WO2005053604A2 - Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees - Google Patents

Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees Download PDF

Info

Publication number
WO2005053604A2
WO2005053604A2 PCT/US2004/017855 US2004017855W WO2005053604A2 WO 2005053604 A2 WO2005053604 A2 WO 2005053604A2 US 2004017855 W US2004017855 W US 2004017855W WO 2005053604 A2 WO2005053604 A2 WO 2005053604A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
human
variable region
set forth
Prior art date
Application number
PCT/US2004/017855
Other languages
English (en)
Other versions
WO2005053604A3 (fr
Inventor
Jing Ma
Yajun Guo
Original Assignee
Oncomax Acquisition Corp.
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
Priority claimed from CNB2003101199264A external-priority patent/CN1279056C/zh
Application filed by Oncomax Acquisition Corp. filed Critical Oncomax Acquisition Corp.
Priority to CA002528182A priority Critical patent/CA2528182A1/fr
Priority to EP04776307A priority patent/EP1631238A2/fr
Priority to AU2004294893A priority patent/AU2004294893A1/en
Priority to JP2006533570A priority patent/JP2007535905A/ja
Publication of WO2005053604A2 publication Critical patent/WO2005053604A2/fr
Priority to IL172242A priority patent/IL172242A0/en
Publication of WO2005053604A3 publication Critical patent/WO2005053604A3/fr

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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3015Breast
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/303Liver or Pancreas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen

Definitions

  • the present invention relates generally to the field of cancer biology and immunotherapy. More specifically, it relates to a tumor antigen specifically expressed in melanoma, breast cancer and hepatocellular carcinoma, antibodies directed against the tumor antigen, and methods of diagnosing and/or treating cancers associated with the antigen.
  • Malignancy is one of the key diseases threatening the life of human kind. Melanoma, breast cancer and hepatocellular carcinoma are some of the most common malignancies.
  • Melanoma is a type of cancer with high degree of malignancy. About 20% of the melanoma cases occur in neck and head, with the majority on skin and mucus membranes of cavities and sinuses. Generally human melanomas originate from pigmentary naevi, which become destmctive metastatic melanoma cells when stimulated by irradiation. Melanomas are quite liable to spread through the body and hard to control. Chemotherapy and radiotherapy are not effective to melanomas.
  • HMB-45 The most commonly used antibodies for melanoma immunohistochemical diagnosis are HMB-45, anti-S-100, and NKI/C3.(Smoller BR, Pathology: state of the art reviews, 2: 371-383, 1994). These antibodies have their disadvantages.
  • the sensitivity of HMB-45 is not high enough (sensitivity between 67%-93%), and may leave some melanoma cases undiagnosed (Wick MR et al., J Cutan Pathol 15: 201-207, 1988; Ordonez NG et al. Am J. Clin. Pathol. 90: 385-390, 1988).
  • NKI/C3 binds to a lot of benign as well as malignant tumors, and this non-specificity greatly limits the application of NKI/C3 in melanoma diagnosis.
  • Breast cancer is one of the most frequent malignancies for women. About 1.2 million women are affected by the disease each year, and 0.5 million are dying from the disease each year in the world. Developed countries in North America, Western and Northern Europe have the highest occurrence of the disease, and countries in Africa have the lowest occurrence. The new cases of breast cancer are increasing significantly all across the world and are growing at a rate of 5-20% both in high-occurring and low-occurring areas.
  • Hepatocellular carcinoma is one of the most frequent malignancies in China and its occurrence is related to hepatitis.
  • Liver cancer antigens that are most studied include PHC, F062, 25T and Habl ⁇ et al (Lian-Jun Yang, Wen-Liang Wang., World J. Gastroenterol. 8(5): 808-814, 2002).
  • AFP is found not only expressed in the blood of liver cancer patients, but also expressed in ovarian and testis cancer cells.
  • Hepatoma-specific monoclonal antibody Hab 18 has also been studied, but it fails to be an effective target for hepatoma therapy.
  • the invention disclosed herein concerns antibodies (such as monoclonal antibodies) and polypeptides that specifically bind to an antigen SM5-1 which is expressed in melanoma, breast cancer, and hepatocellular carcinoma.
  • the molecular weight of this antigen is 230 kD and 180 kD.
  • This antigen can be isolated from a melanoma, breast cancer and/or hepatocellular carcinoma cell using antibodies of the invention.
  • the invention provides a human SM5-1 specific monoclonal antibody (huSM5-l).
  • the variable region of heavy chain and light chain of the human SM5-1 specific monoclonal antibody (huSM5-l) are shown in Table 1.
  • the antibody huSM5-l is produced by a host cell having an accessing number or progeny thereof.
  • the invention is an antibody or a polypeptide comprising a fragment or a region of the antibody huSM5-l .
  • the fragment is a heavy chain of the antibody huSM5-l as shown in Table 1.
  • the fragment is a light chain of the antibody huSM5-l as shown in Table 1.
  • the fragment contains one or more variable regions from a heavy chain and/or a light chain of the antibody huSM5-l as set forth in SEQ ID NO:9 and SEQ ID NO: 10.
  • the fragment contains one or more complementarity determining regions (CDRs) from a heavy chain and/or a light chain of the antibody huSM5-l as shown in Table 1.
  • CDRs complementarity determining regions
  • the invention provides antibodies that competitively inhibits the immunospecific binding of antibody huSM5-l to a SM5-1 target antigen.
  • the variable region of heavy chain of the antibody comprises the amino acid sequences 31-35, 50-66 and 99-108 set forth in SEQ ID NO:9 and the variable region of light chain of the antibody comprises the amino acid sequences 24-40, 56-62 and 95-102 set forth in SEQ ID NO: 10.
  • variable region of heavy chain of the antibody comprises the amino acid sequence set forth in SEQ ID NO:9.
  • variable region of light chain of the antibody comprises the amino acid sequence set forth in SEQ ID NO:10.
  • variable region of heavy chain of the antibody comprises the amino acid sequence set forth in SEQ ID NO:9 and the variable region of light chain of the antibody comprises the amino acid sequence set forth in SEQ ID NO: 10.
  • the antibody is a human antibody.
  • the invention provides a mouse SM5-1 specific antibody (mSM5-l)
  • variable region of the heavy chain and the light chain are shown in Table 2.
  • the variable region of heavy chain of antibody mSM5-l comprises the amino acid sequence set forth in SEQ
  • variable region of light chain of the antibody mSM5-l comprises the amino acid sequence set forth in SEQ ID NO:4.
  • the invention is an antibody or a polypeptide comprising a fragment or a region of the antibody mSM5-l .
  • the fragment contains one or more variable regions from a heavy chain and/or a light chain of the antibody mSM5-l as set forth in SEQ ID NO:3 and SEQ ID NO:4.
  • the fragment contains one or more complementarity determining regions (CDRs) from a heavy chain and/or a light chain of the antibody mSM5-l as set forth in SEQ ID NO:3 and SEQ ID NO:4.
  • CDRs complementarity determining regions
  • the invention provides an antibody that competitively inhibits the immunospecific binding of the antibody mSM5-l to a SM5-1 target antigen.
  • the variable region of heavy chain of the antibody comprises the amino acid sequences 31-35, 50-66 and 99-108 set forth in SEQ ID NO:3 and the variable region of light chain of the antibody comprises the amino acid sequences 24-40, 56-62 and 95-102 set forth in
  • the antibody is a humanized antibody.
  • the antibody immunospecific for a SM5-1 target antigen is a chimeric antibody, wherein the variable region of heavy chain of the antibody comprises the amino acid sequence set forth in SEQ ID NO:3 and the variable region of light chain of the antibody comprises the amino acid sequence set forth in SEQ ID NO:4.
  • the antibody of the invention is a humanized antibody, wherein the variable region of heavy chain of the humanized antibody comprises the amino acid sequence set forth in SEQ ID NO:l and/or the variable region of light chain of the humanized antibody comprises the amino acid sequence set forth in SEQ ID NO:2.
  • the humanized antibody is produced by a host cell with an accession number or progeny thereof.
  • the invention is an antibody or a polypeptide comprising a fragment or a region of the humanized antibody.
  • the fragment contains one or more variable regions from a heavy chain and/or a light chain of the humanized antibody as set forth in SEQ ID NO: 1 and SEQ ID NO:2.
  • the fragment contains one or more CDRs from a heavy chain and/or a light chain of the humanized antibody as set forth in SEQ ID NO: 1 and SEQ ID NO:2.
  • the invention also provides antibodies that competitively inhibits the immunospecific binding of the humanized antibody to a SM5-1 target antigen.
  • the antibody of the invention is a polyclonal antibody, a monoclonal antibody, a Fab fragment, a Fab' fragment, a F(ab') 2 fragment, a Fv fragment, a diabody, a single-chain antibody, or a multi-specific antibody formed from antibody fragments described herein.
  • the invention also provides isolated nucleic acid comprising a nucleotide sequence encoding the heavy chain and/or the light chain, or a fragment thereof, of any of the antibody described herein.
  • the nucleic acid comprises the nucleotide sequence encoding amino acid sequence set forth in SEQ ID NO:9 and/or SEQ ID NO: 10.
  • the nucleic acid comprises the nucleotide sequence set forth in SEQ ID NO: 11 and/or SEQ ID NO: 12.
  • the nucleic acid comprises the nucleotide sequence encoding amino acid sequence set forth in SEQ ID NO: 3 and/or SEQ ID NO:4.
  • the nucleic acid comprises the nucleotide sequence set forth in SEQ ID NO:7 and/or SEQ ID NO:8. In other embodiments, the nucleic acid comprises the nucleotide sequence encoding amino acid sequence set forth in SEQ ID NO: 1 and/or SEQ ID NO:2. In other embodiments, the nucleic acid comprises the nucleotide sequence set forth in SEQ ID NO:5 and/or SEQ ID NO:6. [0020] In another aspect, the invention provides an isolated nucleic acid comprising a nucleotide sequence complementary to any of the nucleotide sequence described herein. [0021] In another aspect, the invention provides a vector containing any of the nucleic acid described herein.
  • the vector may further comprises expression modulation sequence operatively linked to the nucleic acid encoding any of the antibody described herein.
  • the invention also provides a recombinant cell containing any of the nucleic acid described herein.
  • the recombinant cell is an eukaryote cell.
  • the recombinant cell is a CHO cell.
  • the cell has an accession number .
  • the cell has an accession number .
  • the invention is a method of producing any of the antibody described herein, or a fragment thereof, comprising growing a recombinant cell containing the nucleic acid such that the encoded antibody, or a fragment thereof, is expressed by the cell; and recovering the expressed antibody, or a fragment thereof. In some embodiments, the method further comprises isolating and/or purifying the recovered antibody, or a fragment thereof.
  • the invention provides a pharmaceutical composition comprising an effective amount of any of the antibody described herein and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition comprising an effective amount of a human SM5-1 specific monoclonal antibody and a pharmaceutically acceptable carrier or excipient, wherein the variable region of heavy chain of the human SM5-1 specific monoclonal antibody comprises the amino acid sequences 31-35, 50-66 and 99-108 set forth in SEQ ID NO:9 and the variable region of light chain of the human SM5-1 specific monoclonal antibody comprises the amino acid sequences 24-40, 56-62 and 95-102 set forth in SEQ ID NO: 10.
  • the pharmaceutical composition comprising an effective amount of a humanized SM5-1 specific monoclonal antibody and a pharmaceutically acceptable carrier or excipient, wherein the variable region of heavy chain of the humanized SM5-1 specific monoclonal antibody comprises the amino acid sequences 31-35, 50-66 and 99-108 set forth in SEQ ID NO:l and the variable region of light chain of the humanized SM5-1 specific monoclonal antibody comprises the amino acid sequences 24-40, 56-62 and 95-102 set forth in
  • the invention also provides a kit comprising an effective amount of any of the antibody described herein, and an instruction means for administering the antibody.
  • the invention provides a method for treating neoplasm in a mammal, which method comprises administering to a mammal to wliich such treatment is needed or desirable, an effective amount of any of the antibody described herein.
  • the mammal is a human.
  • the neoplasm is melanoma, breast cancer or hepatocellular carcinoma.
  • the antibody exerts its anti-neoplasm effect via antibody dependent cell mediated cytotoxicity (ADCC) or complement dependent cell mediated cytotoxicity (CDC).
  • ADCC antibody dependent cell mediated cytotoxicity
  • CDC complement dependent cell mediated cytotoxicity
  • the antibody is a human antibody.
  • the antibody is a humanized antibody.
  • the invention provides a combination, which combination comprises: a) an effective amount of an antibody described herein; and b) an effective amount of' an anti-neoplasm agent.
  • the anti-neoplasm agent is an agent that treats melanoma, breast cancer or hepatocellular carcinoma.
  • the invention also provides a method for treating neoplasm in a mammal, which method comprises administering to a mammal to which such treatment is needed or desirable, an effective amount of a combination described herein.
  • the invention provides a method for inducing caspase-10 mediated apoptosis in a cell, which method comprises administering to a cell to which such induction is needed or desirable, an effective amount of any of the antibody described herein.
  • the cell is a mammalian cell. In some embodiments, the cell is contained in a mammal.
  • the invention also provides a conjugate, which conjugate comprises any of the antibody described herein conjugated to a toxin and/or a radioactive isotope.
  • the invention provides a method for assaying for SM5-1 target antigen (e.g., human SM5-1 target antigen) in a sample, which method comprises: a) obtaining a sample from a subject to be tested; b) contacting said sample with any of the antibody described herein under suitable conditions to allow binding between said SM5-1 target antigen, if present in said sample, to said antibody; and c) assessing binding between said SM5-1 target antigen, if present in said sample, to said antibody to determine presence, absence and/or amount of said SM5-1 target antigen in said sample.
  • the method is used in the prognosis or diagnosis of a neoplasm.
  • the neoplasm is melanoma, breast cancer or hepat
  • the invention also provides a kit for assaying for SM5-1 target antigen (e.g., human SM5-1 target antigen) in a sample, which method comprises: a) any of the antibody described herein; and b) means for assessing binding between said SM5-1 target antigen, if present in said sample, to said antibody to determine presence, absence and/or amount of said SM5-1 target antigen in said sample.
  • SM5-1 target antigen e.g., human SM5-1 target antigen
  • Figure 1 illustrates the expression vector pMGl 8-3K. Regions of the vector encoding different functions are indicated.
  • HCMV pro human cytomegalovirus Major Immediate Early promoter
  • C the human K chain constant region gene
  • CH the human ⁇ l chain constant region gene
  • pA polyadenylation signal
  • DHFR dihydrofolate reductase gene
  • pUC origin plasmid origin of replication
  • Amp designates the ⁇ -lactamase gene.
  • Figure 2 illustrates amino acid sequences of the heavy and light chain variable regions of the humanized anti-SM5-l antibody (ReSM5-l).
  • the V H of human antibody KOL was chosen as framework for the humanized heavy chain and the V L of human Bence-Jones protein REI was chosen for the humanized light chain.
  • the dashes represent amino acids that are the same as the corresponding residues in human antibodies KOL or REI.
  • the CDRs are enclosed in brackets. Amino acids (in one-letter notation) are numbered according to Kabat
  • Figure 3 illustrates the FACS graph showing human anti-SM5-l antibody (huSM5-l) binding to breast cancer cell lines and melanoma cell lines.
  • Figure 4 illustrates the immunofootprinting with human SM5-1 antigen.
  • Figure 5 illustrates the changes of activity of caspase- 10 in human anti-SM5- 1 antibody (huSM5-l) treated QYC and XJC cells.
  • Figure 6 illustrates the inhibition of proliferation/growth curve lines for QYC cells treated with humanized and chimeric anti-SM5-l antibodies.
  • A growth inhibition curve lines for chSM5-l treated cells;
  • B growth inhibition curve lines for ReSM5-l treated cells.
  • Figure 7 illustrates anti-neoplasm effect by chSM5-l antibody and humanized anti-SM5-l antibody (ReSM5-l) on QYC cells via ADCC.
  • Figure 8 illustrates anti-neoplasm effect by chSM5-l antibody (A) and humanized anti-SM5-l antibody (ReSM5-l) (B) on QYC cells via CDC.
  • Figure 9 illustrates the therapeutic effect of chSM5-l and ReSM5-l for QYC bearing nude mice.
  • Figure 10 illustrates the distribution of 125 I labeled ReSM5-l and chSM5-l .
  • the present invention is based on a discovery of a new antigen, SM5-1 antigen, which is over expressed in melanoma, breast cancer and hepatocellular carcinoma.
  • the antigen is present in glycosylated and non-glycosylated form.
  • the antibodies of the invention specifically bound to two proteins having a molecular weight of 230 kD (designated as A230) and 180 kD (designated as A180).
  • the invention also provides antibodies specific for SM5-1 antigen such as the human antibody (huSM5-l, the variable regions shown in Table 1), the humanized antibody (ReSM5-l, the variable regions are shown in Table 3), and the chimeric antibody (chSM5-l, the variable regions are shown in Table 2).
  • SM5-1 antigen can bind to the antigen present in melanoma, breast cancer and hepatocellular carcinoma.
  • These antibodies also inhibit growth and/or proliferation, induce caspase-10 mediated apoptosis in these cancer cells.
  • SM5-1 antigen provides a target for treating these malignancies.
  • an “antibody” is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • the term encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab') 2 , Fv), single chain (ScFv), a diabody, a multi-specific antibody formed from antibody fragments, mutants thereof, fusion proteins comprising an antibody portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity.
  • An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes.
  • immunoglobulins There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • a “monoclonal antibody” refers to a homogeneous antibody population wherein the monoclonal antibody is comprised of amino acids (naturally occurring and non-naturally occurring) that are involved in the selective binding of an antigen. Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • the term "monoclonal antibody” encompasses not only intact monoclonal antibodies and full-length monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab') 2 , Fv), single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity and the ability to bind to an antigen. It is not intended to be limited as regards to the source of the antibody or the manner in which it is made (e.g., by hybridoma, phage selection, recombinant expression, transgenic animals, etc.).
  • variable region of an antibody refers to the variable region of the antibody light ⁇ ; chain or the variable region of the antibody heavy chain, either alone or in combination.
  • the variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions.
  • the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies.
  • CDRs complementarity determining regions
  • a CDR may refer to CDRs defined by either approach or by a combination of both approaches.
  • a "constant region" of an antibody refers to the constant region of the antibody light chain or the constant region of the antibody heavy chain, either alone or in combination.
  • Humanized antibodies refer to a molecule having an antigen binding site substantially derived from an immunoglobulin from a non-human species and the remaining immunoglobulin structure of the molecule based upon the structure and/or sequence of a human immunoglobulin.
  • the antigen binding site may comprise either complete variable domains fused onto constant domains or only the complementarity determining regions (CDRs) grafted onto appropriate framework regions in the variable domains.
  • Antigen binding sites may be wild type or modified by one or more amino acid substitutions; e.g., modified to resemble human immunoglobulin more closely.
  • Some forms of humanized antibodies preserve all CDR sequences (for example, a humanized mouse antibody which contains all six CDRs from the mouse antibodies).
  • CDRs one, two, three, four, five, six
  • CDRs derived from
  • CDRs “derived from” one or more CDRs of the original antibody.
  • “Chimeric antibodies” refers to those antibodies wherein one portion of each of the amino acid sequences of heavy and light chains is homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular class, while the remaining segment of the chains is homologous to corresponding sequences in another.
  • variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals, while the constant portions are homologous to the sequences in antibodies derived from another.
  • variable regions can conveniently be derived from presently known sources using readily available hybridomas or B cells from non human host organisms in combination with constant regions derived from, for example, human cell preparations. While the variable region has the advantage of ease of preparation, and the specificity is not affected by its source, the constant region being human, is less likely to elicit an immune response from a human subject when the antibodies are injected than would the constant region from a non-human source.
  • the definition is not limited to this particular example.
  • polypeptide oligopeptide
  • peptide protein
  • polymers of amino acids of any length may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • the polypeptides of this invention are based upon an antibody, the polypeptides can occur as single chains or associated chains.
  • nucleic acid refers to deoxyribonucleic acid (DNA) and/or ribonucleic acid (RNA) in any form, including inter alia, single-stranded, duplex, triplex, linear and circular forms. It also includes polynucleotides, oligonucleotides, chimeras of nucleic acids and analogues thereof.
  • the nucleic acids described herein can be composed of the well-known deoxyribonucleotides and ribonucleotides composed of the bases adenosine, cytosine, guanine, thymidine, and uridine, or may be composed of analogues or derivatives of these bases.
  • oligonucleotide derivatives with nonconventional phosphodiester backbones are also included herein, such as phosphotriester, polynucleopeptides (PNA), methylphosphonate, phosphorothioate, polynucleotides primers, locked nucleic acid (LNA) and the like.
  • PNA polynucleopeptides
  • LNA locked nucleic acid
  • a "host cell” includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts.
  • Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation.
  • a host cell includes cells transfected in vivo with a polynucleotide(s) of this invention.
  • “treatment” or “treating” is an approach for obtaining beneficial or desired results including and preferably clinical results.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) cancerous cells, reducing metastasis of cancerous cells found in cancers, shrinking the size of the tumor, decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals.
  • an "effective amount" of an antibody, drug, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results including clinical results such as shrinking the size of the tumor (in the cancer context, for example, breast or liver cancer), retardation of cancerous cell growth, decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival of individuals.
  • An effective amount can be administered in one or more administrations.
  • an effective amount of drug, compound, or pharmaceutical composition is an amount sufficient to reduce the proliferation of (or destroy) cancerous cells and to reduce and/or delay the development, or growth, of metastases of cancerous cells, either directly or indirectly.
  • an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pha ⁇ naceutical composition.
  • an "effective amount" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • a "biological sample” encompasses a variety of sample types obtained from an individual and can be used in a diagnostic or monitoring assay.
  • the definition encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom, and the progeny thereof.
  • the definition also includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as proteins or polynucleotides, or embedding in a semi-solid or solid matrix for sectioning purposes.
  • the term "biological sample” encompasses a clinical sample, and also includes cells in culture, cell supernatants, cell lysates, serum, plasma, biological fluid, and tissue samples.
  • the present invention provides antibodies or polypeptides that specifically bind to SM5-1 antigen.
  • the antibody is a monoclonal antibody.
  • the antibody is a human, a humanized, or a chimeric antibody.
  • the invention is an antibody or a polypeptide that specifically binds to SM5-1 antigen, wherein the variable region of heavy chain of the antibody comprises the amino acid sequences 31-35, 50-66 and 99-108 set forth in SEQ ID NO:9, and/or the variable region of light chain of the antibody comprises the amino acid sequences 24-40, 56-62 and 95-102 set forth in SEQ ID NO: 10.
  • the heavy chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO:9. In other embodiments, the light chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO: 10. In other embodiments, the heavy chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO:9, and the light chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO: 10. In some embodiments, the antibody is huSM5-l shown in Table 1.
  • the invention is an antibody (e.g., a monoclonal antibody) or a polypeptide, wherein the heavy chain variable region of the antibody comprises the amino acid sequences 31-35, 50-66 and 99-108 set forth in SEQ ID NO:3, and/or the light chain variable region of the antibody comprises the amino acid sequences 24-40, 56-62 and 95-102 set forth in SEQ ID NO:4.
  • the heavy chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO:3.
  • the light chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO:4.
  • the heavy chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO:3, and the light chain variable region of the antibody comprises the amino acid sequence set forth in SEQ ID NO:4.
  • the antibody is a humanized antibody.
  • the heavy chain variable region of the humanized antibody comprises the amino acid sequence set forth in SEQ ID NO: 1
  • the light chain variable region of the humanized antibody comprises the amino acid sequence set forth in SEQ ID NO:2.
  • the antibody is a chimeric antibody, wherein the variable region of heavy chain of said chimeric antibody comprises the amino acid sequences set forth in SEQ ID NO:3. In other embodiments, the antibody is a chimeric antibody, wherein the variable region of light chain of said chimeric antibody comprises the amino acid sequences set forth in SEQ ID NO:4. In other embodiments, the antibody is a chimeric antibody, wherein the variable region of heavy chain of said chimeric antibody comprises the amino acid sequences set forth in SEQ ID NO:3 and the variable region of light chain of said chimeric antibody comprises the amino acid sequences set forth in SEQ ID NO:4.
  • the invention also provides antibodies (e.g., a monoclonal antibody) and polypeptides that competitively inhibit the immunospecific binding of any of SM5-1 specific monoclonal antibody described herein to a SM5-1 target antigen.
  • Competition assays can be used to determine whether two antibodies bind the same epitope by recognizing identical or sterically overlapping epitopes. Competition assays are known in the art. Typically, antigen is immobilized on a multi-well plate and the ability of unlabeled antibodies to block the binding of labeled antibodies is measured. Common labels for such competition assays are radioactive labels or enzyme labels.
  • the present invention also encompasses various formulations of the antibodies describe above and equivalent antibodies or polypeptide fragments (e.g., Fab, Fab', F(ab') 2 , Fv, Fc, etc.), single chain (ScFv), a diabody, a multi-specific antibody formed from antibody fragments, mutants thereof, fusion proteins comprising an antibody portion, and any other modified configuration of the antibodies that comprises a SM5-1 antigen recognition site of the required specificity.
  • Fab fragment antigen binding
  • the host cell that produces the human antibody (huSM5-l) having sequences of the variable regions shown in Table 1 is deposited at having accessing number on .
  • the host cell that produces the humanized antibody (ReSM5-l) having sequences of the variable regions shown in Table 3 is deposited at having accessing number on .
  • This deposit was made under the provisions of the Budapest Treaty on the international Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder. Accordingly, the invention provides antibodies, antibody fragments, and polypeptides derived from antibodies produced by the host cells described herein.
  • the invention also provides any of the following, or compositions (including pharmaceutical compositions) comprising any of the following: (a) antibody huSM5-l (variable regions shown in Table 1 , or produced by the host cell having an accessing number or progeny thereof); (b) a fragment or a region of the antibody huSM5-l ; (c) a heavy chain of the antibody huSM5-l; (d) a light chain of the antibody huSM5-l; (e) one or more variable region(s) from a light chain and/or a heavy chain of the antibody huSM5-l ; (f) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody huSM5-l ; (g) three CDRs from the light chain of antibody huSM5-l; (h) three CDRs from the heavy chain of antibody huSM5-l; (i) three CDRs from the light chain and three CDR
  • the invention also provides any of the following, or compositions (including pharmaceutical compositions) comprising any of the following: (a) antibody mSM5-l (variable regions are shown in Table 2, or produced by the host cell having an ATCC Designition No. HB- 12588 or progeny thereof); (b) a fragment or a region of the antibody mSM5-l; (c) one or more variable region(s) from a light chain and/or a heavy chain of the antibody mSM5-l; (d) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody mSM5-l ; (e) three CDRs from the light chain of antibody mSM5-l; (f) three CDRs from the heavy chain of antibody mSM5-l ; (g) three CDRs from the light chain and three CDRs from the heavy chain, of antibody mSM5-l; and (h) an antibody comprising any one of (b) through (g).
  • the invention also provides any of the following, or compositions (including pharmaceutical compositions) comprising any of the following: (a) antibody ReSM5-l (variable regions are shown in Table 3, or produced by the host cell having an accessing number or progeny thereof); (b) a fragment or a region of the antibody ReSM5-l; (c) one or more variable region(s) from a light chain and/or a heavy chain of the antibody ReSM5-l ; (d) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody ReSM5-l; (e) three CDRs from the light chain of antibody ReSM5-l; (f) three CDRs from the heavy chain of antibody ReSM5-l; (g) three CDRs from the light chain and three CDRs from the heavy chain, of antibody ReSM5-l; and (h) an antibody comprising any one of (b) through (g). [0071] It is understood that in some embodiments, the CDR can be
  • the invention provides an antibody which comprises at least one CDR that is substantially homologous to at least one CDR, at least two, at least three, at least four, at least five CDRs of huSM5-l, mSM5-l, or ReSM5-l (or, in some embodiments substantially homologous to all 6 CDRs of huSM5-l, mSM5-l, or ReSM5-l, or derived from huSM5-l, mSM5-l, or ReSM5-l).
  • antibodies which have at least two, three, four, five, or six CDR(s) that are substantially homologous to at least two, three, four, five or six CDRs of huSM5-l, mSM5-l, or ReSM5-l, or derived from huSM5-l, mSM5-l, or ReSM5-l. It is understood that, for purposes of this invention, binding specificity and/or overall activity (which may be in terms of treating cancer (e.g., melanoma, breast cancer, and hepatocellular carcinoma) is generally retained, although the extent of activity may vary compared to huSM5-l, mSM5-l, or ReSM5-l (may be greater or lesser)).
  • cancer e.g., melanoma, breast cancer, and hepatocellular carcinoma
  • the invention also provides a polypeptide (which may or may not be an antibody) which comprises an amino acid sequence that has any of the following: at least 5 contiguous amino acids, at least 8 contiguous amino acids, at least about 10 contiguous amino acids, at least about 15 contiguous amino acids, at least about 20 contiguous amino acids, at least about 25 contiguous amino acids, at least about 30 contiguous amino acids of a variable sequence of the antibody described herein (such huSM5-l, mSM5-l, and ReSM5-l).
  • the invention also provides methods of making any of these antibodies or polypeptides.
  • the antibodies of this invention can be made by procedures known in the art, some of which are illustrated in the Examples.
  • the method comprises growing a recombinant cell containing the nucleic acid encoding any of the antibody described herein or a fragment thereof (such as nucleic acid encoding huSM5-l shown in Table 1, and ReSM5-l shown in Table 3) such that the encoded antibody or a fragment thereof is expressed, and recovering the expressed antibody or a fragment thereof.
  • the method further comprises isolating and/or purifying the recovered antibody or a fragment thereof.
  • the polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis. Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available.
  • Monoclonal antibodies may be prepared using hybridoma methods, such as those described by Kohler and Milstein, 1975, Nature 256:495.
  • a hybridoma method a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent.
  • the lymphocytes may be immunized in vitro.
  • the antibodies or fragments of the invention may also be made by recombinant DNA methods, such as those described in U.S. Patent No. 4,816,567.
  • DNA encoding the antibodies or fragments is isolated and sequenced using conventional procedures, such as by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibodies.
  • the DNA for example, SEQ ID NO:5 and SEQ ID NO:6
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of the antibodies in the recombinant host cells.
  • Vectors (including expression vectors) and host cells are further described herein.
  • the invention includes modifications to antibodies described herein, including functionally equivalent antibodies which do not significantly affect their properties and variants which have enhanced or decreased activity. Modification of potypeptides is routine practice in the art and need not be described in detail herein. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or use of chemical analogs.
  • Amino acid residues which can be conservatively substituted for one another include but are not limited to: glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid; serine/threonine; lysine/arginine; and phenylalanine/tyrosine.
  • These polypeptides also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation.
  • the amino acid substitutions would be conservative, i.e., the substituted amino acid would possess similar chemical properties as that of the original amino acid.
  • conservative substitutions are known in the art, and examples have been provided above.
  • Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the variable region. Changes in the variable region can alter binding affinity and/or specificity. Other methods of modification include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay.
  • the invention also encompasses fusion proteins comprising one or more fragments or regions from the antibodies of this invention.
  • the fusion polypeptide contains a light chain variable region and/or a heavy chain variable region set forth in SEQ ID NO:2 and/or SEQ ID NO: 1. In other embodiments, the fusion polypeptide contains a light chain variable region and/or a heavy chain variable region set forth in SEQ ID NO: 10 and/or SEQ ID NO:9.
  • a fusion protein contains one or more antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region. Exemplary heterologous sequences include, but are not limited to a "tag" such as a FLAG tag or a 6His tag. Tags are well known in the art.
  • antibodies or fragments thereof disclosed herein may be used to make anti-tumor bifunctional fusion proteins, such as chimeric proteins as described in co-pending U.S. Application Serial No. (Attorney Docket No. 54906-2000200; Title:
  • a fusion polypeptide can be created by methods known in the art, for example, synthetically or recombinantly.
  • the fusion proteins of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, although they may also be prepared by other means known in the art, including, for example, chemical synthesis.
  • the chimeric antibody of the invention are provided in which the heavy and/or light chains are fusion proteins.
  • the constant domain of the chains is from one particular species and/or class, and the variable domains are from a different species and/or class.
  • a chimeric antibody in some embodiments is one in which the constant regions are derived from human origin, and the variable regions are homologous or derived from a murine antibody (for example, SEQ ID NO:3 and SEQ ID NO:4).
  • an antibody with a humanized variable region in which (in some embodiments) the CDR regions comprise murine amino acid sequences, while the framework regions are derived from human sequences.
  • humanized antibodies are known in the art and described herein. Also embodied are functional fragments of chimeras.
  • An example is a humanized Fab fragment, which contains a human hinge region, a human first constant region, a human kappa light or heavy chain constant region, and the variable region of light and/or heavy chain from a mouse antibody (for example, SEQ ID NO:3 and SEQ ID NO:4).
  • the humanized Fab fragments can in turn be made to form Fab dimers.
  • the fusion proteins and chimeras of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, althougli they may also be prepared by other means known in the art, including, for example, chemical synthesis. See, for example, U.S. Pat. Nos. 5,807,715; 4,816,567; and 6,331,415.
  • the invention also encompasses humanized antibodies.
  • the polynucleotide sequence of an antibody (for example, SEQ ID NO: 7 and SEQ ID NO: 8) or other equivalent > antibodies may be used for genetic manipulation to generate a "humanized” antibody, or to improve the affinity, or other characteristics of the antibody.
  • the general principle in humanizing an antibody involves retaining the basic sequence of the antigen-binding portion of the antibody, while swapping the non-human remainder of the antibody with human antibody sequences. There are four general steps to humanize a monoclonal antibody.
  • the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. See, for example, U.S. Patent Nos. 5,997,867 and 5,866,692.
  • a number of "humanized" antibody molecules comprising an antigen-binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent or modified rodent V regions and their associated complementarity determining regions (CDRs) fused to human constant domains.
  • CDRs complementarity determining regions
  • rodent CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody constant domain See, for example, Riechmann et al. Nature 332:323-327 (1988), Verhoeyen et al. Science 239:1534-1536 (1988), and Jones et al. Nature 321 :522-525 ( 1986).
  • Another reference describes rodent CDRs supported by recombinantly veneered rodent framework regions. See, for example, European Patent Publication No. 519,596. These "humanized" molecules are designed to minimize unwanted immunological response toward rodent anti-human antibody molecules which limits the duration and effectiveness of therapeutic applications of those moieties in human recipients.
  • antibodies may be made recombinantly by phage display technology. See, for example, U.S. Patent Nos. 5,565,332; 5,580,717; 5,733,743 and 6,265,150; and Winter et al, Annu. Rev. Immunol. 12:433-455 (1994), and Example 2.
  • the phage display technology McCafferty et ah, Nature 348:552-553 (1990)
  • V immunoglobulin variable
  • antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as Ml 3 or fd, and displayed as functional antibody fragments on the surface of the phage particle.
  • a filamentous bacteriophage such as Ml 3 or fd
  • the filamentous particle contains a single-stranded DNA copy of the phage genome
  • selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage mimics some of the properties of the B cell.
  • Phage display can be performed in a variety of formats; for review see, e.g., Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3, 564-571 (1993).
  • V-gene segments can be used for phage display.
  • Clackson et al. Nature 352:624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice.
  • a repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Mark et al., J. Mol. Biol. 222:581-597 (1991), or Griffith et al., EMBOJ. 12:725-734 (1993).
  • This invention also provides antibodies or polypeptides described herein conjugated (for example, linked) to a therapeutic agent, such as a radioactive moiety, a toxin (e.g., calicheamicin), or a chemotherapeutic molecule, a prodrug-activating enzyme which converts a prodrug to an active anti-cancer drug, or to liposomes or other vesicles containing chemotherapeutic compounds (or compositions comprising these antibodies or polypeptides).
  • a therapeutic agent such as a radioactive moiety, a toxin (e.g., calicheamicin), or a chemotherapeutic molecule, a prodrug-activating enzyme which converts a prodrug to an active anti-cancer drug, or to liposomes or other vesicles containing chemotherapeutic compounds (or compositions comprising these antibodies or polypeptides).
  • compositions when administered to an individual, can target these agents to a cancer cell expressing SM5-1 antigen recognized by the antibody or polypeptide(s) and thus can, for example, eliminate (or reduce the number of) cancerous cells and/or suppress proliferation and/or growth of cancerous cells.
  • conjugation generally refers to linking these components as described herein.
  • the linking (which is generally fixing these components in proximate association at least for administration) can be achieved in any number of ways, as described below.
  • a radioactive moiety or molecule of this invention includes any radioisotope which is effective in destroying a cancerous cell. Examples include, but not limited to, cobalt-60, 131 I, and X-rays. Additionally, naturally occurring radioactive elements such as uranium, radium, and thorium which typically represent mixtures of radioisotopes, are suitable examples of a radioactive molecule.
  • a toxin of the invention include, but not limited to, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • the antibodies or polypeptides of the invention can be conjugated (linked) to a radioactive moiety or molecule, a toxin, or other therapeutic agents, a prodrug-activating enzyme which converts a prodrug to an active anti-cancer drug, or to liposomes or other vesicles containing therapeutic agents covalently or non-covalently, directly or indirectly.
  • the antibody may be linked to the radioactive molecule, the toxin, the therapeutic molecule, or a prodrug-activating enzyme at any location along the antibody so long as the antibody is able to bind its target antigen.
  • a toxin or a therapeutic agent may be coupled (e.g., covalently bonded) to a suitable monoclonal antibody either directly or indirectly (e.g., via a linker group, or, alternatively, via a linking molecule with appropriate attachment sites, such as a platform molecule as described in U.S. patent 5,552,391).
  • the toxin and therapeutic agent of the present invention can be coupled directly to the particular targeting proteins using methods known in the art. For example, a direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other.
  • a nucleophilic group such as an amino or sulfhydryl group
  • a carbonyl-containing group such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.
  • the antibody or polypeptide conjugates of the present invention may include a bifunctional linker which contains both a group capable of coupling to a toxic agent or therapeutic agent and a group capable of coupling to the antibody.
  • a linker can function as a spacer to distance an antibody from an agent in order to avoid interference with binding capabilities.
  • a linker can be cleavable or non-cleavable.
  • a linker can also serve to increase the chemical reactivity of a substituent on an agent or an antibody, and thus increase the coupling efficiency. An increase in chemical reactivity may also facilitate the use of agents, or functional groups on agents, which otherwise would not be possible.
  • the bifunctional linker can be coupled to the antibody by means which are known in the art.
  • a linker containing an active ester moiety such as an N-hydroxysuccinimide ester
  • a linker containing a nucleophilic amine or hydrazine residue can be coupled to aldehyde groups produced by glycolytic oxidation of antibody carbohydrate residues.
  • the linker can be indirectly coupled to the antibody by means of an intermediate carrier such as an aminodextran.
  • the modified linkage is via either lysine, carbohydrate, or an intermediate carrier.
  • the linker is coupled site-selectively to free thiol residues in the protein.
  • Moieties which are suitable for selective coupling to thiol groups on proteins are well known in the art. Examples include disulfide compounds, ⁇ -halocarbonyl and ⁇ -halocarboxyl compounds, and maleimides.
  • a nucleophilic amine function is present in the same molecule as an ⁇ -halo carbonyl or carboxyl group the potential exists for cyclization to occur via intramolecular alkylation of the amine.
  • Methods to prevent this problem are well known to one of ordinary skill in the art, for example by preparation of molecules in which the amine and ⁇ -halo functions are separated by inflexible groups, such as aryl groups or trans-alkenes, that make the undesired cyclization stereochemically disfavored. See, for example, U.S. Patent No. 6,441,163 for preparation of conjugates of maytansinoids and antibody via a disulfide moiety.
  • An antibody (or polypeptide) of this invention may be conjugated (linked) to a radioactive moiety or molecule by any method known to the art.
  • a radioactive moiety or molecule for a discussion of methods for radiolabeling antibody see "Cancer Therapy with Monoclonal AntibodiesT", D. M. Goldenberg ed. (CRC Press, Boca Raton, 1995).
  • the antibodies (or polypeptides) of the invention may be linked to an agent (including a prodrug-activating enzyme) which converts a prodrug to an active anti-cancer drug.
  • the antibodies (or polypeptides) of this invention may be used in Antibody Dependent Enzyme Mediated Prodrug Therapy (ADEPT) by conjugating (linking) the antibody to a prodrug-activating enzyme which converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see WO81/01145) to an active-cancer drug. See, for example, WO 88/07378 and U.S. Patent No. 4,975,278.
  • ADEPT Antibody Dependent Enzyme Mediated Prodrug Therapy
  • an antibody (or polypeptide) of this invention may be linked to a labeling agent (alternatively termed "label") such as a fluorescent molecule, a radioactive molecule or any others labels known in the art.
  • label such as a fluorescent molecule, a radioactive molecule or any others labels known in the art.
  • Labels are known in the art which generally provide (either directly or indirectly) a signal.
  • compositions including pharmaceutical compositions, comprising effective amount of antibodies (including antibody conjugates) and polypeptides that bind to SM5-1 antigen, and polynucleotides comprising sequences encoding antibodies, polypeptides described herein.
  • compositions comprise one or more antibodies that bind to SM5-1 antigen, and/or one or more polynucleotides comprising sequences encoding one or more antibodies that bind to SM5-1.
  • compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • This invention also encompasses a combination comprising an effective amount of any of the antibodies described herein and an effective amount of an anti-neoplasm agent.
  • the anti-neoplasm agent can be an agent that treats melanoma, breast cancer, or hepatocellular carcinoma.
  • the invention also provides an isolated SM5-1 target antigen, which comprises a protein that specifically binds to the antibodies described herein.
  • the isolated SM5-1 antigen is a human antigen.
  • the isolated SM5-1 antigen is a glycosylated protein.
  • the isolated SM5-1 antigen is a non-glycosylated protein.
  • the isolated SM5-1 antigen is a fragment of A230 or A 180 described herein.
  • the isolated SM5-1 antigen is isolated from a melanoma, breast cancer and/or hepatocellular carcinoma cell.
  • the invention also provides isolated polynucleotides encoding the antibodies of the invention (for example, an antibody comprising the polypeptide sequences of the light chain and heavy chain variable regions set forth in SEQ ID NO:2 and SEQ ID NO:l, and an antibody comprising the polypeptide sequences of the light chain and heavy chain variable regions set forth in SEQ ID NO: 10 and SEQ ID NO:9), and vectors and host cells comprising the polynucleotide.
  • the invention provides polynucleotides encoding any of the polypeptides (including antibody fragments) described herein.
  • the invention provides compositions (such as a pharmaceutical compositions) comprising any of the polynucleotides of the invention.
  • the polynucleotides comprises nucleotide sequences set forth in SEQ ID NO: 11 or SEQ ID NO:
  • the polynucleotides comprises nucleotide sequences set forth in
  • the composition comprises an expression vector comprising a polynucleotide encoding the antibody as described herein. In still other embodiments, the composition comprises either or both of the polynucleotides described herein. Expression vectors, and administration of polynucleotide compositions are further described herein.
  • the invention provides a method of making any of the polynucleotides described herein.
  • Polynucleotides complementary to any such sequences are also encompassed by the present invention.
  • Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules.
  • RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence.
  • Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule.
  • the effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein.
  • Variants preferably exhibit at least about 70% identity, more preferably at least about 80% identity and most preferably at least about 90% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M.O. (1978) A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol.
  • the "percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e. gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e.
  • Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof. Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence).
  • Suitable “moderately stringent conditions” include prewashing in a solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65°C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0. 1 % SDS.
  • highly stringent conditions or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50°C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42°C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/ml), 0.1% SDS, and 10% dextran s
  • formamide for example, 50% (v
  • the resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • the polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
  • a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein.
  • Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained witliin the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome.
  • the polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al. (1989).
  • PCR allows reproduction of DNA sequences.
  • PCR technology is well known in the art and is described in U.S. Patent Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston (1994).
  • RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., (1989), for example.
  • Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may var ⁇ ' according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector.
  • Suitable examples include plasmids and bacterial viruses, e.g., pUCl 8, ⁇ UC19, pUC57, pMG18-3K, Bluescript (e.g., pBS SK+, pBS SK-) and its derivatives, mpl8, mpl9, pBR322, pMB9, ColEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
  • Bluescript e.g., pBS SK+, pBS SK-
  • mpl8 mpl9
  • pBR322 pMB9
  • ColEl pCRl
  • RP4 phage DNAs
  • shuttle vectors such as pSA3 and pAT28.
  • shuttle vectors such as pSA3 and pAT28.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, and cosmids. Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • the vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus).
  • electroporation employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances
  • microprojectile bombardment e.g., where the vector is an infectious agent such as vaccinia virus.
  • infection e.g., where the vector is an infectious agent such as vaccinia virus.
  • the choice of introducing vectors or polynucleotides will often depend on features of the host cell.
  • the invention also provides host cells comprising any of the polynucleotides described herein. Any host cells* capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest.
  • mammalian host cells include but not limited to COS, HeLa, and CHO cells. Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe; or K. lactis).
  • the host cells express the cDNAs at a level of about 5 fold higher, more preferably 10 fold liigher, even more preferably 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells.
  • Screening the host cells for a specific antibody binding to SM5-1 target antigen is effected by an immunoassay or FACS.
  • a cell overexpressing the antibody or protein of interest can be identified.
  • the invention provides methods for assaying for human SM5-1 target antigen in a sample, which method comprises: a) obtaining a sample from a subject to be tested; b) contacting said sample with an antibody specific for SM5- 1 target antigen; and c) assessing binding between said human SM5-1 target antigen, if present in said sample, to said antibody to determine presence, absence and/or amount of said human SM5-1 target antigen in said sample.
  • Antibodies specific for SM5-1 target antigen described herein may be used to identify the presence or absence of cancerous cells, including but not limited to, melanoma, breast cancer, and hepatocellular carcinoma for purposes of diagnosis. Detection generally involves contacting cells with an antibody specific for SM5-1 target antigen described herein that binds to the antigen and the formation of a complex between the antigen and the antibody. The formation of such a complex can be in vitro or in vivo.
  • the invention provides methods of aiding diagnosis of cancer using any antibodies or polypeptides described herein.
  • methods for "aiding diagnosis” means that these methods assist in making a clinical determination regarding the classification, or nature, of cancer, and may or may not be conclusive with respect to the definitive diagnosis.
  • a method of aiding diagnosis of cancer can comprise the step of detecting the level of SM5-1 target antigen in a biological sample from the individual and/or determining the level of SM5-1 target antigen expression in the sample.
  • One method of using the antibodies for diagnosis is in vivo tumor imaging by linking the antibody to a labeling moiety (e.g., a fluorescent agent, a radioactive or radioopaque agent), administering the antibody to the individual and using an x-ray or other imaging machine to visualize the localization of the labeled antibody at the surface of cancer cells expressing the antigen.
  • a labeling moiety e.g., a fluorescent agent, a radioactive or radioopaque agent
  • the antibody is administered at a concentration that promotes binding at physiological conditions.
  • Labeling moieties are known in the art.
  • the cancerous cells are removed and the tissue prepared for immunohistochemistry by methods well known in the art (e.g., embedding in a freezing compound, freezing and sectioning, with or without fixation; fixation and paraffin embedding with or without various methods of antigen retrieval and counterstaining).
  • the antibodies may also be used to identify cancerous cells at different stages of development.
  • the antibodies may also be used to determine which individuals' tumors express the antigen on their surface at a pre-determined level and are thus candidates for immunotherapy using antibodies directed against said antigen.
  • Antibodies (or polypeptides) recognizing the antigen may also be used to create diagnostic immunoassays for detecting antigen released or secreted from living or dying cancer cells in bodily fluids, including but not limited to, blood, saliva, urine, pulmonary fluid, or ascites fluid. Methods of using antibodies of the invention for diagnostic purposes is useful both before and after any form of anti-cancer treatment, e.g., chemotherapy or radiation therapy, to determine which tumors are most likely to respond to a given treatment, prognosis for individual with cancer, tumor subtype or origin of metastatic disease, and progression of the disease or response to treatment.
  • any form of anti-cancer treatment e.g., chemotherapy or radiation therapy
  • the present invention also provides a method for treating neoplasm in a mammal, which method comprises administering to a mammal to which such treatment is needed or desirable, an effective amount of an antibody specific to SM5-1 antigen described herein (for example, huSM5-l and ReSM5-l).
  • the antibodies described in this invention may be used for therapeutic purposes in individuals with cancer in a variety of tissues, including but not limited to, melanoma, breast cancer, and hepatocellular carcinoma. In some embodiments, the antibody is used for passive immunity of cancer patients.
  • the antibody administered exerts its anti-neoplasm effect via antibody dependent cell mediated cytotoxicity (ADCC) and/or complement dependent cell mediated cytotoxicity (CDC).
  • ADCC antibody dependent cell mediated cytotoxicity
  • CDC complement dependent cell mediated cytotoxicity
  • the antibody is used for treating neoplasm in human.
  • the invention also provides a method for treating neoplasm in a mammal comprising administering to a mammal to which such treatment is needed or desirable, an effective amount of a combination which comprises an effective amount of an antibody specific to SM5-1 described herein and an effective amount of an anti -neoplasm agent.
  • the anti-neoplasm agent is an agent that treats melanoma, breast cancer, or hepatocellular carcinoma.
  • the invention also provides a method for inducing caspase-10 mediated apoptosis in cell, which method comprises administering to a cell to which such induction is needed or desirable, an effective amount of an antibody specific to SM5-1 described herein.
  • the cell is a mammalian cell. In some embodiments, the cell is contained in a mammal.
  • anti-SM5-l antibodies described herein and equivalent antibodies or fragments e.g., Fab, Fab', F(ab') 2 , Fv, Fc, etc.
  • Fab, Fab', F(ab') 2 , Fv, Fc, etc. such as chimeric antibodies, single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion, humanized antibodies, antibodies conjugated to a toxin or a radioactive isotope and any other modified configuration that comprises the required specificity, thereof
  • the antibodies or various formulations thereof may be administered neat.
  • the antibodies or various formulations (including any composition embodiment described herein) thereof and a pharmaceutically acceptable excipient are administered, and may be in various formulations.
  • excipients are known in the art, and are relatively inert substances that facilitate administration of a pharmacologically effective substance.
  • an excipient can give form or consistency, or act as a diluent.
  • Suitable excipients include but are not limited to stabilizing agents, wetting and emulsifying agents, salts for varying osmolarity, encapsulating agents, buffers, and skin penetration enhancers.
  • Excipients as well as formulations for parenteral and nonparenteral drug delivery are set forth in Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing (2000).
  • these agents are formulated for administration by injection (e.g., intraperitoneally, intravenously, subcutaneously, intramuscularly, etc.), although other forms of administration (e.g., oral, mucosal, etc) can be also used.
  • antibody and equivalents thereof are preferably combined with pharmaceutically acceptable vehicles such as saline, Ringer's solution, dextrose solution, and the like.
  • the particular dosage regimen i.e., dose, timing and repetition, will depend on the particular individual and that individual's medical history.
  • a dose of at least about 100 ug/kg body weight, at least about 250 ug/kg body weight, at least about 750 ug/kg body weight, at least about 3 mg /kg body weight, at least about 5 mg /kg body weight, at least about 10 mg/kg body weight is administered.
  • a dose of 1-200 mg/per day may be administered.
  • the antibody may be injected into the tumor in situ (Irie et al., Proc. Natl. Acad. Sci. USA 83:8694-8698 (1986)) or administered systematically (especially for metastasis).
  • Empirical considerations such as the half-life, generally will contribute to the determination of the dosage.
  • Antibodies which are compatible with the human immune system such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's immune system. Frequency of administration may be determined and adjusted over the course of therapy, and is based on reducing the number of cancerous cells, maintaining the reduction of cancerous cells, reducing the proliferation of cancerous cells, or delaying the development of metastasis.
  • the presence of cancerous cells can be identified by any number of methods known to one of skill in the art or discussed herein (e.g., detection by immunohistochemistry or flow cytometry of biopsies or biological samples).
  • sustained continuous release formulations of antibodies may be appropriate.
  • Various formulations and devices for achieving sustained release are known in the art.
  • dosages for antibodies may be determined empirically in individuals who have been given one or more administration(s). Individuals are given incremental dosages of the antibodies.
  • a marker of the specific cancer disease state can be followed. These include direct measurements of tumor size via palpation or visual observation, indirect measurement of tumor size by x-ray or other imaging techniques; an improvement as assessed by direct tumor biopsy and microscopic examination of the tumor sample; the measurement of an indirect tumor marker, a decrease in pain or paralysis; improved speech, vision, breathing or other disability associated with the tumor; increased appetite; or an increase in quality of life as measured by accepted tests or prolongation of survival. It will be apparent to one of skill in the art that the dosage will vary depending on the individual, the type of cancer, the stage of cancer, whether the cancer has begun to metastasize to other location in the individual, and the past and concurrent treatments being used.
  • compositions include suitable delivery forms known in the art including, but not limited to, carriers such as liposomes. See, for example, Mahato et al. (1997) Pharm. Res. 14:853-859.
  • Liposomal preparations include, but are not limited to, cytofectins, multilamellar vesicles and unilamellar vesicles.
  • more than one antibody or other agent may be present.
  • the antibodies can be monoclonal or polyclonal.
  • Such compositions may contain at least one, at least two, at least three, at least four, at least five different antibodies that are reactive against carcinomas, adenocarcinomas, sarcomas, or adenosarcomas.
  • a mixture of antibodies, as they are often denoted in the art, may be particularly useful in treating a broader range of population of individuals.
  • kits comprising antibodies for use in detection and/or therapy.
  • the kit comprises any antibodies described herein.
  • the kits of tins invention are in suitable packaging, and may optionally provide additional components such as, buffers and instructions for use of the antibody in any of the methods described herein.
  • the kits may be used for any of the methods described herein, including, for example, to treat an individual with a neoplasm.
  • the kit comprises an effective amount of an antibody described herein, and an instruction means for administering said antibody.
  • the invention provides a kit for assaying for human SM5-1 target antigen in a sample, which kit comprises an antibody described herein and means for assessing binding between the human SM5-1 target antigen, if present in the sample, to the antibody to determine presence, absence, and/or amount of the target antigen in the sample.
  • the kit further comprises an instruction means for performing the assay.
  • RNAs were extracted from hepatocellular carcinoma cell line QYC with Trizol reagents. Then mRNAs were isolated and cDNA was synthesized as described (Marken JS. PNAS, 1992, 89:3503-3507). The cDNA was inserted into mammalian transient expression vector pCDMS (from Invitrogen.) after ligation of the non-self-complementary BstXI adaptors and transformed into the E.coli. MC1061/P3 (from Invitrogen) by electroporation to construct the cDNA library.
  • pCDMS from Invitrogen.
  • MC1061/P3 from Invitrogen
  • COS-7 (Invitrogen) cells were transfected with the above acquired cDNA library using Lipofection method. After twelve hours, the cells were digested and plated in new flasks. Seventy-two hours after transfection, the cells were harvested and re-suspended in PBS/0.5 mM EDTA/5%FBS containing mouse monoclonal antibody specific for SM5-1 antigen (designated as mSM5-l, the hybridoma producing this antibody was deposited in the American Type Culture Collection (ATCC) on October 10, 1998, with a Patent Deposit Designation of HB-12588).
  • ATCC American Type Culture Collection
  • Plasmid DNA was recovered from the adherent cells by Hirt method (Hirt B. J Mol Biol, 1967,26:365-369). The recovered plasmid DNA was transformed into E.coli MC1061/p3 cells and the transformed E.coli were used to prepare a second cDNA library.
  • the final harvested plasmid acquired with Hirt method was transformed into E.coli. MC 1061/P3. Many clones were then randomly selected. Plasmid was extracted from these clones and used to transfect COS-7 cells by Lipofectin method. Twelve hours after transfection, the cells were digested with trypsin and plated on new plastic dishes. Seventy-two hours after transfection, mSM5-l was added into dish, and stained with FITC-labeled goat anti-mouse Ig secondary antibody. Positive clones were identified under fluorescent microscope. Finally, the plasmid cDNA was isolated from a single postive clone.
  • SM5-1 antigen The extracellur region of SM5-1 antigen was cloned into a mammalian expression vector and the construted vector was transfected into CHO cells for expression.
  • the extracellur region of SM5-1 antigen was purified by affinity chromatography (mouse SM5-1 antibody immobilized on Sepharose-4B) from the serum-free culture supernatant.
  • Example 2 Screening for variable region gene of a human anti-human SM5-1 antibody from a human antibody library
  • the human antibody library was constructed according to methods described by
  • Haidais CG et al J. Immunol. Methods., 2001, Nov 1; 257(1-2): 185-202), Griffiths, A.D. et al.
  • the recovered antibody library was added into 14 ml fresh LB media and cultured for
  • the bacteria were centrifuged at 12,000 rpm for 10 min. The supernatant was transferred to a sterile 50 ml centrifuge tube and stored for later use and the titer should be higher than 2xl ⁇ ".
  • Cell culture flask was coated with the purified antigen SM5-1 acquired in Example 1. No less than 3xl0 10 phage particles were added into the flask and incubated at 37°C for lh.
  • the flask was washed with 10 ml PBS containing 1% Tween-20 for 10 times.
  • the adherent particles were eluted by elution buffer and added into 1 ml TGI cells growing in logarithmic stage.
  • the cells were cultured in a shaker at 37°C for 16 h.
  • agar plate 1.5% agar plate to acquire single clones.
  • the clones were cultured on a deep-well 96-well plate, one well for each clone; totaling 960 clones (10 plates) were obtained.
  • the plates were centrifuged at 5000 rpm for 20 min, and the supernatant was transferred to a sterile deep-well plate and covered and stored at 4°C.
  • TMB substrate solution 100 ⁇ l TMB substrate solution were added into the well and the plates were incubated at room temperature without light for 5-20 min to develop the color. And 50ul stop solution was added into each well. The plates were read at 450 nm.
  • anti-SM5-l protein was isolated and purified , which is a human antibody against human SM5-1 antigen.
  • This human antibody (huSM5-l) was purified for affinity dete ⁇ nination and the positive clone with the highest affinity was selected for further research.
  • the amino acid sequence and the nucleotide sequence for the heavy chain variable region (SEQ ID NOS: 9 and 11) and the light chain variable region (SEQ ID NOS: 10 and 12) of this antibody are shown in Table 1 below.
  • VH heavy chain variable region gene
  • MDFQVQIFSFLLISASVIISRG (SEQ ID NO: 13), and the nucleotide sequence of mAb OKT3 signal peptide is ATGGATTTTCAGGTGCAGATTTTCAGCTTCCTGCT
  • VL light chain variable region gene
  • VL was excised by Hindlll and BsiWI digestion and then, inserted into the expression vector pMG18-3K at the position of Hindlll/ BsiWI.
  • CHOdhfr- cells Prior to transfection, CHOdhfr- cells were maintained in complete DMEM medium containing glycin, hypoxanthine and thymidine (GHT).
  • GHT hypoxanthine and thymidine
  • the expression vector described above was transfected into CHOdhfr- cells using Lipofectamine 2000 reagent (Invitrogen, Garlsbad,
  • the transfected cells were then selected in
  • GHT free DMEM medium containing stepwise increments in MTX level up to 1.0 M.
  • Drug resistant clones were picked and expanded for further analysis.
  • the culture supematants from cell clones were analyzed for antibody production by the sandwich ELISA which used goat anti-human IgG(Fc) (KPL) as capture antibody and goat anti-human kappa-HRP (KPL) as detector antibody.
  • Purified human IgGl/Kappa (Sigma) was used as a standard in the ELISA assay.
  • the clone producing the highest amount of antibody was selected and grown in serum-free medium.
  • the recombinant antibodies were purified by Protein A affinity chromatography from the serum-free culture supernatant.
  • the heavy and light variable region cDNAs of mSM5-l were cloned from hybridoma cells using 5'RACE system (Gibco BRL, Gaithersburg, MD) according to the manufacture's instruction.
  • the final PCR products were cloned into pGEM-T vector (Promega, Madison, WI) for sequence determination.
  • the nucleotide sequence and the deduced amino acid sequences of heavy (mSM5-l VH) and light (mSM5-l VL) variable region are shown in Table 2 below.
  • PKLLIY ASTRESGVPDRFTGSGSGTDFTLTISSVQAEDLAVYYCHQYF SSYTFGGGTKLEIKR mSM5-l heavy chain variable region nucleotide sequence (SEQ ID NO:7)
  • variable regions of the heavy and the light chain of mSM5-l shown above were used to construct mouse-human chimeric antibody.
  • the chimeric antibody expression vector was constmcted in an identical manner to huSM5-l described in Example 3.
  • CHOdhfr- cells Prior to transfection, CHOdhfr- cells were maintained in complete DMEM medium containing glycin, hypoxanthine and thymidine (GHT).
  • GGT hypoxanthine and thymidine
  • the expression vector pMGl 8-3 K containing heavy and light chain of chSM5-l was transfected into CHOdhfr- cells using Lipofectamine 2000 reagent (Invitrogen, Garlsbad, CA) according to the manufacture's instruction.
  • the transfected cells were then selected in GHT free DMEM medium containing stepwise increments in MTX level up to 1.0 M. Drug resistant clones were picked and expanded for further analysis.
  • the culture supematants from cell clones were analyzed for antibody production by the sandwich ELISA which used goat anti-human IgG(Fc) (KPL) as capture antibody and goat anti-human kappa-HRP (KPL) as detector antibody.
  • Purified human IgGl/Kappa (Sigma) was used as a standard in the ELISA assay.
  • the clone producing the highest amount of antibody was selected and grown in serum-free medium.
  • the recombinant antibodies were purified by Protein A affinity chromatography from the serum-free culture supernatant.
  • the V H of human antibody KOL was chosen as framework for the humanized heavy chain and the V L of human Bence-Jones protein REI was chosen for the humanized light chain.
  • the three CDRs from mSM5-l light chain or heavy chain were directly grafted into human antibody light chain or heavy chain framework regions to generate a humanized antibody genes.
  • the light and heavy variable region genes of humanized antibodies were synthesized by overlapping PCR method.
  • the expression vectors for humanized antibodies were constmcted in an identical manner to the chimeric antibody described above.
  • the three CDRs from mSM5-l light chain or heavy chain were directly grafted into human antibody light chain or heavy chain framework regions to generate humanized antibody genes.
  • the humanized V and V H were cloned into pMG18-3K expression vector and was expressed transiently in COS cells, yielding humanized version.
  • Humanized antibody in COS cell culture supernatant was quantitated by ELISA and the binding of this version to hepatocelluer carcinoma cell line QYC was determined by FCM.
  • the antigen binding activity assay indicated that this antibody bound poorly to human melanoma cells. This suggested that some human FR residues must be altered to reconstitute the full binding activity.
  • ReSM5-l displayed equivalent avidity as the murine SM5-1 or chimeric SM5-1 antibody.
  • ReSM5-l heavy chain variable region amino acid sequence SEO ID NO:l Gin Val Gin Leu Val Gin Ser Gly Gly Gly Val Val Gin Pro Gly Arg 1 5 10 15 Ser Leu Arg Leu Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr 20 25 30 Val Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp He 35 40 45 Gly Tyr lie Val Pro Tyr Asn Asp Gly Thr Lys Tyr Asn Glu Lys Phe 50 55 60 Lys Gly Arg Phe Thr lie Ser Ser Asp Lys Ser Lys Ser Thr Ala Phe 65 70 75 80 Leu Gin Met Asp Ser Leu Arg Pro Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95 Ala Arg Gly Ser Arg Tyr Asp Trp Tyr Leu Asp Tyr Trp Gly Gin Gly 100 105 110 Thr Pro Val Thr Val Ser Ser 115
  • ReSM5-l light chain variable region amino acid sequence (SEO ID NO:2) Asn lie Met Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15 Asp Arg Val Thr lie Thr Cys Lys Ser Ser Gin Ser Val Leu Tyr Ser 20 25 30 Ser Asn Gin Lys Asn Tyr Leu Ala Trp Tyr Gin Gin Thr Pro Gly Lys 35 40 45 Ala Pro Lys Leu Leu He Tyr Trp Ala Ser Thr Arg Glu Ser Gly Val 50 ' 55 60 Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Tyr Thr Phe Thr 65 70 75 80 lie Ser Ser Leu Gin Pro Glu Asp lie Ala Thr Tyr Tyr Cys His Gin 85 90 95 Tyr Phe Ser Ser Tyr Thr Phe Gly Gin Gly Thr Lys Leu Gin lie Thr 100 105 110 Arg
  • ReSM5-l heavy chain variable region nucleotide sequence (SEQ ID NO:5)
  • CHOdhfr- cells Prior to transfection, CHOdhfr- cells were maintained in complete DMEM medium containing glycin, hypoxanthine and thymidine (GHT). Appropriate expression vector was transfected into CHOdhfr- cells using Lipofectamine 2000 reagent (Invitrogen, Garlsbad, CA) according to the manufacture's instmction. The transfected cells were then selected in GHT free DMEM medium containing stepwise increments in MTX level up to 1.0 M. Drug resistant clones were picked and expanded for further analysis.
  • GHT glycin, hypoxanthine and thymidine
  • the culture supematants from cell clones were analyzed for antibody production by the sandwich ELISA which used goat anti-human IgG(Fc) (KPL) as capture antibody and goat anti-human kappa-HRP (KPL) as detector antibody.
  • KPL goat anti-human IgG(Fc)
  • KPL goat anti-human kappa-HRP
  • Purified human IgGl/Kappa Sigma was used as a standard in the ELISA assay.
  • the clone producing the highest amount of antibody was selected and grown in serum-free medium.
  • the recombinant antibody (ReSM5-l) was purified by Protein A affinity chromatography from the serum-free culture supernatant.
  • the melanoma cell line A2058 was expanded to 10 6 /ml in RPMI-1640 culture medium.
  • the cell suspension (20 ⁇ l) and different amount of huSM5-l (0, 1, 5, 10, 20, 50, 100 ⁇ l) were added into each well of the 96 well plate.
  • the antibody huSM5-l was diluted in RPMI-1640 medium contianing 10% FCS to reach a final concentration of 1 ⁇ g/ ⁇ l before it was added into the well.
  • RPMI-1640 medium contianing 10% FCS was added to each well to make a final volume of 500 ⁇ l in each well. Each condition was performed in triplicate. Cells were incubated at 37°C in a humidified incubator at 5% CO 2 for 24 h.
  • Viable cell count [0158] Viable cells in 0.2 ml cell suspension from each well were counted. The results are shown in Table 4. Viable cells are expressed as percentage of viable cells compared to the wells without addition of huSM5-l . As shown in Table 4, antibody huSM5-l significantly reduced number of viable A2058 cells in a dose dependent manner after 24 h incubation.
  • MTT solution 5 mg/ml was added into 0.2 ml cell suspension describe above at 1 : 10 dilution. The cells were then incubated at 37°C for 30 min and optical adsorption at 570 nm for each sample was read. As shown in Table 5, antibody huSM5-l treated cells had significant lower optical abso ⁇ tion at 570 nm compared to control, indicating viable cells were significantly reduced after treatment with antibody huSM5-l .
  • huSM5- 1 antibody can prevent proliferation of human melanoma cells effectively in vitro and may be used for treating melanoma.
  • SM5-1 antigen on cancer cells' surface were conducted using flow cytometry with huSM5-l antibody.
  • Cancer cells used in the study included breast cancer cell lines SK-BR-3, MDA-MB-231, BT-20-T, MDA-MB-468 and MCF-7; melanoma cell lines CRL-1872 and U10; hepatocellular carcinoma cell lines QYC, LYX and XJC.
  • SM5-1 antigen is not only expressed characteristically by melanoma cells but also over-expressed by breast cancer and hepatocellular carcinoma cells.
  • Cancer cells to be tested were incubated with appropriately diluted huSM5-l for 1 h, and then washed 5 times with PBS. Cells were incubated with FITC labeled goat-anti-human IgG (2 mg/ml, Jackson Immunoresearch Laboratories, West Grove, PA) and then fixed in PBS containing 1% formalin. Expression of the antigen in these cells were then analyzed using flow cytometry.
  • SM5-1 antigen was highly or moderately expressed in breast cancer cell lines (MDA-MB-231, MDA-MB-468, and MCF-7), melanoma cell lines
  • hepatocellular carcinoma cell lines (CRL-1872), and hepatocellular carcinoma cell lines (QYC and LYX). But the antigen had a lower level expression in hepatocellular carcinoma cell line XJC, breast cancer cell lines
  • huSM5-l antibody induces caspase 10-related apoptosis
  • caspase inhibitors were tested to observe the inhibitory rate of apoptosis.
  • the caspase inhibitors tested included common caspase inhibitor (Z-VAD-FMK), caspase- 1 inhibitor
  • inhibitory rates of these inhibitors were: 72% for pan-caspase inhibitor, 52% for caspase-10 inhibitor, 28% for caspase-6 inhibitor, 27% for caspase- 1 inhibitor, 17% for caspase-S inhibitor, 15% for caspase- 13 inhibitor, 14% for caspase-4 inhibitor, 5% for caspase-9 inhibitor, 1% for caspase-2 inhibitor, 1% for caspase-3 inhibitor.
  • pan caspase inhibitor had the highest inhibitory effect on huSM5-l induced apoptosis, and caspase 10 inhibitor also significantly inhibited the huSM5-l induced apoptosis, indicating that huSM5-l induced apoptosis was related to caspase-mediated pathway, and caspase-10 was one of the caspases which affected huSM5-l induced apoptosis mostly.
  • caspase-10 color comparing analysis kit was used to examine the increase of caspase-10 bioactivity in QYC and XJC cells.
  • Caspase-10 activity was examined with caspase-10 analysis kit (R&D, USA) according to manufacture's instmction. Cells were incubated with 50 ng/ml huSM5-l antibody for a certain period of time. Cells were then centrifuged and lysed in a lysis buffer (25 ul/10 6 cells) on ice for 10 min. The lysates were centrifuged, and the supernatant was transferred into new tubes and kept on ice.
  • the enzyme activity of caspase was examined on 96-well micro-titer plate. Each reaction included 50 ⁇ l supernatant from cell lysate, 50 ⁇ l 2x reaction buffer, 10 ⁇ l fresh DTT storage solution. In addition, 5 ul caspase-10 color-comparing substrate(AEVD-pNA) was added into the reaction. The plate was incubated at 37°C for 1-2 h and absorbance at 405 nm was then measured.
  • Caspase-10 activity (%) (B-CV(A-C) x 100% [0168] "A” represents OD value of supernatant from cell lysates without huSM5-l antibody treatment; “B” represents OD value of supernatant from cell lysate treated with huSM5-l; “C” represents OD value of negative control. Each test was done in triplicate.
  • Fig. 5 shows that the caspase-10 activity increased from 13% (48h) to 51% (96h) for huSM5-l treated QYC cells and from 17% (48h) to 38% (72h) for huSM5-l treated XJC cells.
  • MTT assay is based on the principle that viable cells can reduce yellow MTT into blue pu ⁇ le crystals.
  • Inhibition (%) (B-CV(A-C) ⁇ l00 [0171] "A” represents the OD value of cells without huSM5-l treatment; "B” represents the
  • huSM5-l Growth inhibition of huSM5-l were tested in four tumor cell lines (hepatocellular carcinoma cell QYC and XJC, breast cancer cell line MDA-MB-231, and melanoma cell line
  • huSM5-l antibody could significantly inhibited the growth of tumor cells in a dose- and time-dependent manner.
  • the growth inhibition of huSM5-l antibody is related to apoptosis induction through a caspase-10 mediated pathway, and the apoptotic process may involve DNA fragmentation.
  • Example 6 The in vitro effect on tumor cells of anti-human SM5-1 chimeric antibody
  • the cell lines used in viability examination was QYC cells which were obtained from
  • the QYC cells was cultured in 25 cm 2 flask with
  • the assay was performed in 96-well plate.
  • the anti-human SM5-1 humanized and chimeric antibody described above were diluted with RPMI- 1640/DMEM containing 10% FCS.
  • the antibody to be tested (20 mg/ml) was diluted to to 8 ⁇ g/ml in serial dilution, with each dilution no more than 10-fold. The antibody were further diluted 1 :2 serially with
  • RPMI- 1640/DMEM containing 10% FCS for fourteen serial concentrations in 96-well plate, leaving 100 ⁇ l/well.
  • Cells 100 ⁇ l were added into each well containing 200 ⁇ l various concentration of antibody or 200 ⁇ l of RPMI- 1640/DMEM containing 10% FCS as control.
  • wells that were at the edge of the plate were not used for the assay but added 200 ⁇ l PBS. The plate was incubated at 37°C with 7% CO 2 for seven days.
  • PBL peripheral blood lymphocytes
  • Venous blood was taken sterilely from healthy donor and put into sterile 15 ml centrifuge tube containing 20 U/ml heparin. The solution was carefully mixed and equal volume of steriled PBS was added to dilute the blood.
  • lymphocytes isolation fluid obtained from CACS, Cellular Biology Institute
  • the tube was centrifuged at 20°C, 800g for 30 min with brake turned off. After the centrifuge, three layers were formed. The three layers (from above to the bottom) were blood plasma layer, cell separation liquid layer, red and granular cell layer. A white frosted glass-like layer between the plasma layer and cell separation liquid layer was the layer containing lymphocytes and mononuclear cells. This white layer was taken out using a pipette and put into another 15 ml sterile centrifuge tube. PBS was added into the centrifuge tube to dilute the PBL suspension and then centrifuged at 200g for 5 min. The pellet was washed 2 times with PBS.
  • the anti-SM5-l antibodies was diluted to 40 ⁇ g/ml, and was then diluted serially 1 :2 in 1.5 ml centrifuge tube with non-phenol red RPMI- 1640/DMEM, totaling 14 concentrations and 300 ul/tube.
  • QYC cells 300 ⁇ l were added into each tube. Cells were incubated at 4°C for 30 min. Cells were then centrifuged at 200g for 5 min. . Cell pellets were washed 2 times with PBS. Cells were then suspended in 300 ul non-phenol red RPMI- 1640/DMEM. Cells reacted with anti-SM5-l antibody were added into a well of a 96-well plate at 100 ul/well. Effector cells at 100 ul/well were added into each well of the 96-well plate with effector to target ratio of 20: 1. The plate was incubated at 37°C with 7% CO 2 for 7 h.
  • Antigen recognized by anti-SM5-l is highly expressed on the surface of human hepatocellular carcinoma cell line QYC. With human complement in the culture fluid, the target cells bound with chimeric antibodies will be lysed by so-called complement dependent cell mediated cytotoxicity (CDC). When there is an over dose of complement (provided by fresh normal human sera), in a certain range, the degree of lysis is related to antibody concentration.
  • CDC complement dependent cell mediated cytotoxicity
  • Degree of lysis can be determined by detecting lactate dehydrogenase (LDH) released by the lysed cells.
  • LDH lactate dehydrogenase
  • Cell line for bioactivity determination is QYC , which is without any pathogen.
  • the cells were cultured in 25-75 cm 2 flasks with RPMI-1640/DMEM(1:1) containing 10%NBS.
  • the following culture medium were prepared and stored at 4°C: A, RPMI- 1640/DMEM (1 :1) containing 10% NBS; B, non-phenol red RPMI-1640 without sera; C, culture fluid B with 5% normal human sera.
  • the sera were freshly isolated from healthy donor, and was stored at
  • the culture condition was 37°C, 5% CO 2 , and saturation humidity.
  • a standard sample and the protein to be tested were diluted to 40 ⁇ g/ml in serial dilution, with each dilution no more than 10-fold.
  • the standard and the protein to be tested were further diluted 1 :2 in fourteen 1.5-ml sterile centrifuge tubes with the final dilution volume
  • Culture medium C was used as negative control.
  • 0.1 ml of the diluted standard protein or the protein to be tested described above or the negative control was added into each well. Each condition was done in duplicate. The plate was incubated at 37°C, 5% CO 2 for 3-4 h.
  • Fig. 8 shows the result.
  • Bioactivity percentage (%) half effective dosage of standard product(ED50)/HALF effective dosage of samples (ED50) x 100%
  • mice Forty female nude mice were inoculated s.c with QYC. After seven weeks, tumor masses reached 0.5 cm in diameter. These mice were randomly divided into 5 groups: 8 mice for PBS group; 8 mice for non-related antibody, chimeric anti-human CD3 mAb, 4 mg/kg; 8 mice for humanized anti-human CD3 mAb, 4 mg/kg; 8 mice for anti-human SM5-1 chimeric mAb, 4 mg/kg; 8 mice for anti-human SM5-1 humanized mAb, 4 mg/kg.
  • injection volume was about 250 ul for each nude mouse.
  • Fig. 9 indicated that both chimeric and humanized anti-human SM5-1 monoclonal antibodies were effective in controlling the size of the tumor mass formed by human hepatocellular carcinoma cell line QYC. These antibodies may function via ADCC and/or CDC.
  • Example 10 Tissue distribution of 12S I labeled anti-human SM5-1 chimeric and humanized mAbs after tail vein injection into nude mice
  • the labeling efficiency was 682823 cpm/ ⁇ l (0.52 ⁇ g/ ⁇ l) for anti-human SM5-1 chimeric antibody, 681012 cpm/ ⁇ l (0.52 ⁇ g/ ⁇ l) for anti-human SM5-1 humanized mAbs.
  • the nude mice were weighed and 125 I labeled humanized and chimeric anti-human SM5-1 mAbs were injected through tail vein at about 180 ul for each mouse. According to literature, distribution measurement was generally done within 24-72h. Forty-eight hours was taken as the measure time in this experiment.
  • the blood was taken from eyeballs with eye forceps, and then the following 21 kinds of tissues (blood, thyroid gland, lung, heart, skin, gallbladder, spleen, fat, adrenal gland, kidney, liver, stomach, intestine, intestinal content, mesentery lymphnode, bladder, testis, muscle, bone, brain, and tumor) were taken sequentially. Tumor tissue was taken at last. Cross pollution should be avoided during the process.
  • Fig. 10 shows the results. As shown in Fig. 10, both chimeric and humanized anti-human SM5-1 mAbs were selectively concentrated at tumor mass. Therefore, drugs that derived from chimeric and humamzed anti-human SM5-1 mAbs may be applied for tumor radiotherapy.
  • the reaction mixture was loaded on a Sephadex G-50 gel column for gelfiltration purification. After testing, the radioactivity of the labeled antibody was 126 MBq/mg, which was enough for radioimmunotherapy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

L'invention concerne des anticorps qui sont spécifiques de l'antigène SM5-1 exprimé dans les mélanomes, le cancer du sein et les carcinomes hépatocellulaires ; ainsi que des polynucléotides codant ces anticorps. L'invention concerne en outre l'utilisation desdits anticorps et/ou polynucléotides dans le diagnostic et le traitement de tumeurs malignes.
PCT/US2004/017855 2003-06-06 2004-06-04 Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees WO2005053604A2 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA002528182A CA2528182A1 (fr) 2003-06-06 2004-06-04 Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees
EP04776307A EP1631238A2 (fr) 2003-06-06 2004-06-04 Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees
AU2004294893A AU2004294893A1 (en) 2003-06-06 2004-06-04 Antibodies specific for cancer associated antigen SM5-1 and uses thereof
JP2006533570A JP2007535905A (ja) 2003-06-06 2004-06-04 癌関連抗原sm5−1の特異抗体およびその用途
IL172242A IL172242A0 (en) 2003-06-06 2005-11-29 Antibodies specific for cancer associated antigen sm5-1 and uses thereof

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
CN03129123.6 2003-06-06
CN03129123 2003-06-06
CNB2003101199264A CN1279056C (zh) 2003-06-06 2003-11-25 肿瘤相关抗原sm5-1的特异性抗体及其应用
CN2003101199264 2003-11-25
US10/722,849 US20050031617A1 (en) 2003-06-06 2003-11-26 Antibodies specific for cancer associated antigen SM5-1 and uses thereof
US10/722,849 2003-11-26
TW092133571A TW200517122A (en) 2003-06-06 2003-11-28 Antibodies specific for cancer associated antigen SM5-1 and uses thereof
TW092133571 2003-11-28

Publications (2)

Publication Number Publication Date
WO2005053604A2 true WO2005053604A2 (fr) 2005-06-16
WO2005053604A3 WO2005053604A3 (fr) 2006-02-09

Family

ID=42331655

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/017855 WO2005053604A2 (fr) 2003-06-06 2004-06-04 Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees

Country Status (5)

Country Link
JP (1) JP2007535905A (fr)
AU (1) AU2004294893A1 (fr)
CA (1) CA2528182A1 (fr)
IL (1) IL172242A0 (fr)
WO (1) WO2005053604A2 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007017686A3 (fr) * 2005-08-11 2007-07-19 Matossian-Rogers Arpi Peptides pour traitement et diagnostic d'une maladie auto-immune
JP2012139219A (ja) * 2007-01-05 2012-07-26 Univ Of Zuerich 疾病に特異的な結合分子および標的を提供する方法
US8796439B2 (en) 2006-07-14 2014-08-05 Ac Immune S.A. Nucleic acid molecules encoding a humanized antibody
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
RU2556816C1 (ru) * 2014-03-12 2015-07-20 Открытое акционерное общество "Фармацевтический импорт, экспорт" (ОАО "Фармимэкс") Штамм клеток яичников китайского хомячка - продуцент рекомбинантного антитела против фактора некроза опухоли альфа человека
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
CN109627337A (zh) * 2018-12-29 2019-04-16 上海复宏汉霖生物技术股份有限公司 一种抗prlr的单克隆抗体及其制备方法和应用
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL1954718T3 (pl) 2005-11-30 2015-04-30 Abbvie Inc Przeciwciała skierowane przeciwko A globulomerowi, ich reszty wiążące antygeny, odpowiednie hybrydomy, kwasy nukleinowe, wektory, komórki gospodarze, sposoby wytwarzania tych przeciwciał, kompozycje zawierające te przeciwciała, zastosowania tych przeciwciał i sposoby stosowania tych przeciwciał
EP2124952A2 (fr) 2007-02-27 2009-12-02 Abbott GmbH & Co. KG Méthode de traitement d'amyloïdoses
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
AU2008311367B2 (en) 2007-10-05 2014-11-13 Ac Immune S.A. Use of anti-amyloid beta antibody in ocular diseases
CA2806909C (fr) 2010-07-30 2019-12-17 Ac Immune S.A. Anticorps humanises surs et fonctionnels

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6180377B1 (en) * 1993-06-16 2001-01-30 Celltech Therapeutics Limited Humanized antibodies

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6180377B1 (en) * 1993-06-16 2001-01-30 Celltech Therapeutics Limited Humanized antibodies

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
REINKE S ET AL: 'Differential expression of MART-1, tyrosinase, and SM5-1 in primary and metastatic melanoma.' AMERICAN JOURNAL OF DERMATOLOGY. vol. 27, no. 5, 2005, pages 401 - 406, XP008058050 *
TREFZER U ET AL: 'SM5-1: a new monoclonal antibody which is highly sensitive and specific for melanocytic lesions.' ARCH DERMATOL RES. vol. 292, 2000, pages 583 - 589, XP002993817 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
EP2578602A3 (fr) * 2005-08-11 2013-07-31 Arpi Matossian-Rogers Peptides associés au TCR-V beta pour traitement et diagnostic d'une maladie auto-immune
US9243062B2 (en) 2005-08-11 2016-01-26 Arpi Matossian-Rogers Peptides for treatment and diagnosis of autoimmune disease
WO2007017686A3 (fr) * 2005-08-11 2007-07-19 Matossian-Rogers Arpi Peptides pour traitement et diagnostic d'une maladie auto-immune
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US8796439B2 (en) 2006-07-14 2014-08-05 Ac Immune S.A. Nucleic acid molecules encoding a humanized antibody
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
JP2012139219A (ja) * 2007-01-05 2012-07-26 Univ Of Zuerich 疾病に特異的な結合分子および標的を提供する方法
US9585956B2 (en) 2007-06-12 2017-03-07 Ac Immune S.A. Polynucleotides encoding anti-amyloid beta monoclonal antibodies
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
RU2556816C1 (ru) * 2014-03-12 2015-07-20 Открытое акционерное общество "Фармацевтический импорт, экспорт" (ОАО "Фармимэкс") Штамм клеток яичников китайского хомячка - продуцент рекомбинантного антитела против фактора некроза опухоли альфа человека
CN109627337A (zh) * 2018-12-29 2019-04-16 上海复宏汉霖生物技术股份有限公司 一种抗prlr的单克隆抗体及其制备方法和应用
CN109627337B (zh) * 2018-12-29 2023-07-21 上海复宏汉霖生物技术股份有限公司 一种抗prlr的单克隆抗体及其制备方法和应用

Also Published As

Publication number Publication date
CA2528182A1 (fr) 2005-06-16
IL172242A0 (en) 2006-04-10
JP2007535905A (ja) 2007-12-13
WO2005053604A3 (fr) 2006-02-09
AU2004294893A1 (en) 2005-06-16

Similar Documents

Publication Publication Date Title
CN109963591B (zh) B7h3抗体-药物偶联物及其医药用途
CA2600505C (fr) Anticorps diriges contre la mesotheline
CN113544156B (zh) 抗Claudin18.2抗体及其应用
US20060228366A1 (en) Tumor specific monoclonal antibodies
WO2005053604A2 (fr) Anticorps specifiques de l'antigene sm5-1 associe au cancer et utilisations associees
MXPA04004857A (es) ANTICUERPOS DE COLáGENA HUMANIZADOS Y METODOS RELACIONADOS.
US20030077277A1 (en) Human antibodies that have MN binding and cell adhesion-neutralizing activity
JP2022539344A (ja) 抗cea抗体及びその応用
US20050031617A1 (en) Antibodies specific for cancer associated antigen SM5-1 and uses thereof
US6787638B1 (en) Tumor specific human monoclonal antibodies and methods of use
US20070244304A1 (en) Methods of Identifying Neoplasm-Specific Antibodies and Uses Thereof
EP1611156A2 (fr) Anticorps specifiques des neoplasmes et leurs utilisations
US20050232926A1 (en) Antibodies specific for cancer associated antigen SM5-1 and uses thereof
AU2012254877B2 (en) Anti-mesothelin antibodies
CA2467020A1 (fr) Anticorps monoclonaux specifiques de tumeur
EP3619237A1 (fr) Anticorps dirigés contre l'antigène carcino-embryonnaire pour la thérapie et le diagnostic du cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 172242

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2528182

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004776307

Country of ref document: EP

Ref document number: 2006533570

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2004294893

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2622/KOLNP/2005

Country of ref document: IN

WWP Wipo information: published in national office

Ref document number: 2004294893

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2004776307

Country of ref document: EP