EP0831880A1 - Antibody and antibody fragments for inhibiting the growth of tumors - Google Patents

Antibody and antibody fragments for inhibiting the growth of tumors

Info

Publication number
EP0831880A1
EP0831880A1 EP96921457A EP96921457A EP0831880A1 EP 0831880 A1 EP0831880 A1 EP 0831880A1 EP 96921457 A EP96921457 A EP 96921457A EP 96921457 A EP96921457 A EP 96921457A EP 0831880 A1 EP0831880 A1 EP 0831880A1
Authority
EP
European Patent Office
Prior art keywords
amino acid
variable region
chain variable
acid sequence
molecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP96921457A
Other languages
German (de)
French (fr)
Other versions
EP0831880A4 (en
Inventor
Neil I. Goldstein
Nicholas A. Giorgio
Steven Tarran Jones
Jose William Saldanha
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ImClone LLC
Original Assignee
ImClone Systems 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 ImClone Systems Inc filed Critical ImClone Systems Inc
Publication of EP0831880A1 publication Critical patent/EP0831880A1/en
Publication of EP0831880A4 publication Critical patent/EP0831880A4/en
Withdrawn legal-status Critical Current

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention is directed to antibodies and antibody fragments useful in inhibiting the growth of certain tumor cells.
  • Recent research has uncovered the important role of growth factor receptor tyrosine kinases in the etiology and progression of human malignancies. These biological receptors are anchored by means of a transmembrane domain in the membranes of cells that express them. An extracellular domain binds to a growth factor. The binding of the growth factor to the extracellular domain results in a signal being transmitted to the intracellular kinase domain. The transduction of this signal contributes to the events that are responsible for the proliferation and differentiation of the cells.
  • EGF epidermal growth factor
  • TGF-alpha transforming growth factor alpha
  • EGF receptor tyrosine kinases are found in unusually high numbers on human tumors. For example, many tumors of epithelial origin express increased levels of EGF receptor on their cell membranes. Examples of tumors that express EGF receptors include glioblastomas, as well as cancers of the lung, breast, head and neck, and bladder. The amplification and or overexpression of the EGF receptors on the membranes of tumor cells is associated with a poor prognosis.
  • Antibodies especially monoclonal antibodies, raised against tumor antigens have been investigated as potential anti-tumor agents. Such antibodies may inhibit the growth of tumors through a number of mechanisms. For example, antibodies may inhibit the growth of tumors immunologically through antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • antibodies may compete with growth factors in binding to their receptors. Such competition inhibits the growth of tumors that express the receptor.
  • toxins are conjugated to antibodies raised against tumor antigens.
  • the antibody portion directs the conjugate to the tumor, which is killed by the toxin portion.
  • U.S. Patent No. 4,943,533 describes a murine monoclonal antibody called 225 that binds to the EGF receptor.
  • the patent is assigned to the University of California and licensed exclusively to ImClone Systems Incorporated.
  • the 225 antibody is able to inhibit the growth of cultured EGFR-expressing tumor lines as well as the growth of these tumors in vivo when grown as xenografts in nude mice.
  • no clinical response was observed when up to 300 mg of murine 225 antibodies was administered to humans. See Divgi et al., J. Natl. Cancer Inst. £1, 97-104 (1991). See Masui et al, Cancer Res. 44, 5592-5598 (1986).
  • a disadvantage of using murine monoclonal antibodies in human therapy is the possibility of a human anti-mouse antibody (HAMA) response due to the presence of mouse Ig sequences.
  • HAMA human anti-mouse antibody
  • This disadvantage can be minimized by replacing the entire constant region of a murine (or other non-human mammalian) antibody with that of a human constant region. Replacement of the constant regions of a murine antibody with human sequences is usually referred to as chimerization.
  • the chimerization process can be made more effective by also replacing the variable regions - other than the hypervariable regions or the complementarity- determining regions (CDRs), of a murine antibody with the corresponding human sequences.
  • the variable regions other than the CDRs are also known as the variable framework regions (FRs).
  • the replacement of the constant regions and non-CDR variable regions with human sequences is usually referred to as humanization.
  • the humanized antibody is less immunogenic (i.e. elicits less of a HAMA response) as more murine sequences are replaced by human sequences.
  • both the cost and effort increase as more regions of a murine antibodies are replaced by human sequences.
  • Another approach to reducing the immunogenicity of antibodies is the use of antibody fragments.
  • an article by Aboud-Pirak et al, Journal of the National Cancer Institute £0, 1605-1611 (1988) compares the anti-tumor effect of an anti-EGF receptor antibody called 108.4 with fragments of the antibody.
  • the tumor model was based on KB cells as xenografts in nude mice. KB cells are derived from human oral epidermoid carcinomas, and express elevated levels of EGF receptors.
  • Aboud-Pirak et al. found that both the antibody and the bivalent F(ab') 2 fragment retarded tumor growth in vivo, although the F(ab') 2 fragment was less efficient.
  • An object of the present invention is the discovery of such new anti-tumor agents that combine the advantageous features of monoclonal antibodies, antibody fragments and single chain antibodies.
  • polypeptide lacking the constant region and the variable light chain of an antibody, the polypeptide comprising the amino acid sequence N Y G V H (SEQ IDNO: 1),GVIWSGGNTDYNTPFTSR (SEQ ID NO: 2), or V I W S G G N T D Y N T P F T S (SEQ ID NO: 3).
  • the polypeptide may be conjugated to an effector molecule, such as a molecule that inhibits tumor growth.
  • the invention further is directed to DNA encoding such polypeptides.
  • the invention also includes polypeptides consisting of the amino acid sequence N
  • the invention also includes a molecule having the constant region of a human antibody and the variable region of monoclonal antibody 225 conjugated to a cytoxic agent such as doxorubicin, taxol, or cis-diamminedichloroplatinum (cisplatin).
  • a cytoxic agent such as doxorubicin, taxol, or cis-diamminedichloroplatinum (cisplatin).
  • the invention further includes a method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of a polypeptide lacking the constant region of the variable light chain of an antibody, the polypeptide comprising the amino acid sequence NYGVH, GVIWSGGNTD YNTPFTSR,orVIWSGGNTDYNTPFTS.
  • Another aspect of the invention is a method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of a polypeptide consisting of the amino acid sequence NYGVH, GVIWSGGNTDYNTPF TSR,orVIWSGGNTDYNTPFTS.
  • the invention further includes a method for significantly inhibiting the growth of tumor cells that express the EGF receptor in a human.
  • the method comprises administering to the human an effective amount of a molecule having the constant region of a human antibody and the variable region of monoclonal antibody 225, both in the prescence of and, in particular, in the absence of, cytotoxic molecules, such as chemotherapeutic agents.
  • Figure 1 Effect of 225 on the growth of established A431 tumor xenografts in nude mice. Animals were injected with 10 7 cells in the flank. Treatments, consisting of PBS or 1 mg/animal of 225 twice weekly for 5 weeks, were begun when tumors reached an average volume of 2-300 mm 3 . Volumes and Remission Index (RI) were determined as described in the "Examples" section.
  • FIG. 1 Effect of 225 and chimerized 225 (C225) on the growth of established A431 tumor xenografts in nude mice. Animals were treated with lmg/mouse of PBS twice weekly for 5 weeks. A: Average tumor volumes; B: Remission Index. The apparent tumor regression in the PBS control group at day 37 was due to the death of 3 out of the 10 animals within the group at this time and the concommitant decrease in overall tumor volume.
  • C225 chimerized 225
  • FIG. 3 Effect of C225 on the growth of established A431 xenografts in nude mice. Animals were treated with lmg of C225 or PBS twice weekly for 5 weeks. The average tumor volume of the C225 group showed statistically significant biological effects compared to control (see text) A: Average tumor volumes (asterisks show statistical significance with respect to control); B: Remission Index.
  • FIG. 4 Dose response of C225 on the growth of established A431 xenografts in nude mice. Animals were treated with PBS, 1, 0.5, or 0.25 mg/animal twice weekly for 5 weeks as described in Materials Methods. Animals treated with 1 mg/dose of C225 showed statistically significant biological effects compared to control (see text). A: Average tumor volumes (asterisks define statistical signifiance with respect to control); B: Remission Index. The drop in RI for the 250 ug dose group on day 47 resulted from the re-appearance of a tumor in an apparent tumor-free animal. (In this instance, the effect of C225 was transient.)
  • FIG. 7 FACS analysis of EGFR expression on human prostatic carcinoma cell lines.
  • LNCaP human prostatic carcinoma, androgen-dependent
  • DU 145 and PC-3 human prostatic carcinoma, androgen-independent
  • A431 human epidermoid carcinoma
  • FIG. 8 Inhibition of EGF-induced phosphorylation of the EGFR by C225.
  • LNCaP, DU 145, and PC-3 monolayers were stimulated with EGF in the presence or absence of C225.
  • Cells were lysed, subjected to SDS PAGE, blotted, and screened with a mouse monoclonal antibody to PTyr (UBI, Lake Placid).
  • Lane A no additions (basal level of EGFR phosphorylation);
  • Lane B stimulation of EGFR with 10 ng/ml EGF for 15 minutes at room temperature in the absence of C225;
  • Lane C stimulation of EGFR with EGF in the presence of 10 ug/ml of C225.
  • Figure 9 Growth inhibition of established DU 145 xenografts by C225.
  • One million DU 145 cells in matrigel were innoculated into nude mice (males, nu nu). After tumors reached an average volume of approximately 100 mm 3 (day 20), animals were randomized (10 animals per group) and treated with either PBS (control) or C225 (0.5 mg/dose, lOx). Animal were treated for 35 days and followed for an additional 3 weeks. Mice that were tumor-free or carrying small tumors were maintained for an additional 3 months. Significance (shown by astericks in Figure 3A) was determined by a Student's T-test and a p value ⁇ 0.5 was considered significant.
  • B growth characteristics for tumors in the PBS group
  • C growth characteristics for tumors in the C225-treated groups.
  • Figure 10 Effects of C225 on tumor elimination and surviva .
  • the complete elimination of tumors during the course of the study was defined by a Remission Index (RI). Animal mortality during the study was considered a treatment failure and included in the analysis.
  • B Survival curve.
  • the empty and filled circles in Figure 10 have the same meanings as in Figure 9.
  • Figure 11. Schematic representation of the pKNIOO mammalian expression vector used for the expression of the kapp light chains of the chimeric C225 and reshaped human H225 antibody.
  • Figure 12 Schematic representation of the pGlD105 mammalian expression vector used for the expression of the heavy chains of the chimeric C225 and reshaped human H225 antibody.
  • Figure 15 DNA (SEQ ID NO: 10) and peptide (SEQ ID NO: 11) sequences of the kappa light chain variable region of theC225 antibody.
  • Figure 17 DNA (SEQ ID NO: 14) and peptide (SEQ ID NO: 15) sequences of the kappa light chain variable region of the C225 antibody with the modified leader sequence from the kappa light chain of L7'CL antibody (28).
  • Figure 18 Typical example of the results of a cell ELISA to measure the binding affinty of chimeric C225 and reshaped human H225 (225RK A /225RH A ) antibodies to epidermal growth factor receptor expressed on the surface of A431 cells.
  • Figure 20 DNA (SEQ ID NO: 18) and peptide (SEQ ID NO: 19) sequences of the first version (225RH A ) of the heavy chain variable region of the reshaped human H225 antibody.
  • Figure 21 Amino acid sequences of the two versions (225RK A and 225RK B ) of the kappa light chain variable region of the reshaped human H225 antibody (SEQ ID NO: 20), (SEQ ID NO: 21), (SEQ ID NO: 22), (SEQ ID NO: 23). Residues are numbered according to Kabat et al (20). Mouse framework residues conserved in the reshaped human frameworks are highlighted in bold.
  • 225RH C , 225RH D , 225RH E of the heavy chain variable region of the reshaped human H225 antibody (SEQ ID NO: 24), (SEQ ID NO: 25), (SEQ ID NO: 26), (SEQ ID NO: 27), (SEQ ID NO: 28), (SEQ ID NO: 29), (SEQ ID NO: 30).
  • Residues are numbered according to Kabat et al. (20). Mouse framework residues conserved in the reshaped human frameworks are highlighted in bold.
  • a polypeptide lacking the constant region and the variable light chain of an antibody comprises the first and second heavy chain complementarity determining regions of monoclonal antibody 225. These regions have the following amino acid sequences:
  • the peptide comprising the first and second complementarity determining regions mentioned above may be obtained by methods well known in the art.
  • the polypeptides may be expressed in a suitable host by DNA that encodes the polypeptides and isolated.
  • the DNA may be synthesized chemically from the four nucleotides in whole or in part by methods known in the art. Such methods include those described by Caruthers in Science 230, 281-285 (1985).
  • the DNA may also be obtained from murine monoclonal antibody 225, which was described by Mendelsohn, et al U.S. Patent No. 4,943,533. This antibody was deposited in the American Type Culture Collection, Bethesda, Maryland on June 7, 1995. (Accession number 11935).
  • Methods for obtaining the variable heavy chain region of antibodies are known in the art. Such methods include, for example, those described in U.S. patents by Boss (Celltech) and by Cabilly (Genentech). See U.S. Patent Nos. 4,816,397 and 4,816,567, respectively.
  • the DNA encoding the protein of the invention may be replicated and used to express recombinant protein following insertion into a wide variety of host cells in a wide variety of cloning and expression vectors.
  • the host may be prokaryotic or eukaryotic.
  • the polypeptide may contain either NYGVH, GVIWSGGNTDYNTPF TSR, orVIWSGGNTDYNTPFTS.
  • the polypeptide may contain the sequence N Y G V H, and either of the sequences GVIWSGGNTD YNTPFTSR, orVIWSGGNTDYNTPFTS.
  • the polypeptide may also be conjugated to an effector molecule.
  • the effector molecule performs various useful functions such as, for example, inhibiting tumor growth, permitting the polypeptide to enter a cell such as a tumor cell, and directing the polypeptide to the appropriate location within a cell.
  • the effector molecule may be a cytotoxic molecule.
  • the cytotoxic molecule may be a protein, or a non-protein organic chemotherapeutic agent.
  • suitable chemotherapeutic agents include, for example, doxorubicin, taxol, and cisplatin.
  • effector molecules suitable for conjugation to the polypeptides of the invention include signal transduction inhibitors, ras inhibitors, and cell cycle inhibitors.
  • signal transduction inhibitors include protein tyrosine kinase inhibitors, such as quercetin (Grazieri et al , Biochim. Biophs. Acta 714.415 (1981)); lavendustin A (Onoda et al, J. Nat. Prod.52, 1252 (1989)); and herbimycin A (Ushara et al, Biochem. Int., 41, 831 (1988)).
  • Ras inhibitors include inhibitors of ras farnesylation, such as the benzodiazepine peptidomimetics described by James et al in Science 2601937 (1993), which have the formula shown below:
  • R is H or CH 3 ; and X is Methione, Serine, Leucine, or an ester or amide derivative thereof.
  • Proteins and non-protein chemotherapeutic agents may be conjugated to the polypeptides by methods that are known in the art. Such methods include, for example, that described by Greenfield et al , Cancer Research £0, 6600-6607 (1990) for the conjugation of doxorubicin and those described by Arnon et al, Adv. Exp. Med. Biol. 201 79-90 (1991) and by Kiseleva et al, Mol. Biol. (USSR) 25, 508-514 (1991) for the conjugation of platinum compounds.
  • the invention further includes a modified antibody having the constant region of a human antibody, and the hypervariable region of monoclonal antibody 225.
  • modified antibodies are optionally conjugated to an effector molecule, such as a cytotoxic agent.
  • the variable region other than the hypervariable region may also be derived from the variable region of a human antibody. Such an antibody is said to be humanized. Methods for making humanized antibodies are known in the art. Methods are described, for example, in Winter, U.S. Patent No. 5,225,539.
  • the most thorough method for humanization of the 225 antibodies is CDR- grafting.
  • the regions of the mouse antibody that are directly involved in binding to antigen are grafted into human variable regions to create "reshaped human" variable regions.
  • These fully humanized variable regions are then joined to human constant regions to create complete "fully humanized” antibodies.
  • the human variable regions into which the 225 antibodies CDRs will be grafted must be carefully selected, and it is usually necessary to make a few amino acid changes at critical positions within the framework regions (FRs) of the human variable regions.
  • FRs framework regions
  • the reshaped human H225 variable regions include up to a single amino acid change in the FRs of the selected human kappa light chain variable region and as many as twelve amino acid changes in the FRs of the selected human heavy chain variable region.
  • the DNA sequences coding for these reshaped human H225 heavy and kappa light chain variable region genes are joined to DNA sequences coding for the human ⁇ l and human K constant region genes, respectively.
  • the reshaped human H225 antibody is then expressed in mammalian cells and tested, in comparison with mouse M225 antibody, and chimeric C225 antibody for binding to human EGF receptor expressed on the surface of A431 cells.
  • variable region of the antibody outside of the hypervariable region may also be derived from monoclonal antibody 225.
  • the entire variable region is derived from murine monoclonal antibody 225, and the antibody is said to be chimerized, i.e., C225.
  • Methods for making chimerized antibodies are known in the art. Such methods include, for example, those described in U.S. patents by Boss (Celltech) and by Cabilly (Genentech). See U.S. Patent Nos. 4,816,397 and 4,816,567, respectively.
  • the constant region of the modified antibodies may be of any human class, i.e., IgG, IgA, IgM, IgD, and IgE. Any subclass of the above classes is also suitable, e.g., IgGl, IgG2, IgG3 and IgG4, in which IgGl is preferred.
  • any of the effector molecules mentioned above in connection with conjugation to a polypeptide can also be conjugated to chimeric or humanized antibodies of the invention.
  • Doxorubicin, taxol, and cisplatin are prefened.
  • the polypeptides and antibodies of the invention significantly inhibit the growth of tumor cells when administered to a human in an effective amount.
  • the optimal dose can be determined by physicians based on a number of parameters including, for example, age, sex, weight, severity of the condition being treated, the active ingredient being administered, and the route of administration.
  • a serum concentration of polypeptides and antibodies that permits saturation of EGF receptors is desirable.
  • a concentration in excess of approximately 0.1 nM is normally sufficient.
  • a dose of 100 mg/m 2 of C225 provides a serum concentration of approximately 20 nM for approximately eight days.
  • doses of antibodies may be given weekly in amounts of 10- 300 mg/m 2 .
  • Equivalent doses of antibody fragments should be used at more frequent intervals in order to maintain a serum level in excess of the concentration that permits saturation of EGF receptors.
  • Some suitable routes of administration include intravenous, subcutaneous, and intramuscle administration. Intravenous administration is preferred.
  • the peptides and antibodies of the invention may be administered along with additional pharmaceutically acceptable ingredients.
  • additional pharmaceutically acceptable ingredients include, for example, immune system stimulators and chemotherapeutic agents, such as those mentioned above.
  • the chimeric and humanized antibodies significantly inhibit tumor growth in humans, even in the absence of other anti-tumor agents, including other chemotherapeutic agents, such as cisplatin, doxorubicin, taxol, and their derivatives.
  • Significant inhibition may mean the shrinkage of tumors by at least 20%, preferably 30%, and more preferably 50%. In optimal cases, 90% and even 100% shrinkage of tumors is achieved.
  • significant inhibition may mean an RI greater than 0.3, preferably greater than 0.4, and more preferably greater than 0.5.
  • the significant inhibition of tumor growth and or increase in RI manifests itself in numerous ways. For example, there is an increase in life expectency and/or a stabilization of previously aggresive tumor growth.
  • C225 may be substituted for the chemotherapeutic agents, and achieve comparable results.
  • Example III-l the results shown in Example III-l indicate that, while the in vitro inhibitory properties of 225 and C225 are comparable, the in vivo effects of the antibodies differ considerably.
  • Antibody isotype does not play a significant role in the differences seen between 225 and C225 (e.g., mouse IgGl vs. human IgGl).
  • a recent report indicates that neither 225 nor C225 induced complement mediated lysis to any degree and the ADCC reactivity of these antibodies appeared to be species specific. Naramura et al, Immunol. Immunother. 22, 343-349 (1993). Therefore, if inhibition of A431 xenografts was mediated through immune responses, 225 should be the more potent antibody because of its ability to activate the murine effector cells involved in ADCC. The opposite is, in fact, the case.
  • Example III-2 describes the biological effects of C225 on the activation of the
  • the EGFR expressed by A431 cells can be stimulated by exogenously added ligand (EGF) and C225 can abrogate activation of the receptor.
  • Figure 8 shows the results of similar studies with the prostatic lines. The addition of EGF to LNCaP, PC-3, and DU 145 induced phosphorylation of the EGFR that was blocked by C225 with high efficiency. These data indicate that C225 effectively inhibits ligand-activated EGFR signalling pathways, and has anti-tumor activity when EGFR activation is required for growth in vivo.
  • C225 The ability of C225 to inhibit tumor growth in vivo was tested against established DU 145 xenografts in athymic nude mice. DU 145 cells were innoculated at 10 6 cells per animals in combination with matrigel. Tumors developed in 100% of the animals within 20 days. Preliminary experiments had shown that a dose level of 1 mg (lOx) induced significant tumor inhibition. For these studies, C225 was injected at a 0.5 mg (lOx) dose level.
  • Example III-2 clearly shows that C225 was capable of inhibiting the growth of established, EGFR-positive DU 145 xenografts and could induce long-lived tumor remissions in a high percentage of treated animals. These results could not be predicted from the in vitro data.
  • KB cells human epidermoid carcinoma
  • KB xenografts did not respond to a treatment regimen including a 1 mg dose (xlO) of C225, a level able to induce complete remissions in 100% of animals carrying established A431 tumors.
  • xlO 1 mg dose
  • Example III-2 treatment of mice innoculated with DU 145 tumor cells with C225 alone at a 0.5 mg dose (xlO) led to significant tumor regressions in all treated animals. Sixty percent of the mice were in complete remission following termination of the treatment.
  • Blockage of receptor activaton by C225 also has clinical implications for the treatment of metastatic prostatic carcinoma in humans, especially during the late stages of the disease.
  • Example 1-1 Cell Lines and Media
  • A431 cells were routinely grown in a 1:1 mixture of Dulbecco's modified Eagle's medium and Ham's F-12 supplemented with 10% fetal bovine serum, 2mM L- glutamine, and antibiotics.
  • the androgen-independent and dependent human prostatic carcinoma cell lines (DU 145, PC-3 and LNaP) were obtained from the ATCC (Rockville MD) and routinely maintained in RPMI 1640 medium (Sigma, St. Louis, MO) supplemented with 10% fetal bovine serum (Intergen, Purchase NY) and 2 mM L-gluatmine (Sigma). Cells were checked regularly for the presence of mycoplasma.
  • the 225 antibody was grown as ascites in pristane primed Balb/c mice. Ascites fluid was purified by HPLC (ABX and Protein G) and determined to be >95% pure by SDS PAGE.
  • C225 doxorubicin conjugates (C225-DOX) were prepared using a modification of the method described by Greenfield et al. , Cancer Research 50, 6600-6607 (1990). Briefly, Doxorubicin was reacted with the crosslinking agent PDPH (3-[2- pyridyldithio]propionyl hydrazide) (Pierce Chemical Co.) to form the acyl hydrazone derivative doxorubicin 13-[3-(2-pyridyldithiol) propionyl] hydrazone hydrochloride.
  • PDPH 3-[2- pyridyldithio]propionyl hydrazide
  • C225 was thiolated with the reagent N-succinimydyl 3-(pyridyldithio) propionate and reacted with doxorubicin hydrazone to form a conjugate containing a hydrazide as well as a disulfide bond.
  • the complex was purified by gel filtration at neutral pH.
  • the C225-doxorubicin conjugate was stable at neutral to alkaline pH (pH 7-8) and was stored at 4C. The conjugate was readily hydrolyzed at pH 6, releasing active Doxorubicin.
  • the media containing the 225 mouse hybridoma cell line was expanded to one liter in tissue culture flasks.
  • Total cell RNA was prepared by lysing washed cells in guanidine isothiocyanate containing 2-mercaptoethanol, shearing the solution in a dounce homogenizer to degrade cell DNA and layering the preparation on a 10 ml cesium chloride cushion. After centrifugation at 24,000 rpm for 16 hr. the pellet was resuspended in Tris-EDTA (TE) buffer and precipitated with ethanol. The poly A(+) mRNA fraction was isolated by binding to and elution from oligo dT cellulose.
  • a cDNA library was prepared using the poly A (+) mRNA as template and oligo dT as the primer.
  • the second strand was synthesized by nick translation using RNase H and DNA polymerase I.
  • the double-stranded DNA was passed through a 2 ml Sepharose G75 column to remove oligo dT and small entities.
  • the purified DNA was then ligated into a polylinker with the sequence:
  • Isolates with the longest cDNA inserts were subcloned in a plasmid vector (Eco RI-Bam HI fragments for heavy (H) chain V regions and Eco RI-Hpa I fragments for light (L) chain variable (V) regions) and DNA sequenced.
  • the subcloned fragments contained the complete V region and a small portion of associated mouse constant (C) region.
  • C mouse constant
  • a total of eight L chain cDNAs were sequenced and represent four different mRNAs.
  • Three full-length H chain cDNAs were sequenced encoding the same V region and a portion of the correct gamma 1 C region.
  • Three other isolates containing gamma 2a sequence were also identified but were not studied further.
  • L chain cDNA To identify the correct L chain cDNA, a sample of mouse 225 antibody was sequenced by automated Edman degradation after first separating the H and L chains by SDS reducing gel electrophoresis and blotting to membranes. The sequence obtained for the L chain matched one of the cDNAs. This isolate was rearranged to J5 and was found to be 91% homologous with Vk T2. The H chain V region was found to be 96% homologous with VH 101 subgroup VII- 1.
  • Example I-4B Adaption of cDNAs and Construction of Expression Vectors
  • V regions were adapted for expression by ligating the body of each to a synthetic DNA duplex encoding the sequence between the closest unique restriction site to the V/C junction and the exact boundary of the V region. To this was ligated a second, short intron sequence which, when joined, restores a functional splice donor site to the V region.
  • a Bam HI site At the end of the intron for the L chain is a Bam HI site and at the end of the H chain intron is a Hind III site.
  • the adapted L Chain V region was then isolated as a Xba I-Bam HI fragment (the Xba I site was in the original linker used for cDNA cloning) while the adapted H chain V region was isolated as a Xho I- Hind III fragment.
  • the final vector was identified by restriction mapping and identified as pdHL2-ch225.
  • Example I-4C Expression of Chimeric 225 in Transfected Hybridoma Cells
  • the pdHL2-ch225 plasmid was introduced into hybridoma Sp2/0 Agl4 cells by protoplast fusion.
  • the bacteria harboring the plasmid were grown to an optical density of 0.5 at 600 nm at which time chloramphenicol was added to arrest growth and amplify the plasmid copy number.
  • the following day the bacteria were treated with lysozyme to remove the cell wall and the resulting protoplasts were fused to the hybridoma cells with polyethylene glycol 1500. After fusion, the cells were grown in antibodies to kill any surviving bacteria and were plated in 96-well plates.
  • the selection medium (containing methotrexate (MTX) at 0.1 ⁇ M) was added after 24-48 hours to allow only the transfected cells to grow, by virtue of their expression of the marker gene (dehydrofolate reductase) present on the expression plasmid.
  • MTX methotrexate
  • the productivity of the subclones was tested by seeding cells at 2 X 10 5 cells per ml in growth medium and measuring the accumulated antibody on day 7.
  • the two highest producers from the first subcloning were lines SdER6.25 and SdER14.10. These were subcloned a second time and the final three candidate lines were designated SdER6.25.8, SdER6.25.49, and SdER14.10.1. Clone SdER6.25.8 was selected based on expression of antibody.
  • the antibodies were found to inhibit the proliferation of cultured A431 cells to the same extent (Table 2).
  • 225 and C225 were able to block EGF-induced phosphorylation of the EGFR in A431 cells.
  • the relative binding affinity of the antibodies was determined using an ELISA protocol previously described by Lokker et al J. Immunol. 146, 893-898 (1991). Briefly, A431 cells (10 4 or 10 5 per well) were grown in 96 well microtiter plates overnight at 37°C. Cells were fixed with 3.7% neutral buffered formalin for 10 minutes at room temperature. After washing three times with PBS, wells were blocked with 1% bovine serum albumin in Hank's balanced salt solution for two hours at room temperature. C225 or 225 were added to the wells at various concentrations (serial dilutions starting at 50 nM).
  • the apparent binding affinities of M225 and C225 were also determined using the InAcoreTM (Pharmacia Biosensor, Piscataway NJ; manufacturer's application note 301 and O'Shannessy et al, Anal. Biochem. 212, 457-468 (1993). Briefly, soluble recombinant EGFR was immobilized on sensor chips via amino groups as described by the manufacturer. Real time binding parameters of 225 and C225 to EGFR was established at various antibody concentrations and the apparent Kd was calculated from the binding rate constants obtained via non linear fitting using BiaevaluationTM 2.0 Software.
  • Example II-3 In vitro Inhibition of Cell Growth with 225 and C225
  • the in vitro inhibitory activity of 225 and C225 was determined by plating A431 cells (300-500 per well) in 96 microtiter plates in complete growth medium. After adding C225 or 225 in various concentrations (4 replicates per concentration), plates were incubated for 48 hours at 37°C followed by a 24 hour pulse with 3H-thymidine. Cells were harvested, collected on filter mats and counted in a Wallace Microbeta scintillation counter to determine percent inhibition. Percent inhibition compares the decrease in 3H thymidine incorporation of antibody-treated cells with cells grown in the absence of antibody.
  • Athymic nude mice (nu/nu; 6-8 weeks old females) were obtained from Charles River Laboratories. Animals (10 mice per treatment group) were innoculated in the right flank with 10 7 A431 cells in 0.5 ml of Hank's balanced salt solution. Mice were observed until tumors were visible (about 7-12 days) and had reached an average volume of 150-300 mm 3 . At that time, antibody therapy was begun. The therapy included twice weekly intraperitoneal injections (varying concentrations in 0.5 ml of PBS) over 5 weeks. U 1 animals received injections of PBS. Tumors were measured two times per week and volumes calculated using the following formula: ⁇ /6 x larger diameter x (smaller diameter) 2 .
  • RI Remission Index
  • Example III-l The Capacity of the Antibodies to Inhibit the Growth of A431 Xenografts in Nude Mice
  • mice were innoculated in the flanks with A431 cells. Tumors of 150-300 mm 3 appeared by day 7-10. Refering to Experiments 1-4 in Table 3, animals were then randomized and injected with PBS or 225 (Exp 1), PBS, 225, or C225 (Exp 2); and PBS or C225 (Exp 3 and 4). In Experiments 1-3, animals received injections of 1 mg of antibody (in 0.5 ml PBS) twice weekly over 5 weeks for a total dose of 10 mg of antibody per animal. In Exp 4, animals received one of three possible doses: 1, 0.5, and 0.25 mg/injection for total doses of 10, 5, and 2.5 mg, respectively. Tumors were measured twice weekly over the course of treatment. Tumor-free animals and animals with small tumors continued to be monitored for 2-3 months following the sacrificing of animals with large tumors.
  • Figure 1 shows the effect of 225 on the growth of A431 tumors in nude mice
  • Figure 4 shows the results of the dose reponse experiment (Exp 4). All animals receiving 1 mg/injection underwent complete remission and remained tumor free for over 100 days following termination of the antibody injections ( Figure 4A and B; Table 3). These results are highly significant with p values varying from p ⁇ 0.006 on day 33 to p ⁇ 0.0139 on day 59. In Experiments 2 and 3, about 40% of the animals receiving the 1 mg dose of C225 underwent complete remission although C225 showed significant tumor regression in Exp 3 ( Figure 3).
  • the overall inhibition of tumor growth was not statistically significant because of the large variations in tumor volume among animals of both the PBS and the 0.5 mg groups.
  • the 0.5 mg dose group in Exp 4 had a higher RI than the 1 mg dose group in Exp 3. This result may be attributed to the effects of tumor burden.
  • the average starting volume for tumors in the 0.5 mg dose group was 160 mm 3 , there was great variability in tumor size among individual animals. A number of animals carried smaller tumors ( ⁇ 100 mm 3 ) that are most susceptible to the biological effect of C225.
  • Percent inhibition is defined as the decrease in 3-H thymidine incorporation of antibody-treated samples (4 replicates/concentration) versus cells growing in the absence of antibody.
  • Table 3 represents a comparison of complete tumor remissions in athymic nude mice carrying established A431 tumors following treatment with PBS, 225, or C225 twice weekly for 5 weeks. Animals were treated with 1 mg of antibody in 0.5 ml of PBS by the intraperitoneal route except for study 4, which is a dose response experiment in which mice were given 1, 0.5, or 0.25 mg/injection. Tumor measurements were done as described above. This chart describes the RI at the time when the animals (PBS control and test) carrying large tumors were euthanized. All animals showing complete remissions or small tumors were followed for an additional 2-3 months. The differences in total number of animals results from death of mice within these treatment groups during the course of the experiments. Table 3. REMISSION INDICES FOR ANIMALS INNOCULATED WITH A431 CELLS AND TREATED WITH 225 OR C225
  • RI The Remission Index
  • RI is defined as the fraction of mice that were tumor free on the day when the PBS control mice and test animals with large tumors were euthanized. A complete remission at the 0.25 mg dose level showed a subsequent recurrance of tumor (day 47).
  • Example IH-2 Inhibition of Growth of Established Human Prostatic Carcinoma Xenografts in Nude Mice
  • Example III-2A FACS Analysis of C225 Binding to DU 145. PC-3 and LNCaP
  • EGF receptor on DU 145, PC-3 and LNCaP cells was determined by FACS analysis.
  • Cells were grown to near confluency in complete medium, removed from the flasks with non-enzymatic dissociation buffer (Sigma), and resuspended at 5-10 x 10 5 per tube in 100 ul of cold H-BSA (Hanks balanced salt solution containing 1% BSA).
  • H-BSA Hypothalamic Base
  • Ten micrograms C225 or an irrelevant myeloma- derived human IgGl were added to the tubes and incubated on ice for 60 minutes.
  • MFI Mean Fluroescence Intensity
  • Example III-2B Phosphorylation Assays on PC-3. DU 145. and LNCaP Cells
  • Phosphorylation assays were performed on PC-3, DU 145, and LNCaP cells to determine if the EGF receptors expressed by these cells were functional and inhibited by C225. Assays and Western blot analysis were performed as previously described by Gill et al, Nature 293, 305-307 (1981). Briefly, DU 145, PC-3, and LNCaP cells were grown to 90% confluency in complete medium and then starved in DMEM-0.5% calf serum 24 hours prior to experimentation. Cells were stimulated with EGF in the presence or absence of C225 for 15 minutes at room temperature. Monolayers were then washed with the ice cold PBS containing 1 mM sodium orthovanadate.
  • Athymic nude mice (nu/nu; 6-8 weeks old males; Charles River Labs, Wilmington MA) were innoculated subcutaneously in the right flank with 10 6 DU 145 in 0.2 ml of Hank's balanced salt solution mixed with 0.2 ml of matrigel. Mice were observed until tumors were visible (about 14-20 days post challenge) and had reached an average volume of about 100 mm 3 . Animals were weighed and randomly divided into treatment groups (10 animals per group). Antibody therapy, which included twice weekly intraperitoneal injections of 0.5 mg of C225 over 5 weeks, was begun. Control animals received injections of PBS.
  • Example IH-3 Biological Activity of Peptides Containing CDR Regions of 225
  • peptides were dissolved in PBS at a concentration of 1 mg/ml.
  • A431 cells were plated at 1000 cells per well in 96 well plates. Peptides were added at various concentrations. The chimeric C225 antibody and an irrelevant, isotype- matched immunoglobulin were used as a positive and negative U Is, respectively. Plates were incubated for 72 hours at 37 °C and pulsed overnight with 3 H-thymidine. Cells were harvested and counted in a liquid scintillation counter. Percent inhibition is defined as the decrease in 3-H thymidine incorporation of antibody or peptide treated cells compared to cells grown in the absence of antibody or peptide.
  • A431 cells are inhibited by C225 and by heavy chain CDR-1 and heavy chain CDR-2 of monoclonal antibody 225.
  • isotype- matched irrelevant antibody and U 1 peptide did not inhibit A431 cells.
  • C225-DOX The biological activity of C225-DOX was evaluated in vitro using EGFR expressing cell lines A431 , KB and MDA-468 as well as EGFR non-expressing cell lines Molt-4 and SK-MEL-28. EGF receptor expression was verified by FACS analysis using C225 and C225-DOX conjugate. Assays were conducted over a 72h incubation period using 3 [H] -thymidine and WST-1 as a read out. In all assays with EGFRc expressing cell lines, i.e., A431, KB and MDA-468 cells, C225-DOX exhibited high inhibition of cell proliferation when compared to no treatment or hlgGl U Is.
  • DMEM Dulbecco's Modified Eagles Medium
  • FCS Foetal Calf Serum
  • RNA ribonuceic acid
  • mRNA messenger RNA
  • DNA deoxyribonucleic acid
  • ds-DNA double-stranded DNA
  • PCR polymerase chain reaction
  • ELISA enzyme linked immunoabsorbant assay
  • Materials Dulbecco's Modified Eagles Medium (DMEM); Foetal Calf Serum (FCS); rib
  • RNA isolation kit is obtained from Stratgene (USA) while the 1 st strand cDNA synthesis kit is purchased from Pharmacia (UK). All the constituents and equipment for the PCR-reactions, including AmpliTaq®DNA polymerase, are purchased from Perkin Elmer (USA).
  • the TA Cloning® kit is obtained from Invitrogen (USA) and the Sequenase® DNA sequencing kit is purchased from Amersham International (UK). Agarose (UltraPureTM) is obtained from Life Technologies (UK).
  • the WizardTM PCR Preps DNA Purification Kit, the MagicTM DNA Clean-up System and XL 1 Blue competent cells are purchased from Promega Corporation (USA). All other molecular biological products are purchased from New England Biolabs (USA). Nunc-Immuno Plate MaxiSorpTM immunoplates are obtained from Life Technologies (UK). Both the goat anti-human IgG, Fc ⁇ fragment specific, antibody and the goat anti-human IgG (H+L) / horseradish peroxidase conjugate are purchased from Jackson
  • TMB substrate A and substrate B are obtained from Kirkegaard-Pery (USA). All other products for both ELISAs are obtained from Sigma (UK).
  • Microplate Manager® data analysis software package is purchased from Bio-Rad (UK).
  • the molecular modelling package QUANTA is obtained from the Polygen Corporation (USA) and the IRIS 4D workstation is purchased from Silicon Graphics (USA).
  • Example IV-3 PCR cloning and sequencing of the mouse variable region genes
  • the mouse M225 hybridoma cell line is grown, in suspension, using DMEM supplemented with 10% (v/v) FCS, 50 Units/ml penicillin / 50 ⁇ g/ml streptomycin and 580 ⁇ g/ml L-glutamine. Approximately 10 8 viable cells are harvested, while the supernatent from the hybridoma cells is assayed by ELISA to confirm that they are producing a mouse antibody. From the 10 8 cells total RNA is isolated using a RNA Isolation kit according to the manufacturers instructions. The kit uses a guanidinium thiocyanate phenol-chloroform single step extraction procedure as described by Chomczynski and Sacchi (6).
  • a 1 st Strand cDNA Synthesis kit is employed to produce a single-stranded DNA copy of the M225 hybridoma mRNA using the NotI-(dT) lg primer supplied in the kit. Approximately 5 ⁇ g of total RNA is used in a 33 ⁇ l final reaction volume. The completed reaction mix is then heated to 90 °C for 5 min, to denature the RNA-cDNA duplex and inactivate the reverse transcriptase, before being chilled on ice.
  • PCR- reaction tubes are loaded into a Perkin Elmer 480 DNA thermal cycler and cycled (after an initial melt at 94 °C for 1.5 min) at 94 °C for 1 min, 50 °C for 1 min and 72 °C for 1 min over 25 cycles.
  • a final extension step at 72 °C for 10 min is carried out before the reactions are cooled to 4 °C. Except for between the annealing (50 °C) and extension (72 °C) steps, when an extended ramp time of 2.5 min is used, a 30 sec ramp time between each step of the cycle is employed.
  • Colonies containing the plasmid, with a correctly sized insert are identified by PCR-screening the colonies using the pCRTMII Forward and pCRTMII Reverse oligonucleotide primers described in Table 6 according to the method of G ⁇ ssow and Clackson (8).
  • the putative positive clones identified are finally double-stranded plasmid DNA sequenced using the Sequenase®DNA Sequencing kit according to the method of Redston and Kern (9).
  • Example IV-4 Construction of chimeric genes
  • the cloned mouse leader- variable region genes are both modified at the 5'- and 3'- ends using PCR-primers to create restriction enzyme sites for convenient insertion into the expression vectors, a Kozak sequence for efficient eukaryotic translation of the mRNA encoding the respective immunoglobulin chains (10) and a splice-donor site for the correct RNA splicing of the variable and constant region genes.
  • a Hndlll site is added to the 5 '-end of both mouse variable region genes, however, different restriction sites attached to the 3 '-end of the mouse variable region genes i.e. a BamHl site at the 3'-end of the V ⁇ gene and aXbal site at the 3'-end of the V ⁇ gene.
  • PCR-reactions are prepared according to the method for the construction of chimeric genes in Kettleborough et al. (11), using the primers C225V H 5' and
  • the PCR-products are column purified using a WizardTMPCR Preps DNA Purification kit according to the manufacturers instructions, digested with the appropriate restriction enzymes, as is plasmid pUC19, and separated on a 1% agarose / TBE buffer (pH8.8) gel.
  • the heavy and kappa light chain variable region genes are excised from the agarose gel and purified using a Wizard' PCR Preps DNA Purification kit.
  • the pUC19 is also excised from the agarose gel and purified using the MagicTMDNA Clean-up System as per the manufacturers instructions.
  • the heavy and kappa light chain variable region genes are then separately ligated into the purified pUC19 to produce plasmids pUC-C225V H and pUC-C225V ⁇ , respectively, and transformed into XL 1 Blue competent cells. Putative positive colonies containing the appropriate plasmid are then identified by PCR- screening, using oligonucleotide primers RSP and UP (Table 6) and finally ds-DNA sequenced both to confirm the introduction of the sequence modifications and also to prove that no unwanted changes to the DNA sequence have occured as a consequence of the PCR-reactions.
  • PCR-mutagenesis is used, according to the protocol described by Kettleborough et al (11).
  • PCR-primers C225V ⁇ 5'sp and C225V ⁇ 3'sp (Table 7) are used on pUC-C225V ⁇ template DNA to create the modified gene (C225V ⁇ sp) using the modified two step PCR amplification protocol.
  • the PCR-product is then column purified before digesting both the purified PCR-product and pUC-C225V ⁇ with Hind ⁇ ll and Pstl.
  • PCR-fragment and the plasmid DNA are then agarose gel- purified, ligated together and cloned to create plasmid pUC-C225V ⁇ sp.
  • putative positive transformants are identified via a PCR-screen (using the RSP and UP primers) and then ds-DNA sequenced to confirm both the presence of the modified signal peptide and the absence of PCR-errors.
  • the adapted mouse kappa light and heavy chain leader- variable region genes are then directly inserted, as a H dIII-5 ⁇ wHI fragment in the case of the mouse V H and as a Hin ⁇ lll-Xbal fragment in the case of the mouse V ⁇ , into vectors designed to express chimeric light and heavy chains in mammalian cells.
  • These vectors contain the ⁇ CMV enhancer and promoter to drive the transcription of the immunoglobulin chain, a MCS for the insertion of the immunoglobulin variable region gene, a cDNA clone of the appropriate human kappa light or heavy chain constant region, a synthetic poly(A) + sequence to polyadenylate the immunoglobulin chain mRNA, an artificial sequence designed to terminate the transcription of the immunoglobulin chain, a gene such as dhfr or neo for selection of transformed stable cell lines, and an SV40 origin of replication for transient DNA replication in COS cells.
  • the human kappa light chain mammalian expression vector is called pKN 100 ( Figure 11) and the human ⁇ 1 heavy chain mammalian expression vector is called pGlD105 ( Figure 12).
  • Putative positive colonies are both PCR-screened, using primers ⁇ CMVi and New. ⁇ u ⁇ for the chimeric kappa light chain vector and primers ⁇ CMVi and ⁇ uC ⁇ l for the chimeric heavy chain vector (Table 6), and undergo restriction analysis to confirm the presence of the correct insert in the expression vector constructs.
  • the new constructs containing the mouse variable region genes of the M225 antibody are called pK 100-C225V ⁇ (or pKN100-C225V ⁇ sp ) and pGlD105-C225V ⁇ , respectively.
  • Example IV-5 Molecular modelling of mouse M225 antibody variable regions
  • a molecular model of the variable regions of the mouse M225 antibody is built. Modelling the structures of well-characterized protein families like immunoglobulins is achieved using the established method of modelling by homology. This is done using an IRIS 4D workstation running under the UNIX operating system, the molecular modelling package QUANTA and the Brookhaven crystallographic database of solved protein structures (12). The FRs of the M225 variable regions are modelled on FRs from similar, structurally- solved immunoglobulin variable regions. While identical amino acid side chains are kept in their original orientation, mismatched side chains are substituted using the maximum overlap procedure to maintain chi angles as in the original mouse M225 antibody.
  • CDRs of the M225 variable regions are modelled based on the canonical structures for hypervariable loops which correspond to CDRs at the structural level (13-16).
  • the CDR loop is modelled based on a similar loop structure present in any structurally-solved protein.
  • the model is subjected to energy minimization using the CHARMm potential (17) as implemented in QUANTA.
  • the FRs from the light chain variable region of M225 antibody are modelled on the FRs from the Fab fragment of mouse monoclonal antibody HyHel-10 (18).
  • the FRs from the heavy chain variable region are modelled on the FRs from the Fab fragment of mouse monoclonal antibody D 1.3 (19).
  • Those amino acid side chains which differ between the mouse M225 antibody and the variable regions upon which the model is based are first substituted.
  • the light chain of Fab HyHel-10 antibody is then superimposed onto the light chain of D1.3 by matching residues 35-39, 43-47, 84-88 and 98-102, as defined by Kabat et al, (20).
  • the purpose of this is to place the two heterologous variable regions, i.e. the HyHel-10-based kappa light chain variable region and the D1.3-based heavy variable region, in the correct orientation with respect to each other.
  • CDR1 (LI) of the light chain variable region of mAb M225 fits into the LI canonical group 2, as proposed by Chothia et al. (14), except for the presence of an isoleucine, instead of the more usual leucine, at canonical residue position 33.
  • this substitution is considered too conservative to merit significant concern in assigning a canonical loop structure to this hypervariable loop.
  • the LI loop of mouse Fab HyHel-10 is identical in amino acid length and matches the same canonical group - with a leucine at position 33 - as the LI loop of M225 mAb. Consequently this hypervariable loop is used to model the LI loop of M225 kappa light chain variable region.
  • CDR2 (L2) and CDR3 (L3) of the M225 mAb both match their respective canonical group 1 loop structures.
  • the corresponding hypervariable loop structures of the HyHel-10 Fab fragment are also both group 1. Accordingly, the L2 and L3 loops of the M225 kappa light chain variable region are modelled on L2 and L3 of Fab HyHel-10.
  • CDRl (HI) and CDR 2 (H2) hypervariable loops of the heavy chain variable region of mAb M225 both fit their respective canonical group 1 loop structures as defined by Chothia et al. (14).
  • the corresponding HI and H2 hypervariable loops of mouse D1.3 Fab fragment also match their respective canonical group 1 loop structures. Consequently, as with the light chain, these hypervariable loops are modelled on the HI and H2 loops of the heavy variable region upon which the model is based.
  • the Brookhaven database is searched for a loop of identical length and similar amino acid sequence.
  • Example IV-6 Design of the reshaped human H225 antibody variants.
  • the first step in designing the CDR-grafted variable regions of the H225 antibody is the selection of the human light and heavy chain variable regions that will serve as the basis of the humanized variable regions.
  • the M225 antibody light and heavy chain variable regions are initially compared to the consensus sequences of the four subgroups of human kappa light chain variable regions and the three subgroups of human heavy chain variable regions as defined by Kabat et al. (20).
  • the mouse M225 light chain variable region is most similar to the consensus sequences of both human kappa light chain subgroup I, with a 61.68% identity overall and a 65.00% identity with the FRs only, and subgroup III, with a 61.68%) identity overall and a 68.75% identity with the FRs only.
  • the mouse M225 heavy chain variable region is most similar to the consensus sequence for human heavy chain subgroup II with a 52.10% identity overall and a 57.47% identity between the FRs alone. This analysis is used to indicate which subgroups of human variable regions are likely to serve as good sources for human variable regions to serve as templates for CDR-grafting, however, this is not always the case due to the diversity of individual sequences seen within some of these artificially constructed subgroups.
  • mouse M225 variable regions are also compared to all the recorded examples of individual sequences of human variable regions publically available.
  • the mouse M225 light chain variable region is most similar to the sequence for the human kappa light chain variable region from human antibody LS7'CL (22) - which is not related to the mouse L7'CL sequence.
  • the kappa light chain variable region of human LS7'CL is a member of subgroup III of human kappa light chain variable regions.
  • the overall sequence identity between the mouse M225 and human LS7'CL light chain variable regions is calculated to be 64.42% overall and 71.25% with respect to the FRs alone.
  • the mouse M225 heavy chain variable region is most similar to the sequence for the human heavy chain variable region from human antibody 38P1'CL (23). Surprisingly, the heavy chain variable region of human 38P1'CL is a member of subgroup III and not subgroup II of the human heavy chain variable regions. The overall sequence identity between the mouse M225 and human 38P1'CL heavy chain variable regions is calculated to be 48.74% while the identity between the FRs alone is 58.62%. Based on these comparisons, human LS7'CL light chain variable region is selected as the human FR donor template for the design of reshaped human M225 light chain variable region and human 38P1'CL heavy chain variable region is selected as the human FR donor template for the design of reshaped human M225 heavy chain variable region.
  • the human light and heavy chain variable regions that are selected for the humanization of the M225 antibody are derived from two different human antibodies. Such a selection process allows the use of human variable regions which display the highest possible degree of similarity to the M225 variable regions.
  • CDR-grafted antibodies based on variable regions derived from two different human antibodies.
  • One of the best studied examples is reshaped human CAMPATH-1 antibody (24). Nevertheless, such a strategy also requires a careful analysis of the interdomain packing residues between the kapp light chain and heavy chain variable regions. Any mis-packing in this region can have a dramatic affect upon antigen binding, irrespective of the conformation of the CDR loop structures of the reshaped human antibody.
  • the second step in the design process is to insert the M225 CDRs, as defined by Kabat et al. (20), into the selected human light and heavy chain variable region FRs to create a simple CDR-graft. It is usual that a mouse antibody that is humanized by a simple CDR-graft in this way, will show little or no binding to antigen. Consequently, it is important to study the amino acid sequences of the human FRs to determine if any of these amino acid residues are likely to adversely influence binding to antigen, either directly through interactions with antigen, or indirectly by altering the positioning of the CDR loops.
  • Table 8 describes how the first version (225RK A ) of the reshaped human H225 kappa light chain variable regions is designed.
  • the tyrosine found in human LS7'CL kappa light chain variable region is changed to a lysine, as found in mouse M225 kappa light chain variable region. From the model it appears that the lysine in M225 is located close to CDR3 (H3) of the heavy chain variable region and may be interacting with it.
  • the residue is also positioned adjacent to CDR2 (L2) of the kappa light chain variable region and is rarely seen at this location amonst the members of mouse kappa light chain subgroup V, as defined by Kabat et al. (20), to which the M225 kappa light chain variable region belongs. For these reasons it is felt prudent to conserve the mouse lysine residue in 225RK A .
  • a second version is also made of the reshaped human kappa light chain (225RK B ) which reverses the FR2 modification made in 225RK A , by replacing the lysine at position
  • Table 9 shows the first version (225RH A ).
  • the amino acid present in the human 38P1'CL FRs to the amino acids present in the original mouse M225 FRs (i.e. A24V, T28S, F29L, S30T, V48L, S49G, F67L and R71K).
  • the amino acid residues as present in the mouse sequence are retained in the reshaped human H225 heavy chain variable region because they represent some of the canonical residues important for the HI hypervariable loop structure (14).
  • residue positions 24-30 are considered part of the HI hypervariable loop itself and so are even more critical to the correct conformation and orientation of this loop and justifying their conservation even more strongly.
  • residue position 71 in FR3 is another position in the heavy chain variable region which has been identified by Chothia et al. (14) as one of the locations important for the correct orientation and structure of the H2 hypervariable loop and, as such, is one of the canonical amino acids of CDR2. Consequently, the lysine in the mouse will replace the arginine in the human at this residue position.
  • Version B of the reshaped human H225 heavy chain variable region incorporates all the substitutions made in 225RH A and, in addition, contains a further mouse residue.
  • the human threonine residue is replaced by proline which is invariably seen at this position in the mouse subgroup IB and is also very commonly seen in human subgroup III.
  • threonine is not usually seen at this location in the human subgroup III (only ' V 87 times) and from the model it is appears that the residue is located on a turn located on the surface of the M225 V H region. What effect this may have on hypervariable loop structures is unclear, however, this version of the reshaped human H225 heavy chain variable region should clarify this.
  • Version C of the reshaped human H225 heavy chain variable region incorporates all the substitutions made in 225RH A and, in addition, contains a further two mouse residues located at position 68 and 70 in FR3. From the model of the mouse M225 variable region, both the serine at position 68 and the asparagine at position 70 appear to be on the surface and at the edge of the antigen binding site. Since there is a possibility that either or both amnio acids could directly interact with EGFR, both the threonine at position 68 and the seine at position 70 in the human FRs are replaced with the corresponding mouse residues in 225RH C .
  • Version D of the reshaped human H225 heavy chain variable region simply incorporates all the mouse FR substitutions made in 225RH A , 225RH B and 225RH C to determine the combined effect of these changes.
  • Version E of the reshaped human H225 heavy chain variable region (225RH E ) incoiporates all the substitutions made in 225RH A and, in addition, incorporates another residue change at position 78 in FR3. From the model there is some evidence to suggest that the mouse amino acid (valine) at position 78 could influence the conformation of the HI hypervariable loops from its location buried underneath CDR1. Consequently, the human residue (leucine) is replaced by the mouse amino acid in 225RH E .
  • the construction of the first version of the reshaped human H225 V ⁇ region (225RK A ) is carried out essentially as described by Sato et al. (26). In essence, this involves annealing PCR-primers encoding FR modifcations (Table TO) onto a DNA template of the chimeric C225V K gene using the two step PCR-amplification protocol to synthesize the reshaped human variable region gene. As a consequence, the FR DNA sequence of the chimeric C225V K is modified by the primers to that of the reshaped human kappa light chain variable region gene 225RK A .
  • the newly synthesized reshaped variable region gene following column purification, is digested with Hr ⁇ dlll and Xbal, agarose gel-purified and subcloned into pUC19 (digested and agarose gel-purified in an identical manner).
  • the new plasmid construct, pUC-225RK A is then transformed into XL 1 Blue competent cells. Putative positive clones are identified by PCR-screening (using primers RSP and UP) and then finally ds-DNA sequenced, both to confirm their integrity and discount the presence of PCR-errors.
  • Version B of the reshaped human ⁇ 225 V ⁇ (225RK B ) is constructed using oligonucleotide primers 225RK B .K49Y and APCR40 (Table 11).
  • a 100 ⁇ l PCR-reaction mix comprising 65.5 ⁇ l of sterile distilled/deionized water, 5 ⁇ l of 2 ng/ ⁇ l plasmid pUC-
  • 225RK A template DNA, 10 ⁇ l of 10 X PCR buffer II, 6 ⁇ l of 25 mM MgCl 2 , 2 ⁇ l each of the 10 mM stock solutions of dNTPs, 2.5 ⁇ l aliquots (each of 10 ⁇ M) of primers 225RK B .K49Y and APCR40 and 0.5 ⁇ l of AmpliTaq®DNA polymerase is overlayed with 50 ⁇ l of mineral oil and loaded into a DNA thermal cycler. The PCR-reaction is PCR-amplified, using the two step protocol over 25 cycles, and the PCR-product column purified before it is cut with Msc ⁇ .
  • Plasmid pUC-225RK A is also cut with Mscl and both the digested PCR product and the plasmid fragment are agarose gel-purified. The PCR- product is then cloned into pUC-225RK A , to create pUC-225RK B , before being transformed into XL 1 Blue competent cells. Putative positive transformant are first identified, using primers 225RK B .K49Y and UP in a PCR-screening assay, and then confirmed via ds-DNA sequencing.
  • a selected individual clone is finally sublconed into pKNIOO to produce the plasmid pKN100-225RK B , whose correct construction is confirmed both by using primers HCMVi and New.Hu ⁇ (Table 6) in a PCR-screening assay and restriction analysis.
  • the construction of the first version of the reshaped human H225 V H region (225RH A ) is also carried out essentially as described by Sato et al. (26).
  • the two step PCR-amplification protocol is used and the reshaped variable region gene created is cloned into pUC19 vector, as an agarose gel-purified Hindlll-Bam ⁇ l fragment, to create plasmid pUC-225RH A .
  • Putative positive clones identified by PCR- screening (using primers RSP and UP) are finally ds-DNA sequenced both to confirm the DNA sequence and prove the absence of PCR-errors. From the confirmed positive clones an individual clone is selected and directly inserted, as a Hindffl-BamHI fragment, into the human ⁇ l heavy chain mammalian expression vector pGlD105 to create plasmid pGlD105-225RH A .
  • This plasmid is then confirmed both by using primers HCMVi and ⁇ AS (Table 6) in a PCR-screening assay and restriction analysis. Versions B of the reshaped human H225 V H (225RH B ) is synthesized in a two step PCR-mutagenesis procedure in the following manner.
  • Two separate 100 ⁇ l PCR- reaction mixes are first prepared by combining 65.5 ⁇ l of sterile distilled/deionized water, 5 ⁇ l of 2 ng/ ⁇ l plasmid pUC-225RH A template DNA, 10 ⁇ l of 10 X PCR buffer II, 6 ⁇ l of 25 mM MgCl 2 , 2 ⁇ l each of the 10 mM stock solutions of dNTPs, 2.5 ⁇ l aliquots (each of 10 ⁇ M) of primers APCR10 and 225RH B .T41P-AS in the first PCR- reaction, and primers APCR40 and 225RH B .T41P-S in the second PCR-reaction (Table 13), and finally 0.5 ⁇ l of AmpliTaq®DNA polymerase.
  • Each of the two PCR- reaction mixes are overlayed with 50 ⁇ l of mineral oil, loaded into a DNA thermal cycler and PCR-amplified using the two step protocol over 25 cycles.
  • the two PCR- products are then agarose gel-purified, to separate them from any template DNA remaining in the PCR-reaction, before being resuspended in 50 ⁇ l of distilled/deionized water and their concentration determined.
  • This PCR-reaction is overlayed with mineral oil and PCR-amplified using the two step protocol over 7 cycles only.
  • a third PCR-reaction is then prepared comprising 1 ⁇ l of the product of the second PCR-reaction 69.5 ⁇ l of sterile distilled/deionized water, 10 ⁇ l of 10 X PCR buffer II, 6 ⁇ l of 25 mM MgCl 2 , 2 ⁇ l each of the 10 mM stock solutions of dNTPs, 2.5 ⁇ l aliquots (each of 10 ⁇ M) of the nested primers RSP and UP and 0.5 ⁇ l of AmpliTaq®DNA polymerase.
  • PCR-reaction is overlayed with mineral oil and amplified using the two step protocol for a final 25 cycles.
  • This PCR-product is then column purified, isolated as an agarose gel purified HmdIII-itomHI fragment, subcloned into HindlU-BamHl digested and agarose gel -purified plasmid pUC19, and finally transformed into XL 1 Blue competent cells. Putative positive transformants are first identified and then confirmed as described previously.
  • a selected individual clone is then sublconed into pGlD105 to produce the plasmid pGlD105-225RH B - which is confirmed using primers HCMVi and ⁇ AS (Table 6) in a PCR-screening assay and by restriction analysis.
  • Version C of the reshaped human H225 V H (225RH B ) is synthesized in a similar manner to 225RKc.
  • a 100 ⁇ l PCR-reaction mix containing 65.5 ⁇ l of sterile distilled/deionized water, 5 ⁇ l of 2 ng/ ⁇ l plasmid pUC-225RH A template DNA, 10 ⁇ l of 10 X PCR buffer II, 6 ⁇ l of 25 mM MgCl 2 , 2 ⁇ l each of the 10 mM stock solutions of dNTPs, 2.5 ⁇ l aliquots (each of 10 M) of primers APCR40 and 225RH C .T68S/S70N (Table 13) and 0.5 ⁇ l of AmpliTaq®DNA polymerase.
  • the PCR-reaction is overlayed with mineral oil PCR-amplified, using the two step protocol over 25 cycles, and column purified prior to digestion with Sail and BamHl. Plasmid pUC-225RH A is also cut with with Sail and BamHl and both the digested PCR product and the plasmid are agarose gel-purified. The PCR-product is then cloned into pUC-225RH A , to create pUC-225RH c , before being transformed into XL 1 Blue competent cells. Putative positive transformant are first identified, using primers RSP and UP in a PCR-screennig assay, and later confirmed via ds-DNA sequencing.
  • a selected individual clone is then sublconed into pGlD105 to produce the plasmid pGlD105-225RH c .
  • the correct construction of this vector finally proven both by using primers HCMVi and ⁇ AS (Table 6) in a PCR-screening assay and restriction analysis.
  • Version D of the reshaped human H225 V H is a product of the changes incorporated into versions B and C of the reshaped human heavy chain of H225 antibody. Fortuitously, it is possible to amalgamate the changes made to these heavy chain variable region genes by digesting both pUC-225RH B and pUC-225RH c with Sa l and BamHl. The 2.95 kb vector fragment from pUC-225RH B and the approximately 180 bp insert fragment from pUC-225RH c are then agarose gel- purified before being ligated together and transformed into XL 1 Blue competent cells.
  • Version E of the reshaped human H225 V H (225RH E ) is a derivative of 225RH A and is synthesized in an identical manner to 225RH C using primers APCR40 and 225RH E .L78V (Table 13).
  • a selected 225RH E clone from plasmid pUC-225RH E is then sublconed into pGlD105 to produce the vector pGlD105-225RH E - the correct construction of which is proven in the usual manner.
  • Kettleborough et al. (11) The method of Kettleborough et al. (11) is followed to transfect the mammalian expression vectors into COS cells.
  • Example IV-9 Protein A purification of recombinant 225 antibodies
  • Both the chimeric C225 antibody and the various reshaped human H225 antibody constructs are protein A purified according to the protocol described in Kolbinger et al. (27).
  • Each well of a 96-well Nunc-Immuno Plate MaxiSorpTM immunoplate is first coated with 100 ⁇ l aliquots of 0.5 ng/ ⁇ l goat anti-mouse IgG ( ⁇ -chain specific) antibody, diluted in coating buffer (0.05 M Carbonate-bicarbonate buffer, pH 9.6), and incubated overnight at 4 °C.
  • the wells are blocked with 200 ⁇ l/well of mouse blocking buffer (2.5% (w/v) I _ in PBS) for 1 hr at 37 °C before being washed with 200 ⁇ l/well aliquots of wash buffer (PBS / 0.05% (v/v) tween-20) three times.
  • sample-enzyme conjugate buffer 0.1 M Tris-HCl (pH 7.0), 0.1 M NaCl, 0.02% (v/v) tween-20 and 0.2% (w/v) BSA
  • sample-enzyme conjugate buffer 0.1 M Tris-HCl (pH 7.0), 0.1 M NaCl, 0.02% (v/v) tween-20 and 0.2% (w/v) BSA
  • a purified mouse IgG standard serially diluted 1 :2 from a starting concentration of 1000 ng/ml, is also loaded onto the immunoplate.
  • the immunoplate is incubated at 37 °C for 1 hr and washed three times with 200 ⁇ l/well of wash buffer.
  • 100 ⁇ l aliquots of TMB peroxiodase substrate A:peroxidase substrate B ( 1 : 1 ) are now added to each well and incubated for 10 min at RT in the dark.
  • the reaction is halted by dispensing 50 ⁇ l of 1 N H 2 SO 4 into each well.
  • the optical density at 450 nm is finally determined using a Bio-Rad 3550 microplate reader in conjunction with Microplate ManagerTM.
  • Example IV- 11 Quantification of whole human ⁇ l/ ⁇ antibody via ELISA
  • Each well of a 96-well Nunc-Immuno Plate MaxiSorpTM immunoplate is first coated with 100 ⁇ l aliquots of 0.4 ng/ ⁇ l goat anti-human IgG (Fc ⁇ fragment specific) antibody, diluted in coating buffer (0.05 M Carbonate-bicarbonate buffer, pH 9.6), and incubated overnight at 4 °C.
  • the wells are then each blocked with 200 ⁇ l of human blocking buffer (2% (w/v) BSA in PBS) for 2 hr at RT before being washed with 200 ⁇ l/well aliquots of wash buffer (PBS / 0.05% (v/v) tween-20) three times. 100 ⁇ l/well aliquots of the experimental samples (i.e.
  • sample-enzyme conjugate buffer 0.1 M Tris-HCl (pH 7.0), 0.1 M NaCl, 0.02% (v/v) tween-20 and 0.2% (w/v) BSA
  • sample-enzyme conjugate buffer 0.1 M Tris-HCl (pH 7.0), 0.1 M NaCl, 0.02% (v/v) tween-20 and 0.2% (w/v) BSA
  • a purified human ⁇ l/ ⁇ antibody which is used as a standard and serially diluted 1 :2, is also loaded onto the immunoplate.
  • the immunoplate is incubated at 37 °C for 1 hr before being washed with 200 ⁇ l well of wash buffer three times.
  • Example IV- 12. A4 1 Cell ELISA for the detection of EGFR antigen binding
  • the procedure is based upon the one provided by ImClone Systems Inc. to determine the relative binding affinity of the recombinant 225 antibody constructs, to EGFR expressed on the surface of A431 cells.
  • the A431 cells are plated onto a 96- well flat bottomed tissue culture plate and incubated overnight in DMEM media with 10% (v/v) FBS at 37 °C and 5% CO 2 . The following day the media is removed, the cells are washed once in PBS and then fixed with 100 ⁇ l/well of 0.25% (v/v) gluteraldehyde in PBS.
  • mice antibody in the media from the M225 hybridoma cells at the point of harvesting the cells for RNA purification was proven using the mouse antibody ELISA.
  • the single stranded cDNA template was PCR-amplified with two series of degenerate primers, one series specific for the kappa light chain signal peptide/variable region genes (Table 4) and the second series specific for the heavy chain signal peptide/variable region genes (Table 5).
  • Table 4 the primerst strand synthesis the single stranded cDNA template was PCR-amplified with two series of degenerate primers, one series specific for the kappa light chain signal peptide/variable region genes (Table 4) and the second series specific for the heavy chain signal peptide/variable region genes (Table 5).
  • Table 4 the primers both the V ⁇ gene and the V H gene of the M225 antibody were successfully PCR-cloned from the M225 hybridoma cell line.
  • the M225 kappa light chain variable region gene was PCR-cloned, as an approximately 416bp fragment, using primers MKV4 (which annealed to the 5' end of the DNA sequence of the kappa light chain signal peptide) and MKC (designed to anneal to the 5 'end of the mouse kappa constant region gene).
  • MKV4 which annealed to the 5' end of the DNA sequence of the kappa light chain signal peptide
  • MKC designed to anneal to the 5 'end of the mouse kappa constant region gene.
  • the M225 heavy chain variable region gene was PCR-cloned, as an approximately 446bp fragment, using the MHV6 (which annealed to the 5' end of the DNA sequence of the heavy chain signal peptide) and MHCG1 (designed to anneal to the 5' end of the CH] domain of the mouse ⁇ l heavy chain gene) primers.
  • the mouse M225 antibody V ⁇ and V H genes were determined as shown in Figures 13 and 14, respectively.
  • the amino acid sequences of the M225 V ⁇ and V H regions were compared with other mouse variable regions and also the consensus sequences of the subgroups that the variable regions were subdivided into in the Kabat database (20).
  • the M225 V ⁇ region was found to most closely match the consensus sequence of mouse kappa subgroup V, with an identity of 62.62% and a similarity of 76.64% to the subgroup.
  • the kappa light chain variable region also displayed a close match to mouse kappa subgroup III with a 61.68% identity and a 76.64% similarity to its consensus sequence.
  • Example IV- 14 Construction and expression of chimeric C225 antibody
  • the PCR-products from the two PCR-reactions prepared to construct the C225 V ⁇ and V H genes were separately subcloned into pUC19 as Hind ⁇ ll-BamHl fragments and then PCR-screened to identify putative positive transformants. Those transformants so identified were then ds-DNA sequenced, to confirm their synthesis, and then subcloned into their respective mammalian expression vectors.
  • the DNA and amino acid sequences of the chimeric C225 kappa light chain and heavy chain variable regions are shown in Figures 15 and 16, respectively.
  • the amino acid sequence of the L7'CL kappa light chain signal peptide (i.e. MVSTPQFLVFLLFWIPASRG (SEQ ID NO: 36)) displays all the characteristics thought important in a such a signal sequence - such as a hydrophobic core - and so it was decided to replace the signal peptide of the PCR- cloned 225V K with this new sequence.
  • Another point of interest was that the differences between the M225V K and the L7'CL signal peptides nearly all occured at its 5 '-end where the MKV4 primer annealed (i.e.
  • PCR-mtuagenesis of the C225V K template produced an approximately 390bp product.
  • the HmdIII-.P-.tI digested and purified fragment was then subcloned into identically digested and agarose gel-purified plasmid pUC-C225V ⁇ and transformed into XLlBlue competent cells. Putative positive transformants were identified and then ds-DNA sequenced.
  • the C225V ⁇ sp gene ( Figure 17) was subcloned into pKNIOO and the resulting expression vector (pKN100-C225V ⁇ sp) PCR-screened and restriction digested to confirm the presence of the correct insert.
  • This vector was finally co-transfected into COS cells with pGlD105-C225V H and after 72 hr incubation, the medium was collected, spun to remove cell debri and analysed by ELISA for antibody production and binding to EGFR. This time chimeric C225 antibody was detected in the supernatent of the COS cell co-transfections at an approximate concentration of 150 ng/ml and this antibody bound to EGFR in the cell ELISA.
  • Figure 18 shows a typical example of one such experiment.
  • Example IV-15 Construction and expression of the reshaped H225 antibody (225RK ⁇ /225RH ⁇ )
  • the construction of the first version of the reshaped human H225 kappa light chain variable region produced an approximately 416bp product that was then subcloned into pUC19 as a Hindll-BamHl fragment. Putative positive transformants were identified using the PCR-screening assay and then ds-DNA sequenced.
  • the 225RK A gene ( Figure 19) was subcloned into pKNIOO and the resulting expression vector (pKN100-225RK A ) PCR-screened and restriction digested to confirm the presence of the correct insert.
  • the construction of the first version of the reshaped human H225 heavy chain variable region produced an approximately 446bp product which was then subcloned into pUC 19 as a Hindll- BamHl fragment. Putative positive transformants were again identified in the PCR- screen and then ds-DNA sequenced.
  • the 225RH A gene ( Figure 20) was subcloned into pGlD105 and the resulting expression vector (pGlD105-225RH A ) PCR-screened and restriction digested to confirm the presence of the correct insert.
  • MKV1 (30mer) ATGAAGTTGCCTGTTAGGCTGTTGGTGCTG MKV2 (30mer) ATGGAGACAGACACACTCCTGCTATGGGTG T T
  • MKV8 (31mer) ATGTGGGGACCTTTTTTCCCTTTTTCAATTG T G C AA
  • a MKV indicates primers that hybridize to leader sequences of mouse kappa light chain variable region genes.
  • b MKC indicates the primer that hybridizes to the mouse kappa constant region gene. Table 5. Degenerate and specific PCR-primers used in the cloning of the M225 heavy chain variable region genes.
  • MHV5 (30mer) ATGGACTCCAGGCTCAATTTAGTTTTCCTT MHV6 (27mer) ATGGCTGTCCTAGGGCTACTCTTCTGC T G C G MHV7 (26mer) ATGGGATGGAGCGGGATCTTTCTCTT A T G A
  • a MHV indicates primers that hybridize to leader sequences of mouse heavy chain variable region genes.
  • b MHCG indicates primers that hybridize to mouse constant region genes. Table 6. Primers for PCR screening transformed colonies
  • invariant residues as defined either by the Kabat consensus sequences i.e. 95% or greater occurrence within Kabat subgroup (Kabat et al, 19 1) (in the case of columns 5 and 6) or as part of the canonical structure for the CDR loops (in the case of column 8) as defined by Chothia et al, (1989); (BOLD) positions in Frs and CDRs where the human amino acid residue was replaced by the corresponding mouse residue (UNDERLINE) positions in Frs where the human residue differs from the analogous mouse residue number; ( ⁇ ) numbering of changes in the human Frs; (mouse C225) amino acid sequence of the V H region from chimeric C225 antibody; (mouse IB) consensus sequence of mouse V H regions from subgroup IB (Kabat et al, 1991); (human III) consensus sequence of human V H regions from subgroup III (Kabat et al, 1991); (Human Donors: 38P1) amino acid sequence from human antibody 38P1'

Abstract

Chimerized and humanized versions of anti EGF receptor antibody 225 and fragments thereof for treatment of tumors.

Description

ANTIBODY AND ANTIBODY FRAGMENTS FOR INHIBITING THE GROWTH OF TUMORS
This application is a continuation-in-part of Serial No. 08/573,289 filed December 15, 1995, which was a continuation-in-part of Serial No. 08/482,982 filed June 7, 1995, the disclosures of both of which are incorporated herein by reference.
FIELD OF THE INVENTION
The present invention is directed to antibodies and antibody fragments useful in inhibiting the growth of certain tumor cells.
BACKGROUND OF THE INVENTION
Recent research has uncovered the important role of growth factor receptor tyrosine kinases in the etiology and progression of human malignancies. These biological receptors are anchored by means of a transmembrane domain in the membranes of cells that express them. An extracellular domain binds to a growth factor. The binding of the growth factor to the extracellular domain results in a signal being transmitted to the intracellular kinase domain. The transduction of this signal contributes to the events that are responsible for the proliferation and differentiation of the cells.
Members of the epidermal growth factor (EGF) receptor family are important growth factor receptor tyrosine kinases. The first member of the EGF receptor family to be discovered was the glycoprotein having an apparent molecular weight of approximately 165 kD. This glycoprotein, which was described by Mendelsohn et al. in U.S. Patent No. 4,943,533, is known as the EGF receptor (EGFR). The binding of an EGFR ligand to the EGF receptor leads to cell growth. EGF and transforming growth factor alpha (TGF-alpha) are two known ligands of EGFR.
Many receptor tyrosine kinases are found in unusually high numbers on human tumors. For example, many tumors of epithelial origin express increased levels of EGF receptor on their cell membranes. Examples of tumors that express EGF receptors include glioblastomas, as well as cancers of the lung, breast, head and neck, and bladder. The amplification and or overexpression of the EGF receptors on the membranes of tumor cells is associated with a poor prognosis.
Antibodies, especially monoclonal antibodies, raised against tumor antigens have been investigated as potential anti-tumor agents. Such antibodies may inhibit the growth of tumors through a number of mechanisms. For example, antibodies may inhibit the growth of tumors immunologically through antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
Alternatively, antibodies may compete with growth factors in binding to their receptors. Such competition inhibits the growth of tumors that express the receptor.
In another approach, toxins are conjugated to antibodies raised against tumor antigens. The antibody portion directs the conjugate to the tumor, which is killed by the toxin portion.
For example, U.S. Patent No. 4,943,533 describes a murine monoclonal antibody called 225 that binds to the EGF receptor. The patent is assigned to the University of California and licensed exclusively to ImClone Systems Incorporated. The 225 antibody is able to inhibit the growth of cultured EGFR-expressing tumor lines as well as the growth of these tumors in vivo when grown as xenografts in nude mice. In a phase I clinical trial, however, no clinical response was observed when up to 300 mg of murine 225 antibodies was administered to humans. See Divgi et al., J. Natl. Cancer Inst. £1, 97-104 (1991). See Masui et al, Cancer Res. 44, 5592-5598 (1986). More recently, a treatment regimen combining 225 plus doxorubicin or cis-platin exhibited therapeutic synergy against several well established human xenograft models in mice. Basalga et al, J. Natl. Cancer Inst. £5, 1327-1333 (1993).
A disadvantage of using murine monoclonal antibodies in human therapy is the possibility of a human anti-mouse antibody (HAMA) response due to the presence of mouse Ig sequences. This disadvantage can be minimized by replacing the entire constant region of a murine (or other non-human mammalian) antibody with that of a human constant region. Replacement of the constant regions of a murine antibody with human sequences is usually referred to as chimerization.
The chimerization process can be made more effective by also replacing the variable regions - other than the hypervariable regions or the complementarity- determining regions (CDRs), of a murine antibody with the corresponding human sequences. The variable regions other than the CDRs are also known as the variable framework regions (FRs).
The replacement of the constant regions and non-CDR variable regions with human sequences is usually referred to as humanization. The humanized antibody is less immunogenic (i.e. elicits less of a HAMA response) as more murine sequences are replaced by human sequences. Unfortunately, both the cost and effort increase as more regions of a murine antibodies are replaced by human sequences.
Another approach to reducing the immunogenicity of antibodies is the use of antibody fragments. For example, an article by Aboud-Pirak et al, Journal of the National Cancer Institute £0, 1605-1611 (1988), compares the anti-tumor effect of an anti-EGF receptor antibody called 108.4 with fragments of the antibody. The tumor model was based on KB cells as xenografts in nude mice. KB cells are derived from human oral epidermoid carcinomas, and express elevated levels of EGF receptors. Aboud-Pirak et al. found that both the antibody and the bivalent F(ab')2 fragment retarded tumor growth in vivo, although the F(ab')2 fragment was less efficient. The monovalent Fab fragment of the antibody, whose ability to bind the cell-associated receptor was conserved, did not, however, retard tumor growth.
There is, therefore, a continuing need for improved anti-tumor agents that can be efficiently and inexpensively produced, have little or no immunogenicity in humans, are capable of binding to receptors that are expressed in high numbers on tumor cells, and are capable of blocking the binding of such growth factors to such receptors. An object of the present invention is the discovery of such new anti-tumor agents that combine the advantageous features of monoclonal antibodies, antibody fragments and single chain antibodies.
SUMMARY OF THE INVENTION
These and other objects, as will be apparent to those having ordinary skill in the art, have been met by providing a polypeptide lacking the constant region and the variable light chain of an antibody, the polypeptide comprising the amino acid sequence N Y G V H (SEQ IDNO: 1),GVIWSGGNTDYNTPFTSR (SEQ ID NO: 2), or V I W S G G N T D Y N T P F T S (SEQ ID NO: 3). The polypeptide may be conjugated to an effector molecule, such as a molecule that inhibits tumor growth. The invention further is directed to DNA encoding such polypeptides.
The invention also includes polypeptides consisting of the amino acid sequence N
YGVH, GVIWSGGNTDYNTPFTSRorVIWSGGNTDYNTPF TS.
The invention also includes a molecule having the constant region of a human antibody and the variable region of monoclonal antibody 225 conjugated to a cytoxic agent such as doxorubicin, taxol, or cis-diamminedichloroplatinum (cisplatin). The invention further includes a method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of a polypeptide lacking the constant region of the variable light chain of an antibody, the polypeptide comprising the amino acid sequence NYGVH, GVIWSGGNTD YNTPFTSR,orVIWSGGNTDYNTPFTS. Another aspect of the invention is a method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of a polypeptide consisting of the amino acid sequence NYGVH, GVIWSGGNTDYNTPF TSR,orVIWSGGNTDYNTPFTS.
The invention further includes a method for significantly inhibiting the growth of tumor cells that express the EGF receptor in a human. The method comprises administering to the human an effective amount of a molecule having the constant region of a human antibody and the variable region of monoclonal antibody 225, both in the prescence of and, in particular, in the absence of, cytotoxic molecules, such as chemotherapeutic agents.
DESCRIPTION OF FIGURES
Figure 1. Effect of 225 on the growth of established A431 tumor xenografts in nude mice. Animals were injected with 107 cells in the flank. Treatments, consisting of PBS or 1 mg/animal of 225 twice weekly for 5 weeks, were begun when tumors reached an average volume of 2-300 mm3. Volumes and Remission Index (RI) were determined as described in the "Examples" section.
Figure 2. Effect of 225 and chimerized 225 (C225) on the growth of established A431 tumor xenografts in nude mice. Animals were treated with lmg/mouse of PBS twice weekly for 5 weeks. A: Average tumor volumes; B: Remission Index. The apparent tumor regression in the PBS control group at day 37 was due to the death of 3 out of the 10 animals within the group at this time and the concommitant decrease in overall tumor volume.
Figure 3. Effect of C225 on the growth of established A431 xenografts in nude mice. Animals were treated with lmg of C225 or PBS twice weekly for 5 weeks. The average tumor volume of the C225 group showed statistically significant biological effects compared to control (see text) A: Average tumor volumes (asterisks show statistical significance with respect to control); B: Remission Index.
Figure 4. Dose response of C225 on the growth of established A431 xenografts in nude mice. Animals were treated with PBS, 1, 0.5, or 0.25 mg/animal twice weekly for 5 weeks as described in Materials Methods. Animals treated with 1 mg/dose of C225 showed statistically significant biological effects compared to control (see text). A: Average tumor volumes (asterisks define statistical signifiance with respect to control); B: Remission Index. The drop in RI for the 250 ug dose group on day 47 resulted from the re-appearance of a tumor in an apparent tumor-free animal. (In this instance, the effect of C225 was transient.)
Figure 5. Inhibition of A431 cells by C225 and by heavy chain CDR-1 and heavy chain CDR-2 of monoclonal antibody 225
Figure 6. Inhibition by C225-Doxorubicin conjugate of A431 cells in vivo as a function of concentration.
Figure 7. FACS analysis of EGFR expression on human prostatic carcinoma cell lines. LNCaP (human prostatic carcinoma, androgen-dependent), DU 145 and PC-3 (human prostatic carcinoma, androgen-independent), and A431 (human epidermoid carcinoma) cells were removed with EDTA from the growth flasks and stained with C225. Data are presented as MFI (Mean Fluorescence Intensity), an indirect measure of antigen expression. The results shown in this figure are representative of at least 5 experiments.
Figure 8. Inhibition of EGF-induced phosphorylation of the EGFR by C225. LNCaP, DU 145, and PC-3 monolayers were stimulated with EGF in the presence or absence of C225. Cells were lysed, subjected to SDS PAGE, blotted, and screened with a mouse monoclonal antibody to PTyr (UBI, Lake Placid). Lane A: no additions (basal level of EGFR phosphorylation); Lane B: stimulation of EGFR with 10 ng/ml EGF for 15 minutes at room temperature in the absence of C225; Lane C: stimulation of EGFR with EGF in the presence of 10 ug/ml of C225.
Figure 9. Growth inhibition of established DU 145 xenografts by C225. One million DU 145 cells in matrigel were innoculated into nude mice (males, nu nu). After tumors reached an average volume of approximately 100 mm3 (day 20), animals were randomized (10 animals per group) and treated with either PBS (control) or C225 (0.5 mg/dose, lOx). Animal were treated for 35 days and followed for an additional 3 weeks. Mice that were tumor-free or carrying small tumors were maintained for an additional 3 months. Significance (shown by astericks in Figure 3A) was determined by a Student's T-test and a p value < 0.5 was considered significant. A: average tumor volume; B: growth characteristics for tumors in the PBS group; C: growth characteristics for tumors in the C225-treated groups.
Figure 10. Effects of C225 on tumor elimination and surviva . The complete elimination of tumors during the course of the study was defined by a Remission Index (RI). Animal mortality during the study was considered a treatment failure and included in the analysis. A: Remission Index; B: Survival curve. The empty and filled circles in Figure 10 have the same meanings as in Figure 9. Figure 11. Schematic representation of the pKNIOO mammalian expression vector used for the expression of the kapp light chains of the chimeric C225 and reshaped human H225 antibody.
Figure 12. Schematic representation of the pGlD105 mammalian expression vector used for the expression of the heavy chains of the chimeric C225 and reshaped human H225 antibody.
Figure 13. DNA (SEQ ID NO: 4) and peptide (SEQ ID NO: 5) sequences of the kappa light chain variable region of the M225 antibody. The PCR-clones from which this information was obtained were amplified using the degenerate primer MKV4 (SEQ ID NO: 6)(7).
Figure 14. DNA (SEQ ID NO: 7) and peptide (SEQ ID NO: 8) sequences of the heavy chain variable region of the M225 antibody. The PCR-clones from which this information was obtained were amplified using the degenerate primer MHV6 (SEQ ID NO: 9)(7).
Figure 15. DNA (SEQ ID NO: 10) and peptide (SEQ ID NO: 11) sequences of the kappa light chain variable region of theC225 antibody.
Figure 16. DNA (SEQ ID NO: 12) and peptide (SEQ ID NO: 13) sequences of the heavy chain variable region of the C225 antibody.
Figure 17. DNA (SEQ ID NO: 14) and peptide (SEQ ID NO: 15) sequences of the kappa light chain variable region of the C225 antibody with the modified leader sequence from the kappa light chain of L7'CL antibody (28). Figure 18. Typical example of the results of a cell ELISA to measure the binding affinty of chimeric C225 and reshaped human H225 (225RKA/225RHA) antibodies to epidermal growth factor receptor expressed on the surface of A431 cells.
Figure 19. DNA (SEQ ID NO: 16) and peptide (SEQ ID NO: 17) sequences of the first version (225RKA) of the kappa light chain variable region of the reshaped human H225 antibody.
Figure 20. DNA (SEQ ID NO: 18) and peptide (SEQ ID NO: 19) sequences of the first version (225RHA) of the heavy chain variable region of the reshaped human H225 antibody.
Figure 21. Amino acid sequences of the two versions (225RKA and 225RKB) of the kappa light chain variable region of the reshaped human H225 antibody (SEQ ID NO: 20), (SEQ ID NO: 21), (SEQ ID NO: 22), (SEQ ID NO: 23). Residues are numbered according to Kabat et al (20). Mouse framework residues conserved in the reshaped human frameworks are highlighted in bold.
Figure 22. Amino acid sequences of the five versions (225RHA, 225RHB,
225RHC, 225RHD, 225RHE) of the heavy chain variable region of the reshaped human H225 antibody (SEQ ID NO: 24), (SEQ ID NO: 25), (SEQ ID NO: 26), (SEQ ID NO: 27), (SEQ ID NO: 28), (SEQ ID NO: 29), (SEQ ID NO: 30). Residues are numbered according to Kabat et al. (20). Mouse framework residues conserved in the reshaped human frameworks are highlighted in bold.
DETAILED DESCRIPTION OF THE INVENTION
In one aspect of the invention, a polypeptide lacking the constant region and the variable light chain of an antibody comprises the first and second heavy chain complementarity determining regions of monoclonal antibody 225. These regions have the following amino acid sequences:
CDR-1 N Y G V H (SEQ ID NO: 1)
CDR-2 G V I W S G G N T D Y N T P F T S R (SEQ ID NO: 2)
The peptide comprising the first and second complementarity determining regions mentioned above may be obtained by methods well known in the art. For example, the polypeptides may be expressed in a suitable host by DNA that encodes the polypeptides and isolated. The DNA may be synthesized chemically from the four nucleotides in whole or in part by methods known in the art. Such methods include those described by Caruthers in Science 230, 281-285 (1985).
The DNA may also be obtained from murine monoclonal antibody 225, which was described by Mendelsohn, et al U.S. Patent No. 4,943,533. This antibody was deposited in the American Type Culture Collection, Bethesda, Maryland on June 7, 1995. (Accession number 11935). Methods for obtaining the variable heavy chain region of antibodies are known in the art. Such methods include, for example, those described in U.S. patents by Boss (Celltech) and by Cabilly (Genentech). See U.S. Patent Nos. 4,816,397 and 4,816,567, respectively.
The DNA encoding the protein of the invention may be replicated and used to express recombinant protein following insertion into a wide variety of host cells in a wide variety of cloning and expression vectors. The host may be prokaryotic or eukaryotic. The polypeptide may contain either NYGVH, GVIWSGGNTDYNTPF TSR, orVIWSGGNTDYNTPFTS. Alternatively, the polypeptide may contain the sequence N Y G V H, and either of the sequences GVIWSGGNTD YNTPFTSR, orVIWSGGNTDYNTPFTS.
The polypeptide may also be conjugated to an effector molecule. The effector molecule performs various useful functions such as, for example, inhibiting tumor growth, permitting the polypeptide to enter a cell such as a tumor cell, and directing the polypeptide to the appropriate location within a cell.
The effector molecule, for example, may be a cytotoxic molecule. The cytotoxic molecule may be a protein, or a non-protein organic chemotherapeutic agent. Some examples of suitable chemotherapeutic agents include, for example, doxorubicin, taxol, and cisplatin.
Some additional examples of effector molecules suitable for conjugation to the polypeptides of the invention include signal transduction inhibitors, ras inhibitors, and cell cycle inhibitors. Some examples of signal transduction inhibitors include protein tyrosine kinase inhibitors, such as quercetin (Grazieri et al , Biochim. Biophs. Acta 714.415 (1981)); lavendustin A (Onoda et al, J. Nat. Prod.52, 1252 (1989)); and herbimycin A (Ushara et al, Biochem. Int., 41, 831 (1988)). Ras inhibitors include inhibitors of ras farnesylation, such as the benzodiazepine peptidomimetics described by James et al in Science 2601937 (1993), which have the formula shown below:
in which R is H or CH3; and X is Methione, Serine, Leucine, or an ester or amide derivative thereof.
Proteins and non-protein chemotherapeutic agents may be conjugated to the polypeptides by methods that are known in the art. Such methods include, for example, that described by Greenfield et al , Cancer Research £0, 6600-6607 (1990) for the conjugation of doxorubicin and those described by Arnon et al, Adv. Exp. Med. Biol. 201 79-90 (1991) and by Kiseleva et al, Mol. Biol. (USSR) 25, 508-514 (1991) for the conjugation of platinum compounds.
The invention further includes a modified antibody having the constant region of a human antibody, and the hypervariable region of monoclonal antibody 225. These modified antibodies are optionally conjugated to an effector molecule, such as a cytotoxic agent. The variable region other than the hypervariable region may also be derived from the variable region of a human antibody. Such an antibody is said to be humanized. Methods for making humanized antibodies are known in the art. Methods are described, for example, in Winter, U.S. Patent No. 5,225,539.
The most thorough method for humanization of the 225 antibodies is CDR- grafting. As described in Example IV, the regions of the mouse antibody that are directly involved in binding to antigen, the complementarity determining region or CDRs, are grafted into human variable regions to create "reshaped human" variable regions. These fully humanized variable regions are then joined to human constant regions to create complete "fully humanized" antibodies. In order to create a fully humanized antibody that binds well to antigen, it is essential to carefully design the reshaped human variable regions. The human variable regions into which the 225 antibodies CDRs will be grafted must be carefully selected, and it is usually necessary to make a few amino acid changes at critical positions within the framework regions (FRs) of the human variable regions. The reshaped human H225 variable regions, as designed, include up to a single amino acid change in the FRs of the selected human kappa light chain variable region and as many as twelve amino acid changes in the FRs of the selected human heavy chain variable region. The DNA sequences coding for these reshaped human H225 heavy and kappa light chain variable region genes are joined to DNA sequences coding for the human γl and human K constant region genes, respectively. The reshaped human H225 antibody is then expressed in mammalian cells and tested, in comparison with mouse M225 antibody, and chimeric C225 antibody for binding to human EGF receptor expressed on the surface of A431 cells.
The variable region of the antibody outside of the hypervariable region may also be derived from monoclonal antibody 225. In such case, the entire variable region is derived from murine monoclonal antibody 225, and the antibody is said to be chimerized, i.e., C225. Methods for making chimerized antibodies are known in the art. Such methods include, for example, those described in U.S. patents by Boss (Celltech) and by Cabilly (Genentech). See U.S. Patent Nos. 4,816,397 and 4,816,567, respectively.
The constant region of the modified antibodies may be of any human class, i.e., IgG, IgA, IgM, IgD, and IgE. Any subclass of the above classes is also suitable, e.g., IgGl, IgG2, IgG3 and IgG4, in which IgGl is preferred.
Any of the effector molecules mentioned above in connection with conjugation to a polypeptide can also be conjugated to chimeric or humanized antibodies of the invention. Doxorubicin, taxol, and cisplatin are prefened.
The polypeptides and antibodies of the invention significantly inhibit the growth of tumor cells when administered to a human in an effective amount. The optimal dose can be determined by physicians based on a number of parameters including, for example, age, sex, weight, severity of the condition being treated, the active ingredient being administered, and the route of administration. In general, a serum concentration of polypeptides and antibodies that permits saturation of EGF receptors is desirable. A concentration in excess of approximately 0.1 nM is normally sufficient. For example, a dose of 100 mg/m2 of C225 provides a serum concentration of approximately 20 nM for approximately eight days.
As a rough guideline, doses of antibodies may be given weekly in amounts of 10- 300 mg/m2. Equivalent doses of antibody fragments should be used at more frequent intervals in order to maintain a serum level in excess of the concentration that permits saturation of EGF receptors.
Some suitable routes of administration include intravenous, subcutaneous, and intramuscle administration. Intravenous administration is preferred.
The peptides and antibodies of the invention may be administered along with additional pharmaceutically acceptable ingredients. Such ingredients include, for example, immune system stimulators and chemotherapeutic agents, such as those mentioned above.
It has now surprisingly been found that, unlike the murine 225 antibody, the chimeric and humanized antibodies significantly inhibit tumor growth in humans, even in the absence of other anti-tumor agents, including other chemotherapeutic agents, such as cisplatin, doxorubicin, taxol, and their derivatives. Significant inhibition may mean the shrinkage of tumors by at least 20%, preferably 30%, and more preferably 50%. In optimal cases, 90% and even 100% shrinkage of tumors is achieved. Alternatively, significant inhibition may mean an RI greater than 0.3, preferably greater than 0.4, and more preferably greater than 0.5. The significant inhibition of tumor growth and or increase in RI manifests itself in numerous ways. For example, there is an increase in life expectency and/or a stabilization of previously aggresive tumor growth.
In cases where the side effects of chemotherapeutic agents are too severe for a patient to continue such treatments, C225 may be substituted for the chemotherapeutic agents, and achieve comparable results.
For example, the results shown in Example III-l indicate that, while the in vitro inhibitory properties of 225 and C225 are comparable, the in vivo effects of the antibodies differ considerably. Antibody isotype does not play a significant role in the differences seen between 225 and C225 (e.g., mouse IgGl vs. human IgGl). A recent report indicates that neither 225 nor C225 induced complement mediated lysis to any degree and the ADCC reactivity of these antibodies appeared to be species specific. Naramura et al, Immunol. Immunother. 22, 343-349 (1993). Therefore, if inhibition of A431 xenografts was mediated through immune responses, 225 should be the more potent antibody because of its ability to activate the murine effector cells involved in ADCC. The opposite is, in fact, the case.
In addition, there were differences in the way individual animals within a group responded to treatment with either 225 or C225. It appeared that C225 alone was very effective in inducing complete tumor remission at the 1 mg dose whereas 225 at this dose level showed marginal effects. In Experiments 2 and 3 of Example III-l, about 40% of the animals were tumor free at the end of each study. The animals responding in those groups usually had smaller tumors at the beginning of the treatment protocols, once again indicating that initial tumor burden plays a role in the biological efficacy of C225. Significantly, animals treated with either 225 or C225 showed greater survival characteristics compared to the PBS control group in all studies. As demonstrated in Example III-2, prostatic carcinoma is also an appropriate target for anti-EGFR immunotherapeutic intervention with C225. Since the metastatic prostatic carcinoma cells coexpress TGF-α as well as the EGFR, late stage prostatic carcinoma is an especially appropriate target.
Example III-2 describes the biological effects of C225 on the activation of the
EGFR in cultured human prostatic carcinoma cells and the growth of prostate xenografts in nude mice. The in vitro experiments were designed to determine the expression levels of the EGFR on three human prostatic carcinoma cell lines and the ability of C225 to block the functional activation of the receptor. Figure 7 shows the results of a FACS analysis comparing EGFR expression on A431 cells to levels seen on LNCaP (androgen-dependent) and PC-3 and DU 145 (androgen-independent) cells. Both PC-3 (MFI = 135) and DU-145 (MFI = 124) cells expressed about 7 fold less receptor than A431 cells (MFI = 715). Since MFI is an indirect measure of antigen density, both PC-3 and DU 145 cells would appear to express about 105 receptors each. LNCaP cells, on the other hand, expressed very low levels of surface receptor (MFI = 12).
As shown above, the EGFR expressed by A431 cells can be stimulated by exogenously added ligand (EGF) and C225 can abrogate activation of the receptor. Figure 8 shows the results of similar studies with the prostatic lines. The addition of EGF to LNCaP, PC-3, and DU 145 induced phosphorylation of the EGFR that was blocked by C225 with high efficiency. These data indicate that C225 effectively inhibits ligand-activated EGFR signalling pathways, and has anti-tumor activity when EGFR activation is required for growth in vivo.
The ability of C225 to inhibit tumor growth in vivo was tested against established DU 145 xenografts in athymic nude mice. DU 145 cells were innoculated at 106 cells per animals in combination with matrigel. Tumors developed in 100% of the animals within 20 days. Preliminary experiments had shown that a dose level of 1 mg (lOx) induced significant tumor inhibition. For these studies, C225 was injected at a 0.5 mg (lOx) dose level.
As shown in Figure 9, C225 alone was effective in significantly inhibiting the growth of established DU 145 xenografts (p < 0.5). The overall therapeutic effect was apparent by day 34 and significant with respect to the control group by day 36 (Figure 9A). All tumors in the sham-injected group continued to grow throughout the course of the study (Figure 9B) but the anti-tumor effect of the antibody was seen throughout the study (Figure 9C). Although spontaneous remissions in PBS-treated animals were never seen in this model, 60% of the C225 treated animals were tumor free by day 60 (Figure 10 A) and remained tumor-free for an additional 90 days after termination of the antibody injections. In addition, tumors that did not disappear in the C225 group grew extremely slowly after treatment was stopped (day 55; Figure 9C) suggesting a long-lived effect of the antibody. There was no significant difference in the survival curves during the course of treatment (Figure 10B).
Example III-2 clearly shows that C225 was capable of inhibiting the growth of established, EGFR-positive DU 145 xenografts and could induce long-lived tumor remissions in a high percentage of treated animals. These results could not be predicted from the in vitro data.
Not all cell lines that express EGFR at levels similar to those seen in DU 145 cells respond to C225 in vivo. For example, KB cells (human epidermoid carcinoma) express about 2 x 105 EGFR per cell and activation of the receptors by EGF was blocked by C225 in vitro. However, KB xenografts did not respond to a treatment regimen including a 1 mg dose (xlO) of C225, a level able to induce complete remissions in 100% of animals carrying established A431 tumors. As surprisingly shown in Example III-2, treatment of mice innoculated with DU 145 tumor cells with C225 alone at a 0.5 mg dose (xlO) led to significant tumor regressions in all treated animals. Sixty percent of the mice were in complete remission following termination of the treatment. Blockage of receptor activaton by C225 also has clinical implications for the treatment of metastatic prostatic carcinoma in humans, especially during the late stages of the disease.
EXAMPLES
Example I. Materials
Example 1-1. Cell Lines and Media
A431 cells were routinely grown in a 1:1 mixture of Dulbecco's modified Eagle's medium and Ham's F-12 supplemented with 10% fetal bovine serum, 2mM L- glutamine, and antibiotics.
The androgen-independent and dependent human prostatic carcinoma cell lines (DU 145, PC-3 and LNaP) were obtained from the ATCC (Rockville MD) and routinely maintained in RPMI 1640 medium (Sigma, St. Louis, MO) supplemented with 10% fetal bovine serum (Intergen, Purchase NY) and 2 mM L-gluatmine (Sigma). Cells were checked regularly for the presence of mycoplasma.
Example 1-2. Preparation and Purification of M225 and C225
The 225 antibody was grown as ascites in pristane primed Balb/c mice. Ascites fluid was purified by HPLC (ABX and Protein G) and determined to be >95% pure by SDS PAGE.
Human clinical grade C225 was grown in proprietary serum free medium in 300 liter lots. After clarification, the concentrated broth was purified on a series of chromatographic columns and vialed under asceptic conditions. Purity was determined to be >99% by SDS PAGE.
Example 1-3. Preparation of Doxorubicin-C225 Conjugates
C225 doxorubicin conjugates (C225-DOX) were prepared using a modification of the method described by Greenfield et al. , Cancer Research 50, 6600-6607 (1990). Briefly, Doxorubicin was reacted with the crosslinking agent PDPH (3-[2- pyridyldithio]propionyl hydrazide) (Pierce Chemical Co.) to form the acyl hydrazone derivative doxorubicin 13-[3-(2-pyridyldithiol) propionyl] hydrazone hydrochloride. C225 was thiolated with the reagent N-succinimydyl 3-(pyridyldithio) propionate and reacted with doxorubicin hydrazone to form a conjugate containing a hydrazide as well as a disulfide bond. The complex was purified by gel filtration at neutral pH. The C225-doxorubicin conjugate was stable at neutral to alkaline pH (pH 7-8) and was stored at 4C. The conjugate was readily hydrolyzed at pH 6, releasing active Doxorubicin.
Example 1-4. Chimerization of Antibody 225
Example I-4A. Cloning of H and L Chain cDNAs
The media containing the 225 mouse hybridoma cell line was expanded to one liter in tissue culture flasks. Total cell RNA was prepared by lysing washed cells in guanidine isothiocyanate containing 2-mercaptoethanol, shearing the solution in a dounce homogenizer to degrade cell DNA and layering the preparation on a 10 ml cesium chloride cushion. After centrifugation at 24,000 rpm for 16 hr. the pellet was resuspended in Tris-EDTA (TE) buffer and precipitated with ethanol. The poly A(+) mRNA fraction was isolated by binding to and elution from oligo dT cellulose. A cDNA library was prepared using the poly A (+) mRNA as template and oligo dT as the primer. The second strand was synthesized by nick translation using RNase H and DNA polymerase I. The double-stranded DNA was passed through a 2 ml Sepharose G75 column to remove oligo dT and small entities. The purified DNA was then ligated into a polylinker with the sequence:
5' -AATTCTCGAGTCTAGA -3' (SEQ ID NO: 31)
which encodes an Eco RI four base sticky end for ligation to the cloning vector, and the restriction sites for Xho I and Xba I for subsequent manipulations of the cDNAs. The ligated cDNA was then size-selected by electrophoresis on a 5% polyacrylamide gel. The appropriate size fractions (-1500 bp for H chain and -900 bp for L chain cDNA) were electroeluted from gel slices and ligated to Eco Rl-digested lambda gtlO phage DNA. Libraries were generated by packaging the ligation products in vitro and plating recombinant phage on lawns of E. coli strain C600 HFL. Phage containing H and L cDNAs were identified by phage filter lifts that were hybridized with radiolabeled oligonucleotides of the mouse kappa and gamma constant region. The identified phage were restriction mapped.
Isolates with the longest cDNA inserts were subcloned in a plasmid vector (Eco RI-Bam HI fragments for heavy (H) chain V regions and Eco RI-Hpa I fragments for light (L) chain variable (V) regions) and DNA sequenced. The subcloned fragments contained the complete V region and a small portion of associated mouse constant (C) region. A total of eight L chain cDNAs were sequenced and represent four different mRNAs. Three full-length H chain cDNAs were sequenced encoding the same V region and a portion of the correct gamma 1 C region. Three other isolates containing gamma 2a sequence were also identified but were not studied further. To identify the correct L chain cDNA, a sample of mouse 225 antibody was sequenced by automated Edman degradation after first separating the H and L chains by SDS reducing gel electrophoresis and blotting to membranes. The sequence obtained for the L chain matched one of the cDNAs. This isolate was rearranged to J5 and was found to be 91% homologous with Vk T2. The H chain V region was found to be 96% homologous with VH 101 subgroup VII- 1.
Example I-4B. Adaption of cDNAs and Construction of Expression Vectors
The V regions were adapted for expression by ligating the body of each to a synthetic DNA duplex encoding the sequence between the closest unique restriction site to the V/C junction and the exact boundary of the V region. To this was ligated a second, short intron sequence which, when joined, restores a functional splice donor site to the V region. At the end of the intron for the L chain is a Bam HI site and at the end of the H chain intron is a Hind III site. The adapted L Chain V region was then isolated as a Xba I-Bam HI fragment (the Xba I site was in the original linker used for cDNA cloning) while the adapted H chain V region was isolated as a Xho I- Hind III fragment.
The expression vector pdHL2, containing human kappa and human gamma 1 constant regions, was used for insertion of the adapted L chain V region. The resulting plasmid, pdHL2-Vk(225), was then digested with Xba I and Bam HI and used for the insertion of the adapted L chain V region. The resulting plasmid, pdHL2- Vk(225), was then digested with Xho I and Hind III and used for the insertion of the adapted H chain V region. The final vector was identified by restriction mapping and identified as pdHL2-ch225.
Example I-4C. Expression of Chimeric 225 in Transfected Hybridoma Cells
The pdHL2-ch225 plasmid was introduced into hybridoma Sp2/0 Agl4 cells by protoplast fusion. The bacteria harboring the plasmid were grown to an optical density of 0.5 at 600 nm at which time chloramphenicol was added to arrest growth and amplify the plasmid copy number. The following day the bacteria were treated with lysozyme to remove the cell wall and the resulting protoplasts were fused to the hybridoma cells with polyethylene glycol 1500. After fusion, the cells were grown in antibodies to kill any surviving bacteria and were plated in 96-well plates. The selection medium (containing methotrexate (MTX) at 0.1 μM) was added after 24-48 hours to allow only the transfected cells to grow, by virtue of their expression of the marker gene (dehydrofolate reductase) present on the expression plasmid.
After two weeks, several MTX-resistant clones were obtained that were then tested for antibody expression. Culture supernatants were added to wells coated with an anti-human Ig (Fc-specific) antibody as the capture reagent. The detection system was an HRP-conjugated goat anti-human kappa antibody. The majority of clones were found to be secreting human antibody determinants and the three highest producers were further adapted to grow at 1 μM and then 5 μM methotrexate. Two of the lines, designated SdER6 and SdER14, continued to grow well at the higher levels of MTX and were subcloned by limiting dilution. The productivity of the subclones was tested by seeding cells at 2 X 105 cells per ml in growth medium and measuring the accumulated antibody on day 7. The two highest producers from the first subcloning were lines SdER6.25 and SdER14.10. These were subcloned a second time and the final three candidate lines were designated SdER6.25.8, SdER6.25.49, and SdER14.10.1. Clone SdER6.25.8 was selected based on expression of antibody.
Example 1-5. Analysis of C225 Expressed from SdER6.25.8
Studies with antibody produced from the clone SdER6.25.8 were conducted to characterize the nature of the antibody. Culture supernatants from the transfected cell clones expressing C225 antibody were tested for their ability to bind human tumor cells expressing different levels of EGF receptor. A431 epidermal carcinoma cells (high expressors) were intensely stained while M24 melanoma cells (expressing 10- fold fewer receptors) were moderately stained. A neuroblastoma line, IMR-32, which does not express EGF receptor, was not stained. Example 1-6. Effects of Chimerizing the C225 Antibody
The apparent Kd was found to be 0.1 and 0.201 nM for C225 and 1.17 and 0.868 nM for 225, using ELISA and SPR methods, respectively (Table 1). These results were similar to published data for C225 (Kd = 0.39 nM) and 225 (Kd = 0.79 nM, Kd = 1 nM) as shown in Table 1. The antibodies were found to inhibit the proliferation of cultured A431 cells to the same extent (Table 2). In addition, 225 and C225 were able to block EGF-induced phosphorylation of the EGFR in A431 cells. These results indicated that chimerization of 225 did not affect the biological properties of the antibody and increased the relative binding affinity of C225 for EGFR.
Example II. Methods and Assays
Example II- 1. Relative Affinity Measurements bv ELISA
The relative binding affinity of the antibodies was determined using an ELISA protocol previously described by Lokker et al J. Immunol. 146, 893-898 (1991). Briefly, A431 cells (104 or 105 per well) were grown in 96 well microtiter plates overnight at 37°C. Cells were fixed with 3.7% neutral buffered formalin for 10 minutes at room temperature. After washing three times with PBS, wells were blocked with 1% bovine serum albumin in Hank's balanced salt solution for two hours at room temperature. C225 or 225 were added to the wells at various concentrations (serial dilutions starting at 50 nM). After a two hour incubation at 37°C, plates were extensively washed with PBS and incubated with goat anti-human antibody (Sigma, St Louis MO; 1:1000) for one hour at 37°C. Plates were washed and the chromogen TMB (Kirkegaard and Perry, Gaithersburg MD) added for 30 minutes in the dark. The color reaction was stopped with 1 N sulfuric acid and the plates read in an ELISA reader at 450 nm. The relative binding affinity is defined as the concentration giving the half maximal OD. Example II -2. Affinity Constants of 225 and C225 using Surface Plasmon Resonance Technology fSPR.
The apparent binding affinities of M225 and C225 were also determined using the InAcore™ (Pharmacia Biosensor, Piscataway NJ; manufacturer's application note 301 and O'Shannessy et al, Anal. Biochem. 212, 457-468 (1993). Briefly, soluble recombinant EGFR was immobilized on sensor chips via amino groups as described by the manufacturer. Real time binding parameters of 225 and C225 to EGFR was established at various antibody concentrations and the apparent Kd was calculated from the binding rate constants obtained via non linear fitting using Biaevaluation™ 2.0 Software.
Example II-3. In vitro Inhibition of Cell Growth with 225 and C225
The in vitro inhibitory activity of 225 and C225 was determined by plating A431 cells (300-500 per well) in 96 microtiter plates in complete growth medium. After adding C225 or 225 in various concentrations (4 replicates per concentration), plates were incubated for 48 hours at 37°C followed by a 24 hour pulse with 3H-thymidine. Cells were harvested, collected on filter mats and counted in a Wallace Microbeta scintillation counter to determine percent inhibition. Percent inhibition compares the decrease in 3H thymidine incorporation of antibody-treated cells with cells grown in the absence of antibody.
Example H-4. Animal Studies
Athymic nude mice (nu/nu; 6-8 weeks old females) were obtained from Charles River Laboratories. Animals (10 mice per treatment group) were innoculated in the right flank with 107 A431 cells in 0.5 ml of Hank's balanced salt solution. Mice were observed until tumors were visible (about 7-12 days) and had reached an average volume of 150-300 mm3. At that time, antibody therapy was begun. The therapy included twice weekly intraperitoneal injections (varying concentrations in 0.5 ml of PBS) over 5 weeks. U 1 animals received injections of PBS. Tumors were measured two times per week and volumes calculated using the following formula: π/6 x larger diameter x (smaller diameter)2. Animals were followed at least 3 weeks after the final antibody treatment (8 weeks after the start of therapy) at which time U 1 and test animals with extremely large tumors were euthanized. Tumor free animals and animal with small tumors were followed for an additional 2-3 months. Statistical analysis of tumor growth in each of the studies was done using a two tailed Student's T-test.
In addition to demonstrating growth inhibitory effects of the antibodies, many animals were found to be in complete remission (i.e., tumor free). This biological effect was quantified as a Remission Index (RI), defined as the number of tumor free mice/total animals within a treatment group. Termination occured at the time of euthanasia for animals with large tumors, and 2-3 months later for other animals. Animals that died during treatment were excluded from this analysis. For example, one complete remission among eight surviving animals equals an RI of 0.125.
Example III. Biological Activity of C225
Example III-l . The Capacity of the Antibodies to Inhibit the Growth of A431 Xenografts in Nude Mice
Animals were innoculated in the flanks with A431 cells. Tumors of 150-300 mm3 appeared by day 7-10. Refering to Experiments 1-4 in Table 3, animals were then randomized and injected with PBS or 225 (Exp 1), PBS, 225, or C225 (Exp 2); and PBS or C225 (Exp 3 and 4). In Experiments 1-3, animals received injections of 1 mg of antibody (in 0.5 ml PBS) twice weekly over 5 weeks for a total dose of 10 mg of antibody per animal. In Exp 4, animals received one of three possible doses: 1, 0.5, and 0.25 mg/injection for total doses of 10, 5, and 2.5 mg, respectively. Tumors were measured twice weekly over the course of treatment. Tumor-free animals and animals with small tumors continued to be monitored for 2-3 months following the sacrificing of animals with large tumors.
Figure 1 shows the effect of 225 on the growth of A431 tumors in nude mice (Exp
1). The average tumor volumes of the experimental and U 1 groups were similar (Figure 1 A) and only one complete tumor remission was observed (Remission Index (RI) of 0.17; Figure IB and Table 3). A comparison of 225 and C225 is shown in Figure 2 (Exp 2 in Table 3). Although there was no significant difference in average tumor size between the groups, animals treated with C225 had an RI of 0.44 (i.e., 4/9 complete remissions) compared to an RI of 0.11 for 225 (Figure 2B and Table 3). The apparent tumor regression for the PBS U 1 group at day 37 (Figure 2 A) was attributable to the death of 3/10 animals at this time and the concommitant decrease in overall tumor volume. A similar RI for C225 was seen in Exp 3 (Figure 3B; RI = 0.4). In addition, inhibition of tumor growth by C225 was also found to be significant when compared to the growth of xenografts in PBS-treated mice (Figure 3A; p < 0.02 following day 32).
Because a number of animals receiving C225 showed tumor regressions at the 1 mg/injection level, the lowest biologically effective dose was defined. Figure 4 shows the results of the dose reponse experiment (Exp 4). All animals receiving 1 mg/injection underwent complete remission and remained tumor free for over 100 days following termination of the antibody injections (Figure 4A and B; Table 3). These results are highly significant with p values varying from p < 0.006 on day 33 to p < 0.0139 on day 59. In Experiments 2 and 3, about 40% of the animals receiving the 1 mg dose of C225 underwent complete remission although C225 showed significant tumor regression in Exp 3 (Figure 3). The increased efficacy of the 1 mg dose in Experiments 3 and 4 in significantly reducing average tumor volume versus U 1 may have occured because mice carrying smaller tumors were used at the start of the treatment protocols in these experiments (152 mm3 [Exp 4] and 185 mm3 [Exp 3] vs. 267 mm3 [Exp 2]). These data suggest that the clinical effectiveness of C225 may be related to tumor burden.
At the 0.5 mg dose in Exp 4, the overall inhibition of tumor growth was not statistically significant because of the large variations in tumor volume among animals of both the PBS and the 0.5 mg groups. However, the RI was high for the 0.5 mg group (RI = 0.63; Figure 4b and Table 2) indicating that the antibody induced anti-tumor responses in individual animals. Interestingly, the 0.5 mg dose group in Exp 4 had a higher RI than the 1 mg dose group in Exp 3. This result may be attributed to the effects of tumor burden. Although the average starting volume for tumors in the 0.5 mg dose group was 160 mm3, there was great variability in tumor size among individual animals. A number of animals carried smaller tumors (<100 mm3) that are most susceptible to the biological effect of C225. At 0.25 mg dose, average tumor growth appeared to be greater than the PBS U 1. This was due to the inclusion within this group of two animals with large tumors (760 and 1140 mm3) at the start of the treatments which resulted in an increase in average tumor volume during the course of Exp 4. Overall, there is no significant difference between these groups but it is interesting to note that one animal (1/8) at the 0.25 mg dose was tumor free at the end of the study (RI = 0.13). At day 47, there appeared to be a drop in the RI. At this time, a tumor reappeared in one mouse that had apparently undergone a complete remission. In this single case, C225 had a transient biological effect. This animal is not included in Table 3. As with the 1 mg dose group, tumor-free animals in the 0.5 and 0.25 mg groups remained tumor free a minimum of 2-3 months after the PBS control mice were sacrificed.
Table 1. DISSOCIATION CONSTANTS (Kd) FOR 225 AND C225
AS DETERMINED BY VARIOUS METHODS
Kd(nM)
* Scatchard results are expressed as Kd, SPR results as apparent Kd, and ELISA data as the apparent affinity, a relative measure of the Kd. See Materials and Methods for description of the generation of the ELISA and SPR data.
Table 2. IN VITRO INHIBITION OF A431 CELLS BY C225 AND 225
The results shown in Table 2 represent a typical experiment in which the ability of
225 and C225 to inhibit the growth of A431 was tested in vitro. Details are described above. Percent inhibition is defined as the decrease in 3-H thymidine incorporation of antibody-treated samples (4 replicates/concentration) versus cells growing in the absence of antibody.
Table 3 represents a comparison of complete tumor remissions in athymic nude mice carrying established A431 tumors following treatment with PBS, 225, or C225 twice weekly for 5 weeks. Animals were treated with 1 mg of antibody in 0.5 ml of PBS by the intraperitoneal route except for study 4, which is a dose response experiment in which mice were given 1, 0.5, or 0.25 mg/injection. Tumor measurements were done as described above. This chart describes the RI at the time when the animals (PBS control and test) carrying large tumors were euthanized. All animals showing complete remissions or small tumors were followed for an additional 2-3 months. The differences in total number of animals results from death of mice within these treatment groups during the course of the experiments. Table 3. REMISSION INDICES FOR ANIMALS INNOCULATED WITH A431 CELLS AND TREATED WITH 225 OR C225
EXP
* Tumor free animals/total number of surviving animals. Differences in the number of animals presented are the result of mice dying during the five week course of the various treatment regimens, and these were not included in the statistical analysis.
** The Remission Index (RI) is defined as the fraction of mice that were tumor free on the day when the PBS control mice and test animals with large tumors were euthanized. A complete remission at the 0.25 mg dose level showed a subsequent recurrance of tumor (day 47).
Example IH-2. Inhibition of Growth of Established Human Prostatic Carcinoma Xenografts in Nude Mice
Example III-2A. FACS Analysis of C225 Binding to DU 145. PC-3 and LNCaP
The relative expression levels of EGF receptor on DU 145, PC-3 and LNCaP cells was determined by FACS analysis. Cells were grown to near confluency in complete medium, removed from the flasks with non-enzymatic dissociation buffer (Sigma), and resuspended at 5-10 x 105 per tube in 100 ul of cold H-BSA (Hanks balanced salt solution containing 1% BSA). Ten micrograms C225 or an irrelevant myeloma- derived human IgGl (Tago, Burlingame CA) were added to the tubes and incubated on ice for 60 minutes. After washing with cold H-BSA, goat anti-human IgG conjugated to FITC (Tago, Burlingame CA) was added for an additional 30 minutes on ice. Cells were washed 2 times with cold H-BSA, resuspended in 1 ml of H-BSA, and analyzed using a Coulter Epics Elite cell sorter (Coulter, Hialeah FL). Baseline fluroescence was determined using the FITC-labelled secondary antibody alone and non-specific flurorescence was defined by the irrelevant isotype control. Data is presented as the Mean Fluroescence Intensity (MFI), which is an indirect measure of antigen density. MFI is defined as the mean channel fluorescence multiplied by the percentage of positive cells for each sample.
Example III-2B. Phosphorylation Assays on PC-3. DU 145. and LNCaP Cells
Phosphorylation assays were performed on PC-3, DU 145, and LNCaP cells to determine if the EGF receptors expressed by these cells were functional and inhibited by C225. Assays and Western blot analysis were performed as previously described by Gill et al, Nature 293, 305-307 (1981). Briefly, DU 145, PC-3, and LNCaP cells were grown to 90% confluency in complete medium and then starved in DMEM-0.5% calf serum 24 hours prior to experimentation. Cells were stimulated with EGF in the presence or absence of C225 for 15 minutes at room temperature. Monolayers were then washed with the ice cold PBS containing 1 mM sodium orthovanadate. Cells were lysed and subjected to SDS PAGE followed by Western blot analysis. The phosphorylation patterns were determined by probing the blot with a monoclonal antibody to phosphotyrosine (UBI, Lake Placid NY) followed by detection using the ECL method (Amersham).
Example III-2C Animal Studies
Athymic nude mice (nu/nu; 6-8 weeks old males; Charles River Labs, Wilmington MA) were innoculated subcutaneously in the right flank with 106 DU 145 in 0.2 ml of Hank's balanced salt solution mixed with 0.2 ml of matrigel. Mice were observed until tumors were visible (about 14-20 days post challenge) and had reached an average volume of about 100 mm3. Animals were weighed and randomly divided into treatment groups (10 animals per group). Antibody therapy, which included twice weekly intraperitoneal injections of 0.5 mg of C225 over 5 weeks, was begun. Control animals received injections of PBS. Preliminary studies established that there was no significant difference between the growth of DU 145 xenografts in animals treated with polyclonal, DU 145-absorbed human IgG compared to PBS. Tumors were measured two times per week and volumes calculated using the following formula: π/6 x larger diameter x (smaller diameter)2. Animals were followed for at least 3 weeks following the final antibody injection (8 weeks after the start of therapy), at which time control animals were euthanized. Tumor free animals and mice with small tumors were followed for an additional 2-3 months. Statistical analysis of tumor growth in each study was determined with a two tailed Student's T- test using the computer program SigmaStat (Jandel, San Rafael CA). A p value of < 0.05 was considered significant.
Example IH-3. Biological Activity of Peptides Containing CDR Regions of 225
This example demonstrates that peptides constructed using 225-CDR sequences had biological activity against cell lines that express EGF receptors. A series of six peptides were generated with the following sequences:
Heavy Chain
CDR-1 NYGVH
CDR-2 GVIWSGGNTDYNTPFTSR
CDR-3 RALTYYDYEFAYW (SEQ ID NO: 32)
Light Chain CDR-1 RASQSIGTNIH (SEQ ID NO: 33)
CDR-2 YASESIS (SEQ ID NO: 34) CDR-3 QQNNWP (SEQ ID NO: 35)
These peptides were dissolved in PBS at a concentration of 1 mg/ml. A431 cells were plated at 1000 cells per well in 96 well plates. Peptides were added at various concentrations. The chimeric C225 antibody and an irrelevant, isotype- matched immunoglobulin were used as a positive and negative U Is, respectively. Plates were incubated for 72 hours at 37 °C and pulsed overnight with 3H-thymidine. Cells were harvested and counted in a liquid scintillation counter. Percent inhibition is defined as the decrease in 3-H thymidine incorporation of antibody or peptide treated cells compared to cells grown in the absence of antibody or peptide.
As can be seen in Figure 5, A431 cells are inhibited by C225 and by heavy chain CDR-1 and heavy chain CDR-2 of monoclonal antibody 225. In contrast, isotype- matched irrelevant antibody and U 1 peptide did not inhibit A431 cells. These results indicate that heavy chain CDR-1 and -2 are able to inhibit the growth of A431 cells by interfering with the binding of ligand to the EGFR.
The biological activity of C225-DOX was evaluated in vitro using EGFR expressing cell lines A431 , KB and MDA-468 as well as EGFR non-expressing cell lines Molt-4 and SK-MEL-28. EGF receptor expression was verified by FACS analysis using C225 and C225-DOX conjugate. Assays were conducted over a 72h incubation period using 3[H] -thymidine and WST-1 as a read out. In all assays with EGFRc expressing cell lines, i.e., A431, KB and MDA-468 cells, C225-DOX exhibited high inhibition of cell proliferation when compared to no treatment or hlgGl U Is. Comparisons of equimolar concentrations of C225-DOX with doxorubicin alone or mixtures of C225 and doxorubicin showed a 4-5 fold higher inhibition using the C225-DOX conjugate. Inhibition of cell proliferation by C225- DOX was also seen in EGFRc nonexpressing cell lines at higher doses. The C225- DOX inhibition in EGFRc-negative cell lines was 5-15 fold lower than EGFRc- positive cell lines and was similar to inhibition seen with equimolar concentrations of doxorubicin alone. Representative results are shown in Figure 6 for activity of C225- DOX on 431 cells.
Example IV. Humanization of M225.
Example IV- 1. Abbreviations
Dulbecco's Modified Eagles Medium (DMEM); Foetal Calf Serum (FCS); ribonuceic acid (RNA); messenger RNA (mRNA); deoxyribonucleic acid (DNA); double-stranded DNA (ds-DNA); polymerase chain reaction (PCR); enzyme linked immunoabsorbant assay (ELISA); hour (hr); minute (min); second (sec); human cytomegalovirus (HCMV); polyadenylation (poly(A)+); immunoglobulin (IgG); monoclonal antibody (mAb); complementarity determining region (CDR); framework region (FR); Tris-borate buffer (TBE); bovine serum albumin (BSA); phosphate buffered saline (PBS); room temperature (RT); nanometre (nm); epidermal growth factor receptor (EGFR); Example IV-2. Materials
Media components and all other tissue culture materials are obtained from Life Technologies (UK), except for FCS which is purchased from JRH Biosciences (USA). The RNA isolation kit is obtained from Stratgene (USA) while the 1st strand cDNA synthesis kit is purchased from Pharmacia (UK). All the constituents and equipment for the PCR-reactions, including AmpliTaq®DNA polymerase, are purchased from Perkin Elmer (USA). The TA Cloning® kit is obtained from Invitrogen (USA) and the Sequenase® DNA sequencing kit is purchased from Amersham International (UK). Agarose (UltraPure™) is obtained from Life Technologies (UK). The Wizard™ PCR Preps DNA Purification Kit, the Magic™ DNA Clean-up System and XL 1 Blue competent cells are purchased from Promega Corporation (USA). All other molecular biological products are purchased from New England Biolabs (USA). Nunc-Immuno Plate MaxiSorp™ immunoplates are obtained from Life Technologies (UK). Both the goat anti-human IgG, Fcγ fragment specific, antibody and the goat anti-human IgG (H+L) / horseradish peroxidase conjugate are purchased from Jackson
ImmunoResearch Laboratories Inc. (USA). TMB substrate A and substrate B are obtained from Kirkegaard-Pery (USA). All other products for both ELISAs are obtained from Sigma (UK). Microplate Manager® data analysis software package is purchased from Bio-Rad (UK). The molecular modelling package QUANTA is obtained from the Polygen Corporation (USA) and the IRIS 4D workstation is purchased from Silicon Graphics (USA).
Example IV-3. PCR cloning and sequencing of the mouse variable region genes
The mouse M225 hybridoma cell line is grown, in suspension, using DMEM supplemented with 10% (v/v) FCS, 50 Units/ml penicillin / 50μg/ml streptomycin and 580 μg/ml L-glutamine. Approximately 108 viable cells are harvested, while the supernatent from the hybridoma cells is assayed by ELISA to confirm that they are producing a mouse antibody. From the 108 cells total RNA is isolated using a RNA Isolation kit according to the manufacturers instructions. The kit uses a guanidinium thiocyanate phenol-chloroform single step extraction procedure as described by Chomczynski and Sacchi (6). Also following the manufacturers instructions, a 1st Strand cDNA Synthesis kit is employed to produce a single-stranded DNA copy of the M225 hybridoma mRNA using the NotI-(dT)lg primer supplied in the kit. Approximately 5 μg of total RNA is used in a 33 μl final reaction volume. The completed reaction mix is then heated to 90 °C for 5 min, to denature the RNA-cDNA duplex and inactivate the reverse transcriptase, before being chilled on ice.
To PCR-amplify the mouse variable region genes the method described by Jones and Bendig (7) is followed. Essentially, two series of degenerate primers, one series designed to anneal to the leader sequences of mouse kappa light chain genes (i.e. MKV1-11; Table 4) and one series designed to anneal to the leader sequences of mouse heavy chain genes (i.e. MHV1-12; Table 5), are used in conjunction with primers designed to anneal to the 5 '-end of the mouse kappa light chain constant region gene (MKC; Table 4) and the 5'-end of the mouse γl heavy chain constant region gene (MHCG1; Table 5), respectively, to PCR-clone the mouse variable region genes of the M225 antibody. Separate reactions are prepared for each of the MKV and MHV degenerate primers, with their respective constant region primer. The PCR- reaction tubes are loaded into a Perkin Elmer 480 DNA thermal cycler and cycled (after an initial melt at 94 °C for 1.5 min) at 94 °C for 1 min, 50 °C for 1 min and 72 °C for 1 min over 25 cycles. At the completion of the last cycle a final extension step at 72 °C for 10 min is carried out before the reactions are cooled to 4 °C. Except for between the annealing (50 °C) and extension (72 °C) steps, when an extended ramp time of 2.5 min is used, a 30 sec ramp time between each step of the cycle is employed.
20 μl aliquots from each PCR-reaction are run on agarose gels to determine which have produced a PCR-product of the correct size. Those PCR-reactions that do appear to amplify full-length variable domain genes are repeated to produce independent PCR-clones and thereby minimise the effect of PCR-errors. 6 μl aliquots of those PCR-products of the correct size are directly cloned into the pCR ™II vector, provided by the TA Cloning® kit, and transformed into INVαF' competent cells as described in the manufacturers instructions. Colonies containing the plasmid, with a correctly sized insert, are identified by PCR-screening the colonies using the pCR™II Forward and pCR™II Reverse oligonucleotide primers described in Table 6 according to the method of Gϋssow and Clackson (8). The putative positive clones identified are finally double-stranded plasmid DNA sequenced using the Sequenase®DNA Sequencing kit according to the method of Redston and Kern (9).
Example IV-4. Construction of chimeric genes
The cloned mouse leader- variable region genes are both modified at the 5'- and 3'- ends using PCR-primers to create restriction enzyme sites for convenient insertion into the expression vectors, a Kozak sequence for efficient eukaryotic translation of the mRNA encoding the respective immunoglobulin chains (10) and a splice-donor site for the correct RNA splicing of the variable and constant region genes. A Hndlll site is added to the 5 '-end of both mouse variable region genes, however, different restriction sites attached to the 3 '-end of the mouse variable region genes i.e. a BamHl site at the 3'-end of the VΗ gene and aXbal site at the 3'-end of the Vκ gene.
PCR-reactions are prepared according to the method for the construction of chimeric genes in Kettleborough et al. (11), using the primers C225VH5' and
C225VH3' for the heavy chain, and C225VK5' and C225VK3' for the kappa light chain (Table 7). Following an initial melting step at 94 °C for 90 sec the mixes are PCR- amplified at 94 °C for 2 min and 72 °C for 4 min over 25 cycles. This two step PCR- cycle, as opposed to the more usual three step cycle, is possible because each of the primers is designed to anneal to the template DNA over 24 bases which allows them to anneal at the relatively high temperature of 72 °C. A 30 sec ramp time is used between each step and at the end of the last cycle, the PCR-reactions are completed with a final extension step at 72 °C for 10 min before cooling to 4 °C. The PCR-products are column purified using a Wizard™PCR Preps DNA Purification kit according to the manufacturers instructions, digested with the appropriate restriction enzymes, as is plasmid pUC19, and separated on a 1% agarose / TBE buffer (pH8.8) gel. The heavy and kappa light chain variable region genes are excised from the agarose gel and purified using a Wizard' PCR Preps DNA Purification kit. The pUC19 is also excised from the agarose gel and purified using the Magic™DNA Clean-up System as per the manufacturers instructions. The heavy and kappa light chain variable region genes are then separately ligated into the purified pUC19 to produce plasmids pUC-C225VH and pUC-C225Vκ, respectively, and transformed into XL 1 Blue competent cells. Putative positive colonies containing the appropriate plasmid are then identified by PCR- screening, using oligonucleotide primers RSP and UP (Table 6) and finally ds-DNA sequenced both to confirm the introduction of the sequence modifications and also to prove that no unwanted changes to the DNA sequence have occured as a consequence of the PCR-reactions.
To modify the signal peptide sequence at the 5 '-end of the kappa light chain variable region PCR-mutagenesis is used, according to the protocol described by Kettleborough et al (11). PCR-primers C225Vκ5'sp and C225Vκ3'sp (Table 7) are used on pUC-C225Vκ template DNA to create the modified gene (C225Vκsp) using the modified two step PCR amplification protocol. The PCR-product is then column purified before digesting both the purified PCR-product and pUC-C225Vκ with Hindϊll and Pstl. The PCR-fragment and the plasmid DNA are then agarose gel- purified, ligated together and cloned to create plasmid pUC-C225Vκsp. As before, putative positive transformants are identified via a PCR-screen (using the RSP and UP primers) and then ds-DNA sequenced to confirm both the presence of the modified signal peptide and the absence of PCR-errors.
The adapted mouse kappa light and heavy chain leader- variable region genes are then directly inserted, as a H dIII-5 τwHI fragment in the case of the mouse VH and as a Hinάlll-Xbal fragment in the case of the mouse Vκ, into vectors designed to express chimeric light and heavy chains in mammalian cells. These vectors contain the ΗCMV enhancer and promoter to drive the transcription of the immunoglobulin chain, a MCS for the insertion of the immunoglobulin variable region gene, a cDNA clone of the appropriate human kappa light or heavy chain constant region, a synthetic poly(A)+ sequence to polyadenylate the immunoglobulin chain mRNA, an artificial sequence designed to terminate the transcription of the immunoglobulin chain, a gene such as dhfr or neo for selection of transformed stable cell lines, and an SV40 origin of replication for transient DNA replication in COS cells. The human kappa light chain mammalian expression vector is called pKN 100 (Figure 11) and the human γ 1 heavy chain mammalian expression vector is called pGlD105 (Figure 12). Putative positive colonies are both PCR-screened, using primers ΗCMVi and New.Ηuκ for the chimeric kappa light chain vector and primers ΗCMVi and ΗuCγl for the chimeric heavy chain vector (Table 6), and undergo restriction analysis to confirm the presence of the correct insert in the expression vector constructs. The new constructs containing the mouse variable region genes of the M225 antibody are called pK 100-C225Vκ (or pKN100-C225Vκsp ) and pGlD105-C225VΗ, respectively.
Example IV-5. Molecular modelling of mouse M225 antibody variable regions
To assist in the design of the CDR-grafted variable regions of the H225 antibody, a molecular model of the variable regions of the mouse M225 antibody is built. Modelling the structures of well-characterized protein families like immunoglobulins is achieved using the established method of modelling by homology. This is done using an IRIS 4D workstation running under the UNIX operating system, the molecular modelling package QUANTA and the Brookhaven crystallographic database of solved protein structures (12). The FRs of the M225 variable regions are modelled on FRs from similar, structurally- solved immunoglobulin variable regions. While identical amino acid side chains are kept in their original orientation, mismatched side chains are substituted using the maximum overlap procedure to maintain chi angles as in the original mouse M225 antibody. Most of the CDRs of the M225 variable regions are modelled based on the canonical structures for hypervariable loops which correspond to CDRs at the structural level (13-16). However, in cases such as CDR3 of the heavy chain variable region, where there are no known canonical structures, the CDR loop is modelled based on a similar loop structure present in any structurally-solved protein. Finally, in order to relieve unfavourable atomic contacts and to optimize Van der Waals and electrostatic interactions, the model is subjected to energy minimization using the CHARMm potential (17) as implemented in QUANTA.
The FRs from the light chain variable region of M225 antibody are modelled on the FRs from the Fab fragment of mouse monoclonal antibody HyHel-10 (18). The FRs from the heavy chain variable region are modelled on the FRs from the Fab fragment of mouse monoclonal antibody D 1.3 (19). Those amino acid side chains which differ between the mouse M225 antibody and the variable regions upon which the model is based are first substituted. The light chain of Fab HyHel-10 antibody is then superimposed onto the light chain of D1.3 by matching residues 35-39, 43-47, 84-88 and 98-102, as defined by Kabat et al, (20). The purpose of this is to place the two heterologous variable regions, i.e. the HyHel-10-based kappa light chain variable region and the D1.3-based heavy variable region, in the correct orientation with respect to each other.
CDR1 (LI) of the light chain variable region of mAb M225 fits into the LI canonical group 2, as proposed by Chothia et al. (14), except for the presence of an isoleucine, instead of the more usual leucine, at canonical residue position 33. However, this substitution is considered too conservative to merit significant concern in assigning a canonical loop structure to this hypervariable loop. The LI loop of mouse Fab HyHel-10 is identical in amino acid length and matches the same canonical group - with a leucine at position 33 - as the LI loop of M225 mAb. Consequently this hypervariable loop is used to model the LI loop of M225 kappa light chain variable region. Similarly, CDR2 (L2) and CDR3 (L3) of the M225 mAb both match their respective canonical group 1 loop structures. In addition, the corresponding hypervariable loop structures of the HyHel-10 Fab fragment are also both group 1. Accordingly, the L2 and L3 loops of the M225 kappa light chain variable region are modelled on L2 and L3 of Fab HyHel-10.
Likewise, CDRl (HI) and CDR 2 (H2) hypervariable loops of the heavy chain variable region of mAb M225 both fit their respective canonical group 1 loop structures as defined by Chothia et al. (14). Moreover, the corresponding HI and H2 hypervariable loops of mouse D1.3 Fab fragment also match their respective canonical group 1 loop structures. Consequently, as with the light chain, these hypervariable loops are modelled on the HI and H2 loops of the heavy variable region upon which the model is based. To identify a matching loop structure to the CDR3 (H3) hypervariable loop of the heavy chain variable region of M225 the Brookhaven database is searched for a loop of identical length and similar amino acid sequence. This analysis found that the H3 loop of the mouse Fab 26/9 (21) exhibited the closest match to the H3 loop of M225 mAb and is consequently used as the basis for this hypervariable loop in the mouse M225 variable region model. After adjusting the whole of the model for obvious steric clashes it is finally subjected to energy minimization, as implemented in QUANTA, both to relieve unfavourable atomic contacts and to optimize van der Waals and electrostatic interactions.
Example IV-6. Design of the reshaped human H225 antibody variants.
The first step in designing the CDR-grafted variable regions of the H225 antibody is the selection of the human light and heavy chain variable regions that will serve as the basis of the humanized variable regions. As an aid to this process the M225 antibody light and heavy chain variable regions are initially compared to the consensus sequences of the four subgroups of human kappa light chain variable regions and the three subgroups of human heavy chain variable regions as defined by Kabat et al. (20). The mouse M225 light chain variable region is most similar to the consensus sequences of both human kappa light chain subgroup I, with a 61.68% identity overall and a 65.00% identity with the FRs only, and subgroup III, with a 61.68%) identity overall and a 68.75% identity with the FRs only. The mouse M225 heavy chain variable region is most similar to the consensus sequence for human heavy chain subgroup II with a 52.10% identity overall and a 57.47% identity between the FRs alone. This analysis is used to indicate which subgroups of human variable regions are likely to serve as good sources for human variable regions to serve as templates for CDR-grafting, however, this is not always the case due to the diversity of individual sequences seen within some of these artificially constructed subgroups.
For this reason the mouse M225 variable regions are also compared to all the recorded examples of individual sequences of human variable regions publically available. With respect to human antibody sequences, the mouse M225 light chain variable region is most similar to the sequence for the human kappa light chain variable region from human antibody LS7'CL (22) - which is not related to the mouse L7'CL sequence. The kappa light chain variable region of human LS7'CL is a member of subgroup III of human kappa light chain variable regions. The overall sequence identity between the mouse M225 and human LS7'CL light chain variable regions is calculated to be 64.42% overall and 71.25% with respect to the FRs alone. The mouse M225 heavy chain variable region is most similar to the sequence for the human heavy chain variable region from human antibody 38P1'CL (23). Surprisingly, the heavy chain variable region of human 38P1'CL is a member of subgroup III and not subgroup II of the human heavy chain variable regions. The overall sequence identity between the mouse M225 and human 38P1'CL heavy chain variable regions is calculated to be 48.74% while the identity between the FRs alone is 58.62%. Based on these comparisons, human LS7'CL light chain variable region is selected as the human FR donor template for the design of reshaped human M225 light chain variable region and human 38P1'CL heavy chain variable region is selected as the human FR donor template for the design of reshaped human M225 heavy chain variable region.
As is commonly seen, the human light and heavy chain variable regions that are selected for the humanization of the M225 antibody are derived from two different human antibodies. Such a selection process allows the use of human variable regions which display the highest possible degree of similarity to the M225 variable regions. In addition, there are many successful examples of CDR-grafted antibodies based on variable regions derived from two different human antibodies. One of the best studied examples is reshaped human CAMPATH-1 antibody (24). Nevertheless, such a strategy also requires a careful analysis of the interdomain packing residues between the kapp light chain and heavy chain variable regions. Any mis-packing in this region can have a dramatic affect upon antigen binding, irrespective of the conformation of the CDR loop structures of the reshaped human antibody. Consequently, the amino acids located at the VK/VH interface, as defined by Chothia et al. (25), are checked for unusual or rare residues. Any residues so identified are then considered for mutagenesis to an amino acid more commonly seen at the specific residue position under investigation.
The second step in the design process is to insert the M225 CDRs, as defined by Kabat et al. (20), into the selected human light and heavy chain variable region FRs to create a simple CDR-graft. It is usual that a mouse antibody that is humanized by a simple CDR-graft in this way, will show little or no binding to antigen. Consequently, it is important to study the amino acid sequences of the human FRs to determine if any of these amino acid residues are likely to adversely influence binding to antigen, either directly through interactions with antigen, or indirectly by altering the positioning of the CDR loops.
This is the third step of the design process where decisions are made as to which amino acids in the human donor FRs should be changed to their corresponding mouse M225 rsidues in order to achieve good binding to antigen. This is a difficult and critical step in the humanization procedure and it is at this stage that the model of the M225 variable regions becomes most useful to the design process. In conjunction with the model the following points are now addressed.
It is of great importance that the canonical structures for the hypervariable loops (13- 16) are conserved. It is therefore crucial to conserve in the humanized H225 variable regions any of the mouse FR residues that are part of these canonical structures. It is also helpful to compare the sequence of the M225 antibody to similar sequences from other mouse antibodies to determine if any of the amino acids are unusual or rare as this may indicate that the mouse residue has an important role in antigen binding. By studying the model of the M225 variable regions, it is then possible to make a prediction as to whether any of these amino acids, or any other residues at particular positions, could or could not influence antigen binding. Comparing the individual human donor sequences for the kappa light and heavy chain variable regions to the consensus sequence of human variable regions subgroups to which the donor sequences belong, and identifying amino acids that are particularly unusual is also important. By following this design process a number of amino acids in the human FRs are identified that should be changed from the amino acid present at that position in the human variable region to the amino acid present at that position in the Mouse M225 variable region.
Table 8 describes how the first version (225RKA) of the reshaped human H225 kappa light chain variable regions is designed. There is only one residue in the reshaped human FRs where it is considered necessary to change the amino acid present in the human FRs to the amino acid present in the original mouse FRs. This change is at position 49 in FR2, as defined by Kabat et al. (20). The tyrosine found in human LS7'CL kappa light chain variable region is changed to a lysine, as found in mouse M225 kappa light chain variable region. From the model it appears that the lysine in M225 is located close to CDR3 (H3) of the heavy chain variable region and may be interacting with it. The residue is also positioned adjacent to CDR2 (L2) of the kappa light chain variable region and is rarely seen at this location amonst the members of mouse kappa light chain subgroup V, as defined by Kabat et al. (20), to which the M225 kappa light chain variable region belongs. For these reasons it is felt prudent to conserve the mouse lysine residue in 225RKA.
A second version is also made of the reshaped human kappa light chain (225RKB) which reverses the FR2 modification made in 225RKA, by replacing the lysine at position
49 with the original human tyrosine amino acid. Consequently, this version of the reshaped human kappa light chain will contain no mouse residues in the FRs whatsoever.
With respect to the design of reshaped human H225 heavy chain variable region, Table 9 shows the first version (225RHA). In all there are eight residues in the reshaped human FRs where it is considered necessary to change the amino acid present in the human 38P1'CL FRs to the amino acids present in the original mouse M225 FRs (i.e. A24V, T28S, F29L, S30T, V48L, S49G, F67L and R71K). At positions 24, 28, 29 and 30 in FR1 the amino acid residues as present in the mouse sequence are retained in the reshaped human H225 heavy chain variable region because they represent some of the canonical residues important for the HI hypervariable loop structure (14). Since canonical residues are so critical for the correct orientation and structure of hypervariable loops that they are generally always conserved in the reshaped variable region. Moreover, residue positions 24-30 are considered part of the HI hypervariable loop itself and so are even more critical to the correct conformation and orientation of this loop and justifying their conservation even more strongly. Similarly, residue position 71 in FR3 is another position in the heavy chain variable region which has been identified by Chothia et al. (14) as one of the locations important for the correct orientation and structure of the H2 hypervariable loop and, as such, is one of the canonical amino acids of CDR2. Consequently, the lysine in the mouse will replace the arginine in the human at this residue position. At positions 48 and 49 in FR2 and 67 in FR3, the valine, serine and phenylalanine residues (respectively) present in the human 38P1'CL VH sequence are changed to leucine, glycine and leucine (respectively) as present in the mouse M225 VH sequence. This descision is made on the basis of the model which shows that all three residues are buried underneath the H2 loop and so could influence the conformation of the hypervariable loop and hence interfere with antigen binding. These are then the mouse residues conserved in the first version of the reshaped human H225 heavy chain variable region.
Version B of the reshaped human H225 heavy chain variable region (225RHB) incorporates all the substitutions made in 225RHA and, in addition, contains a further mouse residue. At position 41 in FR2 the human threonine residue is replaced by proline which is invariably seen at this position in the mouse subgroup IB and is also very commonly seen in human subgroup III. In contrast, threonine is not usually seen at this location in the human subgroup III (only ' V87 times) and from the model it is appears that the residue is located on a turn located on the surface of the M225 VH region. What effect this may have on hypervariable loop structures is unclear, however, this version of the reshaped human H225 heavy chain variable region should clarify this.
Version C of the reshaped human H225 heavy chain variable region (225RHC) incorporates all the substitutions made in 225RHA and, in addition, contains a further two mouse residues located at position 68 and 70 in FR3. From the model of the mouse M225 variable region, both the serine at position 68 and the asparagine at position 70 appear to be on the surface and at the edge of the antigen binding site. Since there is a possibility that either or both amnio acids could directly interact with EGFR, both the threonine at position 68 and the seine at position 70 in the human FRs are replaced with the corresponding mouse residues in 225RHC.
Version D of the reshaped human H225 heavy chain variable region (225RHD) simply incorporates all the mouse FR substitutions made in 225RHA, 225RHB and 225RHC to determine the combined effect of these changes.
Version E of the reshaped human H225 heavy chain variable region (225RHE) incoiporates all the substitutions made in 225RHA and, in addition, incorporates another residue change at position 78 in FR3. From the model there is some evidence to suggest that the mouse amino acid (valine) at position 78 could influence the conformation of the HI hypervariable loops from its location buried underneath CDR1. Consequently, the human residue (leucine) is replaced by the mouse amino acid in 225RHE.
Example IV-7. Construction of the humanized antibody variable region genes
The construction of the first version of the reshaped human H225 Vκ region (225RKA) is carried out essentially as described by Sato et al. (26). In essence, this involves annealing PCR-primers encoding FR modifcations (Table TO) onto a DNA template of the chimeric C225VK gene using the two step PCR-amplification protocol to synthesize the reshaped human variable region gene. As a consequence, the FR DNA sequence of the chimeric C225VK is modified by the primers to that of the reshaped human kappa light chain variable region gene 225RKA. The newly synthesized reshaped variable region gene, following column purification, is digested with Hrødlll and Xbal, agarose gel-purified and subcloned into pUC19 (digested and agarose gel-purified in an identical manner). The new plasmid construct, pUC-225RKA, is then transformed into XL 1 Blue competent cells. Putative positive clones are identified by PCR-screening (using primers RSP and UP) and then finally ds-DNA sequenced, both to confirm their integrity and discount the presence of PCR-errors. From the confirmed postive clones an individual clone is selected and directly inserted, as a Hindlϊl-Xbal fragment, into the human kappa light chain mammalian expression vector (pKNl 00) to create the plasmid pKN100-225RKA. The integrity of this vector construct is confirmed via PCR-screening (using primers ΗCMVi and New.Ηuκ) and restriction digest analysis.
Version B of the reshaped human Η225 Vκ (225RKB) is constructed using oligonucleotide primers 225RKB.K49Y and APCR40 (Table 11). A 100 μl PCR-reaction mix comprising 65.5 μl of sterile distilled/deionized water, 5 μl of 2 ng/μl plasmid pUC-
225RKA template DNA, 10 μl of 10 X PCR buffer II, 6 μl of 25 mM MgCl2, 2 μl each of the 10 mM stock solutions of dNTPs, 2.5 μl aliquots (each of 10 μM) of primers 225RKB.K49Y and APCR40 and 0.5 μl of AmpliTaq®DNA polymerase is overlayed with 50 μl of mineral oil and loaded into a DNA thermal cycler. The PCR-reaction is PCR-amplified, using the two step protocol over 25 cycles, and the PCR-product column purified before it is cut with Mscϊ. Plasmid pUC-225RKA is also cut with Mscl and both the digested PCR product and the plasmid fragment are agarose gel-purified. The PCR- product is then cloned into pUC-225RKA, to create pUC-225RKB, before being transformed into XL 1 Blue competent cells. Putative positive transformant are first identified, using primers 225RKB.K49Y and UP in a PCR-screening assay, and then confirmed via ds-DNA sequencing. A selected individual clone is finally sublconed into pKNIOO to produce the plasmid pKN100-225RKB, whose correct construction is confirmed both by using primers HCMVi and New.Huκ (Table 6) in a PCR-screening assay and restriction analysis.
The construction of the first version of the reshaped human H225 VH region (225RHA) is also carried out essentially as described by Sato et al. (26). In the case of the reshaped human 225RHA gene this involves annealing PCR-primers (Table 12) onto both a DNA template of a prevoiusly humanized mAb, to create the 5 '-half of the reshaped human kappa light chain variable region gene, and the chimeric C225VH gene, to synthesize the 3 '-half of the reshaped human kappa light chain variable region gene. Again, the two step PCR-amplification protocol is used and the reshaped variable region gene created is cloned into pUC19 vector, as an agarose gel-purified Hindlll-BamΑl fragment, to create plasmid pUC-225RHA. Putative positive clones identified by PCR- screening (using primers RSP and UP) are finally ds-DNA sequenced both to confirm the DNA sequence and prove the absence of PCR-errors. From the confirmed positive clones an individual clone is selected and directly inserted, as a Hindffl-BamHI fragment, into the human γl heavy chain mammalian expression vector pGlD105 to create plasmid pGlD105-225RHA. The construction of this plasmid is then confirmed both by using primers HCMVi and γAS (Table 6) in a PCR-screening assay and restriction analysis. Versions B of the reshaped human H225 VH (225RHB ) is synthesized in a two step PCR-mutagenesis procedure in the following manner. Two separate 100 μl PCR- reaction mixes are first prepared by combining 65.5 μl of sterile distilled/deionized water, 5 μl of 2 ng/μl plasmid pUC-225RHA template DNA, 10 μl of 10 X PCR buffer II, 6 μl of 25 mM MgCl2, 2 μl each of the 10 mM stock solutions of dNTPs, 2.5 μl aliquots (each of 10 μM) of primers APCR10 and 225RHB.T41P-AS in the first PCR- reaction, and primers APCR40 and 225RHB.T41P-S in the second PCR-reaction (Table 13), and finally 0.5 μl of AmpliTaq®DNA polymerase. Each of the two PCR- reaction mixes are overlayed with 50 μl of mineral oil, loaded into a DNA thermal cycler and PCR-amplified using the two step protocol over 25 cycles. The two PCR- products are then agarose gel-purified, to separate them from any template DNA remaining in the PCR-reaction, before being resuspended in 50μl of distilled/deionized water and their concentration determined.
In a second PCR-reaction 20pmol aliquots of each of the two PCR-products from the first PCR-reaction (equivalent to 8 μl of the APCR10/225RHB.T41P-AS PCR product and 10 μl of the APCR40/225RHB.T41P-S PCR-product) are added to 57.5 μl of sterile distilled/deionized water, 10 μl of 10 X PCR buffer II, 6 μl of 25 mM MgCl2, 2 μl each of the 10 mM stock solutions of dNTPs and 0.5 μl of AmpliTaq®DNA polymerase. This PCR-reaction is overlayed with mineral oil and PCR-amplified using the two step protocol over 7 cycles only. A third PCR-reaction is then prepared comprising 1 μl of the product of the second PCR-reaction 69.5 μl of sterile distilled/deionized water, 10 μl of 10 X PCR buffer II, 6 μl of 25 mM MgCl2, 2 μl each of the 10 mM stock solutions of dNTPs, 2.5 μl aliquots (each of 10 μM) of the nested primers RSP and UP and 0.5 μl of AmpliTaq®DNA polymerase. The PCR-reaction is overlayed with mineral oil and amplified using the two step protocol for a final 25 cycles. This PCR-product is then column purified, isolated as an agarose gel purified HmdIII-itomHI fragment, subcloned into HindlU-BamHl digested and agarose gel -purified plasmid pUC19, and finally transformed into XL 1 Blue competent cells. Putative positive transformants are first identified and then confirmed as described previously. A selected individual clone is then sublconed into pGlD105 to produce the plasmid pGlD105-225RHB - which is confirmed using primers HCMVi and γAS (Table 6) in a PCR-screening assay and by restriction analysis.
Version C of the reshaped human H225 VH (225RHB ) is synthesized in a similar manner to 225RKc. A 100 μl PCR-reaction mix containing 65.5 μl of sterile distilled/deionized water, 5 μl of 2 ng/μl plasmid pUC-225RHA template DNA, 10 μl of 10 X PCR buffer II, 6 μl of 25 mM MgCl2, 2 μl each of the 10 mM stock solutions of dNTPs, 2.5 μl aliquots (each of 10 M) of primers APCR40 and 225RHC.T68S/S70N (Table 13) and 0.5 μl of AmpliTaq®DNA polymerase. The PCR-reaction is overlayed with mineral oil PCR-amplified, using the two step protocol over 25 cycles, and column purified prior to digestion with Sail and BamHl. Plasmid pUC-225RHA is also cut with with Sail and BamHl and both the digested PCR product and the plasmid are agarose gel-purified. The PCR-product is then cloned into pUC-225RHA, to create pUC-225RHc, before being transformed into XL 1 Blue competent cells. Putative positive transformant are first identified, using primers RSP and UP in a PCR-screennig assay, and later confirmed via ds-DNA sequencing. A selected individual clone is then sublconed into pGlD105 to produce the plasmid pGlD105-225RHc. The correct construction of this vector finally proven both by using primers HCMVi and γAS (Table 6) in a PCR-screening assay and restriction analysis.
Version D of the reshaped human H225 VH(225RHD) is a product of the changes incorporated into versions B and C of the reshaped human heavy chain of H225 antibody. Fortuitously, it is possible to amalgamate the changes made to these heavy chain variable region genes by digesting both pUC-225RHB and pUC-225RHc with Sa l and BamHl. The 2.95 kb vector fragment from pUC-225RHB and the approximately 180 bp insert fragment from pUC-225RHc are then agarose gel- purified before being ligated together and transformed into XL 1 Blue competent cells. Positive transformant are identified and ds-DNA sequenced before a selected individual clone is sublconed into pGlD105 to produce the plasmid pGlD105- 225RHD. The correct construction of this vector is finally confirmed as described previously.
Version E of the reshaped human H225 VH(225RHE) is a derivative of 225RHA and is synthesized in an identical manner to 225RHC using primers APCR40 and 225RHE.L78V (Table 13). A selected 225RHE clone from plasmid pUC-225RHE is then sublconed into pGlD105 to produce the vector pGlD105-225RHE - the correct construction of which is proven in the usual manner.
Example IV-8. Transfection of DNA into COS cells
The method of Kettleborough et al. (11) is followed to transfect the mammalian expression vectors into COS cells.
Example IV-9. Protein A purification of recombinant 225 antibodies
Both the chimeric C225 antibody and the various reshaped human H225 antibody constructs are protein A purified according to the protocol described in Kolbinger et al. (27).
Example IV-10. Mouse Antibody ELISA
Each well of a 96-well Nunc-Immuno Plate MaxiSorp™ immunoplate is first coated with 100 μl aliquots of 0.5 ng/μl goat anti-mouse IgG (γ-chain specific) antibody, diluted in coating buffer (0.05 M Carbonate-bicarbonate buffer, pH 9.6), and incubated overnight at 4 °C. The wells are blocked with 200 μl/well of mouse blocking buffer (2.5% (w/v) I _ in PBS) for 1 hr at 37 °C before being washed with 200 μl/well aliquots of wash buffer (PBS / 0.05% (v/v) tween-20) three times. 100 μl/well aliquots of the experimental samples (i.e. harvested media from the M225 hybridoma cell line - spun to remove cell debris) and 1 :2 sample dilutions, diluted in sample-enzyme conjugate buffer (0.1 M Tris-HCl (pH 7.0), 0.1 M NaCl, 0.02% (v/v) tween-20 and 0.2% (w/v) BSA), are now dipensed onto the immunoplate. In addition, a purified mouse IgG standard, serially diluted 1 :2 from a starting concentration of 1000 ng/ml, is also loaded onto the immunoplate. The immunoplate is incubated at 37 °C for 1 hr and washed three times with 200 μl/well of wash buffer. 100 μl of goat anti-mouse IgG/horseradish peroxidase conjugate, diluted 1000-fold in sample- enzyme conjugate buffer, is now added to each well, following which the immunoplate is incubated at 37 °C for 1 hr before it is washed as before. 100 μl aliquots of TMB peroxiodase substrate A:peroxidase substrate B ( 1 : 1 ) are now added to each well and incubated for 10 min at RT in the dark. The reaction is halted by dispensing 50 μl of 1 N H2SO4 into each well. The optical density at 450 nm is finally determined using a Bio-Rad 3550 microplate reader in conjunction with Microplate Manager™.
Example IV- 11. Quantification of whole human γl/κ antibody via ELISA
Each well of a 96-well Nunc-Immuno Plate MaxiSorp™ immunoplate is first coated with 100 μl aliquots of 0.4 ng/μl goat anti-human IgG (Fcγ fragment specific) antibody, diluted in coating buffer (0.05 M Carbonate-bicarbonate buffer, pH 9.6), and incubated overnight at 4 °C. The wells are then each blocked with 200 μl of human blocking buffer (2% (w/v) BSA in PBS) for 2 hr at RT before being washed with 200 μl/well aliquots of wash buffer (PBS / 0.05% (v/v) tween-20) three times. 100 μl/well aliquots of the experimental samples (i.e. harvested cos cell supernatents - spun to remove cell debris) and 1 :2 sample dilutions, diluted in sample-enzyme conjugate buffer (0.1 M Tris-HCl (pH 7.0), 0.1 M NaCl, 0.02% (v/v) tween-20 and 0.2% (w/v) BSA), are now dipensed onto the immunoplate. In addition, a purified human γl/κ antibody, which is used as a standard and serially diluted 1 :2, is also loaded onto the immunoplate. The immunoplate is incubated at 37 °C for 1 hr before being washed with 200 μl well of wash buffer three times. 100 μl of goat anti-human kappa light chain/horseradish peroxidase conjugate, diluted 5000-fold in sample- enzyme conjugate buffer, is added to each well, following which the immunoplate is incubated at 37 °C for 1 hr before it is washed as before. The remainder of the protocol is identical to the mouse antibody ELISA.
Example IV- 12. A4 1 Cell ELISA for the detection of EGFR antigen binding
The procedure is based upon the one provided by ImClone Systems Inc. to determine the relative binding affinity of the recombinant 225 antibody constructs, to EGFR expressed on the surface of A431 cells. The A431 cells are plated onto a 96- well flat bottomed tissue culture plate and incubated overnight in DMEM media with 10% (v/v) FBS at 37 °C and 5% CO2. The following day the media is removed, the cells are washed once in PBS and then fixed with 100 μl/well of 0.25% (v/v) gluteraldehyde in PBS. This is removed and the plate is washed again in PBS before it is blocked with 200 μl/well of 1% (w/v) BSA in PBS for 2 hr at 37 °C. The blocking solution is removed and 100 μl/well aliquots of the experimental samples (i.e. harvested COS cell supernatents - spun to remove cell debris) and 1 :2 sample dilutions thereof (diluted in 1% (w/v) BSA in PBS) are dispensed onto the tissue culture plate. In addition, 80 μl well aliquots of purified human γl/κ antibody, which is used as a standard and serially diluted 1 :5 from a starting concentration of 20 μg/ml, is also loaded onto the plate. The plate is incubated at 37 °C for 1 hr and then washed with 200 μl/well of 0.5% (v/v) tween-20 in PBS, three times. 100 μl of goat anti-human IgG (H+L)/horseradish peroxidase conjugate, diluted 5000-fold in 1% (w/v) BSA in PBS, is now added to each well, following which the plate is incubated at 37 °C for 1 hr before being washed first with 200 μl/well of 0.5% (v/v) tween-20 in PBS (three times) and then distilled deionized water (twice). The remainder of the protocol is identical to the mouse antibody ELISA. Example IV-13. Cloning and sequencing of the variable regions of the M225 antibody
The presence of mouse antibody in the media from the M225 hybridoma cells at the point of harvesting the cells for RNA purification was proven using the mouse antibody ELISA. Following 1st strand synthesis the single stranded cDNA template was PCR-amplified with two series of degenerate primers, one series specific for the kappa light chain signal peptide/variable region genes (Table 4) and the second series specific for the heavy chain signal peptide/variable region genes (Table 5). Using these primers both the Vκgene and the VH gene of the M225 antibody were successfully PCR-cloned from the M225 hybridoma cell line.
The M225 kappa light chain variable region gene was PCR-cloned, as an approximately 416bp fragment, using primers MKV4 (which annealed to the 5' end of the DNA sequence of the kappa light chain signal peptide) and MKC (designed to anneal to the 5 'end of the mouse kappa constant region gene). Likewise the M225 heavy chain variable region gene was PCR-cloned, as an approximately 446bp fragment, using the MHV6 (which annealed to the 5' end of the DNA sequence of the heavy chain signal peptide) and MHCG1 (designed to anneal to the 5' end of the CH] domain of the mouse γl heavy chain gene) primers.
To minimize the possibility of introducing errors into the wild-type sequences of the mouse M225 variable region genes, either caused by AmpliTaq® DNA polymerase itself or changes introduced by reverse transcriptase (which has an error frequency approximately V-0 that of AmpliTaq®), a strict protocol was followed. At least two separate PCR-products, each from a different total RNA preparation and subsequent 1st strand cDNA synthesis reaction, were PCR-cloned and then completely DNA sequenced on both DNA strands for both the kappa light chain and heavy chain variable region genes of M225 mAb. From DNA sequence analysis of several individual clones from each of these PCR-reactions the mouse M225 antibody Vκ and VH genes were determined as shown in Figures 13 and 14, respectively. The amino acid sequences of the M225 Vκ and VH regions were compared with other mouse variable regions and also the consensus sequences of the subgroups that the variable regions were subdivided into in the Kabat database (20). From this analysis the M225 Vκ region was found to most closely match the consensus sequence of mouse kappa subgroup V, with an identity of 62.62% and a similarity of 76.64% to the subgroup. However, the kappa light chain variable region also displayed a close match to mouse kappa subgroup III with a 61.68% identity and a 76.64% similarity to its consensus sequence. When only the
FRs of the M225 kappa light chain variable region (i.e. without the amino acids in the CDRs) were compared to mouse subgroups III and V the identity increased to 66.25% for both subgroups while the similarity rose to 78.75% for subgroup III and to exactly 80.00% for subgroup V. Similar analysis of the M225 VH region found that it exhibited the closest match to the consensus sequence of mouse heavy chain subgroup IB in the Kabat database (20). Identity between the mouse heavy chain variable region amino acid sequence of M225 and the consensus sequence of subgroup IB was measured at 78.15% while the similarity was calculated to be 84.87%, with no other consensus sequence coming even remotely near these values. These results confirm that the mouse M225 variable regions appear to be typical of mouse variable regions.
Example IV- 14. Construction and expression of chimeric C225 antibody
The PCR-products from the two PCR-reactions prepared to construct the C225 Vκ and VH genes were separately subcloned into pUC19 as Hindϊll-BamHl fragments and then PCR-screened to identify putative positive transformants. Those transformants so identified were then ds-DNA sequenced, to confirm their synthesis, and then subcloned into their respective mammalian expression vectors. The DNA and amino acid sequences of the chimeric C225 kappa light chain and heavy chain variable regions are shown in Figures 15 and 16, respectively. Once the integrity of the expression vectors had also been confirmed, by PCR-screening and restriction analysis to confirm the presence of the correct insert, the vectors were co-transfected into COS cells. After 72 hr incubation, the medium was collected, spun to remove cell debri and analysed by ELISA for antibody production and binding to EGFR.
Unfortunately, no chimeric antibody could be detected in the supernatent of the COS cell co-transfections.
An analysis of the leader sequence of C225VK established that it was unusual, compared to the leader sequences of other kappa light chain variable regions in mouse kappa light chain subgroups III and V (20). To try and find a more suitable leader squence, the Kabat database was analysed to identify an individual kappa light chain which both matched C225 Vκ amino acid sequence and whose signal peptide sequence was known. This search identified the kappa light chain of mouse antibody L7'CL (28) which exhibited a 94.79% identity and a 94.79% similarity to the C225VK region and a perfect match with resepct to FRl , which play an important role in the excision of the signal peptide during secretion. The amino acid sequence of the L7'CL kappa light chain signal peptide (i.e. MVSTPQFLVFLLFWIPASRG (SEQ ID NO: 36)) displays all the characteristics thought important in a such a signal sequence - such as a hydrophobic core - and so it was decided to replace the signal peptide of the PCR- cloned 225VK with this new sequence. Another point of interest was that the differences between the M225VK and the L7'CL signal peptides nearly all occured at its 5 '-end where the MKV4 primer annealed (i.e. the first 33 bases which is eqiuvalent to the first 11 amino acids of the signal peptide) when the M225VK gene was originally PCR-cloned. Thus, these differences could well be primer induced errors in the DNA sequence of the signal peptide. PCR-mtuagenesis of the C225VK template produced an approximately 390bp product. The HmdIII-.P-.tI digested and purified fragment was then subcloned into identically digested and agarose gel-purified plasmid pUC-C225Vκ and transformed into XLlBlue competent cells. Putative positive transformants were identified and then ds-DNA sequenced. The C225Vκsp gene (Figure 17) was subcloned into pKNIOO and the resulting expression vector (pKN100-C225Vκsp) PCR-screened and restriction digested to confirm the presence of the correct insert. This vector was finally co-transfected into COS cells with pGlD105-C225VH and after 72 hr incubation, the medium was collected, spun to remove cell debri and analysed by ELISA for antibody production and binding to EGFR. This time chimeric C225 antibody was detected in the supernatent of the COS cell co-transfections at an approximate concentration of 150 ng/ml and this antibody bound to EGFR in the cell ELISA. Figure 18 shows a typical example of one such experiment.
Example IV-15. Construction and expression of the reshaped H225 antibody (225RKΛ/225RHΛ)
The construction of the first version of the reshaped human H225 kappa light chain variable region (225RKA) produced an approximately 416bp product that was then subcloned into pUC19 as a Hindll-BamHl fragment. Putative positive transformants were identified using the PCR-screening assay and then ds-DNA sequenced. The 225RKA gene (Figure 19) was subcloned into pKNIOO and the resulting expression vector (pKN100-225RKA) PCR-screened and restriction digested to confirm the presence of the correct insert. Likewise, the construction of the first version of the reshaped human H225 heavy chain variable region (225RHA) produced an approximately 446bp product which was then subcloned into pUC 19 as a Hindll- BamHl fragment. Putative positive transformants were again identified in the PCR- screen and then ds-DNA sequenced. The 225RHA gene (Figure 20) was subcloned into pGlD105 and the resulting expression vector (pGlD105-225RHA) PCR-screened and restriction digested to confirm the presence of the correct insert.
These vectors were then co-transfected together into COS cells and after 72 hr incubation, the medium was collected, spun to remove cell debri and analysed by ELISA for antibody production and binding to EGFR. The concentration of reshaped human antibody in the COS cell supernatents was slightly higher than those following transient expression of the C225 chimeric antibody (approximately 200 ng/ml). In addition, a significant level of binding to EGFR was shown in the cell ELISA. Figure 8 shows a typical example of one such experiment which appears to show that the reshaped human H225 antibody (225RKA/225RHA) bound to EGFR expressed on the surface of A431 cells with about 65% of the relative affinity of the chimeric C225 antibody.
The amino acid sequences of the two versions of the kappa light chain reshaped human H225 variable regions constructed are shown in Figure 21, while the amino acid sequences of the five versions of the heavy chain reshaped human H225 variable regions constructed are shown in Figure 22.
Example IV- 16. References
1. Mendelsohn, J. (1988). In: Waldmann, H. (ed). Monoclonal antibody therapy. Prog. Allergy Karger, Basel, pl47.
2. Aboud-Pirak, E., Hurwitz, E., Pirak, M.E., Bellot, F., Schlessinger, J., and Sela, M. ( 1988). J. Natl. Cancer Inst.80: 1605.
3. Masui, H., Kawamoto, T., Sato, J.D., Wolf, B., Sato, G., and Mendelsohn, J. (1984). Cancer Research 44:1002.
4. Mueller, B.M., Romerdahl, C.A., Trent, J.M., and Reisfeld, R.A. (1991). Cancer Research 51:2193. 5. Rodeck, U., Herlyn, M., Herlyn, D., Molthoff, C, Atkinson, B., Varello, M., Steplewski, Z., and Koprowski, H. (1987). Cancer Research 47:3692.
6. Chomczynski, P., and Sacchi, N. (1987). Anal. Biochem. 162:156.
7. Jones, S ., and Bendig, M.M. (1991). Bio/Technology 9:88.
8. Gussow, D., and Clackson, T. (1989). Nucleic Acids Res.17:4000. 9. Redston, M.S., and Kern, S.E. (1994). Biotechniques 17:286.
10. Kozak, M. (1987). J. Mol Biol. 196:947.
11. Kettleborough, C.A., Saldanha, J., Heath, V.J., Morrison, C.J., and Bendig, M.M. (1991). Protein Eng. 4:773.
12. Bernstein, F.C., Koetzle, T.F., Willliams, G.J., Meyer, E.F., Brice, M.D., Todgers, J.R., Kennard, O., Shimanouchi, T. and Tasumi, M. (1977). J.Mol Biol.
112:535.
13. Chothia, C, and Lesk, A.M. (1987). J. Mol Biol. 196:901.
14. Chothia, C, Lesk, A.M., Tramontano, A., Levitt, M., Smith-Gill, S.J., Air, G., Sheriff, S., Padlan, E.A., Davies, A., Tulip, W.R., Colman, P.M., Spinelli, S., Alzari, P.M., and Poljak, R.J. (1989). Nature 34:877.
15. Tramontano, A., Chothia, C, and Lesk, A.M. (1990). J. Mol Biol. 215:175.
16. Chothia, C, Lesk, A.M., Gherardi, E., Tomlinson, I.M., Walter, G., Marks, J.D., Llewelyn, M.B., and Winter, G. (1992). J. Mol. Biol 227:799. 17. Brooks, B.R., Bruccoleri, R.E., Olafson, B.D., States, D.J., Swaminathan, S., and Karplus, M. (1983). J. Comp. Chem. 4:187.
18. Padlan, E.A., Silverton, E.W., Sheriff, S., Cohen, G.H., Smith-Gill, S.J., and Davies, D.R. (1989). Proc. Nat. Acad. Sci. USA 86:5938. 19. Fischmann, T.O., Bentley, G.A., Bhat, T.N., Boulot, G., Mariuzza, R.A., Phillips, S.E.V., Tello, D. and Poljak, R.J. (1991) J.Biol.Chem. 266:12915. 20. Kabat, E.A., Wu, T.T., Perry, H.M., Gottesman, K.S., and Foeller, C. (1991). Sequences of proteins ofimmunological interest, Fifth Edition, U.S. Department of Health and Human Services, U.S. Government Printing Office. 21. Schulze-Gahmen, U., Rini, J.M., and Wilson, A. (1993). J.MolBiol 234:1098.
22. Silberstein, L.E., Litwin, S., and Carmack, C.E. (1989). J. Exp. Med. 169: 1631.
23. Schroeder Jr., H.W., Hillson, J.L., and Perlmutter, R.M. (1987). Science 238:791.
24. Riechmann, L., Clark, M., Waldmann, H., and Winter, G. (1988). Nature 322:21.
25. Chothia, C, Novotny, J., Bruccoleri, R., and Karplus, M. (1985). J. Mol Biol. 186:651.
26. Sato, K., Tsuchiya, M., Saldanha, J., Koishihara, Y., Ohsugi, Y., Kishimoto, T., and Bendig, M.M. (1993). Cancer Research 53:851.
27. Kolbinger, F., Saldanha, J., Hardman, N., and Bendig, M.M. (1993). Protein Eng. 6:971. 28. Pech, M., Hochtl, J., Schnell, H., and Zachau, H.G. (1981). Nature 291:668.
Example IV- 17. Tables
Table 4. Degenerate and specific PCR-primers used in the cloning of the M225 kappa light chain variable region genes.
Table 5. Degenerate and specific PCR-primers used in the cloning of the M225 heavy chain variable region genes.
Table 6. Primers for PCR screening transformed colonies
Table 7. Primers for constructing chimeric C225 antibody kappa light chain and heavy chain variable region genes and also for modifying the signal peptide sequence of the C225 antibody kappa light chain.
Table 8. Alignment of amino acid sequences leading to the design of the first version of the reshaped human H225 antibody kappa light chain variable region (225RKA).
Table 9. Alignment of .unino acid sequences leading to the design of the first version of the reshaped human H225 antibody heavy chain variable region (225RHA).
Table 10. Primers for constructing reshaped human antibody H225 kappa light chain variable region gene 225RKA.
Table 11. Primers for constructing reshaped human antibody H225 kappa light chain variable region gene 225RKB.
Table 12. Primers for constructing reshaped human antibody H225 heavy chain variable region gene 225RHA. Table 13. Primers for constructing reshaped human antibody H225 heavy chain variable region genes 225RHB, 225RHC, 225RHD and 225RHE.
Table 4. Degenerate and specific PCR-primers used in the cloning of the M225 kappa light chain variable region genes.
Name Sequence (5' - 3')
MKV1« (30mer) ATGAAGTTGCCTGTTAGGCTGTTGGTGCTG MKV2 (30mer) ATGGAGACAGACACACTCCTGCTATGGGTG T T
MKV3 (30mer) ATGAGTGTGCTCACTCAGGTCCTGGCGTTG
G
MKV4 (33mer) ATGAGGGCCCCTGCTCAGTTTTTTGGCTTCTTG A A C AA
MKV5 (30mer) ATGGATTTTCAGGTGCAGATTATCAGCTTC A T
MKV6 (27mer) ATGAGGTGCCCTGTTCAGTTCCTGGGG T TT C G C T A
MKV7 (31mer) ATGGGCATCAAGATGGAGTCACAGACCCAGG
MKV8 (31mer) ATGTGGGGACCTTTTTTCCCTTTTTCAATTG T G C AA
MKV9 (25mer) ATGGTATCCACACCTCAGTTCCTTG G T G
MKV10 (27mer) ATGTATATATGTTTGTTGTCTATTTCT MKVl l (28mer) ATGGAAGCCCCAGCTCAGCTTCTCTTCC MKC6 (20mer) ACTGGATGGTGGGAAGATGG
a MKV indicates primers that hybridize to leader sequences of mouse kappa light chain variable region genes. b MKC indicates the primer that hybridizes to the mouse kappa constant region gene. Table 5. Degenerate and specific PCR-primers used in the cloning of the M225 heavy chain variable region genes.
Name Sequence (5' - 3')
MHVla (27mer) ATGAAATGCAGCTGGGGCATCTTCTTC
G MHV2 (26mer) ATGGGATGGAGCTGTATCATGTTCTT
A CC
MHV3 (27mer) ATGAAGTTGTGGTTAAACTGGGTTTTT A
MHV4 (25mer) ATGAACTTTGGGCTCAGCTTGATTT G T G
MHV5 (30mer) ATGGACTCCAGGCTCAATTTAGTTTTCCTT MHV6 (27mer) ATGGCTGTCCTAGGGCTACTCTTCTGC T G C G MHV7 (26mer) ATGGGATGGAGCGGGATCTTTCTCTT A T G A
MHV8 (23mer) ATGAGAGTGCTGATTCTTTTGTG MHV9 (30mer) ATGGATTGGGTGTGGACCTTGCTATTCCTG C A
MHV10(27mer) ATGGGCAGACTTACATTCTCATTCCTG MHVll(28mer) ATGGATTTTGGGCTGATTTTTTTTATTG MHV12 (27mer) ATGATGGTGTTAAGTCTTCTGTACCTG MHCGl*(21mer) CAGTGGATAGACAGATGGGGG
a MHV indicates primers that hybridize to leader sequences of mouse heavy chain variable region genes. b MHCG indicates primers that hybridize to mouse constant region genes. Table 6. Primers for PCR screening transformed colonies
Name Sequence (5' - 3') pCR ™I Forward Primer (18mer) C TAGATGCATGC TC GAGC pCR™H Reverse Primer(21mer) TAC CGAGC T CGGAT C CAC T
AG RSP (Reverse Sequencing Primer) AGCGGATAACAAT T T CACA (24mer) C A G G A
UP (Universal Primer) (24mer) CGCCAGGGT T T TCCCAGTC
AC GAC γAS (20mer) ACGACAC CGT CAC CGGT T C
G
HCMVi (28mer) GT CAC CGT C C T TGACAC GC
GT C T CGGGA New.Hu (25mer) GTTGTTTGCGC ATAATCAC
AGGGC A
Huγl(17mer) T T GGAGGAGGGT GC CAG
225RKB.K49Y (60mer) CAGCAAAGAC C TGGCCAGG
CTCCAAGGCTTCTCATATA TTATGCTTCTGAGTCTATC TCT
Table 7. Primers for constructing chimeric C225 antibody kappa light chain and heavy chain variable region genes and also for modifying the signal peptide sequence of the C225 antibody kappa light chain.
Name Sequence (5' - 3')
C225VH5' (36mer) AAGCTTGCCGCCACCATGG
CTGTCTTGGGGCTGCTC
C225VH3' (34mer) GGATCCACTCACCTGCAGA
GACAGTGACCAGAGT
C225VK5' (36mer) AAGCTTGCCGCCACCATGG
TATCCACACCTCAGAAC
C225VK3' (40mer) TCTAGAAGGATCCACTCAC
GTTTCAGCTCCAGCTTGGT CC
C225Vκ5'sp (99mer) AAGCTTGCCGCCACCATGG
TATCCACACCTCAGTTCCT TGTATTTTTGCTTTTCTGG ATTCCAGCCTCCAGAGGTG ACATCTTGCTGACTCAGTC T C C A C225Vκ3'sp (21mer) AGAGATAGACTCAGAAGCA
T A
Table 8. Alignment of amino acid sequences leading to the design of the first version of the reshaped human H225 antibody kappa light chain variable region (225RKA).
Table 8 - Continued
68
2 Table 8 - Continued
Table 8 - Continued
Table 8 - Continued
Table 8 - Continued
Legend: (*) invariant residues as defined either by the Kabat consensus sequences i.e. 95% or greater occurrence within Kabat subgroup (Kabat et al, 1991) (in the case of columns 5 and 6) or as part of the canonical structure for the CDR loops (in the case of column 8) as defined by Chothia et al, (1989); (BOLD) positions in Frs and CDRs where the human amino acid residue was replaced by the corresponding mouse residue (UNDERLINE) positions in Frs where the human residue differs from the analogous mouse residue number; (δ) numbering of changes in the human Frs; (mouse C225) amino* acid sequence of the VL region from chimeric C225 antibody; (mouse -V) consensus sequence of mouse kappa VL regions from subgroup V (Kabat etal., 1991); (human -HI) consensus sequence of human VL regions from subgroup IH (Kabat etal., 1991); (Human Donor LS7'CL) amino acid sequence from human LS7'CL antibody (Silberstein, L.E. et al., 1989); (Surface or Buried) position of amino acid in relation to the rest of the residues in both chains of the antibody variable regions; (225RKA) amino acid sequence of the first version of the reshaped human mAb H225 Vκ region; (Core packing AA/Packing AA) amino acids located at the VL/VH interface as defined by Chothia et al. (1985); (Canonical AA) amino acids defined by Chothia and Lesk (1987), Chothia etal. (1989), Tramontano et al. (1990) and Chothia et al. (1992) as being important for CDR loop conformation.
Table 9. Alignment of amino acid sequences leading to the design of the first version of the reshaped human H225 antibody kappa light chain variable region (225RK .
Table 9 - Continued
Table 9 - Continued
Table 9 - Continued
76
gyBSπTUTE SHEET (RULE 26) Table 9 - Continued
Table 9 - Continued
Legend: (*) invariant residues as defined either by the Kabat consensus sequences i.e. 95% or greater occurrence within Kabat subgroup (Kabat et al, 19 1) (in the case of columns 5 and 6) or as part of the canonical structure for the CDR loops (in the case of column 8) as defined by Chothia et al, (1989); (BOLD) positions in Frs and CDRs where the human amino acid residue was replaced by the corresponding mouse residue (UNDERLINE) positions in Frs where the human residue differs from the analogous mouse residue number; (δ) numbering of changes in the human Frs; (mouse C225) amino acid sequence of the VH region from chimeric C225 antibody; (mouse IB) consensus sequence of mouse VH regions from subgroup IB (Kabat et al, 1991); (human III) consensus sequence of human VH regions from subgroup III (Kabat et al, 1991); (Human Donors: 38P1) amino acid sequence from human antibody 38P1'CL (Schroeder Jr et al 1987); (Surface or Buried) position of amino acid in relation to the rest of the residues in both chains of the antibody variable regions; (225KΑp) amino acid sequence of the first version of the reshaped human mAb H225 VH region. (Core packing of the first version of the reshaped human mAb H225 VH region (Core packing AA Packing AA) amino acids located at the VL/VH interface as defined by Chothia et al. (1985); (Canonical AA) amino acids defined by Chothia and Lesk (1987), Chothia et al. (1989), Tramontano et al. (1990) and Chothia et al. (1992) as being important for CDR loop conformation.
Table 10. Primers for constructing reshaped human antibody H225 kappa light chain variable region gene 225RKA.
Name Sequence (5' - 3')
225RKA.LEAD (88mer) C TGGAGAC TGAGT CA G TACG
AT T TCAC T TC TGGAG G CTCG AATCCAGAAAAGCAA A AATA C TTGGT TC TGAGGTG T GGA T ACCATGGT
225RKA.FR1 (80mer) TCGTAC TGAC TCAGT C TCCA
GCCACCC TGTC T T TG A GTCC AGGAGAAAGAGCCAC C CTCT CC TGCAGGGCCAGTC A GAG T
225RKA.FR2a (74mer) GAGATAGAC TCAGAA G CATA
C T T TATGAGAAGCC T T GGAG C CTGGCCAGGTC T TT G CTGA TACCAGTGTATGT T
225RKA.FR3 (71mer) GGC T TC TCATAAAGT A TGCT
TCTGAGTC TATC TC T G GAA T CCCTGCCAGGTTTAG T GGC A GTGGATCAGGG
225RKA.FR3a (77mer) T T TTGT TGACAGTAA T A AA C
TGCAAAATC T TCAGG C TCCA CAC TGCTGATGGTAA G AGTA AAATC TGTCCC TGAT C C
225RKA.CDR3 (33mer) GAT T T TGCAGT T TAT T ACTG
T C AAC A A AAT A AT
225RKA.FR4a (68mer) TCTAGAAGGATCCAC T CACG
T TTCAGC TCCACC T T G GTCC CTCCACCGAACGTGG T TGGC C AGT T AT T
225RKA. V78L (42mer) AC TC T TACCATCAGC A GTC T
GGAGCC TGAAGAT T T T GCAG T T
225RKA.L108I (57mer) TCTAGAAGGATCCAC T CACG
T TTGATC TCCACC T T G GTCC C TCCACCGAACGTGG T T
78/1 Table 10. continued...
225RKA.LS7 leader AAGCTTGCCGCCACCATGGA
(99mer) AGCCCCAGCTCAGCTTCTCT
TCCTCTTGCTTCTCTGGCTC CCAGATACCACCGGAGAAAT CGTACTGACTCAGTCTCCA
79 Table 11. Primers for constructing reshaped human antibody H225 kappa light chain variable region gene 225RKB.
Name Sequence (5' - 3')
225RKB.K49Y (60mer) CAGCAAAGACC TGGCCAGGC
TCCAAGGCTTCTCATATATT ATGCTTCTGAGTCTATCTCT
APCR40(25mer) CTGAGAGTGCACCATATGCG
GTGTG
80 Table 12. Primers for constructing reshaped human antibody H225 heavy chain variable region gene 225RHA.
Name Sequence (5' - 3')
225RHA.FR1 (37mer) GGTGCAGCTGGTCGAGTC TG
GGGGAGGCTTGGTACAG
225RHA.FRla (50mer) GGC TGTACCAAGCC TCCCCC
AGACTCGACCAGCTGCACCT C AC ACTGGAC
225RHA.CDRla (64mer) CCCAGTGTACACCATAGT TA
GTTAATGAGAATCCGGAGAC TGCACAGGAGAGTCTCAGGG AC CC
225RHA.FR2 (63mer) TTAACTAACTATGGTGTACA
CTGGGTTCGCC AGGC T AC AG GAAAGGGTCTGGAGTGGCTG GGA
225RHA.FR3a (74mer) CTGTTCAT T TGCAGATACAG
GGAGTTCTTGGCATTTTCCT TGGAGATGGTCAGTCGACTT GTGAAAGGTGTATT
225RHA.FR3 (73mer) C TCCC TGTATCTGCAAATGA
ACAGTCTCAGAGCCGGGGAC ACAGCCGTGTATTACTGTGC CAGAGCCCTCACC
225RHA.FR4a (65mer) GGATCCAC TCACC TGAAGAG
ACAGTGACCATAGTCCCTTG GCCCCAGTAAGCAAA
81 Table 13. Primers for constructing reshaped human antibody H225 heavy chain variable region genes 225RHB, 225RHC, 225RHD and 225RHE.
Name Sequence (5' - 3')
225RHB.T41P-S (35mer) G G G T T C G C C AGGCTCCAGGA
AAGGGTCTGGAGTGG
225RHB.T41P-AS (30mer) T C C T G G A G C C T G G C G A A C C C
AGTGTACACC
225RHC.T68S/S70N (46mer) CACAAGTCGACTGAGCATCA
ACAAGGAAAATGCCAAGAAC T CCC TG
225RHE.L78V (72mer) CACAAGTCGACTGACCATCT
TCAAGGAAAATGCCAAGAAC TCCGTTTATCTGCAAATGAA CAGTCTCAGAGC
APCR10(25mer) TACGCAAACCGCCTCTCCCC
GCGCG
APCR40 (25mer) CTGAGAGTGCACCATATGCG
GTGTG
RSP (Reverse Sequencing Primer) AGCGGATAACAATTTCACAC (24mer) A G G A
UP (Universal Primer) (24mer) CGCCAGGGTTTTCCCAGTCA
CGAC
82

Claims

WHAT WE CLAIM IS:
1. A polypeptide lacking the constant region and the variable light chain of an antibody, the polypeptide comprising the amino acid sequence N Y G V H, G V I W S G G N T D Y N T P F T S R, or V I W S G G N T D Y N T P F T S.
2. A polypeptide according to Claim 1, comprising amino acid sequences N Y G V H and G V I W S G G N T D Y N T P F T S R or V I W S G G N T D Y N T P F T S.
3. A polypeptide consisting of the amino acid sequence N Y G V H or G V I W S G G N T D Y N T P F T S R.
4. A polypeptide consisting of the amino acid sequence N Y G V H or V I W S G G N T D Y N T P F T S.
5. A polypeptide according to Claim 1 conjugated to an effector molecule.
6. A polypeptide according to Claim 5 wherein the effector molecule inhibits tumor growth.
7. A polypeptide according to Claim 5 wherein the effector molecule is cytotoxic.
8. A polypeptide according to Claim 5 wherein the effector molecule is doxorubicin.
9. A polypeptide according to Claim 5 wherein the effector molecule is cisplatin.
10. A polypeptide according to Claim 5 wherein the effector molecule is taxol.
11. A polypeptide according to Claim 5 wherein the effector molecule is a signal transduction inhibitor.
12. A polypeptide according to Claim 5 wherein the effector molecule is a ras inhibitor.
13. A polypeptide according to Claim 5 wherein the effector molecule is a cell cycle inhibitor.
14. DNA encoding a polypeptide lacking the constant region and the variable light chain of an antibody, the polypeptide comprising the amino acid sequence N Y G V H, G V I W S G G N T D Y N T P F T S R or V I W S G G N T D Y N T P F T S.
15. DNA encoding the polypeptide of Claim 14 comprising amino acid sequences N Y G V H and G V I W S G G N T D Y N T P F T S R or V I W S G G N T D Y N T P F T S.
16. DNA encoding a polypeptide according to Claim 14 conjugated to an effector molecule.
17. DNA encoding a polypeptide according to Claim 16 wherein the effector molecule inhibits tumor growth.
18. A molecule having the constant region of a human antibody and the hypervariable region of monoclonal antibody 225 conjugated to an effector molecule.
19. A molecule according to Claim 18 wherein the effector molecule is a cytotoxic agent.
20. A molecule according to Claim 19 wherein the cytotoxic agent is doxorubicin.
21. A molecule according to Claim 19 wherein the cytotoxic agent is taxol.
22. A molecule according to Claim 19 wherein the cytotoxic agent is cisplatin.
23. A molecule comprising: a constant region of a human antibody; a variable region other than the CDRs of a human antibody, the variable region comprising a kappa light chain and a heavy chain, and the CDRs of monoclonal antibody 225.
24. A molecule according to claim 23 wherein the constant region has an amino acid sequence of an IgG.
25. A molecule according to claim 24 wherein the IgG is IgG1.
26. A molecule according to claim 23 that is reshaped according to Example IV.
27. A molecule according to claim 23, wherein the heavy chain has at least one amino acid, according to the Kabat numbering system, at an amino acid position selected from the group consisting of 24, 28, 29, 30, 41, 48, 49, 67, 68, 70, 71 and 78, substituted with a murine amino acid selected from the corresponding Kabat amino acid position.
28. A molecule according to claim 23, wherein the kappa light chain has an amino acid, according to the Kabat numbering system, at position 49 substituted with a murine amino acid selected from the corresponding Kabat amino acid position.
29. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence selected from 225RKA or 225RKB.
30. A molecule according to claim 23 wherein the heavy chain variable region has an amino acid sequence selected from 225RHA, 225RHB, 225RHC, 225RHD, or 225RHE.
31. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHA.
32. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHB.
33. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHC.
34. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHD.
35. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHE.
36. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHA.
37. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHB.
38. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHC.
39. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHD.
40. A molecule according to claim 23 wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHE.
41. A molecule according to Claim 23, wherein the molecule is attached to an effector molecule.
42. A molecule according to Claim 39, wherein the effector molecule is a cytotoxic agent.
43. A molecule according to Claim 40, wherein the cytotoxic agent is doxorubicin.
44. A molecule according to Claim 40, wherein the cytotoxic agent is taxol.
45. A molecule according to Claim 40, wherein the cytotoxic agent is cisplatin
46. A method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of the polypeptide according to Claim 1.
47. A method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of the polypeptide according to Claim 3 or Claim 4.
48. A method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of a molecule having the constant region of a human antibody and the variable region of monoclonal antibody
225.
49. A method for significantly inhibiting the growth of tumor cells in a human comprising administering to the human an effective amount of a molecule having a constant region of a human antibody; a variable region other than the CDRs of a human antibody, the variable region comprising a kappa light chain and heavy chain, and the CDRs of monoclonal antibody 225.
50. A method according to claim 47, wherein the kappa light chain variable region has an amino acid sequence selected from 225RKA or 225RKB.
51. A method according to claim 47, wherein the heavy chain variable region has an amino acid sequence selected from 225RHA, 225RHB, 225RHC, 225RHD, or 225RHE.
52. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHA.
53. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHB.
54. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHC.
55. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHD.
56. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKA and the heavy chain variable region has amino acid sequence 225RHE.
57. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHA.
58. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHB.
59. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHC.
60. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHD.
61. A method according to claim 47, wherein the kappa light chain variable region has amino acid sequence 225RKB and the heavy chain variable region has amino acid sequence 225RHE.
62. A method according to any of Claims 44-47, further comprising administering a cytotoxic agent.
63. A molecule according to Claim 60, wherein the cytotoxic agent is doxorubicin.
64. A molecule according to Claim 60, wherein the cytotoxic agent is taxol.
65. A molecule according to Claim 60, wherein the cytotoxic agent is cisplatin
66. The method according to Claim 44 or Claim 45, wherein the polypeptide is conjugated to an effector molecule.
67. The method according to Claim 46 or Claim 47, wherein the molecule is conjugated to an effector molecule.
68. The method according to Claim 64, wherein the effector molecule is cytotoxic.
69. The method according to Claim 64, wherein the effector molecule is doxorubicin.
70. The method according to Claim 64, wherein the effector molecule is cisplatin.
71. The method according to Claim 64, wherein the effector molecule is taxol.
72. The method according to Claim 64, wherein the effector molecule is a signal transduction inhibitor.
73. The method according to Claim 64, wherein the effector molecule is a ras inhibitor.
74. The method according to Claim 64, wherein the effector molecule is a cell cycle inhibitor.
75. The method according to Claim 65, wherein the effector molecule is cytotoxic.
76. The method according to Claim 65, wherein the effector molecule is doxorubicin.
77. The method according to Claim 65, wherein the effector molecule is cisplatin.
78. The method according to Claim 65, wherein the effector molecule is taxol.
79. The method according to Claim 65, wherein the effector molecule is a signal transduction inhibitor.
80. The method according to Claim 65, wherein the effector molecule is a ras inhibitor.
81. The method according to Claim 65, wherein the effector molecule is a cell cycle inhibitor.
82. The method according to any of Claims 44-47, wherein the tumor cells are prostatic tumor cells.
83. The method according to Claim 80, wherein the prostatic tumor cells are late stage prostatic tumor cells.
84. A nucleic acid molecule that encodes a molecule comprising: a constant region of a human antibody; a variable region other than the CDRs of a human antibody, the variable region comprising a kappa light chain and a heavy chain, and the CDRs of monoclonal antibody 225.
85. A vector comprising the nucleic acid molecule claim of 84.
86. A vector according to claim 85, wherein the vector is an expressible vector.
87. A vector according to claim 86, wherein the vector is expressible in a prokaryotic cell.
88. A vector according to claim 86, wherein the vector is expressible in a eukaryotic cell.
89. A prokaryotic cell comprising the expressible vector of claim 87.
90. An eukaryotic cell comprising the expressible vector of claim 88.
91. A pharmaceutical composition, comprising the molecule of claim 23 and a pharmaceutically acceptable carrier.
EP96921457A 1995-06-07 1996-06-07 Antibody and antibody fragments for inhibiting the growth of tumors Withdrawn EP0831880A4 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US48298295A 1995-06-07 1995-06-07
US482982 1995-06-07
US57328995A 1995-12-15 1995-12-15
US573289 1995-12-15
PCT/US1996/009847 WO1996040210A1 (en) 1995-06-07 1996-06-07 Antibody and antibody fragments for inhibiting the growth of tumors

Publications (2)

Publication Number Publication Date
EP0831880A1 true EP0831880A1 (en) 1998-04-01
EP0831880A4 EP0831880A4 (en) 2004-12-01

Family

ID=27047489

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96921457A Withdrawn EP0831880A4 (en) 1995-06-07 1996-06-07 Antibody and antibody fragments for inhibiting the growth of tumors

Country Status (5)

Country Link
EP (1) EP0831880A4 (en)
JP (3) JPH11507535A (en)
AU (1) AU6267896A (en)
CA (1) CA2222231A1 (en)
WO (1) WO1996040210A1 (en)

Families Citing this family (427)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6811779B2 (en) * 1994-02-10 2004-11-02 Imclone Systems Incorporated Methods for reducing tumor growth with VEGF receptor antibody combined with radiation and chemotherapy
US20030108545A1 (en) * 1994-02-10 2003-06-12 Patricia Rockwell Combination methods of inhibiting tumor growth with a vascular endothelial growth factor receptor antagonist
US7122636B1 (en) 1997-02-21 2006-10-17 Genentech, Inc. Antibody fragment-polymer conjugates and uses of same
NZ337413A (en) * 1997-03-10 2003-02-28 Univ California Antibodies that bind to Prostate Stem Cell Antigen (PSCA) to treat prostate cancer.
US20020173629A1 (en) 1997-05-05 2002-11-21 Aya Jakobovits Human monoclonal antibodies to epidermal growth factor receptor
US6235883B1 (en) * 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998051702A1 (en) * 1997-05-14 1998-11-19 Sloan-Kettering Institute For Cancer Research Methods and compositions for destruction of selected proteins
US6417168B1 (en) 1998-03-04 2002-07-09 The Trustees Of The University Of Pennsylvania Compositions and methods of treating tumors
US6706721B1 (en) 1998-04-29 2004-03-16 Osi Pharmaceuticals, Inc. N-(3-ethynylphenylamino)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine mesylate anhydrate and monohydrate
MXPA00011248A (en) * 1998-05-15 2004-09-06 Imclone Systems Inc Treatment of human tumors with radiation and inhibitors of growth factor receptor tyrosine kinases.
AU768027B2 (en) * 1999-02-24 2003-11-27 Uab Research Foundation, The Taxane derivatives for targeted therapy of cancer
BR0010524A (en) * 1999-05-14 2002-05-28 Imclone Systems Inc Treatment of refractory human tumors with epidermal growth factor receptor antagonists
EP1189641B1 (en) 1999-06-25 2009-07-29 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
ES2330301T3 (en) 1999-08-27 2009-12-09 Genentech, Inc. DOSAGES FOR TREATMENT WITH ANTI-ERBB2 ANTIBODIES.
US7087613B2 (en) 1999-11-11 2006-08-08 Osi Pharmaceuticals, Inc. Treating abnormal cell growth with a stable polymorph of N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine hydrochloride
UA74803C2 (en) 1999-11-11 2006-02-15 Осі Фармасьютікалз, Інк. A stable polymorph of n-(3-ethynylphenyl)-6,7-bis(2-methoxyetoxy)-4-quinazolinamine hydrochloride, a method for producing thereof (variants) and pharmaceutical use
ES2528794T3 (en) 2000-04-11 2015-02-12 Genentech, Inc. Multivalent antibodies and uses thereof
CN102698265A (en) 2000-05-19 2012-10-03 杰南技术公司 Gene detection assay for improving the likelihood of an effective response to an erbb antagonist cancer therapy
JP2004527456A (en) * 2000-08-09 2004-09-09 イムクローン システムズ インコーポレイティド Treatment of hyperproliferative diseases with EGF receptor antagonists
RU2297245C2 (en) * 2001-02-19 2007-04-20 Мерк Патент Гмбх Modified anti-egfr antibodies with reduced ammonogenicity
JP2004528851A (en) * 2001-05-07 2004-09-24 ハイブリドン・インコーポレイテッド Epidermal growth factor receptor antisense oligonucleotides
ES2552281T3 (en) 2001-05-11 2015-11-26 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
US20110313230A1 (en) 2001-05-11 2011-12-22 Terrance Grant Johns Specific binding proteins and uses thereof
US20100056762A1 (en) 2001-05-11 2010-03-04 Old Lloyd J Specific binding proteins and uses thereof
DE10133394A1 (en) * 2001-07-13 2003-01-30 Merck Patent Gmbh Liquid formulation containing cetuximab
JP4606739B2 (en) * 2002-01-18 2011-01-05 ピエール、ファーブル、メディカマン Novel anti-IGF-IR antibodies and uses thereof
US7241444B2 (en) 2002-01-18 2007-07-10 Pierre Fabre Medicament Anti-IGF-IR antibodies and uses thereof
AU2003210802B2 (en) 2002-02-05 2009-09-10 Genentech Inc. Protein purification
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
JP2005535572A (en) * 2002-04-12 2005-11-24 メディミューン,インコーポレーテッド Recombinant anti-interleukin-9 antibody
CA2478925C (en) 2002-04-26 2016-06-07 Robert Lee Fahrner Non-affinity purification of proteins
US7696320B2 (en) 2004-08-24 2010-04-13 Domantis Limited Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
PT1585966E (en) 2002-07-15 2012-02-20 Hoffmann La Roche Treatment of cancer with the anti-erbb2 antibody rhumab 2c4
BRPI0314814C1 (en) 2002-09-27 2021-07-27 Xencor Inc antibody comprising an fc variant
JP2006524036A (en) * 2002-11-08 2006-10-26 アブリンクス エン.ヴェー. Single domain antibodies targeting tumor necrosis factor alpha and uses thereof
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US20060034845A1 (en) 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
EP1583562B1 (en) 2003-01-06 2011-06-15 Angiochem Inc. Angiopep-1, related compounds, and uses thereof
ES2574993T3 (en) 2003-01-22 2016-06-23 Roche Glycart Ag Fusion constructs and use thereof to produce antibodies with higher Fc receptor binding affinity and effector function
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
WO2004093807A2 (en) 2003-04-22 2004-11-04 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Somatostatin vectors
ES2786568T3 (en) 2003-07-28 2020-10-13 Genentech Inc Reduction of protein A leaching during protein A affinity chromatography
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
KR101520209B1 (en) 2003-11-06 2015-05-13 시애틀 지네틱스, 인크. Monomethylvaline compounds capable of conjugation to ligands
DE10355904A1 (en) * 2003-11-29 2005-06-30 Merck Patent Gmbh Solid forms of anti-EGFR antibodies
EP1701979A2 (en) * 2003-12-03 2006-09-20 Xencor, Inc. Optimized antibodies that target the epidermal growth factor receptor
EP2221315A1 (en) 2003-12-04 2010-08-25 Xencor, Inc. Methods of generating variant proteins with increased host string content and compositions thereof
JP4969440B2 (en) 2004-04-08 2012-07-04 デビッド, ビー. エイガス, ErbB antagonist for the treatment of pain
NZ551180A (en) 2004-06-01 2009-10-30 Genentech Inc Antibody drug conjugates and methods
EP1919950A1 (en) 2004-07-15 2008-05-14 Xencor, Inc. Optimized fc variants
US7662926B2 (en) 2004-09-02 2010-02-16 Genentech, Inc. Anti-Fc-gamma receptor antibodies, bispecific variants and uses therefor
US7655229B2 (en) 2004-09-02 2010-02-02 Chan Andrew C Anti-FC-gamma RIIB receptor antibody and uses therefor
EP1791565B1 (en) 2004-09-23 2016-04-20 Genentech, Inc. Cysteine engineered antibodies and conjugates
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US7939267B2 (en) 2004-11-04 2011-05-10 Laboratory Corporation Of America Holdings Detection of activation of endothelial cells as surrogate marker for angiogenesis
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
CN102746404B (en) 2004-11-12 2016-01-20 赞科股份有限公司 To FcRn in conjunction with reformed Fc variant
EP1812064A4 (en) 2004-11-19 2009-07-08 Cornell Res Foundation Inc Use of vascular endothelial growth factor receptor 1+ cells in treating and monitoring cancer and in screening for chemotherapeutics
EP3698807A1 (en) 2005-01-21 2020-08-26 Genentech, Inc. Fixed dosing of her antibodies
US20080161251A1 (en) 2005-01-21 2008-07-03 Astex Therapeutics Limited Pharmaceutical Compounds
PT2360258E (en) 2005-02-18 2015-01-13 Angiochem Inc Aprotinin polypeptides for transporting a compound across the blood-brain barrier
AU2006216732C1 (en) 2005-02-23 2017-07-20 Genentech, Inc. Extending time to disease progression or survival in cancer patients using a HER dimerization inhibitor
AR053272A1 (en) 2005-05-11 2007-04-25 Hoffmann La Roche DETERMINATION OF RESPONSIVES TO CHEMOTHERAPY
DK2433653T3 (en) 2005-07-15 2019-08-19 Angiochem Inc Use of aprotinin polypeptides as carriers in pharmaceutical conjugates
ES2530265T3 (en) 2005-07-21 2015-02-27 Genmab A/S Binding potency assays of an antibody drug substance to an FC receptor
RU2515108C2 (en) 2005-08-19 2014-05-10 Эббви Инк Immunoglobulin with double variable domains and its applications
EP2500356A3 (en) 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP1931709B1 (en) 2005-10-03 2016-12-07 Xencor, Inc. Fc variants with optimized fc receptor binding properties
DE602006013029D1 (en) 2005-11-12 2010-04-29 Lilly Co Eli ANTI-EGFR ANTIBODY
CA2641310C (en) 2006-02-03 2013-08-20 Imclone Systems Incorporated Igf-ir antagonists as adjuvants for treatment of prostate cancer
AR060635A1 (en) 2006-04-27 2008-07-02 Banyu Pharma Co Ltd DERIVATIVES OF 1,2-DIHIDRO-3H-PIRAZOLO [3,4-D] PIRIMIDIN-3-ONA, PHARMACEUTICAL COMPOSITIONS THAT INCLUDE THEM AND THEIR USE IN THE TREATMENT OF CANCER
FR2902799B1 (en) 2006-06-27 2012-10-26 Millipore Corp METHOD AND UNIT FOR PREPARING A SAMPLE FOR THE MICROBIOLOGICAL ANALYSIS OF A LIQUID
JP2008081492A (en) 2006-08-31 2008-04-10 Banyu Pharmaceut Co Ltd New aminopyridine derivative having aurora a selective inhibitory action
CA2662236A1 (en) 2006-09-12 2008-03-20 Genentech, Inc. Methods and compositions for the diagnosis and treatment of cancer
US8916552B2 (en) 2006-10-12 2014-12-23 Astex Therapeutics Limited Pharmaceutical combinations
EP2073807A1 (en) 2006-10-12 2009-07-01 Astex Therapeutics Limited Pharmaceutical combinations
US7820159B2 (en) * 2006-10-31 2010-10-26 Instiute of Virology of the Slovak Academy of Sciences MN/CA IX and EGFR pathway inhibition
US8362217B2 (en) 2006-12-21 2013-01-29 Emd Millipore Corporation Purification of proteins
US8569464B2 (en) 2006-12-21 2013-10-29 Emd Millipore Corporation Purification of proteins
WO2008079280A1 (en) 2006-12-21 2008-07-03 Millipore Corporation Purification of proteins
US7977336B2 (en) 2006-12-28 2011-07-12 Banyu Pharmaceutical Co. Ltd Aminopyrimidine derivatives as PLK1 inhibitors
EP2123676A4 (en) * 2007-01-05 2011-01-05 Univ Tokyo Diagnosis and treatment of cancer by using anti-prg-3 antibody
ES2426158T3 (en) 2007-01-22 2013-10-21 Genentech, Inc. Precipitation with polyelectrolyte and antibody purification
US9090693B2 (en) 2007-01-25 2015-07-28 Dana-Farber Cancer Institute Use of anti-EGFR antibodies in treatment of EGFR mutant mediated disease
WO2008101177A2 (en) * 2007-02-16 2008-08-21 University Of Virginia Patent Foundation Ige antibodies to chimeric or humanized igg therapeutic monoclonal antibodies as a screening test for anaphylaxis
EP2132229B1 (en) 2007-03-01 2016-05-11 Symphogen A/S Recombinant anti-epidermal growth factor receptor antibody compositions
WO2008109440A2 (en) 2007-03-02 2008-09-12 Genentech, Inc. Predicting response to a her dimerisation inhibitor based on low her3 expression
MX2009009782A (en) 2007-03-15 2010-09-10 Ludwig Inst Cancer Res Treatment method using egfr antibodies and src inhibitors and related formulations.
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
WO2008154249A2 (en) 2007-06-08 2008-12-18 Genentech, Inc. Gene expression markers of tumor resistance to her2 inhibitor treatment
SI3597659T1 (en) 2007-07-09 2023-04-28 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
WO2009023265A1 (en) 2007-08-14 2009-02-19 Ludwig Institute For Cancer Research Monoclonal antibody 175 targeting the egf receptor and derivatives and uses thereof
EP2044949A1 (en) 2007-10-05 2009-04-08 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
AU2008315048A1 (en) 2007-10-23 2009-04-30 Msd K.K. Pyridone-substituted-dihydropyrazolopyrimidinone derivative
RS53850B2 (en) 2007-10-30 2018-07-31 Genentech Inc Antibody purification by cation exchange chromatography
MX2010005222A (en) 2007-11-12 2010-09-28 Bipar Sciences Inc Treatment of breast cancer with a parp inhibitor alone or in combination with anti-tumor agents.
SI2808343T1 (en) 2007-12-26 2019-10-30 Xencor Inc Fc variants with altered binding to FcRn
US8454960B2 (en) * 2008-01-03 2013-06-04 The Scripps Research Institute Multispecific antibody targeting and multivalency through modular recognition domains
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
SG190572A1 (en) 2008-04-29 2013-06-28 Abbott Lab Dual variable domain immunoglobulins and uses thereof
RU2010153578A (en) 2008-06-03 2012-07-20 Эбботт Лэборетриз (Us) IMMUNOGLOBULINS WITH DOUBLE VARIABLE DOMAINS AND THEIR APPLICATION
CA2726087A1 (en) 2008-06-03 2009-12-10 Tariq Ghayur Dual variable domain immunoglobulins and uses thereof
US8999702B2 (en) 2008-06-11 2015-04-07 Emd Millipore Corporation Stirred tank bioreactor
RU2011104348A (en) 2008-07-08 2012-08-20 Эбботт Лэборетриз (Us) IMMUNOGLOBULINS WITH DOUBLE VARIABLE DOMAIN AGAINST PROSTAGLANDINE E2 AND THEIR APPLICATION
HUE047316T2 (en) 2008-08-14 2020-04-28 Genentech Inc Methods for removing a contaminant using indigenous protein displacement ion exchange membrane chromatography
CN102137874B (en) 2008-08-29 2015-02-18 西福根有限公司 Recombinant anti-epidermal growth factor receptor antibody compositions
JP5705115B2 (en) 2008-09-10 2015-04-22 ジェネンテック, インコーポレイテッド Compositions and methods for prevention of oxidative degradation of proteins
KR20190025057A (en) 2008-10-14 2019-03-08 제넨테크, 인크. Immunoglobulin variants and uses thereof
CA2740316A1 (en) 2008-10-15 2010-04-22 Angiochem Inc. Conjugates of glp-1 agonists and uses thereof
BRPI0922689A2 (en) 2008-12-05 2018-11-06 Angiochem Inc. neurotensin conjugates or neurotensin analogs and uses thereof
CN105037535A (en) 2008-12-16 2015-11-11 Emd密理博公司 Stirred tank reactor and method
JP2012512244A (en) 2008-12-16 2012-05-31 イー・エム・デイー・ミリポア・コーポレイシヨン Protein purification
US8288396B2 (en) 2009-02-25 2012-10-16 Msdkk Pyrimidopyrimidoindazole derivative
US8435488B2 (en) 2009-02-27 2013-05-07 Genentech, Inc. Methods and compositions for protein labelling
EP2810652A3 (en) 2009-03-05 2015-03-11 AbbVie Inc. IL-17 binding proteins
EP2403339B1 (en) 2009-03-06 2017-01-18 Merck Sharp & Dohme Corp. Combination cancer therapy with an akt inhibitor and other anticancer agents
ES2572728T3 (en) 2009-03-20 2016-06-02 F. Hoffmann-La Roche Ag Bispecific anti-HER antibodies
MX2011010169A (en) * 2009-04-07 2011-10-11 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies.
RU2011146654A (en) 2009-04-20 2013-05-27 Ангиокем Инк. METHODS FOR TREATING OVARIAN CANCER USING CONJUGATED PRODUCT
CA2765287C (en) 2009-06-15 2018-12-11 Bayer Bioscience N.V. Nicotiana benthamiana plants deficient in xylosyltransferase activity
IN2012DN00248A (en) 2009-07-02 2015-05-01 Angiochem Inc
US9345661B2 (en) 2009-07-31 2016-05-24 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
HUE048980T2 (en) 2009-08-11 2020-08-28 Hoffmann La Roche Production of proteins in glutamine-free cell culture media
KR20120060877A (en) 2009-09-01 2012-06-12 아보트 러보러터리즈 Dual variable domain immunoglobulins and uses thereof
US9428548B2 (en) 2009-09-01 2016-08-30 Genentech, Inc. Enhanced protein purification through a modified protein A elution
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
ES2354922B1 (en) 2009-09-02 2012-02-07 Fundacion Institut De Recerca De L'hospital Universitari Vall D'hebron MARKERS FOR THE SELECTION OF PERSONALIZED THERAPIES FOR THE TREATMENT OF THE C�? NCER.
AR078161A1 (en) 2009-09-11 2011-10-19 Hoffmann La Roche VERY CONCENTRATED PHARMACEUTICAL FORMULATIONS OF AN ANTIBODY ANTI CD20. USE OF THE FORMULATION. TREATMENT METHOD
US20110076232A1 (en) 2009-09-29 2011-03-31 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
AU2010306677B2 (en) 2009-10-15 2013-05-23 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
EP2507381A4 (en) 2009-12-04 2016-07-20 Hoffmann La Roche Multispecific antibodies, antibody analogs, compositions, and methods
CN102712640A (en) 2010-01-12 2012-10-03 弗·哈夫曼-拉罗切有限公司 Tricyclic heterocyclic compounds, compositions and methods of use thereof
US20120309056A1 (en) 2010-02-04 2012-12-06 Leon Arnaud Fed-batch process using concentrated cell culture medium for the efficient production of biologics in eb66 cells
WO2011097527A2 (en) 2010-02-04 2011-08-11 Xencor, Inc. Immunoprotection of therapeutic moieties using enhanced fc regions
JP5981853B2 (en) 2010-02-18 2016-08-31 ジェネンテック, インコーポレイテッド Neuregulin antagonists and their use in the treatment of cancer
EP2536432B1 (en) 2010-02-19 2018-08-08 Cornell University Method to treat autoimmune demyelinating diseases and other autoimmune or inflammatory diseases
AU2011222012C1 (en) 2010-03-02 2015-02-26 Kyowa Kirin Co., Ltd. Modified antibody composition
EP2547338A2 (en) 2010-03-17 2013-01-23 F. Hoffmann-La Roche AG Imidazopyridine compounds, compositions and methods of use
PL2550018T3 (en) 2010-03-22 2019-08-30 F.Hoffmann-La Roche Ag Compositions and methods useful for stabilizing protein-containing formulations
WO2011118739A1 (en) 2010-03-26 2011-09-29 協和発酵キリン株式会社 Novel antibody having modification site introduced therein, and antibody fragment
WO2011120135A1 (en) 2010-03-29 2011-10-06 Zymeworks, Inc. Antibodies with enhanced or suppressed effector function
MX2012011887A (en) 2010-04-16 2012-11-30 Genentech Inc Fox03a as predictive biomarker for pi3k/akt kinase pathway inhibitor efficacy.
EP2566510A1 (en) 2010-05-03 2013-03-13 F. Hoffmann-La Roche AG Compositions and methods useful for reducing the viscosity of protein-containing formulations
SI2571532T1 (en) 2010-05-14 2017-10-30 Abbvie Inc. Il-1 binding proteins
EP2571903B1 (en) 2010-05-17 2019-09-04 EMD Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
JP6050226B2 (en) 2010-05-28 2016-12-21 ジェネンテック, インコーポレイテッド Lowering lactic acid levels and increasing polypeptide production by downregulating LDH and PDHK expression
CA2802756C (en) 2010-06-24 2021-05-04 Genentech, Inc. Compositions and methods for stabilizing protein-containing formulations
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
JP5953303B2 (en) 2010-07-29 2016-07-20 ゼンコア インコーポレイテッド Antibodies with modified isoelectric points
KR20130100118A (en) 2010-08-03 2013-09-09 아비에 인코포레이티드 Dual variable domain immunoglobulins and uses therof
US9051370B2 (en) * 2010-08-10 2015-06-09 Glycotope Gmbh Humanized EGFR antibodies
EP2603528B1 (en) 2010-08-10 2016-10-12 Glycotope GmbH Fab-glycosylated antibodies
BR112013003279A2 (en) 2010-08-13 2016-06-14 Genentech In "Methods for treating a disease, method for neutralizing or blocking il-1ß and / or il-18 activity, antibody, uses of an antibody and uses of a monoclonal antibody"
CA2809433A1 (en) 2010-08-26 2012-03-01 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
SG10201408229WA (en) 2010-08-31 2015-02-27 Genentech Inc Biomarkers and methods of treatment
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
BR112013006016A2 (en) 2010-09-15 2016-06-07 Hoffmann La Roche azabenzothiazole compounds, compositions and methods of use
WO2012048332A2 (en) * 2010-10-08 2012-04-12 John Williams A monoclonal antibody framework binding interface for meditopes, meditope delivery systems and methods for their use
US8962804B2 (en) 2010-10-08 2015-02-24 City Of Hope Meditopes and meditope-binding antibodies and uses thereof
US9309322B2 (en) 2010-11-12 2016-04-12 Scott & White Healthcare (Swh) Antibodies to tumor endothelial marker 8
WO2012066061A1 (en) 2010-11-19 2012-05-24 F. Hoffmann-La Roche Ag Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
KR20130133247A (en) 2010-12-21 2013-12-06 애브비 인코포레이티드 Il-1-alpha and -beta bispecific dual variable domain immunoglobulins and their use
WO2012085176A1 (en) 2010-12-23 2012-06-28 F. Hoffmann-La Roche Ag Tricyclic pyrazinone compounds, compositions and methods of use thereof as janus kinase inhibitors
JP5882913B2 (en) 2010-12-27 2016-03-09 協和発酵キリン株式会社 Method for preparing aqueous solution containing medium and chelating agent
WO2012095514A1 (en) 2011-01-14 2012-07-19 Vivalis Recombinant protein production system
WO2012100346A1 (en) * 2011-01-24 2012-08-02 Ym Biosciences Inc. Antibodies selective for cells presenting egfr at high density
EA028804B1 (en) 2011-03-25 2018-01-31 Гленмарк Фармасьютикалс С.А. Hetero-dimeric immunoglobulins
EP2714738B1 (en) 2011-05-24 2018-10-10 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
RU2014101492A (en) 2011-06-20 2015-07-27 Траслэшионал Кэнсэр Драгз Фарма, С.Л. METHOD FOR PREDICTING A CLINICAL RESPONSE TO CHEMOTHERAPY IN A SUBJECT WITH CANCER
WO2019071023A1 (en) 2017-10-04 2019-04-11 Yale University Compositions and methods for making selenocysteine containing polypeptides
US9464288B2 (en) 2011-07-11 2016-10-11 Yale University Compositions and methods for making selenocysteine containing polypeptides
WO2013007768A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors
WO2013007765A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Fused tricyclic compounds for use as inhibitors of janus kinases
EP2551348B1 (en) 2011-07-29 2014-09-24 Icon Genetics GmbH Production of galactosylated N-glycans in plants
WO2013022855A1 (en) 2011-08-05 2013-02-14 Xencor, Inc. Antibodies with modified isoelectric points and immunofiltering
CN103889976A (en) 2011-08-12 2014-06-25 弗·哈夫曼-拉罗切有限公司 Indazole compounds, compositions and methods of use
MX2014001766A (en) 2011-08-17 2014-05-01 Genentech Inc Neuregulin antibodies and uses thereof.
KR101870555B1 (en) 2011-08-23 2018-06-22 로슈 글리카트 아게 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
CA2845409A1 (en) 2011-09-20 2013-03-28 Yingjie Lai Imidazopyridine compounds, compositions and methods of use
US10196664B2 (en) 2011-10-04 2019-02-05 Icon Genetics Gmbh Nicotiana benthamiana plants deficient in fucosyltransferase activity
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
JP6310394B2 (en) 2011-10-10 2018-04-11 ゼンコア インコーポレイテッド Methods for purifying antibodies
MX2014004977A (en) 2011-10-24 2014-09-11 Abbvie Inc Immunobinders directed against sclerostin.
CA2857114A1 (en) 2011-11-30 2013-06-06 Genentech, Inc. Erbb3 mutations in cancer
TR201815709T4 (en) 2011-12-22 2018-11-21 Hoffmann La Roche Ion exchange membrane chromatography.
TW201333035A (en) 2011-12-30 2013-08-16 Abbvie Inc Dual specific binding proteins directed against IL-13 and/or IL-17
US9428553B2 (en) 2012-02-10 2016-08-30 City Of Hope Meditopes and meditope-binding antibodies and uses thereof
EP3296320A1 (en) 2012-03-08 2018-03-21 Halozyme, Inc. Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
JP2015514710A (en) 2012-03-27 2015-05-21 ジェネンテック, インコーポレイテッド Diagnosis and treatment of HER3 inhibitors
CN109880867A (en) 2012-03-27 2019-06-14 弗·哈夫曼-拉罗切有限公司 Improved harvest for recombinant protein operates
BR112014028600B1 (en) 2012-05-18 2022-11-22 Genentech, Inc SUSPENSION FORMULATIONS COMPRISING HIGH CONCENTRATION MONOCLONAL ANTIBODY, ITS METHOD OF PREPARATION, ITS USE AND DEVICE FOR SUBCUTANEOUS ADMINISTRATION, AND METHOD FOR PRODUCING AN ARTICLE OF MANUFACTURING
MX2014014356A (en) 2012-05-24 2015-07-06 Fundació Inst D Investigació Biomédica De Bellvitge Idibell Method for the identification of the origin of a cancer of unknown primary origin by methylation analysis.
AU2013270684B2 (en) 2012-06-08 2018-04-19 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
JP6103832B2 (en) 2012-06-25 2017-03-29 Hoya株式会社 EGFR binding peptide
WO2014011955A2 (en) 2012-07-12 2014-01-16 Abbvie, Inc. Il-1 binding proteins
ES2907763T3 (en) 2012-08-31 2022-04-26 Sutro Biopharma Inc Modified amino acids comprising an azido group
WO2014056783A1 (en) 2012-10-08 2014-04-17 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
AU2013334493B2 (en) 2012-10-26 2018-11-29 The University Of Queensland Use of endocytosis inhibitors and antibodies for cancer therapy
CA2890263C (en) 2012-11-01 2020-03-10 Abbvie Inc. Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
SG10201802044RA (en) 2012-11-01 2018-05-30 Abbvie Inc Stable dual variable domain immunoglobulin protein formulations
CN104994885B (en) 2012-12-19 2017-09-29 基因泰克公司 Method and composition for radiohalogen protein labeling
AU2014205086B2 (en) 2013-01-14 2019-04-18 Xencor, Inc. Novel heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
EP2945969A1 (en) 2013-01-15 2015-11-25 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
EP2954068B8 (en) 2013-02-05 2018-10-31 Pierian Holdings, Inc. Drug selection for non-small cell lung cancer therapy
JP2016509045A (en) 2013-02-22 2016-03-24 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト How to treat cancer and prevent drug resistance
MY192312A (en) 2013-02-26 2022-08-17 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
RU2015140915A (en) 2013-02-26 2017-04-03 Роше Гликарт Аг BSPECIFIC ANTI-BINDING MOLECULES ACTIVATING T-CELLS
US9925240B2 (en) 2013-03-06 2018-03-27 Genentech, Inc. Methods of treating and preventing cancer drug resistance
CN105980386B (en) 2013-03-13 2021-08-13 基因泰克公司 Pyrazolo compounds and uses thereof
EP2968540A2 (en) 2013-03-14 2016-01-20 Genentech, Inc. Combinations of a mek inhibitor compound with an her3/egfr inhibitor compound and methods of use
CA2905070A1 (en) 2013-03-14 2014-09-25 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
EP3421495A3 (en) 2013-03-15 2019-05-15 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions
MX2015011899A (en) 2013-03-15 2016-05-05 Genentech Inc Methods of treating cancer and preventing cancer drug resistance.
WO2014144280A2 (en) 2013-03-15 2014-09-18 Abbvie Inc. DUAL SPECIFIC BINDING PROTEINS DIRECTED AGAINST IL-1β AND / OR IL-17
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
CN105143257B (en) 2013-03-15 2020-10-27 艾伯维生物医疗股份有限公司 FC variants
ES2658039T3 (en) 2013-07-10 2018-03-08 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods for their preparation and methods of use
WO2015035044A2 (en) 2013-09-04 2015-03-12 Abbvie Biotherapeutics Inc. Fc VARIANTS WITH IMPROVED ANTIBODY-DEPENDENT CELL-MEDIATED CYTOTOXICITY
WO2015035062A1 (en) 2013-09-05 2015-03-12 Genentech, Inc. Antiproliferative compounds
TW201605857A (en) 2013-10-03 2016-02-16 赫孚孟拉羅股份公司 Therapeutic inhibitors of CDK8 and uses thereof
WO2015054658A1 (en) 2013-10-11 2015-04-16 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
MX2016004802A (en) 2013-10-18 2016-07-18 Genentech Inc Anti-rsp02 and/or anti-rsp03 antibodies and their uses.
PE20160724A1 (en) 2013-11-04 2016-08-04 Glenmark Pharmaceuticals Sa PRODUCTION OF T-CELL REDIRECTING HETERODIMERIC IMMUNOGLOBULINS
EP3083692B1 (en) 2013-12-17 2020-02-19 F.Hoffmann-La Roche Ag Methods of treating her2-positive cancers using pd-1 axis binding antagonists and anti-her2 antibodies
CA2934028A1 (en) 2013-12-17 2015-06-25 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
GB201322626D0 (en) 2013-12-19 2014-02-05 Immutep S A Combined preparations for the treatment of cancer
BR112016021383A2 (en) 2014-03-24 2017-10-03 Genentech Inc METHOD TO IDENTIFY A PATIENT WITH CANCER WHO IS LIKE OR LESS LIKELY TO RESPOND TO TREATMENT WITH A CMET ANTAGONIST, METHOD TO IDENTIFY A PATIENT WITH PREVIOUSLY TREATED CANCER, METHOD TO DETERMINE THE EXPRESSION OF THE HGF BIOMARKER, ANTI-C-MET ANTAGONIST AND ITS USE, DIAGNOSTIC KIT AND ITS PREPARATION METHOD
EP3954713A3 (en) 2014-03-28 2022-03-30 Xencor, Inc. Bispecific antibodies that bind to cd38 and cd3
EP3632934A1 (en) 2014-03-31 2020-04-08 F. Hoffmann-La Roche AG Anti-ox40 antibodies and methods of use
WO2015153514A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
ES2825574T3 (en) 2014-07-09 2021-05-17 Hoffmann La Roche PH adjustment to improve thaw recovery of cell banks
PE20170263A1 (en) 2014-08-04 2017-03-30 Hoffmann La Roche T-CELL ACTIVATING ANTIGEN-BINDING BI-SPECIFIC MOLECULES
WO2016036873A1 (en) 2014-09-05 2016-03-10 Genentech, Inc. Therapeutic compounds and uses thereof
EP3193866A1 (en) 2014-09-19 2017-07-26 Genentech, Inc. Use of cbp/ep300 and bet inhibitors for treatment of cancer
EP3204379B1 (en) 2014-10-10 2019-03-06 Genentech, Inc. Pyrrolidine amide compounds as histone demethylase inhibitors
SG10202006685XA (en) 2014-10-23 2020-08-28 Innate Pharma Treatment of Cancers Using Anti-NKG2A Agents
CN114381521A (en) 2014-11-03 2022-04-22 豪夫迈·罗氏有限公司 Methods and biomarkers for efficacy prediction and assessment of OX40 agonist treatment
CA2966523A1 (en) 2014-11-03 2016-05-12 Genentech, Inc. Assays for detecting t cell immune subsets and methods of use thereof
US11773166B2 (en) 2014-11-04 2023-10-03 Ichnos Sciences SA CD3/CD38 T cell retargeting hetero-dimeric immunoglobulins and methods of their production
RU2017119428A (en) 2014-11-06 2018-12-06 Дженентек, Инк. COMBINED THERAPY, INCLUDING THE USE OF OX40-CONNECTING AGONISTS AND TIGIT INHIBITORS
MA40940A (en) 2014-11-10 2017-09-19 Constellation Pharmaceuticals Inc SUBSTITUTED PYRROLOPYRIDINES USED AS BROMODOMA INHIBITORS
MA40943A (en) 2014-11-10 2017-09-19 Constellation Pharmaceuticals Inc SUBSTITUTED PYRROLOPYRIDINES USED AS BROMODOMA INHIBITORS
JP6639497B2 (en) 2014-11-10 2020-02-05 ジェネンテック, インコーポレイテッド Bromodomain inhibitors and uses thereof
WO2016081384A1 (en) 2014-11-17 2016-05-26 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
BR112017010324A2 (en) 2014-11-20 2018-05-15 F. Hoffmann-La Roche Ag method for treating or slowing cancer progression in an individual, molecules, methods for enhancing immune function in an individual and for selecting a patient for treatment, kits, pharmaceutical composition and uses of a combination of one molecule
CA2967426A1 (en) 2014-11-26 2016-06-02 Xencor, Inc. Heterodimeric antibodies that bind cd3 and tumor antigens
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
BR112017011166A2 (en) 2014-11-26 2018-02-27 Xencor, Inc. heterodimeric antibodies that bind to cd3 and cd38
EP3224258B1 (en) 2014-11-27 2019-08-14 Genentech, Inc. 4,5,6,7-tetrahydro-1h-pyrazolo[4,3-c]pyridin-3-amine compounds as cbp and/or ep300 inhibitors
ES2764299T3 (en) 2014-12-09 2020-06-02 Inst Nat Sante Rech Med Human monoclonal antibodies against AXL
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2016105450A2 (en) 2014-12-22 2016-06-30 Xencor, Inc. Trispecific antibodies
RU2710735C2 (en) 2014-12-23 2020-01-10 Дженентек, Инк. Compositions and methods of treating and diagnosing cancer-resistant cancer
EP3240908A2 (en) 2014-12-30 2017-11-08 F. Hoffmann-La Roche AG Methods and compositions for prognosis and treatment of cancers
CN107406414B (en) 2015-01-09 2022-04-19 基因泰克公司 (piperidin-3-yl) (naphthalen-2-yl) methanone derivatives as inhibitors of histone demethylase KDM2B for the treatment of cancer
JP6889661B2 (en) 2015-01-09 2021-06-18 ジェネンテック, インコーポレイテッド 4,5-Dihydroimidazole derivative and its use as a histone dimethylase (KDM2B) inhibitor
CN107406429B (en) 2015-01-09 2021-07-06 基因泰克公司 Pyridazinone derivatives and their use in the treatment of cancer
GB201500374D0 (en) 2015-01-09 2015-02-25 Immutep S A Combined preparations for the treatment of cancer
KR101776879B1 (en) 2015-01-19 2017-09-08 주식회사 녹십자 Pharmaceutical formulation comprising anti-egfr antibody
WO2016123391A1 (en) 2015-01-29 2016-08-04 Genentech, Inc. Therapeutic compounds and uses thereof
WO2016123387A1 (en) 2015-01-30 2016-08-04 Genentech, Inc. Therapeutic compounds and uses thereof
MA41598A (en) 2015-02-25 2018-01-02 Constellation Pharmaceuticals Inc PYRIDAZINE THERAPEUTIC COMPOUNDS AND THEIR USES
WO2016135041A1 (en) 2015-02-26 2016-09-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Fusion proteins and antibodies comprising thereof for promoting apoptosis
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
CN107709364A (en) 2015-04-07 2018-02-16 豪夫迈·罗氏有限公司 Antigen binding complex and application method with agonist activity
ES2835866T3 (en) 2015-05-12 2021-06-23 Hoffmann La Roche Therapeutic and diagnostic procedures for cancer
KR20180012753A (en) 2015-05-29 2018-02-06 제넨테크, 인크. Treatment and Diagnosis Methods for Cancer
EP3303399A1 (en) 2015-06-08 2018-04-11 H. Hoffnabb-La Roche Ag Methods of treating cancer using anti-ox40 antibodies
CA2988420A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists
CA2989400A1 (en) 2015-06-15 2016-12-22 Angiochem Inc. Ang1005 for the treatment of leptomeningeal carcinomatosis
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
MX2017016353A (en) 2015-06-17 2018-05-02 Genentech Inc Methods of treating locally advanced or metastatic breast cancers using pd-1 axis binding antagonists and taxanes.
DK3341376T3 (en) 2015-08-26 2021-03-29 Fundacion Del Sector Publico Estatal Centro Nac De Investigaciones Oncologicas Carlos Iii F S P Cnio CONDENSED TRICYCLIC COMPOUNDS AS PROTEINKINASE INHIBITORS
MX2018002226A (en) 2015-08-28 2018-03-23 Amunix Operating Inc Chimeric polypeptide assembly and methods of making and using the same.
WO2017041027A1 (en) 2015-09-04 2017-03-09 Obi Pharma, Inc. Glycan arrays and method of use
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
CN107849137B (en) 2015-10-02 2021-11-26 豪夫迈·罗氏有限公司 Bispecific anti-CEAXCD 3T cell activating antigen binding molecules
EP3387013B1 (en) 2015-12-07 2022-06-08 Xencor, Inc. Heterodimeric antibodies that bind cd3 and psma
JP7325186B2 (en) 2015-12-09 2023-08-14 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Type II anti-CD20 antibody for reducing the formation of anti-drug antibodies
CN113999249A (en) 2015-12-16 2022-02-01 基因泰克公司 Methods for making tricyclic PI3K inhibitor compounds and methods of treating cancer therewith
MX2018008347A (en) 2016-01-08 2018-12-06 Hoffmann La Roche Methods of treating cea-positive cancers using pd-1 axis binding antagonists and anti-cea/anti-cd3 bispecific antibodies.
CN109196121B (en) 2016-02-29 2022-01-04 基因泰克公司 Methods for treatment and diagnosis of cancer
UA125378C2 (en) 2016-03-14 2022-03-02 Універшітетет І Осло Engineered immunoglobulins with altered fcrn binding
FI3433280T3 (en) 2016-03-22 2023-06-06 Hoffmann La Roche Protease-activated t cell bispecific molecules
US10980894B2 (en) 2016-03-29 2021-04-20 Obi Pharma, Inc. Antibodies, pharmaceutical compositions and methods
KR20180121786A (en) 2016-03-29 2018-11-08 오비아이 파머 인코퍼레이티드 Antibodies, pharmaceutical compositions and methods
EP3436610A1 (en) 2016-03-29 2019-02-06 Geltor, Inc. Expression of proteins in gram-negative bacteria wherein the ratio of periplasmic volume to cytoplasmic volume is between 0.5:1 and 10:1
WO2017180864A1 (en) 2016-04-14 2017-10-19 Genentech, Inc. Anti-rspo3 antibodies and methods of use
JP2019515670A (en) 2016-04-15 2019-06-13 ジェネンテック, インコーポレイテッド Methods for monitoring and treating cancer
CN109072311A (en) 2016-04-15 2018-12-21 豪夫迈·罗氏有限公司 Diagnostic and therapeutic method for cancer
ES2850428T3 (en) 2016-04-15 2021-08-30 Hoffmann La Roche Cancer monitoring and treatment procedures
KR20230110820A (en) 2016-04-22 2023-07-25 오비아이 파머 인코퍼레이티드 Cancer immunotherapy by immune activation or immune modulation via globo series antigens
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
CN109476641B (en) 2016-05-24 2022-07-05 基因泰克公司 Heterocyclic inhibitors of CBP/EP300 and their use in the treatment of cancer
CN109476663B (en) 2016-05-24 2021-11-09 基因泰克公司 Pyrazolopyridine derivatives for the treatment of cancer
CN109415441B (en) 2016-05-24 2023-04-07 英斯梅德股份有限公司 Antibodies and methods of making same
SG11201810793XA (en) 2016-06-03 2018-12-28 Giordano Caponigro Pharmaceutical combinations
EP3469099A1 (en) 2016-06-08 2019-04-17 F. Hoffmann-La Roche AG Diagnostic and therapeutic methods for cancer
MX2018015592A (en) 2016-06-14 2019-04-24 Xencor Inc Bispecific checkpoint inhibitor antibodies.
US11098107B2 (en) 2016-06-15 2021-08-24 Sutro Biopharma, Inc. Antibodies with engineered CH2 domains, compositions thereof and methods of using the same
CA3029328A1 (en) 2016-06-28 2018-01-04 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
CN110072545A (en) 2016-07-27 2019-07-30 台湾浩鼎生技股份有限公司 Immunogenicity/therapeutic glycan pool object and application thereof
KR102528998B1 (en) 2016-07-29 2023-05-03 오비아이 파머 인코퍼레이티드 Human Antibodies, Pharmaceutical Compositions and Methods
EP3494139B1 (en) 2016-08-05 2022-01-12 F. Hoffmann-La Roche AG Multivalent and multiepitopic anitibodies having agonistic activity and methods of use
JP7250674B2 (en) 2016-08-08 2023-04-03 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト CANCER TREATMENT AND DIAGNOSTIC METHOD
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
EP3519437B1 (en) 2016-09-30 2021-09-08 F. Hoffmann-La Roche AG Bispecific antibodies against p95her2
AU2017339517B2 (en) 2016-10-06 2024-03-14 Foundation Medicine, Inc. Therapeutic and diagnostic methods for cancer
AU2017342559B2 (en) 2016-10-14 2022-03-24 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15/IL-15Ralpha Fc-fusion proteins and PD-1 antibody fragments
EP3532091A2 (en) 2016-10-29 2019-09-04 H. Hoffnabb-La Roche Ag Anti-mic antibidies and methods of use
JP2019535731A (en) 2016-11-21 2019-12-12 オービーアイ ファーマ,インコーポレイテッド Conjugated biological molecules, pharmaceutical compositions and methods
JP2020503316A (en) 2016-12-22 2020-01-30 ジェネンテック, インコーポレイテッド Methods and formulations for reducing the reconstitution time of lyophilized polypeptides
TW201837467A (en) 2017-03-01 2018-10-16 美商建南德克公司 Diagnostic and therapeutic methods for cancer
AU2018250875A1 (en) 2017-04-13 2019-10-03 F. Hoffmann-La Roche Ag An interleukin-2 immunoconjugate, a CD40 agonist, and optionally a PD-1 axis binding antagonist for use in methods of treating cancer
CN108948206B (en) * 2017-05-23 2022-08-23 赵磊 anti-EGFR/PD-1 double-targeting antibody, preparation method and application thereof
JP2020529832A (en) 2017-06-30 2020-10-15 ゼンコア インコーポレイテッド Targeted heterodimer Fc fusion protein containing IL-15 / IL-15Rα and antigen binding domain
JP2020527351A (en) 2017-07-21 2020-09-10 ジェネンテック, インコーポレイテッド Cancer treatment and diagnosis
WO2019033043A2 (en) 2017-08-11 2019-02-14 Genentech, Inc. Anti-cd8 antibodies and uses thereof
KR102208378B1 (en) 2017-08-17 2021-01-28 주식회사 녹십자 Pharmaceutical formulation comprising anti-egfr antibody
CA3073073A1 (en) 2017-09-08 2019-03-14 F. Hoffmann-La Roche Ag Diagnostic and therapeutic methods for cancer
US11180541B2 (en) 2017-09-28 2021-11-23 Geltor, Inc. Recombinant collagen and elastin molecules and uses thereof
JP2021502066A (en) 2017-11-06 2021-01-28 ジェネンテック, インコーポレイテッド Cancer diagnosis and therapy
EP3706793A1 (en) 2017-11-08 2020-09-16 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-pd-1 sequences
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
WO2019125732A1 (en) 2017-12-19 2019-06-27 Xencor, Inc. Engineered il-2 fc fusion proteins
AU2019218959A1 (en) 2018-02-08 2020-09-03 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
WO2019165434A1 (en) 2018-02-26 2019-08-29 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
AU2019247415A1 (en) 2018-04-04 2020-10-22 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
CA3097593A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof
AU2019256529A1 (en) 2018-04-18 2020-11-26 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and TIM-3 antigen binding domains
EP3794041B1 (en) 2018-05-18 2023-07-12 Glycotope GmbH Anti-muc1 antibody
AU2019275404A1 (en) 2018-05-21 2020-12-03 Nanostring Technologies, Inc. Molecular gene signatures and methods of using same
WO2019246557A1 (en) 2018-06-23 2019-12-26 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
US11203645B2 (en) 2018-06-27 2021-12-21 Obi Pharma, Inc. Glycosynthase variants for glycoprotein engineering and methods of use
BR112020025623A2 (en) 2018-07-03 2021-03-23 Bristol-Myers Squibb Company methods of producing recombinant proteins
CA3104147A1 (en) 2018-07-18 2020-01-23 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
WO2020028909A1 (en) 2018-08-03 2020-02-06 Brown University Oral formulations with increased uptake
JP2021535169A (en) 2018-09-03 2021-12-16 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Carboxamide and sulfonamide derivatives useful as TEAD modulators
AU2019342099A1 (en) 2018-09-19 2021-04-08 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
EP4249917A3 (en) 2018-09-21 2023-11-08 F. Hoffmann-La Roche AG Diagnostic methods for triple-negative breast cancer
JP2022503959A (en) 2018-10-03 2022-01-12 ゼンコア インコーポレイテッド IL-12 heterodimer FC-fusion protein
US20210380923A1 (en) 2018-10-10 2021-12-09 Boehringer Ingelheim International Gmbh Method for membrane gas transfer in high density bioreactor culture
WO2020076849A1 (en) 2018-10-11 2020-04-16 The Scripps Research Institute Antibody compounds with reactive arginine and related antibody drug conjugates
MX2021004348A (en) 2018-10-18 2021-05-28 Genentech Inc Diagnostic and therapeutic methods for sarcomatoid kidney cancer.
WO2020136147A1 (en) 2018-12-26 2020-07-02 Innate Pharma Compounds and methods for treatment of head and neck cancer
MX2021007797A (en) 2018-12-28 2021-10-26 Kyowa Kirin Co Ltd BISPECIFIC ANTIBODY BINDING TO TfR.
CN113396230A (en) 2019-02-08 2021-09-14 豪夫迈·罗氏有限公司 Methods of diagnosis and treatment of cancer
JP2022522185A (en) 2019-02-27 2022-04-14 エピアクシス セラピューティクス プロプライエタリー リミテッド Methods and agents for assessing T cell function and predicting response to treatment
CN113710706A (en) 2019-02-27 2021-11-26 豪夫迈·罗氏有限公司 Administration for anti-TIGIT antibody and anti-CD 20 antibody or anti-CD 38 antibody treatment
WO2020180726A1 (en) 2019-03-01 2020-09-10 Xencor, Inc. Heterodimeric antibodies that bind enpp3 and cd3
SG11202109884WA (en) * 2019-03-29 2021-10-28 Green Cross Corp Fusion protein comprising anti-mesothelin antibody, anti-cd3 antibody or anti-egfr antibody, bispecific or trispecific antibody comprising same, and uses thereof
KR20210145152A (en) 2019-04-01 2021-12-01 제넨테크, 인크. Compositions and methods for stabilization of protein-containing formulations
CN113993885A (en) 2019-04-12 2022-01-28 格尔托公司 Recombinant elastin and production thereof
WO2020223233A1 (en) 2019-04-30 2020-11-05 Genentech, Inc. Prognostic and therapeutic methods for colorectal cancer
AU2020270376A1 (en) 2019-05-03 2021-10-07 Genentech, Inc. Methods of treating cancer with an anti-PD-L1 antibody
CN114269749A (en) 2019-06-10 2022-04-01 苏特罗生物制药公司 5H-pyrrolo [3,2-d ] pyrimidine-2, 4-diamino compounds and antibody conjugates thereof
JP2022536800A (en) 2019-06-17 2022-08-18 ストロ バイオファーマ インコーポレーテッド 1-(4-(aminomethyl)benzyl)-2-butyl-2H-pyrazolo[3,4-C]quinolin-4-amine derivatives and related compounds as TOLL-like receptor (TLR) 7/8 agonists, and antibody drug conjugates thereof for use in cancer therapy and diagnosis
CN112300279A (en) 2019-07-26 2021-02-02 上海复宏汉霖生物技术股份有限公司 Methods and compositions directed to anti-CD 73 antibodies and variants
KR20220039777A (en) 2019-08-01 2022-03-29 브리스톨-마이어스 스큅 컴퍼니 Methods for improving protein productivity in fed-batch cell culture
MX2022002738A (en) 2019-09-04 2022-06-27 Genentech Inc Cd8 binding agents and uses thereof.
CR20220127A (en) 2019-09-27 2022-05-27 Genentech Inc Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
US20230024096A1 (en) 2019-10-29 2023-01-26 Hoffmann-La Roche Inc. Bifunctional compounds for the treatment of cancer
US20220389103A1 (en) 2019-11-06 2022-12-08 Genentech, Inc. Diagnostic and therapeutic methods for treatment of hematologic cancers
CA3155989A1 (en) 2019-11-13 2021-05-20 Jason Robert ZBIEG Therapeutic compounds and methods of use
WO2021100034A1 (en) 2019-11-19 2021-05-27 Protalix Ltd. Removal of constructs from transformed cells
PE20221511A1 (en) 2019-12-13 2022-10-04 Genentech Inc ANTI-LY6G6D ANTIBODIES AND METHODS OF USE
AU2020408562A1 (en) 2019-12-20 2022-06-23 Erasca, Inc. Tricyclic pyridones and pyrimidones
WO2021194481A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
EP4114852A1 (en) 2020-03-03 2023-01-11 Sutro Biopharma, Inc. Antibodies comprising site-specific glutamine tags, methods of their preparation and methods of their use
WO2021177980A1 (en) 2020-03-06 2021-09-10 Genentech, Inc. Combination therapy for cancer comprising pd-1 axis binding antagonist and il6 antagonist
CN115698717A (en) 2020-04-03 2023-02-03 基因泰克公司 Methods of treatment and diagnosis of cancer
EP4143345A1 (en) 2020-04-28 2023-03-08 Genentech, Inc. Methods and compositions for non-small cell lung cancer immunotherapy
WO2021231976A1 (en) 2020-05-14 2021-11-18 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (psma) and cd3
EP3915576A1 (en) 2020-05-28 2021-12-01 Fundació Privada Institut d'Investigació Oncològica de Vall-Hebron Chimeric antigen receptors specific for p95her2 and uses thereof
KR20230025691A (en) 2020-06-16 2023-02-22 제넨테크, 인크. Methods and compositions for treating triple negative breast cancer
EP4168118A1 (en) 2020-06-18 2023-04-26 Genentech, Inc. Treatment with anti-tigit antibodies and pd-1 axis binding antagonists
AU2021291011A1 (en) 2020-06-19 2023-01-05 F. Hoffmann-La Roche Ag Antibodies binding to CD3 and CD19
EP4172621A1 (en) 2020-06-30 2023-05-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting the risk of recurrence and/or death of patients suffering from a solid cancer after preoperative adjuvant therapies
CN115843335A (en) 2020-06-30 2023-03-24 国家医疗保健研究所 Method for predicting the risk of relapse and/or death of a patient with solid cancer after preoperative adjuvant and radical surgery
EP3939999A1 (en) 2020-07-14 2022-01-19 Fundación del Sector Público Estatal Centro Nacional de Investigaciones Oncológicas Carlos III (F.S.P. CNIO) Interleukin 11 receptor alpha subunit (il11ra) neutralizing antibodies and uses thereof
US11787775B2 (en) 2020-07-24 2023-10-17 Genentech, Inc. Therapeutic compounds and methods of use
JP2023536602A (en) 2020-08-03 2023-08-28 ジェネンテック, インコーポレイテッド Diagnostic and therapeutic methods for lymphoma
EP4196612A1 (en) 2020-08-12 2023-06-21 Genentech, Inc. Diagnostic and therapeutic methods for cancer
AU2021329378A1 (en) 2020-08-19 2023-03-23 Xencor, Inc. Anti-CD28 compositions
KR20230073270A (en) 2020-09-22 2023-05-25 브리스톨-마이어스 스큅 컴퍼니 Methods of producing therapeutic proteins
IL301524A (en) 2020-09-23 2023-05-01 Erasca Inc Tricyclic pyridones and pyrimidones
CN116406291A (en) 2020-10-05 2023-07-07 基因泰克公司 Administration of treatment with anti-FCRH 5/anti-CD 3 bispecific antibodies
US20230374447A1 (en) 2020-10-05 2023-11-23 Protalix Ltd. Dicer-like knock-out plant cells
WO2022103983A2 (en) 2020-11-11 2022-05-19 Sutro Biopharma, Inc. Fluorenylmethyloxycarbonyl and fluorenylmethylaminocarbonyl compounds, protein conjugates thereof, and methods for their use
US20230107642A1 (en) 2020-12-18 2023-04-06 Erasca, Inc. Tricyclic pyridones and pyrimidones
PE20231505A1 (en) 2021-02-12 2023-09-26 Hoffmann La Roche BICYCLIC TETRAHYDROAZEPINE DERIVATIVES FOR THE TREATMENT OF CANCER
EP4305067A1 (en) 2021-03-09 2024-01-17 Xencor, Inc. Heterodimeric antibodies that bind cd3 and cldn6
WO2022192586A1 (en) 2021-03-10 2022-09-15 Xencor, Inc. Heterodimeric antibodies that bind cd3 and gpc3
AU2022280025A1 (en) 2021-05-25 2023-12-07 Erasca, Inc. Sulfur-containing heteroaromatic tricyclic kras inhibitors
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
WO2023010060A2 (en) 2021-07-27 2023-02-02 Novab, Inc. Engineered vlrb antibodies with immune effector functions
WO2023015234A1 (en) 2021-08-05 2023-02-09 Bristol-Myers Squibb Company Cell culture methods for producing therapeutic proteins
WO2023018699A1 (en) 2021-08-10 2023-02-16 Erasca, Inc. Selective kras inhibitors
WO2023049687A1 (en) 2021-09-21 2023-03-30 Bristol-Myers Squibb Company Methods of controlling the level of dissolved oxygen (do) in a solution comprising a recombinant protein in a storage container
WO2023097195A1 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic indazole compounds and methods of use in the treatment of cancer
US20230203062A1 (en) 2021-11-24 2023-06-29 Genentech, Inc. Therapeutic compounds and methods of use
WO2023164487A1 (en) 2022-02-22 2023-08-31 Brown University Compositions and methods to achieve systemic uptake of particles following oral or mucosal administration
EP4253418A1 (en) 2022-03-29 2023-10-04 Fundació Privada Institut d'Investigació Oncològica de Vall-Hebron Immune cells expressing chimeric antigen receptors and bispecific antibodies and uses thereof
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2024006272A1 (en) 2022-06-27 2024-01-04 Sutro Biopharma, Inc. β-GLUCURONIDE LINKER-PAYLOADS, PROTEIN CONJUGATES THEREOF, AND METHODS THEREOF
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024015229A1 (en) 2022-07-15 2024-01-18 Sutro Biopharma, Inc. Protease/enzyme cleavable linker-payloads and protein conjugates
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024033389A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydrothiazepine derivatives
WO2024033457A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydrothiazepine derivatives
WO2024033458A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydroazepine derivatives
WO2024033388A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydrothiazepine derivatives

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0739984A1 (en) * 1995-04-26 1996-10-30 San Tumorforschungs-Gmbh Bivalent polypeptides containing at least two domains

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) * 1986-03-27 1986-04-30 Winter G P Recombinant dna product
ES2206447T3 (en) * 1991-06-14 2004-05-16 Genentech, Inc. HUMANIZED ANTIBODY FOR HEREGULINE.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0739984A1 (en) * 1995-04-26 1996-10-30 San Tumorforschungs-Gmbh Bivalent polypeptides containing at least two domains

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FAN Z ET AL: "Blockade of Epidermal Growth Factor REceptor function by bivalent and monovalent fragments of 225 anti-EGFR monoclonal antibodies" CANCER RESEARCH, vol. 53, , pages 4322-4328, XP008014409 *
See also references of WO9640210A1 *

Also Published As

Publication number Publication date
CA2222231A1 (en) 1996-12-19
EP0831880A4 (en) 2004-12-01
JP2009062375A (en) 2009-03-26
AU6267896A (en) 1996-12-30
JP2006246896A (en) 2006-09-21
WO1996040210A1 (en) 1996-12-19
JPH11507535A (en) 1999-07-06

Similar Documents

Publication Publication Date Title
US7060808B1 (en) Humanized anti-EGF receptor monoclonal antibody
EP0831880A1 (en) Antibody and antibody fragments for inhibiting the growth of tumors
KR101676622B1 (en) Recombinant anti-epidermal growth factor receptor antibody compositions
KR101706255B1 (en) Recombinant anti-epidermal growth factor receptor antibody compositions
EP1527100B1 (en) Human monoclonal antibodies to interleukin-5 and methods and compositions comprising same
EP2516469B1 (en) ANTI-HER3 Antibodies and uses thereof
EP2220121B1 (en) Axl antibodies
JP5513888B2 (en) Compositions and methods for glucagon receptor antibodies
CN105968206B (en) anti-ERBB 3 antibodies
CN110545844A (en) Cancer treatment using antibodies that bind cytotoxic T lymphocyte antigen-4 (CTLA-4)
EP2476427B1 (en) A method of treating cancer comprising a VEGF-B antagonist
EP2943508B1 (en) Combination therapy of anti-her3 and anti-her2 antibodies
KR20180004277A (en) PDL-1 antibody, pharmaceutical composition thereof and uses thereof
JP2002514421A (en) Antibodies to CD23, their derivatives, and their therapeutic use
RU2766582C2 (en) Immunotherapy using antibodies binding programmed cell death protein ligand 1 (pd-l1)
EP2420514A1 (en) Targeted binding agents directed to PDGFR-alpha and uses thereof
SK332792A3 (en) Humanized and chimeric monoclonal antibodies
US20170002084A1 (en) Monoclonal antibodies directed to cd20
KR20230034960A (en) Antibodies that bind to LAG3 and uses thereof
CA3067597C (en) Chimeric antibodies for treatment of amyloid deposition diseases
US20030224001A1 (en) Antibody and antibody fragments for inhibiting the growth of tumors
CA2791560A1 (en) Monoclonal antibodies directed to cd20
CN115702003A (en) Chemokine receptor 4 (CXCR 4) antagonist antibodies
WO2023241656A1 (en) Bispecific antibody containing anti-cldn18.2 antibody, and pharmaceutical composition and use thereof
EP4059963A1 (en) Molecule capable of binding to human 4-1bb, and application of molecule

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19971204

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL PAYMENT 971204;LT PAYMENT 971204;LV PAYMENT 971204;SI PAYMENT 971204

A4 Supplementary search report drawn up and despatched

Effective date: 20041015

17Q First examination report despatched

Effective date: 20050502

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SALDANHA, JOSE WILLIAM

Inventor name: JONES, STEVEN TARRAN

Inventor name: GIORGIO, NICHOLAS A.

Inventor name: GOLDSTEIN, NEIL I.

17Q First examination report despatched

Effective date: 20050502

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090519