US20220378956A1 - Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use - Google Patents

Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use Download PDF

Info

Publication number
US20220378956A1
US20220378956A1 US17/749,574 US202217749574A US2022378956A1 US 20220378956 A1 US20220378956 A1 US 20220378956A1 US 202217749574 A US202217749574 A US 202217749574A US 2022378956 A1 US2022378956 A1 US 2022378956A1
Authority
US
United States
Prior art keywords
targeted
cancer
antibody
radiopharmaceutical
pcta
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.)
Pending
Application number
US17/749,574
Other languages
English (en)
Inventor
James T. Harvey
Andrew Mazar
R. Keith Frank
Jaime Simon
Jason Rogers
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.)
Northstar Medical Technologies LLC
Monopar Therapeutics Inc
Original Assignee
Northstar Medical Technologies LLC
Monopar Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Northstar Medical Technologies LLC, Monopar Therapeutics Inc filed Critical Northstar Medical Technologies LLC
Priority to US17/749,574 priority Critical patent/US20220378956A1/en
Assigned to NORTHSTAR MEDICAL TECHNOLOGIES LLC, Monopar Therapeutics Inc. reassignment NORTHSTAR MEDICAL TECHNOLOGIES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ISO THERAPEUTICS GROUP LLC, HARVEY, JAMES T., FRANK, R. KEITH, ROGERS, JASON, SIMON, JAIME, MAZAR, ANDREW
Publication of US20220378956A1 publication Critical patent/US20220378956A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K51/1093Antibodies 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 conjugates with carriers being antibodies
    • A61K51/1096Antibodies 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 conjugates with carriers being antibodies radioimmunotoxins, i.e. conjugates being structurally as defined in A61K51/1093, and including a radioactive nucleus for use in radiotherapeutic applications
    • 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
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • 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
    • A61K51/0474Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group
    • A61K51/0482Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group chelates from cyclic ligands, e.g. DOTA
    • 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
    • A61K51/1045Antibodies 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 against animal or human tumor cells or tumor cell determinants
    • 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
    • A61K51/1045Antibodies 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 against animal or human tumor cells or tumor cell determinants
    • A61K51/1054Antibodies 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 against animal or human tumor cells or tumor cell determinants the tumor cell being from lung
    • 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
    • A61K51/1093Antibodies 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 conjugates with carriers being antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/121Solutions, i.e. homogeneous liquid formulation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • Radiopharmaceuticals typically contain a radioisotope attached to a targeting moiety or carrier.
  • the radioisotope is carried to the target by the carrier where it decays.
  • the mode of isotope decay determines the type of radiopharmaceutical.
  • gamma emitting isotopes are used to detect the fate of the construct and are used for diagnostic purposes. Constructs with particle emitters are preferred for therapy. Although beta-emitting radionuclides were used previously, alpha-emitting radionuclides have shown excellent efficacy in recent years.
  • Alpha-emitting radionuclides are effective at killing cells in part due to the short particle range and high linear energy transfer (LET).
  • LET linear energy transfer
  • the relatively long half-life of the alpha-emitting radionuclide actinium-225 (Ac-225) compared to other alpha emitters is one of the reasons that it has become popular as a therapeutic radioisotope for the treatment of cancer. Clinical trials with constructs using the isotope have shown excellent results. The about 10-day half-life is a good match for the in vivo biological half-life of monoclonal antibodies, and the four alpha emissions produced by Ac-225 and its daughters were responsible for a high rate of tumor cell kill. However, the chemistry necessary to attach Ac-225 to a targeting moiety was lacking.
  • Ac-225 ions exhibit a valence of +3, with a documented ionic radius of 112 pm. Due to its lack of polarizability, Ac +3 is classified as a “hard” Lewis acid according to the Hard and Soft Acids and Bases (HSAB) [Pearson, J Am Chem Soc 1963, 85:3533-3539] theory and is therefore likewise predicted to prefer “hard,” nonpolarizable, electronegative Lewis bases such as anionic oxygen donors.
  • HSAB Hard and Soft Acids and Bases
  • the hard/soft acid-base properties of a specific ion can be quantified using the concept of absolute (h) chemical hardness.
  • Absolute chemical hardness of Ac +3 and La +3 so calculated are 14.4 eV and 15.4 eV, respectively.
  • Soft ions such as Au + , Ag + and Cu + exhibit absolute chemical hardness values that's range from 5.7 to 6.3 eV, whereas conventional hard ions, like Sc +3 and Al +3 are characterized by absolute chemical hardness values of greater than 24 eV. Thiele et al., Cancer Biother Radio, 2018 33(8):336-348.
  • the large ionic size of Ac +3 is suited to large polydentate chelators of high denticities, because most commonly used chelates for Ac(III) range between 8-12 coordinate.
  • Actinium is similar to other actinides and rare earth elements, and can undergo hydrolysis in solution in the absence of a chelating agent to form [Ac(OH) 3-x ] x- ; the sub-picomolar concentrations of Ac-225 cause the hydroxide species in turn to form radiocolloids that bind to surfaces such as reaction vessels.
  • Emission of multiple alpha-particles in the Ac-225 decay chain makes Ac-225 a particularly effective isotope to kill cancer cells, yet also makes the directed delivery of the nuclide and its decay daughters a challenge. Due to the conservation of momentum, the emission of an energetic alpha particle imparts a recoil energy to the daughter nucleus often >100 keV, 1000 times larger than the binding energy for any chemical bond. This results in release of the daughter nuclide from the chelator of the original delivery vector. The subsequent redistribution of the alpha-emitting daughter nuclides in vivo can cause substantial harm to untargeted healthy tissues and reduce the therapeutic effect.
  • Ac-225-CHX-DTPA-monoclonal antibody (Mab) complexes used to determine biokinetic behavior on tumor-bearing nude mice showed successful in vitro complexing but poor stability in vivo.
  • Ac-225 may prove useful in radiotherapeutic models, information regarding potentially effective chelators and the relative stability of such Ac-225 complexes in vivo is lacking.
  • CHX-A′′-DTPA provides inadequate chelation of Ac(III).
  • PCTA 3,6,9,15-tetraazabicyclo-[9.3.1]pentadeca-1(15),11,13-triene-3,6,9-acetic acid
  • the method in the Simon disclosure required the use of high concentrations of acetate ion and a high chelator to antibody ratio (CAR). Starting reactions were conducted using a molar reactant ratio of 100 chelators per antibody to yield a CAR number of 10-12.
  • Bi-213 is a radioactive decay product of Ac-225, whereas Bi-212 produced by the radioactive decay of lead-212 (Pb-212) after step-wise decay of uranium-234 (U-234).
  • Pb-212 the radioactive decay of lead-212
  • U-234 step-wise decay of uranium-234
  • Bismuth isotopes Bi-212 and Bi-213, are also candidates for use in radioimmunotherapy.
  • Several preclinical studies have been published utilizing one, the other or both isotopes. Both have valences of +3, and as such tend to remain complexed after actinium has decayed.
  • Bi-212 has a half-life of about one hour and emits both alpha and beta particles in an almost 1:2 ratio.
  • Bi-213 has a half-life of about 45 minutes and decays almost completely by beta emission to polonium-213, which then emits an alpha particle to form lead-209 as is shown in FIG. 1 herein.
  • TAT targeted a-particle therapy
  • TCMC 2-(4-isothio-cyanatobenzyl-1,4,7,10-tetraaza-1,4,7,10-tetra-(2-carbamonylmetyl)cyclododecane
  • Zirconium-89 is another useful radioisotope in that zirconium has a valence and the Zr-89 emits a gamma ray (909 keV) and also a positron at about 397 keV, both emissions being useful in diagnostics.
  • the half-life of Zr-89 is 3.3 days, which is similar to the circulation half-lives of many monoclonal antibodies used in medicine. Those isotopes have been used in radiolabelling and evaluation of mAbs in positron emission tomography (Immuno-PET). [Saleem et al., Sci World J 2014, Article ID 269605, 9 pages.]
  • the final decay product of Zr-89 is yttrium-89, a stable non-radioactive isotope.
  • a further useful isotope in the present invention is indium-111.
  • Indium also has a valence of +3, and In-111 has a half-life of about 2.8 days.
  • Indium-111 decay provides gamma rays of 0.171 MeV and 0.245 MeV, that can be used in diagnostic scans such as single photon emission computed tomography (SPECT) imaging.
  • SPECT single photon emission computed tomography
  • In-111 decays to cadmium-111, which is non-radioactive and stable.
  • the invention disclosed below teaches the using various targeting species and a single chelating agent for both therapeutic and diagnostic (theranostic) uses, providing a single chelator-linked targeting system for both uses.
  • a theranostic has significant benefits in development and manufacturing as the targeting species and chelation manufacturing steps can be common with the labeling of the radioisotope being distinct. This provides some time and cost advantages in development, toxicity studies with the unlabeled targeted-chelator, common stability and bulk drug substance.
  • This invention relates to the use of a chelating agent containing a 12-membered macrocyclic amine with a pyridine ring imbedded in the structure surprisingly that easily makes stable metal ligand complexes with trivalent radioactive isotope ions such as Ac-225, Bi-212, Bi-213, Zr-89 and In-111, and also with a targeting species molecule to form a radiotherapeutic agent or a radiodiagnostic agent (or generically, a radiopharmaceutical agent). These radiopharmaceuticals can also be referred to as radiotheranostic agents.
  • the chelating agent is bonded to a targeting species molecule, permitting the chelation of the Q +3 ion to that part of the molecule.
  • the chelating agent is bonded to a part of the targeting molecule that does not interfere with the ability of the targeting molecule to reach its target.
  • the targeting species binds the radiopharmaceutical agent to cells that are to be killed or one or more of whose presence, location, size and shape are to be determined.
  • a targeting species is chemically-bonded to a PCTA chelator with its chelated trivalent radioactive isotope ion, Q +3 , to form the theranostic radiopharmaceutical that has the general structural formula shown below in Formula I and which, depending on the radioactive isotope that is chelated can be used therapeutically to kill targeted cells or to bind to targeted cells to signal the one or more of the presence, location, size or shape of the bound cells
  • the chelated Q +3 ion is a radioactive isotope having a valence of +3, six of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are H, and the seventh contains a reacted functionality, Z, that forms the chemical bond with the targeting species, T.
  • X 1 , X 2 , and X 3 are the same or different substituent groups that can coordinate to that ion and/or help neutralize the ionic charge of the chelated Q +3 ion such as trivalent Ac-225, Bi-213, Bi-212, Zr-89 or In-111.
  • g is a number whose average value is 1 to about 12 that indicates the average number of chelated PCTA-chelated trivalent radioactive ions, Q +3 , per each molecule of targeting species, T.
  • An optional anion, Y ⁇ can be present in an amount needed to balance the ionic charge.
  • the chelation reaction with the Q +3 ion can be performed first followed by attachment to the targeting species molecule, T. This is referred to as a two-step process because the isotope is handled twice.
  • the conjugation reaction attaching the chelating agent to the targeting species
  • 225 Ac +3 is a preferred Q +3 ion.
  • a pharmaceutical composition is contemplated that comprises a theranostic effective amount of a targeted radiopharmaceutical of Formula I dissolved or dispersed in a pharmaceutically acceptable diluent.
  • the pharmaceutically acceptable diluent is an aqueous liquid at ambient temperature and is adapted for parenteral administration.
  • composition in a method for treating a mammalian host having a disease, disorder or condition characterized by undesired angiogenesis, tumor growth and/or tumor metastasis comprising administering to the host a targeted cell-killing (therapeutic) effective amount of the targeted radiopharmaceutical.
  • a targeted cell-killing (therapeutic) effective amount of the targeted radiopharmaceutical comprising administering to the host a targeted cell-killing (therapeutic) effective amount of the targeted radiopharmaceutical.
  • a contemplated targeted radiopharmaceutical is used as a diagnostic agent.
  • the invention contemplates a method for assaying a mammalian host thought or known to have a disease, disorder or condition characterized by undesired angiogenesis, tumor growth and/or tumor metastasis by administering to the host a target cell-binding effective amount of the targeted radiopharmaceutical followed by scanning the host to detect and locate the radiation emitted by the bound targeted radiopharmaceutical.
  • This invention relates to a targeted radiopharmaceutical that comprises PCTA-chelated Q +3 ion chemically-bonded to a targeting species.
  • a contemplated targeted radiopharmaceutical of this invention has the general structural formula shown below in Formula I
  • Q +3 is a trivalent radioactive isotope ion; six of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are H and the seventh contains a reacted linking functionality, Z, that forms a chemical bond with the targeting species, T.
  • the X 1 , X 2 , and X 3 groups are the same or different substituent groups that can coordinate to the Q +3 ion such as 225 Ac +3 , 212 Bi +3 , 213 Bi +3 , 89 Zr +3 or 111 In +3 and help neutralize the ionic charge of the chelated Q +3 ion.
  • g is a number whose average value is about 1 to about 12 that indicates the average number of chelated PCTA-chelated trivalent radioactive ions, Q +3 , per each molecule of targeting species, T.
  • An optional anion, Y ⁇ can be present in an amount needed to balance the ionic charge.
  • Illustrative targeted radiopharmaceutical chelates are illustrated in Formulas Ia, Ib, Ic and Id below without depicting a specific targeting species, the number of chelates bonded to each target species, or a reacted linking functionality, Z.
  • the two bismuth isotopes ( 212 Bi +3 and 213 Bi +3 ) are written together as 212/213 Bi +3 for convenience.
  • Actinium-225 is a preferred radiopharmaceutical isotope because it has a half-life of almost ten days and as shown in FIG. 1 , decays to release four alpha-particles and three beta-particles to form bismuth-209, a stable isotope.
  • One of the daughter decay products from Ac-225 is Bi-213, and the final decay product is Bi-209, which is not radioactive and is stable.
  • Bismuth-212 is a decay product of lead-212. Once obtained, the bismuth-212 can be separated from the lead-212 and complexed with an appropriately linked PCTA to form a chelated Q +3 ion chemically-bonded to a targeting species. Bi-212 ultimately decays to lead-208, which is not radioactive and is stable.
  • Some versions of the chelating agent are referred to as pyridine-based 12-membered tetraaza-macrocyclic ligands or PCTA (First published: 2 Apr. 2019 chemistry-europe.onlinelibrary-.wiley.com/-doi/abs/10.1002/ejoc.201900280.
  • the reacted functionality, Z is selected from the group consisting of one or more of a reacted Michael reaction acceptor, a reacted isocyanato group, a reacted carboxyl group, and a 1,4-disubstituted-1,2,3-triazine formed by the reaction of an azide and an alkyne.
  • a reacted isothiocyanate [—NH—C( ⁇ S)—NH—; a thiourea] is one preferred reacted functionality.
  • the number of chelators bonded per antibody molecule is an average number because some antibody molecules in a given composition do not react whereas others do react. Average numbers of chelators bonded per antibody molecule are 1 to about 12, preferably about 3 to about 12, and more preferably about 8 to about 10 when an isothiocyanate group is being bonded to an intact antibody. Where a paratope-containing portion (or antigen-binding fragment) thereof is the targeting species, the number of PCTA chelators per targeting species molecule tend to be fewer such as about 1 to about 5 as there are fewer lysine amino groups with which the isothiocyanate group can react when the two pairs of CH2 and CH3 portions of the heavy chain are absent.
  • An illustrative chelator reacted functionality, Z, prior to reaction can be a Michael reaction acceptor such as maleimide can link up to about 8 chelators to a reduced intact antibody thiol groups.
  • a Michael reaction acceptor such as maleimide
  • a reduced intact antibody thiol groups can link up to about 8 chelators to a reduced intact antibody thiol groups.
  • smaller targeting species such as a the peptidomimetic cyclic-(RGDyK) discussed hereinafter has only one amine that can bond to the chelator, thereby limiting the number of radiopharmaceutical chelates to which it can be linked.
  • a Michael reaction acceptor contains an a,b-unsaturated carbonyl group that can react with a nucleophile such as an amine or a mercaptan.
  • a nucleophile such as an amine or a mercaptan.
  • Illustrative Michael reaction acceptor groups include acryloyl, methacryloyl and maleimido groups.
  • Precursors for the formation of the 1,4-disubstituted-1,2,3-triazine, an azide and an alkyne can be present, one each, on either the pre-reacted functionality of the chelator or on the targeting species.
  • the coupling reaction can be catalyzed by a copper(II) ion or by irradiation with UV light.
  • the targeting species, T is selected from the group consisting of one or more of a chemically-bonded antibody or paratope-containing portion of an antibody, a chemically-bonded hormone, a chemically bonded non-antibody protein, a chemically-bonded cytokine, a chemically bonded aptamer, a chemically bonded oligonucleotide, a chemically-bonded cytokine, a straight chain or cyclic oligopeptide or peptide mimetic, and a straight chain or branched chain oligosaccharide.
  • a monoclonal antibody (mAb) or a paratope-containing portion thereof is a preferred targeting group, and a humanized monoclonal antibody or a paratope-containing portion thereof is particularly preferred.
  • the X 1 , X 2 , and X 3 groups are the same or different substituent that is a functional group useful for chelation that can coordinate to the Q +3 ion and/or help neutralize the ionic charge of the targeted radiopharmaceutical.
  • exemplary X substituents include a —(CH 2 ) n CO 2 M group, a phosphonic acid (—PO 3 M 2 ) group and half-esters thereof, as well as carboxamides —(CH 2 ) n CONH 2 , and —(CH 2 ) n CH 2 NR 10 R 11 primary, secondary or tertiary amines where R 10 and R 11 are the same or different H or C 1 -C 4 alkyl.
  • M is a proton (H + ), an ammonium ion or an alkali metal ion. It is preferred that each of the X 1 , X 2 , and X 3 groups is the same, and more preferably each is a —COOM group. “n” is zero or 1, preferably zero so that an X group is —CO 2 M. It is to be understood that once in an aqueous composition such as a buffer, the cationic M is likely exchanged for another cation present in the aqueous composition.
  • PCTA A preferred chelator is referred to in the art as PCTA.
  • the chemical formula for a particularly preferred form of PCTA is the (4-isothiocyanato-phenyl)methyl derivative that enables the chelator to be bifunctional, and is shown in Formula II, below, where M is as before described.
  • the chelator of Formula II is commercially available from Macrocyclics Inc. (Dallas, Tex.) under the designation p-SCN-Bn-PCTA.
  • Antibodies are one group of preferred targeting species molecules in one aspect of the invention because many bind to cell surface antigens of undesirable cells in the body such as cancer cells. Once bound, the antibody and its bonded chelated Q +3 ion can be taken into the unwanted cell (cell to be treated) at which time the Q +3 ion such as Ac-225 or one of its daughter atoms such as 213 Bi +3 can decompose to release its cytotoxic alpha particle within the unwanted cell.
  • the particular combination of a Q +3 ion with a PCTA chelator forms particularly stable chelation products as compared to those formed using DOTA as the chelator with a Q +3 ion. As a consequence, there is a greater concentration of radioisotope at the target cell and a lower concentration of radioisotope elsewhere in the recipient body than when a chelator such as DOTA is utilized.
  • mAb MNBR-101 is a humanized version of mouse mAb ATN-658, whose hybridoma has ATCC Accession Number BTA-8191, disclosed and claimed in U.S. Pat. No. 8,101,726.
  • Mouse mAb ATN-615 that is also disclosed and claimed in U.S. Pat. No. 8,101,726, is secreted by a hybridoma that has ATCC Accession Number BTA-8192.
  • uPA-uPAR urokinase plasminogen activator
  • uPAR cell surface receptor
  • U.S. Pat. No. 8,101,726 notes that expression of uPA and uPAR has been demonstrated in numerous tumor types.
  • the mAb MNPR-101 paratopic amino acid residue sequence (CDR; complementarity determining region; variable region) binds to its uPA-uPAR antigen very similarly to the binding of mAb ATN-658 Table 3, hereinafter).
  • the heavy chain constant regions (CH1, CH2 and CH3) are those of a human IgG1 antibody.
  • HETM Human EngineerTM
  • XOMA Corp. Engelbreviations: XOMA Corp. (Emeryville, Calif.) proprietary HETM method to generate the low risk and low plus moderate risk HETM variants;
  • the phrase “low risk” discussed above and hereinafter relates to whether a mouse-to-human amino acid residue change results in a major reduction in therapeutic immunogenicity with little chance of affecting binding affinity.
  • the second phrase “high risk” relates to modifying positions at which a mouse-to-human amino acid residue change results in a degradation or abolition of binding activity with little or no actual reduction in therapeutic immunogenicity.
  • antibody is meant to include both intact immunoglobulin (Ig) molecules as well as fragments and derivative thereof, that can be produced by proteolytic cleavage of Ig molecules or engineered genetically or chemically.
  • Paratope-containing portions or fragments include, for example, Fab, Fab′, F(ab′) 2 and Fv, each of which is capable of binding antigen. These fragments lack the Fc fragment of intact antibody (Ab) and have an additional advantage, if used therapeutically, of clearing more rapidly from the circulation and undergoing less non-specific tissue binding than intact antibodies.
  • Papain treatment of Ig's produces Fab fragments; pepsin treatment produces F(ab′) 2 fragments. These fragments can also be produced by genetic or protein engineering using methods well known in the art.
  • a Fab fragment or portion is a multimeric protein consisting of the portion of an Ig molecule containing the immunologically active portions of an Ig heavy (H) chain and an Ig light (L) chain covalently coupled together and capable of specifically combining with antigen.
  • Fab fragments are typically prepared by proteolytic digestion of substantially intact Ig molecules with papain using methods that are well known in the art. However, a Fab fragment can also be prepared by expressing in a suitable host cell the desired portions of Ig H chain and L chain using methods well known in the art.
  • a F(ab′) 2 fragment is a tetramer that includes a fragment of two H and two L chains.
  • the Fv fragment is a multimeric protein consisting of the immunologically active portions of an Ig H chain variable (V) region (V H ) and an Ig L chain V region (V L ) covalently coupled together and capable of specifically combining with antigen.
  • Fv fragments are typically prepared by expressing in suitable host cell the desired portions of Ig V H region and V L region using methods well known in the art.
  • Single-chain antigen-binding protein or single chain Ab is a polypeptide composed of an Ig V L amino acid residue sequence tethered to an Ig V H amino acid residue sequence by a peptide that links the C-terminus of the V L sequence to the N-terminus of the V H sequence.
  • the Ab is a mouse monoclonal antibody (mAb) designated ATN-615 (Creative Biolabs, Inc., Shirley, N.Y.) or ATN-658 (hybridoma B cell:ATCC PTA-8191; Manassas, Va.), both of which are IgG1 antibodies.
  • An Ab of the present invention can be produced as a single chain Ab or scFv instead of the normal multimeric structure.
  • Single chain Abs include the hypervariable regions from an Ig of interest and recreate the antigen binding site of the native Ig while being a fraction of the size of the intact Ig (Skerra et al., Science, 1988 240:1038-1041; Pluckthun et al. Methods Enzymol 1989 178:497-515; Winter et al., Nature 1989 349:293-299); Bird et al., Science 1988 242:423-426; Huston et al. Proc. Natl. Acad. Sci. USA 1988 85:5879-5883; Jost et al., J Biol Chem. 1994 269:26267-26273; U.S. Pat. Nos. 4,704,692, 4,853,871, 4,946,778, 5,260,203, and 5,455,030).
  • DNA sequences encoding the V regions of the H chain and the L chain are ligated to a linker that encodes a sequence of at least about 4 amino acid residues (typically small neutral amino acids).
  • the protein encoded by this fusion permits assembly of a functional variable region that retains the specificity and affinity of the original Ab.
  • a different type of single chain Ab is an antibody induced in a camelid such as a dromedary, llama, alpaca, of vicuna. These animals produce single heavy-chain-only antibodies (HcAbs) that have a variable region and a constant region, with many unique properties such as small size, excellent solubility, superior stability, quick clearance from blood, and deep tissue penetration.
  • HcAbs single heavy-chain-only antibodies
  • Sanofi's affiliate Ablynx N.V. is the holder worldwide of the NANOBODY® trademark.
  • CDR3 complementarity-determining region 3
  • the former consists of 3 to 28 amino acids (AAs), whereas the latter only 8 to 15 AAs.
  • AAs amino acids
  • nanobodies enable a targeted therapy by lesion-specific delivery of drugs and effector domains, thereby improving the specificity and efficacy of the therapy.
  • Human genes that encode the constant (C) regions of the chimeric antibodies of the present invention can be derived from a human fetal liver library or from any human cell including those which express and produce human Igs.
  • the human C H region can be derived from any of the known classes or isotypes of human H chains, including g, m, a, d or e, and subtypes thereof, such as G1, G2, G3 and G4.
  • the choice of C H region will be guided by the desired effector functions, such as complement fixation, or activity in Ab-dependent cellular cytotoxicity (ADCC).
  • the C H region is derived from g1 (IgG1), g3 (IgG3), g4 (IgG4), or m (IgM).
  • the human C L region can be derived from either human L chain isotype, k or 1.
  • Human Ig C regions are obtained from human cells by standard cloning techniques [Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1989)]. Human C region genes are readily available from known clones containing genes representing the two classes of L chains, the five classes of H chains and subclasses thereof.
  • the chimeric antibodies of the present invention are produced by cloning DNA segments encoding the H and L chain antigen-binding regions of a specific Ab of the invention, preferably non-human, and joining these DNA segments to DNA segments encoding human C H and C L regions, respectively, to produce chimeric Ig-encoding genes.
  • a fused gene is created that comprises a first DNA segment that encodes at least the antigen-binding region of non-human origin, such as CDR1, CDR2 and CDR3 of the V region with joining (J) segment, linked to a second DNA segment encoding at least a part of a human C region.
  • Chimeric Ab fragments such as F(ab′) 2 and Fab
  • a chimeric gene encoding an H chain portion of an F(ab′) 2 fragment would include DNA sequences encoding the CH 1 domain and hinge region of the H chain, followed by a translational stop codon to yield the truncated molecule.
  • H and L chain J regions have different sequences, but a high degree of sequence homology exists (greater than 80%) among each group, especially near the C region. This homology is exploited in this method and consensus sequences of H and L chain J regions can be used to design oligonucleotides for use as primers for introducing useful restriction sites into the J region for subsequent linkage of V region segments to human C region segments.
  • C region cDNA vectors prepared from human cells can be modified by site-directed mutagenesis to place a restriction site at the analogous position in the human sequence. For example, one can clone the complete human k chain C (C k ) region and the complete human g-1 C region (C g-1 ). In this case, the alternative method based upon genomic C region clones as the source for C region vectors would not permit these genes to be expressed in bacterial systems where enzymes needed to remove intervening sequences are absent. Cloned V region segments are excised and ligated to L or H chain C region vectors. Alternatively, the human C g-1 region can be modified by introducing a termination codon thereby generating a gene sequence that encodes the H chain portion of an Fab molecule. The coding sequences with linked V and C regions are then transferred into appropriate expression vehicles for expression in appropriate hosts, prokaryotic or eukaryotic.
  • the targeting molecule is a relatively small molecule such as a straight chain or cyclic oligopeptide or peptidomimetic having a molecular weight of about 400 to about 1000 amu.
  • a relatively small molecule such as a straight chain or cyclic oligopeptide or peptidomimetic having a molecular weight of about 400 to about 1000 amu.
  • One illustrative cyclic oligopeptide is the cyclic tetrapeptide referred to as cyclic-(RGDyK) that binds to the a v b 3 receptor expressed on some tumors and on the endothelial cells of tumor neovasculature [Yapp et al., Mol Imaging June 2013 12(4):263-272].
  • a peptidomimetic is a compound whose essential elements (pharmacophore) mimic a natural peptide or protein in 3D space and retain the ability to interact with the biological target and produce the same biological effect as the natural peptide or protein [Vagner et al., Curr Opin Chem Biol June 2008 12(3):292-296].
  • An illustrative peptidomimetic of interest here is an inhibitor of prostate-specific membrane antigen (PSMA).
  • PSMA is a surface type 2 integral membrane glycoprotein with folate hydrolase and carboxypeptidase, and internalization activities [Cimadamore et al., Front Oncol Dec. 21, 2018 8:article 653]. PSMA is highly expressed in prostate cancer tumor cells as well as vessels of various non-prostatic solid tumors.
  • Monoclonal antibody J591 and the three other anti-PSMA monoclonals are each mouse monoclonals.
  • One or more of those mAbs is the subject disclosed and/or claimed in the following U.S. Pat. Nos. 6,107,090; 6,136,311; 6,649,163; 6,770,450; 7,045,605; 7,112,412; 7,163,680; 7,192,586; 7,514,078; 7,666,414; 7,666,425; and 8,951,737.
  • N-acetylaspartylglutamate peptidase N-acetylated-alpha-linked acid dipeptidase;NAAALDASE
  • urea-based PSMA ligands usually consist of 3 components: a binding motif (glutamate-urea-lysine [Glu-urea-Lys]), a linker and a radiolabel-bearing moiety (chelator molecule for radiolabeling).
  • chelator molecule for radiolabeling A particularly useful such molecule is illustrated below without the chelator.
  • Illustrative of branched oligosaccharide targeting species are the sialyl-Lewis a (sLe a ) sialyl-Lewis x (sLe x ) antigens that bind to selectin receptors on endothelial cells and others that can lead to metastisis.
  • the anti-CD33 mAb, lintuzumab, binds to the sialoadhesin receptor CD33.
  • Folic acid and derivatives can be used as a targeting species for cancerous kidney cells that overexpress the folic acid receptor.
  • the folate receptor is also overexpressed in cancers of the brain, kidney, lung, ovary, and breast relative to lower levels in normal cells (see, e.g., Sudimack et al., Adv Drug Deliv Rev 2000 41:147-162).
  • Figliola et al., RSC Adv 2019 9:14078-14092 provides a synthetic route for preparation of folate targeting species with the drug prodigiosene using several ⁇ , ⁇ -amino linking groups such as ethylene diamine, an ethylene oxide, cystamine, and a diamino oligo oxytheylene.
  • Immune cells have an affinity for mannose, and several RGD-containing targeting peptides that contain 4 to 30 amino acid residues are known and many are described by Beer et al., Methods Mol. Biol. 2011 680:183-200; Beer et al., Theranostics 2011 1:48-57; Morrison et al., Theranostics 2011 1:149-153; Zhou et al., Theranostics 2011 1:58-82; and by Auzzas et al., Curr. Med. Chem. 2010 17:1255-1299, and Goonewardena et al., U.S. Pat. No. 9,931,412.
  • a pharmaceutical composition containing a theranostic effective amount of a contemplated targeted radiopharmaceutical dissolved or dispersed in a pharmaceutically acceptable diluent is utilized in a contemplated method.
  • a theranostic effective amount is a targeted cell-killing effective amount as the treatment is therapeutic.
  • Such a composition is administered in vivo into in a mammalian host animal to bind to and kill unwanted targeted cells such as cancer cells and aberrant immune cells.
  • Illustrative unwanted targeted cells include cells associated with undesired cell migration, invasion, proliferation, immune response or angiogenesis.
  • Illustrative of such cells are aberrant immune cells and, cancer cells such as those of lung cancer, ovarian cancer, prostate cancer, brain cancer, bladder cancer, head and neck cancer, pancreatic cancer and colon cancer.
  • Treatment of blood cancers such as acute myeloid leukemia that express the CD33 marker, and breast cancers that express the HER2 marker is also contemplated.
  • a theranostic amount of targeted radiopharmaceutical Q +3 ion administered therapeutically to provide a targeted cell-killing effective amount usually varies with the patient and the severity of the disease such a tumor load in cancer situations that the patient has.
  • two to about four cycles of about 80 to about 120 kBq/kg body weight every other month typically shows positive results.
  • the use of three cycles of about 100 kBq/kg body weight with the same administration regimen was reported to provide positive results using 225 AC-PSMA-617 that utilizes a DOTA-based chelating agent in prostate cancer patients leading to complete remissions in some patients.
  • Such dosages can be used to provide a basis for dosages for therapeutic treating of other conditions.
  • the host is administered a theranostic amount that is a target cell-binding (diagnostic) effective amount of the targeted radiopharmaceutical.
  • the host is thereafter maintained for a time period of about 1 hour to several days, more usually about 1 to about 4 hours, for the radiopharmaceutical to bind to the targeted cells.
  • the maintenance times can depend on several factors such as the decay rate of the trivalent isotope used and the clearance rate of the targeted radiopharmaceutical.
  • the maintained host mammal is thereafter scanned as by a PET scan for positron emissions (PET scan) or by a gamma ray detector (e.g., SPECT scan) to detect and locate the radiation emitted by the target cell-bound targeted radiopharmaceutical, and thereby identify one or more of the following 1) that targeted cells were present in the host, 2) the location in the host body of the targeted cells, 3) the size and possibly 4) the shape of the mass of cells bound by the targeting species.
  • PET scan positron emissions
  • a gamma ray detector e.g., SPECT scan
  • the diagnostically-effective amount of targeted radiopharmaceutical administered is typically enough radioisotope to provide about 0.5 to about 6 mCi for an adult, and appropriately less for a child.
  • In-111 is typically used at about 111 MBq (3 mCi) to about 222 MBq (6 mCi) for intravenous administration to an average adult (70 kg).
  • Patients can receive Zr-89 at about 0.5 to about 2 mCi by intravenous administration for a whole-body PET scan.
  • a contemplated targeted radiopharmaceutical pharmaceutical composition is intended for parenteral administration as by injection, such a composition should contain an electrolyte, and preferably have approximately physiological osmolality and pH value of the mammalian species intended as the recipient.
  • a preferred concentration of singly charged electrolyte ions in a targeted radiopharmaceutical pharmaceutical composition is about 0.5 to about 1.5% (w/v), more preferably at about 0.8 to about 1.2% (w/v), and most preferably at a concentration of about 0.9% (w/v).
  • the about 0.9% (w/v) concentration is particularly preferred because it corresponds to an approximately isotonic solution for a human.
  • the electrolyte in a chemoablative pharmaceutical composition is sodium chloride.
  • Electrolytes at such levels increase the osmolality of the targeted radiopharmaceutical pharmaceutical composition.
  • osmolality can be used to characterize, in part, the electrolyte level of the composition. It is preferred that the osmolality of a composition be greater than about 100 mOsm/kg and less that about 520 mOsm/kg, more preferably that the osmolality of the composition be greater than about 250 mOsm/kg, and most preferably that it be about 300 to about 500 mOsm/kg.
  • the pH value of the targeted radiopharmaceutical composition be about 4 to about 9, to yield maximum solubility of the targeted radiopharmaceutical in an aqueous vehicle and assure compatibility with biological tissue.
  • a particularly preferred pH value is about 5 to about 8, and more preferably between about 6 to about 7.5.
  • the pH value of the targeted radiopharmaceutical pharmaceutical composition can be regulated or adjusted by any suitable means known to those of skill in the art.
  • the composition can be buffered or the pH value adjusted by addition of acid or base or the like.
  • a contemplated targeted radiopharmaceutical pharmaceutical composition is intended for parenteral administration route, it is further preferred that it be sterile, such as required for conformance to U.S. Pharmacopeia (USP) ⁇ 71>, and further that it contains negligible levels of pyrogenic material, such that it conforms to USP ⁇ 85> (limulus amebocyte lysate assay) or to USP ⁇ 151> (rabbit pyrogen test), or to substantially equivalent requirements, at a pyrogen or endotoxin level equivalent to not more than (NMT) 10 endotoxin units (EU) per mL.
  • USP U.S. Pharmacopeia
  • the pharmaceutical composition should conform to requirements limiting content of particulate matter as defined in USP ⁇ 788> (i.e., NMT 3000 particulates greater than 10 microns in size, and NMT 300 particulates greater than 25 microns in size, per container) or substantially equivalent requirements.
  • Illustrative mammalian animal hosts to which a contemplated targeted radiopharmaceutical composition can be administered include a primate such as a human, an ape such as a chimpanzee or gorilla, a monkey such as a cynomolgus monkey or a macaque, a laboratory animal such as a rat, mouse or rabbit, a companion animal such as a dog, cat, horse, or a food animal such as a cow or steer, sheep, lamb, pig, goat, llama or the like.
  • a contemplated pharmaceutical composition is usually administered a plurality of times to a mammalian host over a period of weeks, or months. As noted, a usual administration regimen is carried out every other month. Screenings of the host between administrations provides updates from which an attending physician can make determinations concerning further treatments. As noted before, a series of three bimonthly (about 60-days apart) administrations of a composition of a different Ac-225-containing targeted radiopharmaceutical pharmaceutical at 100 kBq/kg each produced complete remissions in some prostate cancer patients.
  • parenteral compositions are discussed in, for example, Hoover, John E., Remington's Pharmaceutical Sciences , Mack Publishing Co., Easton, Pa.; 1975 and Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms , Marcel Decker, New York, N.Y., 1980.
  • sterile injectable aqueous suspensions can be formulated according to the known art using a suitable dispersing or wetting compound and suspending materials.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that can be employed are aqueous liquids at ambient temperature such as water, Ringer's solution, and isotonic sodium chloride solution, phosphate-buffered saline.
  • Liquid pharmaceutical compositions include, for example, solutions suitable for parenteral administration. Sterile water solutions of targeted radiopharmaceutical or sterile solution of the targeted radiopharmaceutical in solvents comprising water, ethanol, DMSO or propylene glycol are examples of liquid compositions suitable for parenteral administration.
  • Sterile solutions can be prepared by dissolving the targeted radiopharmaceutical component in the desired solvent system, and then passing the resulting solution through a membrane filter to sterilize it or, alternatively, by dissolving the sterile compound in a previously sterilized solvent under sterile conditions.
  • Two bifunctional chelators were purchased from Macrocyclics, Dallas, Tex. The structure of the two are shown below.
  • the Macrocyclics catalog names them as: S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid and 3,6,9,15-tetraazabicyclo[9.3.1]pentadeca-1(15),11,13-triene-4-S-(4-isothiocyanatobenzyl)-3,6,9-triacetic acid.
  • They are also named p-SCN-Bn-DOTA and p-SCN-Bn-PCTA, respectively, as unreacted precursors. Once reacted as with a targeting species, they are more simply referred to as DOTA and PCTA, and are so named herein.
  • Monoclonal antibody (mAb) MNPR-101 is a humanized version of mouse IgG1, k, mAb ATN-658 having ATCC Accession Number PTA-8191 that is disclosed and claimed in U.S. Pat. No. 8,101,726.
  • the mAb MNPR-101 paratope amino acid residue sequence (CDR; complementarity determining region) is the same as that of ATN-658, whereas the framework portions of the variable region are humanized and the Fc portion is that of a human IgG1 antibody.
  • the molar reaction ratios were 1, 3, 5, 10 and 20.
  • the molar reaction ratios were 3, 10, 25, 50 and 100.
  • the pH of the solutions was adjusted to 9.2 with 1 M Na 2 CO 3 . Reactions were run at 37° C. for 1.5 hours.
  • Bio-Rad 10DG gravity-fed columns with a 6,000 Dalton molecular weight cut-off were used purify the conjugates.
  • the columns were rinsed with 15 mL of 0.1 M HEPES buffer in 0.1M NaCl.
  • the pH of the buffer was 7.3.
  • the total contents of the reaction vials were introduced to the top of the column and collected in 2 mL tubes. Multiple 0.5 mL elutions with the same buffer were also captured in separate tubes.
  • the UV absorbance at 280 nm of each fraction was measured to determine the fractions containing protein. Typically, protein eluted in 4 fractions that were combined.
  • the protein content of the combined fractions was measured using a Pierce BCA assay kit.
  • the concentrations of the protein conjugates produced was about 1 mg/mL.
  • Conjugates were prepared by the method of Example 1 but with a chelant to protein molar reaction ratios of 12 and 25. Typical reaction yields are about 30%. Thus, average CAR numbers of about 4 and 8 are expected for the reactions.
  • Ac-225 was obtained from ORNL.
  • the reaction vial contained solid Ac-225 which was dissolved using 0.2 M HCl.
  • the same 4 conjugates as described in the previous section were used to prepare Ac-225 chelates.
  • a ratio of 50 ⁇ Ci of Ac-225 to 50 ⁇ g MNPR-101-PCTA chelant conjugate was used such that if there were a 100% yield, the specific activity would be 1 mCi/mg.
  • Reactions were run in 100 ⁇ L volumes. That volume included about 4 ⁇ L Ac-225 in 0.2M HCl, 60 ⁇ L 0.1 M ammonium acetate buffer, and 36 ⁇ L MNPR-101-PCTA or -DOTA conjugate. Reactions were incubated at pH 5.8 and 37° C. for 60 minutes.
  • the radiochemical yield of the reactions was determined by diluting a 50 ⁇ L aliquot of reaction to 3 mL in buffer and passing through a 30 kDa Amicon® filter. Small, non-chelated Ac-225 ions pass through the filter, whereas the conjugate is retained by the filter. Samples were counted on a Ge detector after 45 minutes using the first daughter of Ac-225 (Fr-221). In addition, samples were counted using a dose calibrator after overnight (about 18 hours) equilibration of Ac with its daughters. The results of the chelation are shown below.
  • the specific activity of the chelates formed from PCTA was 1,000 ⁇ Ci/g, whereas the specific activity for the chelates from conjugates prepared from DOTA ranged from about 216 to 284 mCi/g.
  • This head-to-head comparison between DOTA and PCTA shows the superiority of the PCTA chelators compared to DOTA chelators for chelating Ac.
  • HPLC High Performance Liquid Chromatography
  • bovine serum albumin with no chelants added was used as a negative control.
  • the total volume of each of the reactions was 150 ⁇ L.
  • Each of the reactions was measured for yield using the filtration method of Example 2.
  • the percent of the activity in the retentate was used as the yield of the reaction.
  • MNPR-PCTA(12) had an initial yield of 99.1%. After DTPA challenge, the chelate lost only about 1% of the activity. In contrast, the MNPR-DOTA (25) only had an initial yield of 8.9% and that decreased to 5.6% after the DTPA challenge. In addition, the control BSA only showed 3.4% of the activity associated with the protein (non-specific binding) decreasing to 1.3% after DTPA wash. The data are consistent with PCTA outperforming DOTA in the ability to chelate Ac-225 even at a lower CAR ration. In addition, the lack of binding with naked BSA shows that non-specific binding is not an issue.
  • MNPR-101 MNPR
  • PCTA conjugate has been shown to efficiently chelate Ac-225.
  • Ac-225 chelated much more efficiently to the PCTA conjugate than with the DOTA conjugate.
  • MNPR-PCTA(12) refers to the MNPR-101 conjugate made with PCTA with a starting molar reaction ratio of 12:1 p-SCN-Bn-PCTA (PCTA) to antibody.
  • MNPR-DOTA(25) refers to the conjugate of MNPR-101 with a starting molar reaction ratio of 25 p-SCN-Bn-DOTA (DOTA) chelators to antibody.
  • Reactions were targeted to produce 1 mCi/mg assuming 100% incorporation of the Ac into the antibody.
  • the reactions were run in 150 ⁇ L volumes and incubated at pH 5.8 and 37° C. for 60 minutes. Following the reaction, 35 ⁇ L aliquots of each reaction were mixed with 35 ⁇ L of 1M diethylenetriamine pentaacetate (DTPA) and allowed to stand at room temperature for 1 hour.
  • DTPA diethylenetriamine pentaacetate
  • the percentage designates the relative amount of activity in the filter compared to the total (filter+filtrate).
  • MNPR-PCTA(12) gave the best results with 99% and 98% attached to the antibody (on the filter) after 1 and 24 hour incubation with DTPA. The purity dropped to 73% after 72 hours. Note that there is no radioprotectant added and Ac-225 gives a high radiation dose to the solution. The fact that the isotope remained associated with the protein shows a high degree of stability, and that there was little loss of the bismuth decay product to the solution during that time periods examined.
  • the antibody MNPR-101 conjugated with PCTA with a starting chelator to antibody molar reaction ratio of 12:1 was shown to reproducibly chelate Ac-225 in high yield and high specific activity (1,000 ⁇ Ci/mg). Incubation of the material in excess DTPA showed a high degree of stability even when the formulation did not contain any radioprotectant.
  • a head-to-head comparison with the same antibody conjugated with DOTA with a starting ratio of 25:1 ligand to protein molar ratio gave much lower yields showing the advantage of PCTA over DOTA for chelating Ac-225. Naked BSA was used as a control showing low amounts of non-specific binding.
  • the starting molar ratio of chelator to antibody was 12 to 1.
  • Fifty ⁇ Ci of Ac-225 was combined with 50 ⁇ g of the MNPR-PCTA conjugate and the pH value adjusted to 5.8 with ammonium acetate for 60 minutes at 37 C. The total volume of the reaction was 100 ⁇ L.
  • a volume of 25 ⁇ L of the reaction mixture was analyzed on high performance liquid chromatography using a size exclusion column.
  • Bismuth ions can form very insoluble compounds that could precipitate carrying both bismuth and actinium ions. Prevention of bismuth compounds precipitation by the mAb-linked-PCTA chelating functionality provides another benefit of this invention.
  • PCTA conjugates were prepared with humanized mAb MNPR-101 in parallel with two other illustrative mouse monoclonal antibodies: mAb ATN-616 and mAb ATN-292.
  • the chelator to protein molar ratios of 12 and 75 were used to optimize subsequent chelation of Ac-225.
  • MNPR-101 and ATN-616 were conjugated with PCTA at the molar reaction ratio of 12:1, whereas ATN-292 was conjugated at a 75:1 excess.
  • the pH values of the solutions were adjusted to 9.2 with 1 M NaH 2 CO 3 and 0.2 M HCl. Reactions were run at 37° C. for 1.5 hours.
  • the conjugates so formed were purified using Bio-Rad 10DG gravity-fed columns (6,000 Dalton (Da) molecular weight cut-off) in which each conjugate was eluted with 0.1 M ammonium acetate buffer, pH 5.77. Eluted fractions (0.5 mL) were collected in 1.5 mL metal-free tubes and were measured at UV absorbance 280 nm. 3 or 4 fractions were combined, depending on concentration of protein in the eluant, and re-concentrated using Amicon® concentrators (30 kDa). Combined fractions were analyzed using a PierceTM BCA Assay Kit (Thermofisher; Final protein concentrations were about 2-3 mg/mL).
  • a reaction vial containing solid Ac-225 was obtained from ORNL and was dissolved using 0.2 M HCl.
  • the three conjugates from Example 6 were used to prepare Ac-225 chelates.
  • a ratio of 100 ⁇ Ci of Ac-225 to 100 ⁇ g mAb-PCTA chelant conjugate for all reactions was used such that if there were a 100% yield, the specific activity would be 1 mCi/mg.
  • Reactions were run in about 110 ⁇ L volumes including approximately 10 ⁇ L Ac-225 in 0.2 M HCl, 60 ⁇ L 0.1 M ammonium acetate buffer, and 40 ⁇ L mAb-PCTA conjugate, dependent upon and normalized against each protein concentration. Reactions were incubated at pH 5.7 and 37° C. for 60 minutes.
  • Peak fractions from each reaction were measured at time points 24 hours and 48 hours post-purification to determine the chelants' ability to retain Ac-225 daughter isotopes. Peak activity increasing as a function of time provides evidence that the chelants did not effectively control the daughter isotopes. However, if activity decreased at a rate consistent with Ac-225 degradation, evidence suggests that the chelants were able to retain Ac-225 and its daughters.
  • Peak Yields of the three reactions when analyzed by the dose calibrator show 80.6%, 77.0%, and 75.5%, respectively, as described in Example 7, these same reactions show Reaction Yields of 96.2%, 92.7%, and 97.8%, respectively, when analyzed using HPLC purification and NaI detection.
  • This variance is understood to stem from a lack of the ability to measure activity remaining on the size-exclusion column, thus observing the more conservative yields from the Bio-Rad gravity-fed columns and dose calibrator values.
  • V L light chain variable region
  • V H heavy chain variable region
  • VK2 V Kappa light chain subgroup 2
  • VH1 human heavy chain subgroup 1
  • Two sequences for each of the light chain and the heavy chain variable regions were prepared.
  • One sequence for each chain contained only low risk changes and the other sequence that contained both the low risk and the moderate risk changes were prepared for the VK2 and VH1 regions, providing a total of four sequences.
  • Ten low risk and 1 moderate risk changes were introduced into the light chain framework sequences and 11 low risk and 5 moderate risk changes were introduced into the heavy chain framework sequences.
  • Low risk residue position changes those exposed to solvent but not contributing to antigen binding or antibody structure, are likely to decrease immunogenicity with little or no effect on binding affinity.
  • the amino acid residue sequences were sent to Blue Heron Biotech LLP, (Bothell, Wash.) for codon (Chinese Hamster Ovary cells) and expression optimization.
  • the optimized DNA sequences were received and sent back to Blue Heron for gene synthesis.
  • Codon- and expression-optimized low risk and low plus moderate risk Human EngineeredTM light chains and heavy chains were cloned in-frame into XOMA's proprietary transient antibody expression vectors that contain human Kappa and Gamma-1 constant region modules. The DNA sequences were verified (at ELIM Biopharmaceuticals, Inc., Hayward, Calif.) prior to initiating expression.
  • HETM ATN-658 variants (referred to as HETM ATN-1, HETM ATN-2, HETM ATN-3 and HETM ATN-4) were produced by transient transfection in HEK293E cells.
  • XOMA's transient transfection approach is described in detail in a poster presented at the 2005 ASCB Annual Meeting.
  • the light and heavy chains were co-transfected into XOMA's suspension-adapted HEK293E cells grown in IS293 medium (Irvine Scientific, Irvine, Calif.) using 2 liter shake flasks. After 24 hours in shake flasks, 200 ml of transfected cells were centrifuged, resuspended in 40 ml of fresh medium and transferred to Integra flasks (Wilson Wolf Manufacturing, Inc., New Brighton, Minn.) for production. After incubation for seven days, the cell suspensions were removed from the Integra flasks, centrifuged and the culture supernatants retained. Antibodies in the culture supernatants were purified on protein A spin columns (Pro-Chem), dialyzed against PBS, concentrated and sterile filtered.
  • IS293 medium Irvine Scientific, Irvine, Calif.
  • variable region constituent sequences of those four antibodies are illustrated in Table 1, below.
  • Mouse parental mAb ATN-658 was diluted in PBS to 2 mg/mL and injected over a rabbit anti-mouse capture surface.
  • the HETM variants were diluted to 1 mg/mL and injected over a protein A/G surface.
  • Antibody injections were optimized to produce antibody densities of 100-200 RU.
  • CDRs complementarity-determining regions
  • a SalI restriction site was placed in frame and up-stream of the encoded N-terminus of each of the heavy and light chains and a XhoI site was inserted in frame and down-stream from the encoded C-terminus of each chain for insertion of coding nucleic acids into their expression vectors.
  • the heavy and light chain of the monoclonal antibody candidate were packaged in a pUC19 plasmid.
  • cDNA inserts encoding the monoclonal antibodies were cloned out and heavy and light chains were inserted into expression vectors.
  • the DHFR-deficient CHO cell line DUX B11 was transfected with light chain and heavy chain containing vectors and a cationic liposome mixture (Lipofectamine® 2000; Invitrogen Corp., Carlsbad, Calif.). Forty-eight hours after transfection, cells were subcloned in 96 well dishes using a purine-free growth medium in the presence of geneticin (G418) and 20 nM methotrexate (MTX).
  • mice mAb ATN-658 and the above discussed Human EngineeredTM ATN-658 antibodies were measured once.
  • the sensorgram results of individual assays indicated that all of the transiently-expressed antibodies displayed a similar affinity with mAb ATN-658 as well as among themselves. Results for the four combinations of two VL and two VH chains are shown below in Table 3.
  • Antibody HETM ATN-4 was renamed MNPR-101.
  • PCTA-MNPR-101 produced at 12:1) freshly prepared in an aqueous solution at a pH value of 9.2 (1M NaHCO 3 and HCl) that contained 4.0 mg/mL by protein analysis was incubated for 1.5 hours at 37° C.
  • the conjugate 220.0 mL MNPR-101-PCTA was purified by passage through a PD10 column with elution using 0.1M ammonium acetate. Samples containing the conjugate were collected and concentrated using a 30 kDa Amicon® concentrator (4000 rpm for 20 minutes).
  • aqueous chelation reactions were set up, each with activity of about 200 ⁇ Ci for a target specific activity of 10 mCi/mg. Each was mixed with In-111 chloride obtained from BWXT Medical, Ottawa, ON, Canada. All reactions were stored at 4° C. and assayed for stability after 24, 48 and 72 hours.
  • Stability in this context is the maintenance of radioactive ion chelation over time. Stability was determined by gravity fed SEC column (PD10 6,000 Dalton cut off), HPLC and TLC for comparison.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Optics & Photonics (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Dispersion Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nuclear Medicine (AREA)
  • Peptides Or Proteins (AREA)
US17/749,574 2021-05-21 2022-05-20 Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use Pending US20220378956A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/749,574 US20220378956A1 (en) 2021-05-21 2022-05-20 Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202163191499P 2021-05-21 2021-05-21
US202163191506P 2021-05-21 2021-05-21
US17/749,574 US20220378956A1 (en) 2021-05-21 2022-05-20 Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use

Publications (1)

Publication Number Publication Date
US20220378956A1 true US20220378956A1 (en) 2022-12-01

Family

ID=84140859

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/749,574 Pending US20220378956A1 (en) 2021-05-21 2022-05-20 Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use
US17/749,763 Pending US20220409751A1 (en) 2021-05-21 2022-05-20 Urokinase Plasminogen Activator Receptor-Targeted Radiopharmaceutical

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/749,763 Pending US20220409751A1 (en) 2021-05-21 2022-05-20 Urokinase Plasminogen Activator Receptor-Targeted Radiopharmaceutical

Country Status (9)

Country Link
US (2) US20220378956A1 (ko)
EP (2) EP4341296A1 (ko)
JP (2) JP2024519970A (ko)
KR (1) KR20240035757A (ko)
AU (2) AU2022276433A1 (ko)
CA (1) CA3223227A1 (ko)
IL (1) IL308731A (ko)
MX (1) MX2023013836A (ko)
WO (2) WO2022246183A1 (ko)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024147853A1 (en) * 2023-01-05 2024-07-11 NorthStar Medical Technologies, LLC Method for stacked elution of mother-daughter radionuclides

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024153756A1 (en) * 2023-01-18 2024-07-25 Curasight A/S Radionuclide labelled peptide conjugate for site-specific upar-targeting

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5334371A (en) * 1988-07-20 1994-08-02 Schering Aktiengesellschaft Marcocyclic polyaza bicyclo compounds containing 5 or 6 membered rings, and method for MRI
US20090180952A1 (en) * 2004-05-25 2009-07-16 Melexis Nv Ligands binding the complex of urokinase-type plasminogen activator (uppa) and its receptor (upar) that inhibit downstream upar interactions: identification and use in diagnosis or therapy
US20210009715A1 (en) * 2017-05-24 2021-01-14 ITM Isotopen Technologien München AG Novel psma-binding agents and uses thereof
WO2022123462A1 (en) * 2020-12-09 2022-06-16 3B Pharmaceuticals Gmbh Radiolabelled prostate specific membrane antigen (psma) inhibitors and use thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2182022C1 (ru) * 2001-05-24 2002-05-10 Российская медицинская академия последипломного образования МЗ РФ Способ лечения онкологических заболеваний щитовидной железы
AU2005249490B2 (en) * 2004-06-01 2010-07-29 Genentech, Inc. Antibody drug conjugates and methods
FR2883562B1 (fr) * 2005-03-24 2009-02-27 Guerbet Sa Chelates lipophiles et leur utilisation en imagerie
ES2636451T3 (es) * 2006-12-08 2017-10-05 Monopar Therapeutics Inc. Epítopo del receptor del activador del plasminógeno tipo uroquinasa
EP3456700A1 (en) * 2013-10-18 2019-03-20 Deutsches Krebsforschungszentrum Labeled inhibitors of prostate specific membrane antigen (psma), their use as imaging agents and pharmaceutical agents for the treatment of prostate cancer
DK3261681T3 (da) * 2015-02-26 2020-04-27 Sciencons AS Radiofarmaceutiske opløsninger med fordelagtige egenskaber
ES2802234T3 (es) * 2015-05-22 2021-01-18 Memorial Sloan Kettering Cancer Center Sistemas y métodos para determinar la dosis óptima y específica para el paciente de anticuerpos dirigidos al tumor
WO2017223565A1 (en) * 2016-06-24 2017-12-28 University Of Iowa Research Foundation Compositions and methods of treating melanoma
CA3182769A1 (en) * 2020-06-15 2021-12-23 Andrew P. Mazar Precision radioimmunotherapeutic targeting of the urokinase plasminogen activator receptor (upar) for treatment of severe covid-19 disease

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5334371A (en) * 1988-07-20 1994-08-02 Schering Aktiengesellschaft Marcocyclic polyaza bicyclo compounds containing 5 or 6 membered rings, and method for MRI
US20090180952A1 (en) * 2004-05-25 2009-07-16 Melexis Nv Ligands binding the complex of urokinase-type plasminogen activator (uppa) and its receptor (upar) that inhibit downstream upar interactions: identification and use in diagnosis or therapy
US20210009715A1 (en) * 2017-05-24 2021-01-14 ITM Isotopen Technologien München AG Novel psma-binding agents and uses thereof
WO2022123462A1 (en) * 2020-12-09 2022-06-16 3B Pharmaceuticals Gmbh Radiolabelled prostate specific membrane antigen (psma) inhibitors and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Song et al., J. Nucl. Med., 2016, 57, p. 1105–1111. (Year: 2016) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024147853A1 (en) * 2023-01-05 2024-07-11 NorthStar Medical Technologies, LLC Method for stacked elution of mother-daughter radionuclides

Also Published As

Publication number Publication date
KR20240035757A (ko) 2024-03-18
JP2024520180A (ja) 2024-05-21
WO2022246183A1 (en) 2022-11-24
EP4341265A1 (en) 2024-03-27
WO2022246210A1 (en) 2022-11-24
US20220409751A1 (en) 2022-12-29
AU2022276444A1 (en) 2023-12-21
CA3223227A1 (en) 2022-11-24
MX2023013836A (es) 2024-01-11
EP4341296A1 (en) 2024-03-27
JP2024519970A (ja) 2024-05-21
AU2022276433A1 (en) 2023-12-07
IL308731A (en) 2024-01-01

Similar Documents

Publication Publication Date Title
KR101893720B1 (ko) 방사 면역 접합체 및 그 용도
US20220378956A1 (en) Trivalent Radioisotope Bio-Targeted Radiopharmaceutical, Methods Of Preparation And Use
JP2023078366A (ja) ポリペプチドの放射性標識
Qu et al. Development of humanized antibodies as cancer therapeutics
US20210017295A1 (en) Bispecific binding agents and uses thereof
CN111093693A (zh) 抗l1-cam抗体及其用途
KR20240014479A (ko) 전립선암의 치료를 위한 조성물 및 방법
CN117794950A (zh) 靶向尿激酶型纤溶酶原激活物受体的放射性药物
EP4327831A1 (en) Radioactive complex of anti-cd20 antibody, and radiopharmaceutical
Boswell et al. In Vitro–In Vivo Correlations of Pharmacokinetics, Pharmacodynamics, and Metabolism for Antibody Therapeutics
EP4397320A1 (en) Radioactive complex of deglycosylated antibody and radioactive pharmaceutical product
US20240316228A1 (en) Radioactive complex of deglycosylated antibody and radioactive pharmaceutical product
WO2023157822A1 (ja) 抗vegf抗体の放射性複合体、及び、放射性医薬
EA045797B1 (ru) Радиоактивное мечение полипептидов

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: MONOPAR THERAPEUTICS INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HARVEY, JAMES T.;MAZAR, ANDREW;FRANK, R. KEITH;AND OTHERS;SIGNING DATES FROM 20220620 TO 20221127;REEL/FRAME:061894/0370

Owner name: NORTHSTAR MEDICAL TECHNOLOGIES LLC, WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HARVEY, JAMES T.;MAZAR, ANDREW;FRANK, R. KEITH;AND OTHERS;SIGNING DATES FROM 20220620 TO 20221127;REEL/FRAME:061894/0370

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION