EP0881910A1 - Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines - Google Patents

Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines

Info

Publication number
EP0881910A1
EP0881910A1 EP96943706A EP96943706A EP0881910A1 EP 0881910 A1 EP0881910 A1 EP 0881910A1 EP 96943706 A EP96943706 A EP 96943706A EP 96943706 A EP96943706 A EP 96943706A EP 0881910 A1 EP0881910 A1 EP 0881910A1
Authority
EP
European Patent Office
Prior art keywords
antibody
vaccine
cea
administering
mammal
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP96943706A
Other languages
German (de)
French (fr)
Other versions
EP0881910A4 (en
Inventor
Hans J. Hansen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunomedics Inc
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 US08/577,106 external-priority patent/US7354587B1/en
Application filed by Immunomedics Inc filed Critical Immunomedics Inc
Priority to EP09002533A priority Critical patent/EP2057999A3/en
Publication of EP0881910A1 publication Critical patent/EP0881910A1/en
Publication of EP0881910A4 publication Critical patent/EP0881910A4/en
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001182Carcinoembryonic antigen [CEA]
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/208IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/217IFN-gamma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • C07K16/3007Carcino-embryonic Antigens
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to methods for inducing humoral and cellular immune responses against malignant cells and infectious agents.
  • this invention is directed to methods for producing an integrated i munologic response against tumor cells or infectious agents using immunoconjugates compri ⁇ ing antibodies and anti-idiotype antibodies that mimic an epitope of an antigen that is associated with a tumor or infectious agent.
  • the present invention also is directed to a method for augmenting such an integrated response using immunoconjugates, antibodies, anti-idiotype antibodies and cytokines.
  • TAA tumor associated antigens
  • CEA carcinoembryonic antigen
  • Ab2 anti-idiotype antibodies
  • Goldenberg Amer. J . Med . 94 : 297 (1993).
  • Ab2 are antibodies directed against the variable regions of conventional antibodies (Abl) . Since Ab2 and antigen can bind with the same regions of the Abl-combining site, certain Ab2 (termed “Ab2 ⁇ ” or "internal-image” antibodies) can mimic the three dimensional structure of the nominal antigen. Jerne et al ., EMBO J . 1 : 243 (1982); Losman et al . , Int . J. Cancer 46 : 310 (1990); Losman et al . , Proc . Nat 'l Acad .
  • antigen mimicry properties of anti-idiotype antibodies have led to the use of Ab2 ⁇ as surrogate antigens (or idiotype vaccines) , when the nominal antigen is not readily available or when the host is tolerant to the nominal antigen.
  • immunization with Ab2fi mimicking certain TAA creates specific immunity to the TAA and protect against subsequent tumor growth. See, for example, Nepom et al . , Proc . Nat 'l Acad . Sci . USA 81 : 2864 (1984); Raychaudhuri et al., J . Immunol . 139 : 271 (1987).
  • anti- idiotype vaccines have been developed against infectious organisms, such as Streptococcus pneumoniae [McNamara et al . , Science 226 : 1325 (1984)], hepatitus B virus [Kennedy et al . , Science 223 : 930 (1984) ], Escherichia coli K13 [Stein et al ., J . Exp . Med . 160 : 1001 (1984)], Schistosomiasis man ⁇ oni [Kresina et al . , J . Clin . Invest 83 : 912 (1989)], and Moloney murine sarcoma virus [Powell et al., J. Immunol . 142 : 1318 (1989)].
  • Cancer patients receiving an anti-TAA of animal origin will usually produce antibodies to the Abl and these anti-immunoglobulin antibodies include Ab2.
  • the anti-idiotype response also may include the generation of T cells (T2) . Fager erg et al . , Cancer Immunol . Immunother. 37 : 264 (1993) .
  • Ab2 may subsequently induce a humoral and cellular anti-anti-idiotypic response, Ab3 and T3, respectively, which may recognize the same epitope as Abl. Id .
  • Another object of this invention i ⁇ to provide methods for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen comprising the administration of a vaccine comprising an antibody component that binds with the HLA-DR-complex and an antigenic peptide that induces a major histocompatibility (MHC) -restricted immune response.
  • a vaccine comprising an antibody component that binds with the HLA-DR-complex and an antigenic peptide that induces a major histocompatibility (MHC) -restricted immune response.
  • MHC major histocompatibility
  • a method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen (TAA) or against a disease caused by an infectious agent comprising: (a) administering a first vaccine intradermally to the mammal, wherein the first vaccine comprises an immunoconjugate that comprises: (i) an antibody component that binds with the HLA-DR-complex, and ( ⁇ ) an antigenic peptide, wherein the antigenic peptide comprises at least one epitope of a TAA or an antigen associated with the infectious agent, and
  • the antibody component of the fir ⁇ t vaccine may be selected from the group consisting of (a) a murine monoclonal antibody; (b) a humanized antibody derived from a murine monoclonal antibody; (c) a human monoclonal antibody; and (d) an antibody fragment derived from (a) , (b) or (c) , wherein the antibody fragment is selected from the group consisting of F(ab') 2 , F(ab) 2 , Fab', Fab, Fv, sFv and minimal recognition unit.
  • the present invention also is directed to a method further comprising administering at least one of interferon-7, interleukin-2, or interleukin-12 prior to and during the administering of the vaccine intravenously to the mammal.
  • the present invention is further directed to a method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen (TAA) , comprising: (a) administering a first vaccine intradermally to the mammal, wherein the first vaccine comprises an immunoconjugate that comprises: (I) an antibody component that binds with the HLA-DR-complex, and
  • the antibody component of the first vaccine is selected from the group consisting of (a) a murine monoclonal antibody; (b) a humanized antibody derived from a murine monoclonal antibody; (c) a human monoclonal antibody; and (d) an antibody fragment derived from (a) , (b) or (c) , in which the antibody fragment is selected from the group consisting of F(ab') 2 , F(ab) 2 , Fab', Fab, Fv, sFv and minimal recognition unit.
  • a suitable antigenic peptide for example, is tetanus toxin P2 peptide.
  • the present invention also is directed to a method further comprising administering at least one of ⁇ nterferon-7, mterleuk ⁇ n-2, or inte ieukm-12 prior to and during the administering of the vaccine intravenously to the mammal.
  • the present invention is further directed to a method comprising administering a second vaccine intravenously to the mammal, wherein the second vaccine comprises an immunoconjugate that comprises:
  • the present invention also is direct to a method further comprising administering at least one cytokine selected from the group consisting of ⁇ nterleukm-2 , mterleukm-12 and interferon-7 prior to and during the administering of the second vaccine intravenously to the mammal.
  • the present invention also is directed to a method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses carcinoembryonic antigen (CEA) , comprising:
  • the second vaccine comprises an anti- idiotype antibody component that mimics an epitope of the CEA, and wherein the anti- idiotype antibody component is conjugated with a soluble immunogenic carrier protein, and
  • the third vaccine comprises an immunoconjugate comprising an antigenic peptide that comprises an epitope of CEA, and an antibody component that binds with the HLA-DR complex.
  • a suitable antigenic peptide of the third vaccine comprises the A3B3 domain of CEA.
  • the antigenic peptide of the third vaccine can comprise a minimal recognition unit of an anti-idiotype antibody that mimics an epitope of CEA.
  • the antibody component of the first vaccine is selected from the group consisting of:
  • the anti-idiotype antibody component is selected from the group consisting of: (a) a polyclonal antibody that binds with the variable region of a Class III anti-CEA antibody;
  • the present invention also is directed to methods further comprising administering at lea ⁇ t one of interferon-7, interleukin-2, or interleukin-12 prior to and during the administration of the second vaccine.
  • a structural gene is a DNA sequence that is transcribed into messenger RNA (mRNA) which is then translated into a sequence of amino acids characteristic of a specific polypeptide.
  • a promoter is a DNA sequence that directs the transcription of a structural gene. Typically, a promoter is located in the 5' region of a gene, proximal to the transeriptional start site of a structural gene. If a promoter is an inducible promoter, then the rate of transcription increases in response to an inducing agent. In contrast, the rate of transcription is not regulated by an inducing agent if the promoter is a constitutive promoter.
  • An isolated DNA molecule is a fragment of DNA that is not integrated in the genomic DNA of an organism.
  • a cloned T cell receptor gene is a DNA fragment that has been separated from the genomic DNA of a mammalian cell.
  • Another example of an isolated DNA molecule is a chemically-synthesized DNA molecule that is not integrated in the genomic DNA of an organism.
  • An enhancer is a DNA regulatory element that can increase the efficiency of transcription, regardless of the distance or orientation of the enhancer relative to the start site of transcription.
  • cDNA Complementary DNA
  • cDNA is a single-stranded DNA molecule that is formed from an mRNA template by the enzyme reverse transcriptase. Typically, a primer complementary to portions of mRNA is employed for the initiation of reverse transcription.
  • cDNA refers to a double- stranded DNA molecule consisting of such a single- stranded DNA molecule and its complementary DNA strand.
  • expression refers to the biosynthesis of a gene product.
  • expression involves transcription of the structural gene into mRNA and the translation of mRNA into one or more polypeptides.
  • a cloning vector is a DNA molecule, such as a plasmid, cosmid, or bacteriophage, that has the capability of replicating autonomously in a host cell.
  • Cloning vectors typically contain one or a small number of restriction endonuclease recognition sites at which foreign DNA sequences can be inserted in a determinable fashion without loss of an essential biological function of the vector, as well as a marker gene that is suitable for use in the identification and selection of cells transformed with the cloning vector.
  • Marker genes typically include genes that provide tetracycline resistance or ampicillin resistance.
  • 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 include ⁇ those prokaryotic or eukaryotic cells that have been genetically engineered to contain the cloned gene(s) in the chromosome or genome of the host cell.
  • a tumor associated antigen is a protein normally not expressed, or expressed at very low levels, by a normal counterpart.
  • tumor associated antigens include ⁇ -fetoprotein and carcinoembryonic antigen (CEA) .
  • an infectious agent denotes both microbe ⁇ and parasites.
  • a "microbe” includes viruses, bacteria, rickettsia, ycoplasma, protozoa, fungi and like microorganisms.
  • a "parasite” denotes infectious, generally microscopic or very small multicellular invertebrates, or ova or juvenile forms thereof, which are susceptible to antibody-induced clearance or lytic or phagocytic destruction, such a ⁇ malarial parasites, spirochetes, and the like.
  • an anti-CEA MAb is a Class III MAb, a ⁇ described by Primu ⁇ et al . , Cancer Research 43 : 686 (1983) and by Primus et al . , U.S. patent No. 4,818,709, which are incorporated by reference.
  • an Abl is an antibody that binds with a tumor associated antigen or an antigen as ⁇ ociated with an infectious agent.
  • An anti-idiotype antibody is an antibody that binds with an Abl. Importantly, an Ab2 binds with the variable region of Abl and thus, an Ab2 mimics an epitope of a tumor associated antigen or an epitope of an infectious agent associated antigen.
  • An antibody fragment is a portion of an antibody such a ⁇ F(ab') 2 , F(ab) 2 , Fab', Fab, and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an anti-CEA Mab (Abl) fragment binds with CEA, while an Ab2 fragment binds with the variable region of the Abl and mimics an epitope of CEA.
  • antibody fragment also includes any synthetic or genetically engineered protein that acts like an antibody by binding to a specific antigen to form a complex.
  • antibody fragments include isolated fragments consisting of the light chain variable region, "Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant ⁇ ingle chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker ("sFv proteins”) , and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
  • Humanized antibodies are recombinant proteins in which murine complementarity determining regions of MAb have been transferred from heavy and light variable chains of the murine immunoglobulin into a human variable domain.
  • antibody component includes both an entire antibody and an antibody fragment.
  • Rodent monoclonal antibodies to specific antigens may be obtained by methods known to those skilled in the art. See , for example, Kohler and Milstein, Nature 256 : 495
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising an antigen, 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 the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
  • CEA-related antigen ⁇ The major members of this family of CEA-related antigen ⁇ are (1) the normal cross-reactive antigen (NCA) , which shares a similar ti ⁇ ue di ⁇ tribution with CEA, and (2) meconium antigen (MA) , which shares almost identical physiochemical properties with CEA.
  • NCA normal cross-reactive antigen
  • MA meconium antigen
  • the first panel of monoclonal antibodie ⁇ (MAb) that defined NCA-cross-reactive, MA-cross-reactive, and CEA- ⁇ pecific epitopes on the CEA molecule were described by Primus et al . , Cancer Research 43 : 686 (1983) .
  • three classes of anti-CEA antibody were identified: 1) Class I antibodies, which react with CEA, NCA and MA;
  • Class II antibodies which react with CEA and MA, but not with NCA
  • Class III antibodies which are ⁇ pecific for CEA and do not bind with NCA or MA.
  • Methods for obtaining Class III anti-CEA MAb ⁇ are disclosed by Primus et al . , Cancer Research 43 : 686 (1983), and Primus et al . , U.S. patent No. 4,818,709.
  • the production of second generation Class III anti-CEA MAbs i ⁇ disclosed by Hansen et al . , Cancer 71 : 3478 (1993) , which is incorporated by reference.
  • MAbs 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-exclu ⁇ ion 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) .
  • an antibody of the present invention is a subhuman primate antibody.
  • an antibody of the present invention is a "humanized" monoclonal antibody. That is, mouse complementarity determining regions are transferred from heavy and light variable chains of the mouse immunoglobulin into a human variable domain, followed by the replacement of some human residues in the framework regions of their murine counterparts.
  • Humanized monoclonal antibodies in accordance with this invention are suitable for use in therapeutic methods. General techniques for cloning murine immunoglobulin variable domains are described, for example, by the publication of Orlandi et al . , Proc . Nat ' l Acad . Sci . USA 86 : 3833 (1989) , which i ⁇ incorporated by reference in its entirety.
  • an antibody of the present invention is a human monoclonal antibody.
  • Such antibodies are obtained from tran ⁇ genic mice that have been "engineered” to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas.
  • Methods for obtaining human antibodies from transgenic mice are de ⁇ cribed 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), which are incorporated by reference.
  • Antibody fragments can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of the DNA coding for the fragment.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • antibody fragment ⁇ can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S 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 3.5S Fab' monovalent fragments.
  • an enzymatic cleavage using pepsin produces two monovalent Fab fragments and an Fc fragment directly.
  • Fv fragments comprise an association of V H and V L chains. This association can be noncovalent, as described in Inbar et al . , Proc . Nat 'l Acad . Sci . USA 69 : 2659 (1972) .
  • the variable chains can be linked by an intermolecular disulfide bond or cross- linked by chemicals such as glutaraldehyde. See, for example, Sandhu, supra .
  • the Fv fragment ⁇ compri ⁇ e V H and V L chains which are connected by a peptide linker.
  • These single-chain antigen binding proteins are prepared by constructing a structural gene comprising DNA sequences encoding the V H and V L domains which are connected by an oligonucleotide. The structural gene is inserted into an expression vector which is sub ⁇ equently introduced into a host cell, such as E . coli . The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains. Methods for producing sFvs are described, for example, by Whitlow et al .
  • Another form of an antibody fragment is a peptide coding for a single complementarity-determining region (CDR) .
  • CDR peptides (“minimal recognition units") can be obtained by constructing genes encoding the CDR of an antibody of interest.
  • Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody- producing cells. See, for example, Larrick et al . , Methods : A Companion to Methods in Enzymology 2 : 106 (1991) ; Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING AND CLINICAL APPLICATION, Ritter et al . (eds.) , pages 166-179 (Cambridge University Pres ⁇ 1995) ; and Ward et al .
  • Polyclonal Ab2 can be prepared by immunizing animals with Abl or fragments, using standard techniques. See, for example, Green et al . , "Production of Polyclonal
  • monoclonal Ab2 can be prepared u ⁇ ing
  • humanized Ab2 or subhuman primate Ab2 can be prepared using the above-described techniques. 5. Production of Bispecific Antibodies
  • Bispecific antibodies can be used to recruit and target T cells to a tumor cell.
  • a bispecific antibody is a hybrid molecule that consists of nonidentical light and heavy chain pairs, providing two distinct antibody specificities.
  • bispecific antibodies have been produced with one binding site recognizing the CD3 signal transducing protein on T cells and a second binding site for a tumor-associated antigen. See, for example, Canevari et al . , Int . J . Cancer 42 : 18 (1988); Lanzaveccia et al . , Eur . J. Immunol . 17 : 105 (1987) ; Van Dijk et al . , Int . J . Cancer 43 : 344 (1989); and Renner et al . , Science 264 : 833 (1994) .
  • Bispecific antibodies can be made by a variety of conventional methods, e . g . , disulfide cleavage and reformation of mixtures of whole antibody or, preferably F(ab') 2 fragments, fusions of more than one hybridoma to form polyo as that produce antibodies having more than one specificity, and by genetic engineering. Bispecific antibodies have been prepared by oxidative cleavage of Fab' fragments resulting from reductive cleavage of different antibodies. See, for example, Winter et al . , Nature 349 : 293 (1991) .
  • 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 antibodies containing a Fab portion specific to each of the original epitopes.
  • General technique ⁇ for the preparation of ⁇ uch antibody composites may be found, for example, in Nisonhoff et al . , Arch Biochem . Biophys . 93 : 470 (1961) , Ham erling et al . , J . Exp . Med .
  • More selective linkage can be achieved by using a heterobifunctional linker such as malei ide- hydroxy ⁇ uccinimide e ⁇ ter. Reaction of the ester with an antibody or fragment will derivatize amine group ⁇ 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' ⁇ Reagent) . Such a linker i ⁇ less likely to crosslink groups in the same antibody and improves the selectivity of the linkage.
  • a heterobifunctional linker such as malei ide- hydroxy ⁇ uccinimide e ⁇ ter. Reaction of the ester with an antibody or fragment will derivatize amine group ⁇ on the antibody or fragment, and the derivative can then be reacted with, e . g . , an antibody Fab fragment having free s
  • a bispecific antibody comprises binding moieties for T cell ⁇ and an antigen that i ⁇ a ⁇ sociated with a tumor cell or infectious agent.
  • a CEA binding moiety can be derived from a Class III Mab and the T cell-binding moiety can be derived from anti-CD3 Mab.
  • Methods for preparing anti- CD3 antibodies are well-known to those of skill in the art. See, for example, Canevari et al . , supra , Van Dijk et al . , supra , Hansen et al .
  • anti-CD3 antibodie ⁇ can be obtained from commercial sources such as Boehringer Mannheim Corp. (Indianapolis, IN; Cat. No. 1273 485) and the American Type Culture Collection (Rockville, MD; ATCC CRL 8001 [OKT-3]) .
  • a bispecific antibody can be prepared by obtaining an F(ab') 2 fragment from an anti-CEA Class III Mab, as described above.
  • the interchain disulfide bridges of the anti-CEA Clas ⁇ III F(ab') 2 fragment are gently reduced with cysteine, 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 (1, 1'- (methylenedi-4, 1- phenylene)bis-malemide) .
  • the anti-CD3 Mab is converted to Fab'-SH and then reacted with the activated anti-CEA Class III Fab'-SH fragment to obtain a bispecific antibody.
  • bispecific antibodies can be produced by fusing two hybridoma cell lines that produce anti-CD3 Mab and anti-CEA Class III Mab.
  • Techniques for producing tetradomas are described, for example, by Milstein et al . , Nature 305 : 537 (1983) and Pohl et al ., Int . J . Cancer 54 : 418 (1993) .
  • bispecific antibodies can be produced by genetic engineering.
  • plasmid ⁇ containing DNA coding for variable domains of an anti-CEA Class III Mab can be introduced into hybridomas that secrete anti- CD3 antibodie ⁇ .
  • the re ⁇ ulting "transfectoma ⁇ ” produce bispecific antibodies that bind CEA and CD3.
  • chimeric genes can be designed that encode both anti-CD3 and anti-CEA binding domains.
  • General techniques for producing bispecific antibodies by genetic engineering are described, for example, by Songsivilai et al., Biochem . Biophys . Res . Commun . 164 : 271 (1989); Traunecker et al . , EMBO J. 10 : 3655 (1991); and Weiner et al . , J . Immunol . 147 : 4035 (1991) .
  • an "immunoconjugate” is a molecule comprising an antibody component and an antigenic peptide.
  • An immunoconjugate retains the immunoreactivity of the antibody component, i.e., the antibody moiety has about the same, or slightly reduced, ability to bind the cognate antigen after conjugation as before conjugation.
  • Suitable antigenic peptides comprise either at least one epitope of a tumor associated antigen or at least one epitope of an antigen associated with an infectious agent.
  • the A3B3 epitope of CEA i ⁇ an example of a preferred tumor-associated, antigenic peptide. Jessup et al . , Int . J . Cancer 55 : 262 (1993) ; Zhou et al . , Cancer Res . 53 : 3817 (1993); and Hefta et al . , Cancer Res . 52 : 5647 (1992) .
  • Peptides containing CEA epitopes can be produced by recombinant DNA methodology. Id .
  • synthetic peptide ⁇ can be produced u ⁇ ing the general technique ⁇ described below.
  • Useful antigenic peptides also include epitopes of antigens from infectious agents, such as E . coli endotoxin core polysaccharide. See, for example, Greenman et al . , J . Am . Med . Assoc . 266 : 1097 (1991) .
  • particularly useful immunoconjugate ⁇ deliver antigenic peptide ⁇ to cell ⁇ for antigen pre ⁇ entation. See, for example, Wy ⁇ -Coray et al . , Cell . Immunol . 139 : 268 (1992) , which describes the use of an antibody-peptide construct to deliver antigenic peptides to T cells.
  • antigenic peptides include the tetanus toxoid peptide P2 with an N-terminal cysteine, CQYIKANSKFIGITEL (C + tt830-844; C-ttp2; SEQ ID NO:l), and tetanus toxoid peptide P30 with a C-terminal cysteine, FNNFTVSFWLRVPKVSASHLEC (tt947-967 + C; SEQ ID N0:2).
  • Additional antigenic peptides can be derived from single complementarity-determining regions (CDR ⁇ ) of an anti-idiotype antibody.
  • CDR ⁇ complementarity-determining regions
  • Such CDR peptides, or “minimal recognition units,” can be obtained, for example, using the polymerase chain reaction to synthe ⁇ ize the variable region from RNA of antibody-producing cells. See, for example, Larrick et al . , Methods : A Companion to Methods in Enzymology 2 : 106 (1991) ; Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodie ⁇ ," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING AND CLINICAL APPLICATION, Ritter et al .
  • Minimal recognition units also can be obtained by synthesizing peptides having amino acid sequences of known antibodie ⁇ . See, for example, Kabat et al . , SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, U.S. Department of Health and Human Service ⁇ (1983) .
  • Antigenic peptides can be attached at the hinge region of a reduced antibody component via disulfide bond formation.
  • the tetanus toxoid peptides described above were constructed with a single cysteine residue that is used to attach the peptide to an antibody component.
  • such peptides can be attached to the antibody component using a heterobifunctional cross-linker, such as W-succinyl 3-(2- pyridyldithio)proprionate (SPDP) . Yu et al . , Int . J. Cancer 56 : 244 (1994) .
  • SPDP W-succinyl 3-(2- pyridyldithio)proprionate
  • an antigenic peptide can be attached to a reduced thiol group in the hinge region of an antibody component.
  • the antigenic peptide can be conjugated via a carbohydrate moiety in the Fc region of the antibody.
  • the carbohydrate group can be used to increase the loading of the same peptide that is bound to a thiol group, or the carbohydrate moiety can be used to bind a different peptide.
  • the Fc region is absent if an antibody fragment is used as the antibody component of the immunoconjugate.
  • a carbohydrate moiety into the light chain variable region of an antibody or antibody fragment. See, for example, Leung et al . , J . Immunol . 154 : 5919 (1995); Hansen et al . , U.S. patent No. 5,443,953 (1995) .
  • the engineered carbohydrate moiety is used to attach the antigenic peptide. 7.
  • the present invention contemplates the therapeutic use of immunoconjugates, Abl, Ab2 generated against Abl, and fragments of either Abl or Ab2.
  • immunoconjugates, antibodies and antibody fragments can be used as vaccines to induce both humoral and cellular immune responses in the recipient mammal.
  • administration of immunoconjugates, Abl and/or bispecific antibodies can be used to amplify the integrated immune response.
  • a mammal is immunized with a vaccine comprising Abl or fragments thereof, to induce the production of Ab2 and T cells (T2 cells) .
  • T2 cells Ab2 and T cells
  • the mammal may be given Abl, or fragments thereof, by intravenous administration to expand the T2 cell mass.
  • An additional advantage of this second administration is that the antibodies or fragments bind with cognate antigen on cancer cell ⁇ or infectiou ⁇ organisms and thus, serve as targets for T2 cells.
  • Methods for detecting the production of T cells that react with specific antibodies are well-known to those of ordinary skill in the art. See, for example, Fagerberg et al . , Cancer Immunol . Immunother . 37 : 264 (1993) , which is incorporated by reference.
  • a mammal is subsequently immunized with a vaccine comprising Ab2, or fragments thereof, to induce the formation of Ab3 and T cells that recognize Ab2 (T3 cells) .
  • T3 cells T cells that recognize Ab2
  • An advantage of this subsequent Ab2 vaccination is that cells expressing a tumor associated antigen or infectious agent antigen are destroyed by T3 cells directed to the antigen, and by T2 cells directed to Ab3 , which also is bound by the antigen.
  • Example 4 illustrate ⁇ a method of treatment compri ⁇ ing the administration of an Abl vaccine, Abl (or fragments) , and an Ab2 vaccine.
  • a MAb conjugated to a cytokine or lymphokine is administered by intravenous injection subsequent to immunization with Abl.
  • This step amplifies the cytotoxic lymphocyte clones that are induced by the intradermal immunization with Abl and accrete in the targeted cells.
  • the MAb portion of the conjugate can be directed to the same antigen as Abl used in the immunization, or to a different antigen. If the MAb is directed to the same epitope or antigenic determinant on the antigen as the Abl used in the immunization, there should be no cross- reactivity between the idiotype of the Abl used in the vaccine and the idiotype of the MAb in the conjugate.
  • MN-14 and NP-4 are both Class III, anti-CEA MAb that react with the same epitope on CEA, but the two have different idiotypes.
  • the cytokine or lymphokine to which the MAb is conjugated is one that drives induction of immune cytotoxic lymphocytes.
  • exemplary MAb-cytokine/lymphokine conjugates include IL-1, IL-2, 11-12, IL-15, CSF and
  • GM-CSF GM-CSF, with IL-2 and IL-15 being particularly preferred.
  • the T2 response may be further amplified by the intravenous administration of Abl antibodies or fragments after Ab2 vaccination. It is possible that the efficacy of an Ab2 vaccine may be decreased by the presence of circulating Abl antibody components, which have been administered intravenously. Therefore, it is advantageous to clear circulating Abl components prior to the administration of an Ab2 vaccine.
  • One method that can be used to achieve Abl clearance is to use Abl antibodies that have been conjugated with biotin. In this way, circulating biotinylated Abl can be cleared prior to Ab2 vaccination by the intravenous administration of avidin. Preferably, clearance with avidin is performed one to two days after the intravenous administration of Abl (or fragments thereof) . This antibody clearance technique is described by Goldenberg, international application publication No. WO 94/04702 (1994) .
  • an antibody or antibody fragment is conjugated with a peptide capable of inducing a strong major histocompatibility complex (MHC)- restricted immune response.
  • MHC major histocompatibility complex
  • An example of a suitable antigenic peptide is the tetanus toxin P2 peptide, described above.
  • Such a peptide can be conjugated, for example, to the IMMU-LL1 (EPB-1) antibody, which binds with the HLA-DR-complex on the plasma membrane of macrophages, monocytes, and B-lymphocytes. Palak- Byczkow ⁇ ka et al . , Cancer Res. 49 : 4568 (1989) .
  • An IMMU- LL1 vaccine fir ⁇ t is injected intradermally to establish primary sensitization and then, the vaccine is administered intravenously to boost the immune response.
  • an immunoconjugate such as an IMMU-LL1-P2 vaccine
  • the mammal can be treated with an immunoconjugate that directs the immune response to tumor cells.
  • an immunoconjugate comprising humanized LL2 and P2 can be used to target CD22-bearing tumor cells.
  • LL2 is described by Goldenberg et al . , J . Clin . Oncol . 9 : 548 (1991) , and by Murthy et al . , Eur . J . Nucl . Med . 19 : 394 (1992).
  • the sensitizing peptide e . g . , P2
  • the sensitizing peptide is cleaved from the antibody component after internalization, bound to class II MHC heterodimers, and transported to the cell surface.
  • Cytotoxic T cells generated with the LL1-P2 vaccine will then recognize the HLA-II-peptide complex on the cellular membrane and destroy the tumor cell.
  • This general approach can be used to treat other tumors that expres ⁇ the HLA-DR complex, or to treat autoimmune disease ⁇ that are caused by cells expressing the HLA-DR complex.
  • Immunoconjugates also can be used to induce or to boost the immune response to a tumor cell or to an infectious agent using a peptide that contains a suitable epitope.
  • a peptide containing the A3B3 domain of CEA can be conjugated to IMMU-LLl antibody (or fragment) and injected subcutaneously to establish primary sensitization against CEA, or injected intravenously to boost the immune response to CEA.
  • immunoconjugates comprising CDR ⁇ of anti- idiotype antibodie ⁇ can be used to induce or to boost the immune response.
  • a peptide containing the amino acid sequence of a CDR is conjugated with an antibody or antibody fragment.
  • the minimal recognition unit of IMMU-14 Ab2 antibody can be conjugated with IMMU-LLl antibody or antibody fragment.
  • the immune response is further amplified by the administration of cytokines.
  • cytokines include the interferons (INF ⁇ ) , interleukin ⁇ (IL ⁇ ) and tumor necrosis factors.
  • INF-7 induces macrophages, as well a ⁇ cell-surface class II histocompatibility antigen ⁇ on lymphoid cell ⁇ and monocyte ⁇ . See, for example, Kleger an et al . , "Lymphokines and Monokines," in BIOTECHNOLOGY AND PHARMACY, Pezzuto et al . (eds.) , pages 53-70 (Chapman & Hall 1993) , and Roitt et al .
  • IL-2 is a T cell growth factor and a stimulator of natural killer cells and tumor-reactive T cells. Id .
  • INF-7 and IL-2 are preferred cytokines for the augmentation of the immune response.
  • IL-12 is another preferred cytokine for enhancing the immune response to the immunoconjugates of the present invention.
  • This cytokine is produced by phagocytic cells in response to bacteria, bacterial products and intracellular parasites. See, for example, Trinchieri, Annu . Rev . Immunol . 13 : 251 (1995) .
  • IL-12 induces cytokine production, primarily INF-7, by natural killer cells and by T cells, and IL-12 acts a ⁇ a growth factor for activated natural killer cells and T cell ⁇ , enhances the cytotoxic activity of natural killer cells, and stimulates cytotoxic T cell generation. Jd.
  • IL-12 has been used to treat Schistosoma mansoni, Mycobacterium avium, Histoplas a capsulatum , as well as sarcoma, lung metastases.
  • Wynn et al . Nature 376 : 594 (1995) ; Castro et al . , J . Immunol . 155 : 2013 (1995) ; Zhou et al . , J . Immunol . 155 : 785 (1995) ; Zitvogel et al . , J . Immunol . 155 : 1393 (1995) .
  • the antibodies and fragments of the present invention can be used as vaccines by conjugating the antibodie ⁇ or fragments to a soluble immunogenic carrier protein.
  • Suitable carrier proteins include keyhole lympet hemocyanin, which is the preferred carrier protein.
  • the antibodies and fragments can be conjugated to the carrier protein using standard methods. See, for example, Hancock et al , "Synthesis of Peptide ⁇ for Use as Immunogens," in METHODS IN MOLECULAR BIOLOGY: IMMUNOCHEMICAL PROTOCOLS, Manson (ed.), pages 23-32 (Humana Press 1992) . Immunoconjugates comprising one of the above-described antigenic peptides do not require the addition of an immunogenic carrier protein.
  • a preferred vaccination composition compri ⁇ es an antibody conjugate or fragment conjugate, and an adjuvant.
  • suitable adjuvants include aluminum hydroxide and lipid.
  • Methods of formulating vaccine compositions are well-known to those of ordinary skill in the art. See, for example, Rola, "Immunizing Agents and Diagnostic Skin Antigens," in REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, Gennaro (ed.) , pages 1389-1404 (Mack Publishing Company 1990) . Additional pharmaceutical methods may be employed to control the duration of action of a vaccine in a therapeutic application. Control release preparations can be prepared through the use of polymers to complex or adsorb the immunoconjugates, antibodies or fragments.
  • 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, antibody or antibody fragment from such a matrix depends upon the molecular weight of the immunoconjugate, antibody or antibody fragment, the amount of immunoconjugate, antibody or antibody fragment within the matrix, and the size of dispersed particles. Saltzman et al . , Biophys . J . 55: 163 (1989); Sherwood et al . , supra .
  • the therapeutic preparations of the present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby immunoconjugates, antibodies or antibody fragments are combined in a mixture with a pharmaceutically acceptable carrier.
  • a composition is said to be a "pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient mammal.
  • Sterile phosphate- buffered saline is one example of a pharmaceutically acceptable carrier.
  • Other suitable carriers are well- known to those in the art. See, for example, Ansel et al.
  • the immunoconjugates, antibodies or fragments may be administered to a mammal intravenously or subcutaneously. Moreover, the administration may be by continuous infusion or by single or multiple boluse ⁇ . Preferably, an antibody vaccine is administered subcutaneously, while an antibody preparation that is not a vaccine i ⁇ administered intravenously. In general, the dosage of administered immunoconjugates, antibodies or fragments for humans will vary depending upon such factors as the patient's age, weight, height, sex, general medical condition and previous medical history.
  • immunoconjugates, antibodies or fragments which is in the range of from about 1 pg/kg to 10 mg/kg (amount of agent/body weight of patient) , although a lower or higher dosage also may be administered as circumstances dictate.
  • immunoconjugates, antibodie ⁇ or fragments are administered to a mammal in a therapeutically effective amount.
  • An antibody preparation is said to be administered in a "therapeutically effective amount” if the amount administered is physiologically significant.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient mammal.
  • an antibody preparation of the present invention is physiologically significant if it ⁇ presence invokes a humoral and/or cellular immune response in the recipient mammal.
  • a cytokine such as INF-7, IL-2 , or IL-12 may be administered before and during the administration of an Abl vaccine or an Ab2 vaccine.
  • cytokines may be administered together before and during the administration of an antibody vaccine.
  • Cytokines are administered to the mammal intravenously, intramuscularly or subcutaneously.
  • recombinant IL-2 may be administered intravenously as a bolus at 6 x io 1 iu/kg or a ⁇ a continuou ⁇ infu ⁇ ion at a dose of 18 x 10 6 IU/m 2 /d.
  • recombinant IL-2 may be administered subcutaneously at a dose of 12 x 10 6 IU. Vogelzang et al . , J. Clin . Oncol . 11 : 1809 (1993) .
  • INF-7 may be administered subcutaneously at a dose of 1.5 x IO 6 U. Lienard et al . , J. Clin . Oncol . 10 : 52 (1992) .
  • Nadeau et al . , J. Pharmacol . Exp . Ther. 274 : 78 (1995) have shown that a single intravenous dose of recombinant IL-12 (42.5 ⁇ g/kilogram) elevated IFN-7 levels in rhesus monkeys.
  • Suitable IL-2 formulations include PROLEUKIN (Chiron Corp./Cetus Oncology Corp.; Emeryville, CA) and TECELEUKIN (Hoffman-La Roche, Inc.; Nutley, NJ) .
  • ACTIMMUNE Genentech, Inc.; South San Francisco, CA is a suitable INF-7 preparation.
  • bispecific antibodies may be administered after the initial Abl treatment .
  • the function of the bispecific antibodies is to bridge lymphocytes with CEA-bearing tumor cells and to trigger the lymphocyte-mediated cytolysis.
  • Bispecific antibodies can be administered according to above-described general guidelines. However, bispecific antibodies, unlike antibody vaccines, are not conjugated with immunogens.
  • the above-described methods can be used to provide prophylaxis against infectious agents.
  • the present invention contemplates the use of methods described herein to provide protection to a mammal before exposure to an infectious agent.
  • MN-14 a Class III, anti-CEA MAb
  • MN-14 a Class III, anti-CEA MAb
  • a 20 gram BALB/c female mouse was immunized subcutaneou ⁇ ly with 7.5 ⁇ g of partially-purified CEA in complete Freund adjuvant.
  • the mouse was boosted subcutaneously with 7.5 ⁇ g of CEA in incomplete Freund adjuvant and then, the mouse was boosted intravenously with 7.5 ⁇ g of CEA in saline on days 6 and 9.
  • the mouse On day 278, the mouse was given 65 ⁇ g of CEA intravenously in saline and 90 ⁇ g of CEA in saline on day 404.
  • the mouse On day 407, the mouse was sacrificed, a cell suspension of the spleen was prepared, the spleen cells were fused with murine myeloma cells, SP2/0-Ag 14 (ATCC CRL 1581) using polyethylene glycol, and the cells were cultured in medium containing 8- azaguanine.
  • Hybridoma supematants were screened for CEA-reactive antibody using an 125 I-CEA radioimmunoas ⁇ ay (Roche; Nutley, NJ) . Positive clones were recloned.
  • MN-14 One clone, designated MN-14 , had properties similar to the Cla ⁇ III anti-CEA- ⁇ pecific MAb, NP-4, being unreactive with normal cros ⁇ -reactive antigen and meconium antigen. However, MN-14, compared with NP-4 , demonstrated significantly superior tumor targeting in a human colon tumor xenograft model and consistently stronger staining of frozen section ⁇ of colon cancer.
  • a modified antibody was prepared in which the complementarity determining regions (CDR) of MN-14 were engrafted to the framework regions of human IgG ! antibody.
  • CDR-grafted (“humanized") MN-14 antibody was designated "hMN-14.”
  • General techniques for producing humanized antibodies are described, for example, by Jones et al . , Nature 321 : 522 (1986), Riechmann et al . , Nature 332 : 323 (1988) , Verhoeyen et al . , Science 239 : 1534 (1988), Carter et al ., Proc . Nat 'l Acad . Sci . USA 89 : 4285 (1992), Sandhu, Crit . Rev . Biotech . 12 : 437 (1992), and Singer et al . , J . Immun . 150 : 2844 (1993) .
  • hMN-14 was conjugated with keyhole lympet hemocyanin. Typically, patients are immunized with subcutaneous injections of the conjugate
  • Rat Ab2 to MN-14 was prepared as described by Losman et al . , Int . J . Cancer 56 : 580 (1994) , which is incorporated by reference. Briefly, female 3-week-old Copenhagen rats were injected intraperitoneally with 200 ⁇ g of MN-14 F(ab') 2 fragments emulsified in Freund's complete adjuvant. Animals were boosted at days 200, 230, and 235 with the same amount of antigen in Freund's incomplete adjuvant. Four days after the last injection, animals were sacrificed, spleen cell su ⁇ pen ⁇ ions were prepared, and the cells were fused with murine non- secreting plasmocytoma SP2/0 using standard techniques. Hybridoma cells were cultured in the presence of rat peritoneal feeder cells (10,000 cells/200 ⁇ l culture well) .
  • WI2 is an IgG u Ab2 which is specific for MN-14 and does not react with other isotype-matched anti-CEA MABs. Immunization of mice or rabbits with WI2 (but not with control rat IgG) induced the production of Abl' anti-CEA antibodies. Thus, WI2 can be used as an idiotype vaccine for patients with CEA-producing tumors.
  • WI2 vaccine is prepared from WI2 as de ⁇ cribed for the preparation of hMN-14 vaccine.
  • a patient with Dukes C colon carcinoma underwent a primary tumor resection for cure and then, was placed on fluorouracil and Levamisole adjuvant therapy.
  • the pre- operative CEA titer was 15.5 ng/ml.
  • Three months after primary surgery, the CEA titer was in the normal range, that is, below 2.5 ng/ml.
  • the patient wa ⁇ found to have a CEA titer of 25 ng/ml and a CAT scan showed a 5 cm tumor in the left lobe of liver and a 2 cm tumor in the right lobe.
  • the CEA titer was 25 ng/ml and the patient was immunized subcutaneou ⁇ ly with 2 mg of hAbl vaccine (day 0) . Immunization was repeated at day 7.
  • the patient wa ⁇ found to have lymphocytes reactive with the Abl (T2 cells) .
  • the patient was given 100 mg of the hAbl intravenously.
  • the CEA titer was 5 ng/ml and a CAT scan showed that the left lobe tumor had decreased to 2 cm in size, while the right lobe tumor had completely regres ⁇ ed.
  • the left lobe tumor had increa ⁇ ed in size, and a large tumor mass was found in the abdomen, as confirmed by needle biopsy.
  • the CEA titer had increased to 50 ng/ml.
  • the CEA titer was found to be less than 2.5 ng/ml, and the left lobe tumor had completely resolved.
  • the mass in the abdomen was reduced in size and a needle biopsy failed to reveal the presence of a tumor, demonstrating only fibrou ⁇ ti ⁇ ue infiltrated with lymphocytes.
  • IMMU-LLl (EPB-1) is a murine monoclonal antibody that binds with the HLA-DR complex on the plasma membrane of macrophages, monocytes, and B-lymphocytes and then, rapidly internalizes.
  • the preparation of IMMU-LLl is described by Pawak-Byczkowska et al . , Cancer Res . 49 : 4568 (1989) .
  • F(ab') 2 fragments are prepared from intact IMMU-LLl by conventional proteolysi ⁇ techniques, and conjugated with the P2 peptide [SEQ ID NO: 1] of tetanus toxin at the hinge region, a ⁇ de ⁇ cribed above.
  • the P2 peptide i ⁇ conjugated via an engineered carbohydrate moiety on the light chain of the antibody fragment ⁇ using the techniques of Leung et al . , J . Immunol 154 : 5919 (1995) .
  • the IMMU-LL1-P2 vaccine is administered ⁇ ubcutaneously to establish primary sen ⁇ itization due to the strong MHC-restricted immune response induced by the
  • the IMMU-LL1-P2 vaccine also can be administered intravenously to boost the immune response.
  • LL2 is a murine monoclonal antibody that binds with
  • CD22 on B-cell lymphomas See, for example, Goldenberg et al . , J . Clin . Oncol . 9 : 548 (1991) ; Murthy et al . ,
  • Humanized LL2 is prepared as described by Leung et al . , Hybridoma 13:469 (1994), and antibody fragments of humanized LL2 are prepared using standard techniques.
  • An LL2-P2 conjugate is prepared as described above and administered intravenously to the sensitized subject to direct the immune response against tumor cells bearing the CD22 antigen.
  • the A3B3 epitope of CEA is produced recombinantly or by peptide synthesis using the known amino acid sequence. Jessup et al . , int . J . Cancer 55 : 262 (1993); Zhou et al . , Cancer Res . 53 : 3817 (1993); and Hefta et al . , Cancer Res . 52 : 5647 (1992) .
  • A3B3 peptides are conjugated to IMMU-LLl antibody or fragment using standard techniques described above.
  • the IMMU-LL1-A3B3 vaccine is administered subcutaneously to induce the immune response against CEA- bearing tumor cells.
  • the vaccine also can be administered intravenously to boost the immune response against such tumor cells.
  • Peptides having the amino acid sequence of minimal recognition units of the Ab2 antibody described in Example 2 are prepared using the techniques described in section 6 above.
  • the peptides are conjugated with IMMU- LL1 antibodies or fragments to produce immunoconjugates that are suitable for inducing (via subcutaneous administration) or boosting (via intravenous administration) the immune response.
  • a patient with an adenocarcinoma of the lung undergoes resection of the primary tumor, and CEA is demonstrated to be present on the cancer cells by immunohistology.
  • the blood CEA increases from 5 ng/ml to 20 ng/ml and a bone scan demonstrates recurrent carcinoma at numberous sites. Standard chemotherapy is given, however, the CEA titer continues to rise, and a repeat bone scan demonstrates tumor progression.
  • the patient then is immunized with hMN14 and hW12 intradermally, as in Example 4.
  • IL-2 is administered during the immunization to divert the immune response to the Tl helper pathway.
  • a conjugate of IL-15 and hNP-4, a Class III anti-CEA- specific MAb (hNP-4-IL-15) is administered by intravenous infusion.
  • hNP-4-IL-15 a Class III anti-CEA- specific MAb
  • a hNP-4-IL-2 conjugate is administered by intravenous infusion.

Abstract

Humoral and cellular immune responses against tumor cells and infectious agents are induced in a mammal using a vaccine comprising immunoconjugates that comprise antibodies and anti-idiotype antibodies that mimic an epitope of an antigen that is associated with a tumor or an infectious agent. These immunoconjugates also comprise a peptide that contains an epitope of a tumor associated antigen or infectious agent antigen, a peptide that contains a minimal recognition unit of an anti-idiotype antibody, or a peptide that induces a strong major histocompatibility complex-restricted immune response. Antibodies and cytokines also may be used to amplify the immune cascade.

Description

USE OF IMMUNOCONJUGATES TO ENHANCE THE EFFICACY OF MULTI-STAGE CASCADE BOOSTING VACCINES
BACKGROUND OF THE INVENTION
1. Field of the Invention
The present invention relates to methods for inducing humoral and cellular immune responses against malignant cells and infectious agents. In particular, this invention is directed to methods for producing an integrated i munologic response against tumor cells or infectious agents using immunoconjugates compriεing antibodies and anti-idiotype antibodies that mimic an epitope of an antigen that is associated with a tumor or infectious agent. The present invention also is directed to a method for augmenting such an integrated response using immunoconjugates, antibodies, anti-idiotype antibodies and cytokines.
2. Background
One of the major goals of immunotherapy is to harness a patient's immune system against tumor cells or infectious organisms. With regard to cancer therapy, the objective is to direct the patient's immune system against tumor cells by targeting antigens that are associated with tumor cells, but not normal counterparts. Although these tumor associated antigens (TAA) have been difficult to identify, certain tumor cells express antigens that are normally not expressed, or expressed at very low levels, in adult life but present during fetal development. One example of such oncofetal TAA is α-fetoprotein, which is expressed by liver cancer cells. Another oncofetal TAA is the carcinoembryonic antigen (CEA) , which is expressed in most adenocarcinomas of entodermally-derived digestive system epithelia, as well as in breast tumor cells and non-small-cell lung cancer cells. Thomas et al . , Biochim . Biophys . Acta 1032 : 177 (1990) .
The administration of anti-idiotype antibodies (Ab2) mimicking TAA represents one of the most promising approaches to cancer immunotherapy. Goldenberg, Amer. J . Med . 94 : 297 (1993). Ab2 are antibodies directed against the variable regions of conventional antibodies (Abl) . Since Ab2 and antigen can bind with the same regions of the Abl-combining site, certain Ab2 (termed "Ab2β" or "internal-image" antibodies) can mimic the three dimensional structure of the nominal antigen. Jerne et al ., EMBO J . 1 : 243 (1982); Losman et al . , Int . J. Cancer 46 : 310 (1990); Losman et al . , Proc . Nat 'l Acad . Sci . USA 88 : 3421 (1991); Losman et al . , Int . J . Cancer 56 : 580 (1994) . Individuals immunized with Ab2B can develop anti-anti-antibodies (Ab3), some of which (Abl') can bind the nominal antigen.
The antigen mimicry properties of anti-idiotype antibodies have led to the use of Ab2β as surrogate antigens (or idiotype vaccines) , when the nominal antigen is not readily available or when the host is tolerant to the nominal antigen. In experimental systems, immunization with Ab2fi mimicking certain TAA creates specific immunity to the TAA and protect against subsequent tumor growth. See, for example, Nepom et al . , Proc . Nat 'l Acad . Sci . USA 81 : 2864 (1984); Raychaudhuri et al., J . Immunol . 139 : 271 (1987). Similarly, anti- idiotype vaccines have been developed against infectious organisms, such as Streptococcus pneumoniae [McNamara et al . , Science 226 : 1325 (1984)], hepatitus B virus [Kennedy et al . , Science 223 : 930 (1984) ], Escherichia coli K13 [Stein et al ., J . Exp . Med . 160 : 1001 (1984)], Schistosomiasis manεoni [Kresina et al . , J . Clin . Invest . 83 : 912 (1989)], and Moloney murine sarcoma virus [Powell et al., J. Immunol . 142 : 1318 (1989)].
Cancer patients receiving an anti-TAA of animal origin will usually produce antibodies to the Abl and these anti-immunoglobulin antibodies include Ab2. Herlyn et al . , J . Immunol . Methods 85 : 27 (1985) ; Traub et al . , Cancer Res . 48 : 4002 (1988) . The anti-idiotype response also may include the generation of T cells (T2) . Fager erg et al . , Cancer Immunol . Immunother. 37 : 264 (1993) . Moreover, Ab2 may subsequently induce a humoral and cellular anti-anti-idiotypic response, Ab3 and T3, respectively, which may recognize the same epitope as Abl. Id .
Thus, an opportunity exists to provide an approach to immunotherapy utilizing both humoral and cellular immune systems. The applicant has developed methods to provoke an integrated response against tumor cells, aε well aε against infectious agents. Furthermore, the applicant has developed methods to amplify the immune cascade.
SUMMARY OF THE INVENTION
Accordingly, it is an object of the present invention to provide a method for inducing humoral and cellular immune responses against tumor cells and infectious agents using vaccines comprising an antibody that binds with the HLA-DR-complex and an antigenic peptide that comprises at least one epitope of a TAA or an antigen associated with an infectious agent. It is a further object of this invention to provide a method to amplify such an integrated response using antibodies and cytokines.
Another object of this invention iε to provide methods for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen comprising the administration of a vaccine comprising an antibody component that binds with the HLA-DR-complex and an antigenic peptide that induces a major histocompatibility (MHC) -restricted immune response. These and other objects are achieved, in accordance with one embodiment of the present invention by the provision of a method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen (TAA) or against a disease caused by an infectious agent, comprising: (a) administering a first vaccine intradermally to the mammal, wherein the first vaccine comprises an immunoconjugate that comprises: (i) an antibody component that binds with the HLA-DR-complex, and (ϋ) an antigenic peptide, wherein the antigenic peptide comprises at least one epitope of a TAA or an antigen associated with the infectious agent, and
(b) administering the vaccine intravenously to the mammal. The antibody component of the firεt vaccine may be selected from the group consisting of (a) a murine monoclonal antibody; (b) a humanized antibody derived from a murine monoclonal antibody; (c) a human monoclonal antibody; and (d) an antibody fragment derived from (a) , (b) or (c) , wherein the antibody fragment is selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
The present invention also is directed to a method further comprising administering at least one of interferon-7, interleukin-2, or interleukin-12 prior to and during the administering of the vaccine intravenously to the mammal.
The present invention is further directed to a method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen (TAA) , comprising: (a) administering a first vaccine intradermally to the mammal, wherein the first vaccine comprises an immunoconjugate that comprises: (I) an antibody component that binds with the HLA-DR-complex, and
(II) an antigenic peptide that i n d u c e s a m a j o r histocompatibility (MHC)- restricted immune response, and (b) administering the vaccine intravenously to the mammal. The antibody component of the first vaccine is selected from the group consisting of (a) a murine monoclonal antibody; (b) a humanized antibody derived from a murine monoclonal antibody; (c) a human monoclonal antibody; and (d) an antibody fragment derived from (a) , (b) or (c) , in which the antibody fragment is selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit. A suitable antigenic peptide, for example, is tetanus toxin P2 peptide.
The present invention also is directed to a method further comprising administering at least one of ιnterferon-7, mterleukιn-2, or inte ieukm-12 prior to and during the administering of the vaccine intravenously to the mammal.
The present invention is further directed to a method comprising administering a second vaccine intravenously to the mammal, wherein the second vaccine comprises an immunoconjugate that comprises:
(i) an antibody component that binds with a TAA, and (ii) an antigenic peptide that induces a MHC-restricted immune response. An example of a suitable antigenic peptide of the second vaccine is tetanus toxin P2 peptide. The present invention also is direct to a method further comprising administering at least one cytokine selected from the group consisting of ιnterleukm-2 , mterleukm-12 and interferon-7 prior to and during the administering of the second vaccine intravenously to the mammal.
The present invention also is directed to a method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses carcinoembryonic antigen (CEA) , comprising:
(a) administering a first vaccine to the mammal, wherein the first vaccine comprises an antibody component that binds with CEA, and wherein the antibody component is conjugated with a soluble immunogenic carrier protein; and
(b) administering a second vaccine to the mammal, wherein the second vaccine comprises an anti- idiotype antibody component that mimics an epitope of the CEA, and wherein the anti- idiotype antibody component is conjugated with a soluble immunogenic carrier protein, and
(c) administering a third vaccine to the mammal, wherein the third vaccine comprises an immunoconjugate comprising an antigenic peptide that comprises an epitope of CEA, and an antibody component that binds with the HLA-DR complex.
A suitable antigenic peptide of the third vaccine comprises the A3B3 domain of CEA. Moreover, the antigenic peptide of the third vaccine can comprise a minimal recognition unit of an anti-idiotype antibody that mimics an epitope of CEA. In these methods, the antibody component of the first vaccine is selected from the group consisting of:
(a) a murine monoclonal Class III anti-CEA antibody;
(b) a humanized antibody derived from a murine monoclonal Class III anti-CEA antibody; (c) a human monoclonal anti-CEA antibody; and
(d) an antibody fragment derived from (a) , (b) or (c) , wherein the antibody fragment is selected from the group consisting of F(ab')2, F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit. Furthermore, the anti-idiotype antibody component is selected from the group consisting of: (a) a polyclonal antibody that binds with the variable region of a Class III anti-CEA antibody;
(b) a murine monoclonal antibody that binds with the variable region of a Class III anti-CEA antibody;
(c) a humanized antibody derived from (b) ;
(d) a human monoclonal antibody that binds with the variable region of a Clasε III anti-CEA antibody; (e) a subhuman primate antibody that binds with the variable region of a Clasε III anti-CEA antibody; and
(f) an antibody fragment derived from (a) , (b) ,
(c) , (d) or (e) , in which the antibody fragment iε selected from the group consisting of
F(ab')2, F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
The present invention also is directed to methods further comprising administering at leaεt one of interferon-7, interleukin-2, or interleukin-12 prior to and during the administration of the second vaccine.
DETAILED DESCRIPTION
1. Definitions
In the description that follows, a number of terms are used extensively. The following definitions are provided to facilitate understanding of the invention.
A structural gene is a DNA sequence that is transcribed into messenger RNA (mRNA) which is then translated into a sequence of amino acids characteristic of a specific polypeptide. A promoter is a DNA sequence that directs the transcription of a structural gene. Typically, a promoter is located in the 5' region of a gene, proximal to the transeriptional start site of a structural gene. If a promoter is an inducible promoter, then the rate of transcription increases in response to an inducing agent. In contrast, the rate of transcription is not regulated by an inducing agent if the promoter is a constitutive promoter. An isolated DNA molecule is a fragment of DNA that is not integrated in the genomic DNA of an organism. For example, a cloned T cell receptor gene is a DNA fragment that has been separated from the genomic DNA of a mammalian cell. Another example of an isolated DNA molecule is a chemically-synthesized DNA molecule that is not integrated in the genomic DNA of an organism.
An enhancer is a DNA regulatory element that can increase the efficiency of transcription, regardless of the distance or orientation of the enhancer relative to the start site of transcription.
Complementary DNA (cDNA) is a single-stranded DNA molecule that is formed from an mRNA template by the enzyme reverse transcriptase. Typically, a primer complementary to portions of mRNA is employed for the initiation of reverse transcription. Those skilled in the art also use the term "cDNA" to refer to a double- stranded DNA molecule consisting of such a single- stranded DNA molecule and its complementary DNA strand.
The term expression refers to the biosynthesis of a gene product. For example, in the case of a εtructural gene, expression involves transcription of the structural gene into mRNA and the translation of mRNA into one or more polypeptides.
A cloning vector is a DNA molecule, such as a plasmid, cosmid, or bacteriophage, that has the capability of replicating autonomously in a host cell.
Cloning vectors typically contain one or a small number of restriction endonuclease recognition sites at which foreign DNA sequences can be inserted in a determinable fashion without loss of an essential biological function of the vector, as well as a marker gene that is suitable for use in the identification and selection of cells transformed with the cloning vector. Marker genes typically include genes that provide tetracycline resistance or ampicillin resistance.
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 includeε those prokaryotic or eukaryotic cells that have been genetically engineered to contain the cloned gene(s) in the chromosome or genome of the host cell.
A tumor associated antigen is a protein normally not expressed, or expressed at very low levels, by a normal counterpart. Examples of tumor associated antigens include α-fetoprotein and carcinoembryonic antigen (CEA) . As used herein, an infectious agent denotes both microbeε and parasites. A "microbe" includes viruses, bacteria, rickettsia, ycoplasma, protozoa, fungi and like microorganisms. A "parasite" denotes infectious, generally microscopic or very small multicellular invertebrates, or ova or juvenile forms thereof, which are susceptible to antibody-induced clearance or lytic or phagocytic destruction, such aε malarial parasites, spirochetes, and the like.
In the present context, an anti-CEA MAb is a Class III MAb, aε described by Primuε et al . , Cancer Research 43 : 686 (1983) and by Primus et al . , U.S. patent No. 4,818,709, which are incorporated by reference. As used herein, an Abl is an antibody that binds with a tumor associated antigen or an antigen asεociated with an infectious agent.
An anti-idiotype antibody (Ab2) , as used herein, is an antibody that binds with an Abl. Importantly, an Ab2 binds with the variable region of Abl and thus, an Ab2 mimics an epitope of a tumor associated antigen or an epitope of an infectious agent associated antigen.
An antibody fragment is a portion of an antibody such aε F(ab')2 , F(ab)2, Fab', Fab, and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an anti-CEA Mab (Abl) fragment binds with CEA, while an Ab2 fragment binds with the variable region of the Abl and mimics an epitope of CEA.
The term "antibody fragment" also includes any synthetic or genetically engineered protein that acts like an antibody by binding to a specific antigen to form a complex. For example, antibody fragments include isolated fragments consisting of the light chain variable region, "Fv" fragments consisting of the variable regions of the heavy and light chains, recombinant εingle chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker ("sFv proteins") , and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
Humanized antibodies are recombinant proteins in which murine complementarity determining regions of MAb have been transferred from heavy and light variable chains of the murine immunoglobulin into a human variable domain.
As used herein, the term antibody component includes both an entire antibody and an antibody fragment. 2. Production of Monoclonal Antibodies, Humanized Antibodies, Primate Antibodies and Human Antibodies
Rodent monoclonal antibodies to 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, pageε 2.5.1-2.6.7 (John Wiley & Sons
1991) [hereinafter "Coligan"]. Briefly, monoclonal antibodies can be obtained by injecting mice with a composition comprising an antigen, 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 the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
A wide variety of monoclonal antibodies against tumor associated antigens or infectious agentε have been developed. See, for example, Goldenberg et al . , international application publication No. WO 91/11465 (1991) , and Goldenberg, international application publication No. WO 94/04702 (1994) , each of which iε incorporated by reference in itε entirety. An example of a suitable Mab is a Class III anti-CEA Mab. Conventional antisera raised against CEA usually contain antibodies that react with a group of substances closely related to CEA. The major members of this family of CEA-related antigenε are (1) the normal cross-reactive antigen (NCA) , which shares a similar tiεεue diεtribution with CEA, and (2) meconium antigen (MA) , which shares almost identical physiochemical properties with CEA. The first panel of monoclonal antibodieε (MAb) that defined NCA-cross-reactive, MA-cross-reactive, and CEA-εpecific epitopes on the CEA molecule were described by Primus et al . , Cancer Research 43 : 686 (1983) . In particular, three classes of anti-CEA antibody were identified: 1) Class I antibodies, which react with CEA, NCA and MA;
2) Class II antibodies, which react with CEA and MA, but not with NCA; and 3) Class III antibodies, which are εpecific for CEA and do not bind with NCA or MA. Methods for obtaining Class III anti-CEA MAbε are disclosed by Primus et al . , Cancer Research 43 : 686 (1983), and Primus et al . , U.S. patent No. 4,818,709. Moreover, the production of second generation Class III anti-CEA MAbs iε disclosed by Hansen et al . , Cancer 71 : 3478 (1993) , which is incorporated by reference.
MAbs 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-excluεion 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) . In another embodiment, an antibody of the present invention is a subhuman primate antibody. General techniques for raising therapeutically useful antibodies in baboons may be found, for example, in Goldenberg et al . , international patent publication No. WO 91/11465 (1991), and in Losman et al . , Int . J . Cancer 46 : 310 (1990) , which iε incorporated by reference.
In yet another embodiment, an antibody of the present invention is a "humanized" monoclonal antibody. That is, mouse complementarity determining regions are transferred from heavy and light variable chains of the mouse immunoglobulin into a human variable domain, followed by the replacement of some human residues in the framework regions of their murine counterparts. Humanized monoclonal antibodies in accordance with this invention are suitable for use in therapeutic methods. General techniques for cloning murine immunoglobulin variable domains are described, for example, by the publication of Orlandi et al . , Proc . Nat ' l Acad . Sci . USA 86 : 3833 (1989) , which iε incorporated by reference in its entirety. Techniques for producing humanized MAbs are described, for example, by Jones et al . , Nature 321 : 522 (1986), Riechmann et al . , Nature 332 : 323 (1988), Verhoeyen et al ., Science 239 : 1534 (1988), Carter et al . , Proc . Nat 'l Acad . Sci . USA 89 : 4285 (1992), Sandhu, Crit . Rev . Biotech . 12 : 437 (1992), and Singer et al . , J . Immun . 150 : 2844 (1993), each of which is hereby incorporated by reference. In another embodiment, an antibody of the present invention is a human monoclonal antibody. Such antibodies are obtained from tranεgenic mice that have been "engineered" to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. The transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas. Methods for obtaining human antibodies from transgenic mice are deεcribed 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), which are incorporated by reference.
3. Production of Antibody Fragments
The present invention contemplates the use of fragments of Abl or Ab2. Antibody fragments can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of the DNA coding for the fragment.
Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods. For example, antibody fragmentε can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S 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 3.5S Fab' monovalent fragments. Alternatively, an enzymatic cleavage using pepsin produces two monovalent Fab fragments and an Fc fragment directly. These methods are described, for example, by Goldenberg, U.S. patent Nos. 4,036,945 and 4,331,647 and references contained therein, which patents are incorporated herein in their entireties by reference. Also, see Nisonoff et al . , Arch Biochem . Biophys . 89 : 230 (1960); Porter, Biochem . J . 73 : 119 (1959), Edelman et al . , in METHODS IN ENZYMOLOGY VOL. 1, page 422 (Academic Press 1967), and Coligan at pages 2.8.1-2.8.10 and 2.10.-2.10.4.
Other methods of cleaving antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody. For example, Fv fragments comprise an association of VH and VL chains. This association can be noncovalent, as described in Inbar et al . , Proc . Nat 'l Acad . Sci . USA 69 : 2659 (1972) . Alternatively, the variable chains can be linked by an intermolecular disulfide bond or cross- linked by chemicals such as glutaraldehyde. See, for example, Sandhu, supra .
Preferably, the Fv fragmentε compriεe VH and VL chains which are connected by a peptide linker. These single-chain antigen binding proteins (sFv) are prepared by constructing a structural gene comprising DNA sequences encoding the VH and VL domains which are connected by an oligonucleotide. The structural gene is inserted into an expression vector which is subεequently introduced into a host cell, such as E . coli . The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains. Methods for producing sFvs are described, for example, by Whitlow et al . , Methods : A Companion to Methods in Enzymology 2 : 97 (1991) . Also see Bird et al . , Science 242:423-426 (1988), Ladner et al . , U.S. Patent No. 4,946,778, Pack et al . , Bio /Technology 11:1271-1277 (1993) , and Sandhu, supra . Another form of an antibody fragment is a peptide coding for a single complementarity-determining region (CDR) . CDR peptides ("minimal recognition units") can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody- producing cells. See, for example, Larrick et al . , Methods : A Companion to Methods in Enzymology 2 : 106 (1991) ; Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING AND CLINICAL APPLICATION, Ritter et al . (eds.) , pages 166-179 (Cambridge University Presε 1995) ; and Ward et al . , "Genetic Manipulation and Expression of Antibodies," in MONOCLONAL ANTIBODIES: PRINCIPLES AND APPLICATIONS, Birch et al . , (edε.) , pageε 137-185 (Wiley-Liss, Inc. 1995) .
4. Production of Anti-idiotype Antibodies (Ab2)
Polyclonal Ab2 can be prepared by immunizing animals with Abl or fragments, using standard techniques. See, for example, Green et al . , "Production of Polyclonal
Antisera," in METHODS IN MOLECULAR BIOLOGY:
IMMUNOCHEMICAL PROTOCOLS, Manson (ed.), pages 1-12
(Humana Press 1992) . Also, see Coligan at pages 2.4.1-
2.4.7. Alternatively, monoclonal Ab2 can be prepared uεing
Abl or fragmentε aε i munogenε with the techniqueε, deεcribed above. The preparation of a rat monoclonal Ab2 is illustrated in Example 3.
As another alternative, humanized Ab2 or subhuman primate Ab2 can be prepared using the above-described techniques. 5. Production of Bispecific Antibodies
Bispecific antibodies can be used to recruit and target T cells to a tumor cell. A bispecific antibody is a hybrid molecule that consists of nonidentical light and heavy chain pairs, providing two distinct antibody specificities. For example, bispecific antibodies have been produced with one binding site recognizing the CD3 signal transducing protein on T cells and a second binding site for a tumor-associated antigen. See, for example, Canevari et al . , Int . J . Cancer 42 : 18 (1988); Lanzaveccia et al . , Eur . J. Immunol . 17 : 105 (1987) ; Van Dijk et al . , Int . J . Cancer 43 : 344 (1989); and Renner et al . , Science 264 : 833 (1994) .
Bispecific antibodies can be made by a variety of conventional methods, e . g . , disulfide cleavage and reformation of mixtures of whole antibody or, preferably F(ab')2 fragments, fusions of more than one hybridoma to form polyo as that produce antibodies having more than one specificity, and by genetic engineering. Bispecific antibodies have been prepared by oxidative cleavage of Fab' fragments resulting from reductive cleavage of different antibodies. See, for example, Winter et al . , Nature 349 : 293 (1991) . 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 antibodies containing a Fab portion specific to each of the original epitopes. General techniqueε for the preparation of εuch antibody composites may be found, for example, in Nisonhoff et al . , Arch Biochem . Biophys . 93 : 470 (1961) , Ham erling et al . , J . Exp . Med . 128 : 1461 (1968) , and U.S. patent No. 4,331,647. More selective linkage can be achieved by using a heterobifunctional linker such as malei ide- hydroxyεuccinimide eεter. Reaction of the ester with an antibody or fragment will derivatize amine groupε 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'ε Reagent) . Such a linker iε less likely to crosslink groups in the same antibody and improves the selectivity of the linkage.
It iε advantageouε to link the antibodieε or fragmentε at sites remote from the antigen binding sites. This can be accomplished by, e . g . , linkage to cleaved interchain sulfydryl groupε, as noted above. Another method involves reacting an antibody having an oxidized carbohydrate portion with another antibody which has at lease one free amine function. This results in an initial Schiff base (imine) linkage, which is preferably stabilized by reduction to a secondary amine, e . g . , by borohydride reduction, to form the final composite. Such site-εpecific linkageε are diεcloεed, for small molecules, in U.S. patent No. 4,671,958, and for larger addends in U.S. patent No. 4,699,784.
In the present context, a bispecific antibody comprises binding moieties for T cellε and an antigen that iε aεsociated with a tumor cell or infectious agent. For example, a CEA binding moiety can be derived from a Class III Mab and the T cell-binding moiety can be derived from anti-CD3 Mab. Methods for preparing anti- CD3 antibodies are well-known to those of skill in the art. See, for example, Canevari et al . , supra , Van Dijk et al . , supra , Hansen et al . , "Human T Lymphocyte Cell Surface Molecules Defined by the Workshop Monoclonal Antibodies (T Cell Protocol)," in LEUKOCYTE TYPING: HUMAN LEUKOCYTE MARKERS DETECTED BY MONOCLONAL ANTIBODIES, Bernard et al . , (eds.) pages 195-212 (Springer-Verlag 1984); and U.S. patent No. 4,361,549. Alternatively, anti-CD3 antibodieε can be obtained from commercial sources such as Boehringer Mannheim Corp. (Indianapolis, IN; Cat. No. 1273 485) and the American Type Culture Collection (Rockville, MD; ATCC CRL 8001 [OKT-3]) . For example, a bispecific antibody can be prepared by obtaining an F(ab')2 fragment from an anti-CEA Class III Mab, as described above. The interchain disulfide bridges of the anti-CEA Clasε III F(ab')2 fragment are gently reduced with cysteine, 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 (1, 1'- (methylenedi-4, 1- phenylene)bis-malemide) . The anti-CD3 Mab is converted to Fab'-SH and then reacted with the activated anti-CEA Class III Fab'-SH fragment to obtain a bispecific antibody.
Alternatively, such bispecific antibodies can be produced by fusing two hybridoma cell lines that produce anti-CD3 Mab and anti-CEA Class III Mab. Techniques for producing tetradomas are described, for example, by Milstein et al . , Nature 305 : 537 (1983) and Pohl et al ., Int . J . Cancer 54 : 418 (1993) .
Finally, bispecific antibodies can be produced by genetic engineering. For example, plasmidε containing DNA coding for variable domains of an anti-CEA Class III Mab can be introduced into hybridomas that secrete anti- CD3 antibodieε. The reεulting "transfectomaε" produce bispecific antibodies that bind CEA and CD3. Alternatively, chimeric genes can be designed that encode both anti-CD3 and anti-CEA binding domains. General techniques for producing bispecific antibodies by genetic engineering are described, for example, by Songsivilai et al., Biochem . Biophys . Res . Commun . 164 : 271 (1989); Traunecker et al . , EMBO J. 10 : 3655 (1991); and Weiner et al . , J . Immunol . 147 : 4035 (1991) .
6. Preparation of Immunoconjugates
The present invention contemplates the use of immunoconjugates to augment the immune response. In the present context, an "immunoconjugate" is a molecule comprising an antibody component and an antigenic peptide. An immunoconjugate retains the immunoreactivity of the antibody component, i.e., the antibody moiety has about the same, or slightly reduced, ability to bind the cognate antigen after conjugation as before conjugation. Suitable antigenic peptides comprise either at least one epitope of a tumor associated antigen or at least one epitope of an antigen associated with an infectious agent. A general overview of useful tumor associated antigens and of infectious agent antigens is provided above. The A3B3 epitope of CEA iε an example of a preferred tumor-associated, antigenic peptide. Jessup et al . , Int . J . Cancer 55 : 262 (1993) ; Zhou et al . , Cancer Res . 53 : 3817 (1993); and Hefta et al . , Cancer Res . 52 : 5647 (1992) . Peptides containing CEA epitopes can be produced by recombinant DNA methodology. Id . Alternatively, synthetic peptideε can be produced uεing the general techniqueε described below.
Useful antigenic peptides also include epitopes of antigens from infectious agents, such as E . coli endotoxin core polysaccharide. See, for example, Greenman et al . , J . Am . Med . Assoc . 266 : 1097 (1991) .
In the present context, particularly useful immunoconjugateε deliver antigenic peptideε to cellε for antigen preεentation. See, for example, Wyεε-Coray et al . , Cell . Immunol . 139 : 268 (1992) , which describes the use of an antibody-peptide construct to deliver antigenic peptides to T cells. Examples of such antigenic peptides include the tetanus toxoid peptide P2 with an N-terminal cysteine, CQYIKANSKFIGITEL (C + tt830-844; C-ttp2; SEQ ID NO:l), and tetanus toxoid peptide P30 with a C-terminal cysteine, FNNFTVSFWLRVPKVSASHLEC (tt947-967 + C; SEQ ID N0:2).
Additional antigenic peptides can be derived from single complementarity-determining regions (CDRε) of an anti-idiotype antibody. Such CDR peptides, or "minimal recognition units," can be obtained, for example, using the polymerase chain reaction to syntheεize the variable region from RNA of antibody-producing cells. See, for example, Larrick et al . , Methods : A Companion to Methods in Enzymology 2 : 106 (1991) ; Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodieε," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING AND CLINICAL APPLICATION, Ritter et al . (eds.) , pages 166-179 (Cambridge University Press 1995); and Ward et al . , "Genetic Manipulation and Expression of Antibodies," in MONOCLONAL ANTIBODIES: PRINCIPLES AND APPLICATIONS, Birch et al . , (eds.), pages 137-185 (Wiley-Lisε, Inc. 1995) . Minimal recognition units also can be obtained by synthesizing peptides having amino acid sequences of known antibodieε. See, for example, Kabat et al . , SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, U.S. Department of Health and Human Serviceε (1983) . General methods for peptide synthesis can be found, for example, in Bodanszky et al . , THE PRACTICE OF PEPTIDE SYNTHESIS (Springer-Verlag 1984); Bodanszky, PRINCIPLES OF PEPTIDE SYNTHESIS (Springer-Verlag 1984) ; Hancock et al . , "Syntheεis of Peptides for Use as Immunogens," in METHODS IN MOLECULAR BIOLOGY, VOL. 10: IMMUNOCHEMICAL PROTOCOLS,
Manson (ed.) pages 23-32 (The Humana Press, Inc. 1992).
Antigenic peptides can be attached at the hinge region of a reduced antibody component via disulfide bond formation. For example, the tetanus toxoid peptides described above were constructed with a single cysteine residue that is used to attach the peptide to an antibody component. As an alternative, such peptides can be attached to the antibody component using a heterobifunctional cross-linker, such as W-succinyl 3-(2- pyridyldithio)proprionate (SPDP) . Yu et al . , Int . J. Cancer 56 : 244 (1994) . General techniqueε for εuch conjugation are well-known in the art. See, for example, Wong, CHEMISTRY OF PROTEIN CONJUGATION AND CROSS-LINKING (CRC Preεs 1991); Upeslaciε et al . , "Modification of Antibodies by Chemical Methods," in MONOCLONAL ANTIBODIES: PRINCIPLES AND APPLICATIONS, Birch et al . (eds.), pages 187-230 (Wiley-Lisε, Inc. 1995); Price, "Production and Characterization of Synthetic Peptide- Derived Antibodieε, " in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING AND CLINICAL APPLICATION, Ritter et al . (eds.), pages 60-84 (Cambridge University Press 1995) . As discussed above, an antigenic peptide can be attached to a reduced thiol group in the hinge region of an antibody component. Alternatively, the antigenic peptide can be conjugated via a carbohydrate moiety in the Fc region of the antibody. The carbohydrate group can be used to increase the loading of the same peptide that is bound to a thiol group, or the carbohydrate moiety can be used to bind a different peptide.
Methods for conjugating peptides to antibody components via an antibody carbohydrate moiety are well- known to those of skill in the art. See, for example, Shih et al . , Int . J . Cancer 41 : 832 (1988) ; Shih et al . , Int . J . Cancer 46 : 1101 (1990) ; and Shih et al . , U.S. patent No. 5,057,313. The general method involves reacting an antibody component having an oxidized carbohydrate portion with a carrier polymer that has at least one free amine function and that is loaded with a plurality of peptide. This reaction resultε in an initial Schiff base (imine) linkage, which can be stabilized by reduction to a secondary amine to form the final conjugate.
The Fc region is absent if an antibody fragment is used as the antibody component of the immunoconjugate. However, it is possible to introduce a carbohydrate moiety into the light chain variable region of an antibody or antibody fragment. See, for example, Leung et al . , J . Immunol . 154 : 5919 (1995); Hansen et al . , U.S. patent No. 5,443,953 (1995) . The engineered carbohydrate moiety is used to attach the antigenic peptide. 7. The use of Immunoconjugates, Antibodies and Cytokines to Amplify the Humoral and Cellular Immune Response Against Tumor Cells and Infectious Agents
The present invention contemplates the therapeutic use of immunoconjugates, Abl, Ab2 generated against Abl, and fragments of either Abl or Ab2. These immunoconjugates, antibodies and antibody fragments can be used as vaccines to induce both humoral and cellular immune responses in the recipient mammal. Moreover, the administration of immunoconjugates, Abl and/or bispecific antibodies can be used to amplify the integrated immune response.
According to one method of the present invention, a mammal is immunized with a vaccine comprising Abl or fragments thereof, to induce the production of Ab2 and T cells (T2 cells) . After the mammal begins to produce T2 cells, the mammal may be given Abl, or fragments thereof, by intravenous administration to expand the T2 cell mass. An additional advantage of this second administration is that the antibodies or fragments bind with cognate antigen on cancer cellε or infectiouε organisms and thus, serve as targets for T2 cells. Methods for detecting the production of T cells that react with specific antibodies are well-known to those of ordinary skill in the art. See, for example, Fagerberg et al . , Cancer Immunol . Immunother . 37 : 264 (1993) , which is incorporated by reference.
According to a preferred method, a mammal is subsequently immunized with a vaccine comprising Ab2, or fragments thereof, to induce the formation of Ab3 and T cells that recognize Ab2 (T3 cells) . An advantage of this subsequent Ab2 vaccination is that cells expressing a tumor associated antigen or infectious agent antigen are destroyed by T3 cells directed to the antigen, and by T2 cells directed to Ab3 , which also is bound by the antigen. Example 4 illustrateε a method of treatment compriεing the administration of an Abl vaccine, Abl (or fragments) , and an Ab2 vaccine. According to a preferred embodiment of this method, a MAb conjugated to a cytokine or lymphokine is administered by intravenous injection subsequent to immunization with Abl. This step amplifies the cytotoxic lymphocyte clones that are induced by the intradermal immunization with Abl and accrete in the targeted cells. The MAb portion of the conjugate can be directed to the same antigen as Abl used in the immunization, or to a different antigen. If the MAb is directed to the same epitope or antigenic determinant on the antigen as the Abl used in the immunization, there should be no cross- reactivity between the idiotype of the Abl used in the vaccine and the idiotype of the MAb in the conjugate. For example, MN-14 and NP-4 are both Class III, anti-CEA MAb that react with the same epitope on CEA, but the two have different idiotypes.
The cytokine or lymphokine to which the MAb is conjugated is one that drives induction of immune cytotoxic lymphocytes. Exemplary MAb-cytokine/lymphokine conjugates include IL-1, IL-2, 11-12, IL-15, CSF and
GM-CSF, with IL-2 and IL-15 being particularly preferred.
In addition, the T2 response may be further amplified by the intravenous administration of Abl antibodies or fragments after Ab2 vaccination. It is possible that the efficacy of an Ab2 vaccine may be decreased by the presence of circulating Abl antibody components, which have been administered intravenously. Therefore, it is advantageous to clear circulating Abl components prior to the administration of an Ab2 vaccine. One method that can be used to achieve Abl clearance is to use Abl antibodies that have been conjugated with biotin. In this way, circulating biotinylated Abl can be cleared prior to Ab2 vaccination by the intravenous administration of avidin. Preferably, clearance with avidin is performed one to two days after the intravenous administration of Abl (or fragments thereof) . This antibody clearance technique is described by Goldenberg, international application publication No. WO 94/04702 (1994) .
In an alternative method of immunotherapy, a mammal is immunized with an Abl vaccine, treated with Abl (or fragments) to saturate a high percentage of tumor or infectious agent antigen siteε and then, hyperimmunized with Abl vaccine to generate large numbers of cytotoxic lymphocytes directed against cells coated with Abl (or fragments thereof) . The immunoconjugates of the present invention are used to further enhance the efficacy of antibody vaccine administration. According to one method, an antibody or antibody fragment is conjugated with a peptide capable of inducing a strong major histocompatibility complex (MHC)- restricted immune response. An example of a suitable antigenic peptide is the tetanus toxin P2 peptide, described above. Such a peptide can be conjugated, for example, to the IMMU-LL1 (EPB-1) antibody, which binds with the HLA-DR-complex on the plasma membrane of macrophages, monocytes, and B-lymphocytes. Palak- Byczkowεka et al . , Cancer Res. 49 : 4568 (1989) . An IMMU- LL1 vaccine firεt is injected intradermally to establish primary sensitization and then, the vaccine is administered intravenously to boost the immune response. Once a mammal has been sensitized by treatment with an immunoconjugate, such as an IMMU-LL1-P2 vaccine, the mammal can be treated with an immunoconjugate that directs the immune response to tumor cells. For example, an immunoconjugate comprising humanized LL2 and P2 can be used to target CD22-bearing tumor cells. LL2 is described by Goldenberg et al . , J . Clin . Oncol . 9 : 548 (1991) , and by Murthy et al . , Eur . J . Nucl . Med . 19 : 394 (1992). In this way, the sensitizing peptide (e . g . , P2) is cleaved from the antibody component after internalization, bound to class II MHC heterodimers, and transported to the cell surface. Cytotoxic T cells generated with the LL1-P2 vaccine will then recognize the HLA-II-peptide complex on the cellular membrane and destroy the tumor cell. This general approach can be used to treat other tumors that expresε the HLA-DR complex, or to treat autoimmune diseaseε that are caused by cells expressing the HLA-DR complex. Immunoconjugates also can be used to induce or to boost the immune response to a tumor cell or to an infectious agent using a peptide that contains a suitable epitope. As an illustration, a peptide containing the A3B3 domain of CEA can be conjugated to IMMU-LLl antibody (or fragment) and injected subcutaneously to establish primary sensitization against CEA, or injected intravenously to boost the immune response to CEA.
Similarly, immunoconjugates comprising CDRε of anti- idiotype antibodieε can be used to induce or to boost the immune response. In this approach, a peptide containing the amino acid sequence of a CDR is conjugated with an antibody or antibody fragment. For example, the minimal recognition unit of IMMU-14 Ab2 antibody can be conjugated with IMMU-LLl antibody or antibody fragment. The preparation of IMMU-14 anti-idiotype antibodieε iε described in Example 2.
According to preferred methods of immunotherapy, the immune response is further amplified by the administration of cytokines. Examples of cytokines include the interferons (INFε) , interleukinε (ILε) and tumor necrosis factors. INF-7 induces macrophages, as well aε cell-surface class II histocompatibility antigenε on lymphoid cellε and monocyteε. See, for example, Kleger an et al . , "Lymphokines and Monokines," in BIOTECHNOLOGY AND PHARMACY, Pezzuto et al . (eds.) , pages 53-70 (Chapman & Hall 1993) , and Roitt et al . , IMMUNOLOGY, 3rd Edition, pageε 7.8-7.14 (Mosby 1993). IL-2 is a T cell growth factor and a stimulator of natural killer cells and tumor-reactive T cells. Id . Thus, INF-7 and IL-2 are preferred cytokines for the augmentation of the immune response.
IL-12 is another preferred cytokine for enhancing the immune response to the immunoconjugates of the present invention. This cytokine is produced by phagocytic cells in response to bacteria, bacterial products and intracellular parasites. See, for example, Trinchieri, Annu . Rev . Immunol . 13 : 251 (1995) . IL-12 induces cytokine production, primarily INF-7, by natural killer cells and by T cells, and IL-12 acts aε a growth factor for activated natural killer cells and T cellε, enhances the cytotoxic activity of natural killer cells, and stimulates cytotoxic T cell generation. Jd. In experimental animal model systems, IL-12 has been used to treat Schistosoma mansoni, Mycobacterium avium, Histoplas a capsulatum , as well as sarcoma, lung metastases. Wynn et al . , Nature 376 : 594 (1995) ; Castro et al . , J . Immunol . 155 : 2013 (1995) ; Zhou et al . , J . Immunol . 155 : 785 (1995) ; Zitvogel et al . , J . Immunol . 155 : 1393 (1995) .
The antibodies and fragments of the present invention can be used as vaccines by conjugating the antibodieε or fragments to a soluble immunogenic carrier protein. Suitable carrier proteins include keyhole lympet hemocyanin, which is the preferred carrier protein. The antibodies and fragments can be conjugated to the carrier protein using standard methods. See, for example, Hancock et al , "Synthesis of Peptideε for Use as Immunogens," in METHODS IN MOLECULAR BIOLOGY: IMMUNOCHEMICAL PROTOCOLS, Manson (ed.), pages 23-32 (Humana Press 1992) . Immunoconjugates comprising one of the above-described antigenic peptides do not require the addition of an immunogenic carrier protein. A preferred vaccination composition compriεes an antibody conjugate or fragment conjugate, and an adjuvant. Examples of suitable adjuvants include aluminum hydroxide and lipid. Methods of formulating vaccine compositions are well-known to those of ordinary skill in the art. See, for example, Rola, "Immunizing Agents and Diagnostic Skin Antigens," in REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, Gennaro (ed.) , pages 1389-1404 (Mack Publishing Company 1990) . Additional pharmaceutical methods may be employed to control the duration of action of a vaccine in a therapeutic application. Control release preparations can be prepared through the use of polymers to complex or adsorb the immunoconjugates, antibodies or fragments. For example, 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, antibody or antibody fragment from such a matrix depends upon the molecular weight of the immunoconjugate, antibody or antibody fragment, the amount of immunoconjugate, antibody or antibody fragment within the matrix, and the size of dispersed particles. Saltzman et al . , Biophys . J . 55: 163 (1989); Sherwood et al . , supra . Other solid doεage 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) .
The therapeutic preparations of the present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby immunoconjugates, antibodies or antibody fragments are combined in a mixture with a pharmaceutically acceptable carrier. A composition is said to be a "pharmaceutically acceptable carrier" if its administration can be tolerated by a recipient mammal. Sterile phosphate- buffered saline is one example of a pharmaceutically acceptable carrier. Other suitable carriers 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 PHARMACEUTICAL SCIENCES, 18th Edition (Mack Publishing Company 1990) .
The immunoconjugates, antibodies or fragments may be administered to a mammal intravenously or subcutaneously. Moreover, the administration may be by continuous infusion or by single or multiple boluseε. Preferably, an antibody vaccine is administered subcutaneously, while an antibody preparation that is not a vaccine iε administered intravenously. In general, the dosage of administered immunoconjugates, antibodies or fragments for humans will vary depending upon such factors as the patient's age, weight, height, sex, general medical condition and previous medical history. Typically, it is desirable to provide the recipient with a dosage of immunoconjugates, antibodies or fragments which is in the range of from about 1 pg/kg to 10 mg/kg (amount of agent/body weight of patient) , although a lower or higher dosage also may be administered as circumstances dictate. For purposes of therapy, immunoconjugates, antibodieε or fragments are administered to a mammal in a therapeutically effective amount. An antibody preparation is said to be administered in a "therapeutically effective amount" if the amount administered is physiologically significant. An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient mammal. In particular, an antibody preparation of the present invention is physiologically significant if itε presence invokes a humoral and/or cellular immune response in the recipient mammal.
A cytokine, such as INF-7, IL-2 , or IL-12 may be administered before and during the administration of an Abl vaccine or an Ab2 vaccine. Alternatively, cytokines may be administered together before and during the administration of an antibody vaccine. Cytokines are administered to the mammal intravenously, intramuscularly or subcutaneously. For example, recombinant IL-2 may be administered intravenously as a bolus at 6 x io1 iu/kg or aε a continuouε infuεion at a dose of 18 x 106 IU/m2/d. Weiss et al . , J . Clin . Oncol . 10 : 275 (1992). Alternatively, recombinant IL-2 may be administered subcutaneously at a dose of 12 x 106 IU. Vogelzang et al . , J. Clin . Oncol . 11 : 1809 (1993) . Moreover, INF-7 may be administered subcutaneously at a dose of 1.5 x IO6 U. Lienard et al . , J. Clin . Oncol . 10 : 52 (1992) . Furthermore, Nadeau et al . , J. Pharmacol . Exp . Ther. 274 : 78 (1995) , have shown that a single intravenous dose of recombinant IL-12 (42.5 μg/kilogram) elevated IFN-7 levels in rhesus monkeys.
Suitable IL-2 formulations include PROLEUKIN (Chiron Corp./Cetus Oncology Corp.; Emeryville, CA) and TECELEUKIN (Hoffman-La Roche, Inc.; Nutley, NJ) . ACTIMMUNE (Genentech, Inc.; South San Francisco, CA) is a suitable INF-7 preparation.
In addition, bispecific antibodies may be administered after the initial Abl treatment . The function of the bispecific antibodies is to bridge lymphocytes with CEA-bearing tumor cells and to trigger the lymphocyte-mediated cytolysis. Bispecific antibodies can be administered according to above-described general guidelines. However, bispecific antibodies, unlike antibody vaccines, are not conjugated with immunogens.
Those of ordinary skill in the art will appreciate that the above-described methods can be used to provide prophylaxis against infectious agents. Thus, the present invention contemplates the use of methods described herein to provide protection to a mammal before exposure to an infectious agent.
The present invention, thus generally described, will be understood more readily by reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present invention. EX AMPLE 1 Production of Murine Anti-CEA MAb (MN-14)
The production of MN-14, a Class III, anti-CEA MAb, has been described by Hansen et al . , Cancer 71 : 3478 (1993), which is incorporated by reference. Briefly, a 20 gram BALB/c female mouse was immunized subcutaneouεly with 7.5 μg of partially-purified CEA in complete Freund adjuvant. On day 3, the mouse was boosted subcutaneously with 7.5 μg of CEA in incomplete Freund adjuvant and then, the mouse was boosted intravenously with 7.5 μg of CEA in saline on days 6 and 9. On day 278, the mouse was given 65 μg of CEA intravenously in saline and 90 μg of CEA in saline on day 404. On day 407, the mouse was sacrificed, a cell suspension of the spleen was prepared, the spleen cells were fused with murine myeloma cells, SP2/0-Ag 14 (ATCC CRL 1581) using polyethylene glycol, and the cells were cultured in medium containing 8- azaguanine. Hybridoma supematants were screened for CEA-reactive antibody using an 125I-CEA radioimmunoasεay (Roche; Nutley, NJ) . Positive clones were recloned.
One clone, designated MN-14 , had properties similar to the Claεε III anti-CEA-εpecific MAb, NP-4, being unreactive with normal crosε-reactive antigen and meconium antigen. However, MN-14, compared with NP-4 , demonstrated significantly superior tumor targeting in a human colon tumor xenograft model and consistently stronger staining of frozen sectionε of colon cancer.
EXAMPLE 2
Preparation of CDR-Grafted MN-14 (hMN-14) and hAbl Vaccine (hMN-14 Vaccine)
A modified antibody was prepared in which the complementarity determining regions (CDR) of MN-14 were engrafted to the framework regions of human IgG! antibody. The CDR-grafted ("humanized") MN-14 antibody was designated "hMN-14." General techniques for producing humanized antibodies are described, for example, by Jones et al . , Nature 321 : 522 (1986), Riechmann et al . , Nature 332 : 323 (1988) , Verhoeyen et al . , Science 239 : 1534 (1988), Carter et al ., Proc . Nat 'l Acad . Sci . USA 89 : 4285 (1992), Sandhu, Crit . Rev . Biotech . 12 : 437 (1992), and Singer et al . , J . Immun . 150 : 2844 (1993) .
To prepare hMN-14 vaccine, hMN-14 was conjugated with keyhole lympet hemocyanin. Typically, patients are immunized with subcutaneous injections of the conjugate
(2 mg/injection) mixed with 100 μl (107 organisms) of
Tice Bacillus Calmette-Guerin (Organon; West Orange, NJ) .
EXAMPLE 3 Preparation of Rat Monoclonal Ab2 to MN-14 (WI2) and Ab2 Vaccine (WI2 Vaccine)
Rat Ab2 to MN-14 was prepared as described by Losman et al . , Int . J . Cancer 56 : 580 (1994) , which is incorporated by reference. Briefly, female 3-week-old Copenhagen rats were injected intraperitoneally with 200 μg of MN-14 F(ab')2 fragments emulsified in Freund's complete adjuvant. Animals were boosted at days 200, 230, and 235 with the same amount of antigen in Freund's incomplete adjuvant. Four days after the last injection, animals were sacrificed, spleen cell suεpenεions were prepared, and the cells were fused with murine non- secreting plasmocytoma SP2/0 using standard techniques. Hybridoma cells were cultured in the presence of rat peritoneal feeder cells (10,000 cells/200 μl culture well) .
Culture εupernatantε were screened by ELISA for reactivity with MN-14 and absence of reactivity with control murine MAbs. Positive hybridomas were cloned at least twice by limiting dilution in the presence of rat peritoneal feeder cells. WI2 is an IgGu Ab2 which is specific for MN-14 and does not react with other isotype-matched anti-CEA MABs. Immunization of mice or rabbits with WI2 (but not with control rat IgG) induced the production of Abl' anti-CEA antibodies. Thus, WI2 can be used as an idiotype vaccine for patients with CEA-producing tumors.
WI2 vaccine is prepared from WI2 as deεcribed for the preparation of hMN-14 vaccine.
EXAMPLE 4 T_reat_ne_at with hMN-14 Vaccine (hAbl -Vaccine) and WI2 Vaccine (Ab2 Vaccine)
A patient with Dukes C colon carcinoma underwent a primary tumor resection for cure and then, was placed on fluorouracil and Levamisole adjuvant therapy. The pre- operative CEA titer was 15.5 ng/ml. Three months after primary surgery, the CEA titer was in the normal range, that is, below 2.5 ng/ml.
Two yearε later, the patient waε found to have a CEA titer of 25 ng/ml and a CAT scan showed a 5 cm tumor in the left lobe of liver and a 2 cm tumor in the right lobe. One month later, the CEA titer was 25 ng/ml and the patient was immunized subcutaneouεly with 2 mg of hAbl vaccine (day 0) . Immunization was repeated at day 7. On day 30, the patient waε found to have lymphocytes reactive with the Abl (T2 cells) . On day 40, the patient was given 100 mg of the hAbl intravenously. Two monthε later, the CEA titer was 5 ng/ml and a CAT scan showed that the left lobe tumor had decreased to 2 cm in size, while the right lobe tumor had completely regresεed.
Six monthε later, the left lobe tumor had increaεed in size, and a large tumor mass was found in the abdomen, as confirmed by needle biopsy. The CEA titer had increased to 50 ng/ml. The patient waε given the WI2 Ab2 vaccine (2 mg) subcutaneously on day 0 and on day 30. A severe reaction occurred at the injection site on day 35, which slowly resolved.
Three months later, the CEA titer was found to be less than 2.5 ng/ml, and the left lobe tumor had completely resolved. The mass in the abdomen was reduced in size and a needle biopsy failed to reveal the presence of a tumor, demonstrating only fibrouε tiεεue infiltrated with lymphocytes.
Two years later, a CAT scan showed that tumor recurrence had not occurred, and the CEA titer was less than 2.5 ng/ml.
EXAMPLE 5
Preparation and Use of an Immunoconjugate to Induce Primary Sensitization
IMMU-LLl (EPB-1) is a murine monoclonal antibody that binds with the HLA-DR complex on the plasma membrane of macrophages, monocytes, and B-lymphocytes and then, rapidly internalizes. The preparation of IMMU-LLl is described by Pawak-Byczkowska et al . , Cancer Res . 49 : 4568 (1989) . F(ab')2 fragments are prepared from intact IMMU-LLl by conventional proteolysiε techniques, and conjugated with the P2 peptide [SEQ ID NO: 1] of tetanus toxin at the hinge region, aε deεcribed above. Alternatively, the P2 peptide iε conjugated via an engineered carbohydrate moiety on the light chain of the antibody fragmentε using the techniques of Leung et al . , J . Immunol 154 : 5919 (1995) .
The IMMU-LL1-P2 vaccine is administered εubcutaneously to establish primary senεitization due to the strong MHC-restricted immune response induced by the
P2 moiety. The IMMU-LL1-P2 vaccine also can be administered intravenously to boost the immune response.
LL2 is a murine monoclonal antibody that binds with
CD22 on B-cell lymphomas. See, for example, Goldenberg et al . , J . Clin . Oncol . 9 : 548 (1991) ; Murthy et al . ,
Eur . J . Nucl . Med . 19 : 394 (1992). Humanized LL2 is prepared as described by Leung et al . , Hybridoma 13:469 (1994), and antibody fragments of humanized LL2 are prepared using standard techniques. An LL2-P2 conjugate is prepared as described above and administered intravenously to the sensitized subject to direct the immune response against tumor cells bearing the CD22 antigen.
EXAMPLE 6
Preparation and Use of an Immunoconjugate Comprising an Epitope of a Tumor Associated Antigen
To target CEA-expressing tumor cells, the A3B3 epitope of CEA is produced recombinantly or by peptide synthesis using the known amino acid sequence. Jessup et al . , int . J . Cancer 55 : 262 (1993); Zhou et al . , Cancer Res . 53 : 3817 (1993); and Hefta et al . , Cancer Res . 52 : 5647 (1992) . A3B3 peptides are conjugated to IMMU-LLl antibody or fragment using standard techniques described above. The IMMU-LL1-A3B3 vaccine is administered subcutaneously to induce the immune response against CEA- bearing tumor cells. The vaccine also can be administered intravenously to boost the immune response against such tumor cells.
EXAMPLE 7
Preparation and Use of an Immunoconjugate Comprising a Minimal Recognition Unit
Peptides having the amino acid sequence of minimal recognition units of the Ab2 antibody described in Example 2 are prepared using the techniques described in section 6 above. The peptides are conjugated with IMMU- LL1 antibodies or fragments to produce immunoconjugates that are suitable for inducing (via subcutaneous administration) or boosting (via intravenous administration) the immune response. EXAMPLE 8
Treatment with hMN-14 Vaccine (hABl-Vaccine) , hW12 vaccine (hAb2 Vaccine) and hNP-4-IL-15 conjugate
A patient with an adenocarcinoma of the lung undergoes resection of the primary tumor, and CEA is demonstrated to be present on the cancer cells by immunohistology. Three months later, the blood CEA increases from 5 ng/ml to 20 ng/ml and a bone scan demonstrates recurrent carcinoma at numberous sites. Standard chemotherapy is given, however, the CEA titer continues to rise, and a repeat bone scan demonstrates tumor progression. The patient then is immunized with hMN14 and hW12 intradermally, as in Example 4. IL-2 is administered during the immunization to divert the immune response to the Tl helper pathway. Subsequently, a conjugate of IL-15 and hNP-4, a Class III anti-CEA- specific MAb (hNP-4-IL-15) , is administered by intravenous infusion. Over the next three months the blood CEA titer decreases to a non-detectable level, and a bone scan demonstrates the therapy has produced a complete response.
EXAMPLE 9
Treatment with an Abl specific for MUCl (MA5) , an Anti- Id raised to MA5 (Ab2-W5) and and hNP-4-IL-2 conjugate A patient with primary breast cancer is found to have metastatic disease in the lungs, liver and bone. Immunohistology demonstrates that the cancer cells are negative for estrogen receptor, but produce both CEA and MUCl. The primary tumor is resected, and the patient is placed on chemotherapy, but only a minimal response is achieved. The patient then is immunized intradermally with a vaccine that contains an Abl specific for MUCl (MA5) and an anti-Id raised to MA5 (Ab2-W5) . W5 has been proven to induce an Ab3 in rabbits that reacts strongly with MUCl. IL-2 is administered with the vaccine. Subsequently, a hNP-4-IL-2 conjugate is administered by intravenous infusion. Three months later the tumor deposits in the lungs and bone have resolved, and the tumor nodules in the liver have decreased in size. The patient again is treated with the Abl/Ab2 vaccine, and with the hNP-4-IL-2 conjugate. Three months later all tumor nodules in the liver have disappeared, and there is no evidence of tumor in other organs.
Although the foregoing refers to particular preferred embodiments, it will be understood that the present invention is not so limited. It will occur to those of ordinary skill in the art that various modifications may be made to the disclosed embodiments and that such modifications are intended to be within the scope of the present invention, which is defined by the following claims.
All publications and patent applications mentioned in this specification are indicative of the level of skill of those in the art to which the invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference in its entirety.

Claims

What Is Claimed Is:
1. A method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen (TAA) or against a disease caused by an infectious agent, said method comprising:
(a) administering a first vaccine intradermally to the mammal, wherein said first vaccine compriseε an immunoconjugate that compriεeε: (i) an antibody component that binds with the
HLA-DR-complex, and (ii) an antigenic peptide, wherein said antigenic peptide compriseε at least one epitope of a TAA or an antigen associated with said infectious agent, and
(b) administering said vaccine intravenously to said mammal.
2. The method of claim 1, wherein said antibody component is selected from the group conεisting of:
(a) a murine monoclonal antibody;
(b) a humanized antibody derived from a murine monoclonal antibody;
(c) a human monoclonal antibody; and
(d) an antibody fragment derived from (a) , (b) or (c) .
3. The method of claim 2, wherein said antibody fragment is selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
4. The method of claim 1, wherein said method further comprises administering interleukin-2, interferon-7 and interleukin-12 prior to and during the administering of said vaccine intravenously to said mammal.
5. The method of claim 1, wherein said method further comprises administering a conjugate of a monoclonal antibody with a cytokine or lymphokine after intradermal administration of said first vaccine.
6. A method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses a tumor associated antigen (TAA) , said method comprising
(a) administering a first vaccine intradermally to the mammal, wherein said first vaccine compriseε an immunoconjugate that compriεes: (i) an antibody component that binds with the
HLA-DR-complex, and (ii) an antigenic peptide that induces a major histocompatibility (MHC) -restricted immune response, and
(b) administering said vaccine intravenously to said mammal.
7. The method of claim 6, wherein said antibody component is selected from the group consiεting of:
(a) a murine monoclonal antibody;
(b) a humanized antibody derived from a murine monoclonal antibody;
(c) a human monoclonal antibody; and
(d) an antibody fragment derived from (a) , (b) or (c) .
8. The method of claim 6, wherein εaid antibody fragment iε selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
9. The method of claim 6, wherein said method further comprises administering at least one cytokine selected from the group consisting of interleukin-2, interferon-7 and interleukin-12 prior to and during the administering of said vaccine intravenously to said mammal.
10. The method of claim 6, wherein said antigenic peptide is tetanus toxin P2 peptide.
11. The method of claim 6, wherein said method further comprises administering a conjugate of a monoclonal antibody with a cytokine or lymphokine after intradermal administration of said first vaccine.
12. The method of claim 6, further comprising administering a second vaccine intravenously to said mammal, wherein said second vaccine comprises an immunoconjugate that comprises:
(i) an antibody component that binds with a TAA, and
(ii) an antigenic peptide that induces a MHC- restricted immune response.
13. The method of claim 12, wherein said antigenic peptide of said second vaccine is tetanuε toxin P2 peptide.
14. The method of claim 13 , further compriεing administering at least one cytokine selected from the group consisting of interleukin-2, interleukin-12 and interferon-7 prior to and during the administering of the second vaccine intravenously to said mammal.
15. A method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses carcinoembryonic antigen (CEA) , said method comprising: (a) administering a first vaccine to the mammal, wherein said first vaccine comprises an antibody component that binds with CEA, and wherein said antibody component is conjugated with a soluble immunogenic carrier protein;
(b) administering a second vaccine to said mammal, wherein said second vaccine comprises an anti- idiotype antibody component that mimics an epitope of said CEA, and wherein said anti- idiotype antibody component is conjugated with a soluble immunogenic carrier protein; and
(c) administering a third vaccine to said mammal, wherein said third vaccine comprises an immunoconjugate comprising an antigenic peptide that comprises an epitope of CEA, and an antibody component that binds with the HLA-DR complex.
16. The method of claim 15, wherein said antigenic peptide of said third vaccine comprises the A3B3 domain of CEA.
17. The method of claim 14, wherein εaid antigenic peptide of εaid third vaccine compriεeε a minimal recognition unit of an anti-idiotype antibody that mimicε an epitope of said CEA.
18. The method of claim 15, wherein said antibody component of said first vaccine is selected from the group consisting of:
(a) a murine monoclonal Claεs III anti-CEA antibody;
(b) a humanized antibody derived from a murine monoclonal Class III anti-CEA antibody;
(c) a human monoclonal anti-CEA antibody; and
(d) an antibody fragment derived from (a) , (b) or (c).
19. The method of claim 18, wherein said antibody fragment is selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
20. The method of claim 15, wherein said anti- idiotype antibody component is selected from the group consisting of:
(a) a polyclonal antibody that binds with the variable region of a Clasε III anti-CEA antibody;
(b) a murine monoclonal antibody that bindε with the variable region of a Claεε III anti-CEA antibody;
(c) a humanized antibody derived from (b) ;
(d) a human monoclonal antibody that binds with the variable region of a Class III anti-CEA antibody;
(e) a subhuman primate antibody that binds with the variable region of a Clasε III anti-CEA antibody; and
(f) an antibody fragment derived from (a) , (b) , (c), (d) or (e) .
21. The method of claim 20, wherein said antibody fragment is selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
22. The method of claim 15, wherein εaid method further comprises administering at least one cytokine selected from the group consisting of interleukin-2, interleukin-12 and interferon-7 prior to and during said administration of said second vaccine.
23. The method of claim 15, wherein said method further comprises administering a conjugate of a monoclonal antibody with a cytokine or lymphokine.
24. Use of a vaccine comprising an immunoconjugate that comprises:
(i) an antibody component that binds with the
HLA-DR-complex, and (ii) an antigenic peptide, wherein said antigenic peptide compriseε at least one epitope of a TAA or an antigen associated with said infectious agent, in the preparation of a medicament for use in an improved method for inducing humoral and cellular immune responses in a patient against a tumor that expresεeε a tumor associated antigen (TAA) or against a disease caused by an infectious agent, wherein in said method said patient is:
(a) administered said vaccine intradermally, and
(b) administered said vaccine intravenously.
25. Use of a vaccine comprising an immunoconjugate that comprises:
(i) an antibody component that binds with the
HLA-DR-complex, and (ii) an antigenic peptide that induces a major histocompatibility (MHC) -restricted immune response, in the preparation of a medicament for use in an improved method for inducing humoral and cellular immune responεeε in a mammal against a tumor that expresεeε a tumor associated antigen (TAA) , wherein in said method said patient is:
(a) administered said vaccine intradermally, and
(b) administered said vaccine intravenously.
26. Use of:
(1) a first vaccine comprising an immunoconjugate that comprises:
(i) an antibody component that binds with the
HLA-DR-complex, and (ii) an antigenic peptide that induces a major histoco patibility (MHC) -restricted immune response, and
(2) a second vaccine comprising an immunoconjugate that comprises:
(i) an antibody component that binds with a
TAA, and (ii) an antigenic peptide that induces a MHC- restricted immune response, in the preparation of a medicament for use in an improved method for inducing humoral and cellular immune reεponεeε in a mammal against a tumor that expresseε a tumor associated antigen (TAA) , wherein in said method said patient is:
(a) administered said first vaccine intradermally,
(b) administered said first vaccine intravenously, and
(c) administered said second vaccine intravenously.
27. Use of:
(1) a first vaccine comprising an antibody component that binds with CEA and is conjugated with a soluble immunogenic carrier protein;
(2) a second vaccine comprising an anti-idiotype antibody component that mimics an epitope of said CEA, and wherein said anti-idiotype antibody component is conjugated with a soluble immunogenic carrier protein; and
(3) a third vaccine comprising an immunoconjugate comprising an antigenic peptide that compriseε an epitope of CEA, and an antibody component that bindε with the HLA-DR complex, in the preparation of a medicament for use in an improved method for inducing humoral and cellular immune responses in a mammal against a tumor that expresses carcinoembryonic antigen (CEA) , wherein in said method said patient is:
(a) administered said first vaccine,
(b) administered said second vaccine, and
(c) administered said third vaccine.
28. The use of any of claims 24 through 27, wherein said antibody component is selected from the group consisting of:
(a) a murine monoclonal antibody;
(b) a humanized antibody derived from a murine monoclonal antibody;
(c) a human monoclonal antibody; and
(d) an antibody fragment derived from (a) , (b) or (c) .
29. The use of any of claims 24 through 27, wherein said antibody fragment is selected from the group consisting of F(ab')2 , F(ab)2, Fab', Fab, Fv, sFv and minimal recognition unit.
30. The use of either of claim 24 or 25, wherein said medicament further comprises at least one cytokine selected from the group consisting of interleukin-2, interleukin-12 and interferon-7, and said patient is administered said at least one cytokine prior to and during the administering of said vaccine intravenously.
31. The use of either of claim 26 or 27, wherein said medicament further comprises at least one cytokine selected from the group consisting of interleukin-2, interleukin-12 and interferon-7, and said patient is administered said at least one cytokine prior to and during the administering said second vaccine intravenously.
32. The use of either of claims 25 or 26, wherein said antigenic peptide is tetanus toxin P2 peptide.
33. The use of claim 27, wherein said antigenic peptide of said third vaccine comprises the A3B3 domain of CEA.
34. The use of claim 27, wherein said antigenic peptide of said third vaccine comprises a minimal recognition unit of an anti-idiotype antibody that mimics an epitope of εaid CEA.
35. The use of claim 27, wherein said antibody component of said first vaccine is selected from the group consisting of:
(i) a murine monoclonal Class III anti-CEA antibody; (ii) a humanized antibody derived from a murine monoclonal Class III anti-CEA antibody; (iii) a human monoclonal anti-CEA antibody; and (iv) an antibody fragment derived from (i) ,
(ii) or (iii) .
36. The use of claim 27, wherein said anti-idiotype antibody component of said second vaccine is selected from the group consisting of:
(i) a polyclonal antibody that binds with the variable region of a Clasε III anti-CEA antibody; (ii) a murine monoclonal antibody that bindε with the variable region of a Claεε III anti-CEA antibody; (iii) a humanized antibody derived from (b) ; (iv) a human monoclonal antibody that bindε with the variable region of a Claεε III anti-CEA antibody; (v) a subhuman primate antibody that binds with the variable region of a Class III anti-CEA antibody; and
(vi) an antibody fragment derived from (i) , (ii) , (iii) , (iv) or (v) .
37. The use of either of claim 24 or 25, wherein said medicament further comprises administering at least one cytokine selected from the group consisting of interleukin-2, interferon-7 and interleukin-12 prior to and during the administering of said vaccine intravenously to said mammal.
38. The use of any of claim 24 through 26, wherein said medicament further comprises a conjugate of a monoclonal antibody with a cytokine or lymphokine and said patient is administered said at MAb- cytokine/lymphokine conjugate subsequent to the administering of said vaccine intradermally.
EP96943706A 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines Ceased EP0881910A4 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP09002533A EP2057999A3 (en) 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/577,106 US7354587B1 (en) 1994-07-06 1995-12-22 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
US577106 1995-12-22
PCT/US1996/019755 WO1997023237A1 (en) 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP09002533A Division EP2057999A3 (en) 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines

Publications (2)

Publication Number Publication Date
EP0881910A1 true EP0881910A1 (en) 1998-12-09
EP0881910A4 EP0881910A4 (en) 2006-05-03

Family

ID=24307306

Family Applications (2)

Application Number Title Priority Date Filing Date
EP09002533A Ceased EP2057999A3 (en) 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
EP96943706A Ceased EP0881910A4 (en) 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP09002533A Ceased EP2057999A3 (en) 1995-12-22 1996-12-20 Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines

Country Status (5)

Country Link
EP (2) EP2057999A3 (en)
JP (2) JP2000503003A (en)
AU (1) AU721927B2 (en)
CA (1) CA2240834C (en)
WO (1) WO1997023237A1 (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2301328A1 (en) * 1997-08-13 1999-02-25 The Uab Research Foundation Vaccination by topical application of genetic vectors
AU2994599A (en) * 1998-03-06 1999-09-20 Imclone Systems Incorporated Active immunization against angiogenesis-associated antigens
ATE473242T1 (en) 1998-05-20 2010-07-15 Immunomedics Inc THERAPEUTIC AGENTS CONTAINING A BISPECIFIC ANTI-HLA CLASS II INVARIANT CHAIN X ANTI-PATHOGEN ANTIBODIES
JP4286483B2 (en) * 1999-06-09 2009-07-01 イムノメディクス, インコーポレイテッド Immunotherapy for autoimmune diseases using antibodies targeting B cells
AU8024100A (en) * 1999-10-13 2001-04-23 Roswell Park Memorial Institute Induction of a strong immune response to a self-tumor associated antigen
JP4212921B2 (en) 2002-03-29 2009-01-21 独立行政法人科学技術振興機構 Therapeutic agents using protein hollow nanoparticles presenting antibodies and protein hollow nanoparticles
TWI434855B (en) * 2006-11-21 2014-04-21 Hoffmann La Roche Conjugate and its use as a standard in an immunoassay
US9562075B2 (en) 2007-05-31 2017-02-07 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Intradermal HPV peptide vaccination
TWI438675B (en) 2010-04-30 2014-05-21 Ibm Method, device and computer program product for providing a context-aware help content
JP6130307B2 (en) * 2011-03-17 2017-05-17 ザ ユニバーシティ オブ バーミンガム Redirected immunotherapy
GB201203442D0 (en) 2012-02-28 2012-04-11 Univ Birmingham Immunotherapeutic molecules and uses
CN104168918A (en) * 2012-03-19 2014-11-26 德国癌症研究中心 B-cell receptor complex binding proteins containing T-cell epitopes
GB201216649D0 (en) * 2012-09-18 2012-10-31 Univ Birmingham Agents and methods
CA2971288A1 (en) 2015-02-02 2016-08-11 The University Of Birmingham Targeting moiety peptide epitope complexes having a plurality of t-cell epitopes
EP3377103B1 (en) 2015-11-19 2021-03-17 Revitope Limited Functional antibody fragment complementation for a two-components system for redirected killing of unwanted cells

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0324625A1 (en) * 1988-01-12 1989-07-19 Bunge (Australia) Proprietary Limited Antigen antibody conjugate
US5194254A (en) * 1986-05-06 1993-03-16 Connaught Laboratories Limited Enhancement of antigen immunogenicity
WO1995031483A1 (en) * 1994-05-13 1995-11-23 Eclagen Limited Improvements in or relating to peptide delivery
WO1996004313A1 (en) * 1994-08-05 1996-02-15 Immunomedics, Inc. Polyspecific immunoconjugates and antibody composites for targeting the multidrug resistant phenotype
WO1996037224A1 (en) * 1995-05-25 1996-11-28 Ludwig Institute For Cancer Research Methods of treating colon cancer utilizing tumor-specific antibodies
WO1996040941A1 (en) * 1995-06-07 1996-12-19 Connaught Laboratories Limited Chimeric antibodies for delivery of antigens to selected cells of the immune system

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4036945A (en) 1976-05-03 1977-07-19 The Massachusetts General Hospital Composition and method for determining the size and location of myocardial infarcts
US4361549A (en) 1979-04-26 1982-11-30 Ortho Pharmaceutical Corporation Complement-fixing monoclonal antibody to human T cells, and methods of preparing same
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4671958A (en) 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
US4818709A (en) 1983-01-21 1989-04-04 Primus Frederick J CEA-family antigens, Anti-CEA antibodies and CEA immunoassay
US5525338A (en) 1992-08-21 1996-06-11 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin conjugates
US5057313A (en) 1986-02-25 1991-10-15 The Center For Molecular Medicine And Immunology Diagnostic and therapeutic antibody conjugates
US4699784A (en) 1986-02-25 1987-10-13 Center For Molecular Medicine & Immunology Tumoricidal methotrexate-antibody conjugate
GB8610983D0 (en) * 1986-05-06 1986-06-11 Connaught Lab Enhancement of antigen immunogenicity
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
DE69030172T2 (en) 1990-01-26 1997-06-19 Immunomedics Inc Vaccines against cancer and infectious diseases
US5443953A (en) 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5194254A (en) * 1986-05-06 1993-03-16 Connaught Laboratories Limited Enhancement of antigen immunogenicity
EP0324625A1 (en) * 1988-01-12 1989-07-19 Bunge (Australia) Proprietary Limited Antigen antibody conjugate
WO1995031483A1 (en) * 1994-05-13 1995-11-23 Eclagen Limited Improvements in or relating to peptide delivery
WO1996004313A1 (en) * 1994-08-05 1996-02-15 Immunomedics, Inc. Polyspecific immunoconjugates and antibody composites for targeting the multidrug resistant phenotype
WO1996037224A1 (en) * 1995-05-25 1996-11-28 Ludwig Institute For Cancer Research Methods of treating colon cancer utilizing tumor-specific antibodies
WO1996040941A1 (en) * 1995-06-07 1996-12-19 Connaught Laboratories Limited Chimeric antibodies for delivery of antigens to selected cells of the immune system

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
BECKER S: "Interferon-gamma accelerates immune proliferation via its effect on monocyte HLA - DR expression." CELLULAR IMMUNOLOGY, (1985 MAR) 91 (1) 301-7., March 1985 (1985-03), XP000939351 *
BOHLEN H ET AL: "Idiotype vaccination strategies against a murine B-cell lymphoma: dendritic cells loaded with idiotype and bispecific idiotype x anti-class II antibodies can protect against tumor growth." CYTOKINES AND MOLECULAR THERAPY, (1996 DEC) 2 (4) 231-8., December 1996 (1996-12), XP000946037 *
BURTON J.D. ET AL: 'CD74 is expressed by multiple myeloma and is a promising target for therapy' CLIN CANCER RES vol. 10, 01 October 2004, pages 6606 - 6611 *
CARAYANNIOTIS G ET AL: "Delivery of synthetic peptides by anti-class II MHC monoclonal antibodies induces specific adjuvant-free Ig responses in vivo." MOLECULAR IMMUNOLOGY, (1988 SEP) 25 (9) 907-11., September 1988 (1988-09), XP000939329 *
DATABASE WPI Week 199601, Derwent Publications Ltd., London, GB; Class B04, AN 1996-010883 'IMPROVEMENT IN OR RELATING TO PEPTIDE DELIVERY' & WO 95 31483 A1 (ECLAGEN LIMITED) 23 November 1995 *
HANSEN H. ET AL: 'Internalization and catabolism of radiolabelled antibodies to the MHC class - II invariant chain by B-cell lymphomas' BIOCHEM JOURNAL vol. 320, 15 November 1996, pages 293 - 300, XP002110872 *
HANSEN H. ET AL: 'MAb LL1 reacts with the cell-surface invariant chain of HLA-Dr complexes on plasma membranes: In vitro and in vivo results' FASEB JOURNAL vol. 6, no. 10, 30 April 1996, page A1211 *
LOSMAN M J ET AL: "Human response against NP-4, a mouse antibody to carcinoembryonic antigen: human anti-idiotype antibodies mimic an epitope on the tumor antigen." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, (1991 APR 15) 88 (8) 3421-5., 15 April 1991 (1991-04-15), XP002147167 *
NEPOM G T ET AL: "Induction of immunity to a human tumor marker by in vivo administration of anti-idiotypic antibodies in mice." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, (1984 MAY) 81 (9) 2864-7., May 1984 (1984-05), XP002147166 *
PAWLAK-BYCZKOWSKA E.J. ET AL: 'Two new monoclonal antibodies, EPB-1 and EPB-2, reactive with human lymphoma' CANCER RES vol. 49, no. 16, 15 August 1989, pages 4568 - 4577 *
ROCHE P.A. ET AL: 'Cell surface HLA-DR-invariant chain complexes are targeted to endosomes by rapid internalization' PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA vol. 90, no. 18, 15 September 1993, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, pages 8581 - 8585, XP002520502 *
See also references of WO9723237A1 *
YU Z ET AL: "Peptide-antibody conjugates for tumour therapy: a MHC-class-II-restricted tetanus toxin peptide coupled to an anti-Ig light chain antibody can induce cytotoxic lysis of a human B-cell lymphoma by specific CD4 T cells." INTERNATIONAL JOURNAL OF CANCER, (1994 JAN 15) 56 (2) 244-8., 15 January 1994 (1994-01-15), XP000946044 *

Also Published As

Publication number Publication date
AU721927B2 (en) 2000-07-20
EP2057999A2 (en) 2009-05-13
JP2000503003A (en) 2000-03-14
EP2057999A3 (en) 2009-07-29
JP2008115196A (en) 2008-05-22
CA2240834A1 (en) 1997-07-03
EP0881910A4 (en) 2006-05-03
CA2240834C (en) 2013-12-03
AU1287197A (en) 1997-07-17
WO1997023237A1 (en) 1997-07-03

Similar Documents

Publication Publication Date Title
US5798100A (en) Multi-stage cascade boosting vaccine
US8163887B2 (en) Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
JP2008115196A (en) Immunoconjugates for enhancing efficacy of multi-stage cascade boosting vaccines
US7348419B2 (en) Humanization of an anti-carcinoembryonic antigen anti-idiotype antibody as a tumor vaccine and for targeting applications
US6306393B1 (en) Immunotherapy of B-cell malignancies using anti-CD22 antibodies
AU728325B2 (en) Immunotherapy of B-cell malignancies using anti-CD22 antibodies
Bhattacharya-Chatterjee et al. Anti-idiotype vaccine against cancer
JPH11500607A (en) Mouse monoclonal anti-idiotype antibody 3H1
Bhattacharya-Chatterjee et al. Anti-idiotype antibody vaccine therapies of cancer
MXPA98009586A (en) Method and composition for the reconformation of multi-peptide antigens to start an animal response

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19980721

AK Designated contracting states

Kind code of ref document: A1

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

A4 Supplementary search report drawn up and despatched

Effective date: 20060316

17Q First examination report despatched

Effective date: 20061121

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

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20091016