WO2014165506A1 - Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas - Google Patents

Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas Download PDF

Info

Publication number
WO2014165506A1
WO2014165506A1 PCT/US2014/032513 US2014032513W WO2014165506A1 WO 2014165506 A1 WO2014165506 A1 WO 2014165506A1 US 2014032513 W US2014032513 W US 2014032513W WO 2014165506 A1 WO2014165506 A1 WO 2014165506A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
antibodies
pancreatic cancer
pam4
Prior art date
Application number
PCT/US2014/032513
Other languages
English (en)
Inventor
Donglin Liu
David V. Gold
Chien-Hsing Chang
Serengulam V. Govindan
David M. Goldenberg
Original Assignee
Immunomedics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US14/036,765 external-priority patent/US8795662B2/en
Application filed by Immunomedics, Inc. filed Critical Immunomedics, Inc.
Priority to CN201480019479.1A priority Critical patent/CN105074469A/zh
Priority to CA2899811A priority patent/CA2899811A1/fr
Priority to EP14778990.3A priority patent/EP2981829A4/fr
Publication of WO2014165506A1 publication Critical patent/WO2014165506A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/303Liver or Pancreas
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [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 against structure-related tumour-associated moieties
    • C07K16/3092Immunoglobulins [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 against structure-related tumour-associated moieties against tumour-associated mucins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4725Mucins, e.g. human intestinal mucin

Definitions

  • This invention relates to anti-pancreatic cancer antibodies and antigen-binding fragments thereof that bind to MUC5ac mucin in pancreatic cancer. More preferably, the antibodies or fragments thereof bind to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052).
  • the subject antibodies or antibody fragments bind with high selectivity to pancreatic cancer cells to allow detection and/or diagnosis of pancreatic adenocarcinoma at the earliest stages of the disease. Most preferably, antibody -based assays are capable of detecting about 85% or more of pancreatic adenocarcinomas, with a false positive rate of about 5% or less for healthy controls.
  • the methods and compositions can be used to detect and/or diagnose pancreatic adenocarcinoma by screening serum samples from subjects and preferably can detect 60% or more of stage I pancreatic cancers and 80% or more of stage II cancers by serum sample analysis.
  • immunoassay with an anti-MUC5ac antibody may be combined with immunodetection using other pancreatic cancer markers, such as CA19.9, to provide improved detection rates for pancreatic cancer without decreasing specificity.
  • reactivity with the anti-pancreatic cancer antibody can be used to detect occult pancreatic cancer or neoplastic precursor lesions against a background of pancreatitis or benign pancreatic hyperplasia.
  • the anti-pancreatic cancer antibody binds to the same epitope as, or competes for binding to MUC5ac with a PAM4 antibody comprising the light chain variable region complementarity-determining region (CDR) sequences CDRl
  • the anti-pancreatic cancer antibody is a humanized PAM4 (hPAM4) antibody comprising the light chain CDR sequences CDRl (SASSSVSSSYLY, SEQ ID NO: l); CDR2 (STSNLAS, SEQ ID NO:2); and CDR3 (HQWNRYPYT, SEQ ID NO:3); and the heavy chain CDR sequences CDRl (SYVLH, SEQ ID NO:4); CDR2 (YINPYNDGTQYNEKFKG, SEQ ID NO:5) and CDR3 (GFGGSYGFAY, SEQ ID NO:6), along with human antibody framework (FR) and constant region sequences.
  • hPAM4 humanized PAM4
  • Pancreatic cancer is a malignant growth of the pancreas that mainly occurs in the cells of the pancreatic ducts. This disease is the ninth most common form of cancer, yet it is the fourth and fifth leading cause of cancer deaths in men and women, respectively.
  • the number of patients who succumb to pancreatic cancer each year continues to rise, unlike other leading cancers where surveillance and/or screening technologies have led to a decrease in cancer- related mortality rates (Jemal et al, 2009, CA Cancer J Clin 59:225-49).
  • the overall survival rate is only 20% after one year and less than 4% after 5 years.
  • the major reasons for this poor prognosis include the inability to detect the disease at an early- stage, when curative measures may have greater opportunity to provide successful outcomes, and the lack of an effective treatment for advanced disease.
  • pancreatic cancer The most common symptoms of pancreatic cancer include jaundice, abdominal pain, and weight loss, which, together with other presenting factors, are nonspecific in nature. Thus, diagnosing pancreatic cancer at an early stage of tumor growth is often difficult and requires extensive diagnostic work-up, often times including exploratory surgery.
  • Endoscopic ultrasonography and computed tomography are the best noninvasive means available today for diagnosis of pancreatic cancer.
  • reliable detection of small tumors, as well as differentiation of pancreatic cancer from focal pancreatitis is difficult.
  • the vast majority of patients with pancreatic cancer are presently diagnosed at a late stage when the tumor has already extended outside of the capsule to invade surrounding organs and/or has metastasized extensively. (Gold et al., 2001, Crit. Rev. Oncology/Hematology, 39: 147-54).
  • pancreatic cancer Early detection and diagnosis of pancreatic cancer, as well as appropriate staging of the disease, would provide an increased survival advantage.
  • a number of laboratories have attempted to develop a diagnostic procedure based upon the release of a tumor-associated marker into the bloodstream, as well as detection of the marker substance within biopsy specimens.
  • the best previously -characterized tumor associated marker for pancreatic cancer has been the immunoassay for CA19.9. Elevated levels of this sialylated Le a epitope structure were found in 70% of pancreatic cancer patients but were not found in any of the focal pancreatitis specimens examined. However, CA19.9 levels were found to be elevated in a number of other malignant and benign conditions, and currently the assay cannot be used for diagnosis.
  • the assay is useful for monitoring, with continued increase in CA19.9 serum levels after surgery being indicative of a poor prognosis.
  • Many other monoclonal antibodies have been reported with immunoassays for diagnosis in varying stages of development. These include but are not limited to DUPAN2, SPAN1, B72.3, Ia3, and various anti-CEA (carcinoembryonic antigen, or CEACAM5) antibodies.
  • Antibodies in particular MAbs and engineered antibodies or antibody fragments, have been tested widely and shown to be of value in detection and treatment of various human disorders, including cancers, autoimmune diseases, infectious diseases, inflammatory diseases, and cardiovascular diseases [Filpula and McGuire, Exp. Opin. Ther. Patents (1999) 9: 231-245].
  • the clinical utility of an antibody or an antibody-derived agent is primarily dependent on its ability to bind to a specific targeted antigen associated with a particular disorder.
  • Selectivity is valuable for delivering a diagnostic or therapeutic agent, such as drugs, toxins, cytokines, hormones, hormone antagonists, enzymes, enzyme inhibitors, oligonucleotides, growth factors, radionuclides, angiogenesis inhibitors or metals, to a target location during the detection and treatment phases of a human disorder, particularly if the diagnostic or therapeutic agent is toxic to normal tissue in the body.
  • Radiolabeled antibodies have been used with some success in numerous malignancies, including ovarian cancer, colon cancer, medullary thyroid cancer, and lymphomas. This technology may also prove useful for pancreatic cancer.
  • previously reported antibodies against pancreatic cancer antigens have not been successfully employed to date for the effective therapy or early detection and/or diagnosis of pancreatic cancer.
  • pancreatic cancer-associated antigens in body fluids, particularly blood, can enable improved earlier diagnosis of this disease, so long as it differentiates well from benign diseases, and can also be used for monitoring response to therapy and potentially also to enhance prognosis by indicating disease burden.
  • the present invention concerns antibodies, antigen-binding antibody fragments and fusion proteins that bind to the MUC5ac pancreatic cancer mucin, preferably to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052). More preferably, the subject antibodies or fragments thereof bind specifically to pancreatic cancer cells, with little or no binding to normal or non-neoplastic pancreatic cells. The antibodies are capable of binding to the earliest stages of pancreatic cancer, with detection rates of about 50-60% for PanTN-lA, 70- 80% for Pan IB and 80-90% for PanI -2.
  • the antibodies bind to 80 to 90% or more of human invasive pancreatic adenocarcinoma, intraductal papillary mucinous neoplasia, PanI - ⁇ , PanI -lB and PanIN-2 lesions. Most preferably, the antibodies can distinguish between early stage pancreatic cancer and non-malignant conditions such as pancreatitis.
  • Such antibodies are of particular use for early detection of cancer and differential diagnosis between early stage pancreatic cancer and benign pancreatic conditions.
  • such antibodies are of use for in vivo or ex vivo analysis of samples from individuals suspected of having early stage pancreatic or certain other cancers. More preferably, the antibodies are of use for detection and diagnosis of early stage pancreatic cancer by analysis of serum samples.
  • the antibodies, antibody fragments or fusion proteins are capable of binding to synthetic peptide sequences, for example to phage display peptides, such as WTW ITKAYPLP (SEQ ID NO:7) and ACPEWWGTTC (SEQ ID NO:8).
  • synthetic peptides may be linear or cyclic and may or may not compete with antibody binding to the endogenous pancreatic cancer antigen.
  • Amino acids in certain positions of the synthetic peptide sequences may be less critical for antibody binding than others.
  • the residues K, A and L at positions 7, 8 and 11 of the peptide sequence may be varied while still retaining antibody binding.
  • SEQ ID NO: 8 the threonine residues at positions 8 and 9 of the sequence may be varied, although substitution of the threonine at position 9 may significantly affect antibody binding to the peptide.
  • Binding of the antibodies to a target pancreatic cancer antigen may be inhibited by treatment of the target antigen with reagents such as dithiothreitol (DTT) and/or periodate.
  • DTT dithiothreitol
  • binding of the antibodies to a pancreatic cancer antigen may be dependent upon the presence of disulfide bonds and/or the glycosylation state of the target antigen.
  • the epitope recognized by the subject antibodies is not cross-reactive with other reported mucin-specific antibodies, such as the MA5 antibody, the CLH2-2 antibody and/or the 45M1 antibody (see, e.g., Major et al, J Histochem Cytochem. 35: 139- 48, 1987; Dion et al, Hybridoma 10:595-610, 1991).
  • the subject antibodies or fragments may be naked antibodies or fragments or preferably are conjugated to at least one therapeutic and/or diagnostic agent for delivery of the agent to target tissues.
  • the subject antibodies or fragments may be part of a bispecific antibody with a first binding site for an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052) and a second binding site for a hapten conjugated to a targetable construct.
  • the targetable construct may in turn be attached to at least one therapeutic and/or diagnostic agent, of use in pretargeting techniques.
  • the subject antibody, antibody fragment or fusion protein is a humanized PAM4 antibody or fragment, comprising the light chain variable region CDR sequences CDR1 (SASSSVSSSYLY, SEQ ID NO: l); CDR2 (STSNLAS, SEQ ID NO:2); and CDR3 (HQWNRYPYT, SEQ ID NO: 3); and the heavy chain variable region CDR sequences CDR1 (SYVLH, SEQ ID NO:4); CDR2 (YINPYNDGTQYNEKFKG, SEQ ID NO:5) and CDR3 (GFGGSYGFAY, SEQ ID NO:6) and human antibody framework region (FR) and constant region sequences.
  • CDR1 SASSSVSSSYLY, SEQ ID NO: l
  • CDR2 STSNLAS, SEQ ID NO:2
  • CDR3 HQWNRYPYT, SEQ ID NO: 3
  • CDR1 SYVLH, SEQ ID NO:4
  • CDR2 YINPYNDGTQY
  • the FRs of the light and heavy chain variable regions of the humanized PAM4 antibody or fragment thereof comprise at least one amino acid substituted from amino acid residues 5, 27, 30, 38, 48, 66, 67 and 69 of the murine PAM4 heavy chain variable region (SEQ ID NO: 12) and/or at least one amino acid selected from amino acid residues 21, 47, 59, 60, 85, 87 and 100 of the murine PAM4 light chain variable region (SEQ ID NO: 10).
  • the antibody or fragment thereof comprises the hPAM4 VH amino acid sequence of SEQ ID NO: 19 and the hPAM4 VK amino acid sequence of SEQ ID NO: 16.
  • the anti-pancreatic cancer antibody may be a chimeric, humanized or human antibody that binds to the same antigenic determinant (epitope) as, or competes for binding to MUC5ac with, a chimeric PAM4 (cPAM4) antibody.
  • cPAM4 chimeric PAM4
  • the cPAM4 antibody is one that comprises the light chain variable region CDR sequences CDR1 (SASSSVSSSYLY, SEQ ID NO: l); CDR2 (STSNLAS, SEQ ID NO:2); and CDR3 (HQWNRYPYT, SEQ ID NO: 3); and the heavy chain variable region CDR sequences CDR1 (SYVLH, SEQ ID NO:4); CDR2 (YINPYNDGTQYNEKFKG, SEQ ID NO:5)and CDR3 (GFGGSYGFAY, SEQ ID NO:6).
  • Antibodies that bind to the same antigenic determinant may be identified by a variety of techniques known in the art, such as by competitive binding studies using the cPAM4 antibody as the competing antibody and human pancreatic mucin or MUC5ac as the target antigen.
  • Antibodies that block (compete for) binding to human pancreatic mucin by a cPAM4 antibody are referred to as cross- blocking antibodies.
  • cross-blocking antibodies are ones that bind to an epitope located within the second to fourth cysteine-rich domains of MUC5ac, or that compete for binding to amino acid residues 1575-2052 with a PAM4 antibody.
  • cancer cell-targeting therapeutic immunoconjugates comprising an antibody or fragment thereof or fusion protein bound to at least one therapeutic agent.
  • the therapeutic agent is selected from the group consisting of a radionuclide, an immunomodulator, a hormone, a hormone antagonist, an enzyme, an oligonucleotide such as an anti-sense oligonucleotide or a siRNA, an enzyme inhibitor, a photoactive therapeutic agent, a cytotoxic agent such as a drug or toxin, an angiogenesis inhibitor and a pro-apoptotic agent.
  • the therapeutic agents may comprise multiple copies of the same therapeutic agent or else combinations of different therapeutic agents.
  • an oligonucleotide such as an antisense molecule or siRNA inhibiting bcl-2 expression as described in U.S. Pat. No. 5,734,033 (the Examples section of which is incorporated herein by reference), may be attached to, or form the therapeutic agent portion of an immunoconjugate or antibody fusion protein.
  • the oligonucleotide may be administered concurrently or sequentially with a naked or conjugated anti-pancreatic cancer antibody or antibody fragment, such as a PAM4 antibody.
  • the oligonucleotide is an antisense oligonucleotide that is directed against an oncogene or oncogene product, such as bcl-2, p53, ras or other well-known oncogenes.
  • an oncogene or oncogene product such as bcl-2, p53, ras or other well-known oncogenes.
  • the therapeutic agent is a cytotoxic agent, such as a drug or a toxin.
  • the drug is selected from the group consisting of nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas, gemcitabine, triazenes, folic acid analogs, anthracyclines, taxanes, COX-2 inhibitors, pyrimidine analogs, purine analogs, antibiotics, enzyme inhibitors, epipodophyllotoxins, platinum coordination complexes, vinca alkaloids, substituted ureas, methyl hydrazine derivatives, adrenocortical suppressants, hormone antagonists, endostatin, taxols, camptothecins, SN-38, doxorubicins and their analogs, antimetabolites, alkylating agents, antimitotics, anti-angiogenic agents, tyrosine kinase inhibitors, Bruton t
  • the therapeutic agent is a toxin selected from the group consisting of ricin, abrin, alpha toxin, saporin, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin and combinations thereof.
  • an immunomodulator selected from the group consisting of a cytokine, a stem cell growth factor, a lymphotoxin, a hematopoietic factor, a colony stimulating factor (CSF), an interferon (IFN), a stem cell growth factor, erythropoietin, thrombopoietin and a
  • the therapeutic agent is an enzyme selected from the group consisting of malate dehydrogenase, staphylococcal nuclease, delta-V-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta- galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • malate dehydrogenase staphylococcal nuclease
  • delta-V-steroid isomerase yeast alcohol dehydrogenase
  • alpha-glycerophosphate dehydrogenase alpha-glycerophosphate dehydrogenase
  • triose phosphate isomerase horseradish peroxidas
  • Such enzymes may be used, for example, in combination with prodrugs that are administered in relatively non-toxic form and converted at the target site by the enzyme into a cytotoxic agent.
  • a drug may be converted into less toxic form by endogenous enzymes in the subject but may be reconverted into a cytotoxic form by the therapeutic enzyme.
  • Radionuclides such as 14 C, 13 N, 15 0, 32 P, 33 P, 47 Sc,
  • tyrosine kinase inhibitors include, but are not limited to canertinib, dasatinib, erlotinib, gefitinib, imatinib, lapatinib, leflunomide, nilotinib, pazopanib, semaxinib, sorafenib, sunitinib, sutent and vatalanib.
  • a specific class of tyrosine kinase inhibitor is the Bruton tyrosine kinase inhibitor.
  • Bruton tyrosine kinase (Btk) has a well- defined role in B-cell development.
  • Bruton kinase inhibitors include, but are not limited to, PCI- 32765 (ibrutinib), PCI-45292, GDC-0834, LFM-A13 and R 486.
  • the subject antibody or fragment may be conjugated to at least one diagnostic (or detection) agent.
  • the diagnostic agent is selected from the group consisting of a radionuclide, a contrast agent, a fluorescent agent, a chemiluminescent agent, a
  • the diagnostic agent is a radionuclide with an energy between 20 and 4,000 keV or is a radionuclide selected from the group consisting of 110 In, m In, 177 Lu, 18 F, 52 Fe, 62 Cu, 64 Cu, 67 Cu, 67 Ga, 68 Ga, 86 Y, 90 Y, 89 Zr, 94m Tc, 94 Tc, 99m Tc, 120 1, 123 I, 124 I, 125 1, 131 I, 154"158 Gd, 32 P, n C, 13 N, 15 0, 186 Re, 188 Re, 51 Mn, 52m Mn, 55 Co, 72 As, 75 Br, 76 Br, 82m Rb, 83 Sr, or other gamma-, beta-, or positron-emitters.
  • the diagnostic agent is a radionuclide with an energy between 20 and 4,000 keV or is a radionuclide selected from the group consisting of 110 In, m In, 177 Lu, 18 F,
  • the diagnostic radionuclide 18 F is used for labeling and PET imaging, as described in the Examples below.
  • the 18 F may be attached to an antibody, antibody fragment or peptide by complexation to a metal, such as aluminum, and binding of the 18 F-metal complex to a chelating moiety that is conjugated to a targeting protein, peptide or other molecule.
  • the diagnostic agent is a paramagnetic ion, such as chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III) and erbium (III), or a radiopaque material, such as barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric
  • the diagnostic agent is a fluorescent labeling compound selected from the group consisting of fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine, a chemiluminescent labeling compound selected from the group consisting of luminol, isoluminol, an aromatic acridinium ester, an imidazole, an acridinium salt and an oxalate ester, or a bioluminescent compound selected from the group consisting of luciferin, luciferase and aequorin.
  • the diagnostic immunoconjugates are used in intraoperative, endoscopic, or intravascular tumor diagnosis.
  • multivalent, multispecific antibodies or fragments thereof comprising at least one binding site that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052) and one or more hapten binding sites having affinity towards hapten molecules.
  • the antibody or fragment thereof is a chimeric, humanized or fully human antibody or fragment thereof.
  • the hapten molecule may be conjugated to a targetable construct for delivery of one or more therapeutic and/or diagnostic agents.
  • the multivalent antibodies or fragments thereof may be prepared by the DOCK-AND-LOCKTM (DNLTM) technique, as described below.
  • DNLTM construct incorporating hPAM4 antibody fragments is designated TF10, as described below.
  • a bispecific antibody or fragment thereof comprising at least one binding site with an affinity toward an epitope located within the second to fourth cysteine- rich domains of MUC5ac (amino acid residues 1575-2052) and at least one binding site with an affinity toward a targetable construct which is capable of carrying at least one diagnostic and/or therapeutic agent.
  • Targetable constructs suitable for use are disclosed, for example, in U.S. Patent Nos. 6,576,746; 6,962,702; 7,052,872; 7, 138, 103; 7, 172,751; 7,405,320;
  • fusion proteins comprising at least two anti-pancreatic cancer antibodies and fragments thereof as described herein.
  • the fusion protein or fragment thereof may comprise at least one first antibody or fragment thereof that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), and at least one second MAb or fragment thereof.
  • the second MAb binds to a tumor-associated antigen, for example selected from the group consisting of CA19.9, DUPAN2, SPAN1, Nd2, B72.3, CC49, CEA (CEACAM5),
  • CEACAM6, Le a the Lewis antigen Le(y), CSAp, insulin-like growth factor (IGF), epithelial glycoprotein- 1 (EGP-1), epithelial glycoprotein-2 (EGP-2), CD-80, placental growth factor (P1GF), carbonic anhydrase IX, tenascin, IL-6, HLA-DR, CD40, CD74 (e.g., milatuzumab), CD 138 (syndecan-1), MUC1, MUC2, MUC3, MUC4, MUC5ac, MUC16, MUC17, TAG-72, EGFR, platelet-derived growth factor (PDGF), angiogenesis factors (e.g., VEGF and P1GF), products of oncogenes (e.g., bcl-2, Kras, p53), cMET, HER2/neu, and antigens associated with gastric cancer and colorectal cancer.
  • IGF insulin-like growth factor
  • EBP-1 epithelial
  • the second MAb may also bind to a different epitope of MUC5ac than the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052).
  • the antibody fusion protein or fragments thereof may further comprise at least one diagnostic and/or therapeutic agent.
  • DNA sequences comprising a nucleic acid encoding an anti-pancreatic cancer antibody, fusion protein, multispecific antibody, bispecific antibody or fragment thereof as described herein.
  • Other embodiments concern expression vectors and/or host cells comprising the antibody-encoding DNA sequences.
  • the host cell may be an Sp2/0 cell line transformed with a mutant Bcl-2 gene, for example with a triple mutant Bcl-2 gene (T69E, S70E, S87E), that has been adapted to cell transformation and growth in serum free medium.
  • a mutant Bcl-2 gene for example with a triple mutant Bcl-2 gene (T69E, S70E, S87E)
  • T69E, S70E, S87E triple mutant Bcl-2 gene
  • Another embodiment concerns methods of delivering a diagnostic or therapeutic agent, or a combination thereof, to a target comprising (i) providing a composition that comprises an anti-pancreatic cancer antibody or fragment that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575- 2052), conjugated to at least one diagnostic and/or therapeutic agent and (ii) administering to a subject in need thereof the diagnostic or therapeutic conjugate of any one of the antibodies, antibody fragments or fusion proteins claimed herein.
  • the carrier molecule binds to more than one binding site of the antibody.
  • Described herein is a method for diagnosing or treating cancer, comprising: (a) administering to a subject any one of the multivalent, multispecific antibodies or fragments thereof claimed herein that have an affinity toward an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052) and comprise one or more hapten binding sites; (b) waiting a sufficient amount of time for an amount of the non-bound antibody to clear the subject's blood stream; and (c) administering to said subject a carrier molecule comprising a diagnostic agent, a therapeutic agent, or a combination thereof, that binds to a binding site of the antibody.
  • the cancer is pancreatic cancer.
  • the method can be used for intraoperative identification of diseased tissues, endoscopic identification of diseased tissues, or intravascular identification of diseased tissues.
  • Another embodiment is a method of treating a malignancy in a subject comprising administering to said subject a therapeutically effective amount of an antibody or fragment thereof that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), optionally conjugated to at least one therapeutic agent.
  • the antibody or fragment thereof may alternatively be a naked antibody or fragment thereof.
  • the antibody or fragment is administered either before, simultaneously with, or after administration of another therapeutic agent as described above.
  • [037] Contemplated herein is a method of diagnosing a malignancy in a subject, particularly a pancreatic cancer, comprising (a) administering to said subject a diagnostic conjugate comprising an antibody or fragment thereof that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), wherein said MAb or fragment thereof is conjugated to at least one diagnostic agent, and (b) detecting the presence of labeled antibody bound to pancreatic cancer cells or other malignant cells, wherein binding of the antibody is diagnostic for the presence of pancreatic cancer or another malignancy.
  • a diagnostic conjugate comprising an antibody or fragment thereof that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), wherein said MAb or fragment thereof is conjugated to at least one diagnostic agent, and (b) detecting the presence of labeled antibody bound to pancreatic cancer cells
  • the antibody or fragment binds to pancreatic cancer and not to normal pancreatic tissue, pancreatitis or other non-malignant conditions. In less preferred embodiments, the antibody or fragment binds at a significantly higher level to cancer cells than to non-malignant cells, allowing differential diagnosis of cancer from non- malignant conditions.
  • the diagnostic agent may be an F-18 labeled molecule that is detected by PET imaging.
  • the use of anti-pancreatic cancer antibodies that bind to an epitope located within the second to fourth cysteine-rich domains of MUC5ac allows the detection and/or diagnosis of pancreatic cancer with high specificity and sensitivity at the earliest stages of malignant disease.
  • the diagnostic antibody or fragment is capable of labeling at least 70%, more preferably at least 80%, more preferably at least 90%, more preferably at least 95%, most preferably about 100% of well differentiated, moderately differentiated and poorly differentiated pancreatic cancer and 90% or more of invasive pancreatic adenocarcinomas.
  • the anti-pancreatic cancer antibody of use is preferably capable of detecting 85% or more of PanIN-lA, PanIN-lB, PanTN-2, IPMN and MCN precursor lesions. Most preferably, immunoassays using the anti- pancreatic cancer antibody are capable of detecting 89% or more of total PanIN, 86% or more of IPMN, and 92% or more of MCN.
  • An alternative embodiment is a method of detecting the presence of PAM4-binding MUC5ac and/or diagnosing pancreatic cancer in an individual by in vitro analysis of blood, plasma or serum samples.
  • the sample is subjected to an organic phase extraction, using an organic solvent such as butanol, before it is processed for immunodetection using an anti-pancreatic cancer antibody, such as a PAM4 antibody.
  • organic phase extraction the extracted aqueous phase is analyzed for the presence of the epitope of MUC5ac to which PAM4 binds in the sample, using any of a variety of immunoassay techniques known in the art, such as ELISA, sandwich immunoassay, solid phase RIA, and similar techniques.
  • the organic phase extraction results in the removal of an inhibitor of PAM4 binding to MUC5ac, allowing detection of MUC5ac in fresh serum samples.
  • serum samples may be analyzed to detect and/or diagnose pancreatic cancer in a subject at the earliest stages of pancreatic adenocarcinoma.
  • Another embodiment is a method of treating a cancer cell in a subject comprising administering to said subject a composition comprising a naked antibody or fragment thereof or a naked antibody fusion protein or fragment thereof that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052).
  • the method further comprises administering a second naked antibody or fragment thereof selected from the group consisting of CA19.9, DUPAN2, SPANl, Nd2, B72.3, CC49, anti-CEA, anti-CEACAM6, anti-EGP-1, anti-EGP-2, anti-Le a , antibodies defined by the Lewis antigen Le(y), and antibodies against CSAp, MUC1, MUC2, MUC3, MUC4,
  • angiogenesis factors e.g., VEGF and placenta-like growth factor (P1GF), insulin-like growth factor (IGF), tenascin, platelet-derived growth factor, IL-6, products of oncogenes, cMET, and HER2/neu.
  • Still other embodiments concern a method of diagnosing a malignancy in a subject comprising (i) performing an in vitro diagnosis assay on a specimen from said subject with a composition comprising an antibody or fragment thereof that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575- 2052); and (ii) detecting the presence of antibody or fragment bound to malignant cells in the specimen.
  • the malignancy is a cancer. More preferably, the cancer is pancreatic cancer.
  • FIG. 1A Variable region cDNA sequences (SEQ ID NO: 9) and the deduced amino acid sequences (SEQ ID NO: 10) of the murine PAM4 Vk.
  • Amino acid sequences encoded by the corresponding DNA sequences are given as one-letter codes below the nucleotide sequence. Numbering of the nucleotide sequence is on the right side. The amino acid residues in the CDR regions are shown in bold and underlined. Kabat's Ig molecule numbering is used for amino acid residues as shown by the numbering above the amino acid residues. The amino acid residues numbered by a letter are the insertion residues defined by Kabat numbering scheme. The insertion residues have the same preceding digits as that of the previous residue.
  • FIG. IB Variable region cDNA sequence (SEQ ID NO: 11) and the deduced amino acid sequence (SEQ ID NO: 12) of the murine PAM4 VH.
  • Amino acid sequences encoded by the corresponding DNA sequences are given as one-letter codes below the nucleotide sequence. Numbering of the nucleotide sequence is on the right side. The amino acid residues in the CDR regions are shown in bold and underlined. Kabat's Ig molecule numbering is used for amino acid residues as shown by the numbering above the amino acid residues. The amino acid residues numbered by a letter are the insertion residues defined by Kabat numbering scheme. The insertion residues have the same preceding digits as that of the previous residue.
  • FIG. 2A Amino acid sequence (SEQ ID NO: 13) of the chimeric PAM4 (cPAM4) Vk. The sequences are given as one letter codes. The amino acid residues in the CDR regions are shown in bold and underlined. Kabat's Ig molecule number scheme is used to number the residues.
  • FIG. 2B Amino acid sequence (SEQ ID NO: 14) of the cPAM4 VH. The sequences are given as one letter codes. The amino acid residues in the CDR regions are shown in bold and underlined. Kabat's Ig molecule number scheme is used to number the residues.
  • FIG. 3 A Alignment of the VK amino acid sequences of the human antibody Walker (SEQ ID NO: 15) with PAM4 (SEQ ID NO: 10) and hPAM4 (SEQ ID NO: 16). Dots indicate the residues of PAM4 that are identical to the corresponding residues of the human or humanized antibodies. Boxed regions represent the CDR regions. Both N- and C-terminal residues (underlined) of hPAM4 are fixed by the staging vectors used. Kabat's Ig molecule number scheme is used to number the residues. [047] FIG. 3B.
  • FIG. 4A DNA (SEQ ID NO:20) and amino acid (SEQ ID NO: 16) sequences of the humanized PAM4 (hPAM4) Vk. Numbering of the nucleotide sequence is on the right side. Amino acid sequences encoded by the corresponding DNA sequences are given as one-letter codes. The amino acid residues in the CDR regions are shown in bold and underlined.
  • FIG. 4B DNA (SEQ ID NO:21) and amino acid (SEQ ID NO: 19) sequences of the hPAM4 VH. Numbering of the nucleotide sequence is on the right side. Amino acid sequences encoded by the corresponding DNA sequences are given as one-letter codes. The amino acid residues in the CDR regions are shown in bold and underlined. Kabat's Ig molecule numbering scheme is used for amino acid residues.
  • FIG. 5 Binding activity of humanized PAM4 antibody, hPAM4, as compared to the chimeric PAM4, cPAM4. hPAM4 is shown by diamonds and cPAM4 is shown by closed circles. Results indicate comparable binding activity of the hPAM4 antibody and cPAM4 when competing with 125 I-cPAM4 binding to CaPanl antigens.
  • FIG. 6 PET/CT fusion images for a patient with inoperable metastatic pancreatic cancer treated with fractionated 90 Y-hPAM4 plus gemcitabine, before therapy (left side) and post-therapy (right side).
  • the circle indicates the location of the primary lesion, which shows a significant decrease in PET/CT intensity following therapy.
  • FIG. 7 3D PET images for a patient with inoperable metastatic pancreatic cancer treated with fractionated 90 Y-hPAM4 plus gemcitabine, before therapy (left side) and post- therapy (right side). Arrows point to the locations of the primary lesion (on right) and metastases (on left), each of which shows a significant decrease in PET image intensity after therapy with radiolabeled hPAM4 plus gemcitabine.
  • FIG. 8A In vivo imaging of tumors using an m In-labeled diHSG peptide (IMP 288) with or without pretargeting TF10 bispecific anti-pancreatic cancer MUC5ac antibody.
  • FIG. 8A illustrates mice showing the location of tumors (arrow).
  • FIG. 8B In vivo imaging of tumors using an m In-labeled diHSG peptide (IMP 288) with or without pretargeting TF10 bispecific anti-pancreatic cancer MUC5ac antibody.
  • FIG. 8B shows the detected tumors with U 1 ln-labeled IMP 288 in the presence (above) or absence (below) of TF10 bispecific antibody.
  • FIG. 9 Exemplary binding curves for TF10, PAM4-IgG, PAM4-F(ab') 2 and a monovalent bsPAM4 chemical conjugate (PAM4-Fab' x anti-DTPA-Fab'). Binding to target mucin antigen was measured by ELISA assay.
  • FIG. 10A Immunoscintigraphy of CaPanl human pancreatic cancer xenografts (- 0.25 g). An image of mice that were injected with bispecific TF 10 (80 ⁇ g, 5.07 x 10 ⁇ 10 mol) followed 16 h later by administration of m In-IMP-288 (30 ⁇ , 5.07 x 10 _11 mol). The image was taken 3 h later. The intensity of the image background was increased to match the intensity of the image obtained when m In-IMP-288 was administered alone (30 ⁇ , 5.07 x 10 ⁇ n mol).
  • FIG. 10B Immunoscintigraphy of CaPanl human pancreatic cancer xenografts (- ⁇ 0.25 g). No targeting was observed in mice given u l In-IMP-288 alone.
  • FIG. IOC Immunoscintigraphy of CaPanl human pancreatic cancer xenografts ( ⁇ 0.25 g). An image of mice that were given u l In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) with imaging done 24 h later. Although tumors are visible, considerable background activity is still present at this time point.
  • FIG. 11A Extended biodistribution of m In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ , 5.07 x 10 "11 mol m In-IMP-288) in nude mice bearing CaPanl human pancreatic cancer xenografts (mean tumor weight +/- SD, 0.28 +/- 0.21 and 0.10 +/- 0.06 g for the pretargeting and IgG groups of animals, respectively) .
  • FIG. 11A Extended biodistribution of m In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ , 5.07 x 10 "11
  • 11A shows percent of initial dose per gram of tissue in tumor with PAM4 IgG (open circles), blood with PAM4 IgG (open squares), tumor with pretargeted peptide (closed circles) and blood with pretargeted peptide (closed squares).
  • FIG. 11B Extended biodistribution of u l In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ , 5.07 x 10 "11 mol m In-IMP-288) in nude mice bearing CaPanl human pancreatic cancer xenografts (mean tumor weight +/- SD, 0.28 +/- 0.21 and 0.10 +/- 0.06 g for the pretargeting and IgG groups of animals, respectively).
  • FIG. 11B Extended biodistribution of u l In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ , 5.07 x 10 "
  • 11B shows percent of initial dose per per gram of tissue in liver with PAM4 IgG (open triangles), kidney with PAM4 IgG (open diamonds), liver with pretargeted peptide (closed triangles) and kidney with pretargeted peptide (closed diamonds).
  • FIG. IIC Extended biodistribution of m In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ 3 ⁇ 4 5.07 x 10 "11 mol m In-IMP-288) in nude mice bearing CaPanl human pancreatic cancer xenografts (mean tumor weight +/- SD, 0.28 +/- 0.21 and 0.10 +/- 0.06 g for the pretargeting and IgG groups of animals, respectively).
  • 11C shows microcuries per gram of tissue in tumor with PAM4 IgG (open circles), blood with PAM4 IgG (open squares), tumor with pretargeted peptide (closed circles) and blood with pretargeted peptide (closed squares).
  • FIG. 11D Extended biodistribution of m In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ 3 ⁇ 4 5.07 x 10 "11 mol m In-IMP-288) in nude mice bearing CaPanl human pancreatic cancer xenografts (mean tumor weight +/- SD, 0.28 +/- 0.21 and 0.10 +/- 0.06 g for the pretargeting and IgG groups of animals, respectively).
  • FIG. 11D Extended biodistribution of m In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ g) and TFlO-pretargeted u l In-IMP-288 (80 ⁇ g, 5.07 x 10 "10 mol TF10 followed 16 h later with 30 ⁇ 3 ⁇ 4 5.07 x 10 "
  • 11D shows microcuries per per gram of tissue in liver with PAM4 IgG (open triangles), kidney with PAM4 IgG (open diamonds), liver with pretargeted peptide (closed triangles) and kidney with pretargeted peptide (closed diamonds).
  • FIG. 12 Therapeutic activity of a single treatment of established (-0.4 cm 3 ) CaPanl tumors with 0.15 mCi of 90 Y-hPAM4 IgG, or 0.25 or 0.50 mCi of TF lO-pretargeted 90 Y-IMP- 288.
  • FIG. 13 Effect of gemcitabine potentiation of PT-RAIT therapy.
  • FIG. 14 Effect of combination of cetuximab with gemcitabine and PT-RAIT.
  • FIG. 15 Differential diagnosis of pancreatic cancer using PAM4-based
  • the horizontal line shows the cutoff level selected for a positive result, based on ROC analysis.
  • FIG. 16 Frequency distribution of PAM4 antigen in patient sera from healthy volunteers and individuals with varying stages of pancreatic cancer.
  • FIG. 17 ROC curve for PAM4 serum immunoassay, showing sensitivity for detection of 81.6% and specificity of 84.6%.
  • FIG. 20 Receiver Operator Characteristics (ROC) curve for the performance of the PAM4-based immunoassay; pancreatic adenocarcinoma vs. healthy adults. Values for the area under the curves (AUC) and 95% confidence limits are provided.
  • ROC Receiver Operator Characteristics
  • FIG. 21A Circulating PAM4 antigen levels correlated with progression/regression of tumor volume (CT) following treatment with 90 Y-PAM4-IgG plus gemcitabine.
  • CT tumor volume
  • Patient 076- 001 was responsive to therapy and serum PAM4 antigen decreased.
  • Serum PAM4 levels correlated with tumor volume.
  • FIG. 21B Circulating PAM4 antigen levels correlated with progression/regression of tumor volume (CT) following treatment with 90 Y-PAM4-IgG plus gemcitabine.
  • CT tumor volume
  • Serum PAM4 levels correlated with tumor volume.
  • FIG. 22 Reactivity of PAM4 with mucin standards in the presence or absence of palmitic acid.
  • FIG. 23A Sensitivity and specificity for PAM4 detection of PDAC vs. chronic pancreatitis (CP).
  • FIG. 23B Sensitivity and specificity for PAM4 detection of PDAC vs. all benign tissue samples.
  • FIG. 24 Comparative labeling of PDAC vs. non-neoplastic prostate tissue with PAM4 vs. antibodies against MUC1, MUC4, CEACAM6 and CA19-9.
  • FIG. 25 Reactivity of several anti-mucin MAbs with a high molecular weight mucin containing fraction (CPM1) isolated from the Capan-1, human pancreatic adenocarcinoma. MAbs are identified by clone name with reactive species of mucin indicated by horizontal bars beneath MAb clone names. In addition to PAM4, substantial reactions were observed for anti-MUCl, MUC5ac, and CEACAM6 antibodies. All MAbs were employed at a constant 10 ⁇ g/mL.
  • CPM1 mucin containing fraction
  • FIG. 26 Reaction of several anti-mucin MAbs with PAM4-captured antigen. Mucin antigens were captured on hPAM4 coated plates, and then probed with several murine anti- mucin MAbs for reaction signal. Both anti-MUC5ac MAbs (2-1 1M1 and 45M1) bound to the hPAM4-captured mucin, whereas the anti-MUCl MAbs (MA5 and KC4) did not bind. The homologous hPAM4/mPAM4, capture/probe immunoassay gave no signal, suggesting the density of PAM4 epitopes within the mucin may be low, possibly only a single site. A rabbit polyclonal anti-CPMl, IgG, was used as a positive control for reaction with hPAM4- captured antigen.
  • FIG. 27A Inhibition of hPAM4/antigen binding reaction by murine anti-mucin MAbs.
  • Anti-mucin mMAbs purified IgG
  • IC max 25.5%
  • Neither 2-1 1M1, anti-MUC5ac nor MA5 and KC4, anti- MUC1 MAbs were able to inhibit the specific hPAM4/antigen reaction.
  • FIG. 27B Inhibition of hPAM4/antigen binding reaction by murine anti-mucin MAbs.
  • a similar inhibition study was performed with several anti-MUC5ac MAbs obtained as ascites fluids.
  • MAbs 21M1, 62M1, and 463M1, anti-MUC5ac provided substantial inhibitory affect similar to that observed with mPAM4, IgG, self-inhibition.
  • the ascites form of 45M1 yielded an inhibitory affect similar to that of the purified IgG.
  • Ascites containing anti-alpha fetoprotein was employed as a negative control.
  • FIG. 28 Representation of the domains of the MUC5ac glycoprotein with reactive epitopes indicated for several anti-MUC5ac MAbs.
  • Specific blocking studies suggest the PAM4-epitope resides within the cysteine-rich C-terminus domain.
  • the term "PAM4 antibody” includes murine, chimeric, humanized and human PAM4 antibodies.
  • the PAM4 antibody or antigen-binding fragment thereof comprises the CDR sequences of SEQ ID NO: l to SEQ ID NO:6.
  • an “anti-pancreatic cancer antibody” is an antibody that exhibits the same diagnostic, therapeutic and binding characteristics as the PAM4 antibody.
  • the "anti-pancreatic cancer antibody” binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052).
  • a “non-endocrine pancreatic cancer” generally refers to cancers arising from the exocrine pancreatic glands.
  • the term excludes pancreatic insulinomas and includes pancreatic carcinoma, pancreatic adenocarcinoma, adenosquamous carcinoma, squamous cell carcinoma and giant cell carcinoma and precursor lesions such as pancreatic intra-epithelial neoplasia (Panl ), mucinous cyst neoplasms (MCN) and intrapancreatic mucinous neoplasms (IPMN), which are neoplastic but not yet malignant.
  • Pancreatic cancer and “non-endocrine pancreatic cancer” are used interchangeably herein.
  • An antibody refers to a full-length (i.e., naturally occurring or formed by normal immunoglobulin gene fragment recombinatorial processes)
  • immunoglobulin molecule e.g., an IgG antibody
  • immunologically active (i.e., specifically binding) portion of an immunoglobulin molecule like an antibody fragment.
  • an antibody fragment is a portion of an antibody such as F(ab')2, Fab', Fab, Fv, sFv and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the full-length antibody.
  • antibody fragment also includes isolated fragments consisting of the variable regions of antibodies, such as the "Fv” fragments consisting of the variable regions of the heavy and light chains and recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker ("scFv proteins").
  • Another form of antibody fragment is a single domain antibody (nanobody).
  • a naked antibody is an antibody or fragment thereof that is not conjugated to a therapeutic or diagnostic agent.
  • the Fc portion of the antibody molecule provides effector functions, such as complement-mediated cytotoxicity (CDC) and ADCC (antibody- dependent cellular cytotoxicity), which set mechanisms into action that may result in cell lysis.
  • CDC complement-mediated cytotoxicity
  • ADCC antibody- dependent cellular cytotoxicity
  • the Fc portion may not be required for therapeutic function, with other mechanisms, such as signaling-induced apoptosis, coming into play.
  • naked antibodies include both polyclonal and monoclonal antibodies, as well as fusion proteins and certain recombinant antibodies, such as chimeric, humanized or human antibodies.
  • a chimeric antibody is a recombinant protein that contains the variable domains including the complementarity determining regions (CDRs) of an antibody derived from one species, preferably a rodent antibody, while the constant domains of the antibody molecule are derived from those of a human antibody.
  • the constant domains of the chimeric antibody may be derived from that of other species, such as a cat or dog.
  • a humanized antibody is a recombinant protein in which the CDRs from an antibody from one species; e.g., a rodent antibody, are transferred from the heavy and light variable chains of the rodent antibody into human heavy and light variable domains (e.g., framework region sequences).
  • the constant domains of the antibody molecule are derived from those of a human antibody.
  • a limited number of framework region amino acid residues from the parent (rodent) antibody may be substituted into the human antibody framework region sequences.
  • a human antibody is, e.g., an antibody obtained from transgenic mice that have been "engineered” to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain loci are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous murine heavy chain and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for particular antigens, and the mice can be used to produce human antibody-secreting hybridomas. Methods for obtaining human antibodies from transgenic mice are described by Green et al, Nature Genet. 7: 13 (1994), Lonberg et al, Nature 368:856 (1994), and Taylor et al., Int. Immun. 6:579 (1994).
  • a fully human antibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art. See for example, McCafferty et al, Nature 348:552-553 (1990) for the production of human antibodies and fragments thereof z ' w vitro, from immunoglobulin variable domain gene repertoires from unimmunized donors.
  • antibody variable domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, and displayed as functional antibody fragments on the surface of the phage particle.
  • the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. In this way, the phage mimics some of the properties of the B cell.
  • Phage display can be performed in a variety of formats, for review, see e.g. Johnson and Chiswell, Current Opinion in Structural Biology 3 :5564-571 (1993).
  • Human antibodies may also be generated by in vitro activated B cells. See U.S. Pat. Nos. 5,567,610 and 5,229,275, the Examples sections of which are incorporated herein by reference.
  • a therapeutic agent is a compound, molecule or atom which is administered separately, concurrently or sequentially with an antibody moiety or conjugated to an antibody moiety, i.e., antibody or antibody fragment, or a sub fragment, and is useful in the treatment of a disease.
  • therapeutic agents include antibodies, antibody fragments, cytotoxic agents, drugs, toxins, nucleases, hormones, immunomodulators, pro-apoptotic agents, anti-angiogenic agents, boron compounds, photoactive agents or dyes and
  • Radioisotopes are described in more detail below.
  • a diagnostic agent is a molecule, atom or other detectable moiety which may be administered conjugated to an antibody moiety or targetable construct and is useful in detecting or diagnosing a disease by locating cells containing the target antigen.
  • useful diagnostic agents include, but are not limited to, radioisotopes, dyes, contrast agents, fluorescent compounds or molecules and enhancing agents (e.g., paramagnetic ions) for magnetic resonance imaging (MRI) or positron emission tomography (PET) scanning.
  • the diagnostic agents are selected from the group consisting of radioisotopes, enhancing agents for use in magnetic resonance or PET imaging, and fluorescent compounds.
  • a reagent having a long tail to which are attached a multiplicity of chelating groups for binding the ions.
  • a tail can be a polymer such as a polylysine, polysaccharide, or other derivatized or derivatizable chain having pendant groups to which can be bound chelating groups such as, e.g.,
  • EDTA ethylenediaminetetraacetic acid
  • DTP A diethylenetriaminepentaacetic acid
  • DOTA diethylenetriaminepentaacetic acid
  • NOTA DOTA
  • NETA NETA
  • porphyrins polyamines, crown ethers, bis-thiosemicarbazones, polyoximes, and like groups known to be useful for this purpose.
  • Chelates are coupled to the antibodies using standard chemistries.
  • the chelate is normally linked to the antibody by a group which enables formation of a bond to the molecule with minimal loss of
  • Particularly useful metal-chelate combinations include 2-benzyl-DTPA and its monomethyl and cyclohexyl analogs, used with diagnostic isotopes in the general energy range of 60 to u 125 T 131 T 123 T 124 T 62 / -, 64 / -, 18 ⁇ 111 T 67 ⁇ 68 ⁇ 99 ⁇ ⁇ 94 ⁇ ⁇ 11,-, 13
  • 4,000 kev such as I, I, I, I, Cu, Cu, F, In, Ga, Ga, Tc, Tc, C, N, 15 0, 76 Br, for radioimaging.
  • non-radioactive metals such as manganese, iron and gadolinium are useful for MRI, when used along with the antibodies of the invention.
  • Macrocyclic chelates such as NOTA (1,4,7- triazacyclononane-N,N',N"-triacetic acid), DOTA (1,4,7, 10-tetraazacyclododecanetetraacetic acid), and TETA (p-bromoacetamido-benzyl-tetraethylaminetetraacetic acid) are of use with a variety of metals and radiometals, most particularly with radionuclides of gallium, yttrium and copper, respectively. Such metal-chelate complexes can be made very stable by tailoring the ring size to the metal of interest.
  • NOTA 1,4,7- triazacyclononane-N,N',N"-triacetic acid
  • DOTA 1,4,7, 10-tetraazacyclododecanetetraacetic acid
  • TETA p-bromoacetamido-benzyl-tetraethylaminetetraacetic acid
  • ring-type chelates such as macrocyclic polyethers, which are of interest for stably binding nuclides, such as 223 Ra for radioimmunotherapy (RAIT) are encompassed by the invention. More recently, techniques of general utility for labeling virtually any molecule with an 18 F atom of use in PET imaging have been described in U.S. Patent Nos. 7,563,433; 7,597,876 and 7,993,626, the Examples section of each incorporated herein by reference.
  • An immunoconjugate is an antibody, antibody fragment or antibody fusion protein conjugated to at least one therapeutic and/or diagnostic agent.
  • the diagnostic agent and/or therapeutic agent are as defined above.
  • An expression vector is a DNA molecule comprising a gene that is expressed in a host cell. Typically, gene expression is placed under the control of certain regulatory elements, including constitutive or inducible promoters, tissue-specific regulatory elements and enhancers. Such a gene is said to be "operably linked to" the regulatory elements.
  • a recombinant host may be any prokaryotic or eukaryotic cell that contains either a cloning vector or expression vector. This term also includes those prokaryotic or eukaryotic cells, as well as transgenic animals, that have been genetically engineered to contain the cloned gene(s) in the chromosome or genome of the host cell.
  • Suitable mammalian host cells include myeloma cells, such as SP2/0 cells, and NSO cells, as well as Chinese Hamster Ovary (CHO) cells, hybridoma cell lines and other mammalian host cell useful for expressing antibodies.
  • Sp2/0 cells transfected with an apoptosis inhibitor, such as a Bcl-EEE gene, and adapted to grow and be further transfected in serum free conditions, as described in U.S. Patent Nos. 7,531,327; 7,537,930; and 7,608,425, the Examples section of each of which is incorporated herein by reference.
  • an apoptosis inhibitor such as a Bcl-EEE gene
  • Various embodiments of the invention concern antibodies that react with very high selectivity with pancreatic cancer as opposed to normal or benign pancreatic tissues.
  • the anti-pancreatic cancer antibodies and fragments thereof are preferably raised against a crude mucin preparation from a tumor of the human pancreas, although partially purified or even purified MUC5ac may be utilized.
  • a non-limiting example of such antibodies is the PAM4 antibody.
  • the murine PAM4 (mPAM4) antibody was developed by employing pancreatic cancer mucin derived from the xenografted RIP- 1 human pancreatic carcinoma as immunogen. (Gold et al, Int. J. Cancer, 57:204-210, 1994.) As discussed below, antibody cross-reactivity and immunohistochemical staining studies indicate that the PAM4 antibody recognizes a unique and novel epitope on MUC5ac. Immunohistochemical staining studies, such as those described in Example 2, have shown that the PAM4 MAb binds to an antigen expressed by breast, pancreas and other cancer cells, with limited binding to normal human tissue. However, the highest expression is usually by pancreatic cancer cells.
  • the PAM4 antibodies are relatively specific to pancreatic cancer and preferentially bind pancreatic cancer cells.
  • the PAM4 antibody is reactive with a target epitope which can be internalized. This epitope is expressed primarily by antigens associated with pancreatic cancer and not with focal pancreatitis or normal pancreatic tissue. Binding of PAM4 antibody to the PAM4 epitope is inhibited by treatment of the antigen with DTT or periodate. Localization and therapy studies using a radiolabeled PAM4 MAb in animal models have demonstrated tumor targeting and therapeutic efficacy.
  • the PAM4 antibodies bind to an epitope of MUC5ac (located within the second to fourth cysteine-rich domains), which is expressed by many organs and tumor types, but is preferentially expressed in pancreatic cancer cells. Studies with a PAM4 MAb, such as in Example 3, indicate that the antibody exhibits several important properties, which make it a good candidate for clinical diagnostic and therapeutic applications.
  • the epitope provides a useful target for diagnosis and therapy of pancreatic and other cancers.
  • the PAM4 antibody apparently recognizes an epitope of MUC5ac that is distinct from the epitopes recognized by non-PAM4 anti-pancreatic cancer antibodies (e.g., CA19.9, DUPAN2, SPAN1, Nd2, CEACAM5, B72.3, anti-Le a , and other anti-Lewis antigens).
  • non-PAM4 anti-pancreatic cancer antibodies e.g., CA19.9, DUPAN2, SPAN1, Nd2, CEACAM5, B72.3, anti-Le a , and other anti-Lewis antigens.
  • the Examples below indicate that the MUC5ac epitope to which PAM4 binds is present in detectable concentrations in serum of patients with very early stage pancreatic cancer. Also surprisingly, it appears that an endogenous inhibitor of PAM4 antibody binding to MUC5ac is present in fresh human serum. The inhibitor is removed by long-term frozen storage of serum samples, or by organic phase extraction of fresh serum. These unexpected results provide the basis of a relatively non-invasive, early detection test for pancreatic cancer, using blood, serum or plasma samples.
  • the PAM4 antibody may be used alone, or else in conjunction with one or more other antibodies, such as CA19.9 antibody, to detect pancreatic cancer markers in serum.
  • antibodies suitable for use in combination or conjunction with PAM4 antibodies include, for example, the antibodies CA19.9, DUPAN2, SPAN1, Nd2, B72.3, CC49, anti-CEA, anti-CEACAM6, anti-Le a , anti-HLA-DR, anti-CD40, anti-CD74, anti-CD 138, and antibodies defined by the Lewis antigen Le(y), or antibodies against colon- specific antigen-p (CSAp), MUC1, MUC2, MUC3, MUC4, MUC5ac, MUC16, MUC17, EGP-1, EGP-2, HER2/neu, EGFR, angiogenesis factors (e.g., VEGF and P1GF), insulin-like growth factor (IGF), tenascin, platelet-derived growth factor, and IL-6, as well as products of oncogenes (bcl-2, Kras, p53), cMET, and antibodies against tumor necrosis substances, such as described in patents by Epstein et
  • immunomodulators involved in effector cell function against tumor cells could also be useful in combination with PAM4 antibodies alone or in combination with other tumor-associated antibodies, one example being antibodies against CD40.
  • PAM4 antibodies alone or in combination with other tumor-associated antibodies, one example being antibodies against CD40.
  • markers or products of oncogenes e.g., bcl-2, Kras, p53, cMET
  • angiogenesis factors such as VEGFR and placenta-like growth factor (P1GF).
  • the murine, chimeric, humanized and fully human PAM4 antibodies and fragments thereof described herein are exemplary of anti-pancreatic cancer antibodies of use for diagnostic and/or therapeutic methods.
  • the Examples below disclose a preferred
  • a humanized PAM4 antibody Because non- human monoclonal antibodies can be recognized by the human host as a foreign protein, and repeated injections can lead to harmful hypersensitivity reactions, humanization of a murine antibody sequences can reduce the adverse immune response that patients may experience. For murine-based monoclonal antibodies, this is often referred to as a Human Anti-Mouse Antibody (HAMA) response. Preferably some human residues in the framework regions of the humanized anti-pancreatic cancer antibody or fragments thereof are replaced by their murine counterparts. It is also preferred that a combination of framework sequences from two different human antibodies is used for VH. The constant domains of the antibody molecule are derived from those of a human antibody.
  • HAMA Human Anti-Mouse Antibody
  • Monoclonal antibodies for specific antigens may be obtained by methods known to those skilled in the art. See, for example, Kohler and Milstein, Nature 256: 495 (1975), and Coligan et al. (eds.), CURRENT PROTOCOLS IN IMMUNOLOGY, VOL. 1, pages 2.5.1- 2.6.7 (John Wiley & Sons 1991) (hereinafter "Coligan").
  • anti-pancreatic cancer MAbs can be obtained by injecting mice with a composition comprising a mixture of pancreatic cancer mucins comprising MUC5ac, or a purified MUC5ac, or a peptide or protein corresponding to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B-lymphocytes, fusing the B- lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones which produce antibodies to MUC5ac, culturing the clones that produce antibodies to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), and isolating anti-pancreatic cancer antibodies from the hybridoma cultures.
  • the antibodies can be sequenced and subsequently prepared by recombinant techniques to produce chimeric or humanized antibodies. Chimerization of murine antibodies and antibody fragments are well known to those skilled in the art. The use of antibody components derived from chimerized monoclonal antibodies reduces potential problems associated with the immunogenicity of murine constant regions.
  • V (variable light chain) and VH (variable heavy chain) sequences for murine antibodies can be obtained by a variety of molecular cloning procedures, such as RT-PCR, 5'-RACE, and cDNA library screening.
  • VH and V sequences of the murine PAM4 MAb were cloned by PCR amplification from the hybridoma cells by RT-PCR, and their sequences determined by DNA sequencing.
  • V and VH genes can be expressed in cell culture as a chimeric Ab as described by Orlandi et al., (Proc Natl. Acad. Sci., USA, 86: 3833, 1989).
  • a chimerized PAM4 antibody or antibody fragment comprises the complementarity-determining regions (CDRs) and framework regions (FR) of a murine PAM4 MAb and the light and heavy chain constant regions of a human antibody, wherein the CDRs of the light chain variable region of the chimerized PAM4 comprises CDR1 (SASSSVSSSYLY, SEQ ID NO: l); CDR2 (STSNLAS, SEQ ID NO:2); and CDR3 (HQWNRYPYT, SEQ ID NO:3); and the CDRs of the heavy chain variable region of the chimerized PAM4 MAb comprises CDR1 (SYVLH, SEQ ID NO:4); CDR2
  • humanized monoclonal antibodies may be produced by transferring murine complementary determining regions from heavy and light variable chains of the mouse immunoglobulin into a human variable domain, and then substituting selected human residues in the framework regions with their the murine FR counterparts.
  • the use of human framework region sequences, in addition to human constant region sequences, further reduces the chance of inducing HAMA reactions.
  • Humanized antibodies can be designed and constructed as described by Leung et al. (Mol Immunol. 32: 1413 (1995)).
  • Example 1 describes the humanization process utilized for construction of the hPAM4 MAb.
  • a humanized PAM4 antibody or antibody fragment comprises the light and heavy chain CDR sequences (SEQ ID NO: 1 to SEQ ID NO: 6) disclosed above.
  • the FRs of the light and heavy chain variable regions of the humanized antibody comprise at least one amino acid substituted from said corresponding FRs of the murine PAM4 MAb.
  • a fully human antibody e.g., human PAM4 can be obtained from a transgenic non- human animal. See, e.g., Mendez et al, Nature Genetics, 15: 146-156 (1997); U.S. Pat. No. 5,633,425.
  • a human antibody can be recovered from a transgenic mouse possessing human immunoglobulin loci.
  • the mouse humoral immune system is humanized by inactivating the endogenous immunoglobulin genes and introducing human
  • the human immunoglobulin loci are exceedingly complex and comprise a large number of discrete segments which together occupy almost 0.2% of the human genome.
  • large portions of human heavy- and light-chain loci must be introduced into the mouse genome. This is accomplished in a stepwise process beginning with the formation of yeast artificial chromosomes (YACs) containing either human heavy - or light-chain immunoglobulin loci in germline configuration. Since each insert is approximately 1 Mb in size, YAC construction requires homologous recombination of overlapping fragments of the immunoglobulin loci.
  • the two YACs, one containing the heavy-chain loci and one containing the light-chain loci are introduced separately into mice via fusion of YAC-containing yeast spheroblasts with mouse embryonic stem cells.
  • Embryonic stem cell clones are then microinjected into mouse blastocysts. Resulting chimeric males are screened for their ability to transmit the YAC through their germline and are bred with mice deficient in murine antibody production. Breeding the two transgenic strains, one containing the human heavy -chain loci and the other containing the human light- chain loci, creates progeny which produce human antibodies in response to immunization.
  • these techniques are not limiting and other methods known in the art for producing human antibodies, such as the use of phage display, may also be utilized to produce human anti-pancreatic cancer antibodies.
  • Antibodies can be produced by cell culture techniques using methods known in the art.
  • transfectoma cultures are adapted to serum-free medium.
  • cells may be grown as a 500 ml culture in roller bottles using HSFM. Cultures are centrifuged and the supernatant filtered through a 0.2 ⁇ membrane. The filtered medium is passed through a protein-A column (1 x 3 cm) at a flow rate of 1 ml/min. The resin is then washed with about 10 column volumes of PBS and protein A-bound antibody is eluted from the column with 0.1 M glycine buffer (pH 3.5) containing 10 mM EDTA.
  • 0.1 M glycine buffer pH 3.5
  • Fractions of 1.0 ml are collected in tubes containing 10 ⁇ of 3 M Tris (pH 8.6), and protein concentrations determined from the absorbance at 280/260 nm. Peak fractions are pooled, dialyzed against PBS, and the antibody concentrated, for example, with a Centricon 30 filter (Amicon, Beverly, MA). The antibody concentration is determined by ELISA and its concentration adjusted to about 1 mg/ml using PBS. Sodium azide, 0.01% (w/v), is conveniently added to the sample as preservative.
  • Antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography. See, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3. Also, see Baines et al, "Purification of Immunoglobulin G (IgG)," in METHODS IN MOLECULAR
  • BIOLOGY, VOL. 10, pages 79-104 The Humana Press, Inc. 1992.
  • Anti-pancreatic cancer MAbs can be characterized by a variety of techniques that are well-known to those of skill in the art. For example, the ability of an antibody to bind to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052) can be verified using an indirect enzyme immunoassay, flow cytometry analysis, ELISA or Western blot analysis.
  • Antibody fragments are antigen binding portions of an antibody, such as F(ab') 2, Fab', F(ab)2, Fab, Fv, sFv, scFv and the like. Antibody fragments which recognize specific epitopes can be generated by known techniques. F(ab')2 fragments, for example, can be produced by pepsin digestion of the antibody molecule. These and other methods are described, for example, by Goldenberg, U.S. Pat. Nos. 4,036,945 and 4,331,647 and references contained therein. Also, see Nisonoff et al, Arch Biochem. Biophys. 89: 230 (1960); Porter, Biochem. J. 73 : 1 19 (1959), Edelman et al, in METHODS ⁇
  • Fab' expression libraries can be constructed (Huse et al, 1989, Science, 246: 1274-1281) to allow rapid and easy identification of monoclonal Fab' fragments with the desired specificity.
  • a single chain Fv molecule comprises a VL domain and a VH domain.
  • the VL and VH domains associate to form a target binding site. These two domains are further covalently linked by a peptide linker (L).
  • L peptide linker
  • a scFv molecule is denoted as either VL-L-VH if the VL domain is the N-terminal part of the scFv molecule, or as VH-L-VL if the VH domain is the N-terminal part of the scFv molecule.
  • VHH Single domain antibodies
  • Single domain antibodies may be obtained, for example, from camels, alpacas or llamas by standard immunization techniques.
  • the VHH may have potent antigen-binding capacity and can interact with novel epitopes that are inaccessible to conventional VH-VL pairs.
  • Alpaca serum IgG contains about 50% camelid heavy chain only IgG antibodies (HCAbs) (Maass et al, 2007).
  • Alpacas may be immunized with known antigens, such as TNF-a, and VHHs can be isolated that bind to and neutralize the target antigen (Maass et al, 2007).
  • PCR primers that amplify virtually all alpaca VHH coding sequences have been identified and may be used to construct alpaca VHH phage display libraries, which can be used for antibody fragment isolation by standard biopanning techniques well known in the art (Maass et al, 2007).
  • An antibody fragment can also be prepared by proteolytic hydrolysis of a full-length antibody or by expression in E. coli or another host of the DNA coding for the fragment.
  • An antibody fragment can be obtained by pepsin or papain digestion of full-length antibodies by conventional methods.
  • an antibody fragment can be produced by enzymatic cleavage of antibodies with pepsin to provide an approximate 100 Kd fragment denoted F(ab')2.
  • This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce an approximate 50 Kd Fab' monovalent fragment.
  • an enzymatic cleavage using papain produces two monovalent Fab fragments and an Fc fragment directly.
  • Fusion proteins comprising the anti-pancreatic cancer antibodies of interest can be prepared by a variety of conventional procedures, ranging from glutaraldehyde linkage to more specific linkages between functional groups.
  • the antibodies and/or antibody fragments that comprise the fusion proteins described herein are preferably covalently bound to one another, directly or through a linker moiety, through one or more functional groups on the antibody or fragment, e.g., amine, carboxyl, phenyl, thiol, or hydroxyl groups.
  • linkers in addition to glutaraldehyde can be used, e.g., diisocyanates, diiosothiocyanates, bis(hydroxysuccinimide)esters, carbodiimides, maleimidehydroxy succinimide esters, and the like.
  • an antibody fusion protein comprises an anti-pancreatic cancer MAb, or fragment thereof, wherein the MAb binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052).
  • This fusion protein and fragments thereof preferentially bind pancreatic cancer cells.
  • This monovalent, monospecific MAb is useful for direct targeting of an antigen, where the MAb is attached to a therapeutic agent, a diagnostic agent, or a combination thereof, and the protein is administered directly to a patient.
  • the fusion proteins may instead comprise at least two anti-pancreatic cancer MAbs that bind to distinct epitopes of MUC5ac.
  • the MAbs can produce antigen specific diabodies, triabodies and tetrabodies, which are multivalent but monospecific to the MUC5ac.
  • the non-covalent association of two or more scFv molecules can form functional diabodies, triabodies and tetrabodies.
  • Monospecific diabodies are homodimers of the same scFv, where each scFv comprises the VH domain from the selected antibody connected by a short linker to the VL domain of the same antibody.
  • a diabody is a bivalent dimer formed by the non-covalent association of two scFvs, yielding two Fv binding sites.
  • a triabody results from the formation of a trivalent trimer of three scFvs, yielding three binding sites, and a tetrabody is a tetravalent tetramer of four scFvs, resulting in four binding sites.
  • Several monospecific diabodies have been made using an expression vector that contains a recombinant gene construct comprising VHI -linker- Vi l . See Holliger et al, Proc Natl. Acad.
  • the multivalent, monospecific antibody fusion protein binds to two or more of the same type of epitopes that can be situated on the same antigen or on separate antigens.
  • the increased valency allows for additional interaction, increased affinity, and longer residence times.
  • These antibody fusion proteins can be utilized in direct targeting systems, where the antibody fusion protein is conjugated to a therapeutic agent, a diagnostic agent, or a combination thereof, and administered directly to a patient in need thereof.
  • a preferred embodiment is a multivalent, multispecific antibody or fragment thereof comprising one or more antigen binding sites having an affinity toward a PAM4 target epitope and one or more additional binding sites for other epitopes associated with pancreatic cancer.
  • This fusion protein is multispecific because it binds at least two different epitopes, which can reside on the same or different antigens.
  • the fusion protein may comprise more than one antigen binding site, the first with an affinity toward an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052) epitope and the second with an affinity toward another target antigen such as TAG-72 or CEA.
  • bispecific antibody fusion protein which may comprise a CA19.9 MAb (or fragment thereof) and a PAM4 MAb (or fragment thereof).
  • a fusion protein will have an affinity toward CA19.9 as well as MUC5ac.
  • the antibody fusion proteins and fragments thereof can be utilized in direct targeting systems, where the antibody fusion protein is conjugated to a therapeutic agent, a diagnostic agent, or a combination thereof, and administered directly to a patient in need thereof.
  • a multivalent, multispecific antibody comprising at least one binding site having affinity toward a PAM4 target epitope and at least one hapten binding site having affinity towards hapten molecules.
  • a bispecific fusion protein may comprise the 679 MAb (or fragment thereof) and the PAM4 MAb (or fragment thereof).
  • the monoclonal 679 antibody binds with high affinity to molecules containing the tri-peptide moiety histamine succinyl glycyl (HSG).
  • HSG histamine succinyl glycyl
  • Such a bispecific PAM4 antibody fusion protein can be prepared, for example, by obtaining a F(ab') 2 fragment from 679, as described above.
  • the interchain disulfide bridges of the 679 F(ab3 ⁇ 4 fragment are gently reduced with DTT, taking care to avoid light-heavy chain linkage, to form Fab'-SH fragments.
  • the SH group(s) is (are) activated with an excess of bis-maleimide linker (l, l'-(methylenedi-4,l- phenylene)b-is-maleimide).
  • the PAM4 MAb is converted to Fab'-SH and then reacted with the activated 679 Fab'-SH fragment to obtain a bispecific antibody fusion protein.
  • Bispecific antibody fusion proteins such as this one can be utilized in affinity enhancing systems, where the target antigen is pretargeted with the fusion protein and is subsequently targeted with a diagnostic or therapeutic agent attached to a carrier moiety (targetable construct) containing one or more HSG haptens.
  • a DNLTM-based hPAM4- 679 construct such as TF10, may be prepared and used as described in the Examples below.
  • Bispecific antibodies can be made by a variety of conventional methods, e.g., disulfide cleavage and reformation of mixtures of whole IgG or, preferably F(ab') 2 fragments, fusions of more than one hybridoma to form polyomas that produce antibodies having more than one specificity, and by genetic engineering.
  • Bispecific antibody fusion proteins have been prepared by oxidative cleavage of Fab' fragments resulting from reductive cleavage of different antibodies.
  • This is advantageously carried out by mixing two different F(ab') 2 fragments produced by pepsin digestion of two different antibodies, reductive cleavage to form a mixture of Fab' fragments, followed by oxidative reformation of the disulfide linkages to produce a mixture of F(ab') 2 fragments including bispecific antibody fusion proteins containing a Fab' portion specific to each of the original epitopes.
  • General techniques for the preparation of antibody fusion proteins may be found, for example, in Nisonoff et al, Arch Biochem Biophys. 93: 470 (1961), Hammerling et al, J Exp Med. 128: 1461 (1968), and U.S. Pat. No. 4,331,647.
  • More selective linkage can be achieved by using a heterobifunctional linker such as maleimidehydroxysuccinimide ester. Reaction of the ester with an antibody or fragment will derivatize amine groups on the antibody or fragment, and the derivative can then be reacted with, e.g., an antibody Fab fragment having free sulfhydryl groups (or, a larger fragment or intact antibody with sulfhydryl groups appended thereto by, e.g., Traut's Reagent). Such a linker is less likely to crosslink groups in the same antibody and improves the selectivity of the linkage.
  • a heterobifunctional linker such as maleimidehydroxysuccinimide ester.
  • ScFvs with linkers greater than 12 amino acid residues in length allow interactions between the VH and VL domains on the same chain and generally form a mixture of monomers, dimers (termed diabodies) and small amounts of higher mass multimers, (Kortt et al., Eur J Biochem. (1994) 221 : 151-157). ScFvs with linkers of 5 or less amino acid residues, however, prohibit intramolecular pairing of the VH and VL domains on the same chain, forcing pairing with VH and VL domains on a different chain.
  • Linkers between 3- and 12-residues form predominantly dimers (Atwell et al., Protein Engineering (1999) 12: 597-604). With linkers between 0 and 2 residues, trimeric (termed triabodies), tetrameric (termed tetrabodies) or higher oligomeric structures of scFvs are formed; however, the exact patterns of oligomerization appear to depend on the composition as well as the orientation of the V-domains, in addition to the linker length.
  • DOCK-AND-LOCKTM involves the production of fusion proteins that comprise at their N- or C-terminal ends one of two complementary peptide sequences, called dimerization and docking domain (DDD) and anchoring domain (AD) sequences.
  • the DDD sequences are derived from the regulatory subunits of cAMP-dependent protein kinase and the AD sequence is derived from the sequence of A- kinase anchoring protein (AKAP).
  • the DDD sequences form dimers that bind to the AD sequence, which allows formation of trimers, tetramers, hexamers or any of a variety of other complexes.
  • effector moieties such as antibodies or antibody fragments
  • complexes may be formed of any selected combination of antibodies or antibody fragments.
  • the DNLTM complexes may be covalently stabilized by formation of disulfide bonds or other linkages.
  • Bispecific or multispecific antibodies may be utilized in pre-targeting techniques. Pre-targeting is a multistep process originally developed to resolve the slow blood clearance of directly targeting antibodies, which contributes to undesirable toxicity to normal tissues such as bone marrow. With pre-targeting, a radionuclide or other therapeutic agent is attached to a small delivery molecule (targetable construct or targetable conjugate) that is cleared within minutes from the blood. A pre-targeting bispecific or multispecific antibody, which has binding sites for the targetable construct as well as a target antigen, is administered first, free antibody is allowed to clear from circulation and then the targetable construct is administered.
  • Pre-targeting methods are well known in the art, for example, as disclosed in Goodwin et al, U.S. Pat. No. 4,863,713; Goodwin et al, J. Nucl. Med. 29:226, 1988;
  • a pre-targeting method of treating or diagnosing a disease or disorder in a subject may be provided by: (1) administering to the subject a bispecific antibody or antigen binding antibody fragment; (2) optionally administering to the subject a clearing composition, and allowing the composition to clear the antibody from circulation; and (3) administering to the subject the targetable construct, containing one or more chelated or chemically bound therapeutic or diagnostic agents.
  • the technique may also be utilized for antibody dependent enzyme prodrug therapy (ADEPT) by administering an enzyme conjugated to a targetable construct, followed by a prodrug that is converted into active form by the enzyme.
  • ADPT antibody dependent enzyme prodrug therapy
  • the claimed methods and compositions may utilize any of a variety of antibodies known in the art, for example for combination antibody therapy.
  • Antibodies of use may be commercially obtained from a number of known sources. For example, a variety of antibody secreting hybridoma lines are available from the American Type Culture Collection (ATCC, Manassas, VA). A large number of antibodies against various disease targets, including but not limited to tumor-associated antigens, have been deposited at the ATCC and/or have published variable region sequences and are available for
  • antibodies that may be of use for therapy of cancer within the scope of the claimed methods and compositions include, but are not limited to, LL1 (anti-CD74), LL2 or RFB4 (anti-CD22), veltuzumab (hA20, anti-CD20), rituxumab (anti-CD20), obinutuzumab (GA101, anti-CD20), lambrolizumab (anti-PD-1 receptor), nivolumab (anti-PD-1 receptor), ipilimumab (anti-CTLA-4), RS7 (anti-epithelial glycoprotein- 1 (EGP-1, also known as TROP-2)), KC4 (anti-mucin), MN-14 (anti-carcinoembryonic antigen (anti-CEA, also known as CD66e or CEACAM5), MN-15
  • panitumumab (anti-EGFR); tositumomab (anti-CD20); PAM4 (aka clivatuzumab, anti- MUC5ac) and trastuzumab (anti-ErbB2).
  • anti-EGFR panitumumab
  • anti-CD20 tositumomab
  • PAM4 aka clivatuzumab, anti- MUC5ac
  • trastuzumab anti-ErbB2
  • Other useful antigens that may be targeted using the described conjugates include carbonic anhydrase IX, B7, CCL19, CCL21, CSAp, HER-2/wew, BrE3, CD 1, CDla, CD2, CD3, CD4, CD5, CD 8, CD1 1A, CD14, CD15, CD16, CD18, CD19, CD20 (e.g., C2B8, hA20, 1F5 MAbs), CD21, CD22, CD23, CD25, CD29, CD30, CD32b, CD33, CD37, CD38, CD40, CD40L, CD44, CD45, CD46, CD47, CD52, CD54, CD55, CD59, CD64, CD67, CD70, CD74, CD79a, CD80, CD83, CD95, CD126, CD133, CD138, CD147, CD154, CEACAM5, CEACAM6, CTLA-4, CXCR4, alpha-fetoprotein (AFP), VEGF (e.g.,
  • AVASTIN® fibronectin splice variant
  • ED-B fibronectin e.g., L19
  • EGP-1 TROP-2
  • EGP-2 e.g., 17-1A
  • EGF receptor ErbBl
  • ErbB2 ErbB3, Factor H, FHL-1, Flt-3, folate receptor, Ga 733,GRO- , HMGB-1, hypoxia inducible factor (HIF), HM1.24, HER-2/wew, histone H2B, histone H3, histone H4, insulin-like growth factor (ILGF), IFN- ⁇ , IFN-a, IFN- ⁇ , IFN- ⁇ , IL-2R, IL-4R, IL-6R, IL-13R, IL-15R, IL-17R, IL-18R, IL-2, IL-6, IL-8, IL-12, IL-15, IL-17, IL-18, IL-25, IP-10, IGF-1R, la,
  • CD Cluster Designation
  • the CD66 antigens consist of five different glycoproteins with similar structures, CD66a-e, encoded by the carcinoembryonic antigen (CEA) gene family members, BCG, CGM6, NCA, CGM1 and CEA, respectively. These CD66 antigens (e.g., CEACAM6) are expressed mainly in granulocytes, normal epithelial cells of the digestive tract and tumor cells of various tissues. Also included as suitable targets for cancers are cancer testis antigens, such as NY-ESO-1 (Theurillat et al, Int. J. Cancer 2007; 120(11):2411-7), as well as CD79a in myeloid leukemia (Kozlov et al, Cancer Genet. Cytogenet.
  • CEACAM6 carcinoembryonic antigen
  • Macrophage migration inhibitory factor is an important regulator of innate and adaptive immunity and apoptosis. It has been reported that CD74 is the endogenous receptor for MIF (Leng et al, 2003, J Exp Med 197: 1467-76).
  • the therapeutic effect of antagonistic anti-CD74 antibodies on MIF-mediated intracellular pathways may be of use for treatment of a broad range of disease states, such as cancers of the bladder, prostate, breast, lung, colon and chronic lymphocytic leukemia (e.g., Meyer-Siegler et al, 2004, BMC Cancer 12:34; Shachar & Haran, 2011, Leuk Lymphoma 52: 1446-54); autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus (Morand & Leech, 2005, Front Biosci 10: 12-22; Shachar & Haran, 2011, Leuk Lymphoma 52: 1446-54); kidney diseases such as renal allograft rejection (Lan, 2008, Nephron Exp Nephrol.
  • a broad range of disease states such as cancers of the bladder, prostate, breast, lung, colon and chronic lymphocytic leukemia (e.g., Meyer-Siegler et al, 2004, BMC Cancer 12:34
  • Anti-TNF-a antibodies are known in the art and may be of use to treat immune diseases, such as autoimmune disease, immune dysfunction (e.g., graft-versus-host disease, organ transplant rejection) or diabetes.
  • Known antibodies against TNF-a include the human antibody CDP571 (Ofei et al, 2011, Diabetes 45:881-85); murine antibodies MTNFAI, M2TNFAI, M3TNFAI, M3TNFABI, M302B and M303 (Thermo Scientific, Rockford, IL); infliximab (Centocor, Malvern, PA); certolizumab pegol (UCB, Brussels, Belgium); and adalimumab (Abbott, Abbott Park, IL).
  • anti-B-cell antibodies such as veltuzumab, epratuzumab, milatuzumab or hL243 ; tocilizumab (anti-IL-6 receptor); basiliximab (anti-CD25); daclizumab (anti-CD25); efalizumab (anti-CD 11 a); muromonab-CD3 (anti-CD3 receptor); anti-CD40L (UCB, Brussels, Belgium); natalizumab (anti-a4 integrin) and omalizumab (anti-IgE).
  • anti-B-cell antibodies such as veltuzumab, epratuzumab, milatuzumab or hL243 ; tocilizumab (anti-IL-6 receptor); basiliximab (anti-CD25); daclizumab (anti-CD25); efalizumab (anti-CD 11 a); muromonab-CD3 (anti-CD3 receptor); anti-CD40L
  • Immune checkpoint inhibitor antibodies have been used primarily in cancer therapy. Immune checkpoints refer to inhibitory pathways in the immune system that are responsible for maintaining self-tolerance and modulating the degree of immune system response to minimize peripheral tissue damage. However, tumor cells can also activate immune system checkpoints to decrease the effectiveness of immune response against tumor tissues.
  • Exemplary checkpoint inhibitor antibodies against cytotoxic T-lymphocyte antigen 4 (CTLA4, also known as CD 152), programmed cell death protein 1 (PD1, also known as CD279) and programmed cell death 1 ligand 1 (PD-Ll, also known as CD274), may be used in combination with one or more other agents to enhance the effectiveness of immune response against disease cells, tissues or pathogens.
  • Exemplary anti-PDl antibodies include lambrolizumab (MK-3475, MERCK), nivolumab (BMS-936558, BRISTOL-MYERS SQUIBB), AMP-224 (MERCK), and pidilizumab (CT-011, CURETECH LTD.).
  • Anti-PDl antibodies are commercially available, for example from ABCAM® (AB137132),
  • BIOLEGEND® EH12.2H7, RMP1-14
  • AFFYMETRIX EBIOSCIENCE J105, Jl 16, MIH4
  • Exemplary anti-PD-Ll antibodies include MDX-1 105 (MEDAREX), MEDI4736 (MEDIMMU E) MPDL3280A (GENENTECH) and BMS-936559 (BRISTOL-MYERS SQUIBB).
  • Anti-PD-Ll antibodies are also commercially available, for example from AFFYMETRIX EBIOSCIENCE (MIH1).
  • Exemplary anti-CTLA4 antibodies include ipilimumab (Bristol-Myers Squibb) and tremelimumab (PFIZER).
  • Anti-PD 1 antibodies are commercially available, for example from ABCAM® (AB 134090), SI O BIOLOGICAL INC. (1 1159-H03H, 11 159-H08H), and THERMO SCIENTIFIC PIERCE (PA5-29572, PA5- 23967, PA5-26465, MA1-12205, MA1-35914). Ipilimumab has recently received FDA approval for treatment of metastatic melanoma (Wada et al, 2013, J Transl Med 1 1 :89).
  • antibodies of use may include anti-histone antibodies and/or antigen-binding fragments thereof, such as the BWA-3 (anti-H4), LG2-1 (anti-H3) and LG2-2 (anti-H2B) antibodies.
  • anti-histone antibodies are disclosed, for example, in U.S. Patent Application Serial No. 14/180,646, filed 2/14/14 (the Examples section of which is incorporated herein by reference).
  • antibodies are used that internalize rapidly and are then re-expressed, processed and presented on cell surfaces, enabling continual uptake and accretion of circulating conjugate by the cell.
  • An example of a most-preferred antibodies are used that internalize rapidly and are then re-expressed, processed and presented on cell surfaces, enabling continual uptake and accretion of circulating conjugate by the cell.
  • an anti-CD74 MAb invariant chain, class II-specific chaperone, Ii
  • the CD74 antigen is highly expressed on B-cell lymphomas (including multiple myeloma) and leukemias, certain T-cell lymphomas, melanomas, colonic, lung, and renal cancers, glioblastomas, and certain other cancers (Ong et al, Immunology 95:296-302 (1999)).
  • a review of the use of CD74 antibodies in cancer is contained in Stein et al, Clin Cancer Res. 2007 Sep 15; 13(18 Pt 2):5556s-5563s, incorporated herein by reference.
  • the diseases that are preferably treated with anti-CD74 antibodies include, but are not limited to, non-Hodgkin's lymphoma, Hodgkin's disease, melanoma, lung, renal, colonic cancers, glioblastome multiforme, histiocytomas, myeloid leukemias, and multiple myeloma.
  • Continual expression of the CD74 antigen for short periods of time on the surface of target cells, followed by internalization of the antigen, and re-expression of the antigen enables the targeting LL1 antibody to be internalized along with any chemotherapeutic moiety it carries. This allows a high, and therapeutic, concentration of LL1 -chemotherapeutic drug conjugate to be accumulated inside such cells. Internalized LL1 -chemotherapeutic drug conjugates are cycled through lysosomes and endosomes, and the chemotherapeutic moiety is released in an active form within the target cells.
  • Certain embodiments concern methods of diagnosing or treating a malignancy in a subject, comprising administering to the subject an anti-pancreatic cancer MAb, fusion protein or fragment thereof, wherein the MAb, fusion protein or fragment is bound to at least one diagnostic and/or therapeutic agent.
  • a method for diagnosing or treating cancer comprising administering to a subject a multivalent, multispecific antibody or fragment thereof comprising one or more antigen binding sites toward an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575- 2052) and one or more hapten binding sites, waiting a sufficient amount of time for non- bound antibody to clear the subject's blood stream; and then administering to the subject a carrier molecule comprising a diagnostic agent, a therapeutic agent, or a combination thereof, that binds to the hapten-binding site of the localized antibody.
  • the cancer is a non-endocrine pancreatic cancer.
  • MAbs for in vitro diagnosis. See, for example, Carlsson et al, Bio/Technology 7 (6): 567 (1989).
  • MAbs can be used to detect the presence of a tumor-associated antigen in tissue from biopsy samples.
  • MAbs also can be used to measure the amount of tumor-associated antigen in clinical fluid samples, such as blood or serum, using techniques such as radioimmunoassay, enzyme-linked immunosorbent assay, and fluorescence immunoassay. In vitro and in vivo methods of diagnosis are discussed in further detail below.
  • Conjugates of tumor-targeted MAbs and toxins can be used to selectively kill cancer cells in vivo (Spalding, Bio/Technology 9(8): 701 (1991); Goldenberg, Scientific American Science & Medicine 1(1): 64 (1994)).
  • therapeutic studies in experimental animal models have demonstrated the anti-tumor activity of antibodies carrying cytotoxic radionuclides. (Goldenberg et al, Cancer Res. 41 : 4354 (1981), Cheung et al, J. Nat'l Cancer Inst. 77: 739 (1986), and Senekowitsch et al, J. Nucl. Med. 30: 531 (1989)).
  • the conjugate comprises a 90 Y-labeled hPAM4 antibody.
  • the conjugate may optionally be administered in conjunction with one or more other therapeutic agents.
  • 90 Y-labeled hPAM4 is administered together with gemcitabine or 5- fluorouracil to a patient with pancreatic cancer.
  • 90 Y is conjugated to a DOTA chelate for attachment to hPAM4.
  • the 90 Y-DOTA-hPAM4 is combined with gemcitabine in fractionated doses comprising a treatment cycle, such as with repeated, lower, less-toxic doses of gemcitabine combined with lower, fractionated doses of 90 Y-DOTA-hPAM4.
  • a radiolabeled or other conjugated PAM4 antibody may be administered in combination with another therapeutic agent.
  • immunoconjugate such as an SN-38 conjugated antibody.
  • a particularly preferred combination is 90 Y-hPAM4 and SN-38-hRS7 (anti-TROP2 antibody) (see, e.g., U.S. Patent No. 8,586,050, the Examples section incorporated herein by reference).
  • Chimeric, humanized and human antibodies and fragments thereof have been used for in vivo therapeutic and diagnostic methods. Accordingly contemplated is a method of delivering a diagnostic or therapeutic agent, or a combination thereof, to a target comprising (i) providing a composition that comprises an anti-pancreatic cancer antibody or fragment thereof, such as a chimeric, humanized or human PAM4 antibody, conjugated to at least one diagnostic and/or therapeutic agent and (ii) administering to a subject the diagnostic or therapeutic antibody conjugate.
  • the anti-pancreatic cancer antibodies and fragments thereof are humanized or fully human.
  • Another embodiment concerns a method for treating a malignancy comprising administering a naked or conjugated anti-pancreatic cancer antibody, antibody fragment or fusion protein that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052), such as a PAM4 antibody, either alone or in conjunction with one or more other therapeutic agents.
  • the other therapeutic agent may be added before, simultaneously with or after the antibody.
  • the therapeutic agent is gemcitabine, and in a more preferred embodiment, gemcitabine is given with the hPAM4 radioconjugate in a fractionated dose schedule at lower doses than the conventional 800-1,000 mg/m 2 doses of gemcitabine given weekly for 6 weeks.
  • fractionated therapeutic doses of 90 Y-PAM4 when combined with fractionated therapeutic doses of 90 Y-PAM4, repeated fractionated doses intended to function as a radiosensitizing agent of 200-380 mg/m 2 gemcitabine are infused.
  • the antibodies, fusion proteins and/or fragments thereof described and claimed herein may be administered with any known or described therapeutic agent, including but not limited to heat shock protein 90 (Hsp90).
  • Hsp90 heat shock protein 90
  • subjects receive immunoconjugates in conjunction with standard cancer chemotherapy.
  • standard cancer chemotherapy For example, "CVB" (1.5 g/m 2
  • cyclophosphamide 200-400 mg/m 2 etoposide, and 150-200 mg/m 2 carmustine
  • a regimen used to treat non-Hodgkin's lymphoma Patti et ah, Eur. J. Haematol. 51: 18 (1993).
  • Other suitable combination chemotherapeutic regimens are well-known to those of skill in the art. See, for example, Freedman et at, "Non-Hodgkin's Lymphomas," in CANCER MEDICINE, VOLUME 2, 3rd Edition, Holland et al. (eds.), pages 2028-2068 (Lea & Febiger 1993).
  • first generation chemotherapeutic regimens for treatment of intermediate- grade non-Hodgkin's lymphoma include C-MOPP (cyclophosphamide, vincristine, procarbazine and prednisone) and CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone).
  • a useful second generation chemotherapeutic regimen is m-BACOD
  • a suitable third generation regimen is MACOP-B (methotrexate, doxorubicin, cyclophosphamide, vincristine, prednisone, bleomycin and leucovorin).
  • Additional useful drugs include phenyl butyrate, bendamustine, and bryostatin- 1.
  • the present invention contemplates the administration of anti-pancreatic cancer antibodies and fragments thereof, including fusion proteins and fragments thereof, alone, as a naked antibody or antibody fragment, or administered as a multimodal therapy.
  • the antibody is a humanized or fully human PAM4 antibody or fragment thereof.
  • Multimodal therapies further include immunotherapy with a naked anti-pancreatic cancer antibody supplemented with administration of other antibodies in the form of naked antibodies, fusion proteins, or as immunoconjugates.
  • a humanized or fully human PAM4 antibody may be combined with another naked antibody, or a humanized PAM4 or other antibody conjugated to an isotope, one or more chemotherapeutic agents, cytokines, toxins or a combination thereof.
  • the present invention contemplates treatment of a naked or conjugated PAM4 antibody or fragments thereof before, in combination with, or after other pancreatic tumor associated antibodies such as CA19.9, DUPAN2, SPAN1, Nd2, B72.3, CC49, anti-Le a antibodies, and antibodies to other Lewis antigens (e.g., Le(y)), as well as antibodies against carcinoembryonic antigen (CEA or CEACAM5), CEACAM6, colon- specific antigen-p (CSAp), MUC1, MUC2, MUC3, MUC4, MUC5ac, MUC16, MUC17, HLA-DR, CD40, CD74, CD 138, HER2/neu, EGFR, EGP-1, EGP-2, angiogenesis factors (e.g., VEGF, PIGF), insulin-like growth factor (IGF), tenascin, platelet-derived growth factor, and IL-6, as well as products of oncogenes (e.g., bcl-2, Kra
  • These solid tumor antibodies may be naked or conjugated to, inter alia, drugs, toxins, isotopes, radionuclides or immunomodulators. Many different antibody combinations may be constructed and used in either naked or conjugated form. Alternatively, different naked antibody combinations may be employed for administration in combination with other therapeutic agents, such as a cytotoxic drug or with radiation, given consecutively, simultaneously, or sequentially.
  • Administration of the antibodies and their fragments can be effected by intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, intrapleural, intrathecal, perfusion through a regional catheter, or direct intralesional injection.
  • the administration may be by continuous infusion or by single or multiple boluses.
  • the immunoconjugate of the present invention can be formulated for intravenous administration via, for example, bolus injection or continuous infusion.
  • the antibody of the present invention is infused over a period of less than about 4 hours, and more preferably, over a period of less than about 3 hours.
  • the first 25-50 mg could be infused within 30 minutes, preferably even 15 min, and the remainder infused over the next 2-3 hrs.
  • Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • Control release preparations can be prepared through the use of polymers to complex or adsorb the immunoconjugate.
  • biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebacic acid. Sherwood et al, Bio/Technology 10: 1446 (1992). The rate of release of an immunoconjugate or antibody from such a matrix depends upon the molecular weight of the immunoconjugate or antibody, the amount of immunoconjugate or antibody within the matrix, and the size of dispersed particles.
  • the dosage of an administered immunoconjugate for humans will vary depending upon such factors as the patient's age, weight, height, sex, general medical condition and previous medical history. It may be desirable to provide the recipient with a dosage of immunoconjugate, antibody fusion protein that is in the range of from about 1 mg/kg to 25 mg/kg as a single intravenous infusion, although a lower or higher dosage also may be administered as circumstances dictate.
  • the dosage may be repeated as needed, for example, once per week for 4-10 weeks, once per week for 8 weeks, or once per week for 4 weeks. It may also be given less frequently, such as every other week for several months, or monthly or quarterly for many months, as needed in a maintenance therapy.
  • an antibody may be administered as one dosage every 2 or 3 weeks, repeated for a total of at least 3 dosages.
  • the antibodies may be administered twice per week for 4-6 weeks. If the dosage is lowered to approximately 200-300 mg/m 2 (340 mg per dosage for a 1.7-m patient, or 4.9 mg/kg for a 70 kg patient), it may be administered once or even twice weekly for 4 to 10 weeks.
  • the dosage schedule may be decreased, namely every 2 or 3 weeks for 2-3 months. It has been determined, however, that even higher doses, such as 20 mg/kg once weekly or once every 2-3 weeks can be administered by slow i.v. infusion, for repeated dosing cycles.
  • the dosing schedule can optionally be repeated at other intervals and dosage may be given through various parenteral routes, with appropriate adjustment of the dose and schedule.
  • Anti-pancreatic cancer antibodies and fragments thereof may be conjugated to at least one therapeutic and/or diagnostic agent for therapy or diagnosis.
  • the objective is to deliver cytotoxic doses of radioactivity, toxin, antibody and/or drug to target cells, while minimizing exposure to non-target tissues.
  • anti-pancreatic cancer antibodies are used to diagnose and/or treat pancreatic tumors.
  • any of the antibodies, antibody fragments and fusion proteins can be conjugated with one or more therapeutic or diagnostic agents, using a variety of techniques known in the art.
  • One or more therapeutic or diagnostic agents may be attached to each antibody, antibody fragment or fusion protein, for example by conjugating an agent to a carbohydrate moiety in the Fc region of the antibody. If the Fc region is absent (for example with certain antibody fragments), it is possible to introduce a carbohydrate moiety into the light chain variable region of either an antibody or antibody fragment to which a therapeutic or diagnostic agent may be attached. See, for example, Leung et al, J Immunol. 154: 5919 (1995); Hansen et al, U.S. Pat. No. 5,443,953 (1995), Leung et al, U.S. Pat. No. 6,254,868, the Examples section of each patent incorporated herein by reference.
  • Antibody fusion proteins or multispecific antibodies comprise two or more antibodies or fragments thereof, each of which may be attached to at least one therapeutic agent and/or diagnostic agent. Accordingly, one or more of the antibodies or fragments thereof of the antibody fusion protein can have more than one therapeutic and/or diagnostic agent attached. Further, the therapeutic agents do not need to be the same but can be different therapeutic agents, for example, one can attach a drug and a radioisotope to the same fusion protein.
  • an IgG can be radiolabeled with 131 I and attached to a drug. The 131 I can be incorporated into the tyrosine of the IgG and the drug attached to the epsilon amino group of the IgG lysines.
  • Both therapeutic and diagnostic agents also can be attached to reduced SH groups and to the carbohydrate side chains of antibodies.
  • a bispecific antibody may comprise one antibody or fragment thereof against a disease antigen and another against a hapten attached to a targetable construct, for use in pretargeting techniques as discussed above.
  • a therapeutic or diagnostic agent can be attached at the hinge region of a reduced antibody component via disulfide bond formation.
  • such agents can be attached to the antibody component using a heterobifunctional cross-linker, such as N- succinyl 3-(2-pyridyldithio)proprionate (SPDP). Yu et al, Int. J. Cancer 56: 244 (1994). General techniques for such conjugation are well-known in the art.
  • the azide alkyne Huisgen cycloaddition reaction uses a copper catalyst in the presence of a reducing agent to catalyze the reaction of a terminal alkyne group attached to a first molecule.
  • a second molecule comprising an azide moiety
  • the azide reacts with the activated alkyne to form a 1,4-disubstituted 1,2,3-triazole.
  • the copper catalyzed reaction occurs at room temperature and is sufficiently specific that purification of the reaction product is often not required.
  • a copper-free click reaction has been proposed for covalent modification of biomolecules.
  • the copper- free reaction uses ring strain in place of the copper catalyst to promote a [3 + 2] azide-alkyne cycloaddition reaction (Id.)
  • cyclooctyne is a 8-carbon ring structure comprising an internal alkyne bond.
  • the closed ring structure induces a substantial bond angle deformation of the acetylene, which is highly reactive with azide groups to form a triazole.
  • cyclooctyne derivatives may be used for copper- free click reactions (Id.)
  • a wide variety of therapeutic reagents can be administered concurrently or sequentially, or advantageously conjugated to the antibodies of the invention, for example, drugs, toxins, oligonucleotides (e.g., siR A), immunomodulators, hormones, hormone antagonists, enzymes, enzyme inhibitors, radionuclides, angiogenesis inhibitors, pro- apoptotic agents, etc.
  • the therapeutic agents recited here are those agents that are useful for either conjugated to an antibody, fragment or fusion protein or for administration separately with a naked antibody as described above.
  • Therapeutic agents include, for example, chemotherapeutic drugs such as vinca alkaloids, anthracyclines, gemcitabine, epipodophyllotoxins, taxanes, antimetabolites, alkylating agents, antibiotics, SN-38, COX-2 inhibitors, antimitotics, antiangiogenic and apoptotic agents, particularly doxorubicin, methotrexate, taxol, CPT-1 1, camptothecans, proteosome inhibitors, mTOR inhibitors, HDAC inhibitors, tyrosine kinase inhibitors, and others from these and other classes of anticancer agents.
  • chemotherapeutic drugs such as vinca alkaloids, anthracyclines, gemcitabine, epipodophyllotoxins, taxanes, antimetabolites, alkylating agents, antibiotics, SN-38, COX-2 inhibitors, antimitotics, antiangiogenic and apoptotic agents, particularly doxorubicin, methotrexate, taxol,
  • cancer chemotherapeutic drugs include nitrogen mustards, alkyl sulfonates, nitrosoureas, triazenes, folic acid analogs, COX-2 inhibitors, antimetabolites, pyrimidine analogs, purine analogs, platinum coordination complexes, mTOR inhibitors, tyrosine kinase inhibitors, proteosome inhibitors, HDAC inhibitors, camptothecins and hormones. Suitable chemotherapeutic agents are described in REMINGTON'S
  • Specific drugs of use may include 5-fluorouracil, afatinib, aplidin, azaribine, anastrozole, anthracyclines, axitinib, AVL-101, AVL-291, bendamustine, bleomycin, bortezomib, bosutinib, bryostatin- 1 , busulfan, calicheamycin, camptothecin, carboplatin, 10- hydroxycamptothecin, carmustine, Celebrex, chlorambucil, cisplatin (CDDP), Cox-2 inhibitors, irinotecan (CPT-1 1), SN-38, carboplatin, cladribine, camptothecans, crizotinib, cyclophosphamide, cytarabine, dacarbazine, dasatinib, dinaciclib, docetaxel, dactinomycin, daunorubicin, doxor
  • conjugates of camptothecins and related compounds may be conjugated to hPAM4 or other anti-pancreatic cancer antibodies, for example as disclosed in U.S. Patent No. 7,591,994; and U.S. Patent Application Serial No. 1 1/388,032, filed March 23, 2006, the Examples section of each of which is incorporated herein by reference.
  • prodrug forms of 2-PDOX as disclosed in U.S. Patent Application Serial No.
  • 14/175,089 may be used as an immunoconjugate with an anti-pancreatic cancer antibody that binds to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052).
  • an hPAM4 antibody is given with gemcitabine, which may be given before, after, or concurrently with a naked or conjugated chimeric, humanized or human PAM4 antibody.
  • the conjugated hPAM4 antibody or antibody fragment is conjugated to a radionuclide.
  • a toxin can be of animal, plant or microbial origin.
  • a toxin, such as Pseudomonas exotoxin, may also be complexed to or form the therapeutic agent portion of an
  • toxins suitably employed in the preparation of such conjugates or other fusion proteins, include ricin, abrin, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin, ranpirnase, Pseudomonas exotoxin, and Pseudomonas endotoxin.
  • RNase ribonuclease
  • DNase I DNase I
  • Staphylococcal enterotoxin-A Staphylococcal enterotoxin-A
  • pokeweed antiviral protein pokeweed antiviral protein
  • gelonin gelonin
  • diphtheria toxin diphtheria toxin
  • ranpirnase Pseudomonas exotoxin
  • Pseudomonas endotoxin Pseudomonas endotoxin.
  • An immunomodulator such as a cytokine
  • cytokine may also be conjugated to, or form the therapeutic agent portion of the immunoconjugate, or may be administered with, but unconjugated to, an antibody, antibody fragment or fusion protein.
  • the fusion protein may comprise one or more antibodies or fragments thereof binding to different antigens.
  • the fusion protein may bind to an epitope located within the second to fourth cysteine-rich domains of MUC5ac (amino acid residues 1575-2052) as well as to
  • immunomodulator includes a cytokine, a lymphokine, a monokine, a stem cell growth factor, a lymphotoxin, a hematopoietic factor, a colony stimulating factor (CSF), an interferon (IFN), parathyroid hormone, thyroxine, insulin, proinsulin, relaxin, prorelaxin, follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), luteinizing hormone (LH), hepatic growth factor, prostaglandin, fibroblast growth factor, prolactin, placental lactogen, OB protein, a transforming growth factor (TGF), TGF-a, TGF- ⁇ , insulin-like growth factor (IGF), erythropoietin, erythropoietin, TGF, TGF-a, TGF- ⁇ , insulin-like growth factor (IGF), erythropoietin, TGF, TGF-a, TGF- ⁇ , insulin-like growth factor (I
  • the therapeutic agent may comprise one or more radioactive isotopes useful for treating diseased tissue.
  • Particularly useful therapeutic radionuclides include, but are not limited to m In, 177 Lu, 212 Bi, 213 Bi, 211 At, 62 Cu, "Cu, 67 Cu, 90 Y, 125 I, 131 1, 32 P, 33 P, 47 Sc, i n Ag,
  • therapeutic radionuclide preferably has a decay energy in the range of 20 to 6,000 keV, preferably in the ranges 60 to 200 keV for an Auger emitter, 100-2,500 keV for a beta emitter, and 4,000-6,000 keV for an alpha emitter.
  • Maximum decay energies of useful beta- particle-emitting nuclides are preferably 20-5,000 keV, more preferably 100-4,000 keV, and most preferably 500-2,500 keV. Also preferred are radionuclides that substantially decay with Auger-emitting particles.
  • beta- particle-emitting nuclides are preferably ⁇ 1,000 keV, more preferably ⁇ 100 keV, and most preferably ⁇ 70 keV. Also preferred are radionuclides that substantially decay with generation of alpha-particles.
  • Such radionuclides include, but are not limited to: Dy-152, At- 211, Bi-212, Ra-223, Rn-219, Po-215, Bi-21 1, Ac-225, Fr-221, At-217, Bi-213, Th-227 and Fm-255. Decay energies of useful alpha-particle-emitting radionuclides are preferably 2,000- 10,000 keV, more preferably 3,000-8,000 keV, and most preferably 4,000-7,000 keV.
  • 67 Cu considered one of the more promising radioisotopes for radioimmunotherapy due to its 61.5-hour half-life and abundant supply of beta particles and gamma rays, can be conjugated to an antibody using the chelating agent, p-bromoacetamido- benzyl-tetraethylaminetetraacetic acid (TETA).
  • TETA p-bromoacetamido- benzyl-tetraethylaminetetraacetic acid
  • 90 Y which emits an energetic beta particle, can be coupled to an antibody, antibody fragment or fusion protein, using diethylenetriaminepentaacetic acid (DTPA), or more preferably using DOTA.
  • DTPA diethylenetriaminepentaacetic acid
  • Additional potential therapeutic radioisotopes include U C, 13 N, 15 0, 75 Br, 198 Au, 224 Ac, 126 I, 133 1, 77 Br, 113m In, 95 Ru, 97 Ru, 103 Ru, 105 Ru, 107 Hg, 203 Hg, 121m Te, 122m Te, 125m Te, 165 Tm, 167 Tm, 168 Tm, 197 Pt, 109 Pd, 105 Rh, 142 Pr, 143 Pr, 161 Tb, 166 Ho, 199 Au, 57 Co, 58 Co, 51 Cr, 59 Fe, 75 Se, 201 T1, 225 Ac, 76 Br, 169 Yb, and the like.
  • a radiosensitizer can be used in combination with a naked or conjugated antibody or antibody fragment.
  • the radiosensitizer can be used in combination with a radiolabeled antibody or antibody fragment.
  • the addition of the radiosensitizer can result in enhanced efficacy when compared to treatment with the radiolabeled antibody or antibody fragment alone. Radiosensitizers are described in D. M. Goldenberg (ed.), CANCER THERAPY WITH RADIOLABELED ANTIBODIES, CRC Press (1995).
  • radionsensitizers of interest for use with this technology include gemcitabine, 5-fluorouracil, and cisplatin, and have been used in combination with external irradiation in the therapy of diverse cancers, including pancreatic cancer. Therefore, we have studied the combination of gemcitabine at what is believed to be radiosensitizing doses (once weekly 200 mg/m 2 over 4 weeks) of gemcitabine combined with fractionated doses of 90 Y- hPAM4, and have observed objective evidence of pancreatic cancer reduction after a single cycle of this combination that proved to be well-tolerated (no grade 3-4 toxicities by NCI- CTC v. 3 standard).
  • Antibodies or fragments thereof that have a boron addend-loaded carrier for thermal neutron activation therapy will normally be affected in similar ways. However, it will be advantageous to wait until non-targeted immunoconjugate clears before neutron irradiation is performed. Clearance can be accelerated using an anti-idiotypic antibody that binds to the anti-pancreatic cancer antibody. See U.S. Pat. No. 4,624,846 for a description of this general principle.
  • boron addends such as carboranes, can be attached to antibodies. Carboranes can be prepared with carboxyl functions on pendant side chains, as is well-known in the art.
  • Attachment of carboranes to a carrier can be achieved by activation of the carboxyl groups of the carboranes and condensation with amines on the carrier.
  • the intermediate conjugate is then conjugated to the antibody.
  • a boron addend is activated by thermal neutron irradiation and converted to radioactive atoms which decay by alpha-emission to produce highly toxic, short- range effects.
  • RNAi RNA interference
  • RISC RNA-induced silencing complex
  • Types of RNAi molecules include microRNA (miRNA) and small interfering RNA (siRNA).
  • mRNA messenger RNA
  • siRNA small interfering RNA
  • RNAi Upon binding to a complementary mRNA species, RNAi induces cleavage of the mRNA molecule by the argonaute component of RISC.
  • miRNA and siRNA differ in the degree of specificity for particular gene targets, with siRNA being relatively specific for a particular target gene and miRNA inhibiting translation of multiple mRNA species.
  • RNAi by inhibition of selected gene expression has been attempted for a variety of disease states, such as macular degeneration and respiratory syncytial virus infection (Sah, 2006, Life Sci 79: 1773-80). It has been suggested that siRNA functions in host cell defenses against viral infection and siRNA has been widely examined as an approach to anti-viral therapy (see, e.g., Zhang et al, 2004, Nature Med 1 1 :56-62; Novina et al, 2002, Nature Med 8:681-86; Palliser et al, 2006, Nature 439:89-94). The use of siRNA for cancer therapy has also been attempted. Fujii et al.
  • siRNA or interference RNA species may be attached to the subject antibodies.
  • siRNA and RNAi species against a wide variety of targets are known in the art, and any such known oligonucleotide species may be utilized in the claimed methods and compositions.
  • siRNA species of potential use include those specific for IKK-gamma (U.S. Patent 7,022,828); VEGF, Flt-1 and Flk-l/KDR (U.S. Patent 7, 148,342); Bcl2 and EGFR (U.S. Patent 7,541,453); CDC20 (U.S. Patent 7,550,572); transducin (beta)-like 3 (U.S. Patent 7,576, 196); KRAS (U.S. Patent 7,576, 197); carbonic anhydrase II (U.S. Patent 7,579,457); complement component 3 (U.S.
  • Patent 7,582,746 interleukin- 1 receptor- associated kinase 4 (IRAK4) (U.S. Patent 7,592,443); survivin (U.S. Patent 7,608,7070); superoxide dismutase 1 (U.S. Patent 7,632,938); MET proto-oncogene (U.S. Patent
  • amyloid beta precursor protein U.S. Patent 7,635,771
  • IGF-1R U.S. Patent 7,638,621
  • ICAM1 U.S. Patent 7,642,349
  • complement factor B U.S. Patent 7,696,344
  • p53 7,781,575)
  • apolipoprotein B 7,795,421
  • siRNA species are available from known commercial sources, such as Sigma-Aldrich (St Louis, MO), Invitrogen (Carlsbad, CA), Santa Cruz Biotechnology (Santa Cruz, CA), Ambion (Austin, TX), Dharmacon (Thermo Scientific, Lafayette, CO), Promega (Madison, WI), Minis Bio (Madison, WI) and Qiagen (Valencia, CA), among many others.
  • Other publicly available sources of siRNA species include the siRNAdb database at the Swedish Bioinformatics Centre, the MIT/ICBP siRNA Database, the RNAi Consortium shRNA Library at the Broad Institute, and the Probe database at NCBI.
  • siRNA species there are 30,852 siRNA species in the NCBI Probe database.
  • the skilled artisan will realize that for any gene of interest, either an siRNA species has already been designed, or one may readily be designed using publicly available software tools. Any such siRNA species may be delivered using the subject DNLTM complexes.
  • siRNA species that have been reported are listed in Table 1. Although siRNA is delivered as a double-stranded molecule, for simplicity only the sense strand sequences are shown in Table 1. Table 1. Exemplary siRNA Sequences
  • VEGF R2 AATGCGGCGGTGGTGACAGTA SEQ ID NO:22
  • VEGF R2 AAGCTCAGCACACAGAAAGAC SEQ ID NO:23
  • Sortilin 1 AGGTGGTGTTAACAGCAGAG SEQ ID NO:32 Apolipoprotein E AAGGTGGAGCAAGCGGTGGAG SEQ ID NO:33 Apolipoprotein E AAGGAGTTGAAGGCCGACAAA SEQ ID NO:34 Bcl-X UAUGGAGCUGCAGAGGAUGdTdT SEQ ID NO:35 Raf-1 TTTGAATATCTGTGCTGAGAACACA SEQ ID NO:36
  • IGFBP3 AAUCAUCAUCAAGAAAGGGCA SEQ ID NO:38
  • CEACAM1 AACCTTCTGGAACCCGCCCAC SEQ ID NO:47
  • diagnosis In the context of this application, the terms “diagnosis” or “detection” can be used interchangeably. Whereas diagnosis usually refers to defining a tissue's specific histological status, detection recognizes and locates a tissue, lesion or organism containing a particular antigen.
  • the subject antibodies and fragments can be detectably labeled by linking the antibody to an enzyme. When the antibody-enzyme conjugate is incubated in the presence of the appropriate substrate, the enzyme moiety reacts with the substrate to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric or visual means.
  • enzymes that can be used to detectably label antibody include malate dehydrogenase, staphylococcal nuclease, delta-V-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, alpha- galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • the immunoconjugate may comprise one or more radioactive isotopes useful for detecting diseased tissue.
  • Particularly useful diagnostic radionuclides include, but are not limited to, 110 In, m In, 177 Lu, 18 F, 52 Fe, 62 Cu, 64 Cu, 67 Cu, 67 Ga, 68 Ga, 86 Y, 90 Y, 89 Zr, 94m Tc, 94 Tc, 99m Tc, 120 I, 123 I, 124 1, 125 I, 131 I, 154"158 Gd, 32 P, n C, 13 N, 15 0, 186 Re, 188 Re, 51 Mn, 52m Mn, 55 Co, 72 As, 75 Br, 76 Br, 82m Rb, 83 Sr, or other gamma-, beta-, or positron-emitters, preferably with a decay energy in the range of 20 to 4,000 keV, more preferably in the range of 25 to 4,000 keV, and even more preferably in the range of 25 to 1,000
  • Total decay energies of useful positron-emitting radionuclides are preferably ⁇ 2,000 keV, more preferably under 1,000 keV, and most preferably ⁇ 700 keV.
  • Radionuclides useful as diagnostic agents utilizing gamma-ray detection include, but are not limited to: 51 Cr, 57 Co, 58 Co, 59 Fe, 67 Cu, 67 Ga, 75 Se, 97 Ru, 99m Tc, m In, 114m In, 123 1, 125 I, 131 I, 169 Yb, 197 Hg, and 201 T1.
  • Decay energies of useful gamma-ray emitting radionuclides are preferably 20-2000 keV, more preferably 60-600 keV, and most preferably 100-300 keV.
  • Methods of diagnosing cancer in a subject may be accomplished by administering a diagnostic immunoconjugate and detecting the diagnostic label attached to an
  • a suitable non-radioactive diagnostic agent is a contrast agent suitable for magnetic resonance imaging, X-rays, computed tomography or ultrasound.
  • Magnetic imaging agents include, for example, nonradioactive metals, such as manganese, iron and gadolinium, complexed with metal-chelate combinations that include 2-benzyl-DTPA and its monomethyl and cyclohexyl analogs. See U.S. Ser. No. 09/921,290 (now abandoned) filed on Oct. 10, 2001, the Examples section of which is incorporated herein by reference.
  • Other imaging agents such as PET scanning nucleotides, preferably 18 F, may also be used.
  • Contrast agents such as MRI contrast agents, including, for example, gadolinium ions, lanthanum ions, dysprosium ions, iron ions, manganese ions or other comparable labels, CT contrast agents, and ultrasound contrast agents may be used as diagnostic agents.
  • MRI contrast agents including, for example, gadolinium ions, lanthanum ions, dysprosium ions, iron ions, manganese ions or other comparable labels, CT contrast agents, and ultrasound contrast agents may be used as diagnostic agents.
  • Paramagnetic ions suitable for use include chromium (III), manganese (II), iron (III), iron
  • gadolinium is particularly preferred.
  • Ions useful in other contexts include but are not limited to lanthanum (III), gold (III), lead (II) and bismuth (III).
  • Fluorescent labels include rhodamine, fluorescein and renographin. Rhodamine and fluorescein are often linked via an
  • Metals are also useful in diagnostic agents, including those for magnetic resonance imaging techniques. These metals include, but are not limited to: gadolinium, manganese, iron, chromium, copper, cobalt, nickel, dysprosium, rhenium, europium, terbium, holmium and neodymium. In order to load an antibody with radioactive metals or paramagnetic ions, it may be necessary to react it with a reagent having a long tail to which are attached a multiplicity of chelating groups for binding the ions.
  • Such a tail can be a polymer such as a polylysine, polysaccharide, or other derivatized or derivatizable chain having pendant groups to which can be bound chelating groups such as, e.g., ethylenediaminetetraacetic acid (EDTA), diethylenetriaminepentaacetic acid (DTPA), porphyrins, polyamines, crown ethers, bis-thiosemicarbazones, polyoximes, and like groups known to be useful for this purpose.
  • chelating groups such as, e.g., ethylenediaminetetraacetic acid (EDTA), diethylenetriaminepentaacetic acid (DTPA), porphyrins, polyamines, crown ethers, bis-thiosemicarbazones, polyoximes, and like groups known to be useful for this purpose.
  • Chelates are coupled to an antibody, fusion protein, or fragments thereof using standard chemistries.
  • the chelate is normally linked to the antibody by a group which enables formation of a bond to the molecule with minimal loss of immunoreactivity and minimal aggregation and/or internal cross-linking.
  • Other, more unusual, methods and reagents for conjugating chelates to antibodies are disclosed in U.S. Pat. No. 4,824,659 to Hawthorne, entitled “Antibody Conjugates", issued Apr. 25, 1989, the Examples section of which is incorporated herein by reference.
  • Particularly useful metal-chelate combinations include 2-benzyl-DTPA and its monomethyl and cyclohexyl analogs, used with diagnostic isotopes in the general energy range of 20 to 2,000 keV.
  • the same chelates, when complexed with non-radioactive metals, such as manganese, iron and gadolinium are useful for MRI.
  • Macrocyclic chelates such as NOTA, DOTA, and TETA are of use with a variety of metals and radiometals, most particularly with radionuclides of gallium, yttrium and copper, respectively.
  • Such metal-chelate complexes can be made very stable by tailoring the ring size to the metal of interest.
  • Other ring-type chelates such as macrocyclic polyethers, which are of interest for stably binding nuclides, such as 223Ra for RAIT are encompassed by the invention.
  • Radiopaque and contrast materials are used for enhancing X-rays and computed tomography, and include iodine compounds, barium compounds, gallium compounds, thallium compounds, etc. Specific compounds include barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric acid, iosulamide meglumine, iosemetic acid, iotasul, iotetric acid, iothalamic acid, iotroxic acid, ioxaglic acid, ioxotrizoic acid, ipodate, meglumine, metrizamide,
  • the antibodies, antibody fragments and fusion proteins also can be labeled with a fluorescent compound.
  • the presence of a fluorescent-labeled MAb is determined by exposing the antibody to light of the proper wavelength and detecting the resultant fluorescence.
  • Fluorescent labeling compounds include Alexa 350, Alexa 430, AMCA, aminoacridine, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, 5-carboxy-4',5'-dichloro-2',7'-dimethoxy fluorescein, 5- carboxy-2',4',5',7'-tetrachlorofluorescein, 5-carboxyfluorescein, 5-carboxyrhodamine, 6- carboxyrhodamine, 6-carboxytetramethyl amino, Cascade Blue, Cy2, Cy3, Cy5,6-FAM, dansyl chloride, Fluoresc
  • the antibodies, antibody fragments and fusion proteins can be detectably labeled by coupling the antibody to a chemiluminescent compound.
  • the presence of the chemiluminescent-tagged MAb is determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of
  • chemiluminescent labeling compounds include luminol, isoluminol, an aromatic acridinium ester, an imidazole, an acridinium salt and an oxalate ester.
  • Bioluminescent compound can be used to label the antibodies and fragments there.
  • Bioluminescence is a type of chemiluminescence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Bioluminescent compounds that are useful for labeling include luciferin, luciferase and aequorin.
  • a method of diagnosing a malignancy in a subject comprising performing an in vitro diagnosis assay on a specimen (fluid, tissue or cells) from the subject with a composition comprising an anti-pancreatic cancer MAb, fusion protein or fragment thereof.
  • Immunohistochemistry can be used to detect the presence of PAM4 antigen in a cell or tissue by the presence of bound antibody.
  • the malignancy that is being diagnosed is a cancer.
  • the cancer is pancreatic cancer.
  • a chelator such as DTPA, DOTA, TETA, or NOTA or a suitable peptide, to which a detectable label, such as a fluorescent molecule, or cytotoxic agent, such as a heavy metal or radionuclide, can be conjugated to a subject antibody.
  • a detectable label such as a fluorescent molecule, or cytotoxic agent, such as a heavy metal or radionuclide
  • a therapeutically useful immunoconjugate can be obtained by conjugating a photoactive agent or dye to an antibody fusion protein.
  • Fluorescent compositions, such as fluorochrome, and other chromogens, or dyes, such as porphyrins sensitive to visible light have been used to detect and to treat lesions by directing the suitable light to the lesion.
  • Fluorescent and radioactive agents conjugated to antibodies or used in bispecific, pretargeting methods are particularly useful for endoscopic, intraoperative or intravascular detection of the targeted antigens associated with diseased tissues or clusters of cells, such as malignant tumors, as disclosed in Goldenberg U.S. Pat. Nos. 5,716,595; 6,096,289 and 6,387,350, the Examples section of each incorporated herein by reference, particularly with gamma-, beta- and positron-emitters.
  • Endoscopic applications may be used when there is spread to a structure that allows an endoscope, such as the colon.
  • Radionuclides useful for positron emission tomography include, but are not limited to: F-18, Mn-51, Mn-52m, Fe-52, Co-55, Cu-62, Cu-64, Ga-68, As-72, Br-75, Br-76, Rb-82m, Sr-83, Y-86, Zr-89, Tc-94m, In- 1 10, 1-120, and 1-124.
  • Total decay energies of useful positron-emitting radionuclides are preferably ⁇ 2,000 keV, more preferably under 1,000 keV, and most preferably ⁇ 700 keV.
  • Radionuclides useful as diagnostic agents utilizing gamma-ray detection include, but are not limited to: Cr-51, Co-57, Co-58, Fe-59, Cu-67, Ga-67, Se-75, Ru-97, Tc-99m, In-I l l, In- 1 14m, 1-123, 1-125, 1-131, Yb-169, Hg-197, and Tl-201. Decay energies of useful gamma- ray emitting radionuclides are preferably 20-2000 keV, more preferably 60-600 keV, and most preferably 100-300 keV.
  • the present invention contemplates the use of anti-pancreatic cancer antibodies to screen biological samples in vitro for the presence of the PAM4 antigen.
  • the antibody, antibody fragment or fusion protein may be utilized in liquid phase or bound to a solid-phase carrier, as described below.
  • any type of antibody such as murine, chimeric, humanized or human may be utilized, since there is no host immune response to consider.
  • a screening method for determining whether a biological sample contains MUC5ac is the radioimmunoassay (RIA).
  • RIA radioimmunoassay
  • the substance under test is mixed with PAM4 MAb in the presence of radiolabeled MUC5ac.
  • concentration of the test substance will be inversely proportional to the amount of labeled MUC5ac bound to the MAb and directly related to the amount of free, labeled MUC5ac.
  • in vitro assays can be performed in which an anti-pancreatic cancer antibody, antibody fragment or fusion protein is bound to a solid-phase carrier.
  • MAbs can be attached to a polymer, such as aminodextran, in order to link the MAb to an insoluble support such as a polymer-coated bead, a plate or a tube.
  • the presence of the PAM4 antigen in a biological sample can be determined using an enzyme-linked immunosorbent assay (ELISA) (e.g., Gold et al. J Clin Oncol. 24:252-58, 2006).
  • ELISA enzyme-linked immunosorbent assay
  • a pure or semipure antigen preparation is bound to a solid support that is insoluble in the fluid or cellular extract being tested and a quantity of detectably labeled soluble antibody is added to permit detection and/or quantitation of the binary complex formed between solid-phase antigen and labeled antibody.
  • a double-determinant ELISA also known as a “two-site ELISA” or “sandwich assay”
  • the double-determinant ELISA is preferred to the direct competitive ELISA for the detection of an antigen in a clinical sample. See, for example, the use of the double-determinant ELISA for quantitation of the c-myc oncoprotein in biopsy specimens. Field et al, Oncogene 4: 1463 (1989); Spandidos et al, Anticancer Res. 9: 821 (1989).
  • a quantity of unlabeled MAb or antibody fragment (the "capture antibody") is bound to a solid support, the test sample is brought into contact with the capture antibody, and a quantity of detectably labeled soluble antibody (or antibody fragment) is added to permit detection and/or quantitation of the ternary complex formed between the capture antibody, antigen, and labeled antibody.
  • an antibody fragment is a portion of an anti-pancreatic cancer MAb that binds to an epitope of MUC5ac.
  • the soluble antibody or antibody fragment must bind to a MUC5ac epitope that is distinct from the epitope recognized by the capture antibody.
  • the double-determinant ELISA can be performed to ascertain whether the PAM4 antigen is present in a biopsy sample.
  • the assay can be performed to quantitate the amount of MUC5ac that is present in a clinical sample of body fluid.
  • the quantitative assay can be performed by including dilutions of purified MUC5ac.
  • kits may comprise a carrier means that is compartmentalized to receive in close confinement one or more container means such as vials, tubes and the like, each of said container means comprising the separate elements of the immunoassay.
  • the subject antibodies, antibody fragments and fusion proteins also can be used to detect the presence of the PAM4 antigen in tissue sections prepared from a histological specimen.
  • Such in situ detection can be used to determine the presence of MUC5ac and to determine the distribution of MUC5ac in the examined tissue.
  • In situ detection can be accomplished by applying a detectably-labeled antibody to frozen tissue sections. Studies indicate that the PAM4 antigen is preserved in paraffin-embedded sections. General techniques of in situ detection are well-known to those of ordinary skill. See, for example, Ponder, "Cell Marking Techniques and Their Application," in MAMMALIAN
  • Antibodies, antibody fragments and fusion proteins can be detectably labeled with any appropriate marker moiety, for example, a radioisotope, an enzyme, a fluorescent label, a dye, a chromogen, a chemiluminescent label, a bioluminescent labels or a paramagnetic label.
  • a marker moiety for example, a radioisotope, an enzyme, a fluorescent label, a dye, a chromogen, a chemiluminescent label, a bioluminescent labels or a paramagnetic label.
  • the marker moiety can be a radioisotope that is detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.
  • a radioisotope that is detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.
  • the diagnostic conjugate is a gamma-, beta- or a positron-emitting isotope.
  • a marker moiety in the present description refers to a molecule that will generate a signal under predetermined conditions.
  • marker moieties include radioisotopes, enzymes, fluorescent labels, chemiluminescent labels, bioluminescent labels and paramagnetic labels.
  • the binding of marker moieties to anti-pancreatic cancer antibodies can be accomplished using standard techniques known to the art. Typical methodology in this regard is described by Kennedy et al, Clin Chim Acta 70: 1 (1976), Schurs et al, Clin. Chim. Acta 81 : 1 (1977), Shih et al, Int J Cancer 46: 1 101 (1990).
  • the above-described in vitro and in situ detection methods may be used to assist in the diagnosis or staging of a pathological condition.
  • such methods can be used to detect tumors that express the PAM4 antigen such as pancreatic cancer.
  • radioisotopes may be bound to antibody either directly or indirectly by using an intermediary functional group.
  • intermediary functional groups include chelators such as ethylenediaminetetraacetic acid and diethylenetriaminepentaacetic acid. For example, see Shih et al, supra, and U.S. Pat. No. 5,057,313.
  • the radiation dose delivered to the patient is maintained at as low a level as possible through the choice of isotope for the best combination of minimum half-life, minimum retention in the body, and minimum quantity of isotope which will permit detection and accurate measurement.
  • radioisotopes that can be bound to anti -pancreatic cancer antibody and are appropriate for diagnostic imaging include 99m Tc, m In and 18 F.
  • the subject antibodies, antibody fragments and fusion proteins also can be labeled with paramagnetic ions and a variety of radiological contrast agents for purposes of in vivo diagnosis.
  • Contrast agents that are particularly useful for magnetic resonance imaging comprise gadolinium, manganese, dysprosium, lanthanum, or iron ions. Additional agents include chromium, copper, cobalt, nickel, rhenium, europium, terbium, holmium, or neodymium.
  • Antibodies and fragments thereof can also be conjugated to ultrasound contrast/enhancing agents.
  • one ultrasound contrast agent is a liposome.
  • the ultrasound contrast agent is a liposome that is gas filled.
  • a bispecific antibody can be conjugated to a contrast agent.
  • the bispecific antibody may comprise more than one image-enhancing agent for use in ultrasound imaging.
  • the contrast agent is a liposome.
  • the liposome comprises a bivalent DTPA-peptide covalently attached to the outside surface of the liposome.
  • Control release preparations can be prepared through the use of polymers to complex or adsorb the antibody, antibody fragment or fusion protein.
  • biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebacic acid. Sherwood et al., Bio/Technology 10: 1446 (1992).
  • the rate of release of an antibody, antibody fragment or fusion protein from such a matrix depends upon the molecular weight of the antibody, antibody fragment or fusion protein, the amount of antibody within the matrix, and the size of dispersed particles. Saltzman et al, Biophys. J. 55: 163 (1989); Sherwood et al, supra. Other solid dosage forms are described in Ansel et al, PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th Edition (Lea & Febiger 1990), and Gennaro (ed.), REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition (Mack Publishing Company 1990), and revised editions thereof.
  • the antibodies, fragments thereof or fusion proteins to be delivered to a subject can comprise one or more pharmaceutically suitable excipients, one or more additional ingredients, or some combination of these.
  • the antibody can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the
  • a pharmaceutically suitable excipient Sterile phosphate-buffered saline is one example of a pharmaceutically suitable excipient.
  • Other suitable excipients are well-known to those in the art. See, for example, Ansel et al, PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th Edition (Lea & Febiger 1990), and Gennaro (ed.), REMINGTON'S
  • the immunoconjugate or naked antibody can be formulated for intravenous administration via, for example, bolus injection or continuous infusion.
  • Formulations for injection can be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the immunoconjugate, naked antibody, fragment thereof or fusion protein may also be administered to a mammal subcutaneous ly or by other parenteral routes.
  • the antibody or fragment thereof is administered in a dosage of 20 to 2000 milligrams protein per dose.
  • the administration may be by continuous infusion or by single or multiple boluses.
  • the dosage of an administered immunoconjugate, fusion protein or naked antibody for humans will vary depending upon such factors as the patient's age, weight, height, sex, general medical condition and previous medical history.
  • a dosage of immunoconjugate, antibody fusion protein or naked antibody that is in the range of from about 1 mg/kg to 20 mg/kg as a single intravenous or infusion, although a lower or higher dosage also may be administered as circumstances dictate.
  • This dosage may be repeated as needed, for example, once per week for four to ten weeks, preferably once per week for eight weeks, and more preferably, once per week for four weeks. It may also be given less frequently, such as every other week for several months, or more frequently, such as two- or three-time weekly.
  • the dosage may be given through various parenteral routes, with appropriate adjustment of the dose and schedule.
  • kits containing components suitable for treating or diagnosing diseased tissue in a patient may contain at least one antibody, antigen binding fragment or fusion protein as described herein. If the composition containing components for administration is not formulated for delivery via the alimentary canal, such as by oral delivery, a device capable of delivering the kit components through some other route may be included.
  • a device capable of delivering the kit components through some other route may be included.
  • an anti-pancreatic cancer antibody or antigen binding fragment thereof may be provided in the form of a prefilled syringe or autoinjection pen containing a sterile, liquid formulation or lyophilized preparation of antibody (e.g., Kivitz et al, Clin. Ther. 2006, 28: 1619-29).
  • the kit components may be packaged together or separated into two or more containers.
  • the containers may be vials that contain sterile, lyophilized formulations of a composition that are suitable for reconstitution.
  • a kit may also contain one or more buffers suitable for reconstitution and/or dilution of other reagents.
  • Other containers that may be used include, but are not limited to, a pouch, tray, box, tube, or the like. Kit components may be packaged and maintained sterilely within the containers. Another component that can be included is instructions for use of the kit.
  • the claimed methods and compositions utilize the antibody hPAM4 which is a humanized IgG of the murine PAM4 MAb raised against pancreatic cancer mucin. Humanization of the murine PAM4 sequences was utilized to reduce the human antimouse antibody (HAMA) response. To produce the humanized PAM4, murine complementarity determining regions (CDR) were transferred from heavy and light variable chains of the mouse immunoglobulin into human framework region (FR) antibody sequences, followed by the replacement of some human FR residues with their murine counterparts. Humanized monoclonal antibodies are suitable for use in in vitro and in vivo diagnostic and therapeutic methods.
  • HAMA human antimouse antibody
  • CDR murine complementarity determining regions
  • variable region framework sequences of the murine PAM4 MAb (FIGS. 1A and IB) to known human antibodies in the Kabat database showed that the FRs of PAM4 VK and VH exhibited the highest degree of sequence homology to that of the human antibodies Walker VK (FIG. 3A) and Wil2 V H (FIG. 3B), respectively. Therefore, the Walker VK (FIG. 3 A) and Wil2 VH (FIG. 3B) FRs were selected as the human frameworks into which the murine CDRs for PAM4 VK and VH were grafted, respectively.
  • the FR4 sequence of the human antibody, NEWM was used to replace the Wil2 FR4 sequence for the humanization of the PAM4 heavy chain (FIG. 3B).
  • a few amino acid residues in PAM4 FRs that flank the putative CDRs were maintained in hPAM4 based on the consideration that these residues have more impact on Ag binding than other FR residues. These residues were 21M, 47W, 59P, 60A, 85S, 87F, and 100G of VK (FIG. 3A) and 27Y, 30P, 38K, 481, 66K, 67A, and 69L of V H (FIG. 3B).
  • the DNA and amino acid sequences of hPAM4 VK (SEQ ID NO: 16) and V H (SEQ ID NO: 19) are shown in FIGS. 4A and 4B, respectively.
  • hPAM4 V H A represents nt 17 to 189 of the hPAM4 V H domain.
  • hPAM4 VHB represents the minus strand of the hPAM4 VH domain complementary to nt 169 to 341.
  • oligonucleotides Each annealed end serves as a primer for the transcription of the single stranded DNA, resulting in a double strand DNA composed of the nt 17 to 341 of hPAM4 VH.
  • This DNA was further amplified in the presence of two short oligonucleotides, hPAM4 VHBACK and hPAM4 V H FOR to form the full-length hPAM4 V H .
  • the underlined portions are restriction sites for subcloning as shown in FIG. 4B.
  • hPAM4 VHA and VHB were amplified in the presence of 10 ⁇ , of 10X PCR Buffer (500 mM KCl, 100 mM Tris HCl buffer, pH 8.3, 15 mM MgCl 2 ), 2 ⁇ of hPAM4 VHBACK and hPAM4 V K FOR, and 2.5 units of Taq DNA polymerase (Perkin Elmer Cetus, Norwalk, Conn.).
  • This reaction mixture was subjected to three cycles of polymerase chain reaction (PCR) consisting of denaturation at 94°C for 1 minute, annealing at 45°C for 1 minute, and polymerization at 72°C for 1.5 minutes.
  • PCR polymerase chain reaction
  • Double-stranded PCR-amplified product for hPAM4 V H was gel-purified, restriction-digested with Pstl and BstEII restriction sites and cloned into the complementary Pstl/BstEII restriction sites of the heavy chain staging vector, VnpBS2, in which the VH sequence was fully assembled with the DNA sequence encoding the translation initiation codon and a secretion signal peptide in-frame ligated at the 5 '-end and an intron sequence at the 3'-end.
  • VnpBS2 is a modified staging vector of VnpBS (Leung et al, Hybridoma, 13:469, 1994), into which a Xhol restriction site was introduced at sixteen bases upstream of the translation initiation codon to facilitate the next subcloning step.
  • the assembled VH gene was subcloned as a XhoI-BamHI restriction fragment into the expression vector, pdHL2, which contains the expression cassettes for both human IgG heavy and light chains under the control of IgH enhancer and MT1 promoter, as well as a mouse d/fr gene as a marker for selection and amplification.
  • hPAM4 VnpdHL2 Since the heavy chain region of pdHL2 lacks a BamHI restriction site, this ligation requires use of a linker to provide a bridge between the BamHI site of the variable chain and the Hindlll site present in the pdHL2 vector.
  • the resulting expression vectors were designated as hPAM4 VnpdHL2.
  • hPAM4 V A (157-mer) and hPAM4 V B (156-mer) were synthesized as described above.
  • hPAM4 V K A and V K B were amplified by two short oligonucleotides hPAM4 V K BACK and hPAM4 V K FOR as described above.
  • hPAM4 V K A represents nt 16 to 172 of the hPAM4 VK domain.
  • hPAM4 V B represents the minus strand of the hPAM4 VK domain complementary to nt 153 to 308.
  • oligonucleotides Each annealed end served as a primer for the transcription of the single stranded DNA, resulting in a double strand DNA composed of nt 16 to 308 of hPAM4 VK.
  • This DNA was further amplified in the presence of two short oligonucleotides, hPAM4 V K BACK and hPAM4 V K FOR to form the full-length hPAM4 VK.
  • the underlined portions are restriction sites for subcloning as described below.
  • V K PBR2 is a modified staging vector of V K PBR (Leung et al, Hybridoma, 13:469, 1994), into which a Xbal restriction site was introduced at sixteen bases upstream of the translation initiation codon.
  • the assembled VK genes were subcloned as Xbal-BamHI restriction fragments into the expression vector containing the V H sequence, hPAM4
  • V H pdHL2 The resulting expression vectors were designated as hPAM4pdHL2.
  • hPAM4pdHL2 was linearized by digestion with Sail and transfected into Sp2/0-Agl4 cells by electroporation at 450 V and 25 ⁇ .
  • the transfected cells were plated into 96-well plates and incubated in a CO 2 cell culture incubator for two days and then selected for MTX resistance. Colonies surviving selection emerged in two to three weeks and were screened for human antibody secretion by ELISA assay. Briefly, supernatants (-100 ul) from the surviving colonies were added into the wells of an ELISA microplate precoated with goat anti-human IgG F(ab')2 fragment-specific Ab. The plate was incubated for one hour at room temperature.
  • Unbound proteins were removed by washing three times with wash buffer (PBS containing 0.05% Tween-20). Horseradish peroxidase- conjugated goat anti-human IgG Fc fragment-specific Ab was added to the wells. Following incubation for one hour, a substrate solution (100 ⁇ / ⁇ ) containing 4 mM o- phenylenediamine dihydrochloride (OPD) and 0.04% H2O2 in PBS was added to the wells after washing. Color was allowed to develop in the dark for 30 minutes and the reaction was stopped by the addition of 50 ⁇ , of 4 N H2SO4 solution. The bound human IgG was measured by reading the absorbance at 490 nm on an ELISA reader. Positive cell clones were expanded and hPAM4 was purified from cell culture supernatant by affinity chromatography on a Protein A column.
  • the Ag-binding activity of hPAM4 was confirmed by ELISA assay in a microtiter plate coated with pancreas cancer cell extracts.
  • An ELISA competitive binding assay using PAM4-antigen coated plates was developed to assess the Ag-binding affinity of hPAM4 in comparison with that of a chimeric PAM4 composed of murine V and human C domains.
  • Constant amounts of the HRP-conjugated cPAM4 mixed with varying concentrations of cPAM4 or hPAM4 were added to the coated wells and incubated at room temperature for 1 -2 h.
  • the amount of HRP-conjugated cPAM4 bound to the CaPanl Ag was revealed by reading the absorbance at 490 nm after the addition of a substrate solution containing 4 mM 0- phenylenediamine dihydrochloride and 0.04% 3 ⁇ 4(3 ⁇ 4. As shown by the competition assays in FIG. 5, hPAM4 and cPAM4 antibodies exhibited similar binding activities.
  • PAM4 was reactive with twenty one out of twenty five (85%) pancreatic cancers (Table 4 and Table 5) and ten out of twenty six colon cancers, but only limited reactivity with tumors of the stomach, lung, breast, ovary, prostate, liver or kidney (Table 5). PAM4 reactivity appeared to correlate with the stage of tumor differentiation, with a greater percentage of staining observed in well differentiated pancreatic cancers than in moderately differentiated or poorly differentiated tumors. Generally, poorly differentiated tumors represent less than 10% of all pancreatic cancers.
  • PAM4 was found to be only weakly reactive with three out of twelve chronic pancreatitis specimens, whereas CA19.9 and DUPAN2 were strongly reactive with all twelve specimens. Although it is recognized that specificity is dependent in part upon the type of assay employed and the range and number of tissues examined, the ability of PAM4 to discriminate between normal and neoplastic pancreatic tissue, its ability to react with a large percentage of the cancer specimens, the high intensity of the reactions, and the ability to distinguish between early stage pancreatic cancer and benign conditions such as pancreatitis are important characteristics of this exemplary anti-pancreatic cancer antibody.
  • a - values are mean from two autopsy specimens
  • the biodistribution data were used to estimate potential radiation doses to the tumor of 12,230; 10,684; 6,835; and 15,843 cGy/mCi of injected dose to AsPcl, BxPc3, Hs766T and CaPanl, respectively.
  • MTD maximum tolerated dose
  • PAM4 could provide substantial rad dose to each of the xenografted tumor models.
  • the blood levels of radiolabeled PAM4 were significantly ( ⁇ 0.01-0.001) lower than the nonspecific Ag8.
  • Potential radiation doses to the blood from PAM4 were 1.4-4.4 fold lower than from Ag8.
  • the tumors received doses that were 2.2; 3.3; 3.4; and 13.1-fold higher than blood, respectively.
  • PAM4 showed no evidence of targeting to normal tissues, except in the CaPanl tumor model, where a small but statistically significant splenic uptake was observed (range 3.1-7.5% ID/g on day-3). This type of splenic targeting has been observed in the clinical application of the anti-mucin antibodies B72.3 and CC49. Importantly, these studies also reported that splenic targeting did not affect tumor uptake of antibody nor did it interfere with interpretation of the nuclear scans.
  • splenic targeting was not due to crossreactive antigens in the spleen, nor to binding by Fc receptors, but rather to one or more of the following possibilities: direct targeting of antigen trapped in the spleen, or indirect uptake of antigen:antibody complexes formed either in the blood or released from the tumor site. The latter would require the presence of immune complexes in the blood.
  • Radiolabeled 131 I-PAM4 administered to animals bearing four week old orthotopic tumors (approximately 0.2 g) showed specific targeting to the primary tumor with localization indices of 7.9 ⁇ 3.0 at day one increasing to 22.8 ⁇ 15.3 at day fourteen. No evidence of specific targeting to other tissues was noted. In one case where tumor metastases to the liver and spleen were observed, both metastases were targeted, and had high concentrations of radiolabeled antibody. In addition, approximately half of the animals developed a subcutaneous tumor at the incision site. No significant differences were noted in the targeting of orthotopic and subcutaneous tumors within the same animal, and no significant differences were observed in the targeting of orthotopic tumor whether or not the animal had an additional subcutaneous tumor. The estimated radiation doses from PAM4 were 6,704 and 1,655 cGy/mCi to the primary tumor and blood, respectively. Example 5. Radioimmunotherapy of Pancreatic Cancer
  • Toxicity studies indicate the ability to give multiple cycles of radioimmunotherapy, which may be more effective with a larger tumor burden.
  • the untreated group had a mean survival time of 3.7 ⁇ 1.0 weeks (survival defined as time for tumor to reach 5 cm 3 ). Animals died as early as three weeks, with no animal surviving past six weeks.
  • a single dose of 350 ⁇ 131 I- PAM4 produced a significant increase in the survival time to 18.8 ⁇ 4.2 weeks ( O.0001).
  • the range of animal deaths extended from weeks thirteen to twenty five. None of the animals were alive at the end of the study period of twenty six weeks.
  • Radiolabeled PAM4 provided a significant survival advantage ( ⁇ 0.001) as compared to the untreated or Ag8 treated animals, with 70% survival at sixteen weeks, the end of the experiment. At this time the surviving animals were sacrificed to determine tumor size. All animals had tumor with an average weight of 1.2 g, as well as one or two small ( ⁇ 0.1 g) metastases evident in four of the seven animals. At sixteen weeks of growth, these tumors were more
  • periodate studies have shown as high as a 60% loss of immunoreactivity with PAM4 antibody (not shown).
  • the results of periodate and DTT studies suggest that the PAM4 epitope is conformationally dependent upon some minimal form of glycosylation, and may be affected by intermolecular disulfide bond formation.
  • the PAM4 antibody appears to bind to the same antigenic protein as the 45M1 anti-MUC5ac MAb, it is noted that MUC5ac is not specific to pancreas cancer and it is found in a number of normal tissues (other than the gastric mucosa with which PAM4 is reactive). For example, MUC5ac is found in normal lung, colon and other tissues. PAM4 antibody does not bind to normal lung tissues, except as indicated above in few samples and to a limited or minimal amount.
  • PAM4 antibody binding was examined with two different phage display peptide libraries. The first was a linear peptide library consisting of 12 amino acid sequences and the second was a cyclic peptide consisting of 7 amino acid sequences cyclized by a disulfide bridge. We panned the individual libraries alternately against hPAM4 and hLL2 (negative selection with anti-CD22 antibody) for a combined total of 4 rounds, and then screened the phage displayed peptide for reactivity with both hPAM4 and mPAM4 with little to no reactivity against hLL2. Phage binding in a non-specific manner (i.e., binding to
  • the sequence WTWNITKAYPLP (SEQ ID NO:7) was identified 30 times (in 35 sequenced phage), each of which were shown to have reactivity with PAM4 antibodies.
  • a mutational analysis was conducted in which a library based on this sequence and having 7.5% degeneracy at each position, was constructed, panned and screened as before. Variability was noted in the 19 obtained peptide sequences that were positive for PAM4 binding with 7 being identical to the parental sequence, 5 having the sequence WTW TKEYPQP (SEQ ID NO:62) and the rest being uniquely present. Table 6 shows the results of this mutational analysis.
  • the upper row lists the sequences identified and the lower row lists the frequency with which each of the amino acids was identified in that position.
  • the parent sequence is most frequent (bold) with the next highest variation a substitution of E for A at position 8 and a substitution of Q for L at position 1 1. It does not appear that these substitutions had any great effect upon immunoreactivity.
  • GTTGTTC (SEQ ID NO: 64) is present within the MUC5ac protein towards the amino terminus as compared to the cyclic peptide sequence shown above, which shows homology at the C-terminal end of the consensus peptide sequence.
  • the cyclic peptide only showed approximately 10% of the immunoreactivity of the linear sequence with the PAM4 antibody.
  • Both linear and cyclic consensus sequences are associated with a cysteine, which may or may not relate to the effect of DTT on MUC5ac immunoreactivity.
  • pancreatic carcinoma such as pancreatitis, diabetes, smoking and others.
  • screening measures are particularly important for the early detection of pancreatic neoplasia.
  • MAb-MA5 as a positive control for pancreatic ductal and acinar cells.
  • the two control MAbs provided immunohistologic evidence consistent with pancreatitis, in no instance did PAM4 react with inflamed pancreatic tissue.
  • pancreatic adenocarcinoma was also present within the tissue specimen.
  • PAM4 gave an intense stain of the neoplastic cells within this tumor.
  • PAM4 a small PanlN precursor lesion was identified that was labeled with PAM4. Labeling of the PanlN within this latter specimen is consistent with early detection of pancreatic neoplasia in a patient diagnosed with a non-malignant disease.
  • Example 11 Therapy of a Patient With Inoperable and Metastatic Pancreatic
  • Patient 1 18-001, CWG is a 63 -year-old man with Stage-IV pancreatic adenocarcinoma with multiple liver metastases, diagnosed in November of 2007. He agreed to undertake combined radioimmunotherapy and gemcitabine chemotherapy as a first treatment strategy, and was then given a first therapy cycle of 6.5 mCi/m 2 of 90 Y-hPAM4, combined with 200 mg/m 2 gemcitabine, whereby the gemcitabine was given once weekly on weeks 1-4 and 90 Y- hPAM4 was given once-weekly on weeks 2-4 (3 doses). Two months later, the same therapy cycle was repeated, because no major toxicities were noted after the first cycle.
  • Example 12 Therapy of a Patient with Inoperable Metastatic Pancreatic Carcinoma
  • 90 Y-DOTA-hPAM4 radiolabeled humanized antibody is administered at a dose of 10 mCi/m 2 of 90 Y and 20 mg antibody protein, in a two-hour i.v. infusion.
  • the patient is given a course of gemcitabine chemotherapy consisting of 3 weekly doses of 600 mg/m 2 by i.v. infusion.
  • the patient is then evaluated 4 weeks later, and has a mild leukopenia (grade-2), no other major blood or enzyme changes over baseline, but shows an improvement in the blood CA19.9 titer from 5,700 to 1,200 and a decrease in jaundice, with an overall subjective improvement.
  • grade-2 mild leukopenia
  • This follows 3 weeks later with a repeat of the cycle of lower-dose gemcitabine (weekly x 4), with 3 doses of 90 Y- DOTA-hPAM4.
  • the patient is reevaluated, and the CT and PET scans confirm an approximately 40% reduction of total tumor mass (primary cancer and metastases), with a further reduction of the CA19.9 titer to 870.
  • the patient regains appetite and activity, and is able to return to more usual daily activities without the need for pain medication. He gains 12 lbs after beginning this experimental therapy. A repeat of the scans and blood values indicates that this response is maintained 6 weeks later.
  • the DNLTM technique can be used to make dimers, trimers, tetramers, hexamers, etc. comprising virtually any antibodies or fragments thereof or other effector moieties.
  • IgG antibodies or Fab antibody fragments may be produced as fusion proteins containing either a dimerization and docking domain (DDD) or anchoring domain (AD) sequence.
  • DDD and AD moieties are produced as fusion proteins, the skilled artisan will realize that other methods of conjugation, such as chemical cross-linking or click chemistry, may be utilized within the scope of the claimed methods and compositions.
  • Bispecific antibodies may be formed by combining a Fab-DDD fusion protein of a first antibody with a Fab-AD fusion protein of a second antibody.
  • constructs may be made that combine IgG-AD fusion proteins with Fab-DDD fusion proteins.
  • the technique is not limiting and any protein or peptide of use may be produced as an AD or DDD fusion protein for incorporation into a DNLTM construct.
  • the AD and DDD conjugates are not limited to proteins or peptides and may comprise any molecule that may be cross-linked to an AD or DDD sequence using any cross- linking technique known in the art.
  • a polyethylene glycol (PEG) or other polymeric moiety may be incorporated into a DNLTM construct, as described in further detail below.
  • an antibody or fragment containing a binding site for an antigen associated with a diseased tissue such as a tumor-associated antigen (TAA)
  • TAA tumor-associated antigen
  • a second antibody or fragment that binds a hapten on a targetable construct to which a therapeutic and/or diagnostic agent is attached.
  • TAA tumor-associated antigen
  • the DNLTM-based bispecific antibody is administered to a subject, circulating antibody is allowed to clear from the blood and localize to target tissue, and the conjugated targetable construct is added and binds to the localized antibody for diagnosis or therapy.
  • independent transgenic cell lines may be developed for each Fab or IgG fusion protein. Once produced, the modules can be purified if desired or maintained in the cell culture supernatant fluid. Following production, any DDD-fusion protein module can be combined with any AD-fusion protein module to generate a bispecific DNLTM construct. For different types of constructs, different AD or DDD sequences may be utilized. Exemplary DDD and AD sequences are provided below.
  • DDD1 SHIQIPPGLTELLQGYTVEVLRQQPPDLVEFAVEYFTRLREARA (SEQ ID NO:65)
  • DDD2 CGHIQIPPGLTELLQGYTVEVLRQQPPDLVEFAVEYFTRLREARA (SEQ ID NO:66)
  • AD1 QIEYLAKQIVDNAIQQA (SEQ ID NO: 67)
  • AD2 CGQIEYLAKQIVDNAIQQAGC (SEQ ID NO:68)
  • DDDl and DDD2 comprise the DDD sequence of the human Rlla form of protein kinase A.
  • the DDD and AD moieties may be based on the DDD sequence of the human RIa form of protein kinase A and a corresponding AKAP sequence, as exemplified in DDD3, DDD3C and AD3 below.
  • the plasmid vector pdHL2 has been used to produce a number of antibodies and antibody-based constructs. See Gillies et al., J Immunol Methods (1989), 125: 191-202; Losman et al, Cancer (Phila) (1997), 80:2660-6.
  • the di-cistronic mammalian expression vector directs the synthesis of the heavy and light chains of IgG.
  • the vector sequences are mostly identical for many different IgG-pdHL2 constructs, with the only differences existing in the variable domain (VH and VL) sequences. Using molecular biology tools known to those skilled in the art, these IgG expression vectors can be converted into Fab-DDD or Fab- AD expression vectors.
  • Fab-DDD expression vectors To generate Fab-DDD expression vectors, the coding sequences for the hinge, CH2 and CH3 domains of the heavy chain are replaced with a sequence encoding the first 4 residues of the hinge, a 14 residue Gly-Ser linker and the first 44 residues of human Rlla (referred to as DDDl).
  • AD1 AKAP-ZS
  • Two shuttle vectors were designed to facilitate the conversion of IgG-pdHL2 vectors to either Fab-DDD 1 or Fab-AD 1 expression vectors, as described below.
  • the CHI domain was amplified by PCR using the pdHL2 plasmid vector as a template.
  • the left PCR primer consisted of the upstream (5') end of the CHI domain and a SacII restriction endonuclease site, which is 5' of the CHI coding sequence.
  • the right primer consisted of the sequence coding for the first 4 residues of the hinge (PKSC (SEQ ID NO:
  • the 410 bp PCR amplimer was cloned into the PGEMT® PCR cloning vector (PROMEGA®, Inc.) and clones were screened for inserts in the T7 (5') orientation.
  • a duplex oligonucleotide designated (G 4 S) 2 DDD 1 ('(G 4 S) 2 ' disclosed as SEQ ID NO:
  • oligonucleotides designated RIIA1-44 top and RIIA1-44 bottom, which overlap by 30 base pairs on their 3' ends, were synthesized and combined to comprise the central 154 base pairs of the 174 bp DDD l sequence.
  • the oligonucleotides were annealed and subjected to a primer extension reaction with Taq polymerase. Following primer extension, the duplex was amplified by PCR. The amplimer was cloned into PGEMT® and screened for inserts in the T7 (5') orientation.
  • a duplex oligonucleotide was synthesized to code for the amino acid sequence of AD1 preceded by 11 residues of the linker peptide with the first two codons comprising a BamHI restriction site. A stop codon and an Eagl restriction site are appended to the 3 'end. The encoded polypeptide sequence is shown below.
  • AKAP-IS Top and AKAP-IS Bottom Two complimentary overlapping oligonucleotides encoding the above peptide sequence, designated AKAP-IS Top and AKAP-IS Bottom, were synthesized and annealed. The duplex was amplified by PCR. The amplimer was cloned into the PGEMT® vector and screened for inserts in the T7 (5') orientation.
  • a 190 bp fragment encoding the DDD1 sequence was excised from PGEMT® with BamHI and Notl restriction enzymes and then ligated into the same sites in CHI -PGEMT® to generate the shuttle vector CH1-DDD1 -PGEMT®.
  • a 110 bp fragment containing the AD1 sequence was excised from PGEMT® with BamHI and Notl and then ligated into the same sites in CHI -PGEMT® to generate the shuttle vector CHI -AD 1 -PGEMT®.
  • CH1-DDD1 or CH1-AD1 can be incorporated into any IgG construct in the pdHL2 vector.
  • the entire heavy chain constant domain is replaced with one of the above constructs by removing the SacII/EagI restriction fragment (CH1-CH3) from pdHL2 and replacing it with the SacII/EagI fragment of CH1-DDD1 or CH1-AD 1, which is excised from the respective pGemT shuttle vector.
  • h679-Fd-ADl-pdHL2 is an expression vector for production of h679 Fab with AD1 coupled to the carboxyl terminal end of the CHI domain of the Fd via a flexible Gly/Ser peptide spacer composed of 14 amino acid residues.
  • a pdHL2 -based vector containing the variable domains of h679 was converted to h679-Fd-ADl-pdHL2 by replacement of the SacII/EagI fragment with the CH1-AD1 fragment, which was excised from the CH1-AD1- SV3 shuttle vector with SacII and Eagl.
  • C-DDDl-Fd-hMN-14-pdHL2 is an expression vector for production of a stable dimer that comprises two copies of a fusion protein C-DDDl-Fab-hMN-14, in which DDD1 is linked to hMN-14 Fab at the carboxyl terminus of CHI via a flexible peptide spacer.
  • the plasmid vector hMN-14(I)-pdHL2 which has been used to produce hMN-14 IgG, was converted to C-DDDl-Fd-hMN-14-pdHL2 by digestion with SacII and Eagl restriction endonucleases to remove the CH1-CH3 domains and insertion of the CH1-DDD1 fragment, which was excised from the CH1-DDD 1-SV3 shuttle vector with SacII and Eagl.
  • AD- and DDD-fusion proteins comprising a Fab fragment of any of such antibodies may be combined, in an approximate ratio of two DDD-fusion proteins per one AD-fusion protein, to generate a trimeric DNLTM construct comprising two Fab fragments of a first antibody and one Fab fragment of a second antibody.
  • C-DDD2-Fd-hMN-14-pdHL2 is an expression vector for production of C-DDD2-Fab- hMN-14, which possesses a dimerization and docking domain sequence of DDD2 appended to the carboxyl terminus of the Fd of hMN-14 via a 14 amino acid residue Gly/Ser peptide linker.
  • the fusion protein secreted is composed of two identical copies of hMN-14 Fab held together by non-covalent interaction of the DDD2 domains.
  • the expression vector was engineered as follows. Two overlapping, complimentary oligonucleotides, which comprise the coding sequence for part of the linker peptide and residues 1-13 of DDD2, were made synthetically. The oligonucleotides were annealed and phosphorylated with T4 PNK, resulting in overhangs on the 5' and 3' ends that are compatible for ligation with DNA digested with the restriction endonucleases BamHI and Pstl, respectively.
  • the duplex DNA was ligated with the shuttle vector CH1-DDD1 -PGEMT®, which was prepared by digestion with BamHI and Pstl, to generate the shuttle vector CH1-DDD2- PGEMT®.
  • a 507 bp fragment was excised from CH 1 -DDD2-PGEMT® with SacII and EagI and ligated with the IgG expression vector hMN-14(I)-pdHL2, which was prepared by digestion with SacII and EagI.
  • the final expression construct was designated C-DDD2-Fd- hMN-14-pdHL2. Similar techniques have been utilized to generated DDD2-fusion proteins of the Fab fragments of a number of different humanized antibodies.
  • h679-Fab-AD2 was designed to pair as B to C-DDD2-Fab-hMN- 14 as A.
  • h679-Fd- AD2-pdHL2 is an expression vector for the production of h679-Fab-AD2, which possesses an anchoring domain sequence of AD2 appended to the carboxyl terminal end of the CH 1 domain via a 14 amino acid residue Gly/Ser peptide linker.
  • AD2 has one cysteine residue preceding and another one following the anchor domain sequence of AD 1.
  • the expression vector was engineered as follows. Two overlapping, complimentary oligonucleotides (AD2 Top and AD2 Bottom), which comprise the coding sequence for AD2 and part of the linker sequence, were made synthetically. The oligonucleotides were annealed and phosphorylated with T4 PNK, resulting in overhangs on the 5' and 3' ends that are compatible for ligation with DNA digested with the restriction endonucleases BamHI and Spel, respectively.
  • duplex DNA was ligated into the shuttle vector CHl-ADl-PGEMT®, which was prepared by digestion with BamHI and Spel, to generate the shuttle vector CH1-AD2- PGEMT®.
  • a 429 base pair fragment containing CHI and AD2 coding sequences was excised from the shuttle vector with SacII and Eagl restriction enzymes and ligated into h679-pdHL2 vector that prepared by digestion with those same enzymes.
  • the final expression vector is h679-Fd-AD2-pdHL2.
  • the IgG and Fab fusion proteins shown in Table 9 were constructed and incorporated into DNLTM constructs.
  • the fusion proteins retained the antigen-binding characteristics of the parent antibodies and the DNLTM constructs exhibited the antigen-binding activities of the incorporated antibodies or antibody fragments.
  • AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the AD and DDD sequences incorporated into the
  • DNLTM construct comprise the amino acid sequences of ADl, AD2, AD3, DDDl, DDD2, DDD3 or DDD3C as discussed above.
  • sequence variants of AD and/or DDD moieties may be utilized in construction of the DNLTM complexes.
  • the Rlla DDD sequence is the basis of DDD 1 and DDD2 disclosed above.
  • the four human PKA DDD sequences are shown below.
  • the DDD sequence represents residues 1-44 of Rlla, 1-44 of RII , 12-61 of RIa and 13-66 of Rip. (Note that the sequence of DDD1 is modified slightly from the human PKA Rlla DDD moiety.)
  • Alto et al. (2003) performed a bioinformatic analysis of the AD sequence of various AKAP proteins to design an RII selective AD sequence called AKAP-IS (SEQ ID NO:67), with a binding constant for DDD of 0.4 nM.
  • the AKAP-IS sequence was designed as a peptide antagonist of AKAP binding to PKA. Residues in the AKAP-IS sequence where substitutions tended to decrease binding to DDD are underlined in SEQ ID NO:67.
  • sequence variants of the AD sequence one would desirably avoid changing any of the underlined residues, while conservative amino acid substitutions might be made for residues that are less critical for DDD binding.
  • the SuperAKAP-IS sequence may be substituted for the AKAP-IS AD moiety sequence to prepare DNLTM constructs.
  • Other alternative sequences that might be substituted for the AKAP-IS AD sequence are shown in SEQ ID NO:79-81. Substitutions relative to the AKAP-IS sequence are underlined. It is anticipated that, as with the AD2 sequence shown in SEQ ID NO:68, the AD moiety may also include the additional N-terminal residues cysteine and glycine and C-terminal residues glycine and cysteine.
  • Figure 2 of Gold et al. disclosed additional DDD-binding sequences from a variety of AKAP proteins, shown below.
  • LIEEAASRIVDAVIEQVK (SEQ ID NO: 84)
  • LAWKIAKMIVSDVMQQ (SEQ ID NO:91)
  • AKAPIS QIEYLAKQIVDNAIQQA (SEQ ID NO: 67)
  • AKAP7 ⁇ -P-pep PEDAELVRLSKRLPENAVLKAVQQY (SEQ ID NO: 101)
  • AKAP7 ⁇ 5-PP-pep PEDAELVRLSKRLPENAPLKAVQQY (SEQ ID NO: 102)
  • AKAP 1 -pep EEGLDRNEEIKRAAFQIISQVISEA (SEQ ID NO: 104)
  • AKAP10-pep NTDEAQEELAWKIAKMIVSDIMQQA (SEQ ID NO: 108)
  • AKAP12-pep NGILELETKSSKLVQNIIQTAVDQF (SEQ ID NO: l 10)
  • Rab32-pep ETSAKDNINIEEAARFLVEKILVNH (SEQ ID NO:l 12) [0304] Residues that were highly conserved among the AD domains of different AKAP proteins are indicated below by underlining with reference to the AKAP IS sequence (SEQ ID NO:67). The residues are the same as observed by Alto et al. (2003), with the addition of the C-terminal alanine residue. (See FIG. 4 of Hundsrucker et al.
  • sequences of peptide antagonists with particularly high affinities for the RII DDD sequence were those of AKAP -IS, AKAP75-wt-pep, AKAP75-L304T-pep and AKAP75-L308D-pep.
  • Carr et al. examined the degree of sequence homology between different AKAP -binding DDD sequences from human and non-human proteins and identified residues in the DDD sequences that appeared to be the most highly conserved among different DDD moieties. These are indicated below by underlining with reference to the human PKA Rlla DDD sequence of SEQ ID NO:65. Residues that were particularly conserved are further indicated by italics. The residues overlap with, but are not identical to those suggested by Kinderman et al. (2006) to be important for binding to AKAP proteins.
  • a trimeric DNLTM construct designated TF2 was obtained by reacting C-DDD2-Fab- hMN-14 with h679-Fab-AD2.
  • a pilot batch of TF2 was generated with >90% yield as follows.
  • Protein L-purified C-DDD2-Fab-hMN-14 200 mg was mixed with h679-Fab-AD2 (60 mg) at a 1.4: 1 molar ratio.
  • the total protein concentration was 1.5 mg/ml in PBS containing 1 mM EDTA.
  • Subsequent steps involved TCEP reduction, HIC chromatography, DMSO oxidation, and IMP 291 affinity chromatography. Before the addition of TCEP, SE- HPLC did not show any evidence of a2b formation.
  • TF2 was purified to near homogeneity by IMP 291 affinity chromatography (not shown).
  • IMP 291 is a synthetic peptide containing the HSG hapten to which the 679 Fab binds (Rossi et al, 2005, Clin Cancer Res 1 l :7122s-29s).
  • SE-HPLC analysis of the IMP 291 unbound fraction demonstrated the removal of a 4 , a2 and free kappa chains from the product (not shown).
  • Non-reducing SDS-PAGE analysis demonstrated that the majority of TF2 exists as a large, covalent structure with a relative mobility near that of IgG (not shown). The additional bands suggest that disulfide formation is incomplete under the experimental conditions (not shown). Reducing SDS-PAGE shows that any additional bands apparent in the non-reducing gel are product-related (not shown), as only bands representing the constituent polypeptides of TF2 were evident (not shown). However, the relative mobilities of each of the four polypeptides were too close to be resolved.
  • MALDI-TOF mass spectrometry revealed a single peak of 156,434 Da, which is within 99.5% of the calculated mass (157,319 Da) of TF2.
  • TF2 The functionality of TF2 was determined by BIACORE® assay.
  • TF2, C-DDD1- hMN-14+h679-ADl (used as a control sample of noncovalent a2b complex), or C-DDD2- hMN-14+h679-AD2 (used as a control sample of unreduced a 2 and b components) were diluted to 1 ⁇ g/ml (total protein) and passed over a sensorchip immobilized with HSG.
  • the response for TF2 was approximately two-fold that of the two control samples, indicating that only the h679-Fab-AD component in the control samples would bind to and remain on the sensorchip.
  • TF10 DNLTM construct comprising two copies of a C-DDD2-Fab-hPAM4 and one copy of C-AD2-Fab-679.
  • the cancer- targeting antibody component in TF10 was derived from hPAM4, a humanized anti-MUC5ac MAb that has been studied in detail as a radiolabeled MAb (e.g., Gold et al, Clin. Cancer Res. 13: 7380-7387, 2007).
  • the hapten-binding component was derived from h679, a humanized anti-histaminyl-succinyl-glycine (HSG) MAb.
  • the TFIO bispecific ([hPAM4]2 x h679) antibody was produced using the method disclosed for production of the (anti CEA) 2 x anti HSG bsAb TF2, as described above.
  • the TFIO construct bears two humanized PAM4 Fabs and one humanized 679 Fab.
  • tissue culture supernatant fluids were combined, resulting in a two-fold molar excess of hPAM4-DDD.
  • the reaction mixture was incubated at room temperature for 24 hours under mild reducing conditions using 1 mM reduced glutathione. Following reduction, the DNLTM reaction was completed by mild oxidation using 2 mM oxidized glutathione.
  • TFIO was isolated by affinity chromatography using IMP 291-affigel resin, which binds with high specificity to the h679 Fab.
  • the following study demonstrates the feasibility of in vivo imaging using the pretargeting technique with bispecific antibodies incorporating hPAM4 and labeled peptides.
  • the TF10 bispecific antibody comprising two copies of a C-DDD2-Fab-hPAM4 and one copy of C-AD2-Fab-679, was prepared as described in the preceding Example. Nude mice bearing 0.2 to 0.3 g human pancreatic cancer xenografts were imaged, using pretargeting with TF10 and i n In-IMP-288 peptide. The results, shown in FIG. 8A and FIG.
  • FIG. 8B demonstrate how clearly delineated tumors can be detected in animal models using a bsMAb pretargeting method, with m In-labeled di-HSG peptide, IMP-288.
  • Tumor uptake averaged 20- 25% ID/g with tumor/blood ratios exceeding 2000: 1, tumor/liver ratios of 170: 1, and tumor/kidney ratios of 18/1.
  • 18 F-labeled proteins or peptides are prepared by a novel technique and used for diagnostic and/or imaging studies, such as PET imaging.
  • the novel technique for 18 F labeling involves preparation of an 18 F-metal complex, preferably an 18 F- aluminum complex, which is chelated to a chelating moiety, such as DOTA, NOTA or NETA or derivatives thereof.
  • Chelating moieties may be attached to proteins, peptides or any other molecule using conjugation techniques well known in the art.
  • the 18 F-A1 complex is formed in solution first and then attached to a chelating moiety that is already conjugated to a protein or peptide.
  • the aluminum may be first attached to the chelating moiety and the 18 F added later.
  • Peptides were synthesized by solid phase peptide synthesis using the Fmoc strategy. Groups were added to the side chains of diamino amino acids by using Fmoc/Aloc protecting groups to allow differential deprotection. The Aloe groups were removed by the method of Dangles et. al. (J. Org. Chem. 1987, 52:4984-4993) except that piperidine was added in a 1 : 1 ratio to the acetic acid used. The unsymmetrical tetra-t-butyl DTPA was made as described in McBride et al. (US Patent Application Pub. No. 2005/0002945, the Examples section of which is incorporated herein by reference).
  • DiBocTACN, NODA-GA(?Bu) 3 and the N02A?Bu were purchased from CheMatech (Dijon, France).
  • the Aloc/Fmoc Lysine and Dap (diaminopropionic acid derivatives (also Dpr)) were obtained from CREOSALUS® (Louisville, KY) or BACHEM® (Torrance, CA).
  • the Sieber Amide resin was obtained from NOVABIOCHEM® (San Diego, CA). The remaining Fmoc amino acids were obtained from CREOSALUS®, BACHEM®, PEPTECH®
  • IMP 272 was synthesized as described (US Patent No. 7,534,431, the Examples section of which is incorporated herein by reference).
  • Acetate buffer solution - Acetic acid 1.509 g was diluted in ⁇ 160 mL water and the pH was adjusted by the addition of 1 M NaOH then diluted to 250 mL to make a 0.1 M solution at pH 4.03.
  • IMP 272 acetate buffer solution - Peptide, 0.001 1 g, 7.28 x 10 "7 mol IMP 272 was dissolved in 364 ⁇ of the 0.1 M pH 4 acetate buffer solution to obtain a 2 mM stock solution of the peptide.
  • the labeled peptide was purified by applying the labeled peptide solution onto a 1 cc (30 mg) WATERS® HLB column (Part # 186001879) and washing with 300 ⁇ , water to remove unbound F-18.
  • the peptide was eluted by washing the column with 2 x 100 ⁇ ⁇ 1 : 1 EtOH/H 2 0.
  • the purified peptide was incubated in water at 25 °C and analyzed by reverse phase HPLC (not shown). The HPLC analysis showed that the 18 F-labeled IMP 272 was not stable in water. After 40 min incubation in water about 17% of the 18 F was released from the peptide, while 83% was retained (not shown).
  • the peptide (16 2 mM IMP 272, 48 ⁇ g) was labeled with 18 F and analyzed for antibody binding by size exclusion HPLC.
  • the size exclusion HPLC showed that the peptide bound hM -14 x 679 but did not bind to the irrelevant bispecific antibody hM -14 x 734 (not shown).
  • the peptide, IMP 448 D-Ala-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH 2 MH + 1009 was made on Sieber Amide resin by adding the following amino acids to the resin in the order shown: Aloc-D-Lys(Fmoc)-OH, Trt-HSG-OH, the Aloe was cleaved, Fmoc-D-Tyr(But)-OH, Aloc-D-Lys(Fmoc)-OH, Trt-HSG-OH, the Aloe was cleaved, Fmoc-D-Ala-OH with final Fmoc cleavage to make the desired peptide.
  • the peptide was then cleaved from the resin and purified by HPLC to produce IMP 448, which was then coupled to ITC-benzyl NOTA.
  • the peptide, IMP 448, 0.0757g (7.5 x 10 "5 mol) was mixed with 0.0509 g (9.09 x 10 "5 mol) ITC benzyl NOTA and dissolved in 1 mL water.
  • Potassium carbonate anhydrous (0.2171 g) was then slowly added to the stirred peptide/NOTA solution.
  • the reaction solution was pH 10.6 after the addition of all the carbonate.
  • the reaction was allowed to stir at room temperature overnight.
  • the reaction was carefully quenched with 1 M HC1 after 14 hr and purified by HPLC to obtain 48 mg of IMP 449.
  • the peptide IMP 449 (0.002 g, 1.37 x 10 "6 mol) was dissolved in 686 ⁇ . (2 mM peptide solution) 0.1 M NaOAc pH 4.02. Three microliters of a 2 mM solution of Al in a pH 4 acetate buffer was mixed with 15 ⁇ ⁇ , 1.3 mCi of 18 F. The solution was then mixed with 20 ⁇ . of the 2 mM IMP 449 solution and heated at 105 °C for 15 min.
  • Reverse Phase HPLC analysis showed 35% (3 ⁇ 4 ⁇ 10 min) of the activity was attached to the peptide and 65% of the activity was eluted at the void volume of the column (3.1 min, not shown) indicating that the majority of activity was not associated with the peptide.
  • the crude labeled mixture (5 yl ) was mixed with pooled human serum and incubated at 37 °C. An aliquot was removed after 15 min and analyzed by HPLC. The HPLC showed 9.8% of the activity was still attached to the peptide (down from 35%). Another aliquot was removed after 1 hr and analyzed by HPLC. The HPLC showed 7.6% of the activity was still attached to the peptide (down from 35%), which was essentially the same as the 15 min trace (data not shown).
  • the crude reaction solution was pulled through the column into a waste vial and the column was washed with 3 x 1 mL fractions of water (18.97 mCi).
  • the HLB column was then placed on a new vial and eluted with 2 x 200 1 : 1 EtOH/H 2 0 to collect the labeled peptide (1.83 mCi).
  • the column retained 0.1 mCi of activity after all of the elutions were complete.
  • An aliquot of the purified 18 F-labeled peptide (20 ⁇ ) was mixed with 200 ⁇ ⁇ of pooled human serum and heated at 37 °C. Aliquots were analyzed by reverse phase HPLC.
  • 18 F-labeled IMP 449 was prepared as follows. The 18 F, 54.7 mCi in ⁇ 0.5 mL was mixed with 3 ⁇ , 2 mM Al in 0.1 M NaOAc pH 4 buffer. After 3 min 10 of 0.05 M IMP 449 in 0.5 M pH 4 NaOAc buffer was added and the reaction was heated in a 96 °C heating block for 15 min. The contents of the reaction were removed with a syringe. The crude labeled peptide was then purified by HPLC on a C 18 column. The flow rate was 3 mL/min. Buffer A was 0.1% TFA in water and Buffer B was 90% acetonitrile in water with 0.1% TFA.
  • the gradient went from 100% A to 75/25 A:B over 15 min. There was about 1 min difference in retention time (3 ⁇ 4) between the labeled peptide, which eluted first and the unlabeled peptide.
  • the HPLC eluent was collected in 0.5 min (mL) fractions.
  • the labeled peptide had a t R between 6 to 9 min depending on the column used.
  • the HPLC purified peptide sample was further processed by diluting the fractions of interest two fold in water and placing the solution in the barrel of a 1 cc WATERS® HLB column. The cartridge was eluted with 3 x 1 mL water to remove acetonitrile and TFA followed by 400 ⁇ ⁇ 1 : 1
  • TACONIC® nude mice bearing the four slow-growing sc CaPanl xenografts were used for in vivo studies. Three of the mice were injected with TF10 (162 ⁇ g) followed with [A1 18 F] IMP 449 18 h later.
  • TF 10 is a humanized bispecific antibody of use for tumor imaging studies, with divalent binding to the PAM-4 defined tumor antigen and monovalent binding to HSG (see, e.g., Gold et al, 2007, J. Clin. Oncol.
  • mice 25(18S):4564).
  • One mouse was injected with peptide alone. All of the mice were necropsied at 1 h post peptide injection. Tissues were counted immediately. Comparison of mean distributions showed substantially higher levels of 18 F-labeled peptide localized in the tumor than in any normal tissues in the presence of tumor-targeting bispecific antibody (data not shown).
  • Tissue uptake was similar in animals given the [A1 18 F] IMP 449 alone or in a pretargeting setting (data not shown).
  • Uptake in the human pancreatic cancer xenograft, CaPanl, at 1 h was increased 5 -fold in the pretargeted animals as compared to the peptide alone (4.6 ⁇ 0.9% ID/g vs. 0.89% ID/g).
  • Exceptional tumor/nontumor ratios were achieved at this time (e.g., tumor/blood and liver ratios were 23.4 ⁇ 2.0 and 23.5 ⁇ 2.8, respectively).
  • Tumor uptake was 3.47 ⁇ 0.66% ID/g at this time point with no accumulation in any normal tissue.
  • i n In-IMP-288 was administered 16 hours after TF10.
  • imaging showed intense uptake within the tumors and no evidence of accretion in any normal tissue. No targeting was observed in animals given only the m In-peptide.
  • Tumor uptake of the TF10- pretargeted 111 In-IMP-288 was 24.3 ⁇ 1.7% ID/g, whereas for i n In-IMP-288 alone it was only 0.12 ⁇ 0.002% ID/g at 16 hours.
  • TF10 pretargeting may provide improved imaging for early detection, diagnosis, and treatment of pancreatic cancer as compared with directly radiolabeled PAM4-IgG.
  • PAM4 for pancreatic cancer is of use for the detection and diagnosis of early disease.
  • 90 Y-PAM4 IgG was found to be effective in treating large human pancreatic cancer xenografts in nude mice (Cardillo et al., Clin Cancer Res 2001, 7:3186-92), and when combined with gemcitabine, further improvements in therapeutic response were observed (Gold et al, Clin Cancer Res 2004, 10:3552-61 ; Gold et al, Int J Cancer 2004, 109:618-26).
  • TF2 and TF10 bispecific DNLTM constructs and the IMP 288 targeting peptide were prepared as described above.
  • Sodium iodide ( 125 I) and indium chloride ( m In) were obtained from PERKIN-ELMER®.
  • TF 10 was routinely labeled with 125 I by the iodogen method, with purification by use of size-exclusion spin columns. Radiolabeling of DOTA- peptide and DOTA-PAM4-IgG with m InCl was done as previously described (Rossi et al, Proc Natl Acad Sci U S A 2006, 103:6841-6; McBride et al, J Nucl Med 2006, 47: 1678-88). Purity of the radiolabeled products was examined by size-exclusion high-performance liquid chromatography with the amount of free, unbound isotope determined by instant TLC.
  • mice -20 g (TACONIC® Farms), bearing s.c. CaPanl human pancreatic cancer xenografts, were injected with 125 I-TF10 (10 ⁇ ; 40 ⁇ g, 2.50 x 10 ⁇ 10 mol).
  • a bispecific mAb/radiolabeled peptide molar ratio of 10: 1 was used.
  • TF10 80 ⁇ g, 5.07 x 10 ⁇ 10 mol
  • a second group was left untreated.
  • u l In-IMP-288 hapten-peptide (30 ⁇ , 5.07 x 10 ⁇ u mol) was administered.
  • mice were necropsied at several time points, with tumor and nontumor tissues removed and counted in a gamma counter to determine the %ID/g. Tumor/nontumor ratios were calculated from these data.
  • groups of mice were given u l In-DOTA-PAM4-IgG (20 ⁇ , 50 ⁇ , 3.13 x 10 ⁇ 10 mol) for the purpose of comparing biodistribution, nuclear imaging, and potential therapeutic activity. Radiation dose estimates were calculated from the time-activity curves with the assumption of no activity at zero time. Student's t test was used to assess significant differences.
  • the immunoreactive fraction of 125 I-TF 10 bound to MUC5ac was 87%, with 9% found as unbound TF 10 and 3% as free iodide (not shown).
  • Ninety percent ofthe m In-IMP-288 bound to TF10 (not shown).
  • An additional 5% ofthe radiolabeled peptide eluted in the free peptide volume. None of the radiolabeled peptide bound to the mucin antigen in the absence of TF10 (not shown).
  • Activity in the stomach most likely reflects the accretion and excretion of radioiodine, suggesting that the radioiodinated TF10 was actively catabolized, presumably in the liver and spleen, thereby explaining its rapid clearance from the blood. Nevertheless, by 16 hours, the concentration of radioiodine within the stomach was below 1% ID/g.
  • Tumor weight 0.12 ⁇ 0.03 0.32 ⁇ 0.09 0.27 ⁇ 0.01 0.35 ⁇ 0.03
  • the 10: 1 ratio was chosen for further study because the absolute difference in tumor uptake of radiolabeled peptide was not substantially different between the 10: 1 (24.3 ⁇ 1.71% ID/g) and 20: 1 (28.6 ⁇ 0.73% ID/g) ratios.
  • FIG. 10 A Images of the animals given TF 10-pretargeted m In-IMP-288 at a bispecific mAb/peptide ratio of 10: 1, or the u l In-IMP-288 peptide alone, are shown in FIG. 10 A, FIG. 10B and FIG. IOC. The majority of these tumors were ⁇ 0.5 cm in diameter, weighing -0.25 g.
  • the images show highly intense uptake in the tumor of the TF 10-pretargeted animals (FIG. 10A).
  • the intensity of the image background for the TF 10-pretargeted animals was increased to match the intensity of the image taken of the animals given the u l In-IMP-288 alone (FIG. 10B).
  • FIG. IOC shows the images of the animals at 24 hours postadministration of m In-PAM4-IgG, illustrating that tumors could be visualized at this early time, but there was still considerable activity within the abdomen.
  • Tumor/nontumor ratios were mostly higher for TF lO-pretargeted m In-labeled hapten-peptide as compared with u l In-hPAM4-IgG, except forthe kidneys, where tumor/kidney ratios with the m In-IMP-288 and u l In-hPAM4-IgG were similar at later times.
  • tumor/kidney ratios for the TFlO-pretargeted m In-IMP-288 were high enough (e.g., -7: 1) at 3 hours to easily discern tumor from normal tissue.
  • FIG. 11 A to FIG. 11D illustrates the potential therapeutic capability of the direct and pretargeted methods to deliver radionuclide ( 90 Y). Although the concentration (%ID/g) of radioisotope within the tumor seems to be much greater when delivered by PAM4-IgG than by pretargeted TF 10 at their respective maximum tolerated dose (0.15 mCi for 90 Y-hPAM4 and 0.9 mCi for TFlO-pretargeted 90 Y-IMP-288) (FIG.
  • the radiation dose to tumor would be similar (10,080 and 9,229 cGy for 90 Y-PAM4-IgG and TFlO-pretargeted 90 Y-IMP- 288, respectively) (FIG. 11C).
  • the advantage for the pretargeting method would be the exceptionally low activity in blood (9 cGy), almost 200-fold less than with the 90 Y-hPAM4 IgG (1,623 cGy) (FIG. 11C). It is also important to note that the radiation dose to liver, as well as other nontumor organs, would be much lower with the TFlO-pretargeted 90 Y-IMP-288 (FIG. 11B, FIG. 11D).
  • the exception would be the kidneys, where the radiation dose would be similar for both protocols at their respective maximum dose (612 and 784 cGy for 90 Y- PAM4-IgG and TF10- 90 Y-IMP-288, respectively) (FIG. 11B, FIG. 11D).
  • the data suggest that for 90 Y-PAM4-IgG, as with most other radiolabeled whole-IgG mAbs, the dose-limiting toxicity would be hematologic; however, for the TF 10 pretargeting protocol, the dose-limiting toxicity would be the kidneys.
  • Imaging with a mAb-targeted approach such as is described herein with mAb-PAM4, may provide for the diagnosis of these small, early cancers.
  • mAb-PAM4 may provide for the diagnosis of these small, early cancers.
  • mAb-PAM4 may provide for the diagnosis of these small, early cancers.
  • mAb-PAM4 may provide for the diagnosis of these small, early cancers.
  • PAM4 although not reactive with normal adult pancreatic tissues nor active pancreatitis, is reactive with the earliest stages of neoplastic progression within the pancreas (pancreatic intraepithelial neoplasia 1 and intraductal papillary mucinous neoplasia) and that the biomarker remains at high levels of expression throughout the progression to invasive pancreatic adenocarcinoma (Gold et al, Clin Cancer Res
  • Time for blood levels of the bispecific constructs to reach less than 1% ID/g was 40 hours postinjection for the chemical construct versus 16 hours for TF10.
  • a more rapid clearance of the pretargeting agent has provided a vast improvement of the tumor/blood ratio, while maintaining high signal strength at the tumor site (%ID/g).
  • the results support the use of the TF10 pretargeting system for cancer therapy.
  • Consideration of the effective radiation dose to tumor and nontumor tissues favors the pretargeting method over directly radiolabeled PAM4-IgG.
  • the dose estimates suggest that the two delivery systems have different dose-limiting toxicities: myelotoxicity for the directly radiolabeled PAM4 versus the kidney for the TF 10 pretargeting system. This is of significance for the future clinical development of radiolabeled PAM4 as a therapeutic agent.
  • Gemcitabine the frontline drug of choice for pancreatic cancer, can provide significant radiosensitization of tumor cells.
  • TF10 pretargeting may provide a better opportunity for control of tumor growth than directly radiolabeled PAM4-IgG.
  • Example 22 Therapy of Pancreatic Cancer Xenografts with Gemcitabine and
  • PT-RAIT Monthly fractionated PT-RAIT (0.25 mCi/dose given at the start of each gemcitabine cycle) added to a standard gemcitabine regimen (6 mg wkly x 3; 1 wk off; repeat 3 times) significantly increased the median time for tumors to reach 3.0 cm 3 over PT-RAIT alone.
  • Other treatment plans examining non-cytotoxic radiosensitizing dose regimens of gemcitabine added to PT-RAIT also showed significant improvements in treatment response over PT-RAIT alone.
  • the results show that PT-RAIT is a promising new approach for treating pancreatic cancer. Current data indicate combining PT-RAIT with gemcitabine will enhance therapeutic responses.
  • TF10 bispecific antibody was prepared as described above.
  • TF10 was given to nude mice bearing the human pancreatic adenocarcinoma cell line, CaPanl. After allowing sufficient time for TF10 to clear from the blood (16h), the radiolabeled divalent HSG-peptide was administered.
  • the small molecular weight HSG-peptide (-1.4 kD) clears within minutes from the blood, entering the extravascular space where it can bind to anti-HSG arm of the pretargeted TF10 bsMAb.
  • >80% of the radiolabeled HSG-peptide is excreted in the urine, leaving the tumor localized peptide and a trace amount in the normal tissues.
  • FIG. 12 illustrates the therapeutic activity derived from a single treatment of established (-0.4 cm 3 ) CaPanl tumors with 0.15 mCi of 90 Y-hPAM4 IgG, or 0.25 or 0.50 mCi of TF lO-pretargeted 90 Y-IMP-288. Similar anti-tumor activity was observed for the 0.5- mCi pretargeted dose vs. 0.15-mCi dose of the directly radiolabeled IgG, but hematological toxicity was severe at this level of the direct conjugate (not shown), while the pretargeted dose was only moderately toxic (not shown). Indeed, the MTD for pretargeting using 90Y- IMP-288 is at least 0.9 mCi in nude mice.
  • FIG. 13 shows that the combination of gemcitabine and PT-RAIT has a synergistic effect on anti-tumor therapy.
  • Human equivalent doses of 1000 mg/m 2 (6 mg) of gemcitabine (GEM) were given intraperitoneally to mice weekly for 3 weeks, then after resting for 1 week, this regimen was repeated 2 twice.
  • PT-RAIT (0.25 mCi of TFlO-pretargeted 90 Y-IMP- 288) was given 1 day after the first GEM dose in each of the 3 cycles of treatment. Gem alone had no significant impact on tumor progression (survival based on time to progress to 3.0 cm 3 ).
  • PT-RAIT alone improved survival compared to untreated animals, but the combined GEM with PT-RAIT regimen increased the median survival by nearly 10 weeks. Because hematological toxicity is NOT dose-limiting for PT-RAIT, but it is one of the limitations for gemcitabine therapy, these studies suggest that PT-RAIT could be added to a standard GEM therapy with the potential for enhanced responses. The significant synergistic effect of gemcitabine plus PT-RAIT was surprising and unexpected.
  • TF 10- pretargeted 90 Y-IMP-288 was given [A] once (0.6mCi on wk 0) or [B] fractionated (0.3 mCi on wks 0 and 1), [C] (0.2 mCi on wks 0, 1 and 2) or [D] (0.2 mCi on wks 0, 1 and 4).
  • Example 24 90 Y-hPAM4 Radioimmunotherapy (RAIT) Plus Radiosensitizing Gemcitabine (GEM) Treatment in Advanced Pancreatic Cancer (PC)
  • RAIT Radioimmunotherapy
  • GEM Radiosensitizing Gemcitabine
  • 90 Y-hPAM4 a humanized antibody highly specific for PC, showed transient activity in patients with advanced disease, and GEM enhanced RAIT in preclinical studies.
  • This study evaluated repeated treatment cycles of 90 Y-hPAM4 plus GEM in patients with untreated, unresectable PC.
  • the 90 Y-dose was escalated by cohort, with patients repeating 4- wk cycles (once weekly 200 mg/m 2 GEM, 90 Y-hPAM4 once-weekly wks 2-4) until progression or unacceptable toxicity.
  • Response assessments used CT, FDG-PET, and CA19.9 serum levels.
  • Example 25 Early Detection of Pancreatic Carcinoma Using Mab-PAM4 and
  • Table 12 shows the results of immunohistochemical analysis with PAM4 MAb in pancreatic adenocarcinoma samples of various stages of differentiation. Overall, there was an 87% detection rate for all pancreatic cancer samples, with 100% detection of well differentiated and almost 90% detection of moderately differentiated pancreatic cancers.
  • Table 13 shows that PAM4 immunohistochemical staining also detected a very high percentage of precursor lesions of pancreatic cancer, including Panln-1A to PanI -3, IPMN (intraductal papillary mucinous neoplasms) and MCN (mucinous cystic neoplasms). Overall, PAM4 staining detected 89% of all pancreatic precursor lesions. These results demonstrate that PAM4 antibody -based immunodetection is capable of detecting almost 90% of pancreatic cancers and precursor lesions by in vitro analysis. PAM4 expression was observed in the earliest phases of PanlN development. Intense staining was observed in IPMN and MCN samples (not shown).
  • the PAM4 epitope was present at high concentrations (intense diffuse stain) in the great majority of pancreatic adenocarcinomas.
  • PAM4 showed diffuse, intense reactivity with the earliest stages of pancreatic carcinoma precursor lesions, including PanIN-1, IPMN and MCN, yet was non-reactive with normal pancreatic tissue.
  • ROC curve (not shown) was constructed with the data from Table 14. Examining a total of 283 patients, including 53 with pancreatic carcinoma, and comparing the presence of circulating MUC5ac in patients with pancreatic cancer to all other samples, the ROC curve provided an AUC of 0.88 ⁇ 0.03 (95% ci, 0.84-0.92) with a P value ⁇ 0.0001, a highly significant difference for discrimination of pancreatic carcinoma from non-pancreatic carcinoma samples. Comparing pancreatic CA with other tumors and normal tissue, the PAM4 based serum assay showed a sensitivity of 77% and a specificity of 95%.
  • the mucin biomarker can also serve as a target for in vivo targeting of imaging and therapeutic agents.
  • ImmunoPET imaging for detection of "early" pancreatic carcinoma is of use for the early diagnosis of pancreatic cancer, when it can be more effectively treated.
  • Use of radioimmunotherapy with a humanized PAM4 antibody construct, preferably in combination with a radiosensitizing agent, is of use for the treatment of pancreatic cancer.
  • the presence of MUC5ac and/or to diagnose the presence of pancreatic cancer in a subject by in vitro analysis of samples that can be obtained by non-invasive techniques, such as blood, plasma or serum samples. Such ex vivo analysis may be preferred, for example, in screening procedures where there is no a priori reason to believe that an individual has a pancreatic tumor in a specific location.
  • the objective of the present study was to develop a reliable, accurate, serum-based assay for detection of pancreatic cancer at the earliest stages of the disease.
  • a human pancreatic mucin preparation was isolated from CaPanl, a human pancreatic cancer grown as xenografts in athymic nude mice. Briefly, 1 g of tissue was homogenized in 10 mL of 0.1 M ammonium bicarbonate containing 0.5 M sodium chloride. The sample was then centrifuged to obtain a supernatant that was fractionated on a
  • IgG fraction was purified and assessed for purity by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and molecular-sieve high- performance liquid chromatography.
  • Murine MA5 antibody reactive with the MUC1 protein core was obtained from Immunomedics, Inc. (Morris Plains, NJ).
  • a non-binding isotype- matched control antibody, Ag8 was purified from the P3X63-Ag8 murine myeloma.
  • the top aqueous layer was removed to a clean tube and the sample diluted 1 :2 in 2.0% (w/v) casein-sodium salt in 0.1 M sodium phosphate buffer, pH 7.2, containing 0.15 M sodium chloride (PBS) for immunoassay.
  • PBS sodium chloride
  • Enzyme immunoassay The immunoassay was performed in a 96-well polyvinyl plate that had been coated with 100 ⁇ ⁇ of humanized-PAM4 IgG at 20 ⁇ g/mL in PBS with incubation at 4°C overnight. The wells were then blocked by addition of 200 ⁇ ⁇ of a 2.0% (w/v) solution of casein in PBS and incubated for 1.5 h at 37°C. The blocking solution was removed from the wells and the plate washed 5-times with 250 ⁇ ⁇ of PBS containing 0.1% (v/v) Tween-20. The standards, or unknown specimens, 100 ⁇ ⁇ in triplicate, were added to the appropriate wells and incubated at 37°C for 1.5 h. The plate was then washed 5-times with PBS-Tween-20 as above.
  • polyclonal, rabbit anti-mucin antibody diluted to 5 ⁇ g/mL in 1.0% (w/v) casein in PBS containing 50 ⁇ g/mL non-specific, human IgG, was added to each well and incubated for 1 h at 37°C.
  • the polyclonal antibody was then washed from the wells as above, and peroxidase-labeled donkey anti-rabbit IgG (Jackson ImmunoResearch Laboratories, West Grove, PA), at a 1 :2000 dilution in 1.0% (w/v) casein in PBS, also containing 50 ⁇ g/mL human IgG, was added to the wells and incubated at 37°C for 1 h.
  • the two healthy controls gave average results of 0.27 ⁇ 0.06 and 0.30 ⁇ 0.27 units/mL, each of which was close to the minimum absolute error for the EIA with consequent high CV of 21.65% and 88.19%, respectively.
  • the other patient serum gave an average of 19.45 ⁇ 2.51 units/mL with a CV of 12.9%.
  • Specimens evaluated for PAM4-reactive antigen included 68 patients with confirmed pancreatic adenocarcinoma divided by stage: 21 from stage-1 ; 14 from stage-2; and 33 from stages-3 and -4 (advanced).
  • 19 sera collected from healthy adult volunteers and 29 patients diagnosed with chronic pancreatitis were included as control groups.
  • the maximum concentration shown in the dot-plot (FIG. 19) was 80 units/mL, because there were insufficient volumes of sera to perform additional dilution studies.
  • stage-1 pancreatic adenocarcinoma As shown in Table 15, sensitivity for detection of early, stage-1 pancreatic adenocarcinoma was relatively high, with 13 of 21 (62%) specimens above the cutoff value. As expected, this detection rate was lower than that observed for the stage-2 (86%) and advanced stage-3 and -4 (91%) patient groups. A statistically significant trend (P ⁇ 0.01) was noted for detection rate vs. stage of disease. We considered that this was most likely due to tumor size or burden.
  • the average tumor sizes for stage-1, stage-2, and stage-3/4 groups were 2.14 ⁇ 1.02 cm 3 , 3.36 ⁇ 1.18 cm 3 , and 3.45 ⁇ 1.06 cm 3 , respectively.
  • the average tumor size for stage-1 A was 1.41 ⁇ 0.58 cm 3 (range: 0.4 cm 3 - 2.0 cm 3 ) and for stage- IB was 3.15 ⁇ 0.44 cm 3 (range: 2.5 cm 3 - 4 cm 3 ); P ⁇ 0.001 for comparison of the two groups.
  • pancreatitis serum was 1.28 units/mL, comparable to the healthy volunteer group (1.18 units/mL), but considerably lower (3.5-fold) than the stage-1 pancreatic adenocarcinoma group (4.53 units/mL). It should be noted that our earlier results for pancreatitis specimens suggested a considerably lower false-positive rate, only 5%.
  • pancreatitis specimens were stored frozen for less than 5 years, and were not organic phase extracted prior to analysis.
  • Biopsy and/or surgical specimens were available from 14 of the chronic pancreatitis specimens, 6 of which were from patients who were considered positive for circulating MUC5ac. In 3 of these 6 positive cases, precursor lesions were identified within the tissue sections. It was then considered whether the positive serum test was due to pancreatitis or the presence of neoplastic precursor lesions. We performed immunohistochemistry on an additional 30 biopsy specimens from patients diagnosed with pancreatitis.
  • pancreatic adenocarcinoma and one large PanIN-2-3 lesion were identified (in separate specimens) by use of PAM4 staining, while surrounding acinar-ductal metaplasia (ADM) and normal tissues were negative (data not shown).
  • ADM acinar-ductal metaplasia
  • Table 16 presents an interim analysis based upon studies completed to date; overall, the data are remarkably similar to our earlier data.
  • adenocarcinoma was 62% for patients with stage- 1 and 86% for patients with stage-2 disease and serum levels generally increased with advancing stage of disease. A high percentage of patients with stage- 1 and -2 disease are clinically asymptomatic. We conclude that detection of tumor growth at these early stages using a PAM4 serum assay could provide improved prospects for survival.
  • pancreatic cancers have been developed.
  • pancreatic cancers are suspected of having micrometastatic disease at presentation, even if they do not have histologically-apparent regional lymph node involvement.
  • pancreatic adenocarcinoma The accrual of specimens that are well-defined is problematic. Further complicating the issue is that many of these pancreatic cancers occur in the presence of chronic pancreatitis, cholecystitis, and neoplastic precursor lesions, amongst other conditions.
  • pancreatic cancer screening the general population for pancreatic cancer is not considered medically or economically worthwhile, because the disease is simply too infrequent.
  • pancreatic adenocarcinoma there is considerable interest in screening patients predicted to have an increased risk of developing pancreatic adenocarcinoma.
  • screening individuals with strong family histories of pancreatic cancer can identify precursor neoplasms of the pancreas that are amenable to surgical resection (Canto et al, 2006, Clin Gastroenterol Hepatol 4:766-81; Canto, 2005, Clin Gastroenterol Hepatol 3 :S46- 58; Brentnall et al, 1999, Ann Intern Med 131 :247-55).
  • pancreatic cancer patients have a significantly higher risk of developing pancreatic cancer than the general population (Shi et al, 2009, Arch Pathol Lab Med 133 :365-74).
  • a small percentage of patients with familial pancreatic cancer harbor mutations oiPALB2 (partner and localizer of BRCA2), a susceptibility gene for pancreatic cancer (Tischkowitz et al, 2009,

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Hospice & Palliative Care (AREA)
  • Microbiology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des compositions et des méthodes d'utilisation d'anticorps dirigés contre le cancer du pancréas ou de fragments de ceux-ci, tels que les anticorps PAM4 murins, chimériques, humanisés ou humains. Les anticorps de l'invention présentent de nouvelles caractéristiques utiles en matière de diagnostic, telles qu'une liaison avec une grande spécificité au cancer du pancréas et à d'autres cancers, mais pas à des tissus pancréatiques normaux ou bénins, et une liaison à un pourcentage élevé de cancers du pancréas à un stade précoce. De préférence, les anticorps se lient à un épitope situé à l'intérieur des second, troisième et quatrième domaines riches en cystéine de MUC5ac (restes d'acides aminés 1575-2052) et ils sont utilisés pour la détection et le diagnostic du cancer du pancréas à un stade précoce. Dans des modes de réalisation préférés, les anticorps dirigés contre le cancer du pancréas peuvent être utilisés pour un immuno-essai d'échantillons de sérum, ledit immuno-essai permettant de détecter un marqueur du cancer du pancréas à un stade précoce dans le sérum. Mieux encore, le sérum est extrait avec une phase organique telle que le butanol avant l'immuno-essai.
PCT/US2014/032513 2013-04-01 2014-04-01 Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas WO2014165506A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN201480019479.1A CN105074469A (zh) 2013-04-01 2014-04-01 用于胰腺癌早期检测和治疗的抗粘蛋白抗体
CA2899811A CA2899811A1 (fr) 2013-04-01 2014-04-01 Anticorps anti-mucines permettant la detection precoce et le traitement du cancer du pancreas
EP14778990.3A EP2981829A4 (fr) 2013-04-01 2014-04-01 Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201361807176P 2013-04-01 2013-04-01
US61/807,176 2013-04-01
US201361818708P 2013-05-02 2013-05-02
US61/818,708 2013-05-02
US14/036,765 2013-09-25
US14/036,765 US8795662B2 (en) 2002-06-14 2013-09-25 Anti-mucin antibodies for early detection and treatment of pancreatic cancer
US201361896909P 2013-10-29 2013-10-29
US61/896,909 2013-10-29

Publications (1)

Publication Number Publication Date
WO2014165506A1 true WO2014165506A1 (fr) 2014-10-09

Family

ID=51659171

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/032513 WO2014165506A1 (fr) 2013-04-01 2014-04-01 Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas

Country Status (4)

Country Link
EP (1) EP2981829A4 (fr)
CN (1) CN105074469A (fr)
CA (1) CA2899811A1 (fr)
WO (1) WO2014165506A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105738492A (zh) * 2014-12-10 2016-07-06 人福医药集团股份公司 Lc-ms/ms联用测定拉帕替尼中杂质含量的方法
WO2016176332A1 (fr) * 2015-04-27 2016-11-03 The Regents Of The University Of Colorado Promédicaments d'anthracyne et procédés de production et d'utilisation de ces derniers
WO2017007807A1 (fr) * 2015-07-07 2017-01-12 Immunomedics, Inc. Procédés améliorés d'imagerie avec des molécules marquées au ga-68
WO2017091512A1 (fr) * 2015-11-23 2017-06-01 Sangamo Biosciences, Inc. Méthodes et compositions pour modifier l'immunité
WO2017201449A1 (fr) * 2016-05-20 2017-11-23 Genentech, Inc. Conjugués anticorps-protac et procédés d'utilisation
CN107847593A (zh) * 2015-04-20 2018-03-27 美侬米克国际有限公司 治疗抗体及其用途

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016003869A1 (fr) * 2014-06-30 2016-01-07 Immunomedics, Inc. Anticorps réagissant avec un épitope situé dans la région n-terminale de muc5ac comprenant un sous-domaine 2 riche en cystéine (cys2)
CN107362370B (zh) * 2016-05-13 2022-07-26 国家纳米科学中心 一种基于金纳米簇联合NGF siRNA治疗胰腺癌的方法
CN107153908A (zh) * 2017-03-24 2017-09-12 国家计算机网络与信息安全管理中心 移动新闻App影响力评级方法
CN113198018A (zh) * 2021-04-30 2021-08-03 张志刚 靶向白三烯受体在胰腺癌治疗组合物中的应用
CN116804221B (zh) * 2023-08-23 2023-11-03 汲迈生命科技(苏州)有限公司 Lmo7表达水平在预测胰腺癌吉西他滨耐药中的应用
CN117275744B (zh) * 2023-11-22 2024-02-13 北京大学人民医院 一种综合基因突变特征与mIF图像特征的肺癌预后多模态预测模型构建方法

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011133609A2 (fr) * 2010-04-19 2011-10-27 The Translational Genomics Research Institute Méthodes et nécessaires de prédiction de résultat thérapeutique associée à l'utilisation d'inhibiteurs de la btk
WO2012112443A2 (fr) * 2011-02-15 2012-08-23 Immunomedics, Inc. Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas
US20120301400A1 (en) * 2010-10-08 2012-11-29 Williams John C Meditopes and meditope-binding antibodies and uses thereof
US20130005598A1 (en) * 2009-12-31 2013-01-03 The Regents Of The University Of Michigan Methods for Diagnosing The Malignant Potential of Pancreatic Cystic Lesions
US20140227179A1 (en) * 2003-06-16 2014-08-14 Immunomedics, Inc. Anti-mucin antibodies for early detection and treatment of pancreatic cancer

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2301968A3 (fr) * 2002-06-14 2011-06-29 Immunomedics, Inc. Anticorps monoclonal humanisé HPAM4
CN102137681B (zh) * 2008-08-08 2014-12-03 埃姆诺医药有限公司 抗胰腺癌抗体
CA2787753A1 (fr) * 2010-01-22 2011-07-28 Immunomedics, Inc. Detection d'un adenocarcinome pancreatique a un stade precoce
NZ610267A (en) * 2010-10-08 2015-05-29 Hope City A monoclonal antibody framework binding interface for meditopes, meditope delivery systems and methods for their use

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8795662B2 (en) * 2002-06-14 2014-08-05 Immunomedics, Inc. Anti-mucin antibodies for early detection and treatment of pancreatic cancer
US20140227179A1 (en) * 2003-06-16 2014-08-14 Immunomedics, Inc. Anti-mucin antibodies for early detection and treatment of pancreatic cancer
US20130005598A1 (en) * 2009-12-31 2013-01-03 The Regents Of The University Of Michigan Methods for Diagnosing The Malignant Potential of Pancreatic Cystic Lesions
WO2011133609A2 (fr) * 2010-04-19 2011-10-27 The Translational Genomics Research Institute Méthodes et nécessaires de prédiction de résultat thérapeutique associée à l'utilisation d'inhibiteurs de la btk
US20120301400A1 (en) * 2010-10-08 2012-11-29 Williams John C Meditopes and meditope-binding antibodies and uses thereof
WO2012112443A2 (fr) * 2011-02-15 2012-08-23 Immunomedics, Inc. Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2981829A4 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105738492A (zh) * 2014-12-10 2016-07-06 人福医药集团股份公司 Lc-ms/ms联用测定拉帕替尼中杂质含量的方法
CN105738492B (zh) * 2014-12-10 2017-12-19 人福医药集团股份公司 Lc‑ms/ms联用测定拉帕替尼中杂质含量的方法
CN107847593A (zh) * 2015-04-20 2018-03-27 美侬米克国际有限公司 治疗抗体及其用途
US11198738B2 (en) 2015-04-20 2021-12-14 Minomic International Ltd. Therapeutic antibodies and uses thereof
WO2016176332A1 (fr) * 2015-04-27 2016-11-03 The Regents Of The University Of Colorado Promédicaments d'anthracyne et procédés de production et d'utilisation de ces derniers
US10912767B2 (en) 2015-04-27 2021-02-09 The Regents Of The University Of Colorado, A Body Corporate Anthracycline prodrugs and methods of making and using the same
WO2017007807A1 (fr) * 2015-07-07 2017-01-12 Immunomedics, Inc. Procédés améliorés d'imagerie avec des molécules marquées au ga-68
WO2017091512A1 (fr) * 2015-11-23 2017-06-01 Sangamo Biosciences, Inc. Méthodes et compositions pour modifier l'immunité
US10639383B2 (en) 2015-11-23 2020-05-05 Sangamo Therapeutics, Inc. Methods and compositions for engineering immunity
US20170196992A1 (en) * 2015-11-23 2017-07-13 Sangamo Biosciences, Inc. Methods and compositions for engineering immunity
US11793888B2 (en) 2015-11-23 2023-10-24 Sangamo Therapeutics, Inc. Methods and compositions for engineering immunity
JP2019522633A (ja) * 2016-05-20 2019-08-15 ジェネンテック, インコーポレイテッド Protac抗体コンジュゲート及び使用方法
WO2017201449A1 (fr) * 2016-05-20 2017-11-23 Genentech, Inc. Conjugués anticorps-protac et procédés d'utilisation
US11666581B2 (en) 2016-05-20 2023-06-06 Genentech, Inc. PROTAC antibody conjugates and methods of use

Also Published As

Publication number Publication date
CA2899811A1 (fr) 2014-10-09
CN105074469A (zh) 2015-11-18
EP2981829A4 (fr) 2016-10-26
EP2981829A1 (fr) 2016-02-10

Similar Documents

Publication Publication Date Title
US9238084B2 (en) Anti-mucin antibodies for early detection and treatment of pancreatic cancer
US9089618B2 (en) Anti-mucin antibodies for early detection and treatment of pancreatic cancer
US8491896B2 (en) Anti-pancreatic cancer antibodies
US9513293B2 (en) Detection of early-stage pancreatic adenocarcinoma
CA2731438C (fr) Anticorps anti-cancer pancreatique
WO2014165506A1 (fr) Anticorps anti-mucines permettant la détection précoce et le traitement du cancer du pancréas
US9452228B2 (en) Antibodies reactive with an epitope located in the N-terminal region of MUC5AC comprising cysteine-rich subdomain 2 (Cys2)
CA2948013A1 (fr) Anticorps reagissant avec un epitope situe dans la region n-terminale de muc5ac comprenant un sous-domaine 2 riche en cysteine (cys2)
US8974784B2 (en) Anti-pancreatic cancer antibodies
US9599619B2 (en) Anti-pancreatic cancer antibodies
US20170137534A1 (en) Anti-Pancreatic Cancer Antibodies

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480019479.1

Country of ref document: CN

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

Ref document number: 14778990

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2899811

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2014778990

Country of ref document: EP