WO1993025240A2 - Pretargeting methods and compounds - Google Patents

Pretargeting methods and compounds Download PDF

Info

Publication number
WO1993025240A2
WO1993025240A2 PCT/US1993/005406 US9305406W WO9325240A2 WO 1993025240 A2 WO1993025240 A2 WO 1993025240A2 US 9305406 W US9305406 W US 9305406W WO 9325240 A2 WO9325240 A2 WO 9325240A2
Authority
WO
WIPO (PCT)
Prior art keywords
biotin
conjugate
ligand
targeting moiety
antibody
Prior art date
Application number
PCT/US1993/005406
Other languages
French (fr)
Other versions
WO1993025240A3 (en
Inventor
Donald B. Axworthy
Louis J. Theodore
Linda M. Gustavson
John M. Reno
Original Assignee
Neorx Corporation
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 US07/895,588 external-priority patent/US5283342A/en
Priority to AT93915235T priority Critical patent/ATE210464T1/en
Priority to CA002134239A priority patent/CA2134239C/en
Priority to JP6501638A priority patent/JPH07507804A/en
Priority to US08/351,469 priority patent/US5608060A/en
Priority to EP93915235A priority patent/EP0646019B9/en
Priority to DE69331319T priority patent/DE69331319T2/en
Application filed by Neorx Corporation filed Critical Neorx Corporation
Publication of WO1993025240A2 publication Critical patent/WO1993025240A2/en
Publication of WO1993025240A3 publication Critical patent/WO1993025240A3/en
Priority to US08/350,551 priority patent/US6075010A/en
Priority to US08/468,513 priority patent/US5976535A/en
Priority to US08/788,339 priority patent/US6287536B1/en
Priority to US09/316,452 priority patent/US6358490B2/en
Priority to US09/561,736 priority patent/US6416738B1/en
Priority to US09/920,454 priority patent/US6709652B2/en
Priority to US10/076,031 priority patent/US20030099640A1/en
Priority to US10/125,788 priority patent/US20030129191A1/en
Priority to US10/775,897 priority patent/US7078013B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/555Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound pre-targeting systems involving an organic compound, other than a peptide, protein or antibody, for targeting specific cells
    • A61K47/557Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound pre-targeting systems involving an organic compound, other than a peptide, protein or antibody, for targeting specific cells the modifying agent being biotin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • A61K47/665Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells the pre-targeting system, clearing therapy or rescue therapy involving biotin-(strept) avidin systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6887Antibody-chelate conjugates using chelates for therapeutic purposes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • A61K47/6893Pre-targeting systems involving an antibody for targeting specific cells clearing therapy or enhanced clearance, i.e. using an antibody clearing agents in addition to T-A and D-M
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • A61K47/6897Pre-targeting systems with two or three steps using antibody conjugates; Ligand-antiligand therapies
    • A61K47/6898Pre-targeting systems with two or three steps using antibody conjugates; Ligand-antiligand therapies using avidin- or biotin-conjugated antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1268Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules host-guest, closed hollow molecules, inclusion complexes, e.g. with cyclodextrins, clathrates, cavitates, fullerenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2123/00Preparations for testing in vivo

Definitions

  • the present invention relates to methods
  • administered dose migrating to bone marrow is low. Also, the absolute dose of radiation or therapeutic agent delivered to the tumor is insufficient in many cases to elicit a significant tumor response.
  • the present invention is directed to diagnostic and therapeutic pretargeting methods, moieties useful therein and methods of making those moieties.
  • Such pretargeting methods are characterized by an improved targeting ratio or increased absolute dose to the target cell sites in comparison to conventional cancer therapy.
  • the present invention describes chelate-biotin compounds and radiohalogenated biotin compounds useful in diagnostic and therapeutic pretargeting methods.
  • the present invention also provides targeting moiety-ligand, such as biotin, compounds useful in diagnostic and therapeutic pretargeting methods. Selection of moieties and methodologies used to enhance targeting moiety-ligand, such as biotin, compounds useful in diagnostic and therapeutic pretargeting methods. Selection of moieties and methodologies used to enhance targeting moiety-ligand, such as biotin, compounds useful in diagnostic and therapeutic pretargeting methods. Selection of moieties and methodologies used to enhance targeting moiety-ligand, such as biotin, compounds useful in diagnostic and therapeutic pretargeting methods. Selection of moieties and methodologies used to enhance
  • the present invention provides targeting moiety-anti-ligand, such as avidin or streptavidin, compounds useful in diagnostic and therapeutic pretargeting methods.
  • targeting moiety-anti-ligand such as avidin or streptavidin
  • Other ligand-anti-ligand systems including the zinc finger protein-dsDNA fragment binding pair are also contemplated.
  • the present invention also provides clearing agents to facilitate the removal of circulating targeting moiety-ligand (two-step) or targeting moiety-anti-ligand (two-step) or anti-ligand (three-step) from the mammalian recipient.
  • clearing agents are classifiable as galactose-based and non-galactose-based. Within each category, preferable clearing agents are polymeric or protein based. Particulate agents, extracorporeal procedures and in vivo devices are also contemplated for use in the practice of the present invention.
  • the present invention is directed to methods using streptavidin as an anti-ligand to enhance retention of radionuclide at target cell sites, with pretargeting protocols constituting one such method. More specifically, these embodiments of the present invention involve either (1) targeting moiety-streptavidin-radionuclide (with the radionuclide bound to streptavidin directly or through a chelate or linker), as well as (2) targeting moiety-biotin administered prior to streptavidin-radionuclide, or (3) biotin-radionuclide bound to a pretargeted
  • streptavidin containing molecule streptavidin containing molecule
  • the present invention further provides
  • pretargeting methods employing intraarterial or other local administration of the therapeutic moiety-containing molecule to achieve greater localization thereof to artery-supplied target cell populations.
  • Other methods of the present invention involve
  • monovalent targeting moieties such as Fv or Fab antibody fragments
  • Delivery of other, non-radioactive therapeutic agents such as
  • chemotherapeutic drugs e.g., cytokines
  • anti-tumor agents e.g., cytokines
  • toxins to target cells using the pretargeting methods of the present invention is also described.
  • Figure 2 depicts radiorhenium tumor uptake in a three-step pretargeting protocol, as compared to administration of radiolabeled antibody (conventional means involving antibody that is covalently linked to chelated radiorhenium).
  • Figure 3 depicts the tumor uptake profile of NR-LU-10-streptavidin conjugate (LU-10-StrAv) in comparison to a control profile of native NR-LU-10 whole antibody.
  • Figure 4 depicts the tumor uptake and blood clearance profiles of NR-LU-10-streptavidin conjugate.
  • Figure 5 depicts the rapid clearance from the blood of asialoorosomucoid in comparison with
  • orosomucoid in terms of percent injected dose of I-125-labeled protein.
  • Figure 7 depicts NR-LU-10-streptavidin conjugate blood clearance upon administration of three controls (O, ⁇ , ⁇ ) and two doses of a clearing agent ( , ⁇ ) at 25 hours post-conjugate administration.
  • Figure 8 shows limited biodistribution data for LU-10-StrAv conjugate upon administration of three controls (Groups 1, 2 and 5) and two doses of clearing agent (Groups 3 and 4) at two hours post-clearing agent administration.
  • Figure 9 depicts NR-LU-10-streptavidin conjugate serum biotin binding capability at 2 hours post-clearing agent administration.
  • Figure 10 depicts NR-LU-10-streptavidin conjugate blood clearance over time upon administration of a control (O) and three doses of a clearing agent ( ⁇ , ⁇ , ⁇ ) at 24 hours post-conjugate administration.
  • Figure 11A depicts the blood clearance of LU-10-StrAv conjugate upon administration of a control (PBS) and three doses (50, 20 and 10 ⁇ g) of clearing agent at two hours post-clearing agent administration.
  • Figure 11B depicts LU-10-StrAv conjugate serum biotin binding capability upon administration of a control (PBS) and three doses (50, 20 and 10 ⁇ g) of clearing agent at two hours post-clearing agent administration.
  • Figure 12 depicts the prolonged tumor retention of NR-LU-10-streptavidin conjugate ( ⁇ ) relative to NR-LU-10 whole antibody ( ⁇ ) over time.
  • Figure 13 depicts the prolonged liver retention of a pre-formed complex of NR-LU-10-biotin ( O ;
  • Figure 14 depicts the prolonged liver retention of Biotin-PIP-I-131 label relative to the streptavidin-NR-LU-10-(PIP-I-125) label.
  • Figure 15A depicts tumor uptake for increasing doses of PIP-Biocytin in terms of %ID/G.
  • Figure 15B depicts tumor uptake for increasing doses of PIP-Biocytin over time in terms of pMOL/G.
  • Figure 16A depicts tumor versus blood localization of a 0.5 ⁇ g dose of PIP-Biocytin over time in terms of %ID/G.
  • Figure 16B depicts tumor versus blood localization of a 0.5 ⁇ g dose of PIP-Biocytin in terms of % ID.
  • Figure 17A depicts tumor uptake of LU-10-StrAv and PIP-Biocytin over time in terms of %ID/G.
  • Figure 17B depicts blood clearance of LU-10-StrAv and PIP-Biocytin over time in terms of %ID/G.
  • Figure 18 depicts PIP-Biocytin:LU-10-StrAv ratio in tumor and blood over time.
  • Targeting moiety A molecule that binds to a defined population of cells.
  • the targeting moiety may bind a receptor, an oligonucleotide, an enzymatic substrate, an antigenic determinant, or other binding site present on or in the target cell population.
  • Antibody is used throughout the specification as a prototypical example of a targeting moiety.
  • Tumor is used as a prototypical example of a target in
  • Ligand/anti-ljgand pair A complementary/anti-complementary set of molecules that demonstrate specific binding, generally of relatively high
  • Exemplary ligand/anti-ligand pairs include zinc finger protein/dsDNA fragment, enzyme/inhibitor, hapten/antibody, lectin/carbohydrate, ligand/receptor, and biotin/avidin. Biotin/avidin is used throughout the specification as a prototypical example of a ligand/anti-ligand pair.
  • Anti-ligand As defined herein, an "anti-ligand” demonstrates high affinity, and preferably,
  • the anti-ligand is large enough to avoid rapid renal clearance, and contains sufficient
  • Anti-ligands of the present invention may exhibit or be derivitized to exhibit structural features that direct the uptake thereof, e.g., galactose residues that direct liver uptake.
  • Avidin and streptavidin are used herein as
  • Avidin As defined herein, “avidin” includes avidin, streptavidin and derivatives and analogs thereof that are capable of high affinity, multivalent or univalent binding of biotin.
  • Ligand As defined herein, a "ligand” is a relatively small, soluble molecule that exhibits rapid serum, blood and/or whole body clearance when
  • Biotin is used as the prototypical ligand.
  • a diagnostic or therapeutic agent A diagnostic or therapeutic agent
  • the payload including radionuclides, drugs, anti-tumor agents, toxins and the like. Radionuclide therapeutic agents are used as prototypical active agents.
  • N x S y Chelates As defined herein, the term "N x S y chelates” includes bifunctional chelators that are capable of (i) coordinately binding a metal or
  • radiometal and (ii) covalently attaching to a
  • N x S y chelates have N 2 S 2 and N 3 S cores.
  • N x S y chelates are described in Fritzberg et al., Proc. Natl. Acad. Sci. USA 85:4024-29. 1988; in Weber et al., Bioconi. Chem. 1:431-37, 1990; and in the references cited therein, for instance.
  • Pretargeting involves target site localization of a targeting moiety that is conjugated with one member of a
  • Clearing Agent An agent capable of binding, complexing or otherwise associating with an
  • the clearing agent is preferably characterized by physical
  • Target Cell Retention The amount of time that a radionuclide or other therapeutic agent remains at the target cell surface or within the target cell.
  • Catabolism of conjugates or molecules containing such therapeutic agents appears to be primarily responsible for the loss of target cell retention.
  • a conjugate encompasses chemical conjugates (covalently or non-covalently bound), fusion proteins and the like.
  • Permeability Enhancing Moiety An agent capable of increasing the permeability at a target site characterized by a three dimensional cellular matrix.
  • Exemplary permeability enhancing moieties function by one or more of the following mechanisms: inducing gaps in the endothelium of venules through action on the postcapillary bed; inducing such gaps through action on the entire capillary bed; disrupting cell-to-cell associations; mediating target cell
  • Intercellular Junction An area of interacting adjacent plasma membranes. Intercellular junctions can be categorized functionally into: (1) adhering junctions that hold cells tightly together (for example, desmosomes); (2) impermeable junctions that hold cells tightly together and prevent leakage of molecules between cells (i.e., tight junctions); and (3) communicating junctions that mediate passage of small molecules between adjacent cells (for instance, gap junctions).
  • Immunogen A substance which is capable, under appropriate conditions, of inducing a specific immune response and of reacting with the products of that response (e.g., a specific antibody, specifically sensitized T-lymphocytes or both).
  • Hapten Immunogen A specific protein-free
  • Lymphokine Soluble protein mediators released by certain lymphocytes, which in turn can regulate other cell-mediated immune functions, such as lymphocyte transformation, macrophage activation or cytotoxicity on other cells.
  • Mitogen A substance that induces mitosis and cell transformation, especially lymphocyte
  • a recognized disadvantage associated with in vivo administration of targeting moiety-radioisotopic conjugates for imaging or therapy is localization of the attached radioactive agent at both non-target and target sites.
  • the administered radiolabeled conjugate clears from the circulation, normal organs and tissues are transitorily exposed to the attached radioactive agent.
  • radiolabeled whole antibodies that are administered in vivo exhibit relatively slow blood clearance; maximum target site localization generally occurs 1-3 days post-administration.
  • the longer the clearance time of the conjugate from the circulation the greater the radioexposure of non-target organs.
  • radiosensitive is the dose-limiting organ of non-specific toxicity.
  • potential targeting moieties In order to decrease radioisotope exposure of non-target tissue, potential targeting moieties generally have been screened to identify those that display minimal non-target reactivity, while retaining target specificity and reactivity.
  • increased doses of a radiotherapeutic conjugate may be administered; moreover, decreased non-target accumulation of a radiodiagnostic conjugate leads to improved contrast between background and target.
  • Therapeutic drugs administered alone or as targeted conjugates, are accompanied by similar disadvantages. Again, the goal is administration of the highest possible concentration of drug (to
  • targeting moiety-therapeutic drug conjugates it would be advantageous to combine the relative target specificity of a targeting moiety with a means for enhanced target cell internalization of the targeting moiety-drug conjugate.
  • diagnostic agent-targeting moiety conjugates In contrast, enhanced target cell internalization is disadvantageous if one administers diagnostic agent-targeting moiety conjugates. Internalization of diagnostic conjugates results in cellular catabolism and degradation of the conjugate. Upon degradation, small adducts of the diagnostic agent or the
  • diagnostic agent per se may be released from the cell, thus eliminating the ability to detect the conjugate in a target-specific manner.
  • One. method for reducing non-target tissue exposure to a diagnostic or therapeutic agent involves
  • pretargeting the targeting moiety at a target site, and then subsequently administering a rapidly clearing diagnostic or therapeutic agent conjugate that is capable of binding to the "pretargeted" targeting moiety at the target site.
  • a description of some embodiments of the pretargeting technique may be found in US Patent No. 4,863,713 (Goodwin et al.).
  • anti-ligand conjugate which is allowed to localize at a target site and to dilute in the circulation. Subsequently administered anti-ligand binds to the antibody-ligand conjugate and clears unbound antibody-ligand conjugate from the blood.
  • Preferred anti-ligands are large and contain sufficient multivalency to accomplish
  • Anti-ligand clearance of this type is preferably accomplished with a
  • receptor-based clearance mechanisms e.g., Ashwell receptor hexose, e.g., galactose, mannose or the like, residue
  • ligand-anti-ligand pair displays relatively high affinity binding.
  • a diagnostic or therapeutic agent-ligand conjugate that exhibits rapid whole body clearance is then administered.
  • anti-ligand binds the circulating active agent-ligand conjugate and produces an antibody-ligand : anti-ligand : ligand-active agent "sandwich" at the target site.
  • the diagnostic or therapeutic agent is attached to a rapidly clearing ligand (rather than antibody, antibody fragment or other slowly clearing targeting moiety), this technique promises decreased non-target exposure to the active agent.
  • pretargeting methods eliminate the step of parenterally administering an anti-ligand clearing agent.
  • These "two-step” procedures feature targeting moiety-ligand or targeting moiety-anti-ligand
  • a clearing agent preferably other than ligand or anti-ligand alone is administered to facilitate the clearance of circulating targeting moiety-containing conjugate.
  • clearing agent preferably does not become bound to the target cell population, either directly or through the previously administered and target cell bound
  • targeting moiety-anti-ligand or targeting moiety- ligand conjugate An example of two-step pretargeting involves the use of biotinylated human transferrin as a clearing agent for avidin-targeting moiety
  • the two-step pretargeting approach overcomes certain disadvantages associated with the use of a clearing agent in a three-step pretargeted protocol. More specifically, data obtained in animal models demonstrate that in vivo anti-ligand binding to a pretargeted targeting moiety-ligand conjugate (i.e., the cell-bound conjugate) removes the targeting moiety-ligand conjugate from the target cell.
  • a pretargeted targeting moiety-ligand conjugate i.e., the cell-bound conjugate
  • the apparent loss of targeting moiety-ligand from the cell might result from internal degradation of the conjugate and/or release of active agent from the conjugate (either at the cell surface or intracellularly).
  • An alternative explanation for the observed phenomenon is that permeability changes in the target cell's membrane allow increased passive diffusion of any molecule into the target cell.
  • some loss of targeting moiety-ligand may result from alteration in the affinity by subsequent binding of another moiety to the targeting moiety-ligand, e.g., anti-idiotype monoclonal antibody binding causes removal of tumor bound monoclonal antibody.
  • a targeting moiety may be covalently linked to both ligand and therapeutic agent and administered to a recipient. Subsequent administration of anti-ligand crosslinks targeting moiety-ligand-therapeutic agent tripartite conjugates bound at the surface, inducing internalization of the tripartite conjugate
  • targeting moiety-ligand may be delivered to the target cell surface , followed by administration of anti-ligand-therapeutic agent.
  • targeting moiety-anti-ligand conjugate is administered in vivo; upon target localization of the targeting moiety-anti-ligand conjugate (i.e., and clearance of this conjugate from the circulation), an active agent- ligand conjugate is parenterally administered.
  • This method enhances retention of the targeting moiety-anti-ligand : ligand-active agent complex at the target cell (as compared with targeting moiety-ligand : anti-ligand : ligand-active agent complexes and targeting moiety-ligand : anti-ligand-active agent complexes).
  • ligand/anti-ligand pairs may be suitable for use within the claimed invention, a preferred ligand/anti-ligand pair is biotin/avidin.
  • radioiodinated biotin described herein is a low molecular weight compound that has been easily and well characterized.
  • a targeting moiety-ligand conjugate is administered in vivo; upon target localization of the targeting moiety-ligand conjugate (i.e., and clearance of this conjugate from the circulation), a drug-anti-ligand conjugate is parenterally administered.
  • This two-step method not only provides pretargeting of the targeting moiety conjugate, but also induces internalization of the subsequent targeting moiety-ligand-anti-ligand-drug complex within the target cell.
  • another embodiment provides a three-step protocol that produces a targeting moiety-ligand : anti-ligand : ligand-drug complex at the surface, wherein the ligand-drug conjugate is administered simultaneously or within a short period of time after administration of anti-ligand (i.e., before the targeting moietyligand-anti-ligand complex has been removed from the target cell surface). Additional internalization methodologies are contemplated by the present
  • biotin derivatives were radiolabeled with indium-Ill for use in pretargeted immunoscintigraphy (for instance, Virzi et al., Nucl. Med. Biol. 18:719-26, 1991; Kalofonos et al., J. Nucl. Med. 31: 1791-96, 1990; Paganelli et al., Cane. Res. 51:5960-66, 1991).
  • 99m Tc is a particularly preferred radionuclide for
  • Rhenium-186 displays chelating chemistry very similar to 99m Tc, and is considered to be an excellent therapeutic radionuclide (i.e., a 3.7 day half-life and 1.07 MeV maximum particle that is similar to 131 I). Therefore, the claimed methods for technetium and rhenium radiolabeling of biotin provide numerous advantages.
  • the present invention is also directed to
  • the X 90 value of Y-90 is 5.34 mm (i.e., 90% of the energy emitted from a point source is absorbed in a sphere of 5.34 mm radius). Y-90 has a high equilibrium dose constant or mean
  • Y-90 can be manufactured at high specific activity and is available as a generator product. Specific advantages of Y-90 are (1) that it has the capability to kill neighboring target cells not directly targeted by the pretargeted targeting moiety-ligand or targeting moiety-anti-ligand
  • Lu-177 is a particularly preferred radionuclide for targeted nuclide therapy, since it has a
  • Beta av 0.140 MeV
  • the X 90 for Lu-177 is 0.31 mm, i.e., 90% of the energy emitted form a point source is absorbed in a sphere of radius 0.31 mm.
  • Lu-177 has an equilibrium dose constant or mean energy/ nuclear transition of 0.31 Rad-gram/microcuries-hour and an adequate half-life to serve as a targeted therapeutic radionuclide.
  • Specific advantages of Lu-177 are (1) that its emitted energy is efficiently absorbed in smaller targeted tumor volumes such as metastatic tumor foci or involved lymph nodes and (2) that its long physical half-life makes optimal use of the tumor retention property of the pretargeting delivery method.
  • targeting moiety binds to a defined target cell population, such as tumor cells.
  • Oligonucleotides e.g., antisense oligonucleotides that are complementary to portions of target cell nucleic acids (DNA or RNA), are also useful as
  • targeting moieties that retain the capacity to bind to a defined target cell population may also be used within the claimed invention.
  • synthetic targeting moieties may be designed.
  • targeting moieties of the present invention are also useful as targeting moieties of the present invention.
  • One targeting moiety functional equivalent is a "mimetic" compound, an organic compound, and
  • Another targeting moiety designed to mimic the proper configuration and/or orientation for targeting moiety-target cell binding.
  • polypeptides exhibit the binding affinity of the targeting moiety.
  • Preferred targeting moieties of the present invention are antibodies (polyclonal or monoclonal), peptides, oligonucleotides or the like.
  • Polyclonal antibodies useful in the practice of the present invention are polyclonal (Vial and Callahan, Univ. Mich. Med. Bull., 20: 284-6, 1956), affinity-purified polyclonal or fragments thereof (Chao et al., Res. Comm. in Chem. Path. & Pharm., 9: 749-61, 1974).
  • Monoclonal antibodies useful in the practice of the present invention include whole antibody and fragments thereof. Such monoclonal antibodies and fragments are producible in accordance with
  • Useful monoclonal antibodies and fragments may be derived from any species (including humans) or may be formed as chimeric proteins which employ sequences from more than one species. See, generally, Kohler and Milstein, Nature, 256: 495-97, 1975; Eur. J.
  • Murine monoclonal antibodies or "humanized” murine antibody are also useful as targeting moieties in accordance with the present invention.
  • murine monoclonal antibody may be "humanized” by genetically recombining the nucleotide sequence encoding the murine Fv region (i.e., containing the antigen binding sites) or the complementarity
  • Humanized targeting moieties are recognized to decrease the immunoreactivity of the antibody or polypeptide in the host recipient, permitting an increase in the half-life and a
  • annexins and other platelet binding proteins such as PAP-1 (Placental Anticoagulant
  • Annexins are (with the
  • Annexin II single chain, non-glycosylated proteins of approximately 36 kilodaltons.
  • Annexins possess a number of biological properties based on the principle of calcium ion binding.
  • these proteins have an especially high affinity for negatively charged phospholipids such as phosphatidylserine or phosphatidylinosine.
  • Annexins exert anti-coagulatory effects.
  • coagulation inhibition is mediated by the binding of annexins to negatively charged surface phospholipids (i.e., to platelets) which binding blocks the
  • Annexins localize to target sites rapidly, i.e., in a matter of minutes, but remains circulating in the serum for a longer time period.
  • annexins may be employed in two-step or three-step pretargeting protocols for the diagnosis of blood clots or the treatment of blood clots associated with indications, such as DVT (deep vein thrombosis), PE (pulmonary embolism), heart attack, stroke and the like.
  • indications such as DVT (deep vein thrombosis), PE (pulmonary embolism), heart attack, stroke and the like.
  • a chemical conjugate or a fusion protein of annexin with avidin or streptavidin is administered to a recipient for whom such a diagnosis is desired.
  • the annexin portion of the conjugate localizes rapidly to target sites characterized by negatively charged surface
  • phospholipids such as blood clots.
  • a clearing agent such as those discussed herein, may be administered after the passage of a short time (ranging from about 5 minutes to about 1 hour, with within about 15 minutes preferred).
  • the clearing agent serves to rapidly diminish the serum level of the annexin-anti-ligand conjugate.
  • biotin labeled with an imaging radionuclide such as Tc-99m for example, is administered. Biotin directs the localization of the administered moiety to the previously localized annexin-anti-ligand. Non-target bound radiolabeled biotin is rapidly cleared from the recipient.
  • This approach offers the advantages of speed, facilitating imaging of difficult target sites such as lung clots, and improved clot to blood ratios.
  • a chemical conjugate or a fusion protein of annexin with avidin or streptavidin is administered to a recipient for whom such treatment is desired.
  • a clearing agent such as those discussed herein, may be administered after the passage of a short time (ranging from about 5 minutes to about 1 hour, with within about 15 minutes preferred) to diminish the serum level of the annexin-anti-ligand conjugate.
  • Biotin conjugated with therapeutic agents such as fibrolytic agents, tissue plasminogen activator, thrombolytic agents (e.g., streptokinase, anisoylated plasminogen streptokinase activator complex) and the like, are deliverable in this manner.
  • Such a dosing regimen may be repeated, for example, once daily for a number of weeks to address the recipient's physiological condition. Consequently, delivery of a therapeutic agent to target sites proceeds with minimal exposure of nontarget sites to that agent.
  • Annexin V which was isolated by Bohn in 1979 from human placenta, a rich source of annexins, and termed Placenta Protein 4 (PP4).
  • Annexin V consists of four domains, with each made up of five alpha helices wherein the alpha helices serve as connecting elements between the domains. From the side, the molecule appears crown-like with five calcium binding sites on its convex surface, through which annexin-phospholipid interactions are mediated. Other annexins having the characteristics described above are also useful in the practice of the present invention. Annexin V has been expressed in E . coli.
  • Types of active agents (diagnostic or therapeutic) useful herein include toxins, anti-tumor agents, drugs and radionuclides.
  • toxins include toxins, anti-tumor agents, drugs and radionuclides.
  • Several of the potent toxins useful within the present invention consist of an A and a B chain.
  • the A chain is the cytotoxic portion and the B chain is the receptor-binding portion of the intact toxin molecule (holotoxin).
  • toxin B chain may mediate non-target cell binding, it is often advantageous to conjugate only the toxin A chain to a targeting moiety.
  • elimination of the toxin B chain decreases non-specific cytotoxicity, it also generally leads to decreased potency of the toxin A chain-targeting protein conjugate, as compared to the corresponding holotoxin-targeting moiety
  • Preferred toxins in this regard include
  • holotoxins such as abrin, ricin, modeccin,
  • Pseudomonas exotoxin A Diphtheria toxin, pertussis toxin and Shiga toxin
  • a chain or "A chain-like" molecules such as. ricin A chain, abrin, A chain, modeccin A chain, the enzymatic portion of Pseudomonas exotoxin A, Diphtheria toxin A chain, the enzymatic portion of pertussis toxin, the enzymatic portion of Shiga toxin, gelonin, pokeweed antiviral protein, saporin, tritin, barley toxin and snake venom
  • Ribosomal inactivating proteins RIPs
  • naturally occurring protein synthesis inhibitors that lack translocating and cell-binding ability, are also suitable for use herein.
  • Preferred drugs suitable for use herein include conventional chemotherapeutics, such as vinblastine, doxorubicin, bleomycin, methotrexate, 5-fluorouracil, 6-thioguanine, cytarabine, cyclophosphamide and cis-platinum, as well as other conventional chemotherapeutics, such as vinblastine, doxorubicin, bleomycin, methotrexate, 5-fluorouracil, 6-thioguanine, cytarabine, cyclophosphamide and cis-platinum, as well as other conventional chemotherapeutics, such as vinblastine, doxorubicin, bleomycin, methotrexate, 5-fluorouracil, 6-thioguanine, cytarabine, cyclophosphamide and cis-platinum, as well as other conventional chemotherapeutics, such as vinblastine, doxorubicin, ble
  • a particularly preferred drug within the present invention is a trichothecene.
  • Trichothecenes are drugs produced by soil fungi of the class Fungi imperfecti or isolated from Baccharu ⁇ megapotamica (Bamburg, J.R. Proc. Molec. Subcell.
  • trichothecenes may be subdivided into three groups (i.e., Group A, B, and C) as described in U.S. Patent Nos. 4,744,981 and 4,906,452 (incorporated herein by reference).
  • Group A simple trichothecenes include: Scirpene, Roridin C,
  • Group B simple trichothecenes include: Trichothecolone,
  • Trichothecin deoxynivalenol, 3-acetyldeoxynivalenol, 5-acetyldeoxynivalenol, 3,15-diacetyldeoxynivalenol, Nivalenol, 4-acetylnivalenol (Fusarenon-X),
  • Group C simple trichothecenes include: Crotocol and Crotocin.
  • Representative macrocyclic trichothecenes include Verrucarin A, Verrucarin B, Verrucarin J
  • Mytoxin B Myrotoxin A, Myrotoxin B, Myrotoxin C, Myrotoxin D, Roritoxin A, Roritoxin B, and
  • Experimental drugs such as mercaptopurine, N-methylformamide, 2-amino-1,3,4-thiadiazole, melphalan, hexamethylmelamine, gallium nitrate, 3% thymidine, dichloromethotrexate, mitoguazone, suramin,
  • bromodeoxyuridine iododeoxyuridine, semustine, 1-(2-chloroethyl)-3-(2,6-dioxo-3-piperidyl)-1-nitrosourea, N,N'-hexamethylene-bis-acetamide, azacitidine,
  • pentostatin PALA, carboplatin, amsacrine, caracemide, iproplatin, misonidazole, dihydro-5-azacytidine, 4'-deoxy-doxorubicin, menogaril, triciribine phosphate, camrabine, tiazofurin, teroxirone, ethiofos, N-(2-hydroxyethyl)-2-nitro-1H-imidazole-1-acetamide, mitoxantrone, acodazole, amonafide, fludarabine
  • Radionuclides useful within the present invention include gamma-emitters, positron-emitters, Auger electron-emitters. X-ray emitters and fluorescenceemitters, with beta- or alpha-emitters preferred for therapeutic use.
  • Radionuclides are well-known in the art and include 123 I, 125 I, 130 I, 131 I, 133 I, 135 I, 47 Sc, 72 As, 72 Se, 90 Y, 88 Y, 97 Ru, 100 Pd, 101m Rh, 119 Sb, 128 Ba, 197 Hg, 211 At, 212 Bi, 153 Sm, 169 Eu, 212 Pb, 109 Pd, 111 ln, 67 Ga, 68 Ga, 67 Cu, 75 Br, 76 Br, 77 Br, 99m Tc, 11 C, 13 N, 15 O and 18 F.
  • Preferred therapeutic radionuclides include 188 Re, 186 Re, 203 Pb, 212 Pb, 212 Bi, 109 Pd, 64 Cu, 67 CU, 90 Y, 125 I, 131 I, 77 Br, 211At, 97 Ru, 105 Rh, 198 Au and 199 Ag or 177 Lu.
  • anti-tumor agents are administrable in accordance with the present invention.
  • exemplary anti-tumor agents include cytokines, such as IL-2, tumor necrosis factor or the like, lectin inflammatory response promoters (selectins), such as L-selectin, E-selectin, P-selectin or the like, and like molecules.
  • Ligands suitable for use within the present invention include biotin, haptens, lectins, epitopes, dsDNA fragments, enzyme inhibitors and analogs and derivatives thereof.
  • Useful complementary anti-ligands include avidin (for biotin), carbohydrates (for lectins), antibody, fragments or analogs thereof, including mimetics (for haptens and epitopes) and zinc finger proteins (for dsDNA fragments) and enzymes (for enzyme inhibitors).
  • Preferred ligands and anti-ligands bind to each other with an affinity of at least about k D ⁇ 10 -9 M.
  • Zinc finger protein/dsDNA is a ligand-anti-ligand binding pair contemplated by the present invention.
  • Zinc finger proteins are a class of proteins containing repeating subunits of
  • Zinc finger proteins are a family of proteins consisting of variable and conserved regions. Zinc finger proteins, as a class, contain from about 2 to about 39 repeating subunits (fingers), with each such finger capable of binding specific nucleotide sequences. Each finger is capable of binding to a "cleft" in the conformation of the dsDNA/RNA structure that is dependent upon
  • zinc is incorporated into the proteins within each subunit and is important in maintaining proper conformation for dsDNA binding. To illustrate the use of the zinc finger
  • a two-step approach is described below.
  • This ligand-anti-ligand binding pair may also be used in three-step pretargeting protocols of the present invention.
  • a zinc finger protein or a dsDNA fragment is conjugated to a targeting moiety in any convenient manner therefor, including, for
  • DNA or oligonucleotides to proteins are DNA or oligonucleotides to proteins.
  • One simple method is oxidation of the sugar moiety of the DNA.
  • the resultant aldehyde groups are available for reaction with amines on the protein.
  • the amines generated by such reactions are then reduced by reaction with a reducing agent such as NaCNBH ⁇ to form the more stable amine bonds.
  • Another protein-oligonucleotide conjugation method involves initial oxidation of the sugar residue on DNA using an oxidizing agent such as NaIO 4 .
  • the dialdehyde formed is treated with 5-pyridylcystamine
  • the conjugates are administered to a recipient by a route chosen in view of the patient's physical condition and known or anticipated illness. Following target site localization of the administered conjugate (or clearing agent treatment as described herein), administration of the other member of the ligand pair conjugated to a therapeutic agent (e.g., radionuclide, drug or anti-tumor agent) is undertaken,
  • a therapeutic agent e.g., radionuclide, drug or anti-tumor agent
  • therapeutic agent-containing conjugate binds to the previously localized targeting agent containing conjugate.
  • Therapeutic agent-containing conjugate that does not bind at the target site will be eliminated from the recipient's body at a much more rapid rate than a monoclonal antibody-therapeutic agent conjugate would be removed.
  • such therapeutic agent-containing conjugates can be
  • the targeting agent used in a pretargeting protocol may be human or humanized, and all of the other administered components
  • One embodiment of this aspect of the present invention involves the use of pretargeting approaches to target double stranded DNA itself.
  • Such protocols of the present invention are useful, for example, for gene therapy, delivery of tumor suppressive DNA, deleted genes and the like. More specifically, a targeting moiety-zinc finger protein is pretargeted to a target site. Subsequently, a dsDNA fragment that is complementary to the zinc finger protein is injected and allowed to bind to the localized zinc finger protein. Because zinc finger protein-DNA interaction is a normal cell physiologic mechanism, DNA will be delivered free of encumbering chemistry to the
  • the dsDNA is then internalized by . the target cell via the normal mechanisms therefor, i.e., via endocytosis or the like.
  • One component to be administered in a preferred two-step pretargeting protocol involving the biotin-avidin ligand-anti-ligand system is a targeting moiety-avidin or a targeting moiety-streptavidin conjugate.
  • the preferred targeting moiety useful in these embodiments of the present invention is a monoclonal antibody. Protein-protein conjugations are generally problematic due to the formation of
  • covalent streptavidin-antibody conjugation is preferred, but high molecular weight byproducts are often obtained.
  • the degree of crosslinking and aggregate formation is dependent upon several factors, including the level of protein derivitization using heterobifunctional crosslinking reagents.
  • SMCC succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate
  • Streptavidin-proteinaceous targeting moiety conjugates are preferably prepared as described in Example XI below, with the preparation involving the steps of:. preparation of SMCC-derivitized
  • the purified fraction is preferably further characterized by one or more of the following techniques: HPLC size exclusion, SDS-PAGE,
  • thioether conjugates useful in the practice of the present invention may be formed using other thiolating agents, such as SPDP, iminothiolate, SATA or the like, or other thio-reactive
  • heterobifunctional cross linkers such as m-maleimidobenzoyl-N-hydroxysuccinimide ester, N-succinimidyl (4-iodoacetyl) aminobenzoate or the like.
  • Streptavidin-proteinaceous targeting moiety conjugates of the present invention can also be formed by conjugation of a lysine epsilon amino group of one protein with a maleimide-derivitized form of the other protein.
  • a lysine epsilon amino group of one protein with a maleimide-derivitized form of the other protein.
  • maleimide-derivitized form of the other protein For example, at pH 8-10, lysine epsilon amino moieties react with protein maleimides,
  • conjugates can be prepared by reaction of lysine epsilon amino moieties of one protein with aldehyde functionalities of the other protein.
  • the resultant imine bond is reducible to generate the corresponding stable amine bond.
  • Aldehyde functionalities may be generated, for example
  • Another method of forming streptavidin-targeting moiety conjugates involves immobilized iminobiotin that binds SMCC-derivitized streptavidin.
  • iminobiotin immobilized to streptavidin is exploited to readily separate conjugate from the unreacted targeting moiety.
  • Iminobiotin binding can be reversed under conditions of lower pH and elevated ionic strength, e.g., NH 2 OAc, pH 4 (50 mM) with 0.5 M NaCl.
  • ionic strength e.g., NH 2 OAc, pH 4 (50 mM) with 0.5 M NaCl.
  • DTT-reduced antibody preferably free of reductant
  • a molar excess (with respect to streptavidin) of DTT-reduced antibody is added to the nitrogen-purged, phosphate-buffered iminobiotin column wherein the SMCC-streptavidin is bound (DTT-reduced antibody will saturate the bound SMCC-streptavidin, and unbound reduced antibody passing through the column can be reused);
  • targeting moiety-mediated ligand-anti-ligand pretargeting involves the
  • the first approach allows the targeting moiety-containing conjugate to clear from the blood by "natural” or endogenous clearance
  • conjugate target-to-blood ratio "chases” the conjugate from the circulation through in vivo complexation of conjugate with a molecule constituting or containing the complementary anti-ligand or ligand.
  • biotinylated antibodies are used as a ligand-targeting moiety conjugate, for example, avidin forms relatively large aggregated species upon complexation with the circulating biotinylated antibody, which aggregated species are rapidly cleared from the blood by the RES uptake. See, for example, U.S. Patent No. 4,863,713.
  • One problem with this method is the
  • poly-biotinylated transferrin When avidin-targeting moiety conjugates are employed, poly-biotinylated transferrin has been used to form relatively large aggregated species that are cleared by RES uptake. See, for example, Goodwin, J. Nucl. Med. 33(10):1816-18. 1992). Poly-biotinylated transferrin also has the potential for cross-linking and internalizing tumor-bound avidinylated-targeting moiety, however. In addition, both "chase"
  • the present invention provides clearing agents of protein and non-protein composition having physical properties facilitating use for in vivo complexation and blood clearance of anti-ligand/ligand (e.g., avidin/biotin) -targeting moiety (e.g., antibody) conjugates. These clearing agents are useful in improving the target:blood ratio of targeting moiety conjugate. Other applications of these clearing agents include lesional imaging or therapy involving blood clots and the like, employing antibody-active agent delivery modalities. For example, efficacious anti-clotting agent provides rapid target localization and high target:non-target targeting ratio. Active agents administered in pretargeting protocols of the present invention using efficient clearing agents are targeted in the desirable manner and are, therefore, useful in the imaging/therapy of conditions such as pulmonary embolism and deep vein thrombosis.
  • anti-ligand/ligand e.g., avidin/biotin
  • targeting moiety e.g., antibody
  • Other applications of these clearing agents include lesional imaging or therapy involving blood clots and the
  • Preferred clearing agents include hexose-based and non-hexose based moieties.
  • Hexose-based clearing agents are molecules that have been derivatized to incorporate one or more hexoses (six carbon sugar moieties) recognized by Ashwell receptors. Exemplary of such hexoses are galactose, mannose and the like. Galactose is the prototypical clearing agent hexose derivative for the purposes of this description.
  • Protein-type galactose-based clearing agents include proteins having endogenous exposed galactose residues or which have been derivitized to expose or incorporate such galactose residues. Exposed
  • galactose residues direct the clearing agent to rapid clearance by endocytosis into the liver through specific receptors therefor (Ashwell receptors).
  • An exemplary clearing agent of the protein-based/galactose-bearing variety is the
  • galactose residues include, for example, galactosylated albumin, galactosylated-IgM, galactosylated-IgG,
  • HSA Human serum albumin
  • the ligand is biotin and the hexose is galactose. More preferably, HSA is derivatized with from 10-20
  • HSA clearing agents of the present invention are derivatized with from about 12 to about 15 galactoses and 3 biotins. Derivatization with both galactose and biotin are conducted in a manner
  • clearing agents based upon human proteins especially human serum proteins such as, for example, orosomucoid and human serum albumin, are less immunogenic upon administration into the serum of a human recipient.
  • asialoorosomucoid is that human orosomucoid is
  • One way to prevent clearing agent compromise of target-bound conjugate through direct complexation is through use of a clearing agent of a size sufficient to render the clearing agent less capable of diffusion into the extravascular space and binding to target-associated conjugate.
  • This strategy is useful alone or in combination with the aforementioned recognition that exposed galactose residues direct rapid liver uptake.
  • This size-exclusion strategy enhances the effectiveness of non-galactose-based clearing agents of the present invention.
  • the combination (exposed galactose, and size) strategy improves the
  • galactosylated, biotinylated proteins to remove circulating streptavidin-targeting moiety conjugates, for example
  • intermediate molecular weight ranging from about 40,000 to about 200,000 Dal
  • biotinylated asialoorosomucoid galactosyl-biotinyl-human serum albumin or other galactosylated and biotinylated derivatives of non-immunogenic soluble natural proteins, as well as biotin- and galactose-derivitized polyglutamate, polylysine, polyarginine, polyaspartate and the like.
  • High molecular weight moieties ranging from about 200,000 to about
  • Chemically modified polymers of intermediate or high molecular weight ranging from about 40,000 to about 1,000,000 Dal
  • galactose- and biotin-derivitized dextran hydroxypropylmethacrylamide polymers
  • polyvinylpyrrolidone-polystyrene copolymers divinyl ether-maleic acid copolymers, pyran copolymers, or PEG, also have utility as clearing agents in the practice of the present invention.
  • rapidly clearing biotinylated liposomes high
  • molecular weight moieties with poor target access can be derivitized with galactose and biotin to produce clearing agents for use in the practice of the present invention.
  • a further class of clearing agents useful in the present invention involve small molecules (ranging from about 500 to about 10,000 Dal) derivitized with galactose and biotin that are sufficiently polar to be confined to the vascular space as an in vivo volume of distribution. More specifically, these agents exhibit a highly charged structure and, as a result, are not readily distributed into the extravascular volume, because they do not readily diffuse across the lipid membranes lining the vasculature.
  • Such clearing agents are mono- or poly-biotin-derivitized 6,6'-[(3,3'-dimethyl[1,1'-biphenyl]-4,4'-diyl)bis(azo) bis[4-amino-5-hydroxy-1,3-naphthalene disulfonic acid] tetrasodium salt, mono- or poly-biotinyl-galactose-derivitized polysulfated dextran-biotin, mono- or poly-biotinyl-galactose-derivitized dextran-biotin and the like.
  • the galactose-exposed or -derivitized clearing agents are preferably capable of (1) rapidly and efficiently complexing with the relevant ligand- or anti-ligand-containing conjugates via ligand-anti-ligand affinity; and (2) clearing such complexes from the blood via the galactose- receptor, a liver specific degradation system, as opposed to aggregating into complexes that are taken up by the generalized RES system, including the lung and spleen. Additionally, the rapid kinetics of galactose-mediated liver uptake, coupled with the affinity of the ligand-anti-ligand interaction, allow the use of intermediate or even low molecular weight carriers.
  • Non-galactose residue-bearing moieties of low or intermediate molecular weight (ranging from about
  • Example XXII experimentation is (1) asialoorosomucoid-biotin, (2) human serum albumin derivatized with galactose and biotin, and (3) a 70,000 dalton molecular weight dextran derivatized with both biotin and galactose.
  • the experimentation showed that proteins and polymers are derivatizable to contain both galactose and biotin and that the resultant derivatized molecule is
  • Biotin loading was varied to determine the effects on both clearing the blood pool of circulating avidin-containing conjugate and the ability to deliver a subsequently administered biotinylated isotope to a target site recognized by the streptavidin-containing conjugate. The effect of relative doses of the administered components with respect to clearing agent efficacy was also examined.
  • Protein-type and polymer-type non-galactose-based clearing agents include the agents described above, absent galactose exposure or derivitization and the like. These clearing agents act through an
  • the clearing agent used will be selected on the basis of the target organ to which access of the clearing agent is to be excluded. For example, high molecular weight (ranging from about 200,000 to about 1,000,000 Dal) clearing agents will be used when tumor targets or clot targets are involved.
  • Another class of clearing agents includes agents that do not remove circulating ligand or anti-ligand/targeting moiety conjugates, but instead
  • cap-type clearing agents are poly-biotin-derivitized 6,6'-[(3,3'-dimethyl[1,1'-biphenyl]-4,4'-diyl)bis(azo) bis[4-amino-5-hydroxy-1,3-naphthalene disulfonic acid] tetrasodium salt, poly-biotinyl-derivitized
  • polysulfated dextran-biotin mono- or poly-biotinyl-derivitized dextran-biotin and the like.
  • Cap-type clearing agents are derivitized with the relevant anti-ligand or ligand, and then administered to a recipient of previously administered ligand/ or anti-ligand/targeting moiety conjugate. Clearing agent-conjugate binding therefore diminishes the ability of circulating conjugate to bind any
  • active agent-ligand or active agent-anti-ligand conjugate subsequently administered active agent-ligand or active agent-anti-ligand conjugate.
  • the ablation of active agent binding capacity of the circulating conjugate increases the efficiency of active agent delivery to the target, and increases the ratio of target-bound active agent to circulating active agent by preventing the coupling of long-circulating serum protein kinetics with the active agent. Also, confinement of the clearing agent to the plasma compartment prevents compromise of target-associated ligand or anti-ligand.
  • Clearing agents of the present invention may be administered in single or multiple doses.
  • a single dose of biotinylated clearing agent for example, produces a rapid decrease in the level of circulating targeting moiety-streptavidin, followed by a small increase in that level, presumably caused, at least in part, by re-equilibration of targeting moiety-streptavidin within the recipient's physiological compartments.
  • a second or additional clearing agent doses may then be employed to provide supplemental clearance of targeting moiety-streptavidin.
  • clearing agent may be infused
  • clearing agents and clearance systems are also useful in the practice of the present invention to remove circulating targeting moiety-ligand or -anti-ligand conjugate from the recipient's circulation.
  • Particulate-based clearing agents for example, are discussed in Example IX.
  • extracorporeal clearance systems are discussed in Example IX.
  • One embodiment of the present invention in which rapid acting clearing agents are useful is in the delivery of Auger emitters, such as 1-125, 1-123, Er-165, Sb-119, Hg-197, Ru-97, Tl-201 and Br-77, or nucleus-binding drugs to target cell nuclei.
  • Auger emitters such as 1-125, 1-123, Er-165, Sb-119, Hg-197, Ru-97, Tl-201 and Br-77
  • nucleus-binding drugs to target cell nuclei.
  • targeting moieties that localize to internalizing receptors on target cell surfaces are employed to deliver a
  • targeting moiety-containing conjugate i.e., a targeting moiety-anti-ligand -conjugate in the
  • Such internalizing receptors include EGF receptors, transferrin receptors, HER2 receptors, IL-2 receptors, other interleukins and cluster
  • an active agent-containing ligand or anti-ligand is administered.
  • conjugate such as a biotin-Auger emitter or a biotin- nucleus acting drug
  • a biotin-Auger emitter or a biotin- nucleus acting drug is administered as soon as the clearing agent has been given an opportunity to complex with circulating targeting moiety-containing conjugate, with the time lag between clearing agent and active agent administration being less than about 24 hours.
  • active agent is readily internalized through target cell receptor-mediated internalization.
  • Auger emitters While circulating Auger emitters are thought to be non-toxic, the rapid, specific targeting afforded by the pretargeting protocols of the present invention increases the potential of shorter half-life Auger emitters, such as 1-123, which is available and capable of stable binding.
  • DOTA may also be conjugated to other ligands or to anti-ligands in the practice of the present invention.
  • radioimmunotherapy For therapy, it is very important that the radionuclide be stably bound within the DOTA chelate and that the DOTA chelate be stably attached to biotin. Only radiolabeled DOTA-biotin conjugates exhibiting those two characteristics are useful to deliver radionuclides to the targets. Release of the radionuclide from the DOTA chelate or cleavage of the biotin and DOTA conjugate components in serum or at non-target sites renders the conjugate unsuitable for use in therapy.
  • Biotinidase is a
  • hydrolytic enzyme that catalyzes the cleavage of biotin from biotinyl peptides. See, for example, Evangelatos, et al., "Biotinidase Radioassay Using an I-125-Biotin Derivative, Avidin, and Polyethylene
  • Drug-biotin conjugates which structurally resemble biotinyl peptides are potential substrates for
  • DOTA-SC-biotin exhibits significantly improved serum stability in comparison to DOTA-LC-biotin. This result does not appear to be explainable on the basis of biotinidase activity alone. The experimentation leading to this conclusion is summarized in the Table set forth below.
  • biotinidase activity measuring method uses N-(d-biotinyl)-4-aminobenzoate (BPABA) as a substrate, with the hydrolysis of BPABA resulting in the liberation of biotin and 4-aminobenzoate (PABA).
  • BPABA N-(d-biotinyl)-4-aminobenzoate
  • PABA 4-aminobenzoate
  • Some general strategies for improving serum stability of peptides with respect to enzymatic action are the following: incorporation of D-amino acids, N-methyl amino acids and alpha-substituted amino acids.
  • linkage between DOTA and biotin may also have a significant impact on biodistribution (including normal organ uptake, target uptake and the like) and pharmacokinetics.
  • the DOTA-biotin conjugates of the present invention reflect the implementation of one or more of the following strategies:
  • DOTA-biotin conjugates in accordance with the present invention may be generally characterized as follows: conjugates that retain the biotin carboxy group in the structure thereof and those that do not (i.e., the terminal carboxy group of biotin has been reduced or otherwise chemically modified. Structures of such conjugates represented by the following
  • L may alternatively be substituted in one of the following ways on one of the -CH 2 -COOH branches of. the DOTA structure: -CH(L)-COOH or -CH 2 COOL or
  • functional group for binding with the DOTA conjugate component is selected for the capability to interact with either the carbon or the carboxy in the branch portions of the DOTA structure, with the serum
  • L is selected according to the following principles, with the portion of the linker designed to bind to the DOTA conjugate component selected for the capability to bind to an amine.
  • linkers incorporating a D-amino acid spacer between a DOTA aniline amine and the biotin carboxy group shown above. Substituted amino acids are preferred for these embodiments of the present
  • L moieties of this embodiment of the present invention may be represented as follows:
  • R 1 is selected from lower alkyl, lower alkyl substituted with hydrophilic groups (preferably,
  • glucuronide-substituted amino acids or other glucuronide derivatives
  • R 2 is selected from hydrogen, lower alkyl,
  • substituted lower alkyl e.g., hydroxy, sulfate, phosphonate or a hydrophilic moiety (preferably OH).
  • lower alkyl indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with from one to five carbon atoms. Also, the term “lower alkyl” indicates an alkyl group with
  • substituted includes one or several substituent groups, with a single substituent group preferred.
  • R 2 H ( a D- serine -O-
  • n 1 or 2 as well as molecules wherein R 1 is ; or —(CH 2 ) 5 -NH 2 ; or
  • Preferred moieties incorporating the glucuronide of D-lysine and the glucuronide of amino pimelate are shown below as I and II, respectively.
  • a particularly preferred linker of this embodiment of the present invention is the D-alanine derivative set forth above.
  • Linkers incorporating alkyl substitution on one or more amide nitrogen atoms are also encompassed by the present invention, with some embodiments of such linkers preparable from L-amino acids. Amide bonds having a substituted amine moiety are less susceptible to enzymatic cleavage. Such linkers exhibit the following general formula:
  • R 4 is selected from hydrogen, lower alkyl, lower alkyl substituted with hydroxy, sulfate, phosphonate or the like and ;
  • R 3 is selected from hydrogen; an amine; lower alkyl; an amino- or a hydroxy-, sulfate- or
  • n ranges from 0-4.
  • n 4 (Bis-DOTA-LC-biotin), synthesizable from
  • n 0 (bis-DOTA-SC-biotin), synthesizable from
  • Bis-DOTA-LC-biotin offers the following advantages:
  • Another linker embodiment incorporates a thiourea moiety therein.
  • Exemplary thiourea adducts of the present invention exhibit the following general formula :
  • R 5 is selected from hydrogen or lower alkyl
  • R 6 is selected from H and a hydrophilic moiety; and n ranges from 0-4.
  • the second preferred linker recited above can be prepared using either L-lysine or D-lysine.
  • the third preferred linker can be prepared using either N-methyl-D-lysine or N-methyl-L-lysine.
  • biotinhydrazide commercially available from Sigma Chemical Co., St. Louis, Missouri
  • DOTA-benzylisothiocyanate a known compound synthesized in one step from DOTA-aniline
  • Z is -(CH 2 ) 2 -, conveniently synthesized form glutamic acid
  • Z - (CH 2 ) n -CO-O-CH 2 -, where n ranges from 1-4 and which is synthesizable from serine.
  • Another exemplary linker embodiment of the present invention has the general formula set forth below:
  • n ranges from 1-5.
  • Another embodiment involves disulfide-contaicing linkers, which provide a metabolically cleavable moiety (-S-S-) to reduce non-target
  • n and n' preferably range between 0 and 5
  • the advantage of using conditionally cleavable linkers is an improvement in target/non-target
  • Conditionally cleavable linkers include enzymatically cleavable linkers, linkers that are cleaved under acidic
  • linkers that are cleaved under basic conditions and the like More specifically, use of linkers that are cleaved by enzymes, which are present in non-target tissues but reduced in amount or absent in target tissue, can increase target cell retention of active agent relative to non-target cell retention.
  • Such conditionally cleavable linkers are useful, for example, in delivering therapeutic radionuclides to target cells, because such active agents do not require internalization for efficacy, provided that the linker is stable at the target cell surface or protected from target cell degradation.
  • Q is a bond, a methylene group, a -CO- group or -CO- (CH 2 ) n -NH-;
  • n ranges from 1-5.
  • Another amino-containing linker of the present invention is structured as follows: , where R 7 is lower alkyl,
  • Polymeric linkers are also contemplated by the present invention. Dextran and cyclodextran are preferred polymers useful in this embodiment of the present invention as a result of the hydrophilicity of the polymer, which leads to favorable excretion of conjugates containing the same.
  • dextran polymers are substantially non-toxic and non-immunogenic, that they are commercially available in a variety of sizes and that they are easy to conjugate to other relevant molecules.
  • dextran-linked conjugates exhibit advantages when non-target sites are accessible to dextranase, an enzyme capable of cleaving dextran polymers into smaller units while non-target sites are not so accessible.
  • linkers of the present invention have the following general formula:
  • An ether linkage as shown below may be formed in a DOTA-biotin conjugate in accordance with the procedure indicated below.
  • L' -NH-CO-(CH 2 ) n -O- where n ranges from 1 to 5 , with 1 preferred.
  • This linker has only one amide moiety which is bound directly to the DOTA aniline (as in the structure of DOTA-SC-biotin).
  • the ether linkage imparts hydrophilicity, an important factor in
  • This linker contains no amide moieties and the
  • unalkylated amine may impart favorable biodistribution properties since unalkylated DOTA-aniline displays excellent renal clearance.
  • L. Substituted amine linkers, which can form conjugates via amino-biotin intermediates, are shown below.
  • R 8 is H; -(CH 2 ) 2 -OH or a sulfate or phosphonate
  • R 9 is a bond or -(CH 2 ) n -CO-NH-, where n ranges from
  • Amino biotin may also be used as an
  • Conjugates containing this thiourea linker have the following advantages: no cleavable amide and a short, fairly polar linker which favors renal excretion.
  • a bis-DOTD derivative of the following formula can also be formed from amino-biotin.
  • Such a linker is conveniently synthesizable from lysine.
  • L -(CH 2 ) -CO-NH-, wherein q is 1 or 2, and wherein the amine group is attached to the methylene group corresponding to the reduced biotin carboxy moiety and the methylene group (s) are attached to a core carbon in the DOTA ring. This moiety is
  • linkers set forth above are useful to produce conjugates having one or more of the following
  • step six of the prior art synthesis procedure in which a tetra amine alcohol is converted to a tetra-toluenesulfonamide
  • toluenesulfonate as shown below, is the likely result of premature formation of the toluenesulfonate functionality (before all of the amine groups have been converted to their corresponding sulfonamides.
  • Trifluoroacetic anhydride Trifluoroacetates, being much poorer leaving groups than toluenesulfonates, are not vulnerable to analogous side reactions.
  • the easy hydrolysis of trifluoroacetate groups as reported in Greene and Wuts, "Protecting Groups in Organic Synthesis," John Wiley and Sons, Inc., New York, p. 94, 1991., suggests that addition of methanol to the reaction mixture following consumption of all amines should afford the tetra-fluoroacetamide alcohol as a substantially exclusive product.
  • toluenesulfonate of tetra-fluoroacetamide alcohol in methanolic sodium hydroxide at 15-25°C for 1 hour, should afford nitro-benzyl-DOTA as a substantially exclusive product.
  • trifluoracetamide protecting groups circumvents the difficulties associated with cleavage of the very stable toluenesulfonamide protecting group, which involves heating with a large excess of sulfuric acid followed by neutralization with copious volumes of barium hydroxide .
  • This alternative procedure involves the cyclizaton of p-nitrophenylalanyltriglycine using a coupling agent , such as diethylycyanophosphate , to give the cyclic tetraamide .
  • a coupling agent such as diethylycyanophosphate
  • borane reduction provides 2 - (p-nitrobenzyl) -1 , 4 , 7 , 10-tetraazacyclododecane , a common precursor used in published routes to DOTA including the Renn and Meares article referenced above .
  • This alternative procedure of the present invention offers a synthetic pathway that is
  • the resultant chelate-biotin conjugate shows superior kidney excretion. Although the net overall charge of the conjugate is neutral, the polycarboxylate nature of the molecule generates regions of hydrophilicity and hydrophobicity. By altering the number and nature of the carboxylate groups within the conjugate, excretion may be shifted from kidney to
  • anionic compounds are generally cleared through the GI system.
  • Conjugates containing polylysine may also exhibit beneficial biodistribution properties. With whole antibodies, derivitization with polylysine may skew the biodistribution of conjugate toward liver uptake. In contrast, derivitization of Fab fragments with polylysine results in lower levels of both liver and kidney uptake; blood clearance of these conjugates is similar to that of Fab covalently linked to chelate.
  • An exemplary polylysine derivitized chelate-biotin conjugate is illustrated below.
  • polylysine derivatives are preferably succinylated following biotinylation.
  • Polylysine derivatives offer the further advantages of: (1) increasing the specific activity of the radiometal-chelate-biotin conjugate; (2) permitting control of rate and route of blood clearance by varying the molecular weight of the polylysine polymer; and (3) increasing the circulation half-life of the conjugate for optimal tumor
  • poly-L-lysine is acylated according to standard amino group acylation procedures (aqueous bicarbonate buffer, pH 8, added biotin-NHS ester, followed by chelate NHS ester).
  • the number of biotins attached to polylysine is determined by the HABA assay. Spectrophotometric titration is used to assess the extent of amino group derivitization.
  • conjugate is characterized by size exclusion.
  • linkers that are cleaved by enzymes present in normal tissue but deficient or absent in tumor tissue can increase tumor retention.
  • the kidney has high levels of ⁇ -glutamyl transferase; other normal tissues exhibit in vivo cleavage of ⁇ -glutamyl prodrugs.
  • tumors are generally deficient in enzyme peptidases.
  • the glutamyl-linked biotin conjugate depicted below is cleaved in normal tissue and retained in the tumor.
  • biocytin is condensed with N-t-BOC-(O-sulfonate or O-glucose) serine NHS ester to give N-t-BOC-(O-sulfonate or O-glucose) serine biocytinamide.
  • N-t-BOC-(O-sulfonate or O-glucose) serine NHS ester to give N-t-BOC-(O-sulfonate or O-glucose) serine biocytinamide.
  • ligand-amidoserine O-sulfonate or O-glucose
  • Radioiodinated biotin derivatives prepared by exposure of poly-L-lysine to excess NHS-LC-biotin and then to Bolton-Hunter N-hydroxysuccinimide esters in
  • radioiodinated biotin according to the present invention provides certain advantages. First, the radioiodobiotin derivative is a low
  • the disclosed methods for preparation involve a single step and eliminate the need for a purification step.
  • "X" may be any radiohalogen, including 1 25 I, 131 I, 123 I, 211 At and the like.
  • radionuclides resulting from transmutation processes (without non-radioactive carrier forms present) are preferred.
  • exemplary radionuclides include Y-90, Re-188, At-211, Bi-212 and the like.
  • a therapeutically effective amount of radiation ranges from about 1500 to about 10,000 cGy depending upon several factors known to nuclear medicine practitioners.
  • Intraarterial administration pretargeting can be applied to targets present in organs or tissues for which supply arteries are accessible.
  • Exemplary applications for intraarterial delivery aspects of the pretargeting methods of the present invention include treatment of liver tumors through hepatic artery administration, brain primary tumors and metastases through carotid artery administration, lung carcinomas through bronchial artery administration and kidney carcinomas through renal artery administration.
  • Intraarterial administration pretargeting can be conducted using chemotherapeutic drug, toxin and anti-tumor active agents as discussed below.
  • High potency drugs, lymphokines, such as IL-2 and tumor necrosis factor, drug/lymphokine-carrier-biotin molecules, biotinylated drugs/lymphokines, and drug/lymphokine/ toxin-loaded, biotin-derivitized liposomes are exemplary of active agents and/or dosage forms useful for the delivery thereof in the practice of this embodiment of the present invention.
  • a problem associated with solid tumor target sites is penetration of the therapeutic agent into the active site. If homogeneous
  • One method that may be employed to enhance target site penetration of ligand or anti-ligand conjugated to a targeting agent or anti-ligand or ligand conjugated to a therapeutic moiety is the administration of a permeability
  • permeability enhancing moieties of the present disclosure are enhancing moiety that induces or promotes vascular leakiness, disrupts cell-to-cell associations or similarly facilitates more homogeneous delivery of an administered conjugate to a target site characterized by a three dimensional cellular array.
  • inventions achieve their effects rapidly (e.g., within from about 30 seconds to about 1 hour following administration).
  • invention involves anti-ligand-targeting moiety conjugate administration (e.g., systemically, intra-arterially, locally via catheter or the like)
  • a permeability enhancing moiety e.g., an agent that induces gaps in the endothelium of venules through action on the permeability enhancing moiety
  • postcapillary venules such as histamine, serotonin or bradykinin; or through action on the entire capillary bed such as bacterial endotoxins; or the like
  • One method to achieve site specificity for the therapeutic agent conjugate administration is to administer that agent via catheter.
  • a clearing agent may be administered prior or subsequent to the permeability enhancing moiety, with prior administration preferred.
  • this aspect of the present invention also facilitates the delivery of higher molecular weight therapeutic agent-bearing conjugates to target sites characterized by a three dimensional cellular array.
  • one or more ligand or anti-ligand molecules as well as a plurality of therapeutic agent molecules may be conjugated to a polymer to form an entity of the following formula:
  • Therapeutic Agent 1-30 Polymer-Ligand or
  • arterial supply as discussed herein, involves intraarterial administration of permeability enhancing agent followed by such
  • therapeutic agent-polymer-ligand or -anti-ligand conjugate is administered.
  • An exemplary conjugate suitable for such delivery is a biotinylated
  • the permeability enhancing moiety may be administered prior to administration of the targeting moiety-ligand or -anti-ligand conjugate to facilitate more homogenous targeting of that
  • permeability enhancing moieties may also be employed in three-step pretargeting protocols of the present invention.
  • Preferred permeability enhancing moieties of the present invention induce gaps in the endothelium of venules through "specific” or “non-specific"
  • non-specific permeability enhancing moieties are those that act upon the entire capillary bed.
  • Exemplary permeability enhancing moieties of the non-specific type are bacterial endotoxins, metabolic inhibitors, drugs that alter microfilaments or microtubules and the like.
  • Specific permeability enhancing moieties act on a portion of the capillary bed (e.g., on the postcapillary venules, capillaries or the like).
  • Exemplary permeability enhancing agents of the specific type are histamine, serotonin, bradykinin, and the like.
  • Other permeability enhancing moieties are mannitol, tumor necrosis factor, nitric oxide, prostaglandin E2, leukotriene, leukokinin and the like.
  • enhanced delivery of a diagnostic or therapeutic substance, for example, into a target tumor mass can be achieved by systemically
  • a conjugate containing a targeting moiety, a member of a ligand/anti-ligand binding pair and an amount of a permeability enhancing agent sufficient to achieve disruption of cell-to-cell association within the tumor mass.
  • permeability enhancing moiety may also be locally administered, for example, in the practice of the local or intraarterial administration protocols described above.
  • Exemplary permeability enhancing moieties of the present invention include any substance capable of disrupting cell-to-cell association within a three dimensional target cell array.
  • moieties include trichothecenes, which are small molecule protein synthesis inhibitors, ionophores, membrane-active compounds (particularly inhibitors of microtubule or microfilament polymerization,
  • polymyxins and polyene antibiotics polymyxins and polyene antibiotics
  • cyclic toxin peptides cyclic toxin peptides
  • cytochalasins and combinations thereof.
  • cytotoxic compounds that are devoid of macrocyclic rings may also be suitable for use herein.
  • Small permeability enhancing moieties having a molecular weight less than or equal to about 2,000 daltons, and particularly those having a molecular weight less than or equal to 1,000 daltons, are also preferred. Such small permeability enhancing moieties are able to pass freely between adjacent cells whose cytoplasms are coupled via gap junctional complexes.
  • a systemic administration embodiment In a systemic administration embodiment, a
  • permeability enhancing moiety having a molecular weight of about 1,000 daltons or less is conjugated to a targeting moiety (preferably also having a ligand or an anti-ligand bound thereto) through a selectively cleavable covalent linkage.
  • a targeting moiety preferably also having a ligand or an anti-ligand bound thereto
  • the permeability enhancing moiety is released at the target cell, and is then distributed to adjacent cells through gap junctions. Free diffusion of the released moiety will increase the efficiency, for example, of tumor mass disruption.
  • carrier molecule includes, but is not limited to, a large protein or polymer capable of binding many permeability enhancing moieties to a single targeting moiety.
  • Preferred carrier molecules include albumin, dextran, hydroxy-propylmethacrylamide, poly-L-lysine and polyglutamate, all of which have a molecular weight in excess of 5,000 daltons.
  • Carrier molecules may also be produced through chemical syntheses or recombinant DNA
  • a carrier molecule generally is
  • permeability enhancing moieties to the carrier.
  • the "permeability enhancing moiety loaded” carrier is then either locally or intraarterially administered or systemically administered after being covalently attached to a targeting moiety (directly or through a linker) .
  • a permeability enhancing moiety which is a target cell inflammatory response mediator.
  • a preferred permeability enhancing moiety of this type is a substance that is capable of localizing at a target site and capable of mediating complement- dependent inflammation at the target site within a patient. Such moieties are recognized by their ability to mediate complement-dependent cytotoxicity in vitro with the recipient's serum. Permeability enhancing moieties may be monoclonal antibodies, polyclonal antibodies, chimeric antibodies, human or humanized antibodies or other moiety having the capabilities described above.
  • Permeability enhancing moieties of this type may also be administered as a conjugate with a vasoactive complement-derived peptide, such as a peptide derived from C2.
  • a vasoactive complement-derived peptide such as a peptide derived from C2.
  • Complement-derived peptides are linked to the permeability enhancing moieties in a manner permitting release of the complement-derived peptide at the target site, such as through a labile linkage (i.e., a Schiff base linkage).
  • a subset of antibodies such as mouse antibodies, human/mouse chimeric antibodies or humanized antibodies are used to induce target site-specific increases in vascular permeability.
  • These monoclonal antibodies mediate target cell lysis and/or target cell inflammatory responses in vitro which contribute to target site-specific vascular endothelial growth factor receptors.
  • the invention exploits one attribute of antibodies ⁇ possession of isotype- and/or subclass-specific functions.
  • murine IgG 3 and human/mouse chimeric antibodies having human IgG, or IgG 3 constant portions are generally considered superior to other immunoglobulins of murine or murine/human nature for mediating antibody-dependent cell-mediated
  • ADCC complement-dependent cellular toxicity
  • cytotoxicity must be capable of utilizing the serum complement native to the ultimate recipient having confirmed br potential target sites. Consequently, human antibodies are expected to be capable of utilizing human serum. Antibodies of other types are therefore tested for this ability when administration to human recipients is contemplated.
  • moieties known to mediate dependent cytotoxicity are NR-CO-04, a murine IgG 3 antibody directed to colon carcinoma; NR-LU-13, a murine/human chimeric antibody featuring a human constant region and murine variable region of NR-LU-10, an antibody directed to a 37-40 kilodalton pancarcinoma
  • NR-LU-03 an IgG 3 antibody generated by immune complex immunization with NR-LU-10
  • R24 an IgG 3 antibody directed against GD3 disclosed by Houghton et al., Proc. Natl. Acad. Sci (USA). 82: 1242-1246, 1985
  • antibody 3F8 a mouse IgG 3 directed against GD2
  • Some polymers useful in the practice of this permeability enhancing aspect of the present invention serve solely as a carrier for multiple therapeutic agents. Preferred polymers also direct the
  • renal excretion is preferred, especially for therapeutic protocols.
  • therapeutic agents e.g., radionuclides
  • Exemplary polymers for use in the practice of the present invention are excreted or which metabolites thereof are excreted through a renal pathway when administered to a mammalian recipient and that is capable of covalently or non-covalently binding to one or more drugs, anti-tumor agents, peptides, chelates, ligands, anti-ligands or other small molecules and imposing a renal route of excretion upon the
  • Such polymers include polar molecules having a molecular weight ranging from about 3 kD to about 70 kD).
  • dextran dextran
  • dextran derivatives including carboxymethyl dextran, anionic or polar derivatives thereof such as carboxymethy1-dextran, 3-mercapto-2-hydroxypropyl dextran and the like
  • hyaluronic acid inulin
  • polyaspartate polyglutamate
  • polyethyleneglycol polyethyleneglycol
  • dextran polymers preferably range between about 5 and about 15 kD in size, although larger moieties may also be used.
  • larger (from about 40 to about 70 kD) polymers are employed in accordance with the present invention, the rate of clearance of the conjugate from a recipient is slowed, but the renal pathway for excretion is
  • the moieties bound to the polymer exhibit increased bioavailability as a result of the increased circulation time thereof. Such bound molecules remain directed to renal excretion, however.
  • Exemplary dextran-containing conjugates of the present invention are dextran-biotin conjugates.
  • Chelate-biotin-dextran conjugates of the present invention are formed, for example, from oxidized dextran by conjugating biocytin hydrazide thereto followed by reaction with a chelate active ester, such as a N-hydroxy succinimidyl ester, a tetrafluorophenyl ester and the like.
  • chelate-biotin-dextran can be formed by reacting dextran hydrazide with the carboxy terminus of a chelate-biocytin conjugate.
  • the carboxy terminus has been derivitized to form an active ester, such as an N- hydroxysuccinimidyl ester or the like.
  • Biotin-dextran, lysine fixable (BDLF, available from Sigma Chemical Company, St. Louis, Missouri) is a preferred polymer-ligand for use in the practice of the present invention.
  • Active agents can be bound to dextran polymers characterized by short term serum stable linkages disposed between monomeric, dimeric, trimeric or other convenient unit thereof. Such conjugates are also preferably large (exhibiting a molecular weight ranging from about 40 to about 70 kD).
  • One advantage of the use of such polymers is that the molecules affixed thereto will exhibit an increased circulation time as well as decreased liver uptake. More
  • the circulation time of the bound molecules is dictated by the maintenance of polymeric structural integrity in vivo.
  • Depolymerization releases bound molecule-monomer, -dimer, -trimer or like moieties that are themselves rapidly cleared from the recipient's circulation, preferably via the renal pathway.
  • Depolymerization may be controlled in any order
  • appropriate circulation time to the small molecules bound thereto e.g., 1 to 3 hours for active agent in both the pretargeting and targeted, direct labeled protocols
  • linkages between dextran units that are enzymatically cleaved upon administration of enzyme after the passage of an appropriate amount of bound molecule circulation time.
  • groups such as esters, acetals, disulfides, thioacetals or the like are employed.
  • an ester linkage having a serum stability of 1-3 hours such as phenyl or
  • activated phenyl or phthalyl e.g., chloro-substituted, fluoro-substituted, multi-halogen-substituted, nitro-substituted or the like
  • a linkage is stable in serum for a time sufficient to facilitate localization of the molecules bound to the polymer to the target site.
  • the second approach includes the use of dextran units susceptible to cleavage by an administrable enzyme that is not found in large amounts in human serum.
  • exemplary enzymes useful in the practice of this aspect of the present invention include
  • dextranase alpha-amylase
  • pullulanase a bacterial alpha-1,6-polysaccharidase
  • the enzymes are administered by any convenient route in any convenient dosage form therefor.
  • Such enzymes are optionally conjugated or formed as fusion proteins with long circulating proteins, including albumin, immunoglobulins or portions thereof, and the like. In this manner, the administered enzymes remain in circulation for a time sufficient to effect
  • biotin/avidin ligand/anti-ligand pair can be described as follows: 1) (two-step) administer targeting moiety-biotin-therapeutic agent conjugate (or biotin-targeting moiety-therapeutic agent conjugate); and
  • a variation of this protocol involves administration of targeting moiety-biotin conjugate followed by administration of therapeutic agent-avidin or -streptavidin conjugate.
  • streptavidin-targeting moiety-therapeutic agent conjugate or therapeutic agent-avidin- or
  • streptavidin-targeting moiety conjugate or a
  • therapeutic agent-containing conjugate that also incorporates Antibody-(avidin or streptavidin) 2 or (Antibody) 2 -avidin or -streptavidin);
  • a variation of this protocol involves administration of a targeting moiety-avidin or -streptavidin conjugate, Antibody-(avidin or streptavidin)-, conjugate or
  • biotin-polymer-therapeutic agent conjugate to either cross-link the previously localized avidin or streptavidin or to be internalized into the cell as a result of the avidin or streptavidin cross-linking of previously localized biotin-containing conjugate.
  • cross-linking/internalization embodiments of the present invention are exemplified below with reference to the biotin/avidin ligand/anti-ligand pair, monoclonal antibody targeting moieties and trichothecene therapeutic agents; however, the embodiments are amenable to use with other
  • Exemplary simple trichothecenes useful in the practice of this and other aspects of the present invention are as follows:
  • R 1 is H, OH, , SH or L 1 , L 2 , L 3 , or L 4 ;
  • R 2 is H, OH, or ;
  • R 3 is H, OH, or ;
  • R 4 is H, or forms an epoxide group with R5 ;
  • R 5 is H, OH, , , SH,
  • L 3 , L 4 or forms an epoxide group with R4 ; and R 6 is H, OH, SH, OCH3 , SCH 3 , NH 2 , NHCH3 , N(CH 3 ) 2 . L 3 , or L4
  • R 1 , R 5 and R 6 is L 1 , L 2 , L 3 , or L 4 and further provided that R 5 and R 6 are not L 1 or L 2 and wherein y , , , or , and wherein
  • Y is O or NH
  • R 7 or R 7' is H or CH 3 ,
  • R 8 or R 8' is H or OH
  • n one to ten
  • n' is zero to ten.
  • Preferred simple trichothecenes are compounds
  • R 1 is L 1 or L 2 and Y is NH, R 7 and R 7 , are independently either H or CH 3 , R 8 and R 8 , are
  • n' is zero to ten
  • R 2 and R 3 are both ;
  • R 4 is H; and R 5 and R 6 are selected from the group consisting of H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 ;
  • R 1 is H, OH or SH; and R 5 and R 6 are selected from the group consisting of H, OH, SH, OCH 3 , SCH 3 ,
  • R 7 and R 7 are independently either H or CH 3 and n is one to ten;
  • R 1 is H, OH or SH; and R 5 or R 6 is selected from the group consisting of H, OH, SH, OCH 3 , SCH 3 ,
  • n is one to ten, and R 7 and R 7 , are independently either H or CH 3 ;
  • R 1 is L 3 or L 4 wherein R 7 and R 7 , and
  • R 5 and R 6 are selected from the group consisting of H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 .
  • trichothecene may be utilized in the protocols
  • trichothecene is characterized as follows: R 1 is L 1 and Y is NH, R 7 and R 7 , are both H, and n' is zero; R 4 ,
  • R 5 , R 6 are H; and R 2 and R 3 are both
  • Exemplary macrocyclic trichothecenes useful in the practice of this and other aspects of the present invention are as follows:
  • R' is selected from the group consisting of , ,
  • W and Z is L 1 , L 2 , L 3 , or L 4 , and further provided W and Z are not both L 1 , L 2 , L 3 , or L 4 wherein ,
  • Y is O or NH
  • R 7 or R 7' is H or CH 3 ,
  • R 8 or R 8' is H or OH
  • n one to ten
  • n' is zero to ten; and further provided that W can be an epoxide group between 2' and 3' and still further provided that W and Z can be independently either H, OH, or SH when W and Z are not L 1 , L 2 , L 3 , or L 4 ; R 1 is H, OH, or SH when Rl is not L 1 , L 2 , L 3 , or L4; and R 2 and R 3 are selected from the group consisting of, H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 , L 3 or L 4 .
  • Preferred macrocyclic trichothecenes are compounds wherein R' is
  • Such preferred compounds are derivatives of Roridin A. More preferred macrocyclic trichothecenes of this type are characterized as follows:
  • R 1 is L 1 , L 2 , L 3 , or L 4 wherein
  • Y is O or NH
  • R 7 and R 7' are independently either H or CH 3
  • R 8 and R 8 ' are independently either H or OH
  • n is one to ten
  • n' is zero to ten
  • R 2 and R 3 are selected from the group consisting of, H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 ; and W and Z are independently either H, OH, or SH;
  • R 1 and W are independently either H, OH, or SH;
  • R 2 and R 3 are selected from the group consisting of, H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 ; and
  • Z is L 1 , L 2 , L 3 , or L 4 and wherein
  • Y is 0 or NH
  • R7 or R 7' is H or CH 3 ,
  • R 8 or R 8' is H or OH
  • n one to ten
  • n' is zero to ten
  • R 1 and Z are H, SH, or OH;
  • R 2 and R 3 are selected from the group consisting of, H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 ; and W is either L 1 L 2 , L 3 , or
  • Y is 0 or NH
  • R 7 or R 7 is H or CH 3 ,
  • R 8 or R 8 is H or OH
  • n one to ten
  • n' is zero to ten
  • R 1 , W and Z are independently either SH or OH;
  • R 2 and R 3 are selected from the group consisting of H, OH, SH, OCH 3 , SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 , L 3 , or L 4 wherein
  • R 7 and R 7 are either H or CH 3 , and
  • n one to ten
  • R 2 and R 3 are not both simultaneously L 3 or L 4 .
  • these macrocyclic trichothecenes may be utilized in the protocols numbered 2 and 3 above as, for example, (Biotin) 18 . 20 -Dextran-2'-oxo Roridin A.
  • Prototypical Roridin A derivative macrocyclic trichothecenes may be characterized as follows: R 1 and R 2 are both H; R 3 is CH 3 ; W is L 1 , Y is NH, R 7 and R 7 , are both H, n' is zero; and Z is OH;
  • R 1 and R 2 are both H; R 3 is CH 3 ; W is ; and
  • Z is L 1 , Y is NH, R 7 and R 7 , are both H, n' is zero; and R 1 and R 2 are both H; R 3 is CH 3 ; W is L 3 , R 7 and R 7 , are both H, n is one; and Z is OH.
  • Acid labile linker technology e.g., hydrazone linkers, facilitate release of therapeutic agent in target cell endosomes and lysosomes (pH 3.5-5.5) where the released agent can exert its therapeutic effect (e.g., inhibition of protein synthesis).
  • Disulfide linkages also promote release of therapeutic agent in endosomes and lysosomes of the target cells.
  • Example XVIII Exemplary protocols for trichothecene-1inker preparation are discussed in Example XVIII. Exemplary protocols for trichothecene-antibody conjugation are set forth in Example XIX (A and B). Also, an
  • the rapid clearance of nontargeted therapeutic agent decreases the rapid clearance of nontargeted therapeutic agent
  • pretargeting methods of the present invention optionally include administration of short duration bone marrow
  • a protocol such as administration of
  • streptavidin-targeting moiety conjugate followed by administration of biotinylated cytokine, is also contemplated by the present invention.
  • pretargeting of anti-ligand serves to improve the performance of cytokine therapeutics by increasing the amount of cytokine localized to target cells.
  • Streptavidin-antibody conjugates generally exhibit pharraacokinetics similar to the native antibody and localize well to target cells, depending upon their construction.
  • Biotinylated cytokines retain a short in vivo half-life; however, cytokine may be localized to the target as a result of the affinity of biotin for avidin.
  • biotin-avidin experience a pH-dependent dissociation which occurs at a slow rate, thereby permitting a relatively constant, sustained release of cytokine at the target site over time.
  • cytokines complexed to target cells through biotin-avidin association are available for extraction and internalization by cells involved in cellular-mediated cytotoxicity.
  • a pre-formed antibody-streptavidin-biotin-cytokine preparation may also be employed in the practice of these methods of the present invention.
  • a three-step protocol of the present invention may also be employed to deliver a cytokine, such as IL-2, to a target site.
  • selectins include L-selectin, P-selectin and E-selectin.
  • cytokines stimulates cells, such as endothelial cells, to express selectins on the surfaces thereof.
  • Selectins bind to white blood cells and aid in delivering white blood cells where they are needed. Consequently, a protocol, such as administration of streptavidin- or avidin-targeting moiety conjugate followed by
  • pretargeting of anti-ligand serves to improve the performance of selectin therapeutics by increasing the amount of selectin localized to target cells. In this manner, the necessity of cytokine induction of
  • selectin expression is obviated by the localization and retention of selectin at a target cell population.
  • a three-step protocol may also be employed to deliver selectins to a target site.
  • Chemotherapeutic drugs also generally exhibit short in vivo half-lives at a therapeutically
  • a three-step protocol of the present invention may also be employed to deliver a chemotherapeutic drug, such as methotrexate, adriamycin, high potency adriamycin analogs,
  • trichothecenes such as esperamycins and calicheamycins, cytoxan, vinca alkaloids,
  • actinamycin D, taxol, taxotere or the like to a target site.
  • An additional aspect of the present invention is a method of eradicating target cells within a mammal, involving the steps of:
  • immunogenic molecule comprising an antibody, an antibody fragment or another targeting moiety which binds to the target cells and a member of a
  • step 5 administering an effector cell growth factor bound to a member of the ligand/anti-ligand pair that is complementary to the member administered in step 2.
  • the targeting moiety serves as a carrier to implant a strong predetermined antigenic signal on the target cells, ensuring that those target cells are
  • the engineered antigen bound to the target cells in vivo attracts subsequently administered presensitized effector cells to the target cells. In effect, this procedure constitutes an amplification of the tumor-associated antigen.
  • the ligand/anti-ligand pair conjugate components are employed both to deliver effector cell growth factor to the target site and to avoid toxicity associated with circulating effector cell growth factor by decoupling the slow targeting moiety localization step from delivery of the growth factor.
  • An alternative method for eradicating target cells within a mammal comprises administering to the mammal an immunogenic molecule comprising an antibody- or other targeting moiety-ligand/anti-ligand pair member-immunogen conjugate which binds to the target cells in vivo. Subsequently, the complementary member of the ligand/anti-ligand pair is administered while
  • an effector cell growth factor conjugated to an effector cell growth factor.
  • This in vivo embodiment may be employed when the immunogenic molecule is capable of inducing a therapeutic response in vivo.
  • the antigenic signal is not predetermined, but is sufficiently strong to recruit a therapeutically effective number of effector cells to the target site.
  • the binding of the immunogenic molecule to the target cells within the mammal induces a cytotoxic effector cell response directed at the target cells. Delivery of effector cell growth factor to the target site enhances the activity of the effector cells at that site.
  • effector cells are extracted from a mammal or recruited from within a mammal.
  • These effector cells are cells capable of being "sensitized” such that the sensitized effector cells recognize a particular antigen, and have a cytotoxic effect on target cells bearing that antigen.
  • the effector cells useful in the practice of this aspect of the present invention are cells
  • cytotoxic cells designated as “committed T-cells,” along with a variety of cytotoxic cells designated otherwise, including LAK cells, natural killer (NK) cells and other leukocytes and macrophages which could
  • the effector cells are cytotoxic T-lymphocytes, also called cytotoxic T-cells.
  • Effector cells can be extracted from the mammal to be treated by any suitable procedure for collecting those cells from the patient and separating the desired effector cells from other cell types or biological materials which may be simultaneously extracted. Conventional cell harvesting or
  • cytopheresis e.g., leukopheresis
  • the effector cell extraction procedure may be repeated to obtain a larger quantity of effector cells.
  • the mammal may be subjected to one leukapheresis procedure per day for one or more days (e.g., for five days to collect a large number of effector cells such as T-cells).
  • the effector cell growth factor used may vary according to the type of effector cells employed, and may be any growth factor which stimulates the
  • cytotoxic effector cells are produced.
  • suitable growth factors are monokines for monocytes and lymphokines (e.g., IL-2) for lymphocytes such as T-lymphocytes).
  • the first immunogenic molecule (that with which the effector cells are incubated) may be the same as or different from the second immunogenic molecule (that which is administered to the mammal), provided that the two immunogenic molecules have at least one moiety in common.
  • the proviso ensures that effector cells sensitized in vitro in the presence of the first immunogenic molecule will recognize the second
  • the targeting moiety the ligand/anti-ligand pair member or the combination thereof is sufficiently immunogenic (i.e., is capable of inducing
  • the targeting moiety-ligand/anti-ligand pair member conjugate may serve as the first as well as the second immunogenic molecule.
  • a sufficiently immunogenic portion of the conjugate may be used as the first immunogenic molecule with the entire conjugate serving as the second immunogenic molecule.
  • various compounds may be attached to the targeting moiety or ligand/anti-ligand pair member to increase the immunogenicity thereof.
  • the immunogen-targeting moiety-ligand/anti-ligand pair member conjugate is administered as the second immunogenic molecule.
  • haptens or polypeptides which are "foreign" to the intended mammalian recipient (such as antigens, mitogens, other foreign proteins or fragments thereof, peptides that activate cytotoxic T-cells or the like).
  • attachment of the immunogen to the targeting moiety or ligand/anti-ligand pair member through a spacer may increase the immunogenicity of the linked first immunogenic molecule.
  • Haptens that are useful in the practice of the present invention include benzoate groups, nitrophenol groups, other small molecules such as acetic acid and derivatives thereof, penicillinic acid and derivatives thereof, sulfanilic acid derivatives, hexoseamines, ribonucleotides, ribonucleosides, isocyanates,
  • haptens are benzoic acid, dinitro-chlorobenzene, dinitrofluorobenzene, picrylchloride, p-aminobenzenearsonate, p-azo-benzenearsonate, acetic acid, dinitrophenol, trinitrophenol, trinitrophenol-epsilon-aminocaproic acid, 3-iodo-4-hydroxyl-5-nitrophenylacetic acid, p-hydroxyphenylacetic acid, p-sulfanilic acid, 2,4-diisocyanate, fluorescein isothiocyanate, N-acetyl-glucosamine and the like.
  • T-cell mitogens such as the proteins pokeweed antiviral protein and phytohemagglutinin also may be used.
  • streptavidin thereby producing sensitized effector cells recognizing avidin or streptavidin or a moiety thereof;
  • a three-step pretargeting approach may be designed for the practice of this aspect- of the present invention.
  • steps 3 and 4 may be conducted in reverse order.
  • a clearing agent may be administered between steps 4 and 5.
  • a three-step pretargeting protocol may be employed.
  • a three-step pretargeting protocol may also be
  • HLA matching is conducted in
  • organ transplants selecting candidates for organ transplants.
  • Persons with more similar HLA i.e., twins, siblings, parents and the like
  • organ transplant donors are the most preferably organ transplant donors, because the donated organ is less likely to be recognized as non-self by the recipient.
  • HLA antijgens are relatively rare in humans, i.e. f are infrequently present in the human HLA display.
  • HLA-B7 antigen is present on only 5% of the human population. Consequently, HLA-B7 is recognized as non-self in 95% of humans.
  • Such relatively rare antigens can therefore be
  • the target will be recognized as non-self by the recipient, thereby recruiting the recipient's immune system to combat the target. Since some targets (e.g., certain tumors) are poorly recognized as non-self, the presence of a recognized non-self antigen at that site will improve the recipient's immune system response to the target.
  • HLA antigen that is not present in a recipient can be delivered to a target site by conjugation with a targeting moiety, as part of a fusion protein with a targeting moiety or as part of a conjugate or fusion protein with a ligand or an anti-ligand in the two-step or three- step pretargeting protocols described herein .
  • Radiometal labeling of an exemplary chelate-biotin conjugate is illustrated below.
  • the spacer group "X" permits the biotin portion of the conjugate to be sterically available for avidin binding.
  • R 1 " is a carboxylic acid substituent (for instance, CH 2 COOH)
  • the conjugate exhibits improved water solubility, and further directs in vivo excretion of the radiolabeled biotin conjugate toward renal rather than hepatobiliary clearance.
  • N- ⁇ -Cbz-N- ⁇ -t-BOC protected lysine was converted to the succinimidyl ester with NHS and DCC, and then condensed with aspartic acid ⁇ - -t-butyl ester.
  • the resultant dipeptide was activated with NHS and DCC, and then condensed with glycine t-butyl ester.
  • the Cbz group was removed by hydrogenolysis, and the amine was acylated using tetrahydropyranyl mercaptoacetic acid succinimidyl ester, yielding S-(tetrahydropyranyl)-mercaptoacetyl-lysine.
  • the chelate-biotin conjugate of Example I was radiolabeled with either 99m Tc pertechnetate or 186 Re perrhenate. Briefly, 99m Tc pertechnetate was reduced with stannous chloride in the presence of sodium
  • 186 Re perrhenate was spiked with cold ammonium perrhenate, reduced with stannous chloride, and complexed with citrate.
  • the chelate-biotin conjugate of Example I was added and heated to 90°C for 30 min at a pH of about 2 to 3.
  • the solution was neutralized to a pH of about 6 to about 8, and yielded an N 3 S-coordinated 18o Re-chelate-biotin conjugate.
  • C-18 HPLC gradient elution using 5-60% acetonitrile in 1% acetic acid resulted in radiochemical yields of 85-90%. Subsequent purification over a C-18 reverse phase hydrophobic column yielded material of 99% purity.
  • a 99m Tc-biotin conjugate was subjected to various chemical challenge conditions. Briefly, 99m Tc-chelate-biotin conjugates were combined with avidin and passed over a 5 cm size exclusion gel filtration column. The radiolabeled biotin-avidin complexes were subjected to various chemical challenges (see Table 1), and the incubation mixtures were centrifuged through a size exclusion filter. The percent of radioactivity retained (indicating avidin-biotin-associated
  • each radiolabeled biotin conjugate was incubated at about 50 ⁇ g/ml with serum; upon completion of the incubation, the samples were
  • Each radiolabeled biotin conjugate was further examined using a competitive biotin binding assay. Briefly, solutions containing varying ratios of D-biotin to radiolabeled biotin conjugate were combined with limiting avidin at a constant total biotin: avidin ratio. Avidin binding of each radiolabeled biotin conjugate was determined by ITLC, and was compared to the theoretical maximum stoichiometric binding (as determined by the HABA spectrophotometric assay of Green, Biochem. J. 94:23c-24c, 1965). No significant difference in avidin binding was observed between each radiolabeled biotin conjugate and D-biotin.
  • Example IV Example IV
  • antibody pretargeting protocol involves: (i) prelocalization of biotinylated monoclonal antibody; (ii) administration of avidin for formation of a "sandwich" at the target site and for clearance of residual circulating biotinylated
  • Biotinylated NR-LU-10 was prepared according to either of the following procedures. The first procedure involved derivitization of antibody via lysine s-amino groups. NR-LU-10 was radioiodinated at tyrosines using chloramine T and either 125 I or 131 I sodium iodide. The radioiodinated antibody (5-10 mg/ml) was then biotinylated using biotinamido caproate NHS ester in carbonate buffer, pH 8.5, containing 5% DMSO, according to the scheme below.
  • biotin antibody ratio of 10:1.
  • NR-LU-10 was biotinylated using thiol groups generated by reduction of cystines.
  • NR-LU-10 was radioiodinated using p-aryltin phenylate NHS ester (PIP-NHS) and either 125 I or 131 I sodium iodide. Radioiodinated NR-LU-10 was incubated with 25 mM dithiothreitol and purified using size exclusion chromatography. The reduced antibody (containing free thiol groups) was then reacted with a 10- to 100-fold molar excess of N-iodoacetyl-n'-biotinyl hexylene diamine in phosphate-buffered saline (PBS), pH 7.5, containing 5% DMSO (v/v). TABLE 3
  • PBS phosphate-buffered saline
  • biotin:antibody molar offering only 6 biotins per antibody were incorporated. No significant impact on immunoreactivity was observed.
  • Biotinylated antibody (lysine) had an apparent molecular weight of 160 kD, while
  • biotinylated antibody lysine
  • biotinylated antibody thiol
  • radiolabeled streptavidin migrates as a 60 kD tetramer.
  • radiolabeled streptavidin was combined with 50 ⁇ g/ml biotinylated antibody (analogous to "sandwiching" conditions in vivo), both antibody species formed large molecular weight complexes.
  • biotinylated antibody (thiol)-streptavidin complex moved from the stacking gel into the resolving gel, indicating a decreased molecular weight as compared to the biotinylated antibody (lysine)-streptavidin complex.
  • Radioiodinated biotinylated NR-LU-10 (lysine or thiol) was intravenously administered to non-tumored nude mice at a dose of 100 ⁇ g.
  • mice were intravenously injected with either saline or 400 ⁇ g of avidin. With saline
  • blood clearances for both biotinylated antibody species were biphasic and similar to the clearance of native NR-LU-10 antibody.
  • biotinylated antibody lysine 2 h post-avidin or post-saline administration were performed.
  • Avidin administration significantly reduced the level of biotinylated antibody in the blood (see Figure 1), and increased the level of biotinylated antibody in the liver and spleen. Kidney levels of biotinylated antibody were similar.
  • 18-22 g female nude mice were implanted subcutaneously with LS-180 human colon tumor xenografts, yielding 100-200 mg tumors within 10 days of implantation.
  • NR-LU-10 antibody (MW ⁇ 150 kD) was radiolabeled-with 125 I/Chloramine T and biotinylated via lysine residues (as described in Example IV.A, above).
  • Group 1 Time 0, inject 100 ⁇ g 125 I-labeled
  • Group 2 Time 0, inject 400 ⁇ g 131 I-labeled avidin (control) Time 2 h, inject 60 ⁇ g 186 Re-chelate- biotin conjugate
  • Group 3 Time 0, inject 60 ⁇ g 186 Re-chelate- (control) biotin conjugate
  • the three radiolabels employed in this protocol are capable of detection in the presence of each other. It is also noteworthy that the sizes of the three elements involved are logarithmically different ⁇ antibody ⁇ 150,000-; avidin ⁇ 66,000; and biotin ⁇
  • biotinylated antibody and avidin on blood clearance of the 186 Re-chelate-biotin conjugate was examined; blood clearance was similar in the presence or absence of biotinylated antibody and avidin. Further, antibody immunoreactivity was found to be uncorapromised by biotinylation at the level tested.
  • Fig. 1 At 25 h, about 350 pmol/g biotinylated antibody was present at the tumor; at 32 h the level was about 300 pmol/g; at 48 h, about 200 pmol/g; and at 120 h, about 100 pmol/g. Avidin uptake at the same time points was about 250, 150, 50 and 0 pmol/g, respectively. From the same experiment, tumor to blood ratios were determined for biotinylated antibody and for avidin. From 32 h to 120 h, the ratios of tumor to blood were very similar.
  • Fig. 1 a 10-fold reduction in blood pool antibody concentration was noted (Fig. 1), resulting in a sharp increase in tumor to blood ratios.
  • Avidin is cleared rapidly, with greater than 90% of the injected dose cleared from the blood within 1 hour after administration.
  • the Re-186-biotin chelate is also very rapidly cleared, with greater than 99% of the injected dose cleared from the blood by 1 hour after administration.
  • the three-step pretargeting protocol (described for Group 1, above) was then examined. More
  • tumor uptake of the 186 Re-chelate-biotin conjugate in the presence or absence of biotinylated antibody and avidin was determined.
  • the 186 Re-chelate- biotin conjugate displayed a slight peak 2 h post-injection, which was substantially cleared from the tumor by about 5 h.
  • the 186 Re-chelate-biotin conjugate reached a peak in tumor approximately 7 times greater than that observed in the absence of biotinylated antibody and avidin.
  • T:B ⁇ 8 at 30 h; ⁇ 15 at 100 h; ⁇ 35 at 140 h).
  • Tumor uptake of the 186 Re-chelate-biotin conjugate has further been shown to be dependent on the dose of biotinylated antibody administered.
  • biotinylated antibody about 200 pmol/g of 186 Re-chelate-biotin conjugate was present at the tumor at 2 h after administration; at 50 ⁇ g antibody, about 500 pmol/g of 1B6 Re-chelate-biotin conjugate; and at 100 ⁇ g antibody, about 1,300 pmol/g of 186 Re-chelate-biotin conjugate.
  • the biodistribution of 111 In-labeled-biotin derivatives varies greatly with structural changes in the chelate and the conjugating group. Similar structural changes may affect the biodistribution of technetium- and rhenium-biotin conjugates.
  • a neutral MAMA chelate-biotin conjugate is prepared according to the following scheme.
  • the resultant chelate-biotin conjugate shows superior kidney excretion. Although the net overall charge of the conjugate is neutral, the polycarboxylate nature of the molecule generates regions of hydrophilicity and hydrophobicity. By altering the number and nature of the carboxylate groups within the conjugate, excretion may be shifted from kidney to
  • anionic compounds are generally cleared through the GI system.
  • Conjugates containing polylysine may also exhibit beneficial biodistribution properties. With whole antibodies, derivitization with polylysine may skew the biodistribution of conjugate toward liver uptake. In contrast, derivitization of Fab fragments with polylysine results in lower levels of both liver and kidney uptake; blood clearance of these conjugates is similar to that of Fab covalently linked to chelate.
  • An exemplary polylysine derivitized chelate-biotin conjugate is illustrated below.
  • polylysine derivatives are preferably succinylated following biotinylation.
  • Polylysine derivatives offer the further advantages of: (1) increasing the specific activity of the radiometal-chelate-biotin conjugate; (2) permitting control of rate and route of blood clearance by varying the molecular weight of the polylysine polymer; and (3) increasing the circulation half-life of the conjugate for optimal tumor
  • poly-L-lysine is acylated according to standard amino group acylation procedures (aqueous bicarbonate buffer, pH 8, added biotin-NHS ester, followed by chelate NHS ester).
  • the number of biotins attached to polylysine is determined by the HABA assay. Spectrophotometric titration is used to assess the extent of amino group derivitization.
  • conjugate is characterized by size exclusion.
  • linkers that are cleaved by enzymes present in normal tissue but deficient or absent in tumor tissue can increase tumor retention.
  • the kidney has high levels of ⁇ -glutamyl transferase; other normal tissues exhibit in vivo cleavage of ⁇ -glutamyl prodrugs.
  • tumors are generally deficient in enzyme peptidases.
  • the glutamyl-linked biotin conjugate depicted below is cleaved in normal tissue and retained in the tumor.
  • biocytin is condensed with N-t-BOC-(O-sulfonate or O-glucose) serine NHS ester to give N-t-BOC-(O-sulfonate or O-glucose) serine biocytinamide.
  • N-t-BOC-(O-sulfonate or O-glucose) serine NHS ester to give N-t-BOC-(O-sulfonate or O-glucose) serine biocytinamide.
  • ligand-amidoserine O-sulfonate or O-glucose
  • Radioiodinated biotin derivatives prepared by exposure of poly-L-lysine to excess NHS-LC-biotin and then to Bolton-Hunter N-hydroxysuccinimide esters in
  • radioiodinated biotin according to the present invention provides certain advantages. First, the radioiodobiotin derivative is a low
  • the disclosed methods for preparation involve a single step and eliminate the need for a purification step.
  • "X" may be any radiohalogen, including 1 25 I, 131 I, 123 I, 211 At and the like.
  • tributyltin intermediate Water soluble carbodiimide was used in the above-depicted reaction, since the NHS ester 1 formed intractable mixtures with DCU.
  • the NHS ester was not compatible with chromatography; it was insoluble in organic and aqueous solvents and did not react with biocytin in DMF or in buffered aqueous acetonitrile.
  • the reaction between 1 and biocytin or 5-(biotinamido) pentylamine was sensitive to base.
  • the reaction of 1 and biocytin or the pentylamine was performed in the presence of triethylamine in hot DMSO, formation of more than one biotinylated product resulted.
  • the reaction was extremely clean and complete when a suspension of 1 and biocytin (4 mg/ml) or the pentylamine (4 mg/ml) was heated in DMSO at 117°C for about 5 to about 10 min.
  • the resultant 125 I-biotin derivatives were obtained in 94% radiochemical yield.
  • the radioiodinated products may be purified using C-18 HPLC and a reverse phase hydrophobic column.
  • biodistribution properties of 2 provided enhanced whole body clearance of radioisotope and indicate the advantageous use of 2 within pretargeting protocols.
  • radiohalogenated biotin compounds are amenable to the same types of modifications described in Example VI above for 186 Re-chelate-biotin conjugates.
  • the following PIP-polylysine-biotin molecule is made by trace labeling polylysine with 125 I-PIP, followed by extensive biotinylation of the polylysine.
  • Certain antibodies have available for reaction endogenous sulfhydryl groups. If the antibody to be biotinylated contains endogenous sulfhydryl groups, such antibody is reacted with N-iodoacetyl-n'-biotinyl hexylene diamine (as described in Example IV.A., above).
  • N-iodoacetyl-n'-biotinyl hexylene diamine as described in Example IV.A., above.
  • DTT reducing agent
  • one or more sulfhydryl groups are attached to a targeting moiety through the use of chemical compounds or linkers that contain a terminal sulfhydryl group.
  • An exemplary compound for this purpose is iminothiolane.
  • a NR-LU-13-avidin conjugate is prepared as
  • SMCC-derived avidin is then incubated with NR-LU-13 in a 1:1 molar ratio at pH 8.5 for 16 h. Unreacted NR-LU-13 and SMCC-derived avidin are removed from the mixture using preparative size exclusion HPLC. Two conjugates are obtained as products ⁇ the desired 1 : 1 NR-LU-13 -avidin conjugate as the major product; and an incompletely
  • a 99m Tc-chelate-biotin conjugate is prepared as in Example II, above.
  • the NR-LU-13-avidin conjugate is administered to a recipient and allowed to clear from the circulation.
  • One of ordinary skill in the art of radioimmunoscintigraphy is readily able to determine the optimal time for NR-LU-13-avidin conjugate tumor localization and clearance from the circulation.
  • the 99m Tc-chelate-biotin conjugate is administered to the recipient. Because the 99m Tc- chelate-biotin conjugate has a molecular weight of ⁇ 1,000, crosslinking of NR-LU-13-avidin molecules on the surface of the tumor cells is dramatically reduced or eliminated. As a result, the 99m Tc diagnostic agent is retained at the tumor cell surface for an extended period of time. Accordingly, detection of the
  • diagnostic agent by imaging techniques is optimized; further, a lower dose of radioisotope provides an image comparable to that resulting from the typical three-step pretargeting protocol.
  • NR-LU-13-avidin clearance of NR-LU-13-avidin from the circulation may be accelerated by plasmapheresis in combination with a biotin affinity column.
  • a biotin affinity column Through use of such column, circulating NR-LU-13-avidin will be retained extracorporeally, and the recipient's immune system exposure to a large, proteinaceous immunogen (i.e., avidin) is minimized.
  • an example of an extracorporeal clearance methodology may include the following steps:
  • withdrawing blood from the recipient by, for example,
  • NR-LU-13-avidin Clearance of NR-LU-13-avidin is also facilitated by administration of a particulate-type clearing agent (e.g., a polymeric particle having a plurality of biotin molecules bound thereto).
  • a particulate clearing agent e.g., a polymeric particle having a plurality of biotin molecules bound thereto.
  • Such a particulate clearing agent preferably constitutes a biodegradable polymeric carrier having a plurality of biotin
  • Particulate clearing agents of the present invention exhibit the capability of binding to circulating administered conjugate and removing that conjugate from the recipient.
  • Particulate clearing agents of this aspect of the present invention may be of any configuration suitable for this purpose.
  • Preferred particulate clearing agents exhibit one or more of the following
  • microparticulate e.g., from about 0.5
  • micrometers to about 100 micrometers in diameter, with from about 0.5 to about 2 micrometers more preferred), free flowing powder structure;
  • biodegradable structure designed to biodegrade over a period of time between from about 3 to about 180 days, with from about 10 to about 21 days more preferred, or non-biodegradable structure;
  • binding moieties preferably, the complementary member of the ligand/anti-ligand pair
  • the total molar binding capacity of the particulate clearing agents depends upon the particle size selected and the ligand or anti-ligand substitution ratip.
  • the binding moieties are capable of coupling to the surface structure of the particulate dosage form through covalent or non-covalent modalities as set forth herein to provide accessible ligand or anti-ligand for binding to its previously administered circulating binding pair member.
  • Preferable particulate clearing agents of the present invention are biodegradable or non-biodegradable microparticulates. More preferably, the particulate clearing agents are formed of a polymer containing matrix that biodegrades by random,
  • Polymers derived from the condensation of alpha hydroxycarboxylic acids and related lactones are more preferred for use in the present invention.
  • thermoplastic polyesters e.g., polylactide or polyglycolide
  • a copolymer of lactide and glycolide components such as poly (lactide-co-glycolide).
  • An exemplary structure, a random poly (DL-lactide-co-glycolide), is shown below, with the values of x and y being manipulable by a practitioner in the art to achieve desirable microparticulate properties.

Abstract

Methods, compounds, compositions and kits that relate to pretargeted delivery of diagnostic and therapeutic agents are disclosed. In particular, methods for radiometal labeling of biotin and for improved radiohalogenation of biotin, as well as related compounds, are described. Also, clearing agents, anti-ligand-targeting moiety conjugates, target cell retention enhancing moieties and additional methods are discussed.

Description

PRETARGETING METHODS AND COMPOUNDS Technical Field
The present invention relates to methods,
compounds, compositions and kits useful for delivering to a target site a targeting moiety that is conjugated to one member of a ligand/anti-ligand pair. After localization and clearance of the targeting moiety conjugate, direct or indirect binding of a diagnostic or therapeutic agent conjugate at the target site occurs. Methods for radiometal labeling of biotin and for improved radiohalogenation of biotin, as well as the related compounds, are also disclosed. Also, clearing agents, anti-ligand-targeting moiety
conjugates, target cell retention enhancing moieties and additional methods are set forth. Background of the Invention
Conventional cancer therapy is plagued by two problems. The generally attainable targeting ratio (ratio of administered dose localizing to tumor versus administered dose circulating in blood or ratio of administered dose localizing to tumor versus
administered dose migrating to bone marrow) is low. Also, the absolute dose of radiation or therapeutic agent delivered to the tumor is insufficient in many cases to elicit a significant tumor response.
Improvement in targeting ratio or absolute dose to tumor is sought.
Summary of the Invention
The present invention is directed to diagnostic and therapeutic pretargeting methods, moieties useful therein and methods of making those moieties. Such pretargeting methods are characterized by an improved targeting ratio or increased absolute dose to the target cell sites in comparison to conventional cancer therapy.
The present invention describes chelate-biotin compounds and radiohalogenated biotin compounds useful in diagnostic and therapeutic pretargeting methods.
The present invention also provides targeting moiety-ligand, such as biotin, compounds useful in diagnostic and therapeutic pretargeting methods. Selection of moieties and methodologies used to enhance
internalization (of chemotherapeutic drugs, for example) or to enhance retention at the target cell surface (of radionuclides, for example) is also discussed.
In addition, the present invention provides targeting moiety-anti-ligand, such as avidin or streptavidin, compounds useful in diagnostic and therapeutic pretargeting methods. Other ligand-anti-ligand systems including the zinc finger protein-dsDNA fragment binding pair are also contemplated.
Preparation and purification of such anti-ligand-targeting moiety compounds are also discussed.
The present invention also provides clearing agents to facilitate the removal of circulating targeting moiety-ligand (two-step) or targeting moiety-anti-ligand (two-step) or anti-ligand (three-step) from the mammalian recipient. Preferred
clearing agents are classifiable as galactose-based and non-galactose-based. Within each category, preferable clearing agents are polymeric or protein based. Particulate agents, extracorporeal procedures and in vivo devices are also contemplated for use in the practice of the present invention.
Also, the present invention is directed to methods using streptavidin as an anti-ligand to enhance retention of radionuclide at target cell sites, with pretargeting protocols constituting one such method. More specifically, these embodiments of the present invention involve either (1) targeting moiety-streptavidin-radionuclide (with the radionuclide bound to streptavidin directly or through a chelate or linker), as well as (2) targeting moiety-biotin administered prior to streptavidin-radionuclide, or (3) biotin-radionuclide bound to a pretargeted
streptavidin containing molecule.
The present invention further provides
pretargeting methods employing intraarterial or other local administration of the therapeutic moiety-containing molecule to achieve greater localization thereof to artery-supplied target cell populations. Other methods of the present invention involve
administration of short duration bone marrow
protecting agents or vascular permeability enhancing agents prior to radionuclide-ligand molecule or radionuclide-anti-ligand molecule administration.
Further, monovalent targeting moieties, such as Fv or Fab antibody fragments, are useful in the inventive pretargeting methods. Delivery of other, non-radioactive therapeutic agents, such as
chemotherapeutic drugs, anti-tumor agents (e.g., cytokines) and toxins, to target cells using the pretargeting methods of the present invention is also described.
Brief Description of the Drawings
Figure 1 illustrates blood clearance of
biotinylated antibody following intravenous
administration of avidin.
Figure 2 depicts radiorhenium tumor uptake in a three-step pretargeting protocol, as compared to administration of radiolabeled antibody (conventional means involving antibody that is covalently linked to chelated radiorhenium).
Figure 3 depicts the tumor uptake profile of NR-LU-10-streptavidin conjugate (LU-10-StrAv) in comparison to a control profile of native NR-LU-10 whole antibody.
Figure 4 depicts the tumor uptake and blood clearance profiles of NR-LU-10-streptavidin conjugate.
Figure 5 depicts the rapid clearance from the blood of asialoorosomucoid in comparison with
orosomucoid in terms of percent injected dose of I-125-labeled protein.
Figure 6 depicts the 5 minute limited
biodistribution of asialoorosomucoid in comparison with orosomucoid in terms of percent injected dose of I-125-labeled protein.
Figure 7 depicts NR-LU-10-streptavidin conjugate blood clearance upon administration of three controls (O,●,■) and two doses of a clearing agent (
Figure imgf000006_0001
,⃞) at 25 hours post-conjugate administration.
Figure 8 shows limited biodistribution data for LU-10-StrAv conjugate upon administration of three controls (Groups 1, 2 and 5) and two doses of clearing agent (Groups 3 and 4) at two hours post-clearing agent administration.
Figure 9 depicts NR-LU-10-streptavidin conjugate serum biotin binding capability at 2 hours post-clearing agent administration.
Figure 10 depicts NR-LU-10-streptavidin conjugate blood clearance over time upon administration of a control (O) and three doses of a clearing agent (∇, Δ,⃞) at 24 hours post-conjugate administration.
Figure 11A depicts the blood clearance of LU-10-StrAv conjugate upon administration of a control (PBS) and three doses (50, 20 and 10 μg) of clearing agent at two hours post-clearing agent administration.
Figure 11B depicts LU-10-StrAv conjugate serum biotin binding capability upon administration of a control (PBS) and three doses (50, 20 and 10 μg) of clearing agent at two hours post-clearing agent administration. Figure 12 depicts the prolonged tumor retention of NR-LU-10-streptavidin conjugate (▲ ) relative to NR-LU-10 whole antibody (Δ ) over time.
Figure 13 depicts the prolonged liver retention of a pre-formed complex of NR-LU-10-biotin ( O ;
chloramine T labeled with 1-125) complexed with streptavidin ( ● ; PIP-I-131 labeled).
Figure 14 depicts the prolonged liver retention of Biotin-PIP-I-131 label relative to the streptavidin-NR-LU-10-(PIP-I-125) label.
Figure 15A depicts tumor uptake for increasing doses of PIP-Biocytin in terms of %ID/G.
Figure 15B depicts tumor uptake for increasing doses of PIP-Biocytin over time in terms of pMOL/G.
Figure 16A depicts tumor versus blood localization of a 0.5 μg dose of PIP-Biocytin over time in terms of %ID/G.
Figure 16B depicts tumor versus blood localization of a 0.5 μg dose of PIP-Biocytin in terms of % ID.
Figure 17A depicts tumor uptake of LU-10-StrAv and PIP-Biocytin over time in terms of %ID/G.
Figure 17B depicts blood clearance of LU-10-StrAv and PIP-Biocytin over time in terms of %ID/G.
Figure 18 depicts PIP-Biocytin:LU-10-StrAv ratio in tumor and blood over time.
Detailed Description of the Invention
Prior to setting forth the invention, it may be helpful to set forth definitions of certain terms to be used within the disclosure.
Targeting moiety: A molecule that binds to a defined population of cells. The targeting moiety may bind a receptor, an oligonucleotide, an enzymatic substrate, an antigenic determinant, or other binding site present on or in the target cell population.
Antibody is used throughout the specification as a prototypical example of a targeting moiety. Tumor is used as a prototypical example of a target in
describing the present invention.
Ligand/anti-ljgand pair: A complementary/anti-complementary set of molecules that demonstrate specific binding, generally of relatively high
affinity. Exemplary ligand/anti-ligand pairs include zinc finger protein/dsDNA fragment, enzyme/inhibitor, hapten/antibody, lectin/carbohydrate, ligand/receptor, and biotin/avidin. Biotin/avidin is used throughout the specification as a prototypical example of a ligand/anti-ligand pair.
Anti-ligand: As defined herein, an "anti-ligand" demonstrates high affinity, and preferably,
multivalent binding of the complementary ligand.
Preferably, the anti-ligand is large enough to avoid rapid renal clearance, and contains sufficient
multivalency to accomplish crosslinking and
aggregation of targeting moiety-ligand conjugates. Univalent anti-ligands are also contemplated by the present invention. Anti-ligands of the present invention may exhibit or be derivitized to exhibit structural features that direct the uptake thereof, e.g., galactose residues that direct liver uptake. Avidin and streptavidin are used herein as
prototypical anti-ligands.
Avidin: As defined herein, "avidin" includes avidin, streptavidin and derivatives and analogs thereof that are capable of high affinity, multivalent or univalent binding of biotin.
Ligand: As defined herein, a "ligand" is a relatively small, soluble molecule that exhibits rapid serum, blood and/or whole body clearance when
administered intravenously in an animal or human.
Biotin is used as the prototypical ligand.
Active Agent: A diagnostic or therapeutic agent
("the payload"), including radionuclides, drugs, anti-tumor agents, toxins and the like. Radionuclide therapeutic agents are used as prototypical active agents.
NxSy Chelates: As defined herein, the term "NxSy chelates" includes bifunctional chelators that are capable of (i) coordinately binding a metal or
radiometal and (ii) covalently attaching to a
targeting moiety, ligand or anti-ligand. Particularly preferred NxSy chelates have N2S2 and N3S cores.
Exemplary NxSy chelates are described in Fritzberg et al., Proc. Natl. Acad. Sci. USA 85:4024-29. 1988; in Weber et al., Bioconi. Chem. 1:431-37, 1990; and in the references cited therein, for instance.
Pretargeting: As defined herein, pretargeting involves target site localization of a targeting moiety that is conjugated with one member of a
ligand/anti-ligand pair; after a time period
sufficient for optimal target-to-non-target
accumulation of this targeting moiety conjugate, active agent conjugated to the opposite member of the ligand/anti-ligand pair is administered and is bound (directly or indirectly) to the targeting moiety conjugate at the target site (two-step pretargeting). Three-step and other related methods described herein are also encompassed.
Clearing Agent: An agent capable of binding, complexing or otherwise associating with an
administered moiety (e.g., kargeting moiety-ligand, targeting moiety-anti-ligand or anti-ligand alone) present in the recipient's circulation, thereby facilitating circulating moiety clearance from the recipient's body, removal from blood circulation, or inactivation thereof in circulation. The clearing agent is preferably characterized by physical
properties, such as size, charge, configuration or a combination thereof, that limit clearing agent access to the population of target cells recognized by a targeting moiety used in the same treatment protocol as the clearing agent.
Target Cell Retention: The amount of time that a radionuclide or other therapeutic agent remains at the target cell surface or within the target cell.
Catabolism of conjugates or molecules containing such therapeutic agents appears to be primarily responsible for the loss of target cell retention.
Conjugate: A conjugate encompasses chemical conjugates (covalently or non-covalently bound), fusion proteins and the like.
Permeability Enhancing Moiety: An agent capable of increasing the permeability at a target site characterized by a three dimensional cellular matrix. Exemplary permeability enhancing moieties function by one or more of the following mechanisms: inducing gaps in the endothelium of venules through action on the postcapillary bed; inducing such gaps through action on the entire capillary bed; disrupting cell-to-cell associations; mediating target cell
inflammatory responses; or the like.
Intercellular Junction: An area of interacting adjacent plasma membranes. Intercellular junctions can be categorized functionally into: (1) adhering junctions that hold cells tightly together (for example, desmosomes); (2) impermeable junctions that hold cells tightly together and prevent leakage of molecules between cells (i.e., tight junctions); and (3) communicating junctions that mediate passage of small molecules between adjacent cells (for instance, gap junctions).
Immunogen: A substance which is capable, under appropriate conditions, of inducing a specific immune response and of reacting with the products of that response (e.g., a specific antibody, specifically sensitized T-lymphocytes or both). Hapten Immunogen: A specific protein-free
substance which has a chemical configuration such that it can interact with specific combining groups on an antibody or with the recognition site on a T- lymphocyte but which, unlike antigenic determinants, does not itself elicit an immune response (e.g., a detectable T-cell response or the formation of a detectable amount of antibody). When coupled with a carrier protein, it does elicit an immune response.
Lymphokine. Soluble protein mediators released by certain lymphocytes, which in turn can regulate other cell-mediated immune functions, such as lymphocyte transformation, macrophage activation or cytotoxicity on other cells.
Mitogen. A substance that induces mitosis and cell transformation, especially lymphocyte
transformation.
A recognized disadvantage associated with in vivo administration of targeting moiety-radioisotopic conjugates for imaging or therapy is localization of the attached radioactive agent at both non-target and target sites. Until the administered radiolabeled conjugate clears from the circulation, normal organs and tissues are transitorily exposed to the attached radioactive agent. For instance, radiolabeled whole antibodies that are administered in vivo exhibit relatively slow blood clearance; maximum target site localization generally occurs 1-3 days post-administration. Generally, the longer the clearance time of the conjugate from the circulation, the greater the radioexposure of non-target organs.
These characteristics are particularly problematic with human radioimmunotherapy. In human clinical trials, the long circulating half-life of radioisotope bound to whole antibody causes relatively large doses of radiation to be delivered to the whole body. In particular, the bone marrow, which is very
radiosensitive, is the dose-limiting organ of non-specific toxicity.
In order to decrease radioisotope exposure of non-target tissue, potential targeting moieties generally have been screened to identify those that display minimal non-target reactivity, while retaining target specificity and reactivity. By reducing non-target exposure (and adverse non-target localization and/or toxicity), increased doses of a radiotherapeutic conjugate may be administered; moreover, decreased non-target accumulation of a radiodiagnostic conjugate leads to improved contrast between background and target.
Therapeutic drugs, administered alone or as targeted conjugates, are accompanied by similar disadvantages. Again, the goal is administration of the highest possible concentration of drug (to
maximize exposure of target tissue), while remaining below the threshold of unacceptable normal organ toxicity (due to non-target- tissue exposure). Unlike radioisotopes, however, therapeutic drugs need to be taken into a target cell to exert a cytotoxic effect. In the case of targeting moiety-therapeutic drug conjugates, it would be advantageous to combine the relative target specificity of a targeting moiety with a means for enhanced target cell internalization of the targeting moiety-drug conjugate.
In contrast, enhanced target cell internalization is disadvantageous if one administers diagnostic agent-targeting moiety conjugates. Internalization of diagnostic conjugates results in cellular catabolism and degradation of the conjugate. Upon degradation, small adducts of the diagnostic agent or the
diagnostic agent per se may be released from the cell, thus eliminating the ability to detect the conjugate in a target-specific manner. One. method for reducing non-target tissue exposure to a diagnostic or therapeutic agent involves
"pretargeting" the targeting moiety at a target site, and then subsequently administering a rapidly clearing diagnostic or therapeutic agent conjugate that is capable of binding to the "pretargeted" targeting moiety at the target site. A description of some embodiments of the pretargeting technique may be found in US Patent No. 4,863,713 (Goodwin et al.).
A typical pretargeting approach ("three-step") is schematically depicted below.
Figure imgf000013_0001
Briefly, this three-step pretargeting protocol
features administration of an antibody-ligand
conjugate, which is allowed to localize at a target site and to dilute in the circulation. Subsequently administered anti-ligand binds to the antibody-ligand conjugate and clears unbound antibody-ligand conjugate from the blood. Preferred anti-ligands are large and contain sufficient multivalency to accomplish
crosslinking and aggregation of circulating antibody-ligand conjugates. The clearing by anti-ligand is probably attributable to anti-ligand crosslinking and/or aggregation of antibody-ligand conjugates that are circulating in the blood, which leads to
complex/aggregate clearance by the recipient's RES (reticuloendothelial system). Anti-ligand clearance of this type is preferably accomplished with a
multivalent molecule; however, a univalent molecule of sufficient size to be cleared by the RES on its own could also be employed. Alternatively, receptor-based clearance mechanisms, e.g., Ashwell receptor hexose, e.g., galactose, mannose or the like, residue
recognition mechanisms, may be responsible for anti-ligand clearance. Such clearance mechanisms are less dependent upon the valency of the anti-ligand with respect to the ligand than the RES complex/aggregate clearance mechanisms. It is preferred that the ligand-anti-ligand pair displays relatively high affinity binding.
A diagnostic or therapeutic agent-ligand conjugate that exhibits rapid whole body clearance is then administered. When the circulation brings the active agent-ligand conjugate in proximity to the target cell-bound antibody-ligand-anti-ligand complex, anti-ligand binds the circulating active agent-ligand conjugate and produces an antibody-ligand : anti-ligand : ligand-active agent "sandwich" at the target site. Because the diagnostic or therapeutic agent is attached to a rapidly clearing ligand (rather than antibody, antibody fragment or other slowly clearing targeting moiety), this technique promises decreased non-target exposure to the active agent.
Alternate pretargeting methods eliminate the step of parenterally administering an anti-ligand clearing agent. These "two-step" procedures feature targeting moiety-ligand or targeting moiety-anti-ligand
administration, followed by administration of active agent conjugated to the opposite member of the ligand-anti-ligand pair. As an optional step "1.5" in the two-step pretargeting methods of the present
invention, a clearing agent (preferably other than ligand or anti-ligand alone) is administered to facilitate the clearance of circulating targeting moiety-containing conjugate.
In the two-step pretargeting approach, the
clearing agent preferably does not become bound to the target cell population, either directly or through the previously administered and target cell bound
targeting moiety-anti-ligand or targeting moiety- ligand conjugate. An example of two-step pretargeting involves the use of biotinylated human transferrin as a clearing agent for avidin-targeting moiety
conjugate, wherein the size of the clearing agent results in liver clearance of transferrin-biotin-circulating avidin-targeting moiety complexes and substantially precludes association with the avidin-targeting moiety conjugates bound at target cell sites. (See, Goodwin, D.A., Antibod. Immunoconj.
Radiopharm., 4: 427-34, 1991).
The two-step pretargeting approach overcomes certain disadvantages associated with the use of a clearing agent in a three-step pretargeted protocol. More specifically, data obtained in animal models demonstrate that in vivo anti-ligand binding to a pretargeted targeting moiety-ligand conjugate (i.e., the cell-bound conjugate) removes the targeting moiety-ligand conjugate from the target cell. One explanation for the observed phenomenon is that the multivalent anti-ligand crosslinks targeting moiety-ligand conjugates on the cell surface, thereby
initiating or facilitating internalization. of the resultant complex. The apparent loss of targeting moiety-ligand from the cell might result from internal degradation of the conjugate and/or release of active agent from the conjugate (either at the cell surface or intracellularly). An alternative explanation for the observed phenomenon is that permeability changes in the target cell's membrane allow increased passive diffusion of any molecule into the target cell. Also, some loss of targeting moiety-ligand may result from alteration in the affinity by subsequent binding of another moiety to the targeting moiety-ligand, e.g., anti-idiotype monoclonal antibody binding causes removal of tumor bound monoclonal antibody.
The present invention recognizes that this
phenomenon (apparent loss of the targeting moiety-ligand from the target cell) may be used to advantage with regard to in vivo delivery of therapeutic agents generally, or to drug delivery in particular. For instance, a targeting moiety may be covalently linked to both ligand and therapeutic agent and administered to a recipient. Subsequent administration of anti-ligand crosslinks targeting moiety-ligand-therapeutic agent tripartite conjugates bound at the surface, inducing internalization of the tripartite conjugate
(and thus the active agent). Alternatively, targeting moiety-ligand may be delivered to the target cell surface , followed by administration of anti-ligand-therapeutic agent.
In one aspect of the present invention, a
targeting moiety-anti-ligand conjugate is administered in vivo; upon target localization of the targeting moiety-anti-ligand conjugate (i.e., and clearance of this conjugate from the circulation), an active agent- ligand conjugate is parenterally administered. This method enhances retention of the targeting moiety-anti-ligand : ligand-active agent complex at the target cell (as compared with targeting moiety-ligand : anti-ligand : ligand-active agent complexes and targeting moiety-ligand : anti-ligand-active agent complexes). Although a variety of ligand/anti-ligand pairs may be suitable for use within the claimed invention, a preferred ligand/anti-ligand pair is biotin/avidin.
In a second aspect of the invention,
radioiodinated biotin and related methods are
disclosed. Previously, radioiodinated biotin
derivatives were of high molecular weight and were difficult to characterize. The radioiodinated biotin described herein is a low molecular weight compound that has been easily and well characterized.
In a third aspect of the invention, a targeting moiety-ligand conjugate is administered in vivo; upon target localization of the targeting moiety-ligand conjugate (i.e., and clearance of this conjugate from the circulation), a drug-anti-ligand conjugate is parenterally administered. This two-step method not only provides pretargeting of the targeting moiety conjugate, but also induces internalization of the subsequent targeting moiety-ligand-anti-ligand-drug complex within the target cell. Alternatively, another embodiment provides a three-step protocol that produces a targeting moiety-ligand : anti-ligand : ligand-drug complex at the surface, wherein the ligand-drug conjugate is administered simultaneously or within a short period of time after administration of anti-ligand (i.e., before the targeting moietyligand-anti-ligand complex has been removed from the target cell surface). Additional internalization methodologies are contemplated by the present
invention and are discussed herein.
In a fourth aspect of the invention, methods for radiolabeling biotin with technetium-99m, rhenium-186 and rhenium-188 are disclosed. Previously, biotin derivatives were radiolabeled with indium-Ill for use in pretargeted immunoscintigraphy (for instance, Virzi et al., Nucl. Med. Biol. 18:719-26, 1991; Kalofonos et al., J. Nucl. Med. 31: 1791-96, 1990; Paganelli et al., Cane. Res. 51:5960-66, 1991). However, 99mTc is a particularly preferred radionuclide for
immunoscintigraphy due to (i) low cost, (ii)
convenient supply and (iii) favorable nuclear
properties. Rhenium-186 displays chelating chemistry very similar to 99mTc, and is considered to be an excellent therapeutic radionuclide (i.e., a 3.7 day half-life and 1.07 MeV maximum particle that is similar to 131I). Therefore, the claimed methods for technetium and rhenium radiolabeling of biotin provide numerous advantages.
The present invention is also directed to
radiolabeling with yttrium-90, lutetium-177, sumarium-153, and other appropriate +3 metals. Y-90 is a particularly preferred radionuclide for therapy, because it exhibits favorable nuclear properties including high specific activity, long path length with respect to deposition of radiation in tissue, high equilibrium dose constant and favorable half-life properties. More specifically, the beta emission of Y-90 (Betaav=0.937 MeV) is one of the most energetic of all beta emitters. The X90 value of Y-90 is 5.34 mm (i.e., 90% of the energy emitted from a point source is absorbed in a sphere of 5.34 mm radius). Y-90 has a high equilibrium dose constant or mean
energy/nuclear transition. Delta = 1.99 Rad-gram/microcurie-hour, and a 64 hour half-life suitable for targeted therapy. Y-90 can be manufactured at high specific activity and is available as a generator product. Specific advantages of Y-90 are (1) that it has the capability to kill neighboring target cells not directly targeted by the pretargeted targeting moiety-ligand or targeting moiety-anti-ligand
conjugate and (2) that more radiation is deposited per microcurie localized than for other beta emitters of lower mean particle energy (provided that a
sufficiently large target volume is available).
Lu-177 is a particularly preferred radionuclide for targeted nuclide therapy, since it has a
moderately energetic beta emission (Betaav=0.140 MeV); it is available in high specific activity; its
radiochemical production is efficient; it emits two gammas of ideal energy and abundance for imaging (208 keV, 11% and 113 keV, 7%); and it has a relatively long half-life (161 hours). The X90 for Lu-177 is 0.31 mm, i.e., 90% of the energy emitted form a point source is absorbed in a sphere of radius 0.31 mm. Lu-177 has an equilibrium dose constant or mean energy/ nuclear transition of 0.31 Rad-gram/microcuries-hour and an adequate half-life to serve as a targeted therapeutic radionuclide. Specific advantages of Lu-177 are (1) that its emitted energy is efficiently absorbed in smaller targeted tumor volumes such as metastatic tumor foci or involved lymph nodes and (2) that its long physical half-life makes optimal use of the tumor retention property of the pretargeting delivery method.
The "targeting moiety" of the present invention binds to a defined target cell population, such as tumor cells. Preferred targeting moieties useful in this regard include antibody and antibody fragments, peptides, and hormones. Proteins corresponding to known cell surface receptors (including low density lipoproteins, transferrin and insulin), fibrinolytic enzymes, anti-HER2, platelet binding proteins such as annexins, and biological response modifiers (including interleukin, interferon, erythropoietin and colony-stimulating factor) are also preferred targeting moieties. Also, anti-EGF receptor antibodies, which internalize following binding to the receptor and traffic to the nucleus to an extent, ar«= preferred targeting moieties for use in the present invention to facilitate delivery of Auger emitters and nucleus binding drugs to target cell nuclei.
Oligonucleotides, e.g., antisense oligonucleotides that are complementary to portions of target cell nucleic acids (DNA or RNA), are also useful as
targeting moieties in the practice of the present invention. Oligonucleotides binding to cell surfaces are also useful. Analogs of the above-listed
targeting moieties that retain the capacity to bind to a defined target cell population may also be used within the claimed invention. In addition, synthetic targeting moieties may be designed.
Functional equivalents of the aforementioned molecules are also useful as targeting moieties of the present invention. One targeting moiety functional equivalent is a "mimetic" compound, an organic
chemical construct designed to mimic the proper configuration and/or orientation for targeting moiety- target cell binding. Another targeting moiety
functional equivalent is a short polypeptide
designated as a "minimal" polypeptide, constructed using computer-assisted molecular modeling and mutants having altered binding affinity, which minimal
polypeptides exhibit the binding affinity of the targeting moiety.
Preferred targeting moieties of the present invention are antibodies (polyclonal or monoclonal), peptides, oligonucleotides or the like. Polyclonal antibodies useful in the practice of the present invention are polyclonal (Vial and Callahan, Univ. Mich. Med. Bull., 20: 284-6, 1956), affinity-purified polyclonal or fragments thereof (Chao et al., Res. Comm. in Chem. Path. & Pharm., 9: 749-61, 1974).
Monoclonal antibodies useful in the practice of the present invention include whole antibody and fragments thereof. Such monoclonal antibodies and fragments are producible in accordance with
conventional techniques, such as hybridoma synthesis, recombinant DNA techniques and protein synthesis. Useful monoclonal antibodies and fragments may be derived from any species (including humans) or may be formed as chimeric proteins which employ sequences from more than one species. See, generally, Kohler and Milstein, Nature, 256: 495-97, 1975; Eur. J.
Immunol., 6: 511-19, 1976.
Human monoclonal antibodies or "humanized" murine antibody are also useful as targeting moieties in accordance with the present invention. For example, murine monoclonal antibody may be "humanized" by genetically recombining the nucleotide sequence encoding the murine Fv region (i.e., containing the antigen binding sites) or the complementarity
determining regions thereof with the nucleotide sequence encoding a human constant domain region and an Fc region, e.g., in a manner similar to that disclosed in European Patent Application
No. 0,411,893 A2. Some murine residues may also be retained within the human variable region framework domains to ensure proper target site binding
characteristics. Humanized targeting moieties are recognized to decrease the immunoreactivity of the antibody or polypeptide in the host recipient, permitting an increase in the half-life and a
reduction in the possibility of adverse immune reactions.
Another preferred targeting moiety of the present invention are annexins and other platelet binding proteins, such as PAP-1 (Placental Anticoagulant
Protein or Annexin V) . Annexins are (with the
exception of annexin II) single chain, non-glycosylated proteins of approximately 36 kilodaltons. Annexins possess a number of biological properties based on the principle of calcium ion binding.
Investigations have shown that annexins bind with high affinity to membrane lipids in the presence of
micromolar quantities of calcium. In the presence of calcium, these proteins have an especially high affinity for negatively charged phospholipids such as phosphatidylserine or phosphatidylinosine.
Annexins exert anti-coagulatory effects. In a manner analogous to anti-inflammatory mechanisms, coagulation inhibition is mediated by the binding of annexins to negatively charged surface phospholipids (i.e., to platelets) which binding blocks the
activation of clotting factors by such negatively charged surface phospholipids. Annexins localize to target sites rapidly, i.e., in a matter of minutes, but remains circulating in the serum for a longer time period.
As a result of these properties, annexins may be employed in two-step or three-step pretargeting protocols for the diagnosis of blood clots or the treatment of blood clots associated with indications, such as DVT (deep vein thrombosis), PE (pulmonary embolism), heart attack, stroke and the like.
Exemplary diagnostic and treatment protocols employing the two-step pretargeting approach are set forth below to further elucidate this aspect of the present invention.
For the visualization of blood clots associated with a number of pathological conditions, a chemical conjugate or a fusion protein of annexin with avidin or streptavidin is administered to a recipient for whom such a diagnosis is desired. The annexin portion of the conjugate localizes rapidly to target sites characterized by negatively charged surface
phospholipids, such as blood clots. As a consequence of the rapid target site uptake, a clearing agent, such as those discussed herein, may be administered after the passage of a short time (ranging from about 5 minutes to about 1 hour, with within about 15 minutes preferred). The clearing agent serves to rapidly diminish the serum level of the annexin-anti-ligand conjugate. Next, biotin labeled with an imaging radionuclide, such as Tc-99m for example, is administered. Biotin directs the localization of the administered moiety to the previously localized annexin-anti-ligand. Non-target bound radiolabeled biotin is rapidly cleared from the recipient.
Consequently, imaging of the target sites proceeds with minimal exposure of non-target sites to
radioactivity. This approach offers the advantages of speed, facilitating imaging of difficult target sites such as lung clots, and improved clot to blood ratios.
For therapeutic applications involving blood clots associated with a number of pathological conditions, a chemical conjugate or a fusion protein of annexin with avidin or streptavidin is administered to a recipient for whom such treatment is desired. A clearing agent, such as those discussed herein, may be administered after the passage of a short time (ranging from about 5 minutes to about 1 hour, with within about 15 minutes preferred) to diminish the serum level of the annexin-anti-ligand conjugate. Biotin conjugated with therapeutic agents, such as fibrolytic agents, tissue plasminogen activator, thrombolytic agents (e.g., streptokinase, anisoylated plasminogen streptokinase activator complex) and the like, are deliverable in this manner. Such a dosing regimen may be repeated, for example, once daily for a number of weeks to address the recipient's physiological condition. Consequently, delivery of a therapeutic agent to target sites proceeds with minimal exposure of nontarget sites to that agent.
An example of an annexin useful in the practice of the present invention is Annexin V which was isolated by Bohn in 1979 from human placenta, a rich source of annexins, and termed Placenta Protein 4 (PP4).
Annexin V consists of four domains, with each made up of five alpha helices wherein the alpha helices serve as connecting elements between the domains. From the side, the molecule appears crown-like with five calcium binding sites on its convex surface, through which annexin-phospholipid interactions are mediated. Other annexins having the characteristics described above are also useful in the practice of the present invention. Annexin V has been expressed in E . coli.
Types of active agents (diagnostic or therapeutic) useful herein include toxins, anti-tumor agents, drugs and radionuclides. Several of the potent toxins useful within the present invention consist of an A and a B chain. The A chain is the cytotoxic portion and the B chain is the receptor-binding portion of the intact toxin molecule (holotoxin). Because toxin B chain may mediate non-target cell binding, it is often advantageous to conjugate only the toxin A chain to a targeting moiety. However, while elimination of the toxin B chain decreases non-specific cytotoxicity, it also generally leads to decreased potency of the toxin A chain-targeting protein conjugate, as compared to the corresponding holotoxin-targeting moiety
conjugate.
Preferred toxins in this regard include
holotoxins, such as abrin, ricin, modeccin,
Pseudomonas exotoxin A, Diphtheria toxin, pertussis toxin and Shiga toxin; and A chain or "A chain-like" molecules, such as. ricin A chain, abrin, A chain, modeccin A chain, the enzymatic portion of Pseudomonas exotoxin A, Diphtheria toxin A chain, the enzymatic portion of pertussis toxin, the enzymatic portion of Shiga toxin, gelonin, pokeweed antiviral protein, saporin, tritin, barley toxin and snake venom
peptides. Ribosomal inactivating proteins (RIPs), naturally occurring protein synthesis inhibitors that lack translocating and cell-binding ability, are also suitable for use herein.
Preferred drugs suitable for use herein include conventional chemotherapeutics, such as vinblastine, doxorubicin, bleomycin, methotrexate, 5-fluorouracil, 6-thioguanine, cytarabine, cyclophosphamide and cis-platinum, as well as other conventional
chemotherapeutics as described in Cancer: Principles and Practice of Oncology, 2d ed., V.T. DeVita, Jr., S. Hellman, S.A. Rosenberg, J.B. Lippincott Co.,
Philadelphia, PA, 1985, Chapter 14. A particularly preferred drug within the present invention is a trichothecene.
Trichothecenes are drugs produced by soil fungi of the class Fungi imperfecti or isolated from Baccharuε megapotamica (Bamburg, J.R. Proc. Molec. Subcell.
Biol. 8 : 41-110, 1983; Jarvis & Mazzola, Acc. Chem. Res. 15:338-395, 1982). They appear to be the most toxic molecules that contain only carbon, hydrogen and oxygen (Tamm, C. Fortschr. Chem. Org.
Naturst. 31: 61-117, 1974). They are all reported to act at the level of the ribosome as inhibitors of protein synthesis at the initiation, elongation, or termination phases.
There are two broad classes of trichothecenes:
those that have only a central sesquiterpenoid
structure and those that have an additional
macrocyclic ring (simple and macrocyclic
trichothecenes, respectively). The simple
trichothecenes may be subdivided into three groups (i.e., Group A, B, and C) as described in U.S. Patent Nos. 4,744,981 and 4,906,452 (incorporated herein by reference). Representative examples of Group A simple trichothecenes include: Scirpene, Roridin C,
dihydrotrichothecene, Scirpen-4, 8-diol, Verrucarol, Scirpentriol, T-2 tetraol, pentahydroxyscirpene,
4-deacetylneosolaniol, trichodermin,
deacetylcalonectrin, calonectrin, diacetylverrucarol, 4-monoacetoxyscirpenol, 4,15-diacetoxyscirpenol,
7-hydroxydiacetoxyscirpenol,
8-hydroxydiacetoxy-scirpenol (Neosolaniol),
7,8-dihydroxydiacetoxyscirpenol,
7-hydroxy-8-acetyldiacetoxyscirpenol,
8-acetylneosolaniol, NT-1, NT-2, HT-2, T-2, and acetyl T-2 toxin. Representative examples of Group B simple trichothecenes include: Trichothecolone,
Trichothecin, deoxynivalenol, 3-acetyldeoxynivalenol, 5-acetyldeoxynivalenol, 3,15-diacetyldeoxynivalenol, Nivalenol, 4-acetylnivalenol (Fusarenon-X),
4,15-idacetylnivalenol, 4,7, 15-triacetylnivalenol, and tetra-acetylnivalenol. Representative examples of Group C simple trichothecenes include: Crotocol and Crotocin. Representative macrocyclic trichothecenes include Verrucarin A, Verrucarin B, Verrucarin J
(Satratoxin C), Roridin A, Roridin D, Roridin E
(Satratoxin D), Roridin H, Satratoxin F, Satratoxin G, Satratoxin H, Vertisporin, Mytoxin A, Mytoxin C,
Mytoxin B, Myrotoxin A, Myrotoxin B, Myrotoxin C, Myrotoxin D, Roritoxin A, Roritoxin B, and
Roritoxin D. In addition, the general "trichothecene" sesquiterpenoid ring structure is also present in compounds termed "baccharins" isolated from the higher plant Baccharis megapotamica, and these are described in the literature, for instance as disclosed by
Jarvis et al. (Chemistry of Alleopathy, ACS Symposium Series No. 268: ed. A.C. Thompson, 1984, pp. 149-159).
Experimental drugs, such as mercaptopurine, N-methylformamide, 2-amino-1,3,4-thiadiazole, melphalan, hexamethylmelamine, gallium nitrate, 3% thymidine, dichloromethotrexate, mitoguazone, suramin,
bromodeoxyuridine, iododeoxyuridine, semustine, 1-(2-chloroethyl)-3-(2,6-dioxo-3-piperidyl)-1-nitrosourea, N,N'-hexamethylene-bis-acetamide, azacitidine,
dibromodulcitol, Erwinia asparaginase, ifosfamide, 2-mercaptoethane sulfonate, teniposide, taxol, 3-deazauridine, soluble Baker's antifol,
homoharringtonine, cyclocytidine, acivicin, ICRF-187, spiromustine, levamisole, chlorozotocin, aziridinyl benzoquinone, spirogermanium, aclarubicin,
pentostatin, PALA, carboplatin, amsacrine, caracemide, iproplatin, misonidazole, dihydro-5-azacytidine, 4'-deoxy-doxorubicin, menogaril, triciribine phosphate, fazarabine, tiazofurin, teroxirone, ethiofos, N-(2-hydroxyethyl)-2-nitro-1H-imidazole-1-acetamide, mitoxantrone, acodazole, amonafide, fludarabine
phosphate, pibenzimol, didemnin B, merbarone,
dihydrolenperone, flavone-8-acetic acid, oxantrazole, ipomeanol, trimetrexate, deoxyspergualin, echinomycin, and dideoxycytidine (see NCI Investigational Drugs, Pharmaceutical Data 1987. NIH Publication No. 88-2141, Revised November 1987) are also preferred.
Radionuclides useful within the present invention include gamma-emitters, positron-emitters, Auger electron-emitters. X-ray emitters and fluorescenceemitters, with beta- or alpha-emitters preferred for therapeutic use. Radionuclides are well-known in the art and include 123I, 125I, 130I, 131I, 133I, 135I, 47Sc, 72As,72Se, 90Y, 88Y, 97Ru, 100Pd, 101mRh, 119Sb, 128Ba, 197Hg, 211At, 212Bi, 153Sm, 169Eu, 212Pb, 109Pd, 111ln, 67Ga, 68Ga, 67Cu, 75Br, 76Br, 77Br, 99mTc, 11C, 13N, 15O and 18F. Preferred therapeutic radionuclides include 188Re, 186Re, 203Pb, 212Pb, 212Bi, 109Pd, 64Cu, 67CU, 90Y, 125I, 131I, 77Br, 211At, 97Ru, 105Rh, 198Au and 199Ag or 177Lu.
Other anti-tumor agents are administrable in accordance with the present invention. Exemplary anti-tumor agents include cytokines, such as IL-2, tumor necrosis factor or the like, lectin inflammatory response promoters (selectins), such as L-selectin, E-selectin, P-selectin or the like, and like molecules.
Ligands suitable for use within the present invention include biotin, haptens, lectins, epitopes, dsDNA fragments, enzyme inhibitors and analogs and derivatives thereof. Useful complementary anti-ligands include avidin (for biotin), carbohydrates (for lectins), antibody, fragments or analogs thereof, including mimetics (for haptens and epitopes) and zinc finger proteins (for dsDNA fragments) and enzymes (for enzyme inhibitors). Preferred ligands and anti-ligands bind to each other with an affinity of at least about kD ≥ 10-9 M.
As mentioned above, zinc finger protein/dsDNA is a ligand-anti-ligand binding pair contemplated by the present invention. Zinc finger proteins are a class of proteins containing repeating subunits of
approximately 30 amino acids that bind to specific promoter regions on dsDNA and aid in transcription. These proteins are also capable of binding to dsRNA with unknown function. Like antibodies, zinc finger proteins are a family of proteins consisting of variable and conserved regions. Zinc finger proteins, as a class, contain from about 2 to about 39 repeating subunits (fingers), with each such finger capable of binding specific nucleotide sequences. Each finger is capable of binding to a "cleft" in the conformation of the dsDNA/RNA structure that is dependent upon
nucleotide sequence. The present inventors
hypothesize that greater numbers of subunits
correspond to greater affinity and specificity. In addition, zinc is incorporated into the proteins within each subunit and is important in maintaining proper conformation for dsDNA binding. To illustrate the use of the zinc finger
protein/dsDNA binding pair in pretargeting protocols, a two-step approach is described below. This ligand-anti-ligand binding pair may also be used in three-step pretargeting protocols of the present invention. For two-step pretargeting, a zinc finger protein or a dsDNA fragment is conjugated to a targeting moiety in any convenient manner therefor, including, for
example, conjugation methods described herein.
Several methods have been developed to conjugate
DNA or oligonucleotides to proteins. One simple method is oxidation of the sugar moiety of the DNA. The resultant aldehyde groups are available for reaction with amines on the protein. The amines generated by such reactions are then reduced by reaction with a reducing agent such as NaCNBH^ to form the more stable amine bonds.
Another protein-oligonucleotide conjugation method involves initial oxidation of the sugar residue on DNA using an oxidizing agent such as NaIO4. The dialdehyde formed is treated with 5-pyridylcystamine
hydrochloride and NaCNBH3. This dithiopyridine
derivative can then be reacted with tributylphosphine to generate the thiol adduct. Reaction of the free thiol-bearing DNA adduct with a maleimide-derivatized protein results in a thioether oligonucleotide-protein conjugate. See, for example, Kuijpers et al.,
"Specific Recognition of Antibody-Oligonucleotide Conjugates by Radiolabeled Antisense Nucleotides: A Novel Approach for Two-Step Radioimmunotherapy of Cancer," Bioconjugate Chem., 4: 94-102 (1993).
The conjugates are administered to a recipient by a route chosen in view of the patient's physical condition and known or anticipated illness. Following target site localization of the administered conjugate (or clearing agent treatment as described herein), administration of the other member of the ligand pair conjugated to a therapeutic agent (e.g., radionuclide, drug or anti-tumor agent) is undertaken, The
administered therapeutic agent-containing conjugate binds to the previously localized targeting agent containing conjugate. Therapeutic agent-containing conjugate that does not bind at the target site will be eliminated from the recipient's body at a much more rapid rate than a monoclonal antibody-therapeutic agent conjugate would be removed. Moreover, such therapeutic agent-containing conjugates can be
chemically altered for more rapid excretion, if necessary, to reduce non-target exposure to the therapeutic agent.
A schematic of the administered components and ultimate in vivo formed "sandwich" for a monoclonal antibody-zinc finger protein/dsDNA-therapeutic agent two-step pretargeting protocol are shown below.
Figure imgf000030_0001
Figure imgf000031_0001
Use of the zinc finger protein/dsDNA fragment ligand/anti-ligand binding pair offers advantages with respect to immunogenicity. The targeting agent used in a pretargeting protocol may be human or humanized, and all of the other administered components
(exclusive of the therapeutic agent) are human in origin. An additional advantage of this approach is that zinc finger proteins can be engineered to
accommodate specific, high affinity interactions with synthetic or cloned dsDNA fragments, thereby
eliminating non-specific interactions of the
administered components with normal tissue.
One embodiment of this aspect of the present invention involves the use of pretargeting approaches to target double stranded DNA itself. Such protocols of the present invention are useful, for example, for gene therapy, delivery of tumor suppressive DNA, deleted genes and the like. More specifically, a targeting moiety-zinc finger protein is pretargeted to a target site. Subsequently, a dsDNA fragment that is complementary to the zinc finger protein is injected and allowed to bind to the localized zinc finger protein. Because zinc finger protein-DNA interaction is a normal cell physiologic mechanism, DNA will be delivered free of encumbering chemistry to the
cellular milieu. The dsDNA is then internalized by . the target cell via the normal mechanisms therefor, i.e., via endocytosis or the like.
One component to be administered in a preferred two-step pretargeting protocol involving the biotin-avidin ligand-anti-ligand system is a targeting moiety-avidin or a targeting moiety-streptavidin conjugate. The preferred targeting moiety useful in these embodiments of the present invention is a monoclonal antibody. Protein-protein conjugations are generally problematic due to the formation of
undesirable byproducts, including high molecular weight and cross-linked species, however. A non-covalent synthesis technique involving reaction of biotinylated antibody with streptavidin has been reported to result in substantial byproduct formation. Also, at least one of the four biotin binding sites on the streptavidin is used to link the antibody and streptavidin, while another such binding site may be sterically unavailable for biotin binding due to the configuration of the streptavidin-antibody conjugate.
Thus, covalent streptavidin-antibody conjugation is preferred, but high molecular weight byproducts are often obtained. The degree of crosslinking and aggregate formation is dependent upon several factors, including the level of protein derivitization using heterobifunctional crosslinking reagents. Sheldon et al., Appl. Radiat. Isot. 43: 1399-1402, 1992, discuss preparation of covalent thioether conjugates by reacting succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC)-derivitized antibody and iminothiolane-derivitized streptavidin.
Streptavidin-proteinaceous targeting moiety conjugates are preferably prepared as described in Example XI below, with the preparation involving the steps of:. preparation of SMCC-derivitized
streptavidin; preparation of DTT-reduced proteinaceous targeting moiety; conjugation of the two prepared moieties; and purification of the monosubstituted conjugate. The purified fraction is preferably further characterized by one or more of the following techniques: HPLC size exclusion, SDS-PAGE,
immunoreactivity, biotin binding capacity and in vivo studies.
Alternatively, thioether conjugates useful in the practice of the present invention may be formed using other thiolating agents, such as SPDP, iminothiolate, SATA or the like, or other thio-reactive
heterobifunctional cross linkers, such as m-maleimidobenzoyl-N-hydroxysuccinimide ester, N-succinimidyl (4-iodoacetyl) aminobenzoate or the like.
Streptavidin-proteinaceous targeting moiety conjugates of the present invention can also be formed by conjugation of a lysine epsilon amino group of one protein with a maleimide-derivitized form of the other protein. For example, at pH 8-10, lysine epsilon amino moieties react with protein maleimides,
prepared, for instance, by treatment of the protein with SMCC, to generate stable amine covalent
conjugates. In addition, conjugates can be prepared by reaction of lysine epsilon amino moieties of one protein with aldehyde functionalities of the other protein. The resultant imine bond is reducible to generate the corresponding stable amine bond.
Aldehyde functionalities may be generated, for
example, by oxidation of protein sugar residues or by reaction with aldehyde-containing heterobifunctional cross linkers.
Another method of forming streptavidin-targeting moiety conjugates involves immobilized iminobiotin that binds SMCC-derivitized streptavidin. In this conjugation/purification method, the reversible binding character of iminobiotin (immobilized) to streptavidin is exploited to readily separate conjugate from the unreacted targeting moiety.
Iminobiotin binding can be reversed under conditions of lower pH and elevated ionic strength, e.g., NH2OAc, pH 4 (50 mM) with 0.5 M NaCl.
For streptavidin, for example, the conjugation/purification proceeds as follows:
- SMCC-derivitized streptavidin is bound to immobilized iminobiotin (Pierce Chemical Co., St.
Louis, Missouri), preferably in column format;
- a molar excess (with respect to streptavidin) of DTT-reduced antibody (preferably free of reductant) is added to the nitrogen-purged, phosphate-buffered iminobiotin column wherein the SMCC-streptavidin is bound (DTT-reduced antibody will saturate the bound SMCC-streptavidin, and unbound reduced antibody passing through the column can be reused);
- the column is washed free of excess antibody; and
- a buffer that lowers the pH and increases ionic strength is added to the column to elute streptavidin-antibody conjugate in pure form.
As indicated above, targeting moiety-mediated ligand-anti-ligand pretargeting involves the
localization of either targeting moiety-ligand or targeting moiety-anti-ligand at target tissue. Often, peak uptake to such target tissue is achieved before the circulating level of targeting moiety-containing conjugate in the blood is sufficiently low to permit the attainment of an optimal target-to-non-target conjugate ratio. To obviate this problem, two
approaches are useful. The first approach allows the targeting moiety-containing conjugate to clear from the blood by "natural" or endogenous clearance
mechanisms. This method is complicated by variations in systemic clearance of proteins and by endogenous ligand or anti-ligand. For. example, endogenous biotin may interfere with the preservation of biotin binding sites on a streptavidin-targeting moiety conjugate.
The second approach for improving targeting moiety-ligand or targeting moiety-anti-ligand
conjugate target-to-blood ratio "chases" the conjugate from the circulation through in vivo complexation of conjugate with a molecule constituting or containing the complementary anti-ligand or ligand. When
biotinylated antibodies are used as a ligand-targeting moiety conjugate, for example, avidin forms relatively large aggregated species upon complexation with the circulating biotinylated antibody, which aggregated species are rapidly cleared from the blood by the RES uptake. See, for example, U.S. Patent No. 4,863,713. One problem with this method, however, is the
potential for cross-linking and internalizing tumor-bound biotinylated antibody by avidin.
When avidin-targeting moiety conjugates are employed, poly-biotinylated transferrin has been used to form relatively large aggregated species that are cleared by RES uptake. See, for example, Goodwin, J. Nucl. Med. 33(10):1816-18. 1992). Poly-biotinylated transferrin also has the potential for cross-linking and internalizing tumor-bound avidinylated-targeting moiety, however. In addition, both "chase"
methodologies involve the prolonged presence of aggregated moieties of intermediate, rather than large, size (which are not cleared as quickly as large size particles by RES uptake), thereby resulting in serum retention of subsequently administered ligand-active agent or anti-ligand-active agent. Such serum retention unfavorably impacts the target cell-to-blood targeting ratio.
The present invention provides clearing agents of protein and non-protein composition having physical properties facilitating use for in vivo complexation and blood clearance of anti-ligand/ligand (e.g., avidin/biotin) -targeting moiety (e.g., antibody) conjugates. These clearing agents are useful in improving the target:blood ratio of targeting moiety conjugate. Other applications of these clearing agents include lesional imaging or therapy involving blood clots and the like, employing antibody-active agent delivery modalities. For example, efficacious anti-clotting agent provides rapid target localization and high target:non-target targeting ratio. Active agents administered in pretargeting protocols of the present invention using efficient clearing agents are targeted in the desirable manner and are, therefore, useful in the imaging/therapy of conditions such as pulmonary embolism and deep vein thrombosis.
Clearing agents useful in the practice of the present invention preferably exhibit one or more of the following characteristics:
- rapid, efficient complexation with targeting moiety-ligand (or anti-ligand) conjugate .in vivo;
- rapid clearance from the blood of targeting moiety conjugate capable of binding a subsequently administered complementary anti-ligand or ligand containing molecule;
- high capacity for clearing (or inactivating) large amounts of targeting moiety conjugate; and
- low immunogenicity.
Preferred clearing agents include hexose-based and non-hexose based moieties. Hexose-based clearing agents are molecules that have been derivatized to incorporate one or more hexoses (six carbon sugar moieties) recognized by Ashwell receptors. Exemplary of such hexoses are galactose, mannose and the like. Galactose is the prototypical clearing agent hexose derivative for the purposes of this description.
Thus, galactose-based and non-galactose based
molecules are discussed below. Protein-type galactose-based clearing agents include proteins having endogenous exposed galactose residues or which have been derivitized to expose or incorporate such galactose residues. Exposed
galactose residues direct the clearing agent to rapid clearance by endocytosis into the liver through specific receptors therefor (Ashwell receptors).
These receptors bind the clearing agent, and induce endocytosis into the hepatocyte, leading to fusion with a lysosome and recycle of the receptor back to the cell surface. This clearance mechanism is
characterized by high efficiency, high capacity and rapid kinetics.
An exemplary clearing agent of the protein-based/galactose-bearing variety is the
asialoorosomucoid derivative of human alpha-1 acid glycoprotein (orosomucoid, molecular weight = 41,000 Dal, isoelectric point = 1.8-2.7). The rapid
clearance from the blood of asialoorosomucoid has been documented by Galli, et al., J. of Nucl. Med. Allied Sci. 32(2) : 110-16, 1988.
Treatment of orosomucoid with neuraminidase removes sialic acid residues, thereby exposing
galactose residues. Other such derivitized clearing agents include, for example, galactosylated albumin, galactosylated-IgM, galactosylated-IgG,
asialohaptoglobin, asialofetuin, asialoceruloplasmin and the like.
Human serum albumin (HSA), for example, may be employed in a clearing agent of the present invention as follows:
(Hexose)m╌Human Serum Albumin (HSA)╌ (Ligand)n , wherein n is an integer from 1 to about 10 and m is an integer from 1 to about 25 and wherein the hexose is recognized by Ashwell receptors.
In a preferred embodiment of the present invention the ligand is biotin and the hexose is galactose. More preferably, HSA is derivatized with from 10-20
galactose residues and 1-5 biotin residues. Still more preferably, HSA clearing agents of the present invention are derivatized with from about 12 to about 15 galactoses and 3 biotins. Derivatization with both galactose and biotin are conducted in a manner
sufficient to produce individual clearing agent molecules with a range of biotinylation levels that averages a recited whole number, such as 1, biotin. Derivatization with 3 biotins, for example, produces a product mixture made up of individual clearing agent molecules, substantially all of which having at leastone biotin residue. Derivatization with 1 biotin produces a clearing agent product mixture, wherein a significant portion of the individual molecules are not biotin derivatized. The whole numbers used in this description refer to the average biotinylation of the clearing agents under discussion.
In addition, clearing agents based upon human proteins, especially human serum proteins such as, for example, orosomucoid and human serum albumin, are less immunogenic upon administration into the serum of a human recipient. Another advantage of using
asialoorosomucoid is that human orosomucoid is
commercially available from, for example, Sigma
Chemical Co, St. Louis, Missouri.
One way to prevent clearing agent compromise of target-bound conjugate through direct complexation is through use of a clearing agent of a size sufficient to render the clearing agent less capable of diffusion into the extravascular space and binding to target-associated conjugate. This strategy is useful alone or in combination with the aforementioned recognition that exposed galactose residues direct rapid liver uptake. This size-exclusion strategy enhances the effectiveness of non-galactose-based clearing agents of the present invention. The combination (exposed galactose, and size) strategy improves the
effectiveness of "protein-type" or "polymer-type" galactose-based clearing agents.
Galactose-based clearing agents include
galactosylated, biotinylated proteins (to remove circulating streptavidin-targeting moiety conjugates, for example) of intermediate molecular weight (ranging from about 40,000 to about 200,000 Dal), such as biotinylated asialoorosomucoid, galactosyl-biotinyl-human serum albumin or other galactosylated and biotinylated derivatives of non-immunogenic soluble natural proteins, as well as biotin- and galactose-derivitized polyglutamate, polylysine, polyarginine, polyaspartate and the like. High molecular weight moieties (ranging from about 200,000 to about
1,000,000 Dal) characterized by poor target access, including galactosyl-biotinyl-IgM or -IgG
(approximately 150,000 Dal) molecules, as well as galactose- and biotin-derivitized transferrin
conjugates of human serum albumin, IgG and IgM
molecules and the like, can also be used as clearing agents of the claimed invention. Chemically modified polymers of intermediate or high molecular weight (ranging from about 40,000 to about 1,000,000 Dal), such as galactose- and biotin-derivitized dextran, hydroxypropylmethacrylamide polymers,
polyvinylpyrrolidone-polystyrene copolymers, divinyl ether-maleic acid copolymers, pyran copolymers, or PEG, also have utility as clearing agents in the practice of the present invention. In addition, rapidly clearing biotinylated liposomes (high
molecular weight moieties with poor target access) can be derivitized with galactose and biotin to produce clearing agents for use in the practice of the present invention.
A further class of clearing agents useful in the present invention involve small molecules (ranging from about 500 to about 10,000 Dal) derivitized with galactose and biotin that are sufficiently polar to be confined to the vascular space as an in vivo volume of distribution. More specifically, these agents exhibit a highly charged structure and, as a result, are not readily distributed into the extravascular volume, because they do not readily diffuse across the lipid membranes lining the vasculature. Exemplary of such clearing agents are mono- or poly-biotin-derivitized 6,6'-[(3,3'-dimethyl[1,1'-biphenyl]-4,4'-diyl)bis(azo) bis[4-amino-5-hydroxy-1,3-naphthalene disulfonic acid] tetrasodium salt, mono- or poly-biotinyl-galactose-derivitized polysulfated dextran-biotin, mono- or poly-biotinyl-galactose-derivitized dextran-biotin and the like.
The galactose-exposed or -derivitized clearing agents are preferably capable of (1) rapidly and efficiently complexing with the relevant ligand- or anti-ligand-containing conjugates via ligand-anti-ligand affinity; and (2) clearing such complexes from the blood via the galactose- receptor, a liver specific degradation system, as opposed to aggregating into complexes that are taken up by the generalized RES system, including the lung and spleen. Additionally, the rapid kinetics of galactose-mediated liver uptake, coupled with the affinity of the ligand-anti-ligand interaction, allow the use of intermediate or even low molecular weight carriers.
Non-galactose residue-bearing moieties of low or intermediate molecular weight (ranging from about
40,000 to about 200,000 Dal) localized in the blood may equilibrate with the extravascular space and, therefore, bind directly to target-associated
conjugate, compromising target localization. In addition, aggregation-mediated clearance mechanisms operating through the RES system are accomplished using a large stoichiometric excess of clearing agent. In contrast, the rapid blood clearance of galactose- based clearing agents used in the present invention prevents equilibration, and the high affinity ligand-anti-ligand binding allows the use of low
stoichiometric amounts of such galactose-based
clearing agents. This feature further diminishes the potential for galactose-based clearing agents to compromise target-associated conjugate, because the absolute amount of such clearing agent administered is decreased.
Clearing agent evaluation experimentation
involving galactose- and biotin-derivatized clearing agents of the present invention is detailed in Example XXII. Specific clearing agents of the present
invention that were examined during the Example XXII experimentation are (1) asialoorosomucoid-biotin, (2) human serum albumin derivatized with galactose and biotin, and (3) a 70,000 dalton molecular weight dextran derivatized with both biotin and galactose. The experimentation showed that proteins and polymers are derivatizable to contain both galactose and biotin and that the resultant derivatized molecule is
effective in removing circulating streptavidin-protein conjugate from the serum of the recipient. Biotin loading was varied to determine the effects on both clearing the blood pool of circulating avidin-containing conjugate and the ability to deliver a subsequently administered biotinylated isotope to a target site recognized by the streptavidin-containing conjugate. The effect of relative doses of the administered components with respect to clearing agent efficacy was also examined.
Protein-type and polymer-type non-galactose-based clearing agents include the agents described above, absent galactose exposure or derivitization and the like. These clearing agents act through an
aggregation-mediated RES mechanism. In these embodiments of the present invention, the clearing agent used will be selected on the basis of the target organ to which access of the clearing agent is to be excluded. For example, high molecular weight (ranging from about 200,000 to about 1,000,000 Dal) clearing agents will be used when tumor targets or clot targets are involved.
Another class of clearing agents includes agents that do not remove circulating ligand or anti-ligand/targeting moiety conjugates, but instead
"inactivate" the circulating conjugates by blocking the relevant anti-ligand or ligand binding sites thereon. These "cap-type" clearing agents are
preferably small (500 to 10,000 Dal) highly charged molecules, which exhibit physical characteristics that dictate a volume of distribution equal to that of the plasma compartment (i.e., do not extravasate into the extravascular fluid volume). Exemplary cap-type clearing agents are poly-biotin-derivitized 6,6'-[(3,3'-dimethyl[1,1'-biphenyl]-4,4'-diyl)bis(azo) bis[4-amino-5-hydroxy-1,3-naphthalene disulfonic acid] tetrasodium salt, poly-biotinyl-derivitized
polysulfated dextran-biotin, mono- or poly-biotinyl-derivitized dextran-biotin and the like.
Cap-type clearing agents are derivitized with the relevant anti-ligand or ligand, and then administered to a recipient of previously administered ligand/ or anti-ligand/targeting moiety conjugate. Clearing agent-conjugate binding therefore diminishes the ability of circulating conjugate to bind any
subsequently administered active agent-ligand or active agent-anti-ligand conjugate. The ablation of active agent binding capacity of the circulating conjugate increases the efficiency of active agent delivery to the target, and increases the ratio of target-bound active agent to circulating active agent by preventing the coupling of long-circulating serum protein kinetics with the active agent. Also, confinement of the clearing agent to the plasma compartment prevents compromise of target-associated ligand or anti-ligand.
Clearing agents of the present invention may be administered in single or multiple doses. A single dose of biotinylated clearing agent, for example, produces a rapid decrease in the level of circulating targeting moiety-streptavidin, followed by a small increase in that level, presumably caused, at least in part, by re-equilibration of targeting moiety-streptavidin within the recipient's physiological compartments. A second or additional clearing agent doses may then be employed to provide supplemental clearance of targeting moiety-streptavidin.
Alternatively, clearing agent may be infused
intravenously for a time period sufficient to clear targeting moiety-streptavidin in a continuous manner.
Other types of clearing agents and clearance systems are also useful in the practice of the present invention to remove circulating targeting moiety-ligand or -anti-ligand conjugate from the recipient's circulation. Particulate-based clearing agents, for example, are discussed in Example IX. In addition, extracorporeal clearance systems are discussed in Example IX. In vivo clearance protocols employing arterially inserted proteinaceous or polymeric
multiloop devices are also described in Example IX.
One embodiment of the present invention in which rapid acting clearing agents are useful is in the delivery of Auger emitters, such as 1-125, 1-123, Er-165, Sb-119, Hg-197, Ru-97, Tl-201 and Br-77, or nucleus-binding drugs to target cell nuclei. In these embodiments of the present invention, targeting moieties that localize to internalizing receptors on target cell surfaces are employed to deliver a
targeting moiety-containing conjugate (i.e., a targeting moiety-anti-ligand -conjugate in the
preferred two-step protocol) to the target cell population. Such internalizing receptors include EGF receptors, transferrin receptors, HER2 receptors, IL-2 receptors, other interleukins and cluster
differentiation receptors, somatostatin receptors, other peptide binding receptors and the like.
After the passage of a time period sufficient to achieve localization of the conjugate to target cells, but insufficient to induce internalization of such targeted conjugates by those cells through a receptor- mediated event, a rapidly acting clearing agent is administered. In a preferred two-step protocol, an active agent-containing ligand or anti-ligand
conjugate, such as a biotin-Auger emitter or a biotin- nucleus acting drug, is administered as soon as the clearing agent has been given an opportunity to complex with circulating targeting moiety-containing conjugate, with the time lag between clearing agent and active agent administration being less than about 24 hours. In this manner, active agent is readily internalized through target cell receptor-mediated internalization. While circulating Auger emitters are thought to be non-toxic, the rapid, specific targeting afforded by the pretargeting protocols of the present invention increases the potential of shorter half-life Auger emitters, such as 1-123, which is available and capable of stable binding.
The 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''- tetra acetic acid (DOTA) -biotin conjugate (DOTA-LC- biotin) depicted below has been reported to have desirable in vivo biodistribution and is cleared primarily by renal excretion.
Figure imgf000044_0001
DOTA may also be conjugated to other ligands or to anti-ligands in the practice of the present invention.
Because DOTA strongly binds Y-90 and other
radionuclides, it has been proposed for use in
radioimmunotherapy. For therapy, it is very important that the radionuclide be stably bound within the DOTA chelate and that the DOTA chelate be stably attached to biotin. Only radiolabeled DOTA-biotin conjugates exhibiting those two characteristics are useful to deliver radionuclides to the targets. Release of the radionuclide from the DOTA chelate or cleavage of the biotin and DOTA conjugate components in serum or at non-target sites renders the conjugate unsuitable for use in therapy.
Serum stability of DOTA-LC-biotin (where LC refers to the "long chain" linker, including an aminocaproyl spacer between the biotin and. the DOTA conjugate components) shown above, while reported in the
literature to be good, has proven to be problematic. Experimentation has revealed that DOTA-LC-biotin is rapidly cleared from the blood and excreted into the urine as fragments, wherein the biotinamide bond rather than the DOTA-amide bond has been cleaved, as shown below.
Figure imgf000045_0001
Additional experimentation employing PIP-biocytin conjugates produced parallel results as shown below.
Figure imgf000046_0001
Cleavage of the benzamide was not observed as
evidenced by the absence of detectable quantities of iodobenzoic acid in the serum.
It appears that the cleavage results from the action of serum biotinidase. Biotinidase is a
hydrolytic enzyme that catalyzes the cleavage of biotin from biotinyl peptides. See, for example, Evangelatos, et al., "Biotinidase Radioassay Using an I-125-Biotin Derivative, Avidin, and Polyethylene
Glycol Reagents," Analytical Biochemistry, 196 : 385-89, 1991.
Drug-biotin conjugates which structurally resemble biotinyl peptides are potential substrates for
cleavage by plasma biotinidase. Poor in. vivo
stability therefore limits the use of drug-biotin conjugates in therapeutic applications. The use of peptide surrogates to overcome poor stability of peptide therapeutic agents has been an area of intense research effort. See, for example, Spatola, Peptide Backbone Modification: A Structure-Activity Analysis of Peptide Containing Amide Bond Surrogates,
"Chemistry and Biochemistry of Amino Acids, Peptides and Proteins," vol. 7, Weinstein, ed., Marcel Dekker, New York, 1983; and Kim et al., "A New Peptide Bond Surrogate: 2-Isoxazoline in Pseudodipeptide
Chemistry," Tetrahedron Letters, 45: 6811-14, 1991.
Elimination of the aminocaproyl spacer of DOTA-LC-biotin gives DOTA-SC-biotin (where the SC indicates the "short chain" linker between the DOTA and biotin conjugate components), which molecule is shown below:
Figure imgf000047_0001
DOTA-SC-biotin exhibits significantly improved serum stability in comparison to DOTA-LC-biotin. This result does not appear to be explainable on the basis of biotinidase activity alone. The experimentation leading to this conclusion is summarized in the Table set forth below.
Time Dependent Cleavage of DOTA-Biotin Conjugates
Time at 37°C % Avidin Binding
PIP-Biocytin Y-90-LC Y-90-SC
DOTA-Biotin DOTA-Biotin
5 Minutes 75% 50% -
15 Minutes 57% 14% -
30 Minutes 31% 12% -
6600 MMiinnuutteess - 00%% 98%
20 Hours - 0% 60% where "-" indicates that the value was not measured.
The difference in serum stability between DOTA-LC-biotin and DOTA-SC-biotin might be explained by the fact that the SC derivative contains an aromatic amide linkage in contrast to the aliphatic amide linkage of the LC derivative, with the aliphatic amide linkage being more readily recognized by enzymes as a
substrate therefor. This argument cannot apply to biotinidase, however, because biotinidase very
efficiently cleaves aromatic amides. In fact, it is recognized that the simplest and most commonly
employed biotinidase activity measuring method uses N-(d-biotinyl)-4-aminobenzoate (BPABA) as a substrate, with the hydrolysis of BPABA resulting in the liberation of biotin and 4-aminobenzoate (PABA). See, for example, B. Wolf, et al., "Methods in Enzymology," pp. 103-111, Academic Press Inc., 1990. Consequently, one would predict that DOTA-SC-biotin, like its LC counterpart, would be a biotinidase substrate. Since DOTA-SC-biotin exhibits serum stability, biotinidase activity alone does not adequately explain why some conjugates are serum stable while others are not. A series of DOTA-biotin conjugates was therefore
synthesized by the present inventors to determine which structural features conferred serum stability to the conjugates.
Some general strategies for improving serum stability of peptides with respect to enzymatic action are the following: incorporation of D-amino acids, N-methyl amino acids and alpha-substituted amino acids.
In vivo stable biotin-DOTA conjugates are useful within the practice of the present invention. In vivo stability imparts the following advantages:
1) increased tumor uptake in that more of the radioisotope will be targeted to the previously localized targeting moiety-streptavidin; and
2) increased tumor retention, if biotin is more stably bound to the radioisotope.
In addition, the linkage between DOTA and biotin may also have a significant impact on biodistribution (including normal organ uptake, target uptake and the like) and pharmacokinetics.
The strategy for design of the DOTA-containing molecules and conjugates of the present invention involved three primary considerations:
1) in vivo stability (including biotinidase and general peptidase activity resistance), with an initial cut of 100% stability for 1 hour;
2) renal excretion; and
3) ease of synthesis. The DOTA-biotin conjugates of the present invention reflect the implementation of one or more of the following strategies:
1) substitution of the carbon adjacent to the cleavage susceptible amide nitrogen;
2) alkylation of the cleavage susceptible amide nitrogen;
3) substitution of the amide carbonyl with an alkyl amino group;
4) incorporation of D-amino acids as well as analogs or derivatives thereof; or
5) incorporation of thiourea linkages.
DOTA-biotin conjugates in accordance with the present invention may be generally characterized as follows: conjugates that retain the biotin carboxy group in the structure thereof and those that do not (i.e., the terminal carboxy group of biotin has been reduced or otherwise chemically modified. Structures of such conjugates represented by the following
general formula have been devised:
Figure imgf000049_0001
wherein L may alternatively be substituted in one of the following ways on one of the -CH2-COOH branches of. the DOTA structure: -CH(L)-COOH or -CH2COOL or
-CH2COL). When these alternative structures are employed, the portion of the linker bearing the
functional group for binding with the DOTA conjugate component is selected for the capability to interact with either the carbon or the carboxy in the branch portions of the DOTA structure, with the serum
stability conferring portion of the linker structure being selected as described below. In the case where the linkage is formed on the core of the DOTA structure as shown above, L is selected according to the following principles, with the portion of the linker designed to bind to the DOTA conjugate component selected for the capability to bind to an amine.
A. One embodiment of the present invention
includes linkers incorporating a D-amino acid spacer between a DOTA aniline amine and the biotin carboxy group shown above. Substituted amino acids are preferred for these embodiments of the present
invention, because alpha-substitution also confers enzymatic cleavage resistance. Exemplary L moieties of this embodiment of the present invention may be represented as follows:
Figure imgf000050_0002
where R1 is selected from lower alkyl, lower alkyl substituted with hydrophilic groups (preferably,
(CH2)n-OH, (CH2)n-OSO3, (CH2)n-SO3, , where n
Figure imgf000050_0001
is 1 or 2), glucuronide-substituted amino acids or other glucuronide derivatives; and
R2 is selected from hydrogen, lower alkyl,
substituted lower alkyl (e.g., hydroxy, sulfate, phosphonate or a hydrophilic moiety (preferably OH).
For the purposes of the present disclosure, the term "lower alkyl" indicates an alkyl group with from one to five carbon atoms. Also, the term
"substituted" includes one or several substituent groups, with a single substituent group preferred.
Preferred L groups of this embodiment of the present invention include the following:
R1 = CH3 and R2 = H (a D-alanine derivative, with a synthetic scheme therefor shown in Example XXI); R1 = CH3 and R2 = CH3 (an N-methyl-D-alanine derivative);
R1 = CH2-OH and R2 = H (a D-serine derivative);
R1 = CH2OSO3 and R2 = H (a D-serine-O-sulfate-derivative); and
and R2 = H ( a D- serine -O-
Figure imgf000051_0001
phosphonate-derivative);
Other preferred moieties of this embodiment of the present invention include molecules wherein R1 is hydrogen and R2 = -(CH2)nOH or a sulfate or
phosphonate derivative thereof and n is 1 or 2 as well as molecules wherein R1 is ; or —(CH2)5-NH2 ; or
Figure imgf000051_0002
; when R2 = H
Figure imgf000051_0003
Preferred moieties incorporating the glucuronide of D-lysine and the glucuronide of amino pimelate are shown below as I and II, respectively.
Figure imgf000051_0004
Figure imgf000051_0005
A particularly preferred linker of this embodiment of the present invention is the D-alanine derivative set forth above. B. Linkers incorporating alkyl substitution on one or more amide nitrogen atoms are also encompassed by the present invention, with some embodiments of such linkers preparable from L-amino acids. Amide bonds having a substituted amine moiety are less susceptible to enzymatic cleavage. Such linkers exhibit the following general formula:
Figure imgf000052_0001
where R4 is selected from hydrogen, lower alkyl, lower alkyl substituted with hydroxy, sulfate, phosphonate or the like and ;
Figure imgf000052_0002
R3 is selected from hydrogen; an amine; lower alkyl; an amino- or a hydroxy-, sulfate- or
phosphonate-substituted lower alkyl; a glucuronide or a glucuronide-derivatized amino groups; and
n ranges from 0-4.
Preferred linkers of this embodiment of the present invention include:
R3 = H and R4 = CH3 when n = 4, synthesizable as discussed in Example XXI;
R3 = H and R4 = CH3 when n = 0, synthesizable from N-methyl-glycine (having a trivial name of sarcosine) as described in Example XXI;
R3 = NH2 and R4 = CH3 , when n = 0;
R3 = H and R4 = when
Figure imgf000052_0003
n = 4 (Bis-DOTA-LC-biotin), synthesizable from
bromohexanoic acid as discussed in Example XXI; and
R3 = H and R4 = when
Figure imgf000052_0004
n = 0 (bis-DOTA-SC-biotin), synthesizable from
iminodiacetic acid.
The synthesis of a conjugate including a linker wherein R3 is H and R4 is -CH2CH2OH and n is 0 is also described in Example XXI. Schematically, the
synthesis of a conjugate of this embodiment of the present invention wherein n is 0, R3 is H and R4 is -CH2-COOH is shown below.
Figure imgf000053_0001
Bis-DOTA-LC-biotin, for example, offers the following advantages:
1) incorporation of two DOTA molecules on one biotin moiety increases the overall hydrophilicity of the biotin conjugate and thereby directs in vivo distribution to urinary excretion; and
2) substitution of the amide nitrogen adjacent to the biotin carboxyl group blocks peptide and/or biotinidase cleavage at that site.
Bis-DOTA-LC-biotin, the glycine-based linker and the N-methylated linker where R3 = H, R4 = CH3, n = 4 are particularly preferred linkers of this embodiment of the present invention. C. Another linker embodiment incorporates a thiourea moiety therein. Exemplary thiourea adducts of the present invention exhibit the following general formula :
Figure imgf000054_0003
where R5 is selected from hydrogen or lower alkyl;
R6 is selected from H and a hydrophilic moiety; and n ranges from 0-4.
Preferred linkers of this embodiment of the
present invention are as follows:
R5 = H and R6 = H when n = 5;
R5 = H and R6 = COOH when n = 5; and
R5 = CH3 and R6 = COOH when n = 5.
The second preferred linker recited above can be prepared using either L-lysine or D-lysine.
Similarly, the third preferred linker can be prepared using either N-methyl-D-lysine or N-methyl-L-lysine.
An.ther thiourea adduct of minimized lipophilicity is
Figure imgf000054_0002
which may be formed via the addition of
biotinhydrazide (commercially available from Sigma Chemical Co., St. Louis, Missouri) and DOTA-benzylisothiocyanate (a known compound synthesized in one step from DOTA-aniline), with the thiourea-containing compound formed as shown below.
Figure imgf000054_0001
Figure imgf000055_0004
D. Amino acid-derived linkers of the present invention with substitution of the carbon adjacent to the cleavage susceptible amide have the general formula set forth below:
Figure imgf000055_0003
wherein Z is -(CH2)2-, conveniently synthesized form glutamic acid; or
Z = -CH2-S-CH2-, synthesizable from cysteine and iodo-acetic acid; or
Z = -CH2-, conveniently synthesized form aspartic acid; or
Z = - (CH2)n-CO-O-CH2-, where n ranges from 1-4 and which is synthesizable from serine.
E. Another exemplary linker embodiment of the present invention has the general formula set forth below:
Figure imgf000055_0002
and n ranges from 1-5.
F. Another embodiment involves disulfide-contaicing linkers, which provide a metabolically cleavable moiety (-S-S-) to reduce non-target
retention of the biotin-DOTA conjugate. Exemplary linkers of this type exhibit the following formula:
Figure imgf000055_0001
where n and n' preferably range between 0 and 5 The advantage of using conditionally cleavable linkers is an improvement in target/non-target
localization of the active agent. Conditionally cleavable linkers include enzymatically cleavable linkers, linkers that are cleaved under acidic
conditions, linkers that are cleaved under basic conditions and the like. More specifically, use of linkers that are cleaved by enzymes, which are present in non-target tissues but reduced in amount or absent in target tissue, can increase target cell retention of active agent relative to non-target cell retention. Such conditionally cleavable linkers are useful, for example, in delivering therapeutic radionuclides to target cells, because such active agents do not require internalization for efficacy, provided that the linker is stable at the target cell surface or protected from target cell degradation.
G. Ether, thioether, ester and thioester linkers are also useful in the practice of the present
invention, because such linkages are acid cleavable and therefore facilitate improved non-target
retention. Exemplary linkers of this type have the following general formula:
Figure imgf000056_0001
where X is O or S; and
Q is a bond, a methylene group, a -CO- group or -CO- (CH2)n-NH-; and
n ranges from 1-5.
Other such linkers have the general formula:
-CH2-X-Q, where Q and X are defined as set forth above.
H. Another amino-containing linker of the present invention is structured as follows: , where R7 is lower alkyl,
Figure imgf000057_0001
preferably methyl.
In this case, resistance to enzymatic cleavage is conferred by the alkyl substitution on the amine.
I. Polymeric linkers are also contemplated by the present invention. Dextran and cyclodextran are preferred polymers useful in this embodiment of the present invention as a result of the hydrophilicity of the polymer, which leads to favorable excretion of conjugates containing the same. Other advantages of using dextran polymers are that such polymers are substantially non-toxic and non-immunogenic, that they are commercially available in a variety of sizes and that they are easy to conjugate to other relevant molecules. Also, dextran-linked conjugates exhibit advantages when non-target sites are accessible to dextranase, an enzyme capable of cleaving dextran polymers into smaller units while non-target sites are not so accessible.
Other linkers of the present invention are
produced prior to conjugation to DOTA and following the reduction of the biotin carboxy moiety. These linkers of the present invention have the following general formula:
Figure imgf000057_0002
Embodiments of linkers of this aspect of the present invention include the following:
J. An ether linkage as shown below may be formed in a DOTA-biotin conjugate in accordance with the procedure indicated below.
L' = -NH-CO-(CH2)n-O- where n ranges from 1 to 5 , with 1 preferred.
Figure imgf000058_0001
This linker has only one amide moiety which is bound directly to the DOTA aniline (as in the structure of DOTA-SC-biotin). In addition, the ether linkage imparts hydrophilicity, an important factor in
facilitating renal excretion.
K. An amine linker formed from reduced biotin (hydroxybiotin or aminobiotin) is shown below, with conjugates containing such a linker formed, for example, in accordance with the procedure described in Example XXI.
L' = -NH-
This linker contains no amide moieties and the
unalkylated amine may impart favorable biodistribution properties since unalkylated DOTA-aniline displays excellent renal clearance. L. Substituted amine linkers, which can form conjugates via amino-biotin intermediates, are shown below.
Figure imgf000059_0001
where R8 is H; -(CH2)2-OH or a sulfate or phosphonate
derivative thereof; or ; or the like;
Figure imgf000059_0002
and R9 is a bond or -(CH2)n-CO-NH-, where n ranges from
0-5 and is preferably 1 and where q is 0 or 1.
These moieties exhibit the advantages of an amide only directly attached to DOTA-aniline and either a non-amide amine imparting a positive charge to the linker in vivo or a N-alkylated glucuronide hydrophilic group, each alternative favoring renal excretion.
M. Amino biotin may also be used as an
intermediate in the production of conjugates linked by linkers having favorable properties, such as a
thiourea-containing linker of the formula:
L' = -NH-CS-NH-
Conjugates containing this thiourea linker have the following advantages: no cleavable amide and a short, fairly polar linker which favors renal excretion.
A bis-DOTD derivative of the following formula can also be formed from amino-biotin.
Figure imgf000059_0003
where n ranges from 1 to 5, with 1 and 5 preferred. This molecule offers the advantages of the previously discussed bis-DOTA derivatives with the added
advantage of no cleavable amides.
Additional linkers of the present invention which are employed in the production of conjugates
characterized by a reduced biotin carboxy moiety are the following:
L = -(CH2)4-NH-, wherein the amine group is
attached to the methylene group corresponding to the reduced biotin carboxy moiety and the methylene chain is attached to a core carbon in the DOTA ring. Such a linker is conveniently synthesizable from lysine.
L = -(CH2) -CO-NH-, wherein q is 1 or 2, and wherein the amine group is attached to the methylene group corresponding to the reduced biotin carboxy moiety and the methylene group (s) are attached to a core carbon in the DOTA ring. This moiety is
synthesizable from amino-biotin.
The linkers set forth above are useful to produce conjugates having one or more of the following
advantages:
- bind avidin or streptavidin with the same or substantially similar affinity as free biotin;
- bind metal M+3 ions efficiently and with high kinetic stability;
- are excreted primarily through the kidneys into urine;
- are stable to bodily fluid amidases;
- penetrate tissue rapidly and bind to pretargeted avidin or streptavidin; and
- are excreted rapidly with a whole body residence half-life of less than about 5 hours.
Synthetic routes to an intermediate of the DOTA-biotin conjugates depicted above, nitrobenzyl-DOTA, have been. proposed. These proposed synthetic routes produce the intermediate compound in suboptimal yield, however. For example, Renn and Meares, "Large Scale Synthesis of Bifunctional Chelating Agent Q-(p-nitrobenzyl)-1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetra acetic acid, and the Determination of its Enantiomeric Purity by Chiral Chromatography," Bioconj. Chem., 3: 563-9, 1992, describe a nine-step synthesis of nitrobenzyl-DOTA, including reaction steps that either proceed in low yield or involve cumbersome transformations or purifications. More specifically, the sixth step proceeds in only 26% yield, and the product must be purified by preparative HPLC. Additionally, step eight proceeds in good yield, but the process involves copious volumes of the coreactants.
These difficulties in steps 6-8 of the prior art synthesis are overcome in the practice of the present invention through the use of the following synthetic alternative therefor.
Figure imgf000061_0001
The poor yield in step six of the prior art synthesis procedure, in which a tetra amine alcohol is converted to a tetra-toluenesulfonamide
toluenesulfonate as shown below, is the likely result of premature formation of the toluenesulfonate functionality (before all of the amine groups have been converted to their corresponding sulfonamides.
Figure imgf000062_0001
Such a sequence of events would potentially result in unwanted intra- or inter-molecular displacement of the reactive toluene sulfonate by unprotected amine groups, thereby generating numerous undesirable side-products.
This problem is overcome in the aforementioned alternative synthesis scheme of the present invention by reacting the tetra-amine alcohol with
trifluoroacetic anhydride. Trifluoroacetates, being much poorer leaving groups than toluenesulfonates, are not vulnerable to analogous side reactions. In fact, the easy hydrolysis of trifluoroacetate groups, as reported in Greene and Wuts, "Protecting Groups in Organic Synthesis," John Wiley and Sons, Inc., New York, p. 94, 1991., suggests that addition of methanol to the reaction mixture following consumption of all amines should afford the tetra-fluoroacetamide alcohol as a substantially exclusive product. Conversion of the tetra-fluoroacetamide alcohol to the corresponding toluenesulfonate provides a material which is expected to cyclize analogously to the tetra-toluenesulfonamide toluenesulfonate of the prior art. The cyclic tetraamide product of the cyclization of the
toluenesulfonate of tetra-fluoroacetamide alcohol, in methanolic sodium hydroxide at 15-25°C for 1 hour, should afford nitro-benzyl-DOTA as a substantially exclusive product. As a result, the use of trifluoracetamide protecting groups circumvents the difficulties associated with cleavage of the very stable toluenesulfonamide protecting group, which involves heating with a large excess of sulfuric acid followed by neutralization with copious volumes of barium hydroxide .
Another alternative route to nitro-benzyl-DOTA is shown below.
Figure imgf000063_0001
This alternative procedure involves the cyclizaton of p-nitrophenylalanyltriglycine using a coupling agent , such as diethylycyanophosphate , to give the cyclic tetraamide . Subsequent borane reduction provides 2 - (p-nitrobenzyl) -1 , 4 , 7 , 10-tetraazacyclododecane , a common precursor used in published routes to DOTA including the Renn and Meares article referenced above . This alternative procedure of the present invention offers a synthetic pathway that is
considerably shorter than the prior art Renn and Meares route , requiring two rather than four steps
Figure imgf000064_0001
The resultant chelate-biotin conjugate shows superior kidney excretion. Although the net overall charge of the conjugate is neutral, the polycarboxylate nature of the molecule generates regions of hydrophilicity and hydrophobicity. By altering the number and nature of the carboxylate groups within the conjugate, excretion may be shifted from kidney to
gastrointestinal routes. For instance, neutral compounds are generally cleared by the kidneys;
anionic compounds are generally cleared through the GI system.
B. Polylysine Derivitization
Conjugates containing polylysine may also exhibit beneficial biodistribution properties. With whole antibodies, derivitization with polylysine may skew the biodistribution of conjugate toward liver uptake. In contrast, derivitization of Fab fragments with polylysine results in lower levels of both liver and kidney uptake; blood clearance of these conjugates is similar to that of Fab covalently linked to chelate. An exemplary polylysine derivitized chelate-biotin conjugate is illustrated below.
Figure imgf000065_0001
Inclusion of polylysine in radiometal-chelate-biotin conjugates is therefore useful for minimizing or eliminating RES sequestration while maintaining good liver and kidney clearance of the conjugate. For improved renal excretion properties, polylysine derivatives are preferably succinylated following biotinylation. Polylysine derivatives offer the further advantages of: (1) increasing the specific activity of the radiometal-chelate-biotin conjugate; (2) permitting control of rate and route of blood clearance by varying the molecular weight of the polylysine polymer; and (3) increasing the circulation half-life of the conjugate for optimal tumor
interaction.
Polylysine derivitization is accomplished by standard methodologies. Briefly, poly-L-lysine is acylated according to standard amino group acylation procedures (aqueous bicarbonate buffer, pH 8, added biotin-NHS ester, followed by chelate NHS ester).
Alternative methodology involves anhydrous conditions using nitrophenyl esters in DMSO and triethyl amine. The resultant conjugates are characterized by UV and NMR spectra.
The number of biotins attached to polylysine is determined by the HABA assay. Spectrophotometric titration is used to assess the extent of amino group derivitization. The radiometal-chelate-biotin
conjugate is characterized by size exclusion.
C. Cleavable Linkage
Through insertion of a cleavable linker between the chelate and biotin portion of a radiometal- chelate-biotin conjugate, retention of the conjugate at the tumor relative to normal tissue may be
enhanced. More specifically, linkers that are cleaved by enzymes present in normal tissue but deficient or absent in tumor tissue can increase tumor retention. As an example, the kidney has high levels of γ-glutamyl transferase; other normal tissues exhibit in vivo cleavage of γ-glutamyl prodrugs. In contrast, tumors are generally deficient in enzyme peptidases.
The glutamyl-linked biotin conjugate depicted below is cleaved in normal tissue and retained in the tumor.
Figure imgf000066_0001
* D. Serine Linker With O-Polar Substituent
Sugar substitution of N3S chelates renders such chelates water soluble. Sulfonates, which are fully ionized at physiological pH, improve water solubility of the chelate-biotin conjugate depicted below.
Figure imgf000067_0001
This compound is synthesized according to the standard reaction procedures. Briefly, biocytin is condensed with N-t-BOC-(O-sulfonate or O-glucose) serine NHS ester to give N-t-BOC-(O-sulfonate or O-glucose) serine biocytinamide. Subsequent cleavage of the N-t-BOC group with TFA and condensation with ligand NHS ester in DMF with triethylamine provides ligand-amidoserine (O-sulfonate or O-glucose) biocytinamide.
Example VII
Preparation and Characterization of PIP- Radioiodinated Biotin
Radioiodinated biotin derivatives prepared by exposure of poly-L-lysine to excess NHS-LC-biotin and then to Bolton-Hunter N-hydroxysuccinimide esters in
DMSO has been reported. After purification, this product was radiolabeled by the iodogen method (see, for instance, Del Rosario et al., J. Nucl. Med. 32:5, 1991, 993 (abstr.)). Because of the high molecular weight of the resultant radioiodinated biotin
derivative, only limited characterization of product
(i.e., radio-HPLC and binding to immobilized
streptavidin) was possible. Preparation of radioiodinated biotin according to the present invention provides certain advantages. First, the radioiodobiotin derivative is a low
molecular weight compound that is amenable to complete chemical characterization. Second, the disclosed methods for preparation involve a single step and eliminate the need for a purification step.
Briefly, iodobenzamide derivatives corresponding to biocytin (R = COOH) and biotinamidopentylamine (R = H) were prepared according to the following scheme. In this scheme, "X" may be any radiohalogen, including 125I, 131I, 123I, 211At and the like.
Figure imgf000068_0001
Preparation of 1 was generally according to Wilbur et al., J. Nucl. Med. 30:216-26, 1989, using a
tributyltin intermediate. Water soluble carbodiimide was used in the above-depicted reaction, since the NHS ester 1 formed intractable mixtures with DCU. The NHS ester was not compatible with chromatography; it was insoluble in organic and aqueous solvents and did not react with biocytin in DMF or in buffered aqueous acetonitrile. The reaction between 1 and biocytin or delivered therefore becomes the biotin binding
capacity at the target (i.e., the amount of antibody at the target and the avidin equivalent attached thereto).
For these embodiments of the pretargeting methods of the present invention, particle emitting
therapeutic radionuclides resulting from transmutation processes (without non-radioactive carrier forms present) are preferred. Exemplary radionuclides include Y-90, Re-188, At-211, Bi-212 and the like.
Other reactor-produced radionuclides are useful in the practice of these embodiments of the present
invention, if they are able to bind in amounts
delivering a therapeutically effective amount of radiation to the target. A therapeutically effective amount of radiation ranges from about 1500 to about 10,000 cGy depending upon several factors known to nuclear medicine practitioners.
Intraarterial administration pretargeting can be applied to targets present in organs or tissues for which supply arteries are accessible. Exemplary applications for intraarterial delivery aspects of the pretargeting methods of the present invention include treatment of liver tumors through hepatic artery administration, brain primary tumors and metastases through carotid artery administration, lung carcinomas through bronchial artery administration and kidney carcinomas through renal artery administration.
Intraarterial administration pretargeting can be conducted using chemotherapeutic drug, toxin and anti-tumor active agents as discussed below. High potency drugs, lymphokines, such as IL-2 and tumor necrosis factor, drug/lymphokine-carrier-biotin molecules, biotinylated drugs/lymphokines, and drug/lymphokine/ toxin-loaded, biotin-derivitized liposomes are exemplary of active agents and/or dosage forms useful for the delivery thereof in the practice of this embodiment of the present invention.
A problem associated with solid tumor target sites, for example, is penetration of the therapeutic agent into the active site. If homogeneous
penetration of such target sites is achieved, more effective therapy is possible. One method that may be employed to enhance target site penetration of ligand or anti-ligand conjugated to a targeting agent or anti-ligand or ligand conjugated to a therapeutic moiety is the administration of a permeability
enhancing moiety that induces or promotes vascular leakiness, disrupts cell-to-cell associations or similarly facilitates more homogeneous delivery of an administered conjugate to a target site characterized by a three dimensional cellular array. Preferably, permeability enhancing moieties of the present
invention achieve their effects rapidly (e.g., within from about 30 seconds to about 1 hour following administration).
One example of this aspect of the present
invention involves anti-ligand-targeting moiety conjugate administration (e.g., systemically, intra-arterially, locally via catheter or the like)
followed, for example, by intravascular perfusion catheter administration of a permeability enhancing moiety (e.g., an agent that induces gaps in the endothelium of venules through action on the
postcapillary venules such as histamine, serotonin or bradykinin; or through action on the entire capillary bed such as bacterial endotoxins; or the like) which disrupts the target three dimensional structure and induces microvascular leakiness, thereby permitting simultaneously or subsequently administered ligand-therapeutic agent conjugates to achieve a higher interstitial concentration at the target site. One method to achieve site specificity for the therapeutic agent conjugate administration is to administer that agent via catheter. The optimal route of
administration for each administered component will be dictated by the recipient's physiological condition and the specific treatment selected therefor by an experienced medical practitioner. Optionally, a clearing agent may be administered prior or subsequent to the permeability enhancing moiety, with prior administration preferred.
The use of this aspect of the present invention also facilitates the delivery of higher molecular weight therapeutic agent-bearing conjugates to target sites characterized by a three dimensional cellular array. For example, one or more ligand or anti-ligand molecules as well as a plurality of therapeutic agent molecules may be conjugated to a polymer to form an entity of the following formula:
Therapeutic Agent1-30-Polymer-Ligand or
Anti-ligand 1 -10
A specific embodiment of this aspect of the present invention, useful particularly for target sites characterized by an accessible (for
administration purposes) arterial supply as discussed herein, involves intraarterial administration of permeability enhancing agent followed by such
administration of therapeutic agent-polymer-ligand or -anti-ligand conjugate. An exemplary conjugate suitable for such delivery is a biotinylated
derivative of the hydroxy-propylmethacrylate-adriamycin conjugate discussed by R. Duncan and J. Kopecek, Advances in Polymer Science, 57 : 52-101 (1984) and R. Duncan, Anticancer Drugs. 3 : 175-210 (1992).
Alternatively, the permeability enhancing moiety may be administered prior to administration of the targeting moiety-ligand or -anti-ligand conjugate to facilitate more homogenous targeting of that
administered conjugate which serves as a receptor for subsequently administered anti-ligand- or ligand-therapeutic agent conjugate. Additionally,
permeability enhancing moieties may also be employed in three-step pretargeting protocols of the present invention.
Preferred permeability enhancing moieties of the present invention induce gaps in the endothelium of venules through "specific" or "non-specific"
interaction. For the purposes of this discussion, non-specific permeability enhancing moieties are those that act upon the entire capillary bed. Exemplary permeability enhancing moieties of the non-specific type are bacterial endotoxins, metabolic inhibitors, drugs that alter microfilaments or microtubules and the like. Specific permeability enhancing moieties act on a portion of the capillary bed (e.g., on the postcapillary venules, capillaries or the like).
Exemplary permeability enhancing agents of the specific type are histamine, serotonin, bradykinin, and the like. Other permeability enhancing moieties are mannitol, tumor necrosis factor, nitric oxide, prostaglandin E2, leukotriene, leukokinin and the like.
Alternatively, enhanced delivery of a diagnostic or therapeutic substance, for example, into a target tumor mass, can be achieved by systemically
administering a conjugate containing a targeting moiety, a member of a ligand/anti-ligand binding pair and an amount of a permeability enhancing agent sufficient to achieve disruption of cell-to-cell association within the tumor mass. Such a
permeability enhancing moiety may also be locally administered, for example, in the practice of the local or intraarterial administration protocols described above.
Exemplary permeability enhancing moieties of the present invention include any substance capable of disrupting cell-to-cell association within a three dimensional target cell array. Some preferred
moieties include trichothecenes, which are small molecule protein synthesis inhibitors, ionophores, membrane-active compounds (particularly inhibitors of microtubule or microfilament polymerization,
polymyxins and polyene antibiotics), cyclic toxin peptides, cytochalasins and combinations thereof.
Verrucarin A, microcystin (from Microcystin
aeruσinosa), cycloheximide and puromycin are
particularly preferred permeability enhancing
moieties.
Moieties possessing a macrocyclic ring, a
functional, epoxide group or both are also preferred within the present invention. However, cytotoxic compounds that are devoid of macrocyclic rings may also be suitable for use herein.
Small permeability enhancing moieties having a molecular weight less than or equal to about 2,000 daltons, and particularly those having a molecular weight less than or equal to 1,000 daltons, are also preferred. Such small permeability enhancing moieties are able to pass freely between adjacent cells whose cytoplasms are coupled via gap junctional complexes.
In a systemic administration embodiment, a
permeability enhancing moiety having a molecular weight of about 1,000 daltons or less is conjugated to a targeting moiety (preferably also having a ligand or an anti-ligand bound thereto) through a selectively cleavable covalent linkage. In this embodiment, after delivery of the conjugate to a target cell, the permeability enhancing moiety is released at the target cell, and is then distributed to adjacent cells through gap junctions. Free diffusion of the released moiety will increase the efficiency, for example, of tumor mass disruption.
For some permeability enhancing moieties, it may be advantageous to conjugate multiple moieties to a carrier molecule through a selectively cleavable covalent linkage. As used herein, "carrier molecule" includes, but is not limited to, a large protein or polymer capable of binding many permeability enhancing moieties to a single targeting moiety. Preferred carrier molecules include albumin, dextran, hydroxy-propylmethacrylamide, poly-L-lysine and polyglutamate, all of which have a molecular weight in excess of 5,000 daltons. Carrier molecules may also be produced through chemical syntheses or recombinant DNA
technology. A carrier molecule generally is
derivatized with small selectively cleavable linking groups, which enable binding of one or more
permeability enhancing moieties to the carrier. The "permeability enhancing moiety loaded" carrier is then either locally or intraarterially administered or systemically administered after being covalently attached to a targeting moiety (directly or through a linker) .
Alternatively, specific target site vascular permeability is achievable by administering an
effective amount of a permeability enhancing moiety which is a target cell inflammatory response mediator. A preferred permeability enhancing moiety of this type is a substance that is capable of localizing at a target site and capable of mediating complement- dependent inflammation at the target site within a patient. Such moieties are recognized by their ability to mediate complement-dependent cytotoxicity in vitro with the recipient's serum. Permeability enhancing moieties may be monoclonal antibodies, polyclonal antibodies, chimeric antibodies, human or humanized antibodies or other moiety having the capabilities described above.
Permeability enhancing moieties of this type may also be administered as a conjugate with a vasoactive complement-derived peptide, such as a peptide derived from C2. Complement-derived peptides are linked to the permeability enhancing moieties in a manner permitting release of the complement-derived peptide at the target site, such as through a labile linkage (i.e., a Schiff base linkage).
Preferably, a subset of antibodies, such as mouse antibodies, human/mouse chimeric antibodies or humanized antibodies are used to induce target site-specific increases in vascular permeability. These monoclonal antibodies mediate target cell lysis and/or target cell inflammatory responses in vitro which contribute to target site-specific vascular
permeability increases. Both direct and indirect action on target cells may be accomplished by
mediation of complement-dependent cytotoxicity. The ability to mediate complement-dependent cytotoxicity in vitro is indicative of the ability to mediate complement-dependent inflammation in vivo.
The invention exploits one attribute of antibodies ╌ possession of isotype- and/or subclass-specific functions. For example, murine IgG3 and human/mouse chimeric antibodies having human IgG, or IgG3 constant portions are generally considered superior to other immunoglobulins of murine or murine/human nature for mediating antibody-dependent cell-mediated
inflammation (ADCC) or complement-dependent cellular toxicity. Antibodies or other moieties effective in mediating target-site complement-dependent
cytotoxicity must be capable of utilizing the serum complement native to the ultimate recipient having confirmed br potential target sites. Consequently, human antibodies are expected to be capable of utilizing human serum. Antibodies of other types are therefore tested for this ability when administration to human recipients is contemplated.
Exemplary moieties known to mediate dependent cytotoxicity are NR-CO-04, a murine IgG3 antibody directed to colon carcinoma; NR-LU-13, a murine/human chimeric antibody featuring a human constant region and murine variable region of NR-LU-10, an antibody directed to a 37-40 kilodalton pancarcinoma
glycoprotein; NR-LU-03, an IgG3 antibody generated by immune complex immunization with NR-LU-10; R24, an IgG3 antibody directed against GD3 disclosed by Houghton et al., Proc. Natl. Acad. Sci (USA). 82: 1242-1246, 1985; antibody 3F8, a mouse IgG3 directed against GD2
disclosed by Munn et al., Cancer Res., 47:6600-6605, 1987 and Cheung et al., J. Clin. Invest., 81:1122-1128, 1988; and the like.
Some polymers useful in the practice of this permeability enhancing aspect of the present invention serve solely as a carrier for multiple therapeutic agents. Preferred polymers also direct the
biodistribution of the ligand or anti-ligand and therapeutic agents to which the polymer is bound to renal rather than, for example, hepatobiliary
excretion. For many administered therapeutic agents (e.g., radionuclides), renal excretion is preferred, especially for therapeutic protocols.
Exemplary polymers for use in the practice of the present invention are excreted or which metabolites thereof are excreted through a renal pathway when administered to a mammalian recipient and that is capable of covalently or non-covalently binding to one or more drugs, anti-tumor agents, peptides, chelates, ligands, anti-ligands or other small molecules and imposing a renal route of excretion upon the
associated molecule(s). Such polymers include polar molecules having a molecular weight ranging from about 3 kD to about 70 kD).
Exemplary polymers useful in permeability
enhancing aspects of the present invention are
dextran; dextran derivatives including carboxymethyl dextran, anionic or polar derivatives thereof such as carboxymethy1-dextran, 3-mercapto-2-hydroxypropyl dextran and the like; hyaluronic acid, inulin,
carboxymethyl cellulose; hydroxy-propylmethacrylamide (HPMA) polymers; succinylated polylysine;
polyaspartate; polyglutamate; polyethyleneglycol
(PEG); and the like. Dextran polymers are described herein as the prototypical polymers for use in the permeability enhancing aspects of the invention.
For use in the present invention, dextran polymers preferably range between about 5 and about 15 kD in size, although larger moieties may also be used. When larger (from about 40 to about 70 kD) polymers are employed in accordance with the present invention, the rate of clearance of the conjugate from a recipient is slowed, but the renal pathway for excretion is
maintained. In this manner, the moieties bound to the polymer exhibit increased bioavailability as a result of the increased circulation time thereof. Such bound molecules remain directed to renal excretion, however.
Exemplary dextran-containing conjugates of the present invention are dextran-biotin conjugates.
Chelate-biotin-dextran conjugates of the present invention are formed, for example, from oxidized dextran by conjugating biocytin hydrazide thereto followed by reaction with a chelate active ester, such as a N-hydroxy succinimidyl ester, a tetrafluorophenyl ester and the like. Alternatively, chelate-biotin-dextran can be formed by reacting dextran hydrazide with the carboxy terminus of a chelate-biocytin conjugate. Preferably, the carboxy terminus has been derivitized to form an active ester, such as an N- hydroxysuccinimidyl ester or the like. an alternative protocol of general applicability to different
polymers is discussed in Example XVII.
Biotin-dextran, lysine fixable (BDLF, available from Sigma Chemical Company, St. Louis, Missouri) is a preferred polymer-ligand for use in the practice of the present invention.
Active agents can be bound to dextran polymers characterized by short term serum stable linkages disposed between monomeric, dimeric, trimeric or other convenient unit thereof. Such conjugates are also preferably large (exhibiting a molecular weight ranging from about 40 to about 70 kD). One advantage of the use of such polymers is that the molecules affixed thereto will exhibit an increased circulation time as well as decreased liver uptake. More
specifically, the circulation time of the bound molecules is dictated by the maintenance of polymeric structural integrity in vivo. Depolymerization releases bound molecule-monomer, -dimer, -trimer or like moieties that are themselves rapidly cleared from the recipient's circulation, preferably via the renal pathway.
Depolymerization may be controlled in any
convenient manner therefor, including, for example, the following methods:
- Use of linkages between dextran units
containing chemical groups that are stable in serum for a period of time sufficient to provide an
appropriate circulation time to the small molecules bound thereto (e.g., 1 to 3 hours for active agent in both the pretargeting and targeted, direct labeled protocols); or
- Use of linkages between dextran units that are enzymatically cleaved upon administration of enzyme after the passage of an appropriate amount of bound molecule circulation time. In the first approach, groups such as esters, acetals, disulfides, thioacetals or the like are employed. For example, an ester linkage, having a serum stability of 1-3 hours such as phenyl or
activated phenyl or phthalyl (e.g., chloro-substituted, fluoro-substituted, multi-halogen-substituted, nitro-substituted or the like), may be employed to attach dextran polymer units. Such a linkage is stable in serum for a time sufficient to facilitate localization of the molecules bound to the polymer to the target site.
The second approach includes the use of dextran units susceptible to cleavage by an administrable enzyme that is not found in large amounts in human serum. Exemplary enzymes useful in the practice of this aspect of the present invention include
dextranase, alpha-amylase, pullulanase (a bacterial alpha-1,6-polysaccharidase) and the like. The enzymes are administered by any convenient route in any convenient dosage form therefor. Such enzymes are optionally conjugated or formed as fusion proteins with long circulating proteins, including albumin, immunoglobulins or portions thereof, and the like. In this manner, the administered enzymes remain in circulation for a time sufficient to effect
depolymerization of the polymer.
As discussed previously, cross-linking of moieties bound to a target cell surface results in
internalization of those bound moieties by the cell. This cross-linking phenomena can be exploited in several ways using the 2-step and 3-step pretargeting protocols of the present invention. Three exemplary protocols of the cross-linking/internalization aspect of the present invention using the exemplary
biotin/avidin ligand/anti-ligand pair can be described as follows: 1) (two-step) administer targeting moiety-biotin-therapeutic agent conjugate (or biotin-targeting moiety-therapeutic agent conjugate); and
administer avidin or streptavidin to cross-link the previously localized conjugate. A variation of this protocol involves administration of targeting moiety-biotin conjugate followed by administration of therapeutic agent-avidin or -streptavidin conjugate.
2) (two-step) administer avidin- or
streptavidin-targeting moiety-therapeutic agent conjugate (or therapeutic agent-avidin- or
streptavidin-targeting moiety conjugate; or a
therapeutic agent-containing conjugate that also incorporates Antibody-(avidin or streptavidin)2 or (Antibody)2-avidin or -streptavidin); and
administer multiple biotin-polymer conjugate to cross-link the previously localized conjugate. A variation of this protocol involves administration of a targeting moiety-avidin or -streptavidin conjugate, Antibody-(avidin or streptavidin)-, conjugate or
(Antibody)2-avidin or -streptavidin conjugate followed by administration of multiple biotin-polymer
therapeutic agent conjugate.
3) (three-step) administer targeting moiety-biotin conjugate;
administer avidin or streptavidin;
administer multiple biotin-polymer-therapeutic agent conjugate to either cross-link the previously localized avidin or streptavidin or to be internalized into the cell as a result of the avidin or streptavidin cross-linking of previously localized biotin-containing conjugate.
These cross-linking/internalization embodiments of the present invention are exemplified below with reference to the biotin/avidin ligand/anti-ligand pair, monoclonal antibody targeting moieties and trichothecene therapeutic agents; however, the embodiments are amenable to use with other
administered components.
Exemplary simple trichothecenes useful in the practice of this and other aspects of the present invention are as follows:
Figure imgf000081_0003
wherein:
R1 is H, OH,
Figure imgf000081_0004
, SH or L1 , L2, L3, or L4;
R2 is H, OH, or
Figure imgf000081_0005
; R3 is H, OH, or
Figure imgf000081_0006
;
R4 is H, or forms an epoxide group with R5 ;
R5 is H, OH,
Figure imgf000081_0007
,
Figure imgf000081_0008
, SH,
OCH3, SCH3, NH2, NHCH3, N(CH3)2. L3, L4, or forms an epoxide group with R4 ; and R6 is H, OH, SH, OCH3 , SCH3 , NH2 , NHCH3 , N(CH3)2. L3, or L4
provided that at least one of R1, R5 and R6 is L1, L2, L3, or L4 and further provided that R5 and R6 are not L 1 or L2 and wherein y ,
Figure imgf000081_0001
,
Figure imgf000081_0002
, or
Figure imgf000082_0001
,
Figure imgf000082_0002
and wherein
Y is O or NH,
R7 or R7' is H or CH3,
R8 or R8' is H or OH,
n is one to ten; and
n' is zero to ten.
Preferred simple trichothecenes are compounds
where R2 and R3 are both , and R4 and R5
Figure imgf000082_0003
are both H. Other preferred trichothecenes are
characterized as follows:
(1) R1 is L1 or L2 and Y is NH, R7 and R7, are independently either H or CH3 , R8 and R8, are
independently either H or OH, and n' is zero to ten;
further provided that R2 and R3 are both ;
Figure imgf000082_0004
R4 is H; and R5 and R6 are selected from the group consisting of H, OH, SH, OCH3, SCH3, NH2 , NHCH3, N(CH3)2;
(2) R1 is H, OH or SH; and R5 and R6 are selected from the group consisting of H, OH, SH, OCH3, SCH3,
NH2, NHCH3, N(CH3)2, or L3, wherein R7 and R7, are independently either H or CH3 and n is one to ten;
(3) R1 is H, OH or SH; and R5 or R6 is selected from the group consisting of H, OH, SH, OCH3, SCH3,
NH2, NHCH3, N(CH3)2. or L4, wherein n is one to ten, and R7 and R7, are independently either H or CH3;
(4) R1 is L3 or L4 wherein R7 and R7, and
independently either H or CH3 and n is one to ten; and R5 and R6 are selected from the group consisting of H, OH, SH, OCH3, SCH3, NH2, NHCH3, N(CH3)2.
A prototypical simple trichothecene is anguidine or the 3-keto derivative thereof which may be used in protocol number 1 set forth above as, for example, Biotin-NR-LU-10 monoclonal antibody-S-S-(CH2)2-CONH- N=(3-keto anguidine). Similarly, this simple
trichothecene may be utilized in the protocols
numbered 2 and 3 above as, for example, (Biotin) 18-20-Dextran-anguidine. Within the context of the above presented structures a prototypical simple
trichothecene is characterized as follows: R1 is L1 and Y is NH, R7 and R7, are both H, and n' is zero; R4,
R5, R6 are H; and R2 and R3 are both
Figure imgf000083_0005
Exemplary macrocyclic trichothecenes useful in the practice of this and other aspects of the present invention are as follows:
Figure imgf000083_0004
wherein R' is selected from the group consisting of
Figure imgf000083_0001
, ,
Figure imgf000083_0002
,
Figure imgf000083_0003
Figure imgf000084_0001
, ,
Figure imgf000084_0002
provided at least one of %, W and Z is L1, L2, L3, or L4, and further provided W and Z are not both L1, L2, L3, or L4 wherein ,
,
Figure imgf000084_0003
, or
Figure imgf000084_0004
and wherein
Y is O or NH,
R7 or R7' is H or CH3,
R8 or R8' is H or OH,
n is one to ten; and
n' is zero to ten; and further provided that W can be an epoxide group between 2' and 3' and still further provided that W and Z can be independently either H, OH, or SH when W and Z are not L 1 , L2, L3, or L4; R1 is H, OH, or SH when Rl is not L1, L2, L3, or L4; and R2 and R3 are selected from the group consisting of, H, OH, SH, OCH3, SCH3, NH2, NHCH3, N(CH3)2, L3 or L4.
Preferred macrocyclic trichothecenes are compounds wherein R' is
Figure imgf000085_0001
Such preferred compounds are derivatives of Roridin A. More preferred macrocyclic trichothecenes of this type are characterized as follows:
(1) R1 is L1, L2, L3, or L4 wherein
Y is O or NH,
R7 and R7' are independently either H or CH3, R8 and R8 ' are independently either H or OH, n is one to ten,
n' is zero to ten;
when R2 and R3 are selected from the group consisting of, H, OH, SH, OCH3, SCH3, NH2, NHCH3, N(CH3)2; and W and Z are independently either H, OH, or SH;
(2) R1 and W are independently either H, OH, or SH; R2 and R3 are selected from the group consisting of, H, OH, SH, OCH3, SCH3, NH2, NHCH3, N(CH3)2; and Z is L1, L2 , L3, or L4 and wherein
Y is 0 or NH,
R7 or R7' is H or CH3 ,
R8 or R8' is H or OH,
n is one to ten; and
n' is zero to ten;
(3) R1 and Z are H, SH, or OH; R2 and R3 are selected from the group consisting of, H, OH, SH, OCH3, SCH3, NH2, NHCH3, N(CH3)2; and W is either L1 L2, L3, or
L4 and wherein
Y is 0 or NH,
R7 or R7, is H or CH3,
R8 or R8, is H or OH,
n is one to ten, and
n' is zero to ten; and
(4) R1, W and Z are independently either SH or OH; R2 and R3 are selected from the group consisting of H, OH, SH, OCH3, SCH3, NH2, NHCH3, N(CH3)2, L3, or L4 wherein
R7 and R7, are either H or CH3, and
n is one to ten;
provided that R2 and R3 are not both simultaneously L3 or L4.
Prototypical macrocyclic trichothecenes are
Roridin A and derivatives thereof such as the 2'-oxo or the 13'-oxo derivatives. These trichothecenes may be used in protocol number 1 set forth above as, for example, Biotin-NR-LU-10 monoclonal antibody-S-S-(CH2)2-CONH-N=(2'-oxo Roridin A), Biotin-NR-LU-10 monoclonal antibody-S-S-(CH2)2-CONH-N=(13'-oxo Roridin A), 2'-oxo-Roridin A-S-S-(CH2)2-CONH-lysine-NR-LU-10 monoclonal antibody-biotin and the like. Similarly, these macrocyclic trichothecenes may be utilized in the protocols numbered 2 and 3 above as, for example, (Biotin)18.20-Dextran-2'-oxo Roridin A.
Prototypical Roridin A derivative macrocyclic trichothecenes may be characterized as follows: R1 and R2 are both H; R3 is CH3; W is L1, Y is NH, R7 and R7, are both H, n' is zero; and Z is OH;
R1 and R2 are both H; R3 is CH3; W is ; and
Figure imgf000086_0001
Z is L1, Y is NH, R7 and R7, are both H, n' is zero; and R1 and R2 are both H; R3 is CH3; W is L3, R7 and R7, are both H, n is one; and Z is OH. Acid labile linker technology, e.g., hydrazone linkers, facilitate release of therapeutic agent in target cell endosomes and lysosomes (pH 3.5-5.5) where the released agent can exert its therapeutic effect (e.g., inhibition of protein synthesis). Disulfide linkages also promote release of therapeutic agent in endosomes and lysosomes of the target cells.
Exemplary protocols for trichothecene-1inker preparation are discussed in Example XVIII. Exemplary protocols for trichothecene-antibody conjugation are set forth in Example XIX (A and B). Also, an
exemplary protocol for the preparation of
trichothecene-polymer conjugates is set forth in
Example XIX (C).
In embodiments of the present invention employing radionuclide therapeutic agents, the rapid clearance of nontargeted therapeutic agent decreases the
exposure of non-target organs, such as bone marrow, to the therapeutic agent. Consequently, higher doses of radiation can be administered absent dose limiting bone marrow toxicity. In addition, pretargeting methods of the present invention optionally include administration of short duration bone marrow
protecting agents, such as WR 2721. As a result, even higher doses of radiation can be given, absent dose limiting bone marrow toxicity.
While the pretargeting protocols set forth above have been described primarily in combination with delivery of a radionuclide diagnostic or therapeutic moiety, the protocols are amenable to use for delivery of other moieties, including anti-tumor agents, chemotherapeutic drugs and the like. For example, most naturally occurring and recombinant cytokines have short in vivo half lives. This characteristic limits the clinical effectiveness of these molecules, because near toxic doses are often required. Dose-limiting toxicities in humans have been observed upon high dose IL-2 or tumor necrosis factor
administrations, for example.
A protocol, such as administration of
streptavidin-targeting moiety conjugate followed by administration of biotinylated cytokine, is also contemplated by the present invention. Such
pretargeting of anti-ligand serves to improve the performance of cytokine therapeutics by increasing the amount of cytokine localized to target cells.
Streptavidin-antibody conjugates generally exhibit pharraacokinetics similar to the native antibody and localize well to target cells, depending upon their construction. Biotinylated cytokines retain a short in vivo half-life; however, cytokine may be localized to the target as a result of the affinity of biotin for avidin. In addition, biotin-avidin experience a pH-dependent dissociation which occurs at a slow rate, thereby permitting a relatively constant, sustained release of cytokine at the target site over time.
Also, cytokines complexed to target cells through biotin-avidin association are available for extraction and internalization by cells involved in cellular-mediated cytotoxicity.
A pre-formed antibody-streptavidin-biotin-cytokine preparation may also be employed in the practice of these methods of the present invention. In addition, a three-step protocol of the present invention may also be employed to deliver a cytokine, such as IL-2, to a target site.
Other anti-tumor agents that may be delivered in accordance with the pretargeting techniques of the present invention are selectins, including L-selectin, P-selectin and E-selectin. The presence of cytokines stimulates cells, such as endothelial cells, to express selectins on the surfaces thereof. Selectins bind to white blood cells and aid in delivering white blood cells where they are needed. Consequently, a protocol, such as administration of streptavidin- or avidin-targeting moiety conjugate followed by
administration of biotinylated selectins, is also contemplated by the present invention. Such
pretargeting of anti-ligand serves to improve the performance of selectin therapeutics by increasing the amount of selectin localized to target cells. In this manner, the necessity of cytokine induction of
selectin expression is obviated by the localization and retention of selectin at a target cell population. A three-step protocol may also be employed to deliver selectins to a target site.
Chemotherapeutic drugs also generally exhibit short in vivo half-lives at a therapeutically
effective dose. Consequently, another example of a protocol of the present invention includes
administration of avidin-targeting moiety conjugate followed by administration of a biotin-chemotherapeutic drug conjugate or complex, such as a drug-carrier-biotin complex. A three-step protocol of the present invention may also be employed to deliver a chemotherapeutic drug, such as methotrexate, adriamycin, high potency adriamycin analogs,
trichothecenes, potent enediynes, such as esperamycins and calicheamycins, cytoxan, vinca alkaloids,
actinamycin D, taxol, taxotere or the like to a target site.
An additional aspect of the present invention is a method of eradicating target cells within a mammal, involving the steps of:
1) extracting effector cells (or precursors thereof) from the mammal;
2) cultivating the extracted effector cells in vitro with both an effector cell growth factor and a first immunogenic molecule, thereby producing
sensitized effector cells recognizing the first immunogenic molecule or a moiety thereof; 3) administering to the mammal a second
immunogenic molecule comprising an antibody, an antibody fragment or another targeting moiety which binds to the target cells and a member of a
ligand/anti-ligand pair, and having at least one moiety in common with the first immunogenic molecule;
4) administering the sensitized effector cells produced in step 2 to the mammal; and
5) administering an effector cell growth factor bound to a member of the ligand/anti-ligand pair that is complementary to the member administered in step 2.
In this aspect of the present invention, the targeting moiety serves as a carrier to implant a strong predetermined antigenic signal on the target cells, ensuring that those target cells are
sufficiently "immunogenic" for subsequent therapy.
The engineered antigen bound to the target cells in vivo attracts subsequently administered presensitized effector cells to the target cells. In effect, this procedure constitutes an amplification of the tumor-associated antigen. The ligand/anti-ligand pair conjugate components are employed both to deliver effector cell growth factor to the target site and to avoid toxicity associated with circulating effector cell growth factor by decoupling the slow targeting moiety localization step from delivery of the growth factor.
An alternative method for eradicating target cells within a mammal comprises administering to the mammal an immunogenic molecule comprising an antibody- or other targeting moiety-ligand/anti-ligand pair member-immunogen conjugate which binds to the target cells in vivo. Subsequently, the complementary member of the ligand/anti-ligand pair is administered while
conjugated to an effector cell growth factor. This in vivo embodiment may be employed when the immunogenic molecule is capable of inducing a therapeutic response in vivo. In this case, the antigenic signal is not predetermined, but is sufficiently strong to recruit a therapeutically effective number of effector cells to the target site. The binding of the immunogenic molecule to the target cells within the mammal induces a cytotoxic effector cell response directed at the target cells. Delivery of effector cell growth factor to the target site enhances the activity of the effector cells at that site.
In accordance with the present invention, effector cells are extracted from a mammal or recruited from within a mammal. These effector cells are cells capable of being "sensitized" such that the sensitized effector cells recognize a particular antigen, and have a cytotoxic effect on target cells bearing that antigen. The effector cells useful in the practice of this aspect of the present invention are cells
designated as "committed T-cells," along with a variety of cytotoxic cells designated otherwise, including LAK cells, natural killer (NK) cells and other leukocytes and macrophages which could
potentially be sensitized and manipulated similarly. In a preferred embodiment of the present invention, the effector cells are cytotoxic T-lymphocytes, also called cytotoxic T-cells.
Effector cells can be extracted from the mammal to be treated by any suitable procedure for collecting those cells from the patient and separating the desired effector cells from other cell types or biological materials which may be simultaneously extracted. Conventional cell harvesting or
cytopheresis (e.g., leukopheresis) procedures may be used, and the effector cell extraction procedure may be repeated to obtain a larger quantity of effector cells. For example,, the mammal may be subjected to one leukapheresis procedure per day for one or more days (e.g., for five days to collect a large number of effector cells such as T-cells).
The effector cell growth factor used may vary according to the type of effector cells employed, and may be any growth factor which stimulates the
propagation of the cultured cells or enhances the activation thereof so that sensitized cytotoxic effector cells are produced. Among suitable growth factors are monokines for monocytes and lymphokines (e.g., IL-2) for lymphocytes such as T-lymphocytes).
The first immunogenic molecule (that with which the effector cells are incubated) may be the same as or different from the second immunogenic molecule (that which is administered to the mammal), provided that the two immunogenic molecules have at least one moiety in common. The proviso ensures that effector cells sensitized in vitro in the presence of the first immunogenic molecule will recognize the second
immunogenic molecule in vivo.
When the targeting moiety, the ligand/anti-ligand pair member or the combination thereof is sufficiently immunogenic (i.e., is capable of inducing
sensitization of effector cells), the targeting moiety-ligand/anti-ligand pair member conjugate may serve as the first as well as the second immunogenic molecule. Alternatively, a sufficiently immunogenic portion of the conjugate may be used as the first immunogenic molecule with the entire conjugate serving as the second immunogenic molecule.
When the targeting moiety-ligand/anti-ligand pair member conjugate or any portion thereof is not
sufficiently immunogenic (i.e., is incapable of inducing sensitization of effector cells) various compounds may be attached to the targeting moiety or ligand/anti-ligand pair member to increase the immunogenicity thereof. In this case, the immunogen-targeting moiety-ligand/anti-ligand pair member conjugate is administered as the second immunogenic molecule.
Among the types of compounds that may be attached to a targeting moiety or ligand/anti-ligand pair member to increase the immunogenicity thereof are haptens or polypeptides which are "foreign" to the intended mammalian recipient (such as antigens, mitogens, other foreign proteins or fragments thereof, peptides that activate cytotoxic T-cells or the like). In some circumstances, attachment of the immunogen to the targeting moiety or ligand/anti-ligand pair member through a spacer (i.e., a linker molecule that serves to physically separate the components) may increase the immunogenicity of the linked first immunogenic molecule.
Haptens that are useful in the practice of the present invention include benzoate groups, nitrophenol groups, other small molecules such as acetic acid and derivatives thereof, penicillinic acid and derivatives thereof, sulfanilic acid derivatives, hexoseamines, ribonucleotides, ribonucleosides, isocyanates,
isothiocyanates and the like. Preferred haptens are benzoic acid, dinitro-chlorobenzene, dinitrofluorobenzene, picrylchloride, p-aminobenzenearsonate, p-azo-benzenearsonate, acetic acid, dinitrophenol, trinitrophenol, trinitrophenol-epsilon-aminocaproic acid, 3-iodo-4-hydroxyl-5-nitrophenylacetic acid, p-hydroxyphenylacetic acid, p-sulfanilic acid, 2,4-diisocyanate, fluorescein isothiocyanate, N-acetyl-glucosamine and the like.
"Foreign" polypeptides useful in the practice of this aspect of the present invention include
flagellins, fragments of keyhole limpet hemocyanin, histocompatibility antigens, bacterial cell surface proteins, viral coat proteins, fragments thereof and the like. Small polypeptide fragments are generally preferred in order to avoid interference with the biodistribution of the targeting moiety-containing conjugate. T-cell mitogens such as the proteins pokeweed antiviral protein and phytohemagglutinin also may be used.
A specific example of the in vitro sensitization embodiment of this aspect of the present invention may be described as follows:
1) extracting effector cells (or precursors thereof) from the mammal;
2) cultivating the extracted effector cells in vitro with interleukin-2 (IL-2) and avidin or
streptavidin, thereby producing sensitized effector cells recognizing avidin or streptavidin or a moiety thereof;
3) administering to the mammal a targeting moiety-avidin or -streptavidin conjugate;
4) administering the sensitized effector cells produced in step 2 to the mammal; and
5) administering an IL-2-biotin conjugate.
A three-step pretargeting approach may be designed for the practice of this aspect- of the present invention.
Another specific example of an in vitro embodiment of the present invention involves the following steps:
1) extracting effector cells (or precursors thereof) from a mammal;
2) cultivating the effector cells with IL-2;
3) conjugating the cultivated effector cells with biotin;
4) administering a targeting moiety-avidin or targeting moiety-streptavidin to the mammal, wherein the targeting moiety is specific for the cells upon which effector cell action is desired; and
5) administering effector cell-biotin conjugate to the mammal.
In this embodiment of the present invention, steps 3 and 4 may be conducted in reverse order. Also, a clearing agent may be administered between steps 4 and 5. In addition, a three-step pretargeting protocol may be employed.
A specific example of the in vivo sensitization embodiment of this aspect of the present invention may be described as follows:
1) administering to the mammal a hapten-targeting moiety-avidin or -streptavidin conjugate to induce sensitization of effector cells; and
2) administering to the mammal an IL-2-biotin conjugate.
A three-step pretargeting protocol may also be
designed for the practice of this embodiment of the present invention.
Another protocol employing the concepts of the effector cell sensitization involves principles incident to organ transplantation. The HLA system in humans, including class I and class II antigens, corresponds to the MHC complex of mice, for example, and is a major factor in self/non-self recognition. For this reason, HLA matching is conducted in
selecting candidates for organ transplants. Persons with more similar HLA (i.e., twins, siblings, parents and the like) are the most preferably organ transplant donors, because the donated organ is less likely to be recognized as non-self by the recipient.
Some HLA antijgens are relatively rare in humans, i.e. f are infrequently present in the human HLA display. For example, the HLA-B7 antigen is present on only 5% of the human population. Consequently, HLA-B7 is recognized as non-self in 95% of humans. Such relatively rare antigens can therefore be
targeted to target sites within an individual that does not exhibit the antigen. Due to the presence of the antigen at the target site, the target will be recognized as non-self by the recipient, thereby recruiting the recipient's immune system to combat the target. Since some targets (e.g., certain tumors) are poorly recognized as non-self, the presence of a recognized non-self antigen at that site will improve the recipient's immune system response to the target.
HLA antigen that is not present in a recipient can be delivered to a target site by conjugation with a targeting moiety, as part of a fusion protein with a targeting moiety or as part of a conjugate or fusion protein with a ligand or an anti-ligand in the two-step or three- step pretargeting protocols described herein .
The invention is further described through
presentation of the following examples. These
examples are offered by way of illustration, and not by way of limitation.
Example I
Synthesis of a Chelate-Biotin Conjugate A chelating compound that contains an N3S chelating core was attached via an amide linkage to biotin.
Radiometal labeling of an exemplary chelate-biotin conjugate is illustrated below.
Figure imgf000096_0001
The spacer group "X" permits the biotin portion of the conjugate to be sterically available for avidin binding. When "R1" is a carboxylic acid substituent (for instance, CH2COOH), the conjugate exhibits improved water solubility, and further directs in vivo excretion of the radiolabeled biotin conjugate toward renal rather than hepatobiliary clearance.
Briefly, N-α-Cbz-N-Σ-t-BOC protected lysine was converted to the succinimidyl ester with NHS and DCC, and then condensed with aspartic acid β- -t-butyl ester. The resultant dipeptide was activated with NHS and DCC, and then condensed with glycine t-butyl ester. The Cbz group was removed by hydrogenolysis, and the amine was acylated using tetrahydropyranyl mercaptoacetic acid succinimidyl ester, yielding S-(tetrahydropyranyl)-mercaptoacetyl-lysine.
Trifluoroacetic acid cleavage of the N-t-BOC group and t-butyl esters, followed by condensation with LC-biotin-NHS ester provided (Σ-caproylamide biotin)-aspartyl glycine. This synthetic method is
illustrated below.
Figure imgf000097_0001
Figure imgf000098_0001
1H NMR: (CD3OD, 200 MHz Varian) : 1.25-1.95 (m, 24H),
2.15-2.25 (broad t, 4H), 2.65-3.05 (m, 4H),
3.30-3.45 (dd, 2H), 3.50-3.65 (ddd, 2H), 3.95 (broad s, 2H), 4.00-4.15 (m, 1H), 4.25-4.35
(m, 1H), 4.45-4.55 (m, 1H), 4.7-5.05 (m overlapping with HOD).
Elemental Analysis: C, H, N for C35H57N7O11S2·H2O
calculated: 50.41, 7.13, 11.76
found: 50.13, 7.14, 11.40
Example II
Preparation of a Technetium or Rhenium Radiolabeled Chelate-Biotin Conjugate
The chelate-biotin conjugate of Example I was radiolabeled with either 99mTc pertechnetate or 186Re perrhenate. Briefly, 99mTc pertechnetate was reduced with stannous chloride in the presence of sodium
gluconate to form an intermediate Tc-gluconate
complex. The chelate-biotin conjugate of Example I was added and heated to 100°C for 10 min at a pH of about 1.8 to about 3.3. The solution was neutralized to a pH of about 6 to about 8, and yielded an N3S-coordinated 99mTc-chelate-biotin conjugate. C-18 HPLC gradient elution using 5-60% acetonitrile in 1% acetic acid demonstrated two anomers at 97% or greater
radiochemical yield using δ (gamma ray) detection.
Alternatively, 186Re perrhenate was spiked with cold ammonium perrhenate, reduced with stannous chloride, and complexed with citrate. The chelate-biotin conjugate of Example I was added and heated to 90°C for 30 min at a pH of about 2 to 3. The solution was neutralized to a pH of about 6 to about 8, and yielded an N3S-coordinated 18oRe-chelate-biotin conjugate. C-18 HPLC gradient elution using 5-60% acetonitrile in 1% acetic acid resulted in radiochemical yields of 85-90%. Subsequent purification over a C-18 reverse phase hydrophobic column yielded material of 99% purity.
Example III
In Vitro Analysis of Radiolabeled
Chelate-Biotin Conjugates
Both the 99mTc- and 186Re-chelate-biotin conjugates were evaluated in vitro. When combined with excess avidin (about 100-fold molar excess), 100% of both radiolabeled biotin conjugates complexed with avidin.
A 99mTc-biotin conjugate was subjected to various chemical challenge conditions. Briefly, 99mTc-chelate-biotin conjugates were combined with avidin and passed over a 5 cm size exclusion gel filtration column. The radiolabeled biotin-avidin complexes were subjected to various chemical challenges (see Table 1), and the incubation mixtures were centrifuged through a size exclusion filter. The percent of radioactivity retained (indicating avidin-biotin-associated
radiolabel) is presented in Table 1. Thus, upon chemical challenge, the radiometal remained associated with the macromolecular complex. TABLE 1
Chemical Challenge of 99mTc-Chelate- Biotin- •Avidin Complexes
Challenge pH % Radioactivity Retained Medium 1 h, 37ºC 18 h. RT
PBS 7.2 99 99
Phosphate 8.0 97 97
10 mM cysteine 8.0 92 95
10 mM DTPA 8.0 99 98
0.2 M carbonate 10.0 97 94
In addition, each radiolabeled biotin conjugate was incubated at about 50 μg/ml with serum; upon completion of the incubation, the samples were
subjected to instant thin layer chromatography (ITLC) in 80% methanol. Only 2-4% of the radioactivity remained at the origin (i.e., associated with
protein); this percentage was unaffected by the addition of exogenous biotin. When the samples were analyzed using size exclusion H-12 FPLC with 0.2 M phosphate as mobile phase, no association of
radioactivity with serum macromolecules was observed.
Each radiolabeled biotin conjugate was further examined using a competitive biotin binding assay. Briefly, solutions containing varying ratios of D-biotin to radiolabeled biotin conjugate were combined with limiting avidin at a constant total biotin: avidin ratio. Avidin binding of each radiolabeled biotin conjugate was determined by ITLC, and was compared to the theoretical maximum stoichiometric binding (as determined by the HABA spectrophotometric assay of Green, Biochem. J. 94:23c-24c, 1965). No significant difference in avidin binding was observed between each radiolabeled biotin conjugate and D-biotin. Example IV
In Vivo Analysis of Radiolabeled Chelate-Biotin Conjugates Administered After Antibody Pretargeting
The 186Re-chelate-biotin conjugate of Example I was studied in an animal model of a three-step
antibody pretargeting protocol. Generally, this protocol involved: (i) prelocalization of biotinylated monoclonal antibody; (ii) administration of avidin for formation of a "sandwich" at the target site and for clearance of residual circulating biotinylated
antibody; and (iii) administration of the 186Re-biotin conjugate for target site localization and rapid blood clearance. A . Preparation and Characterization of
Biotinylated Antibody
Biotinylated NR-LU-10 was prepared according to either of the following procedures. The first procedure involved derivitization of antibody via lysine s-amino groups. NR-LU-10 was radioiodinated at tyrosines using chloramine T and either 125 I or 131I sodium iodide. The radioiodinated antibody (5-10 mg/ml) was then biotinylated using biotinamido caproate NHS ester in carbonate buffer, pH 8.5, containing 5% DMSO, according to the scheme below.
Figure imgf000101_0001
The impact of lysine biotinylation on antibody immunoreactivity was examined. As the molar offering of biotin: antibody increased from 5:1 to 40:1, biotin incorporation increased as expected (measured using the HABA assay and pronase-digested product) (Table 2, below). Percent of biotinylated antibody
immunoreactivity as compared to native antibody was assessed in a limiting antigen ELISA assay. The immunoreactivity percentage dropped below 70% at a measured derivitization of 11.1:1; however, at this level of derivitization, no decrease was observed in antigen-positive cell binding (performed with LS-180 tumor cells at antigen excess). Subsequent
experiments used antibody derivitized at a
biotin: antibody ratio of 10:1.
TABLE 2
Effect of Lysine Biotinylation on Immunoreactivity
Molar Measured Immunoassessment (%)
Offering Derivitization
(Biotins/Ab) (Biotins/Ab) ELISA Cell Binding
5:1 3.4 86
10:1 8.5 73 100
13:1 11.1 69 102
20:1 13.4 36 106
40:1 23.1 27 Alternatively, NR-LU-10 was biotinylated using thiol groups generated by reduction of cystines.
Derivitization of thiol groups was hypothesized to be less compromising to antibody immunoreactivity.
NR-LU-10 was radioiodinated using p-aryltin phenylate NHS ester (PIP-NHS) and either 125I or 131I sodium iodide. Radioiodinated NR-LU-10 was incubated with 25 mM dithiothreitol and purified using size exclusion chromatography. The reduced antibody (containing free thiol groups) was then reacted with a 10- to 100-fold molar excess of N-iodoacetyl-n'-biotinyl hexylene diamine in phosphate-buffered saline (PBS), pH 7.5, containing 5% DMSO (v/v). TABLE 3
Effect of Thiol Biotinylation on Immunoreactivity
Molar Measured Immunoassessment (%) Offering Derivitization
(Biotins/Ab) (Biotins/Ab) ELISA Cell Binding
10:1 4.7 114
50:1 6.5 102 100
100:1 6.1 95 100
As shown in Table.3, at a 50:1 or greater
biotin:antibody molar offering, only 6 biotins per antibody were incorporated. No significant impact on immunoreactivity was observed.
The lysine- and thiol-derivitized biotinylated antibodies ("antibody (lysine)" and "antibody
(thiol)", respectively) were compared. Molecular sizing on size exclusion FPLC demonstrated that both biotinylation protocols yielded monomolecular
(monomeric) IgGs. Biotinylated antibody (lysine) had an apparent molecular weight of 160 kD, while
biotinylated antibody (thiol) had an apparent
molecular weight of 180 kD. Reduction of endogenous sulfhydryls (i.e., disulfides) to thiol groups, followed by conjugation with biotin, may produce a somewhat unfolded macromolecule. If so, the antibody (thiol) may display a larger hydrodynamic radius and exhibit an apparent increase in molecular weight by chromatographic analysis. Both biotinylated antibody species exhibited 98% specific binding to immobilized avidin-agarose.
Further comparison of the biotinylated antibody species was performed using non-reducing SDS-PAGE, using a 4% stacking gel and a 5% resolving gel. Biotinylated samples were either radiolabeled or unlabeled and were combined with either radiolabeled or unlabeled avidin or streptavidin. Samples were not boiled prior to SDS-PAGE analysis. The native
antibody and biotinylated antibody (lysine) showed similar migrations; the biotinylated antibody (thiol) produced two species in the 50-75 kD range. These species may represent two thiol-capped species. Under these SDS-PAGE conditions, radiolabeled streptavidin migrates as a 60 kD tetramer. When 400 μg/ml
radiolabeled streptavidin was combined with 50 μg/ml biotinylated antibody (analogous to "sandwiching" conditions in vivo), both antibody species formed large molecular weight complexes. However, only the biotinylated antibody (thiol)-streptavidin complex moved from the stacking gel into the resolving gel, indicating a decreased molecular weight as compared to the biotinylated antibody (lysine)-streptavidin complex.
B. Blood Clearance of Biotinylated Antibody
Species
Radioiodinated biotinylated NR-LU-10 (lysine or thiol) was intravenously administered to non-tumored nude mice at a dose of 100 μg. At 24 h post-administration of radioiodinated biotinylated NR-LU-10, mice were intravenously injected with either saline or 400 μg of avidin. With saline
administration, blood clearances for both biotinylated antibody species were biphasic and similar to the clearance of native NR-LU-10 antibody.
In the animals that received avidin intravenously at 24 h, the biotinylated antibody (lysine) was cleared (to a level of 5% of injected dose) within 15 min of avidin administration (avidin:biotin = 10:1). With the biotinylated antibody (thiol), avidin
administration (10:1 or 25:1) reduced the circulating antibody level to about 35% of injected dose after two hours. Residual radiolabeled antibody activity in the circulation after avidin administration was examined in vitro using immobilized biotin. This analysis revealed that 85% of the biotinylated antibody was complexed with avidin. These data suggest that the biotinylated antibody (thiol)-avidin complexes that were formed were insufficiently crosslinked to be cleared by the RES.
Blood clearance and biodistribution studies of biotinylated antibody (lysine) 2 h post-avidin or post-saline administration were performed. Avidin administration significantly reduced the level of biotinylated antibody in the blood (see Figure 1), and increased the level of biotinylated antibody in the liver and spleen. Kidney levels of biotinylated antibody were similar.
Example V
In Vivo Characterization of 186Re- Chelate-Biotin Conjugates In a Three-Step
Pretargeting Protocol
A 186Re-chelate-biotin conjugate of Example I (MW ≈
1000; specific activity = 1-2 mCi/mg) was examined in a three-step pretargeting protocol in an animal model.
More specifically, 18-22 g female nude mice were implanted subcutaneously with LS-180 human colon tumor xenografts, yielding 100-200 mg tumors within 10 days of implantation.
NR-LU-10 antibody (MW ≈ 150 kD) was radiolabeled-with 125I/Chloramine T and biotinylated via lysine residues (as described in Example IV.A, above).
Avidin (MW ≈ 66 kD) was radiolabeled with 131I/PIP-NHS (as described for radioiodination of NR-LU-10 in
Example IV.A., above). The experimental protocol was as follows:
Group 1: Time 0, inject 100 μg 125I-labeled,
biotinylated NR-LU-10
Time 24 h, inject 400 μg 131I-labeled avidin Time 26 h, inject 60 μg 186Re-chelate- biotin conjugate
Group 2: Time 0, inject 400 μg 131I-labeled avidin (control) Time 2 h, inject 60 μg 186Re-chelate- biotin conjugate
Group 3: Time 0, inject 60 μg 186Re-chelate- (control) biotin conjugate
The three radiolabels employed in this protocol are capable of detection in the presence of each other. It is also noteworthy that the sizes of the three elements involved are logarithmically different ╌ antibody ≈ 150,000-; avidin ≈ 66,000; and biotin ≈
1,000. Biodistribution analyses were performed at 2, 6, 24, 72 and 120 h after administration of the 186Re-chelate-bibtin conjugate.
Certain preliminary studies were performed in the animal model prior to analyzing the 186Re-chelate-biotin conjugate in a three-step pretargeting
protocol. First, the effect of biotinylated antibody on blood clearance of avidin was examined. These experiments showed that the rate and extent of avidin clearance was similar in the presence or absence of biotinylated antibody. Second, the effect of
biotinylated antibody and avidin on blood clearance of the 186Re-chelate-biotin conjugate was examined; blood clearance was similar in the presence or absence of biotinylated antibody and avidin. Further, antibody immunoreactivity was found to be uncorapromised by biotinylation at the level tested.
Third, tumor uptake of biotinylated antibody administered at time 0 or of avidin administered at time 24 h was examined. The results of this
experimentation are shown in Fig. 1. At 25 h, about 350 pmol/g biotinylated antibody was present at the tumor; at 32 h the level was about 300 pmol/g; at 48 h, about 200 pmol/g; and at 120 h, about 100 pmol/g. Avidin uptake at the same time points was about 250, 150, 50 and 0 pmol/g, respectively. From the same experiment, tumor to blood ratios were determined for biotinylated antibody and for avidin. From 32 h to 120 h, the ratios of tumor to blood were very similar.
Rapid and efficient removal of biotinylated antibody from the blood by complexation with avidin was observed. Within two hours of avidin
administration, a 10-fold reduction in blood pool antibody concentration was noted (Fig. 1), resulting in a sharp increase in tumor to blood ratios. Avidin is cleared rapidly, with greater than 90% of the injected dose cleared from the blood within 1 hour after administration. The Re-186-biotin chelate is also very rapidly cleared, with greater than 99% of the injected dose cleared from the blood by 1 hour after administration.
The three-step pretargeting protocol (described for Group 1, above) was then examined. More
specifically, tumor uptake of the 186Re-chelate-biotin conjugate in the presence or absence of biotinylated antibody and avidin was determined. In the absence of biotinylated antibody and avidin, the 186Re-chelate- biotin conjugate displayed a slight peak 2 h post-injection, which was substantially cleared from the tumor by about 5 h. In contrast, at 2 h post-injection in the presence of biotinylated antibody and avidin (specific), the 186Re-chelate-biotin conjugate reached a peak in tumor approximately 7 times greater than that observed in the absence of biotinylated antibody and avidin. Further, the specifically bound 186Re-chelate-biotin conjugate was retained at the tumor at significant levels for more than 50 h. Tumor to blood ratios determined in the same experiment increased significantly over time (i.e., T:B = ≈ 8 at 30 h; ≈ 15 at 100 h; ≈ 35 at 140 h).
Tumor uptake of the 186Re-chelate-biotin conjugate has further been shown to be dependent on the dose of biotinylated antibody administered. At 0 μg of biotinylated antibody, about 200 pmol/g of 186Re-chelate-biotin conjugate was present at the tumor at 2 h after administration; at 50 μg antibody, about 500 pmol/g of 1B6Re-chelate-biotin conjugate; and at 100 μg antibody, about 1,300 pmol/g of 186Re-chelate-biotin conjugate.
Rhenium tumor uptake via the three-step
pretargeting protocol was compared to tumor uptake of the same antibody radiolabeled through chelate
covalently attached to the antibody (conventional procedure) . The results of this comparison are depicted in Figure 2. Blood clearance and tumor uptake were compared for the chelate directly labeled rhenium antibody conjugate and for the three-step pretargeted sandwich. Areas under the curves (AUC) and the ratio of AUCtumor/AUCblood were determined. For the chelate directly labeled rhenium antibody
conjugate, the ratio of AUCtumor/AUCblood = 24055/10235 or 2.35; for the three-step pretargeted sandwich, the ratio of AUCtumor/AUCblood = 46764/6555 or 7.13.
Tumor uptake. results are best taken in context with radioactivity exposure to the blood compartment, which directly correlates with bone marrow exposure. Despite the fact that 100-fold more rhenium was administered to animals in the three-step protocol, the very rapid clearance of the small molecule (Re-186-biotin) from the blood minimizes the exposure to Re-186 given in this manner. In the same matched antibody dose format, direct labeled (conventional procedure) NR-LU-10 whole antibody yielded greater exposure to rhenium than did the 100-fold higher dose given in the three-step protocol. A clear increase in the targeting ratio (tumor exposure to
radioactivity:blood, exposure to radioactivity╌
AUCtunor:AUCblood) was observed for three-step
pretargeting (approximately 7:1) in comparison to the direct labeled antibody approach (approximately
2.4:1).
Example VI
Preparation of Chelate-Biotin Conjugates
Having Improved Biodistribution Properties
The biodistribution of 111In-labeled-biotin derivatives varies greatly with structural changes in the chelate and the conjugating group. Similar structural changes may affect the biodistribution of technetium- and rhenium-biotin conjugates.
Accordingly, methods for preparing technetium- and rhenium-biotin conjugates having optimal clearance from normal tissue are advantageous.
A. Neutral MAMA Chelate/Coniugate
A neutral MAMA chelate-biotin conjugate is prepared according to the following scheme.
Figure imgf000109_0001
Figure imgf000110_0001
The resultant chelate-biotin conjugate shows superior kidney excretion. Although the net overall charge of the conjugate is neutral, the polycarboxylate nature of the molecule generates regions of hydrophilicity and hydrophobicity. By altering the number and nature of the carboxylate groups within the conjugate, excretion may be shifted from kidney to
gastrointestinal routes. For instance, neutral compounds are generally cleared by the kidneys;
anionic compounds are generally cleared through the GI system.
B. Polylysine Derivitization
Conjugates containing polylysine may also exhibit beneficial biodistribution properties. With whole antibodies, derivitization with polylysine may skew the biodistribution of conjugate toward liver uptake. In contrast, derivitization of Fab fragments with polylysine results in lower levels of both liver and kidney uptake; blood clearance of these conjugates is similar to that of Fab covalently linked to chelate. An exemplary polylysine derivitized chelate-biotin conjugate is illustrated below.
Figure imgf000111_0001
Inclusion of polylysine in radiometal-chelate-biotin conjugates is therefore useful for minimizing or eliminating RES sequestration while maintaining good liver and kidney clearance of the conjugate. For improved renal excretion properties, polylysine derivatives are preferably succinylated following biotinylation. Polylysine derivatives offer the further advantages of: (1) increasing the specific activity of the radiometal-chelate-biotin conjugate; (2) permitting control of rate and route of blood clearance by varying the molecular weight of the polylysine polymer; and (3) increasing the circulation half-life of the conjugate for optimal tumor
interaction.
Polylysine derivitization is accomplished by standard methodologies. Briefly, poly-L-lysine is acylated according to standard amino group acylation procedures (aqueous bicarbonate buffer, pH 8, added biotin-NHS ester, followed by chelate NHS ester).
Alternative methodology involves anhydrous conditions using nitrophenyl esters in DMSO and triethyl amine. The resultant conjugates are characterized by UV and NMR spectra.
The number of biotins attached to polylysine is determined by the HABA assay. Spectrophotometric titration is used to assess the extent of amino group derivitization. The radiometal-chelate-biotin
conjugate is characterized by size exclusion.
C. Cleavable Linkage
Through insertion of a cleavable linker between the chelate and biotin portion of a radiometal-chelate-biotin conjugate, retention of the conjugate at the tumor relative to normal tissue may be
enhanced. More specifically, linkers that are cleaved by enzymes present in normal tissue but deficient or absent in tumor tissue can increase tumor retention. As an example, the kidney has high levels of γ-glutamyl transferase; other normal tissues exhibit in vivo cleavage of γ-glutamyl prodrugs. In contrast, tumors are generally deficient in enzyme peptidases.
The glutamyl-linked biotin conjugate depicted below is cleaved in normal tissue and retained in the tumor.
Figure imgf000112_0001
D. Serine Linker With O-Polar Substituent
Sugar substitution of N3S chelates renders such chelates water soluble. Sulfonates, which are fully ionized at physiological pH, improve water solubility of the chelate-biotin conjugate depicted below.
Figure imgf000113_0001
This compound is synthesized according to the standard reaction procedures. Briefly, biocytin is condensed with N-t-BOC-(O-sulfonate or O-glucose) serine NHS ester to give N-t-BOC-(O-sulfonate or O-glucose) serine biocytinamide. Subsequent cleavage of the N-t-BOC group with TFA and condensation with ligand NHS ester in DMF with triethylamine provides ligand-amidoserine (O-sulfonate or O-glucose) biocytinamide.
Example VII
Preparation and Characterization of PIP- Radioiodinated Biotin
Radioiodinated biotin derivatives prepared by exposure of poly-L-lysine to excess NHS-LC-biotin and then to Bolton-Hunter N-hydroxysuccinimide esters in
DMSO has been reported. After purification, this product was radiolabeled by the iodogen method (see, for instance, Del Rosario et al., J. Nucl. Med. 32 :5, 1991, 993 (abstr.)). Because of the high molecular weight of the resultant radioiodinated biotin
derivative, only limited characterization of product
(i.e., radio-HPLC and binding to immobilized
streptavidin) was possible. Preparation of radioiodinated biotin according to the present invention provides certain advantages. First, the radioiodobiotin derivative is a low
molecular weight compound that is amenable to complete chemical characterization. Second, the disclosed methods for preparation involve a single step and eliminate the need for a purification step.
Briefly, iodobenzamide derivatives corresponding to biocytin (R = COOH) and biotinamidopentylamine (R = H) were prepared according to the following scheme. In this scheme, "X" may be any radiohalogen, including 125I, 131I, 123I, 211At and the like.
Figure imgf000114_0001
Preparation of 1 was generally according to Wilbur et al., J. Nucl. Med. 30:216-26, 1989, using a
tributyltin intermediate. Water soluble carbodiimide was used in the above-depicted reaction, since the NHS ester 1 formed intractable mixtures with DCU. The NHS ester was not compatible with chromatography; it was insoluble in organic and aqueous solvents and did not react with biocytin in DMF or in buffered aqueous acetonitrile. The reaction between 1 and biocytin or 5-(biotinamido) pentylamine was sensitive to base.
When the reaction of 1 and biocytin or the pentylamine was performed in the presence of triethylamine in hot DMSO, formation of more than one biotinylated product resulted. In contrast, the reaction was extremely clean and complete when a suspension of 1 and biocytin (4 mg/ml) or the pentylamine (4 mg/ml) was heated in DMSO at 117°C for about 5 to about 10 min. The resultant 125I-biotin derivatives were obtained in 94% radiochemical yield. Optionally, the radioiodinated products may be purified using C-18 HPLC and a reverse phase hydrophobic column. Hereinafter, the resultant radioiodinated products 2 are referred to as PIP-biocytin (R = COOH) and PIP-pentylamine (R = H).
Both iodobiotin derivatives 2. exhibited ≥95% binding to immobilized avidin. Incubation of the products 2 with mouse serum resulted in no loss of the ability of 2 to bind to immobilized avidin.
Biodistribution studies of 2 in male BALB/c mice showed rapid clearance from the blood (similar to
186Re-chelate-biotin conjugates described above). The radioiodobiotin 2 had decreased hepatobiliary
excretion as compared to the 186Re-chelate-biotin conjugate; urinary excretion was increased as compared to the 186Re-chelate-biotin conjugate. Analysis of urinary metabolites of 2 indicated deiodination and cleavage of the biotin amide bond; the metabolites showed no binding to immobilized avidin. In contrast, metabolites of the 186Re-chelate-biotin conjugate appear to be excreted in urine as intact biotin conjugates. Intestinal uptake of 2 is <50% that of the 186Re-chelate-biotin conjugate. These
biodistribution properties of 2 provided enhanced whole body clearance of radioisotope and indicate the advantageous use of 2 within pretargeting protocols.
131I-PIP-biocytin was evaluated in a two-step pretargeting procedure in tumor-bearing mice. Briefly, female nude mice were injected subcutaneously with LS-180 tumor cells; after 7 d, the mice displayed 50-100 mg tumor xenografts. At t = 0, the mice were injected with 200 μg of NR-LU-10-avidin conjugate labeled with 125I using PIP-NHS (see Example IV.A.). At t = 36 h, the mice received 42 μg of 131I-PIP-biocytin. The data showed immediate, specific tumor localization, corresponding to ≈ 1.5 131I-PIP-biocytin molecules per avidin molecule.
The described radiohalogenated biotin compounds are amenable to the same types of modifications described in Example VI above for 186Re-chelate-biotin conjugates. In particular, the following PIP-polylysine-biotin molecule is made by trace labeling polylysine with 125I-PIP, followed by extensive biotinylation of the polylysine.
Figure imgf000116_0001
Assessment of 125I binding to immobilized avidin ensures that all radioiodinated species also contain at least an equivalent of biotin.
Example VIII
Preparation of Biotinylated Antibody (Thiol) Through Endogenous Antibody Sulfhydryl Groups
Or Sulfhydryl-Generating Compounds
Certain antibodies have available for reaction endogenous sulfhydryl groups. If the antibody to be biotinylated contains endogenous sulfhydryl groups, such antibody is reacted with N-iodoacetyl-n'-biotinyl hexylene diamine (as described in Example IV.A., above). The availability of one or more endogenous sulfhydryl groups obviates the need to expose the antibody to a reducing agent, such as DTT, which can have other detrimental effects on the biotinylated antibody.
Alternatively, one or more sulfhydryl groups are attached to a targeting moiety through the use of chemical compounds or linkers that contain a terminal sulfhydryl group. An exemplary compound for this purpose is iminothiolane. As with endogenous
sulfhydryl groups (discussed above), the detrimental effects of reducing agents on antibody are thereby avoided.
Example IX
Two-Step Pretargeting Methodology That
Does Not Induce Internalization
A NR-LU-13-avidin conjugate is prepared as
follows. Initially, avidin is derivitized with N-succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC). SMCC-derived avidin is then incubated with NR-LU-13 in a 1:1 molar ratio at pH 8.5 for 16 h. Unreacted NR-LU-13 and SMCC-derived avidin are removed from the mixture using preparative size exclusion HPLC. Two conjugates are obtained as products ╌ the desired 1 : 1 NR-LU-13 -avidin conjugate as the major product; and an incompletely
characterized component as the minor product.
A 99mTc-chelate-biotin conjugate is prepared as in Example II, above. The NR-LU-13-avidin conjugate is administered to a recipient and allowed to clear from the circulation. One of ordinary skill in the art of radioimmunoscintigraphy is readily able to determine the optimal time for NR-LU-13-avidin conjugate tumor localization and clearance from the circulation. At such time, the 99mTc-chelate-biotin conjugate is administered to the recipient. Because the 99mTc- chelate-biotin conjugate has a molecular weight of ≈ 1,000, crosslinking of NR-LU-13-avidin molecules on the surface of the tumor cells is dramatically reduced or eliminated. As a result, the 99mTc diagnostic agent is retained at the tumor cell surface for an extended period of time. Accordingly, detection of the
diagnostic agent by imaging techniques is optimized; further, a lower dose of radioisotope provides an image comparable to that resulting from the typical three-step pretargeting protocol.
Optionally, clearance of NR-LU-13-avidin from the circulation may be accelerated by plasmapheresis in combination with a biotin affinity column. Through use of such column, circulating NR-LU-13-avidin will be retained extracorporeally, and the recipient's immune system exposure to a large, proteinaceous immunogen (i.e., avidin) is minimized.
Exemplary methodology for plasmapheresis/column purification useful in the practice of the present invention is discussed in the context of reducing radiolabeled antibody titer in imaging and in treating tumor target sites in U.S. Patent 5,078,673. Briefly, for the purposes of the present invention, an example of an extracorporeal clearance methodology may include the following steps:
administering a ligand- or anti-ligand-targeting moiety conjugate to a recipient;
after a time sufficient for localization of the administered conjugate to the target site, withdrawing blood from the recipient by, for example,
plasmapheresis;
separating cellular element from said blood to produce a serum fraction and returning the cellular elements to the recipient; and
reducing the titer of the administered conjugate in the serum fraction to produce purified serum;
infusing the purified serum back into the
recipient. Clearance of NR-LU-13-avidin is also facilitated by administration of a particulate-type clearing agent (e.g., a polymeric particle having a plurality of biotin molecules bound thereto). Such a particulate clearing agent preferably constitutes a biodegradable polymeric carrier having a plurality of biotin
molecules bound thereto. Particulate clearing agents of the present invention exhibit the capability of binding to circulating administered conjugate and removing that conjugate from the recipient.
Particulate clearing agents of this aspect of the present invention may be of any configuration suitable for this purpose. Preferred particulate clearing agents exhibit one or more of the following
characteristics:
- microparticulate (e.g., from about 0.5
micrometers to about 100 micrometers in diameter, with from about 0.5 to about 2 micrometers more preferred), free flowing powder structure;
- biodegradable structure designed to biodegrade over a period of time between from about 3 to about 180 days, with from about 10 to about 21 days more preferred, or non-biodegradable structure;
- biocompatible with the recipients physiology over the course of distribution, metabolism and excretion of the clearing agent, more preferably including biocompatible biodegradation products;
- and capability to bind with one or more
circulating conjugates to facilitate the elimination or removal thereof from the recipient through one or more binding moieties (preferably, the complementary member of the ligand/anti-ligand pair) . The total molar binding capacity of the particulate clearing agents depends upon the particle size selected and the ligand or anti-ligand substitution ratip. The binding moieties are capable of coupling to the surface structure of the particulate dosage form through covalent or non-covalent modalities as set forth herein to provide accessible ligand or anti-ligand for binding to its previously administered circulating binding pair member.
Preferable particulate clearing agents of the present invention are biodegradable or non-biodegradable microparticulates. More preferably, the particulate clearing agents are formed of a polymer containing matrix that biodegrades by random,
nonenzymatic, hydrolytic scissioning.
Polymers derived from the condensation of alpha hydroxycarboxylic acids and related lactones are more preferred for use in the present invention. A
particularly preferred moiety is formed of a mixture of thermoplastic polyesters (e.g., polylactide or polyglycolide) or a copolymer of lactide and glycolide components, such as poly (lactide-co-glycolide). An exemplary structure, a random poly (DL-lactide-co-glycolide), is shown below, with the values of x and y being manipulable by a practitioner in the art to achieve desirable microparticulate properties.
Figure imgf000120_0001
Other agents suitable for forming particulate clearing agents of the present invention include polyorthoesters and polyacetals (Polymer Letters,
18.: 293, 1980) and polyorthocarbonates (U.S. Patent No. 4,093,709) and the like.
Preferred lactic acid/glycolic acid polymer containing matrix particulates of the present
invention are prepared by emulsion-based processes, that constitute modified solvent extraction processes such as those described by Cowsar et al.,
"Poly(Lactide-Co-Glycolide) Microcapsules for
Controlled Release of Steroids," Methods Enzymology, 112:101-116, 1985 (steroid entrapment in
microparticulates); Eldridge et al., "Biodegradable and Biocompatible Poly(DL-Lactide-Co-Glycolide)
Microspheres as an Adjuvant for Staphylococcal
Enterotoxin B Toxoid Which Enhances the Level of
Toxin-Neutralizing Antibodies," Infection and
Immunity, 59: 2978-2986, 1991 (toxoid entrapment);
Cohen et al., "Controlled Delivery Systems for
Proteins Based on Poly (Lactic/Glycolic Acid)
Microspheres," Pharmaceutical Research. 8(6) :713-720, 1991 (enzyme entrapment); and Sanders et al.,
"Controlled Release of a Luteinizing Hormone-Releasing Hormone Analogue from Poly (D,L-Lactide-Co-Glycolide) Microspheres," J. Pharmaceutical Science, 73(9) : 1294-1297, 1984 (peptide entrapment).
In general, the procedure for forming particulate clearing agents of the present invention involves dissolving the polymer in a halogenated hydrocarbon solvent and adding an additional agent that acts as a solvent for the halogenated hydrocarbon solvent but not for the polymer. The polymer precipitates out from the polymer-halogenated hydrocarbon solution. Following particulate formation, they are washed and hardened with an organic solvent. Water washing and aqueous non-ionic surfactant washing steps follow, prior to drying at room temperature under vacuum.
For biocompatibility purposes, particulate
clearing agents are sterilized prior to packaging, storage or administration. Sterilization may be conducted in any convenient manner therefor. For example, the particulates can be irradiated with gamma radiation, provided that exposure to such radiation does not adversely impact the structure or function of the binding moiety attached thereto. If the binding moiety is so adversely impacted, the particulate clearing agents can be produced under sterile
conditions.
The preferred lactide/glycolide structure is biocompatible with the mammalian physiological
environment. Also, these preferred sustained release dosage forms have the advantage that biodegradation thereof forms lactic acid and glycolic acid, both normal metabolic products of mammals.
Functional groups required for binding moiety -particulate bonding, are optionally included in the particulate structure, along with the non-degradable or biodegradable polymeric units. Functional groups that are exploitable for this purpose include those that are reactive with ligands or anti-ligands, such as carboxyl groups, amine groups, sulfhydryl groups and the like. Preferred binding enhancement moieties include the terminal carboxyl groups of the preferred (lactide-glycolide) polymer containing matrix or the like. A practitioner in the art is capable of
selecting appropriate functional groups and monitoring conjugation reactions involving those functional groups.
Advantages garnered through the use of particulate clearing agents of the type described above are as follows:
- particles in the "micron" size range localize in the RES and liver, with galactose derivatization or charge modification enhancement methods for this capability available, and, preferably, are designed to remain in circulation for a time sufficient to perform the clearance function;
- the size of the particulates facilitates central vascular compartment retention thereof, substantially precluding equilibration into the peripheral or extravascular compartment; - desired substituents for ligand or anti-ligand binding to the particulates can be introduced into the polymeric structure;
- ligand- or anti-ligand-particulate linkages having desired properties (e.g., serum biotinidase resistance thereby reducing the release of biotin metabolite from a particle-biotin clearing agent) and
- multiple ligands or anti-ligands can be bound to the particles to achieve optimal cross-linking of circulating targeting agent-ligand or -anti-ligand conjugate and efficient clearance of cross-linked species. This advantage is best achieved when care is taken to prevent particulate aggregation both in storage and upon in vivo administration.
Clearance of NR-LU-13-avidin may also be
accelerated by an arterially inserted proteinaceous or polymeric multiloop device. A catheter-like device, consisting of thin loops of synthetic polymer or protein fibers derivitized with biotin, is inserted into a major artery (e.g., femoral artery) to capture NR-LU-13-avidin. Since the total blood volume passes through a major artery every 70 seconds, the in situ clearing device is effective to reduce circulating NR-LU-13-avidin within a short period of time. This device offers the advantages that NR-LU-13-avidin is not processed through the RES; removal of NR-LU-13-avidin is controllable and measurable; and fresh devices with undiminished binding capacity are
insertable as necessary. This methodology is also useful with intraarterial administration embodiments of the present invention.
An alternative procedure for clearing NR-LU-13-avidin from the circulation without induction of internalization involves administration of
biotinylated, high molecular weight molecules, such as liposomes, IgM and other molecules that are size excluded from ready permeability to tumor sites. When such biotinylated, high molecular weight molecules aggregate with NR-LU-13-avidin, the aggregated
complexes are readily cleared from the circulation via the RES.
Example X
Enhancement of Therapeutic Agent Internalization
Through Avidin Crosslinking
The ability of multivalent avidin to crosslink two or more biotin molecules (or chelate-biotin
conjugates) is advantageously used to improve delivery of therapeutic agents. More specifically, avidin crosslinking induces internalization of crosslinked complexes at the target cell surface.
Biotinylated NR-CO-04 (lysine) is prepared
according to the methods described in Example IV.A., above. Doxorubicin-avidin conjugates are prepared by standard conjugation chemistry. The biotinylated NR-CO-04 is administered to a recipient and allowed to clear from the circulation. One of ordinary skill in the art of radioimmunotherapy is readily able to determine the optimal time for biotinylated NR-CO-04 tumor localization and clearance from the circulation. At such time, the doxorubicin-avidin conjugate is administered to the recipient. The avidin portion of the doxorubicin-avidin conjugate crosslinks the biotinylated NR-CO-04 on the cell surface, inducing internalization of the complex. Thus, doxorubicin is more efficiently delivered to the target cell.
In a first alternative protocol, a standard three-step pretargeting methodology is used to enhance intracellular delivery of a drug to a tumor target cell. By analogy to the description above,
biotinylated NR-LU-05 is administered, followed by avidin (for blood clearance and to form the middle layer of the sandwich at the target cell-bound
biotinylated antibody). Shortly, thereafter, and prior to internalization of the biotinylated NR-LU-05-avidin complex, a methotrexate-biotin conjugate is
administered.
In a second alternative protocol, biotinylated NR-LU-05 is further covalently linked to methotrexate. Subsequent administration of avidin induces
internalization of the complex and enhances
intracellular delivery of drug to the tumor target cell.
In a third alternative protocol, NR-CO-04-avidin is administered to a recipient and allowed to clear from the circulation and localize at the target site. Thereafter, a polybiotinylated species (such as biotinylated poly-L-lysine, as in Example IV. B., above) is administered. In this protocol, the drug to be delivered may be covalently attached to either the antibody-avidin component or to the polybiotinylated species. The polybiotinylated species induces
internalization of the (drug)-antibody-avidin-polybiotin-(drug) complex.
Example XI
Targeting Moiety-Anti-Ligand Conjugate for Two-Step
Pretargeting In vivo
A. Preparation of SMCC-derivitized streptavidin. 31 mg (0.48 μmol) streptavidin was dissolved in 9.0 ml PBS to prepare a final solution at 3.5 mg/ml. The pH of the solution was adjusted to 8.5 by addition of 0.9 ml of 0.5 M borate buffer, pH 8.5. A DMSO solution of SMCC (3.5 mg/ml) was prepared, and 477 μl (4.8 μmol) of this solution was added dropwise to the vortexing protein solution. After 30 minutes of stirring, the solution was purified by G-25 (PD-10, Pharmacia, Piscataway, New Jersey) column
chromatography to remove unreacted or hydrolyzed SMCC. The purified SMCC-derivitized streptavidin was
isolated (28 mg, 1.67 mg/ml). B. Preparation of DTT-reduced NR-LU-10. To 77 mg NR-LU-10 (0.42 μmol) in 15.0 ml PBS was added 1.5 ml of 0.5 M borate buffer, pH 8.5. A DTT solution, at 400 mg/ml (165 μl) was added to the protein solution. After stirring at room temperature for 30 minutes, the reduced antibody was purified by G-25 size exclusion chromatography. Purified DTT-reduced NR-LU-10 was obtained (74 mg, 2.17 mg/ml).
C. Conjugation of SMCC-streptavidin to DTT- reduced NR-LU-10. DTT-reduced NR-LU-10 (63 mg, 29 ml, 0.42 μmol) was diluted with 44.5 ml PBS. The solution of SMCC-streptavidin (28 mg, 17 ml, 0.42 μmol) was added rapidly to the stirring solution of NR-LU-10. Total protein concentration in the reaction mixture was 1.0 mg/ml. The progress of the reaction was monitored by HPLC (Zorbax® GF-250, available from MacMod) . After approximately 45 minutes, the reaction was quenched by adding solid sodium tetrathionate to a final concentration of 5 mM.
D. Purification of conjugate. For small scale reactions, monosubstituted conjugate was obtained using HPLC Zorbax (preparative) size exclusion
chromatography. The desired monosubstituted conjugate product eluted at 14.0-14.5 min (3.0 ml/min flow rate), while unreacted NR-LU-10 eluted at 14.5-15 min and unreacted derivitized streptavidin eluted at 19-20 min.
For larger scale conjugation reactions,
monosubstituted adduct is isolatable using DEAE ion exchange chromatography. After concentration of the crude conjugate mixture, free streptavidin was removed therefrom by eluting the column with 2.5% xylitol in sodium borate buffer, pH 8.6. The bound unreacted antibody and desired conjugate were then sequentially eluted from the column using an increasing salt gradient in 20 mM diethanolamine adjusted to pH 8.6 with sodium hydroxide. E. Characterization of Conjugate.
1. HPLC size exclusion was conducted as described above with respect to small scale purification.
2. SDS-PAGE analysis was performed using 5% polyacrylamide gels under non-denaturing conditions. Conjugates to be evaluated were not boiled in sample buffer containing SDS to avoid dissociation of
streptavidin into its 15 kD subunits. Two product bands were observed on the gel, which correspond to the mono- and di- substituted conjugates.
3. Immunoreactivity was assessed, for example, by competitive binding ELISA as compared to free
antibody. Values obtained were within 10% of those for the free antibody.
4. Biotin binding capacity was assessed, for example, by titrating a known quantity of conjugate with p-[I-125]iodobenzoylbiocytin. Saturation of the biotin binding sites was observed upon addition of 4 equivalences of the labeled biocytin.
5. In vivo studies are useful to characterize the reaction product, which studies include, for example, serum clearance profiles, ability of the conjugate to target antigen-positive tumors, tumor retention of the conjugate over time and the ability of a biotinylated molecule to bind streptavidin conjugate at the tumor. These data facilitate determination that the synthesis resulted in the formation of a 1:1 streptavidin-NR-LU-10 whole antibody conjugate that exhibits blood clearance properties similar to native NR-LU-10 whole antibody, and tumor uptake and retention properties at least equal to native NR-LU-10.
For example, Figure 3 depicts the tumor uptake profile of the NR-LU-10-streptavidin conjugate (LU-10-StrAv) in comparison to a control profile of native NR-LU-10 whole antibody. LU-10-StrAv was radiolabeled on the streptavidin component only, giving a clear indication that LU-10-StrAv localizes to target cells as efficiently as NR-LU-10 whole antibody itself.
Example XII
Two-Step Pretargeting In Vivo
A 186Re-chelate-biotin conjugate (Re-BT) of Example I (MW a 1000; specific activity = 1-2 mCi/mg) and a biotin-iodine-131 small molecule, PIP-Biocytin (PIP-BT, MW approximately equal to 602; specific activity = 0.5-1.0 mCi/mg), as discussed in Example VII above, were examined in a three-step pretargeting protocol in an animal model, as described in Example V above.
Like Re-BT, PIP-BT has the ability to bind well to avidin and is rapidly cleared from the blood, with a serum half-life of about 5 minutes. Equivalent results were observed for both molecules in the two-step pretargeting experiments described herein.
NR-LU-10 antibody (MW ≈ 150 kD) was conjugated to streptavidin (MW ≈ 66 kD) (as described in Example XI above) and radiolabeled with 125I/PIP-NHS (as described for radioiodination of NR-LU-10 in Example IV.A., above). The experimental protocol was as follows:
Time 0 inject (i.v.) 200 μg NR-LU-10-StrAv
conjugate;
Time 24-48 h inject (i.v.) 60-70 fold
molar excess of
radiolabeled biotinyl
molecule ;
and perform biodistributions at 2, 6, 24, 72, 120 hours after injection of radiolabeled biotinyl molecule
NR-LU-10-streptavidin has shown very consistent patterns of blood clearance and tumor uptake in the LS-180 animal model. A representative profile is shown in Figure 4. When either PIP-BT or Re-BT is administered after allowing the LU-10-StrAv conjugate to localize to target cell sites for at least 24 hours, the tumor uptake of therapeutic radionuclide is high in both absolute amount and rapidity. For PIP-BT administered at 37 hours following LU-10-StrAv (1-125) administration, tumor uptake was above 500 pMOL/G at the 40 hour time point and peaked at about 700 pMOL/G at 45 hours post-LU-10-StrAv administration.
This almost instantaneous uptake of a small molecule therapeutic into tumor in stoichiometric amounts comparable to the antibody targeting moiety facilitates utilization of the therapeutic
radionuclide at its highest specific activity. Also, the rapid clearance of radionuclide that is not bound to LU-10-StrAv conjugate permits an increased
targeting ratio (tumor:blood) by eliminating the slow tumor accretion phase observed with directly labeled antibody conjugates. The pattern of radionuclide tumor retention is that of whole antibody, which is very persistent.
Experimentation using the two-step pretargeting approach and progressively lower molar doses of radiolabeled biotinyl molecule was also conducted. Uptake values of about 20% ID/G were achieved at no-carrier added (high specific activity) doses of radiolabeled biotinyl molecules. At less than
saturating doses, circulating LU-10-StrAv was observed to bind significant amounts of administered
radiolabeled biotinyl molecule in the blood
compartment. Example XIII
Asialoorosomucoid Clearing Agent and Two-Step
Pretargeting
In order to maximize the targeting ratio
(tumor:blood), clearing agents were sought that are capable of clearing the. blood pool of targeting moiety-anti-ligand conjugate (e.g., LU-10-StrAv), without compromising the ligand binding capacity thereof at the target sites. One such agent,
biotinylated asialoorosomucoid, which employs the avidin-biotin interaction to conjugate to circulating LU-10-StrAv, was tested.
A. Derivitization of orosomucoid. 10 mg human orosomucoid (Sigma N-9885) was dissolved in 3.5 ml of pH 5.5 0.1 M sodium acetate buffer containing 160 mM NaCl. 70 μl of a 2% (w/v) CaCl solution in deionized (D.I.) water was added and 11 μl of neuraminidase (Sigma N-7885), 4.6 U/ml, was added. The mixture was incubated at 37°C for 2 hours, and the entire sample was exchanged over a Centricon-10® ultrafiltration device (available from Amicon, Danvers, Massachusetts) with 2 volumes of PBS. The asialoorosomucoid and orosomucoid starting material were radiolabeled with
1-125 using PIP technology, as described in Example IV above.
The two radiolabeled preparations were injected i.v. into female BALB/c mice (20-25 g), and blood clearance was assessed by serial retro-orbital eye bleeding of each group of three mice at 5, 10, 15 and 30 minutes, as well as at 1, 2 and 4 hours post-administration. The results of this experiment are shown in Figure 5, with asialoorosomucoid clearing more rapidly than its orosomucoid counterpart.
. In addition, two animals receiving each compound were sacrificed at 5 minutes post-administration and limited biodistributions were performed. These results are shown in Figure 6. The most striking aspects of these data are the differences in blood levels (78% for orosomucoid and 0.4% for
asialoorosomucoid) and the specificity of uptake of asialoorosomucoid in the liver (86%), as opposed to other tissues.
B. Biotinylation of asialoorosomucoid clearing agent and orosomucoid control. 100 μl of 0.2 M sodium carbonate buffer, pH 9.2, was added to 2 mg (in 1.00 ml PBS) of PIP-125-labeled orosomucoid and to 2 mg PIP-125-labeled asialoorosomucoid. 60 μl of a 1.85 mg/ml solution of NHS-amino caproate biotin in DMSO was then added to each compound. The reaction
mixtures were vortexed and allowed to sit at room temperature for 45 minutes. The material was purified by size exclusion column chromatography (PD-10,
Pharmacia) and eluted with PBS. 1.2 ml fractions were taken, with fractions 4 and 5 containing the majority of the applied radioactivity (>95%). Streptavidin-agarose beads (Sigma S-1638) or -pellets were washed with PBS, and 20 μg of each biotinylated, radiolabeled protein was added to 400 μl of beads and 400 μl of PBS, vortexed for 20 seconds and centrifuged at 14,000 rpm for 5 minutes. The supernatant was removed and the pellets were washed with 400 μl PBS. This wash procedure was repeated twice more, and the combined supernatants were assayed by placing them in a
dosimeter versus their respective pellets. The values are shown below in Table 4.
TABLE 4
Compound Supernatant Pellet orosomucoid 90% 10% biotin-oroso 7.7% 92.% asialoorosomucoid 92% 8.0% biotin-asialo 10% 90%
C. Protein-Streptavidin Binding in vivo. Biotin-asialoorosomucoid was evaluated for the ability to couple with circulating LU-10-StrAv conjugate in vivo and to remove it from the blood. Female BALB/c mice (20-25 g) were injected i.v. with 200 μg LU-10-StrAv conjugate. Clearing agent (200 μl PBS - group 1; 400 μg non-biotinylated asialoorosomucoid - group 2; 400 μg biotinylated asialoorosomucoid - group 3; and 200 μg biotinylated asialoorosomucoid- group 4) was administered at 25 hours following conjugate
administration. A fifth group received PIP-I-131-LU-10-StrAv conjugate which had been saturated prior to injection with biotin - group 5. The 400 μg dose constituted a 10:1 molar excess of clearing agent over the initial dose of LU-10-StrAv conjugate, while the 200 μg dose constituted a 5:1 molar excess. The saturated PIP-I-131-LU-10-StrAv conjugate was produced by addition of a 10-fold molar excess of D-biotin to 2 mg of LU-10-StrAv followed by size exclusion
purification on a G-25 PD-10 column.
Three mice from each group were serially bled, as described above, at 0.17, 1, 4 and 25 hours (pre-injection of clearing agent), as well as at 27, 28, 47, 70 and 90 hours. Two additional animals from each group were sacrificed at 2 hours post-clearing agent administration and limited biodistributions were performed.
The blood clearance data are shown in Figure 7. These data indicate that circulating LU-10-StrAv radioactivity in groups 3 and 4 was rapidly and significantly reduced, in comparison to those values obtained in the control groups 1, 2 and 5. Absolute reduction in circulating antibody-streptavidin conjugate was approximately 75% when compared to controls.
Biodistribution data are shown in tabular form in Figure 8. The biodistribution data show reduced levels of conjugate for groups 3 and 4 in all tissues except the liver, kidney and intestine, which is consistent with the processing and excretion of radiolabel associated with the conjugate after complexation with biotinylated asialoorosomucoid.
Furthermore, residual circulating conjugate was obtained from serum samples by cardiac puncture (with the assays conducted in serum + PBS) and analyzed for the ability to bind biotin (immobilized biotin on agarose beads), an indicator of functional
streptavidin remaining in the serum. Group 1 animal serum showed conjugate radiolabel bound about 80% to immobilized biotin. Correcting the residual
circulating radiolabel values by multiplying the remaining percent injected dose (at 2 hours after clearing agent administration) by the remaining percent able to bind immobilize biotin (the amount of remaining functional conjugate) leads to the graph shown in Figure 9. Administration of 200 μg
biotinylated asialoorosomucoid resulted in a 50-fold reduction in serum biotin-binding capacity and, in preliminary studies in tumored animals, has not exhibited cross-linking and removal of prelocalized LU-10-StrAv conjugate from the tumor. Removal of circulating targeting moiety-anti-ligand without diminishing biotin-binding capacity at target cell sites, coupled with an increased radiation dose to the tumor resulting from an increase in the amount of targeting moiety-anti-ligand administered, results in both increased absolute rad dose to tumor and
diminished toxicity to non-tumor cells, compared to what is currently achievable using conventional radioimmunotherapy.
A subsequent experiment was executed to evaluate lower doses of asialoorosomucoid-biotin. In the same animal model, doses of 50, 20 and 10 μg
asialoorosomucoid-biotin were injected at 24 hours following administration of the LU-10-StrAv conjugate. Data from animals serially bled are shown in Figure 10, and data from animals sacrificed two hours after clearing agent administration are shown in Figures 11A (blood clearance) and 11B (serum biotin-binding), respectively. Doses of 50 and 20 μg
asialoorosomucoid-biotin effectively reduced
circulating LU-10-StrAv conjugate levels by about 65% (Figure 11A) and, after correction for binding to immobilized biotin, left only 3% of the injected dose in circulation that possessed biotin-binding capacity, compared with about 25% of the injected dose in control animals (Figure 11B). Even at low doses
(approaching 1:1 stoichiometry with circulating LU-10-StrAv conjugate), asialoorosomucoid-biotin was highly effective at reducing blood levels of circulating streptavidin-containing conjugate by an in vivo
complexation that was dependent upon biotin-avidin interaction.
Example XIV
Streptavidin Anti-Ligand in Tumors A set of female nude mice, implanted
subcutaneously with LS-180 human colon carcinoma xenografts as described above, were randomized into groups of 4 animals/timepoint. The mice were
intravenously injected with 200 μg of 1:1 mol/mol NR-LU-10 monoclonal antibody covalently coupled to streptavidin (MAB-STRPT), with the conjugate formed as described in Example XI above. The streptavidin portion of the conjugate was radiolabeled with
paraiodophenyl (PIP) 1-125, as described in Example IV above. Groups of mice were sacrificed at 26, 30, 48, 96 and 144 hours post-conjugate injection. Tissues were isolated, weighed and counted with respect to iodine radionuclide content using conventional
procedures therefor.
A second set of female nude mice bearing LS-180 xenografts were also randomized into groups of 4 animals/timepoint. These mice were intravenously injected with 50 μg of NR-LU-10 monoclonal antibody radiolabeled with paraiodophenyl (PIP) I-131 (MAB), as described in Example IV above. Mice were sacrificed at 4, 24, 48, 128 and 168 hours post-radiolabeled monoclonal antibody injection. Tissues were isolated, weighed and counted with respect to iodine radionuclide content using conventional procedures therefor.
For each data set, a radioactivity standard of the injected dose was also counted, and data were reduced to a percent of the total injected dose per gram of tissue. Figure 12 shows the percent injected
dose/gram of NR-LU-10-streptavidin-PIP-I-125 and NR-LU-10-PIP-I-131 in LS-180 tumors over time. The NR-LU-10-streptavidin conjugate exhibits higher tumor uptake and a longer retention time as compared to NR-LU-10 alone.
Example XV
Streptavidin Anti-Ligand in Liver Female nude mice xenografted with LS-180 tumor cells, as discussed above, were randomized into groups of 4 animals/timepoint. Mice were intravenously injected with 50 μg of biotinylated NR-LU-10
monoclonal antibody that was non-covalently coupled (to form a complex) through biotin-streptavidin binding to 30 μg of streptavidin. Prior to
complexation in vivo, the antibody portion of the complex was radiolabeled with 1-125 using chloramine-T, and the streptavidin portion was labeled with paraiodophenyl (PIP) I-131, both of the labeling procedures having been described above. Mice were sacrificed at 4, 24, 48, 96 and 144 hours post-conjugate injection. Tissues were isolated, weighed and counted with respect to the content of each iodine radionuclide using conventional procedures therefor.
A radioactivity standard of the injected doses of each complex component was also counted, and data were reduced to a percent of the total injected dose per gram of tissue. Figure 13 shows the percent injected dose per gram of streptavidin-PIP-I-131 (STREPT) and NR-LU-10-biotin-Chioramine-T-I-125 (MAB-BT) in liver over time. The complex localized at the liver as a single molecule; however, the processing of the individual components thereof differed in the liver. The I-131-streptavidin label showed prolonged
residence in the liver, while the monoclonal antibody label (1-125) was rapidly lost.
In another liver study, female nude mice
xenografted with xenografted with LS-180 tumor cells, as discussed above, and were intravenously injected with 200 μg of 1:1 mol/mol NR-LU-10 monoclonal
antibody covalently coupled to streptavidin, prepared as described in Example XI above. The antibody portion of the conjugate was radiolabeled with
paraiodophenyl (PIP-I-125). Twenty four hours later, the mice received an injection of 0.5 μg of
paraiodophenyl (PIP 1-131) biocytin. Mice were sacrificed at 28, 48, 120 and 168 hours post-conjugate injection. Tissues were isolated, weighed and counted with respect to the content of each iodine
radionuclide using conventional procedures therefor.
A radioactivity standard of the injected doses of each complex component was also counted, and data were reduced to a percent of the total injected dose per gram of tissue (%ID/G). Figure 14 shows the percent injected dose per gram of streptavidin-monoclonal antibody-PIP-I-125 (STREP-MAB-I-125) and biocytin-PIP-1-131 (BT-I-131) in liver over time. When biocytin-PIP-I-131 was subsequently administered, the retention of streptavidin-bound biotin radiolabel (1-131) was prolonged relative to the retention of the antibody-bound label (1-125) on the same moiety in the liver.
Example XVI
Tumor Uptake of PIP-Biocytin
PIP-Biocytin, as prepared and described in Example VII above, was tested to determine the fate thereof in vivo. The following data are based on experimentation with tumored nude mice (100 mg LS-180 tumor xenografts implanted subcutaneously 7 days prior to study) that received, at time 0, 200 μg of 1-125 labeled NR-LU-10-Streptavidin conjugate (950 pmol), as discussed in Example XI above. At 24 hours, the mice received an i.v. injection of PIP-I-131-biocytin (40 μCi) and an amount of cold carrier PIP-I-127 biocytin
corresponding to doses of 42 μg (69,767 pmol), 21 μg (34,884 pmol), 5.7 μg (9468 pmol), 2.85 μg (4734 pmol) or 0.5 μg (830 pmol). Tumors were excised and counted for radioactivity 4 hours after PIP-biocytin
injection, and the tumor uptake data are shown in Figures 15A (%ID/G v. Dose) and 15B (pMOL/G v. Dose).
The three highest doses produced PIP-biocytin tumor localizations of about 600 pmol/g. Histology conducted on tissues receiving the two highest doses indicated that saturation of tumor-bound streptavidin was achieved. Equivalent tumor localization observed at the 5.7 μg dose (Figure 15B) is indicative of streptavidin saturation as well. In contrast, the two lowest doses produced lower absolute tumor
localization of PIP-biocytin, despite equivalent localization of NR-LU-10-Streptavidin conjugate
(tumors in all groups averaged about 40% ID/G for the conjugate).
The lowest dose group (0.5 μg) exhibited high efficiency tumor delivery of PIP-I-131-biocytin, which efficiency increased over time, as shown in Figure 16A. A peak uptake of 85.0 % ID/G was observed at the 120 hour time point (96 hours after administration of PIP-biocytin). Also, the absolute amount of PIP-biocytin, in terms of % ID, showed a continual
increase in the tumor over all of the sampled time points (Figure 16B). The decrease in uptake on a % ID/G basis (Figure 16A) at the 168 hour time point resulted from significant growth of the tumors between the 120 and 168 hour time points. In addition. Figure 17A shows the co-localization of NR-LU-10-Streptavidin conjugate (LU-10-StrAv) and the subsequently administered PIP-Biocytin at the same tumors over time. The localization of radioactivity at tumors by PIP-biocytin exhibited a pattern of uptake and retention that differed from that of the antibody-streptavidin conjugate (LU-10-StrAv). LU-10-StrAv exhibited a characteristic tumor uptake pattern that is equivalent to historical studies of native NR-LU-10 antibody, reaching a peak value of 40% ID/G between 24 and 48 hours after administration. In contrast, the PIP-Biocytin exhibited an initial rapid accretion in the tumor, reaching levels greater than those of LU-10-StrAv by 24 hours after PIP-Biocytin administration. Moreover, the localization of PIP- Biocytin continued to increase out to 96 hours, when the concentration of radioactivity associated with the conjugate has begun to decrease. The slightly greater amounts of circulating PIP-Biocytin compared to LU-10-StrAv at these time points (shown in Figure 17B) appeared insufficient to account for this phenomenon.
As Figure 18 clearly shows, the ratio of PIP-Biocytin to LU-10-StrAv in the tumor increased
continually during the experiment, while the ratio in the blood decreased continually. This observation is consistent with a process involving continual binding of targeting moiety-containing conjugate (with PIP-Biocytin bound to it) from the blood to the tumor, with subsequent differential processing of the PIP-Biocytin and the conjugate. Since radiolabel
associated with the streptavidin conjugate component (compared to radiolabel associated with the targeting moiety) has shown increased retention in organs of metabolic processing (Examples XIV and XV above), PIP-Biocytin associated with the streptavidin appears to be selectively retained by the tumor cells. Because radiolabel is retained at target cell sites, a greater accumulation of radioactivity at those sites results.
The AUCtumor/AUCblood for PIP-Biocytin is over twice that of the conjugate (4.27 compared to 1.95, where AUC means "area under the curve"). Further, the absolute AUCtunor for PIP-Biocytin is nearly twice that of the conjugate (9220 compared to 4629).
Consequently, an increase in radiation dose to tumor was achieved.
Example XVII
Polymer-Ligand Conjugation Polylysine (approximately 10,000 Dal. molecular weight, available from Sigma Chemical Co., St. Louis, Missouri) and dextran (lysine fixable, available from Sigma Chemical Co.) were derivitized with SPDP and purified from unreacted SPDP using size exclusion chromatography (using a PD-10 column available from Pharmacia, Piscataway, New Jersey). The resultant
SPDP-derivitized adducts were reduced with DTT in pH 4.7 0.2 M NaOAc buffer to generate free reactive thiols. Reduced Tc-99m, generated from stannous gluconate as described, for example, by Carlsson et al., Biochem. J., 173: 723-737, 1978, was added. A 90% incorporation of Tc-99m was obtained for the polylysine adduct within 15 min, as measured by ITLC. 96% of the radioactivity coeluted with the dextran using size exclusion (PD-10) chromatography. These results are indicative of chelation. EXAMPLE XVIII
Preparation of Trichothecene-Linker Molecules A. Preparation of 3-(2-Pyridinyldithio)propanoic acid. 5.00 g (52.2 mmol) of 3-mercaptopropanoic acid (Aldrich Chemical Co., Milwaukee, Wisconsin) in 75 ml of dry methylene chloride was added to a solution of 5.96 g (52.2 mmol) of methoxycarbonylsulfenyl chloride (Fluka Chemika, Long Island, New York) in 150 ml of dry methylene chloride. The mixture was stirred at 15-25ºC for 90 minutes and then concentrated. The residue was redissolved in 150 ml of dry methylene chloride and dropwise treated with 5.80 g (52.2 mmol) of 2-mercaptopyridine (Aldrich Chemical Co.) in 75 ml of dry methylene chloride. The mixture was stirred at 15-25 ºC for 18 hours and concentrated to afford 11.2 g of the product as a pale yellow oil (99%).
B. Preparation of 3-(2-pyridinyldithio)propanoic acid hydrazide. Dry triethylamine (3.76 g, 37.2 mmol) was added to a solution of 8.00 g (37.2 mmol) of 3-(2-pyridinyldithio)propanoic acid in 100 ml of dry tetrahydrofuran (THF). The mixture was cooled in an ice bath followed by the addition of 5.08 g (37.2 mmol) of isobutyl chloroformate. The mixture was stirred for 5-10 minutes, and 4.92 g (37.2 mmol) of tert-butyl carbazate was added. The resultant mixture was stirred at 15-25 ºC for l hour and then
concentrated. The residue was diluted with 200 ml of methylene chloride and washed with water (2 × 100 ml). The organic phase was dried over magnesium sulfate, filtered and concentrated. The residue was dissolved in ice-cold trifluoroacetic acid (160 ml) and stirred for 10 minutes after dissolution was complete. The mixture was concentrated, and the residue was
chromatographed on silica gel, eluting with 85:14:1 chloroform?methanol/ammonium hydroxide, to afford 4.25 g of the product as a pale yellow solid (50%) : TLC-Rf 0.55 (85:15:1 chloroform/methanol/ ammonium
hydroxide).
C. Preparation of Hydrazone Derivative of 3-Dehydroanguidine and 3-(2-Pyridinylithio)propanoic Acid Hydrazide. 100 microliters (0.708 mmol) of trifluoroacetic anhydride at -70ºC was added to a solution of 100 microliters of dry dimethylsulfoxide (1.41 mmol) in 3 ml of dry methylene chloride. The mixture was stirred at -70ºC for 10 minutes and then 70 mg (0.198 mmol) of anguidine (Sigma Chemical Co.) in 2 ml of dry methylene chloride was added over a period of 2-3 minutes. The resultant mixture was stirred at -70 ºC for 15 minutes and then 20
microliters of dry triethylamine was added. The mixture was stirred at -70ºC for 15 minutes and at -70ºC to 15ºC for 60 minutes. The resultant mixture was diluted with 50 ml of methylene chloride and washed with 1 N aqueous HCl (50 ml). The organic phase was dried over magnesium sulfate, filtered and concentrated to afford crude 3-dehydroanguidine. The crude material was dissolved in 3 ml of dry methanol and treated with 100 mg (0.436 mmol) of 3-(2-pyridinyldithio)propanoic acid hydrazide followed by 0.021 mmol of trifluoroacetic acid in 100 microliters of methanol. The mixture was stirred at 15-25 "C for 5 hours and then concentrated. The residue was chromatographed on silica gel, eluting with 75% ethyl acetate/hexane, to afford 67 mg of the product, a foamy white solid, as a mixture of syn and anti isomers (60%): TLC-Rf 0.43 and 0.60 (75% ethyl acetate/hexane).
D. Preparation of a Hydrazone Derivative of 2'-dehydro-Roridin A.
1. Preparation of 13'-O-tert-butyldimethylsilyl-Roridin A. 100 mg (1.47 mmol) of imidazole was added to a solution of 232 mg (0.436 mmol) of Roridin A in 3 ml of dry dimethylformamide. The mixture was cooled to -5 ºC and then 72 mg (0.478 mmol) of tert-butyldimethylsilyl chloride (Aldrich Chemical Co.) was added. The resultant mixture was stirred at -5ºC to 5ºC for 4 hours and then concentrated. The residue was chromatographed on silica gel, eluting first with 30% ethyl acetate/hexane, next with 50% ethyl acetate/hexane and finally with 70% ethyl
acetate/hexane, to afford 137 mg of the product as a foamy white solid (49%): TLC-Rf 0.32 (30% ethyl acetate/hexane).
2. Preparation of 2'-dehydro-13'-O-tert-butyldimethylsilyl-Roridin A. At -70ºC, 150
microliters (1.06 mmol) of trifluoroacetic anhydride was added to a solution of 100 microliters (1.41 mmol) of dry dimethylsulfoxide in 3 ml of dry methylene chloride. The mixture was stirred at -70ºC for 15 minutes and then 100 mg (0.155 mmol) of 13'-O-tert-butyldimethylsilyl-Roridin A in 2 ml of dry methylene chloride was added. The resultant mixture was stirred at -70ºC for 15 minutes and then 300 microliters (2.15 mmol) of dry triethylamine was added. This mixture was stirred at -70ºC for 30 minutes and at -70ºC to 15ºC for 30 minutes. The resultant mixture was then concentrated and the residue was
chromatographed on silica gel, eluting with 30% ethyl acetate/hexane, to afford 82 mg of the product as a foamy white solid (82%): TLC-Rf 0.51 (30% ethyl acetate/hexane).
3. Preparation of 2'-dehydro-Roridin A. 5 ml of 3:1:1 acetic acid:THF:water was added to a 10 ml round bottom flask, charged with 62 mg (0.096 mmol) of 2'-dehydro-13"-O-tert-butyldimethylsilyl-Roridin A. The mixture was stirred at 45-50ºC for 4.5 hours, cooled and concentrated. The residue was chromatographed on silica gel, eluting with 5% methanol/methylene
chloride, to afford 36 mg of the product as a foamy white solid (70%): TLC-Rf 0.48 (5% methanol/methylene chloride).
4. Preparation of a hydrazone derivative of 2'-dehydro-Roridin A and 3-(2-pyridinyldithio)propanoic acid hydrazide. To a solution of 25 mg (0.471 mmol) of 2'-dehydro-Roridin A in 2 ml of dry methanol was added 30 mg (0.131 mmol) of 3-(2-pyridinyldithio) propanoic acid hydrazide followed by 0.013 mmol trifluoroacetic acid. The mixture was stirred at 15-25"C for 4 hours and then concentrated. The residue was chromatographed on silica gel, eluting with 75% ethyl acetate/hexane, to afford 25 mg of the product as a mixture of syn and anti isomers (71%) : TLC-Rf 0.29 and 0.53 (75% ethyl acetate/hexane).
E. Preparation of a Hydrazone Derivative of 2'-O-Acetyl-13'-Dehydro-Roridin A.
1. Preparation of 2'-O-acetyl-13'-O-tert-butyl dimethylsilyl-Roridin A. To a solution of 159 mg (0.246 mmol) of 13'-O-tert-butyldimethylsilyl Roridin A in 4 ml of dry methylene chloride was added 200 microliters (1.43 mmol) of triethylamine, 2 mg (0.019 mmol) of dimethylaminopyridine and 120 microliters
(1.27 mmol) acetic anhydride. The mixture was stirred at 15-25ºC for 16 hours and then concentrated. The residue was chromatographed on silica gel, eluting first with 30% ethyl acetate/hexane and then with 50% ethyl acetate/hexane, to afford 156 mg of the product as a foamy white solid (92%): TLC-Rf 0.50 (30% ethyl acetate/hexane).
2. Preparation of 2'-O-acetyl-Roridin A. To a solution of 156 mg (0.226 mmol) of 2'-O-acetyl-13'-tert-butyldimethylsilyl-Roridin A in 3 ml of dry THF was added 1.5 ml of 1M tetrabutyl ammonium fluoride. The mixture was stirred at 15-25ºC for 4 hours and then concentrated. The residue was chromatographed on silica gel, eluting with 50% ethyl acetate/hexane, to afford 125 mg of the product as a foamy white solid (96%): TLC-Rf 0.17 (50% ethyl acetate/hexane).
3. Preparation of 2'-O-acetyl-13'-dehydro-Roridin A. To a solution of 62 microliters (0.87 mmol) of dry dimethylsulfoxide in 2 ml of dry methylene chloride at -70ºC was added 62 microliters (0.439 mmol)
trifluoroacetic anhydride. The mixture was stirred at -70ºC for 10 minutes followed by the addition of 25 mg (0.044 mmol) of 2'-O-acetyl-Roridin A in 1.5 ml of dry methylene chloride over a 2-3 minute period. This mixture was stirred at -70ºC for 20 minutes and then 180 microliters (1.29 mmol) of dry triethylamine was added. The resultant mixture was stirred at -70ºC for 15 minutes and at -70ºC to 15ºC for 20 minutes and then concentrated. The residue was chromatographed on silica gel, eluting with 5% methanol/methylene
chloride, to afford 21 mg of the product as a foamy white solid.
4. Preparation of a hydrazone derivative of 2'O-acetyl-13'-dehydro-Roridin A and 3-(2-pyridinyldithio) propanoic acid hydrazide. To a solution of 21 mg (0.037 mmol) of 2'-O-acetyl-13'-dehydro-Roridin A in 3 ml of dry methanol was added 25 mg (0.11 mmol) of 3-(2-pyridinyldithio) propanoic acid hydrazide followed by 0.011 mmol of trif-luoroacetic acid in 100
microliters of dry methanol. The mixture was stirred at 15-25ºC for 3 hours and then concentrated. The residue was chromatographed on silica gel, eluting first with 75% ethyl acetate/hexane and then with ethyl acetate, to afford 18 mg of the product as a colorless oil (62%): TLC-Rf 0.30 (75% ethyl
acetate/hexane).
F. Preparation of 2'-Desoxy-2'-alpha-(N-hydroxysuccinimidyl-3-dithiopropanoic acid)-Roridin A.
1. Preparation of 2'-desoxy-2'-beta-iodo-13'-O-tert-butyldimethylsilyl-Roridin A. To a solution of 99 mg (0.153 mmol) of 13'-O-tert-butyldimethylsilyl-Roridin A in 5 ml of dry methylene chloride, at 0ºC, was added 80 microliters (0.459 mmol) of dry diisopropylethylamine followed by 51 microliters (0.303 mmol) of trifluoromethanesulfonic anhydride. The mixture was stirred at 0ºC for 30 minutes and then diluted with 30 ml of methylene chloride and washed with 20 ml of 1N aqueous HCl. The organic phase was dried over magnesium sulfate, filtered and concentrated. The residue was diluted with 5 ml of acetone and then 235 mg (1.72 mmol) of sodium iodide. The mixture was stirred at reflux for 20 minutes and then diluted with 50 ml of methylene chloride and washed with 25 ml of water. The organic phase was dried over magnesium sulfate, filtered and
concentrated. The residue was chromatographed on silica gel, eluting with 30% ethyl acetate/hexane to afford 95 mg of the product as a foamy white solid (87%): TLC-Rf 0.55 (35% ethyl acetate/hexane).
2. Preparation of 2'-desoxy-2'-beta-iodo-Roridin A. To 95 mg (0.126 mmol) of 2'-desoxy-2'-beta-iodo-13'-O-tert-butyldimethylsilyl-Roridin A was added 5 ml of 3:1:1 acetic acid:THF:water. The mixture was stirred at 60-65ºC for 4 hours and then concentrated. The residue was chromatographed on silica gel, eluting with 60% ethyl acetate/hexane, to afford 67 mg of the product as a foamy white solid (83%).
3. Preparation of 2'-desoxy-2'-alpha-thioacetyl-Roridin A. To a solution of 67 mg (0.104 mmol) of 2'-desoxy-2'-beta-iodo-Roridin A in 5 ml of absolute ethanol was added 240 mg (2.10 mmol) of potassium thioacetate. The mixture was stirred at 65-70ºC for 5 hours, cooled, diluted with 50 ml of methylene
chloride and washed with 50 ml of water. The organic phase was dried over magnesium sulfate, filtered and concentrated. The residue was chromatographed on silica gel, eluting with 60% ethyl acetate/hexane, to afford 58 mg of the product as a foamy white solid (94%): TLC-Rf 0.53 (60%ethyl acetate/hexane).
4. Preparation of 2'-desoxy-2'-alpha-mercaptyl-Roridin A. To a solution of 57 mg (0.097 mmol) of 2'-desoxy-2'-alpha-thioacetyl-Roridin A in 4 ml of ethanol was added 2 ml of 28% aqueous ammonium
hydroxide. The mixture was stirred at 65-70ºC for 6 hours. The mixture was concentrated and the residue was chromatographed on silica gel, eluting with 10% ethyl acetate/hexane, to afford 25 mg of an
intermediate disulfide. The disulfide was diluted into 3 ml of methylene chloride and 0.5 ml of methanol and 25 microliters (0.10 mmol) of tributylphosphine was added. The mixture was stirred at 15-25ºC for 15 minutes and then concentrated. The residue was chromatographed on silica gel, eluting with 60% ethyl acetate/hexane, to afford 23 mg of the product as a colorless oil (43%): TLC-Rf 0.39 (60% ethyl
acetate/hexane).
5. Preparation of 2'-desoxy-2'-alpha-(3-dithiopropanoic acid)-Roridin A. To a solution of 21 mg (0.038 mmol) of 2'desoxy-2'-alpha-mercaptyl-Roridin A in 3 ml of ethanol was added 20 mg (0.093 mmol) of 3-(2-pyridinyldithio) propanoic acid followed by 25 microliters (0.179 mmol) of triethylamine. The mixture was stirred at 15-25ºC for 30 minutes and then diluted with 50 ml of ethyl acetate and washed with 10 ml of 1N aqueous HCl. The organic phase was dried over magnesium sulfate, filtered and concentrated.
The residue was chromatographed on silica gel, eluting first with 0.1% acetic acid/ethyl acetate and then with 0.1:5:95 acetic acid:methanol: ethyl acetate, to afford 22 mg of the product as a near colorless oil (88%): TLC-Rf 0.37 (0.1:5:95 acetic
acid:methanol: ethyl acetate).
6. Preparation of 2'-desoxy-2'-alpha-(N-hydroxy succinimidyl-3-dithiopropanoic acid)-Roridin A. To a solution of 22 mg (0.0337 mmol) of 2'-desoxy-2'-alpha- (3-dithiopropanoic acid) -Roridin A in 2 ml of dry THF was added 12 mg (0.104 mmol) of N-hydroxysuccinimide followed by 18 mg (0.087 mmol) of dicyclohexyl
carbodiimide. The mixture was stirred at 15-25ºC for 4 hours and then 20 microliters of acetic acid was added. The mixture was stirred an additional 30 minutes and then filtered through a plug of glass wool. The solids were washed with 3 ml of THF. The filtrates were combined and concentrated. The residue was chromatographed on silica gel, eluting first with 0.1:50:50 acetic acid: ethyl acetate:hexane and then with 0.1% acetic acid/ethyl acetate, to afford 18 mg of the product as a white solid (71%): TLC-Rf 0.69 (ethyl acetate).
EXAMPLE XIX Preparation of Trichothecene-Containing Conjugates
Conjugation of trichothecenes as described in the specification may be conducted through either of two exemplary synthesis schemes: (1) reaction of the amino groups of a targeting moiety with N-hydroxyl succinimidate (NHS)-derivatized trichothecenes or (2) the reaction of reduced disulfides or free sulfhydryls of a targeting moiety with 2-pyridinyl dithio-derivatized trichothecenes.
A. Antibody Conjugation of NHS-Derivatized
Trichothecenes. This conjugation was generally performed in 6.2 mM sodium borate buffer (0.5 M, pH 8), containing the antibody stabilizer molecusol at 1% wt/vol. The NHS-derivatized trichothecene dissolved in 100% DMSO was added dropwise to a stirred solution of antibody at a final antibody concentration of 1.0 mg/ml reaction solution. To achieve a trichothecene loading of 6-9/antibody, NHS-derivatized trichothecene was offered at a 30:1 trichothecene: antibody molar ratio at a final concentration of 25% DMSO vol/vol. The reaction mixture was incubated with continuous stirring at room temperature for 1 hour prior to purification by size exclusion chromatography, extensive dialysis, concentration via membrane
centrifugation and sterile filtration. Conjugates thus prepared were stored refrigerated (e.g., at 5-10ºC) or quick frozen in liquid nitrogen and then stored at -70ºC until use). Specifically for succinimidyl-2'-Roridin A 3-dithiopropionate conjugation to antibody, 0.40 ml of
6.2 mM sodium borate buffer (0.5 M, pH 8.0) and 0.20 ml of 100 mg molecusol/ml H2O were added to 1.0 ml of 2.0 mg NR-LU-10/ml in PBS (i.e., 6.2 mM sodium
phosphate, 150 mM NaCl, pH 7.2). 0.50 ml of DMSO having 290 micrograms of succinimidyl-2'-Roridin A 3-dithiopropionate dissolved therein (for a 30:1
trichothecene: antibody molar offering ratio) was added dropwise and with stirring over a 10 second time period. To purify the resultant product, 1 ml
aliquots of reaction solution were applied to PD-10 Sephadex columns (Pharmacia, Uppsala, Sweden)
equilibrated in PBS containing 1% molecusol wt/vol. The eluted conjugate was collected in the 2.4-4.8 ml fraction. The PD-10 purified aliquots were then pooled, exhaustively dialyzed against PBS containing 1% molecusol (wt/vol) using Spectra/POR molecular porous membrane tubing (Spectrum Medical Industries, Inc., Los Angeles, California), concentrated on a
Centricon PM-30 Microconcentrator (Amicon Div., W.R. Grace & Co., Beverly, Massachusetts), and sterile filtered using a 2.2 micron Acrodisc (Gelman Sciences, Ann Arbor, Michigan). Conjugates thus prepared were stored refrigerated (e.g., at 5-10ºC) or quick frozen in liquid nitrogen and then stored at -70ºC until use).
B. Antibody Conjugation of 2-Pyridinyl-Dithio-Derivatized Trichothecenes. In conjugation of 2-pyridinyl-dithio-derivatized trichothecenes, NR-LU-10 disulfides were first reduced by incubating NR-LU-10 at 5-10 mg in 1 ml of PBS containing 10-15 mM
dithiothreitol for 15 minutes at room temperature. The reduced product was purified over a PD-10 column that had been equilibrated in degassed 0.1 M sodium phosphate, pH 7.5, containing 0.1 M NaCl, 1% molecusol wt/vol and.0.2 mM EDTA (Na salt). Immediately after reduced-disulfide NR-LU-10 purification, .dropwise addition of 2-pyridinyl-dithio-derivatized
trichothecene dissolved in DMSO (0.40 ml of DMSO, containing 432 micrograms of 3-(2-pyridinyldithio) propionate hydrazide 2'-Roridin A hydrazone or
containing 337 micrograms of 3-(2-pyridinyldithio) propionate hydrazide 3-anguidine hydrazone or
containing 459 micrograms of 3-(2-pyridinyldithio) propionate hydrazide 2'-acetyl-13'-Roridin A
hydrazone) to 2 mg of the reduced disulfide NR-LU-10 solution (a 44:1 trichothecene: antibody molar offering ratio) was conducted with stirring. Generally in conducting the conjugation, the reaction concentration of antibody was 0.8 -1.2 mg/ml, DMSO was 16-25% vol/vol, and trichothecene to antibody molar offering ranged from 40:1 to 50:1 (in order to achieve a loading of approximately 6 trichothecenes per
antibody). After 1 hour of incubation at room
temperature, purification proceeded analogously to the purification scheme described in Example XVII (A) above. Conjugates thus prepared were stored
refrigerated (e.g., at 5-10ºC) or quick frozen in liquid nitrogen and then stored at -70ºC until use). C. Trichothecene-Polymer-Ligand Conjugation.
Experimentation involving the use of a biotin-dextran-trichothecene conjugate in a pretargeting approach included:
- trace labeling using an 1-125 PIP NHS ester of an available lysine of a 70,000 dalton dextran
molecule that had been biotinylated in accordance with techniques discussed herein in Example XVII, yielding radiotagged dextran biotin (represented as dextran*-biotin);
- trichothecene drug conjugation to the remaining lysines using an NHS activated trichothecene; - demonstration of binding of the drug-dextran*-biotin molecule to immobilized avidin; and
- assessment of serum clearance in mice.
Biotinylated dextran having a molecular weight of 70,000 daltons, with 18 moles of biotin covalently bound thereto and 18 additional lysine epsilon amino groups, was purchased from Sigma Chemical Co. (St. Louis, Missouri). To radiolabel the material, 4 mC of 1-125 PIP NHS ester of specific activity of 2200 mCi/mmole (New England Nuclear, Boston, Massachusetts) in acetonitrile in a 2 ml glass vial was blown down to dryness in a nitrogen stream. 10 mg of biotinylated, lysine-derivatized dextran lyophilizate, reconstituted with 0.650 ml of 1.0M sodium borate, pH 9.0, was added to the iodinating compound and incubated at room temperature for 10 minutes. Following reaction, the radioiodinated dextran moiety was purified by size exclusion chromatography using a PD-10 column
(Pharmacia, Uppsala, Sweden) equilibrated in phosphate buffered saline (i.e., 6.2M sodium phosphate, 150 mM NaCl, pH.7.2) containing 1% molecusol (Pharmatec, Alachua, Florida). The biotin-dextran* eluted from the column in the 2.4-4.8 ml fractions at a specific activity of 0.2 mCi/mg and a concentration of 3.5 mg/ml.
To derivatize the biotin-dextran* with
trichothecene, 0.9 ml of biotin-dextran* was diluted with 1.3 ml of sodium borate buffer, 0.3M, Ph 8.5, containing 1% molecusol followed by addition, with stirring of 1.2 ml of DMSO containing 1.2 mg of 2'- Desoxy-2'-alpha-(N-hydroxysuccinimidyl-3-dithiopropanoic acid) -Roridin A (i.e., 2:1 drug to available lysine molar ratio). After incubation for 1.5 hours at room temperature, each 1 ml aliquot of reaction mixture was purified as noted above with a PD-10 column equilibrated in PBS. Yields exceeded 85%. To establish that the biotin-dextran*-trichothecene molecule was able to bind to avidin or streptavidin, 1 microgram of biotin-dextran* and 1 microgram of biotin-dextran*-trichothecehe were incubated for 15 minutes at room temperature with 1 unit of avidin insolubilized on agarose beads (Sigma Chemical Co., St. Louis, Missouri) in 0.2 ml of 0.2 M Pi buffer, pH 6.3 containing 150 mM NaCl. Following this incubation, the percent radioactivity bound to the agarose beads was assessed after dilution with 1.4 ml buffer, centrifugation of the agarose suspension and three washings of the pellets with 1.4 ml buffer. 100% binding was observed for both biotin-dextran* and biotin-dextran*-trichothecene.
Serum clearance studies of biotin-dextran* and biotin-dextran*-trichothecene were also performed in Balb C mice. Serial blood samplings revealed that he two molecules exhibited substantially similar serum clearance upon injection of 2 μCi thereof.
EXAMPLE XXI
Synthesis of DOTA-Biotin Conjugates
A. Synthesis of Nitro-Benzyl-DOTA.
The synthesis of aminobenzyl-DOTA was conducted substantially in accordance with the procedure of McMurry et al., Bioconjugate Chem., 3: 108-117, 1992. The critical step in the prior art synthesis is the intermolecular cyclization between disuccinimidyl N-(tert-butoxycarbonyl) iminodiacetate and N-(2-aminoethyl)-4-nitrophenyl alaninamide to prepare 1- (tert-butoxycarbonyl)-5-(4-nitrobenzyl)-3,6,11-trioxo-1,4,7,10-tetraazacyclododecane. In other words, the critical step is the intermolecular cyclization between the bis-NHS ester and the diamine to give the cyclized dodecane. McMurry et al. conducted the cyclization step on a 30 mmol scale, dissolving each of the reagents in 100 ml DMF and adding via a syringe pump over 48 hours to a reaction pot containing 4 liters dioxane.
A 5x scale-up of the McMurry et al . procedure was not practical in terms of reaction volume, addition rate and reaction time. Process chemistry studies revealed that the reaction addition rate could be substantially increased and that the solvent volume could be greatly reduced, while still obtaining a similar yield of the desired cyclization product.
Consequently on a 30 mmol scale, each of the reagents was dissolved in 500 ml DMF and added via addition funnel over 27 hours to a reaction pot containing 3 liters dioxane. The addition rate of the method employed involved a 5.18 mmol/hour addition rate and a 0.047 M reaction concentration.
B. Synthesis of a D-alanine-linked conjugate with a preserved biotin carboxy moiety. A reaction scheme to form a compound of the following formula is
discussed below.
Figure imgf000152_0001
The D-alanine-linked conjugate was prepared by first coupling D-alanine (Sigma Chemical Co.) to biotin-NHS ester. The resultant biotinyl-D-alanine was then activated with 1-(3-dimethylaminopropyl)-3-ethyl-carbodiimide hydrochloride (EDCI) and N-hydroxysuccinimide (NHS). This NHS ester was reacted in situ with DOTA-aniline to give the desired product which was purified by preparative HPLC.
More specifically, a mixture of D-alanine (78 mg, 0.88 mmol, 1.2 equivalents), biotin-NHS ester (250 mg, 0.73 mmol, 1.0 equivalent), triethylamine (0.30 ml, 2.19 mmol, 3.0 equivalents) in DMF (4 ml) was heated at 110ºC for 30 minutes. The solution was cooled to 23ºC and evaporated. The product solid was acidified with glacial acetic acid and evaporated again. The product biotinyl-D-alanine, a white solid, was
suspended in 40 ml of water to remove excess unreacted D-alanine, and collected by filtration. Biotinyl-D-alanine was obtained as a white solid (130 mg, 0.41 mmol) in 47% yield.
NHS (10 mg, 0.08 mmol) and EDCI (15 mg, 0.07 mmol) were added to a solution of biotinyl-D-alanine (27 mg, 0.08 mmol) in DMF (1 ml). The solution was stirred at 23ºC for 60 hours, at which time TLC analysis
indicated conversion of the carboxyl group to the N-hydroxy succinimidyl ester. Pyridine (0.8 ml) was added followed by DOTA-aniline (20 mg, 0.04 mmol).
The mixture was heated momentarily at approximately 100 ºC, then cooled to 23ºC and evaporated. The product, DOTA-aniline-D-alanyl-biotinamide was
purified by preparative HPLC.
C. Synthesis of N-hydroxyethyl-linked conjugate. Iminodiacetic acid dimethyl ester is condensed with biotin-NHS-ester to give biotinyl dimethyl iminodiacetate. Hydrolysis with one equivalent of sodium hydroxide provides the monomethyl ester after purification from under and over hydrolysis products. Reduction of the carboxyl group with borane provides the hydroxyethyl amide. The hydroxyl group is
protected with t-butyl-dimethyl-silylchloride. The methyl ester is hydrolysed, activated with EDCI and condensed with DOTA-aniline to form the final product conjugate. D. Synthesis of N-Me-LC-DOTA-biotin. A reaction scheme is shown below.
Figure imgf000154_0001
Esterification of 6-Aminocaproic acid (Sigma
Chemical Co.) was carried out with methanolic HCI. Trifluoroacetylation of the amino group using
trifluoroacetic anhydride gave N-6-(methylcaproyl)- trifluoroacetamide. The amide nitrogen was methylated using sodium hydride and iodomethane in
tetrahydrofuran. The trifluoroacetyl protecting group was cleaved in acidic methanol to give methyl 6- methylamino-caproate hydrochloride. The amine was condensed with biotin-NHS ester to give methyl N- methyl-caproylamido-biotin. Saponification afforded the corresponding acid which was activated with EDCI and NHS and, in situ, condensed with DOTA-aniline to give DOTA-benzylamido-N-methyl-caproylamido-biotin.
1. Preparation of methyl 6-aminocaproate
hydrochloride. Hydrogen chloride (gas) was added to a solution of 20.0 g (152 mmol) of 6-aminocaproic acid in 250 ml of methanol via rapid bubbling for 2-3 minutes. The mixture was stirred at 15-25ºC for 3 hours and then concentrated to afford 27.5 g of the product as a white solid (99%):
H-NMR (DMSO) 9.35 (1 H, broad t), 3.57 (3H, s), 3.14 (2H, quartet), 2.28 (2H, t), 1.48 (4H, multiplet), and 1.23 ppm (2H, multiplet).
2. Preparation of N-6-(methylcaproyl)-trifluoroacetamide. To a solution of 20.0 g (110 mmol) of methyl 6-aminocaproate hydrochloride in 250 ml of dichloromethane was added 31.0 ml (22.2 mmol) of triethylamine. The mixture was cooled in an ice bath and trifluoroacetic anhydride (18.0 ml, 127 mmol) was added over a period of 15-20 minutes. The mixture was stirred at 0-10ºC for 1 hour and concentrated. The residue was diluted with 300 ml of ethyl acetate and saturated aqueous sodium bicarbonate (3 × 100 ml). The organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated to afford 26.5 g of the product as a pale yellow oil (100%):
H-NMR (DMSO) 3.57 (3H, s), 3.37 (2H, t), 3.08 (1.9H, quartet, N-CH3), 2.93 (1.1H, s, N-CH3), 2.30 (2H, t), 1.52 (4H, multiplet), and 1.23 ppm (2H, multiplet).
3. Preparation of methyl 6-N-methylamino-caproate hydrochloride. To a solution of 7.01 g (29.2 mmol) of N-6-(methylcaproyl)-trifluoroacetamide in 125 ml of anhydrous tetrahydrofuran was slowly added 1.75 g of 60% sodium hydride (43.8 mmol) in mineral oil. The mixture was stirred at 15-25ºC for 30 minutes and then 6.2 g (43.7 mmol) of iodomethane was added. The mixture was stirred at 15-25ºC for 17 hours and then filtered through celite. The solids were rinsed with 50 ml of tetrahydrofuran. The filtrates were combined and concentrated. The residue was diluted with 150 ml of ethyl acetate and washed first with 5% aqueous sodium sulfite (2 × 100 ml) and then with 100 ml of l N aqueous hydrochloric acid. The organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated to afford a yellow oily residue. The residue was diluted with 250 ml of methanol and then hydrogen chloride (gas) was rapidly bubbled into the mixture for 2-3 minutes. The resultant mixture was refluxed for 18 hours, cooled and concentrated. The residue was diluted with 150 ml of methanol and washed with hexane (3 × 150 ml) to remove mineral oil
previously introduced with NaH. The methanol phase was concentrated to afford 4.91 g of the product as a yellow oil (86%):
H-NMR (DMSO) 8.80 (2H, broad s), 3.58 (3H, s), 2.81 (2H, multiplet), 2.48 (3H, s), 2.30 (2H, t), 1.52 (4H, multiplet), and 1.29 ppm (2H, multiplet).
4. Preparation of methyl 6-(N-methylcaproylamidobiotin. N-hydroxysuccinimidyl biotin (398 mg, 1.16 mmol) was added to a solution of methyl 6-(N-methyl) aminocaproate hydrochloride (250 mg, 1.28 mmol) in DMF (4.0 ml) and triethylamine (0.18 ml, 1.28 mmol). The mixture was heated in an oil bath at 100ºC for 10 minutes. The solution was evaporated, acidified with glacial acetic acid and evaporated again. The residue was chromatographed on a 25 mm flash chromatography column manufactured by Ace Glass packed with 50 g silica (EM Science, Gibbstown, New Jersey, particle size 0.40-0.63 mm) eluting with 15% MeOH/EtOAc. The product was obtained as a yellow oil (390 mg) in 79% yield.
5. Preparation of 6-(N-methyl-N-biotinyl) amino caproic acid. To a solution of methyl 6-(N-methylcaproylamido-biotin (391 mg, 1.10 mmol) in methanol (2.5 ml) was added a 0.95 N NaOH solution (1.5 ml). This solution was stirred at 23ºC for 3 hours. The solution was neutralized by the addition of 1.0 M HCI (1.6 ml) and evaporated. The residue was dissolved in water, further acidified with 1.0 M HCI (0.4 ml) and evaporated. The gummy solid residue was suspended in water and agitated with a spatula until it changed into a white powder. The powder was collected by filtration with a yield of 340 mg.
6. Preparation of DOTA-benzylamido-N-methyl-caproylamido-biotin. A suspension of 6-(N-methyl-N-biotinyl) amino caproic acid (29 mg, 0.08 mmol) and N-hydroxysuccinimide (10 mg, 0.09 mmol) in DMF (0.8 ml) was heated over a heat gun for the short time
necessary for the solids to dissolve. To this heated solution was added EDCI (15 mg, 0.08 mmol). The resultant solution was stirred at 23°C for 20 hours. To this stirred solution were added aminobenzyl-DOTA
(20 mg, 0.04 mmol) and pyridine (0.8 ml). The mixture was heated over a heat gun for 1 minute. The product was isolated by preparative HPLC, yielding 3 mg. E. Synthesis of a bis-DOTA conjugate with a preserved biotin carboxy group. A reaction scheme is shown below.
„ l u
y/
Figure imgf000157_0001
Figure imgf000158_0001
1. Preparation of methyl 6-bromocaproate (methyl 6-bromohexanoate). Hydrogen chloride (gas) was added to a solution of 5.01 g (25.7 mmol) of 6-bromocaproic acid in 250 ml of methanol via vigorous bubbling for 2-3 minutes. The mixture was stirred at 15-25°C for 3 hours and then concentrated to afford 4.84 g of the product as a yellow oil (90%):
H-NMR (DMSO) 3.58 (3H, s), 3.51 (2H, t), 2.29 (2H, t), 1.78 (2H, pentet), and 1.62-1.27 ppm
(4H, m).
2. Preparation of N,N-bis-(methyl 6-hexanoyl)-amine hydrochloride. To a solution of 4.01 g (16.7 mmol) of N-(methyl 6-hexanoyl)-trifluoroacetamide (prepared in accordance with section D.2. herein) in 125 ml of anhydrous tetrahydrofuran was added 1.0 g (25 mmol) of 60% sodium hydride in mineral oil. The mixture was stirred at 15-25°C for 1 hour and then 3.50 g (16.7 mmol) of methyl 6-bromocaproate was added and the mixture heated to reflux. The mixture was stirred at reflux for 22 hours. NMR assay of an aliquot indicated the reaction to be incomplete.
Consequently, an additional 1.00 g (4.8 mmol) of methyl 6-bromocaproate was added and the mixture stirred at reflux for 26 hours. MNR assay of an aliquot indicated the reaction to be incomplete. An additional 1.0 g of methyl 6-bromocaproate was added and the mixture stirred at reflux for 24.hours. NMR assay of an aliquot indicated the reaction to be near complete. The mixture was cooled and then directly filtered through celite. The solids were rinsed with 100 ml of tetrahydrofuran. The filtrates were
combined and concentrated. The residue was diluted with 100 ml of methanol and washed with hexane (3x 100 ml) to remove the mineral oil introduced with the sodium hydride. The methanol phase was treated with 6 ml of 10 N aqueous sodium hydroxide and stirred at 15-25ºC for 3 hours. The mixture was concentrated. The residue was diluted with 100 ml of deionized water and acidified to pH 2 with concentrated HCI. The mixture was washed with ether (3 × 100 ml). The aqueous phase was concentrated, diluted with 200 ml of dry methanol and then hydrogen chloride gas was bubbled through the mixture for 2-3 minutes. The mixture was stirred at 15-25ºC for 3 hours and then concentrated. The residue was diluted with 50 ml of dry methanol and filtered to remove inorganic salts. The filtrate was concentrated to afford 1.98 g of the product as a white solid (38%) :
H-NMR (DMSO) 8.62 (2H, m) 3.58 (6H, s), 2.82 (4H, m) 2.30 (4H, t), 1.67-1.45 (8H, m) and
1.38-1.22 ppm (4H, m).
3. Preparation of N,N=bis-(methyl 6-hexanoyl)-biotinamide. To a solution of 500 mg (1.46 mmol) of
N-hydroxysuccinimidyl. biotin in 15 ml of dry dimethyl-formamide was added 600 mg (1.94 mmol) of N,N-bis-(methyl 6-hexanoyl) amine hydrochloride followed by 1.0 ml of triethylamine. The mixture was stirred at 80-85ºCC for 3 hours and then cooled and concentrated.
The residue was chromatographed on silica gel, eluting with 20% methanol/ethyl acetate, to afford 620 mg of the product as a near colorless oil (85%) :
H-NMR (CDCl3) 5.71 (1H, s), 5.22 (1H, s), 4.52 (1H,. m), 4.33 (1H, m), 3.60 (3H, s), 3.58 (3H, s), 3.34-3.13 (5H, m), 2.92 (1H, dd), 2.75 (1H, d), 2.33 (6H, m) and 1.82-1.22 ppm (18H, m);
TLC-Rf 0.39 (20:80 methanol/ ethyl acetate).
4. Preparation of N,N-bis-(6-hexanoyl)-biotinamide. To a solution of 610 mg (0.819 mmol) of N,N-bis-(methyl 6-hexanoyl)-biotinamide in 35 ml of methanol was added 5.0 ml of IN aqueous sodium
hydroxide. The mixture was stirred at 15-25ºC for 4.5 hours and then concentrated. The residue was diluted with 50 ml of deionized water acidified to pH 2 with IN aqueous hydrochloric acid at 4ºC. The product, which precipitated out as a white solid, was isolated by vacuum filtration and dried under vacuum to afford 482 mg (84%) :
H-NMR (DMSO) 6.42 (1H, S), 6.33 (1H, s), 4.29 (1H, m), 4.12 (1H, m), 3.29-3.04 (5H, m), 2.82
(1H, dd), 2.57 (1H, d), 2.21 (6H, m) and 1.70- 1.10 ppm (18H, m).
5. Preparation of N',N'-bis-(N-hydroxy-succinimidyl 6-hexanoyl)-biotinamide. To a solution of 220 mg (0.467 mmol) of N,N-bis-(6-hexanoyl)-biotinamide in 3 ml of dry dimethylformamide was added 160 mg (1.39 mmol) of N-hydroxysuccinimide followed by 210 mg (1.02 mmol) of dicyclohexyl-carbodiimide. The mixture was stirred at 15-25ºC for 17 hours and then concentrated. The residue was chromatographed on silica gel, eluting with 0.1:20:80 acetic
acid/methanol/ethyl acetate, to afford 148 mg of the product as a foamy off-white solid (48%):
H-NMR (DMSO) 6.39 (1H, s), 6.32 (1H, s), 4,29 (1H, m), 4,12 (1H, m), 3.30-3.03 (5H, m), 2.81
(9H, dd and s), 2.67 (4H, m), 2.57 (1H, d),
2.25 (2H, t), 1.75-1.20 (18H, m); TLC-Rf 0.37 (0.1:20:80 acetic acid/methanol/ethyl acetate).
6. Preparation of N,N-bis-(6-hexanoylamidobenzyl-DOTA) -biotinamide. To a mixture of 15 mg of DOTA-benzylamine and 6.0 mg of N',N'-bis-(N-hydroxy-succinimidyl 6-hexanoyl)-biotinamide in 1.0 ml of dry dimethylformamide was added 0.5 ml of dry pyridine. The mixture was stirred at 45-50°C for 4.5 hours and at 15-25°C for 12 hours. The mixture was concentrated and the residue chromatographed on a 2.1 × 2.5 cm octadecylsilyl (ODS) reverse-phase preparative HPLC column eluting with a - -20 minute gradient profile of 0.1:95:5 to 0.1:40:60 trifluoroacetic
acid : water : acetonitrile at 13 ml/minute to afford the desired product. The retention time was 15.97 minutes using the aforementioned gradient at a flow rate of 1.0 ml/minute on a 4.6 mm × 25 cm ODS analytical HPLC column.
F. Synthesis of an N-methyl-glycine linked conjugate. A reaction scheme for this synthesis is shown below .
Figure imgf000161_0001
The N-methyl glycine-linked DOTA-biotin conjugate was prepared by an analogous method to that used to prepare D-alanine-linked DOTA-biotin conjugates. N-methyl-glycine (trivial name sarcosine, available from Sigma Chemical Co.) was condensed with biotin-NHS ester in DMF and triethylamine to obtain N-methyl glycyl-biotin. N-methyl-glycyl biotin was then activated with EDCI and NHS. The resultant NHS ester was not isolated and was condensed in situ with DOTA-aniline and excess pyridine. The reaction solution was heated at 60ºC for 10 minutes and then evaporated. The residue was purified by preparative HPLC to give [(N-methyl-N-biotinyl)-N-glycyl]-aminobenzyl-DOTA.
1. Preparation of (N-methyl) glycyl biotin. DMF (8.0 ml) and triethylamine (0.61 ml, 4.35 mmol) were added to solids N-methyl glycine (182 mg, 2.05 mmol) and N-hydroxy-succinimidyl biotin (500 mg, 1.46 mmol). The mixture was heated for 1 hour in an oil bath at
85ºC during which time the solids dissolved producing a clear and colorless solution. The solvents were then evaporated. The yellow oil residue was acidified with glacial acetic acid, evaporated and
chromatographed on a 27 mm column packed with 50 g silica, eluting with 30% MeOH/EtOAc 1% HOAc to give the product as a white solid (383 mg) in 66% yield.
H-NMR (DMSO) : 1.18-1.25 (m, 6H, (CH2)3), 2.15, 2.35 (2 t's, 2H, CH2CO) , 2.75 (m, 2H, SCH2),
2.80, 3.00 (2 s's, 3H, NCH3) , 3.05-3.15 (m, IH,
SCH), 3.95, 4.05 (2 s's, 2H, CH2N), 4.15, 4.32 (2 m's, 2H, 2CHN's), 6.35 (s, NH), 6.45 (s,
NH).
2. Preparation of [(N-methyl-N-biotinyl)glycyl] aminobenzyl-DOTA. N-hydroxysuccinimide (10 mg, 0.08 mmol) and EDCI (15 mg, 6.08 mmol) were added to a solution of (N-methylglycyl biotin (24 mg, 0.08 mmol) in DMF (1.0 ml). The solution was stirred at 23 'C for 64 hours. Pyridine (0.8 ml) and aminobenzyl-DOTA (20mg, 0.04 mmol) were added. The mixture was heated in an oil bath at 63ºC for 10 minutes, then stirred at 23ºC for 4 hours. The solution was evaporated. The residue was purified by preparative HPLC to give the product as an off white solid (8 mg, 0.01 mmol) in 27% yield.
H-NMR (D2O) : 1.30-1.80 (m, 6H), 2.40, 2.55 (2 t's, 2H, CH2CO), 2.70-4.2 (complex multiplet), 4.35 (m, CHN), 4.55 (m, CHN), 7.30 (m, 2H, benzene hydrogens), 7.40 (m, 2H, benzene hydrogens).
G. Synthesis of a short chain amine-linked conjugate with a reduced biotin carboxy group. A two- part reaction scheme is shown below.
Figure imgf000163_0001
The biotin carboxyl group is reduced with diborane in THF to give a primary alcohol. Tosylation of the alcohol with tosyl chloride in pyridine affords the primary tosylate. Aminobenzyl DOTA is acylated with trifluoroacetic anhydride in pyridine to give (N- trifluoroacetyl) aminobenzyl-DOTA. Deprotonation with 5.0 equivalents of sodium hydride followed by
displacement of the biotin tosylate provides the (N-trifluoracetamido-N-descarboxylbiotinyl)aminobenzyl-DOTA. Acidic cleavage of the N-trifluoroacetamide group with HCl(g) in methanol provides the amine-linked DOTA-biotin conjugate.
EXAMPLE XXII Clearing Agent Evaluation Experimentation
The following experiments conducted on non-tumor-bearing mice were conducted using female BALB/c mice (20-25 g). For tumor-bearing mice experimentation, female nude mice were injected subcutaneously with LS-180 tumor cells, and, after 7 d, the mice displayed 50-100 mg tumor xenografts. The monoclonal antibody used in these experiments was NR-LU-10. When
radiolabeled, the NR-LU-10-streptavidin conjugate was radiolabeled with I-125 using procedures described herein. When radiolabeled, PIP-biocytin was labeled with I-131 or I-125 using procedures described herein.
A. Utility of Asialoorosomucoid-Biotin (AO-Bt) in
Reducing Circulating Radioactivity from a Subseguently
Administered Radiolabeled Biotin Ligand. Mice bearing LS-180 colon tumor xenografts were injected with 200 micrograms NR-LU-10 antibody-streptavidin (MAb-StrAv) conjugate at time 0, which was allowed to prelocalize to tumor for 22 hours. At that time, 20 micrograms of AO-Bt was administered to one group of animals. Two hours later, 90 micrograms of a radioisotope-bearing, ligand-containing small molecule (PIP-biotin-dextran prepared as discussed in part B hereof) was
administered to this group of mice and also to a group which had not received AO-Bt. The results of this experiment with respect to radiolabel uptake in tumor and clearance from the blood indicated that tumor-targeting of the radiolabeled biotin-containing conjugate was retained while blood clearance was enhanced, leading to an overall improvement in amount delivered to target/amount located in serum. The AUC tumor/AUC blood with clearing agent was 6.87, while AUC tumor/AUC blood without clearing agent was 4.45.
Blood clearance of the circulating MAb-StrAv conjugate was enhanced with the use of clearing agent. The clearing agent was radiolabeled in a separate group of animals and found to bind directly to tumor at very low levels (1.7 pmol/g at a dose of 488 total pmoles
(0.35%ID/g), indicating that it does not significantly compromise the ability of tumor-bound MAb-StrAv to bind subsequently administered radiolabeled ligand.
B. Preparation Protocol for PIP-Biotin-Dextran. A solution of 3.0 mg biotin-dextran, lysine fixable
(BDLF, available from Sigma Chemical Co., St. Louis, Missouri, 70,000 dalton molecular weight with
approximately 18 biotins/molecule) in 0.3 ml PBS and 0.15 ml 1 M sodium carbonate, pH 9.25, was added to a dried residue (1.87 mCi) of N-succinimidyl p-I-125-iodobenzoate prepared in accordance with Wilbur, et al., J. Nucl. Med., 30: 216-226, 1989.
C. Dosing Optimization of AO-Bt. Tumored mice receiving StrAv-MAb as above, were injected with increasing doses of AO-Bt (0 micrograms, 20
micrograms, 50 micrograms, 100 micrograms and 200 micrograms). Tumor uptake of I-131-PIP-biocytin (5.7 micrograms, administered 2 hours after AO-Bt
administration) was examined. Increasing doses of AO-Bt had no effect on tumor localization of MAb-StrAv. Data obtained 44 hours after AO-Bt administration showed the same lack of effect. This data indicates that AO-Bt dose not cross-link and internalize MAb-StrAv on the tumor surface, as had been noted for avidin administered following biotinylated antibody.
PIP-biocytin tumor localization was inhibited at higher doses of AO-Bt. This effect is most likely due to reprocessing and distribution to tumor of biotin used to derivatize AO-Bt. Optimal tumor to blood ratios (% injected dose of radiolabeled ligand/gram weight of tumor divided by % injected dose of
radioligand/gram weight of blood were achieved at the 50 microgram dose of AO-Bt. Biodistributions
conducted following completion of the protocols employing a 50 microgram AO-Bt dose revealed low retention of radiolabel in all non-target tissues (1.2 pmol/g in blood; 3.5 pmol/gram in tail; 1.0 pmol/g in lung; 2.2 pmol/g in liver; 1.0 pmol/g is spleen; 7.0 pmol/g in stomach; 2.7 pmol/g in kidney; and 7.7 pmol/g in intestine). With 99.3 pmol/g in tumor, these results indicate effective decoupling of the PIP-biocytin biodistribution from that of the MAb-StrAv at all sites except tumor. This decoupling occurred at all clearing agent doses in excess of 50 micrograms as well. Decreases in tumor localization of PIP-biocytin was the significant result of
administering clearing agent doses in excess of 50 micrograms. In addition, the amount of PIP-biocytin in non-target tissues 44 hours after administration was identical to localization resulting from
administration of PIP-biocytin alone (except for tumor, where negligible accretion was seen when PIP- biocytin was administered alone), indicating effective decoupling.
D. Further Investigation of Optimal Clearing Agent Dose. Tumored mice injected with MAb-StrAv at time 0 as above; 50 micrograms of AO-Bt at time 22 hours; and 545 microcuries of I-131-PIP-biocytin at time 25 hours. Whole body radiation was measured and compared to that of animals that had not received clearing agent. 50 micrograms of AO-Bt was efficient in allowing the injected radioactivity to clear from the animals unimpeded by binding to circulating MAb-StrAv conjugate. Tumor uptake of I-131-PIP-biocytin was preserved at the 50 microgram clearing agent dose, with AUC tumor/AUC blood of 30:1 which is
approximately 15-fold better than the AUC tumor/AUC blood achieved in conventional antibody-radioisotope therapy using this model.
E. Galactose- and Biotin-Derivatization of Human Serum Albumin (HSA). HSA was evaluated because it exhibits the advantages of being both inexpensive and non-immunogenic. HSA was derivatized with varying levels of biotin (1-about 9 biotins/molecule) via analogous chemistry to that previously described with respect to AO. More specifically, to a solution of HSA available from Sigma Chemical Co. (5-10 mg/ml in PBS) was added 10% v/v 0.5 M sodium borate buffer, pH 8.5, followed by dropwise addition of a DMSO solution of NHS-LC-biotin (Sigma Chemical Co.) to the stirred solution at the desired molar offering (relative molar equivalents of reactants). The final percent DMSO in the reaction mixture should not exceed 5%. After stirring for 1 hour at room temperature, the reaction was complete. A 90% incorporation efficiency for biotin on HSA was generally observed. As a result, if 3 molar equivalences of the NHS ester of LC-biotin was introduced, about 2.7 biotins per HSA molecule were obtained. Unreacted biotin reagent was removed from the biotin-derivatized HSA using G-25 size exclusion chromatography. Alternatively, the crude material may be directly galactosylated. The same chemistry is applicable for biotinylating non-previously
biotinylated dextran.
HSA-biotin was then derivatized with from 12 to 15 galactoses/molecule. Galactose derivatization- of the biotinylated HSA was performed according to the procedure of Lee, et al., Biochemiεtry, 15: 3956, 1976. More specifically, a 0.1 M methanolic solution of cyanomethyl-2,3,4,6-tetra-0-acetyl-1-thio-D-galactopyranoside was prepared and reacted with a 10% v/v 0.1 M NaOMe in methanol for 12 hours to generate the reactive galactosyl thioimidate. The
galactosylation of biotinylated HSA began by initial evaporation of the anhydrous methanol from a 300 fold molar excess of reactive thioimidate. Biotinylated
HSA in PBS, buffered with 10% v/v 0.5 M sodium borate, was added to the oily residue. After stirring at room temperature for 2 hours, the mixture was stored at 4ºC for 12 hours. The galactosylated HSA-biotin was then purified by G-25 size exclusion chromatography or by buffer exchange to yield the desired product. The same chemistry is exploitable to galactosylating dextran. The incorporation efficiency of galactose on HSA is approximately 10%.
70 micrograms of Galactose-HSA-Biotin (G-HSA-B), with 12-15 galactose residues and 9 biotins, was administered to mice which had been administered 200 micrograms of StrAv-MAb or 200 microliters of PBS 24 hours earlier. Results indicated that G-HSA-B is effective in removing StrAv-MAb from circulation.
Also, the pharmacokinetics of G-HSA-B is unperturbed and rapid in the presence or absence of circulating MAb-StrAv.
F. Non-Protein Clearing Agent. A commercially available form of dextran, molecular weight of 70,000 daltons, pre-derivatized with approximately 18
biotins/molecule and having an equivalent number of free primary amines was studied. The primary amine moieties were derivatized with a galactosylating reagent, substantially in accordance with the
procedure therefor described above in the discussion of HSA-based clearing agents, at a level of about 9 galactoses/molecule. The molar equivalence offering ratio of galactose to HSA was about 300:1, with about one-third of the galactose being converted to active form. 40 Micrograms of galactose-dextran-biotin (GAL-DEX-BT) was then injected i.v. into one group of mice which had received 200 micrograms MAb-StrAv conjugate intravenously 24 hours earlier, while 80 micrograms of GAL-DEX-BT was injected into other such mice. GAL-DEX-BT was rapid and efficient at clearing StrAv-MAb conjugate, removing over 66% of circulating conjugate in less than 4 hours after clearing agent
administration. An equivalent effect was seen at both clearing agent doses, which correspond to 1.6 (40 micrograms) and 3.2 (80 micrograms) times the
stoichiometric amount of circulating StrAv conjugate present.
G. Dose Ranging for G-HSA-B Clearing Agent. Dose ranging studies followed the following basic format:
200 micrograms MAb-StrAv conjugate administered; 24 hours later, clearing agent administered; and 2 hours later, 5.7 micrograms PIP-biocytin
administered.
Dose ranging studies were performed with the G-HSA-B clearing agent, starting with a loading of 9 biotins per molecule and 12-15 galactose residues per molecule. Doses of 20, 40, 70 and 120 micrograms were administered 24 hours after a 200 microgram dose of MAb-StrAv conjugate. The clearing agent
administrations were followed 2 hours later by
administration of 5.7 micrograms of I-131-PIP- biocytin. Tumor uptake and blood retention of PIP-biocytin was examined 44 hours after administration thereof (46 hours after clearing agent
administration). The results showed that a nadir in blood retention of PIP-biocytin was achieved by all doses greater than or equal to 40 micrograms of G-HSA-B. A clear, dose-dependent decrease in tumor binding of PIP-biocytin at each increasing dose of G-HSA-B was present, however. Since no dose-dependent effect on the localization of MAb-StrAv conjugate at the tumor was observed, this data was interpreted as being indicative of relatively higher blocking of tumor- associated MAb-StrAv conjugate by the release of biotin from catabolized clearing agent. Similar results to those described earlier for the
asialoorosomucoid clearing agent regarding plots of tumor/blood ratio were found with respect to G-HSA-B, in that an optimal balance between blood clearance and tumor retention occurred around the 40 microgram dose.
Because of the relatively large molar amounts of biotin that could be released by this clearing agent at higher doses, studies were undertaken to evaluate the effect of lower levels of biotinylation on the effectiveness of the clearing agent. G-HSA-B,
derivatized with either 9, 5 or 2 biotins/molecule, was able to clear MAb-StrAv conjugate from blood at equal protein doses of clearing agent. All levels of biotinylation yielded effective, rapid clearance of MAb-StrAv from blood.
Comparison of these 9-, 5-, and 2-biotin- derivatized clearing agents with a single biotin G-HSA-B clearing agent was carried out in tumored mice, employing a 60 microgram dose of each clearing agent. This experiment showed each clearing agent to be substantially equally effective in blood clearance and tumor retention of MAb-StrAv conjugate 2 hours after clearing agent administration. The G-HSA-B with a single biotin was examined for the ability to reduce binding of a subsequently administered biotinylated small molecule (PIP-biocytin) in blood, while
preserving tumor binding of PIP-biocytin to
prelocalized MAb-StrAv conjugate. Measured at 44 hours following PIP-biocytin administration, tumor localization of both the MAb-StrAv conjugate and PIP- biocytin was well preserved over a broad dose range of G-HSA-B with one biotin/molecule (90 to 180
micrograms). A progressive decrease in blood
retention of PIP-biocytin was achieved by increasing doses of the single biotin G-HSA-B clearing agent, while tumor localization remained essentially
constant, indicating that this clearing agent, with a lower level of biotinylation, is preferred. This preference arises because the single biotin G-HSA-B clearing agent is both effective at clearing MAb-StrAv over a broader range of doses (potentially eliminating the need for patient-to-patient titration of optimal dose) and appears to release less competing biotin into the systemic circulation than the same agent having a higher biotin loading level.
Another way in which to decrease the effect of clearing agent-released biotin on active agent-biotin conjugate binding to prelocalized targeting moiety-streptavidin conjugate is to attach the protein or polymer or other primary clearing agent component to biotin using a retention linker. A retention linker has a chemical structure that is resistant to agents that cleave peptide bonds and, optionally, becomes protonated when localized to a catabolizing space, such as a lysosome . Preferred retention linkers of the present invention are short strings of D-amino acids or small molecules having both of the
characteristics set forth above. An exemplary
retention linker of the present invention is cyanuric chloride, which may be interposed between an epsilon amino group of a lysine of a proteinaceous primary clearing agent component and an amine moiety of a reduced and chemically altered biotin carboxy moiety (which has been discussed above) to form a compound of the structure set forth below.
Figure imgf000171_0001
When the compound shown above is catabolized in a catabolizing space, the heterocyclic ring becomes protonated. The ring protonation prevents the catabolite from exiting the lysosome. In this manner, biotin catabolites containing the heterocyclic ring are restricted to the site(s) of catabolism and, therefore, do not compete with activs-agent-biotin conjugate for prelocalized targeting moiety-streptavidin target sites.
Comparisons of tumor/blood localization of
radiolabeled PIP-biocytin observed in the G-HSA-B dose ranging studies showed that optimal tumor to
background targeting was achieved over a broad dose range (90 to 180 micrograms), with the results
providing the expectation that even larger clearing agent doses would also be effective. Another key result of the dose ranging experimentation is that G- HSA-B with an average of only 1 biotin per molecule is presumably only clearing the MAb-StrAv conjugate via the Ashwell receptor mechanism only, because too few biotins are present to cause cross-linking and
aggregation of MAb-StrAv conjugates and clearing agents with such aggregates being cleared by the reticuloendothelial system.
H. Tumor Targeting Evaluation Using G-HSA-B. The protocol for this experiment was as follows :
Time 0: administer 400 micrograms MAb-StrAv conjugate;
Time 24 hours: administer 240 micrograms of G-HSA-B with one biotin and 12-15 galactoses and
Time 26 hours : administer 6 micrograms of
Figure imgf000172_0001
Lu-177 is complexed with the DOTA chelate using known techniques therefor.
Efficient delivery of the Lu-177-DOTA-biotin small molecule was observed, 20-25 % injected dose/gram of tumor. These values are equivalent with the
efficiency of the delivery of the MAb-StrAv conjugate. The AUC tumor/AUC blood obtained for this non- optimized clearing agent dose was 300% greater than that achievable by comparable direct MAb-radiolabel administration. In addition, the HSA-based clearing agent is expected to exhibit a low degree of
immunogenicity in humans.
Kits containing one or more of the components described above are also contemplated. For instance, radiohalogenated biotin may be provided in a sterile container for use in pretargeting procedures. A chelate-biotin conjugate provided in a sterile
container is suitable for radiometallation by the consumer; such kits would be particularly amenable for use in pretargeting protocols. Alternatively, radiohalogenated biotin and a chelate-biotin conjugate may be vialed in a non-sterile condition for use as a research reagent.
From the foregoing, it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention.
Accordingly, the invention is not limited except as by the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A biotin-DOTA conjugate of the following formula:
Figure imgf000174_0006
wherein a linker L is selected from the group
comprising:
1) a D-amino acid-containing linker of the formula
Figure imgf000174_0005
;
2) a linker of the formula
Figure imgf000174_0004
; and
3) a linker of the formula
Figure imgf000174_0003
a linker of the formula , wherein L' is selected from
Figure imgf000174_0002
the group comprising:
a) -NH-CO- (CH2)n-O- ;
b) -NH- ; c) ;
Figure imgf000174_0001
d) -NH-CS-NH- ; and
e) -NH-CO- (CH2)n-NH- ,
wherein R1 is hydrogen, lower alkyl; lower alkyl substituted with one or more hydrophilic groups including (CH2)m-OH, (CH2)m-OSO3, (CH2)m-SO3, and
Figure imgf000175_0001
, where m is 1 or 2;
glucuronide-substituted amino acids; or other glucuronide derivatives;
R2 is hydrogen; lower alkyl; substituted lower alkyl having one or more substituents selected from the group comprising hydroxy, sulfate, and
phosphonate; or a hydrophilic moiety;
R3 is hydrogen; an amine; a lower alkyl; a hydroxy-, sulfate- or phosphonate-substituted lower alkyl; a glucuronide; or a glucuronide-derivatized amino acid;
R4 is hydrogen, lower alkyl or ;
Figure imgf000175_0002
is hydrogen; -(CH2)2-OH or a sulfate or
Figure imgf000175_0003
phosphonate derivative thereof; or ;
R'' is a bond or -(CH2)n-CO-NH- ; and
n ranges from 0-5.
2. A biotin-DOTA conjugate of Claim 1 wherein L is a D-amino acid-incorporating a linker of the formula
Figure imgf000175_0004
.
3. A biotin-DOTA conjugate of Claim 2 wherein R1 is CH3 and R2 is H.
4. A biotin-DOTA conjugate of Claim 1 wherein L is a linker of the formula
Figure imgf000175_0005
.
5. A biotin-DOTA conjugate of Claim 4 wherein R3 is hydrogen; R4 is CH3; and n is 4.
6. A biotin-DOTA conjugate of Claim 4 wherein R3 is hydrogen; R4 is CH3; and n is 0.
is
7. A biotin-DOTA conjugate of Claim 4 wherein R3 hydrogen; R4 is ; and n is
Figure imgf000176_0001
8. A biotin-DOTA conjugate of Claim 1 wherein L is a linker of the formula , wherein L' is selected from the
Figure imgf000176_0002
group comprising:
a) -NH-CO- (CH2)n-O- ;
b) -NH- ;
c)
Figure imgf000176_0003
;
d) -NH-CS-NH- ; and
e) -NH-CO- (CH2)n-NH- or the bis-DOTA derivative thereof.
9. A clearing agent useful in practicing
pretargeting methods, which clearing agent has the following structure:
(Hexose ) m- -Human Serum Albumin (HSA) - - (Ligand) n , wherein n is an integer from 1 to 10 and m is an
integer from 1 to 25 and wherein the hexose is
recognized by Ashwell receptors.
10. A clearing agent of Claim 9 wherein the
ligand is biotin and the hexose is galactose or
mannose.
11. A -clearing agent of Claim 10 wherein n is an integer from 1 to 5 and the hexose is galactose.
12. A clearing agent of Claim 10 wherein n is 3 and the hexose is galactose.
13. A clearing agent of Claim 10 wherein m is an integer from 10 to 15.
14. A method of increasing active agent
localization to a target cell population of a
mammalian recipient, which method comprises:
administering to the recipient a targeting moiety-anti-ligand conjugate;
allowing an amount of time to pass that is
sufficient for localization of the targeting moiety-anti-ligand conjugate to target cells;
thereafter administering to the recipient a clearing agent of claim 9, wherein the ligand is complementary to the anti-ligand in the targeting moiety-anti-ligand conjugate; and
subsequently administering to the recipient an active agent-ligand conjugate, wherein the active agent-ligand conjugate localizes to target cell-bound targeting moiety-anti-ligand conjugate.
15. A method according to claim 14, wherein the targeting moiety-anti-ligand conjugate comprises an avidin or a streptavidin anti-ligand component
covalently bound to the targeting moiety.
16. A method according to claim 14, wherein the targeting agent-anti-ligand conjugate is administered at a substantially tumor saturating dose.
17. A method according to claim 14, wherein the clearing agent administering step is conducted after a time period of about 20 to about 72 hours following the targeting moiety-anti-ligand administering step.
18. A method according to Claim 14, further comprising a second administering step of a clearing agent, involving administration of a second clearing agent of the formula
(Hexose)m╌Human Serum Albumin (HSA) - - (Ligand)n , wherein the second clearing agent administering step occurs prior to the active agent-ligand conjugate administering step, with a time period ranging from about 1 to about 24 hours elapsing between clearing agent administrations and wherein the second clearing agent is the same or different from previously administered clearing agent.
19. A method according to Claim 14 wherein the active agent-ligand conjugate is radiolabeled biotin.
20. A method according the Claim 19 wherein the active agent-ligand conjugate is an active agent-biotin-DOTA conjugate of the formula wherein R3 is H ; R4 is CH3 and n is 0 .
PCT/US1993/005406 1973-12-07 1993-06-07 Pretargeting methods and compounds WO1993025240A2 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
AT93915235T ATE210464T1 (en) 1992-06-09 1993-06-07 BIOTIN-DOTA CONJUGATES AND THEIR USE IN PRETARGETING PROCEDURES
CA002134239A CA2134239C (en) 1992-06-09 1993-06-07 Pretargeting methods and compounds
JP6501638A JPH07507804A (en) 1992-06-09 1993-06-07 Pre-targeting methods and compounds
US08/351,469 US5608060A (en) 1992-06-09 1993-06-07 Biotinidase-resistant biotin-DOTA conjugates
EP93915235A EP0646019B9 (en) 1992-06-09 1993-06-07 Biotin-DOTA conjugates and their use in pretargeting methods
DE69331319T DE69331319T2 (en) 1992-06-09 1993-06-07 Biotin-DOTA conjugates and their use in "pretargeting" processes
US08/350,551 US6075010A (en) 1992-06-09 1994-12-07 Small molecular weight ligand-hexose containing clearing agents
US08/468,513 US5976535A (en) 1992-06-09 1995-06-06 Pretargeting protocols for the enhanced localization of cytotoxins to target sites and cytotoxic combinations useful therefore
US08/788,339 US6287536B1 (en) 1992-06-09 1997-01-27 Two-step pretargeting methods using improved bidtin-active agent conjugates
US09/316,452 US6358490B2 (en) 1992-06-09 1999-05-21 Three-step pretargeting methods and compounds
US09/561,736 US6416738B1 (en) 1973-12-07 2000-04-25 Pretargeting methods and compounds
US09/920,454 US6709652B2 (en) 1992-06-09 2001-08-01 Pretargeting methods and compounds
US10/076,031 US20030099640A1 (en) 1992-06-09 2002-02-12 Three-step pretargeting methods and compounds
US10/125,788 US20030129191A1 (en) 1992-12-23 2002-04-17 Pretargeting methods and compounds
US10/775,897 US7078013B2 (en) 1992-06-09 2004-02-10 Pretargeting methods and compounds

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US07/895,588 1992-06-09
US07/895,588 US5283342A (en) 1992-06-09 1992-06-09 Biotinylated small molecules
US99538192A 1992-12-23 1992-12-23
US99538392A 1992-12-23 1992-12-23
US07/995,383 1992-12-23
US07/995,381 1992-12-23

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US99538392A Continuation-In-Part 1992-06-09 1992-12-23
US99538192A Continuation-In-Part 1973-12-07 1992-12-23

Related Child Applications (8)

Application Number Title Priority Date Filing Date
US16318473A Continuation-In-Part 1973-12-07 1973-12-07
US08/122,979 Continuation US5630996A (en) 1992-06-09 1993-09-16 Two-step pretargeting methods using improved biotin-active agent conjugates
US08/156,565 Continuation-In-Part US6022966A (en) 1992-06-09 1993-11-22 Pretargeting methods and compounds
US08/156,614 Continuation-In-Part US5578287A (en) 1992-06-09 1993-11-23 Three-step pretargeting methods using improved biotin-active agent
US16318893A Continuation-In-Part 1992-06-09 1993-12-07
US16318493A Continuation-In-Part 1992-06-09 1993-12-07
US35100594A Continuation-In-Part 1992-06-09 1994-12-07
US09/316,452 Continuation-In-Part US6358490B2 (en) 1992-06-09 1999-05-21 Three-step pretargeting methods and compounds

Publications (2)

Publication Number Publication Date
WO1993025240A2 true WO1993025240A2 (en) 1993-12-23
WO1993025240A3 WO1993025240A3 (en) 1994-02-17

Family

ID=27420561

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/005406 WO1993025240A2 (en) 1973-12-07 1993-06-07 Pretargeting methods and compounds

Country Status (7)

Country Link
US (2) US5608060A (en)
EP (2) EP0646019B9 (en)
JP (1) JPH07507804A (en)
AT (1) ATE210464T1 (en)
CA (1) CA2134239C (en)
DE (1) DE69331319T2 (en)
WO (1) WO1993025240A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994016734A1 (en) * 1993-01-21 1994-08-04 President And Fellows Of Harvard College Amplified direction of effector groups to specific target cells in an animal
EP0656115A1 (en) * 1992-08-21 1995-06-07 Immunomedics, Inc. Improved detection and therapy of lesions with biotin/avidin conjugates
EP0683676A1 (en) * 1993-02-02 1995-11-29 Neorx Corporation Directed biodistribution of small molecules
WO1995033491A1 (en) * 1994-06-06 1995-12-14 Elmaleh David R Biotin compounds for targetting tumors and sites of infection
US5482698A (en) * 1993-04-22 1996-01-09 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin polymer conjugates
EP0724601A1 (en) * 1993-08-17 1996-08-07 Neorx Corporation S 3n chelating compounds for the radiolabeling of ligands, anti-ligands or other proteins
EP0732943A1 (en) * 1993-11-23 1996-09-25 Neorx Corporation Three-step pretargeting methods and compounds
EP0733066A1 (en) * 1993-12-07 1996-09-25 Neorx Corporation Pretargeting methods and compounds
EP0736035A1 (en) * 1993-11-22 1996-10-09 Neorx Corporation Pretargeting methods and compounds
EP0743956A1 (en) * 1993-12-07 1996-11-27 NeoRx Pretargeting methods and compounds
EP0743958A1 (en) * 1994-02-07 1996-11-27 Research Development Foundation Non-viral vector
EP0813425A1 (en) * 1993-07-12 1997-12-29 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
US5716594A (en) * 1994-06-06 1998-02-10 The Jmde Trust Biotin compounds for targetting tumors and sites of infection
EP0832111A1 (en) * 1995-06-07 1998-04-01 Scott F. Rosebrough Modified avidin and streptavidin and methods of use thereof
EP0837696A1 (en) * 1995-06-07 1998-04-29 Immunomedics, Inc. Improved delivery of diagnostic and therapeutic agents to a target site
EP0906015A1 (en) * 1996-06-06 1999-04-07 Neorx Corporation Liver retention clearing agents
US5922302A (en) * 1993-05-17 1999-07-13 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin-metal chelating protein conjugates
US6015897A (en) * 1993-12-07 2000-01-18 Neorx Corporation Biotinamido-n-methylglycyl-seryl-o-succinamido-benzyl dota
US6075010A (en) * 1992-06-09 2000-06-13 Neorx Corporation Small molecular weight ligand-hexose containing clearing agents
US6120768A (en) * 1993-05-17 2000-09-19 Immunomedics, Inc. Dota-biotin derivatives
EP0914042A4 (en) * 1996-06-06 2000-10-25 Neorx Corp Cluster clearing agents
US6217869B1 (en) 1992-06-09 2001-04-17 Neorx Corporation Pretargeting methods and compounds
EP1121455A1 (en) * 1998-10-06 2001-08-08 Isis Pharmaceuticals, Inc. Zinc finger peptide cleavage of nucleic acids
US6358490B2 (en) 1992-06-09 2002-03-19 Neorx Corporation Three-step pretargeting methods and compounds
US6416738B1 (en) 1973-12-07 2002-07-09 Neorx Corporation Pretargeting methods and compounds
WO2002062398A2 (en) * 2001-01-08 2002-08-15 Neorx Corporation Radioactively labelled conjugates of phosphonates
WO2002066075A2 (en) * 2001-02-16 2002-08-29 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Biotin-derivatives and their conjugates with chelating agents
US6908903B1 (en) 1994-12-07 2005-06-21 Aletheon Pharmaceuticals, Inc. Cluster clearing agents
US10093741B1 (en) 2017-05-05 2018-10-09 Fusion Pharmaceuticals Inc. IGF-1R monoclonal antibodies and uses thereof
US10758636B2 (en) 2013-11-12 2020-09-01 Centre For Probe Development And Commercialization Residualizing linkers and uses thereof
US10925852B2 (en) 2015-06-30 2021-02-23 The Trustees Of Columbia University In The City Of New York Talc-bound compositions and uses thereof
US11191854B2 (en) 2017-05-05 2021-12-07 Centre For Probe Development And Commercialization Pharmacokinetic enhancements of bifunctional chelates and uses thereof
US11433148B2 (en) 2017-05-05 2022-09-06 Centre For Probe Development And Commercialization IGF-1R monoclonal antibodies and uses thereof
US11819551B2 (en) 2020-01-31 2023-11-21 Avilar Therapeutics, Inc. ASGPR-binding compounds for the degradation of extracellular proteins

Families Citing this family (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5670347A (en) 1994-05-11 1997-09-23 Amba Biosciences Llc Peptide-mediated gene transfer
JPH0889278A (en) * 1994-09-29 1996-04-09 Ajinomoto Co Inc Modified protein for introducing gene and its production
US5955605A (en) * 1995-02-21 1999-09-21 Neorx Corporation Biotinidase resistant biotin-DOTA conjugates
US6155266A (en) * 1995-05-08 2000-12-05 Maxim Pharmaceuticals, Inc. Method for treatment of cancer and infectious disease
US5739313A (en) * 1995-11-13 1998-04-14 Regents Of The University Of Minnesota Radionuclide labeling of vitamin B12 and coenzymes thereof
RU2219949C2 (en) * 1996-01-11 2003-12-27 Перигрин Фармасьютикалс Инк. Antibodies with reduced total positive charge
US20010023288A1 (en) * 1999-07-07 2001-09-20 Wilbur D. Scott Trifunctional reagent for conjugation to a biomolecule
JP4981197B2 (en) * 1996-10-08 2012-07-18 クレアテク バイオテクノロジー ベー.ファー. Methods for labeling nucleotides, labeled nucleotides and useful intermediates
EP0977770A4 (en) * 1997-03-14 2000-04-05 Univ Boston Multiflavor streptavidin
AU9692198A (en) * 1997-10-10 1999-05-03 Kevin J. Donahue Gene delivery compositions and methods
CA2341742A1 (en) * 1998-08-24 2000-03-02 Maxim Pharmaceuticals, Inc. Activation and protection of t-cells (cd4+ and cd8+) using an h2 receptor agonist and other t-cell activating agents
IL131266A0 (en) 1999-08-05 2001-01-28 N S T Neurosurvival Technologi Peptides and pharmaceutical compositions comprising same
IL125908A (en) 1998-08-24 2005-05-17 Nst Neurosurvival Technologies Peptides and pharmaceutical compositions comprising same
US6498181B1 (en) * 1999-01-06 2002-12-24 Maxim Pharmaceuticals Synergistic tumorcidal response induced by histamine
EP1141011A2 (en) 1999-01-12 2001-10-10 Neorx Corporation Compounds and methods to inhibit or augment an inflammatory response
DE60001629T2 (en) * 1999-01-15 2003-12-18 Light Sciences Corp THERAPEUTIC COMPOSITIONS FOR BONE METABOLISM DISORDERS OR BONE METASTASES CONTAINING A PHOTOSENSITIZER AND A BISPHOSPHONATE
JP2002534218A (en) * 1999-01-15 2002-10-15 ライト サイエンシーズ コーポレイション Non-invasive vascular therapy
US6602274B1 (en) * 1999-01-15 2003-08-05 Light Sciences Corporation Targeted transcutaneous cancer therapy
US6454789B1 (en) * 1999-01-15 2002-09-24 Light Science Corporation Patient portable device for photodynamic therapy
US6806363B1 (en) * 1999-04-16 2004-10-19 Mayo Foundation For Medical Education & Research Cobalamin conjugates useful as antitumor agents
AU5032400A (en) * 1999-05-21 2000-12-12 Wake Forest University Sigma-1 ligands useful for determining the proliferative status of cancer cells
US20030103948A1 (en) * 1999-06-07 2003-06-05 Neorx Corporation Streptavidin expressed gene fusions and methods of use thereof
US7144991B2 (en) 1999-06-07 2006-12-05 Aletheon Pharmaceuticals, Inc. Streptavidin expressed gene fusions and methods of use thereof
US20030143233A1 (en) * 1999-06-07 2003-07-31 Neorx Corporation Streptavidin expressed gene fusions and methods of use thereof
AU5597500A (en) * 1999-06-07 2000-12-28 Neorx Corporation Streptavidin expressed gene fusions and methods of use thereof
CA2371728C (en) * 1999-06-11 2009-06-02 Neorx Corporation High dose radionuclide complexes for bone marrow suppression
US7094885B2 (en) * 1999-07-11 2006-08-22 Neorx Corporation Skeletal-targeted radiation to treat bone-associated pathologies
JP2003512338A (en) * 1999-10-15 2003-04-02 メイオウ・フアウンデーシヨン・フオー・メデイカル・エジユケイシヨン・アンド・リサーチ Cobalamin conjugates useful as contrast agents and antitumor agents
CA2387767A1 (en) 1999-10-15 2001-04-26 Henricus P. C. Hogenkamp Cobalamin conjugates useful as imaging and therapeutic agents
US7591995B2 (en) * 1999-10-15 2009-09-22 Mayo Foundation For Medical Education And Research Cobalamin conjugates useful as imaging and therapeutic agents
US7067111B1 (en) * 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
DE19956029A1 (en) * 1999-11-22 2001-05-23 Aventis Pharma Gmbh Active substance carrier system based on coating a substance with enzymatically cleavable side groups, useful for gene therapy especially of cancer and inflammation
EP1267935A2 (en) 2000-01-12 2003-01-02 Light Sciences Corporation Novel treatment for eye disease
NO312708B1 (en) * 2000-02-21 2002-06-24 Anticancer Therapeutic Inv Sa Radioactive liposomes for therapy
CN1292798C (en) * 2000-06-02 2007-01-03 德克萨斯州立大学董事会 Ethylenedicysteine (EC)-drug conjugates
EP1315777B1 (en) * 2000-09-06 2007-10-31 AP Pharma, Inc. Degradable polyacetal polymers
CA2427146A1 (en) * 2000-10-25 2002-07-18 Mayo Foundation For Medical Education And Research Transcobalamin binding conjugates useful for treating abnormal cellular proliferation
ATE359807T1 (en) * 2001-02-21 2007-05-15 Surromed Inc MODIFIED ANNEXIN PROTEINS AND PREVENTION AND TREATMENT OF THROMBOSIS
US20090291086A1 (en) * 2001-02-21 2009-11-26 Alavita Pharmaceuticals, Inc. Compositions and Methods for Treating Cerebral Thrombosis and Global Cerebral Ischemia
US7635676B2 (en) * 2001-02-21 2009-12-22 Alavita Pharmaccuticals, Inc. Modified annexin proteins and methods for their use in organ transplantation
US7635680B2 (en) * 2001-02-21 2009-12-22 Alavita Pharmaceuticals, Inc. Attenuation of reperfusion injury
US7645739B2 (en) * 2001-02-21 2010-01-12 Alavita Pharmaceuticals, Inc. Modified annexin compositions and methods of using same
US6441163B1 (en) * 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
WO2003000010A2 (en) * 2001-06-20 2003-01-03 Mayo Foundation For Medical Education And Research Adenosyl-cobalamin fortified compositions
US20060074034A1 (en) * 2001-09-17 2006-04-06 Collins Douglas A Cobalamin mediated delivery of nucleic acids, analogs and derivatives thereof
JP2005508332A (en) * 2001-09-28 2005-03-31 メイオウ・フアウンデーシヨン・フオー・メデイカル・エジユケイシヨン・アンド・リサーチ Co-administration of transport protein and conjugated cobalamin for drug delivery
WO2003061696A2 (en) * 2002-01-23 2003-07-31 Light Sciences Corporation Systems and methods for photodynamic therapy
US6982154B2 (en) * 2002-02-21 2006-01-03 Surromed, Inc. Modified annexin proteins and methods for treating vaso-occlusive sickle-cell disease
ITRM20020128A1 (en) * 2002-03-08 2003-09-08 Sigma Tau Ind Farmaceuti DIMERI OF AVIDINA EFFECTIVE IN INCREASING THE CONCENTRATION OF RADIOACTIVE BIOTIN IN IMMUNOTHERAPY WITH "PRETARGETING".
WO2003077864A2 (en) * 2002-03-15 2003-09-25 Department Of Veterans Affairs, Rehabilitation R & D Service Methods and compositions for directing cells to target organs
KR100440725B1 (en) * 2002-06-20 2004-07-15 주식회사 그린진 바이오텍 A Method for Increasing an Abiotic-Resistance in Monocot Plants
KR20050062605A (en) * 2002-10-10 2005-06-23 디파트먼트 오브 베테랑스 어페어스 오피스 오브 더 제네럴 카운셀 (024) Detection, localization and staging of tumors using labeled activated lymphocytes directed to a tumor specific epitope
EP1587550B1 (en) * 2003-01-31 2011-07-27 Immunomedics, Inc. Methods and compositions for administering therapeutic and diagnostic agents
WO2005051429A2 (en) * 2003-11-19 2005-06-09 Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Targeted conjugates with a modified saccharide linker
US9050378B2 (en) * 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
CA2566662C (en) * 2004-05-13 2013-05-07 Centre National De La Recherche Scientifique A device for binding a target entity to a bait entity and detection methods using the same
US20050271585A1 (en) * 2004-06-07 2005-12-08 General Electric Company Extended conjugated polymers
US20060057062A1 (en) * 2004-09-10 2006-03-16 Trotter Dinko G Compositions and methods useful in pretargeted imaging
US20080181847A1 (en) * 2004-10-07 2008-07-31 Koninklijke Philips Electronics, N.V. Targeted Imaging and/or Therapy Using the Staudinger Ligation
US8535750B2 (en) * 2005-05-17 2013-09-17 Cargill, Incorporated Granular lecithins, granular lysolecithins, process for their production and compositions containing them
TWI381852B (en) * 2005-09-27 2013-01-11 Sigma Tau Ind Farmaceuti Biotin diaminoderivatives and their conjugates with macrocyclic chelating agents
US8758723B2 (en) * 2006-04-19 2014-06-24 The Board Of Regents Of The University Of Texas System Compositions and methods for cellular imaging and therapy
US10925977B2 (en) 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
EP2399993B1 (en) 2009-02-20 2017-09-20 The University of Tokyo Hypo-immunogenic streptavidin and use thereof
FI20090100A0 (en) 2009-03-16 2009-03-16 Wallac Oy Biotinidaasimääritys
GB0922369D0 (en) 2009-12-22 2010-02-03 Hammersmith Imanet Ltd Labelled biotin conjugates
PE20140406A1 (en) 2010-11-18 2014-04-16 Univ Yale BIFUNCTIONAL MOLECULES WITH ANTIBODY RECRUITMENT ACTIVITY AND INITIAL INHIBITORY ACTIVITY AGAINST HUMAN IMMUNODEFICIENCY VIRUS
GB201205360D0 (en) 2012-03-27 2012-05-09 Univ Edinburgh Biotinidase resistant biotinyl compounds
JP6178586B2 (en) 2013-02-20 2017-08-09 サヴィッド・セラピューティックス株式会社 Biotin variants, streptavidin variants and their use
CN106459089B (en) 2014-02-18 2021-01-08 赛威德医疗公司 Biotin variants, streptavidin mutants and uses thereof
CN109983024A (en) 2016-11-25 2019-07-05 赛威德医疗公司 Scavenger
WO2019230905A1 (en) 2018-05-30 2019-12-05 国立大学法人 東京大学 Halogenated biotin-modified dimer and use thereof
WO2019241609A1 (en) * 2018-06-15 2019-12-19 Trustees Of Boston University Polypeptide compositions and methods for site-specific targeting of therapeutic agents
JPWO2020032165A1 (en) 2018-08-08 2021-08-12 国立大学法人 東京大学 Conjugation of biotin-modified dimer and phthalocyanine dye
JPWO2021210573A1 (en) 2020-04-14 2021-10-21
JPWO2021215534A1 (en) 2020-04-24 2021-10-28

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0251494A2 (en) * 1986-06-23 1988-01-07 The Board Of Trustees Of The Leland Stanford Junior University Therapeutic or radiodiagnostic compound
WO1988008422A1 (en) * 1987-04-22 1988-11-03 Schering Aktiengesellschaft Substituted cyclic complex-forming substances, complexes and complex salts, process for manufacturing same, and pharmaceutical agents which contain them

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4647447A (en) * 1981-07-24 1987-03-03 Schering Aktiengesellschaft Diagnostic media
NZ211453A (en) * 1984-03-22 1989-01-06 Biotechnology Research Enterpr Aryl azides, their preparation and use in the detection, localisation and isolation of polynucleotides
US4639365A (en) * 1984-10-18 1987-01-27 The Board Of Regents, The University Of Texas System Gadolinium chelates as NMR contrast agents
US4678667A (en) * 1985-07-02 1987-07-07 501 Regents of the University of California Macrocyclic bifunctional chelating agents
DE3782169T2 (en) * 1986-07-03 1993-02-25 Upjohn Co METHOD FOR PRODUCING ANTIBIOTICA 10381B.
US5246692A (en) * 1986-09-05 1993-09-21 The United States Of America As Represented By The Secretary Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
FR2604092B1 (en) * 1986-09-19 1990-04-13 Immunotech Sa IMMUNOREACTIVES FOR TARGETING ANIMAL CELLS FOR VISUALIZATION OR DESTRUCTION IN VIVO
DE3722647A1 (en) * 1987-07-09 1989-01-19 Gerhard Ohlenschlaeger GALENIC USE OF A TRIPEPTID AS A MEDICINAL PRODUCT
DE68916052T2 (en) * 1988-02-03 1994-11-10 Hybritech Inc Modified haptens, useful as an image-forming agent and as a medicament.
GB8809616D0 (en) * 1988-04-22 1988-05-25 Cancer Res Campaign Tech Further improvements relating to drug delivery systems
US4923985A (en) * 1988-05-25 1990-05-08 The United States Of America As Represented By The Department Of Health & Human Services Process for synthesizing macrocyclic chelates
US4895955A (en) * 1988-05-27 1990-01-23 Lifecodes Corporation Non-radioactive carbodiimide precursor to nucleic acid probes
EP0378666A4 (en) * 1988-07-05 1992-01-22 Amgen Inc. Interleukin ii analogs
US5364613A (en) * 1989-04-07 1994-11-15 Sieving Paul F Polychelants containing macrocyclic chelant moieties
SE465155B (en) * 1989-12-19 1991-08-05 Exploaterings Ab Tbf METAL CHELET FORMING HYDROPHIL POLYMER FOR ADSORPTION ETC AND A SAT FOR PREPARATION OF THE POLYMER
DE59010815D1 (en) * 1989-12-27 1998-04-23 Boehringer Ingelheim Int USE OF AN ANTI-COAGULANT AS A DIAGNOSTIC
JP2776953B2 (en) * 1990-04-10 1998-07-16 日本メジフィジックス株式会社 Multifunctional metal chelate compounds and their uses
DE4035760A1 (en) * 1990-11-08 1992-05-14 Schering Ag MONO-N-SUBSTITUTED 1,4,7,10-TETRAAZACYCLODODECAN DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND PHARMACEUTICAL AGENTS CONTAINING THEM
IT1245748B (en) * 1990-12-21 1994-10-14 Mini Ricerca Scient Tecnolog PREPARATION INCLUDING BIOTINYLATE MONOCLONAL ANTIBODIES, AVIDIN AND BIOTIN, FOR DIAGNOSIS OF CANCER DISEASES AND ITS USE
US5326778A (en) * 1992-03-03 1994-07-05 Research Corporation Technologies, Inc. Conjugates of biotin and deferoxamine for radioimmunoimaging and radioimmunotherapy
CA2154667A1 (en) * 1993-02-02 1994-08-18 Linda M. Gustavson Directed biodistribution of small molecules

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0251494A2 (en) * 1986-06-23 1988-01-07 The Board Of Trustees Of The Leland Stanford Junior University Therapeutic or radiodiagnostic compound
WO1988008422A1 (en) * 1987-04-22 1988-11-03 Schering Aktiengesellschaft Substituted cyclic complex-forming substances, complexes and complex salts, process for manufacturing same, and pharmaceutical agents which contain them

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 106, no. 8, 23 February 1987, Columbus, Ohio, US; abstract no. 55742f, & HEPATOLOGY vol. 6, no. 4 , 1986 , BALTIMORE pages 723 - 728 P. VAN DER SLUIJS ET AL. 'DRUG TARGETING TO THE LIVER WITH LACTOSYLATED ALBUMINS: DOES THE GLYCOPROTEIN TARGET THE DRUG OR IS IT THE DRUG TARGETING THE GLYCOPROTEIN?' *
NUCL. MED. BIOL. vol. 18, no. 7 , 1991 pages 719 - 726 F. VIRZI ET AL. 'NEW INDIUM-111 LABELED BIOTIN DERIVATES FOR IMPROVED IMMUNOTARGETING.' cited in the application *
See also references of EP0646019A1 *

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6416738B1 (en) 1973-12-07 2002-07-09 Neorx Corporation Pretargeting methods and compounds
US5698405A (en) * 1985-07-05 1997-12-16 Immunomedics, Inc. Method of reducing immunogenicity
US6217869B1 (en) 1992-06-09 2001-04-17 Neorx Corporation Pretargeting methods and compounds
US6358490B2 (en) 1992-06-09 2002-03-19 Neorx Corporation Three-step pretargeting methods and compounds
US6075010A (en) * 1992-06-09 2000-06-13 Neorx Corporation Small molecular weight ligand-hexose containing clearing agents
EP0656115A1 (en) * 1992-08-21 1995-06-07 Immunomedics, Inc. Improved detection and therapy of lesions with biotin/avidin conjugates
EP0656115A4 (en) * 1992-08-21 1998-09-09 Immunomedics Inc Improved detection and therapy of lesions with biotin/avidin conjugates.
US5525338A (en) * 1992-08-21 1996-06-11 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin conjugates
WO1994016734A1 (en) * 1993-01-21 1994-08-04 President And Fellows Of Harvard College Amplified direction of effector groups to specific target cells in an animal
EP0683676A1 (en) * 1993-02-02 1995-11-29 Neorx Corporation Directed biodistribution of small molecules
EP0683676A4 (en) * 1993-02-02 1998-09-30 Neorx Corp Directed biodistribution of small molecules.
EP0695194A1 (en) * 1993-04-22 1996-02-07 Immunomedics, Inc. Improved detection and therapy of lesions with biotin/avidin polymer conjugates
EP0695194A4 (en) * 1993-04-22 1999-03-31 Immunomedics Inc Improved detection and therapy of lesions with biotin/avidin polymer conjugates
US5482698A (en) * 1993-04-22 1996-01-09 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin polymer conjugates
US6120768A (en) * 1993-05-17 2000-09-19 Immunomedics, Inc. Dota-biotin derivatives
US5922302A (en) * 1993-05-17 1999-07-13 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin-metal chelating protein conjugates
EP0813425A1 (en) * 1993-07-12 1997-12-29 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
EP0813425A4 (en) * 1993-07-12 1998-09-09 Neorx Corp Metal radionuclide labeled proteins for diagnosis and therapy
EP0724601A1 (en) * 1993-08-17 1996-08-07 Neorx Corporation S 3n chelating compounds for the radiolabeling of ligands, anti-ligands or other proteins
EP0724601A4 (en) * 1993-08-17 1999-02-03 Neorx Corp S 3n chelating compounds for the radiolabeling of ligands, anti-ligands or other proteins
EP0736035A1 (en) * 1993-11-22 1996-10-09 Neorx Corporation Pretargeting methods and compounds
EP0736035A4 (en) * 1993-11-22 1999-09-01 Neorx Corp Pretargeting methods and compounds
EP0732943A1 (en) * 1993-11-23 1996-09-25 Neorx Corporation Three-step pretargeting methods and compounds
EP0732943A4 (en) * 1993-11-23 1998-08-12 Neorx Corp Three-step pretargeting methods and compounds
EP0743956A4 (en) * 1993-12-07 1999-03-24 Neorx Corp Pretargeting methods and compounds
EP0743956A1 (en) * 1993-12-07 1996-11-27 NeoRx Pretargeting methods and compounds
EP0733066A4 (en) * 1993-12-07 1999-04-21 Neorx Corp Pretargeting methods and compounds
EP0733066A1 (en) * 1993-12-07 1996-09-25 Neorx Corporation Pretargeting methods and compounds
EP1346730A1 (en) * 1993-12-07 2003-09-24 Neorx Corporation Pretargeting methods and novel pretargeting conjugates
US6015897A (en) * 1993-12-07 2000-01-18 Neorx Corporation Biotinamido-n-methylglycyl-seryl-o-succinamido-benzyl dota
EP0743958A1 (en) * 1994-02-07 1996-11-27 Research Development Foundation Non-viral vector
EP0743958A4 (en) * 1994-02-07 1999-07-28 Res Dev Foundation Non-viral vector
US5716594A (en) * 1994-06-06 1998-02-10 The Jmde Trust Biotin compounds for targetting tumors and sites of infection
WO1995033491A1 (en) * 1994-06-06 1995-12-14 Elmaleh David R Biotin compounds for targetting tumors and sites of infection
US6172045B1 (en) * 1994-12-07 2001-01-09 Neorx Corporation Cluster clearing agents
US6908903B1 (en) 1994-12-07 2005-06-21 Aletheon Pharmaceuticals, Inc. Cluster clearing agents
EP0837696A4 (en) * 1995-06-07 2005-09-28 Immunomedics Inc Improved delivery of diagnostic and therapeutic agents to a target site
EP0832111A4 (en) * 1995-06-07 1999-03-03 Scott F Rosebrough Modified avidin and streptavidin and methods of use thereof
EP0837696A1 (en) * 1995-06-07 1998-04-29 Immunomedics, Inc. Improved delivery of diagnostic and therapeutic agents to a target site
EP0832111A1 (en) * 1995-06-07 1998-04-01 Scott F. Rosebrough Modified avidin and streptavidin and methods of use thereof
US6429297B1 (en) 1995-06-07 2002-08-06 University Of Rochester Modified avidin and streptavidin and methods of use thereof
EP0906015A4 (en) * 1996-06-06 2004-05-12 Neorx Corp Liver retention clearing agents
EP0914042A4 (en) * 1996-06-06 2000-10-25 Neorx Corp Cluster clearing agents
EP0906015A1 (en) * 1996-06-06 1999-04-07 Neorx Corporation Liver retention clearing agents
EP1121455A1 (en) * 1998-10-06 2001-08-08 Isis Pharmaceuticals, Inc. Zinc finger peptide cleavage of nucleic acids
EP1121455A4 (en) * 1998-10-06 2004-03-24 Isis Pharmaceuticals Inc Zinc finger peptide cleavage of nucleic acids
WO2002062398A2 (en) * 2001-01-08 2002-08-15 Neorx Corporation Radioactively labelled conjugates of phosphonates
WO2002062398A3 (en) * 2001-01-08 2003-12-18 Neorx Corp Radioactively labelled conjugates of phosphonates
CN100457192C (en) * 2001-02-16 2009-02-04 希格马托制药工业公司 Biotin-derivatives and their conjugates with chelating agents
AU2002237517B2 (en) * 2001-02-16 2006-11-30 Alfasigma S.p.A Biotin-derivatives and their conjugates with chelating agents
US7390828B2 (en) * 2001-02-16 2008-06-24 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Aminoderivatives of biotin and their conjugates with macrocyclic chelating agents
WO2002066075A2 (en) * 2001-02-16 2002-08-29 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Biotin-derivatives and their conjugates with chelating agents
US8044081B2 (en) 2001-02-16 2011-10-25 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Aminoderivative of biotin and their conjugates with macrocyclic chelating agents
CZ305442B6 (en) * 2001-02-16 2015-09-23 Sigma-Tau Industrie Farmaceutiche Riunite S. P. A. Biotin amino derivatives and their conjugates with macrocyclic gelling agents
WO2002066075A3 (en) * 2001-02-16 2003-01-30 Sigma Tau Ind Farmaceuti Biotin-derivatives and their conjugates with chelating agents
US11793895B2 (en) 2013-11-12 2023-10-24 Centre For Probe Development And Commercialization Residualizing linkers and uses thereof
US10758636B2 (en) 2013-11-12 2020-09-01 Centre For Probe Development And Commercialization Residualizing linkers and uses thereof
US10925852B2 (en) 2015-06-30 2021-02-23 The Trustees Of Columbia University In The City Of New York Talc-bound compositions and uses thereof
US10093741B1 (en) 2017-05-05 2018-10-09 Fusion Pharmaceuticals Inc. IGF-1R monoclonal antibodies and uses thereof
US11433148B2 (en) 2017-05-05 2022-09-06 Centre For Probe Development And Commercialization IGF-1R monoclonal antibodies and uses thereof
US11191854B2 (en) 2017-05-05 2021-12-07 Centre For Probe Development And Commercialization Pharmacokinetic enhancements of bifunctional chelates and uses thereof
US11819551B2 (en) 2020-01-31 2023-11-21 Avilar Therapeutics, Inc. ASGPR-binding compounds for the degradation of extracellular proteins

Also Published As

Publication number Publication date
WO1993025240A3 (en) 1994-02-17
EP1138334A2 (en) 2001-10-04
US5608060A (en) 1997-03-04
EP0646019A1 (en) 1995-04-05
ATE210464T1 (en) 2001-12-15
JPH07507804A (en) 1995-08-31
CA2134239A1 (en) 1993-12-23
DE69331319T2 (en) 2002-08-08
EP0646019B9 (en) 2002-12-18
DE69331319D1 (en) 2002-01-24
EP1138334A3 (en) 2002-04-03
CA2134239C (en) 2004-11-23
EP0646019B1 (en) 2001-12-12
US5630996A (en) 1997-05-20

Similar Documents

Publication Publication Date Title
US5630996A (en) Two-step pretargeting methods using improved biotin-active agent conjugates
US5847121A (en) Production of nitro-benzyl-dota via direct peptide cyclization
US5955605A (en) Biotinidase resistant biotin-DOTA conjugates
US6022966A (en) Pretargeting methods and compounds
US5976535A (en) Pretargeting protocols for the enhanced localization of cytotoxins to target sites and cytotoxic combinations useful therefore
US6015897A (en) Biotinamido-n-methylglycyl-seryl-o-succinamido-benzyl dota
US5578287A (en) Three-step pretargeting methods using improved biotin-active agent
US6709652B2 (en) Pretargeting methods and compounds
US5283342A (en) Biotinylated small molecules
WO1997046098A1 (en) Cluster clearing agents
US5624896A (en) Clearing agents useful in pretargeting methods
EP0794788B1 (en) Hepatic-directed compounds and reagents for preparation thereof
WO1995015979A1 (en) Pretargeting methods and compounds
WO1995015770A1 (en) Pretargeting methods and compounds
US6075010A (en) Small molecular weight ligand-hexose containing clearing agents
US5616690A (en) Hexose derivatized human serum albumin clearing agents
US6416738B1 (en) Pretargeting methods and compounds
US20030129191A1 (en) Pretargeting methods and compounds
WO1995015978A1 (en) Pretargeting methods and compounds
US5914312A (en) Pretargeting methods and compounds
US6358490B2 (en) Three-step pretargeting methods and compounds

Legal Events

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

Ref document number: 08122979

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 08133613

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 08156565

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 08156614

Country of ref document: US

AK Designated states

Kind code of ref document: A2

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A2

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

AK Designated states

Kind code of ref document: A3

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A3

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2134239

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1993915235

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 08351469

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1993915235

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 1996 753445

Country of ref document: US

Date of ref document: 19961125

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 1997 788339

Country of ref document: US

Date of ref document: 19970127

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 1997 845833

Country of ref document: US

Date of ref document: 19970428

Kind code of ref document: A

WWG Wipo information: grant in national office

Ref document number: 1993915235

Country of ref document: EP