer treatment with c-met antagonists and correlation of the latter with hgf expression.
TWI782930B (en) 2016-11-16 2022-11-11 美商再生元醫藥公司 Anti-met antibodies, bispecific antigen binding molecules that bind met, and methods of use thereof
KR20240017986A (en) * 2017-09-26 2024-02-08 더 리전트 오브 더 유니버시티 오브 캘리포니아 Compositions and methods for treating cancer
JP2022547274A (en) 2019-09-16 2022-11-11 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Radiolabeled MET binding protein for immunoPET imaging

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RS51326B (en) * 2004-08-05 2010-12-31 Genentech Inc. Humanized anti-cmet antagonists
MX2010012290A (en) * 2008-05-14 2011-02-21 Amgen Inc Combinations vegf(r) inhibitors and hepatocyte growth factor (c-met) inhibitors for the treatment of cancer.

Also Published As

Publication number Publication date
AR073852A1 (en) 2010-12-09
WO2010045344A1 (en) 2010-04-22

Similar Documents

Publication Publication Date Title
RU2601892C2 (en) Combined c-met and egfr antagonists therapy
US20230082122A1 (en) Diagnostic and therapeutic methods for cancer
TW201019961A (en) Combination therapy
JP6821693B2 (en) Treatment and diagnosis for cancer
US20110262436A1 (en) Treatment method
KR102129636B1 (en) Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
US20180256711A1 (en) Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists
CN110418851A (en) The treatment of cancer and diagnostic method
KR20080003422A (en) Anti-egfr antibody therapy based on an increased copy number of the egfr gene in tumor tissues
EP3355902A1 (en) Combination of a pd-1 axis binding antagonist and an alk inhibitor for treating alk-negative cancer
JP2019515670A (en) Methods for monitoring and treating cancer
TW202027784A (en) Diagnostic and therapeutic methods for kidney cancer
US20220041734A1 (en) Methods of treating cancer with an anti-pd-l1 antibody
ES2502217T3 (en) NLRR-1 antagonists and their uses
AU2015213411A1 (en) Combination therapy with c-met and EGFR antagonists