WO2021207086A1 - Tem-1-targeted radioimmunoconjugates and uses thereof - Google Patents

Tem-1-targeted radioimmunoconjugates and uses thereof Download PDF

Info

Publication number
WO2021207086A1
WO2021207086A1 PCT/US2021/025800 US2021025800W WO2021207086A1 WO 2021207086 A1 WO2021207086 A1 WO 2021207086A1 US 2021025800 W US2021025800 W US 2021025800W WO 2021207086 A1 WO2021207086 A1 WO 2021207086A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid sequence
amino acid
seq
cdr
radioimmunoconjugate
Prior art date
Application number
PCT/US2021/025800
Other languages
English (en)
French (fr)
Inventor
Eric S. Burak
John R. Forbes
Ryan W. SIMMS
Original Assignee
Fusion Pharmaceuticals 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 Fusion Pharmaceuticals Inc. filed Critical Fusion Pharmaceuticals Inc.
Publication of WO2021207086A1 publication Critical patent/WO2021207086A1/en

Links

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
    • A61K51/1027Antibodies 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 receptors, cell-surface antigens or cell-surface 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
    • 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
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • A61N5/1001X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy using radiation sources introduced into or applied onto the body; brachytherapy
    • A61N2005/1019Sources therefor
    • A61N2005/1021Radioactive fluid

Definitions

  • TEM-1 tumor epithelial marker-1
  • endosialin or CD248 has been identified as a target for cancer therapeutics as a result of its expression across the stroma of many human tumors and because of its low or absent expression in normal tissues.
  • TEM-1 tumor epithelial marker-1
  • ontuxizumab MORAb-004 by Morphotek
  • TEM-1 monoclonal antibody a TEM-1 monoclonal antibody
  • Radiotherapy has been pursued as a treatment for cancer for many years, far longer than immunotherapy.
  • the clinical use of radiotherapy has been limited by the toxicity caused by harmful radiation near or in healthy tissues, which are exposed to radiation that generally continues until, ultimately, the radionuclide decays.
  • dose reductions may be necessary, reducing efficacy; otherwise, severe side- effects, including permanent organ or tissue damage, are clinical risks.
  • improved therapeutics e.g., cancer therapeutics
  • the present invention includes the discovery of an effective therapy for TEM-1 expressing cancerous cells.
  • the invention employs the administration of radioimmunoconjugates that target TEM-1 (e.g., human TEM-1).
  • target TEM-1 e.g., human TEM-1
  • the radioimmunoconjugates of the invention exhibit dramatic and specific TEM-1 tumor killing and yet demonstrate lower, not higher, off target effects. This is a seemingly contradictory combination of high efficacy and reduced toxicity.
  • the treatment effect is unexpected for TEM-1 tumors, relative to a) TEM-1 antibodies alone or b) radiotherapeutics employing alternative structural combinations, relative to those of the invention. It is the combination of the specificity for the TEM-1-expressing tumor cells and improved excretion of the radioimmunoconjugates provided by the structural combinations of the invention that allow for therapeutics that are uniquely effective for treating TEM-1-associated cancer cells.
  • the lowered total radiation resident in the treated subject’s healthy tissue as a result of the radioimmunoconjugates of the invention lowers the deleterious “off target” effects but also allows for a level of dosing that delivers a high degree of therapeutic efficacy.
  • radioimmunoconjugate comprising the following structure: A-L-B (Formula I-a) or a pharmaceutically acceptable salt thereof, wherein: A is a chelating moiety or metal complex thereof; B is a TEM-1 targeting moiety; and L is a linker.
  • the chelating moiety is selected from the group consisting of: DOTA, DOTMA, DOTAM, DOTPA, DO3AM-acetic acid, DOTA-GA anhydride, DOTP, DOTMP, DOTA-4AMP, CB-TE2A, NOTA, NOTP, TETPA, TETA, HEHA, PEPA, H 4 octapa, H 2 dedpa, H 6 phospa, TTHA, DO2P, HP-DO3A, EDTA, Deferoxamine, DTPA, DTPA-BMA, octadentate-HOPO, and porphyrins.
  • A is a metal complex of a chelating moiety.
  • the metal complex comprises a radionuclide.
  • the radionuclide is an alpha emitter, e.g., an alpha emitter selected from the group consisting of Astatine-211 ( 211 At), Bismuth-212 ( 212 Bi), Bismuth-213 ( 213 Bi), Actinium-225 ( 225 Ac), Radium-223 ( 223 Ra), Lead-212 ( 212 Pb), Thorium- 227 ( 227 Th), and Terbium-149 ( 149 Tb), or a progeny thereof.
  • the radionuclide is 225 Ac or a progeny thereof.
  • L 3 is C 5 -C 20 polyethylene glycol. In some embodiments, L 3 is (CH 2 CH 2 O) m (CH 2 ) w , and m and w are independently an integer between 0 and 10 (inclusive). [0011] In some embodiments, the radioimmunoconjugate or a pharmaceutically acceptable salt thereof comprises the following structure: , wherein B is a TEM-1 targeting moiety.
  • the TEM-1 targeting moiety is at least 100 kDa in size, e.g., at least 150 kDa in size, at least 200 kDa in size, at least 250 kDa in size, or at least 300 kDa in size.
  • the TEM-1 targeting moiety is capable of binding to human TEM-1.
  • the TEM-1 targeting moiety comprises an antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is humanized.
  • the antibody or antigen-binding fragment thereof is capable of binding an epitope comprising an amino acid sequence of SRDHQIPVIAAN (SEQ ID NO: 2).
  • the antibody or antigen-binding fragment thereof comprises at least one complementarity determining region (CDR) selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; or CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or 2 amino acids therefrom.
  • CDR-H1 compris
  • the antibody or antigen-binding fragment thereof comprises at least two CDRs selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; or CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or 2 amino acids therefrom.
  • CDR-H1 comprising the amino acid sequence of S
  • the antibody or antigen-binding fragment thereof comprises at least three CDRs selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; or CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or 2 amino acids therefrom.
  • CDR-H1 comprising the amino acid sequence of S
  • the antibody or antigen-binding fragment thereof comprises at least four CDRs selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; or CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or 2 amino acids therefrom.
  • CDR-H1 comprising the amino acid sequence of S
  • the antibody or antigen-binding fragment thereof comprises at least five CDRs selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; or CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or 2 amino acids therefrom.
  • CDR-H1 comprising the amino acid sequence of S
  • the antibody or antigen-binding fragment thereof comprises: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; or CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or 2 amino acids therefrom.
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain comprising at least one CDR selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; and (ii) a light chain variable domain comprising at least one CDR selected from the group consisting of: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; and CDR-L
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain comprising at least one CDR selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6; and (ii) a light chain variable domain comprising at least one CDR selected from the group consisting of: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8; and CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9.
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain comprising at least two CDRs selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; and (ii) a light chain variable domain comprising at least two CDRs selected from the group consisting of: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; and CDR
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain comprising at least two CDRs selected from the group consisting of: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6; and (ii) a light chain variable domain comprising at least two CDRs selected from the group consisting of: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8; and CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9.
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain comprising: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4, or an amino acid sequence differing in 1 or 2 amino acids from SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence differing in 1 or 2 amino acids therefrom; and (ii) a light chain variable domain comprising: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7, or an amino acid sequence differing in 1 or 2 amino acids therefrom; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8, or an amino acid sequence differing in 1 or 2 amino acids therefrom; and CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence differing in 1 or
  • the antibody or antigen-binding fragment thereof comprises (i) a heavy chain variable domain comprising: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5; CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6; and (ii) a light chain variable domain comprising: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8; and CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9.
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain having an amino acid sequence with at least 85% identity with the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain variable domain having an amino acid sequence with at least 85% identity with the amino acid sequence of SEQ ID NO: 21. [0029] In some embodiments, the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain having an amino acid sequence with at least 90% identity with the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain variable domain having an amino acid sequence with at least 90% identity with the amino acid sequence of SEQ ID NO: 21.
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain having an amino acid sequence with at least 95% identity with the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain variable domain having an amino acid sequence with at least 95% identity with the amino acid sequence of SEQ ID NO: 21. [0031] In some embodiments, the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain variable domain comprising the amino acid sequence of SEQ ID NO: 21. [0032] In some embodiments, the antibody is hMP-E-8.3.
  • the proportion of radiation excreted by the intestinal routes, renal route, or both routes is at least 2-fold greater than the proportion of radiation excreted by the same route(s) by a comparable mammal that has been administered a reference radioimmunoconjugate.
  • the proportion of radiation excreted by the intestinal routes, renal route, or both routes is at least 3-fold greater than the proportion of radiation excreted by the same route(s) by a comparable mammal that has been administered a reference radioimmunoconjugate.
  • A-L- is a metal complex of a moiety selected from the group consisting of:
  • A-L- is a metal complex of Moiety 1: [0037] In some embodiments, A-L- is a metal complex of Moiety 1:
  • the metal complex comprises a radionuclide, such as an alpha emitter (e.g., Astatine- 211 ( 211 At), Bismuth-212 ( 212 Bi), Bismuth-213 ( 213 Bi), Actinium-225 ( 225 Ac), Radium-223 ( 223 Ra), Lead-212 ( 212 Pb), Thorium-227 ( 227 Th), and Terbium-149 ( 149 Tb), or a progeny thereof).
  • the TEM-1 targeting moiety comprises an antibody or antigen- binding fragment thereof (e.g., a humanized antibody or antigen-binding fragment thereof).
  • A-L- is a metal complex of Moiety 1: wherein the metal complex comprises 225 Ac or a progeny thereof, and the TEM-1 targeting moiety is hMP-E-8.3 or an antigen-binding fragment thereof.
  • the TEM-1 targeting moiety is an antibody or antigen-binding fragment thereof comprising (i) a heavy chain variable domain comprising: CDR-H1 comprising the amino acid sequence of SEQ ID NO: 3 or 4; CDR-H2 comprising the amino acid sequence of SEQ ID NO: 5; and CDR-H3 comprising the amino acid sequence of SEQ ID NO: 6; and (ii) a light chain variable domain comprising: CDR-L1 comprising the amino acid sequence of SEQ ID NO: 7; CDR-L2 comprising the amino acid sequence of SEQ ID NO: 8; and CDR-L3 comprising the amino acid sequence of SEQ ID NO: 9.
  • A-L- is a metal complex of Moiety 1:
  • the metal complex comprises 225 Ac or a progeny thereof
  • the TEM-1 targeting moiety is hMP-E-8.3 or an antigen-binding fragment thereof.
  • the TEM-1 targeting moiety is hMP-E-8.3.
  • the radioimmunoconjugate or a pharmaceutically acceptable salt thereof comprises the following structure: , wherein is hMP-E-8.3.
  • pharmaceutical compositions comprising a radioimmunoconjugate or a pharmaceutically acceptable salt thereof as described herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising an effective amount of a radioimmunoconjugate or a pharmaceutically acceptable salt thereof as described herein.
  • the subject is a mammal, e.g., a human.
  • tumor stromal cells associated with the cancer express TEM-1.
  • the cancer comprises cells that express TEM-1.
  • the cancer is a solid tumor cancer.
  • the solid tumor cancer may be a sarcoma, e.g., a sarcoma is selected from the group consisting of angiosarcoma or hemangioendothelioma, astrocytoma, chondrosarcoma, Ewing’s sarcoma, fibrosarcoma, glioma, leiomyosarcoma, liposarcoma, malignant fibrous histiocytoma (MFH), mesenchymous or mixed mesodermal tumor, mesothelial sarcoma or mesothelioma, myxosarcoma, osteosarcoma, rhabdomyosarcoma, and synovial sarcoma.
  • a sarcoma is selected from the group consisting of angiosarcoma or hemangioendothelioma, astrocytoma, chondrosarcoma, Ewing’s sarcoma,
  • the sarcoma is osteosarcoma. In some embodiments, the sarcoma is a Ewing’s sarcoma.
  • the solid tumor cancer is a carcinoma, e.g., a carcinoma is selected from the group consisting of adenoid cystic carcinoma, adrenocortical carcinoma, bladder cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, gallbladder carcinoma, gastric cancer, head and neck cancer, lung cancer (e.g., small cell lung cancer or non-small cell lung cancer, or adenocarcinoma of the lung), neuroblastoma, neuroendocrine cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, testicular cancer.
  • a carcinoma is selected from the group consisting of adenoid cystic carcinoma, adrenocortical carcinoma, bladder cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, gallbladder carcinoma, gastric cancer, head and neck cancer,
  • the solid tumor cancer is pancreatic cancer. [0049] In some embodiments, the solid tumor cancer is breast cancer. [0050] In some embodiments, the solid tumor cancer is head and neck cancer. [0051] In some embodiments, the solid tumor cancer is liver cancer. [0052] In some embodiments, the solid tumor cancer is lung cancer. [0053] In some embodiments, the solid tumor cancer is a brain cancer. [0054] In some embodiments, the solid tumor cancer is neuroblastoma. [0055] In some embodiments, the solid tumor cancer is melanoma. [0056] In some embodiments, the pharmaceutical composition is administered systemically.
  • the pharmaceutical composition is administered parenterally, e.g., intravenously, intraarterially, intraperitoneally, subcutaneously, or intradermally.
  • the pharmaceutical composition is administered enterically, e.g., trans-gastrointestinally or orally.
  • the pharmaceutical composition is administered locally, e.g., by peritumoral injection or by intratumoral injection.
  • FIG.1A is a schematic depicting the general structure of a bifunctional chelate comprising a chelate, a linker, and a cross-linking group.
  • FIG.1B is a schematic depicting the general structure of a bifunctional conjugate comprising a chelate, a linker, and a targeting moiety.
  • FIG.2 is a schematic depicting the synthesis of the bifunctional chelate, 4- ⁇ [11- oxo-11-(2,3,5,6-tetrafluorophenoxy)undecyl]carbamoyl ⁇ -2-[4,7,10-tris(carboxymethyl)- 1,4,7,10-tetraazacyclododecan-1-yl]butanoic acid (Compound B). Synthesis of Compound B is described in Example 2.
  • FIG.3 is a schematic depicting the synthesis of the bifunctional chelate, 4- ⁇ [2-(2- ⁇ 2-[3-oxo-3-(2,3,5,6-tetrafluorophenoxy)propoxy]ethoxy ⁇ ethoxy)ethyl]carbamoyl ⁇ -2- [4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl]butanoic acid (Compound C). Synthesis of Compound C is described in Example 4.
  • FIG.4A and FIG.4B show flow cytometry results for SK-N-AS (neuroblastoma) cells and SJSA-1 (osteosarcoma) cells, respectively, stained with a commercially available anti-human TEM-1 (CD248) antibody.
  • the left-most peaks in Figs.4A and 4B represent cells stained with an isotype-matched control antibody instead of anti-human TEM-1.
  • FIG.5 shows flow cytometry results from SJSA-1 cells stained with 1) hMP-E- 8.3 from an original stock solution; 2) hMP-E-8.3 from a re-purified stock solution; 3) Compound C-hMP-E-8.3 conjugate; or 4) an isotype-matched control antibody, then stained with a FITC-labeled secondary antibody. The left-most peak indicates unstained ells.
  • FIG.6 shows results from saturation binding study using SJSA-1 cells to determine the binding affinity for [ 177 Lu]-Compound C-hMP-E-8.3.
  • FIG.7 shows results from a biodistribution study in an SJSA-1 osteosarcoma mouse model. Mice were injected with [ 177 Lu]-Compound C-hMP-E-8.3 and sacrificed at 4 h, 24 h, 48 h, 96 h, or 168 h. The percentage of injected dose/ gram (%ID/g) was assessed in blood, bone, heart, intestines, kidneys + adrenals, liver + gall bladder, lungs, muscle, pancreas, spleen, stomach, or tumor.
  • FIG.8 shows tumor growth curves in an SJSA-1 osteosarcoma mouse model for mice injected with [ 225 Ac]-Compound C-hMP-E-8.3 (10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi) or with vehicle or antibody controls.
  • FIG.9 shows tumor growth curves in an SK-N-AS neuroblastoma mouse model for mice injected with [ 225 Ac]-Compound C-hMP-E-8.3 (10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi) or with vehicle or antibody controls.
  • FIG.10 shows tumor growth curves in an A673 Ewing’s sarcoma mouse model for mice injected with [ 225 Ac]-Compound C-hMP-E-8.3 (10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi) or with vehicle or antibody controls.
  • [ 225 Ac]-Compound C-hMP-E-8.3 10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi
  • vehicle or antibody controls See Example 10.
  • DETAILED DESCRIPTION Radioimmunoconjugates are designed to target a protein or receptor that is upregulated in a disease state to deliver a radioactive payload to damage and kill cells of interest (radioimmunotherapy).
  • Radioimmunoconjugates typically contain a biological targeting moiety (e.g., an antibody or antigen binding fragment thereof that is capable of specifically binding to human TEM-1), a radioisotope, and a molecule that links the two. Conjugates are formed when a bifunctional chelate is appended to the biological targeting molecule so that structural alterations are minimal while maintaining target affinity. Once radiolabelled, the final radioimmunoconjugate is formed.
  • a biological targeting moiety e.g., an antibody or antigen binding fragment thereof that is capable of specifically binding to human TEM-1
  • a radioisotope e.g., a radioisotope
  • Bifunctional chelates structurally contain a chelate, the linker, and a cross-linking group (FIG.1A).
  • FOG.1A cross-linking group
  • Radioimmunoconjugates do not need to block a receptor, as needed with a therapeutic antibody, or release the cytotoxic payload intracellularly, as required with an antibody drug conjugate, in order to have therapeutic efficacy.
  • the emission of the toxic particle is an event that occurs as a result of first-order (radioactive) decay and can occur at random anywhere inside the body after administration. Once the emission occurs, damage could occur to surrounding cells within the range of the emission leading to the potential of off-target toxicity. Therefore, limiting exposure of these emissions to normal tissue is the key to developing new drugs.
  • One potential method for reducing off-target exposure is to remove the radioactivity more effectively from the body (e.g., from normal tissue in the body).
  • One mechanism is to increase the rate of clearance of the biological targeting agent. This approach likely requires identifying ways to shorten the half-life of the biological targeting agent, which is not well described for biological targeting agents. Regardless of the mechanism, increasing drug clearance will also negatively impact the pharmacodynamics/efficacy in that the more rapid removal of drug from the body will lower the effective concentration at the site of action, which, in turn, would require a higher total dose and would not achieve the desired results of reducing total radioactive dose to normal tissue.
  • Other efforts have focused on accelerating the metabolism of the portion of the molecule that contains the radioactive moiety.
  • Cleavable linkers have been taken on different meaning as it relates to radioimmunoconjugates.
  • Cornelissen, et al. has described cleavable linkers as those by which the bifunctional chelate attaches to the biologic targeting agent through a reduced cysteine, whereas others have described the use of enzyme-cleavable systems that require the co-administration of the radioimmunoconjugate with a cleaving agent/enzyme to release [Mol Cancer Ther.2013, 12(11), 2472-2482; Methods Mol Biol.
  • Disclosed immunoconjugates may, in some embodiments, achieve a reduction of total body radioactivity, for example, by increasing the extent of excretion of the catabolic/metabolic products while maintaining the pharmacokinetics of the intact molecule when compared to known bifunctional conjugates. In some embodiments, this reduction in radioactivity results from the clearance of catabolic/metabolic by-products without impacting other in vitro and in vivo properties such as binding specificity (in vitro binding), cellular retention, and tumor uptake in vivo. Thus, in some embodiments, provided radioimmunoconjugates achieve reduced radioactivity in the human body while maintaining on-target activity.
  • antibody refers to a polypeptide whose amino acid sequence includes immunoglobulins and fragments thereof which specifically bind to a designated antigen, or fragments thereof.
  • Antibodies in accordance with the present invention may be of any type (e.g., IgA, IgD, IgE, IgG, or IgM) or subtype (e.g., IgA1, IgA2, IgG1, IgG2, IgG3, or IgG4).
  • a characteristic sequence or portion of an antibody may include amino acids found in one or more regions of an antibody (e.g., variable region, hypervariable region, constant region, heavy chain, light chain, and combinations thereof).
  • a characteristic sequence or portion of an antibody may include one or more polypeptide chains, and may include sequence elements found in the same polypeptide chain or in different polypeptide chains.
  • “antigen-binding fragment” refers to a portion of an antibody that retains the binding characteristics of the parent antibody.
  • bind or “binding” of a targeting moiety means an at least temporary interaction or association with or to a target molecule, e.g., to human TEM-1, e.g., wild type or mutant TEM-1 as described herein.
  • bifunctional chelate refers to a compound that comprises a chelate, a linker, and a cross-linking group. See, e.g., FIG.1A.
  • a “cross-linking group” is a reactive group that is capable of joining two or more molecules, e.g., joining a bifunctional chelate and a targeting moiety, by a covalent bond.
  • the term “bifunctional conjugate,” as used herein, refers to a compound that comprises a chelate or metal complex thereof, a linker, and a targeting moiety, e.g., an antibody or antigen-binding fragment thereof.
  • cancer refers to any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, and lymphomas.
  • a “solid tumor cancer” is a cancer comprising an abnormal mass of tissue, e.g., sarcomas, carcinomas, and lymphomas.
  • a “hematological cancer” or “liquid cancer,” as used interchangeably herein, is a cancer present in a body fluid, e.g., lymphomas and leukemias.
  • chelate refers to an organic compound or portion thereof that can be bonded to a central metal or radiometal atom at two or more points.
  • conjugate refers to a molecule that contains a chelating group or metal complex thereof, a linker group, and which optionally contains a targeting moiety, e.g., an antibody or antigen-binding fragment thereof.
  • targeting moiety e.g., an antibody or antigen-binding fragment thereof.
  • compound is meant to include all stereoisomers, geometric isomers, and tautomers of the structures depicted.
  • the compounds recited or described herein can be asymmetric (e.g., having one or more stereocenters).
  • detection agent refers to a molecule or atom which is useful in diagnosing a disease by locating the cells containing the antigen.
  • Various methods of labeling polypeptides with detection agents are known in the art.
  • detection agents include, but are not limited to, radioisotopes and radionuclides, dyes (such as with the biotin-streptavidin complex), contrast agents, luminescent agents (e.g., fluorescein isothiocyanate or FITC, rhodamine, lanthanide phosphors, cyanine, and near IR dyes), and magnetic agents, such as gadolinium chelates.
  • radioisotopes and radionuclides include, but are not limited to, radioisotopes and radionuclides, dyes (such as with the biotin-streptavidin complex), contrast agents, luminescent agents (e.g., fluorescein isothiocyanate or FITC, rhodamine, lanthanide phosphors, cyanine, and near IR dyes), and magnetic agents, such as gadolinium chelates.
  • luminescent agents e.g., fluorescein isothio
  • the term “radionuclide,” refers to an atom capable of undergoing radioactive decay (e.g., 3 H, 14 C, 15 N, 18 F, 35 S, 47 Sc, 55 Co, 60 Cu, 61 Cu, 62 Cu, 64 Cu, 67 Cu, 75 Br, 76 Br , 77 Br , 89 Zr, 86 Y, 87 Y, 90 Y, 97 Ru, 99 Tc, 99m Tc 105 Rh, 109 Pd, 111 In, 123 I, 124 I, 125 I, 131 I, 149 Pm, 149 Tb, 153 Sm, 166 Ho, 177 Lu, 186 Re, 188 Re, 198 Au, 199 Au, 203 Pb, 211 At, 212 Pb , 212 Bi, 213 Bi, 223 Ra, 225 Ac, 227 Th, 229Th , 66 Ga, 67 Ga, 68 Ga, 82 Rb, 117m Sn, 201 Tl
  • radioactive nuclide may also be used to describe a radionuclide.
  • Radionuclides may be used as detection agents, as described herein.
  • the radionuclide may be an alpha-emitting radionuclide.
  • an “effective amount” of an agent e.g., any of the foregoing conjugates, as used herein, is that amount sufficient to effect beneficial or desired results, such as clinical results, and, as such, an “effective amount” depends upon the context in which it is being applied.
  • an “effective amount” may be an amount sufficient to cure or at least partially arrest the symptoms of the disorder and its complications, and/or to substantially improve at least one symptom associated with the disease or a medical condition.
  • an agent or compound that decreases, prevents, delays, suppresses, or arrests any symptom of the disease or condition would be therapeutically effective.
  • a therapeutically effective amount of an agent or compound is not required to cure a disease or condition but may, for example, provide a treatment for a disease or condition such that the onset of the disease or condition is delayed, hindered, or prevented, such that the disease or condition symptoms are ameliorated, or such that the term of the disease or condition is changed.
  • the disease or condition may become less severe and/or recovery is accelerated in an individual.
  • An effective amount may be administered by administering a single dose or multiple (e.g., at least two, at least three, at least four, at least five, or at least six) doses.
  • a targeting moiety such as an antibody (or antigen-binding fragment thereof), nanobody, affibody, or a consensus sequence from Fibronectin type III domain.
  • the immunoconjugate comprises an average of at least 0.10 conjugates per targeting moiety (e.g., an average of at least 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 4, 5, or 8 conjugates per targeting moiety).
  • the term “radioconjugate,” as used herein, refers to any conjugate that includes a radioisotope or radionuclide, such as any of the radioisotopes or radionuclides described herein.
  • radioimmunoconjugate refers to any immunoconjugate that includes a radioisotope or radionuclide, such as any of the radioisotopes or radionuclides described herein.
  • a radioimmunoconjugate provided in the present disclosure typically refers to a bifunctional conjugate that comprises a metal complex formed from a radioisotope or radionuclide.
  • radiationoimmunotherapy refers a method of using a radioimmunoconjugate to produce a therapeutic effect.
  • radioimmunotherapy may include administration of a radioimmunoconjugate to a subject in need thereof, wherein administration of the radioimmunoconjugate produces a therapeutic effect in the subject.
  • radioimmunotherapy may include administration of a radioimmunoconjugate to a cell, wherein administration of the radioimmunoconjugate kills the cell.
  • radioimmunotherapy involves the selective killing of a cell, in some embodiments the cell is a cancer cell in a subject having cancer.
  • pharmaceutical composition represents a composition containing a radioimmunoconjugate described herein formulated with a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.
  • Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other formulation described herein.
  • a “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being nontoxic and non- inflammatory in a patient.
  • Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, radioprotectants, sorbents, suspending or dispersing agents, sweeteners, or waters of hydration.
  • excipients include, but are not limited to: ascorbic acid, histidine, phosphate buffer, butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid,
  • salts represent those salts of the compounds described here that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, or allergic response.
  • Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. Salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.
  • the compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts.
  • These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention be prepared from inorganic or organic bases.
  • the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases.
  • Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as hydrochloric, sulphuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines for forming basic salts.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.
  • polypeptide refers to a string of at least two amino acids attached to one another by a peptide bond.
  • a polypeptide may include at least 3-5 amino acids, each of which is attached to others by way of at least one peptide bond.
  • polypeptides can include one or more “non-natural” amino acids or other entities that nonetheless are capable of integrating into a polypeptide chain.
  • a polypeptide may be glycosylated, e.g., a polypeptide may contain one or more covalently linked sugar moieties.
  • a single “polypeptide” e.g., an antibody polypeptide
  • subject is meant a human or non-human animal (e.g., a mammal).
  • substantially identical is meant a polypeptide sequence that has the same polypeptide sequence, respectively, as a reference sequence, or has a specified percentage of amino acid residues, respectively, that are the same at the corresponding location within a reference sequence when the two sequences are optimally aligned.
  • an amino acid sequence that is “substantially identical” to a reference sequence has at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to the reference amino acid sequence.
  • the length of comparison sequences will generally be at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 50, 75, 90, 100, 150, 200, 250, 300, or 350 contiguous amino acids (e.g., a full- length sequence).
  • Sequence identity may be measured using sequence analysis software on the default setting (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, WI 53705). Such software may match similar sequences by assigning degrees of homology to various substitutions, deletions, and other modifications.
  • sequence analysis software e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, WI 53705
  • Such software may match similar sequences by assigning degrees of homology to various substitutions, deletions, and other modifications.
  • targeting moiety refers to any molecule or any part of a molecule that is capable of binding to a given target.
  • TEM-1 targeting moiety refers to a targeting moiety that is capable of binding a TEM-1 molecule, e.g., a human TEM- 1 molecule, e.g., a wild type or mutant TEM-1 molecule.
  • a condition or “treatment” of the condition is an approach for obtaining beneficial or desired results, such as clinical results.
  • Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e., not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable.
  • “Palliating” a disease, disorder, or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment.
  • Radioimmunoconjugates [0103] In one aspect, the present disclosure provides radioimmunoconjugates having structure of Formula I-a: A-L-B (Formula I-a) wherein A is a chelating moiety or metal complex thereof, wherein B is a TEM-1 targeting moiety, and wherein L is a linker.
  • the radioimmunoconjugate has or comprises the structure shown in Formula II: , (Formula II) wherein B is the TEM-1 targeting moiety.
  • B is the TEM-1 targeting moiety.
  • A-L- is a metal complex of a moiety selected from the group consisting of:
  • the radioimmunoconjugate comprises a chelating moiety or metal complex thereof, which metal complex may comprise a radionuclide.
  • the average ratio or median ratio of the chelating moiety to the TEM-1 targeting moiety is eight or less, seven or less, six or less, five or less, four or less, three or less, two or less, or about one.
  • the average ratio or median ratio of the chelating moiety to the TEM-1 targeting moiety is about one.
  • the proportion of radiation (of the total amount of radiation that is administered) that is excreted by the intestinal route, the renal route, or both is greater than the proportion of radiation excreted by a comparable mammal that has been administered a reference radioimmunoconjugate.
  • reference immunoconjugate it is meant a known radioimmunoconjugate that differs from a radioimmunoconjugate described herein at least by (1) having a different linker; (2) having a targeting moiety of a different size and/or (3) lacking a targeting moiety.
  • the reference radioimmunoconjugate is selected from the group consisting of [ 90 Y]-ibritumomab tiuxetan (Zevalin (90Y)) and [ 111 In]- ibritumomab tiuxetan (Zevalin ( 111 In)).
  • the proportion of radiation excreted by a given route or set of routes is at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% greater than the proportion of radiation excreted by the same route(s) by a comparable mammal that has been administered a reference radioimmunoconjugate.
  • the proportion of radiation excreted is at least 1.5-fold, at least 2-fold, at least 2.5-fold, at least 3-fold, at least 3.5 fold, at least 4-fold, at least 4.5 fold, at least 5 fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold greater than proportion of radiation excreted by a comparable mammal that has been administered a reference radioimmunoconjugate.
  • the extent of excretion can be measured by methods known in the art, e.g., by measuring radioactivity in urine and/or feces and/or by measuring total body radioactivity over a period time. See also, e.g., International Patent Publication WO 2018/024869.
  • the extent of excretion is measured at a time period of at least or about 12 hours after administration, at least or about 24 hours after administration, at least or about 2 days after administration, at least or about 3 days after administration, at least or about 4 days after administration, at least or about 5 days after administration, at least or about 6 days after administration, or at least or about 7 days, after administration.
  • a radioimmunoconjugate after a radioimmunoconjugate has been administered to a mammal, the radioimmunoconjugate exhibits decreased off-target binding effects (e.g., toxicities) as compared to a reference conjugate (e.g., a reference immunoconjugate such as a reference radioimmunoconjugate).
  • this decreased off-target binding effect is a feature of a radioimmunoconjugate that also exhibits a greater excretion rate as described herein.
  • Targeting moieties include any molecule or any part of a molecule that is capable of binding to a given target, e.g., TEM-1.
  • the targeting moiety is capable of binding to an epitope within human TEM-1, an exemplary sequence for which is shown in SEQ ID NO: 1: (SEQ ID NO: 1) [0112]
  • the targeting moiety comprises a protein or polypeptide.
  • the targeting moiety is selected from the group consisting of antibodies or antigen binding fragments thereof, nanobodies, affibodies, and consensus sequences from Fibronectin type III domains (e.g., Centyrins or Adnectins).
  • a moiety is both a targeting and a therapeutic moiety, i.e., the moiety is capable of binding to a given target and also confers a therapeutic benefit.
  • the targeting moiety comprises a small molecule.
  • the targeting moiety has a molecular weight of at least 50 kDa, at least 75 kDa, at least 100 kDa, at least 125 kDa, at least 150 kDa, at least 175 kDa, at least 200 kDa, at least 225 kDa, at least 250 kDa, at least 275 kDa, or at least 300 kDa.
  • the targeting moiety is capable of binding to TEM-1, e.g., wild type and/or mutant TEM-1.
  • the targeting moiety is capable of binding to human TEM-1, e.g., wild type and/or mutant human TEM-1.
  • the targeting moiety is capable of binding specifically to TEM-1 (e.g., is capable of binding to TEM-1 while exhibiting comparatively little or no binding to other proteins, such as other C-lectin family proteins).
  • the targeting moiety is capable of binding to an extracellular region of TEM-1, e.g., the C-type lectin domain, the Sushi-like domain, one of the three EGF repeat domains, or the mucin domain.
  • the targeting moiety is capable of binding to a portion of the extracellular region that is not the C-type lectin domain (e.g., a Sushi-like domain, an EGF repeat domain, or the mucin domain).
  • the targeting moiety inhibits TEM-1.
  • inhibits it is meant that the targeting moiety at least partially inhibits one or more functions of TEM-1 (e.g., human TEM-1).
  • the targeting moiety at least partially inhibits one or more functions of wild type TEM-1, e.g., wild type human TEM-1.
  • the targeting moiety blocks binding of TEM-1 to one or more of its protein binding partner.
  • the targeting moiety blocks binding of TEM-1 to an extracellular matrix protein (e.g., an extracellular matrix protein that mediates cell adhesion and migration, such as fibronectin or collagen I).
  • the targeting moiety blocks binding of TEM-1 to LGALS3BP6. [0119] In some embodiments, the targeting moiety impairs signaling downstream of TEM-1.
  • Antibodies typically comprise two identical light polypeptide chains and two identical heavy polypeptide chains linked together by disulfide bonds. The first domain located at the amino terminus of each chain is variable in amino acid sequence, providing the antibody-binding specificities of each individual antibody. These are known as variable heavy (VH) and variable light (VL) regions. The other domains of each chain are relatively invariant in amino acid sequence and are known as constant heavy (CH) and constant light (CL) regions. Light chains typically comprise one variable region (VL) and one constant region (CL).
  • An IgG heavy chain includes a variable region (VH), a first constant region (CH1), a hinge region, a second constant region (CH2), and a third constant region (CH3).
  • the heavy chain includes an additional constant region (CH4).
  • Antibodies suitable for use with the present disclosure can include, for example, monoclonal antibodies, polyclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies, camelid antibodies, chimeric antibodies, single-chain Fvs (scFv), disulfide-linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies, and antigen-binding fragments of any of the above.
  • the antibody or antigen-binding fragment thereof is humanized. In some embodiments, the antibody or antigen-binding fragment thereof is chimeric. Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • antigen binding fragment refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen.
  • binding fragments encompassed within the term “antigen binding fragment” of an antibody include a Fab fragment, a F(ab')2 fragment, a Fd fragment, a Fv fragment, a scFv fragment, a dAb fragment (Ward et al., (1989) Nature 341:544-546), and an isolated complementarity determining region (CDR).
  • an “antigen binding fragment” comprises a heavy chain variable region and a light chain variable region.
  • Antibodies or antigen-binding fragments described herein can be produced by any method known in the art for the synthesis of antibodies (see, e.g., Harlow et al., Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 2nd ed.1988); Brinkman et al., 1995, J. Immunol. Methods 182:41-50; WO 92/22324; WO 98/46645). Chimeric antibodies can be produced using the methods described in, e.g., Morrison, 1985, Science 229:1202, and humanized antibodies by methods described in, e.g., U.S. Pat. No.6,180,370.
  • Additional antibodies described herein are bispecific antibodies and multivalent antibodies, as described in, e.g., Segal et al., J. Immunol. Methods 248:1-6 (2001); and Tutt et al., J. Immunol.147: 60 (1991), or any of the molecules described herein.
  • “Avimer” relates to a multimeric binding protein or peptide engineered using, for example, in vitro exon shuffling and phage display. Multiple binding domains are linked, resulting in greater affinity and specificity compared to single epitope immunoglobin domains.
  • Nanobodies are antibody fragments consisting of a single monomeric variable antibody domain.
  • Nanobodies may also be referred to as single-domain antibodies. Like antibodies, nanobodies bind selectively to a specific antigen. Nanobodies may be heavy- chain variable domains or light chain domains. Nanobodies may occur naturally or be the product of biological engineering. Nanobodies may be biologically engineered by site- directed mutagenesis or mutagenic screening (e.g., phage display, yeast display, bacterial display, mRNA display, ribosome display).“Affibodies” are polypeptides or proteins engineered to bind to a specific antigen. As such, affibodies may be considered to mimic certain functions of antibodies.
  • Affibodies may be engineered variants of the B-domain in the immunoglobulin- binding region of staphylococcal protein A.
  • Affibodies may be engineered variants of the Z- domain, a B-domain that has lower affinity for the Fab region.
  • Affibodies may be biologically engineered by site-directed mutagenesis or mutagenic screening (e.g., phage display, yeast display, bacterial display, mRNA display, ribosome display).
  • site-directed mutagenesis or mutagenic screening e.g., phage display, yeast display, bacterial display, mRNA display, ribosome display.
  • Diabodies are antibody fragments with two antigen- binding sites that may be bivalent or bispecific. See for example Hudson et al., (2003). Single-chain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all, or a portion of the light chain variable domain of an antibody.
  • Antibody fragments can be made by various techniques including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant hosts (e.g., E. coli or phage) as described herein.
  • the antibody or antigen-binding fragment thereof is a multispecific, e.g. bispecific.
  • Multispecific antibodies (or antigen-binding fragments thereof) include monoclonal antibodies (or antigen-binding fragments thereof) that have binding specificities for at least two different sites.
  • amino acid sequence variants of antibodies or antigen- binding fragments thereof are contemplated; e.g., variants that bind to TEM-1, e.g., human TEM-1.
  • Amino acid sequence variants of an antibody or antigen-binding fragment thereof may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody or antigen-binding fragment thereof, or by peptide synthesis. Such modifications include, for example, deletions from and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody or antigen-binding fragment thereof. Any combination of deletion, insertion and substitution can be made to arrive at the final construct, provided that the final construct possesses desired characteristics, e.g. antigen binding.
  • the antibody or antigen binding fragment thereof is an inhibitory antibody (also called “antagonistic antibody”) or antigen-binding fragment thereof, e.g., the antibody or antigen binding fragment thereof at least partially inhibits one or more functions of the target molecule (e.g., TEM-1) as explained further herein.
  • inhibitory antibodies include humanized monoclonal antibodies such as hMP-E-8.3 (Mediapharma), ontuxizumab (also known as MORAb-004) (Morphotek), and anti-TEM-1 antibodies from Kirin Brewery. (See, e.g., U.S. Pat. No. 8,895,000, and International Patent Publication Nos.
  • the antibody or antigen binding fragment thereof is an agonistic antibody (also known as stimulatory antibody).
  • the antibody or antigen binding fragment thereof is neither agonistic or antagonistic, or has not been characterized as either agonistic or antagonistic.
  • Additional known TEM-1 antibodies include, for example, humanized antibodies such as humanized FB5; mouse monoclonal antibodies such as, for example, mMP-E-8.3 (Mediapharma) (see, e.g., WO 2017/134234), FB5, 1F9B4 (Proteintech Group), G-9 (sc- 377221) (Santa Cruz Biotechnology), and B1/35 (MilliporeSigma); rat monoclonal antibodies such as, e.g., 9G5, 16D2, 7H12, 15F8, 9A5, 11D1, 15D10, 7D3, or 16D2 from Morphotek (see, e.g., WO 2013/148250), clone 458606 (R&D systems), CBFYE-0326 (Creative Biolabs); rabbit monoclonal antibodies such as EPR17081 (Abcam); and chimeric antibodies such as cMP-E-8.3 (Mediapharma) (see, e.g., WO 2017/
  • the TEM-1 antibody or antigen-binding fragment thereof is capable of binding an epitope comprising an amino acid sequence of SRDHQIPVIAAN (SEQ ID NO: 2).
  • the antibody or antigen-binding fragment thereof comprises specific heavy chain complementarity determining regions CDR- H1, CDR-H2 and/or CDR-H3 as described herein.
  • the complementarity determining regions (CDRs) of the antibody or antigen-binding fragment thereof are flanked by framework regions.
  • a heavy or light chain of an antibody or antigen- binding fragment thereof containing three CDRs typically contains four framework regions.
  • the heavy chain variable region of the TEM-1 antibody or antibody-binding fragment thereof comprises one, two, or three complementarity determining regions (CDRs) CDR-H1, CDR-H2, and/or CDR-H3, with amino acid sequences shown below,, or CDR region(s) having an amino acid sequence(s) differing in 1 or 2 amino acids therefrom: [0139]
  • the light chain variable region of the TEM-1 antibody or antibody-binding fragment thereof comprises one, two, or three complementarity determining regions (CDRs) CDR-L1, CDR-L2, and/or CDR-L3.
  • the antibody or antigen-binding fragment thereof comprises: (i) a heavy chain comprising: a heavy chain complementarity determining region 1 (CDR-H1) having the amino acid sequence as shown in SEQ ID NO: 3 or an amino acid sequence differing in 1 or 2 amino acids therefrom, a heavy chain complementarity determining region 2 (CDR-H2) having the amino acid sequence as shown in SEQ ID NO: 5 or an amino acid sequence differing in 1 or 2 amino acids therefrom, and a heavy chain complementarity determining region 3 (CDR-H3) having the amino acid sequence as shown in SEQ ID NO: 6 or an amino acid sequence differing in 1 or 2 amino acids therefrom, and (ii) a light chain comprising: a light chain complementarity determining region 1 (CDR-L1) having the amino acid sequence as shown in SEQ ID NO: 7 or an amino acid
  • the antibody or antigen-binding fragment thereof has CDR sequences having amino acid sequences of SEQ ID NOs: 3 and 5-9 without any variation.
  • the antibody or antigen-binding fragment thereof comprises heavy chain complementary determining regions CDR-H1, CDR-H2, and CDR-H3 having the amino acid sequences of SEQ ID NOs: 3, 5, and 6, and the chain complementarity determining regions CDR-L1, CDR-L2, and CDR-L3 having the amino acid sequences of SEQ ID NOs: 7, 8, and 9.
  • the antibody or antigen-binding fragment thereof has CDR sequences having amino acid sequences of SEQ ID NOs: 4-9 without any variation.
  • the antibody or antigen-binding fragment thereof comprises heavy chain complementary determining regions CDR-H1, CDR-H2, and CDR-H3 having the amino acid sequences of SEQ ID NOs: 4, 5, and 6, and the chain complementarity determining regions CDR-L1, CDR-L2, and CDR-L3 having the amino acid sequences of SEQ ID NOs: 7, 8, and 9.
  • the heavy chain variable region of the TEM-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 10 or an amino acid sequence differing in 1, 2, 3, or 4 amino acids therefrom, or an amino acid sequence having at least 85%, at least 90%, at least 95%, at least 97%, or at least 99% identity to SEQ ID NO: 10: (SEQ ID NO: 10)
  • the light chain variable region of the TEM-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 11 or an amino acid sequence differing in 1, 2, 3, or 4 amino acids therefrom, or an amino acid sequence having at least 85%, at least 90%, at least 95%, at least 97%, or at least 99% identity to SEQ ID NO: 11: (SEQ ID NO: 11)
  • the heavy chain variable region of the TEM-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence of SEQ ID NO: 12 or 13 or an amino acid sequence having at least 85%, at least 90%, at least 95%, at least 97%,
  • the heavy chain variable region of the TEM-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 16, 17, 18, and 19, or an amino acid sequence differing in 1, 2, 3, or 4 amino acids therefrom, or an amino acid sequence having at least 85%, at least 90%, at least 95%, at least 97%, or at least 99% identity to SEQ ID NO: 16, 17, 18, and 19: Humanized VH1: (SEQ ID NO: 16) Humanized VH2: (SEQ ID NO: 17) Humanized VH3: (SEQ ID NO: 18) Humanized VH4: (SEQ ID NO: 19) [0149]
  • the light chain variable region of the TEM-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 20, 21, 22, and 23, or an amino acid sequence differing in 1, 2, 3, or 4 amino acids therefrom, or an amino acid sequence having at least 85%, at least 90%, at least 95%,
  • the antibody, or antibody-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO: 16, and a light chain variable domain comprising the amino acid sequence of SEQ ID NO: 21.
  • the antibody or antigen-binding fragment thereof has a dissociation constant (Kd) of ⁇ 1 ⁇ M, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM.
  • the antibody or antigen-binding fragment thereof has a dissociation constant (Kd) of between 1 nM and 10 nM (inclusive of endpoints) or between 0.1 nM and 1 nM (inclusive of endpoints).
  • Kd is measured by a radio-labeled antigen binding assay (radioimmunoassay, RIA) performed with the Fab version of an antibody or antigen-binding fragment thereof of interest and its antigen.
  • Kd is measured using surface plasmon resonance assays with immobilized antigen.
  • the antibodies or antigen- binding fragments thereof are human monoclonal antibodies directed against an epitope of TEM-1, e.g., human TEM-1.
  • the antibody or antigen-binding fragment thereof may be any antibody or antigen-binding fragment thereof of natural and/or synthetic origin, e.g. an antibody of mammalian origin.
  • the constant domain if present, is a human constant domain.
  • the variable domain is a mammalian variable domain, e.g., a humanized or a human variable domain.
  • antibodies used in accordance with this disclosure are monoclonal antibodies.
  • antibodies are recombinant murine antibodies, chimeric, humanized or fully human antibodies, multispecific antibodies (e.g., bispecific antibodies), or antigen-binding fragments thereof.
  • the antibody or antigen- binding fragment thereof is labelled, i.e. coupled to a labelling group.
  • suitable labels include radioactive labels, fluorescent labels, suitable dye groups, enzyme labels, chromogenes, chemiluminescent groups, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter etc.
  • one or more labels are covalently bound to the antibody or antigen-binding fragment thereof.
  • Those labelled antibodies or antigen-binding fragments thereof may in particular be used in immunohistochemistry assays or for molecular imaging in vivo.
  • the antibody or antigen- binding fragment thereof is further conjugated with an effector group, in particular, a therapeutic effector group such as a cytotoxic agent or a radioactive group agent.
  • Polypeptides include, for example, any of a variety of hematologic agents (including, for instance, erythropoietin, blood-clotting factors, etc.), interferons, colony stimulating factors, antibodies, enzymes, and hormones. The identity of a particular polypeptide is not intended to limit the present disclosure, and any polypeptide of interest can be a polypeptide in the present methods.
  • a reference polypeptide described herein can include a target-binding domain that is capable of binding to a target of interest (e.g., capable of binding to an antigen, e.g., TEM- 1).
  • a polypeptide such as an antibody, can be capable of binding to a transmembrane polypeptide (e.g., receptor) or ligand (e.g., a growth factor).
  • Modified polypeptides may have a modified amino acid sequence. Modified polypeptides may be substantially identical to the corresponding reference polypeptide (e.g., the amino acid sequence of the modified polypeptide may have at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to the amino acid sequence of the reference polypeptide).
  • the modification does not destroy significantly a desired biological activity (e.g., binding to TEM-1).
  • the modification may reduce (e.g., by at least 5%, 10%, 20%, 25%, 35%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%), may have no effect, or may increase (e.g., by at least 5%, 10%, 25%, 50%, 100%, 200%, 500%, or 1000%) the biological activity of the original polypeptide.
  • the modified polypeptide may have or may optimize a characteristic of a polypeptide, such as in vivo stability, bioavailability, toxicity, immunological activity, immunological identity, and conjugation properties.
  • Modifications include those by natural processes, such as post-translational processing, or by chemical modification techniques known in the art. Modifications may occur anywhere in a polypeptide including the polypeptide backbone, the amino acid side chains and the amino- or carboxy-terminus. The same type of modification may be present in the same or varying degrees at several sites in a given polypeptide, and a polypeptide may contain more than one type of modification. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from post-translational natural processes or may be made synthetically.
  • modifications include pegylation, acetylation, acylation, addition of acetomidomethyl (Acm) group, ADP-ribosylation, alkylation, amidation, biotinylation, carbamoylation, carboxyethylation, esterification, covalent attachment to flavin, covalent attachment to a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of drug, covalent attachment of a marker (e.g., fluorescent or radioactive), covalent attachment of a lipid or lipid derivative, covalent attachment of phosphatidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent crosslinks, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic
  • a modified polypeptide can also include an amino acid insertion, deletion, or substitution, either conservative or non-conservative (e.g., D-amino acids, desamino acids) in the polypeptide sequence (e.g., where such changes do not substantially alter the biological activity of the polypeptide).
  • conservative or non-conservative e.g., D-amino acids, desamino acids
  • the addition of one or more cysteine residues to the amino or carboxy-terminus of a polypeptide herein can facilitate conjugation of these polypeptides by, e.g., disulfide bonding.
  • a polypeptide can be modified to include a single cysteine residue at the amino-terminus or a single cysteine residue at the carboxy-terminus.
  • Amino acid substitutions can be conservative (i.e., wherein a residue is replaced by another of the same general type or group) or non-conservative (i.e., wherein a residue is replaced by an amino acid of another type).
  • a naturally occurring amino acid can be substituted for a non-naturally occurring amino acid (i.e., non-naturally occurring conservative amino acid substitution or a non-naturally occurring non-conservative amino acid substitution).
  • Polypeptides made synthetically can include substitutions of amino acids not naturally encoded by DNA (e.g., non-naturally occurring or unnatural amino acid).
  • non-naturally occurring amino acids include D-amino acids, N-protected amino acids, an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine, a pegylated amino acid, the omega amino acids of the formula NH 2 (CH 2 ) n COOH wherein n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N- methyl isoleucine, and norleucine.
  • Phenylglycine may substitute for Trp, Tyr, or Phe; citrulline and methionine sulfoxide are neutral nonpolar, cysteic acid is acidic, and ornithine is basic. Proline may be substituted with hydroxyproline and retain the conformation conferring properties.
  • Analogs may be generated by substitutional mutagenesis and retain the biological activity of the original polypeptide. Examples of substitutions identified as “conservative substitutions” are shown in Table 1. If such substitutions result in a change not desired, then other type of substitutions, denominated “exemplary substitutions” in Table 1, or as further described herein in reference to amino acid classes, are introduced and the products screened.
  • Chelating moiety or metal complex thereof Chelating moieties
  • suitable chelating moieties include, but are not limited to, DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid), DOTMA (1R,4R,7R,10R)- ⁇ , ⁇ ’, ⁇ ”, ⁇ ’”-tetramethyl-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid, DOTAM (1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane), DOTPA (1,4,7,10- tetraazacyclododecane-1,4,7,10-tetra propionic acid), DO3AM-acetic acid (2-(4,7,10-tris(2- amino-2-oxoethyl)-1,4,7,10-tetrate
  • radioimmunoconjugates comprise a metal complex of a chelating moiety.
  • chelating groups may be used in metal chelate combinations with metals, such as manganese, iron, and gadolinium and isotopes (e.g., isotopes in the general energy range of 60 to 10,000 keV), such as any of the radioisotopes and radionuclides discussed herein.
  • chelating moieties are useful as detection agents, and radioimmunoconjugates comprising such detectable chelating moieties can therefore be used as diagnostic or theranostic agents.
  • the metal complex comprises a radionuclide.
  • suitable radioisotopes and radionuclides include, but are not limited to, 3 H, 14 C, 15 N, 18 F, 35 S, 47 Sc, 55 Co, 60 Cu, 61 Cu, 62 Cu, 64 Cu, 66 Ga, 67 Ga, 67 Cu, 68 Ga, 75 Br, 76 Br , 77 Br , 82 Rb, 89 Zr, 86 Y, 87 Y, 90 Y, 97 Ru, 99 Tc, 99m Tc, 105 Rh, 109 Pd, 111 In, 123 I, 124 I, 125 I, 131 I, 149 Pm, 149 Tb, 153 Sm, 166 Ho, 177 Lu, 117m Sn, 186 Re, 188 Re, 198 Au, 199 Au, 201 Tl, 203 Pb, 211 At, 212 Pb , 212 Bi,
  • the radionuclide is an alpha emitter, e.g., Astatine-211 ( 211 At), Bismuth-212 ( 212 Bi), Bismuth-213 ( 213 Bi), Actinium-225 ( 225 Ac), Radium-223 ( 223 Ra), Lead-212 ( 212 Pb), Thorium-227 ( 227 Th), or Terbium-149 ( 149 Tb), or a progeny thereof.
  • the alpha-emitter is Actinium-225 ( 225 Ac), or a progeny thereof.
  • the linker is as shown within the structure of Formula I-b, as that part of Formula I-b absent A and B: A-L 1 -(L 2 )n-B (Formula I-b) (A and B are as defined in Formula I-a.) [0175]
  • L 1 is substituted C1-C6 alkyl or substituted C1-C6 heteroalkyl, the substituent comprising a heteroaryl group (e.g., six-membered nitrogen- containing heteroaryl).
  • L 3 is substituted C1-C50 alkyl or substituted C1-C50 heteroalkyl, the substituent comprising a heteroaryl group (e.g., six-membered nitrogen- containing heteroaryl).
  • L 3 is C 1 -C 50 heteroalkyl.
  • L 3 is C 5 -C 20 polyethylene glycol.
  • L 3 is (CH2CH2O)m(CH2)w, and m and w are independently an integer between 0 and 10 (inclusive).
  • A is a macrocyclic chelating moiety comprising one or more heteroaryl groups (e.g., six-membered nitrogen-containing heteroaryl).
  • Cross-linking groups [0179]
  • radioimmunoconjugates are synthesized using bifunctional chelates that comprise a chelate, a linker, and a cross-linking group. Once the radioimmunoconjugate is formed, the cross-linking group may be absent from the radioimmunoconjugate.
  • radioimmunoconjugates comprise a cross-linking group instead of or in addition to the targeting moiety (e.g., in some embodiments, B in Formula I comprises a cross-linking group).
  • a cross-linking group is a reactive group that is able to join two or more molecules by a covalent bond.
  • Cross-linking groups may be used to attach the linker and chelating moiety to a therapeutic or targeting moiety.
  • Cross-linking groups may also be used to attach the linker and chelating moiety to a target in vivo.
  • the cross- linking group is an amino-reactive, methionine reactive or thiol-reactive cross-linking group, or comprises a sortase recognition sequence.
  • the amino-reactive or thiol-reactive cross-linking group comprises an activated ester such as a hydroxysuccinimide ester, 2,3,5,6-tetrafluorophenol ester, 4-nitrophenol ester or an imidate, anhydride, thiol, disulfide, maleimide, azide, alkyne, strained alkyne, strained alkene, halogen, sulfonate, haloacetyl, amine, hydrazide, diazirine, phosphine, tetrazine, isothiocyanate, or oxaziridine.
  • the sortase recognition sequence may comprise of a terminal glycine- glycine-glycine (GGG) and/or LPTXG amino acid sequence, where X is any amino acid.
  • GGG terminal glycine- glycine-glycine
  • LPTXG amino acid sequence where X is any amino acid.
  • compositions may be formulated for any of a variety of routes of administration discussed herein (see, e.g., the “Administration and Dosage” subsection herein), Sustained release administration is contemplated, by such means as depot injections or erodible implants or components.
  • compositions that include agents disclosed herein (e.g., radioimmunoconjugates) dissolved or suspended in an acceptable carrier, preferably an aqueous carrier, e.g., water, buffered water, saline, or PBS, among others.
  • an aqueous carrier e.g., water, buffered water, saline, or PBS
  • pharmaceutical compositions contain pharmaceutically acceptable auxiliary substances to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, or detergents, among others.
  • pharmaceutical compositions are formulated for oral delivery and may optionally contain inert ingredients such as binders or fillers for the formulation of a unit dosage form, such as a tablet or a capsule.
  • compositions are formulated for local administration and may optionally contain inert ingredients such as solvents or emulsifiers for the formulation of a cream, an ointment, a gel, a paste, or an eye drop.
  • provided pharmaceutical compositions are sterilized by conventional sterilization techniques, e.g., may be sterile filtered. Resulting aqueous solutions may be packaged for use as is, or lyophilized. Lyophilized preparations can be, for example, combined with a sterile aqueous carrier prior to administration.
  • the pH of preparations typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 6 and 7, such as 6 to 6.5.
  • compositions in solid form may be packaged, for example, in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules.
  • Pharmaceutical compositions in solid form can also be packaged in a container for a flexible quantity, such as in a squeezable tube designed for a topically applicable cream or ointment.
  • Methods of treatment [0185] In one aspect, the present disclosure provides methods of treatment comprising a subject a radioimmunoconjugate as disclosed herein. Subjects [0186] In some disclosed methods, a therapy (e.g., comprising a therapeutic agent) is administered to a subject.
  • the subject is a mammal, e.g., a human.
  • the subject has cancer or is at risk of developing cancer.
  • the subject may have been diagnosed with cancer.
  • the cancer may be a primary cancer or a metastatic cancer.
  • Subjects may have any stage of cancer, e.g., stage I, stage II, stage III, or stage IV with or without lymph node involvement and with or without metastases.
  • Provided radioimmunoconjugates and compositions may prevent or reduce further growth of the cancer and/or otherwise ameliorate the cancer (e.g., prevent or reduce metastases).
  • the subject does not have cancer but has been determined to be at risk of developing cancer, e.g., because of the presence of one or more risk factors such as environmental exposure, presence of one or more genetic mutations or variants, family history, etc. In some embodiments, the subject has not been diagnosed with cancer.
  • the cancer is associated with TEM-1 expression.
  • TEM-1 is expressed in tumor stromal cells (e.g., pericytes, myofibroblasts, or epithelial cells such as vascular epithelial cells).
  • the cancer comprises cancer cells that themselves express TEM-1.
  • the cancer is a solid tumor cancer, e.g., a carcinoma, sarcoma, melanoma, or lymphoma.
  • the solid tumor cancer is a carcinoma, e.g., adenocarcinoma, squamous cell carcinoma, or adenosquamous carcinoma.
  • Non-limiting examples of carcinomas include adenoid cystic carcinoma, adrenocortical carcinoma, bladder cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, gallbladder carcinoma, gastric cancer, head and neck cancer, lung cancer (e.g., small cell lung cancer or non-small cell lung cancer, or adenocarcinoma of the lung), neuroblastoma, neuroendocrine cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, testicular cancer.
  • the solid tumor cancer is a sarcoma.
  • Non-limiting examples of sarcomas include angiosarcoma or hemangioendothelioma, astrocytoma, chondrosarcoma, Ewing’s sarcoma, fibrosarcoma, glioma, leiomyosarcoma, liposarcoma, malignant fibrous histiocytoma (MFH), mesenchymous or mixed mesodermal tumor, mesothelial sarcoma or mesothelioma, myxosarcoma, osteosarcoma, rhabdomyosarcoma, and synovial sarcoma.
  • the cancer is neuroblastoma.
  • the cancer is a brain cancer. In some embodiments, the cancer is a glioma. [0194] In some embodiments, the cancer is melanoma. [0195] In some embodiments, the cancer is a non-solid tumor cancer, e.g., a liquid cancer or hematologic cancer. In some embodiments, the cancer is a myeloma, e.g., multiple myeloma. In some embodiments, the cancer is a leukemia, e.g., acute myeloid leukemia.
  • the cancer is a mixed type cancer, e.g., mixed mesodermal tumor, carcinosarcoma, or teratocarcinoma.
  • Administration and dosage may be administered by any of a variety of routes of administration, including systemic and local routes of administration [0198]
  • Systemic routes of administration include parenteral routes and enteral routes.
  • radioimmunoconjugates or pharmaceutical compositions thereof are administered by a parenteral route, for example, intravenously, intraarterially, intraperitoneally, subcutaneously, or intradermally.
  • radioimmunoconjugates or pharmaceutical compositions thereof are administered intravenously.
  • radioimmunoconjugates or pharmaceutical compositions thereof are administered by an enteral route of administration, for example, trans-gastrointestinal, or orally.
  • enteral route of administration for example, trans-gastrointestinal, or orally.
  • Local routes of administration include, but are not limited to, peritumoral injections and intratumoral injections.
  • Pharmaceutical compositions can be administered for radiation treatment planning, diagnostic, and/or therapeutic treatments. When administered for radiation treatment planning or diagnostic purposes, the radioimmunoconjugate may be administered to a subject in a diagnostically effective dose and/or an amount effective to determine the therapeutically effective dose.
  • compositions may be administered to a subject (e.g., a human) already suffering from a condition (e.g., cancer) in an amount sufficient to cure or at least partially arrest the symptoms of the disorder and its complications.
  • a condition e.g., cancer
  • An amount adequate to accomplish this purpose is defined as a “therapeutically effective amount,” an amount of a compound sufficient to substantially improve at least one symptom associated with the disease or a medical condition.
  • an agent or compound that decreases, prevents, delays, suppresses, or arrests any symptom of the disease or condition would be therapeutically effective.
  • a therapeutically effective amount of an agent or compound is not required to cure a disease or condition but may, for example, provide a treatment for a disease or condition such that the onset of the disease or condition is delayed, hindered, or prevented, such that the disease or condition symptoms are ameliorated, or such that the term of the disease or condition is changed. For example, the disease or condition may become less severe and/or recovery is accelerated in an individual.
  • a subject is administered a first dose of a radioimmunoconjugate or composition in an amount effective for radiation treatment planning, then administered a second dose or set of doses of the radioimmunoconjugate or composition in a therapeutically effective amount.
  • Effective amounts may depend on the severity of the disease or condition and other characteristics of the subject (e.g., weight).
  • Therapeutically effective amounts of disclosed radioimmunoconjugates and compositions for subjects can be determined by the ordinarily-skilled artisan with consideration of individual differences (e.g., differences in age, weight, and the condition of the subject.
  • disclosed radioimmunoconjugates exhibit an enhanced ability to target cancer cells.
  • effective amount of disclosed radioimmunoconjugates are lower than (e.g., less than or equal to about 90%, 75%, 50%, 40%, 30%, 20%, 15%, 12%, 10%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of) the equivalent dose for a therapeutic effect of the unconjugated, and/or non-radiolabeled targeting moiety.
  • Single or multiple administrations of pharmaceutical compositions disclosed herein including an effective amount can be carried out with dose levels and pattern being selected by the treating physician.
  • Dose and administration schedule can be determined and adjusted based on the severity of the disease or condition in the subject, which may be monitored throughout the course of treatment according to the methods commonly practiced by clinicians or those described herein.
  • the following specific examples are to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.
  • EXAMPLES Example 1. General materials and methods [0205] Lutetium-177 can be obtained from Perkin Elmer as lutetium trichloride in a 0.05 N hydrochloric acid solution; indium-111, as a trichloride salt, can be obtained from BWX Technologies and actinium-225 can be obtained as actinium-225 trinitrate from Oak Ridge National Laboratories.
  • Analytical HPLC-MS can be performed using a Waters Acquity HPLC-MS system comprised of a Waters Acquity Binary Solvent Manager, a Waters Acquity Sample Manager (samples cooled to 10 ⁇ C), a Water Acquity Column Manager (column temperature 30 ⁇ C), a Waters Acquity Photodiode Array Detector (monitoring at 254 nm and 214 nm), a Waters Acquity TQD with electrospray ionization and a Waters Acquity BEH C18, 2.1 ⁇ 50 (1.7 ⁇ m) column.
  • a Waters Acquity HPLC-MS system comprised of a Waters Acquity Binary Solvent Manager, a Waters Acquity Sample Manager (samples cooled to 10 ⁇ C), a Water Acquity Column Manager (column temperature 30 ⁇ C), a Waters Acquity Photodiode Array Detector (monitoring at 254 nm and
  • Preparative HPLC can be performed using a Waters HPLC system comprised of a Waters 1525 Binary HPLC pump, a Waters 2489 UV/Visible Detector (monitoring at 254 nm and 214 nm) and a Waters XBridge Prep phenyl or C1819 ⁇ 100 mm (5 ⁇ m) column.
  • a Waters HPLC system comprised of a Waters 1525 Binary HPLC pump, a Waters 2489 UV/Visible Detector (monitoring at 254 nm and 214 nm) and a Waters XBridge Prep phenyl or C1819 ⁇ 100 mm (5 ⁇ m) column.
  • Analytical Size Exclusion Chromatography can be performed using a Waters system comprised of a Waters 1525 Binary HPLC pump, a Waters 2489 UV/Visible Detector (monitoring at 280 nm), a Bioscan Flow Count radiodetector (FC-3300) and TOSOH TSKgel G3000SWxl, 7.8 ⁇ 300 mm column.
  • MALDI-MS positive ion
  • MALDI Bruker Ultraflextreme Spectrometer a MALDI Bruker Ultraflextreme Spectrometer.
  • Radio thin-layer chromatography can be performed with Bioscan AR- 2000 Imaging Scanner, and can be carried out on iTLC-SG glass microfiber chromatography paper (Agilent Technologies, SGI0001) plates using citrate buffer (0.1 M, pH 5.5).
  • the immunoconjugate Compound C-hMP-E-8.3 was eluted from the column with acetate buffer (pH 6.5). Identities of eluates were confirmed by MALDI-TOF. (Throughout, the name “Compound C-hMP-E-8.3” refers to the conjugate formed after reacting Compound C, which contains a crosslinking moiety, with hMP-E-8.3. The conjugate does not contain the cross-linking moiety.) [0228] To make [ 177 Lu]-Compound C-hMP-E-8.3, Lu-177 (0.98 mCi) was added to a solution of Compound C-hMP-E-8.3 (100 ⁇ g in SABST (pH 6.5)).
  • the radiolabeling reaction was incubated at 37° C for 30 minutes.
  • the crude product was purified via a Sephadex G-50 resin packed column eluted with acetate buffer.
  • the chelate-antibody-ratio of [ 177 Lu]- Compound C-hMP-E-8.3 was approximately 4.9.
  • the radiolabeling reaction was incubated at 37° C for 1 hour.
  • the crude product was purified via a Sephadex G-50 resin packed column eluted with acetate buffer.
  • the chelate-antibody-ratio of [ 225 Ac]- Compound C-hMP-E-8.3 was approximately 4.9.
  • [0230] [ 177 Lu]-Compound C-hMP-E-8.3 and [ 225 Ac]-Compound C-hMP-E-8.3 were formulated in SABST (pH 6.5) with 10 mM ascorbate and 1 mM DTPA, and product purity was analyzed by size exclusion chromatography and radio thin layer chromatography.
  • FIG.4A shows results for SK-N-AS cells
  • FIG.4B shows results for SJSA-1 cells.
  • TEM-1 was expressed at high levels in SJSA-1 cells (FIG.4A) and at intermediate levels in SK-N-AS cells (FIG.4B).
  • FIG.7 shows the percentage injected dose per gram (% ID/g) in the blood, bone, heart, intestines, kidneys and adrenals, liver and gallbladder, lungs, muscle, pancreas, spleen, stomach, and tumor at each timepoint.
  • FIG.7 shows rapid blood clearance for a radioimmunoconjugate, in this example, dropping from ⁇ 44% ID/g at 4 h to ⁇ 9% ID/g at 7 days. The distribution in off- target tissues was low, ⁇ 5% ID/g after 7 days.
  • mice were dosed with single doses of 10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi
  • Vehicle and antibody only controls were also included. Tumor volumes were monitored and assessed for 28 days after dosing.
  • FIG.8 shows relative tumor volumes over time.
  • [ 225 Ac]-Compound C-hMP-E-8.3 was highly effective in reducing tumor growth, with tumors appearing to regress at higher doses.
  • Example 9 shows relative tumor volumes over time.
  • mice Female Balb/c mice (5 per group) were injected with SK-N-AS cells to induce subcutaneous tumor xenografts. Once tumors reached a volume of ⁇ 200 mm 3 , mice were dosed with single doses of 10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi [ 225 Ac]- Compound C-hMP-E-8.3.
  • FIG.9 shows relative tumor volumes over time.
  • [ 225 Ac]-Compound C-hMP-E-8.3 was highly effective in reducing tumor growth, with tumors appearing to regress at higher doses.
  • Example 10. Efficacy of [ 225 Ac]-Compound C-hMP-E-8.3 in an Ewing’s sarcoma xenograft model
  • [0241] [ 225 Ac]-Compound C-hMP-E-8.3 was tested in a human Ewing’s sarcoma cancer xenograft model.
  • mice Female Balb/c mice (5 per group) were injected with A673 cells to induce subcutaneous tumor xenografts. Once tumors reached a volume of ⁇ 200 mm 3 , mice were dosed with single doses of 10 nCi, 50 nCi, 100 nCi, 200 nCi, or 400 nCi [ 225 Ac]-Compound C-hMP-E-8.3. Vehicle and antibody only controls were also included. Tumor volumes were monitored and assessed for 28 days after dosing. [0242] FIG.10 shows relative tumor volumes over time. [ 225 Ac]-Compound C-hMP-E- 8.3 reduced tumor growth at the high dose.
  • the Ewing’s sarcoma cells line used in this Example is known to express lower levels of TEM-1 than the osteosarcoma and neuroblastoma cell lines used in Examples 7-9 and therefore demonstrates the ability of [ 225 Ac]-Compound C-hMP-E-8.3 to be effective against tumors that express low levels of TEM-1.
  • Example 11 Effects of [ 225 Ac]-Compound C-hMP-E-8.3 on tumor stroma [0243] In human cancers, TEM-1 is often expressed in stromal cells associated with tumors.
  • TEM-1 may not be expressed (or only expressed at low levels) in the cancerous cells themselves but remains expressed in the stromal compartment.
  • hMP-E-8.3 binds to human TEM-1 but shows little to no cross-reactivity to mouse TEM-1 and is therefore unable to bind to mouse derived stromal cells in a conventional tumor xenograft model where human TEM- 1 is expressed in human cancer cells supported by mouse derived stromal cells.
  • the mice in this model are genetically engineered to express human TEM-1, instead of mouse TEM-1, in cells including the stromal cells.
  • cells of a cancer cell line that do not express endogenous human TEM- 1 are implanted into the transgenic mice.
  • the only human TEM-1 target expressed in the resulting tumor is derived from the humanized mouse stromal cells and the therapeutic effect of targeting human TEM-1 in those stromal cells with [ 225 Ac]-Compound C-hMP-E-8.3 may be determined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Optics & Photonics (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
PCT/US2021/025800 2020-04-06 2021-04-05 Tem-1-targeted radioimmunoconjugates and uses thereof WO2021207086A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063006040P 2020-04-06 2020-04-06
US63/006,040 2020-04-06

Publications (1)

Publication Number Publication Date
WO2021207086A1 true WO2021207086A1 (en) 2021-10-14

Family

ID=78022984

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/025800 WO2021207086A1 (en) 2020-04-06 2021-04-05 Tem-1-targeted radioimmunoconjugates and uses thereof

Country Status (3)

Country Link
AR (1) AR122411A1 (zh)
TW (1) TW202204336A (zh)
WO (1) WO2021207086A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023050008A1 (en) * 2021-09-29 2023-04-06 National Research Council Of Canada Egfrviii-targeted compounds and uses thereof
WO2023122588A3 (en) * 2021-12-20 2023-09-21 Fusion Pharmaceuticals Inc. Egfr-cmet–targeted compounds and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3202788A1 (en) * 2016-02-05 2017-08-09 MediaPharma S.r.l. Endosialin-binding antibody
WO2020115556A1 (en) * 2018-12-03 2020-06-11 Fusion Pharmaceuticals Inc. Radioimmunoconjugates and checkpoint inhibitor combination therapy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3202788A1 (en) * 2016-02-05 2017-08-09 MediaPharma S.r.l. Endosialin-binding antibody
WO2020115556A1 (en) * 2018-12-03 2020-06-11 Fusion Pharmaceuticals Inc. Radioimmunoconjugates and checkpoint inhibitor combination therapy

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CICONE ET AL.: "Preclinical Evaluation and Dosimetry of [111ln]CHX-DTPA-scFv78-Fc Targeting Endosialin/Tumor Endothelial Marker 1 (TEM1", MOLECULAR IMAGING AND BIOLOGY, vol. 22, 28 January 2020 (2020-01-28), pages 979 - 991, XP037186995, DOI: 10.1007/s11307-020-01479-8 *
DATABASE PUBCHEM [online] 12 February 2015 (2015-02-12), XP055866322, Database accession no. 231891810 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023050008A1 (en) * 2021-09-29 2023-04-06 National Research Council Of Canada Egfrviii-targeted compounds and uses thereof
WO2023122588A3 (en) * 2021-12-20 2023-09-21 Fusion Pharmaceuticals Inc. Egfr-cmet–targeted compounds and uses thereof

Also Published As

Publication number Publication date
AR122411A1 (es) 2022-09-07
TW202204336A (zh) 2022-02-01

Similar Documents

Publication Publication Date Title
CN111263747A (zh) 双官能螯合物的药代动力学增强及其用途
CN110891614A (zh) Igf-1r单克隆抗体及其用途
WO2020115548A1 (en) Radioimmunoconjugates and dna damage and repair inhibitor combination therapy
WO2021207086A1 (en) Tem-1-targeted radioimmunoconjugates and uses thereof
US20230091468A1 (en) Sustained immunotherapy
US20240139353A1 (en) Methods of treating cancer
WO2023050008A1 (en) Egfrviii-targeted compounds and uses thereof
WO2023122588A2 (en) Egfr-cmet–targeted compounds and uses thereof
US20230201384A1 (en) Fgfr3-targeted radioimmunoconjugates and uses thereof
WO2023070202A1 (en) Claudin 18.2-targeted compounds and uses thereof
WO2023081698A1 (en) Methods of treating cancer

Legal Events

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

Ref document number: 21785288

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21785288

Country of ref document: EP

Kind code of ref document: A1