EP1576010A2 - Compositions et procedes therapeutiques contre les cancers caracterises par l'expression de l'antigene mn/ca ix associe aux tumeurs - Google Patents

Compositions et procedes therapeutiques contre les cancers caracterises par l'expression de l'antigene mn/ca ix associe aux tumeurs

Info

Publication number
EP1576010A2
EP1576010A2 EP03749129A EP03749129A EP1576010A2 EP 1576010 A2 EP1576010 A2 EP 1576010A2 EP 03749129 A EP03749129 A EP 03749129A EP 03749129 A EP03749129 A EP 03749129A EP 1576010 A2 EP1576010 A2 EP 1576010A2
Authority
EP
European Patent Office
Prior art keywords
carbonic anhydrase
residues
antibody
seq
agent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP03749129A
Other languages
German (de)
English (en)
Other versions
EP1576010A4 (fr
Inventor
Justin Wong
Jill Winter
Guita Lalehzadeh
Robert Warne
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis Vaccines and Diagnostics Inc
Original Assignee
Chiron 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
Application filed by Chiron Corp filed Critical Chiron Corp
Publication of EP1576010A2 publication Critical patent/EP1576010A2/fr
Publication of EP1576010A4 publication Critical patent/EP1576010A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes

Definitions

  • the present invention is directed to compositions and methods of therapy for the treatment of cell proliferative disorders, more particularly antibodies and small molecules that target MN/CA IX enzymatic activity on neoplastic cells that express this protein and methods of their use in treatment of cancers characterized by expression of this tumor-associated antigen.
  • CAs Carbonic anhydrases catalyze the interconversion of carbon dioxide and bicarbonate. These enzymes play a key role in physiological processes involving water and electrolyte balance, CO 2 and HCO 3 " transport, and pH regulation.
  • the novel membrane-associated CA isoenzyme CA IX was initially described as the tumor-associated membrane antigen designated MN. Recognition of MN as being a putative cell-adhesion molecule that has an extracellular domain with the essential structural features and activity of CAs (Pastorek et al. (1994) Oncogene 9:2877-2888; Opavsky et al. (1996) Genomics 33:480-487; Zavada et al. (1997) Intl. J.
  • Oncol. 10:857-863 resulted in the classification of MN as the ninth member of the carbonic anhydrase family, renamed CA IX. Since then, a kidney-cancer-associated antigen designated G250 has been cloned and identified as a transmembrane protein identical to the tumor-associated antigen MN/CA IX (Grabmaier et al. (2000) Int. J. Cancer 85:865-870).
  • CA IX (MN/G250) is also present in normal gastric, intestinal, and biliary mucosa (Pastorekova et al. (1997) Gastroenterology
  • CA IX protein is almost 100% associated with cervical carcinomas (Liao et al. (1994) Am J. Pathol. 145:598-609), esophagus carcinomas (Turner et al. (1997) Hum. Pathol. 28:740-744), and with renal clear cell carcinomas (Liao et al. (1997) Cancer Res. 57:2827-2831) in human patients. It has also been detected in a high percentage of colorectal (Saarnio et al. (1998) Am. J. Pathol. 153:279-285) and lung (Nermylen et al. (1999) Eur. Respir. J. 14:806-811) carcinomas of human patients.
  • CA IX in vitro is upregulated by cell density in HeLa cells and correlates with tumorigenicity in HeLa cell fibroblast cell hybrids (Zavada et al. (1993) Int. J. Cancer 54:268-274). Further, its expression in ⁇ IH3T3 cells promotes cell proliferation (Pastorek et al. (1994) Oncogene 9:2877-2888). Conditions of hypoxia induce expression of CA IX in tumors and cultured tumor cells, indicating that this protein may be a biomarker for hypoxia in some tumors (Ivanov et al. (2001) Am. J. Pathol. 158(3):905-919; Beasley et al. (2001) Cancer Res.
  • CA IX may help to maintain the extracellular acidic pH in tumors, thereby providing a conducive environment for tumor growth and proliferation as well as enhancing tumor resistance to radiotherapy and chemotherapy (Ivanov et al. (2001) Am. J. Pathol. 158(3):905-919; Beasley et al. (2001) Cancer Res. 61(13):5262-5267).
  • compositions and methods for inhibiting the activity of CA IX are needed to provide effective treatment for cancers that are characterized by expression of this protein.
  • compositions and methods useful in inhibiting proliferation of CA IX+ preneoplastic or neoplastic cells in a mammal are provided.
  • the compositions are antagonist anti-CA IX antibodies and other inhibitory agents that target carbonic anhydrase activity of CA IX on these cells.
  • the antagonist anti-CA IX antibodies or antigen-binding fragments thereof specifically react with an inhibitory epitope of CA IX or biologically active variant thereof such that an antibody-antigen complex is formed, whereby the formation of this complex results in inhibition of carbonic anhydrase activity of CA IX or biologically active variant thereof.
  • the antagonist anti-CA DC antibodies are monoclonal antibodies.
  • Suitable monoclonal antibodies are those having human constant regions, those also having wholly or partially humanized framework regions, and those that are fully human antibodies, or antigen-binding fragments thereof that specifically bind to the inhibitory epitope of interest on CA DC or biologically active variant thereof such that the formation of the antibody-antigen complex results in inhibition of carbonic anhydrase activity of the CA IX protein of interest, for example, human CA IX.
  • CA DC tumor-associated antigen Other small molecule agents that inhibit carbonic anhydrase activity of CA DC or biologically active variant thereof and screening assays for identifying such agents are also provided.
  • the antagonist anti-CA DC antibodies, antigen-binding fragments thereof, and other CA IX inhibitory agents identified herein are useful in the treatment of cancers characterized by the expression of the CA DC tumor-associated antigen.
  • Methods for treating a cancer that is characterized by expression of carbonic anhydrase DC (CA DC) in a mammal are also encompassed.
  • the method comprises administering a therapeutically effective amount of an agent that inhibits carbonic anhydrase activity of CA DC in a mammal in need thereof.
  • Agents of the invention include antagonist anti-CA DC antibodies, and antigen-binding fragments of said antagonist anti-CA DC antibody, peptides, peptoids, and small organic molecules.
  • the anti-CA DC antibodies or antigen binding fragments thereof specifically react with an inhibitory epitope of CA DC to form an antibody-antigen complex, whereby the formation of said complex results in inhibition of carbonic anhydrase activity of said CA DC.
  • the agents of the invention find use in inhibiting proliferation of neoplastic cells that are characterized by expression of CA DC protein.
  • the agents are administered in a therapeutically effective dose to inhibit carbonic anhydrase activity ofCA IX.
  • the invention further comprises methods for screening and identifying agents that inhibit carbonic anhydrase activity of a CA IX polypeptide having a functional carbonic anhydrase domain.
  • the agents are identified by combining an agent to be tested with a cell expressing a CA DC polypeptide under conditions suitable for detecting carbonic anhydrase activity and assessing the ability of an agent to inhibit carbonic anhydrase activity, --nhibition of carbonic anhydrase activity by the agent indicates that the agent is an inhibitor.
  • Agents may also be identified by combining an agent to be tested with a composition comprising an isolated or recombinantly produced CA DC polypeptide under conditions suitable for detecting carbonic anhydrase activity and assessing the ability of the agent to inhibit carbonic anhydrase activity. These agents may be used in compositions or formulated into pharmaceutical compositions for use in the methods of the invention.
  • Figure 1 shows a fluorescein diacetate assay used to detect carbonic anhydrase activity of CA DC in a cell-free high-throughput screening assay.
  • compositions and methods for treating a mammal having a cancer characterized by expression of carbonic anhydrase DC also known as MN and G250; referred to herein as CA DC.
  • Compositions include anti- CA DC antibodies and antigen-binding fragments thereof that specifically react with an epitope of CA DC such that an antibody-antigen complex is formed, whereby the formation of this complex results in inhibition of carbonic anhydrase activity of CA DC.
  • Inhibitory epitopes can comprise contiguous amino acid residues (i.e., residues within the epitope are arranged sequentially one after another in a linear fashion), discontinuous amino acid residues (referred to herein as “discontinuous epitopes; residues within these epitopes are not arranged sequentially), or both contiguous and discontinuous amino acid residues of the CA DC polypeptide or variant thereof, and the residues making up the epitope can reside within the CA DC carbonic anhydrase domain and/or outside the CA DC carbonic anhydrase domain, so long as the formation of the antibody-antigen complex with the anti-CA DC antibody or antigen-binding fragment thereof results in reduction or inhibition of the carbonic anhydrase activity of CA DC.
  • the inhibitory epitopes are epitopes comprising contiguous amino acid residues of the carbonic anhydrase domain of CA DC, epitopes comprising discontinuous amino acid residues of the carbonic anhydrase domain of CA DC, and epitopes comprising both contiguous and discontinuous residues of the carbonic anhydrase domain.
  • Binding of an anti-CA DC antibody or antigen-binding fragment thereof to inhibitory epitopes of CA DC as described herein may prevent CA DC from assuming an appropriate structural form or conformation necessary for mediating carbonic anhydrase activity, may physically inhibit CA DC from complexing with inorganic co-factors necessary for carbonic anhydrase activity, or may physically inhibit CA DC from complexing with the appropriate enzymatic substrate, though other mechanisms of inhibition are also encompassed.
  • CA IX also known as the MN and G250 tumor-associated antigens, is an N- glycosylated transmembrane protein encoded by the MN gene.
  • the coding sequence for human CA DC (see SEQ ID NO:l) and the translated amino acid sequence for this protein (SEQ ID NO:2) are known in the art. See, for example, U.S. Patent Nos. 6,204,370 and 5,989,838, herein incorporated by reference.
  • Human CA DC comprises a signal peptide, a proteoglycan-like domain, a carbonic anhydrase domain, a transmembrane domain, and an intracellular (also referred to as intracytoplasmic) domain at the C-terminus.
  • residue 1 the residue positions denoting the location of these domains within the CA DC protein vary within the reported literature, generally by ⁇ 1-3 residues at either end of any given domain. For purposes of the present invention, this variation is accounted for with the use of the term "about.” In this manner, the term "about” with respect to the residue denoting the beginning or end of each of these domains encompasses 1-3 residues on either side of the designated position.
  • a domain is designated a location of about residues 38-134 of the human CA DC amino acid sequence
  • the domain encompasses a starting location at residue 38 ⁇ l-3 residues and encompasses an ending location at residue 134 ⁇ l-3 residues
  • the starting location would encompass residue 35, 36, 37, 38, 39, 40, or 41 of the human sequence
  • the ending location would encompass residue 131, 132, 133, 134, 135, 136, or 137 of the human sequence.
  • the residue locations of the various domains of the human CA DC protein are defined in accordance with this foregoing description.
  • the human CA DC protein consists of a signal peptide (corresponding to residue 1 to about residue 37 of SEQ ID NO:2), a proteoglycan-like domain (corresponding to about residues 38-134 of SEQ ID NO:2), a carbonic anhydrase domain (corresponding to about residues 135-414 of SEQ ID NO:2), a transmembrane domain (corresponding to about residues 415-433 of SEQ ID NO:2), and an intracellular C-terminus (corresponding to about residues 434-459 of SEQ ID NO:2).
  • the functional region of the proteoglycan-like domain resides at about residues 53- 111, while the functional region of the carbonic anhydrase domain resides at about residues 141-389.
  • CA carbonic anhydrase domain
  • residues 226, 228, and 251 of SEQ ID NO:2 Three zinc-liganded histidine residues obligatory for carbonic anhydrase activity are located within the carbonic anhydrase (CA) domain (at residues 226, 228, and 251 of SEQ ID NO:2).
  • Antigenic region analysis of the full-length human CA DC protein predicts an antigenic region within the carbonic anhydrase domain, with the region of antigenicity residing at about residues 229-256 of SEQ ID NO:2.
  • the CA DC tumor-associated antigen was initially identified as being expressed in some human tumor cell lines in vitro, for example, by T24 (bladder carcinoma), HeLa (cervical carcinoma), SK-Mel 1477 (melanoma), and T47D (mammary carcinoma).
  • CA DC is also produced by cells of some human cancers in vivo, for example, by cells of ovarian and endometrial carcinomas, uterine cervical carcinomas, renal cell carcinomas (RCC), colorectal cancer (CRC), and lung cancer, as well as cells of some benign neoplasias such as mammary papillomas.
  • CA DC is not found in non-tumorigenic hybrid cells, and is generally not found in the cells of normal tissues. An exception resides in its expression within normal gastric, intestinal, and biliary mucosa.
  • the CA IX gene is strongly correlated with tumorigenesis and may be a causative agent.
  • Tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth, including tumors and neoplastic growth (i.e., neoplasms).
  • tumors include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
  • cancers include breast cancer, prostate cancer, colon cancer, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, colorectalcancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, vulval cancer, thyroid cancer, hepatic carcinoma, and various types of head and neck cancer.
  • Treatment is an intervention performed with the intention of preventing the development or altering the pathology of a disorder. Accordingly, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures.
  • a therapeutic agent may directly decrease the pathology of tumor cells, or render the tumor cells more susceptible to treatment by other therapeutic agents, e.g., radiation and/or chemotherapy.
  • the "pathology" of cancer includes all phenomena that compromise the well being of the patient. This includes, without limitation, abnormal or uncontrollable cell growth, metastasis, interference with the normal functioning of neighboring cells, release of cytokines or other secretory products at abnormal levels, suppression or aggravation of inflammatory or immunological response, etc.
  • CA DC protein and gene are described in e.g., U.S. Patent Nos. 6,204,370 and 5,989,838.
  • functional domain of this protein is responsible for its role in cell transformation and rumorigenesis.
  • the present invention is based upon the discovery that this role resides within the carbonic anhydrase domain.
  • cells expressing a functional carbonic anhydrase domain of the CA DC protein develop a transformed phenotype.
  • “functional" carbonic anhydrase domain is intended that the domain is expressed and has functional carbonic anhydrase activity.
  • compositions and methods of the invention are directed to the inhibition of carbonic anhydrase activity of CA DC protein or biologically active variant thereof on preneoplastic or neoplastic cells as described herein below.
  • compositions of the invention When administered to a subject in need of treatment, compositions of the invention effectively reduce or inhibit carbonic anhydrase activity of CA DC or biologically active variant thereof by 5%, 10%, 15%, 20%, 25%, 30%, 35%, preferably 40%, 45%, 50%, 55%, 60%, more preferably 70%, 75%, 80%, 85%, and most preferably 90%, 95%, 99%, or 100%.
  • Methods for measuring the ability of a candidate antibody or candidate test agent to inhibit carbonic anhydrase activity of a CA DC protein are known in the art.
  • any assay available in the art for measuring CO 2 release or conversion in the presence of the candidate inhibitory agent can be used to detect the ability of the candidate agent to inhibit CA IX carbonic anhydrase activity.
  • Reduction or inhibition of this activity on preneoplastic cells prevents their phenotypic transformation into tumorogenic cells.
  • Reduction or inhibition of this activity on neoplastic cells can inhibit their continued proliferation or result in overall reduction in tumor size and tumor burden.
  • compositions of the invention can be used to inhibit carbonic anhydrase activity of CA DC at the cell surface of preneoplastic or neoplastic cells expressing this cell-surface antigen, thereby limiting carbonic anhydrase activity in the extracellular environment.
  • the composition is an antagonist antibody of the invention
  • such inhibition occurs by the formation of an antibody-antigen complex as described elsewhere herein.
  • formation of the antibody-antigen complex between the epitope and an antagonist anti-CA DC antibody of the invention can result in a complex that resides extracellularly.
  • formation of the antibody-antigen complex can result in internalization of the CA IX cell-surface protein, thereby sequestering the CA DC molecule inside the cell.
  • the antagonist anti-CA DC antibody can also be used as a delivery mechanism to deliver antibody-conjugated therapeutic moieties to the inside of CA DC-bearing tumor cells.
  • Suitable therapeutic moieties that can be conjugated to the antagonist anti-CA DC antibodies of the invention for subsequent delivery to the inside of CA DC-bearing tumor cells are described herein below and include, but are not limited to, cytotoxins, chemotherapeutics, radio-metal ions, and the like.
  • Any agent that inhibits carbonic anhydrase activity of CA DC or biologically active variant thereof on CA DC+ preneoplastic or neoplastic cells can be used in the methods of the present invention to prevent phenotypic transformation into tumorogenic cells and/or to inhibit proliferation of these cells.
  • Suitable inhibitory agents find use in the treatment of subjects having a cancer characterized by expression of this tumor- associated antigen.
  • Suitable inhibitory agents can be identified using a variety of screening assays, including these disclosed herein, hi one embodiment, suitable agents are identified by monitoring their ability to inhibit cells expressing the CA DC molecule of interest from displaying the transformed phenotype using the soft agar assay described herein below.
  • Suitable inhibitory agents include the antagonist anti- CA DC antibodies disclosed herein, and peptides, peptoids, and small organic molecules that target activity of the carbonic anhydrase (CA) domain of CA DC.
  • Suitable anti-CA DC antibodies of the invention are referred to herein as
  • antagonist anti-CA IX antibodies in view of their ability to inhibit carbonic anhydrase activity of a CA DC polypeptide as noted above.
  • the antagonist anti-CA DC antibodies of the invention and antigen-binding fragments thereof are specifically reactive with an inhibitory epitope of CA IX such that an antibody-antigen complex is formed, whereby the formation of this complex results in inhibition of carbonic anhydrase activity of CA IX.
  • inhibitory epitopes can comprise contiguous amino acid residues, discontinuous amino acid residues, or both contiguous and discontinuous amino acid residues of the CA DC polypeptide or variant thereof, and the residues making up the epitope can reside within the CA IX carbonic anhydrase domain and/or outside the CA DC carbonic anhydrase domain, so long as the formation of the antibody-antigen complex with the anti-CA DC antibody or antigen-binding fragment thereof results in reduction or inhibition of the carbonic anhydrase activity of CA IX.
  • the inhibitory epitopes are epitopes comprising contiguous amino acid residues of the carbonic anhydrase domain of CA DC, epitopes comprising discontinuous amino acid residues of the carbonic anhydrase domain of CA DC, and epitopes comprising both contiguous and discontinuous residues of the carbonic anhydrase domain.
  • specifically reactive or “specifically reacts” is intended that the antibody that recognizes a particular region of the CA DC protein that comprises the inhibitory epitope of interest forms a specific antibody-antigen complex with that portion of the CA DC protein corresponding to the inhibitory epitope of interest when the antibody comes into contact with that protein, either in an in vitro or in vivo setting.
  • candidate antagonist anti-CA DC antibodies that have been prepared against a CA IX protein or biologically active portion thereof that comprises the CA domain of the CA IX protein of interest can be tested for formation of an antibody- antigen complex using a number of well-defined diagnostic assays, such as conventional immunoassay formats to detect and/or quantitate antigen-specific antibodies.
  • diagnostic assays include, for example, enzyme immunoassays, e.g., enzyme- linked immunosorbant assays (ELISA), cell-based assays, flow cytometry, radioimmunoassays, and immunohistochemical staining.
  • a representative assay to detect anti-CA DC antibodies specific to the inhibitory epitopes identified herein, for example inhibitory epitopes comprising residues within the carbonic anhydrase domain is a competition assay in which labeled CA DC polypeptide comprising residues within the CA domain is precipitated by candidate antibodies in a sample, for example, in combination with monoclonal antibodies raised against one or more epitopes comprising residues within this domain.
  • Anti-CA DC antibodies that specifically react with an epitope of interest can be identified by screening a series of antibodies prepared against a CA DC protein or fragment of the protein comprising the particular epitope of the CA DC protein of interest.
  • epitopes of interest include inhibitory epitopes comprising contiguous and/or discontinuous amino acid residues of human CA IX of SEQ ID NO:2.
  • inhibitory epitopes of human CA DC include, for example, those inhibitory epitopes comprising contiguous and/or discontinuous amino acid residues residing within the CA domain corresponding to about residues 135-414 of SEQ ID NO:2.
  • competition assays with previously identified suitable antagonist anti-CA DC antibodies could be used to select monoclonal antibodies comparable to the previously identified antibodies.
  • Antibodies employed in such immunoassays may be labeled or unlabeled. Unlabeled antibodies may be employed in agglutination; labeled antibodies may be employed in a wide variety of assays, employing a wide variety of labels. Detection of the formation of an antibody-antigen complex between an anti-CA DC antibody and an epitope of interest can be facilitated by attaching a detectable substance to the antibody.
  • Suitable detection means include the use of labels such as radionuclides, enzymes, coenzymes, fluorescers, chemiluminescers, chromogens, enzyme substrates or co-factors, enzyme inhibitors, prosthetic group complexes, free radicals, particles, dyes, and the like.
  • labels such as radionuclides, enzymes, coenzymes, fluorescers, chemiluminescers, chromogens, enzyme substrates or co-factors, enzyme inhibitors, prosthetic group complexes, free radicals, particles, dyes, and the like.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material is luminol;
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin; and
  • suitable radioactive material include 125 1, 131 1, 35 S, or 3 H.
  • Such labeled reagents may be used in a variety of well-known assays, such as radioimmunoassays, enzyme immunoassays, e.g., ELISA, fluorescent immunoassays, and the like. See for example, U.S. Patent Nos. 3,766,162; 3,791,932; 3,817,837; and 4,233,402.
  • the antagonist anti-CA DC antibodies of the invention are specifically reactive with an inhibitory epitope comprising contiguous and/or discontinuous residues of the human CA DC protein, and can comprise residues within the carbonic anhydrase (CA) domain of human CA DC (i.e., about residues 135-414 of SEQ ID NO:2), residues outside the CA domain, or residues residing inside and outside the CA domain of human C A DC.
  • Such inhibitory epitopes can correspond to a length of at least 5 residues, at least 8 residues, at least 10, 15, 20, 25, 30, or at least 35 residues of the human CA DC protein.
  • the antagonist anti-CA DC antibodies are specifically reactive with an inhibitory epitope comprising contiguous and/or discontinuous residues of the carbonic anhydrase (CA) domain of human CA DC, which is located at about residues 135-414 of SEQ ID NO:2 and encompasses the functional region of this domain, i.e., about residues 141-389 of SEQ ID NO:2.
  • the antagonist anti-CA IX antibodies are specifically reactive with an inhibitory epitope corresponding to at least 5 residues, at least 8 residues, at least 10, 15, 20, 25, 30, or at least 35 residues of the CA domain of human CA DC protein, i.e., about residues 135- 414 of SEQ ID NO:2.
  • the residues making up the inhibitory epitope can all be contiguous, can all be discontinuous, or can be a mixture of contiguous and discontinuous residues.
  • inhibitory epitopes are those inhibitory epitopes corresponding to a group of contiguous and/or discontinuous amino acid residues selected from residues 200 to 300, residues 210 to 260, residues 220 to 255, and residues 229 to 256 of SEQ ID NO:2.
  • the inhibitory epitope has a length of about 5, 8, 10, 15, 20, 25, 30, or 35 residues, and comprises at least one of the three essential zinc-liganded histidine residues of the CA domain, i.e., at least one of residues 226, 228, and 251 of SEQ ID NO:2.
  • the inhibitory epitope has a length of about 5, 8, 10, 15, 20, 25, 30, or 35 residues, and comprises at least one of residues 229 to 256 of SEQ ID NO:2.
  • the inhibitory epitope can comprise, for example, 1, 3, 5, 8, 10, 12, 15, 18, 20, 22, 25, or even 28 of the residues within residues 229 to 256 of SEQ ID NO:2.
  • inhibitory epitope comprises more than one of, but less than 28 of, residues 229 to 256 of SEQ ID NO:2, these residues can all be contiguous, can all be discontinuous, or can be a mixture of contiguous and discontinuous residues.
  • CA DC protein is a biologically active variant of human CA IX as defined herein below
  • the antagonist anti-CA DC antibodies of the invention are specifically reactive with an inhibitory epitope comprising contiguous and/or discontinuous residues of the CA DC polypeptide variant.
  • Such an inhibitory epitope can comprise residues within the functional CA domain of the CA DC polypeptide variant (i.e., the functional CA domain that is homologous to the functional domain of human CA DC residing at about residues 135-414 of SEQ ID NO:2 when the amino acid sequence for the CA domain of the CA DC polypeptide variant is optimally aligned against the amino acid sequence for the CA domain of human CA IX using sequence alignment methods noted herein below), residues outside the functional CA domain, or residues residing inside and outside the functional CA domain of the CA DC polypeptide variant.
  • Such inhibitory epitopes can correspond to a length of at least 5 residues, at least 8 residues, at least 10, 15, 20, 25, 30, or at least 35 residues of the CA DC polypeptide variant.
  • the antagonist anti-CA DC antibodies of the invention are specifically reactive with an inhibitory epitope of the CA DC polypeptide variant that comprises contiguous and/or discontinuous residues of the functional carbonic anhydrase (CA) domain that is homologous to the functional carbonic CA domain in human CA DC (i.e., about residues 135-414 of SEQ ID NO:2) when the amino acid sequence for the CA domain of the CA DC polypeptide variant is optimally aligned against the amino acid sequence for the CA domain of human CA DC using sequence alignment methods noted herein below.
  • CA functional carbonic anhydrase
  • the antagonist anti- CA DC antibodies are specifically reactive with an inhibitory epitope corresponding to at least 5 residues, at least 8 residues, at least 10, 15, 20, 25, 30, or at least 35 residues of the functional CA domain of the CA DC polypeptide variant.
  • the residues making up the inhibitory epitope can all be contiguous, can all be discontinuous, or can be a mixture of contiguous and discontinuous residues.
  • inhibitory epitopes are those inhibitory epitopes corresponding to a group of contiguous and/or discontinuous amino acid residues selected from the region of the functional CA domain that is homologous to residues 200 to 300, residues 210 to 260, residues 220 to 255, and residues 229 to 256 of SEQ ID NO:2 when the amino acid sequence for the CA domain of the CA DC polypeptide variant is optimally aligned against the amino acid sequence for the CA domain of human CA DC using sequence alignment methods noted herein below.
  • the inhibitory epitope of the CA DC polypeptide variant has a length of about 5, 8, 10, 15, 20, 25, 30, or 35 residues, and comprises at least one of the three essential zinc- liganded histidine residues of the functional CA domain, i.e., at least one of the residues homologous to residues 226, 228, and 251 of SEQ ID NO:2 when the amino acid sequence for the functional CA domain of the CA DC polypeptide variant is optimally aligned against the amino acid sequence for the CA domain of human CA DC using sequence alignment methods noted herein below.
  • the inhibitory epitope of the CA DC polypeptide variant has a length of about 5, 8, 10, 15, 20, 25, 30, or 35 residues, and comprises at least one of the residues of the region of the functional CA domain that is homologous to residues 229 to 256 of SEQ ID NO:2 when the amino acid sequence for the functional CA domain of the CA DC polypeptide variant is optimally aligned against the amino acid sequence for the CA domain of human CA DC using sequence alignment methods noted herein below.
  • the inhibitory epitope can comprise, for example, 1, 3, 5, 8, 10, 12, 15, 18, 20, 22, 25, or even 28 of the residues within the region of the functional CA domain of the CA DC polypeptide variant that is homologous to residues 229 to 256 of SEQ ID NO:2.
  • inhibitory epitope of the CA DC polypeptide variant comprises more than one of, but less than 28 of, the residues in the region of the functional CA domain that is homologous to residues 229 to 256 of SEQ ID NO:2, these residues can all be contiguous, can all be discontinuous, or can be a mixture of contiguous and discontinuous residues.
  • any antagonist anti-CA IX antibody or antigen-binding fragment thereof having the binding characteristics and specificity noted herein is suitable for use in the methods of the present invention.
  • suitable antagonist anti-CA DC antibodies are specifically reactive with an epitope of CA DC such that an antibody-antigen complex is formed, whereby formation of the complex results in inhibition of CA DC carbonic anhydrase activity by 5%, 10%, 15%, 20%, 25%, 30%, 35%, preferably 40%, 45%, 50%, 55%, 60%, more preferably 70%, 75%, 80%, 85%, and most preferably 90%, 95%, 99%, or 100%.
  • Antagonist anti-CA DC antibodies that are specifically reactive with an inhibitory epitope of interest of human CA DC protein or variant thereof can be prepared against human CA DC protein or the variant CA DC polypeptide, for example, antigenic peptides comprising all or a portion of the carbonic anhydrase domain of a CA DC protein of interest, and identified using immunoassays for detecting antibody-antigen complexes and epitope mapping techniques known in the art.
  • immunoassays and epitope mapping techniques include those described in U.S. Patent Nos. 4,708,871 and 5,635, 182, herein incorporated by reference in their entirety, and noted elsewhere herein.
  • antagonist anti-CA IX antibody thus encompasses any antibody that specifically recognizes and binds to an epitope of the CA DC tumor- associated antigen whereby formation of the antibody-antigen complex results in inhibition of carbonic anhydrase activity of CA DC.
  • inhibitory epitopes epitopes that are recognized by the antagonist anti-CA DC antibodies of the invention are referred to herein as “inhibitory epitopes,” as formation of an antibody-antigen complex between the antagonist anti-CA IX antibody and the inhibitory epitope results in inhibition of carbonic anhydrase activity of CA DC.
  • the antagonist anti-CA DC antibodies of the invention include polyclonal antibodies, monoclonal antibodies, single-chain antibodies, and fragments thereof such as Fab, F(ab') 2 , F v , and other fragments that retain the antigen binding function of the parent antagonist anti-CA DC antibody, i.e., binding to an inhibitory epitope of CA DC, whereby carbonic anhydrase activity of this protein is inhibited.
  • Polyclonal sera may be prepared by conventional methods. In general, a solution containing the CA DC antigen is first used to immunize a suitable animal, preferably a mouse, rat, rabbit, or goat.
  • Polyclonal sera can be prepared in a transgenic animal, preferably a mouse bearing human immunoglobulin loci, hi one embodiment, Sf9 cells or Tn5 cells expressing CA DC are used as the immunogen. Immunization can also be performed by mixing or emulsifying the antigen-containing solution, preferably in an adjuvant such as Freund's complete adjuvant, and injecting the mixture or emulsion parenterally (generally subcutaneously, intraperitoneally, or intramuscularly).
  • an adjuvant such as Freund's complete adjuvant
  • a dose of about 10-200, up to about 500 ⁇ g/injection is typically sufficient, --mmunization is generally boosted 2-6 weeks later with one or more injections of the protein, preferably emulsified with Freund's incomplete adjuvant.
  • Polyclonal antisera are obtained by bleeding the immunized animal into a glass or plastic container, incubating the blood at 25°C for one hour, followed by incubating at 4°C for 2-18 hours. The serum is recovered by centrifugation (e.g., 1,000 x g for 10 minutes). About 20-50 ml per bleed may be obtained from rabbits.
  • the antibody is monoclonal in nature.
  • monoclonal antibody an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations or modifications that may be present in minor amounts.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site, i.e., for purposes of the present invention, an epitope of CA DC described herein that when bound to an antagonist antibody of the invention forms an antibody-antigen complex, whereby carbonic anhydrase activity of CA DC is inhibited.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256:495, or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567).
  • the "monoclonal antibodies” may also be isolated from yeast display (Hudson and Souriau (2001) Expert. Opin. Biol. Ther. l(5):845-855), ribosomal display (Hudson and Souriau (2001) Expert. Opin. Biol. Ther. 1(5):845- 855), or phage antibody libraries using the techniques described in, for example, Clackson et al. (1991) Nature 352:624-628; Marks et al. (1991) J. Mol. Biol. 222:581- 597, and U.S. Patent No. 5,514,548; herein inco ⁇ orated by reference.
  • Monoclonal antibodies can be prepared using the method of Kohler et al. (1975) Nature 256:495-496, or a modification thereof.
  • a mouse is immunized with a solution containing an antigen
  • hnmunization can be performed by mixing or emulsifying the antigen-containing solution, preferably in an adjuvant such as Freund's complete adjuvant or Freund's incomplete adjuvant, and injecting the mixture or emulsion parenterally. Any method of immunization known in the art may be used to obtain the monoclonal antibodies of the invention.
  • the spleen (and/or optionally, several large lymph nodes) is removed and dissociated into single cells.
  • the spleen cells may be screened by applying a cell suspension to a plate or well coated with the antigen of interest.
  • the B cells expressing membrane bound immunoglobulin specific for the antigen bind to the plate and are not rinsed away.
  • Resulting B cells, or all dissociated spleen cells are then induced to fuse with myeloma cells to form hybridomas, and are cultured in a selective medium.
  • the resulting cells are plated by limiting dilution and are assayed for the production of antibodies that specifically bind the antigen of interest (and that do not bind to unrelated antigens).
  • the selected monoclonal antibody (mAb)- secreting hybridomas are then cultured either in vitro (e.g., in tissue culture bottles or hollow fiber reactors), or in vivo (as ascites in mice).
  • antibody can be produced in a cell line such as a CHO cell line or myeloma cell line, as disclosed in U.S. Patent Nos. 5,545,403; 5,545,405; 5,998,144; and 5,807,715; incorporated herein by reference.
  • myeloma cell lines include, but are not limited to, Sp2 and NS0 cell lines, and cell lines derived therefrom. Briefly the cell line is transfected with vectors capable of expressing a light chain and a heavy chain, respectively. By transfecting the separate vectors encoding the two proteins, chimeric antibodies can be produced. Another advantage is the correct glycosylation of the antibody.
  • the antibody can be produced by transfecting the cell line with a bicistronic single plasmid encoding the light and heavy chains.
  • anti-CA DC antibody encompasses chimeric anti-CA DC antibodies.
  • chimeric antibodies antibodies that are most preferably derived using recombinant deoxyribonucleic acid techniques and which comprise both human (including immunologically "related" species, e.g., chimpanzee) and non-human components.
  • the constant region of the chimeric antibody is most preferably substantially identical to the constant region of a natural human antibody; the variable region or portion thereof of the chimeric antibody is most preferably derived from a non-human source and has the desired antigenic specificity to the region of the CA DC protein of interest, for example, human CA DC or biologically active variant thereof, that comprises an inhibitory epitope described therein.
  • the non-human source can be any vertebrate source that can be used to generate antibodies to a human CA DC or material comprising a human CA DC, or more particularly an antigenic peptide of human CA DC comprising an inhibitory epitope described herein, for example, an inhibitory epitope residing within the CA domain.
  • Such non-human sources include, but are not limited to, rodents (e.g., rabbit, rat, mouse, etc.; see, for example, U.S. Patent No. 4,816,567, herein incorporated by reference) and non-human primates (e.g., Old World Monkey, Ape, etc.; see, for example, U.S. Patent Nos. 5,750,105 and 5,756,096; herein incorporated by reference).
  • the phrase "immunologically active" when used in reference to chimeric anti-CA DC antibodies means a chimeric antibody that binds human CA DC, more specifically binds to an inhibitory epitope described herein, for example, an inhibitory epitope comprising contiguous and/or discontinuous residues residing within the CA domain of human CA DC or biologically active variant thereof.
  • Humanized anti-CA DC antibodies are also encompassed by the term anti-CA DC antibody as used herein.
  • humanized is intended forms of anti-CA DC antibodies that contain minimal sequence derived from non-human immunoglobulin sequences.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (also known as complementarity determining region or CDR) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and capacity.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance (e.g., to obtain desired affinity).
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • humanized antibodies may include antibodies wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species, h practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some framework residues are substituted by residues from analogous sites in rodent antibodies. See, for example, U.S. Patent ⁇ os. 5,225,539; 5,585,089; 5,693,761; 5,693,762; 5,859,205. See also U.S. Patent No. 6,180,370, and International Publication No. WO 01/27160, where humanized antibodies and techniques for producing humanized antibodies having improved affinity for a predetermined antigen are disclosed.
  • anti-CA DC antibodies are xenogeneic or modified anti-CA DC antibodies produced in a non-human mammalian host, more particularly a transgenic mouse, characterized by inactivated endogenous immunoglobulin (lg) loci.
  • a transgenic mouse characterized by inactivated endogenous immunoglobulin (lg) loci.
  • competent endogenous genes for the expression of light and heavy subunits of host immunoglobulins are rendered non-functional and substituted with the analogous human immunoglobulin loci.
  • These transgenic animals produce human antibodies in the substantial absence of light or heavy host immunoglobulin subunits. See, for example, U.S. Patent Nos. 5,877,397 and 5,939,598, herein inco ⁇ orated by reference.
  • Fragments of the antagonist anti-CA DC antibodies are suitable for use in the methods of the invention so long as they retain the desired affinity and activity of the full-length antibody.
  • a fragment of an anti-CA DC antibody will (1) retain the ability to bind to an inhibitory epitope of CA DC as defined elsewhere herein, for example, an inhibitory epitope comprising contiguous and/or discontinuous residues of the CA domain of the CA DC protein of interest, for example, human CA DC or biologically active variant thereof; and (2) exhibit antagonist activity when bound to such an inhibitory epitope of CA DC, thereby inhibiting carbonic anhydrase activity of the CA DC protein.
  • Such fragments are referred to herein as "antigen-binding" fragments.
  • Suitable antigen-binding fragments of an antibody comprise a portion of a full-length antibody, generally the antigen-binding or variable region thereof.
  • antibody fragments include, but are not limited to, Fab, F(ab 2 , and Fv fragments and single-chain antibody molecules.
  • single-chain Fv or “sFv” antibody fragments is intended fragments comprising the VH and V L domains of an antibody, wherein these domains are present in a single polypeptide chain. See, for example, U.S. Patent Nos. 4,946,778; 5,260,203; 5,455,030; 5,856,456; herein inco ⁇ orated by reference.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains that enables the sFv to form the desired structure for antigen binding.
  • a polypeptide linker between the V H and V L domains that enables the sFv to form the desired structure for antigen binding.
  • Antibodies or antibody fragments can be isolated from antibody yeast libraries, ribosomal expression libraries, or phage libraries generated using the techniques described in, for example, McCafferty et al. (1990) Nature 348:552-554 (1990) and U.S. Patent No. 5,514,548. Clackson et al. (1991) N ⁇ twre 352:624-628 and Marks et al. (1991) J. Mol. Biol. 222:581-597 describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et al.
  • the antibody fragments can be isolated from the antibody yeast, ribosomal, or phage libraries discussed above.
  • Fab'-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab') 2 fragments (Carter et al. (1992) Bio/Technology 10:163-167).
  • F(ab') fragments can be isolated directly from recombinant host cell culture. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
  • the invention also encompasses de-immunized antagonist anti-CA IX antibodies, which can be produced as described in, for example, International Publication ⁇ os. WO 98/52976 and WO 0034317; herein inco ⁇ orated by reference.
  • residues within the antagonist anti-CA DC antibodies of the invention are modified so as to render the antibodies non- or less immunogenic to humans while retaining their antagonist activity toward carbonic anhydrase activity of CA DC, wherein such activity is measured by assays noted elsewhere herein.
  • fusion proteins comprising an antagonist anti-CA DC antibody of the invention, or a fragment thereof, which fusion proteins can be synthesized or expressed from corresponding polynucleotide vectors, as is known in the art. Such fusion proteins are described with reference to conjugation of antibodies as noted below.
  • Antagonist anti-CA DC antibodies identified as having the binding characteristics and specificity described herein for use in the methods of the present invention can have sequence variations produced using methods described in, for example, Patent Publication Nos. EP 0 983 303 Al , WO 00/34317, and WO 98/52976, inco ⁇ orated herein by reference. For example, it has been shown that sequences within the CDR can cause an antibody to bind to MHC Class II and trigger an unwanted helper T cell response. A conservative substitution can allow the antibody to retain binding activity yet lose its ability to trigger an unwanted T cell response. Any such conservative or non-conservative substitutions can be made using art-recognized methods, such as those noted elsewhere herein, and the resulting antibodies will fall within the scope of the invention.
  • the variant antibodies can be routinely tested for antagonist activity, affinity, and specificity using methods described herein.
  • the antibodies of this invention can also have ADCC and CDC activity in addition to neutralizing activity.
  • Antibodies that have ADCC activity interact with "human effector cells” such as leukocytes that express one or more FcRs and perform effector functions.
  • the cells express at least FcyRIII and carry out antigen-dependent cell-mediated cyotoxicity (ADCC) effector function.
  • human leukocytes that mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells, and neutrophils; with PBMCs and NK cells being preferred.
  • Antibodies that have ADCC activity are typically of the IgGl or IgG3 isotype.
  • ADCC-mediating antibodies can be made by engineering a variable region from a non-ADCC antibody or variable region fragment onto an IgGl or IgG3 isotype constant region.
  • Fc receptor or “FcR” are used to describe a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one that binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and FcyRIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • FcyRII receptors include FcyRIIA (an “activating receptor") and FcyRIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor FcyRIIA contains an immunoreceptor tyrosine-based activation motif (IT AM) in its cytoplasmic domain.
  • Inhibiting receptor FcyRlTB contains an immunoreceptor tyrosine-based inhibition motif (ITEVI) in its cytoplasmic domain (see Daeron (1997) Annu. Rev. Immunol 15:203-234).
  • ITEVI immunoreceptor tyrosine-based inhibition motif
  • FcR FcR
  • the term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al. (1976) J Immunol 117:587; and Kim et al. (1994) J. Immunol. 24:249).
  • “Complement dependent cytotoxicity” or “CDC” refers to the ability of a molecule to lyse a target in the presence of complement.
  • the complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule (e.g., an antibody) complexed with a cognate antigen.
  • a CDC assay e.g., as described in Gazzano-Santoro et al. (1996) J. Immunol. Methods 202:163, may be performed.
  • the antagonist anti-CA IX antibody may be labeled using "indirect labeling” or an "indirect labeling approach.
  • indirect labeling or “indirect labeling approach” is intended that a chelating agent is covalently attached to an antibody and at least one radionuclide is inserted into the chelating agent. See, for example, the chelating agents and radionuclides described in Srivagtava and Mease (1991) Nucl. Med. Bio. 18:589-603, herein inco ⁇ orated by reference.
  • the antagonist anti-CA DC antibody may be labeled using "direct labeling” or a "direct labeling approach", where a radionuclide is covalently attached directly to an antibody (typically via an amino acid residue).
  • a radionuclide is covalently attached directly to an antibody (typically via an amino acid residue).
  • Preferred radionuclides are provided in Srivagtava and Mease (1991) supra.
  • the indirect labeling approach is particularly preferred. See also, for example, International Publication Nos. WO 00/52031 and WO 00/52473, where a linker is used to attach a radioactive label to antibodies; and the labeled forms of anti- CA DC antibodies described in U.S. Patent No. 6,015,542; herein inco ⁇ orated by reference.
  • an antagonist anti-CA DC antibody may be conjugated to a therapeutic moiety such as a cytotoxin, a therapeutic agent, or a radioactive metal ion.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include 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.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine.
  • antimetabolites e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine
  • alkylating agents e.g., mechlorethamine.
  • anthracyclines e.g., daunorubicin (formerly daunomycin) and doxorubicin
  • antibiotics e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin
  • the conjugates of the invention can be used for modifying a given biological response; the drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, interferon-alpha, interferon-beta, nerve growth factor, platelet derived growth factor, tissue plasminogen activator; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-1"), interleukin-2 (“IL-2”), interleukin-6 (“IL-6), granulocyte macrophase colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • IL-1 interleukin-1
  • IL-2 interleukin-2
  • the anti-CA DC antibodies of the present invention can bind to amino-acid- residues-specific-epitopes, carbohydrate-specific epitopes, or epitopes formed by both amino acid residues and carbohydrate portions of the molecule, as expressed by the target-bearing tumor cells. If the anti-CA DC antibody carries a cytotoxic or cytostatic agent, it might be preferable that the antibody binds to an epitope that internalizes the antibody-target receptor complex. If the anti-CA DC antibody is to work through ADCC and CDC, then it is preferable that the anti-CA DC antibody remains on the surface of the target tumor cell until the antibody Fc region binds to effector cells. Methods for determining whether an antibody bound to a cognate cell surface antigen remains on a cell surface or is internalized are well known in the art.
  • any agent that specifically inhibits carbonic anhydrase activity of CA DC on CA DC+ tumorigenic cells can be utilized in the methods of the present invention.
  • Candidate agents that inhibit carbonic anhydrase activity of a CA DC protein of interest may be derived from almost any source of chemical libraries, naturally occurring compounds, or mixtures of compounds. Exemplary sources of candidate inhibitors, synthesis of libraries of peptides, peptoids, and small organic molecules are described below.
  • Any agent that is an inhibitor or antagonist of carbonic anhydrase activity of CA DC for example, human CA DC or biologically active variant thereof, can be used in the treatment methods of the present invention.
  • the inhibitor of carbonic anhydrase activity can be a peptide antagonist, a peptoid antagonist, or a small organic molecule antagonist. It is expected that some inhibitors will act at one or more of the three zinc-binding histidine residues within the carbonic anhydrase domain of the CA DC polypeptide (for example, at residues 226, 228, and 251 of human CA DC shown in SEQ ID NO:2), which are obligatory for the catalytic activity of the CA domain (Sly and Hu (1995) Annu. Rev. Biochem. 64:375-401).
  • the use and appropriateness of such inhibitors of CA DC carbonic anhydrase activity for the pu ⁇ oses of the invention are not limited to any theories of mechanism of action of the inhibitor.
  • an inhibitor inhibit the carbonic anhydrase activity of CA DC, for example, human CA DC or biologically active variant thereof as defined herein below.
  • Analogs of peptides as used herein include peptides having one or more peptide mimics, for example peptoids that possess protein-like activity. Included within the definition are, for example, peptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids), peptides with substituted linkages, as well as other modifications known in the art, both naturally occurring and not naturally occurring.
  • small molecule includes any chemical or other moiety that can act to affect biological processes.
  • Small molecules can include any number of therapeutic agents presently known and used, or can be small molecules synthesized in a library of such molecules for the pmpose of screening for function. Small molecules are distinguished from polymers and macromolecules by size and lack of polymerization. Small molecules can include peptides, peptoids, and small organic molecules.
  • the candidate inhibitors of CA DC carbonic anhydrase activity and libraries of candidate inhibitors for screening by the assays disclosed herein can be derived from any of the various possible sources of candidate inhibitors, such as for example, libraries of peptides, peptoids, and small molecules.
  • the inhibitor could be a polypeptide presented by phage display, h general an inhibitor of CA DC carbonic anhydrase activity can be any molecule that may be capable of inhibiting CA DC carbonic anhydrase activity, or carbonic anhydrase activity of a variant CA IX polypeptide.
  • Some libraries for screening can be subdivided into library pools for assaying inhibition of CA IX carbonic anhydrase activity by the assays disclosed herein. Some of each pool is assayed and some is saved for reassay, or to further subdivide into subpools, should a positive be identified. Generation of some of the possible libraries suitable for assay by the methods of the invention is described herein below.
  • a "library” of peptides maybe synthesized and used following the methods disclosed in U.S. Patent No. 5,010,175, (the '175 patent) and in International Publication No. WO 91/17823.
  • a suitable peptide synthesis support for example, a resin
  • the method described in WO 91/17823 is similar but simplifies the process of determining which peptides are responsible for any observed alteration of gene expression in a responsive cell.
  • the methods described in WO 91/17823 and U.S. Patent No. 5,194,392 enable the preparation of such pools and subpools by automated techniques in parallel, such that all synthesis and resynthesis may be performed in a matter of days.
  • peptide analogs and derivatives that can act as inhibitors of gene expression, or as ligands or antagonists.
  • Some general means contemplated for the production of peptides, analogs or derivatives are outlined in Weinstein, ed. (1983) Chemistry and Biochemistry of Amino Acids, Peptides, and
  • Peptoids polymers comprised of monomer units of at least some N-substituted moieties, can act as small molecule inhibitors herein and can be synthesized as described in International Publication No. WO 91/19735.
  • Presently preferred amino acid substitutes are N-alkylated derivatives of glycine, which are easily synthesized and inco ⁇ orated into polypeptide chains.
  • any monomer units that allow for the sequence specific synthesis of pools of diverse molecules are appropriate for use in producing peptoid molecules.
  • the benefits of these molecules for the pu ⁇ ose of the invention are that they occupy different conformational space than a peptide and are more resistant to the action of proteases because their amide linkages are N- substituted.
  • Peptoids are easily synthesized by standard chemical methods.
  • the preferred method of synthesis is the "submonomer” technique described by Zuckermann et al (1992) J. Am. Chem. Soc. 114:10646-7.
  • Synthesis by solid phase techniques of heterocyclic organic compounds in which N-substituted glycine monomer units form a backbone is described in copending application entitled “Synthesis of N-Substituted Oligomers” filed on June 7, 1995 and is herein inco ⁇ orated by reference in full.
  • Combinatorial libraries of mixtures of such heterocyclic organic compounds can then be assayed for the ability to inhibit enzyme activity, specifically carbonic anhydrase activity of CA DC or biologically active variant thereof.
  • the other CA DC inhibitory agents of the invention interact with CA DC or biologically active variant thereof resulting in inhibition of CA IX carbonic anhydrase activity by 5%, 10%, 15%, 20%, 25%, 30%, 35%, preferably 40%, 45%, 50%, 55%, 60%, more preferably 70%, 75%, 80%, 85%, and most preferably 90%, 95%, 99%, or 100%.
  • Assays for detecting inhibition of carbonic anhydrase activity are known in the art, including those assays disclosed elsewhere herein. Screening Assays for Detection of Other --nhibitorv Agents and Antibodies Capable of Inhibiting CA DC Carbonic Anhydrase Activity
  • the present invention thus provides methods for identifying other agents that can serve as therapeutic agents for the treatment of cancers that are characterized by expression of the CA DC protein.
  • the methods comprise screening assays whereby the ability of a test agent to inhibit carbonic anhydrase activity of the CA DC protein of interest is assessed.
  • a candidate test agent is identified as having the ability to inhibit carbonic anhydrase activity in an in vitro assay
  • the agent can be further tested for its ability to inhibit phenotype transformation of cell lines expressing the CA DC protein of interest using cell-based assays known in the art, including the soft agar assay described herein in the Experimental section below.
  • Inhibitory agents identified using in vitro assays can also be further tested using in vivo assays.
  • the invention provides a method (also referred to herein as a "screening assay") for identifying inhibitors, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small organic molecules, or other drugs; and suitable inhibitory antibodies) that bind to the CA DC protein of interest for example, human CA DC or biologically active variant thereof, and which have an inhibitory effect on carbonic anhydrase activity of this protein.
  • inhibitors i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small organic molecules, or other drugs; and suitable inhibitory antibodies) that bind to the CA DC protein of interest for example, human CA DC or biologically active variant thereof, and which have an inhibitory effect on carbonic anhydrase activity of this protein.
  • test compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including biological libraries, spatially addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the "one-bead one- compound” library method, and synthetic library methods using affinity chromatography selection.
  • biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, nonpeptide oligomer, or small molecule libraries of compounds (Lam (1997) Anticancer Drug Des. 12:145).
  • Determining the ability of the test compound to bind to the C A DC protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the CA DC protein, more particularly to the carbonic anhydrase (CA) domain of this protein, can be determined by detecting the labeled compound in a complex.
  • test compounds can be labeled with 125 1, 35 S, 14 C, or 3 H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting.
  • test compounds can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
  • an assay of the present invention is a cell-free assay comprising contacting a CA DC protein, or biologically active portion thereof comprising the CA domain, with a test compound and determining the ability of the test compound to bind to the CA IX protein or biologically active portion thereof, more particularly to the CA domain.
  • Binding of the test compound to the CA DC protein can be determined either directly or indirectly as described above, hi a preferred embodiment, the assay includes contacting the CA IX protein or biologically active portion thereof comprising the CA domain with a known compound that binds CA DC protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to preferentially bind to CA DC protein or biologically active portion thereof as compared to the known compound.
  • a fusion protein can be provided that adds a domain that allows the CA DC protein to be bound to a matrix.
  • glutathione-S-transferase/CA DC fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St.
  • the test compound or the test compound and the nonadsorbed CA DC protein are then combined with the test compound or the test compound and the nonadsorbed CA DC protein, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH).
  • conditions conducive to complex formation e.g., at physiological conditions for salt and pH.
  • the beads or microtitre plate wells are washed to remove any unbound components and complex formation is measured either directly or indirectly, for example, as described above.
  • the complexes can be dissociated from the matrix, and the level of CA DC binding or activity determined using standard techniques.
  • an assay is a cell-free assay comprising contacting CA DC protein or biologically active portion thereof comprising the CA domain with a test compound and determining the ability of the test compound to inhibit the carbonic anhydrase activity of the CA DC protein or biologically active variant thereof. Determining the ability of the test compound to inhibit the carbonic anhydrase activity of a C A DC protein or biologically active portion thereof can be accomplished, for example, by determining the ability of the CA DC protein or portion thereof to bind to a CA DC substrate molecule and inhibit enzymatic activity, as described, for example, in the Experimental section below.
  • a fusion protein can be provided that adds a domain that allows the CA DC protein to be bound to a matrix.
  • glutathione-S-transferase/CA DC fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St.
  • the test compound or the test compound and the nonadsorbed CA DC protein are then combined with the test compound or the test compound and the nonadsorbed CA DC protein, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH).
  • conditions conducive to complex formation e.g., at physiological conditions for salt and pH.
  • the beads or microtitre plate wells are washed to remove any unbound components and complex formation is measured either directly or indirectly, for example, as described above.
  • the complexes can be dissociated from the matrix, and the level of CA DC binding or activity determined using standard techniques.
  • CA DC protein can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated CA DC molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96-well plates (Pierce Chemicals).
  • antibodies reactive with a CA DC protein but which do not interfere with the catalytic action of the CA DC protein on its substrate molecule can be derivatized to the wells of the plate, and unbound CA DC protein trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the CA DC protein, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the CA DC protein or biologically active portion thereof.
  • inhibitors of CA DC carbonic anhydrase activity are identified in a method in which a cell is contacted with a candidate compound and the carbonic anhydrase activity of CA DC protein in the cell is determined relative to carbonic anhydrase activity of CA DC protein in a cell in the absence of the candidate compound.
  • the candidate compound is identified as an inhibitor of CA DC protein carbonic anhydrase activity.
  • the level of CA DC protein carbonic anhydrase activity in the cells can be determined by methods described elsewhere herein for detecting such enzymatic activity.
  • the cells expressing CA DC protein can be derived from a CA IX+ cancer cell line that is positive for expression of this tumor-associated antigen.
  • any suitable host cell can be utilized for recombinant expression of the particular CA DC protein of interest, for example, human CA DC of SEQ ID NO:2 (encoded by SEQ ID NO:l), or biologically active variant thereof as noted elsewhere herein.
  • the CA DC protein of interest can alternatively be expressed as part of a fusion protein as noted herein below.
  • Methods for recombinant expression of CA DC protein or biologically active variants thereof are known in the art. See, for example, the host cell expression systems disclosed in U.S Patent No. 6,051,226, herein inco ⁇ orated by reference in its entirety.
  • Such cell-based assays provide a method for assaying an antibody for the ability to inhibit carbonic anhydrase activity of a CA DC protein of interest.
  • a candidate antibody of interest that is to be tested for its inhibitory activity can be combined with a cell expressing CA DC protein or biologically active variant thereof under conditions suitable for detecting carbonic anhydrase activity, and then the level of the carbonic anhydrase activity of CA DC protein in the cell is determined relative to carbonic anhydrase activity of CA IX protein in a cell in the absence of the candidate antibody. Inhibition of carbonic anhydrase activity by the antibody would be indicative that the antibody is an inhibitor of CA DC carbonic anhydrase activity.
  • Such candidate antibodies that can be tested using this method include existing antibodies as well as candidate antibodies yet to be identified.
  • existing antibodies is intended those antibodies that have been previously identified, including, but not limited to, those antibodies already known in the art.
  • the cell-based screening methods of the present invention provide a means for assaying such antibodies for their ability to inhibit CA DC carbonic anhydrase activity.
  • the CA DC proteins can be used as "bait proteins" in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J. Biol. Chem. 268: 12046-12054; Bartel et al. (1993) Bio/Techniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and PCT Publication No.
  • CA DC-binding proteins or "CA DC-bp”
  • CA DC-bp CA DC-binding proteins
  • the invention includes generating cRNA and cDNA libraries for screening for inhibition of carbonic anhydrase activity of CA DC, can require expression of recombinant CA DC polypeptides, which can alternatively be expressed as a fusion protein as noted elsewhere herein, and can also involve transforming a cell with the gene for a CA DC polypeptide, or fusion protein comprising a CA DC polypeptide, for expression in a screening assay.
  • CA DC is endogenously expressed in cancer cell lines (i.e., CA DC+ neoplastic cells) derived from cancers that are characterized by expression of CA DC.
  • Exemplary systems for generating polypeptides or libraries useful for the screening methods of the invention would include, for example, any standard or useful mammalian, bacterial, yeast, or insect expression system, many of which are described in U.S Patent No. 6,204,370 with regard to production of recombinant CA DC polypeptides.
  • any CA DC polypeptide or peptide useful in the invention can be made by these or other standard methods.
  • plasmids can be constructed and purified using standard recombinant DNA techniques described in, for example, Sambrook et al. (1989), Molecular Cloning, A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Plainview, NY), and Ausubel et al. (1994) Current Protocols in Molecular Biology (Greene Publishing Associates and John Wiley & Sons, New York, NY) under the current regulations described in United States Department of Health and Human Sei"vices (HHS), National Institute of Health (NLH) Guidelines for Recombinant DNA Research.
  • HHS United States Department of Health and Human Sei"vices
  • NLH National Institute of Health
  • references include procedures for the following standard methods: cloning procedures with plasmids, transformation of host cells, cell culture, plasmid DNA purification, phenol extraction of DNA, ethanol precipitation of DNA, agarose gel electrophoresis, purification of DNA fragments from agarose gels, and restriction endonuclease and other DNA-modifying enzyme reactions.
  • CA DC inhibitory agents The antagonist anti-CA IX antibodies, antigen-binding fragments thereof, and other agents that inhibit carbonic anhydrase activity of CA DC or biologically active variant thereof are collectively referred to herein as CA DC inhibitory agents. These CA DC inhibitory agents are useful in the treatment of cancers that are characterized by expression of the CA DC protein.
  • carcinomas such as mammary, bladder, ovarian, uterine, cervical, endometrial, squamous cell and adenosquamous carcinomas; and head and neck cancers; mesodermal tumors, such as neuroblastomas and retinoblastomas; sarcomas, such as osteosarcomas and Ewing's sarcoma; and melanomas.
  • gynecologic cancers including ovarian, cervical, vaginal, endometrial and vulval cancers as well as gynecologic precancerous conditions, such as metaplastic cervical tissues and condylomas; gastrointestinal cancer, such as, stomach, colon and esophageal cancers; urinary tract cancer, such as, bladder and kidney cancers; skin cancer; liver cancer; prostate cancer; lung cancer; and breast cancer.
  • the invention provides methods for treating a subject having a cancer that is characterized by expression of the tumor-associated antigen CA DC.
  • subject is intended mammals, e.g., humans, dogs, cattle, horses, and the like.
  • subject undergoing treatment with the methods of the invention is human.
  • Subjects having a cancer characterized by expression of the CA DC protein can be determined by standard assays known in the art.
  • the presence of CA DC antigen can be detected and/or quantitated using a number of well-defined diagnostic assays.
  • Those in the art can adapt any of the conventional immunoassay formats, for example radioimmunoassays, immunohistochemical staining, immunoelectron and scanning microscopy using immunogold, among other techniques, to detect and/or quantitate MN antigen.
  • Many other formats for detection of CA DC antigen are available.
  • Exemplary immunoassays that are suitable for detecting a serum antigen include those described in U.S. Patent Nos.
  • Exemplary monoclonal antibodies that can be used for detection of CA DC expression in a sample obtained from a subject that is suspected of having existing neoplastic (tumor) cells or newly formed neoplastic cells that express this tumor- associated antigen are the monoclonal antibodies designated M75 and G250.
  • M75 antibody A complete discussion of the M75 antibody is disclosed in U.S. Patent No. 6,051,226, herein inco ⁇ orated by reference in its entirety.
  • a hybridoma that produces this representative CA DC-specific antibody was deposited at the American Type Culture Collection [ATCC; 10801 University Boulevard., Manassas, Virginia 20110-2209 (USA)] under ATCC Number HB 11128.
  • the G250 antibody is well known to those of skill in the art.
  • MN/CA DC-specific monoclonal antibodies can be used to identify the CA IX protein and can be used to readily identify CA DC antigen in Western blots, in radioimmunoassays, and immunohistochemically, for example, in tissue samples that are fresh, frozen, or formalin-, alcohol-, acetone- or otherwised fixed and/or paraffin- embedded and deparaffinized.
  • tissue samples that are fresh, frozen, or formalin-, alcohol-, acetone- or otherwised fixed and/or paraffin- embedded and deparaffinized.
  • tissue samples include tissue specimens, body fluids, tissue extracts, and cell extracts obtained from the subject who is a candidate for treatment with the methods of the invention.
  • Preferred tissue specimens to assay by immunohistochemical staining include cell smears, histological sections from biopsied tissues or organs, and imprint preparations among other tissue samples.
  • Biopsied tissue samples can be, for example, those samples removed by aspiration, bite, brush, cone
  • the treatment methods of the invention comprise administering to a subject in need thereof a therapeutically effective dose or amount of an antagonist anti-CA DC antibody, antigen-binding fragment thereof, or other agent that inhibits carbonic anhydrase activity of CA DC or biologically active variant thereof.
  • these CA DC inhibitory agents exhibit anti-tumor activity.
  • “anti-tumor activity” is intended that the individual undergoing therapy with the CA DC inhibitory agents of the invention will exhibit a favorable response with respect to the neoplastic or preneoplastic condition for which the individual is being treated.
  • favorable responses include, but are not limited to a reduction in the rate of cell proliferation, and hence a decline in growth rate of an existing tumor or in a tumor that arises during therapy, and/or destruction of existing neoplastic (tumor) cells or newly formed neoplastic cells, and hence a decrease in the overall size of a tumor during therapy.
  • these agents that target CA DC carbonic anhydrase activity can be used to promote a positive therapeutic response with respect to a cancer that is characterized by expression of this protein.
  • positive therapeutic response is intended an improvement in the disease in association with the anti-tumor activity of these agents, and/or an improvement in the symptoms associated with the disease.
  • Factors influencing the respective amount of a particular agent to be administered to a subject to achieve inhibition of carbonic anhydrase activity of CA DC, thereby inhibiting proliferation of neoplastic or phenotype transformation of preneoplastic cells expressing this antigen include, but are not limited to, the particular cancer or proliferative disorder or disorder undergoing therapy, the severity of the disease, the history of the disease, and the age, sex, height, weight, health, and physical condition of the individual undergoing therapy.
  • the amount of CA DC inhibitory agent that will constitute an inhibitory amoxmt will also vary depending on such parameters as the particular inhibitory agent and its potency, the half-life of the inhibitory agent in the body, the rate of progression of the cancer or proliferative disorder being treated, the responsiveness of the condition to the dose of treatment or pattern of administration, the formulation, the attending physician's assessment of the medical situation, and other considerations such as prior administration of other therapeutics, or co- administration of any therapeutic that will have an effect on the inhibitory activity of the inhibitory agent or that will have an effect on carbonic anhydrase activity of CA IX.
  • the method comprises administration of multiple doses of antagonist anti-CA DC antibody or antigen-binding fragment thereof, or multiple doses of a small molecule inhibitory agent (e.g., peptide, peptoid, or other small molecule).
  • a small molecule inhibitory agent e.g., peptide, peptoid, or other small molecule
  • the method may comprise administration of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, or more therapeutically effective doses of a pharmaceutical composition comprising an antagonist anti-CA DC antibody or antigen-binding fragment thereof or a pharmaceutical composition comprising a small molecule inhibitory agent identified herein.
  • a pharmaceutical composition comprising an antagonist anti-CA DC antibody or antigen-binding fragment thereof or a pharmaceutical composition comprising a small molecule inhibitory agent identified herein.
  • the frequency and duration of administration of multiple doses of the particular pharmaceutical composition comprising an inhibitor of CA DC carbonic anhydrase activity can be readily determined by one of skill in the art.
  • treatment of a subject with a therapeutically effective amount of such an inhibitory agent can include a single treatment or, preferably, can include a series of treatments.
  • the effective dosage of antibody or antigen- binding fragment thereof or other CA DC inhibitory agent used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of diagnostic assays.
  • Co-administration means administration of an antagonist anti-CA DC antibody, an antigen-binding fragment thereof, or other agent that inhibits carbonic anhydrase activity of CA DC or biologically active variant thereof according to the treatment methods of the invention in combination with a second therapeutic agent.
  • the second therapeutic agent can be any therapeutic agent useful for treatment of the subject's condition.
  • Suitable second therapeutic agents include, but are not limited to, methotrexate, tamoxifen, nelandron, nilutamide, adriamycin, 5FU, interferons, chemokines such as secondary lymphoid-tissue chemokine (CCL21), interferons, and cytokines, including interleukin-2 (IL-2), IL-12, IL-13, and IL-15, and granulocyte-macrophage colony stimulating factor (GM-CSF), and the like.
  • the CA DC inhibitory agents can also be administered in combination with radiation therapy.
  • neoplastic cell proliferation and/or tumor growth using a tumor-associated antigen for the CA IX+ cancer undergoing treatment, or using a vaccine comprising such a tumor-associated antigen, as a second therapeutic agent used in conjunction with a therapeutic agent of the invention that inhibits carbonic anhydrase activity of CA DC or variant thereof is contemplated.
  • a first therapeutic agent can be a small molecule inhibitor of CA DC carbonic anhydrase activity
  • a second therapeutic agent can be an antisense or ribozyme molecule against CA DC that, when administered in a viral or nonviral vector, will facilitate a transcriptional inhibition of CA DC that will complement the inhibitory activity of the small molecule.
  • Peptides that are useful for generating a CA DC+ tumor-specific cytotoxic T-lymphocyte (CTL) response, and vaccines comprising these peptides are known in the art. See, for example, International Publication Nos. WO 01/98363 and WO 01/60317, herein inco ⁇ orated by reference. Also see U.S. Patent No.
  • Co-administration may be simultaneous, for example, by administering a mixture of the therapeutic agents, or may be accomplished by administration of the agents separately, such as within a short time period.
  • Co- administration also includes successive administration of an inhibitor of CA DC carbonic anhydrase activity and one or more of another therapeutic agent.
  • the second therapeutic agent or agents may be administered before or after the inhibitor of CA DC carbonic anhydrase activity.
  • the second therapeutic agent may also be an inhibitor of CA DC carbonic anhydrase activity, which has particular advantages when administered with the first inhibitor.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • Therapeutically effective doses of agents that inhibit carbonic anhydrase activity of CA DC on neoplastic or preneoplastic cells expressing this protein can be administered using any medically acceptable mode of administration that results in an effective amount of the inhibitory agent being presented to the cells that are expressing this protein without incurring unacceptable adverse side effects.
  • routes of administration include parenteral, e.g., intravenous, infusion, intradermal, subcutaneous, intramuscular, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration, h one embodiment, therapeutically effective doses of these therapeutic agents are injected directly into the tumor
  • peritumor site a site less than about 15 cm from an outer edge of the tumor.
  • Administration may be to one or more sites.
  • the therapeutically effective doses of these agents can be administered at multiple sites within a tumor and/or surrounding a tumor.
  • the antagonist anti-CA IX antibodies and other CA DC inhibitory agents of the invention can be inco ⁇ orated into an appropriate pharmaceutical composition that includes a pharmaceutically acceptable carrier for the agent.
  • the pharmaceutical carrier for the agents may be the same or different for each agent.
  • Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycohc acids, polymeric amino acids, amino acid copolymers, and inactive viruses in particles. Such carriers are well known to those of ordinary skill in the art.
  • Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • Pharmaceutically acceptable carriers in therapeutic compositions may contain liquids such as water, saline, glycerol, and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • the therapeutic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared.
  • Liposomes are included within the definition of a pharmaceutically acceptable carrier. Liposomes are described in U.S. Patent Nos. 5,422,120 and 4,762,915, International Publication Nos.
  • the pharmaceutically acceptable carrier or diluent may be combined with other agents to provide a composition either as a liquid solution, or as a solid form
  • composition e.g., lyophilized
  • parenteral or nonparenteral routes can be administered by parenteral or nonparenteral routes.
  • CA DC inhibitory agents of the present invention can be prepared and/or identified using the naturally occurring, full-length CA DC protein of interest, for example, human CA DC (SEQ ID NO:2, encoded by SEQ ID NO:l).
  • these inhibitory agents can be prepared and/or identified using a biologically active variant of the CA DC protein of interest, for example, a biologically active variant of human CA IX.
  • CA DC polypeptide variants refer to polypeptides or proteins derived from the native or naturally occurring CA DC protein, for example, human CA DC protein of SEQ ID NO:2.
  • CA DC polypeptide variants can be naturally occurring or can be generated by deletion or addition of one or more amino acids to the N-terminal and/or C-terminal end of the native protein; deletion or addition of one or more amino acids at one or more sites in the native protein; or substitution of one or more amino acids at one or more sites in the native protein.
  • Such variants include CA DC fragments comprising a functional carbonic anhydrase domain, allelic variants, muteins, homologous orthologues, analogues and fusions of native CA IX polypeptide sequences.
  • analogues is intended analogues of either a native CA DC protein or a fragment of a native CA IX protein that comprise a native CA DC sequence and structure having one or more amino acid substitutions, insertions, or deletions.
  • Peptides having one or more peptoids are also encompassed by the term analogues (WO 91/04282).
  • polypeptide variants of human CA DC of SEQ ID NO:2 which may be naturally occurring (e.g., allelic variants that occur at the CA DC locus) or recombinantly produced (for example, muteins).
  • Polypeptide variants of human CA DC also encompass homologues or orthologues of this native protein.
  • CA DC polypeptide encompasses the native or naturally occurring CA DC protein, for example, human CA DC, and biologically active variants of the native or naturally occurring CA IX protein, including CA DC fragments as noted below, where the biologically active variants have the functional and structural characteristics defined herein below.
  • Fragments or so-called "truncated forms" of a native or naturally occurring CA DC protein or of a CA DC polypeptide variant can be used to prepare and/or identify the inhibitory agents of the present invention as long as the fragments have a functional carbonic anhydrase domain as noted below.
  • CA DC fragments Functional fragments or truncated forms of a CA IX protein, or of a CA DC polypeptide variant, are referred to as "CA DC fragments," or "CA DC polypeptide fragments.”
  • CA DC fragments or “CA DC polypeptide fragments.”
  • These fragments or truncated forms of a CA DC protein or of a CA IX polypeptide variant are generated by removing amino acid residues from the full-length CA DC amino acid sequence (i.e., the sequence for the native CA DC protein or the sequence for the CA DC polypeptide variant), for example, using recombinant DNA techniques well known in the art and described elsewhere herein, and include N-terminal and C-terminal deletions of the full-length CA DC sequence.
  • CA DC fragments are directly or indirectly derived from a native or naturally occurring CA DC protein, they represent one type of CA DC polypeptide variant as noted herein above.
  • screening candidate inhibitory agents including candidate antagonist anti-CA DC antibodies, for their ability to inhibit CA DC carbonic anhydrase activity
  • the full-length CA DC polypeptide variants, and fragments or truncated forms of a full-length CA IX protein or of a full-length CA DC polypeptide variant should retain a functional carbonic anhydrase domain that provides for a carbonic anhydrase activity that is at least 20%, 25%, 30%, 35%, 40%, 45%, preferably at least 50%, 55%, 60%, 65%, 70%, 75%, more preferably at least 80% of the carbonic anhydrase activity of the naturally occurring CA DC protein, for example, human CA DC shown in SEQ ID NO:2.
  • Fragments of a CA DC protein or fragments of a CA DC polypeptide variant suitable for preparing the anti-CA DC antibodies of the invention will comprise at least one inhibitory epitope that can be recognized by an antagonist anti-CA DC antibody, and thus form an antibody-antigen complex in the presence of this antibody whereby carbonic anhydrase activity of the CA IX polypeptide fragment is inhibited.
  • Suitable epitopes to be included in a CA DC polypeptide fragment include those inhibitory epitopes comprising contiguous and/or discontinuous residues of the CA DC polypeptide fragment, for example, inhibitory epitopes comprising contiguous and/or discontinuous residues within the carbonic anhydrase domain.
  • the CA DC inhibitory agents of the invention identified using the methods of the invention are suitable for treating a subject having a cancer characterized by expression of CA DC, or expression of a naturally occurring allelic variant of CA DC.
  • CA DC proteins and CA DC polypeptide variants including CA DC fragments (derived from the native or naturally occurring CA DC protein or derived from a full-length CA DC polypeptide variant) that are useful in the methods of the invention may be modified further so long as they retain CA DC carbonic anhydrase activity as noted below. Further modifications include, but are not limited to, phosphorylation, substitution of non-natural amino acid analogues, and the like.
  • CA DC polypeptide variants have an amino acid sequence that shares at least 70%, generally at least 75%, 80%, 85%, preferably about 90% to 95% or more, and more preferably about 98% or more sequence identity to the native or naturally occurring CA DC protein, where sequence identity is determined as noted herein below.
  • the CA DC polypeptide variant is a variant of human CA DC
  • the CA DC polypeptide variant has an amino acid sequence that shares at least 70%, generally at least 75%, 80%, 85%, preferably about 90% to 95% or more, and more preferably about 98% or more sequence identity to human CA DC of SEQ ID NO:2.
  • suitable CA DC polypeptide variants for pu ⁇ oses of the present invention will have a functional carbonic anhydrase domain that shares at least 70%, generally at least 75%, 80%, 85%, preferably about 90% to 95% or more, and more preferably about 98% or more sequence identity to the carbonic anhydrase domain of human CA IX, i.e., residues 135-414 of SEQ ID NO:2.
  • a variant of the CA IX protein useful in preparing the compositions of the invention may differ from the native or naturally occurring CA DC protein, for example human CA DC of SEQ ID NO:2, by as few as 1-15, as few as 1-10, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue, so long as the functional carbonic anhydrase domain of the variant retains at least 70% sequence identity with residues 135-414 of SEQ ID NO:2.
  • the CA DC polypeptide variant will comprise a functional carbonic anhydrase domain that provides for a carbonic anhydrase activity that is at least 20%, 25%, 30%, 35%, 40%, 45%, preferably at least 50%, 55%, 60%, 65%, 70%, 75%, more preferably at least 80% of the carbonic anhydrase activity of the native CA DC protein.
  • sequences are aligned for optimal comparison pu ⁇ oses.
  • percent identity number of identical positions/total number of positions (e.g., overlapping positions) x 100).
  • a CA DC variant polypeptide having an amino acid sequence at least 95% "identical" to a reference CA DC amino acid sequence is intended that the amino acid sequence of the CA DC variant polypeptide is identical to the reference CA DC polypeptide sequence except that the polypeptide sequence of the CA IX variant can include up to five amino acid alterations per each 100 amino acids of the reference CA DC amino acid sequence.
  • These alterations of the CA DC reference sequence may occur at the amino or carboxy terminal positions of the reference CA DC polypeptide sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference CA DC polypeptide sequence or in one or more contiguous groups within the reference CA DC sequence.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For the pu ⁇ oses of the present invention, percent sequence identity is determined using the Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. The Smith- Waterman homology search algorithm is taught in Smith and Waterman (1981) Adv. Appl. Math. 2:482-489.
  • the CA DC polypeptide variants useful in preparing the compositions of the invention may be obtained by amino acid substitutions, deletions, truncations, and insertions.
  • Preferred CA DC polypeptides variants have one or more conservative amino acid substitutions of the polypeptide of SEQ ID NO:2.
  • conservative amino acid substitutions may be made at one or more amino acid residues.
  • substitutions are made at nonessential amino acid residues, and preferably involve 1-15 residues, 1-10 residues, including 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 residue substitutions.
  • a “nonessential" amino acid residue is a residue that can be altered from the wild-type sequence of a CA DC protein (e.g., the sequence of SEQ ID NO:2) without altering one of the biological activities, whereas an "essential" amino acid residue is required for a given biological activity.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • the contiguous segment of the variant amino acid sequence may have additional amino acid residues or deleted amino acid residues with respect to the reference amino acid sequence.
  • the contiguous segment used for comparison to the reference amino acid sequence will include at least 20 contiguous amino acid residues, and may be 30, 40, 50, or more amino acid residues. Corrections for sequence identity associated with conservative residue substitutions or gaps can be made (see Smith- aterman homology search algorithm, taught in Smith and Waterman (1981) Adv.
  • CA DC polypeptide variants useful in preparing and/or screening for the compositions of the invention may be isolated as naturally occurring variants, isolated after mutagenesis or recombinant manipulation, or be synthetically produced.
  • Naturally occurring allelic variants can be identified with the use of well-known molecular biology techniques, such as, for example, with polymerase chain reaction (PCR) and hybridization techniques. Methods for such manipulations are generally known in the art.
  • variants of the CA IX proteins can be prepared by mutagenesis or recombinant manipulations. Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Kunkel (1985) Proc. Natl. Acad. Sci. USA 82:488-492; Kunkel et al. (1987) Methods Enzymol. 154:367- 382; U.S. Patent No. 4,873,192; Walker and Gaastra, eds. (1983) Techniques in
  • CA DC polypeptides useful in preparing the antagonist anti-CA DC antibodies and identifying other inhibitory agents encompass both naturally occurring proteins as well as variations and modified forms thereof. Such variants will continue to possess the desired CA DC activity, i.e., carbonic anhydrase activity, so that specificity of these inhibitory agents can be tested against the variant polypeptide.
  • a "CA IX polypeptide variant,” including CA DC polypeptide fragments will exhibit at least 20%, 25%, 30%, 35%, 40%, 45%, preferably at least 50% of the CA DC carbonic anhydrase activity of the native CA DC polypeptide.
  • a variant of the CA DC polypeptide of SEQ ID NO:2 will exhibit at least 20%, 25%, 30%, 35%, 40%, 45%, preferably at least 50% of the carbonic anhydrase activity of the corresponding native CA DC polypeptide of SEQ ID NO:2. More typically, variants exhibit at least 60% of the native CA DC carbonic anhydrase activity; even more typically, variants exhibit at least 80% of the native CA DC carbonic anhydrase activity.
  • the deletions, insertions, and substitutions of the CA DC polypeptide sequences useful in preparing the CA DC inhibitory compositions disclosed herein are not expected to produce radical changes in the characteristics of the particular CA DC protein.
  • Specific amino acids within the CA domain that are involved in carbonic anhydrase activity of the CA DC polypeptide that are essential for carbonic anhydrase activity of CA DC can be identified by methods known in the art. Such methods include alanine-scanning mutagenesis, molecular evolution (Crameri et al. (1996) Nat. Biotechnol. 14(3):315-319; Crameri et al. (1998) Nature 15:288-291; Patten et al. (1997) Curr. Opin. Biotechnol. 8:724-733; Stemmer (1994) Proc. Natl. Acad. Sci.
  • the glu-tagged CA domain portion of the human CA DC protein used in the Examples below was expressed in a soluble form in if ⁇ cw/ov-V-.,-- -infected Tn5 cells.
  • Media containing the secreted, glu-tagged CA DC protein (CA IX-glu) was harvested and stored at -80°C.
  • Baculovirus media containing CA IX-glu was thawed (at room temperature) and made 1 mM PMSF. It was concentrated 6 to 7 fold on ice, on a 10 kD Minisette (Filtron) membrane.
  • An Anti Glu-tag Affi-Gel 10 (Biorad) affinity chromatography column was prepared in advance according to the manufacturer's instructions, using Protein G purified monoclonal antibodies. At 4°C, this column was equilibrated in the following equilibration buffer: PBS / 10% Glycerol / 1% Octyl Glucoside. The concentrated media was filtered on a 0.22 ⁇ M CN membrane (Nalgene). Complete, EDTA-free Protease Inhibitor Cocktail (Roche Diagnostics #1873580) supplemented with 5 ⁇ g/ml of Leupeptin, and 10 ⁇ g/ml of E-64, was added.
  • the full-length, glu-tagged human CA DC protein used in Examples below was expressed internally in B aculovirus-inf cted Tn5 cells.
  • Cells were harvested and stored at -80°C. Cells were thawed (at room temperature) and diluted 1/3 with the following lysis buffer: PBS / 10% Glycerol / 0.5% Triton-X, pH 7.4, with Complete, EDTA-free Protease h-hibitor Cocktail (Roche Diagnostics #1873580) supplemented with 5 ⁇ g/ml of Leupeptin, and 10 ⁇ g/ml of E-64. Cells were then lysed in a glass homogenizer on ice. Lysate was centrifuged at
  • the eluate was diafiltered into the following equilibration buffer #2: PBS / 10% Glycerol / 1% Octyl Glucoside, pH 7.4.
  • An Anti Glu-tag Affi-Gel 10 (Biorad) affinity chromatography column was prepared in advance according to the manufacturer's instructions, using Protein G purified monoclonal antibodies. At 4°C, this column was equilibrated in equilibration buffer #2. The diafiltered eluate was loaded on the Anti Glu-tag Affi-Gel 10 column.
  • the following low-throughput assay can be used to screen candidate test agents for their ability to inhibit carbonic anhydrase activity of CA DC.
  • This assay can be conducted using the full-length CA DC protein, or a partial-length CA DC polypeptide that includes a functional carbonic anhydrase domain.
  • the CA DC protein can be glu- tagged as well.
  • Isotype control sets and the M75 set should show no change in acidification kinetics compared to the spontaneous acidification sample (water + enzyme only; no Abs or inhibitors).
  • the Mafenide sample should inhibit (slow down) the rate of acidification.
  • the following high-throughput assay can be used to screen candidate test agents for their ability to inhibit carbonic anhydrase activity of CA DC.
  • the glu- tagged CA DC (CA DC-glu) is purified as noted in the examples above.
  • the full-length CA DC protein or a partial-length CA DC polypeptide that includes a functional carbonic anhydrase can be used.
  • the substrate used in the reaction is fluorescein diacetate (FDA), catalog # F-103 (available from Molecular Probes).
  • FDA fluorescein diacetate
  • catalog # F-103 available from Molecular Probes
  • the reference standard is Acetazolamide, catalog #A-9842
  • Protocol The following assay is carried out in a 384-well black polypropylene plate.
  • test sample 1.25 ⁇ l of test sample in 100% Dimethyl sulfoxide is added to each well.
  • Example 5 Soft Agar Assay for Detection of Inhibition of Carbonic Anhydrase
  • the following protocol can be used to test the ability of a candidate inhibitory agent for its ability to inhibit carbonic anhydrase activity of CA DC.
  • Alamar Blue is metabolized by the cells and reduced over time.
  • the first objective of this study was to determine if expression of the proteoglycan-like (PG) domain of human CA IX (residues 38-134 of SEQ ID NO:2) alone or the carbonic anhydrase (CA) domain of human CA DC (residues 135-414) alone is sufficient to confer a transformed phenotype to NIH-3T3 cells. If the CA domain alone was identified as being sufficient, the second objective was to determine if enzymatic activity is required to confer a transformed phenotype to NQ ⁇ - 3T3 cells.
  • PG proteoglycan-like
  • CA carbonic anhydrase
  • cDNA expression constructs encoding full-length human CA DC (SEQ ID NO:2; encoded by SEQ ID NO:l), the PG domain of human CA DC (residues 38-134 of SEQ ED NO:2), the CA domain of human CA DC (residues 135-414 of SEQ ID NO:2), or the CA domain of CA DC bearing mutations abrogating carbonic anhydrase enzymatic activity were made and transfected into NIH-3T3 cells.
  • the complete constructs were as follows:
  • CA-full Full-length CA DC construct (CA-full). This construct encoded the full-length human CA DC. The coding and translated polypeptide sequences are shown in SEQ ID NOS:l and 2, respectively. Proteoglycan-like (PG) domain construct (CA-PG).
  • This construct encoded the signal peptide (residues 1-37 of SEQ ID NO:2), the PG domain (residues 38-131 of SEQ ID NO:2, which includes most of the PG domain of SEQ ID NO:2 and encompasses the entire functional region of the PG domain, i.e., residues 53-111 of SEQ ID NO:2), a tri-peptide serine linker (included for technical reasons), the transmembrane domain (residues 415-433 of SEQ ID NO:2), and the intracellular or intracytoplasmic domain (residues 434-459 of SEQ ID NO:2).
  • the coding and translated polypeptide sequences are shown in SEQ ID NOS: 3 and 4, respectively.
  • CA-CAH Carbonic anhydrase domain construct
  • This construct encoded the signal peptide (residues 1-37 of SEQ ID NO:2), the CA domain (residues 135-414 of SEQ ID NO:2), the transmembrane domain (residues 415-433 of SEQ TD NO:2), and the intracellular or intracytoplasmic domain (residues 434-459 of SEQ ID NO:2).
  • the coding and polypeptide sequences are shown in SEQ ID NOS: 5 and 6, respectively.
  • This construct encoded the signal peptide (residues 1-37 of SEQ ID NO:2), a short series of residues from the non-functional region of the PG domain (residues 38-45 of SEQ ID NO:2), a single alanine linker (included for technical reasons), residue 47 of SEQ ID NO:2 (from the non-functional region of the PG domain), the glu tag (EYMPME), a short series of residues from the non-functional region of the PG domain (residues 123-134 of SEQ ID NO:2), the CA domain (residues 135-414 of SEQ JD NO:2), the transmembrane domain (residues 415-433 of SEQ ID NO:2), and the intracellular or intracytoplasmic domain (residues 434-459 of SEQ ID NO:2).
  • the coding and polypeptide sequences are shown in SEQ ID NOS : 7 and 8, respectively.
  • CA-CAHglumut Glu-tagged mutant CA domain construct
  • This construct encoded the same amino acid sequence as the glu-tagged CA domain construct with the exception of having the three histidine residues obligatory for carbonic anhydrase activity (i.e., residues 226, 228, and 251 of SEQ ID NO:2) mutated to three glutamine residues.
  • the coding and polypeptide sequences are shown in SEQ ID NOS:9 and 10, respectively.
  • transfected cells an untransfected negative control cell line, and an src- transfected positive control cell line were then tested for their ability to grow in soft agar using the soft agar assay described in Example 5. Results are shown in Table 1 below.
  • protein-based therapeutics such as antibodies which inhibit CA DC carbonic anhydrase activity, or small molecule based therapeutics that inhibit CADC carbonic anhydrase activity, may be effective therapeutic agents for cancers expressing CA DC.
  • Table 1 shows results of the soft agar assay for the indicated cell lines. The units presented are fluorescence units. Results and SD are shown from four separate experiments. Hours 0.1 0.5 1 2.5 3.25 3.75 6.5 23 46 50 93.5 118
  • the antigen used in the ELISA test was a glu-tagged recombinant protein representing the CA domain portion of human CA IX ("CA-glu” protein), isolated as described in Example 1.
  • CA-glu protein was suspended in phosphate-buffered saline (PBS) at a concentration of 1 ⁇ g/ml, and 100 ⁇ l/well of this solution was dispensed into 96-well, flat-bottomed plates (hmmulon 4 HBX). After incubation of the plates overnight at 4°C, the protein solution was removed and the wells were washed with PBST (PBS + 0.05% Tween-20).
  • PBS phosphate-buffered saline
  • the wells received 200 ⁇ l/well of Blocking Buffer (3% Carnation non-fat dry milk in PBS + 0.01% Tween-20), and the plates were incubated for 1 - 2 hours at room temperature.
  • the wells were again washed with PBST, and each well then received 100 ⁇ l of a solution containing an antibody or hybridoma supernatant to be tested.
  • the solutions consisted of 5 - 50 ⁇ l of supernatant brought up to a final volume of 100 ⁇ l with Blocking Buffer. After incubation of the plates for 1 hour at room temperature, the wells were washed with PBST.
  • a secondary antibody solution horseradish peroxidase-conjugated rabbit anti-mouse IgG (gamma); Zymed #61-6020; diluted 1 :4000 in Blocking Buffer
  • 100 ⁇ l/well of ODP peroxidase substrate Sigma #P-9187; prepared according to the manufacturer's instructions
  • Example 8 Fluorescence- Activated Cell Sorting (FACS) Assay to Detect Antibody Reactivity Against Cell-Surface CA IX
  • the cell type utilized in the assay was the HT29 cell line (ATCC #HTB-38), derived from a human colorectal adenocarcinoma.
  • ATCC #HTB-38 derived from a human colorectal adenocarcinoma.
  • the cells were detached from tissue culture plates by treatment for approximately 30 minutes with 3 mM EDTA. The cells were collected by centrifugation and then resuspended in cold incubation buffer (PBS containing 1% bovine serum albumin).
  • Antibodies or hybridoma supernatants to be tested were mixed with 2 - 5x10 ⁇ cells in cold incubation buffer in a final volume of 100 ⁇ l.
  • As a negative control cells were mixed with 10 ⁇ g/ml of nonspecific mouse IgG2b; for a positive control, the cells were mixed with 10 ⁇ g/ml of the anti-CA IX monoclonal antibody M75 (Pastorekova et al. (1992) Virology 187:620-626).
  • the cells were collected by centrifugation, washed three times with cold incubation buffer, and mixed with a detecting antibody (goat anti-mouse IgG antibody conjugated with phycoerythrin; CALTAG M30004-4; 0.2 ⁇ g in 100 ⁇ l cold incubation buffer per cell sample).
  • a detecting antibody goat anti-mouse IgG antibody conjugated with phycoerythrin; CALTAG M30004-4; 0.2 ⁇ g in 100 ⁇ l cold incubation buffer per cell sample.
  • the secondary antibody After incubation of the cells with the secondary antibody for 30 min at 4°C, the cells were again collected by centrifugation and washed three times with cold incubation buffer.
  • Each cell sample was resuspended in 300 - 500 ⁇ l of incubation buffer containing 0.5 ⁇ l/sample of propidium iodide solution (Roche #1 348 639). The samples were then analyzed utilizing a FACScaliber FACS
  • mice Female BALB/c mice were first immunized with cells expressing full-length recombinant human CA IX. To produce the cells for the immunization, Tn5 insect cells were infected with a recombinant baculovirus encoding the human CA IX protein. Two days after the infection, the cells were collected by centrifugation and resuspended in PBS to a final concentration of 5x10 ⁇ cells/ml. The cell suspension was emulsified with an equal volume of Freund's Incomplete Adjuvant (Sigma, F- 5506), and 0.2 ml of the emulsion was injected intraperitoneally into each mouse.
  • Freund's Incomplete Adjuvant Sigma, F- 5506
  • mice were then boosted every 2 - 3 weeks with an intraperitoneal injection of 30 ⁇ g/mouse of recombinant CA-glu protein.
  • the CA-glu protein was emulsified with an equal volume of Freund's Complete Adjuvant (Sigma, F-5881) prior to injection.
  • the protein was emulsified with Freund's Incomplete Adjuvant prior to injection.
  • Eight days following each boost blood was collected from each animal, serum was generated from the blood samples, and the sera were tested for the level of antibodies recognizing the CA-glu protein (titer) using the ELISA described in Example 7.
  • Control wells in the ELISA received either nonspecific mouse IgG2b rather than hybridoma supernatant (negative control for assay background), or samples of hybridoma supernatants previously shown to contain anti-CA-glu antibodies (positive controls). Selected supernatants were also assayed utilizing the FACS protocol described in Example 8.
  • Example 10 Antigenic Region Analysis of Human CA IX
  • the full-length human CA IX protein and the carbonic anhydrase domain i.e., residues 135-414 of SEQ ID NO: 2
  • the antigencity plots for both sequences predicted an antigenic region within the carbonic anhydrase domain, where the 28- residue region of antigenicity corresponded to residues 229 to 256 of SEQ ID NO:2 (data not shown).

Landscapes

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

Abstract

L'invention concerne des compositions et des procédés utiles pour inhiber la prolifération de cellules prénéoplastique ou néoplastiques CA IX + chez un mammifère. Ces compositions sont des anticorps anti-CA IX et d'autres agents inhibiteurs qui ciblent l'activité d'anhydrase carbonique sur ces cellules. Ces anticorps anti-CA IX antagonistes ou fragments de liaison d'antigène de ces derniers réagissent de manière spécifique avec un épitope inhibiteur de CA IX ou un variant biologiquement actif de celui-ci. La formation d'un complexe anticorps-antigène entre les anticorps anti-CA IX antagonistes ou les fragments de liaison d'antigène de ce dernier et les épitopes inhibiteurs respectifs, entraîne l'inhibition de l'activité d'anhydrase carbonique de CA IX ou d'un variant biologiquement actif de ce dernier. L'invention porte sur d'autres agents à petites molécules qui inhibent l'activité d'anhydrase carbonique de CA IX ou d'un variant biologiquement actif de ce dernier et sur des dosages de criblage permettant d'identifier ces agents. Les anticorps anti-CA IX antagonistes, les fragments de liaison d'antigène de ce dernier, et d'autres agents inhibiteurs de CA IX susmentionnés sont utiles dans le traitement de cancers caractérisés par l'expression de l'antigène CA IX associé aux tumeurs.
EP03749129A 2002-08-23 2003-08-25 Compositions et procedes therapeutiques contre les cancers caracterises par l'expression de l'antigene mn/ca ix associe aux tumeurs Withdrawn EP1576010A4 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US40557702P 2002-08-23 2002-08-23
US405577P 2002-08-23
US64693403A 2003-08-22 2003-08-22
US646934 2003-08-22
PCT/US2003/026612 WO2004017923A2 (fr) 2002-08-23 2003-08-25 Compositions et procedes therapeutiques contre les cancers caracterises par l'expression de l'antigene mn/ca ix associe aux tumeurs

Publications (2)

Publication Number Publication Date
EP1576010A2 true EP1576010A2 (fr) 2005-09-21
EP1576010A4 EP1576010A4 (fr) 2006-12-27

Family

ID=31949897

Family Applications (1)

Application Number Title Priority Date Filing Date
EP03749129A Withdrawn EP1576010A4 (fr) 2002-08-23 2003-08-25 Compositions et procedes therapeutiques contre les cancers caracterises par l'expression de l'antigene mn/ca ix associe aux tumeurs

Country Status (5)

Country Link
EP (1) EP1576010A4 (fr)
JP (1) JP2005536220A (fr)
AU (1) AU2003268180A1 (fr)
CA (1) CA2496572A1 (fr)
WO (1) WO2004017923A2 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2477163C (fr) 2002-02-21 2010-07-27 Institute Of Virology Forme soluble d'anhydrase carbonique ix- (s-ca-ix), dosage servant a detecter la co-expression de s-ca ix et ca ix's en utilisant le her-2/neu/c-erbb-2, et des anticorps monoclonales ca ix-specifique a des epitopes non-immunodominants
US7833734B2 (en) 2002-11-26 2010-11-16 Institute Of Virology Of The Slovak Academy Of Sciences CA IX-specific inhibitors
DOP2006000277A (es) * 2005-12-12 2007-08-31 Bayer Pharmaceuticals Corp Anticuerpos anti mn y métodos para su utilización
WO2008091798A2 (fr) * 2007-01-22 2008-07-31 Xencor, Inc. Anticorps de ca9 optimises et methodes d'utilisation associees
AU2008350907A1 (en) * 2007-12-11 2009-08-27 Viamet Pharmaceuticals, Inc. Metalloenzyme inhibitors using metal binding moieties in combination with targeting moieties
EP3966253A4 (fr) * 2019-05-06 2023-02-22 Navi Bio-Therapeutics, Inc. Anticorps anti-anhydrase carbonique ix

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000024913A2 (fr) * 1998-10-23 2000-05-04 Bayer Corporation Gene et proteine mn

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5955075A (en) * 1992-03-11 1999-09-21 Institute Of Virology, Slovak Academy Of Sciences Method of inhibiting tumor growth using antibodies to MN protein

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000024913A2 (fr) * 1998-10-23 2000-05-04 Bayer Corporation Gene et proteine mn

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J. SAARNIO ET EL.: "Immunohistochemical study of colorectal tumors for expression of a novel transmembrane carbonic anhydrase, MN/CA IX, with potential value as a marker of cell proliferation." AMERICAN JOURNAL OF PATHOLOGY, vol. 153, no. 1, 1998, pages 279-285, *
See also references of WO2004017923A2 *

Also Published As

Publication number Publication date
AU2003268180A8 (en) 2004-03-11
WO2004017923A3 (fr) 2005-07-14
AU2003268180A1 (en) 2004-03-11
CA2496572A1 (fr) 2004-03-04
WO2004017923A2 (fr) 2004-03-04
JP2005536220A (ja) 2005-12-02
EP1576010A4 (fr) 2006-12-27

Similar Documents

Publication Publication Date Title
AU2017216513B2 (en) Antibodies against claudin 18.2 useful in cancer diagnosis
US20240117055A1 (en) Antibodies that bind to axl proteins
JP5607530B2 (ja) ポリペプチド並びにポリヌクレオチド、並びに薬剤および生物製剤生産のための薬剤標的としてのその利用
CA2988115A1 (fr) Anticorps anti-gitr pour le diagnostic du cancer
CN107250160B (zh) 人源化cc趋化因子受体4 (ccr4)抗体及其使用方法
JP2009531031A (ja) 抗−腫瘍細胞抗原抗体療法
JP2004520806A (ja) 腫瘍の診断と治療のための組成物と方法
JP2004537534A (ja) 腫瘍の診断及び治療のための組成物及び方法
BRPI0613642A2 (pt) anticorpo, hibridoma murino, ácido nucleico isolado, vetor, célula hospedeira, animal transgênico, processo para a produção de um enticorpo ou um de seus fragmentos funcionais, composição, uso de um anticorpo ou um de seus fragmentos funcionais, métodos de diagnóstico para prognosticar um potencial oncogênico de uma amostra de células da próstata, e para dar seguimento ao processo de um regime terapêutico projetado para aliviar um distúrbio patológico, kit ou conjunto para a realização de um método de diagnóstico de doença, e, métodos para modular e diminuir a atividade de igf-ir em células de mamìferos responsivas ao igf-ir, para identificar um modulador de igf-ir, para detectar um parceiro de ligação para o anticorpo, e para purificar um igf-ir a partir de uma amostra
AU2013251312A1 (en) Anti-GCC antibody molecules and use of same to test for susceptibility to GCC-targeted therapy
US20240076390A1 (en) Antibodies that bind to axl proteins
JP2009527507A (ja) Cd63の表面発現を示す細胞に対する細胞傷害性介入
EP2285403A2 (fr) Anticorps monoclonaux humains dirigés contre le récepteur de chimiokines humaines ccr7
KR20090029276A (ko) 헵신을 표적화하기 위한 방법 및 조성물
EP2160412A1 (fr) Anticorps anti-cd59 humanisés et chimériques qui facilitent la toxicité de cellules cancéreuses
JP2004520810A (ja) 腫瘍の診断と治療のための組成物と方法
CA3203257A1 (fr) Anticorps anti-b7-h3 et ses utilisations
US20090252738A1 (en) Compositions and methods of therapy for cancers characterized by expression of the tumor-associated antigen mn/ca ix
WO2004017923A2 (fr) Compositions et procedes therapeutiques contre les cancers caracterises par l'expression de l'antigene mn/ca ix associe aux tumeurs
EP4265269A1 (fr) Utilisation de protéine taci
JP2015042173A (ja) ポリペプチド並びにポリヌクレオチド、並びに薬剤および生物製剤生産のための薬剤標的としてのその利用
CN115515625A (zh) 与人类ceacam1/3/5特异性结合的新抗体及其用途
US20210403602A1 (en) Anti-ido antibody and uses thereof
JP2004520808A (ja) 腫瘍の診断と治療のための組成物と方法

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20050315

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20061128

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: NOVARTIS VACCINES AND DIAGNOSTICS, INC.

17Q First examination report despatched

Effective date: 20090602

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

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20091123