CA2529027A1 - D-amino acid peptides - Google Patents

D-amino acid peptides Download PDF

Info

Publication number
CA2529027A1
CA2529027A1 CA002529027A CA2529027A CA2529027A1 CA 2529027 A1 CA2529027 A1 CA 2529027A1 CA 002529027 A CA002529027 A CA 002529027A CA 2529027 A CA2529027 A CA 2529027A CA 2529027 A1 CA2529027 A1 CA 2529027A1
Authority
CA
Canada
Prior art keywords
lys
virus
compound
group
hsg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA002529027A
Other languages
French (fr)
Other versions
CA2529027C (en
Inventor
William J. Mcbride
David M. Goldenberg
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunomedics Inc
Original Assignee
Immunomedics, Inc.
William J. Mcbride
David M. Goldenberg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunomedics, Inc., William J. Mcbride, David M. Goldenberg filed Critical Immunomedics, Inc.
Publication of CA2529027A1 publication Critical patent/CA2529027A1/en
Application granted granted Critical
Publication of CA2529027C publication Critical patent/CA2529027C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0815Tripeptides with the first amino acid being basic
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • 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
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1084Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody the antibody being a hybrid immunoglobulin
    • A61K51/109Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody the antibody being a hybrid immunoglobulin immunoglobulins having two or more different antigen-binding sites or multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/02Local antiseptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1008Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1016Tetrapeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1019Tetrapeptides with the first amino acid being basic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1021Tetrapeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The present invention provides compounds of the formula X-R1-D-[Dpr, Orn or Lys](A)-R2(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5; or R1(X)-D-[Dpr, Orn or Lys](A)-R2(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5, in which X is a hard acid cation chelator, a soft acid cation chelator or Ac-, R1, R2 and R3 are independently selected from a covalent bond or one or more D-amino acids that can be the same or different, Y is a hard acid cation chelator, a soft acid cation chelator or absent, Z is a hard acid cation chelator, a soft acid cation chelator or absent, and A and B are haptens or hard acid cation chelators and can be the same or different, and R4 and R5 are independently selected from the group consisting of hard acid cation chelators, soft acid cation chelators, enzymes, therapeutic agents, diagnostic agents and H. The present invention also provides methods of using these compounds and kits containing the compounds.

Description

D-AMINO ACID PEPTIDES
BACKGROUND
Field of the Invention The invention relates to immunological reagents for therapeutic use, for example, in radioimmunotherapy (RAIT), and diagnostic use, for example, in radioimmunodetection (RAID) and magnetic resonance imaging (MRI). This application claims priority to provisional patent application 60/47,403, filed June 13, 2003, the contents of which are hereby incorporated be reference in their entirety. The contents ofU.S.S.N. 60/090,142 and U.S.S.N. 601104,156 also are incorporated herein by reference in their entireties.
Related Art An approach to cancer therapy and diagnosis involves directing antibodies or antibody fragments to disease tissues, wherein the antibody or antibody fragment can target a diagnostic agent or therapeutic agent to the disease site. One approach to this methodology which has been under investigation, involves the use of bsAbs having at least one arm that specifically binds a targeted diseased tissue and at least one other arm that specifically binds a low molecular weight hapten. In this methodology, a bsAb is administered and allowed to localize to target, and to clear normal tissue.
Some time later, a radiolabeled low molecular weight hapten is given, which being recognized by the second specificity of the bsAb, also localizes to the original target.
Although low MW haptens used in combination with bsAbs possess a large number of specific imaging and therapy uses, it is impractical to prepare individual bsAbs for each possible application. Further, the application of a bsAb/low MW
hapten system has to contend with several other issues. First, the arm of the bsAb that binds to the low MW hapten must bind with high affinity, since a low MW hapten is designed to clear the living system rapidly, when not bound by bsAb. Second, the non-bsAb-bound low MW hapten actually needs to clear the living system rapidly to avoid non-target tissue uptake and retention. Third, the detection and/or therapy agent must remain associated with the low MW hapten throughout its application within the bsAb protocol employed.
Of interest with this approach are bsAbs that direct chelators and metal chelate complexes to cancers using Abs of appropriate dual specificity. The chelators and metal chelate complexes used are often radioactive, using radionuclides such as cobalt-57 (Goodwin et al., U.S. Patent No. 4,863,713), indium-111 (Barbet et al.; U.S.
Patent No. 5,256,395 and U.S. Patent No. 5,274,076, Goodwin et al., J. Nucl.
Med., 33:1366-1372 (I992), and Kranenborg et al., Cancer Res (suppl.), 55:5864s-5867s (1995) and Dancer (suppl.) 80:2390-2397 (1997)) and gallium-68 (Boden et al., Bioconjugate Chena., 6:373-379, (1995) and Schuhmacher et al., Cancef-Res., 55:115-123 (1995)) for radioimmuno-imaging. Because the Abs were raised against the chelators and metal chelate complexes, they have remarkable specificity for the complex against which they were originally raised. Indeed, the bsAbs of Boden et al.
have specificity for single enantiomers of enantiomeric mixtures of chelators and metal-chelate complexes. This great specificity has proven to be a disadvantage in one respect, in that othex nuclides such as yttrium-90 and bismuth-213 useful for radioimmunotherapy (BAIT), and gadolinium useful for MRI, cannot be readily substituted into available reagents for alternative uses. As a result iodine-131, a non-metal, has been adopted for BAIT purposes by using an I-131-labeled indium-metal-chelate complex in the second targeting step. A second disadvantage to this methodology requires that antibodies be raised against every agent desired for diagnostic or therapeutic use.
Pretargeting methodologies have received considerable attention for cancer imaging and therapy. Unlike direct targeting systems where an effector molecule (e.g., a radionuclide or a drug linked to a small carrier) is directly linked to the targeting agent, in pretargeting systems, the effector molecule is given some time after the targeting agent. This allows time for the targeting agent to localize in tumor lesions and, more importantly, clear from the body. Since most targeting agents have been antibody proteins, they tend to clear much more slowly from the body (usually days) than the smaller effector molecules (usually in minutes). In direct targeting systems involving therapeutic radionuclides, the body, and in particular the highly vulnerable red marrow, is exposed to the radiation all the while the targeting agent is slowly reaching its peak levels in the tumor and clearing from the body. In a pretargeting system, the radionuclide is usually bound to a small "effector"
molecule, such as a chelate or peptide, which clears very quickly from the body, and thus exposure of normal tissues is minimized. Maximum tumor uptake of the radionuclide is also very rapid because the small molecule efficiently transverses the tumor vasculature and binds to the primary targeting agent. Its small size may also encourage a more uniform distribution in the tumor.
Pretargeting methods have used a number of different strategies, but most often involve an avidin/streptavidin-biotin recognition system or bispecific antibodies that co-recognize a tumor antigen and the effector molecule. The avidin/streptavidin system is highly versatile and has been used in several configurations.
Antibodies can be coupled with streptavidin or biotin, which is used as the primary targeting agent.
This is followed sometime later by the effector molecule, which conjugated with biotin or with avidin/streptavidin, respectively. Another configuration relies on a 3-step approach first targeting a biotin-conjugated antibody, followed by a bridging with streptavidin/avidin, and then the biotin-conjugated effector is given. These systems can be easily converted for use with a variety of effector substances so long as the effector and the targeting agent can be coupled with biotin or streptavidin/avidin depending on the configuration used. With its versatility for use in many targeting situations and high binding affinity between avidinlstreptavidin and biotin, this type of pretargeting has considerable advantages over other proposed systems. However, avidin and streptavidin are foreign proteins and therefore would be imrnunogenic, which would limit the number of times they could be given in a clinical application.
In this respect, bsAbs have the advantage of being able to be engineered as a relatively non-immunogenic humanized protein. Although the binding affinity of a bsAb (typically 10-~ to 10-1° M) cannot compete with the extremely high affinity of the streptavidin/avidin-biotin affinity (~IO-15 M), both pretargeting systems are dependent on the binding affinity of the primary targeting agent, and therefore the higher affinity of the streptavidin/avidin-biotin systems may not offer a substantial advantage over a bsAb pretargeting system. However, most bsAbs have only one arm available for binding the primary target, whereas the streptavidin/avidin-biotin pretargeting systems have typically used a whole IgG with two arms for binding the target, which strengthens target binding. By using a divalent peptide, an affinity enhancement is achieved, which greatly improves the binding of the peptide to the target site compared to a monovalent peptide. Thus, both systems are likely to provide excellent targeting ratios with reasonable retention.
Pretargeting with a bsAb also requires one arm of the antibody to recognize an effector molecule. Most radionuclide targeting systems reported to date have relied on an antibody to a chelate-metal complex, such as antibodies directed indium-loaded DTPA or antibodies to other chelates. Since the antibody is generally highly selective for this particular chelate-metal complex, new bsAbs would need to be constructed with the particular effector antibody. This could be avoided if the antibody was not specific to the effector, but instead reacted with another substance. In this way, a variety of effectors could be made so long as they also contained the antibody recognition substance. We have continued to develop the pretargeting system originally described by Janevik-Ivanovska et al. that used an antibody directed against a histamine derivative, histamine-succinyl-glycl (HSG) as the recognition system on which a variety of effector substances could be prepared. Excellent pretargeting results have been reported using a radioiodinated and a rhenium-labeled divalent HSG-containing peptide. In this work, we have expanded this system to include peptides suitable for radiolabeling 9oY, i l lIn, and 177Lu, as well as an alternative 9~"'Tc-binding peptide.
Thus, there is a continuing need for immunological agents which can be directed to diseased tissue and can specifically bind to a subsequently administered targetable diagnostic or therapeutic conjugate, and a flexible system that accommodates different diagnostic and therapeutic agents without alteration to the bi-specific or mufti-specific antibodies.
In accomplishing the pxesent invention, the present inventors have discovered that it is advantageous to raise mufti-specific Abs against a targetable construct that is capable of carrying one or more diagnostic or therapeutic agents. By utilizing this technique, the characteristics of the chelator, metal chelate complex, therapeutic agent or diagnostic agent can be varied to accommodate differing applications, without raising new mufti-specific Abs for each new application. Further, by using this approach, two or more distinct chelators, metal chelate complexes, diagnostic agents or therapeutic agents can be used with the inventive mufti-specific Ab.
Summary of the Invention One embodiment of the present invention provides a compound comprising the formula:
X-R'-D-[Dpr, Orn or Lys](A)-Rz(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5; or Rl(X)-D-[Dpr, Orn or Lys](A)-RZ(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5, wherein:
X is a hard acid cation chelator, a soft acid cation chelator, an enzyme, a therapeutic agent, a diagnostic agent, or Ac-;
Rl is a covalent bond or one or more D-amino acids that can be the same or different;
R2 is a covalent bond or one or more D-amino acids that can be the same or different;
R3 is a covalent bond or one or more D-amino acids that can be the same or different;
Y is a hard acid cation chelator, a soft acid cation chelator, an enzyme, a therapeutic agent, a diagnostic agent or absent;
Z is a hard acid canon chelator, a soft acid cation chelator, an enzyme, a therapeutic agent, a diagnostic agent, or absent;
A and B are haptens or hard acid cation chelators and can be the same or different; and R4 and RS are independently selected from the group consisting of hard acid cation chelators, soft acid cation chelators, enzymes, therapeutic agents, diagnostic agents and H. In the present formula Dpr is 2,3-diaminoproprionic acid. In some of these embodiments, when Rl or R3 is a covalent bond then the other Rl or R3 can be one or more D-amino acids that can be the same or different. In these and other embodiments, R2 can be one or more D-amino acids that can be the same or different.
In still other of these embodiments, the compound can comprise the formula X-Rl-D-Lys(A)-RZ-D- Lys(B)-R3(Y)-NR4R5. In some of these embodiments RZ is a single D-amino acid. In other embodiments, R2 is two D-amino acids that can be the same or different. In further embodiments, R3 is a D-Lys and Y is a hard acid cation chelator or a soft acid cation chelator. In some of these embodiments R2 is not a D-Lys. In still further embodiments A and B are independently selected from the group consisting of histamine-succinyl-glycine (HSG), DTPA and fluorescein isothiocyanate. In yet other embodiments, Rl is one or more D-amino acids that can be the same or different, Rz is one or more D-amino acids that can be the same or different, R3 is a covalent bond, Y is absent, and A and B are haptens or hard acid cation chelators and can be the same or different. In additional embodiments A
and B
are haptens and can be the same or different. In these and other embodiments R~ and R2 are single D-amino acids and can be the same or different. When Rl is more than one amino acid then one, any or all of the amino acids can be attached to the (X) group. Similarly, when R~ is more than one amino acid then one, any or aII of the amino acids can be attached to the (Z) group. In some embodiments, Z is absent. In some embodiments, only one or two of X, Y, Z R4 or RS is an enzyme, a therapeutic agent or a diagnostic agent. Parentheses indicate substituents on the amino acid side chain. If the molecules are in the main chain of the peptide then they are not surrounded by parentheses.
In additional embodiments RI is selected from the group consisting of D-Tyr, D-Ala, D-Ser, D-Thr, D-Cys, D-Leu, D-Ile, D-Met, D-Gln, D-Asn, D-Val, and D-Phe.
In additional embodiments Rl is selected from the group consisting of D-Pro, D-His, D-Trp, D-Glu, D-Asp, D-Arg, and D-Lys. In these and other embodiments, R2 is selected from the group consisting of D-Asp, D-GIu and D-Tyr. In some of the embodiments described herein R4 and RS are both H. In other embodiments, one of X, R4 and RS is a hard acid cation chelator. In these and other embodiments, one of the remaining X, R4 and RS is a soft acid cation chelator. In some embodiments, X
is a hard acid cation chelator. In additional embodiments, one of R4 and RS is a hard acid cation chelator. In some embodiments, the hard acid cation chelator comprises a carboxylate or amine group. In yet further embodiments the hard acid cation chelator is selected from the group consisting of NOTA, DOTA, DTPA, and TETA. In still other embodiments, one of X, R4 and RS is a soft acid cation chelator, which can comprise a thiol group. The soft acid cation chelator can be selected from the group consisting of Tscg-Cys and Tsca-Cys. In some of the present compounds one of and RS is a soft acid cation chelator and the remaining R4 or RS is H.
In certain embodiments, X is Ac-, A and B are hard acid canon chelators and can be the same or different, R3 is a covalent bond, and Y is absent. In some embodiments, X is Ac-, A and B are haptens and can the same or different, RI
is a covalent bond, and Y is a soft acid cation chelator. Specific embodiments of the present compounds include:
DOTA-D-Asp-D-Lys(HSG)-D-Asp-D-Lys(HSG)-NHZ (IMP 271);
DOTA-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 277);
DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ (IMP 288);
DOTA-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 281);
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NHZ (IMP 284) DOTA-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 301) DOTA-D-Lys(HSG)-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 302) DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Cys-NH2 (IMP 305) Ac-D-Lys(In-DTPA)-D-Tyr-D-Lys(In-DTPA)-D-Lys(Tscg-Cys)-NH2 MH+
1813 (IMP 297) HCO-CO-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 289); and COOH
N
N/~COOH
N
HOOC
S
N"NOD-Ala-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH

In some embodiments of the present compounds, (i) no more than one of X, R4 and RS is a hard acid cation chelator and (ii) no more than one of X, R4 and RS is a soft acid cation chelator, Additional specific examples of the present compounds include:
Ac-D-Phe-D-Lys(DOTA)-D-Tyr-D-Lys(DOTA)-NHZ;

Ac-D-Phe-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-NH2;
Ac-D-Phe-D-Lys(Bz-DTPA)-D-Tyr-D-Lys(Bz-DTPA)-NH2 Ac-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(Tscg-Cys)-NH2;
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(Tscg-Cys)-NH2;
(Tscg-Cys)-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(DOTA)-NH2;
Tscg-D-Cys-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2;
(Tscg-Cys)-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2;
Ac-D-Cys-D-Lys(DOTA)-D-Tyr-D-Ala-D-Lys(DOTA)-D-Cys-NH2;
Ac-D-Cys-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-NH2;
Ac-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-D-Lys(TscG-Cys)-NH2; and Ac-D-Lys(DOTA)-D-Tyr-D-Lys(DOTA)-D-Lys(TscG-Cys)-NH2.
In these and other embodiments the compounds can further comprise at least one radionuclide. Examples of suitable radionuclides include 22sAc, 111Ag, 7zAs, 77AS 211At 198Au 199Au 212Bi 213Bi 758r 76Br llC sSC~ 62Cu 64Cu 67Cu~ 166D
> > > > > > > > > > > > Y
169Er' 18F~ s2Fe' s9Fe' 67Ga' 68Ga' ls4Gd' lssGd' ls6Gd' ls7Gd' lsBGd' 166H~' 120I' I23h 1241 1251 1311 110In I11~ 194, 177Lu slMn s2mMn 99Mo 13N ISO 32P 33P 211Pb a > > > v > > > > > > > o 0 212Pb 109Pd 149Pm 142Pr 143Pr 223Rd $2mRb 186Re I$$Re lg9Re lost 47Sc ls3Sm s o > > > > > a o > > > o 7sse' 83sr~ 89sr' I61~' 94mTC, 94TC' 99mTC, 86y' 90y' 90Y' and 89Zr.
In some of the present compounds the hard acid ration chelator is chelated to a ration selected from the group consisting of Group IIa and Group ITIa metal rations.
In these and other embodiments the soft acid ration chelator is chelated to a ration selected from the group consisting of transition metals, Bi, lanthanides and actinides.
Suitable rations include Tc, Re, and Bi.
In still other embodiments the present compounds R4 or Rs is a therapeutic agent, diagnostic agent or enz5mie. In these embodiments, the therapeutic agent, diagnostic agent or enzyme can be covalently linked to the compound by a linker moiety. In some embodiments, the linker moiety can comprise at least one amino acid. Suitable therapeutic agents for use in the present invention include a drug, prodrug or toxin. Prodrugs can be selected from the group consisting of epirubicin glucuronide, CPT-11, etoposide glucuronide, daunomicin glucuronide and doxorubicin glucuronide. Toxins can be selected from the group consisting of ricin, abrin, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed f antiviral protein, gelonin, diphtherin toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin. The therapeutic agent can comprises doxorubicin, SN-38, camptothecin, etoposide, methotrexate, 6-mercaptopurine or etoposide phosphate. The diagnostic agent can comprise one or more agents for photodynamic therapy, for example a photosensitizer, such as a benzoporphyrin monoacid ring A (BPD-MA), tin etiopurpurin (SnET2), sulfonated aluminum phthalocyanine (AISPc) and lutetium texaphyrin (Lutex). Other suitable diagnostic agents can comprise one or more image enhancing agents for use in magnetic resonance imaging (MRI), such as Mn, Fe, La or Gd. The diagnostic agent can also comprise one or more radiopaque or contrast agents for X-ray or computed tomography, such as barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric acid, iosulamide meglumine, iosemetic acid, iotasul, iotetric acid, iothalamic acid, iotroxic acid, ioxaglic acid, ioxotrizoic acid, ipodate, meglumine, metrizamide, metrizoate, propyliodone, or thallous chloride. The diagnostic agent can also comprises one or more ultrasound contrast agents, for example a liposome or dextran.
In some embodiments the liposome can be gas-filled. In some embodiments an enzyme that is capable of converting a drug intermediate to a toxic form to increase toxicity of the drug at a target site can be included in the compound.
In some of the present compounds the amino acid backbone can have a length of 2 to about 50, 75, 85 or 100 contigous amino acids. For example Rl or R3 can be from about 1,2, 5, 10 or 15 amino acids t about 20, 25, 30 or 35 amino acids in length.
In certain compounds the amino acid chain will be 3, 4, 5, 6, 7, 8, 9 or 10 amino acids in length. In certain embodiments, Rz is l, 2, or 3 amino acids in length. In some embodiments, the present invention also provides a targetable construct comprising any of the present compounds. In some of the present compounds, the amino acid backbone is cyclic, while in others it is linear.
Another embodiment of the present invention provides methods of diagnosing, treating or both diagnosing and treating a disease or a condition that can lead to a disease comprising:
(A) administering a targetable construct comprising any of the present compounds to a subject having, or suspected of having, a disease or a condition, wherein the targetable construct comprises at least one diagnostic or therapeutic cation, and/or one or more chelated or chemically bound therapeutic agents, diagnostic agents, or enzymes; and (B) administering a multispecific antibody or antibody fragment to the S subject, wherein the multispecific antibody or antibody fragment has at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds the targetable construct. These methods can further comprise:
(C) administering a clearing composition to the subject, wherein the clearing composition enhances clearance of non-localized antibodies or antibody fragments from the subject. In the present methods the targetable construct and the multispecific antibody or antibody fragment can be administered at substantially the same time.
Suitable examples of therapeutic canons used in the present methods can emit particles and/or positrons having 20 to 10,000 keV, for example I l lIn, 177Lu, 2lzBi, 1 S 213Bi' 211At' 62~u' 64Cu' 67Cu~ 90Y~ 125I' 131I' 32P' 33P' 47SC' I I IAg~
67Ga, 142Pr' 153~,m~
161' 166Dy' 166Hp' 186Re' 188Re' 189Re' 212Pb' 223Rd' 225AC' S9Fe' 7SSe, 77AS, 89Sr, ~9M0, IOSRh' 109Pd 143Pr 149Pm 169Er 194. 198Au 199Au and 211Pb. Ixl other > > > > > >
embodiments the diagnostic cation emits particles and/or positrons having 2S-10,000 keV, such as IIOIn, IIIIn, 177Lu, IsF, s2Fe, 62Cu, 64Cu, 67Cu, 67Ga, 6sGa, a6Y, 9oY, s9Zr, 94mTC, 94TC, 99mTC, 120I' 123I' 124I' 125I' 131I' ls4-IS8Gd' 32P' llC~ 13N~
lsO' 186Re' 188Re' slMn~ s2m~~ ssCo~ 72As~ 7sBr~ 76Br~ s2mRb and $3Sr.
In some of the present methods, the diagnostic canon or agent is used in positron-emission tomography (PET) or SPECT imaging. In other embodiments, the diagnostic cation or agent comprises one or more image enhancing agents used in 2S magnetic resonance~imaging (MRI), for example Mn, Fe, La and Gd. In other methods the diagnostic agent comprises one or more radiopaque or contrast agents for X-ray or computed tomography, for example comprises barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, 30 iosefamic acid, ioseric acid, iosulamide meglumine, iosemetic acid, iotasul, iotetric acid, iothalamic acid, iotroxic acid, ioxaglic acid, ioxotrizoic acid, ipodate, meglumine, metrizamide, metrizoate, propyliodone, or thallous chloride. In yet additional methods the diagnostic agent comprises one or more ultrasound contrast agents, for example a liposome or dextran. In some methods, the liposome is gas-filled.
In additional methods, the one or more diagnostic agents are selected from the group consisting of fluorescent compounds, chemiluminescent compounds, and bioluminescent compounds. Suitable fluorescent compounds include fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine. Suitable chemiluminescent compounds include luminol, isoluminol, an aromatic acridinium ester, an imidazole, an acridinium salt and an oxalate ester. Suitable bioluminescent compounds ainclude luciferin, luciferase and aequorin.
In some embodiments of the present methods, the targeted tissue is a tumor.
In some embodiments, the tumor produces or is associated with antigens selected from the group consisting of colon-specific antigen-p (CSAp), carcinoembryonic antigen (CEA), CD19, CD20, CD21, CD22, CD23, CD 25, CD30, CD74, CD80, HLA-DR, Ia, MUC l, MUC 2, MUC 3, MLJC 4, EGFR, HER 2/neu, PAM-4, TAG-72, EGP-1, EGP-2, A3, KS-1, Le(y), S 100, PSMA, PSA, tenascin, folate receptor, VEGF, necrosis antigens, 1L,-2, T101, MAGE, IL-6, insulin-like growth factor receptor, carbonic anhydrase IX, and combinations thereof.
In other methods at least one arm that specifically binds the targeted tissue is a monoclonal antibody or a fragment of a monoclonal antibody. In still further methods, the at least one other arm that specifically binds the targetable construct is a monoclonal antibody or a fragment of a monoclonal antibody. In additional methods, the at least one arm that specifically binds the targeted tissue is a human, chimeric or humanized antibody or a fragment of a human, chimeric or humanized antibody.
In additional methods, the at least one other arm that specifically binds the targetable construct is a human, chimeric or humanized antibody or a fragment of a human, chimeric or humanized antibody. In some of the methods, the multispecific antibody or antibody fragment further comprises a therapeutic nuclide, such as those selected from the group consisting of Il lIn, l7~Lu, zlzBi, zl3Bi, zllAt, 6zCu, 6aCu, 67Cu, 9°Y, lzsh 131h 32P' 33P' 47SC' 111Ag~ 67Ga, 142Pr' 153Sm' 161.~a 166Dy~ 166Ho~ la6Re~
lasRe, 189Re, zlzPb~ 223Ra~ 225Ac~ s9Fe~ 75Se~ 77AS' 89Sr~ 99M~~ 105' 109Pd' 143Pr' 149Pm~
169Er~ 194h.~
l9sAu, 199Au and 211Pb.
In certain methods, the multispecific antibody comprises the Fv of MAb Mu-9 and the Fv of MAb 679. In some of these methods, Mu-9 and/or 679 are chimerized or humanized. In other of these methods, Mu-9 and/or 679 are human Mu9 and 679.
In additional methods, the multispecific antibody comprises one or more of the CDRs of Mu-9. In some of the present methods, the multispecific antibody comprises one or more of the CDRs of 679. In some methods, the multispecific antibody is a fusion protein. In yet additional methods, the multispecific antibody comprises the Fv of MAb MN-14 and the Fv of MAb 679. In some of these methods, the MN-14, and/or 679 are chimerized or humanized. In other methods, the multispecific antibody comprises one or more of the CDRs of MN-14. In additional methods, the rnultispecific antibody comprises one or more of the CDRs of 679. Iii still further methods, the multispecific antibody is a fusion protein. In some of the present methods 9 the fusion protein is trivalent, and incorporates the Fv of an antibody reactive with CSAp. In some methods, the multispecific antibody incorporates a Class-III anti-CEA antibody and the Fv of 679.
In further methods disclosed herein the targetable construct comprises 1°B
atoms and the method further comprises:
(C) irradiating the I°B atoms localized at the targeted tissue, thereby effecting boron neutron capture therapy of the targeted tissue.
In additional methods, the targetable construct comprises an enzyme and the method further comprising:
(C) administering a drug to the subject, wherein the enzyme is capable of converting the drug a toxic form thereby increasing the toxicity of the drug at the targeted tissue.
In some of the present methods the disease or condition is selected from the group consisting of cancer, an infectious disease, an inflammatory disease, and autoimmune disease, cardiovascular disease, a metabolic disease, and a neurological disease. Examples of cancer include leukemias, lymphomas, sarcomas, melanomas, carcinomas, gliomas, and skin cancers. Specific examples of cancer include a B-cell malignancy, a B-cell lymphoma, chronic lymphatic leukemia, non-Hodgkin's lymphoma, acute lymphatic leukemias, or a multiple myeloma. Cancers that can be subjected to the present methods include esophageal, gastric, colonic, rectal, pancreatic, lung, breast, ovarian, urinary bladder, endometrial, cervical, testicular, renal, adrenal or liver cancer.
In some of the present methods the disease or condition is an infectious disease caused by a pathogen. Examples of pathogens include a fungus, virus, parasite, bacterium, protozoan, or mycoplasm. In some methods, the pathogen is a fungus selected from the group consisting of Microsporum, Trichophyton, Epidermophyton, Sporothrix schenckii, Cyrptococcus neoformans, Coccidioides immitis, Histoplasma capsulatum, Blastomyces dermatitidis, and Candida albicans. In other methods, the pathogen is a virus selected from the group consisting of human immunodeficiency virus (HIV), herpes virus, cytomegalovirus, rabies virus, influenza virus, hepatitis B virus, Sendai virus, feline leukemia virus, Reo virus, polio virus, human serum parvo-like virus, simian virus 40, respiratory syncytial virus, mouse mammary tumor virus, Varicella-Zoster virus, Dengue virus, rubella virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, papilloma virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus and blue tongue virus. In additional methods, the pathogen is a bacterium selected from the group consisting of Anthrax bacillus, Streptococcus agalactiae, Legionella pneumophilia, Streptococcus pyogenes, Staphylococcus, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Hemophilis influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, Mycobacterium tuberculosis and Clostridium tetani.
In still further methods, the pathogen is a parasite selected from the group consisting of helminth and malarial parasites. In yet other methods, the pathogen is a protozoan selected from the group consisting of Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosome rangeli, Trypanosome cruzi, Trypanosome rhodesiensei, Trypanosorna brucei, Schistosoma mansoni, Schistosoma japanicum, Babesia bovis, Elmeria tenella, Onchocerca volvulus, Leishmania tropica, Trichinella spiralis, Onchocerca volvulus, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus and Mesocestoides cord. In yet further methods, the pathogen is a mycoplasm selected from the group consisting of Mycoplasma arthritidis, Mycoplasma hyorhinis, Mycoplasma orate, Mycoplasma arginini, Acholeplasma laidlawii, Mycoplasma salivarum, and Mycoplasma pneumoniae.
In some of the present methods, the disease or condition is an inflammatory S disease or an autoimmune disease, such as acute idiopathic thrombocytopenic purpura, chronic idiopathic thrombocytopenic purpura, dermatomyositis, Sydenham's chorea, myasthenia gravis, systemic lupus erythematosus, lupus nephritis, rheumatic fever, polyglandular syndromes, bullous pemphigoid, diabetes mellitus, Henoch-Schonlein purpura, post-streptococcalnephritis, erythema nodosurn, Takayasu's arteritis, Addison's disease, rheumatoid arthritis, multiple sclerosis, sarcoidosis, ulcerative colitis, erythema multiforme, IgA nephropathy, polyarteritis nodosa, ankylosing spondylitis, Goodpasture's syndrome, thromboangitisubiterans, Sjogren's syndrome, primary biliary cirrhosis, Hashimoto's thyroiditis,thyrotoxicosis, scleroderma, chronic active hepatitis, polymyositis/dermatomyositis, polychondritis, parnphigus vulgaris, Wegener's granulomatosis, membranous nephropathy, amyotrophic lateral sclerosis, tabes dorsalis, giant cell arteritis/polymyalgia, pernicious anemia, rapidly progressive glomerulonephritis, psoriasis, and fibrosing alveolitis.
In other methods, the disease or condition is cardiovascular disease, such as a myocardial infarction, ischemic heart disease, atherosclerotic plaques, clots, and emboli. In additional methods, the disease or condition is a metabolic disease, such as amyloidosis. In further methods, the disease or condition is a neurological disease, such as Alzheimer's disease.
The present invention also provides methods for detecting, identifying or treating a target cell, tissue or pathogen in a subject, comprising:
(a) administering a targetable construct comprising any of the present compounds to a subject; and (b) administering a multispecific antibody or antibody fragment to the subject, wherein the multispecific antibody or antibody fragment comprises at least one arm that specifically binds a target cell, tissue or pathogen and at least one other arm that specifically binds the targetable construct. In some of these methods, the target comprises a molecule produced by or associated with the target cell, tissue, pathogen. In certein methods, the target tissue is a diseased tissue. The diseased tissue can be identified intraoperatively, endoscopically or intravascularly.
In some methods, the target tissue is normal tissue, such as ovary, thymus, parathyroid, endometrium, bone marrow, or spleen. In additional methods, the pathogen is a fungus, virus, parasite, bacterium, protozoan, or mycoplasm. Examples of fungi include Microsporum, Trichophyton, Epidermophyton, Sporothrix schenckii, Cyrptococcus neoformans, Coccidioides immitis, Histoplasma capsulatum, Blastomyces dermatitidis, and Candida albicans. Examples of viri include human immunodeficiency virus (HIV), herpes virus, cytomegalovirus, rabies virus, influenza virus, hepatitis B virus, Sendai virus, feline leukemia virus, Reo virus, polio virus, human serum parvo-like virus, simian virus 40, respiratory syncytial virus, mouse mammary tumor virus, Varicella-Zoster virus, Dengue virus, rubella virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus and blue tongue virus. Examples of bacterium include Anthrax bacillus, Streptococcus agalactiae, Legionella pneumophilia, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitides, Pneumococcus, Hemophilis influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, Mycobacterium tuberculosis and Tetanus toxin. Parasites can be a helminth or a malarial parasite. Examples of protozoa include Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosoma rangeli, Trypanosoma craze, Trypanosoma rhodesiensei, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japanicum, Babesia bovis, Elmeria tenella, Onchocerca volvulus, Leishmania tropica, Trichinella spiralis, Onchocerca volvulus, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus and Mesocestoides corti.
Examples of mycoplasms include Mycoplasma arthritidis, Mycoplasma hyorhinis, Mycoplasma orate, Mycoplasma arginini, Acholeplasma laidlawii, Mycoplasma salivarum, and Mycoplasma pneumoniae.
In some of the present methods the targetable construct further comprises at least one radionuclide, therapeutic agent, diagnostic agent or enzyme.
Examples of radionuclide can be a2sAc, lllAg, 72As, 77As, 2l~At, I~BAu, 199Au, zl2Bi, Zi3Bi, 75Br, 76Br' l 1C' ssCO' 62Cu' 64Cu' 67Cu' 166Dy~ 169Er' 18F.' s2~.e' s9Fe' 67Ga' 68Ga' 154Gd' ISSGd' ls6Gd' ls7Gd' lsBGd' 166110' l2oh 1231' 1241' 1251' 1311' IIOln' 11111,1' 1941x,' 177Lu' SIMn, 52mMn, 99M0' 13N' 150 32P 33P 211Pb 212Pb 109Pd 149Pm 142Pr 143Pr 223Rd 82mRb > > > > > > > > > > >
186Re 188Re 189Re lOSRh 47SC ls3sm 75Se 83Sr 89Sr 161 94mTC ~4TC ~~mTC 86Y
a a ~ o a ~ s ~ a ~ a > > >
9°Y, 9°Y, and $~Zr.
In some methods, the diagnostic agent comprises an imaging agent. In some of the disclosed methods, the method can further comprise:
(c) administering a clearing composition to the subject, wherein the composition enhances clearance of non-localized antibodies or antibody fragments from the subject. The present methods can be used to treat mammals, including humans, primates, equines, canines and felines. In some of the present methods, the therapeutic agent comprises one or more drugs, toxins, cytokines, hormones, or growth factors. In some methods, the diagnostic agent comprises a contrast agent. In additional methods, the imaging agent is an agent used for PET or SPECT. In some of the present methods, the multispecific antibody or antibody fragment is bispecific.
The present invention also provides kits for treating or identifying diseased tissues in a subject comprising:
(a) a targetable construct comprising one or more of the present compounds; and (b) a multispecific antibody or antibody fragment having at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds the targetable construct. The present kits can further comprise:
(c) a clearing composition for enhancing clearance of non-localized antibodies and antibody fragments. In some of these kits, the diagnostic agent selected from the ou consistin of lloln 11111.1 177Lu 1sF s2Fe 6zCu 64Cu 67Cu p g > > > > > > > >
67Ga, 68Ga, 86Y' 90Y' 89zr' 94mTC' 94TC, 99mTc, 1201' 1231' 1241' 1251' 1311' 154-ls8Gd' 32P' 11C~ 13N~ Iso~ ls6Re~ 188Re, sly s2mMn ssCo 72As 7sBr 76Br 82mRb and $3Sr. In > > > > > >
other kits, the therapeutic agent is selected from the group consisting of 111In, 177Lu, 212$i' 213$i' 211At' 62G,u~ 64Cu' 67Cu' 90Y' 1251' 1311' 32P' 33P' 47SC' 111Ag' 67CTa, 142Pr, ls3sm 161,Lb 1661 166130 186Re 188Re 189Re 212Pb 223Rd 225AC s9Fe 75Se 77AS
> > Y> > > > > > > > > > >
89Sr, 99M0~ IOSRh~ 1o91,d~ 143Pr~ 149Pm' 169Er~ 19411, l9aAu~ 199Au and 211Pb.

In some of the above kits, when the targetable construct comprises an enzyme the kit further comprises a drug which the enzyme is capable of converting to a toxic form to increase the toxicity of the drug.
The present invention can also provide a multi-specific or bi-specific antibody or antibody fragment having at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds a targetable construct.
In the present methods, when the targetable construct comprises an enzyme, the methods can further comprise administering to the patient 1) a prodrug, when the enzyme is capable of converting the prodrug to a drug at the target site;
2) a drug which is capable of being detoxified in the patient to form an intermediate of lower toxicity, when the enzyme is capable of reconverting the detoxified intermediate to a toxic form, and, therefore, of increasing the toxicity of the drug at the target site, 3) a prodrug which is activated in the patient through natural processes and is subject to detoxification by conversion to an intermediate of lower toxicity, when the enzyme is capable of reconverting the detoxified intermediate to a toxic form, and, therefore, of increasing the toxicity of the drug at the target site, or 4) a second targetable construct which comprises a carrier portion which comprises or bears at least one epitope recognizable by the at least one other arm of the bi-specific antibody or antibody fragment, and a prodrug, when the enzyme is capable of converting the prodrug to a drug at the target site.
In another embodiment, kits can further comprise, when the first targetable construct comprises an enzyme, 1) a prodrug, when the enzyme is capable of converting the prodrug to a drug at the target site;
2) a drug which is capable of being detoxified in the patient to form an intermediate of lower toxicity, when the enzyme is capable of reconverting the detoxified intermediate to a toxic form, and, therefore, of increasing the toxicity of the drug at the target site, 3) a prodrug which is activated in the patient through natural processes and is subject to detoxification by conversion to an intermediate of lower toxicity, when the enzyme is capable of reconverting the detoxified intermediate to a toxic form, and, therefore, of increasing the toxicity of the drug at the target site, or 4) a second targetable construct which comprises a Garner portion which comprises or bears at least one epitope recognizable by the at least one other arm of the bi-specific antibody or antibody fragment, and a prodrug, when the enzyme is capable of converting the prodrug to a drug at the target site.
Another embodiment of the invention provides a method of preparing the antibodies or antibody fragments by recombinant technology. In accordance with this aspect of the present invention, there is provided a method of preparing a bi-specific antibody or antibody fragment having at least one arm that specifically binds a targeted tissue and at least one other ann that specifically binds a targetable construct, comprising:
(A) introducing the recombinant DNA construct described above into a host cell;
(B) growing the cell and isolating the antibody or antibody fragment.
In another embodiment of the present invention there is provided a method of preparing a bi-specific fusion protein having at least one arm that specifically binds to a targeted tissue and at least one other ann that is specifically binds to a targetable construct, comprising:
(1) (A) introducing into a host cell a recombinant DNA construct comprising an expression cassette capable of producing in the host cell a fragment of the bi-specific fusion protein, wherein the construct comprises, in the 5' to 3' direction of transcription, a transcriptional initiation regulatory region functional in the host cell, a translational initiation regulatory region functional in the host cell, a DNA sequence encoding a scFv linked to a light-chain antibody fragment, and a transcriptional and translational termination regulatory region functional in the host cell, wherein the fragment of the bi-specific fusion protein is under the control of the regulatory regions;
(B) co-introducing into the host cell a recombinant DNA construct comprising an expression cassette capable of producing in the host cell a Fd fragment which is complementary to the light-chain antibody fragment in (A) and which when associated with the light-chain antibody fragment forms a Fab fragment whose 1~

binding site is specific for the targeted tissue, wherein the construct comprises, in the 5' to 3' direction of transcription, a transcriptional initiation regulatory region functional in the host cell, a translational initiation regulatory region functional in the host cell, a DNA sequence encoding a Fd fragment, and a transcriptional and translational termination regulatory region functional in the host cell, wherein the Fd fragment is under the control of the regulatory regions;
(C) growing the cell and isolating the bi-specific fusion protein, or (2) (A) introducing into a first host cell a recombinant DNA construct comprising an expression cassette capable of producing in the first host cell a fragment of the bi-specific fusion protein, wherein the construct comprises, in the 5' to 3' direction of transcription, a transcriptional initiation regulatory region functional in the first host cell, a translational initiation regulatory region functional in the first host cell, a DNA sequence encoding a scFv linked to a light-chain antibody fragment, and a transcriptional and translatianal termination regulatory region functional in the first host cell, wherein the fragment of the bi-specific fusion protein is under the control of the regulatory regions;
(B) introducing into a second host cell a recombinant DNA construct comprising an expression cassette capable of producing in the second host cell a Fd fragment which is complementary to the light-chain antibody fragment in (2)(A) and which when associated with the light-chain antibody fragment forms a Fab fragment whose binding site is specific for the targeted tissue, wherein the construct comprises, in the 5' to 3' direction of transcription, a transcriptional initiation regulatory region functional in the second host cell, a translational initiation regulatory region functional in the second host cell, a DNA sequence encoding a Fd fragment, and a transcriptional and translational termination regulatory region functional in the second host cell, wherein the Fd fragment is under the control of the regulatory regions;
(C) growing the first and second host cells;
(D) optionally isolating the bi-specific fusion protein fragment and the Fd fragment; and (E) combining the fragments to produce a bi-specific fusion protein and isolating the bi-specific fusion protein.

A variety of host cells can be used to prepare bi-specific antibodies or antibody fragments, including, but not limited to, mammalian cells, insect cells, plant cells and bacterial cells. In one embodiment, the method utilizes a mammalian zygote, and the introduction of the recombinant DNA construct produces a transgenic animal capable of producing a bi-specific antibody or antibody fragment.
The present inventors have discovered that it is advantageous to raise bsAbs against a targetable construct that is capable of carrying one or more diagnostic or therapeutic agents. By utilizing this technique, the characteristics of the chelator, metal chelate complex, therapeutic agent or diagnostic agent can be varied to accommodate differing applications, without raising new bsAbs for each new application. Further, by using this approach, two or more distinct chelators, metal chelate complexes or therapeutic agents can be used with the inventive bsAb.
The invention further relates to a method of screening for a targetable construct comprising:
contacting said targetable construct with a bi-specific antibody or antibody fragment having at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds said targetable construct to give a mixture;
wherein said at least one arm is capable of binding to a complementary binding moiety on the target cells, tissues or pathogen or on a molecule produced by or associated therewith; and optionally incubating said mixture; and analyzing said mixture.
Additional aspects, features and advantages of the invention will be set forth in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention. The embodiments and advantages of the invention may be realized and obtained by means of the instrumentalities and combinations particularly pointed out in the present specification. One skilled in the art will recognize that the embodiments described herein can be suitably used in combination with any other suitable described embodiment.
Brief Description of the Drawings Figure 1 shows a reversed phase HPLC (RP-HPLC)trace of labeled IMP 281.

Figure 2 shows a size-exclusion HPLC (SE-HPLC)trace of labeled IMP 281 Figure 3 shows RP-HPLC traces demonstrating the stability of IMP 281 in human serum.
Figure 4 shows an SE-HPLC of IMP 281 binding to m679xhMN14.
Figures 5 and 6 show SE-HPLC traces of IMP 281 bound to m679xhMN14 after incubation in human serum.
Figures 7 and 8 show RP- and SE-HPLC traces of labeled IMP 284.
Figure 9 shows an SE-HPLC of IMP 284 binding to m679xhMN14 Figure 10 shows RP-HPLC traces demonstrating the stability of IMP 284 in human serum.
Figures 11 and 12 show SE-HPLC traces of IMP 284 bound to m679xhMN14 after incubation in human serum.
Figure 13 shows stability of labeled IMP 284 in mouse serum.
Figures 14 and 15 show stability of labeled IMP 284 bound to m679xhMN14 in human serum.
Figures 16 and 17 show RP- and SE-HPLC traces of labeled IMP 281.
Figure 18 shows an SE-HPLC trace of labeled IMP 281 bound to m679xhMN 14.
Figures 19 and 20 show RP- and SE-HPLC traces of labeled IMP 284.
Figure 21 shows an SE-HPLC trace of IMP 284 bound to m679xhMN14.
Figure 22 shows stability of labeled IMP 281 in human serum.
Figure 23 shows stability of labeled IMP 284 in human serum.
Figure 24 shows stability of labeled IMP 281 bound to m679xhMN14 in human serum Figure 25 shows stability of labeled IMP 284 bound to m679xhMN14 in human serum Figure 26 shows tissue uptake of l2sIhMNl4 x m679 Fab' x Fab' in pretargeting at 3 and 24 h post peptide injection.
Figure 27 shows a comparison of tissue uptake of peptides IMP 241, 281 and 284 in pretargeting, 3 (top) and 24 h (bottom) post peptide injection.
Figure 28 shows a comparison of peptide kidney uptake in. pretargeting at 3 and 24 h post peptide injection.

Figure 29 shows tissue uptake of IMP 241, 281 and 284 peptides at 3 and 24 hours post injection.
Detailed Description Unless otherwise specified, "a" or "an" means "one or more"
'The present invention provides compounds comprising the fornmla:
X-R'-D-[Dpr, Orn or Lys](A)-R2(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5; or R~(X)-D-[Dpr, Orn or Lys](A)-RZ(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5, wherein:
X is a hard acid cation chelator, a soft acid cation chelator, an enzyme, a therapeutic agent, a diagnostic agent, or Ac-;
Rl is a covalent bond or one or more D-amino acids that can be the same or different;
R2 is a covalent bond or one or more D-amino acids that can be the same or different;
R3 is a covalent bond or one or more D-amino acids that can be the same or different;
Y is a hard acid cation chelator, a soft acid cation chelator, an enzyme, a therapeutic agent, a diagnostic agent or absent;
Z is a hard acid cation chelator, a soft acid cation chelator, an enzyme, a therapeutic agent, a diagnostic agent, or absent;
A and B are haptens or hard acid canon chelators and can be the same or different; and R4 and RS are independently selected from the group consisting of hard acid cation chelators, soft acid cation chelators, enzymes, therapeutic agents, diagnostic agents and H. In the present formula Dpr is 2,3-diaminoproprionic acid. In some of these embodiments, when RI or R3 is a covalent bond then the other Rl or R3 can be one or more D-amino acids that can be the same or different. In these and other embodiments, R~ can be one or more D-amino acids that can be the same or different.
In still other of these embodiments, the compound can comprise the formula X-Rt-D-Lys(A)-R2-D- Lys(B)-R3(Y)-NR4R5. In some of these embodiments R2 is a single D-amino acid. In other embodiments, R2 is two D-amino acids that can be the same or different. In further embodiments, R3 is a D-Lys and Y is a hard acid cation chelator or a soft acid canon chelator. In some of these embodiments RZ is not a D-Lys.
In still further embodiments A and B are independently selected from the group consisting of histamine-succinyl-glycine (HSG), DTPA and fluorescein isothiocyanate. In yet other embodiments, R' is one or more D-amino acids that can be the same or different, R2 is one or more D-amino acids that can be the same or different, R3 is a covalent bond, Y is absent, and A and B are haptens or hard acid cation cheIators and can be the same or different. In additional embodiments A
and B
are haptens and can be the same or different. In these and other embodiments Rl and Ra are single D-amino acids and can be the same or different. When Rl is more than one amino acid then one, any or all of the amino acids can be attached to the (X) group. Similarly, when RZ is more than one amino acid then one, any or all of the amino acids can be attached to the (Z) group. In some embodiments, Z is absent. In some embodiments, only one or two of X, Y, Z R4 or RS is an enzyme, a therapeutic agent or a diagnostic agent. Parentheses indicate substituents on the amino acid side chain. If the molecules are in the main chain of the peptide then they are not surrounded by parentheses. In the context of the present invention the skilled artisan will recognize that one or more amino acids may refer to 1-10 amino acids, advantageously 1-5 amino acids, although more than 10 amino acids may be employed as required.
In additional embodiments Rl is selected from the group consisting of D-Tyr, D-Ala, D-Ser, D-Thr, D-Cys, D-Leu, D-Ile, D-Met, D-Gln, D-Asn, D-Val, and D-Phe.
In additional embodiments Rl is selected from the group consisting of D-Pro, D-His, D-Trp, D-Glu, D-Asp, D-Arg, and D-Lys. In these and other embodiments, RZ is selected from the group consisting of D-Asp, D-GIu and D-Tyr. In some of the embodiments described herein R4 and RS are both H. In other embodiments, one of X, R4 and RS is a hard acid cation chelator. In these and other embodiments, one of the remaining X, R4 and RS is a soft acid cation chelator. In some embodiments, X
is a hard acid cation chelator. In additional embodiments, one of R4 and RS is a hard acid cation chelator. In some embodiments, the hard acid cation chelator comprises a carboxylate or amine group. In yet furthex embodiments the hard acid cation chelator is selected from the group consisting of NOTA, DOTA, DTPA, and TETA. In still other embodiments, one of X, R4 and RS is a soft acid cation chelator, which can comprise a thiol group. The soft acid cation chelator can be selected from the group consisting of Tscg-Cys and Tsca-Cys. In some of the present compounds one of and RS is a soft acid cation chelator and the remaining R4 or RS is H.
In certain embodiments, X is Ac-, A and B are hard acid cation chelators and can be the same or different, R3 is a covalent bond, and Y is absent. In some embodiments, X is Ac-, A and B are haptens and can the same or different, Rl is a covalent bond, and Y is a soft acid cation chelator. Specific embodiments of the present compounds include:
DOTA-D-Asp-D-Lys(HSG)-D-Asp-D-Lys(HSG)-NHZ (IMP 271);
DOTA-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ (IMP 277);
DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ (IMP 288);
DOTA-D-AIa-D-Lys(HSG)-D-GIu-D-Lys(HSG)-NH2 (IMP 0281);
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NHZ (IMP 284) DOTA-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 30I) DOTA-D-Lys(HSG)-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 302) DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Cys-NH2 (IMP 305) Ac-D-Lys(In-DTPA)-D-Tyr-D-Lys(In-DTPA)-D-Lys(Tscg-Cys)-NH2 MH+
1813 (IMP 297) HCO-CO-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 289); and COOH
N
N/~COOH
HOO
N
C
S
N~N~D-Ala-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 H H
In some embodiments of the present compounds, (i) no more than one of X, R4 and RS is a hard acid canon chelator and (ii) no more than one of X, R4 and RS
is a soft acid cation chelator. Additional specific examples of the present compounds include:
Ac-D-Phe-D-Lys(DOTA)-D-Tyr-D-Lys(DOTA)-NH2;
Ac-D-Phe-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-NHz;
Ac-D-Phe-D-Lys(Bz-DTPA)-D-Tyr-D-Lys(Bz-DTPA)-NHZ
Ac-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(Tscg-Cys)-NHZ;
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(Tscg-Cys)-NHZ;
(Tscg-Cys)-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(DOTA)-NH2;
Tscg-D-Cys-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2;
IO (Tscg-Cys)-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2;
Ac-D-Cys-D-Lys(DOTA)-D-Tyr-D-Ala-D-Lys(DOTA)-D-Cys-NH2;
Ac-D-Cys-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-NH2;
Ac-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-D-Lys(TscG-Cys)-NHZ; and Ac-D-Lys(DOTA)-D-Tyr-D-Lys(DOTA)-D-Lys(TscG-Cys)-NHZ.
In these and other embodiments the compounds can further comprise at least one radionuclide. In some of the present compounds the hard acid cation chelatox is chelated to a cation selected from the group consisting of Group IIa and Group IITa metal cations. In these and other embodiments the soft acid cation chelator is chelated to a cation selected from the group consisting of transition metals, Bi, lanthanides and actinides.
In still other embodiments the present compounds R4 or RS is a therapeutic agent, diagnostic agent or enzyme.
In some of the present compounds the amino acid backbone can have a length of 2 to about 50, 75, 85 or 100 contigous amino acids. For example Rl or R3 can be from about 1,2, 5, 10 or 15 amino acids t about 20, 25, 30 or 35 amino acids in length.
In certain compounds the amino acid chain will be 3, 4, 5, 6, 7, 8, 9 or 10 amino acids in length. In certain embodiments, RZ is 1, 2, or 3 amino acids in length. In some embodiments, the present invention also provides a targetable construct comprising any of the present compounds. In some of the present compounds, the amino acid backbone is cyclic, while in others it is linear. The present compounds are useful, inter alia, as targetable constructs.

Desirably, the targetable construct includes a peptide having at least 2 units of a recognizable hapten. Examples of recognizable haptens include, but axe not limited to, histamine succinyl glycine (HSG) and fluorescein isothiocyanate. The targetable construct may be conjugated to a variety of agents useful for treating or identifying diseased tissue. Examples of conjugated agents include, but are not limited to, chelators, metal chelate complexes, drugs, toxins (e.g., ricin, abrin, ribonuclease (e.g., RNase), DNase I, Staplaylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtherin toxin, Pseudomonas exotoxin, Pseudornonas endotoxin) and other effector molecules. Additionally, enzymes useful for activating a prodrug or increasing the target-specific toxicity of a drug can be conjugated to the targetable construct. Thus, the use of bsAb which are reactive to a targetable construct allows a variety of therapeutic and diagnostic applications to be performed without raising new bsAb for each application.
Bispecific antibody (bsAb) pretargeting represents a potentially non-immunogenic, highly selective alternative for diagnostic and therapeutic applications.
The bsAb pretargeting system described herein represents an additional significant advantage over other pretargeting systems in that it potentially can be developed for use with a variety of different imaging or therapeutic agents. The flexibility of this system is based on use of an antibody directed against histamine-succinyl-glycl (HSG) and the development of peptides containing the HSG residue. HSG-containing peptides were synthesized with either DOTA for the chelation of 11 iln, 9oY, or'77Lu or a technetium/rhenium chelate. For pretargeting, these peptides were used in combination with bispecific antibodies using the anti-HSG Fab' chemically stabilized with the Fab' of either an anti-carcinoembryonic antigen (CEA) or an anti-colon-specific antigen-p (CSAp) antibody to provide tumor targeting capability for tumors expressing these antigens. However, other antigen targets may include diverse tumor-associated antigens known in the art, such as against CD19, CD20, CD21, CD22, CD23, CD25, CD30, CD74, CD 80, HLA-DR, Ia, MUC 1, MUC 2, MUC 3, MUC 4, EGFR, HER 2/neu, PAM-4, BrE3, TAG-72 (B72.3, CC49), EGP-1 (e.g., RS7), EGP-2 (e.g., 17-lA and other Ep-CAM targets), Le(y) (e.g., B3), A3, KS-1, 5100, IL-2, T101, necrosis antigens, folate receptors, angiogenesis markers (e.g., VEGF), tenascin, PSMA, PSA, tumor-associated cytokines, MAGE and/or fragments thereof.

Tissue-specific antibodies (e.g., against bone marrow cells, such as CD34, CD74, etc., parathyroglobulin antibodies, etc.) as well as antibodies against non-malignant diseased tissues, such as fibrin of clots, macrophage antigens of atherosclerotic plaques (e.g., CD74 antibodies), and also specific pathogen antibodies (e.g., against bacteria, viruses, and parasites) are well known in the art.
The peptides can be radiolabeled to a high specific activity in a facile manner that avoids the need for purification. In some embodiments the peptides will be cleared rapidly from the body with minimal retention in tumor or normal tissues. The pxetargeting system described herein is highly flexible, beirig capable of using a wide array of compounds of diagnostic imaging and therapeutic interest, and by achieving excellent tumor uptake and targeting ratios, is highly promising for use in these applications.
Additionally, encompassed is a method for detecting andlor treating target cells, tissues or pathogens in a mammal, comprising administering an effective amount of one or more of the present compounds as a targetable construct. The method can further comprise administering a bispeciflc antibody or antibody fragment comprising at least one arm that specifically binds a targeted cell, tissue or pathogen and at least one other arm that specifically binds a targetable construct. As used herein, the term "pathogen" includes, but is not limited to fungi (e.g., Microsporum, Trichophyton, Epidermophyton, Sporothrix schenckii, Cryptococcus neoformans, Coccidioides immitis, Histoplasma Capsulatum, Blastomyces dermatitidis, Candida albicans), viruses (e.g., human immunodeficiency virus (H1V), herpes virus, cytomegalovirus, rabies virus, influenza virus, hepatitis B virus, Sendai virus, feline leukemia virus, Reo virus, polio virus, human serum parvo-like virus, simian virus 40, respiratory syncytial virus, mouse mammary tumor virus, Varicella-Zoster virus, Dengue virus, rubella virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choxiomeningitis virus, wart virus and blue tongue virus), parasites, bacteria (e.g., Anthrax bacillus, Streptococcus agalactiae, Legionella pneumophilia, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Hemophilis influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, Mycobacterium tuberculosis and Tetanus toxin), mycoplasma (e.g., Mycoplasma arthritidis, M. hyorhinis, M. orate, M. arginini, Acholeplasma laidlawii, M. salivarum, and M. pneumoniae) and protozoans (e.g., Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosome rangeli, Trypanosome cruzi, Trypanosome rhodesiensei, Trypanosome brucei, Schistosoma mansoni, Schistosoma japanicum, Babesia bovis, Elmeria tenella, Onchocerca volvulus, Leishmania tropica, Trichinella spiralis, Onchocerca volvulus, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus and Mesocestoides corti). See LT.S.
Patent No. 5,332,567.
Also provided herein are antibodies and antibody fragments. The antibody fragments are antigen binding portions of an antibody, such as F(ab')Z, F(ab)2, Fab', Fab, and the like. The antibody fragments bind to the same antigen that is recognized by the intact antibody. For example, an anti-CD22 monoclonal antibody fragment binds to an epitope of CD22.
The term "antibody fragment" also includes any synthetic or genetically engineered protein that acts like an antibody by binding to a specific antigen to form a complex. For example, antibody fragments include isolated fragments, "Fv"
fragments, consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy chain variable regions are connected by a peptide linker ("sFv proteins"), and minimal recognition units consisting of the amino acid residues that mimic the "hypervariable region." Three of these so-called "hypervariable" regions or "complementarity-determining regions" (CDR) are found in each variable region of the light or heavy chain. Each CDR is flanked by relatively conserved framework regions (FR). The FR
are thought to maintain the structural integrity of the variable region. The CDRs of a light chain and the CDRs of a corresponding heavy chain form the antigen-binding site. The "hypervariability" of the CDRs accounts for the diversity of specificity of antibodies.
As used herein, the term "subject" refers to any animal (i.e., vertebrates and invertebrates) including, but not limited to humans and other primates, rodents (e.g., mice, rats, and guinea pigs), lagamorphs (e.g., rabbits), bovines (e.g, cattle), ovines (e.g., sheep), caprines (e.g., goats), porcines (e.g., swine), equines (e.g., horses), canines (e.g., dogs), felines (e.g., cats), domestic fowl (e.g., chickens, turkeys, ducks, geese, other gallinaceous. birds, etc.), as well as feral or wild animals, including, but not limited to, such animals as ungulates (e.g., deer), bear, fish, lagamorphs, rodents, birds, etc. It is not intended that the term be limited to a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are encompassed by the term.
Suitable antibodies and fragments for use with the present compounds include the following: Fvs derived from anti-CD20 antibodies, such as those described in Provisional U.S. Application titled "Anti-CD20 Antibodies And Fusion Proteins Thereof And Methods Of Use", Attorney Docket No. 18733/1073, U.S. Provisional No. 60/356,132, U.S. Provisional Application No. 60/416,232 and Attorney Docket No. 18733/1155 (the contents of which are in their entirety herein by reference);
hMN-14 antibodies, such as those disclosed in U.S. Application No. 5,874,540 (the contents of which are incorporated in their entirety herein by reference), which is a Class III anti-carcinoembryonic antigen antibody (anti-CEA antibody); Mu-9 antibodies, such as those described in U.S. Application No. 10/116,116 (the contents of which are incorporated in their entirety herein by reference); LL1 antibodies, such as those described in U.S. Provisional Application No. 60/360,259 (the contents of which are incozporated in their entirety herein by reference); AFP antibodies, such as those described in U.S. Provisional Application No. 60/399,707 (the contents of which are incorporated in their entirety herein by reference); PAM4 antibodies, such as those described in Provisional U.S. Application titled "Monoclonal Antibody cPAM4", Attorney Docket No. 18733/1102 (the contents of which are incorporated in their entirety herein by reference); RS7 antibodies, such as those described in U.S.
Provisional Application No. 60/360,229 (the contents of which are incorporated in their entirety herein by reference); humanized MN3 antibodies, such as those disclosed in U.S. Provisional Application No. 60/414,341, and CD22 antibodies, such as those disclosed in U.S. Patent Nos. 5,789,554 and 6,187,287 and U.S.
Application Nos. 09/741,843 and 09/988,013 (the contents of which are incorporated in their entirety herein by reference). Many other tumor-associated antigens of hematopoietic and solid tumors are known to those skilled in the art, as contained in the referenced applications, and include (but are not limited to) CD 15, CD 19, CD20, CD21, CD22, CD23, CD25, CD40, CD45, CD66, CD74, CD80, Ii, Ia, HLA-DR, PSMA, PSA, prostastic acid phosphatase, tenascin, Le(y), AFP, HCG, CEA, CSAp, PAM4, MUC1, MUC2, MUC3, MUC4, EGP-1, EGP-2, EGFR, HER2lneu, insulin growth-factor receptors, S 100, VEGF, Placenta Growth Factor (P1GF), placental alkaline phosphatase, necrosis products, oncogene products, and the like. The present compounds can also be used in conjunction with any of the methods disclosed in the above applications.
II. Constructs Targetable to Antibodies The targetable construct can be of diverse structure as described herein. In preferred embodiments, the compounds are selected to diminish the elicitation of immune responses and/or for rapid ifa vivo clearance when used within the bsAb targeting method. Hydrophobic agents are best at eliciting strong immune responses, whereas hydrophilic agents are preferred for rapid ira vivo clearance, thus, a balance between hydrophobic and hydrophilic needs to be established. This is accomplished, in part, by relying on the use of hydrophilic chelating agents to offset the inherent hydrophobicity of many organic moieties. Also, sub-units of the targetable construct may be chosen which have opposite'solution properties, for example, peptides, which contain amino acids, some of which are hydrophobic and some of which are hydrophilic. Aside from peptides, carbohydrates may be used.
Any amino acid can be used in the present compounds, including naturally and non-naturally occurring amino acids.
The targetable construct may include a peptide backbone having as few as two amino-acid residues, with preferably two to ten amino acid residues, and may be coupled to other moieties such as chelating agents. The targetable construct should be a low molecular weight construct, preferably having a molecular weight of less than 50,000 daltons, and advantageously less than about 20,000 daltons, 10,000 daltons or 5,000 daltons, including any metal ions that may be bound to the chelating agents.
For instance, the known peptide DTPA-Tyr-Lys(DTPA)-OH (wherein DTPA is diethylenetriaminepentaacetic acid) has been used to generate antibodies against the indium-DTPA portion of the molecule. However, by use of the non-indium-containing molecule, and appropriate screening steps, new Abs against the tyrosyl-lysine dipeptide can be made. More usually, the antigenic peptide of the targetable construct will have four or more residues, such as the peptide DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2, wherein DOTA is 1,4,7,10-tetraazacyclododecanetetraacetic acid and HSG is the histamine succinyl glycyl group of the formula:
__~ O H
~~.~ N
N N
H H O
The non-metal-containing peptide may be used as an immunogen, with resultant Abs screened for reactivity against the D-Phe-D-Lys-D-Tyr-D-Lys backbone.
The haptens of the targetable construct also provide an immunogenic recognition moiety, for example, a chemical hapten. Using a chemical hapten, preferably the HSG hapten, high specificity of the construct for the antibody is exhibited. This occurs because antibodies raised to the HSG hapten are known and can be easily incorporated into the appropriate bsAb. Thus, binding of the haptens to the peptide backbone would result in a targtable construct that is specific for the bsAb or bsFab.
The present inventors have discovered that peptides having all D-amino acid backbones are stable in vitro and in vivo, even compared against peptides having one or a few D-amino acids incorporated into the backbone. Incorporation of unnatural amino acids, e.g., D-amino acids, into the peptide backbone structure also helps to ensure that, when used with the final bsAb/construct system, the arm of the bsAb which recognizes the targetable construct is specific. The invention further contemplates other backbone structures such as those constructed from non-natural amino acids and peptoids.
The peptides to be used as described can be synthesized conveniently on an automated peptide synthesizer using a solid-phase support and standard techniques of repetitive orthogonal deprotection and coupling. Free amino groups in the peptide, that are to be used later for chelate conjugation, are advantageously blocked with standard protecting groups such as an acetyl group. Such protecting groups will be known to the skilled artisan. See Greene and Wuts Protective Groups in Organic Syn.tlaesis, 1999 (John Wiley and Sons, N.Y.). When the peptides are prepared for later use within the bsAb system, they are advantageously cleaved from the resins to generate the corresponding C-terminal amides, in order to inhibit iya vivo carboxypeptidase activity.
III. Chelate Moieties The presence of hydrophilic chelate moieties on the targetable construct helps to ensure rapid i~ vivo clearance. In addition to hydrophilicity, chelators are chosen for their metal-binding properties, and may be changed at will since, at Ieast for those targetable constructs whose bsAb epitope is part of the peptide or is a non-chelate chemical hapten, recognition of the metal-cheIate complex is no longer an issue.
Particularly useful metal-chelate combinations include 2-benzyl-DTPA and its I O monomethyl and cyclohexyl analogs, used with 47Sc, SZFe, SSCo, 67Ga, ~$Ga, ~ 1 iln, sv~r~ vol,~ i6y~ ~7~Lu~ 212Bi~ zisBi, and 22sAc for radio-imaging and RAIT.
The same chelators, when complexed with non-radioactive metals such as Mn, Fe and Gd for use with MRI, when used along with the bsAbs of the invention. Macrocyclic chelators such as NOTA (1,4,7-triaza-cyclononane-N,N',N"-triacetic acid), DOTA, and TETA (p-bromoacetamido-benzyl-tetraethylaminetetraacetic acid) axe of use with a variety of metals and radiometals, most particularly with xadionuclides of Ga, Y and Cu, respectively.
DTPA and,DOTA-type chelators, where the ligand includes hard base chelating functions such as carboxylate or amine groups, are most effective for chelating hard acid cations, especially Group IIa and Group I)Ta metal cations. Such metal-chelate complexes can be made very stable by tailoring the ring size to the metal of interest. Other ring-type chelators such as macrocyclic polyethers are of interest for stably binding nuclides such as z23Ra for RAIT. Porphyrin chelators may be used with numerous radiometals, and are also useful as certain cold metal complexes for bsAb-directed immuno-phototherapy. Also, more than one type of chelator may be conjugated to the targetable construct to bind multiple metal ions, e.g., cold ions, diagnostic radionuclides and/or therapeutic radionuclides.
Particularly useful diagnostic radionuclides that can be bound to the chelating agents of the targetable construct include, but are not limited to, 11°In, 111Ixi, 177Lu, 18F, 52Fe' 62Cu' 6dCu' 67Cu' 67Ga~ 6aGa~ 86Y~ 90Y' 89zr~ 94mTC' 94TC' 99m'fC~ 120h 123I' 124I' 126I' 131h 164-lseGd 32p uC 13N 1s0 is6Re lBgRe slMn 52'"Mn s6Co 72As 7sBr 76Bx 82mRb $3Sr or > > > > > > > > > > > > > > >
other gamma-, beta-, or positron-emitters. Preferably, the diagnositc radionuclides include a decay energy in the range of 25 to 10,000 keV, more preferably in the range of 25 to 4,000 keV, and even more preferably in the range of 20 to 1,000 keV, and still more preferably in the range of 70 to 700 keV. Total decay energies of useful positron-emitting radionuclides are preferably < 2,000 keV, more preferably under 1,000 keV, and most preferably < 700 keV. Radionuclides useful as diagnostic agents utilizing gamma-ray detection include, but are not limited to: Cr-SI, Co-57, Co-58, Fe-59, Cu-67, Ga-67, Se-75, Ru-97, Tc-99m, In-111, In-114m, I-123, I-125, I-131, Yb-169, Hg-I97, and Tl-ZOI. Decay energies of useful gamma-ray emitting radionuclides are preferably 20-2000 keV, moxe preferably 60-600 keV, and most preferably I00-300 keV.
Particularly useful therapeutic radionuclides that can be bound to the chelating agents of the targetable construct include, but are not limited to "'In, "'Lu, 2'2Bi, 2'3Bi, 211At' 62C''u' 64G"'u' 67C''u' 90I)' 125I' 131h 32P' 33P~ 47SC' 111Ag' 67Ga' la2px~ 153fm' 161'I'b' 166DY~
166H~' 186Re' 188Re' 189Re' 212Pb' 223Rd' 225AC' S9Fe' 75Se' 77AS~ 89Sr' 99M~~
IOS~~ 109Pd~ 143Pr' '49pm, '69Er, '9aIr, 198Au, '99Au, and 2"Pb. The therapeutic radionuclide preferably has a decay energy in the range of 25 to 10,000 keV. Decay energies of useful beta-particle-emitting nuclides are preferably 25-5,000 keV, moxe preferably 100-4,000 keV, and most preferably 500-2,500 keV. Also preferred are radionuclides that substantially decay with Auger-emitting particles. For example, Co-58, Ga-67, Br-80m, Tc-99m, Rh-103m, Pt-109, In-111, Sb-119, I-125, Ho-161, Os-189m and Ir-192. Decay energies of useful beta-particle-emitting nuclides axe preferably < 1,000 keV, moxe preferably < 100 keV, and most preferably < 70 keV. Also preferred are radionuclides that substantially decay with generation of alpha-particles. Such radionuclides include, but are not limited to: Dy-152, At-211, Bi-212, Ra-223, Rn-219, Po-215, Bi-21 l, Ac-225, Fr-221, At-217, Bi-213 and Fm-255. Decay energies of useful alpha-particle-emitting radionuclides are preferably 2,000-9,000 keV, more preferably 3,000-8,000 keV, and most preferably 4,000-7,000 keV.
Chelators such as those disclosed in U.S. Patent 5,753,206, especially thiosemi-carbazonylglyoxylcysteine(Tscg-Cys) and thiosemicarbazinyl-acetylcysteine (Tsca-Cys) chelators are advantageously used to bind soft acid cations of Tc, Re, Bi and other transition metals, lanthanides and actinides that are tightly bound to soft base ligands, especially sulfur- or phosphorus-containing ligands. It can be useful to link more than one type of chelator to a peptide, e.g., a hard acid chelator like DTPA
for In(II~ cations, and a soft acid chelator (e.g, thiol-containing chelator such as Tscg-Cys) for Tc canons. Because antibodies to a di-DTPA hapten are known (Barbet '395, supra) and are readily coupled to a targeting antibody to form a bsAb, it is possible to use a peptide hapten with cold di-DTPA chelator and another chelator for binding a radioisotope, in a pretargeting protocol, for targeting the radioisotope. One example of such a peptide is Ac-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-D-Lys(Tscg-Cys)-NH2. This peptide can be preloaded with In(III) arid then labeled with 99-m-Tc cations, the In(III) ions being preferentially chelated by the DTPA and the Tc cations binding preferentially to the thiol-containing Tscg-Cys. Other hard acid chelators such as NOTA, DOTA, TETA and the like can be substituted for the DTPA groups, and Mabs specific to them can be produced using analogous techniques to those used to generate the anti-di-DTPA Mab.
It will be appreciated that two different hard acid or soft acid chelators can be incorporated into the linker, e.g., with different chelate ring sizes, to bind preferentially to two different hard acid or soft acid cations, due to the differing sizes of the cations, the geometries of the chelate rings and the preferred complex ion structures of the cations. This will permit two different metals, one or both of which may be radioactive or useful for MRI enhancement, to be incorporated into a linker for eventual capture by a pretargeted bsAb.
Preferred chelators include NOTA, DOTA and Tscg and combinations thereof.
These chelators have been incorporated into a chelator-peptide conjugate motif as exemplified in the following constructs:
DOTA-D-Asp-D-Lys(HSG)-D-Asp-D-Lys(HSG)-NHZ (IMP 271);
DOTA-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ (IMP 277);
DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 288);
DOTA-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ (IMP 0281);
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NHZ (IMP 284) DOTA-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 301) DOTA-D-Lys(HSG)-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 302) DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Cys-NH2 (IMP 305) Ac-D-Lys(In-DTPA)-D-Tyr-D-Lys(In-DTPA)-D-Lys(Tscg-Cys)-NH2 MH+
1$13 (IMP 297) HCO-CO-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 289); and COON
C/'~
N
N~COOH
HOOC
S
N~N~D-Ala-D-Lys(HSG -D-T r-D-L s HSG -NH
H H ) y y( ) 2 The chelator-peptide conjugates above, are capable of binding 6sGa and are thus useful in positron emission tomography (PET) applications.
Chelators are coupled to the peptides of the targetable construct using standard chemistries, some of which are discussed more fully in the working examples below.
IV. Genexal Methods for Preparation of Metal Chelates Chelator-peptide conjugates may be stored for long periods as solids. They may be metered into unit doses for metal-binding reactions, and stored as unit doses either as solids, aqueous or semi-aqueous solutions, frozen solutions or lyophilized preparations. They may be labeled by well-known procedures.
Typically, a hard acid cation is introduced as a solution of a convenient salt, and is taken up by the hard acid chelator and possibly by the soft acid chelator.
However, later addition of soft acid cations leads to binding thereof by the soft acid chelator, displacing any hard acid cations which may be chelated therein. For example, even in the presence of an excess of cold 111InC13, labeling with 99m-Tc(V) glucoheptonate or with Tc cations generated ira situ with stannous chloride and Na99m-Tc04 proceeds quantitatively on the soft acid chelator.
Other soft acid canons such as lssRe, isBRe, aisBi and divalent or trivalent cations of Mn, Co, Ni, Pb, Cu, Cd, Au, Fe, Ag (monovalent), Zn and Hg, especially sa.Cu and 67Cu, and the like, some of which are useful for radioimmunodetection or radioimmunotherapy, can be loaded onto the linker peptide by analogous methods. Re cations also can be generated in situ from perrhenate and stannous ions or a prereduced rhenium glucoheptonate or other trans~helator can be used. Because reduction of perrhenate requires more stannous ion (typically above 200 ~.g/mL
final concentration) than is needed for the reduction of Tc, extra care needs to be taken to ensure that the higher levels of stannous ion do not reduce sensitive disulfide bonds S such as those present in disulfide-cyclized peptides. During radiolabeling with rhenium, similar procedures are used as are used with the Tc-99m. One method for the preparation of Re0 metal complexes of the Tscg-Cys- ligands is by reacting the peptide with ReOCl3(P(Ph3)Z but it is also possible to use other reduced species such as Re0(ethylenediamine)2.
V. Methods of Administration It should be noted that much of the discussion presented hereinbelow focuses on the use of the disclosed targetable constructs in the context of treating diseased tissue. The invention contemplates, however, the use of the bispecific antibodies and targetable constructs in treating and/or imaging normal tissue and organs using the methods described in U.S. Patent Nos. 6,126,916; 6,077,499; 6,010,680;
5,776,095;
5,776,094; 5,776,093; 5,772,981; 5,753,206; 5,746,996; 5,697,902; 5,328,679;
5,128,119; 5,101,827; and 4,?35,210, which are incorporated herein by reference. As r used herein, the term "tissue" refers to tissues, including but not limited to, tissues from the ovary, thymus, parathyroid, bone marrow or spleen. An important use when targeting normal tissues is to identify and treat them when they are ectopic (i.e., displaced from their normal location), such as in endometriosis.
The administration of a bsAb and the targetable construct discussed above may be conducted by administering the bsAb at some time prior to administration of the therapeutic agent which is associated with the linker moiety. The doses and timing of the reagents can be readily devised by a skilled artisan, and are dependent on the specific nature of the reagents employed. If a bsAb-F(ab')2 derivative is given first, then a waiting time of 1-6 days before administration of the targetable construct may be appropriate. If an IgG-Fab' bsAb conjugate is the primary targeting vector, then a longer waiting period before administration of the linker moiety may be indicated, in the range of 3-15 days. Alternatively, the bsAb and the targetable construct may be adminsistered substantially at the same time in either a cocktail form or by administering one after the other.

A wide variety of diagnostic and therapeutic reagents can be advantageously conjugated to the targetable construct. Generally, diagnostic and therapeutic agents can include isotopes, drugs, toxins, cytokines, conjugates with cytokines, hormones, growth factors, conjugates, radionuclides, contrast agents, metals, cytotoxic drugs, and immune modulators. For example, gadolinium metal is used for magnetic resonance imaging and fluorochromes can be conjugated for photodynamic therapy.
Moreover, contrast agents can be MRI contrast agents, such as gadolinium ions, lanthanum ions, manganese ions, iron, chromium, copper, cobalt, nickel, dysprosium, rhenium, europium, terbium, holmium, neodymium or other comparable label, CT contrast agents, and ultrasound contrast agents. Additional diagnotic agents can include fluorescent labeling compounds such as fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine, chemiluminescent compounds including luminol, isoluminol, an aromatic acridinium ester, an imidazole, an acridinium salt and an oxalate ester, and bioluminescent compounds including luciferin, luciferase and aequorin. Radionuclides can also be used as diagnostic andlor therapeutic agents, including for example, 9oY, 111In, 1311, 99mTC' is6Re~ iasRe~ l7~Lu, 67Cu, alzBi, 2~3Bi, and ZnAt.
Therapeutic agents also include, for example, chemotherapeutic drugs such as vinca alkaloids, anthracyclines, epidophyllotoxinw, taxanes, antimetabolites, alkylating agents, antibiotics, Cox-2 inhibitors, antimitotics, antiangiogenic and apoptotoic agents, particularly doxorubicin, methotrexate, taxol, CPT-11, camptothecans, and others from these and other classes of anticancer agents.
Other useful therapuetic agents for the preparation of immunoconjugates and antibody fusion proteins include nitrogen mustards, alkyl sulfonates, nitrosoureas, triazenes~
folic acid analogs, COX-2 inhibitors, pyrimidine analogs, purine analogs, platinum coordination complexes, hormones, and the like. Suitable therapeutic agents are described in REMINGTON'S PHARMACEUTICAL SCIENCES, 19th Ed. (Mack Publishing Co. 1995), and in GOODMAN AND GILMAN'S THE
PHARMACOLOGICAL BASIS OF THERAPEUTICS, 7th Ed. (MacMillan Publishing Co. 1985), as well as revised editions of these publications. Other suitable therapeutic agents, such as experimental drugs, are known to those of skill in the art.
Therapeutic agents may also include, without limitation, others drugs, prodrugs and/or toxins. The terms "drug," "prodrug," and "toxin" are defined throughout the specification. The terms "diagnostic agent" or "diagnosis" include, but are not limited to, detection agent, detection, or localization.
When the targetable construct includes a diagnostic agent, the bsAb is preferably administered prior to administration of the threapeutic agent.
After sufficient time has passed for the bsAb to target to the diseased tissue, the diagnostic agent is administered, by means of the targetable construct, so that imaging can be performed. Tumors can be detected in body cavities by means of directly or indirectly viewing various structures to which light of the appropriate wavelength is delivered and then collected, or even by special detectors, such as radiation probes or fluorescent detectors, and the like. Lesions at any body site can be viewed so long as nonionizing radiation can be delivered and recaptured from these structures. For example, PET
which is a high resolution, non-invasive, imaging technique can be used with the inventive antibodies and targetable constructs for the visualization of human disease. In PET, 511 keV gamma photons produced during positron annihilation decay are detected. X-ray, computed tomography (CT), MRI and gamma imaging (e.g., Single Photon Emission Computed Tomography (SPELT)) may also be utlized through use of a diagnostic agent that functions with these modalities.
As discussed earlier, the targetable construct may include radioactive diagnostic' agents that emit 25-10,000 keV gamma-, beta-, alpha- and auger-particles ' and/or positrons. Examples of such agents include, but are not limited to 18F, 52Fe, 62Cu 64Cu 67C~ 67Ga 68Ga 86y 89Zx 94m.L0 94.L0 99mTC lllln 1231 1241 125I 131I

0 > > > > > > s ~ o > > > o s lssGd and 175Lu.
The present bsAbs or bsFabs can be used in a method of photodynamic therapy (PDT) as discussed in U.S. Patent Nos. 6,096,289; 4,331,647;
4,818,709;
4,348,376; 4,361,544; 4,444,744; 5,851,527. In PDT, a photosensitizer, e.g., a hernatoporphyrin derivative such as dihematoporphyrin ether, is administered to a subject. Anti-tumor activity is initiated by the use of light, e.g., 630 nm.
Alternate photosensitizers can be utilized, including those useful at longer wavelengths, where skin is less photosensitized by the sun. Examples of such photosensitizers include, but are not limited to, benzoporphyrin monoacid ring A (BPD-MA), tin etiopurpurin (SnET2), sulfonated aluminum phthalocyanine (AlSPc) and lutetium texaphyrin (Lutex).
Additionally, in PDT, a diagnostic agent may be injected, for example, systemically, and laser-induced fluorescence can be used by endoscopes to detect sites' of cancer which have accreted the light-activated agent. For example, this has been applied to fluorescence bronchoscopic disclosure of early lung tumors. Doiron et al.
Chest 76:32 (1979). In another example, the antibodies and antibody fragments can be used in single photon emission. For example, a Tc-99m-labeled diagnostic agent can be administered to a subject following administration of the inventive antibodies or antibody fragments. The subject is then scanned with a gamma camera which produces single-photon emission computed tomographic images and defines the lesion or tumox site.
Therapeutically useful immunoconjugates can be obtained by conjugating photoactive agents or dyes to an antibody composite. Fluorescent and other chromogens, or dyes, such as porphyrins sensitive to visible light, have been used to detect and to treat lesions by directing the suitable light to the lesion. In therapy, this has been termed photoradiation, phototherapy, or photodynamic therapy (Jori et al.
(eds.), Photodynarnic Therapy of Tumors and Other Diseases (Libreria Progetto 1985); van den Bergh, Chern. Britain 22:430 (1986)). Moreover, monoclonal antibodies have been coupled with photoactivated dyes for achieving phototherapy.
Mew et al., J. I»arnuraol. 130:1473 (1983); idem., Ca»cer Res. 45:4380 (1985);
Oseroff et al., Proc. Natl. Acad. Sci. USA 83:8744 (1986); ide»a., Photoclaem.
Photobiol.
46:83 (1987); Hasan et al., Prog. Cli». Biol. Res. 288:471 (1989); Tatsuta et al., Lasers Surg. Med. 9:422 (1989); Pelegrin et al., Cancer 67:2529 (1991).
However, these earlier studies did not include use of endoscopic therapy applications, especially with the use of antibody fragments or subfragments. Thus, the present invention contemplates the therapeutic use of immunoconjugates comprising photoactive agents or dyes.
Radiopaque and contrast materials are used for enhancing X-rays and computed tomography, and include iodine compounds, barium compounds, gallium compounds, thallium compounds, etc. Specific compounds include barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric acid, iosulamide meglumine, iosemetic acid, iotasul, iotetric acid, iothalamic acid, iotroxic acid, ioxaglic acid, ioxotrizoic acid, ipodate, meglumine, metrizamide, metrizoate, propyliodone, and thallous chloride.
Ultasound contrast material may also by used inlcuding dextran and liposomes, particularily gas-filled liposomes.In one embodiment, an immunomodulator, such as a cytokine, may also be conjugated to the therapeutic constrict. As used herein, the term "immunomodulator" includes cytokines, stem cell growth factors, lymphotoxins, such as tumor necrosis factor (TNF), and hematopoietic factors, such as interleukins (e.g., interleukin-1 (IL-1), IL-2, IL-3, IL-6, IL-10, IL-12 and IL-18), colony stimulating factors (e.g., granulocyte-colony stimulating factor (G-CSF) and granulocyte macrophage-colony stimulating factor (GM-CSF)), interferons (e.g., interferons-a, -(3 and -y), the stem cell growth factor designated "S 1 factor" erythropoietin and thrombopoietin. Examples of suitable immunomodulator moieties include IL-2, IL-6, IL-10, IL-12, IL-18, interferon-y, TNF-a, and the like.
The targetable construct may also be conjugated to an enzyme capable of activating a drug/prodrug at the target site or improving the efficacy of a noimal therapeutic by controlling the body's detoxification pathways. Following administration of the bsAb, an enzyme conjugated to the targetable construct having a low MW hapten is administered. After the enzyme is pretargeted to the target site by bsAbaargetable construct binding, a cytotoxic drug is injected that is known to act at the target site. The drug may be one which is detoxified by the mammal's ordinary detoxification processes to form an intermediate of lower toxicity. For example, the drug may be converted into the potentially less toxic glucuronide in the liver. The detoxified intermediate can then be reconverted to its more toxic fornl by the pretargeted enzyme at the target site, and this enhances cytotoxicity at the target site.
Alternatively, an administered prodrug can be converted to an active drug by the pretargeted enzyme. The pretargeted enzyme improves the efficacy of the treatment by recycling the detoxified drug. This approach can be adopted for use with any enzyme-drug pair. Alternatively, the targetable construct with enzyme can be mixed with the targeting bsAb prior to administration to the patient. After a sufficient time has passed for the bsAbaargetable construct- conjugate to localize to the target site and for unbound targetable construct to clear from circulation, a prodrug is administered. As discussed above, the prodrug is then converted to the drug in situ by the pre-targeted enzyme.
Certain cytotoxic drugs that are useful for anticancer therapy are relatively insoluble in serum. Some are also quite toxic in an unconjugated form, and their toxicity is considerably reduced by conversion to prodrugs. Conversion of a poorly soluble drug to a more soluble conjugate, e.g., a glucuronide, an ester of a hydrophilic acid or an amide of a hydrophilic amine, will improve its solubility in the aqueous phase of serum and its ability to pass through venous, arterial or capillary cell walls and to reach the interstitial fluid bathing the tumor. Cleavage of the prodrug deposits the less soluble drug at the target site. Many examples of such prodrug-to-drug conversions are disclosed in U.S. Patent No. 5,851,527, to Hansen.
Conversion of certain toxic substances such as aromatic or alicyclic alcohols, thiols, phenols and amines to glucuronides in the liver is the body's method of detoxifying them and making them more easily excreted in the urine. One type of antitumor drug that can be converted to such a substrate is epirubicin, a 4-epimer of doxorubicin (Adriamycin), which is an anthracycline glycoside and has been shown to be a substrate for human beta-D-glucuronidase See, e.g., Arcamone Cancer Res.
45:5995 (1985). Other analogues with fewer polar groups are expected to be more lipophilic and show greater promise for such an approach. Other drugs or toxins with aromatic or alicyclic alcohol, thiol or amine groups are candidates for such conjugate formation. These drugs, or other prodrug forms thereof, are suitable candidates for the site-specific enhancement methods of the present invention.
The prodrug CPT- I,1 (irinotecan) is converted in vivo by carboxylesterase to the active metabolite SN-38. One application of the invention, therefore, is to use a bsAb targeted against a tumor and a hapten (e.g. di-DTPA) followed by injection of a di-DTPA-carboxylesterase conjugate. Once a suitable tumor-to-background localization ratio has been achieved, the CPT-11 is given and the tumor-localized carboxylesterase serves to convert CPT-1 I to SN-38 at the tumor. Due to its poor solubility, the active SN-38 will remain in the vicinity of the tumor and, consequently, will exert an effect on adjacent tumor cells that are negative for the antigen being targeted. This is a further advantage of the method. Modified forms of carboxylesterases have been described and are within the scope of the invention. See, e.g., Potter et al., Cancer Res. 58:2646-2651 (1998) and Potter et al., Cancer Res.
58:3627-3632 (1998).
Etoposide is a widely used cancer drug that is detoxified to a major extent by formation of its glucuronide and is within the scope of the invention. See, e.g., Hande et al. Cancer Res. 48:1829-1834 (1988). Glucuronide conjugates can be prepared from cytotoxic drugs and can be injected as therapeutics for tumors pre-targeted with mAb-glucuronidase conjugates. See, e.g., Wang et al. Cancer Res. 52:4484-4491 ,, (1992). Accordingly, such conjugates also can be'used with the pre-targeting approach described here. Similarly, designed prodrugs based on derivatives of daunomycin and doxorubicin have been described for use with carboxylesterases and glucuronidases. See, e.g., Bakina et al. J. Med Chern. 40:4013-4018 (1997).
Other examples of prodrug/enzyme pairs that can be used within the present invention include, but are not limited to, glucuxonide prodrugs of hydroxy derivatives of phenol mustards and beta-glucuronidase; phenol mustards or CPT-11 and carboxypeptidase;
methotrexate-substituted alpha-amino acids and carboxypeptidase A; penicillin or cephalosporin conjugates of drugs such as 6-mercaptopurine and doxorubicin and beta-lactamase; etoposide phosphate and alkaline phosphatase.
The enzyme capable of activating a prodrug at the target site or improving the efficacy of a normal therapeutic by controlling the body's detoxification pathways may aternatively be conjugated to the hapten. The enzyme-hapten conjugate is administered to the subject following administration of the pre-targeting bsAb and is directed to the target site. After the enzyme is localized at the target site, a cytotoxic drug is injected, which is known to act at the target site, or a prodrug form thereof which is converted to the drug ira situ by the pretargeted enzyme. As discussed above, the drug is one which is detoxified to form an intermediate of lower toxicity, most commonly a glucuronide, using the mammal's ordinary detoxification processes.
The detoxified intermediate, e.g., the glucuronide, is reconverted to its more toxic form by the pretargeted enzyme and thus has enhanced cytotoxicity at the target site.
This results in a recycling of the drug. Similarly, an administered prodrug can be converted to an active drug through normal biological processess. The pretargeted enzyme improves the efficacy of the treatment by recycling the detoxified drug. This approach can be adopted for use with any enzyme-drug pair.
In an alternative embodiment, the enzyme-hapten conjugate can be mixed with the targeting bsAb prior to administration to the patient. After a sufficient time has passed for the enzyme-hapten-bsAb conjugate to localize to the target site and for unbound conjugate to clear from circulation, a prodrug is administered. As discussed above, the prodrug is then converted to the drug in situ by the pre-targeted enzyme.
The invention further contemplates the use of the inventive bsAb and the diagnostic agents) in the context of Boron Neutron Capture Therapy (BNCT) protocols. BNCT is a binary system designed to deliver ionizing radiation to tumor cells by neutron irradiation of tumor-localized 1°B atoms. BNCT is based on the nuclear reaction which occurs when a stable isotope, isotopically enriched I°B (present in 19.8% natural abundance), is irradiated with thermal neutrons to produce an alpha particle and a 7Li nucleus. These particles have a path length of about one cell 15' diameter, resulting in high linear energy transfer. Just a few of the short-range 1.7 MeV alpha particles produced in this nuclear reaction are sufficient to target the cell nucleus and destroy it. Success with BNCT of cancer requires methods for localizing a high concentration of I°B at tumor sites, while leaving non-target organs essentially boron-free. Compositions and methods for treating tumors in subjects using pre-targeting bsAb for BNCT are described in U.S. Patent No. 6,228,362 and can easily be modified for the purposes of the present invention.
In another embodiment of the present invention, the peptide backbone of the targetable construct is conjugated to a prodrug. The pre-targeting bsAb is administered to the patient and allowed to localize to the target and substantially clear circulation. At an appropriate later time, a targetable construct comprising a prodrug, for example poly-glutamic acid (SN-38-ester)1°, is given, thereby localizing the prodrug specifically at the tumor target. It is known that tumors have increased amounts of enzymes released from intracellular sources due to the high rate of lysis of cells within and around tumors. A practitioner can capitalize on this fact by appropriately selecting prodrugs capable of being activated by these enzymes.
For example, carboxylesterase activates the prodrug poly-glutamic acid (SN-38-ester)1° by cleaving the ester bond of the poly glutamic acid (SN-38-ester)I°
releasing large concentrations of free SN-38 at the tumor. Alternatively, the appropriate enzyme also can be targeted to the tumor site.
After cleavage from the targetable construct, the drug is internalized by the tumor cells. Alternatively, the drug can be internalized as part of an intact complex by virtue of cross-linking at the target. The targetable construct can induce internalization of tumor-bound bsAb and thereby improve the efficacy of the treatment by causing higher levels of the drug to be internalized.
A variety of peptide carriers are well-suited for conjugation to prodrugs, including polyamino acids, such as polylysine, polyglutamic (E) and aspartic acids (D), including D-amino acid analogs of the same, co-polymers, such as poly(Lys-Glu) f poly[KE]~, advantageously from 1:10 to 10:1. Copolymers based on amino acid mixtures such as poly(Lys-Ala-Glu-Tyr) (KAEY; 5:6:2:1) can also be employed.
Smaller polymeric carriers of defined molecular weight can be produced by solid-phase peptide synthesis techniques, readily producing polypeptides of from 2-SO
residues in chain length. A second advantage of this type of reagent, other than precise structural definition, is the ability to place single or any desired number of chemical handles at certain points in the chain. These can be used later for attachment of recognition and therapeutic haptens at chosen levels of each moiety.
Polyethylene) glycol [PEG] has desirable in vivo properties for a bi-specific antibody prodrug approach. Ester linkages between the hydroxyl group of SN-38 and both ends of a standard di-hydroxyl PEG can be introduced by insertion of diacids such as succinic acid between the SN-38 and PEG hydroxyl groups, to generate species such as SN-38-O-CO(CH2)ZCO-O-PEG-0-CO(CH2)2C0-OSN-38. The di-SN-38-PEG produced can be considered as the shortest member of the class of SN-polymer prodrugs. The desirable in vivo properties of PEG derivatives and the limited loading capacity due to their dimeric functionality led to the preparation of PEG co-polymers having greater hapten-bearing capacity such as those described by Poiani et al. See, e.g., Poiani et al. BiocoT jugate Claern., 5:621-630, 1994. PEG
derivatives are activated at both ends as their bis(succinimidyl)carbonate derivatives and co-polymerized with mufti-functional diamines such as lysine. The product of such co-polymerization, containing (-Lys(COOH)-PEG-Lys(COOH)-PEG-)n repeat units wherein the lysyl carboxyl group is not involved in the polymerization process, can be used for attachment of SN-38 residues. The SN-38 residues are reacted with the free carboxyl groups to produce SN-38 esters of the (-Lys-(COOH)-PEG-Lys(COOH)-PEG-)" chain.
Other synthetic polymers that can be used to carry recognition haptens and prodrugs include N-(2-hydroxypropyl)methacrylamide (HMPA) copolymers, polystyrene-co-malefic acid/anhydride (SMA), poly(divinylether malefic anhydride) (DIVEMA), polyethyleneimine, ethoxylated polyethylene-imine,'starburst dendrimers and poly(N-vinylpyrrolidone) (PVP). As an example, DIVEMA polymer comprised of multiple anhydride units is reacted with a limited amount of SN-38 to produce a desired substitution ratio of drug on the polymer backbone. Remaining anhydride groups are opened under aqueous conditions to produce free carboxylate groups.
A
limited number of the free carboxylate groups are activated using standard water-soluble peptide coupling agents, e.g. 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC), and coupled to a recognition moiety bearing a free amino group.
An example of the latter is histamine, to which antibodies have been raised in the past.
A variety of prodrugs can be conjugated to the targetable construct. The above exemplifications of polymer use are concerned with SN-38, the active metabolite of the prodrug CPT-11 (irinotecan). SN-38 has an aromatic hydroxyl group that was used in the above descriptions to produce aryl esters susceptible to esterase-type enzymes. Similarly the camptothecin analog topotecari, widely used in chemotherapy, has an available aromatic hydroxyl residue that can be used in a similar manner as described for SN-38, producing esterase-susceptible polymer-prodrugs.
Doxorubicin also contains aromatic hydroxyl groups that can be coupled to carboxylate-containing polymeric carriers using acid-catalyzed reactions similar to those described for the camptothecin family. Similarly, doxorubicin analogs like daunomycin, epirubicin and idarubicin can be coupled in the same manner.
Doxorubicin and other drugs with amino 'chemical handles' active enough for chemical coupling to polymeric carriers can be effectively coupled to carrier molecules via these free amino groups in a number of ways. Polymers bearing free carboxylate groups can be activated ira situ (EDC) and the activated polymers mixed with doxorubicin to directly attach the drug to the side-chains of the polymer via amide bonds. Amino-containing drugs can also be coupled to amino-pendant polymers by mixing commercially available and cleavable cross-linking agents, such as ethylene glycobis(succinimidylsuccinate) (EGS,.Pierce Chemical Co., Rockford, IL) or bis-[2-(succinimido-oxycarbonyloxy)ethyl]sulfone (BSOCOES, Molecular Biosciences, Huntsville, AL), to cross-link the two amines as two amides after reaction with the bis(succinimidyl) ester groups. This is advantageous as these groups remain susceptible to enzymatic cleavage. For example, (doxorubicin-EGS)~ poly-lysine remains susceptible to enzymatic cleavage of the diester groups in the EGS
linking chain by enzymes such as esterases. Doxorubicin also can be conjugated to a variety of peptides, for example, HyBnK(DTPA)YK(DTPA)-NH2, using established procedures (HyBn= p-H2NNHC6H4C02H). See Kaneko et al., J. Bioconjugate Chem., 2: 133-141, 1991.
In one preferred embodiment, the therapeutic conjugate comprises doxorubicin coupled to a carrier comprising amine residues and a chelating agent, such as DTPA, to form a DTPA-peptide-doxorubicin conjugate, wherein the DTPA forms the .
recognition moiety for a pretargeted bsAb. In some emboidments, the Garner comprises a tyrosyl-lysine dipeptide, e.g., D-Tyr-D-Lys(DTPA)-NH2, and can comprise D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-NHZ. Doxorubicin phenyl hydrazone conjugates to bis-DPTA containing peptides are particularly desirable in a therapeutic context.
Methotrexate also has an available amino group for coupling to activated carboxylate-containing polymers, in a similar manner to that described for doxorubicin. It also has two glutamyl carboxyl groups (alpha and gamma) that can be activated for coupling to amino-group containing polymers. The free carboxylate groups of methotrexate can be activated in situ (EDC) and the activated drug mixed with an amino-containing polymer to directly attach the drug to the side-chains of the polymer via amide bonds. Excess unreacted or cross-reacted drug is separated readily from the polymer-drug conjugate using size-exclusion or ion-exchange chromatography.
Maytansinoids and calicheamicins (such as esperamycin) contain mixed di-and tri-sulfide bonds that can be cleaved to generate species with a single thiol useful for chemical manipulation. The thiomaytensinoid or thioespera-mycin is first reacted with a cross-linking agent such as a maleimido-peptide that is susceptible to cleavage by peptidases. The C-terminus of the peptide is then activated and coupled to an amino-containing polymer such as polylysine.
In still other embodiments, the bi-specific antibody-directed delivery of therapeutics or prodrug polymers to in vivo targets can be combined with bi-specific antibody delivery of radionuclides, such that combination chemotherapy and radioimmunotherapy is achieved. Each therapy can be conjugated to the targetable construct and administered simultaneously, or the nuclide can be given as part of a first targetable construct and the drug given in a later step as part of a second targetable construct. In one simple embodiment, a peptide containing a single prodrug and a single nuclide is constructed. For example, the tripeptide Ac-D-Glu-D-Gly-D-Lys-NH2 can be used as a carrier portion of a targetable construct, whereby SN-38 is attached to the gamma glutamyl carboxyl group as an aryl ester, while the chelate DOTA is attached to the epsilon amino group as an amide, to produce the complex Ac-D-Glu(SN-38)-D-Gly-D-Lys(DOTA)-NHZ. The DOTA chelate can then be radiolabeled with various metals for imaging and therapy purposes including In-11 l, Y-90, Sm-153, Lu-177 and Zr-89. As the metal-DOTA complex may represent the recognizable hapten on the targetable construct, the only requirement for the metal used as part of the DOTA complex is that the secondary recognition antibody also used recognizes that particular metal-DOTA complex at a sufficiently high affinity.
Generally, this affinity (log I~) is between 6-11. Polymeric peptides such as poly[D-Glu(SN-38)IO-D-Lys(Y-90-DOTA)2] can be given as readily as the more chemically defined lower MW reagent above, and are indeed preferred. Also, triply substituted polymers can be used, such as poly[D-Glu(Sn-38)lo-D-Lys(Y-90-DOTA)"(histamine-succinate)m, where n and m are integers, such that the recognition agent is independent of the radioimmunotherapy agent. The prodrug is activated by carboxylesterases present at the tumor site or by carboxylesterases targeted to the site using a second targetable construct.
Alternatively, a combination therapy can be achieved by administering the chemotherapy and radioimmunotherapy agents in separate steps. For example, a patient expressing CEA-tumors is first administered bsAb with at least one arm which specifically binds CEA and at least one other arm which specifically binds the targetable construct whose hapten is a conjugate of yttrium-DOTA. Later the patient is treated with a targetable construct comprising a conjugate of yttrium-DOTA-beta-glucuronidase. After sufficient time for bsAb and enzyme localization and clearance, a second targetable construct, comprising Ac-D-Glu(SN-38)-D-Gly-D-Lys(Y-90-DOTA)-NHZ, is given. The second targetable construct localizes to the tumor by virtue of bsAb at the tumor that are not already bound to a ftrst targetable construct.
First targetable constructs which are localized to the target site act on the Ac-D-Glu(SN-38)-D-Gly-D-Lys(Y-90-DOTA)-NHZ to liberate the free SN-38 drug.
Localization of both the prodrug and its respective enzyme to the target site enhances the production of active drug by ensuring that the enzyme is not substrate limited.
This embodiment constitutes a marked improvement of current prodrug methodologies currently practiced in the art.
Another advantage of administering the prodrug-polymer in a later step, after the nuclide has been delivered as part of a previously given targetable construct, is that the synergistic effects of radiation and drug therapy can be manipulated and, therefore, maximized. It is hypothesized that tumors become more 'leaky' after BAIT
due to radiation damage. This can allow a polymer-prodrug to enter a tumor more completely and deeply. This results in improved chemotherapy.
Alternatively, the BAIT therapy agent can be attached to bsAb rather than to the targetable construct. For example, an anti-CEA x anti-DTPA bsAb conjugated to Y-90-DOTA is administered first to a patient with CEA-expressing tumors. In this instance, advantage is taken of the selectivity of certain anti-chelate mabs in that an anti-indium-DTPA antibody do not bind to a yttrium-DOTA chelate. After the Y-DOTA-anti-CEA x anti-indium-DTPA has maximized at the tumor and substantially cleared non-target tissue, a conjugate of indium-DTPA-glucuronidase is injected and localized specifically to the CEA tumor sites. The patient is then injected with a polymer-prodrug such as poly(Glu)(SN-38)10. The latter is cleaved selectively at the tumor to active monomeric SN-38, successfully combining chemotherapy with the previously administered BAIT.
It should also be noted that a bispecific antibody or antibody fragment can be used in the present method, with at least one binding site specific to an antigen at a target site and at least one other binding site specific to the enzyme component of the antibody-enzyme conjugate. Such an antibody can bind the enzyme prior to injection, thereby obviating the need to covalently conjugate the enzyme to the antibody, or it can be injected and localized at the target site and, after non-targeted antibody has substantially cleared from the circulatory system of the mammal, enzyme can be injected in an amount and by a route which enables a sufficient amount of the enzyme to reach a localized antibody or antibody fragment and bind to it to form the antibody-enzyme conjugate in situ.
It should also be noted that the invention also contemplates the use of multivalent target binding proteins which have at least three different target binding sites as described in Patent Appl. Serial No. 60/220,782. Multivalent target binding proteins have been made by cross-linking several Fab-like fragments via chemical linkers. See U.S. Patent Nos. 5,262,524; 5,091,542 and Landsdorp et al., Euro.
J.
Immunol. 16: 679-83 (1986). Multivalent target binding proteins also have been made by covalently linking several single chain Fv molecules (scFv) to form a single polypeptide. See U.S. Patent No. 5,892,020. A multivalent target binding protein which is basically an aggregate of scFv molecules has been disclosed in U.S.
Patent Nos. 6,025,165 and 5,837,242. A trivalent target binding protein comprising three scFv molecules has been described in Krott et al., Protein Eyagineerirag 10(4): 423-433 (1997).
A clearing agent may be used which is given between doses of the bsAb and the targetable construct. The present inventors have discovered that a clearing agent of novel mechanistic action may be used with the invention, namely a glycosylated anti-idiotypic Fab' fragment targeted against the disease targeting arms) of the bsAb.
Anti-CEA (MN-14 Ab) x anti-peptide bsAb is given and allowed to accrete in disease targets to its maximum extent. To clear residual bsAb, an anti-idiotypic Ab to MN-14, termed WI2, is given, preferably as a glycosylated Fab' fragment. The clearing agent binds to the bsAb in a monovalent manner, while its appended glycosyl residues direct the entire complex to the liver, where rapid metabolism takes place.
Then the therapeutic or diagnostic agent which is associated with the targetable construct is given to the subject. The WI2 Ab to the MN-14 arm of the bsAb has a high affinity and the clearance mechanism differs from other disclosed mechanisms (see Goodwin et al., ibic~, as it does not involve cross-linking, because the WI2-Fab' is a monovalent moiety.

In accordance with yet another aspect of the present invention, the present invention provides a kit suitable for treating or identifying diseased tissues in a patient, comprising a bi-specific antibody or antibody fragment having at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds a targetable construct, a first targetable construct which comprises a Garner portion which comprises or bears at least one epitope recognizable by the at least one other arm of the bi-specific antibody or antibody fragment, and one or more conjugated therapeutic or diagnostic agents, or enzymes, and, optionally, a clearing composition useful for clearing non-localized antibodies and antibody fragments. The kit may optionally contain a prodrug when the .first targetable construct comprises an enzyme capable of converting the prodrug to a drug at the target site, an enzyme that is capable of reconverting a detoxified intermediate of a drug to a toxic form, and, therefore, of increasing the toxicity of the drug at the target site, or an enzyme capable of reconverting a prodrug which is activated in the patient through natural processes and is subject to detoxification by conversion to an intermediate of lower toxicity from the detoxified intermediate to a toxic form, and, therefore, of increasing the toxicity of the drug at the target site. A second targetable construct may also be used which comprises a carrier portion which comprises or bears at least one epitope recognizable by the at least one other arm of the bi-specific antibody or antibody fragment, and a prodrug, when the enzyme is capable of converting the prodrug to a drug at the target site. Instruments which facilitate identifying or treating diseased tissue also can be included in the kit. Examples include, but are not limited to application devices, such as syringes. Solutions required for utilizing the disclosed invention for identifying or treating diseased tissue also can be included in the kit.
The targetable construct or antibody fragment may be administered intravenously, intraarterially, intraoperatively, endoscopically, intraperitoneally, intramuscularly, subcutaneously, intrapleurally, intrathecally, by perfusion through a regional catheter, or by direct intralesional injection, and can be by continuous infusion or by single or multiple boluses or through other methods known to those skilled in the art for diagnosing (detecting) and treating diseased tissue.
Further, the targetable construct may include agents for other methods of detecting and treating diseased tissue inlcuding, without limitation, conjugating dextran or liposome formulations to the targetable construct for use with ultrasound, or other contrast agents for use with other imaging modalities, such as X-ray, CT, PET, SPECT
and ultrasound, as previously described.
VI. Methods for Raising Antibodies Abs to peptide backbones and/or haptens are generated by well-known methods for Ab production. For example, injection of an immunogen, such as (peptide)n-KLH, wherein KLH is keyhole limpet hemocyanin, and n=1-30, in complete Freund's adjuvant, followed by two subsequent injections of the same immunogen suspended in incomplete Freund's adjuvant into immunocompetent animals, is followed three days after an i.v. boost of antigen, by spleen cell harvesting.
Harvested spleen cells are then fused with Sp2/0-Agl4 myeloma cells and culture supernatants of the resulting clones analyzed for anti-peptide reactivity using a direct-binding ELISA. Fine specificity of generated Abs can be analyzed for by using peptide fragments of the original immunogen. These fragments can be prepared readily using an automated peptide synthesizer. For Ab production, enzyme-deficient hybridomas are isolated to enable selection of fused cell lines. This technique also can be used to 'raise antibodies to one or more of the chelates comprising the linker, e.g., In(III]-DTPA chelates. Monoclonal mouse antibodies to an In(III)-di-DTPA
are known (Barbet '395 supra).
The antibodies used in the present invention are specific to a variety of cell surface or intracellular tumor-associated antigens as marker substances. ~
These markers may be substances produced by the tumor or may be substances which accumulate at a tumor site, on tumor cell surfaces or within tumor cells, whether in the cytoplasm, the nucleus or in various organelles or sub-cellular structures. Among such tumor-associated markers are those disclosed by Herberman, "Irnmunodiagnosis of Cancer", in Fleisher ed., "The Clinical Biochemistry of Cancer", page 347 (American Association of Clinical Chemists, 1979) and in U.S. Patent Nos.
4,150,149; 4,361,544; and 4,444,744. See also U.S. Patent No. 5,965,132, to Thorpe et al., U.S. Patent 6,004;554, to Thorpe et al., U.S. Patent No. 6,071,491, to Epstein et al., U.S. Patent No. 6,017,514, to Epstein et al., U.S. Patent No. 5,882,626, to Epstein et al., U.S. Patent No. 5,019,368, to Epstein et al., and U.S. Patent No.
6,342,221, to Thorpe et al., all of which are incorporated herein by reference.

Tumor-associated markers have been categorized by Herberman, supra, in a number of categories including oncofetal antigens, placental antigens, oncogenic or tumor virus associated antigens, tissue associated antigens, organ associated antigens, ectopic hormones and normal antigens or variants thereof. Occasionally, a sub-unit of a tumor-associated marker is advantageously used to raise antibodies having higher tumor-specificity, e.g., the beta-subunit of human chorionic gonadotropin (HCG) or the gamma region of carcino embryonic antigen (CEA), which stimulate the production of antibodies having a greatly reduced cross-reactivity to non-tumor substances as disclosed in U.S. Patent Nos. 4,361,644 and 4,444,744. Markers of tumor vasculature (e.g., VEGF), of tumor necrosis (Epstein patents), of membrane receptors (e.g., folate receptor, EGFR), of transmembrane antigens (e.g., PSMA), and of oncogene products can also serve as suitable tumor-associated targets for antibodies or antibody fragments. Markers of normal cell constitutents which are expressed copiously on tumor cells, such as B-cell complex antigens (e.g., CD19, CD20, CD21, CD22, CD23, and HLA-DR on B-cell malignancies), as well as cytokines expressed by certain tumor cells (e.g., IL-2 receptor in T-cell malignancies) are also suitable targets for the antibodies and antibody fragments of this invention. Other well-known tumor associated antigens that can be targeted by the antibodies and antibody fragments of this invention include, but are not limited to, CEA, CSAp, TAG-72, MUC-1, MUC-2, MUC-3, MUC-4, EGP-1, EGP-2, BrE3, PAM-4, KC-4, A3, KS-1, PSMA, PSA, tenascin, T101, 5100, MAGE, HLA-DR, CD19, CD20, CD22, CD30, and CD74.
Another marker of interest is transmembrane activator and CAML-interactor (TACI). See Yu et al. Nat. Irnnaunol. 1:252-256 (2000). Briefly, TACI is a marker for B-cell malignancies (e.g., lymphoma). Further it is known that TACI and B-cell maturation antigen (BCMA) are bound by the tumor necrosis factor homolog a proliferation-inducing ligand (APRIL). APRIL stimulates in vitro proliferation of primary B and T cells and increases spleen weight due to accumulation of B
cells in vivo. APRIL, also competes with TALL-I (also called BLyS or BAFF) for receptor binding. Soluble BCMA and TACI specifically prevent binding of APRIL and block APRIL-stimulated proliferation of primary B cells. BCMA-Fc also inhibits production of antibodies against keyhole limpet hemocyanin and Pneumovax in mice, indicating that APRIL and/or TALL-I signaling via BCMA and/or TACI are required for generation of humoral immunity. Thus, APRIL-TALL-I and BCMA-TACI form a two ligand-two receptor pathway involved in stimulation of B and T cell function.
After the initial raising of antibodies to the immunogen, the antibodies can be sequenced and subsequently prepared by recombinant techniques. Humanization and chimerization of murine antibodies and antibody fragments are well known to those skilled in the art. For example, humanized monoclonal antibodies are produced by transfernng mouse complementary determining regions from heavy and light variable chains of the mouse immunoglobulin into a human variable domain, and then, substituting human residues in the framework regions of the murine counterparts. The use of antibody components derived from humanized monoclonal antibodies obviates potential problems associated with the immunogenicity of murine constant regions.
General techniques for cloning murine immunoglobulin variable domains are described, for example, by the publication of Orlandi et al., Proc. Nat'l Acad. Sci. USA
86: 3833 (1989), which is incorporated by reference in its entirety. Techniques for producing humanized Mabs are described, for example, by Jones et al., Natu»e 321: 522 (1986), Riechmann et al., Natu»e 332: 323 (1988), Verhoeyen et al., Scie»ce 239: 1534 (1988), Carter et al., Proc. Nat'l Acad. Sci. USA 89: 4285 (1992), Sandhu, CYit. Rev.
Biotecla.
12: 437 (1992), and Singer et al., J. Isn»aun. 150: 2844 (1993), each of which is hereby incorporated by reference.
Alternatively, fully human antibodies can be obtained from transgenic non-human animals. See, e.g., Mendez et al., Natu»e Genetics, 15: 146-156 (1997);
U.S.
Patent No. 5,633,425. For example, human antibodies can be recovered from transgenic mice possessing human immunoglobulin loci. The mouse humoral immune system is humanized by inactivating the endogenous immunoglobulin genes and introducing human immunoglobulin loci. The human immunoglobulin loci are exceedingly complex and comprise a large number of discrete segments which together occupy almost 0.2% of the human genome. To ensure that transgenic mice are capable of producing adequate repertoires of antibodies, large portions of human heavy- and light-chain loci must be introduced into the mouse genome. This is accomplished in a stepwise process beginning with the formation of yeast artificial chromosomes (YACs) containing either human heavy- or light-chain immunoglobulin loci in germline configuration: Since each insert is approximately 1 Mb in size, YAC
construction requires homologous recombination of overlapping fragments of the immunoglobulin loci. The two YACs, one containing the heavy-chain loci and one containing the light-chain loci, are introduced separately into mice via fusion of YAC-containing yeast spheroblasts with mouse embryonic stem cells. Embryonic stem cell clones are then microinjected into mouse blastocysts. Resulting chimeric males are screened for their ability to transmit the YAC through their germline and are bred with mice deficient in muxine antibody production. Breeding the two transgenic strains, one containing the human heavy-chain loci and the other containing the human light-chain loci, creates progeny which produce human antibodies in response to immunization.
Unrearranged human imrnunoglobulin genes also can be introduced into mouse embryonic stem cells via microcell-mediated chromosome transfer (MMCT).
See, e.g., Tomizuka et al., Nature Genetics, 16: 133 (1997). In this methodology microcells containing human chromosomes are fused with mouse embryonic stem cells. Transferred chromosomes are stably retained, and adult chimeras exhibit proper tissue-specific expression.
As an alternative, an antibody or antibody fragment of the present invention may be derived from human antibody fragments isolated from a combinatorial immunoglobulin library. See, e.g., Barbas et al., METHODS: A Companion to Methods in Enzymology 2: 119 (1991), and Winter et al., Ann. Rev. Imnamiol.
12: 433 (1994), which are incorporated by reference. Many of the difficulties associated with generating monoclonal antibodies by B-cell immortalization can be overcome by engineering and expressing antibody fragments in'E. coli, using phage display.
To 'ensure the recovery of high affinity, monoclonal antibodies a combinatorial immunoglobulin library must contain a large repertoire size. A typical strategy utilizes mRNA obtained from lymphocytes or spleen cells of immunized mice to synthesize cDNA using reverse transcriptase. The heavy- and light-chain genes axe amplified separately by PCR and ligated into phage cloning vectors. Two different libraries are produced, one containing the heavy-chain genes and one containing the light-chain genes. Phage DNA is islolated from each library, and the heavy-and light-chain sequences are ligated together and packaged to form a combinatorial library.

Each phage contains a random pair of heavy- and light-chain cDNAs and upon infection of E. coli directs the expression of the antibody chains in infected cells. To identify an antibody that recognizes the antigen of interest, the phage library is plated, and the antibody molecules present in the plaques are transferred to filters.
The filters are incubated with radioactively labeled antigen and then washed to remove excess unbound ligand. A radioactive spot on the autoradiogram identifies a plaque that contains an antibody that binds the antigen. Cloning and expression vectors that are useful for producing a human immunoglobulin phage library can be obtained, for example, from STRATAGENE Cloning Systems (La Jolla, CA).
A similar strategy can be employed to obtain high-affinity scFv. See, e.g., Vaughn et al., Nat. Biotechnol., 14: 309-314 (1996). An scFv library with a large repertoire can be constructed by isolating V-genes from non-immunized human donors using PCR primers corresponding to all known VHF VK and V~, gene families.
Following amplification, the VK and V~, pools are combined to form one pool.
These fragments are ligated into a phagemid vector. The scFv linker, (Gly4, Ser)3, is then ligated into the phagemid upstream of the VL fragment. The VH and linker-VL
fragments are amplified and assembled on the JH region. The resulting VH-linker-VL
fragments are ligated into a phagemid vector. The phagemid library can be panned using filters, as described above, or using immunotubes (Nunc; Maxisorp).
Similar results can be achieved by constructing a combinatorial immunoglobulin library from lymphocytes or spleen cells of immunized rabbits and by expressing the scFv constructs in P. pastoris. See, e.g., Ridder et al., Biotechnology, 13: 255-260 (1995).
Additionally, following isolation of an appropriate scFv, antibody fragments with higher binding affinities and slower dissociation rates can be obtained through affinity maturation processes such as CDR3 mutagenesis and chain shuffling. See, e.g., Jackson et al., Br. J. Cancer, 78: 181-188 (1998); Osbourn et al., Irnnauraotechraology, 2: 181-196 (1996).
Another form of an antibody fragment is a peptide coding for a single CDR.
CDR peptides ("minimal recognition units") can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells. See, for example, Larnck et al., Methods: A
Companion to Methods ira Enzyrrtology x:106 (1991); Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING AND CLINICAL APPLICATION, Ritter et al. (eds.), pages 166-179 (Cambridge University Press 1995); and Ward et al., "Genetic Manipulation and Expression of Antibodies," in MONOCLONAL ANTIBODIES: PRINCIPLES AND
APPLICATIONS, Birch et al., (eds.), pages 137-185 (Wiley-Liss, Inc. 1995).
The bsAbs can be prepared by techniques known in the art, for example, an anti-CEA tumor Ab and an anti-peptide Ab ark both separately digested with pepsin to their respective F(ab')ZS. The anti-CEA-Ab-F(ab')2 is reduced with cysteine to generate Fab' monomeric units which are further reacted with the cross-linker bis(maleimido) hexane to produce Fab'-maleimide moieties. The anti-peptide Ab-F(ab')2 is reduced with cysteine and the purified, recovered anti-peptide Fab'-SH
reacted with the anti-CEA-Fab'-maleimide to generate the Fab' x Fab' bi-specific Ab.
Alternatively, the anti-peptide Fab'-SH fragment may be coupled with the anti-CEA
F(ab')2 to generate a F(ab')2 x Fab' construct, or with anti-CEA IgG to generate an IgG x Fab' bi-specific construct. In one embodiment, the IgG x Fab' construct can be prepared in a site-specific manner by attaching the antipeptide Fab' thiol group to anti-CEA IgG heavy-chain carbohydrate which has been periodate-oxidized, and subsequently activated by reaction with a commercially available hydrazide-maleimide cross-linker. The component Abs used can be chimerized or humanized by known techniques. A chimeric antibody is a recombinant protein that contains the variable domains and complementary determining regions derived from a rodent antibody, while the remainder of the antibody molecule is derived from a human antibody. Humanized antibodies are recombinant proteins in which murine complementarity determining regions of a monoclonal antibody have been transferred from heavy and light variable chains of the murine inununoglobulin into a human variable domain.
A variety of recombinant methods can be used to produce bi-specific antibodies and antibody fragments. For example, bi-specific antibodies and antibody fragments can be produced in the milk of transgenic livestock. See, e.g., Colman, A., Bioehern. Soc. Symp., 63: 141-147, 1998; U.S. Patent No. 5,827,690. Two DNA
constructs are prepared which contain, respectively, DNA segments encoding paired immunoglobulin heavy and light chains. The fragments are cloned into expression vectors which contain a promoter sequence that is preferentially expressed in mammary epithelial cells. Examples include, but are not limited to, promoters from rabbit, cow and sheep casein genes, the cow cc-lactoglobulin gene, the sheep (3-lactoglobulin gene and the mouse whey acid protein gene. Preferably, the inserted fragment is flanked on its 3' side by cognate genomic sequences from a mammary-specific gene. This provides a polyadenylation site and transcript-stabilizing sequences. The expression cassettes are coinjected into the pronuclei of fertilized, mammalian eggs, which are then implanted into the uterus of a recipient female and allowed to gestate. After birth, the progeny are screened for the presence of both transgenes by Southern analysis. In order for the antibody to be present, both heavy and light chain genes must be expressed concurrently in the same cell. Milk from transgenic females is analyzed for the presence and functionality of the antibody or antibody fragment using standard immunological methods known in the art. The antibody can be purified from the milk using standard methods known in the art.
A chimeric Ab is constructed by ligating the cDNA fragment encoding the mouse light variable and heavy variable domains to fragment encoding the C
domains from a human~antibody. Because the C domains do not contribute to antigen binding, the chimeric antibody will retain the same antigen specificity as the original mouse Ab but will be closer to human antibodies in sequence. Chimeric Abs still contain some mouse sequences, however, and may still be immunogenic. A humanized Ab contains only those mouse amino acids necessary to recognize the antigen. This product'is constructed by building into a human antibody framework the amino acids from mouse complementarity determining regions.
Other methods for producing bsAbs include engineered recombinant Abs which have additional cysteine residues so that they crosslink more strongly than the more common immunoglobulin isotypes. See, e.g., FitzGerald et al., Protein Eng.
10(10):1221-1225, 1997. Another approach is to engineer recombinant fusion proteins linking two or more different single-chain antibody or antibody fragment segments with the needed dual specificities. See, e.g., Coloma et al., Nature Biotech.
15:159-163, 1997. A variety of bi-specific fusion proteins can be produced using molecular engineering. In one form, the bi-specific fusion protein is monovalent, consisting of, for example, a scFv with a single binding site for one antigen and a Fab fragment with a single binding site for a second antigen. In another form, the bi-specific fusion protein is divalent, consisting of, for example, an IgG with two binding sites for one antigen and two scFv with two binding sites for a second antigen.
Functional bi-specific single-chain antibodies (bscAb), also called diabodies, can be produced in mammalian cells using recombinant methods. See, e.g., Mack et al., Proc. Natl. Acad. Sci., 92: 7021-7025, 1995. For example, bscAb are produced by joining two single-chain Fv fragments via a glycine-serine linker using recombinant methods. The V light-chain (VL) and V heavy-chain (VH) domains of two antibodies of interest are isolated using standard PCR methods. The VL and VH cDNA's obtained from each hybridoma are then joined to form a, single-chain fragment in a two-step fusion PCR. The first PCR step introduces the (Gly4-Serl)3 linker, and the second step joins the VL and VH amplicons. Each single chain molecule is then cloned into a bacterial expression vector. Following amplification, one of the single-chain 1 S molecules is excised and sub-cloned into the other vector, containing the second single-chain molecule of interest. The resulting bscAb fragment is subcloned into an eukaryotic expression vector. Functional protein expression can be obtained by transfecting the vector into Chinese hamster ovary cells. Bi-specific fusion proteins are prepared in a similar manner. Bi-specific single-chain antibodies and bi-specific fusion proteins are included within the scope of the present invention.
Bi-specific fusion proteins linking two or more different single-chain antibodies or antibody fragments are produced in similar manner.
Recombinant methods can be used to produce a variety of fusion proteins. For example a fusion protein comprising a Fab fragment derived from a humanized monoclonal anti-CEA antibody and a scFv derived from a murine anti-diDTPA can be produced. A flexible linker, such as GGGS connects the scFv to the constant region of the heavy chain of the anti-CEA antibody. Alternatively, the scFv can be connected to the constant region of the light chain of hMN-14. Appropriate linker sequences necessary for the in-frame connection of the heavy chain Fd to the scFv are introduced into the VL and VK domains through PCR reactions. The DNA fragment encoding the scFv is then ligated into a staging vector containing a DNA
sequence encoding the CH1 domain. The resulting scFv-CHl construct is excised and ligated into a vector containing a DNA sequence encoding the VH region of an anti-CEA
antibody. The resulting vector can be used to transfect mammalian cells for the expression of the bi-specific fusion protein.
Large quantities of bscAb and fusion proteins can be produced using Esclaericlaia coli expression systems. See, e.g., Zhenping et al., Biotechtaology, 14:
192-196, 1996. A functional bscAb can be produced by the coexpression in E.
coli of two "cross-over" scFv fragments in which the VL and VH domains for the two fragments are present on different polypeptide chains. The V light-chain (VL) and V
heavy-chain (VH) domains of two antibodies of interest are isolated using standard PCR methods. The cDNA's are then ligated into a bacterial expression vector such that C-terminus of the VL domain of the first antibody of interest is ligated via a linker to the N-terminus of the VH domain of the second antibody. Similarly, the C-terminus of the VL domain of the second antibody of interest is ligated via a linker to the N-terminus of the VH domain of the first antibody. The resulting dicistronic operon is placed under transcriptional control of a strong promoter, e.g., the E. coli alkaline phosphatase promoter which is inducible by phosphate starvation.
Alternatively, single-chain fusion constructs have successfully been expressed in E. coli using the lac promoter and a medium consisting of 2% glycine and 1% Triton X-100. See, e.g., bang et al., Appl. Eyaviron. Mic~obiol., 64: 2869-2874, 1998. An E. coli, heat-stable, enterotoxin II signal sequence is used to direct the peptides to the periplasmic space.
After secretion, the two peptide chains associate to form a non-covalent heterodimer which possesses both antigen binding specificities. T'he bscAb is purified using standard procedures known in the art, e.g., Staphylococcal protein A
chromatography.
Functional bscAb and fusion proteins also can be produced in the milk of transgenic livestock. See, e.g., Colman, A., Bioclaem. Soc. Symp., 63: 141-147, 1998;
U.S. Patent No. 5,827,690. The bscAb fragment, obtained as described above, is cloned into an expression vector containing a promoter sequence that is preferentially expressed in mammary epithelial cells. Examples include, but are not limited to, promoters from rabbit, cow and sheep casein genes, the cow a-lactoglobulin gene, the sheep (3-lactoglobulin gene and the mouse whey acid protein gene. Preferably, the inserted bscAb is flanked on its 3' side by cognate genomic sequences from a mammary-specific gene. This provides a polyadenylation site and transcript-stabilizing sequences. The expression cassette is then injected into the pronuclei of fertilized, mammalian eggs, which are then implanted into the uterus of a recipient female and allowed to gestate. After birth, the progeny are screened for the presence of the introduced DNA by Southern analysis. Milk from transgenic females is analyzed for the presence and functionality of the bscAb using standard immunological methods known in the art. The bscAb can be purified from the milk using standard methods known in the art. Transgenic production of bscAb in milk provides an efficient method for obtaining large quantities of bscAb.
Functional bscAb and fusion proteins also can be produced in transgenic plants. See, e.g., Fiedler et al., Biotech., 13: 1090-1093, 1995; Fiedler et al., Imnaunotechnology, 3: 205-216, 1997. Such production offers several advantages including low cost, large scale output and stable, long term storage. The bscAb fragment, obtained as described above, is cloned into an expression vector containing a promoter sequence and encoding a signal peptide sequence, to direct the protein to the endoplasmic recticulum. A variety of promoters can be utilized, allowing the practitioner to direct the expression product to particular locations within the plant.
For example, ubiquitous expression in tobacco plants can be achieved by using the strong cauliflower mosaic virus 35S promoter, while organ speciftc expression is achieved via the seed specific legumin B4 promoter. The expression cassette is transformed according to standard methods known in the art. Transformation is verified by Southern analysis. Transgenic plants are!analyzed for the presence and functionality of the bscAb using standard immunological methods known in the art.
The bscAb can be purified from the plant tissues using standard methods known in the ~1 art.
Additionally, transgenic plants facilitate long term storage of bscAb and fusion proteins. Functionally active scFv proteins have been extracted from tobacco' leaves after a week of storage at room temperature. Similarly, transgenic tobacco seeds stored fox 1 year at room temperature show no loss of scFv protein or its antigen binding activity.
Functional bscAb and fusion proteins also can be produced in insect cells.
See, e.g., Mahiouz et al., J. Immunol. Methods, 212: 149-160 (1998). Insect-based expression systems provide a means of producing large quantities of homogenous and properly folded bscAb. The baculovirus is a widely used expression vector for insect cells and has been successfully applied to recombinant antibody molecules.
See, e.g., Miller, L.K., Ann. Rev. Microbiol., 42: 177 (1988); Bei et al., J. Irnmufaol.
Methods, 186: 245 (1995). Alternatively, an inducible expression system can be utilized by generating a stable insect cell line containing the bscAb construct under the transcriptional control of an inducible promoter. See, e.g., Mahiouz et al., J.
Immuraol. Methods, 212: 149-160 (1998). The bscAb fragment, obtained as described above, is cloned into an expression vector containing the Drosphila metallothionein promoter and the human HLA-A2 leader sequence. The construct is then transfected into D. melanogaster SC-2 cells. Expression is induced by exposing the cells to elevated amounts of copper, zinc or cadmium. The presence and functionality of the bscAb is determined using standard immunological methods known in the art.
Purified bscAb is obtained using standard methods known in the art.
Preferred bispecific antibodies of the instant invention are those which incorporate the Fv of MAb Mu-9 and the Fv of MAb 679 or the Fv of MAb MN-14 and the Fv of MAb 679, and their human, chimerized or humanized counterparts.
The MN-14, as well as its chimerized and humanized counterparts, are disclosed in U.S.
Patent No. 5,874,540. Also preferred are bispecific antibodies which incorporate one or more of the CDRs of Mu-9 or 679. The antibody can also be a fusion protein or a bispecific antibody that incorporates a Class-III anti-CEA antibody and the Fv of 679.
Class-III antibodies, including Class-III anti-CEA are discussed in detail in U.S.
Patent No. 4,818,709. 679 is described in US20030113333A and US20020006379.
VII. Other Applications The present invention encompasses the use of the bsAb and a therapeutic or diagnostic agent associated with the targetable construct discussed above in intraoperative, intravascular, and endoscopic tumor and lesion detection, biopsy and therapy as described in U.S. Patent No. 6,096,289.
The antibodies and antibody fragments of the present invention can be employed not only for therapeutic or imaging purposes, but also as aids in performing research in vitro. For example, the bsAbs of the present invention can be used ira vitro to ascertain if a targetable construct can form a stable complex with one or more bsAbs. Such an assay would aid the skilled artisan in identifying targetable constructs which form stable complexes with bsAbs. This would, in turn, allow the skilled artisan to identify targetable constructs which are likely to be superior as therapeutic and/or imaging agents.
The assay is advantageously performed by combining the targetable construct in question with at least two molar equivalents of a bsAb. Following incubation, the mixture is analyzed by size-exclusion HPLC to determine whether or not the construct has bound to the bsAb. Alternatively, the assay is performed using standard combinatorial methods wherein solutions of various bsAbs are deposited in a standard 96-well plate. To each well, is added solutions of targetable construct(s).
Following incubation and analysis, one can readily determine which constructs) binds) best to which bsAb(s).
It should be understood that the order of addition of the bsAb to the targetable construct is not crucial; that is, the bsAb may be added to the construct and vice versa.
Likewise, neither the bsAb nor the construct needs to be in solution; that is, they may be added either in solution or neat, whichever is most convenient. Lastly, the method of analysis for binding is not crucial as long as binding is established.
Thus, one may analyze for binding using standard analytical methods including, but not limited to, FABMS, high-field NMR or other appropriate method in conjunction with, or in place of, size-exclusion HPLC.
The present invention is further illustrated by, though in no way limited to, the following examples.
Examples IMP 281 Labeling ' The following peptide was labeled with I i iIn: (MH+: 1361) DOTA-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 bi addition, the peptide was tested, after labeling, for stability in human serum (previously frozen) over a period of 20 hrs at 37°C. The study showed that the peptide remained stable. IMP 281 was also tested for binding with the humanized antibody, m679xhMNl4. The In-111 labeled peptide was analyzed both on the reverse phase and the size exclusion HPLC systems.
Synthesis of IMP 281 The peptide was synthesized (NB Ref. CN 2-34) by solid phase peptide synthesis on Rink Amide resin (2.0159 g, 0.7 mmol/g) using standard Fmoc synthesis methodology. The following amino acids (6 equivalents per coupling) were added in the order shown; Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Glu(OBut)-OH, Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Ala-OH and DOTA-tris(t-Butyl) ester. Each amino acid was double coupled with two hour couplings first using diisopropylcarbodiimide followed by a coupling using O-Benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluoro-phosphate (HBTU) as the activating agents. The Aloc side chains were then removed with the Pd catalyst in the usual way and the trityl-HSG-OH was double coupled to the lysine side chains. The peptide was cleaved from the resin with TFA, precipitated in ether, and purified by HPLC to obtain the desired peptide. Successful synthesis was confirmed by ESMS analysis, MH+: 1361. The total yield of desired product was 298.8 mg in 6 fractions. Out of these 6 fractions, 4 contain pure peptide amounting to 201.3 mg while the remaining two fractions contain a slight impurity with a total mass of 97.5 mg.
IMP 281 Solution The peptide (3.5 mg) was combined with 1169 ~L of 0.5 M NH40Ac Buffer (pH 3.98, Ref. BM10-91) to reach a final concentration of 2.2x10-3 M.
IMP 281 Labeling (Figures 1 and 2) i i llnCl3 (6.3 ~L) was added to 2.5 ~,L of the IMP 281 solution and 150 ~.L
of 0.5 M NH40Ac Buffer (pH 3.98, Ref. BM10-91) in a screw top plastic vial. This vial was placed in a lead pig which was submerged in a boiling water bath, for 15 minutes.
The plastic vial was removed and allowed to return to r.t. The labeled peptide was subsequently analyzed by reverse phase HPLC and size exclusion HPLC. The chromatograms (Figures 1 & 2) showed that the peptide labeled well. The peptide concentration of this solution was 3.463x10-4 M.
mIn IMP 281 in Human Serum (Fig. 3) A (1:9) mixture of the labeled peptide and human serum (previously frozen) was made and incubated at 37°C. The I l lIn IMP 281 solution (70 ~,L) and 630 ~L of human serum were combined and vortexed. This was maintained at constant temperature for 20 hrs. with injections into the reverse phase HPLC at t0=0 hrs, t1=1.3 hrs, t2=2.5 hrs, and t3=20 hrs. The labeled peptide appears to not have undergone any change as a result of being mixed in human serum. This mixture had a peptide concentration of 3.463x10-5 M.
~ 1 lIn IMP 2g 1 & Antibody Binding (Fig. 4) The labeled peptide (0.5 ~L) mixture was combined with 4.33 ~.L of m679xhMNl4 antibody (antibody/peptide ratio of 10:1) and 995 ~.L of 0.9%
Saline.
The solution was vortexed and analyzed by size exclusion HPLC. The chromatograms indicate that there is mostly bis-antibody bound but some mono-antibody bound peptide.
a lIn IMP 281 w/Antibody & Human Serum (Figs. 5 & 6) "lIn-IMP 281 in human serum (5.0 ~.L) was added to 4.33 ~.L of m679xhMN14 antibody (antibody/peptide ratio of 10:1) and 990 ~.L 0.9% Saline.
The mixture was vortexed and analyzed by size exclusion HPLC at two time points (1 hr and 20 hrs). The results look similar to those of the peptide with the antibody alone.
IMP 284 Labeling The following peptide was labeled with 111In: (MH+: 1471) DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NHZ
In addition, the peptide was tested, after labeling, for stability in human serum over a period of 21 hrs. in human serum and 19 hrs. in mouse serum. The study showed that the peptide remained stable. IMP 284 was also tested for binding with the humanized antibody, m679xhMNl4. Reverse phase and the size exclusion HPLC
systems were utilized in this study to analyze the labeling, antibody binding, and serum stability.
Synthesis of IMP 284 The peptide was synthesized by solid phase peptide synthesis on Rink Amide resin (1.0 g, 0.6 mmol/g) using standard Fmoc synthesis methodology. The following amino acids (6 equivalents per coupling) were added in the order shown; Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Tyr(But)-OH, Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Phe-OH and DOTA-tris(t-Butyl) ester. Each amino acid was coupled overnight using diisopropylcarbodiimide (DIC) as the activating agent. The Aloc side chains were then removed with the Pd catalyst in the usual way and the trityl-HSG-OH (7 Eq) was coupled to the lysine side chains with DIC (7 Eq) in the presence of DIEA (14 Eq).

The peptide was cleaved from the resin with TFA, precipitated in ether, and purified by HPLC to obtain the desired peptide. Successful synthesis was confirmed by ESMS
analysis, MH+: 1471. The total yield of desired product was 248.1 mg in 3 fractions.
IMP 284 Solution The peptide (3.7 mg) was combined with 1143 ~L of 0.5 M NH40Ac Buffer (pH 3.98, Ref. BM10-91) to reach a final concentration of 2.2x10-3 M.
IMP 284 Labeling (Figures 7 & 8) "IInCl3 (7.9 ~.L) was added to 2.5 ~L of the IMP 284 solution and 150 ~.L of 0.5 M NHdOAc Buffer (pH 3.98, Ref. BM10-91) in a "screw top" plastic vial.
This vial was placed in a led pig which was, it self, submerged in a boiling water bath, for minutes. The plastic vial was removed and allowed to return to r.t. and was subsequently analyzed by reverse phase HPLC. The chromatogram showed that the peptide labeled well. The peptide concentration of this solution was 3.429x10-5 M.
11 iIn IMP 284 & Antibody Binding (Figure 9) 15 The labeled peptide (0.5 ~.L) mixture was combined with 3.1 ~.L of m679xhMN14 antibody (antibody/peptide ratio of 19.7:1) and 1000 ~L of 0.9%
Saline. The solution was vortexed and analyzed by HPLC. The chromatograms indicate that there is mostly dibound and a small amount of what may be monobound peptide.
111In IMP 284 in Human Serum (Figure 10) A mixture of the labeled peptide and human serum was made and incubated at 37°C for 19.5 hrs. The human serum was freshly drawn, spun, and filtered through a Millex~-GV 0.22 ~.m Filter Unit. The Indium-labeled peptide solution (50 ~L) and 450 ~L of human serum were combined and vortexed. While incubating, injections were made into the reverse phase HPLC at t0=0 hrs, t1=2.0 hrs, and t2=19.5 hrs. The labeled peptide appears to not have undergone any change as a result of being mixed in human serum. This mixture had a peptide concentration of 3.429x10-6 M.
after dilution in human serum.
1 l lIn IMP 284 in Mouse Serum (Figure 13) A mixture of the labeled peptide and mouse serum was made and incubated at 37°C. The mouse serum was fresh nude mouse serum obtained from (GSCC on the same day). The Indium-labeled peptide solution (50 ~,L) and 450 ~,L of the mouse serum were combined and vortexed. This was incubated at 37°C for 18.0 hrs.
Injections were made into the reverse phase HPLC at t0=0 hrs and t1=18.0 hrs.
The labeled peptide appears to not have undergone any change as a result of being mixed in mouse serum. This mixture had a peptide concentration of 3.429x10- M after dilution in mouse serum.
i i ~In IMP 284 w/Antibody & Human Serum (Figures 11 and 12) "lIn-IMP 284 in human senim (1.0 ~L) was added to 0.7 ~L of m679xhMNl4 antibody (antibody/peptide ratio of 22.3:1) and 60 ~.L 0.9% Saline. The mixture was vortexed and analyzed by size exclusion HPLC. The same procedure was repeated hrs. later. The results of both radiometric chromatograms look similar to those of the peptide with the antibody alone.
i l iIn IMP 284 w/Antibody & Human Serum (Figures 14 and 15) 111In-IMP 284 in human serum (1.0 ~L) was added to 0.7 ~.L of m679xhMNl4 antibody (antibody/peptide ratio of 22.3:1) and 60 ~L 0.9% Saline. The mixture was vortexed and analyzed by size exclusion HPLC. The same procedure was repeated hrs. later. The results of both radiometric chromatograms look similar to those of the peptide with the antibody alone.
Peptide Labeling The following peptides were labeled with 11 lIn:

DOTA-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ (MH+:1361) DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NHZ (MH+:1471) In addition, the peptides were tested, after labeling, for stability in human serum over a period of 21 hrs. The study showed that both peptides remained stable.
IMP281 & IMP284 were also tested for binding with the humanized antibody, m679xhMN14 (100,000 g mol-1, 10.9 mg mL-1). Studies were analyzed both on reverse phase and size exclusion HPLC systems.

This peptide was previously synthesized and due to impurities discovered after the initial purification, was repurified twice more using different mobile phases, columns, flowrates, and gradients. The material used for the labeling study described in this report was ~97% pure (HPLC).

This peptide was previously synthesized and due to impurities discovered after the initial purification, was repurified twice more using different mobile phases, columns, flowrates, and gradients. The material used for the labeling study described in this report was ~97% pure (HPLC).
IMP 281 Stock Solution The peptide (3.9 mg) was combined with 1303 ~.L of 0.5 M NH40Ac Buffer (pH 3.98) to reach a concentration of 2.2x10-3 M.
IMP 284 Stock Solution The peptide (1.5 mg) was combined with 464 ~.L O.SM NH40Ac Buffer (pH
3.98) to reach a concentration of 2.2x10-3 M.
IMP 281 Labeling (Figures 16 & 17) 111InC13 (6.2 ~.L) was added to 2.5 ~,L of the IMP 281 stock solution and 150 ~.L of 0.5 M NH40Ac Buffer (pH 3.98) in a screw top plastic vial. This vial was placed in a lead pig which was submerged in a boiling water bath for 32 minutes. The plastic vial was removed and allowed to return to RT and was subsequently analyzed by reverse phase HPLC. Reverse Phase HPLC shows that the peptide labeled well with only a trace amount of unbound Indium. The peptide concentration of this solution was 3.466x10-5 M.
n iIn IMP 281 ~ Antibody Binding (Figure 18) The labeled peptide (1.0 ~.L) mixture was combined with 3.2 ~L of m679xhMN14 antibody (antibody/peptide ratio of 10:1) and 200 ~.L of 0.9%
Saline.
The solution was vortexed and analyzed by HPLC. The chromatograms indicate that there is mostly dibound but some monobound peptide as well.

IMP 284 Labeling (Figures 19 and 20) "lInCl3 (6.2 ~,L) was added to 2.5 ~,L of the IMP 284 stock solution and 150 ~,L of 0.5 M NH40Ac Buffer (pH 3.98) in a screw top plastic vial. This vial was placed in a lead pig which was submerged in a boiling water bath, for 32 minutes.
The plastic vial was removed and allowed to return to RT and was subsequently analyzed by reverse phase HPLC. Reverse Phase HPLC shows that the peptide labeled well with only a trace amount of unbound Indium. The peptide concentration of this solution was 3.466x10-5 M.
l liIn IMP 284 & Antibody Binding (Fig ure 21) The labeled peptide (1.0 ~,L) mixture was combined with 3.2 ~.L of m679xhMN14 antibody (antibody/peptide ratio of 10:1) and 200 wL of 0.9%
Saline.
The solution was vortexed and analyzed by HPLC. The chromatograms indicate that there is mostly dibound but some monobound peptide as well.
11 lIn IMP 281 in Human Serum (Figure 22) A mixture of the labeled peptide and freshly drawn and filtered human serum was made and incubated at 37°C. The Indium-111 labeled peptide (50 ~.L) and 450 ~,L of human serum were combined and vortexed. This was maintained at constant temperature for 21 hrs with injections into the reverse phase HPLC at t0=2.25 hrs, t1=3.25 hrs, & t2=20.25 hrs. The labeled peptide appears to not have undergone any change as a result of being mixed in human serum. This mixture had a peptide concentration of 3.466x10-6 M.
IllIn IMP 284 in Human Serum (Figure 23) A mixture of the labeled peptide and freshly drawn and filtered human serum was made and incubated at 37°C. The Indium-111 labeled peptide (50 ~,L) and 450 ~.L
of human serum were combined and vortexed. This was maintained at constant temperature for 21 hrs with injections into the reverse phase HPLC at t0=2.5 hrs &
t1=20.5 hrs. The labeled peptide appears to not have undergone any change as a result of being mixed in human serum. This mixture had a peptide concentration of 3.466x10-6 M.
111In IMP 281 w/Antibody & Human Serum (Figure 24) l l lln-IMP 281 in human serum (1.0 ~L) was added to 0.64 ~.L of m679xhMN14 antibody (antibody/peptide ratio of 20:1) and 200 ~L 0.9% Saline.

The mixture was vortexed and analyzed by size exclusion HPLC. The results look similar to those of the peptide with the antibody alone.
i I IIn IMP 284 wlAntibody & Human Serum (Figure 25) I ~ lIn-IMP 284 in human serum (1.0 ~L) was added to 0.64 ~,L of S m679xhMN14 antibody (antibody/peptide ratio of 20:1) and 200 ~L 0.9% Saline.
The mixture was vortexed and analyzed by size exclusion HPLC. The results look similar to those of the peptide with the antibody alone.
The following peptide was synthesized and labeled with 1' 11n: (IMP 277 MH+
1419) DOTA-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 An all D-amino acid was used to improve the stability of the peptide in serum.
Synthesis:
The peptide was synthesized by solid phase peptide synthesis on Sieber Amide resin using the Fmoc procedure. The following amino acids were added in the order shown; Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Glu(OBut)-OH, Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Glu(OBut)-OH, DOTA-tris(tBu) ester. Each amino acid was double coupled with two hour couplings first using diisopropylcarbodiimide followed by a coupling using O-Benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluoro-phosphate (HBTU) as the activating agents. The Aloc side chains were then remove with the Pd catalyst in the usual way and the trityl-HSG-OH was double coupled to the lysine side chains. The peptide was cleaved from the resin with TFA, precipitated in ether, and purified by HPLC to obtain the desired peptide.
Labeling Solution:
The peptide, 0.0015 g (1.06 x 10-6 mol) was dissolved in 480 ~.L, 0.5 M
NH40Ac pH 3.98 for labeling.
Radiolabeling:
The peptide, 2.5 ~,L was mixed with 20 ~L In-111 (2.78 mCi) and 60 ~,L 0.5 M NH40Ac pH 3.98. The solution was heated in a boiling water bath for 15 min.
Another labeling was performed using the same conditions except a different reaction buffer was used 30 ~.L, 0.5 M NH40Ac pH 5.5 buffer Serum Stability:
Human Serum:
The In-111 IMP 277 (30 ~,L) was diluted in 540 pL, of fresh human serum and placed in an incubator at 37°C. Aliquots were withdrawn at various time points and examined by reverse phase HPLC and by size exclusion HPLC.
Mouse Serum:
The In-111 IMP 277 (15 ~L) was diluted in 150 p,L of fresh mouse serum and placed in an incubator at 37°C. Aliquots were withdrawn at various time points and examined by reverse phase HPLC and by size exclusion HPLC.
Results:
The peptide labels well with In-111 but reverse phase HPLC shows that it contains an impurity which varies in amount depending on the labeling conditions used. Antibody binding studies show that the peptide can bind two hMN-14 x 679 bispecific antibodies and that it does not bind hMN-14 x 734. The peptide is very stable in human serum and appears to be just as stable in mouse serum. The mouse serum antibody binding SEC HPLC experiments do show some peaks in the antibody region when there should be none but this is probably due to the presence of the antibody in the injector or syringe from previous injections.
Conclusions:
The peptide labels well and is stable in mouse and human serum.
Example Peptides for Carrying Therapeutic/Imaging Radioisotopes to Tumors via Bispecific Antibody Tumor Pretargeting DOTA-Phe-Lys(HSG)-Tyr-Lys(HSG)-NH2 (IMP 237) was synthesized to deliver therapeutic radioisotopes such as ~°Y or 177Lu to tumors via bispecific antibody tumor pretargeting. The bispecific antibody is composed of one portion which binds to an antigen on the tumor and another portion which binds to the HSG
peptide. The antibody which binds the HSG peptide is 679. This system can also be used to deliver imaging isotopes such as I i lIn-111.
Synthesis of IMP 237 IMP 237 was synthesized on Sieber Amide resin (Nova-Biochem) using standard Fmoc based solid phase peptide synthesis to assemble the peptide backbone with the following protected amino acids, in order: Fmoc-Lys(Aloc)-OH, Fmoc-Tyr(But)-OH, Fmoc-Lys(Aloc)-OH, Fmoc-Phe-OH, (Reagents from Advanced Chemtech) tri-t-butyl DOTA (Macrocyclics). The side lysine side chains were then deprotected with Pd[P(Ph)3]4 by the method of Dangles et.al. J. Org. Chefya.
52:4984-4993 (1987). The HSG ligands were then added as Trityl HSG (synthesis described below) using the BOP/HBTU double coupling procedure used to attach the amino acids. The peptide was cleaved from the resin and the protecting groups were removed by treatment with TFA. The peptide was purified by HPLC to afford 0.6079 g of peptide from 1.823 g of Fmoe-Lys(Aloc)-Tyr(But)-Lys(Aloc)-NH-Sieber amide resin.
Synthesis ofN-Trityl-HSG-OH .
Glycine t-butyl ester hdyrochloride (15.263 g, 9.1 x1 0-2 mol) and 19.760 g NaZC03 were mixed, then suspended in 50 mL H20 and cooled in an ice bath.
Succinic anhydride (9.142 g, 9.14 x 10-2 mol) was then added to the reaction solution which was allowed to warm slowly to room temperature and stir for 18 hr.
Citric acid (39.911 g) was dissolved in 50 mL HZO and slowly added to the reaction solution and then extracted with 2 x 150 mL EtOAc. The organic extracts were dried over Na2S04, filtered and concentrated to afford 25.709 g of a white solid.
The crude product (25.709 g) was dissolved in 125 mL dioxane, cooled in a room temperature water bath and mixed with 11.244 g of N-hydroxysuccinimide.
Diisopropylcarbodiimide 15.0 mL was added to the reaction solution which was allowed to stir for one hour. Histamine dihydrochloride (18.402 g, 1.00 x 10-1 mol) was then dissolved in 100 mL DMF and 35 mL diisopropylethylamine. The histamine mixture was added to the reaction solution which was stirred at room temperature for 21 hr. The reaction was quenched with 100 mL water and filtered to remove a precipitate. The solvents were removed under hi-vacuum on the rotary evaporator.
The crude product was dissolved in 300 mL dichloromethane and extracted with mL saturated NaHC03. The organic layer was dried over Na2S04 and concentrated to afford 34.19 g of crude product as a yellow oil.
The crude product (34.19 g) was dissolved in 50 mL chloroform and mixed with 31 mL diisopropylethylamine. Triphenylmethyl chloride (25.415g) was dissolved in 50 ml chloroform and added dropwise to the stirred reaction solution which was cooled in an ice bath. The reaction was stirred for 45 min and then quenched with 100 mL H20. The layers were separated and the organic solution was dried over Na2S04 and concentrated to form a green gum. 'The gum was triturated with 100 mL Et20 to form a yellow precipitate which was washed with 3 x 50 mL
portions of Et20. The solid was vacuum dried to afford 30.641 g (59.5 %
overall yield) of N-trityl-HSG-t-butyl ester.
N-trityl-HSG-t-butyl ester (20.620 g, 3.64 x 10-2 mol) was dissolved in a solution of 30 mL chloroform and 35 mL glacial acetic acid. The reaction was cooled in an ice bath and 15 mL of BF3~Et20 was slowly added to the reaction solution. The reaction was allowed to warm slowly to room temperature and mix for 5 hr. The reaction was quenched by pouring into 200 mL 1M NaOH and the product was extracted with 200 mL chloroform. The organic layer was dried over Na2S04 and concentrated to afford a crude gum which was triturated with 100 mL Et~,O to form a precipitate. The crude precipitate was poured into 400 mL 0.5 M pH 7.5 phosphate buffer and extracted with 2 x 200 mL EtOAc. The aqueous layer was acidified to pH
3.5 with 1 M HCl and extracted with 2 x 200 mL chloroform. A precipitate formed and was collected by filtration (8.58 g). The precipitate was the desired product by HPLC comparison to a previous sample (ESMS MH+ 511).
Radiolabeling Labeling with I I iIn The lIn 0300 p,Ci/kit) was diluted to 0.5 mL in deionized water and added to the lyophilized kits. The kits were heated in a boiling water bath for 15 min, the vials were cooled and 0.5 mL of 2.56 x 10-5 M In in 0.5 M acetate buffer was added and the kits were again heated in the boiling water bath for 15 min. The labeled peptide vials were cooled to room temperature and evaluated by reverse phase HPLC
(HPLC conditions: Waters Nova-Pak C-18, 8x100 mm RCM column eluted at 3 mLlmin with a linear gradient from 100 % (0.1 % TFA in Hz0) to 100 % (90 CH3CN, 0.1 % TFA, 10 % H20)). The HPLC analysis revealed that the minimum concentration of peptide needed for labeling (4.7 % loose 1 i IIn), with this formulation, was 35 pg/mL. The reverse phase HPLC trace showed a sharp 11 iIn labeled peptide peak. The labeled peptide was completely bound when mixed with excess 679 IgG
by size exclusion HPLC.

In-Vivo Studies Nude mice bearing GW-39 human colonic xenograft tumors (104-500 mg) were injected with the bispecific antibody hMN-14 x m679 (1.5 x 10-x°
mol). The antibody was allowed to clear for 24 hr before the I 1 lIn labeled peptide (8.8 ~.Ci, 1.5 x 10-1' mol) was injected. The animals were sacrificed at 3, 24, 48 hr post injection.
The results of the biodistribution studies of the peptide in the mice pretargeted with hMN-14 x m679 are shown in Table 1. The tumor to non-tumor ratios of the peptides in the pretargeting study are show in Table 2.
Table 1 Pretargeting With' llIn Labeled Peptide 24 hr After Injection of hMN-14 x m679 Injected/g Tissue Tissue 3 hr After "'In24 hr After 48 hr After IMP 237 "'In IMP "'In IMP

GW-39 7.25 2.79 8.38 1.70 5.39 1.46 Liver 0.58 0.13 0.62 0.09 0.61 0.16 Spleen 0.50 0.14 0.71 0.16 0.57 0.15 Kidney 3.59 0.75 2.24 0.40 1.27 0.33 Lungs 1.190;26 0.440.10 0.220.06 Blood 2.42 0.61 0.73 0.17 0.17 0.06 Stomach 0.18 0.03 0.09 0.02 0.07 0.02 Sm. Int. 0.65 0.74 0.18 0.03 0.11 0.02 Lg. Int. 0.30 0.07 0.17 0.03 0.13 0.03 Table 2 Pretargeting With' 1'In Labeled Peptides 24 hr After Injection of hMN-14 x m679 Tumor/Non-Tumor Tissue Ratios Tissue 3 hr After "'In24 hr After 48 hr After IMP 237 "'In IMP "'In IMP

Liver 12.6 4.44 13.6 2.83 8.88 1.78 Spleen 15.1 6.32 12.1 2.86 9.50 1.62 Kidney 2.04 0.74 3.84 1.04 4.25 0.19 Lungs 6.11 1.96 19.6 5.91 25.4 6.00 Blood 3.04 1.13 11.9 3.20 31.9 4.79 Stomach 40.5 16.5 104. 39.6 83.3 16.5 Sm. Int. 18.9 12.6 47.5 10.3 49.5 7.83 Lg. Int. 25.2 10.6 50.1 16.7 43.7 9.35 Serum Stability of DOTA-Phe-Lys(HSG)-Tyr-Lys(HSG)-NH2 (IMP 237) and DOTA-Phe-Lys(HSG)-D-Tyr-Lys(HSG)-NH2 (IMP 241) Peptide Labeling and HPLC Analysis The peptides, IMP 237 and IMP 241, were labeled according to the procedure described by Karacay et. al. Bioconjugate Chem. 11:842-854 (2000). The peptide, IMP 241 (0.0019 g), was dissolved in 587 p,1 0.5 M NH4Cl, pH 5.5. A 1.7 pL
aliquot of the peptide solution was diluted with 165 p1 0.5 M NH4Cl, pH 5.5. The 11 iIn (1.8 mCi) in 10 ~.L, was added to the peptide solution and the mixture was heated in a boiling water bath for 30 min.
The labeled peptide was analyzed by HPLC using a Waters 8x100 mm radial-pak, nova-pak C-18 RCM cartridge column. The column was eluted at 3 mL/min with a linear gradient which started with 100 % of 0.1 % TFA in water and went to 100 % of 0.1 %TFA in 90% acetonitrile and 10 % water over 10 min. There was about 6% loose I I lIn in this labeling which came out at the void volume of the column (1.6 min). There were also some ' 1 IIn labeled peaks at 5 min and 6.6 to 8 min.
The 1 i lIn labeled peptide was eluted at 8.8 min as a single peak. The HPLC profile of 11'In IMP
237 was nearly identical to 11'In IMP 241.
Serum Stability An aliquot (30 ~,L) of "lIn IMP 241 was placed in 300 p.L, of fresh mouse serum and placed in a 37° C incubator. The peptide was monitored as described above by HPLC.
An aliquot (24 p,L) of 1 ~In IMP 237 was placed in 230 p,L of fresh mouse serum and placed in a 37° C incubator. The peptide was monitored as described above by HPLC.
The analysis showed that the I I IIn IMP 241 decomposed slightly (~ 5%) after 22 hr in mouse serum at 37° C. The "lIn IMP 237 was about 70 %
converted to the shorter retention time peak after incubation for 22 hr at 37° C.
Conclusion The D-tyrosine in the IMP 241 peptide slows the decomposition of the peptide in mouse serum compared to IMP 237.
In Vivo Stability of IMP 237 and IMP 241 Compared The in vivo stabilities of lIn IMP 237 and l lIn IMP 241 were compared by examining (by HPLC) urine samples from mice at 30 and 60 min. The peptides, IMP ' 241 and IMP 237, were I1 lIn-111 labeled as described above.
The labeled peptides were injected into Balb/c mice which were sacrificed at min and 60 min post injection of the peptides using one mouse per time point.
The HPLC traces indicated that I l lIn IMP 241 was excreted intact while mIn IMP
237 was almost completely metabolized to a new 1 i lIn labeled peptide.
Conclusion 25 The replacement of Tyr with D-Tyr in the peptide backbone lessened metabolism of the peptide in-vivo.
Synthesis of DOTA-D-Asp-D-Lys(HSG)-D-Asp-D-Lys(HSG)-NHZ (IMP 271) IMP 271 was synthesized on Sieber Amide resin (Nova-Biochem) using standard Frnoc based solid phase peptide synthesis to assemble the peptide backbone 30 with the following protected amino acids, in order: Fmoc-D-Lys(Aloc)-OH, Fmoc-D-Asp(But)-OH, Fmoc-D-Lys(Aloc)-OH, and Fmoc-D-Asp(But)-OH, (Reagents from Advanced Chemtech). The linear pepide was assembled and the DOTA-tris(tBu) ester (Macrocyclics) was added to the N-terminus. The Aloc side chains on the D-lysines were then removed with Pd[P(Ph)3]4 by the method of Dangles et.al. J.
Org.
Chem. 52:4984-4993 (1987). The HSG ligands were then added as Trityl HSG
(previously described) using the BOP/HBTU double coupling procedure used to attach the amino acids. The peptide was cleaved from the resin and the protecting groups were removed by treatment with TFA. The peptide was purified by HPLC.
Comparison biodistribution of the new peptides "IIn-IMP 281 and IMP 284 to IMP 241 in Pretargeting and when injected alone Goal: Tumor targeting of IMP 281 was examined in this example. Reagents:
hMN-14 x m679 Fab' x Fab': GN, LN 073102, 10.9 mg/mL
NCr nudes: (NCI) from Taconic, 4 weeks of age GW39: generation # 8; SC 300 E.~L of 10 %;
IMP 241: DOTA-Phe-Lys(HSG)-D-Tyr-Lys(HSG)-NH2;
IMP 281: DOTA-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NHZ; MH+

IMP 284: DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2; MH+

Group I. I1VIP 241 A. pretargeting I-125 labeled bsMab (15 ~.g) will be injected into 10 GW xenograft bearing mice, with tumors >_0.2 g. After allowing time for localization and clearance from blood, 1 i iIn-IMP-241 will be injected. At the times shown above, 5 mice per time point will be necropsied. Tissues (tumor, liver, spleen, kidney, lungs, blood, stomach, large and small intestines) will be isolated, weighed and counted in the appropriate windows.
B. Peptide alone 111In-IMP-241 (8.8 ~,Ci, 1.5 x 10-I I mol) will be injected into 10 tumor bearing mice. At 3 and 24 h post injection, 5 mice per time point will be necropsied.
Tissues (tumor, liver, spleen, kidney, lungs, blood, stomach, large and small intestines) will be isolated, weighed and counted in the appropriate windows.
Group II. IMP 281 A. pretargeting I-125 labeled bsMab (15 p,g) will be injected into 10 GW xenograft bearing mice, with tumors >_0.2 g. After allowing time for localiza~ion and clearance from blood, 11 lIn-IMP-281 will be injected. At the times shown above, 5 mice per time point will be necropsied. Tissues (tumor, liver, spleen, kidney, lungs, blood, stomach, large and small intestines) will be isolated, weighed and counted in the appropriate windows.
B. Peptide alone "'In-IMP-281 (8.8 p,Ci, 1.5 x 10-11 mol) will be injected into 12 tumor bearing mice. At 3 and 24 h post injection, 5 mice per time point will be necropsied, at 30 min, 2 mice will be necropsied. Tissues (tumor, liver, spleen, kidney, lungs, blood, stomach, large and small intestines) will be isolated, weighed and counted in the appropriate windows.
Group III. IMP 284 A. pretargeting I-125 labeled bsMab (15 fig) will be injected into 10 GW xenograft bearing mice, with tumors ?0.2 g. After allowing time for localization and clearance from blood, I l lIn-IMP-281 will be injected. At the times shown above, 5 mice per time point will be necropsied. Tissues (tumor, liver, spleen, kidney, lungs, blood, stomach, large and small intestines) will be isolated, weighed and counted in the appropriate windows.
B. Peptide alone ;
l l lIn-IMP-284 (8.8 p.Ci, 1.5 x 10-11 mol) will be injected into 13 tumor bearing mice. At 3 and 24 h post injection, 5 mice per time point will be necropsied, at min 3 mice will be necropsied. Tissues (tumor, liver, spleen, kidney, lungs, blood, 25 stomach, large and small intestines) will be isolated, weighed and counted in the appropriate windows.
Radiolabeling:
IMP-241 peptide will be labeled with In-111 according to protocol below.
i l l InCl3 (3mCi) 30 0.5 M ammonium acetate, pH 5.5 (3x volumes of In-111) peptide 2.32 pl., 2.2 x 10-3 M in 0.5 M ammonium acetate, pH 5.5 Centrifuge Heat in boiling water bath for 30 min.
Cool on ice bath for 5 min Centrifuge Add DTPA (0.1 M in 0.1 M NaOAc, pH 6.5) to a final DTPA concentration of 3mM
Let sit at room temperature for 15 min Add 0.1 M sodium acetate, pH 6.5 to 1 mL
Analyze by ITLC and HPLC. ITLC strips will be developed in saturated sodium chloride and water:ethanol:ammonia (5:2:1) Radiolabeling:
IMP-281 peptide will be labeled with In-111 according to protocol below.
lllInCl3 (3mCi) 0.5 M ammonium acetate, pH 5.5 (3x volumes of In-111) peptide 2.32 ~L, 2.2 x 10-3 M in 0.5 M ammonium acetate, pH 5.5 Centrifuge Heat in boiling water bath for 30 min.
Cool on ice bath for 5 min Centrifuge Add DTPA (0.1 M in 0.1 M NaOAc, pH 6.5) to a final DTPA concentration of 3 mM
Let sit at room temperature for 15 min Add 0.1 M sodium acetate, pH 6.5 to 1 mL
Analyze by ITLC and HPLC. ITLC strips will be developed in saturated sodium chloride and water:ethanol:ammonia (5:2:1) Radiolabeling:
IMP-284 peptide will be labeled with In-111 according to protocol below.
n iInCl3 (3mCi) 0.5 M ammonium acetate, pH 5.5 (3x volumes of In-111) peptide 2.32 N,L, 2.2 x 10-3 M in 0.5 M ammonium acetate, pH 5.5 Centrifuge Heat in boiling water bath for 30 min.
Cool on ice bath for 5 min Centrifuge Add DTPA (0.1 M in 0.1 M NaOAc, pH 6.5) to a final DTPA concentration of 3mM
Let sit at room temperature for 15 min Add 0.1 M sodium acetate, pH 6.5 to 1 mL
Analyze by ITLC and HPLC. ITLC strips will be developed in saturated sodium chloride and water:ethanol:ammonia (5:2:1) Results: comparison of the peptides 11 lIn-IMP 241, 281 and 284 alone and pretargeted with hMNl4 x m679 Fab' x Fab' This example compared IMP 241 with the two all D-amino acid backbone peptides described in the previous example, alone and in pretargeting.
Protocol and biodistribution data are attached.
Radiolabeling data for the peptides:
ITLC
I
Peptide % unbound% colloid IMP 241 1.5 0.3 IMP 281 0.6 0.3 IMP 284 0.9 0.3 SE HPLC of "'In labeled peptides peptideRT alone% recoveryRT of Peptide% shift % recovery + bsAb hMN14xm679 IMP 13.81 97.9 ND

IMP 13.46 99 8.80 99 74 IMP 13.88 96.4 8.77 99 72 C18 reverse HPLC of the 111In-labeled peptides:
Peptide RT, min % (area) IMP 241 2.4 1.8 11.51 97.7 IMP 281 2.3 0.75 8.5 98.5 IMP 284 2.3 1.3 11.61 98 Biodistribution:
Biodistribution data is shown below. In comparing the 3 peptides, tumor peptide uptake was similar for all of the 3 peptides in pretargeting after 3 and 24 h post injection, Figure 27. IMP 281 showed lower kidney uptake than IMP 241 and in pretargeting as well as when given alone, Figures 28 and 29.
Conclusions: The bsAb and peptide tumor uptakes were the same in the three Pretargeting groups with the different peptides. IMP 281 showed slightly lower kidney uptake in pretargeting than with IMP 241 and 284. Reduced kidney uptake for IMP
281 was more obvious when the peptides were injected alone.
Table 3 Time post 24 Injection:hr Experiment Pretargeting 1 vs Description: Bio with IMP-284 In-111-IMP-241 vs IMP-28 Group #: IA MAb isotope ("51--> 1 injectedi-125 hrs ->"
In-IMP-241) uCi injected:6 MAb MN-14 1 x injected:hm679 Corrected cpm : 9802182 MAb 1 injected Mean Body 21.09 weight:

Tissue n STD WT STD% %ID/oraSTD% T/NTSTD
Weioht / ID/g ID/p ID/Orp T/NT

Tumor 5 0.054 3.060.61 1.350.22 1.000.00 0.447 Liver 5 0.197 0.190.06 0.220.04 17.043.17 1.223 Spleen 5 0.018 0.200.06 0.020.00 16.053.09 0.081 Kidney 5 0.016 0.140.03 0.020.00 22.273.44 0.139 Lungs 5 0.026 0.130.03 0.020.00 24.527.49 0.153 Blood 5 0.002 0.150.03 0.230.03 20.263.91 0.241 Stomach 5 0.160 0.500.27 0.220.09 8.145.42 0.493 Sm Int 5 0.235 0.080.04 0.090.05 43.6316.32 1.124 Lg Int 5 0.262 0.080.02 0.060.01 41.1012.16 0.875 MAb 2 isotope injectedin-111 MAb 2 cpm injected: 4975201.6 MAb 2 injected: IMP-241 Tissue n WeiohtSTD %ID/pSTD/ %ID/oroSTD% T/NT STD
WT ID/4 ID/Or4 T/NT

Tumor 5 0.4470.05411.311.38 5.050.75 1.00 0.00 Liver 5 1.2230.1970.28 0.09 0.330.06 42.248.25 Spleen 5 0.0810.0180.28 0.11 0.020.01 44.1513.06 Kidney 5 0.1390.0161.41 0.21 0.200.02 8.06 0.83 Lungs 5 0.1530.0260.19 0.08 0.030.01 67.4820.75 Blood 5 0.2410.0020.19 0.06 0.290.07 61.679.61 Stomach5 0.4930.1600.04 0.01 0.020.01 344.86101.23 Sm Int 5 1.1240.2350.08 0.04 0.080.03 168.2253.20 Lg Int 5 0.8750.2620.08 0.02 0.070.02 141.8233.27 8~

Table 4 Time post3 hr Injection:

6tperiment Pretargeting IMP-281 Descripficn: Bio with vs In-111-IMP-241 IMP-284 vs Group IA MAb #: ('u1 1 --> isotope 24 injected:
hrs I-125 ->"'In-IMP241) uCi injected: 6 MAb 1 injected:
hMN-14 x m679 Corrected 9892972 MAb 1 cpm injected:

Mean Body 20.09 weight:

Tissue n WeightSTD WT STD% %ID/orgSTD/ TINTSTDT/NT
%ID/g IDIg ID/Org Tumor 5 0.4890.065 1.50 2.730.62 1.000.00 5.67 Liver 5 1.0690.154 0.12 0.600.12 10.363.25 0.57 Spleen 5 0.0830.009 0.12 0.050.01 8.762.05 0.65 Kidney 5 0.1310.020 0.06 0.050.01 14.363.80 0.40 Lungs 5 0.1390.020 0.04 0.050.01 14.494.16 0.40 Blood 5 0.2330.003 0.07 0.840.13 10.162.89 0.57 Stomach 5 0.3370.046 0.86 0.940.21 2.211.06 2.87 Sm Int 5 0.9620.176 0.05 0.240.06 23.848.09 0.25 Lg Int 5 0.8140.133 0.07 0.290.05 16.184.96 0.36 MAb 2 isotope injected: In-111 MAb 2 cpm injected: 6151305 MAb 2 injected: IMP-241 Tissuen WeightSTD % STD/ %IDlorgSTD% TINT STD
WT ID/g ID/g ID/org TINT

Tumor 5 0.4890.06517.926.32 8.622.65 1.00 0.00 Liver 5 1.0690.1540.38 0.11 0.400.06 47.7114.04 Spleen5 0.0830.0090.31 0.07 0.020.00 57.9511.64 Kidney5 0.131D.0202.78 0.25 0.360.04 6.38 1.78 Lungs 5 0.139O.D200.54 0.10 0.070.01 33.5410.60 Blood 5 0.2330.0030.76 0.17 1.110.18 23.746.56 Stomach5 0.3370.0460.08 0.02 0.030.00 226.8373.39 ' Sm 5 0.9620.1760.16 0.05 0.150.03 120.6046.41 Int Lg 5 0.8140.1330.22 0.09 0.170.06 88.8431.75 Int Table 5 Time post Injection: 24 hr 6cperiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs IMP-Group IB
#:

uCi injected: 8.8 Isotope MAb MP-241 injected: injected:
In-111 I

CPM Injected: 6296515 Mean 19.89 Body weight:

Tissue n WeightSTD %ID/qSTD %ID/orgSTD TINT STD
WT %IDIg %lo~org TINT

Tumor 5 0.3600.0860.0380.0130.0150.008 1.0000.000 Liver 5 1.0080.1250.0710.0160.0700.010 0.5420.154 Spleen 5 0.0670.0150.0370.0090.0020.001 1.0260.196 Kidney 5 0.1400.0391.8570.3460.2550.056 0.0210.006 Lung 5 0.1370.0120.0180.0020.0020.000 2.0780.536 Blood 5 0.2360.0030.0000.0000.0000.000 569.530196.879 Stomach 5 0.3780.0740.0400.0260.0150.008 1.1400.604 Smalllnt.5 0.9520.2040.0440.0190.0390.011 0.8980.136 Large 5 0.7790.1810.1560.0470.1210.042 0.2610.124 Int.

All blood CPMs were 0 All spleen CPMs were 200 or less All lung CPMs were 170 or less Table 6 Time post Injection: 3 hr Experiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs IMP-Group #: IB
uCi injected: 8.8 Isotope injected: In-111 MAb injected: IMP-241 CPM Injected: 7771826.6 Mean Body weight: 18.08 Tissue n WeightSTD / STD % STD TINT STD
WT ID/g %ID/g ID/org/ TINT
ioior9 Tumor 5 0.3760.1270.2090.120 0.0660.008 1.0000.000 Liver 5 0.8750.1300.1490.012 0.1290.011 1,4250.874 Spleen 5 0.0660.0140.0780.007 0.0050.001 2.7381.701 Kidney 5 0.1130.0144.3270.559 0.4880.081 0.0470.020 Lung 5 0.1290.0170.1140.043 0.0150.007 2.0981.561 Blood 5 0.2320.0030.0100.003 0.0130.003 22.73614.408 Stomach5 0.2630.0340.1280.091 0.0320.022 2.4301.763 Smalllnt.5 0.8180.1130.2930.389 0.2290.301 1.7541.473 Large 5 0.6890.1730.3080.089 0.2010.022 0.7560.523 Int.

All blood CPMs were 250 or less Table 7 Time post 24 Injection:hr 6cperiment Pretargeting11-IMP-241iMP-281 Description: Bio with vs vs In-1 IMP-284 Group IIA MAb #: 11251 1 -> isotope 24 injected:
hrs I-125 -a~i~ln-IMP-281) uCi injected:6 MAb injected:
hMN-14 x m679 Corrected 9788320 MAb 1 cpm injected:

Mean Body18.92 weight:

Tissue n WeightSTD WT STD% %ID/orgSTD% TINTSTD
% ID/g ID/g ID/org TINT

Tumor 5 0.5900.042 3.510.74 2.050.36 1.000.00 Liver 5 0.9190.060 0.250.02 0.230.01 14.082.06 Spleen 5 0.0700.005 0.260.03 0.020.00 13.493.14 Kidney 5 0.1260.016 0.160.03 0.020.00 22.052.77 Lungs 5 0.1490.016 0.190.04 0.030.00 19.092.82 Blood 5 0.2390.002 0.210.05 0.290.05 16.631.19 Stomach 5 0.3340.041 0.840.43 0.280.14 4.581.27 Sm Int 5 0.8820.064 0.080.02 0.070.02 44.144.36 Lg Int 5 0.7930.090 0.100.04 0.080.04 35.487.79 MAb 2 isotope injected: In-111 MAb 2 cpm injected: 6659738.3 MAb 2 injected: IMP-281 Tissuen WeightSTD %ID/gSTD% / STD% TINT STD
WT ID/g ID/orgID/Ofg T/NT

Tumor 5 0.5900.04212.662.55 7.411.30 1.00 0.00 Liver 5 0.9190.0600.28 0.07 0.260.05 45.216.11 Spleen5 0.0700.0050.23 0.04 0.020.00 54.6812.53 Kidney5 0.1260.0161.09 0.22 0.140.03 12.214.51 Lungs 5 0.1490.0160.18 0.06 0.030.01 74.2614.28 Blood 5 0.2390.0020.17 0.05 0.230.05 76.2610.04 Stomach5 0.3340.0410.04 0.01 0.010.01 299.0955.84 Sm 5 0.8820.0640.06 0.02 0.060.02 207.3442.11 Int Lg 5 0.7930.0900.13 0.11 0.110.10 126.6755.69 Int Table 8 Time post 3 hr Injection:

6cperiment Pretargeting IMP-281 Description: Bio with vs In-111-IMP-241 IMP-284 vs Group IIA MAbisotope 125 #: (~zsl 1 injected:
--> I-hrs -a~~iln-IMP-281) uCi injected: 6 MAb 1 MN-14 injected:x h m679 Corrected 9853618 MAb 1 cpm injected:

Mean Body 19.01 weight:

Tissue n WeightSTD WT STD% %ID/OrqSTD% TINTSTD
%10/q ID/Q IDlorg TINT

Tumor 4 0.5290.049 5.380.82 2.850.53 1.000.00 Liver 4 0.8740.148 0.900.21 0.770.10 6.121.02 Spleen 4 0.0700.010 1.250.33 0.090.02 4.380.56 ~

Kidney 4 0.1190.011 0.610.15 0.070.01 9.101.40 Lungs 4 0.1380.033 0.650.23 0.080.01 8.741.68 Blood 4 0.2400.002 0.890.26 1.240.26 6.301.03 Stomach 4 0.3370.066 6.983.33 2.261.00 0.940.50 Sm Int 4 0.9800.153 0.360.11 0.340.08 15.873.44 Lg Int 4 0.6940.137 0.590.19 0.400.13 9.832.94 MAb 2 isotope injected: In-111 MAb 2 cpm injected: 8149181.6 MAb 2 injected: IMP-281 Tissue n WeinhtSTD %10/gSTD% %IDIOrqSTD% TINT STD
WT ID/Q ID/Ofp TINT

Tumor 4 0.5290.04917.925.28 9.492.87 1.00 0.00 Liver 4 0.8740.1480.36 0.09 0.300.04 49.602.59 Spleen 4 0.0700.0100.37 0.11 0.030.01 48.462.10 Kidney 4 0.1190.0112.25 0.41 0.270.05 7.95 1.70 Lungs 4 0.1380.0330.56 0.13 0.070.01 31.883.82 Blood 4 0.2400.0020.89 0.28 1.250.29 20.281.19 Stomach4 0.3370.0660.08 0.03 0.030.00 224.0247.19 Sm Int 4 0.9800.1530.14 0.04 0.140.03 129.7537.91 Lg Int 4 0.6940.1370.39 0.38 0.230.17 69.6843.20 Table 9 Time post Injection: 24 hr Experiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs IMP-Group #: 118 uCi injected: 8.8 Isotope injected: In-111 MAb injected: IMP-281 CPM Injected: 8605706.6 Mean Body weight: 20.69 Tissue n WeightSTD / STD / STD TINT STD
WT ID/g %IDIg ID/org/ TINT
iW
ors Tumor 5 0.3060.0850.0260.002 0.0080.002 1.0000.000 Liver 5 1.0850.1360.0350.005 0.0380.006 0.7530.102 Spleen 5 0.0790.0110.0320.003 0.0030.001 0.8050.082 Kidney 5 0.1310.0101.0680.133 0.1400.017 0.0240.003 Lung 5 0.1410.0090.0170.005 0.0020.001 1.6600.423 Blood 5 0.2390.0010.0000.000 0.0000.000 346.489269.842 Stomach5 0.3560.0800.0070.002 0.0030.001 3.8161.387 ' Smalllnt.5 0.9640.1220.0170.002 0.0160.002 1.5430.246 Large 5 0.8470.0750.0310.008 0.0260.008 0.9070.320 Int.

All Blood CPMs are 15 or less All lung CPMs are 300 or less Table 10 Time post Injection: 3 hr F~cperiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs Group #: IIB
uCi injected: 8.8 Isotope injected: In-111 MAb injected: IMP-281 CPM Injected: 10691556.6 Mean Body weight: 18.06 Tissue n Wei4htSTD %1D/4STD%ID/g%ID/orgSTDilolor~T/NT STDT/NT
WT

Tumor 5 0.3010.0620.1000.034 0.0290.008 1.0000.000 Liver 5 0.8770.1480.0600.009 0.0520.008 1.6670.541 Spleen 5 0.0700.0150.0400.005 0.0030.001 2,4810.722 Kidney 5 0.1150.0122.1330.668 0.2460.085 0.0470.014 Lung 5 0.1310.0160.0370.007 0.0050.001 2.6180.528 Blood 5 0.2430.0020.0040.002 0.0060.003 26.5109.697 Stomach5 0.3610.0970.0320.013 0.0110.006 3.7442.828 Smalllnt.5 0.8190.1400.0820.045 0.0690.046 1.6271.183 Large 5 0.6110.0620.2950.119 0.1780.065 0.4180.278 Int.

All Blood CPMs are 180 or less Table 11 Time post Injection: 30 min F~cperiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs Group IIB
#:

uCi injected: 8.8 Isotope : In-111 MAb MP-281 injected injected:
I

CPM Injected: 10598413.3 Mean 17.89 Body weight:

Tissue n WeightSTD %ID/gSTD % STD T/NT STD
WT %ID/g ID/org%W TINT
lore Tumor 3 0.2580.0212.2980.052 0.5940.052 1.0000.000 Liver 3 0.9820.2010.2700.087 0.2660.101 9.0752.628 Spleen 3 0.0800.0180.2570.081 O.o200.008 9.6293.393 Kidney 3 0.1130.0074.9841.280 0.5580.109 0.4810.121 Lung 3 0.1350.0290.6640.139 0.0880.018 3.5820.870 Blood 3 0.2450.0020.8540.290 1.1410.450 2.9341.093 Stomach 3 0.3930.0940.1710.079 0.0630.024 16.75810.840 Smalllnt.3 0.8880.1610.4410.112 0.3870.103 5.4791.562 Large 3 0.6550.0980.2400.196 0.1690.161 14.2238.914 Int.

Table 12 Time post 24 Injection:hr Fxperiment Pretargeting11-IMP-241IMP-281 Description: Bio with vs vs In-1 IMP-264 Group IIIA MAbsotope #: j'~51-a 1 injected:
24 i I-125 hrs -->"'InIMP-284) uCi injected: 6 MAb MN-14m679 1 injected:x h Corrected 9788320 MAb 1 cpm injected:

Mean Body 19.84 weight:

Tissue n WeiohtSTD WT STD% %ID/orgSTD% TINTSTD
%ID/g IDIg IDlorg TINT

Tumor 5 0.6050.049 3.460.75 2.110.54 1.000.00 Liver 5 1.0190.172 0.260.07 0.260.03 13.794.49 Spleen 5 0.0750.015 0.280.05 0.020.00 12.443.45 Kidney 5 0.1260.019 0.160.02 0.020.00 21.374.88 Lungs 5 0.1500.022 0.160.02 0.020.00 21.885.72 Blood 5 0.2420.002 0.200.03 0.300.03 17.284.44 Stomach 5 0.4010.043 0.700.24 0.280.08 5.362.14 Sm Int 5 0.8920.134 0.120.06 0.100.04 35.6716.07 Lg Int 5 0.6970.056 0.110.03 0.060.03 34.7713.25 MAb 2 isotope injected: In-111 MAb 2 cpm injected: 4317976.6 MAb 2 injected: IMP-284 Tissuen WeightSTD %IDIoSTD% %IDIOrgSTD% TINT STD
WT ID/g ID/org TINT

Tumor 5 0.6050.04911.582.17 7.001.45 1.00 0.00 Liver 5 1.0190.1720.40 0.08 0.400.05 29.767.51 Spleen5 0.0750.0150.40 0.11 0.030.00 30.256.78 Kidney5 0.1260.0191.80 0.16 0.220.02 6.45 1.18 Lungs 5 0.1500.0220.21 0.04 0.030.00 55.5711.50 Blood 5 0.2420.0020.22 0.04 0.330.05 52.9512.65 Stomach5 0.4010.0430.04 0.02 0.020.01 303.88103.30 Sm 5 0.8920.1340.08 0.02 0.070.01 155.5434.18 Int Lg 5 0.6970.0560.07 0.01 0.050.01 166.5242.08 Int Table 13 Time post 3 hr Injection:

F~cperiment Pretarge6ng11-IMP-241IMP-281 Description: Bio with vs vs In-1 IMP-284 Group IIIA MAb #: (i~sl 1 -a isotope 24 injected:
hrs 1-125 ->~~~In-IMP-284) uCi injected: 6 MAb MN-14 1 injected:x h m679 Corrected 9853618 MAb 1 cpm injected:

Mean Body 19.39 weight:

Tissue n WeightSTD WT STD% %ID/orgSTD/ T/NT STDT/NT
%IDI4 ID/g ID/Of(1 Tumor 5 0.5710.103 5.061.20 2.890.84 1.00 0.00 Liver 5 0.9010.088 0.730.12 0.660.07 7.13 2.27 Spleen 5 0.0620.011 1.500.60 0.090.02 3.74 1.32 Kidney 5 0.1240.004 0.500.09 0.060.01 10.523.36 Lungs 5 0.1370.010 0.510.09 0.070.01 10.393.38 Blcod 5 0.2340.002 0.770.21 1.100.28 7.13 2.86 Stomach 5 0.4320.163 3.451.91 1.320.53 2.03 1.54 Sm Int 5 0.9060.178 0.320.07 0.290.06 16.605.97 Lg Int 5 0.7160.122 0.450.20 0.320.12 13.567.60 MAb 2 isotope injected: In-111 MAb 2 cpm injected: 5380911.6 MAb 2 injected: IMP-284 Tissue n WeightSTD %ID/gSTD/ %ID/orctSTD% TINT STD
WT ID/g ID/aro TINT

Tumor 5 0.5710.10316.722.35 9.562.20 1.00 0.00 Liver 5 0.9010.0880.46 0.17 0.410.11 39.4712.02 Spleen 5 0.0620.0110.82 0.42 0.050.02 23.558.10 Kidney 5 0.1240.0042.51 0.51 0.310.06 6.87 1.61 Lungs 5 0.1370.0100.68 0.10 0.090.01 24.873.94 Blood 5 0.2340.0021.13 0.40 1.610.53 16.426.27 Stomach5 0.4320.1630.08 0.02 0.030.01 209.9252.53 Sm Int 5 0.9060.1780.17 0.07 0.140.02 109.5935.41 Lg Int 5 0.7160.1220.18 0.05 0.130.02 100.9745.57 Table 14 Time post Injection: 24 hr 6cperiment Description: Pretargeting Bio with in-111-IMP-241 vs IMP-281 vs IMP-Group IIIB
#:

uCi injected: 8.8 Isotope : in-111 MAb injected injected:

CPM Injected: 5380911.6 Mean 19.71 Body weight:

Tissue n WeightSTD %ID/QSTD %ID/orqSTD TINT STD
WT %ID/Q .sl0~om TINT

Tumor 5 0.3150.0530.0530.013 0.0160.004 1.0000.000 Liver 5 1.0610.1030.0950.005 0.1010.014 0.5590.134 Spleen 5 0.0700.0040.0710.009 0.0050.001 0.7440.161 Kidney 5 0.1300.0102.4370.254 0.3170.047 0.0220.006 Lung 5 0.1310.0100.0470.005 0.0060.001 1.1410.348 Blood 5 0.2350.0040.0040.002 0.0060.002 15.1679.733 Stomach 5 0.3670.0660.0270.016 0.0100.006 2.4411.365 Smalllnt.5 0.9700.0710.0380.012 0.0360.011 1.4640.429 Large 5 0.8290.0930.0710.032 0.0600.033 0.8080.253 Int.

All Blood CPMs are 75 or less All Spleen CPMs are 375 or less All Lung CPMs are 350 or less ~6 Table 15 Time post Injection: 3 hr Experiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs IMP-Group IIIB
#:

uCi injected: 8.8 Isotope : In-111 MAb injected injected:

CPM Injected: 6768496.6 Mean 19.16 Body weight:

Tissue n WeightSTD %IDIgSTD %IDlorgSTD TINT STD
/ ) WT %ID/g Woror9 TINT

Tumor 5 0.3060.0760.2550.081 0.0750.023 1.0000.000 L-~

Liver 5 0.9080.0500.1550.024 0.1400.022 1.7080.709 Spleen 5 0.0760.0070.0770.013 0.0060.001 3.5071.588 Kidney 5 0.1220.0124.3590.531 0.5330.086 0.0590.018 Lung 5 0.1510.0190.1490.135 0.0220.019 2.7321.914 Blood 5 0.2350.0020.0210.026 0.0290.036 23.72513.540 Stomach 5 0.2990.0690.0780.032 0.0220.009 3.4830.863 Smalllnt.5 0.9100.0590.1380.040 0.1270.041 2.0020.884 Large 5 0.6460.0710.5220.138 0.3360.094 0.4940.099 Int.

Table 16 Time post Injection: 30 min Experiment Description: Pretargeting Bio with In-111-IMP-241 vs IMP-281 vs IMP-Group IIIB
#:

uGi injected: 8.8 Isotope : In-111 MAb injected injected:

CPM Injected: 6768496.6 Mean 19.92 Body weight:

Tissue n WeightSTD %ID/gSTD %ID/orgSTD TINTSTD
WT / ilororg TINT
ID/g Tumor 2 0.2940.0162.5150.590 0.7430.214 1.0000.000 Liver 2 0.9390.0720.5060.194 0.4820.219 5.1220.797 Spleen 2 0.0850.0180.5200.171 0.0460.024 4.9150.482 Kidney 2 0.1270.0036.7050.699 0.8510.070 0.3730.049 Lung 2 0.1480.0021.3100.367 0.1930.051 1.9330.091 Blood 2 0.2330.0011.7250.965 2.5601.487 1.6150.561 Stomach 2 0.4300.0780.2850.170 0.1160.051 9.9803.876 Smalllnt.2 1.0320.0010.4460.175 0.4600.180 5.8270.968 Large 2 0.5790.0320.3390.134 0.1940.067 7.6701.289 Int.

As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof.
Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as "up to," "at least,"
"greater than," "less than," "more than" and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. In the same manner, all ratios disclosed herein also include all subratios falling within the broader ratio.

One skilled in the art will also readily recognize that where members are grouped together in a common manner, such as in a Markush group, the present invention encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group.
Accordingly, for all purposes, the present invention encompasses not only the main group, but also the main group absent one or more of the group members. The present invention also envisages the explicit exclusion of one or more of any of the group members in the claimed invention.
It will be apparent to those skilled in the art that various modifications and variations can be made to the compositions and processes of this invention.
Thus, it is intended that the present invention cover such modifications and variations, provided they come within the scope of the appended claims and their equivalents.
The disclosure of all publications disclosed herein are expressly incorporated herein by reference in their entireties to the same extent as if each were incorporated by reference individually.
Additional references of interest include the following:
Arano Y, Uezono T, Akizawa H, Ono M, Wakisaka K, Nakayama M, Sakahara H, Konishi J, Yokoyama A., "Reassessment of diethylenetriaminepentaacetic acid (DTPA) as a chelating agent for indium-111 labeling of polypeptides using a newly synthesized monoreactive DTPA
derivative," J
Med Chem. 1996 Aug 30; 39(18):3451-60.
Bamias, A., and Epenetos,A.A. Two-step strategies for the diagnosis and treatment of cancer with bioconjugates. Antibody, Immunoconjugates, Radioplaa~m.
1992; S: 385-395.
Barbet, J., Pettier, P., Bardet, S., Vuillez, JP., Bachelot, L, Denet, S., Olivier, P., Lecia, F., Corcuff, B., Huglo, D., Proye,C., Rouvier, E., Meyer,P., Chatal,J.F.
Radioimmunodetection of rnedullary thyroid carcinoma using indium-111 bivalent hapten and anti-CEA x anti-DTPA-indium bispecifc antibody. J.Nucl.Med. 1998;
39:1172-1178.
Bos, ES., Kuijpers, WHA., Meesters-Winters, M., Pham, DT., deHaan, AS., van Doormalen,Am., Kasperson,F.M.,vanBoeckel, CAA and Gouegeon-Bertrand, F.

In vitro evaluation of DNA-DNA hybridization as a two-step approach in radioimmunotherapy of cancer. Cafacer Res. 1994; 54:3479-3486.
Carr et al., WO00/34317.
Gautherot, E., Bouhou, J., LeDoussal, J-M., Manetti, C., Martin, M., Rouvier, E., Barbet, J. Therapy for colon carcinoma xenografts with bi-specific antibody-targeted, iodine-131-labeled bivalent hapten. Gancer suppl. 1997; 80: 2618-2623.
Gautherot, E., Bouhou, J., Loucif, E., Manetti, C., Martin, M., LeDoussal, J.M., Rouvier, E., Barbet, J. Radioimmunotherapy of LS 174T colon carcinoma in nude mice using an iodine-131-labeled bivalent hapten combined with an anti-CEA x anti-indium-DTPA bi-specific antibody. .LNucl. Med. Suppl. 1997; 38: 7p.
Goodwin, D.A., Meares, CF., McCall, MJ., McTigue,M., Chaovapong, W.
Pre-targeted immunoscintigraphy of murine tumors with indium-111-labeled bifunctional haptens. J.Nucl.Med. 1988; 29:226-234.
Greenwood, F.C. and Hunter, W.M. The preparation of I-131 labeled human growth hormone of high specific radioactivity. Biocherra. 1963; 89:114-123.
Hawkins, G.A., McCabe, R.P., Kim, C.-H., Subramanian,R., Bredehorst, R., McCullers, G.A., Vogel,C.-W., Hanna, M.G.Jr., and Pomata, N. Delivery of radionuclides to pretargeted monoclonal antibodies using dihydrofolate reductase and methotrexate in an affinity system. Cancef° Res. 1993; 53: 2368-2373.
Kranenborg, M.h., Boerman, O.C., Oosterwijk-Wakka, j., weijert, M., Corstens, F., Oosterwijk, E. Development and characterization of anti-renal cell carcinoma x antichelate bi-specific monoclonal antibodies for two-phase targeting of renal cell carcinoma. Cancer Res.(suppl) 1995; 55: 5864s-5867s Losman M.J., Qu Z., Krishnan LS., Wang J., Hansen H.J., Goldenberg D.M., Leung S.O. Clin. CancerRes. 1999; 5(10 Suppl.):3101s-3105s.
Penefsky, H.S. A centrifuged column procedure for the measurement of ligand binding by beef heart F1. Part G. Methods Erazyrnol. 1979; 56:527-530.
Schulnnacher, J., Klivenyi,G., Matys,R., Stadler, M., Regiert, T., Hauser,H., Doll, J., Maier-Borst, W., Zoller, M. Multistep tumor targeting in nude mice using bi-specific antibodies and a gallium chelate suitable for immunocintigraphy with positron emission tomography. Cancer Res. 1995; 55, 115-123.

Sharkey, RM., Karacay, Griffiths, GL., Behr, TM., Blumenthal,RD., Mattes,MJ.,Hansen, HJ., Goldenberg. Development of a streptavidin-anti-carcinoembryonic ntigen antibody, radiolabeled biotin pretargeting method for radioimmunotherapy of colorectal cancer. Studies in a human colon cancer xenograft model. Bioconjugate Claena 1997; 8:595-604.
Stickney, DR., Anderson, LD., Slater, JB., Ahlem, CN.,Kirk, GA., Schweighardt, SA and Frincke, JM. Bifunctional antibody: a binary radiopharmaceutical delivery system for imaging colorectal carcinoma. Cayacen Res.
1991;51: 6650-6655.
All references cited herein are hereby incorporated herein by reference in their entireties.

Claims (151)

1. A compound comprising the formula:
X-R1-D-[Dpr, Orn or Lys](A)-R2(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5; or R1(X)-D-[Dpr, Orn or Lys](A)-R2(Z)-D-[Dpr, Orn or Lys](B)-R3(Y)-NR4R5, wherein:
X is a hard acid cation chelator, a soft acid cation chelator, or Ac-;
R1 is a covalent bond or one or more D-amino acids that can be the same or different;
R2 is a covalent bond or one or more D-amino acids that can be the same or different;
R3 is a covalent bond or one or more D-amino acids that can be the same or different;
Y is a hard acid canon chelator, a soft acid cation chelator or is absent;
Z is a hard acid cation chelator, a soft acid cation chelator or is absent;
A and B independently are haptens or hard acid cation chelators and can be the same or different; and R4 and R5 are independently selected from the group consisting of hard acid cation chelators, soft acid cation chelators, enzymes, therapeutic agents, diagnostic agents and H.
2. The compound of claim 1 wherein the compound comprises the formula:
X-R1-D-Lys(A)-R2-D-Lys(B)-R3(Y)-NR4R5.
3. The compound of claim 1, wherein R2 is a single D-amino acid.
4. The compound of claim 1 wherein R2 is two D-amino acids that can be the same or different.
5. The compound of claim 1 wherein R3 is a D-Lys and Y is a hard acid cation chelator or a soft acid cation chelator.
6. The compound of claim 1 wherein R2 is not a D-Lys.
7. The compound of claim 1, wherein A and B are independently selected from the group consisting of histamine-succinyl-glycine (HSG), DTPA, and a detectable label.
8. The compound of claim 1 wherein:
R1 is one or more D-amino acids that can be the same or different;
R2 is one or more D-amino acids that can be the same or different;
R3 is a covalent bond;
Y is absent; and A and B are haptens or hard acid cation chelators and can be the same or different.
9. The compound of claim 8 wherein A and B are haptens and can be the same or different.
10. The compound of claim 8 wherein R1 and R2 are single D-amino acids and can be the same or different.
11. The compound of claim 8 wherein R1 is selected from the group consisting of D-Tyr, D-Ala, D-Ser, D-Thr, D-Cys, D-Leu, D-Ile, D-Met, D-Gln, D-Asn, D-Val, and D-Phe.
12. The compound of claim 8 wherein R2 is selected from the group consisting of D-Asp, D-Glu and D-Tyr.
13. The compound of claim 8 wherein R4 and R5 are both H.
14. The compound of claim 8 wherein one of X, R4 and R5 is a hard acid cation chelator.
15. The compound of claim 8 wherein one of the remaining X, R4 and R5 is a soft acid cation chelator.
16. The compound of claim 8 wherein X is a hard acid cation chelator.
17. The compound of claim 16 wherein one of R4 and R5 is a hard acid cation chelator.
18. The compound of claim 14, wherein the hard acid cation chelator comprises a carboxylate or amine group.
19. The compound of claim 1, wherein the hard acid cation chelator is selected from the group consisting of NOTA, DOTA, DTPA, and TETA.
20. The compound of claim 8 wherein one of X, R4 and R5 is a soft acid cation chelator.
21. The compound of claim 1, wherein the soft acid cation chelator comprises a thiol group.
22. The compound of claim 21, wherein the soft acid cation chelator is selected from the group consisting of Tscg-Cys and Tsca-Cys.
23. The compound of claim 1 wherein one of R4 and R5 is a soft acid cation chelator and the remaining R4 or R5 is H.
24. The compound of claim 1 wherein:
X is Ac-;
A and B are hard acid cation chelators and can be the same or different;
R3 is a covalent bond; and Y is absent.
25. The compound of claim 1 wherein:
X is Ac-;
A and B are haptens and can the same or different;
R1 is a covalent bond; and Y is a soft acid cation chelator.
26. The compound of claim 1, wherein the compound is selected from the group consisting of:
DOTA-D-Asp-D-Lys(HSG)-D-Asp-D-Lys(HSG)-NH2 (IMP 271);
DOTA-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 277);
DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 288);
DOTA-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 281);
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 284);
DOTA-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 301) DOTA-D-Lys(HSG)-D-Ala-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-NH2 (IMP 302) DOTA-D-Tyr-D-Lys(HSG)-D-Glu-D-Lys(HSG)-D-Cys-NH2 (IMP 305) Ac-D-Lys(In-DTPA)-D-Tyr-D-Lys(In-DTPA)-D-Lys(Tscg-Cys)-NH2 MH+
1813 (IMP 297) HCO-CO-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2 (IMP 289); and
27. ~The compound of claim 1, wherein:
(i) no more than one of X, R4 and R5 is a hard acid cation chelator; and (ii) no more than one of X, R4 and R5 is a soft acid cation chelator.
28. ~The compound of claim 1, wherein the compound is selected from the group consisting of:
Ac-D-Phe-D-Lys(DOTA)-D-Tyr-D-Lys(DOTA)-NH2;
Ac-D-Phe-D-Lys(DTPA)-D-Tyr-D-Lys(DTPA)-NH2;
Ac-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(Tscg-Cys)-NH2;
DOTA-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(Tscg-Cys)-NH2;
Tscg-Cys-D-Phe-D-Lys(HSG)-D-Tyr-D-Lys(HSG)-D-Lys(DOTA)-NH2;
Tscg-D-Cys-D-Glu-D-Lys(HSG)-D-Glu-D-Lys(HSG)-NH2; and Ac-D-Cys-D-Lys(DOTA)-D-Tyr-D-Ala-D-Lys(DOTA)-D-Cys-NH2.
29. ~The compound of claim 1, wherein the compound further comprises at least one radionuclide.
30. ~The compound of claim 29, wherein the radionuclide is selected from the group consisting of 225Ac, 111Ag, 72As, 77As, 211At, 198Au, 199Au, 212Bi, 213Bi, 75Br, 76Br, 11C, 55Co, 62Cu, 64Cu, 67Cu, 166DY, 169Er, 18F, 52Fe, 59Fe, 67Ga, 68Ga, 154Gd, 155Gd, 156Gd, 157Gd, 158Gd, 166Ho, 120I, 123I, 124I, l25I, 131I, 110In, 111In, 194Ir, 177Lu, 51Mn, 52mMn, 99Mo, 13N, 15O, 32P, 33P, 211Pb, 212Pb, 109Pd, 149Pm, 142Pr, 143Pr, 223Ra, 82mRb, 186Re, 188Re, 189Re, 105Rh, 47Sc, 153Sm, 75Se, 83Sr, 89Sr, 161Tb, 94mTC, 94Tc, 99mTc, 86Y, ~
90Y,90Y, and 89Zr.
31. ~The compound of claim 1, wherein the hard acid cation chelator is chelated to a cation selected from the group consisting of Group IIa and Group IIIa metal cations.
32. The compound of claim 1, wherein the soft acid cation chelator is chelated to a cation selected from the group consisting of transition metals, Bi, lanthanides and actinides.
33. The compound of claim 32, wherein the cation is selected from the group consisting of Tc, Re, and Bi.
34. The compound of claim 1 wherein R4 or R5 is a therapeutic agent, diagnostic agent or enzyme.
35. The compound of claim 34, wherein the therapeutic agent, diagnostic agent or enzyme is covalently linked by a linker moiety.
36. The compound of claim 35, wherein the linker moiety comprises at least one amino acid.
37. The compound of claim 34, wherein the therapeutic agent comprises a drug, prodrug or toxin.
38. The compound of claim 37, wherein the prodrug is selected from the group consisting of epirubicin glucuronide, CPT-11, etoposide glucuronide, daunomicin glucuronide and doxorubicin glucuronide.
39. The compound of claim 37, wherein the toxin is selected from the group consisting of ricin, abrin, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtherin toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin.
40. The compound of claim 34, wherein the therapeutic agent comprises doxorubicin, SN-38, camptothecin, etoposide, methotrexate, 6-mercaptopurine or etoposide phosphate.
41. The compound of claim 34, wherein the diagnostic agent comprises one or more agents for photodynamic therapy.
42. The compound of claim 41, wherein the agent for photodynamic therapy is a photosensitizer.
43. The compound of claim 42, wherein the photosensitizer is selected from the group consisting of benzoporphyrin monoacid ring A (BPD-MA), tin etiopurpurin (SnET2), sulfonated aluminum phthalocyanine (A1SPc) and lutetium texaphyrin (Lutex).
44. The compound of claim 34, wherein the diagnostic agent comprises one or more image enhancing agents for use in magnetic resonance imaging (MRI).
45. The compound of claim 44, wherein the enhancing agent comprises Mn, Fe, La or Gd.
46. The compound of claim 34, wherein the diagnostic agent comprises one or more radiopaque or contrast agents for X-ray or computed tomography.
47. The compound of claim 46, wherein the radiopaque or contrast agents comprises barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric acid, iosulamide meglumine, iosemetic acid, iotasul, iotetric acid, iothalamic acid, iotroxic acid, ioxaglic acid, ioxotrizoic acid, ipodate, meglumine, metrizamide, metrizoate, propyliodone, or thallous chloride.
48. The compound of claim 34, wherein the diagnostic agent comprises one or more ultrasound contrast agents.
49. The compound of claim 38, wherein the ultrasound contrast agent comprises a liposome or dextran.
50. The compound of claim 49, wherein the liposome is gas-filled.
51. The compound of claim 34, wherein the enzyme is capable of converting a drug intermediate to a toxic form to increase toxicity of the drug at a target site.
52. A method of diagnosing, treating or both diagnosing and treating a disease or a condition that can lead to a disease comprising:
(A) administering a targetable construct comprising the compound of claim 1 to a subject having, or suspected of having, a disease or a condition, wherein the targetable construct comprises at least one diagnostic or therapeutic cation, andlor one or more chelated or chemically bound therapeutic agents, diagnostic agents, or enzymes; and (B) administering a multispecihc antibody or antibody fragment to the subject, wherein the multispecific antibody or antibody fragment has at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds the targetable construct.
53. The method of claim 52, further comprising:
(C) administering a clearing composition to the subject, wherein the clearing composition enhances clearance of non-localized antibodies or antibody fragments from the subject.
54. The method of claim 52, wherein the targetable construct and the multispecific antibody or antibody fragment are administered at substantially the same time.
55. The method of claim 52, wherein the therapeutic cation emits particles and/or positrons having 20 to 10,000 keV.
56. The method of claim 55, wherein the therapeutic cation is selected from the group consisting of 111In, 177Lu, 212Bi, 213Bi, 211At, 62Cu, 64Cu, 67Cu, 90Y, 125I, 131I, 32P, 33P, 47Sc, 111Ag, 67Ga, 142Pr, 153Sm, 161Tb, 166Dy, 166Ho, 186Re, 188Re, 189Re, 212Pb, 223Ra, 225Ac, 59Fe, 75Se, 77As, 89Sr, 99Mo, 105Rh, 109Pd, 143Pr, 149Pm, 169Er, 194Ir, 198Au, 199Au and 211Pb.
57. The method of claim of claim 56, wherein the diagnostic cation emits particles and/or positrons having 25-10,000 keV.
58. The method of claim 52, wherein the diagnostic cation is selected from the group consisting of 110In, 111In, 177Lu, 18F, 52Fe, 62Cu, 64Cu, 67Cu, 67Ga, 68Ga, 86Y, 90Y, 89Zr, 94mTc, 94Tc, 99mTc, 120I, 123I, 124I, 125I, 131I, 154-158Gd, 32P, 11C, 13N, 15O, 186Re, 188Re, 51Mn, 52mMn, 55Co, 72As, 75Br, 76Br, 82mRb and 83Sr.
59. The method of claim 52, wherein the diagnostic agent is a diagnostic agent used in positron-emission tomography (PET).
60. The method of claim 52, wherein the diagnostic agent is a diagnostic agent used in SPECT imaging.
61. The method of claim 52, wherein the diagnostic cation or agent comprises one or more image enhancing agents used in magnetic resonance imaging (MRI).
62. The method of claim 61, wherein the enhancing agent is selected from the group consisting of Mn, Fe, La and Gd.
63. The method of claim 52, wherein the diagnostic agent comprises one or more radiopaque or contrast agents for X-ray or computed tomography.
64. The method of claim 63, wherein the radiopaque or contrast agent is selected from the group consisting of barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, iogulamide, iohexol, iopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric acid, iosulamide meglumine, iosemetic acid, iotasul, iotetric acid, iothalamic acid, iotroxic acid, ioxaglic acid, ioxotrizoic acid, ipodate, meglumine, metrizamide, metrizoate, propyliodone, and thallous chloride.
65. The method of claim 52, wherein the diagnostic agent comprises one or more ultrasound contrast agents.
66. The method of claim 65, wherein the ultrasound contrast agent comprises a liposorne or dextran.
67. The method of claim 66, wherein the liposome is gas-filled.
68. The method of claim 52, wherein the one or more diagnostic agents are selected from the group consisting of fluorescent compounds, chemiluminescent compounds, and bioluminescent compounds.
69. The method of claim 68, wherein the fluorescent compounds are selected from the group consisting of fluorescein, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.
70. The method of claim 68, wherein the chemiluminescent compounds are selected from the group consisting of luminol, isoluminol, an aromatic acridinium~
ester, an imidazole, an acridinium salt and an oxalate ester.
71. The method of claim 68, wherein the bioluminescent compounds are selected from the group consisting of luciferin, luciferase and aequorin.
72. The method of claim 52, wherein the targeted tissue is a tumor.
73. The method of claim 72, wherein the tumor produces or is associated with antigens selected from the group consisting of colon-specific antigen-p (CSAp), carcinoembryonic antigen (CEA), CD19, CD20, CD21, CD22, CD23, CD 25, CD30, CD74, CD80, HLA-DR, Ia, MUC 1, MUC 2, MUC 3, MUC 4, EGFR, HER 2/neu, PAM-4, TAG-72, EGP-1, EGP-2, A3, KS-1, Le(y), 5100, PSMA, PSA, tenascin, folate receptor, VEGF, necrosis antigens, IL-2, T101, MAGE,1L-6, insulin-like growth factor receptor, carbonic anhydrase IX, and combinations thereof.
74. The method of claim 52, wherein the at least one arm that specifically binds the targeted tissue is a monoclonal antibody or a fragment of a monoclonal antibody.
75. The method of claim 52, wherein the at least one other arm that specifically binds the targetable construct is a monoclonal antibody or a fragment of a monoclonal antibody.
76. The method of claim 52 wherein the at least one arm that specifically binds the targeted tissue is a human, chimeric or humanized antibody or a fragment of a human, chimeric or humanized antibody.
77. The method of claim 52, wherein the at least one other arm that specifically binds the targetable construct is a human, chimeric or humanized antibody or a fragment of a human, chimeric or humanized antibody.
78. The method of claim 52, wherein the multispecific antibody or antibody fragment further comprises a therapeutic nuclide.
79. The method of claim 78, wherein the therapeutic nuclide is selected from the group consisting of 111In, 177Lu, 212Bi, 213Bi, 211At, 62Cu, 64Cu, 67Cu, 90Y, 125I, 131I, 32P, 33P, 47Sc, 111Ag, 67Ga, 142Pr, 153Sm, 161Tb, 166Dy, 166Ho, 186Re, 188Re, 189Re, 212Pb, 223Ra, 225Ac, 59Fe, 75Se, 77As, 89Sr, 99Mo, 105Rh, 109Pd, 143Pr, 149Pm, 169Er, 194Ir, 198Au, 199Au and 211Pb.
80. The method of claim 52, wherein the multispecific antibody comprises the Fv of MAb Mu-9 and the Fv of MAb 679.
81. The method of claim 80, wherein Mu-9 and/or 679 are chimerized or humanized.
82. The method of claim 80, wherein Mu-9 and/or 679 are human Mu9 and 679.
83. The method of claim 80, wherein the multispecific antibody comprises one or more of the CDRs of Mu-9.
84. The method of claim 80, wherein the multispecific antibody comprises one or more of the CDRs of 679.
85. The method of claim 80, wherein the multispecific antibody is a fusion protein.
86. The method of claim 52, wherein the multispecific antibody comprises the Fv of MAb MN-14 and the Fv of MAb 679.
87. The method of claim 86, wherein MN-14, and/or 679 are chimerized or humanized.
88. The method of claim 86, wherein MN-14, and/or 679 are human MN-14 and 679.
89. The method of claim 86, wherein the multispecific antibody comprises one or more of the CDRs of MN-14.
90. The method of claim 86, wherein the multispecific antibody comprises one or more of the CDRs of 679.
91. The method of claim 86, wherein the multispecific antibody is a fusion protein.
92. The method of claim 52, wherein the fusion protein is trivalent, and incorporates the Fv of an antibody reactive with CSAp.
93. The method of claim 52, wherein the multispecific antibody incorporates a Class-III anti-CEA antibody and the Fv of 679.
94. The method of claim 52, wherein the targetable construct comprises one or more radioactive isotopes that is capable of killing diseased tissue.
95. The method of claim 52, wherein the targetable construct comprises 10B atoms and the method further comprises:
(C) irradiating the 10B atoms localized at the targeted tissue, thereby effecting boron neutron capture therapy of the targeted tissue.
96. The method of claim 52, wherein the targetable construct comprises an enzyme and the method further comprising:
(C) administering a drug to the subject, wherein the enzyme is capable of converting the drug a toxic form thereby increasing the toxicity of the drug at the targeted tissue.
97. A method for detecting, identifying or treating a target cell, tissue or pathogen in a subject; comprising:
(a) administering a targetable construct comprising the compound of claim 1 to a subject; and (b) administering a multispecific antibody or antibody fragment to the subject, wherein the multispecific antibody or antibody fragment comprises at least one arm that specifically binds a target cell, tissue or pathogen and at least one other arm that specifically binds the targetable construct.
98. The method of claim 97, wherein the target comprises a molecule produced by or associated with the target cell, tissue, or pathogen.
99. The method of claim 98, wherein the target tissue is a diseased tissue.
100. The method of claim 99, wherein the diseased tissue is identified intraoperatively, endoscopically or intravascularly.
101. The method of claim 98, wherein the target tissue is normal tissue.
102. The method of claim 101, wherein the targeted tissue is ovary, thymus, parathyroid, endometrium, bone marrow, or spleen.
103. The method of claim 98, wherein the pathogen is a fungus, virus, parasite, bacterium, protozoan, or mycoplasma.
104. The method of claim 103, wherein the fungus is selected from the group consisting of Microsporum, Trichophyton, Epidermophyton, Sporothrix schenckii, Cyrptococcus neoformans, Coccidioides immitis, Histoplasma capsulatum, Blastomyces dermatitidis, and Candida albicans.
105. The method of claim 103, wherein the virus is selected from the group consisting of human immunodeficiency virus (HIV), herpes virus, cytomegalovirus, rabies virus, influenza virus, hepatitis B virus, Sendai virus, feline leukemia virus,.
Reo virus, polio virus, human serum parvo-like virus, simian virus 40, respiratory syncytial virus, mouse mammary tumor virus, Varicella-Zoster virus, Dengue virus, rubella virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus and blue tongue virus.
106. The method of claim 103, wherein the bacterium is selected from the group consisting of Anthrax bacillus, Streptococcus agalactiae, Legionella pneumophilia, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Hemophilia influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, Mycobacterium tuberculosis and Tetanus toxin.
107. The method of claim 103, wherein the parasite is a helminth or a malarial parasite.
108. The method of claim 103, wherein the protozoan is selected from the group consisting of Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosoma rangeli, Trypanosoma cruzi, Trypanosoma rhodesiensei, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japanicum, Babesia bovis, Elmeria tenella, Onchocerca volvulus, Leishmania tropica, Trichinella spiralis, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus and Mesocestoides corti.
109. The method of claim 103, wherein the mycoplasma is selected from the group consisting of Mycoplasma arthritidis, Mycoplasma hyorhinis, Mycoplasma orate, Mycoplasma arginini, Acholeplasma laidlawii, Mycoplasma salivarum, and Mycoplasma pneumoniae.
110. The method of claim 98 wherein the targetable construct further comprises at least one radionuclide, therapeutic agent, diagnostic agent or enzyme.
111. The method of claim 110, wherein the radionuclide is selected from the group consisting of 225Ac, 111Ag, 72As, 77As, 211At, 198Au, 199Au, 212Bi, 213Bi, 75Br, 76Br, 11C, 55Co, 62Cu, 64Cu, 67Cu, 166Dy, 169Er, 18F, 52Fe, 59Fe, 67Ga, 68Ga, 154Gd, 155Gd, 156Gd, 157Gd, 158Gd, 166Ho, 120I, 123I, 124I, 125I, 131I, 110In, 111In, 194Ir, 177Lu, 51Mn, 52mMn, 99Mo, 13N, 15O, 32P, 33P, 211Pb, 212Pb, 109Pd, 149Pm, 142Pr, 143Pr, 223Ra, 82mRb, 186Re, 188Re, 189Re, 105Rh, 47Sc, 153Sm, 75Se, 83Sr, 89Sr, 161Tb, 94mTc, 86Y, 90Y, 90Y, and 89Zr.
112. The method of claim 110, wherein the diagnostic agent comprises an imaging agent.
113. The method of claim 112, further comprising:
(c) administering a clearing composition to the subject, wherein the composition enhances clearance of non-localized antibodies or antibody fragments from the subject.
114. The method of claim 98 wherein the subject is a mammal.
115. The method of claim 114, wherein the mammal is selected from the group consisting of humans, primates, equines, canines and felines.
116. The method of claim 110, wherein the therapeutic agent comprises one or more drugs, toxins, cytokines, hormones, or growth factors.
117. The method of claim 110, wherein the diagnostic agent comprises a contrast agent.
118. The method of claim 112, wherein the imaging agent is an agent used for PET.
119. The method of claim 112, wherein the imaging agent is an agent used for SPECT.
120. A kit for treating or identifying diseased tissues in a subject comprising:
(A) a targetable construct comprising the compound of claim 1; and (B) a multispecific antibody or antibody fragment having at least one arm that specifically binds a targeted tissue and at least one other arm that specifically binds the targetable construct.
121. The kit of claim 120, further comprising:
(C) a clearing composition for enhancing clearance of non-localized antibodies and antibody fragments.
122. The kit of claim 120, wherein the diagnostic agent selected from the group consisting of 110In, 111In, 177Lu, 18F, 52Fe, 62Cu, 64Cu, 67Cu, 67Ga, 68Ga, 86Y, 90Y, 89Zr, 94mTc, 94Tc, 99mTc, 120I, 123I, 124I, 125I, 131I, 154-158Gd, 32P, 11C, 13N, 15O, 186Re, 188Re, 51Mn, 52mMn, 55Co, 72As, 75Br, 76Br, 82mRb and 83Sr.
123. The kit of claim 120, wherein the therapeutic agent is selected from the group consisting of 111In, 177Lu, 212Bi, 213Bi, 211At, 62Cu, 64Cu, 67Cu, 90Y, 125I, 131I, 32P, 33P, 47Sc, 111Ag, 67Ga, 142Pr, 153Sm, 161Tb, 166Dy, 166Ho, 156Re, 188Re, 189Re, 212Pb, 223Ra, 225Ac, 59Fe, 75Se, 77As, 89Sr, 99Mo, 105Rh, 109Pd 143Pr, 149Pm, 169Er, 194Ir, 198Au, 199Au and 211Pb
124. The kit of claim 120, wherein said targetable construct comprises an enzyme and said kit further comprises a drug which said enzyme is capable of converting to a toxic form to increase the toxicity of said drug.
125. A targetable construct comprising the compound of claim 1.
126. The method of claim 52 wherein the multispecific antibody or antibody fragment is bispecific.
127. The method of claim 97 wherein the multispecific antibody or antibody fragment is bispecific.
128. The method of claim 52 wherein the disease or condition is selected from the group consisting, of cancer, an infectious disease, an inflammatory disease, and autoimmune disease, cardiovascular disease, a metabolic disease, and a neurological disease.
129. The method of claim 128 wherein the disease or condition is cancer.
130. The method of claim 129 wherein the cancer is selected from the group consisting of leukemias, lymphomas, sarcomas, melanomas, carcinomas, gliomas, and skin cancers.
131. The method of claim 129 wherein the cancer is a B-cell malignancy, a B-cell lymphoma, chronic lymphatic leukemia, non-Hodgkin's lymphoma, acute lymphatic leukemias, or a multiple myeloma.
132. The method of claim 129 wherein the cancer is esophageal, gastric, colonic, rectal, pancreatic, lung, breast, ovarian, urinary bladder, endometrial, cervical, testicular, renal, adrenal or liver cancer.
133. The method of claim 128 wherein the disease or condition is an infectious disease caused by a pathogen.
134. The method of claim 133 wherein the pathogen is a fungus, virus, parasite, bacterium, protozoan, or mycoplasm.
135. The method of claim 134 wherein the pathogen is a fungus selected-from the group consisting of Microsporum, Trichophyton, Epidermophyton, Sporothrix schenckii, Cryptococcus neoformans, Coccidioides immitis, Histoplasma capsulatum, Blastomyces dermatitidis, and Candida albicans.
136. The method of claim 134 wherein the pathogen is a virus selected from the group consisting of human immunodeficiency virus (HIV), herpes virus, cytomegalovirus, rabies virus, influenza virus, hepatitis B virus, Sendai virus, feline leukemia virus, Reo virus, polio virus, human serum parvo-like virus, simian virus 40, respiratory syncytial virus, mouse mammary tumor virus, Varicella-Zoster virus, Dengue virus, rubella virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, papilloma virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus, wart virus and blue tongue virus.
137. The method of claim 134 wherein the pathogen is a bacterium selected from the group consisting of Anthrax bacillus, Streptococcus agalactiae, Legionella pneumophilia, Streptococcus pyogenes, Staphylococcus, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Hemophilis influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, Mycobacterium tuberculosis and Clostridium tetani.
138. The method of claim 134 wherein the pathogen is a parasite selected from the group consisting of helminth and malarial parasites.
139. The method of claim 134 wherein the pathogen is a protozoan selected from the group consisting of Plasmodium falciparum, Plasmodium vivax, Toxoplasma gondii, Trypanosoma rangeli, Trypanosoma cruzi, Trypanosoma rhodesiensei, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japanicum, Babesia bovis, Elmeria tenella, Onchocerca volvulus, Leishmania tropica, Trichinella spiralis, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus and Mesocestoides corti.
140. The method of claim 134 wherein the pathogen is a mycoplasm selected from the group consisting of Mycoplasma arthritidis, Mycoplasma hyorhinis, Mycoplasma orate, Mycoplasma arginini, Acholeplasma laidlawii, Mycoplasma salivarum, and Mycoplasma pneumoniae.
141. The method of claim 128 wherein the disease or condition is an inflammatory disease.
142. The method of claim 141 wherein the inflammatory disease is an autoimmune disease.
143. The method of claim 142 wherein the autoimmune disease is selected from the group consisting of acute idiopathic thrombocytopenic purpura, chronic idiopathic thrombocytopenic purpura, dermatomyositis, Sydenham's chorea, myasthenia gravis, systemic lupus erythematosus, lupus nephritis, rheumatic fever, polyglandular syndromes, bullous pemphigoid, diabetes mellitus, Henoch-Schonlein purpura, post-streptococcalnephritis, erythema nodosurn, Takayasu's arteritis, Addison's disease, rheumatoid arthritis, multiple sclerosis, sarcoidosis, ulcerative colitis, erythema multiforme, IgA nephropathy, polyarteritis nodosa, ankylosing spondylitis, Goodpasture's syndrome, thromboangitisubiterans, Sjögren's syndrome, primary biliary cirrhosis, Hashimoto's thyroiditis,thyrotoxicosis, scleroderma, chronic active hepatitis, polymyositis/dermatomyositis, polychondritis, parnphigus vulgaris, Wegener's granulomatosis, membranous nephropathy, amyotrophic lateral sclerosis, tabes dorsalis, giant cell arteritis/polymyalgia, pernicious anemia, rapidly progressive glomerulonephritis, psoriasis, and fibrosing alveolitis.
144. The method of claim 128 wherein the disease or condition is cardiovascular disease
145. The method of claim 144 wherein the cardiovascular disease is selected from the group consisting of myocardial infarction, ischemic heart disease, atherosclerotic plaques, clots, and emboli.
146. The method of claim 128 wherein the disease or condition is a metabolic disease.
147. The method of claim 146 wherein the metabolic disease is amyloidosis.
148. The method of claim 128 wherein the disease or condition is a neurological disease.
149. The method of claim 148 wherein the neurological disease is Alzheimer's disease.
150. The compound of claim 1 wherein when R1 or R3 is a covalent bond then the other R1 or R3 is one or more D-amino acids that can be the same or different.
151. The compound of claim 1 wherein R2 is one or more D-amino acids that can be the same or different.
CA2529027A 2003-06-13 2004-06-14 D-amino acid peptides Expired - Fee Related CA2529027C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US47840303P 2003-06-13 2003-06-13
US60/478,403 2003-06-13
PCT/US2004/018646 WO2005021494A2 (en) 2003-06-13 2004-06-14 D-amino acid peptides

Publications (2)

Publication Number Publication Date
CA2529027A1 true CA2529027A1 (en) 2005-03-10
CA2529027C CA2529027C (en) 2013-09-10

Family

ID=34272436

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2529027A Expired - Fee Related CA2529027C (en) 2003-06-13 2004-06-14 D-amino acid peptides

Country Status (7)

Country Link
US (3) US7172751B2 (en)
EP (1) EP1633773A4 (en)
JP (1) JP4937751B2 (en)
AU (1) AU2004268932B2 (en)
CA (1) CA2529027C (en)
IL (1) IL172403A (en)
WO (1) WO2005021494A2 (en)

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7138103B2 (en) * 1998-06-22 2006-11-21 Immunomedics, Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
US7591994B2 (en) 2002-12-13 2009-09-22 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US8877901B2 (en) 2002-12-13 2014-11-04 Immunomedics, Inc. Camptothecin-binding moiety conjugates
CA2514277C (en) * 2003-01-31 2015-01-20 Immunomedics, Inc. Methods and compositions for administering therapeutic and diagnostic agents
US7172751B2 (en) * 2003-06-13 2007-02-06 Immunomedics, Inc. D-amino acid peptides
JP2006248978A (en) * 2005-03-10 2006-09-21 Mebiopharm Co Ltd New liposome preparation
JP4537924B2 (en) * 2005-09-29 2010-09-08 株式会社日立製作所 Accelerator system
EP2674440B1 (en) 2005-12-16 2019-07-03 IBC Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
CN100464189C (en) * 2006-04-21 2009-02-25 深圳市新产业生物医学工程有限公司 Magnetic separating direct chemical illuminating reagent and testing method using the same reagent
EP2037954A4 (en) * 2006-06-20 2011-09-14 Janssen Pharmaceutica Nv Method for modulating the pharmacokinetics and metabolism of a therapeutic agent
US20080031866A1 (en) * 2006-06-20 2008-02-07 Eichenbaum Gary M Method for modulating the pharmacokinetics and metabolism of a therapeutic agent
JP5263805B2 (en) * 2007-06-13 2013-08-14 国立大学法人群馬大学 Polymer complexes stabilized by amphiphilic polymer ligands and test and pharmaceutical compositions
DK2187965T3 (en) 2007-08-17 2020-01-20 Purdue Research Foundation PSMA BINDING LIGAND LINKER CONJUGATES AND PROCEDURES FOR USE
EP2396036B1 (en) 2009-02-13 2017-06-28 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
CA2782194C (en) * 2009-12-02 2018-01-16 Immunomedics, Inc. Combination of radiolabelled antibodies (rait) and antibody-drug conjugates (adc) for treatment of pancreatic cancer
US9951324B2 (en) 2010-02-25 2018-04-24 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
JP2014506883A (en) * 2011-02-03 2014-03-20 インディ モレキュラー,インコーポレイテッド PSA scavenger, composition, method and production thereof
RU2632647C2 (en) 2011-04-22 2017-10-06 Аптево Рисерч Энд Девелопмент Ллс Proteins binding specific membrane prostate antigen, and related compositions and methods
EA201890915A1 (en) 2012-11-15 2018-09-28 Эндосайт, Инк. CONJUGATES FOR DELIVERY OF MEDICINES AND METHODS OF TREATMENT OF DISEASES CAUSED BY CELLS EXPRESSING PSMA
WO2017004144A1 (en) 2015-07-01 2017-01-05 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
KR102356017B1 (en) 2012-12-13 2022-01-27 이뮤노메딕스, 인코오포레이티드 Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
EP3495355A1 (en) 2013-10-18 2019-06-12 Deutsches Krebsforschungszentrum Labeled inhibitors of prostate specific membrane antigen (psma), their use as imaging agents and pharmaceutical agents for the treatment of prostate cancer
EP3954373A1 (en) 2014-10-07 2022-02-16 Immunomedics, Inc. Neoadjuvant use of antibody-drug conjugates
US10188759B2 (en) 2015-01-07 2019-01-29 Endocyte, Inc. Conjugates for imaging
CA2983597A1 (en) 2015-06-25 2016-12-29 Immunomedics, Inc. Combining anti-hla-dr or anti-trop-2 antibodies with microtubule inhibitors, parp inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
EA201890613A1 (en) 2015-09-21 2018-10-31 Аптево Рисёрч Энд Девелопмент Ллс POLYPEPTIDES CONNECTING CD3
WO2017181791A1 (en) * 2016-04-19 2017-10-26 南京中硼联康医疗科技有限公司 Application using compound comprising 10b for preparing pharmaceutical product specifically binding amyloid β-protein
CA3036235A1 (en) 2016-09-09 2018-03-15 Irisys, Inc. Lipsomal anticancer compositions
WO2018070524A1 (en) * 2016-10-14 2018-04-19 潤平 笹部 SCREENING METHOD FOR BLOOD IgA PRODUCTION INHIBITOR OR FOR PROPHYLACTIC OR THERAPEUTIC AGENT AGAINST DISEASES CAUSED BY EXCESSIVE IgA IN BLOOD
IL310958A (en) 2021-09-03 2024-04-01 Oncoone Res & Development Gmbh IMPROVED Fc SILENCED ANTI-oxMIF ANTIBODIES WITH REDUCED AGGREGATION POTENTIAL AND REDUCED HYDROPHOBICITY

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5776093A (en) * 1985-07-05 1998-07-07 Immunomedics, Inc. Method for imaging and treating organs and tissues
US5101827A (en) * 1985-07-05 1992-04-07 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4735210A (en) * 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4863713A (en) * 1986-06-23 1989-09-05 The Board Of Trustees Of Leland Stanford Jr. Univ. Method and system for administering therapeutic and diagnostic agents
FR2604092B1 (en) * 1986-09-19 1990-04-13 Immunotech Sa IMMUNOREACTIVES FOR TARGETING ANIMAL CELLS FOR VISUALIZATION OR DESTRUCTION IN VIVO
US5328679A (en) * 1988-04-01 1994-07-12 Immunomedics, Inc. Methods for technetium/rhenium labeling of proteins
US5128119A (en) * 1989-06-12 1992-07-07 Immunomedics, Inc. Methods for technetium/rhenium labeling of f(ab1)2 fragments
FR2652004B1 (en) * 1989-09-21 1994-10-28 Immunotech Partners NOVEL HYDROPHILIC DERIVATIVES, APPLICATION TO DIAGNOSIS AND THERAPEUTICS, DIAGNOSTIC OR THERAPEUTIC KITS AND IMMUNOLOGICAL REAGENTS.
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6120768A (en) * 1993-05-17 2000-09-19 Immunomedics, Inc. Dota-biotin derivatives
IL113610A0 (en) * 1994-06-03 1995-08-31 Immunomedics Inc A method of radiolabeling a protein, radiolabeled proteins prepared thereby and diagnostic kits containing the same
US5746996A (en) * 1994-06-03 1998-05-05 Immunomedics, Inc. Thiolation of peptides for radionuclide-based radiodetection and radiotherapy
US5753206A (en) * 1995-06-07 1998-05-19 Immunomedics, Inc. Radiometal-binding analogues of luteinizing hormone releasing hormone
EP0954340B1 (en) * 1996-05-03 2007-06-27 Immunomedics, Inc. Targeted combination immunotherapy of cancer
EP0975374B1 (en) * 1996-07-12 2007-12-05 Immunomedics, Inc. Radiometal-binding peptide analogues
US6558669B1 (en) * 1996-08-28 2003-05-06 Immunomedics, Inc. Stable radioiodine conjugates and methods for their synthesis
US6663866B1 (en) * 1996-08-28 2003-12-16 Immunomedics, Inc. Stable radioiodine conjugates and methods for their synthesis
US7521531B2 (en) * 1996-08-28 2009-04-21 Immunomedics, Inc. Methods for the purification of stable radioiodine conjugates
US6071490A (en) * 1998-05-07 2000-06-06 Immunomedics, Inc. Position emission tomography using gallium-68 chelates
US7138103B2 (en) * 1998-06-22 2006-11-21 Immunomedics, Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
US7052872B1 (en) * 1999-06-22 2006-05-30 Immunomedics, Inc. Bi-specific antibodies for pre-targeting diagnosis and therapy
US6962702B2 (en) * 1998-06-22 2005-11-08 Immunomedics Inc. Production and use of novel peptide-based agents for use with bi-specific antibodies
LT2857516T (en) * 2000-04-11 2017-09-11 Genentech, Inc. Multivalent antibodies and uses therefor
EP1411987A2 (en) * 2001-07-31 2004-04-28 Immunomedics, Inc. Targeted polymeric delivery systems
AU2002365649A1 (en) * 2001-11-28 2003-06-17 Immunomedics, Inc. Anti-dota antibody
US7011816B2 (en) * 2001-12-26 2006-03-14 Immunomedics, Inc. Labeling targeting agents with gallium-68 and gallium-67
US6735111B2 (en) * 2002-01-16 2004-05-11 Micron Technology, Inc. Magnetoresistive memory devices and assemblies
EP1507562B1 (en) * 2002-05-29 2010-03-24 Immunomedics, Inc. Compositions for radioimmunotherapy of brain
US20040022726A1 (en) * 2002-06-03 2004-02-05 Goldenberg David M. Methods and compositions for intravesical therapy of bladder cancer
WO2003106497A1 (en) * 2002-06-14 2003-12-24 Immunomedics, Inc. Monoclonal antibody pam4 and its use for diagnosis and therapy of pancreatic cancer
AU2003248982B2 (en) * 2002-08-01 2009-12-10 Immunomedics, Inc. Alpha-fetoprotein immu31 antibodies and fusion proteins and methods of use thereof
US7541440B2 (en) * 2002-09-30 2009-06-02 Immunomedics, Inc. Chimeric, human and humanized anti-granulocyte antibodies and methods of use
US7172751B2 (en) * 2003-06-13 2007-02-06 Immunomedics, Inc. D-amino acid peptides

Also Published As

Publication number Publication date
WO2005021494A9 (en) 2005-07-14
CA2529027C (en) 2013-09-10
EP1633773A2 (en) 2006-03-15
US7776311B1 (en) 2010-08-17
US20070142296A1 (en) 2007-06-21
WO2005021494A3 (en) 2005-10-20
JP4937751B2 (en) 2012-05-23
IL172403A (en) 2011-06-30
AU2004268932B2 (en) 2010-06-24
US7521416B2 (en) 2009-04-21
IL172403A0 (en) 2006-04-10
JP2008504214A (en) 2008-02-14
EP1633773A4 (en) 2010-10-20
AU2004268932A1 (en) 2005-03-10
US20050025709A1 (en) 2005-02-03
US20100202961A1 (en) 2010-08-12
US7172751B2 (en) 2007-02-06
WO2005021494A2 (en) 2005-03-10

Similar Documents

Publication Publication Date Title
US7776311B1 (en) D-amino acid peptides
EP1506018B1 (en) Drug pre-targeting by means of bi-specific antibodies and hapten constructs comprising a carrier peptide and the active agent(s)
US6962702B2 (en) Production and use of novel peptide-based agents for use with bi-specific antibodies
CA2555666C (en) Therapeutic and diagnostic conjugates for use with multispecific antibodies

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20190614