[go: up one dir, main page]

WO2025088200A2 - Ligands de la phosphatase acide 3 pour applications d'administration ciblée - Google Patents

Ligands de la phosphatase acide 3 pour applications d'administration ciblée Download PDF

Info

Publication number
WO2025088200A2
WO2025088200A2 PCT/EP2024/080350 EP2024080350W WO2025088200A2 WO 2025088200 A2 WO2025088200 A2 WO 2025088200A2 EP 2024080350 W EP2024080350 W EP 2024080350W WO 2025088200 A2 WO2025088200 A2 WO 2025088200A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
independently selected
independently
optionally substituted
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/EP2024/080350
Other languages
English (en)
Inventor
Samuele CAZZAMALLI
Tony Georgiev
Sebastian OEHLER
Francesca MIGLIORINI
Dario Neri
Young Seo PARK KIM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Philochem AG
Original Assignee
Philochem AG
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 Philochem AG filed Critical Philochem AG
Publication of WO2025088200A2 publication Critical patent/WO2025088200A2/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • 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
    • A61K51/0402Organic compounds carboxylic acid carriers, fatty acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • A61K51/0446Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • 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
    • A61K51/0489Phosphates or phosphonates, e.g. bone-seeking phosphonates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having three nitrogen atoms as the only ring hetero atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6524Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having four or more nitrogen atoms as the only ring hetero atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/6552Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a six-membered ring
    • C07F9/65522Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a six-membered ring condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65615Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing a spiro condensed ring system of the formula where at least one of the atoms X or Y is a hetero atom, e.g. S
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)]
    • C07F9/3804Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)] not used, see subgroups
    • C07F9/3882Arylalkanephosphonic acids

Definitions

  • the present invention relates to ligands against Acid Phosphatase 3 (ACP3 or ACPP), also known as Prostatic Acid Phosphatase (PAP).
  • ACP3 or ACPP Acid Phosphatase 3
  • PAP Prostatic Acid Phosphatase
  • Selective ACP3 ligands may be able to exclusively interact with antigens expressed on the surface of tumor cells for in vivo pharmaco-delivery applications.
  • the ligand may display very high affinity and selectivity towards ACP3 to enable targeted delivery of a payload, including therapeutic and diagnostic payloads, to a site afflicted by or at risk of a disease characterized by the expression of ACP3.
  • cytotoxic agents are at the basis of the treatment of cancer and other pathological conditions. Ideally, cytotoxic agents should accumulate at site of disease, sparing normal tissues. However, many anticancer drugs do not preferentially accumulate in solid tumors. Indeed, it has been demonstrated in tumorbearing mice and in human patients that only a minimal portion of the injected drug reaches the neoplastic mass in comparison to the amount of cytotoxic agent that reaches healthy organs.
  • the targeted delivery of highly potent cytotoxic agents into diseased tissues is therefore desirable for the treatment of cancer and other serious conditions.
  • a therapeutic effector By attaching a therapeutic effector through a site-specific cleavable linker to a binding moiety specific to a marker of disease, the effector preferentially accumulates and acts at the intended site of action, thus increasing the effectively applied dose while reducing side effects.
  • ADCs Antibody-Drug Conjugates
  • Ligand-based pharmacodelivery strategies fundamentally rely on the identification of good-quality markers of pathology, allowing a clear-cut discrimination between diseased tissues and healthy organs.
  • Monoclonal antibodies and their fragments represent the preferred agents for pharmacodelivery applications! 1-2 ], but globular protein mutants 13 ], peptides 14 ] and even small organic binding moieties [5 l are also increasingly being used.
  • SMDCs Small Molecule-Drug Conjugates
  • SMRCs Small Molecule-Radio Conjugates
  • Acid phosphatase 3 is a tyrosine phosphatase expressed as homodimer for which five distinct isoenzymes have been reported in humans, mostly involved in immune defense, growth regulation and bone resorption. I 11-13 ]
  • A. and E. Gutman observed elevated serum levels of acid phosphatases in prostate cancer patients and ACP3 eventually emerged as a clinically validated prognostic marker for prostate cancer which was later replaced by prostate-specific antigen. 114-161 Based on immunohistochemistry, low ACP3 expression has been detected in most human tissues while exceptionally high expression was observed in prostate and prostate cancer.
  • ACP3 isoform 1 is expressed as secreted form or with a transmembrane domain (type I, TM-PAP) as a result of alternative splicing of the same gene.
  • TM-PAP has been shown to localize to the plasma membrane and to vesicles, likely due to internalization as suggested by the presence of a short intracellular lysosomal targeting motif.
  • PCa prostate cancer
  • prostate cancer treatments include surgery, radiation therapy, hormonal therapy (androgen deprivation therapy) and chemotherapy.
  • SMRCs targeting Prostate Specific Membrane Antigen (PSMA) have been developed up to registration in the field of prostate cancer. Those agents are limited by their strong accumulation in healthy tissues such as salivary glands and kidneys. The unwanted accumulation of PSMA-targeting agents in those healthy tissues causes side effects and limits the escalation of their dose to curative regimens.
  • PluvictoTM INN: lutetium ( 177 Lu) vipivotide tetraxetan
  • PluvictoTM does not cure and the accumulation of the radioligand in healthy organs such as salivary glands and kidneys prevents the administration of higher doses. 1211
  • PSMA binders Considering the limitation of prostate cancer therapy using PSMA binders, there is a need for therapeutics that can efficiently accumulate on tumors while sparing normal organs. This is particularly true in the context of radioligand therapeutics delivering alpha emitters.
  • alpha-emitter radionuclide payloads such as Actinium-225 ( 225 Ac) and Lead-212 ( 212 Pb)
  • PSMA-targeting agents are more efficacious in patients relapsing from Lutetium-177-based PSMA targeting agents, but more toxic. 1591
  • the accumulation of PSMA-targeted alpha emitters in the kidney limits the development of those therapeutic candidates, leading to severe and irreversible damage to this vital healthy organ.
  • ACP3 has been recognized as a therapeutic target for the development of vaccines to generate anti-tumor immunity.
  • Ongoing clinical trials investigate the effect of ACP3 immunization in prostate cancer patients (www.clinicaltrials.gov identifier: NCT03600350 and NCT01706458) and a significant survival benefit could be identified in a placebo-controlled phase III trial. 1221
  • antibody fragments and whole IgG antibodies targeting ACP3 have been shown to accumulate in prostate cancer lesions, both in mouse models and in human patients with metastatic prostate cancer. 123-261
  • the present invention aims at solving the problem of identifying and developing binders for ACP3 (PAP, ACPP, TM-ACP3, TM-PAP) which may serve as targeting moiety suitable for applications in the background of targeted delivery of diagnostic or therapeutic payloads to a site afflicted or at risk of a disease characterized by the expression of ACP3.
  • ACP3 binders for ACP3 (PAP, ACPP, TM-ACP3, TM-PAP) which may serve as targeting moiety suitable for applications in the background of targeted delivery of diagnostic or therapeutic payloads to a site afflicted or at risk of a disease characterized by the expression of ACP3.
  • FIG. 1 Enzymatic inhibition measurements with compounds 1 , 2, 3, and 5. The assay was performed according to protocol 1 .
  • FIG. 2 Enzymatic assay experiments with compounds 1 , 5, 6a-c, and 7a-c. DOTA-GA moieties were introduced in the ortho, meta, and para positions of the benzyl (A) and phenyl (B) sides of a- aminophosphonic acid 1 and their IC50 values were compared. IC50 value for compound 7c (meta, not shown): 36 nM. The assay was performed according to protocol 1 .
  • FIG. 3 Advantages of using 4-azido proline as a scaffold extension approach (A). Enzymatic assay experiment with compounds 1 , 9, and 10 were performed according to protocol 1 (B).
  • FIG. 4 Enzymatic assay experiments with compounds O1a-d, O2a-d, 6c, 11 , and 12.
  • the stereochemical impact of the proline scaffold was studied on-DNA along with the gain in activity brought by the N, A/-di benzyl glycine building block.
  • the assay in (A) was performed according to protocol 1 ; in (B) according to protocol 2 as higher sensitivity was needed to detect the sub-nanomolar inhibitor.
  • FIG. 5 (A) Advantages of using 3-lodo-phenylalanine as scaffold extension approach.
  • B Enzymatic assay experiment with on-DNA compounds O3a, b, O4a, b and 05a, b were performed according to protocol 2.
  • C D
  • the 4 new ligands bearing DOTA-GA 14a,b and 16a,b were evaluated against DOTA-GA metaderivative 6b via enzymatic assay- protocol 2, revealing IC50 values in the low nanomolar range.
  • FIG. 6 FP Selectivity screening of FITC labelled compound 4 against a panel of serum proteins and phosphatases
  • A FP binding comparison between FITC labelled compounds 4, 8, and 13 against ACP3.
  • B FP binding comparison between FITC labelled compounds 4, 8, and 13 against ACP3.
  • C FP binding comparison between FITC labelled compounds 8, 15a,b and 17a,b against ACP3.
  • D FP Selectivity screening of FITC labelled compounds 15a against a panel of serum proteins and phosphatases.
  • E FP Selectivity screening of FITC labelled compounds 15b against a panel of serum proteins and phosphatases.
  • F FP Selectivity screening of FITC labelled compounds 17a against a panel of serum proteins and phosphatases.
  • G FP Selectivity screening of FITC labelled compounds 17b against a panel of serum proteins and phosphatases.
  • FIG. 7 Flow cytometry experiment with compounds 8, 13, 15a, b, 17a, b against HT1080.hACP3 cells. A clear shift was observed for all molecules.
  • FIG. 8 Flow cytometry experiment with compounds 8, 13, 15a,b, 17a,b against HT1080 wild type cells. No shift was observed for the compounds.
  • FIG. 9 SPR sensograms of compounds 6c (A), 12a (B), 12b (C), 14a (D), 14b (E), 16a (F), 16b (G) against human ACP3, immobilized on a CM5 chip.
  • the dissociation constants (Kd) of each compound are reported in brackets.
  • FIG. 10 Radiosynthesis of compound 19 and HPLC chromatogram of the final product (Compound 19) as recorded with a radio-detector.
  • FIG. 11 Radiosynthesis of compounds 20a and 20b, and HPLC chromatogram of Compound 20a (SS) as recorded with a radio-detector.
  • FIG. 12 HPLC chromatogram of Compound 20b (RR) as recorded with a radio-detector.
  • FIG. 13 Radiosynthesis of compounds 21a and 21 b, and HPLC chromatogram of Compound 21a (S) as recorded with a radio-detector.
  • FIG. 14 HPLC chromatogram of Compound 21 b (R) as recorded with a radio-detector.
  • FIG. 15 Radiosynthesis of compounds 22a and 22b, and HPLC chromatogram of Compound 22a (S) as recorded with a radio-detector.
  • FIG. 16 HPLC chromatogram of Compound 22b (R) as recorded with a radio-detector.
  • FIG. 25 Therapeutic experiments in male BALB/c nu/nu mice bearing HT1080.hACP3 xenografts.
  • A Compound 20a ( 177 Lu-ProX1-(SS)-DOTAGA) injected at molar activities of 250 MBq/kg or 1000 MBq/kg, and compound 22a ( 177 Lu-ProX3-(S)-DOTAGA) injected at molar activity of 1000 MBq/kg.
  • B Body weight changes of animals throughout the therapy study.
  • FIG. 26 Autoradiography ex vivo results after exposure of (i) HT1080.hACP3 xenografts (ii) HT1080.hPSMA xenografts (iii) human prostate cancer, and (iv) human salivary gland tissues to compounds 19, 20a, 22a. and 177 Lu-PSMA-617 ( 177 Lu vipivotide tetraxetan)
  • FIG. 32 Radiosynthesis of compounds 26a and 26b, and HPLC chromatogram of Compound 26a as recorded with a radio-detector.
  • FIG. 33 Radiosynthesis of compounds 28a and 28b, and HPLC chromatogram of Compound 28a as recorded with a radio-detector.
  • FIG. 34 Radiosynthesis of 68 Ga-ProX1-(SS)-DOTA (compound 44), and HPLC chromatogram of Compound 44 as recorded with a radio-detector.
  • FIG. 37 In vivo biodistribution studies with 68 Ga-ProX1-(SS)-DOTA (44). Mice in the pre-blocking group were injected with cold ProX1-(SS)-DOTA (27a) (50 nmol/mouse, ⁇ 2.5 ⁇ mol/kg - corresponding to a 40-fold molar excess as compared to the radioactive compound) 30 min before the administration of 68 Ga-ProX1-(SS)- DOTA (44). Individual values are represented by circles for which bars display the average group %ID/g values.
  • FIG. 38 Radioligand bead-based assay with 177 Lu-ProX1-(SS)-DOTA (28a) and 177 Lu-ProX3-(S)-DOTAGA (22a). Magnetic Streptavidin-coated DynabeadsTM M-280 were functionalized with recombinant ACP3 and exposed to test compounds 28a and 22a, without (full bars) or with (dashed bars) a 5000-fold molar excess of blocking cold ACP3 ligands.
  • Black arrows indicate intravenous administrations of SMDCs 31 and 36 at 5 nmol/mouse (250 nmol/kg) or 100 ⁇ L of PBS (vehicle). Data is presented as mean ⁇ standard error of the mean (SEM).
  • Black arrows indicate intravenous administrations of SMDC 31 at 5 nmol/mouse (250 nmol/kg) or 100 ⁇ L of PBS (vehicle). Data is presented as mean ⁇ standard error of the mean (SEM).
  • Black arrows indicate intravenous administrations of compound 27a at 5 nmol/mouse (250 nmol/kg) or 100 ⁇ L of PBS (vehicle). Data is presented as mean ⁇ standard error of the mean (SEM).
  • FIG. 42 Enzymatic inhibition measurements with compounds 1 , 47, 48, 49 and 50. The assay was performed according to protocol 2.
  • FIG. 43 Enzymatic inhibition measurements with compounds 27a, 51 , 53, 55a, 55b, 55c and 55d. The assay was performed according to protocol 2.
  • FIG. 44 Enzymatic inhibition measurements with compounds 27a, 56a and 56b. The assay was performed according to protocol 2.
  • FIG. 45 Enzymatic inhibition measurements with compounds 27a, 57a and 57b. The assay was performed according to protocol 2.
  • FIG. 46 Enzymatic inhibition measurements with compounds 27a, 58, 59 and 60. The assay was performed according to protocol 2.
  • FIG. 48 Radiosynthesis of compound 52 and HPLC chromatogram of the final product (Compound 52) as recorded with a radio-detector.
  • FIG. 49 Radiosynthesis of compound 54 and HPLC chromatogram of the final product (Compound 54) as recorded with a radio-detector.
  • the binders of the present invention can rapidly accumulate to ACP3-positive tumors.
  • the compounds do not substantially accumulate in healthy organs such as kidneys, salivary glands, or normal prostate. This is advantageous both for diagnostic applications (e.g., to visualize tumor lesions in normal prostate) and therapeutic applications (e.g., to minimize toxicity in vital healthy organs).
  • the binders of the present invention can achieve surprisingly long residence time in the tumor and promote potent in vivo anti-cancer activity without internalization.
  • radiolabeled conjugates preferably include accumulation to ACP3-positive solid cancer lesions with long residence time (e.g., t1/2 >72 hours) and high selectivity (e.g., tumor-to-blood ratio >148 at 2 hours after administration).
  • long residence time e.g., t1/2 >72 hours
  • high selectivity e.g., tumor-to-blood ratio >148 at 2 hours after administration.
  • bone marrow toxicity has been observed as a common side effect of Lutetium-177-based RLT products.
  • radiolabeled compounds of the present invention tend not to accumulate in healthy bones, while offering highly selective tumor uptake at early time points (e.g., 1 h post-injection).
  • conjugates with cytotoxic or cytostatic payloads, such as MMAE include potent in vivo anti-cancer activity.
  • Compounds of the present invention may provide low healthy organ toxicity and highly selective tumor uptake. Considering the efficient tumor targeting and the lack of uptake observed in healthy organs salivary glands and kidneys, compounds of the present invention may offer an improvement to prostate-specific membrane antigen ligands, e.g., for the targeting of metastatic prostate cancer.
  • Prostate cancer patients with low PSMA levels or relapsing from therapy e.g., with PSMA-617 ( 77 Lu vipivotide tetraxetan), may particularly benefit from the administration of the therapeutics of the present invention.
  • PSMA-617 77 Lu vipivotide tetraxetan
  • a compound that specifically binds ACP3, and has a molecular weight of 5000 Da or less and/or a dissociation constant (Kd) of 50 nM or less.
  • the compound specifically binding ACP3 has a molecular weight of 5000 Da or less and an ACP3 dissociation constant (Kd) of 50 nM or less.
  • the molecular weight and the dissociation constant are as defined below.
  • the molecular weight is 4000 Da or less, or 3000 Da or less. In another embodiment, the molecular weight is 500 Da or more, 600 Da or more, or 800 Da or more. In another embodiment, the molecular weight is of from 500 Da to 5000 Da, from 600 Da to 3000 Da, or from 800 to 3000 Da.
  • the dissociation constant (K ⁇ ) is 45 nM or less, 40 nM or less, 35 nM or less, 30 nM or less, 25 nM or less, 20 nM or less, 15 nM or less, 10 or less, or 5 nM or less. In a preferred embodiment, the dissociation constant is 5 nM or less.
  • the dissociation constant (K ⁇ ) is 0.5 nM or more, 1 nM or more, or 2 nM or more. In a preferred embodiment, the dissociation constant is 2 nM or more.
  • the compound according to the present invention may specifically bind ACP3 on the membrane of tumor cells and/or may be not substantially internalized.
  • the term “not substantially internalized” is used to indicate that a compound or substance is taken up by cells in an amount relative to the sum of internalized and non-internalized amount of s 20%, preferably s 10%, more preferably wherein the internalized amount is non-detectable, e.g., as measured by confocal microscopy, a radioactivity-based internalization assay, mass spectrometry, and the like.
  • the compound according to the present invention may show a higher uptake in tumor(s) than in healthy organ(s) after administration, e.g., intravenous administration.
  • the compound according to the present invention shows an uptake of 10% ID/g or more, 15% ID/g or more, 20% ID/g or more, 25% ID/g or more, 30% ID/g or more, 35% ID/g or more, or 40% ID/g or more in tumor(s) after administration, e.g., intravenous administration, in male mice bearing HT1080.hACP3 xenografts.
  • the compound according to the present invention shows an uptake of less than 10% I D/g , 8% I D/g or less, 5% ID/or less in healthy organ(s) after administration, e.g., intravenous administration.
  • the healthy organ(s) do not include gall bladder and/or urinary bladder.
  • the uptake in tumor(s) and healthy organ(s) may be determined after 1 hour, 2 hours, 6 hours or 24 hours, preferably 1 hour, after administration, e.g., intravenous administration.
  • the compound according to the present invention preferably does not substantially accumulate in kidneys and/or salivary glands and/or healthy prostate after administration, e.g., intravenous administration.
  • the phrase “does not substantially accumulate” is used herein to indicate that the compound is taken up by these organs in an amount of less than 10% I D/g , 8% I D/g or less, more preferably 5% ID/or less after administration, e.g., intravenous administration.
  • Tables 2.1, 2.2 and 2.3 are shown in Tables 2.1, 2.2 and 2.3. These may be particularly useful in imaging/tracing applications (e.g., PET imaging and/or diagnostics), when comprising a suitable nuclide, such as 18 F (Table 2.2.); or as cold versions (Table 2.1) of such compounds (e.g., as standards during GMP manufacturing); or as intermediates or precursors for 18 F labeling (Table 2.3).
  • B-1 bond Table 3.1.
  • Moiety B is a covalent bond or a moiety comprising a chain of atoms that covalently attaches moiety R 1 -Y or R 1 to the payload C, e.g., through one or more covalent bond(s).
  • the moiety B may be cleavable or non- cleavable, multifunctional moiety which can be used to link one or more payload and/or binder moieties to form the targeted conjugate of the invention.
  • moiety B is a multifunctional moiety linking at least one moiety C with at least one moiety R 1 -Y or R 1 .
  • B can be a single bond, or an optionally substituted C-1-50 aliphatic group, in which optionally one or more carbon atoms can be replaced by a heteroatom, a carbocyclic or a C-1-12 heterocyclic group, and which can be saturated, or optionally contain one or more double or triple bonds.
  • a therapeutic effector in particular: a cytotoxic or cytostatic payload
  • a site-specific cleavable linker to a binding moiety specific to a marker of disease
  • the effector preferentially accumulates and acts at the intended site of action, thus increasing the effectively applied dose while reducing side effects.
  • moiety B generally may be cleavable or non-cleavable, yet it is preferred that when moiety C is a cytotoxic or cytostatic payload, e.g., a chemotherapeutic (cytotoxic or cytostatic) agent, such as MMAE, a cleavable moiety B is used, which is contemplated to be advantageous from the viewpoint of payload release, accumulation of (free) payload and/or anti-tumor activity.
  • a cytotoxic or cytostatic payload e.g., a chemotherapeutic (cytotoxic or cytostatic) agent, such as MMAE
  • cleavable linkers can be advantageous where release of the payload C is desirable, e.g., in the case of chemotherapeutic (cytotoxic or cytostatic) agent, cleavable linkers should not be understood to be generally mandatory or essential for the functioning of the compounds of the present invention. For instance, in the cases of radioconjugates useful as radiotherapeutic and/or diagnostic agents, cleavable linkers are not contemplated to be particularly required.
  • Moiety B can comprise or consist of a unit shown in Table 4 below wherein the substituents R and R n shown in the formulae may suitably be independently selected from H, halogen, substituted or unsubstituted (hetero)alkyl, (hetero)alkenyl, (hetero)alkynyl, (hetero)aryl, (hetero)arylalkyl, (hetero)cycloalkyl, (hetero)cycloalkylaryl, heterocyclylalkyl, a peptide, an oligosaccharide or a steroid group.
  • substituents R and R n shown in the formulae may suitably be independently selected from H, halogen, substituted or unsubstituted (hetero)alkyl, (hetero)alkenyl, (hetero)alkynyl, (hetero)aryl, (hetero)arylalkyl, (hetero)cycloalkyl, (
  • each of R, Ri, R2 and R3 is independently selected from H, OH, SH, NH2, halogen, cyano, carboxy, alkyl, cycloalkyl, aryl and heteroaryl, each of which is substituted or unsubstituted.
  • R and R n are independently selected from H, or C1 -C7 alkyl or heteroalkyl. More suitably, R and R n are independently selected from H, methyl or ethyl. Table 4
  • Moiety B, unit(s) BL and/or unit(s) Bs may suitably comprise as a cleavable bond a disulfide linkage since these linkages are stable to hydrolysis, while giving suitable drug release kinetics at the target in vivo, and can provide traceless cleavage of drug moieties including a thiol group.
  • Moiety B, unit(s) BL and/or unit(s) Bs may be polar or charged in order to improve water solubility of the conjugate.
  • the linker may comprise from about 1 to about 20, suitably from about 2 to about 10, residues of one or more known water-soluble oligomers such as peptides, oligosaccharides, glycosaminoglycans, polyacrylic acid or salts thereof, polyethylene glycol, polyhydroxyethyl (meth) acrylates, polysulfonates, etc.
  • the linker may comprise a polar or charged peptide moiety comprising e.g. from 2 to 10 amino acid residues.
  • Amino acids may refer to any natural or non-natural amino acid.
  • the peptide linker suitably includes a free thiol group, preferably a N-terminal cysteine, for forming the said cleavable disulfide linkage with a thiol group on the drug moiety.
  • a free thiol group preferably a N-terminal cysteine
  • Any peptide containing L- or D-aminoacids can be suitable; particularly suitable peptide linkers of this type are Asp-Arg-Asp-Cys and/or Asp-Lys-Asp-Cys.
  • moiety B, unit(s) BL and/or unit(s) Bs may comprise a cleavable or non- cleavable peptide unit that is specifically tailored so that it will be selectively enzymatically cleaved from the drug moiety by one or more proteases on the cell surface or the extracellular regions of the target tissue.
  • the amino acid residue chain length of the peptide unit suitably ranges from that of a single amino acid to about eight amino acid residues.
  • Numerous specific cleavable peptide sequences suitable for use in the present invention can be designed and optimized in their selectivity for enzymatic cleavage by a particular tumor-associated enzyme e.g. a protease.
  • Cleavable peptides for use in the present invention include those which are optimized toward the proteases MMP-1 , 2 or 3, or cathepsin B, C or D. Especially suitable are peptides cleavable by Cathepsin B.
  • Cathepsin B is a ubiquitous cysteine protease. It is an intracellular enzyme, except in pathological conditions, such as metastatic tumors or rheumatoid arthritis.
  • An example for a peptide cleavable by Cathepsin B is containing the sequence Val-Cit.
  • cleavable peptide units include cleavable peptide unit selected from Gly-Pro, Ala-Pro, Val-Pro, Arg-Pro, lle-Pro, Pro- Pro, Gly-Cit, Ala-Cit, Val-Cit, Arg-Cit, lle-Cit, and Pro-Cit; preferably Gly-Pro or Val-Cit.
  • moiety B, unit(s) BL and/or unit(s) Bs may comprise cleavable unit able of being enzymatically cleaved by one or more phosphatases, sulfatases or esterases. This can be particularly advantageous when targeting cells characterized by increased expression of phosphatases, including, e.g., ACP3.
  • Exemplary cleavable units of this type can be selected from: wherein Q 1 is independently selected from OPO3H2, OPO2N(R)2, OPO2NH2, OPO(OH)F, OC(O)OR, OC(O)R, OSO3H, OSO 2 N(R) 2 , and OSO2NH2; preferably OPO3H2, OC(O)OH or OSO3H, more preferably OPO3H2; most preferably OPO3H2; R ⁇ is independently selected from H an electron withdrawing group, preferably from H, NO2, CN, halogen, C(O)R, CF3, and SO3H, more preferably from H and NO2; and each R is independently as defined for R herein.
  • Preferable cleavable units of this type can be selected from:
  • the moiety B and in particular, unit(s) BL suitably further comprised) self- immolative moiety can or cannot be present after the linker.
  • the self-immolative linkers are also known as electronic cascade linkers. These linkers undergo elimination and fragmentation upon enzymatic cleavage of the peptide to release the drug in active, preferably free form.
  • the conjugate is stable extracellularly in the absence of an enzyme capable of cleaving the linker.
  • the linker upon exposure to a suitable enzyme, the linker is cleaved initiating a spontaneous self-immolative reaction resulting in the cleavage of the bond covalently linking the self-immolative moiety to the drug, to thereby effect release of the drug in its underivatized or pharmacologically active form.
  • the self-immolative linker is coupled to the binding moiety through an enzymatically cleavable peptide sequence that provides a substrate for an enzyme to cleave the amide bond to initiate the self-immolative reaction.
  • the drug moiety is connected to the self-immolative moiety of the linker via a chemically reactive functional group pending from the drug such as a primary or secondary amine, hydroxyl, sulfhydryl or carboxyl group.
  • PABC self-immolative linkers
  • PAB para-aminobenzyloxycarbonyl
  • the amide bond linking the carboxy terminus of a peptide unit and the para-aminobenzyl of PAB may be a substrate and cleavable by certain proteases.
  • B comprises a cleavable peptide unit, (e.g., a dipeptide unit as detailed above), is directly bound to a self-immolative moiety (e.g., PABC or PAB), which, in turn, is bound to a drug moiety (e.g., a g a therapeutic effector, in particular: a cytotoxic or cytostatic payload moiety C), e.g., as shown below:
  • the linker comprises a glucuronyl group that is cleavable by glucuronidase present on the cell surface or the extracellular region of the target tissue. It has been shown that lysosomal betaglucuronidase is liberated extracellularly in high local concentrations in necrotic areas in human cancers, and that this provides a route to targeted chemotherapy (Bosslet, K. et al. Cancer Res. 58, 1195-1201 (1998)).
  • the moiety B suitably further comprises a spacer unit.
  • a spacer unit can be the unit Bs, which may be linked to the binding moiety R 1 -Y or R 1 , for example via an amide, amine or thioether bond.
  • the spacer unit is of a length that enables e.g. the cleavable peptide sequence to be contacted by the cleaving enzyme (e. g. cathepsin B) and suitably also the hydrolysis of the amide bond coupling the cleavable peptide to the self-immolative moiety X.
  • Spacer units may for example comprise a divalent radical such as alkylene, arylene, a heteroarylene, repeating units of alkyloxy (e.g., polyethylenoxy, PEG, polymethyleneoxy) and alkylamino (e.g., polyethyleneamino), or diacid ester and amides including succinate, succinamide, diglycolate, malonate, and caproamide.
  • alkylene e.g., polyethylenoxy, PEG, polymethyleneoxy
  • alkylamino e.g., polyethyleneamino
  • diacid ester and amides including succinate, succinamide, diglycolate, malonate, and caproamide.
  • * represents a point of attachment to moiety R 1 -Y or R 1 or a point of attachment for which the shortest path to moiety R 1 -Y or R 1 comprises less atoms than that for •, as the case may be; and • represents a point of attachment a point of attachment to moiety C or a point of attachment to moiety C for which the shortest path to moiety C comprises less atoms than that for *, as the case may be.
  • a reactive moiety L is present rather than payload moiety C.
  • each * represents a point of attachment for which the shortest path to moiety R 1 -Y or R 1 comprises less atoms than that for •; and each • represents a point of attachment for which the shortest path to moiety C comprises less atoms than that for *, with the proviso that when n is > 1 and a respective point of attachment is indicated on any one of R a , R b and R c , then it can be independently present in one or more of the peptide monomeric units, preferably in one peptide monomeric unit most distant from the other point of attachment indicated in the respective structure.
  • peptide refers to peptide mono- or oligomers having a backbone formed by proteinogenic and/or a non-proteinogenic amino acids.
  • aminoacyl or “aminoacid” generally refer to any proteinogenic or a non-proteinogenic amino acid.
  • the side-chain residues of a proteinogenic or a non-proteinogenic amino acid are represented by any of R a , R b and R c , each of which is selected from the following list: wherein each of R, R 1 , R 2 and R 3 is independently selected from H, OH, SH, NH2, halogen, cyano, carboxy, alkyl, cycloalkyl, aryl and heteroaryl, each of which is substituted or unsubstituted; each Xis independently selected from NH, NR, S, O and CH2, preferably NH; and each n and m is independently an integer preferably selected from 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 1 0, 11 , 12, 13, 14, 15, 16, 17, 18, 19 and 20, wherein the definitions of R, R 1 , R 2 , R 3 , X, m, and n here are independent from the definitions of R 1 , R 2 and R 3 , X, m
  • side-chain residues of a proteinogenic or a non- proteinogenic amino acid are represented by any of R a , R ⁇ , R c , R ⁇ and R e , each of which may be part of a 3-, 4-, 5-, 6- or 7-membered ring.
  • the side chain alpha, beta and/or gamma position of said proteinogenic or non-proteinogenic amino acid can be part of a cyclic structure selected from an azetidine ring, pyrrolidine ring and a piperidine ring, such as in the following aminoacids (proline and hydroxyproline): each of which may independently be part of an unsaturated structure (i.e. wherein the H atom geminal to the respective group R a , R b and R c is absent), e.g.:
  • peptide sequences refers to a sequence from N to C terminus, and attachment of group through a horizontal bond (here: moiety C) means covalent attachment to the peptide backbone via amide bond to the respective terminal amino acid (here: AA3):
  • peptide sequences refers to a sequence from N to C terminus, and attachment of group through a vertical bond (here: moiety C) means covalent attachment via the sidechain of the respective amino acid (here: AA3):
  • non-proteinogenic amino acids can be selected from the following list: Particularly preferred embodiments for the moiety B as well as the compound according to the present invention are shown in the appended claims.
  • Moiety C in the present invention represents a payload, which can be generally any atom (including H), molecule or particle.
  • moiety C is not a hydrogen atom.
  • the payload may be a chelator for radiolabeling.
  • the radionuclide is not released.
  • Chelators are well known to those skilled in the art, and for example, include chelators such as sulfur colloid, diethylenetriaminepentaacetic acid (DTPA), ethylenediaminetetraacetic acid (EDTA), 1 ,4,7,10- tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA), 1 ,4,7, 10-tetraazacyclododececane,N-(glutaric acid)-N',N",N"'-triacetic acid (DOTAGA), 1 ,4,7-triazacyclononane-N,N',N"-triacetic acid (NOTA), 1 ,4,8,1 1 - tetraazacyclotetradecane-N,N',N",N"'-tetraacetic acid (TETA), or any of the preferred
  • the payload may be a radioactive group comprising or consisting of radioisotope including isotopes such as 223 Ra, 89 Sr, 94m Tc, 99m Tc, 186 Re, 188 Re, 208 Pb, 212 Pb, 67 Ga, 68 Ga, 47 Sc, 111 1 n , 97 Ru, 62 Cu, 64 Cu, 65 Cu, 67 Cu, 54 Cu, 86 Y j i2i S n A
  • positron emitters such as 18 F and 124 l, or gamma emitters, such as 99m Tc, 111 ln and 123 l
  • beta-emitters such as 89 Sr, 131 l , and 177 Lu
  • Alpha-emitters such as 21 1 At, 225 Ac and 223 Ra may also be used for therapy.
  • the radioisotope is 89 Sr or 223 Ra.
  • the radioisotope is 68 Ga.
  • Cold nuclides may be useful to support GMP manufacturing as reference standards (e.g., 69 Ga, 175 Lu), or as precursors (e.g., 139 La) for other emitter nuclides.
  • the payload may be a chelate of a radioactive isotope, preferably of an isotope listed above, with a chelating agent, preferably a chelating agent listed herein.
  • the payload may be a fluorophore group, preferably selected from a xanthene dye, acridine dye, oxazine dye, cyanine dye, styryl dye, coumarine dye, porphine dye, fluorescent metal-ligand-complex, fluorescent protein, nanocrystals, perylene dye, boron-dipyrromethene dye and phtalocyanine dye, more preferably selected from the structures listed herein.
  • a fluorophore group preferably selected from a xanthene dye, acridine dye, oxazine dye, cyanine dye, styryl dye, coumarine dye, porphine dye, fluorescent metal-ligand-complex, fluorescent protein, nanocrystals, perylene dye, boron-dipyrromethene dye and phtalocyanine dye, more preferably selected from the structures listed herein.
  • the payload may be a cytotoxic and/or cytostatic agent.
  • cytotoxic agents can inhibit or prevent the function of cells and/or cause destruction of cells.
  • cytotoxic agents include radioactive isotopes, chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including synthetic analogues and derivatives thereof.
  • the cytotoxic agent may be selected from the group consisting of an auristatin, a DNA minor groove binding agent, a DNA minor groove alkylating agent, an enediyne, a lexitropsin, a duocarmycin, a taxane, a puromycin, a dolastatin, a maytansinoid and a vinca alkaloid or a combination of two or more thereof.
  • Preferred cytotoxic and/or cytostatic payload moieties are listed herein.
  • the payload is a chemotherapeutic agent selected from the group consisting of a topoisomerase inhibitor, an alkylating agent (e.g., nitrogen mustards; ethylenimes; alkylsulfonates; triazenes; piperazines; and nitrosureas), an antimetabolite (e.g., mercaptopurine, thioguanine, 5-fluorouracil), an antibiotic (e.g., anthracyclines, dactinomycin, bleomycin, adriamycin, mithramycin.
  • a chemotherapeutic agent selected from the group consisting of a topoisomerase inhibitor, an alkylating agent (e.g., nitrogen mustards; ethylenimes; alkylsulfonates; triazenes; piperazines; and nitrosureas), an antimetabolite (e.g., mercaptopurine, thioguanine, 5-fluorour
  • dactinomycin a mitotic disrupter (e.g., plant alkaloids - such as vincristine and/or microtubule antagonists - such as paclitaxel), a DNA methylating agent, a DNA intercalating agent (e.g., carboplatin and/or cisplatin, daunomycin and/or doxorubicin and/or bleomycin and/or thalidomide), a DNA synthesis inhibitor, a DNA-RNA transcription regulator, an enzyme inhibitor, a gene regulator, a hormone response modifier, a hypoxia-selective cytotoxin (e.g., tirapazamine), an epidermal growth factor inhibitor, an anti-vascular agent (e.g., xanthenone 5,6- dimethylxanthenone-4-acetic acid), a radiation-activated prodrug (e.g., nitroarylmethyl quaternary (NMQ) salts) or a bioreductive drug or a combination of two or more thereof.
  • the chemotherapeutic agent may selected from the group consisting of Erlotinib (TARCEVA®), Bortezomib (VELCADE®), Fulvestrant (FASLODEX®), Sutent (SU11248), Letrozole (FEMARA®), Imatinib mesylate (GLEEVEC®), PTK787/ZK 222584, Oxaliplatin (Eloxatin®.), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®.), Lapatinib (GSK572016), Lonafarnib (SCH 66336), Sorafenib (BAY43-9006), and Gefitinib (IRESSA®.), AG1478, AG1571 (SU 5271 ; Sugen) or a combination of two or more thereof.
  • TARCEVA® Erlotinib
  • VELCADE® Bortezomib
  • FASLODEX® Fulvestrant
  • the chemotherapeutic agent may be an alkylating agent - such as thiotepa, CYTOXAN® and/or cyclosphosphamide; an alkyl sulfonate - such as busulfan, improsulfan and/or piposulfan; an aziridine - such as benzodopa, carboquone, meturedopa and/or uredopa; ethylenimines and/or methylamelamines - such as altretamine, triethylenemelamine, triethylenepbosphoramide, triethylenethiophosphoramide and/or trimethylomelamine; acetogenin - such as bullatacin and/or bullatacinone; camptothecin; bryostatin; callystatin; cryptophycins; dolastatin; duocarmycin; eleutherobin; pancratistatin; sarcodictyin; spongistatin; nitrogen mustards
  • doxorubicin - such as morpholinodoxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and/or deoxydoxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins - such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites - such as methotrexate and 5-fluorouracil (5- FU); folic acid analogues - such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogues - such as fludarabine, 6-mercaptopurine, thiamiprine,
  • paclitaxel paclitaxel, abraxane, and/or TAXOTERE®, doxetaxel; chloranbucil; GEMZAR®.
  • gemcitabine 6-thioguanine; mercaptopurine; methotrexate; platinum analogues - such as cisplatin and carboplatin; vinblastine; platinum; etoposide; ifosfamide; mitoxantrone; vincristine; NAVELBINE®, vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids - such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids, derivatives or combinations of two or more of any of the above.
  • platinum analogues - such as
  • the payload may be a tubulin disruptor including but are not limited to: taxanes - such as paclitaxel and docetaxel, vinca alkaloids, discodermolide, epothilones A and B, desoxyepothilone, cryptophycins, curacin A, combretastatin A-4-phosphate, BMS 247550, BMS 184476, BMS 188791 ; LEP, RPR 109881 A, EPO 906, TXD 258, ZD 6126, vinflunine, LU 103793, dolastatin 10, E7010, T138067 and T900607, colchicine, phenstatin, chaicones, indanocine, T138067, oncocidin, vincristine, vinblastine, vinorelbine, vinflunine, halichondrin B, isohomohalichondrin B, ER-86526, pironetin, spongistatin 1 , spiket P,
  • the payload may be a DNA intercalator including but are not limited to: acridines, actinomycins, anthracyclines, benzothiopyranoindazoles, pixantrone, crisnatol, brostallicin, CI-958, doxorubicin (adriamycin), actinomycin D, daunorubicin (daunomycin), bleomycin, idarubicin, mitoxantrone, cyclophosphamide, melphalan, mitomycin C, bizelesin, etoposide, mitoxantrone, SN-38, carboplatin, cisplatin, actinomycin D, amsacrine, DACA, pyrazoloacridine, irinotecan and topotecan and pharmaceutically acceptable salts, acids, derivatives or combinations of two or more of any of the above.
  • a DNA intercalator including but are not limited to: a
  • the payload may be an anti-hormonal agent that acts to regulate or inhibit hormone action on tumors - such as anti-estrogens and selective estrogen receptor modulators, including, but not limited to, tamoxifen, raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and/or fareston toremifene and pharmaceutically acceptable salts, acids, derivatives or combinations of two or more of any of the above.
  • an anti-hormonal agent that acts to regulate or inhibit hormone action on tumors -
  • selective estrogen receptor modulators including, but not limited to, tamoxifen, raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and/or fareston toremifene and pharmaceutically acceptable salts, acids, derivatives or combinations of two or more of
  • the payload may be an aromatase inhibitor that inhibits the enzyme aromatase, which regulates estrogen production in the adrenal glands - such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate, AROMASIN®. exemestane, formestanie, fadrozole, RIVISOR®. vorozole, FEMARA®. letrozole, and ARIMIDEX® and/or anastrozole and pharmaceutically acceptable salts, acids, derivatives or combinations of two or more of any of the above.
  • an aromatase inhibitor that inhibits the enzyme aromatase, which regulates estrogen production in the adrenal glands - such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate, AROMASIN®. exemestane, formestanie, fadrozole, RIVISOR®. vorozole, FEMARA®. letrozole, and ARIM
  • the payload may be an anti-androgen such as flutamide, nilutamide, bicalutamide, leuprolide, goserelin and/or troxacitabine and pharmaceutically acceptable salts, acids, derivatives or combinations of two or more of any of the above.
  • the payload may be or comprise a protein, an antibody or an antibody fragment. Conjugates of the present invention comprising such payloads may be referred to as "bispecific conjugates”.
  • the payload is a cytokine (e.g., an interleukin such as IL2, IL10, IL12, IL15; a member ofthe TNF superfamily; a chemokine; or an interferon such as interferon gamma.).
  • a cytokine e.g., an interleukin such as IL2, IL10, IL12, IL15; a member ofthe TNF superfamily; a chemokine; or an interferon such as interferon gamma.
  • the payload is or comprises an antibody or an antibody fragment.
  • the antibody or antibody fragment is preferably characterized by an activity or function selected from: (i) immune cell engager, such as engager of one or more of T-cells (e.g., anti-CD3, anti-CD28, and/or anti-41 BB), B-cells (e.g., anti-CD40), NK-cells (e.g., anti-NKG2D, anti-CD16), and macrophages; (ii) binding to one or more immune checkpoint inhibitor(s), such as PD1 , CTLA-4, and/or PD-L1 ; (iii) binding to one or more cytokine(s); and (iv) binding to one or more chemokine(s).
  • immune cell engager such as engager of one or more of T-cells (e.g., anti-CD3, anti-CD28, and/or anti-41 BB), B-cells (e.g., anti-CD40), NK-
  • Any payload may be used in unmodified or modified form. Combinations of payloads in which some are unmodified and some are modified may be used.
  • the payload may be chemically modified.
  • One form of chemical modification is the derivatisation of a carbonyl group - such as an aldehyde.
  • the payload moiety C is a topoisomerase inhibitor; preferably camptothecin (CPT) or a derivative thereof; more preferably derived (e.g., by replacing a hydrogen atom) from topotecan, irinotecan, silatecan, cositecan, exatecan, lurtotecan, gimatecan, belotecan, rubitecan; even more preferably exatecan;
  • CPT camptothecin
  • moiety C is an auristatin (i.e., having a structure derived from an auristatin compound family member) or an auristatin derivative. More preferably, moiety C has a structure according to the following formula: wherein: is independently H or C1-6 alkyl; preferably H or CH3; is independently C1-6 alkyl; preferably CH3 or iPr;
  • Rd ⁇ is independently H or C1-6 alkyl; preferably H or CH3;
  • Rd ⁇ is independently H, C1-6 alkyl, COO(C1-6 alkyl), CON(H or C1-6 alkyl), C3-C 10 aryl or C3-C 10 heteroaryl; preferably H, CH3, COOH, COOCH3 or thiazolyl;
  • RdS is independently H, OH, C1-6 alkyl; preferably H or OH;
  • R ⁇ is independently C3-C-1 O aryl or C3-C-1 O heteroaryl; preferably optionally substituted phenyl or pyridyl.
  • moiety C is derived from MMAE or MMAF.
  • moiety C has a structure according to the following formula: wherein: e is 0, 1 , 2, 3, 4 or 5; preferably 1 ;
  • R ⁇ e ’ is independently H, COOH, CONH2, aryl-COOH or heteroaryl-COOH; preferably COOH;
  • R ⁇ 6 ’ is independently H, COOH, CONH2, aryl-COOH or heteroaryl-COOH; preferably COOH; and X is O, NH or S; preferably O.
  • This type of chelator which includes, e.g., DOTAGA, preferably has the configuration:
  • moiety C has a structure according to the following formulae: wherein: f and g are each independently 0, 1 , 2, 3, 4 or 5; preferably 1 ;
  • R ⁇ f is independently H, COOH, CONH2, aryl-COOH or heteroaryl-COOH; preferably COOH;
  • R ⁇ f is independently H, COOH, CONH2, aryl-COOH or heteroaryl-COOH; preferably COOH;
  • R ⁇ f is independently H, COOH, CONH2, aryl-COOH or heteroaryl-COOH; preferably COOH; and X is O, NH or S; preferably O.
  • moiety C has a structure according to any of the following formulae: wherein:
  • Ri h’ and R ⁇ h’ are each independently selected from COOH, CONH2, aryl-COOH, heteroaryl-
  • heteroaryl-CH2COOH wherein the heteroaryl is preferably pyridinyl; and each X is independently O, NH or S;
  • moiety C is a chelator having a structure comprising DOTAM (2-[4,7,10- tris(2-amino-2-oxoethyl)-1 ,4,7,10-tetrazacyclododec-1 -yl]acetamide) or a derivative thereof, e.g.:
  • moiety C comprises two or more therapeutically or diagnostically useful moieties, preferably with different mode of action.
  • Moiety C may be a radiohybrid ligand moiety which can be labeled, e.g., with 18 F via isotopic exchange and/or with (radio)metals (such as 68 Ga, 177 Lu, 225 Ac).
  • exemplary ligands of this class include, e.g. the following structures and (radio)metal chelates thereof:
  • a chelator (or a chelate) can significantly improve the hydrophilicity of an otherwise F-only-based tracer. Additional additional advantages of this radiohybrid concept include that, e.g., both the F-based moiety and the chelator can be labeled in an independent manner using the unprotected precursor, resulting in either a combination of 18F and metal or 19F an radiometal, the latter to be used for imaging (e.g., 68 Ga for PET, 111 ln for SPECT), or for radioligand therapy (e.g., 177 Lu). Corresponding radiopharmaceuticals, for example, 18 F/natGa and 19 F/ 68 Ga, are chemically identical molecules.
  • the resulting twins may be useful to bridge 18 F PET and radioligand therapy.
  • tracers are contemplated as advantageous tools for pretherapeutic patient stratification, pretherapeutic dosimetry, and radioligand therapy with a single tracer by exploiting 18 F and the most suitable therapeutic radioisotope (if also available as nonradioactive isotope).
  • ligands of this class may identify true-positive prostate cancer lesions in patients with negative conventional imaging, may help to better define sites of disease recurrence, and/or may inform salvage therapy decisions than does conventional imaging, potentially leading to improved outcomes.
  • C may also be a dual mode-of-action moiety, e.g., comprising a cytotoxic and a chelating or radioactive moiety as described elsewhere herein, attached to a common scaffold or linker moiety.
  • An exemplary ligand of this class includes, e.g., the following structure and (radio)metal chelates thereof:
  • C may also comprise, in addition to the therapeutic and/or diagnostic agent as described elsewhere herein, also a further targeting moiety, e.g., a PSMA binding moiety, attached to a common scaffold or linker moiety.
  • a further targeting moiety e.g., a PSMA binding moiety
  • An exemplary ligand of this class includes, e.g., the following structure and (radio)metal chelates thereof:
  • R' ⁇ j— z compounds comprising a structure , e.g., wherein R 1a and R 1b are joined together to form a ring:
  • the compounds described herein may be used to treat disease.
  • the treatment may be therapeutic and/or prophylactic treatment, with the aim being to prevent, reduce or stop an undesired physiological change or disorder.
  • the treatment may prolong survival as compared to expected survival if not receiving treatment.
  • the disease that is treated by the compound may be any disease that might benefit from treatment. This includes chronic and acute disorders or diseases including those pathological conditions which predispose to the disorder.
  • cancer and "cancerous” is used in its broadest sense as meaning the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a tumor comprises one or more cancerous cells.
  • the therapeutically effect that is observed may be a reduction in the number of cancer cells; a reduction in tumor size; inhibition or retardation of cancer cell infiltration into peripheral organs; inhibition of tumor growth; and/or relief of one or more of the symptoms associated with the cancer.
  • efficacy may be assessed by physical measurements of the tumor during the treatment, and/or by determining partial and complete remission of the cancer.
  • efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
  • methods for treatment e.g., by therapy or prophylaxis, of a subject suffering from or having risk for a disease or disorder; or by guided surgery practised on a subject suffering from or having risk for a disease or disorder; method for diagnosis of a disease or disorder, e.g., diagnostic method practised on the human or animal body and/or involving a nuclear medicine imaging technique, such as Scintigraphy, Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT); method for targeted delivery of a therapeutic or diagnostic agent to a subject suffering from or having risk for a disease or disorder.
  • a nuclear medicine imaging technique such as Scintigraphy, Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT)
  • the results produced from said nuclear medicine techniques may be analyzed.
  • a baseline PET or SPECT scan is obtained for each individual, optionally at randomization, and utilized to compare subsequent scans for the purpose of monitoring cancer metastasis or progression.
  • the analysis of subsequent scans may show lesions that were not present in the baseline scan such as positive distant lesions, loco-regional lesions which are indicative of progression of the cancer. Said patient subject to such diagnostic scans may then become eligible for a therapeutic treatment.
  • Prostate cancer The course of Prostate cancer from diagnosis to death is best categorized as a series of clinical stages based on the extent of disease, hormonal status, and absence or presence of detectable metastases: localized disease, rising levels of prostate-specific antigen (PSA) after radiation therapy or surgery with no detectable metastases, and clinical metastases in the non-castrate or castrate stage.
  • PSA prostate-specific antigen
  • BCR biochemical recurrence
  • the compound comprises a radioactive group comprising a radioisotope; preferably wherein moiety C is a chelate of a radioactive isotope with a chelating agent; more preferably a beta-emitter; most preferably 177 Lu.
  • moiety C may be a DOTAGA chelate of a beta-emitter, such as 177 Lu.
  • radioactive compounds (“radioconjugates”) may be suitably administered to a subject in need thereof at a dose of £ 250 MBq/kg, s 500 MBq/kg, or s 1000 MBq/kg, each expressed as a mouse dose, or an equivalent human dose.
  • a mouse dose may be recalculated to a corresponding equivalent human dose based on the body surface ratio as follows:
  • R 1 -Y-Z is represented by a structure as defined in claim 13 or 14; preferably by structure A-7; more preferably when the compound is 20a or 28a, the compound may be administered to a subject at a dose of 2 250 MBq/kg; preferably S 500 MBq/kg, each expressed as a mouse dose, or an equivalent human dose, e.g., S 20.3 MBq/kg, preferably S 40.6 MBq/kg, to provide advantageous anticancer activity.
  • R 1 -Y-Z is represented by a structure as defined in any one of claims 15, 16 and 17; preferably by structure A-23; more preferably when the compound is 22a, the compound may be administered to a subject at a dose of S 500 MBq/kg; preferably 2 1000 MBq/kg, each expressed as a mouse dose, or an equivalent human dose, e.g., S 40.6 MBq/kg, preferably S 81 .3 MBq/kg, to provide advantageous anticancer activity.
  • Pharmaceutical compositions are examples of S 500 MBq/kg; preferably 2 1000 MBq/kg, each expressed as a mouse dose, or an equivalent human dose, e.g., S 40.6 MBq/kg, preferably S 81 .3 MBq/kg, to provide advantageous anticancer activity.
  • the compounds described herein may be in the form of pharmaceutical compositions which may be for human or animal usage in human and veterinary medicine (e.g., as therapeutic or diagnostic compositions) and will typically comprise any one or more of a pharmaceutically acceptable diluent, carrier, or excipient.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
  • the choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the pharmaceutical compositions may comprise as - or in addition to - the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • Preservatives may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p- hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used.
  • the pharmaceutical composition may be formulated to be administered using a minipump or by a mucosal route, for example, as a nasal spray or aerosol for inhalation or ingestable solution, or parenterally in which the composition is formulated by an injectable form, for delivery, by, for example, an intravenous, intramuscular or subcutaneous route.
  • the formulation may be designed to be administered by a number of routes.
  • the agent If the agent is to be administered mucosally through the gastrointestinal mucosa, it should be able to remain stable during transit though the gastrointestinal tract; for example, it should be resistant to proteolytic degradation, stable at acid pH and resistant to the detergent effects of bile.
  • the pharmaceutical compositions may be administered by inhalation, in the form of a suppository or pessary, topically in the form of a lotion, solution, cream, ointment or dusting powder, by use of a skin patch, orally in the form of tablets containing excipients such as starch or lactose, or in capsules or ovules either alone or in admixture with excipients, or in the form of elixirs, solutions or suspensions containing flavouring or colouring agents, or the pharmaceutical compositions can be injected parenterally, for example, intravenously, intramuscularly or subcutaneously.
  • compositions may be best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or monosaccharides to make the solution isotonic with blood.
  • compositions may be administered in the form of tablets or lozenges which can be formulated in a conventional manner.
  • the compound of the present invention may be administered in the form of a pharmaceutically acceptable or active salt.
  • Pharmaceutically-acceptable salts are well known to those skilled in the art, and for example, include those mentioned by Berge et al, in J.Pharm.Sci., 66, 1 -19 (1977).
  • Salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1 ,1 '-methylene-bis-(2-hydroxy-3-naphthoate)) salts.
  • pamoate i.e., 1 ,1 '-methylene-bis
  • the routes for administration may include, but are not limited to, one or more of oral (e.g. , as a tablet, capsule, or as an ingestable solution), topical, mucosal (e.g. , as a nasal spray or aerosol for inhalation), nasal, parenteral (e.g., by an injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, ophthalmic (including intravitreal or intracameral), transdermal, rectal, buccal, vaginal, epidural, sublingual.
  • oral e.g. , as a tablet, capsule, or as an ingestable solution
  • mucosal e.g. , as a nasal spray or aerosol for inhalation
  • nasal parenteral (e.g., by an injectable form)
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy.
  • the formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for administration.
  • sterile liquid carrier for example water
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Exemplary unit dosage formulations contain a daily dose or unit daily sub-dose, or an appropriate fraction thereof, of the active ingredient.
  • combination modalities are contemplated, involving using a compound according to the present invention, e.g., a radioconjugates or a compound comprising a chelator moiety C, in combination with one or more other radioligand therapeutics RLTs and/or diagnostics, e.g., PSMA-targeted RLT, such as gozetotide (PSMA-11) or vipivotide tetraxetan (PSMA-617).
  • a compound according to the present invention e.g., a radioconjugates or a compound comprising a chelator moiety C
  • RLTs and/or diagnostics e.g., PSMA-targeted RLT, such as gozetotide (PSMA-11) or vipivotide tetraxetan (PSMA-617).
  • Advantages associated with such approaches may include, e.g., efficient production of radiochelated RLTs in one pots by mixing two precursors in the same vial, and/or improved antitumor activity and/or imaging quality due to the different modes of action (targeting different tumor proteins at the same time).
  • a compound according to the present invention e.g., a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31), in combination with one or more other radioligand therapeutics RLTs and/or diagnostics, e.g., PSMA-targeted RLT, such as gozetotide (PSMA-11) or vipivotide tetraxetan (PSMA-617).
  • RLTs and/or diagnostics e.g., PSMA-targeted RLT, such as gozetotide (PSMA-11) or vipivotide tetraxetan (PSMA-617).
  • Advantages associated with such approaches may include, e.g., improved antitumor activity due to the different modes of action (targeting different tumor proteins at the same time and combining radioactivity with cytotoxicity).
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof), or a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31), in combination with external beam radiation (radiotherapy).
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof), or a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31
  • Advantages associated with such approaches may include, e.g., improved antitumor activity due to the different modes of action.
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof), or a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31), in combination with androgen deprivation therapy (e.g., enzalutamide, abiraterone acetate, flutamide, nilutamide, bicalutamide, leuprolide, goserelin and/or troxacitabine).
  • androgen deprivation therapy e.g., enzalutamide, abiraterone acetate, flutamide, nilutamide, bicalutamide, leuprolide, goserelin and/or troxacitabine.
  • An aim of such type of therapy is to prevent growth of prostate cancer tumors that are castration-resistant (or prevent potential metastasis). Without wishing to be bound by any theory, this is contemplated to work by stopping the growth of androgen-dependent cancer cell by blocking binding of hormones, such as testosterone. However, not all cancer cells are androgen-dependent.
  • a combination partner such as the compounds of the present invention, it is contemplated that evolution of castration-resistant prostate cancer can be significantly further inhibited, preventing aggressive metastasis which results in high death rates of patients.
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof), or a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31), in combination with one or more immunocytokines.
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof)
  • a cytotoxic conjugate preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31)
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof), or a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31), in combination with one or more of: antibodies, bispecific antibodies, trispecific antibodies, and further bi- or trispecific conjugates of the present invention wherein the payload (C) is or comprises (i) an immune cell engager antibody or antibody fragment, such as engager of one or more of T-cells (e.g., anti- CD3, anti-CD28, and/or anti-41 BB), B-cells (e.g., anti-CD40), NK-cells (e.g., anti-NKG2D, anti-CD16), and macrophages, preferably Blinatumomab, Mosunetuzumab, Glofitamab
  • a radioconjugate preferably a ProX1-(SS)-DOTA-
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof), or a cytotoxic conjugate, preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31), in combination with conventional chemotherapy, e.g., as described with respect to chemotherapeutic, cytotoxic and/or cytostatic agents hereinabove.
  • a radioconjugate preferably a ProX1-(SS)-DOTA-based radioconjugate (such as compound 27a and radiochelates thereof)
  • a cytotoxic conjugate preferably a ProX1-(SS) MMAE-based conjugate (such as compound 31)
  • conventional chemotherapy e.g., as described with respect to chemotherapeutic, cytotoxic and/or cytostatic agents hereinabove.
  • the compounds described herein may be prepared by chemical synthesis techniques. It will be apparent to those skilled in the art that sensitive functional groups may need to be protected and deprotected during synthesis of a compound. This may be achieved by conventional techniques, for example as described in "Protective Groups in Organic Synthesis” by T W Greene and P G M Wuts, John Wiley and Sons Inc. (1991), and by P.J.Kocienski, in “Protecting Groups", Georg Thieme Verlag (1994). It is possible during some of the reactions that any stereocentres present could, under certain conditions, be epimerised, for example if a base is used in a reaction with a substrate having an optical centre comprising a base-sensitive group. It should be possible to circumvent potential problems such as this by choice of reaction sequence, conditions, reagents, protection/deprotection regimes, etc. as is well-known in the art.
  • Antibody is used in its broadest sense and covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies), veneered antibodies, antibody fragments and small immune proteins (SIPs) (see Int. J. Cancer (2002) 102, 75-85).
  • An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen.
  • a target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody.
  • An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length immunoglobulin molecule, i.e. a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof.
  • the antibodies may be of any type - such as IgG, IgE, IgM, IgD, and IgA) - any class - such as IgG 1 , lgG2, lgG3, lgG4, lgA1 and lgA2 - or subclass thereof.
  • the antibody may be or may be derived from murine, human, rabbit or from other species.
  • Analog This term encompasses any enantiomers, racemates and stereoisomers, as well as all pharmaceutically acceptable salts and hydrates of such compounds.
  • Alkyl refers to a branched or unbranched saturated hydrocarbyl radical.
  • the alkyl group comprises from 1 to 100, preferably 3 to 30, carbon atoms, more preferably from 5 to 25 carbon atoms.
  • alkyl refers to methyl, ethyl, propyl, butyl, pentyl, or hexyl.
  • Alkenyl refers to a branched or unbranched hydrocarbyl radical containing one or more carbon-carbon double bonds.
  • the alkenyl group comprises from 2 to 30 carbon atoms, preferably from 5 to about 25 carbon atoms.
  • Alkynyl refers to a branched or unbranched hydrocarbyl radical containing one or more carbon-carbon triple bonds.
  • the alkynyl group comprises from about 3 to about 30 carbon atoms, for example from about 5 to about 25 carbon atoms.
  • Halogen refers to fluorine, chlorine, bromine or iodine, preferably fluorine or chlorine.
  • Cycloalkyl refers to an alicyclic moiety, suitably having 3, 4, 5, 6, 7 or 8 carbon atoms.
  • the group may be a bridged or polycyclic ring system. More often cycloalkyl groups are monocyclic. This term includes reference to groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, bicyclo[2.2.2]octyl and the like.
  • Aryl refers to an aromatic carbocyclic ring system, suitably comprising 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15 or 16 ring carbon atoms.
  • Aryl may be a polycyclic ring system, having two or more rings, at least one of which is aromatic. This term includes reference to groups such as phenyl, naphthyl fluorenyl, azulenyl, indenyl, anthryl and the like.
  • a derivative includes the chemical modification of a compound. Examples of such modifications include the replacement of a hydrogen by a halo group, an alkyl group, an acyl group or an amino group and the like. The modification may increase or decrease one or more hydrogen bonding interactions, charge interactions, hydrophobic interactions, van der Waals interactions and/or dipole interactions.
  • Diastereomers or diastereoisomers unless specified otherwise, preferably refer to stereoisomers of a compound having different configurations at one or more stereocenters in parts of the molecule other than moiety R 1 -Y or R 1 . That is, unless specified otherwise, the stereochemical configuration of moiety R 1 -Y or R 1 is as represented in the respective structure, and the individual diastereomers may differ in their stereochemical configuration in the in parts of the molecule other than moiety R 1 -Y or R 1 .
  • Hetero signifies that one or more of the carbon atoms of the group may be substituted by nitrogen, oxygen, phosphorus, silicon or sulfur.
  • Heteroalkyl groups include for example, alkyloxy groups and alkythio groups.
  • Heterocycloalkyl or heteroaryl groups herein may have from 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15 or 16 ring atoms, at least one of which is selected from nitrogen, oxygen, phosphorus, silicon and sulfur.
  • a 3- to 10-membered ring or ring system and more particularly a 5- or 6-membered ring which may be saturated or unsaturated.
  • oxiranyl selected from oxiranyl, azirinyl, 1 ,2-oxathiolanyl, imidazolyl, thienyl, furyl, tetra hydrofury I, pyranyl, thiopyranyl, thianthrenyl, isobenzofuranyl, benzofuranyl, chromenyl, 2H-pyrrolyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, imidazolidinyl, benzimidazolyl, pyrazolyl, pyrazinyl, pyrazolidinyl, thiazolyl, isothiazolyl, dithiazolyl, oxazolyl, isoxazolyl, pyridyl, pyrazinyl, pyrimidinyl, piperidyl, piperazinyl, pyridazinyl, morpholinyl, thiomorpholinyl,
  • “Substituted” signifies that one or more, especially up to 5, more especially 1 , 2 or 3, of the hydrogen atoms in said moiety are replaced independently of each other by the corresponding number of substituents.
  • the term “optionally substituted” as used herein includes substituted or unsubstituted. It will, of course, be understood that substituents are only at positions where they are chemically possible, the person skilled in the art being able to decide (either experimentally or theoretically) without inappropriate effort whether a particular substitution is possible. For example, amino or hydroxy groups with free hydrogen may be unstable if bound to carbon atoms with unsaturated (e.g. , olefinic) bonds.
  • the term “substituted” signifies one or more, especially up to 5, more especially 1 , 2 or 3, of the hydrogen atoms in said moiety are replaced independently of each other by the corresponding number of substituents selected from OH, SH, NH2, halogen, cyano, carboxy, alkyl, cycloalkyl, aryl and heteroaryl.
  • substituents described herein may themselves be substituted by any substituent, subject to the aforementioned restriction to appropriate substitutions as recognised by the skilled person.
  • any of the aforementioned substituents may be further substituted by any of the aforementioned substituents, each of which may be further substituted by any of the aforementioned substituents.
  • substituted means any of the above groups (e.g.., alkyl, alkylene, alkylcycloalkyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, aryloxy, alkylaminyl, alkylcarbonylaminyl, alkylaminy lalkyl, aminylcarbonyl, alkylaminylcarbonyl, aminylcarbonylalkyl, aminylcarbonycycloalkylalkyl, thioalkyl, aryl, aralkyl, carboxyalkyl, cyanoalkyl, cycloalkyl, cyanocycloalkyl, cycloalkylaminylcarbonyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl, A/-heterocyclyl, heterocyclylalkyl, heteroaryl, N- heteroaryl
  • “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • a higher-order bond e.g., a double- or triple-bond
  • nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • R g and Rh are the same or different and independently hydrogen, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, A/-heterocyclyl, heterocyclylalkyl, heteroaryl, A/-heteroaryl and/or heteroarylalkyl.
  • “Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an aminyl, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, A/-heterocyclyl, heterocyclylalkyl, heteroaryl, A/-heteroaryl and/or heteroarylalkyl group.
  • each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
  • substituents suitably include halogen atoms and halomethyl groups such as CF3 and CCH; oxygen containing groups such as oxo, hydroxy, carboxy, carboxyalkyl, alkoxy, alkoyl, alkoyloxy, aryloxy, aryloyl and aryloyloxy; nitrogen containing groups such as amino, alkylamino, dialkylamino, cyano, azide and nitro; sulfur containing groups such as thiol, alkylthiol, sulfonyl and sulfoxide; heterocyclic groups which may themselves be substituted; alkyl groups, which may themselves be substituted; and aryl groups, which may themselves be substituted, such as phenyl and substituted phenyl.
  • Alkyl includes substituted and unsubstituted benzyl.
  • Dissociation constant This term refers to the equilibrium of ligand binding to a specific binding site, that is, the concentration of ligand at which 50% of the ligand is bound to the specific binding site. It is expressed in concentration units and particularly in nM.
  • the dissociation constant may be determined by Surface Plasmon Resonance (SPR).
  • SPR Surface Plasmon Resonance
  • a SPR measurement useful for determining Kd may be performed using a CM5 chip, e.g., with a Biacore X100 instrument.
  • a suitable measurement protocol is as follows.
  • ACP3 is immobilized on both flow cells of a CM5 chip (e.g., Cytiva, #BR100012), reaching 3500 to 4500 RUs, using the EDC/NHS protocol provided by the manufacturer.
  • ACP3 immobilized on the reference flow cell is denatured (e.g., with a denaturing solution: 0.85% H3PO4, 10 mM NaOH, and 50 mM HCI).
  • PBS pH 7.4
  • Test compounds are injected at different concentrations (e.g., 1 pM or 500 nM in running buffer), e.g., according to a multicycle analysis with the following settings: 120 seconds contact time and 15,000 seconds dissociation time at a flow rate of 10 ⁇ L/min.
  • Sensograms may be plotted, e.g. with GraphPad Prism (version 8, GraphPad Software), and fitted, e.g., using the BIAcore Evaluation Software 3.2 RCI (GE Healthcare). Specific binding.
  • “specific binding to ACP3” refers to better ACP3 binding expressed by Kd as compared to binding to other proteins found in mammals, preferably humans, primates and/or rodents, e.g., albumin (e.g., HSA or MSA) and/or other phosphatase(s) (e.g., PLAP, ACPI, TCPTP, and/or TNAP).
  • HPLC Reversed-phase High-Pressure Liquid Chromatography
  • Reversed-phase Medium-Pressure Liquid Chromatography Small organic molecules that could be produced at higher quantities (i.e., >10 mg) were purified by reversed-phase MPLC (BUCHI) on a C18 40 pM irregular 12 g column using mQ millipore water 0.1% formic acid (FA) (eluent A) and acetonitrile 0.1% FA (eluent B) as mobile phase at following gradient: 0-5 min 98% A, 5-45 min 98% to 0% A, 45-50 min 0% A, 50-50.1 min 0% to 98% A and 50.1-55 min 98% A. The flow rate was set to 30 mL/min.
  • Analytical LC-MS Spectra were recorded on an Agilent 6100 Series Single Quadrupole MS system combined with an Agilent 1200 Series LC, using an InfinityLab Poroshell 120 EC-C18 Column, 2.7 pm, 4.6 x 50 mm, at a flow rate of 0.8 mL/min, acetonitrile:water with 0.1 % formic acid. The analysis was performed with the following gradient: 10% to 100% acetonitrile in 5 min.
  • Liquid-Chromatography/Mass-Spectrometry (LC/MS) for oligonucleotides Liquid-Chromatography/Mass-Spectrometry (LC/MS) spectra of oligonucleotides were performed on an Agilent 1260 Series LC coupled to an Agilent 6100 Series Single Quadrupole MS.
  • the system was equipped with an ACQUITY UPLC Oligonucleotide BEH C18 column (130 A, 1.7 pm, 2.1 x 50 mm), using the following gradient of eluent A (15 mM TEA, 400 mM HFIP in mQ H2O) and eluent B (methanol) at a flow rate of 0.4 mL/min and 60°C column temperature: 0-0.2 min 95% A, 0.2-8.2 min 95% to 5% A, 8.2-8.7 min 5% A, 8.7-9.2 min 5 to 95% A, 9.2-13 min 95% A.
  • eluent A 15 mM TEA, 400 mM HFIP in mQ H2O
  • eluent B methanol
  • Fmoc deprotection Resin was incubated two times (15 min) with 20% piperidine in DMF. After deprotection, the resin was washed 5-10 times with DMF to remove residual piperidine.
  • Mini cleavage test for LC-MS analysis (S3): A small portion of resin was transferred to an Eppendorf tube and incubated with 40 ⁇ L trifluoracetic acid (TFA) for 15 min at room temperature. The cleavage was quenched by addition of 80 ⁇ L DMF to centrifuge the suspension (1 min at 10’000 ref) before LC-MS analysis. This method was used to monitor the synthesis after each reaction step on resin.
  • TFA trifluoracetic acid
  • Amide coupling S4: The carboxylic acids, O-(7-azabenzotriazol-1-yl)-A/,A/,A/',A/'-tetramethyluronium hexafluorophosphate (HATU) and diisopropylethylamine (DIPEA) were dissolved in DMF (0.08 M) and added to the resin-bound free amino group. After incubation, the resin was subsequently washed five times with DMF. Coupling efficiency was monitored by LC-MS.
  • HATU O-(7-azabenzotriazol-1-yl)-A/,A/,A/',A/'-tetramethyluronium hexafluorophosphate
  • DIPEA diisopropylethylamine
  • Resin cleavage and purification S5: Cleavage solution was prepared as follows: 95% trifluoracetic acid (TFA), 2.5% water, and 2.5% triisopropylsilane (TIPS). Two consecutive cleavages (1 h at room temperature each) were performed. Cleavage fractions were combined and either directly purified via RP HPLC or precipitated in diethyl ether for subsequent purification (see below).
  • Peptide precipitation S6: Peptides were precipitated from the cleavage solution by addition of 5-10 volumes of ice-cold diethyl ether after most of the TFA was removed under reduced pressure. Precipitation proceeded for 30 min at -20 °C to obtain the peptide as pellet by centrifugation (3200 ref, 5 min, 4 °C). The crude was dissolved in a mixture of watenacetonitrile (1 :1) and purified by reversed-phase chromatography.
  • Phenol (1 equiv.), potassium carbonate (3 equiv.), and potassium iodide (1 equiv.) were loaded into a round-bottom flask coupled with a magnetic stirring bar and suspended in dry acetone (0.13 M). The solution was stirred at room temperature for 30min - 1 h and propargyl bromide (1 .2 equiv.) was added. The mixture was heated to 50 °C and left to stir overnight. The solvent was removed under reduced pressure and the residue was redissolved in ethyl acetate and water and transferred into a separatory funnel.
  • Phosphonate deprotection (GP4): Protected phosphonate (1 equiv.) was loaded into a round-bottom flask coupled with a magnetic stirring bar and was dissolved in dry acetonitrile (0.25 M) under argon. The solution was cooled in an ice bath and bromotrimethylsilane (5 equiv.) was added dropwise. The reaction was monitored by LC-MS. Upon complete conversion, the solvent was removed under reduced pressure and the residue was purified via reverse-phase semi-preparative HPLC.
  • Catalytic Click reaction (GP5): Alkyne (1 equiv.) and Azide (1 equiv.) were loaded into a reaction vessel and dissolved in DMSO. Copper sulfate pentahydrate (0.2 equiv.) and sodium ascorbate (2 equiv.) were loaded into a separate container and dissolved in water. The two solutions were mixed (final DMSO:water ratio - 4:1 , 0.06M) and the reaction was monitored via LC-MS. Upon completion, the reaction mixture was diluted with 3 volumes of DMSO, filtered through a syringe frit, and purified directly via reversed-phase semipreparative HPLC.
  • Nucleophilic substitution with DOTA-GA anhydride (GP7): Amine (1.0 equiv.), DOTA-GA anhydride (1.0 equiv.), and DMAP (0.1 equiv.) were weighed into an Eppendorf tube and dissolved in DMSO (0.01 M). DIPEA (3.0 equiv.) was added and the reaction was incubated in a Thermomixer at 24 °C overnight. The mixture was used as crude for click reaction or diluted with DMSO (3x volume) and was directly purified via reversed-phase semi-preparative HPLC.
  • DOTAGA derivatives (30 nmol) were dissolved in 30 ⁇ L of PBS 2% DMSO and diluted with sodium acetate (198 ⁇ L, 1 M in water, pH 4.5).
  • 24 MBq of 177 Lu solution (12 ⁇ L at an activity of 2 MBq/ ⁇ L) were added and the mixture was heated at 90 °C for 10 min and passively cooled down to rt for 10 min. After cooling to room temperature, an aliquot was analyzed by RP-HPLC (XTerra C18, 5% MeCN in 0.1 % aq. TFA to 80% over 20 min on a Merck-Hitachi D- 7000 HPLC system equipped with a Raytest Gabi Star radiodetector). 177 Lu incorporations >95% were routinely achieved.
  • reaction mixture was diluted with DMF (1 :3) and was directly purified via (Agilent 1200 series system equipped with Synergi 4pm Polar-RP 80A 10 x 150 mm C18 column using a gradient of 90:10 to 0:100 in 14 min water/ACN + 0.1 % TFA).
  • Amino PEG2 Azide (1 .05 mg, 0.006 mmol, 1 .00 equiv) was loaded into an Eppendorf tube and was dissolved in DMSO (100 ⁇ L, 0.06 M). Acetic anhydride (0.6 ⁇ L, 0.0066 mmol, 1.1 equiv.) was added and the reaction was incubated in a ThermoMixer at room temperature overnight. Compound 2 was added to the mixture and GP5 was followed for the click reaction. The product was a white solid after lyophilization (1.3 mg, 0.0024 mmol, 40% yield), m/z calculated for C25H33N5O7P [M - H] _ 546.21 ; observed 546.2.
  • DOTA-GA Anhydride was reacted with I4 according to GP7 (1 .6 ⁇ mol scale). The reaction was incubated in a ThermoMixer at room temperature overnight and the cleavage solution from S5 (200 ⁇ L) was added and the mixture was further incubated until completion by LC-MS. The solution was then further diluted with DMSO (3x volume) and directly purified via reversed-phase semi-preparative HPLC to obtain compound 5 as a white solid after lyophilization (1 .0 mg, 1 ⁇ mol, 65% yield).
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 10 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was directly used in the next step.
  • Compound 8 was prepared via GP6 (1.5 ⁇ mol scale) from azido-PEG4 fluoresceine thiourea and intermediate I6c. The product was an orange solid after lyophilization (1.0 mg, 0.95 ⁇ mol, 63% yield), m/z calculated for C51H55N7O14PS [M - H] _ 1052.33; observed 1052.3.
  • Compound 9 was prepared via GP1 (4 ⁇ mol scale) from compound 2 and a racemic mixture of the O-Me, A/-Boc protected 4-azido proline derivative. The product was a white solid after lyophilization (0.6 mg, 1 ⁇ mol, 25% yield), m/z calculated for C28H35N5O8P [M - H]- 600.22; observed 600.3.
  • I9a (3.9 mg, 0.01 mmol, 1 .0 equiv.) and HATU (3.8 mg, 0.01 mmol, 1 .0 equiv.) were loaded into an Eppendorf tube and dissolved in acetonitrile (0.125 mL, 0.08 M).
  • Tert-butyl (6-aminohexyl)carbamate (3.4 ⁇ L, 0.015 mmol, 1 .5 equiv.) and DIPEA (7.0 ⁇ L, 0.04 mmol, 4.0 equiv.) were added and the reaction was incubated in a ThermoMixer at 37 °C for 2h.
  • the solution was diluted with DMSO (3x volume), was filtered through a syringe frit, and directly purified via reverse d-phase semipreparative HPLC. Upon completion of the purification, the fractions were combined and concentrated under reduced pressure. TFA was added and the purified solution was incubated until complete deprotection of the Boc group was noted by LC-MS. The residue was then lyophilized.
  • I9a (3.9 mg, 0.01 mmol, 1.0 equiv.), HATU (3.8 mg, 0.01 mmol, 1.0 equiv.), and 6-aminohexyl fluoresceine thiourea (5.9 mg, 0.012 mmol, 1.2 equiv.) were loaded into an Eppendorf tube and dissolved in acetonitrile (0.125 mL, 0.08 M). DIPEA (7.0 ⁇ L, 0.04 mmol, 4.0 equiv.) was added and the reaction was incubated in a ThermoMixer at 37 °C for 2h.
  • DIPEA 7.0 ⁇ L, 0.04 mmol, 4.0 equiv.
  • I17a,b were synthesized according to GP9 (0.22 mmol scale) from the respective functionalized 3-lodo- phenylalanines 116a, b. and 114.
  • I22a,b were synthesized according to GP9 (0.22 mmol scale) from the respective functionalized 3-lodo- phenylalanines 121a, b. and 114.
  • 17a (0.7 mg, 0.7 ⁇ mol, 55% yield), m/z calculated for C60H56FN5O12P [M + H] + 1088.36; observed 1088.3.
  • 17b (0.7 mg, 0.7 ⁇ mol, 55% yield), m/z calculated for C60H56FN5O12P [M + H] + 1088.36; observedl 088.3.
  • Amino PEG2 Azide (1.7 mg, 0.01 mmol, 1.0 equiv.) was loaded into an Eppendorf tube and dissolved in acetonitrile (0.2 mL, 0.05 M).
  • Compounds 25a, b are also referred to as ProX1-(SS)-DOTAGA_R and ProX1 -(RR)-DOTAGA_R, respectively.
  • (SS)” or“(RR)” refer to the configuration of the stereogenic centers on the central pyrrolidine moiety;
  • _R refers to the configuration of the stereogenic center on the DOTAGA moiety, if present.
  • 111a, b (1.2 mg, 2.5 ⁇ mol, 1.0 equiv.), DOTA-NHS ester (1.9 mg, 2.5 ⁇ mol, 1.0 equiv.), and DMAP (0.03 mg, 0.25 ⁇ mol, 0.1 equiv.) were weighed into an Eppendorf tube and dissolved in DMSO (125 ⁇ L, 0.02 M). DIPEA (1 .3 ⁇ L, 7.5 ⁇ mol, 3.0 equiv.) was added and the reaction was incubated in a ThermoMixer at 24°C overnight.
  • the mixture was diluted with DMSO (3x volume) and was directly purified via reversed-phase semipreparative HPLC. Products were isolated as white solids after lyophilization.
  • the product 30a was obtained as a red solid after lyophilization (1.0 mg, 0.84 ⁇ mol, 84% yield), m/z calculated for C60H57FN7O13PS2 [M - H] _ 1196.32; observed 1196.2.
  • I29a,b were synthetized according to GP10 (0.23nmol scale) from the respective functionalized 3-iodo- phenylalanines I28a,b and I6b. The crudes were directly used in the next step.
  • I32a,b were synthetized according to GP10 (0.25 nmol scale) from the respective functionalized 3-iodo- phenylalanines 131a, b and I6b. The crude was directly used in the next step.
  • the reaction mixture is worked up by evaporating the solvents at T ⁇ 50 °C under vacuum.
  • the crude was redissolved in EtOAc (50 mL) and washed with water (3 x 50 mL).
  • the organic phase is evaporated at T s 50 °C under vacuum.
  • the product 15c is obtained as a pale-yellow powder.
  • I5c (1.00 eq., 0.018 mol, 2.88 g) is dissolved in ACN (25 mL). Chlorotrimethylsilane (2.00 eq., 0.036 mol, 4.57 mL) and benzylamine (1.00 eq., 0.018 mol, 1.97 mL) are added (T S 37 °C). The reaction mixture is then sonicated at T S 50 °C for 1 h ( ⁇ 10 min). Tris(trimethylsilyl)phosphite (1.50 eq., 0.027 mol, 9.50 mL) and chlorotrimethylsilane (2.00 eq., 0.036 mol, 4.57 mL) are added.
  • reaction mixture is then sonicated at T ⁇ 50 °C for ⁇ 2 h (+10 min), and the reaction conversion is checked (HPLC - MS, I5c ⁇ 5.0%-a/a).
  • the reaction mixture is worked up by evaporating the solvents at T s 50 °C under vacuum.
  • the crude product is purified via preparative RP-HPLC. Fractions containing I6c are combined and lyophilized overnight, and the product is obtained as white powder.
  • (2S,4S)-4-azido-1-(tert-butoxycarbonyl)pyrrolidine-2-carboxylic acid (1 .00 eq., 0.012 mol, 3.00 g) and HATU (1 .00 eq., 0.012 mol, 4.45 g) are dissolved in DMF (50 mL).
  • DIPEA (4.00 eq., 0.048 mol, 8.20 mL) is added at T ⁇ 37 °C.
  • the reaction mixture is stirred at T ⁇ 37 °C for ⁇ 10 minutes.
  • the organic phase is washed with water (3 x 50 mL), saturated solution of ammonium chloride (3 x 50 mL) and water (3 x 50 mL). The washed organic phase is evaporated atT ⁇ 50 °C under vacuum.
  • the crude product is dissolved in DCM (30 mL). TFA (20.00 eq., 0.24 mol, 18.36 mL) is added (T ⁇ 37 °C) to remove the Boc group from Boc-protected I69.
  • the reaction mixture is stirred at T ⁇ 37 °C, and the conversion is checked (HPLC-MS, Boc-protected I69 5 5.0%-a/a).
  • the reaction mixture is worked up by evaporating the solvents at T s 50 °C under vacuum.
  • the crude product is purified via preparative RP-HPLC. Fractions containing I69 are combined and lyophilized overnight. The product is obtained as a pale-yellow powder.
  • Dibenzylglycine (1.00 eq., 0.006 mol, 1.5 g) and HATU (1.00 eq, 0.006 mol, 2.28 g) are dissolved in DMF (12 mL).
  • DIPEA (4.00 eq., 0.024 mol, 4.20 mL) is added at T ⁇ 37 °C.
  • the reaction mixture is stirred at T ⁇ 37 °C for at least 20 minutes.
  • 169 (1.00 eq., 0.006 mol, 2.85 g) is added at T ⁇ 37 °C.
  • the reaction mixture is then stirred at T ⁇ 37 °C, and the conversion is checked by HPLC-MS (Dibenzylglycine ⁇ 5.0%-a/a).
  • reaction mixture is then stirred at T s 37 °C, and the conversion is checked by HPLC-MS (conversion of Dibenzylglycine- I6c intermediate: s 30.0%-a/a).
  • HPLC-MS conversion of Dibenzylglycine- I6c intermediate: s 30.0%-a/a.
  • the reaction mixture is filtered through celite, and the crude product is purified via preparative RP-HPLC. Fractions containing 170 are combined and lyophilized overnight. 170 is obtained as a white powder.
  • the reaction mixture is then stirred at T 37 °C, and the conversion to Prox1-(SS)-DOTA (27a) is checked by HPLC-MS (111a s 5.0%-a/a).
  • the crude product was diluted with ⁇ 1 mL DMF and purified via preparative RP-HPLC. Fractions containing 27a with purity > 98.0% (a/a) are combined and lyophilized overnight. The product 27a is obtained as a white powder.
  • Radiosynthesis of 177 Lu-labelled s38 ( 177 Lu-PSMA-617, i.e., 177 Lu vipivotide tetraxetan) was performed via GP11 from s38. Labelling was performed at the molar activity of 20 MBq/nmol. The HPLC chromatogram of 177 Lu-PSMA-617 as recorded with a radio-detector showed a peak at 12 min. The products were used in autoradiography experiments.
  • Compound I37 was prepared via GP12 (on a 3 ⁇ mol scale) from compound I35 and MMAF as the payload.
  • 6-maleimidohexanoic acid (1.3 mg, 6.0 ⁇ mol, 1.2 equiv.) and HATU (1 .9 mg, 5 ⁇ mol, I .O equiv.) were weighed into an Eppendorftube and dissolved in DMF (0.1 mL, 0.05 M). DIPEA (1 .7 ⁇ L, 10 ⁇ mol, 2.0 equiv.) was added, and the solution was incubated in a shaker incubator for 20 min at room temperature. MMAE (3.6 mg, 5.0 ⁇ mol, 1.00 equiv.) was loaded into a separate Eppendorf tube, with the necessary precaution, and the pre-activated carboxylic acid mixture was added to it.
  • the intermediate was Fmoc deprotected (S2) to afford s40, which was further reacted with 2-(dibenzylamino)acetic acid (191 mg, 0.750 mmol, 3.00 equiv.) as described in S4 (2.9 equiv. HATU, 6 equiv. DIPEA, 2 h).
  • the resin was cleaved according to a modified version of S5, using the following cleavage cocktail is: 50% TFA, 45% DCM, 2.5% TIPS, and 2.5% thioanisole.
  • the cleavage solution was concentrated under reduced pressure, and the crude mixture was precipitated according to S6.
  • SMDC 31 was prepared via GP13 (on a 3 ⁇ mol scale) using I40, 136 and I6c. After lyophilization, the product was obtained as a white solid (1 .00 mg, 0.46 ⁇ mol, 15% yield), m/z calculated for C111H154N17O23PS [M+2H] 2+ 1078.54; m/z observed 1078.6.
  • SMDC 45 was prepared via GP13 (on a 0.4 ⁇ mol scale) using I40, I37 and I6c.
  • SMDC 33 was prepared via GP13 (on a 2 ⁇ mol scale) using I40, Vedotin and I6c. After lyophilization, the product was obtained as a white solid (1 .20 mg, 0.53 ⁇ mol, 27% yield), m/z calculated for C115H164N19O24PS [M+2H] 2+ 1129.58; m/z observed 1129.6.
  • SMDC 46 was prepared via GP13 (on a 2.1 ⁇ mol scale) using I40, I38 and I6c.
  • SMDC 34 was prepared via GP13 (on a 3.0 ⁇ mol scale) using I40, I39 and I6c. After lyophilization, the product was obtained as a white solid (1 .00 mg, 0.54 ⁇ mol, 18% yield), m/z calculated for C96H137N14O19PS
  • dibenzylphosphite 48 ⁇ L, 0.2 mmol, 1 .2 equiv.
  • the reaction was stirred for 30 min at 0 °C.
  • the mixture was diluted with ethyl acetate and washed successively with sat. aq. NaHCO3 (x3), 1 M HCL (x3) and brine, dried over Na2SO4, and concentrated under reduced pressure.
  • Compound 145 was prepared via GP12 (15 ⁇ mol scale) from compound I44 and MMAE as the respective payload. After purification, the solvent was removed under reduced pressure and the residue was treated with TFA (2 mL). The acid was removed under reduced pressure, and the product was lyophilized to yield a white solid (2.8 mg, 2.4 ⁇ mol, 16% yield), m/z calculated for C58H89N7O16P [M+H] + 1170.61 ; observed 1170.6.
  • SMDC 36 was prepared via GP13 (on a 1.2 ⁇ mol scale) using I40, I45 and I6c.
  • Precipitation of the DNA from aqueous phase was achieved by addition of 10% v/v 5 M NaCI or 3 M acetic acid buffer (pH 5). Then, 3 volumes of EtOH were added and the mixture was vortexed and left at -20°C overnight. The DNA was obtained as a pellet Fiby centrifugation (16,100 ref, 4°C, 1 h), the supernatant was discarded, and the pellet dried in a SpeedVac vacuum concentrator.
  • the pre-activation mixture was added to a 5’-amino modified 12-mer oligonucleotide (5’ Ce-amino-TAGTAGCCATCC, 250 nmol in 200 ⁇ L MOPS buffer (100 mM MOPS, 1 M NaCI, pH 8).
  • MOPS buffer 100 mM MOPS, 1 M NaCI, pH 8
  • the reaction proceeded for 2 h at room temperature and was stopped by ethanol precipitation.
  • the DNA-pellet was redissolved in H2O (300 ⁇ L) to which piperidine (35 ⁇ L) was added.
  • the deprotection was complete after 1 h at 40 °C and quenched by the addition of 53 ⁇ L 3 M acetic acid buffer.
  • the oligonucleotide was precipitated by the addition of ethanol for subsequent by RP-HPLC.
  • the modified oligonucleotides (15 nmol each) were dissolved in 50 ⁇ L 250 mM borate buffer, pH 9.4 and CuSO4 (6 ⁇ L, 50 mM in H2O, 20 equiv.), 50 ⁇ L 20 mM intermediate I6c (67 equiv.) and sodium ascorbate (6 ⁇ L, 50 mM in H2O, 20 equiv.) were added.
  • the copper(l)-catalyzed alkyne-azide cycloaddition (CuAAC) proceeded for 30 min at 60 °C and was stopped by the addition of 20 ⁇ L 3 M acetic acid buffer pH 5 for subsequent ethanol precipitation.
  • the crudes were purified by RP-HPLC.
  • 2-(dibenzylamino)acetic acid (24 ⁇ L, 200 mM in DMSO, 160 equiv.) was pre-activated with 4-(4,6-dimethoxy- 1 ,3,5-triazin-2-yl)-4-methyl-morpholinium chloride (DMT-MM, 16 ⁇ L, 200 mM in H2O, 107 equiv.) in 60 ⁇ L DMSO for 30 min at room temperature.
  • the solution was added to the stereo-defined 4-amino-proline- modified oligonucleotides (30 nmol in 40 ⁇ L 250 mM borate buffer, pH 9.4) and the coupling proceeded for 3 h at room temperature.
  • the reaction was stopped by EtOH precipitation and the oligonucleotides were purified by RP-HPLC.
  • the modified oligonucleotides (15 nmol each) were dissolved in 50 ⁇ L 250 mM borate buffer, pH 9.4 and CuSO4 (6 ⁇ L, 50 mM in H2O, 20 equiv.), 50 ⁇ L 20 mM intermediate I6c (67 equiv.) and sodium ascorbate (6 ⁇ L, 50 mM in H2O, 20 equiv.) were added.
  • the copper(l)-catalyzed alkyne-azide cycloaddition (CuAAC) proceeded for 30 min at 60 °C and was stopped by the addition of 20 ⁇ L 3 M acetic acid buffer pH 5 for subsequent ethanol precipitation.
  • the crudes were purified by RP-HPLC. 1.2.2 Synthesis of O3a-b, O4a-b, O5a-b
  • the 2 stereo-defined isomers (S) and (R)-2-((((9/7-Fluoren-9-yl)methoxy)carbonyl)amino)-3-(3- iodophenyl)propanoic acid (200 mM in DMSO, 187.5 ⁇ L, 150 equiv.) were separately activated by the addition of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC, 200 mM in DMSO, 150 ⁇ L, 120 equiv.) and N-hydroxysulfosuccinimide (sNHS, 200mM in DMSO:water (2:1), 125 ⁇ L, 100 equiv.).
  • EDC 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • sNHS N-hydroxysulfosuccinimide
  • the stereo-defined 3-lodo-phenylalanine-modified oligonucleotides (30 nmol each) were dissolved in 40 ⁇ L 250 mM borate buffer, pH 9.4 and acetic anhydride (200 mM in DMSO, 6 ⁇ L, 40 equiv.) was added. The reaction proceeded for 1 h at r.t. The reaction was stopped by EtOH precipitation, and the oligonucleotides were purified by RP-HPLC.
  • the pre-catalyst solution was prepared by mixing 10mM palladium (II) acetate in DMA (100 ⁇ L), 100 mM TPPTS in water (100 ⁇ L), 20mM Copper (II) acetate in water (100 ⁇ L) and diluted up to 1 mL with mQ millipore water, resulting in a 1 mM solution of Pd(0)-TPPTS complex and 2 mM solution of Cu(ll).
  • Each modified oligonucleotide (10 nmol scale) was dissolved in 100 ⁇ L 200 mM potassium carbonate, the pre-catalyst solution (20 ⁇ L of, 20 nmol in Pd) and alkyne I6b (100 mM in DMSO, 20 ⁇ L, 200 equiv.) were subsequently added.
  • the copper was reduced by adding sodium L- ascorbate (10 mM in water, 50 ⁇ L, 50 equiv.) and the resulting solutions were heated at 65 °C for 1 h.
  • the reactions were quenched by adding 100 mM DTT:3M acetate buffer pH 4.7 (1 :1 , 30 ⁇ L).
  • the products O3a, b were precipitated by adding EtOH and purified by RP-HPLC.
  • the 2 stereo-defined isomers (S) and (R)-2-((((9/7-Fluoren-9-yl)methoxy)carbonyl)amino)-3-(3- iodophenyl)propanoic acid (200 mM in DMSO, 187.5 ⁇ L, 150 equiv.) were separately activated by the addition of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC, 200 mM in DMSO, 150 ⁇ L, 120 equiv.) and N-hydroxysulfosuccinimide (sNHS, 200mM in DMSO:water (2:1), 125 ⁇ L, 100 equiv.).
  • EDC 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • sNHS N-hydroxysulfosuccinimide
  • 3-Chloro-4-fluorobenzoic acid (24 ⁇ L, 200 mM in DMSO, 160 equiv.) was pre-activated with 4-(4,6- dimethoxy-1 ,3,5-triazin-2-yl)-4-methyl-morpholinium chloride (DMT-MM, 16 ⁇ L, 200 mM in H2O, 107 equiv.) in 60 ⁇ L DMSO for 30 min at r.t.
  • the solution was added to the stereo-defined 3-lodo-phenylalanine-modified oligonucleotides (30 nmol in 40 ⁇ L 250 mM borate buffer, pH 9.4) and the coupling proceeded for 1 h at room temperature.
  • the reaction was stopped by EtOH precipitation, and the oligonucleotides were purified by RP-HPLC.
  • the pre-catalyst solution was prepared by mixing 10mM palladium (II) acetate in DMA (100 ⁇ L), 100 mM TPPTS in water (100 ⁇ L), 20mM Copper (II) acetate in water (100 ⁇ L) and diluted up to 1 mL with mQ millipore water, resulting in a 1 mM solution of Pd(0)-TPPTS complex and 2 mM solution of Cu(ll).
  • Each modified oligonucleotide (10 nmol scale) was dissolved in 100 ⁇ L 200 mM potassium carbonate, the pre-catalyst solution (20 ⁇ L of, 20 nmol in Pd) and alkyne I6b (100 mM in DMSO, 20 ⁇ L, 200 equiv.) were subsequently added.
  • the copper was reduced by adding sodium L- ascorbate (10 mM in water, 50 ⁇ L, 50 equiv.) and the resulting solutions were heated at 65 °C for 1 h.
  • the reactions were quenched by adding 100 mM DTT:3M acetate buffer (1 :1 , 30 ⁇ L).
  • the products were precipitated by adding EtOH and purified by RP-HPLC.
  • the 2 stereo-defined isomers (S) and (R)-2-((((9H-Fluoren-9-yl)methoxy)carbonyl)amino)-3-(3- iodophenyl)propanoic acid (200 mM in DMSO, 187.5 ⁇ L, 150 equiv.) were separately activated by the addition of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC, 200 mM in DMSO, 150 ⁇ L, 120 equiv.) and N-hydroxysulfosuccinimide (sNHS, 200mM in DMSO:water (2:1), 125 ⁇ L, 100 equiv.).
  • EDC 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • sNHS N-hydroxysulfosuccinimide
  • 4-Fluorophenyl isocyanate (24 ⁇ L, 200 mM in DMSO, 160 equiv.) was pre-activated with 4-(4,6-dimethoxy- 1 ,3,5-triazin-2-yl)-4-methyl-morpholinium chloride (DMT-MM, 16 ⁇ L, 200 mM in H2O, 107 equiv.) in 60 ⁇ L DMSO for 30 min at r.t.
  • the solution was added to the stereo-defined 3-lodo-phenylalanine-modified oligonucleotides (30 nmol in 40 ⁇ L 250 mM borate buffer, pH 9.4) and the coupling proceeded for 1 h at room temperature.
  • the reaction was stopped by EtOH precipitation, and the oligonucleotides were purified by RP-HPLC.
  • the pre-catalyst solution was prepared by mixing 10mM palladium (II) acetate in DMA (100 ⁇ L), 100 mM TPPTS in water (100 ⁇ L), 20mM Copper (II) acetate in water (100 ⁇ L) and diluted up to 1 mL with mQ millipore water, resulting in a 1 mM solution of Pd(0)-TPPTS complex and 2 mM solution of Cu(ll).
  • Enzymatic assay protocol 1 Short assay
  • the plate was left to develop in the dark for 15 to 20 min and was quenched by the addition of 50 ⁇ L of 2M NaOH to each of the wells.
  • the readout was performed using a plate reader (Tecan) and measuring absorption at 405 vs 620 nm.
  • Enzymatic assay protocol 2 Long, sensitive assay
  • the plate was sealed and left to develop in the dark for 18 -20 hours and was quenched by the addition of 50 ⁇ L of 2M NaOH to each of the wells.
  • the readout was performed using a plate reader (Tecan) and measuring absorption at 405 vs 620 nm.
  • the phosphonate-containing group (R 1 ) is considered to be effectively positioned in the active site of the pocket of ACP3. Further surrounding binding sites on ACP3 may be utilized to further improve affinity and inhibitory potency by utilizing suitable groups Y. For instance, using a proline-type scaffold is a viable approach, as indicated by the results in FIGs. 3 and 4.
  • Fluorescence Polarisation (FP) measurements with FITC-labelled compounds The FITC-labelled compounds were diluted to a concentration between 10 and 50 nM (5 pl) and incubated for 15 min in a black 384-well plate (Greiner small volume and non-binding) with serial dilutions of protein (5 pl). The fluorophore was excited at 485 nm and the emission was measured at 535 nm on a Spectra Max Paradigm multimode plate reader (Tecan). The experiments were performed in triplicate.
  • HT1080.hACP3 and wild type HT1080 cells were detached with an Accutase® cell detachment solution, resuspended in a standard culture medium, and transferred to falcon tubes. The cells were counted and the mixtures were centrifuged at 400 ref for 3 min. The supernatant was removed and the cells were resuspended in FACS buffer (PBS 1x, 2% BSA, 2mM EDTA) at a dilution of approx. 300,000 cells/100 ⁇ L; the suspensions were kept on ice. A 96-well plate was used and 100 ⁇ L of the cell solution was loaded in each well.
  • FACS buffer PBS 1x, 2% BSA, 2mM EDTA
  • the plate was left to incubate on ice for 30 min and was then centrifuged at 4 °C for 3 min (400 ref). The supernatant was removed and the cell pellets were resuspended in FACS buffer containing 50 nM concentrations of each of the compounds. The plate was further incubated in the dark at room temperature for 1 h, after which the plate was centrifuged at 4 °C for 3 min (400 ref). A washing step followed and a live/dead cell stain (Zombie NIR) was performed for 20 min at 4°C in the dark. The cells were washed and resuspended in 200 ⁇ L FACS buffer. Data was acquired on a Cytoflex S flow cytometer (Beckman Coulter) and analyzed using the FlowJo software v10 (BD Biosciences).
  • FIG. 7 demonstrates that the FITC-labelled ACP3-binding compounds selectively stained the transfected HT1080.hACP3 cell line, while FIG. 8 shows that they do not bind to the wild type HT1080 cells.
  • Radiolabeling of Compounds 18, 12a,b, 16a,b, 14a,b, 25a and 27a with Lutetium-177 was performed before biodistribution studies at the molar activity of 800 KBq/nmol.
  • 10 ⁇ L of the labeling solution corresponds to one dose (1.25 nmol labeled with 1 MBq).
  • 40 ⁇ L of the labeling solution was diluted with 560 ⁇ L PBS to inject 150 ⁇ L per mouse.
  • Dose escalation studies were performed by the addition of unlabeled precursor to reach the desired amounts per 1 MBq (2.5, 5, 10, and 20 nmol).
  • HT1080.hACP3 cells and PC3.hACP3 cells grown to 80% confluency, were detached with trypsin-EDTA, collected by centrifugation (5 min at 1 ,000g), and resuspended in sterile Hanks’ Balanced Salt Solution (Gibco, #14170-1 12). 100 ⁇ L comprising 5 million cells was injected subcutaneously in the right flank of athymic BALB/c AnNRj-Foxnl nude mice (age 4-8 weeks).
  • Biodistribution analyses were also performed in PC3.hACP3 xenografts.
  • selective tumor accumulation was observed for compound 20a, (FIG. 22A) and for compound 22a (FIG. 22B) with long tumor residence time (tumor half life > 72 hours) and low accumulation in healthy organs including prostate, salivary glands, seminal vesicles, bones, testicles.
  • Test compounds were incubated in the presence of tyrosyl acid phosphatase (Sigma Chemical Co.) and radiolabelled substrate ([ 14 C]phosphotyrosine, (NEN Dupont Custom Synthesis)) plus cold O-phospho-L-tyrosine (10 pM) (Sigma Chemical Company) in a 50 mM sodium acetate buffer (pH 5.5) for 30 min at 37°C.
  • the reaction was stopped by placing the assay on ice and the addition of a 100 ⁇ L aliquot of an enzyme inhibitor solution (1.1 mM sodium orthovanadate, Sigma Chemical Co.; 0.55 M sodium fluoride, Sigma Chemical Co.).
  • the incubation mixture was passed through an ion exchange column ((Ag 1 -x8) (Bio-Rad Laboratories)) and washed with 2.5 mL of distilled deionized water.
  • the total column effluent containing the radiolabelled product [ 14 C]tyrosine) was collected and quantified by liquid scintillation spectroscopy.
  • the test compound IC50 or the concentration of test compound necessary to inhibit 50% of the dephosphorylation was calculated using a quantal dose-response calculation and is reported as an average of at least duplicate determinations using several inhibitor concentrations.
  • the C atom to which the group Q e.g., phosphonic acid (PO3H2) or an isostere thereof
  • the C atom to which the group Q has an absolute configuration corresponding to the ones below; i.e., an (Reconfiguration, e.g. as shown in the structures below:
  • HT1080.hACP3 tumors were implanted into male Balb/c nu/nu mice (age 6-8 weeks) as described in Example 6 and allowed to grow for 7 days to an average volume of -170 mm 3 .
  • Compound 20a 177 Lu-ProX1-DOTAGA
  • compound 22a 177 Lu-ProX3-(S)-DOTAGA
  • tail vein injection 150 ⁇ L, 62.5 nmol/kg, 250 or 1000 MBq/kg.
  • Compounds 20a and 22a mediated strong in vivo anti-cancer activity at the 1000 MBq/kg dose.
  • Compounds were dissolved in DMSO (stock solutions at 1 mM) and diluted in acetate buffer (1 M, pH 4.5) to a final concentration of 200 pM.
  • Compound solutions (5 ⁇ L) were mixed with 177 LuCh (10 ⁇ L, 2 MBq/ ⁇ L) and with acetate buffer (5 ⁇ L, 1 M, pH 4.5).
  • the mixture (20 ⁇ L) was heated at 90 °C for 10 min, left to equilibrate to rt, and diluted to 5 nM with 1 % bovine serum albumin (BSA) in PBS.
  • BSA bovine serum albumin
  • tissue sections were washed two times with PBS (pH 7.4), dried at rt, and a hydrophobic circle was drawn around the tissue with a Dako Pen.
  • the sections were blocked with 20% fetal calf serum and 3% BSA in PBS (pH 7.4) for 30 min and washed three times with PBS (pH 7.4).
  • the tissue slices were separately incubated with 177 Lu-labeled compounds (500 ⁇ L, 5 nM, ⁇ 50 kBq) for 1 h at rt. Sections were subsequently washed three times with PBS (pH 7.4), dried at room temperature, and exposed to the phosphor screen overnight. I mages were recorded on a CR-35 Bio scanner and processed with the AIDA image analysis software.
  • Example 10 Confocal microscopy studies of ProX1-(SS)-FITC (13), ProX2-(S)-Fluo (15a) and ProX2- (R)-Fluo (15b), ProX3-(S)-Fluo (17a) and ProX3-(R)-Fluo (17b)
  • HT1080.hACP3, PC3.hACP3, and corresponding ACP3-negative wild-type cells were seeded into 4-well coverslip chamber plates at a density of 10 4 cells per well in culture medium and allowed to grow for 24 hours at 37 °C (5% CO2).
  • Hoechst 33342 nuclear dye was used to stain nuclear structures.
  • Test compounds 100 nM were incubated in fresh culture medium. Randomly selected colonies were imaged ⁇ 20 min after incubation on an SP8 confocal microscope equipped with an AOBS device (Leica Microsystems).
  • FIG. 27 For ProX1-(SS)-FITC (13), FIG. 28 for ProX2-(S)-Fluo (15a) and ProX2- (R)-Fluo (15b) and FIG. 29 for ProX3-(S)-Fluo (17a) and ProX3-(R)-Fluo (17b).
  • Confocal microscopy studies confirmed membranous staining of ACP3-positive cell lines, while no interaction was detected with the wild- type cells (negative controls HT1080.wt and PC3.wt in FIGs. 27, 28 and 29). Internalization of ACP3 ligands was not observed in confocal microscopy studies.
  • Example 11 Ex vivo biodistribution studies of ProX1-(SS)-AF488 (29a) and ProX3-(S)-AF488 (30a)
  • Tumor-bearing BALB/c nude mice males, tumor model: HT1080.hACP3, tumor size of above 500 mm 3 ) were intravenously injected with ProX1 -(SS)-AF488 (29a) and ProX3-(S)-AF488 (30a) (200 pM, 150 ⁇ L sterile PBS), respectively.
  • Mice were sacrificed 2 h post-injection to isolate tumor, heart, lung, liver, spleen, intestine, kidney, muscle, and salivary glands. The tissues were embedded in Richard-Allan ScientificTM Neg-50TM Frozen Section Medium and cut (10 pm of thickness) with a cryostat microtome. Tissue sections were fixed using mounting medium with DAPI. Images were recorded on an Axioskop 2 fluorescence microscope (Zeiss; 20x/0.7) and processed with lmageJ2 (version 2.14.0).
  • Example 12 In Vivo biodistribution studies and PET-imaging with 68 Ga-ProX1-(SS)-DOTA (44)
  • Galli RD Gallium-68 Generator was used from IRE EliT Radiopharma. Standard elution protocol was followed, which typically yielded -380 MBq of 68 GaCh in ⁇ 1 .5 MBq/ ⁇ L in 0.1 M HCI.
  • ProX1-(SS)-DOTA (27a) (6 ⁇ L, 6 nmol, from 1 mM solution in PBS) was diluted with sodium acetate (34 ⁇ L, 1 M in water, pH 4.5). 24 MBq of 68 Ga solution (20 ⁇ L at an activity of 1.5 MBq/ ⁇ L) were added and the mixture was heated at 90 °C for 10 min and passively cooled down to rt for 5 min. After cooling to room temperature, an aliquot was analyzed by RP-HPLC (XTerra C18, 5% MeCN in 0.1 % aq. TFA to 80% over 20 min on a Merck-Hitachi D-7000 HPLC system equipped with a Raytest Gabi Star radiodetector) (FIG. 34).
  • the mouse was sacrificed 1 h after injection with a dose of 62.5 nmol/kg (600 MBq/kg) and the intact carcass was analysed using an imager (superArgus PET/CT scanner (Sedecal, Madrid, Spain, formerly Vista explore). Results
  • Example 13 In Vitro radioligand bead-based assay with 177 Lu-ProX1-(SS)-DOTA (28a) and 177 Lu- ProX3-(S)-DOTAGA (22a)
  • Radiolabelling of ligands was performed in accordance with Example 6. Specific activities of 20 MBq/nmol were used throughout all in vitro experiments.
  • Magnetic Streptavidin-coated DynabeadsTM M-280 (8 ⁇ L) were loaded into a 1 .5 mL Eppendorf tubes and washed with 390 ⁇ L of phosphate buffer saline solution containing 0.05% Tween-20 (PBS-T). Supernatant removal was performed when the Eppendorf tubes were loaded into a magnetic rack. The washed beads were resuspended in 390 ⁇ L of PBS-T and biotinylated recombinant ACP3 (10 ⁇ L of 3.6 pM solution) was added to the mixtures. After 30 min of incubation time, the supernatant was removed and the beads were washed with 400 ⁇ L PBS-T.
  • PBS-T phosphate buffer saline solution containing 0.05% Tween-20
  • the beads were resuspended with a solution of 177 Lu-ProX1 -(SS)-DOTA (28a) or 177 Lu-ProX3-(S)-DOTAGA (22a) (10 KBq, 1 ⁇ mol, 0.4 mL) in PBS-T and left to incubate for 30 min. A 5000-fold molar excess was added before adding the radioligand in the blocking arm. The supernatants were collected, the beads were washed with 400 ⁇ L PBS-T and then resuspended. The beads suspensions and the supernatants were measured with a Packard Cobra Gamma Counter.
  • Tumor xenografting was performed analogously to Example 6 and masses were allowed to grow to -100 mm 3 . Mice were randomly assigned to different therapy groups:
  • the HT1080.hACP3 model was used to assess the SMDCs of the invention in a therapeutic setting (FIG. 39).
  • the best performing molecule was ProX1 -(SS)-GlyPro-MMAE (31) which resulted in notable tumor shrinkage in all treated animals.
  • ProX1-(SS)-PhoCI1-MMAE (36) bearing the phosphatase cleavable linker also elicited tumor-growth retardation in all mice in the group.
  • ProX1-(SS)-GlyPro-MMAE (31) mediated a therapeutic effect in all treated animals, causing significant tumor growth retardation. Additionally, unlabelled ProX1 -(SS)-DOTA (27a) didn’t elicit a therapeutic response in tumor bearing mice (FIG. 41) due to the lack of a cytotoxic moiety.
  • Example 15 Fluorine-derivatives of a-Benzylaminobenzylphosphonic acid synthesis and in vitro characterization
  • I46 was prepared via GP1 (on a 0.25 mmol scale) from intermediate I5c and 2-Thiophenmethylamin. After lyophilization, the product was obtained as a white solid (31 .5 mg, 93 ⁇ mol, 37% yield), m/z calculated for
  • I48 was prepared via GP14 (on a 20 ⁇ mol scale) from intermediates I9a and I46.
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 5 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was redissolved in acetonitrile:water (1 :1 , 10 mL) mixture. After lyophilization, the product was obtained as a white solid (5.6 mg, 7 ⁇ mol, 33% yield), m/z calculated for C42H53N8O6PS [M-Hp 827.97; m/z observed 827.4.
  • I54 was prepared via GP14 (on a 12 ⁇ mol scale) from intermediate I52 and compound 2.
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 5 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was redissolved in acetonitrile:water (1 :1 , 10 mL) mixture.
  • I54a (S, S) was obtained as a white solid after lyophilization (6.3 mg, 8.0 ⁇ mol, 66% yield), m/z calculated for C40H51N8O7P [M-H]- 785.36; m/z observed 785.4.
  • I54b (S, R) was obtained as a white solid after lyophilization (5.3 mg, 6.7 ⁇ mol, 56% yield), m/z calculated for C40H51N8O7P [M-H]- 785.36; m/z observed 785.4.
  • I54c (R, R) was obtained as a white solid after lyophilization (7.1 mg, 9.0 ⁇ mol, 75% yield), m/z calculated for C40H51N8O7P [M-H]- 785.36; m/z observed 785.4.
  • I55b (S) was obtained as a white solid after lyophilization (24 mg, 65 ⁇ mol, 11 % yield), m/z calculated for C19H21 N5O3 [M-H]- 366.41 ; m/z observed 366.2.
  • I57 was prepared via GP14 (on a 20 ⁇ mol scale) from intermediates I55 and I6c.
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 5 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was redissolved in acetonitrile:water (1 :1 , 10 mL) mixture.
  • I60 was prepared via GP14 (on a 20 ⁇ mol scale) from intermediates I58 and I6c.
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 5 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was redissolved in acetonitrile:water (1 :1 , 10 mL) mixture.
  • I63 was prepared via GP14 (on a 25 ⁇ mol scale) from intermediates 161 and I6c.
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 5 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was redissolved in acetonitrile:water (1 :1 , 10 mL) mixture.
  • I63 was obtained as a white solid after lyophilization (3.3 mg, 4 ⁇ mol, 15% yield), m/z calculated for C45H53N8O8P [M-H]- 863.37; m/z observed 863.4.
  • 59 was prepared via GP15 (on a 4.7 ⁇ mol scale) from intermediate I66.
  • the Boc group was deprotected by dissolving the purified material in a DCM:TFA (1 :1 , 5 mL) mixture and stirring for 2 hours at room temperature. The solvent was removed under reduced pressure and the crude was dissolved in acetonitrile:water (1 :1 , 5 mL) mixture.
  • 59 was obtained as a white solid after lyophilization (1 .4 mg, 1 .3 ⁇ mol, 17% yield), m/z calculated for C53H75N12O13P [M-Hp 11 17.53; m/z observed 1117.6.
  • the Boc group was deprotected by dissolving the intermediate I64 (20 mg, 35 ⁇ mol, 1.0 equiv.) in a
  • 60 was prepared via GP15 (on a 6.4 ⁇ mol scale) from intermediate I68. 60 was obtained as a white solid after lyophilization (2.5 mg, 2 ⁇ mol, 37% yield), m/z calculated for C48H73N12O13P [M-H]- 1055.52; m/z observed 1055.6.
  • the compound 53 has similar high potency compared to compound 27a, while lower inhibitory potencies have been observed for compounds 53, 55a, 55b, 55c and 55d (FIG. 43).
  • the assay was performed according to protocol 2 in Example 2.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des ligands contre la phosphatase acide 3 (ACP3 ou ACPP), également connus sous le nom de phosphatase acide prostatique (PAP). Des ligands sélectifs de l'ACP3 peuvent être capables d'interagir exclusivement avec des antigènes exprimés à la surface de cellules tumorales pour des applications de pharmaco-administration in vivo. Le ligand peut afficher une affinité et une sélectivité très élevées vis-à-vis de l'ACP3 pour permettre l'administration ciblée d'une charge utile, comme des charges utiles thérapeutiques et diagnostiques, à un site atteint d'une maladie caractérisée par l'expression d'ACP3 ou à risque de l'attraper.
PCT/EP2024/080350 2023-10-27 2024-10-25 Ligands de la phosphatase acide 3 pour applications d'administration ciblée Pending WO2025088200A2 (fr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP23206564.9 2023-10-27
EP23206564 2023-10-27
EP24167036.3 2024-03-27
EP24167036 2024-03-27
EP24206215 2024-10-11
EP24206215.6 2024-10-11

Publications (1)

Publication Number Publication Date
WO2025088200A2 true WO2025088200A2 (fr) 2025-05-01

Family

ID=93214931

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2024/080350 Pending WO2025088200A2 (fr) 2023-10-27 2024-10-25 Ligands de la phosphatase acide 3 pour applications d'administration ciblée

Country Status (1)

Country Link
WO (1) WO2025088200A2 (fr)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005082023A2 (fr) 2004-02-23 2005-09-09 Genentech, Inc. Liants et conjugues heterocycliques auto-immolateurs

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005082023A2 (fr) 2004-02-23 2005-09-09 Genentech, Inc. Liants et conjugues heterocycliques auto-immolateurs

Non-Patent Citations (57)

* Cited by examiner, † Cited by third party
Title
"Handbook of Experimental Immunology", vol. I-IV, 1986, BLACKWELL SCIENTIFIC PUBLICATIONS
"Molecular Biology Techniques: An Intensive Laboratory Course", 1998, ACADEMIC PRESS
"PCR (Introduction to Biotechniques Series", 1997, SPRINGER VERLAG
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO.
"Short Protocols in Molecular Biology", 1999, JOHN WILEY & SONS
"The Pharmacological Basis of Therapeutics", 2001, MCGRAW-HILL CO.
A. AHONENK. KAIREMOP. KARNANIJ. HEIKKILAM. NURMIM. TERASO. LUKKARINEN: "Radioimmunodetection of Prostate Cancer by 1111n-Labeled Monoclonal Antibody Against Prostatic Acid Phosphatase", ACTA ONCOL., vol. 32, 1993, pages 723 - 727
A. ANANDP. K. SRIVASTAVA: "A Molecular Description of Acid Phosphatase", APPL. BIOCHEM. BIOTECHNOL., vol. 167, 2012, pages 2174 - 2197, XP035098356, DOI: 10.1007/s12010-012-9694-8
A. B. GUTMANE. B. GUTMAN: "An '' acid '' phosphatase occurring in the serum of patients with metastasizing carcinoma of the prostate gland", J. CLIN. INVEST., vol. 17, 1938, pages 473 - 478
A. I. VOVK ET AL., BIOORG. MED. CHEM. LETT., vol. 18, 2008, pages 4620 - 4623
A. SERGEEVAM. G. KOLONINJ. J. MOLLDREMR. PASQUALINIW. ARAP: "Display technologies: application for the discovery of drug and gene delivery agents", ADV. DRUG DELIV. REV., vol. 58, 2006, pages 1622 - 1654
BEERS, S.A. ET AL., BIOORGANIC & MEDICINAL CHEMISTRY,, vol. 4, no. 10, 1996, pages 1693 - 1701
BERGE ET AL., J.PHARM.SCI., vol. 66, 1977, pages 1 - 19
BERNARDES, G.J. ET AL., ANGEW. CHEM. INT. ED. ENGL, vol. 51, 2012, pages 941
BERNARDES, G.J. ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 51, 2012, pages 941
BOSSLET, K. ET AL., CANCER RES., vol. 58, 1998, pages 1195 - 1201
C. F. SCHWENDERS. A. BEERSE. A. MALLOYJ. J. CINICOLAD. J. WUSTROWK. D. DEMARESTJ. JORDAN: "Benzylphosphonic acid inhibitors of human prostatic acid phosphatase", BIOORG. MED. CHEM. LETT., vol. 6, 1996, pages 311 - 314, XP004135084, DOI: 10.1016/0960-894X(96)00018-2
C. J. ERCOLEP. H. LANGEM. MATHISENR. K. CHIOUP. K. REDDYR. L. VESSELLA: "Prostatic Specific Antigen and Prostatic Acid Phosphatase in the Monitoring and Staging of Patients with Prostatic Cancer", J. UROL., vol. 138, 1987, pages 1181 - 1184
CARL ET AL., J. MED. CHEM., vol. 24, 1981, pages 479 - 480
CASI, G. ET AL., J. AM. CHEM. SOC., vol. 134, 2012, pages 5887
CHAKRAVARTY ET AL., J. MED. CHEM., vol. 26, 1983, pages 638 - 644
D. SCHRAMAR. A. REISFELDJ. C. BECKER: "Antibody targeted drugs as cancer therapeutics", NAT. REV. DRUG DISCOVERY, vol. 5, 2006, pages 147 - 159, XP002566323, DOI: 10.1038/nrd1957
D. W. MOSSF. D. RAYMONDD. B. WILER. REJ, CLINICAL AND BIOLOGICAL ASPECTS OF ACID PHOSPHATASE, vol. 32, 1995, pages 431 - 467
DE GROOT ET AL., JOURNAL OF ORGANIC CHEMISTRY, vol. 66, no. 26, 2001, pages 8815 - 8830
DORONINA S.O. ET AL., BIOCONJUGATE CHEM, vol. 19, 2008, pages 1960
E. J. SMALLP. F. SCHELLHAMMERC. S. HIGANOC. H. REDFERNJ. J. NEMUNAITISF. H. VALONES. S. VERJEEL. A. JONESR. M. HERSHBERG: "Placebo-Controlled Phase III Trial of Immunologic Therapy with Sipuleucel-T (APC8015) in Patients with Metastatic, Asymptomatic Hormone Refractory Prostate Cancer", J. CLIN. ONCOL., vol. 24, 2006, pages 3089 - 3094
E. LUCHTER-WASYLW. OSTROWSKI: "Subunit structure of human prostatic acid phosphatase", BIOCHIM. BIOPHYS. ACTA, vol. 365, pages 349 - 359, XP024556084, DOI: 10.1016/0005-2795(74)90008-7
FLEMING MTHERMSEN RPURYSKO AS ET AL.: "True-Positive F-Flotufolastat Lesions in Patients with Prostate Cancer Recurrence with Baseline-Negative Conventional Imaging: Results from the Prospective, Phase 3, Multicenter SPOTLIGHT Study", J NUCL MED., vol. 65, no. 7, 1 July 2024 (2024-07-01), pages 1080 - 1086
G. S. KILSHEIMERB. AXELROD: "Inhibition of prostatic acid phosphatase by alpha-hydroxycarboxylic acids", J. BIOL., vol. 227, pages 879 - 890
H. K. BINZ, P. AMSTUTZ, A. PLUCKTHUN, BIOTECHNOL., vol. 23, 2005, pages 1257 - 1268
HEO YA.: "Flotufolastat F 18: Diagnostic First Approval", MOLDIAGN THER., vol. 27, no. 5, 2023, pages 631 - 636
I. B. QUINTEROC. S. L. ARAUJOA. E. PULKKAR. S. WIRKKALAA. M. HERRALAE.-L. ESKELINENE. JOKITALOP. A. HELLSTROMH. J. TUOMINENP. P. H: "Prostatic Acid Phosphatase Is Not a Prostate Specific Target", CANCER RES., vol. 67, 2007, pages 6549 - 6554
INT. J. CANCER, vol. 102, 2002, pages 75 - 85
J. A. REDDY, R. DORTON, E. WESTRICK, A. DAWSON, T. SMITH, L.-C. XU, M. VETZEL, P. KLEINDL, I. R. VLAHOV, C. P.LEAMON: "Preclinical Evaluation of EC145, a Folate-Vinca Alkaloid Conjugate", CANCER RES., vol. 67, 2007, pages 4434 - 4442, XP008151835, DOI: 10.1158/0008-5472.CAN-07-0033
J. K. MYERST. S. WIDLANSKI: "Mechanism-Based Inactivation of Prostatic Acid Phosphatase", SCIENCE, vol. 262, 1993, pages 1451 - 1453
J.-N. RYBAKE. TRACHSELJ. SCHEUERMANND. NERI: "Ligand-Based Vascular Targeting of Disease", CHEMMEDCHEM, vol. 2, 2007, pages 22 - 40, XP002573147, DOI: 10.1002/cmdc.200600181
KRATOCHWIL ET AL., JOURNAL OF NUCLEAR MEDICINE, vol. 57, no. 12, December 2016 (2016-12-01), pages 1941 - 1944
LEAMON, C.P. ET AL., BIOCONJUGATE CHEM, vol. 17, 2006, pages 1226
LEAMON, C.P. ET AL., BIOCONJUGATE CHEM., vol. 11, 2006, pages 1226
M. PERÄLÄ-HEAPEP. VIHKOA. LAINEJ. HEIKKILAR. VIHKO: "Effects of tumour mass and circulating antigen on the biodistribution of1111n-labelled F(ab')2 fragments of human prostatic acid phosphatase monoclonal antibody in nude mice bearing PC-82 human prostatic tumour xenografts", EUR. J. NUCL. MED., vol. 18, 1991, pages 339 - 345
M. SRINIVASARAOC. V. GALLIFORDP. S. LOW: "Principles in the design of ligand-targeted cancer therapeutics and imaging agents", NAT. REV. DRUG DISCOV., vol. 14, 2015, pages 203 - 219
N. HEYNICKXK. HERRMANNK. VERMEULENS. BAATOUTA. AERTS: "The salivary glands as a dose limiting organ of PSMA- targeted radionuclide therapy: A review of the lessons learnt so far", NUCL. MED. BIOL., 2021, pages 98 - 99
N. KRALLF. PRETTOD. NERI: "A bivalent small molecule-drug conjugate directed against carbonic anhydrase IX can elicit complete tumour regression in mice", CHEM. SCI., vol. 5, 2014, pages 3640 - 3644
N. KRALLF. PRETTOW. DECURTINSG. J. L. BERNARDESC. T. SUPURAND. NERI: "A Small-Molecule Drug Conjugate for the Treatment of Carbonic Anhydrase IX Expressing Tumors", ANGEW. CHEM., INT. ED., vol. 53, 2014, pages 4231 - 4235, XP055183705, DOI: 10.1002/anie.201310709
P. M. LORUSSOM. J. EDELMANS. L. BEVERK. M. FORMANM. PILATM. F. QUINNJ. LIE. I. HEATHL. M. MALBURGP. J. KLEIN ET AL.: "Phase I Study of Folate Conjugate EC145 (Vintafolide) in Patients With Refractory Solid Tumors", J. CLIN. ONCOL., vol. 30, 2012, pages 4011 - 4016, XP055419700, DOI: 10.1200/JCO.2011.41.4946
P. S. LOWW. A. HENNED. D. DOORNEWEERD: "Discovery and Development of Folic-Acid-Based Receptor Targeting for Imaging and Therapy of Cancer and Inflammatory Diseases", ACC. CHEM. RES., vol. 41, 2008, pages 120 - 129, XP007915422, DOI: 10.1021/ar7000815
P.J. KOCIENSKI: "Protecting Groups", 1994, GEORG THIEME VERLAG
R. J. BABAIANJ. L. MURRAYL. M. LAMKIT. P. HAYNIEE. M. HERSHM. G. ROSENBLUMH. J. GLENNM. W. UNGERD. J. CARLOA. C. VON ESCHENBACH: "Radioimmunological Imaging of Metastatic Prostatic Cancer with 1111ndium-Labeled Monoclonal Antibody Pay 276", J. UROL., vol. 137, 1987, pages 439 - 443
R. J. BABAIANL. M. LAMKI: "Radioimmunoscintigraphy of prostate cancer", SEMIN. NUCL. MED., vol. I-III, 1989, pages 309 - 321
R. J. REBELLO, C. OING, K. E. KNUDSEN, S. LOEB, D. C. JOHNSON, R. E. REITER, S. GILLESSEN, T. VAN DER KWAST, R.G. BRISTOW: "Prostate cancer", NAT. REV. DIS. PRIMERS, vol. 7, 2021, pages 9
S. A. BEERSC. F. SCHWENDERD. A. LOUGHNEYE. MALLOYK. DEMARESTJ. JORDAN: "Phosphatase inhibitors-III. Benzylaminophosphonic acids as potent inhibitors of human prostatic acid phosphatase", BIOORG. MED. CHEM., vol. 4, 1996, pages 1693 - 1701
S. VEERAMANIT.-C. YUANS.-J. CHENF.-F. LINJ. E. PETERSENS. SHAHEDUZZAMANS. SRIVASTAVAR. G. MACDONALDM.-F. LIN: "Cellular prostatic acid phosphatase: a protein tyrosine phosphatase involved in androgen-independent proliferation of prostate cancer", ENDOCR.-RELAT. CANCER, vol. 12, 2005, pages 805 - 822
SARTOR ET AL., N ENGL J MED., vol. 385, no. 12, 16 September 2021 (2021-09-16), pages 1091 - 1103
SUTHERLAND, M.S.K., J. BIOL. CHEM, vol. 281, 2006, pages 10540
WURZER ADI CARLO DSCHMIDT A ET AL.: "Radiohybrid Ligands: A Novel Tracer Concept Exemplified by F- or Ga-Labeled rhPSMA Inhibitors", J NUCL MED., vol. 61, no. 5, 2020, pages 735 - 742, XP055854046, DOI: 10.2967/jnumed.119.234922
WURZER APARZINGER MKONRAD M ET AL.: "Preclinical comparison of four [18F, Ga]rhPSMA-7 isomers: influence of the stereoconfiguration on pharmacokinetics", EJNMMI RES., vol. 10, no. 1, 7 December 2020 (2020-12-07), pages 149, XP021285239, DOI: 10.1186/s13550-020-00740-z
YANG, J. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 13872

Similar Documents

Publication Publication Date Title
ES2910941T3 (es) Ligandos de proteína de activación de fibroblastos para aplicaciones de administración dirigida
US20240148916A1 (en) Bivalent fibroblast activation protein ligands for targeted delivery applications
JP2024503637A (ja) 線維芽細胞活性化タンパク質リガンドを含む化合物およびその使用
JP2024503635A (ja) 線維芽細胞活性化タンパク質リガンドを含む化合物およびその使用
US20250177582A1 (en) High-affinity ligands of fibroblast activation protein for targeted delivery applications
JP2024514528A (ja) 線維芽細胞活性化タンパク質アルファおよび/または前立腺特異的膜抗原を標的とするヘテロ二価およびホモ二価薬剤
CA3171025A1 (fr) Ligands de proteine d'activation des fibroblastes pour applications d'administration ciblee
EP4043452A1 (fr) Ligands de protéines d'activation de fibroblastes bivalents pour des applications d'administration ciblées
JP2025529319A (ja) 標的送達用途のための多価線維芽細胞活性化タンパク質リガンド
TW202409009A (zh) 黑素皮質素2型受體(mc2r)之靶向治療劑及其用途
EP4612144A1 (fr) Ligands d'anhydrase carbonique ix pour applications par administration ciblée
WO2025088200A2 (fr) Ligands de la phosphatase acide 3 pour applications d'administration ciblée
US20230147962A1 (en) Fibroblast activation protein ligands for targeted delivery applications
CN118591549A (zh) 碳酸酐酶ix配位体
CN116917278A (zh) 用于靶向递送应用的二价成纤维细胞活化蛋白配体
US20250213735A1 (en) Neuropeptide y1 receptor (npy1r) targeted therapeutics and uses thereof
US20240226343A1 (en) Neuropeptide y1 receptor (npy1r) targeted therapeutics and uses thereof
BR112023015831B1 (pt) Ligantes de proteína de ativação de fibroblastos bivalentes para aplicações de liberação direcionada, composições farmacêuticas, seus usos e métodos para preparar um conjugado
HK40077413A (en) Fibroblast activation protein ligands for targeted delivery applications
JP2025540658A (ja) 神経ペプチドy1受容体(npy1r)を標的とした治療薬およびその使用
HK40053369B (en) Fibroblast activation protein ligands for targeted delivery applications
HK40053369A (en) Fibroblast activation protein ligands for targeted delivery applications

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: 24794464

Country of ref document: EP

Kind code of ref document: A2