NZ719939B2 - Compounds for positron emission tomography - Google Patents

Compounds for positron emission tomography

Info

Publication number
NZ719939B2
NZ719939B2 NZ719939A NZ71993914A NZ719939B2 NZ 719939 B2 NZ719939 B2 NZ 719939B2 NZ 719939 A NZ719939 A NZ 719939A NZ 71993914 A NZ71993914 A NZ 71993914A NZ 719939 B2 NZ719939 B2 NZ 719939B2
Authority
NZ
New Zealand
Prior art keywords
conjugate
acid
cozh
methyl
nota
Prior art date
Application number
NZ719939A
Other versions
NZ719939A (en
Inventor
Qingshou Chen
Christopher P Leamon
Philip S Low
Garth L Parham
Iontcho R Vlahov
Original Assignee
Endocyte Inc
Purdue Research Foundation
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 Endocyte Inc, Purdue Research Foundation filed Critical Endocyte Inc
Priority to NZ758917A priority Critical patent/NZ758917A/en
Priority claimed from PCT/US2014/065467 external-priority patent/WO2015073678A1/en
Publication of NZ719939A publication Critical patent/NZ719939A/en
Publication of NZ719939B2 publication Critical patent/NZ719939B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • 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
    • A61K51/0459Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having six-membered rings with two nitrogen atoms as the only ring hetero atoms, e.g. piperazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0474Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group
    • A61K51/0482Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group chelates from cyclic ligands, e.g. DOTA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/52Use of compounds or compositions for colorimetric, spectrophotometric or fluorometric investigation, e.g. use of reagent paper and including single- and multilayer analytical elements
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/60Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances involving radioactive labelled substances

Abstract

Described herein are compounds, compositions, and methods for diagnosing and/or monitoring pathogenic disease using positron emission tomography. Also described are conjugates of the formula B-L-P, wherein B is a radical of a targeting agent selected from vitamin receptor binding ligands (such as folate), PSMA binding ligands, or PSMA inhibitors; L is a divalent linker comprising aspartic acid, lysine, or arginine, and P is a radical of an imaging agent or radiotherapy agent, such as a radionuclide or radionuclide containing group, or a radical of a compound capable of binding a radionuclide, such as a metal chelating group. late), PSMA binding ligands, or PSMA inhibitors; L is a divalent linker comprising aspartic acid, lysine, or arginine, and P is a radical of an imaging agent or radiotherapy agent, such as a radionuclide or radionuclide containing group, or a radical of a compound capable of binding a radionuclide, such as a metal chelating group.

Description

COMPOUNDS FOR POSITRON EMISSION APHY REFERENCE TO RELATED APPLICATIONS This application claims the benefit under 35 U.S.C. § 119(e) of United States Provisional ation Serial Nos. 61/904,387, filed November 14, 2013, 61/904400, filed November 14, 2013, and 61/909,822, filed November 27, 2013, the disclosure of each of which is incorporated herein by reference in its entirety.
TECHNICAL FIELD The invention described herein pertains to compounds, compositions, and methods for diagnosing and/or monitoring diseases and disease states using radionuclides. In ular, the invention described herein pertains to nds, compositions, and methods for diagnosing and/or monitoring pathogenic disease states using radionuclides for positron emission tomography (PET).
OUND AND SUMMARY OF THE INVENTION PET is a nuclear imaging methodology that detects pairs of gamma rays emitted indirectly by a positron-producing radionuclide. Because the two d gamma rays travel in exactly opposite directions, it is possible to locate their site of origin and thereby truct a three-dimensional image of all positron emitters from a computer analysis of the origins of emitted gamma rays. Compared to other radioimaging modalities, such as SPECT, PET reportedly shows higher sensitivity (about 2 orders of magnitude), better spatial resolution (about 5 mm), greater signal to noise, and superior tracer quantification in both preclinical and clinical applications. In addition, in st to the about 90 minutes required for body scans for a standard SPECT imaging, PET image acquisition may be routinely performed in about 20 minutes. Moreover, in viva PET imaging generally requires only subnanomolar (10'10 to 10'”) concentrations of racer, which reportedly zes ial damage to other biological systems. y, PET allows for quantitative dynamic imaging, which may facilitate kinetic studies of target engagement through receptor occupancy. It has been discovered herein that PET agents may be ed to predetermined tissues using Vitamin receptors and/or prostate-specific membrane antigen (PSMA).
For example, vitamin receptors are overexpressed on certain pathogenic cells, including many cancer cell types, activated macrophages, and activated monocytes. In particular, folate ors are overexpressed in many cancers. The folate receptor, a 38 KD GPI—anchored protein that binds the vitamin folic acid with high affinity (<1 nM), is overexpressed on many malignant tissues, ing ovarian, breast, bronchial, and brain cancers. It is estimated that 95% of all ovarian carcinomas overexpress the folate receptor.
In contrast, with the exception of kidney, choroid plexus, and placenta, normal tissues express low or nondetectable levels of the folate receptor. Most cells also use an unrelated reduced folate carrier to acquire the necessary folic acid.
Folate receptors are also overexpressed on activated hages, and activated monocytes. Further, it has also been reported that the folate receptor B, the nonepithelial isoform of the folate receptor, is expressed on activated, but not resting, synovial macrophages. Activated hages can participate in the immune response by nonspecifically engulfing and killing foreign pathogens within the macrophage, by displaying degraded peptides from n ns on the macrophage cell surface where they can be recognized by other immune cells, and by secreting cytokines and other factors that modulate the function of T and B lymphocytes, resulting in further stimulation of immune responses.
However, activated macrophages can also bute to the pathophysiology of disease in some instances. For example, activated macrophages can contribute to atherosclerosis, toid tis, autoimmune disease states, and graft versus host disease, among other disease states.
Following or binding of vitamins to vitamin receptors, such as folic acid and analogs and tives of folic acid to folate receptors, rapid endocytosis delivers the vitamin into the cell, where it is unloaded in an endosomal compartment at lower pH.
Importantly, nt conjugation of small molecules, proteins, and even liposomes to vitamins and other vitamin receptor binding ligands does not block the ability of the ligand to bind to its receptor, and therefore, such ligand conjugates can readily be delivered to and can enter cells by receptor-mediated endocytosis. Accordingly, diagnostic, imaging, and eutic agents can be targeted to vitamin receptors, including the folate receptor, for delivery into vitamin receptor expressing cells.
The prostate is a male uctive organ that functions to e and store seminal fluid, which provides nutrients and fluids for the survival of sperm introduced into the vagina during reproduction. Like other tissues, the prostate gland may develop either malignant (cancerous) or benign (non-cancerous) . Prostate cancer is reportedly one of the most common male cancers in western societies, and is the second leading form of malignancy among American men.
Prostate-specific membrane antigen (PSMA) is a biomarker that is pressed on prostate cancer. PSMA is over-expressed in the malignant prostate tissues when compared to other organs in the human body such as , proximal small intestine, and salivary glands. PSMA is also expressed on the neovasculature within many non- prostate solid tumors, including lung, colon, breast, renal, liver and pancreatic carcinomas, but not on normal vasculature. However, PSMA is expressed minimally in brain. PSMA is a type II cell surface membrane-bound glycoprotein with ~110 kD molecular weight, including an intracellular segment (amino acids 1-18), a transmembrane domain (amino acids 19-43), and an extensive extracellular domain (amino acids 44-750). Though the functions of the intracellular segment and the transmembrane domains are currently reported to be insignificant, the extracellular domain is involved in several distinct activities. For example, PSMA plays a role in the central nervous system, where it lizes N—acetyl—aspartyl glutamate (NAAG) into glutamic and N-acetyl aspartic acid. PSMA also plays a role in the proximal small ine where it removes ed glutamate from poly-y—glutamated folate and (x-linked glutamate from peptides and small les.
Though the particular function of PSMA on prostate cancer cells remains unresolved, PSMA is known to undergo rapid internalization into the cell, similar to cell surface bound ors like vitamin receptors. PSMA is internalized through clathrin-coated pits and subsequently can either e to the cell surface or go to lysosomes. Accordingly, stic, g, and therapeutic agents can be targeted to PSMA for delivery into PSMA sing cells, such as prostate cancer cells.
It has been ered herein that the nds and itions described herein are useful for targeting and delivering radionuclides for diagnosing and/or monitoring various diseases and disease states caused by pathogenic cell populations. In addition, it has been discovered that the compounds and compositions described herein are also useful for targeting and delivering radionuclides for treating various diseases and disease states caused by pathogenic cell populations in radiotherapy.
In one illustrative and non-limiting embodiment of the invention described herein, compounds and compositions described herein are used for diagnosing and/or monitoring, or treating various diseases and disease states caused by pathogenic cell populations. In another illustrative embodiment, methods are described herein for stering compounds and compositions described herein for diagnosing and/or monitoring, or treating various es and disease states caused by pathogenic cell populations. In another embodiment, uses of compounds and compositions are described herein for manufacturing ments for diagnosing and/or monitoring, or treating various diseases and disease states caused by pathogenic cell populations. In another embodiment, kits are bed herein for preparing and/or using compounds and compositions described herein for diagnosing andj’or monitoring, or treating various diseases and e states caused by enic cell populations.
BRIEF DESCRIPTION OF THE DRAWINGS shows a rtem biodistribution study of F-QC07017 and 18F-AIF—QC07043 folate—NOTA—Al-lsF conjugates in s tissues at 90 minutes post injection in nude mice bearing KB tumor xenografts. For each tissue, the histogram is in groups of 4 from left to right: 18F—AIF-QC07017, 18F—AIF—QC07017 + excess folic acid, 18F- AIF-QC07043, F-QC07043 + excess folic acid. shows a postmortem tribution study of 18F—AIF-QCO'iOl7 folate-NOTA-Al-ISF conjugate in various tissues at 90 minutes post injection in nude mice bearing KB tumor xenografts or A549 tumor xenografts. It is to be understood that the vertical axis has been expanded and that the kidney data is truncated. For each tissue, the histogram is in groups of 4 from left to right: 18F—AIF-QC07017 against A549 tumor xenografts, 18F—AIF—QCO?017 + excess folic acid against A549 tumor xenografts, 18F—AIF- 7 against KB tumor xenografts, 18F—AIF-QC07017 + excess folic acid against KB tumor xenografts. shows a postmortem biodistribution study of 18F—AIF-QC07O43 folate-NOTA-Al-‘SF conjugate in various tissues at 90 minutes post injection in nude mice bearing KB tumor xenografts or A549 tumor xenografts. It is to be understood that the vertical axis has been expanded and that the kidney data is truncated. For each tissue, the histogram is in groups of 4 from left to right: 18F—AIF-QC07043 against A549 tumor xenografts, 18F—AIF-QCO?043 + excess folic acid t A549 tumor xenografts, 18F-AIF— QC07043 against KB tumor xenografts, F-QC07O43 + excess folic acid against KB tumor xenografts. shows a postmortem biodistribution study of 18F—AIF-QC07017 and 18F-AIF-QC07043 folate—NOTA-Al-ISF conjugates, compared to 99mTc-EC20 in KB tumor xenograft tissues at 90 minutes post ion in nude mice. The histogram from left to right: 99mTc-EC20 against KB tumor xenografts, 99mTc-EC20 + excess folic acid against KB tumor xenografts, 18F—AIF—QCO?017 against KB tumor xenografts, 18F—AIF—QCO?017 + excess folic acid against KB tumor xenografts, 18F—AIF-QC07043 t KB tumor xenografts, 18F-AIF— QC07043 + excess folic acid against KB tumor xenografts. shows a postmortem biodistribution study of 18F—AIF-QC0'1'017 and 18F-AIF-QC07043 folate-NOTA-Al-lSF conjugates, compared to 99mTc-EC20 in A549 tumor xenograft tissues at 90 minutes post injection in nude mice. The histogram from left to right: 99mTc-EC20 against A549 tumor xenografts, 99mTc—EC20 + excess folic acid against A549 tumor xenografts, 18F—AIF—QC07017 against A549 tumor xenografts, 18F-AIF-QC07017 + excess folic acid against A549 tumor afts, 18F—AIF-QCO7043 t A549 tumor xenografts, 18F—AIF-QC07043 + excess folic acid against A549 tumor xenografts.
DETAILED DESCRIPTION In each of the foregoing and each of the ing embodiments, it is to be understood that the formulae include and represent not only all pharmaceutically acceptable salts of the nds, but also e any and all hydrates and/or solvates of the compound formulae. It is appreciated that certain functional groups, such as the hydroxy, amino, and like groups form complexes and/or coordination compounds with water and/or s solvents, in the various al forms of the compounds. Accordingly, the formulae described herein are to be understood to include and represent those s hydrates and/or solvates. It is also to be understood that the non-hydrates and/or non-solvates of the compound formulae are described by such formula, as well as the hydrates and/or solvates of the compound formulae.
As used herein, the term “composition” generally refers to any product sing the ied ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts. It is to be understood that the compositions bed herein may be prepared from isolated compounds described herein or from salts, solutions, hydrates, solvates, and other forms of the compounds described herein. It is appreciated that certain functional groups, such as the hydroxy, amino, and like groups form complexes and/or coordination compounds with water and/or various solvents, in the various al forms of the compounds. It is also to be understood that the compositions may be prepared from various amorphous, non- amorphous, partially crystalline, crystalline, and/or other morphological forms of the compounds described herein. It is also to be understood that the compositions may be prepared from various hydrates and/or solvates of the nds described .
Accordingly, such pharmaceutical itions that recite compounds described herein are to be understood to include each of, or any combination of, the various morphological forms and/or solvate or hydrate forms of the compounds described herein. In addition, it is to be understood that the itions may be prepared from various co—crystals of the compounds bed herein. ratively, compositions may include one or more carriers, diluents, and/or excipients. The compounds described herein, or compositions containing them, may be formulated in a therapeutically effective amount in any conventional dosage forms appropriate for the methods described herein. The compounds described herein, or compositions containing them, including such formulations, may be administered by a wide y of conventional routes for the methods described herein, and in a wide variety of dosage formats, utilizing known procedures (see generally, Remington: The Science and Practice of cy, (21St ed., 2005)).
In each of the foregoing and each of the ing embodiments, it is also to be understood that the formulae include and represent each possible isomer, such as isomers and geometric isomers, both individually and in any and all possible mixtures.
In each of the foregoing and each of the following embodiments, it is also to be understood that the formulae include and represent any and all crystalline forms, partially crystalline forms, and non crystalline and/or ous forms of the compounds.
Illustrative embodiments of the invention are described by the following clauses: A conjugate of the formula B—L—P or a pharmaceutically acceptable salt f, wherein B is a l of a targeting agent selected from vitamin receptor binding ligands, PSMA binding ligands, and PSMA inhibitors, L is a divalent linker, and P is a radical of an imaging agent or radiotherapy agent, such as a radionuclide or radionuclide containing group, or a precursor thereof, or a radical of a nd capable of binding a radionuclide or radionuclide containing group, such as a metal chelating group.
The conjugate of the preceding clause wherein the targeting agent is a l of a folate receptor binding ligand.
The conjugate of any one of the preceding clauses wherein the targeting agent is a radical of a folic acid.
The ate of any one of the preceding clauses comprising —Asp.
The conjugate of any one of the preceding clauses comprising folate—Asp-Arg.
The conjugate of any one of the preceding clauses comprising folate—Arg.
The conjugate of any one of the preceding clauses wherein the linker comprises a polypeptide.
The conjugate of any one of the preceding clauses wherein the linker comprises a polypeptide sing lysine, arginine, or aspartic acid, or a combination thereof.
The conjugate of any one of the preceding clauses n the linker comprises a lysine.
The conjugate of any one of the preceding clauses wherein the linker comprises Lys.
The ate of any one of the preceding clauses wherein the linker comprises s.
The conjugate of any one of the preceding clauses wherein the linker comprises Arg-Arg-Lys.
The conjugate of any one of the preceding clauses wherein the linker comprises Asp-Arg-Arg-Lys.
The conjugate of any one of the preceding clauses wherein the linker does not include a polyamine radical, such as a polyamine diradical of the formula NH—(CH2)2—NH.
The conjugate of any one of the preceding clauses wherein P comprises the formula N\)—\COZH HOZC—/ or a derivative thereof comprising a chelated metal.
The ate of any one of the preceding clauses comprising the formula *QNmN/Rflos <COZH <002H * HNJLN H NV COZH ::—\COZH HOzC—/ HOZC—/ or a derivative thereof comprising a chelated metal.
The conjugate of any one of the preceding clauses comprising folate—PEG.
The conjugate of any one of the ing clauses comprising folate—PEGz.
The conjugate of any one of the ing clauses comprising folate—PEGs.
The conjugate of any one of the preceding clauses sing folate—PEGu.
The conjugate of any one of the ing clauses wherein the linker comprises [(CH2)20]n, [(CHz)zO]n-(CH2)2-C(O), [(CH2)20]n-(CHz)2-C(O)NH, [(CH2)20]n- (CH2)2—C(O)NH—(CH2)2, [(CH2)2O]2—(CH2)n—C(O)NH—(CH2)2NH, where n is an integer from 1 to about 12.
The conjugate of any one of the preceding clauses wherein the linker comprises [(CH2)2O]2, [(CH2)20]6, or [(CH2)2O]12.
The ate of any one of the preceding clauses wherein the linker comprises (CH2)2O-(CH2)2-C(O), 2O]2-(CH2)2-C(O), 2O]6-(CH2)2-C(O), or [(CH2)20]12-(CH2)2—C(O).
The conjugate of any one of the preceding clauses wherein the linker comprises (CH2)zO—(CH2)2—C(O)NH, [(CHz)zO]2—(CHz)z—C(O)NH, [(CH2)zO]6—(CH2)2— C(O)NH, or [(CH2)2O]12—(CH2)z—C(O)NH.
The ate of any one of the preceding clauses wherein the linker comprises (CH2)zO—(CH2)2-C(O)NH—(CH2)2, [(CH2)2O]2—(CH2)z—C(O)NH—(CH2)2, [(CH2)zO]6—(CH2)2—C(O)NH—(CH2)2, or [(CHz)zO]12—(CH2)2—C(O)NH—(CH2)2.
The conjugate of any one of the preceding clauses wherein the linker ses (CH2)2O—(CH2)2—C(O)NH—(CH2)2NH, [(CH2)2O]2—(CH2)2—C(O)NH—(CH2)2NH, [(CHz)20]6—(CH2)2—C(O)NH—(CH2)2NH, or [(CH2)2O]12—(CH2)2—C(O)NH—(CH2)2NH.
The conjugate of any one of the preceding clauses wherein the linker comprises NH[(CH2)2O]n, NH[(CH2)zO]n—(CH2)z—C(O), NH[(CH2)2O]n—(CH2)2—C(O)NH, NH[(CH2)2O]n—(CH2)2—C(O)NH-(CH2)2, NH[(CH2)2O]n—(CH2)2—C(O)NH-(CH2)2NH, where n is an integer from 1 to about 12.
The ate of any one of the preceding clauses wherein the linker comprises NH(CH2)zO, NH[(CH2)2O]2, 2)ZO]6, NH[(CH2)2O]12.
The conjugate of any one of the preceding clauses wherein the linker comprises NH(CH2)zO—(CH2)2—C(O), NH[(CH2)20]2—(CHz)z—C(O), NH[(CH2)zO]6—(CH2)2— C(O), or NH[(CH2)2O]12-(CH2)z-C(O).
The conjugate of any one of the preceding clauses wherein the linker comprises NH(CH2)2O—(CH2)z—C(O)NH, NH[(CH2)2O]2—(CH2)2—C(O)NH, NH[(CH2)2O]6— (CH2)2—C(O)NH, or NH[(CH2)20]12—(CH2)2—C(O)NH.
The conjugate of any one of the preceding clauses wherein the linker ses NH(CH2)2O—(CH2)2—C(O)NH—(CH2)2, NH[(CH2)2O]2—(CH2)2—C(O)NH—(CH2)2, NH[(CH2)20]6-(CH2)2-C(O)NH-(CH2)2, or NH[(CH2)2O]12-(CH2)2—C(O)NH—(CH2)2.
The conjugate of any one of the preceding clauses n the linker comprises NH(CH2)20—(CH2)2—C(O)NH—(CH2)2NH, NH[(CH2)zO]2—(CHz)z—C(O)NH— (CH2)2NH, NH[(CH2)2O]6-(CH2)2—C(O)NH—(CH2)2NH, or NH[(CH2)2O]12—(CH2)2—C(O)NH— (CH2)2NH.
The conjugate of any one of the preceding clauses wherein the linker ses NH[(CH2)2O]n-(CH2)2NH, where n is an integer from 1 to about 12.
The conjugate of any one of the preceding clauses wherein the linker comprises NH(CH2)zO-(CH2)2NH, NH[(CH2)zO]z-(CH2)2NH, NH[(CH2)zO]6-(CH2)2NH, or 2)20]12-(CH2)2NH.
The conjugate of any one of the preceding s wherein the linker comprises NH[(CH2)zO]n-(CHz)2NH-C(O)—(CH2)z-C(O), where n is an integer from 1 to about 12.
The conjugate of any one of the preceding clauses wherein the linker comprises NH(CH2)20—(CH2)2NH—C(O)—(CH2)2—C(O), NH[(CH2)zO]2—(CH2)2NH—C(O)— (CH2)2—C(O), NH[(CH2)zO]6—(CH2)2NH—C(O)—(CH2)2—C(O), or NH[(CH2)zO]12—(CH2)2NH— C(O)—(CH2)2—C(O).
The conjugate of any one of the preceding clauses comprising the a <COZH <C02H * O N/R * HN N/R —\—NH ENN—\\)—\COZH —\—NH ENN—\ViCOZH OW or 0% or a derivative f comprising a chelated metal.
The conjugate of any one of the preceding clauses where P comprises the * HN <COZH “IQ—\COZH or a derivative thereof comprising a chelated metal.
The conjugate of any one of the preceding clauses comprising the formula HOZC/—HOZC> HOZCF or H020> or a derivative f comprising a chelated metal.
The conjugate of any one of the preceding clauses wherein the targeting agent is a radical of a PSMA binding ligand or PSMA inhibitor.
The conjugate of any one of the preceding s wherein the targeting agent is a radical of a PSMA inhibitor.
The ate of any one of the preceding clauses comprising the formula “TNfl’lLNH * H0200, wherein n is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; or YNWHWCJ)‘NH* HOZC/ll,H'hirN COZH or HOZC/II,HH/H wherein n is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; or,JTT COZH ' H020,”HH/N COZH 01.
COZH NTNH * HN\n/N O COZH where W is O or S.
The conjugate of any one of the preceding clauses wherein the linker comprises a polypeptide.
The conjugate of any one of the preceding clauses wherein the linker comprises a polypeptide comprising phenylalanine, , arginine, or aspartic acid, or a combination thereof.
The conjugate of any one of the preceding clauses wherein the linker comprises a lysine.
The conjugate of any one of the ing clauses wherein the linker ses Lys.
The ate of any one of the preceding s wherein the linker comprises Arg-Lys.
The conjugate of any one of the preceding clauses wherein the linker comprises Asp-Arg-Lys.
The conjugate of any one of the preceding clauses wherein the linker comprises Arg-Asp-Arg.
The ate of any one of the preceding clauses wherein the linker comprises Arg-Asp-Arg-Lys.
The conjugate of any one of the preceding clauses wherein the linker comprises Phe-Arg-Asp.
The conjugate of any one of the preceding clauses wherein the linker comprises Phe-Arg—Asp-Arg.
The conjugate of any one of the preceding clauses n the linker comprises Phe-Arg—Asp-Arg-Lys.
The conjugate of any one of the preceding clauses wherein the linker comprises Phe-Phe-Arg.
The ate of any one of the preceding clauses wherein the linker comprises Phe—Phe-Arg—Asp.
WO 73678 The conjugate of any one of the preceding clauses wherein the linker comprises Phe-Phe—Arg-Asp-Arg.
The conjugate of any one of the preceding clauses wherein the linker ses e—Arg-Asp-Arg-Lys.
The ate of any one of the preceding clauses wherein the radical of the radionuclide or radionuclide containing group, or precursor thereof, or compound capable of binding a radionuclide or radionuclide containing group comprises a radical of NOTA.
The conjugate of any one of the preceding clauses wherein P comprises the formula <COZH <COZH [NE * HNWNQWCOZHE“? ,/_/N\) COZH or a derivative thereof comprising a chelated metal.
The conjugate of any one of the preceding clauses comprising the formula , HN <COZH HO2C EN” N;_/N\)—\COZH or a derivative f comprising a chelated metal.
The conjugate of any one of the preceding s comprising the formula O O N n n ‘3 HOZC JLN 0 O wherein n is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
The conjugate of any one of the preceding clauses comprising the formula COZH N N/\/ i0 H H 0 0 H020 N N cogH The conjugate of any one of the preceding s wherein the linker comprises the formula H | * HNNNW The conjugate of any one of the ing clauses wherein the linker comprises the formula H (I) The conjugate of any one of the preceding clauses wherein the linker comprises the formula no u ‘3 0 fiHNWO The conjugate of any one of the preceding clauses wherein one or more of the phenylalanines is L—phenylalanine.
The conjugate of any one of the preceding clauses comprising the formula <COZH (COZH m m *0XNEEM)“COZH M—\—N;"' [,N\)—\COZH O or O 01‘ <COZH * HN HN Phofl—\—NHOEN\)_\COZH or a derivative thereof comprising a chelated metal.
The conjugate of any one of the ing clauses where P comprises the formula * HN <COZH ”Q1002H or a derivative thereof comprising a chelated metal.
The conjugate of any one of the preceding clauses comprising the formula 0 0 NH COZH NH (COZH 0 [mmNV O [NE COzH N\) COZH HOZC/—>N N H020 > HOZC or HozC or * HN\_\ <COZH EN:—\COZH HOZC/—H020> NH <COZH HN [N31CO2H HOZCFH020> or a derivative thereof comprising a chelated metal.
The conjugate of any one of the preceding clauses wherein the radionuclide is a positron emitting uclide.
The conjugate of any one of the preceding clauses wherein the radionuclide is a metal ion.
The conjugate of any one of the ing clauses n the radionuclide is a metal salt.
The conjugate of any one of the preceding clauses comprising an aluminum halide, such as an aluminum fluoride, um chloride, um bromide, or aluminum The conjugate of any one of the preceding clauses comprising an um fluoride.
The conjugate of any one of the preceding clauses comprising an aluminum 18F-fluoride.
The conjugate of any one of the preceding clauses comprising an aluminum iodide.
The conjugate of any one of the preceding clauses comprising an aluminum ”SI-iodide.
The conjugate of any one of the preceding clauses comprising a gallium ion.
The conjugate of any one of the preceding s comprising a 66Ga ion.
The conjugate of any one of the preceding s comprising a 68Ga ion.
The conjugate of any one of the preceding clauses comprising a zirconium ion.
The conjugate of any one of the preceding clauses comprising 21 89Zr ion.
The conjugate of any one of the preceding clauses comprising a copper ion.
The conjugate of any one of the preceding s sing a 64Cu ion.
The conjugate of any one of the preceding clauses wherein the radionuclide is a radiotherapy agent, such as iodine, including 1311, lutetium, including 177Lu, m, including 90Y, strontium, including 89Sr, samarium, including 153Sm, and the like, or a radiotherapy agent containing group.
The conjugate of any one of the preceding clauses comprising a lutetium ion, such as a 177Lu ion.
The conjugate of any one of the preceding clauses comprising a yttrium ion, such as a 90Y ion.
A conjugate of the formulae COZH 0<N\N/> /dNNN/\/firN\/\O/\/o\/\N’lk/N\_/¥C02Hcho7o17 HO {N3fLg HNJE250%22%wa0007029 HN N\ N A I H QC07043 N N O 0 O ul=02H H O H NI N‘ _<OH O H/\/\n/N \/\ /\/ \/\O N 0 O NJ HN \ N /]\\ I H O N N folate-C-NETA or a pharmaceutically acceptable salt thereof.
A conjugate of the formulae 0 §OZH H O o :fi\:§;~nl;j> ”W \/\o/\/ \/\N ‘ (LCOZH “N N\ | IAN folate-NOTA-Al13F (0007017) J\\N N/ COZH o COZH 0 (N3 MNwo’l'VOWNHwnk/NL/ LCOZHM N ZNHNjL/[ZjflH o o FA-PEG12-N0TA-Al-13F ate (oco7o43) M = Al-18F or “Ga folate-C-NETA-M M=Al-15F or 68Ga O¥ or a pharmaceutically acceptable salt thereof.
A conjugate of the formulae 0 flrCOZH N N“ N co H2 N i JL H H 13(— NJ 0 bow H020 N N COZH H H DUPA-EAOA-Phe-Phe-NOTA cogH E H020 N N COZH H H DUPA-C-NETA OH or a pharmaceutically acceptable salt thereof.
A conjugate of the formulae COZH E HOZC H H COZH M=586a64CuorAl-“F DUPA-EAOA-Phe-Phe-NOTA-S‘CuIAl-18F ’ C02H E H020 N N COZH H H DUPA-C-NETA-M OH M = Al-18F,6sGa,177Lu or 90Y or a pharmaceutically acceptable salt thereof.
The conjugate of any one of the ing clauses wherein P comprises the formula N+Me3 x- wherein X" is the conjugate base of an acid, such as trifluoromethanesulfonic acid.
The conjugate of any one of the preceding clauses sing the formula N+Me3 X' CN or * HN\/\O/\/O\/\N N+Me3 X' where X’ is a conjugate base of an acid, such as trifluoromethanesulfonic acid.
The conjugate of any one of the preceding clauses n P comprises the formula The conjugate of any one of the preceding clauses wherein P ses the formula The conjugate of any one of the preceding clauses comprising the formula 0 O * HN\/\o/\/O\/\N * O/\/O\/\N H H F F ON or ON The conjugate of any one of the preceding clauses sing the formula 0 O * HN\/\O/\/O\/\N * O/\/O\/\N H H 18F 18F ON or ON The conjugate of any one of the preceding clauses wherein P comprises the formula *NH-C(CH20H)3.
The conjugate of any one of the ing clauses comprising a boron fluoride.
The conjugate of any one of the preceding clauses sing a boron 18F- fluoride.
A pharmaceutical composition comprising one or more of the conjugates of any one of the preceding clauses, in combination with one or more carriers, diluents, or excipients, or a combination thereof.
A unit dose or unit dosage form composition comprising a diagnostically effective amount of one or more of the conjugates of any one of the preceding clauses, ally in combination with one or more carriers, diluents, or excipients, or a combination thereof for diagnosing and/or ring a pathogenic cell population, such as a cancer or inflammatory disease.
A unit dose or unit dosage form composition comprising a therapeutically effective amount of one or more of the ates of any one of the preceding clauses, optionally in combination with one or more carriers, diluents, or excipients, or a combination thereof for ng a pathogenic cell population, such as a cancer or atory disease.
A composition for diagnosing and/or monitoring a disease or disease state caused at least in part by a pathogenic cell population, such as a cancer or inflammatory disease, in a host animal, the composition comprising a diagnostically effective amount of one or more of the conjugates of any one of the preceding clauses; or a pharmaceutical composition comprising a diagnostically effective amount of one or more of the conjugates of any one of the preceding clauses, optionally further comprising one or more carriers, diluents, or ents, or a combination thereof.
A composition for treating a e or disease state caused at least in part by a pathogenic cell population, such as a cancer or inflammatory disease, in a host animal, the composition comprising a eutically effective amount of one or more of the conjugates of any one of the preceding clauses; or a ceutical composition comprising a therapeutically effective amount of one or more of the conjugates of any one of the preceding clauses, optionally further comprising one or more carriers, diluents, or excipients, or a combination thereof.
A method for diagnosing and/or monitoring a e or disease state caused at least in part by a pathogenic cell population, such as a cancer or inflammatory disease, in a host animal, the method comprising the step of administering to the host animal a diagnostically effective amount of one or more of the conjugates of any one of the preceding clauses; or a pharmaceutical composition comprising a diagnostically effective amount of one or more of the conjugates of any one of the preceding clauses, optionally further comprising one or more carriers, diluents, or excipients, or a combination thereof.
A method for treating a disease or disease state caused at least in part by a pathogenic cell population, such as a cancer or atory disease, in a host animal, the method comprising the step of administering to the host animal a therapeutically effective amount of one or more of the conjugates of any one of the preceding clauses; or a pharmaceutical composition comprising a therapeutically effective amount of one or more of the conjugates of any one of the preceding s, optionally further comprising one or more carriers, diluents, or ents, or a combination thereof.
Use of one or more of the ates of any one of the preceding clauses; or a ceutical ition comprising one or more of the conjugates of any one of the preceding clauses, optionally further comprising one or more carriers, diluents, or excipients, or a combination thereof, in the manufacture of a medicament for diagnosing and/or monitoring a disease or disease state caused at least in part by a pathogenic cell tion, such as a cancer or inflammatory disease, in a host animal.
Use of one or more of the ates of any one of the preceding clauses; or a ceutical composition comprising one or more of the conjugates of any one of the preceding clauses, optionally further comprising one or more carriers, diluents, or excipients, or a combination thereof, in the manufacture of a medicament for treating a disease or e state caused at least in part by a pathogenic cell population, such as a cancer or inflammatory disease, in a host animal.
A kit comprising one or more of the conjugates of any one of the preceding clauses, or a pharmaceutical ition thereof, optionally further comprising one or more carriers, diluents, or excipients, or a combination thereof; an al solvent; an optional reaction container, and a set of instructions for preparing one or more radionuclides and ing the one or more radionuclides with the one or more of the conjugates to prepare an imaging agent, diagnostic agent, or therapeutic agent.
A kit comprising one or more of the conjugates of any one of the preceding s, or a pharmaceutical composition thereof, optionally further comprising one or more carriers, diluents, or excipients, or a combination thereof; an optional solvent; an optional reaction container, and a set of instructions for preparing one or more radionuclides and combining the one or more radionuclides with the one or more of the conjugates to prepare an imaging agent, diagnostic agent, or therapeutic agent.
It is to be understood that in each instance where a compound or al formula includes an atom or locus that is marked with or includes a (*), the (*) indicates that the compound or chemical formula is a radical having an open valence at that atom or locus, and that atom or locus is the location for attachment of another radical.
In another illustrative embodiment, the conjugate, composition, unit dose, method, use, or kit of any other embodiment described herein comprises a compound of formula or a derivative thereof comprising a chelated metal; or a radical of the foregoing, where each R is in each instance independently selected to form a carboxylic acid or salt thereof, ester, or amide, and R1, R2, and R3, are each independently ed from en, and alkyl, cycloalkyl, aryl, arylalkyl, heteroaryl, and heteroarylalkyl, each of which is optionally substituted.
In another illustrative embodiment, the conjugate, composition, unit dose, method, use, or kit of any other ment described herein comprises 1,4,7,10- tetraazacyclododecane-l,4,7,10-tetraacetic acid (DOTA) or a derivative thereof comprising a chelated metal; or a l of the ing.
In r illustrative embodiment, the conjugate, composition, unit dose, method, use, or kit of any other embodiment described herein comprises a compound of a R1< R2 ISM_\ Roc—/ or a derivative thereof comprising a chelated metal; or a radical of the ing, where each R is in each instance independently selected to form a carboxylic acid or salt thereof, ester, or amide, and R1, R2, and R3, are each independently selected from hydrogen, and alkyl, cycloalkyl, aryl, kyl, heteroaryl, and heteroarylalkyl, each of which is optionally substituted, such as the following illustrative compounds: R1 <COZH N/R/R2 H0204 R1 R2 : NH2 H * H20 n H * H20 H H O TNH2S * H20 ONH2 COZH * H2O * H20 W “j rCOZH : | N S * H20, * H20 EOZH H NH2 rCOZH T ,N S * H20 * H C W 2 COZH “WCOZH (COZH : N O * H20, * H20 EOZH COZH COZH f f * H20] W =\- H2C’ W COZH COZH HOchNAcozH rCOzH * HCl; * HZC’NW 002“ COZH or a carboxylic acid salt or carboxamide derivative (CONHz) thereof, or a radical of any of the ing; or a derivative thereof comprising a chelated metal.
In another illustrative embodiment, the conjugate, composition, unit dose, method, use, or kit of any other embodiment described herein comprises a compound of formula N/YR4 R5 HOZC—/ or a derivative thereof comprising a ed metal; or a radical of the foregoing, where R4 and R5 are selected from hydrogen, and alkyl, cycloalkyl, aryl, arylalkyl, heteroaryl, and heteroarylalkyl, each of which is optionally substituted, such as the following illustrative or a carboxylic acid salt or carboxamide derivative (CONHz) thereof, or a radical of any of the ing; or a derivative f comprising a chelated metal.
In another illustrative embodiment, the conjugate, composition, unit dose, method, use, or kit of any other embodiment described herein comprises a compound of formula * H2C-CO2H NODA—HA 0N02 * HC/VCOZH * H20 NODA—MPN * H2C N02 NODA—EPN NODA—MBA * H20 COZH NODA—MPAA NODA—EBA O O OH *HZCMNMN O H / * H20 0 NODA—MPH NODA—BAEM m”“20 mwgfiO / * H20 * H2C O NODA—MPAED NODA—MPAEM Q 0 0 \ flNA/NH o * H2C NODA—MBEM * HZCMN NODA-BM NODA-EA * H20\ NODA-butyne N=N\ COZH * Hzck/ * mNVOTVNa30 NODA-MPAPEG3N3 * HZC/\©\ 0 NODA-EPA-succinyl imozN *HZC N * H20 L—NETA ((R) or (S) or (RS)) or a carboxylic acid salt or carboxamide derivative ) thereof, or a radical of any of the foregoing; or a derivative thereof comprising a chelated metal.
In r illustrative embodiment, the conjugate, composition, unit dose, method, use, or kit of any other embodiment described herein comprises a compound selected from the formulae EN N+\)”_ HO E: N+\)”_ 02N E: NM Q_/ \) COZH Gd COZH 6Q COZH OH COZH COZH or a carboxylic acid salt or carboxamide derivative (CONHz) thereof, or a radical of any of the ing, where n is an integer selected from 1, 2, 3, 4, 5, or 6; or a derivative thereof comprising a chelated metal.
As used herein the term “radical” generally refers to an open valence compound or chemical fragment that results after the removal of a hydrogen atom or a yl group from a carboxylic acid. For example, the following radicals may be formed from L-NETA NAP“ * * 02C—/ *OzC J NH * where each (*) atom is an open e for attachment to a linker and/or targeting agent.
It is to be understood that the foregoing compounds and radicals thereof, may be further functionalized to attach reactive groups for the uent attachment of linkers and/or ing groups. Illustratively, the following reactive intermediates are described herein <C02H <COZH HOZC—/ES:+\_LW2—>HOZC—/CE:+LLW where n is 0 or 1, and NX is O O O 0 O O O N=C=S HNJJ\/SH HNJK/Ns HNJI\/ONH2 HNK b HNJJ\/\N / / o O and the like.
It is to be understood that the following compounds, and metal chelates thereof, are not ates of the invention: H020\| COZH N “1/deWMN\/\/\<(L;>VCOZH fiJK/[Nj/\N COZH H COzH O HN N\ N A l H \ / H2N N N HOZC 0 NH HN NH 0 o /—\ COH )/—NH M 2 NH HN4~<¥(\N/— H020 o o HN N 1 H020 Ph H020 H020 o o HO2C\—)>NH >"‘”/—\—NH HN (\N/—C02H NH HN n( N j Where n is 1 or 3.
The compounds described herein may contain one or more chiral centers, or may otherwise be capable of existing as multiple stereoisomers. It is to be understood that in one embodiment, the invention described herein is not limited to any particular sterochemical requirement, and that the compounds, and compositions, methods, uses, and medicaments that include them may be optically pure, or may be any of a variety of stereoisomeric mixtures, including racemic and other mixtures of omers, other mixtures of diastereomers, and the like. It is also to be understood that such es of stereoisomers may include a single stereochemical configuration at one or more chiral s, while ing mixtures of stereochemical configuration at one or more other chiral centers.
Similarly, the compounds described herein may include geometric centers, such as cis, trans, E, and Z double bonds. It is to be understood that in another embodiment, the invention bed herein is not limited to any particular geometric isomer ement, and that the compounds, and compositions, methods, uses, and medicaments that include them may be pure, or may be any of a variety of geometric isomer mixtures. It is also to be understood that such mixtures of ric isomers may include a single configuration at one or more double bonds, while including mixtures of geometry at one or more other double bonds.
As used herein, the term “alkyl” includes a chain of carbon atoms, which is optionally branched. As used herein, the terms “alkenyl” and “alkynyl” each include a chain of carbon atoms, which is optionally branched, and include at least one double bond or triple bond, respectively. It is to be understood that alkynyl may also include one or more double bonds. It is to be further understood that in certain embodiments, alkyl is advantageously of d length, including C1-Cz4, C1-C1z, C1-C8, C1-C6, and C1-C4. Illustratively, such particularly limited length alkyl , including C1-C8, C1-C6, and C1-C4 may be referred to as lower alkyl. It is to be further understood that in certain embodiments alkenyl and/or l may each be advantageously of limited length, including C2—Cz4, C2—C12, C2—C8, C2— C6, and C2-C4. Illustratively, such ularly limited length alkenyl and/or alkynyl groups, including C2-C8, C2-C6, and C2-C4 may be referred to as lower alkenyl and/or alkynyl. It is appreciated herein that r alkyl, alkenyl, and/or alkynyl groups may add less ilicity to the compound and accordingly will have different pharmacokinetic behavior.
In embodiments of the invention described herein, it is to be understood, in each case, that the tion of alkyl refers to alkyl as defined herein, and optionally lower alkyl. In embodiments of the invention described herein, it is to be understood, in each case, that the recitation of alkenyl refers to l as defined herein, and optionally lower alkenyl. In WO 73678 embodiments of the invention described herein, it is to be understood, in each case, that the tion of alkynyl refers to alkynyl as defined herein, and ally lower alkynyl.
Illustrative alkyl, alkenyl, and alkynyl groups are, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, yl, sec-butyl, tert—butyl, pentyl, yl, 3—pentyl, neopentyl, hexyl, heptyl, octyl, and the like, and the corresponding groups containing one or more double and/or triple bonds, or a combination thereof.
As used herein, the term ene” includes a divalent chain of carbon atoms, which is optionally ed. As used herein, the term “alkenylene” and “alkynylene” includes a divalent chain of carbon atoms, which is optionally branched, and includes at least one double bond or triple bond, respectively. It is to be understood that alkynylene may also include one or more double bonds. It is to be further understood that in certain embodiments, alkylene is ageously of limited length, including C1—C24, C1—C12, C1—C8, C1—C6, and C1- C4. Illustratively, such particularly limited length alkylene groups, ing C1-C8, C1-C6, and C1-C4 may be referred to as lower alkylene. It is to be further understood that in certain embodiments alkenylene and/or alkynylene may each be advantageously of limited length, including C2—Cz4, C2-C12, C2-C8, C2-C6, and C2-C4. Illustratively, such particularly d length alkenylene and/or lene groups, including C2-C8, C2—C6, and C2—C4 may be referred to as lower alkenylene and/or alkynylene. It is appreciated herein that shorter alkylene, alkenylene, and/or lene groups may add less lipophilicity to the compound and accordingly will have different pharmacokinetic behavior. In embodiments of the invention described herein, it is to be understood, in each case, that the recitation of alkylene, alkenylene, and alkynylene refers to alkylene, alkenylene, and alkynylene as defined herein, and optionally lower alkylene, alkenylene, and alkynylene. Illustrative alkyl groups are, but not limited to, methylene, ethylene, n-propylene, isopropylene, n-butylene, isobutylene, sec- butylene, pentylene, 1,2—penty1ene, 1,3-penty1ene, hexylene, heptylene, octylene, and the like.
As used herein, the term “linker” includes is a chain of atoms that connects two or more functional parts of a molecule to form a conjugate. Illustratively, the chain of atoms is selected from C, N, O, S, Si, and P, or C, N, O, S, and P, or C, N, O, and S. The chain of atoms ntly ts different functional capabilities of the conjugate, such as targeting agents, drugs, diagnostic agents, imaging agents, and the like. The linker may have a wide variety of lengths, such as in the range from about 2 to about 100 atoms in the contiguous backbone. The atoms used in forming the linker may be combined in all chemically relevant ways, such as chains of carbon atoms forming alkylene, alkenylene, and alkynylene groups, and the like; chains of carbon and oxygen atoms forming ethers, polyoxyalkylene groups, or when combined with yl groups forming esters and carbonates, and the like; chains of carbon and nitrogen atoms forming amines, imines, polyamines, hydrazines, hydrazones, or when combined with carbonyl groups forming amides, ureas, semicarbazides, carbazides, and the like; chains of carbon, nitrogen, and oxygen atoms forming alkoxyamines, lamines, or when combined with yl groups forming urethanes, amino acids, acyloxylamines, hydroxamic acids, and the like; and many others. In addition, it is to be understood that the atoms forming the chain in each of the foregoing illustrative embodiments may be either saturated or unsaturated, thus forming single, double, or triple bonds, such that for example, s, s, alkynes, imines, and the like may be radicals that are included in the linker. In addition, it is to be understood that the atoms forming the linker may also be cyclized upon each other or be part of cyclic structure to form divalent cyclic structures that form the linker, including cyclo alkanes, cyclic , cyclic amines, and other heterocycles, arylenes, arylenes, and the like in the linker. In this latter arrangement, it is to be tood that the linker length may be defined by any pathway through the one or more cyclic ures. Illustratively, the linker length is defined by the shortest pathway through the each one of the cyclic structures. It is to be understood that the linkers may be optionally substituted at any one or more of the open valences along the chain of atoms, such as optional substituents on any of the carbon, nitrogen, silicon, or phosphorus atoms. It is also to be tood that the linker may connect the two or more functional parts of a le to form a conjugate at any open valence, and it is not necessary that any of the two or more functional parts of a molecule forming the conjugate are attached at any apparent end of the linker.
As used herein, the term “cycloalkyl” includes a chain of carbon atoms, which is ally branched, where at least a portion of the chain in cyclic. It is to be understood that lkylalkyl is a subset of cycloalkyl. It is to be understood that cycloalkyl may be polycyclic. Illustrative cycloalkyl include, but are not limited to, cyclopropyl, cyclopentyl, cyclohexyl, 2-methylcyclopropyl, cyclopentyleth—2—yl, tyl, and the like. As used herein, the term “cycloalkenyl” includes a chain of carbon atoms, which is optionally branched, and includes at least one double bond, where at least a portion of the chain in cyclic. It is to be understood that the one or more double bonds may be in the cyclic portion of cycloalkenyl and/or the non-cyclic portion of cycloalkenyl. It is to be understood that cycloalkenylalkyl and cycloalkylalkenyl are each subsets of cycloalkenyl. It is to be understood that cycloalkyl may be polycyclic. Illustrative cycloalkenyl include, but are not limited to, cyclopentenyl, exylethen—Z-yl, cycloheptenylpropenyl, and the like. It is to be further understood that chain forming cycloalkyl and/or cycloalkenyl is advantageously of limited , including C3-C24, , C3—C8, C3—C6, and C5-C6. It is appreciated herein that shorter alkyl and/or alkenyl chains forming cycloalkyl and/or cycloalkenyl, tively, may add less ilicity to the compound and accordingly will have different cokinetic behavior.
As used herein, the term “heteroalkyl” includes a chain of atoms that includes both carbon and at least one heteroatom, and is optionally branched. Illustrative heteroatoms include nitrogen, oxygen, and sulfur. In certain variations, illustrative heteroatoms also include phosphorus, and selenium. As used herein, the term “cycloheteroalkyl” including cyclyl and heterocycle, includes a chain of atoms that includes both carbon and at least one atom, such as heteroalkyl, and is optionally branched, where at least a portion of the chain is cyclic. Illustrative heteroatoms include nitrogen, , and sulfur. In certain variations, illustrative heteroatoms also include phosphorus, and um. Illustrative cycloheteroalkyl include, but are not limited to, tetrahydrofuryl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, piperazinyl, homopiperazinyl, quinuclidinyl, and the like.
As used herein, the term “aryl” includes monocyclic and polycyclic aromatic carbocyclic groups, each of which may be optionally substituted. Illustrative aromatic carbocyclic groups described herein e, but are not d to, phenyl, naphthyl, and the like. As used , the term “heteroaryl” includes aromatic heterocyclic groups, each of which may be optionally substituted. Illustrative aromatic heterocyclic groups include, but are not limited to, pyridinyl, dinyl, pyrazinyl, triazinyl, tetrazinyl, inyl, quinazolinyl, quinoxalinyl, thienyl, pyrazolyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, triazolyl, benzimidazolyl, benzoxazolyl, benzthiazolyl, benzisoxazolyl, benzisothiazolyl, and the like.
The term "optionally substituted" as used herein includes the replacement of hydrogen atoms with other onal groups on the radical that is optionally substituted.
Such other functional groups illustratively include, but are not limited to, amino, hydroxyl, halo, thiol, alkyl, kyl, heteroalkyl, aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl, heteroarylheteroalkyl, nitro, sulfonic acids and derivatives thereof, carboxylic acids and derivatives thereof, and the like. Illustratively, any of amino, hydroxyl, thiol, alkyl, haloalkyl, heteroalkyl, aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl, heteroarylheteroalkyl, andfor sulfonic acid is optionally substituted.
As used herein, the terms nally tuted aryl" and "optionally substituted heteroaryl" include the replacement of hydrogen atoms with other functional groups on the aryl or heteroaryl that is optionally substituted. Such other functional groups, also referred to herein as aryl subsituents, illustratively include, but are not limited to, amino, hydroxy, halo, thio, alkyl, haloalkyl, heteroalkyl, aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl, heteroarylheteroalkyl, nitro, ic acids and derivatives thereof, carboxylic acids and derivatives thereof, and the like. ratively, any of amino, hydroxy, thio, alkyl, haloalkyl, heteroalkyl, aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl, heteroarylheteroalkyl, and/or sulfonic acid is optionally substituted.
Illustrative substituents include, but are not limited to, a radical -(CH2)XZX, where X is an integer from 0-6 and ZX is selected from halogen, hydroxy, yloxy, including C1-C6 alkanoyloxy, ally substituted aroyloxy, alkyl, including C1—C6 alkyl, alkoxy, including C1-C6 alkoxy, cycloalkyl, including C3-C3 cycloalkyl, cycloalkoxy, including C3-C8 cycloalkoxy, alkenyl, including C2-C6 alkenyl, alkynyl, including C2-C6 l, haloalkyl, including C1-C6 haloalkyl, haloalkoxy, including C1-C6 haloalkoxy, halocycloalkyl, including C3-C8 halocycloalkyl, halocycloalkoxy, including C3-C8 halocycloalkoxy, amino, C1—C6 alkylamino, (C1—C6 alkyl)(C1-C6 alkyl)amino, alkylcarbonylamino, N—(C1—C6 alkyl)alkylcarbonylamino, aminoalkyl, C1—C6 alkylaminoalkyl, (C1-C6 alkyl)(C1—C6 alkyl)aminoalkyl, alkylcarbonylaminoalkyl, N—(C1-C6 alkyl)alkylcarbonylaminoalkyl, cyano, and nitro; or ZX is selected from -C02R4 and -CONR5R6, where R4, R5, and R6 are each independently selected in each occurrence from hydrogen, C1-C6 alkyl, aryl-C1-C6 alkyl, and heteroaryl-Cl—Cg alkyl.
It is to be tood that in every instance disclosed herein, the recitation of a range of integers for any le bes the d range, every individual member in the range, and every possible subrange for that variable. For example, the recitation that n is an integer from 0 to 8, describes that range, the individual and selectable values of 0, 1, 2, 3, 4, , 6, 7, and 8, such as n is 0, or n is l, or n is 2, etc. In addition, the recitation that n is an integer from 0 to 8 also describes each and every subrange, each of which may for the basis of a further embodiment, such as n is an integer from 1 to 8, from 1 to 7, from 1 to 6, from 2 to 8, from 2 to 7, from 1 to 3, from 2 to 4, etc.
As used herein, the term “composition” lly refers to any product comprising the specified ingredients in the specified s, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified s. It is to be understood that the compositions bed herein may be prepared from isolated compounds described herein or from salts, solutions, hydrates, solvates, and other forms of the compounds described herein. It is appreciated that n functional groups, such as the y, amino, and like groups form complexes and/or coordination compounds with water and/or various solvents, in the various physical forms of the compounds. It is also to be understood that the itions may be prepared from various amorphous, non- amorphous, lly lline, crystalline, and/or other morphological forms of the compounds bed herein. It is also to be understood that the compositions may be prepared from s hydrates and/or solvates of the compounds described herein.
Accordingly, such pharmaceutical compositions that recite compounds described herein are to be tood to e each of, or any ation of, the various morphological forms and/or solvate or hydrate forms of the compounds described herein.
Illustratively, compositions may include one or more carriers, diluents, and/or excipients. The compounds described herein, or compositions containing them, may be formulated in a diagnostically or therapeutically effective amount in any conventional dosage forms appropriate for the methods described herein. The compounds described herein, or compositions containing them, including such ations, may be administered by a wide variety of conventional routes for the methods described herein, and in a wide variety of dosage formats, utilizing known procedures (see generally, Remington: The e and Practice of Pharmacy, (21St ed., 2005)).
The term “diagnostically effective amount” as used herein, refers to that amount of active compound or ceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes diagnosis and/or monitoring of the symptoms of the disease or disorder being treated. Illustrative diagnostically effective amounts of the conjugate to be administered to the host animal include about 1 pg/kg to about mg/kg, 1 ng/kg to about 10 mg/kg, or from about 10 pig/kg to about 1 mg/kg, or from about 100 ug/kg to about 500 ug/kg.
The term “therapeutically effective amount” as used herein, refers to that amount of active compound or pharmaceutical agent that s the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated. In one aspect, the therapeutically effective amount is that which may treat or alleviate the e or symptoms of the disease at a reasonable t/risk ratio applicable to any medical treatment. However, it is to be understood that the total daily usage of the compounds and compositions bed herein may be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically-effective dose level for any particular patient will depend upon a variety of factors, including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, gender and diet of the patient: the time of administration, route of administration, and rate of ion of the ic compound employed; the duration of the ent; drugs used in combination or coincidentally with the specific compound ed; and like factors well known to the researcher, veterinarian, medical doctor or other ian of ordinary skill. Illustrative therapeutically effective amounts of the conjugate to be stered to the host animal include about 1 pg/kg to about 10 mg/kg, 1 ng/kg to about mg/kg, or from about 10 pig/kg to about 1 mg/kg, or from about 100 ug/kg to about 500 rig/kg.
The term “administering” as used herein includes all means of introducing the compounds and compositions described herein to the host animal, including, but are not d to, oral (po), intravenous (iv), intramuscular (im), subcutaneous (sc), transdermal, inhalation, buccal, ocular, sublingual, vaginal, rectal, and the like. The compounds and compositions described herein may be administered in unit dosage forms and/or formulations containing conventional nontoxic pharmaceutically—acceptable carriers, nts, and/or vehicles.
As used herein, the term “amino acid” refers generally to beta, gamma, and longer amino acids, such as amino acids of the a: -N(R)-(CR’R")q-C(O)- where R is hydrogen, alkyl, acyl, or a suitable nitrogen protecting group, R’ and R" are hydrogen or a substituent, each of which is independently selected in each occurrence, and q is an integer such as l, 2, 3, 4, or 5. Illustratively, R’ and/or R" independently correspond to, but are not limited to, hydrogen or the side chains t on naturally occurring amino acids, such as methyl, benzyl, ymethyl, thiomethyl, carboxyl, carboxylmethyl, guanidinopropyl, and the like, and derivatives and protected derivatives thereof. The above described a includes all stereoisomeric variations. For example, the amino acid may be ed from alanine, aspartic acid, asparagine, cysteine, glutamic acid, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, glutamine, arginine, , threonine, valine, tryptophan, tyrosine, and ornithine, and the like.
It is to be understood that in every instance disclosed herein, the recitation of a range of integers for any variable describes the recited range, every individual member in the range, and every possible subrange for that variable. For example, the recitation that n is an integer from 0 to 8, describes that range, the individual and selectable values of 0, 1, 2, 3, 4, , 6, 7, and 8, such as n is 0, or n is 1, or n is 2, etc. In addition, the recitation that n is an r from 0 to 8 also describes each and every subrange, each of which may for the basis of a further embodiment, such as n is an integer from 1 to 8, from 1 to 7, from 1 to 6, from 2 to 8, from 2 to 7, from 1 to 3, from 2 to 4, etc.
In another embodiment, the linkers described herein include a polyether, such as the linkers of the following formulae: O]/\/O\/Y N'eO‘PAofVOVYmono]/\/o 0 H02HZnCt.)O p: aer/H“”3?"o erwvw\ '8 )” H020 HOZC H020 ,//N\/”\O/\\/O\//\O/\\/O\/”\O/\~/O+//\oi;\/O\//\n/N\r'\S/” where m is an integer independently selected in each instance from 1 to about 8; p is an integer selected from 1 to about 10; and n is an integer independently selected in each instance from 1 to about 3. In one aspect, In is independently in each instance 1 to about 3.
In another aspect, n is 1 in each instance. In r aspect, p is independently in each instance about 4 to about 6. ratively, the corresponding polypropylene polyethers corresponding to the foregoing are described herein and may be ed in the conjugates as linkers. In addition, it is appreciated that mixed polyethylene and polypropylene polyethers may be included in the conjugates as linkers. Further, cyclic variations of the ing polyether compounds, such as those that include tetrahydrofuranyl, 1,3-dioxanes, 1,4- dioxanes, and the like are bed herein.
In another embodiment, the linkers described herein include a plurality of hydroxyl functional groups, such as linkers that incorporate ccharides, oligosaccharides, polysaccharides, and the like. It is to be understood that the polyhydroxyl containing linkers comprise a plurality of —(CROH)- groups, where R is hydrogen or alkyl.
In another ment, the s include one or more of the ing diradicals: wherein R is H, alkyl, cycloalkyl, or kyl; m is an integer from 1 to about 3; n1 is an integer from 1 to about 5, or n1 is an integer from 2 to about 5, p is an r from 1 to about , and r is an integer selected from 1 to about 3. In one aspect, the r n is 3 or 4. In another aspect, the integer p is 3 or 4. In another aspect, the integer r is 1.
In r embodiment, the linkers include one or more of the following diradicals: O 902H / * H 3 H020 S\ * o QOZH * N N H ? H mm N * N NAC/Irs\ * H H (HOCIJH)n O (Hoc|:H)n (HOCI3H)n R R P wherein R is H, alkyl, cycloalkyl, or arylalkyl; m is an integer from 1 to about 3; n is an integer from 1 to about 5, or from 2 to about 5, p is an integer from 1 to about 5, and r is an integer selected from 1 to about 3. In one , the integer n is 3 or 4. In another aspect, the integer p is 3 or 4. In another aspect, the integer r is 1.
In another embodiment, the linker includes one or more of the following cyclic polyhydroxyl groups: O O H / * a: Z]: / * at u N ['0l4). HO (OH)n HO OH ,3 p wherein n is an integer from 2 to about 5, p is an integer from 1 to about 5, and each r is an independently selected integer from 1 to about 4. In one aspect, the integer n is 3 or 4. In another aspect, the integer p is 3 or 4. In another aspect, each r r is independently 2 or 3. It is understood that all stereochemical forms of such sections of the linkers are described herein. For example, in the above formula, the n may be derived from ribose, xylose, glucose, mannose, galactose, or other sugar and retain the stereochemical arrangements of pendant hydroxyl and alkyl groups present on those molecules. In addition, it is to be tood that in the foregoing formulae, various deoxy compounds are also described.
Illustratively, compounds of the ing ae are described: wherein n is equal to or less than r, such as when r is 2 or 3, n is 1 or 2, or 1, 2, or 3, respectively.
In r embodiment, the linker includes a polyhydroxyl compound of the following formula: wherein n and r are each an integer selected from 1 to about 3. In one aspect, the linker includes one or more polyhydroxyl compounds of the following formulae: 0:3—TOH d/ D/OH H OA/OVOH \o/*/\/O/\/LOH HO / N OH HN/ ‘NH KKL / HO/kKLOH ‘ OH HN\ , OH It is understood that all stereochemical forms of such sections of the linkers are described herein. For example, in the above a, the section may be derived from ribose, xylose, glucose, mannose, galactose, or other sugar and retain the stereochemical arrangements of pendant hydroxyl and alkyl groups present on those molecules.
In another configuration, the linkers L described herein include polyhydroxyl groups that are spaced away from the backbone of the linker. In one embodiment, such carbohydrate groups or polyhydroxyl groups are connected to the back bone by a triazole group, forming triazole-linked linkers. Illustratively, such linkers e diradicals of the following formulae: OH OH OH OH HO HO HO HO HO “‘h‘rhfl” COzHO HO 02 J]rm) wherein n, m, and r are rs and are each independently selected in each ce from 1 to about 5. In one illustrative aspect, m is independently 2 or 3 in each instance. In another aspect, r is l in each instance. In another aspect, n is l in each instance. In one variation, the group connecting the polyhydroxyl group to the backbone of the linker is a different heteroaryl group, including but not limited to, e, pyrazole, l,2,4-triazole, furan, oxazole, isoxazole, thienyl, thiazole, azole, oxadiazole, and the like. Similarly, divalent 6—membered ring heteroaryl groups are described. Other variations of the foregoing illustrative linkers include ylene groups, such as the following formulae: OH OH OH OH HO HO (N?/* (N? N',.
COzHO HO 52 o wherein n and r are integers and are each independently selected in each instance from 1 to about 5; and p is an integer selected from 1 to about 4.
In another embodiment, such carbohydrate groups or droxyl groups are connected to the back bone by an amide group, forming amide-linked linkers. Illustratively, such linkers include diradicals of the following formulae: HO HO OH HO HO COZH Hofi o 0 ”‘(KFO "‘(KFO m ( O NH NH \ *\s "(fifi/ * \S S H A? H \(E)n H n( bj/ \n/\H N /* \n/\H/* N n(N]/N N “Lin/NV /*NH COZHO COzHO COZHO wherein each n is an independently selected r from 1 to about 3, and m is an independently selected integer from 1 to about 22. In one illustrative aspect, each n is independently l or 2. In another illustrative aspect, m is selected from about 6 to about 10, illustratively 8. In one variation, the group connecting the polyhydroxyl group to the backbone of the linker is a different functional group, including but not limited to, esters, ureas, carbamates, drazones, and the like. Similarly, cyclic variations are bed.
Other variations of the foregoing illustrative linkers include oxyalkylene groups, such as the following formulae: HO\%HO 1&0COZH 140%:HO H0 HO NH [le * \s nH’ "(j/NH [HIif MY1m” MW” "erW” COZH O C02H O COZHO wherein n is in each instance an independently selected integer from 1 to about 5; and p is an integer selected from 1 to about 4.
In another embodiment, the linkers include one or more of the ing diradicals: wherein R is H, alkyl, cycloalkyl, or arylalkyl; each m is an ndently selected integer from 1 to about 3; each n is an independently selected integer from 1 to about 6, p is an integer from 1 to about 5, and r is an integer selected from 1 to about 3. In one variation, each n is independently 3 or 4. In another variation, the integer p is 3 or 4. In another variation, the integer r is 1.
In another embodiment, the linkers include one or more of the following diradicals: wherein R is H, alkyl, lkyl, or arylalkyl; each m is an independently selected r from 1 to about 3; each n is an independently selected integer from 2 to about 6, p is an integer from 1 to about 5, and r is an integer selected from 1 to about 3. In one variation, each n is independently 3 or 4. In another variation, the integer p is 3 or 4. In another ion, the integer r is 1.
In another embodiment, the linkers include one or more of the following diradicals: H P N,, N\rj\ N/ * H (H20)m m HN 0 kb HO" OH 'IOH ‘/)/n I‘OH COZHp H 002H NM(‘10ng N H . N r\* (H2C)m A p HN o HMOH H (£0sz H HOC k 0 $OZH 72 EH * of 002H H EMS" N \ *——N N * H )m (Hz/Zr" p HN O kfi/YnOH OH COZHp , wherein each m is an independently selected integer from 1 to about 3; each n is an ndently selected integer from 1 to about 6, p is an integer from 1 to about 5, and r is an integer selected from 1 to about 3. In one variation, each n is independently 3 or 4. In another ion, the integer p is 3 or 4. In another variation, the integer r is 1.
In another embodiment, the s e one or more of the following diradicals: O asco H 2 H _ )m H 002H * N l r N * ifN H NW3“r H H (H C) o HN O HN o m gm“ cozH P cozflp _ wherein each m is an independently selected integer from 1 to about 3; each n is an independently selected integer from 2 to about 6, p is an integer from 1 to about 5, and r is an integer selected from 1 to about 3. In one variation, each n is independently 3 or 4. In another variation, the integer p is 3 or 4. In another variation, the integer r is 1.
In another embodiment, the linkers include one or more of the following diradicals: wherein each m is an independently selected integer from 1 to about 3, p is an integer from 1 to about 5, and r is an integer selected from 1 to about 3. In another variation, the integer p is 3 or 4. In another variation, the integer r is 1.
In another ment, the linker is a ation of backbone and ing side motifs such as is rated by the following formulae H H H N HO H0 HO 902 H >x</ */N */N ) 0 ) o o “0 ) ”O * ) HM“ fir“N/‘jlhl 0 0 n 0 Ho Ho Ho wherein n is an integer independently selected in each instance from 0 to about 3. The above formula are intended to represent 4, 5, 6, and even larger membered cyclic sugars. In addition, it is to be understood that the above formula may be ed to represent deoxy sugars, where one or more of the hydroxy groups present on the formulae are replaced by hydrogen, alkyl, or amino. In addition, it is to be tood that the corresponding carbonyl compounds are described by the above formulae, where one or more of the hydroxyl groups is oxidized to the corresponding carbonyl. In addition, in this illustrative embodiment, the pyranose includes both carboxyl and amino functional groups and (a) can be inserted into the ne and (b) can provide synthetic handles for branching side chains in variations of this embodiment. Any of the pendant hydroxyl groups may be used to attach other al radicals, including additional sugars to prepare the corresponding oligosaccharides. Other variations of this embodiment are also described, including inserting the pyranose or other sugar into the backbone at a single carbon, i.e. a spiro arrangement, at a geminal pair of carbons, and like arrangements. For example, one or two ends of the linker, or the agent P, or the ligand B may be ted to the sugar to be inserted into the backbone in a 1,1; 1,2; 1,3; 1,4; 2,3, or other arrangement.
In another embodiment, the linkers include one or more amino groups of the ing formulae: (EDOZHO NW, co H NW 90 H , 2 * 0 NWTWN : ' \NAMENd o * Nd \N *\ NJ 0 0 O n NAM’n N n NAM? COZH H H H H H H H H H H co H co H 2° * 20 “ ’\ NdNWNTW”O O *\ m o NdNWNTW”O O O n ”M COZH COZH m/\[,,Hkn ”M COZH COZH COQH where each n is an integer independently selected in each instance from 1 to about 3. In one aspect, the each n is independently l or 2 in each instance. In another aspect, the integer n is l in each instance.
In r embodiment, the linker is a ic acid ester, such as an alkyl ester of sulfuric acid. Illustratively, the linker is of the following formula: \/0 HO x/s’ \//o HO\ 0 o \o 0¢s\ /S// g o o/ \o >n M ()n O O HOQC 0 H020 HO n HO\ /0 HO és’ \éo HO\ 0 o \o O¢s\ /s/’ o o/ ‘o >n N ()n I ‘ N‘ ‘ //N | ,/N I o” *\ (— N “(K/“YR”n n( WHWN “Tl/k /* H025 H H220 )n H 0 O o HOQC where each n is an integer independently selected in each instance from 1 to about 3.
Illustratively, each n is independently l or 2 in each ce.
It is understood, that in such polyhydroxyl, polyamino, carboxylic acid, sulfuric acid, and like s that include free hydrogens bound to heteroatoms, one or more of those free hydrogen atoms may be protected with the appropriate hydroxyl, amino, or acid protecting group, respectively, or alternatively may be blocked as the corresponding pro- drugs, the latter of which are selected for the particular use, such as pro-drugs that release the parent drug under general or specific logical conditions.
It is to be understood that in each of the foregoing illustrative examples, the stereochemical configurations shown herein are merely illustrative, and other stereochemical configurations are described. For example in one variation, the corresponding unnatural amino acid urations may be included in the conjugated described herein as follows: H O CO2H AVIS\ T'IOZC N\ o H COZH * N * ‘ r N H ”7N \ /rs\ *r *‘O H H 7\ )r Fol4)r (OH)n p (OH)n , , p 7 _HO C Hozc\E 2 \: o o H H COZH 90sz , ”AWN N rs“ ,,7N/\WN\ NM \, H H H H o 0 qo F0 )r I” (OH): p (OH): p , wherein each n is an ndently ed integer from 2 to about 5, p is an integer from 1 to about 5, and r is an integer from 1 to about 4, as described above.
It is to be further understood that in the foregoing embodiments, open positions, such as (*) atoms are locations for attachment of the targeting agent B or the agent (P). In addition, it is to be understood that such attachment of either or both of B and A may be direct or through an intervening linker. Illustrative additional linkers are described in US. 7,601,332, the disclosure of which is incorporated herein by reference.
Illustrative bivalent radicals forming part of the linker.
H2N NH CO2H HN HOZC O COZH *V*I * K/\>x< | *K/S\N* COZH | f) * OR O gigN *g >z< | | >I< O O O R=H, alkyl, acyl Hozo/jv/Yo I /o o * o o N/Y VN 00 H >x< HO2C 2 >x< OR >I< * OR o |::N’\ * t 8* o R=H, alkyl, acyl NH2 0 COZH COZH * S «k N * ‘ >k * 4‘ l\/S * I >1: 0 O H COZH HO CAN/YO2 HO fO2 J NH J NVCOZH NH N H020 QI/ H020 g H N * * o *N * s * | l * N O O I * O O I I CI) /0 o O O * * o o H * N N co H OR N OR N * N H OR OR o I*| O S >x< O R=H, alkyl» acyl * N R=H, alkyl, acyl COzH COzH H020 O HO:L‘k20 N* I * >l< K/“\N* * >1< N * N Ma:I | COZH H020 H020 o O N as >k N * égN OR * N * N * * O O >x< O R=H, alkyl, acyl O H2NYNH HZNYNH HN HN * N >l< N>l< O * N | o O NH2 NH2 ee >z< N >l< >I< N * * I *N 1I o o O SH SH /0 o N * COZH N >1: * * OR *NX/8* N * >x< | OR o O R=H, alkyl, acyl o o o o o o , * * S S “ N% * N% * N3*n NJn O O O O n=O-3 n= -3 n=1-3 n=1-3 O O 0 9: /\/O it * S Nit *S o :1: N * It is to be understood that the bivalent linkers may be combined in any chemically relevant way, either directly or via an intervening heteroatom to construct the linkers described herein.
In another embodiment, the polyvalent linkers described herein comprise a linker selected from the group consisting of carbonyl, carbonyl, ne, cycloalkylene, necycloalkyl, alkylenecarbonyl, cycloalkylenecarbonyl, carbonylalkylcarbonyl, l alkylenesuccinimidyl, 1 (carbonylalkyl)succinimidy1, alkylenesulfoxyl, sulfonylalkyl, alkylenesulfoxylalkyl, alkylenesulfonylalkyl, carbonyltetrahydro—ZH—pyranyl, yltetrahydrofuranyl, 1—(carbonyltetrahydro-2H— l)succinimidyl, and l-(carbonyltetrahydrofuranyl)succinimidyl.
In r embodiment, the nds described herein comprise one or more amino acids.
The compounds described herein can be used for both human clinical medicine and veterinary applications. Thus, the host animal harboring the population of pathogenic cells and administered the compounds described herein can be human or, in the case of veterinary applications, can be a laboratory, agricultural, domestic, or Wild animal.
The present ion can be applied to host animals including, but not d to, humans, laboratory animals such rodents (e.g., mice, rats, hamsters, etc.), rabbits, monkeys, chimpanzees, ic animals such as dogs, cats, and rabbits, agricultural animals such as cows, horses, pigs, sheep, goats, and Wild animals in captivity such as bears, pandas, lions, tigers, leopards, elephants, zebras, giraffes, gorillas, dolphins, and whales.
The compounds, compositions, s, and uses described herein are useful for diagnosing and/or monitoring diseases caused at least in part by populations of pathogenic cells, which may cause a variety of pathologies in host animals. As used herein, the term “pathogenic cells” or “population of pathogenic cells” gemerally refers to cancer cells, infectious agents such as bacteria and viruses, bacteria- or virus-infected cells, inflammatory cells, activated macrophages capable of causing a disease state, and any other type of pathogenic cells that uniquely express, preferentially express, or overexpress g sites for the targeting agents described herein.
Illustratively, the population of pathogenic cells can be a cancer cell population that is tumorigenic, ing benign tumors and malignant tumors, or it can be non-tumorigenic. The cancer cell population can arise spontaneously or by such processes as mutations present in the germline of the host animal or somatic mutations, or it can be chemically-, virally-, or ion—induced. The invention can be utilized to diagnose, monitor, and/or treat such cancers, ing carcinomas, sarcomas, lymphomas, Hodgekin’s disease, melanomas, mesotheliomas, Burkitt’s lymphoma, nasopharyngeal carcinomas, leukemias, and myelomas. The cancer cell population can include, but is not limited to, oral, d, endocrine, skin, gastric, esophageal, laryngeal, pancreatic, colon, bladder, bone, ovarian, cervical, uterine, breast, testicular, prostate, rectal, kidney, liver, and lung cancers.
Illustratively, the population of pathogenic cells can also be activated monocytes or macrophages associated with disease states such as fibromyalgia, rheumatoid arthritis, osteoarthritis, ulcerative colitis, Crohn’s disease, psoriasis, osteomyelitis, multiple sclerosis, atherosclerosis, pulmonary fibrosis, dosis, systemic sclerosis, organ lant rejection (GVHD), lupus erythematosus, SjOgren’s syndrome, glomerulonephritis, inflammations of the skin, such as psoriasis, and the like, chronic inflammations, and inflammations due to injury, such as head or spinal cord injury, embolisms, and the like.
The conjugates described herein can be formed from, for example, a wide variety of vitamins or receptor-binding n analogs/derivatives, linkers, and g and radiotherapy agents. The conjugates described herein are capable of selectively targeting a tion of pathogenic cells in the host animal due to preferential expression of a receptor for the targeting agent, such as a n, accessible for binding, on the pathogenic cells.
Illustrative vitamin moieties that can be used as the ing agent (B) include carnitine, inositol, lipoic acid, xal, ascorbic acid, niacin, henic acid, folic acid, riboflavin, thiamine, biotin, n B12, and the lipid soluble vitamins A, D, E and K. These vitamins, and their receptor—binding analogs and derivatives, constitute an illustrative targeting entity that can be coupled with the imaging or radiotherapy agent by a bivalent linker (L) to form a targeting agent (B) imaging or radiotherapy agent ate as described herein. The term vitamin is understood to include vitamin s and/or derivatives, unless otherwise indicated. Illustratively, pteroic acid which is a derivative of , biotin analogs such as biocytin, biotin sulfoxide, tin and other biotin receptor-binding compounds, and the like, are considered to be vitamins, n analogs, and vitamin derivatives. It should be appreciated that n analogs or derivatives as described herein refer to vitamins that incorporates an heteroatom through which the vitamin analog or derivative is covalently bound to the bivalent linker (L).
Illustrative vitamin moieties include folic acid, biotin, riboflavin, thiamine, vitamin B12, and receptor—binding analogs and derivatives of these vitamin molecules, and other related vitamin receptor binding molecules.
In one embodiment, the targeting group B is a folate, an analog of folate, or a derivative of folate. It is to be understood as used herein, that the term folate is used both individually and collectively to refer to folic acid itself, and/or to such analogs and derivatives of folic acid that are capable of binding to folate receptors.
Illustrative embodiments of vitamin analogs and/or derivatives include folate and analogs and derivatives of folate such as folinic acid, pteropolyglutamic acid, and folate receptor-binding pteridines such as tetrahydropterins, ofolates, tetrahydrofolates, and their deaza and a analogs. The terms “deaza” and “dideaza” analogs refer to the art- recognized analogs having a carbon atom tuted for one or two nitrogen atoms in the naturally occurring folic acid structure, or analog or derivative thereof. For example, the deaza s include the a, 3-deaza, 5-deaza, 8-deaza, and za analogs of folate, folinic acid, pteropolyglutamic acid, and folate receptor-binding ines such as tetrahydropterins, dihydrofolates, and tetrahydrofolates. The dideaza analogs include, for example, 1,5-dideaza, 5,10-dideaza, 8,10-dideaza, and 5,8-dideaza analogs of folate, folinic acid, pteropolyglutamic acid, and folate receptor-binding pteridines such as tetrahydropterins, dihydrofolates, and tetrahydrofolates. Other folates useful as complex forming s for this invention are the folate receptor-binding analogs aminopterin, pterin (also known as methotrexate), NIO-methylfolate, 2-deamino-hydroxyfolate, deaza analogs such as l- deazamethopterin or 3-deazamethopterin, and 3’,5’—dichloroaminodeoxy-N10- methylpteroylglutamic acid (dichloromethotrexate). The foregoing folic acid analogs and/or derivatives are tionally termed es,” reflecting their ability to bind with folate- receptors, and such ligands when conjugated with exogenous molecules are effective to enhance transmembrane transport, such as via folate—mediated endocytosis as described herein.
Additional analogs of folic acid that bind to folic acid receptors are described in US Patent Application ation Serial Nos. 2005/0227985 and 2004/0242582, the disclosures of which are incorporated herein by reference. Illustratively, radicals of such folate analogs have the general formula: R6 R7 R6 R7 (A2>‘r‘”n\, AW‘16 wherein X and Y are each—independently selected from the group consisting of halo, R2, 0R2, SR3, and NR4R5; U, V, and W represent divalent moieties each independently selected from the group consisting of (R6a)C=, N=, (R6a)C(R7a), and N(R4a); Q is selected from the group consisting of C and CH; T is selected from the group consisting of S, O, N, NH, and —C=C-; A1 and A2 are each independently selected from the group consisting of oxygen, sulfur, C(Z), C(Z)O, OC(Z), N(R4"), C(Z)N(R4b), N(R4b)C(Z), OC(Z)N(R4"), C(Z)O, N(R4b)C(Z)N(R5b), S(O), S(O)2, N(R4a)S(O)2, C(R6b)(R7b), N(CECH), N(CH2CECH), C1—C12 alkylene, and C1—C12 alkyeneoxy, where Z is oxygen or sulfur; R1 is ed-from the group consisting of hydrogen, halo, C1-C12 alkyl, and C1—C12 alkoxy; R2, R3, R4, R4a, R4b, R5, RS", R6b, and R7b are each independently selected from the group consisting of en, halo, C1-C12 alkyl, C1-C12 alkoxy, C1-C12 yl, C1-C1; l, C1-C12 alkynyl, ; alkoxy)carbonyl, and (C1-C12 alkylamino)carbony1; R6 and R7 are each independently selected from the group ting of hydrogen, halo, C1-C12 alkyl, and C1-C12 alkoxy; or, R6 and R7 are taken together to form a carbonyl group; R621 and R721 are each independently selected from the group consisting of hydrogen, halo, C1-C12 alkyl, and C1-C12 alkoxy; or R621 and R721 are taken together to form a carbonyl group; L is one or more, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, amino acids; and n, p, r, s and t are each independently either 0 or 1.
As used herein, it is to be understood that the term folate refers both individually to folic acid used in forming a conjugate, or alternatively to a folate analog or derivative thereof that is capable of binding to folate or folic acid receptors.
In r embodiment, the targeting group is a PSMA ligand or inhibitor, such as a derivative of pentanedioic acid of the formula: H020 X n X is RP(O)(OH)CH2— (US. 5,968,915); RP(O)(OH)N(R1)— (US. 5,863,536); RP(O)(OH)O— (US. 877); RN(OH)C(O)Y— or RC(O)NH(OH)Y, wherein Y is —CR1R2—, —NR3— or —0— (US. 5,962,521); RS(O)Y, RSOzY, or RS(O)(NH)Y, wherein Y is —CR1R2—, —NR3— or —0- (US. 5,902,817); and RS-alkyl, wherein R is for example hydrogen, alkyl, aryl, or arylalkyl, each of which may be optionally substituted (J. Med. Chem. 46:1989—1996 (2003)).
In each of the foregoing formulae, R, R1, R2, and R3 are each independently selected from hydrogen, C1-C9 straight or branched chain alkyl, C2—C9 straight or branched chain alkenyl, C3-C8 cycloalkyl, C5—C7 lkenyl, and aryl. In addition, in each case, each of R, R1, R2, and R3 may be optionally substituted, such as with one or more groups selected from C3-C8 cycloalkyl, C5—C7 lkenyl, halo, hydroxy, nitro, trifluoromethyl, C1-C6 straight or branched chain alkyl, C2—C6 straight or branched chain alkenyl, C1-C4 alkoxy, C2—C4 alkenyloxy, phenoxy, benzyloxy, amino, aryl. In one , aryl is selected from 1—naphthyl, 2-naphthyl, lyl, 3—indolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2—pyridyl, 3—pyridyl, 4-pyridyl, benzyl, and phenyl, and in each case aryl may be optionally substituted with one or more, illustratively with one to three, groups selected from halo, hydroxy, nitro, trifluoromethyl, C1-C6 straight or branched chain alkyl, C2-C6 ht or branched chain alkenyl, C1-C4 alkoxy, C2-C4 alkenyloxy, phenoxy, benzyloxy, and amino. In one ion of each of the above formulae, R is not hydrogen.
Illustrative PSMA ligands (US. 5,968,915) include 2—[[methylhydroxyphosphinyl]methyl]pentanedioic acid; 2—[[ethylhydroxyphosphinyl]methyl]pentanedioic acid; 2—[[propylhydroxyphosphinyl]methyl]pentanedioic acid; 2—[[butylhydroxyphosphinyl]methyl]pentanedioic acid; 2—[[cyclohexylhydroxyphosphinyl]methyl]pentanedioic acid; 2—[[phenylhydroxyphosphinyl]methyl]pentanedioic acid; 2—[[2-(tetrahydrofuranyl)hydroxyphosphinyl]methyl] pentanedioic acid; 2—[[(2-tetrahydropyranyl)hydroxyphosphinyl]methyl] edioic acid; 2—[[((4-pyridyl)methyl)hydroxyphosphinyl]methyl] pentanedioic acid; 2—[[((2-pyridyl)methyl)hydroxyphosphinyl]methyl] pentanedioic acid; 2—[[(phenylmethy1)hydroxyphosphinyl]methyl] pentanedioic acid; 2—[[((2-phenylethyl)methyl)hydroxyphosphinyl]methyl] pentanedioic acid; 3-phenylpropyl)methyl)hydroxyphosphinyl]methyl] pentanedioic acid; 2-[[((3-phenylbutyl)methy1)hydroxyphosphinyl]methyl] pentanedioic acid; 2-phenylbutyl)methyl)hydroxyphosphinyl]methyl] pentanedioic acid; 2-[[(4-phenylbuty1)hydroxyphosphinyl]methyl]pentanedioic acid; and 2—[[(aminomethyl)hydroxyphosphinyl]methyl]pentanedioic acid.
Illustrative PSMA ligands (U.S. 5,863,536) include N-[methylhydroxyphosphinyl]glutamic acid; N-[ethylhydroxyphosphinyl]glutamic acid; N—[propylhydroxyphosphinyl]glutamic acid; N-[butylhydroxyphosphinyl]glutamic acid; N-[phenylhydroxyphosphinyl]glutamic acid; N-[(phenylmethyl)hydroxyphosphinyl]glutarnic acid; N-[((2-phenylethyl)methyl)hydroxyphosphinyl]glutamic acid; and N-methyl—N-[phenylhydroxyphosphinyl]glutamic acid.
Illustrative PSMA ligands (US. 5,795,877) include 2—[[methylhydroxyphosphinyl]0xy]pentanedi0ic acid; 2—[[ethylhydroxyphosphinyl]oxy]pentanedioic acid; 2—[[propylhydroxyphosphinyl]0xy]pentanedi0ic acid; 2—[[butylhydroxyphosphinyl]oxy]pentanedi0ic acid; 2—[[phenylhydroxyphosphinyl]oxy]pentanedi0ic acid; 2—[[((4-pyridyl)methyl)hydr0xyph0sphinyl]oxy]pentanedi0ic acid; 2-[[((2-pyridyl)methyl)hydr0xyph0sphinyl]oxy]pentanedi0ic acid; 2-[[(phenylmethyl)hydr0xyph0sphinyl]oxy]pentanedi0ic acid; and 2[[((2—pheny1ethyl)methyl)hydr0xyph0sphinyl]oxy] pentanedioic acid.
Illustrative PSMA ligands (US. 5,962,521) include 2—[[(N—hydroxy)carbam0yl]methyl]pentanedi0ic acid; —hydroxy-N—methyl)carbamoyl]methyl]pentanedi0ic acid; 2-[[(N-butyl-N-hydr0xy) carbamoyl]methyl]pentanedioic acid; 2—[[(N—benzyl-N-hydroxy)carbanioyl]methyl]pentanedi0ic acid; 2—[[(N—hydroxy-N-phenyl)carbamoyl]methyl]pentanedi0ic acid; 2—[[(N—hydroxy-N—2-phenylethyl)carbanioyl]methyl]pentanedi0ic acid; 2—[[(N—ethyl-N-hydr0xy) carbamoyl]methyl]pentanedi0ic acid; 2—[[(N—hydroxy-N—propyl)carbamoyl]methyl]pentanedi0ic acid; 2—[[(N—hydroxy-N-S-phenylpr0pyl)carbam0yl]methyl]pentanedi0ic acid; 2—[[(N—hydroxy-N—4—pyridyl) carbamoyl]methyl]pentanedi0ic acid; 2—[[(N—hydroxy)carb0xamid0]methyl]pentanedi0ic acid; 2-[[N-hydr0xy (methyl) carboxamido]methyl]pentanedioic acid; 2-[[N-hydr0xy (benzyl) carboxamido]methyl]pentanedi0ic acid; hydroxy(pheny1)carboxamido]methyl]pentanedioic acid; hydroxy(2-phenylethyl)carboxamido]methyl]pentanedi0ic acid; 2—[[N—hydroxy(ethyl)carboxamido]methyl]pentanedi0ic acid; 2-[[N-hydr0xy(propyl) amido]methyl]pentanedi0ic acid; 2-[[N-hydr0xy (3-phenylpr0pyl) carboxamido]methyl]pentanedioic acid; and 2—[[N-hydroxy(4-pyridyl)carboxamido]n1ethyl]pentanedi0ic acid.
Illustrative PSMA s (US. 5,902,817) include 2—[(sulfinyl)methyl]pentanedioic acid; 2-[(methylsulfinyl)methyl]pentanedi0ic acid; 2—[(ethylsulfinyl)methyl]pentanedi0ic acid; 2-[(propylsulfinyl)methyl]pentanedi0ic acid; 2—[(butylsulfinyl)methyl]pentanedi0ic acid; 2-[(phenylsulfinyl]methyl]pentanedi0ic acid; 2—[[(2-phenylethyl)sulfinyl]methyl]pentanedioic acid; 2—[[(3 -phenylpropyl)sulfinyl]methyl]pentanedioic acid; 2—[[(4-pyridyl)sulfinyl]methyl]pentanedioic acid; 2-[(benzylsulfinyl)methyl]pentanedioic acid; 2-[(sulfonyl)methyl]pentanedioic acid; 2-[(methylsulfonyl)methyl]pentanedioic acid; hylsulfonyl)methyl]pentanedioic acid; 2-[(propylsulfonyl)methyl]pentanedioic acid; 2—[(butylsulfonyl)methyl]pentanedioic acid; 2-[(phenylsulfonyl]methyl]pentanedioic acid; -phenylethyl)sulfonyl]methyl]pentanedioic acid; 2-[[(3-pheny1propyl)sulfonyl]methyl]pentanedioic acid; 2-[[(4-pyridyl) sulfonyl]methyl]pentanedioic acid; nzylsulfonyl)methyl]pentanedioic acid; 2—[(sulfoximiny1)methyl]pentanedioic acid; 2-[(methylsulfoximinyl)methyl]pentanedioic acid; 2-[(ethylsulfoximinyl)methyl]pentanedioic acid; opylsulfoximinyl)methyl]pentanedioic acid; 2-[(butylsulfoximinyl)methyl]pentanedioic acid; 2-[(phenylsulfoximinyl]methyl]pentanedioic acid; 2—[[(2-phenylethyl)sulfoximinyl]methyl]pentanedioic acid; 2-[[(3—phenylpropyl) sulfoximinyl]methyl]pentanedioic acid; 2-[[(4-pyridyl)sulfoximinyl]methyl]pentanedioic acid; and 2-[(benzylsulfoximinyl)methyl]pentanedioic acid.
Illustrative PSMA ligands include O HOZC H020 ('DHOH HOZC |\OH P\ OH HOZC I OH H02C CO2H CO2H E O HCO H020 I\/\ HOZC |\/\COZH 2 OH OH c02H OOH2 COZH COZH HOZC COZH HOZC COZH H02C In another embodiment, the PSMAHligand1s a urea of two amino acids. In one aspect, the amino acids include one or more additional carboxylic acids. In another ment, the amino acids include one or more additional phosphoric, phosphonic, phosphinic, sulfinic, sulfonic, or boronic acids. In another aspect, the amino acids include one or more thiol groups or derivatives thereof. In another aspect, the amino acids include one or more carboxylic acid bioisosteres, such as tetrazoles and the like.
In another embodiment, the PSMA ligand is a compound of the formula: AJL/Fv HOZC N N H H where R1 is / \ RSI N\R Bu COzH COZH R=H, CHzCHzCN * N COZH / \N COZH I; R—H, OH_ R=H, OH In another illustrative embodiment, the binding agent is a urea of an amino dicarboxylic acid, such as aspartic acid, glutamic acid, and the like, and another amino dicarboxylic acid, or an analog thereof, such as a binding agent of the formulae ~k * HOOCHim HOOC O ( ”\O O f0 NJLN)m 0 ( n\0 QJLN COOH HOOC NJLN COOH 5 COOH H H H H H H H n Q is a an amino dicarboxylic acid, such as aspartic acid, glutamic acid, or an analog thereof, n and m are each independently selected from an integer between 1 and about 6, and (*) represents the point of attachment for the linker L.
Illustratively, the PSMA ligand is a compound of the formulae: g COOH 0 9 ”0’38” 3J HOPbH9f ”0Pal/V300” OOH COOH HOPbH COOH 00°” C00” COOH COOH COOH ... C00” HOOCN 5... C00” EVEL/fl (PK/6 Hooc P\0H COOH HOOC PbH COOH COOH COOH COOH COOH HNN,NN HJLN fiCOOH HS ° f ° : Hoocrinjku fiCOOHHOOC1H\NfiCHHOoci 8}Ni”: HuirmNfiC DUPA MUPA In another embodiment, the PSMA ligand is 2-[3-(1-Carboxymercapto- ethy1)—ureido]—pentanedioic acid (MUPA) or 2—[3—(1,3—Dicarboxy—propyl)—ureido]— pentanedioic acid .
Other illustrative examples of PSMA ligands include peptide analogs such as quisqualic acid, aspartate glutamate (Asp-Glu), Glu-Glu, Gly-Glu, y-Glu-Glu, beta-N-acetyl— L—aspartate-L—glutamate (B-NAAG), and the like.
In another embodiment, the PSMA ligand comprises a urea or thiourea of lysine and an amino acid, or one or more carboxylic acid tives thereof, including, but not limited to ureas or eas of lysine and aspartic acid, or glutamic acid, or homoglutamic acid.
In another embodiment, the PSMA ligand comprises a urea or thiourea of L- lysine and L-glutamate.
In another embodiment, the PSMA ligand comprises a compound selected from the following COZH COZH K: K o 902H 002H HOZCANJLNWNHZ JOL HOZCAN NWNHZ H H H H COZH COzH In another embodiment, the PSMA ligand comprises the following K o COZH The compounds, s, ediates, and conjugates described herein may be prepared using conventional processes, including the described in ational Patent Publication Nos. WO 02993, 2006/012527, and US. Patent Appl. No. 13/837539 (filed March 15, 2013). The disclosures of each of the foregoing are herein incorporated by reference in their entirety.
Each publication cited herein is incorporated herein by reference.
In another embodiment, a method is described for diagnosing and/or monitoring a e or disease state Where the method comprises the steps of administering to a patient being evaluated for the disease state an effective amount of a conjugate of the general formula B-L-P. The method includes allowing sufficient time for the conjugate to bind to the target tissue, and diagnosing and/or monitoring the disease or disease state extra- corporeally, such as by using on on tomography.
The radionuclide may include a positron-emitting isotope having a suitable half-life and toxicity profile. In various embodiments, the radioisotope has a half-life of more than 30 minutes, more than 70 minutes, more than 80 minutes, more than 90 minutes, more than 100 minutes, less than 8 hours, less than 6 hours, less than 4 hours, or less than 3 hours.
In other embodiments, the radioisotope has a half-life of about 30 minutes to about 4 hours, about 70 minutes to about 4 hours, about 80 minutes to about 4 hours, about 90 minutes to about 4 hours, about 100 minutes to about 4 hours, about 30 minutes to about 6 hours, about 70 s to about 6 hours, about 80 minutes to about 6 hours, about 90 minutes to about 6 hours, about 100 minutes to about 6 hours, about 30 minutes to about 8 hours, about 70 minutes to about 8 hours, about 80 minutes to about 8 hours, about 90 minutes to about 8 hours, or about 100 minutes to about 8 hours.
The uclide may include one or more positron-emitting isotopes, such as but not limited to isotopes selected from 89Zr, 45Ti, 51Mn, 64Cu, 61Cu, 63Zn, 82Rb, 68Ga, 66Ga, 11C, ”N, 15O, 12“I, 34C1, and 18F. In another embodiment, the radionuclide is a halide, such as a positron-emitting halide. In another ment, the radionuclide is a metal ion, such as a positron-emitting metal ion. In another embodiment, the radionuclide is a gallium ion, such as a positron-emitting gallium ion. In another embodiment, the radionuclide is ed from 89Zr, 64Cu, 68Ga, 66Ga, 1241, and 18F. In another illustrative embodiment, the radioisotope is selected from 89Zr, 64Cu, 68Ga, 12“I, and 18F. In another embodiment, the radioisotope is 68Ga, or 89Zr, or 18F. In another embodiment in each of the foregoing and following embodiments described herein, the radioisotope is 68Ga. In another embodiment in each of the foregoing and following embodiments described herein, the sotope is 18F. In another embodiment in each of the foregoing and following embodiments described herein, the radioisotope is 89Zr. In another embodiment in each of the foregoing and ing embodiments described herein, the radioisotope is 64Cu. It is also to be understood that the fluorine isotopes described herein may be selected from various isotopic ations of 18F and 19F. It is understood that factors that may be included during selection of a suitable isotope include sufficient half-life of the positron-emitting isotope to permit ation of a stic composition in a pharmaceutically acceptable carrier prior to administration to the patient, and sufficient remaining half-life to yield sufficient activity to permit extra-corporeal measurement by a PET scan. Further, a suitable e should have a sufficiently short half- life to limit patient exposure to unnecessary radiation. In an illustrative embodiment, 18F, having a ife of 110 minutes, es adequate time for preparation of the diagnostic composition, as well as an acceptable deterioration rate. r, on decay 18F is converted to Illustrative positron—decaying isotopes having suitable half-lives include 34Cl, half-life about 32 s; 45Ti, half-life about 3 hours; 51Mn, ife about 45 minutes; 61Cu, half—life about 3.4 hours; 63Zn, half-life about 38 minutes; 82Rb, half-life about 2 minutes; 68Ga, half-life about 68 minutes, 66Ga, half-life about 9.5 hours, 11C, half-life about minutes, 150, half-life about 2 minutes, 13N, half—life about 10 minutes, or 18F, half-life about 110 minutes.
In another embodiment, the radionuclide is a radiotherapy agent. Illustrative radionuclides for radiotherapy include isotopes of lutetium such as 177Lu, isotopes of yttrium, such as 90Y, isotopes of copper, such as 67Cu and 64Cu, and the like.
The uclide may be covalently attached to the conjugate, such as to an aryl or heteroaryl ic group, ing benzamidyl, benzylic, phenyl, pyridinyl, pyrimidinyl, pyridazinyl, naphthyl, benzothiazolyl, benzimizolyl, azolyl, and like groups. In one illustrative embodiment, the radioisotope is 18F and the radionuclide includes an aryl group to which the radioisotope is covalently attached.
The radionuclide may be non-covalently attached to the conjugate, such as within a chelate.
The methods may also be used in combination with any other methods of cancer diagnosis already developed and known in the art, including methods using other already developed diagnostic agents and utilizing x—ray computed tomography (CT), magnetic resonance imaging (MRI), functional magnetic resonance imaging (fMRI), ultrasound, and single photon emission ed tomography (SPECT).
It is understood that in certain applications of the methods described , each of the processes and synthetic s described herein either substantially complete fluorination, or alternatively only partial fluorination may be desired. Accordingly, the processes and synthetic methods described herein may be performed in various alternative ments. It is therefore understood that in those aspects where only partial fluorination is desired, the processes and syntheses described herein may be performed with less than stoichiometric s of ating agent. Similarly, it is understood that in certain applications of the s described herein, each of the processes and synthetic methods described herein either substantially complete radiofluorination, or alternatively only partial uorination may be desired. Accordingly, the processes and synthetic methods described herein may be performed in various ative embodiments. It is therefore tood that in those aspects where only l radiofluorination is desired, the processes and syntheses described herein may be performed with less than stoichiometric amounts of radiofluorination agent, where the balance is optionally 19F.
The following examples r illustrate specific embodiments of the invention; however, the following illustrative examples should not be interpreted in any way to limit the invention.
EXAMPLES WO 73678 General. Water was distilled and then deionized (18 MQ/cm2) by passing through a Milli-Q water filtration system (Millipore Corp., Milford, MA). All chemicals and solvents, unless specified, were purchased from Sigma (St. Louis, MO) and were used without further purification. Amino acids were purchased from Chem-Impex Int (Chicago, IL). 2,2'—(7-(2-((2,5-dioxopyrrolidinyl)oxy)oxoethyl)—1,4,7-triazonane—1,4-diyl)diacetic acid (NOTA-NHS) was purchased from CheMatech (France). NlO—TFA-Pteroic Acid was provided by Endocyte, Inc. erformance liquid tography (HPLC) analysis and purification of the OTA precursor were performed on an Agilent G6130B instrument. The radioactive HPLC was performed with a y-counter using a Xselect CSH C18 (250x10 mm) column and MeCN and 0.1% Formic Acid as mobile phases.
OZN O NaBH4 OZN BOC-ONO N2 _> —> HZN OMe MeOH H2N OH CHsCN BocHN OH 0004010 0004011 CIH—I:'\\N>_,BOOONCH3CN >—\ 3°C[TEE—$57M ONaBH(OAc)3 60-90% Bil—NE? 02N4< BOCHN CICHZCHZCI 2 800 3 0004001 01B” 0 O 4MHC| ’ ‘WLQNO. O'Bu Br’Y tBuO NJ H; o _. _. y m dioxane </HN N MGOH DIPEA,DMF O N N02 0004014 </N S40 Qc 4o 0 51 O‘Bu O‘Bu i50.1. on 0A 0 13” NAO’BU gmNWNW[Buoh 0 9&0 0004016 06:)WNWNMOH0004018 ‘Buo ‘Buo EXAMPLE. C—NETA. tert—Butyl [2—Hydroxy—1—(4— nitrobenzyl)ethyl]carbamate (QC04011) was prepared from the commercially available methyl 2-amino(4-nitrophenyl)propanoate through NaBH4 reduction and Boc- protection.
Successive artin oxidation and reductive amination with QC04001 afforded tris-Boc protected nd QC04013, which was transformed to QC04014 after Boc- deprotection in 4 M HCl in dioxane. Treatment of QC04014 with tert—butyl bromoacetate, ed by hydrogenolysis of the N02 group provided QC04016. Further reaction of QC04016 with succinic anhydride provided the bifunctional C—NETA (QC04018) as the corresponding tert— butyl ester.
QC04001 EXAMPLE. Di-tert—butyl [1,4,7]Triazanonane-1,4-dicarboxylate (QC04001).
QC04001 was prepared according to a modification of a synthetic procedure reported previously.[19-21] To a solution of 1,4,7-triazonane trihydrogenchloride (TACN'3HCl, 1.85 g, 7.7 mmol, M.W.:238.6) in CHCl3 (25 mL) was added DIPEA (4.0 mL, 3.0 g, 23.1 mmol, M.W.: 129.24, d: 0.742) and BOC-ON (3.77 g, 15.3 mmol, M.W.: ) in portions. The resulting mixture was stirred for 5 days and the t evaporated under vacuum. The residue was partitioned between 10% NaOH solution (10 mL) and diethyl ether (30 mL). The ether layer was separated and washed with 10% NaOH solution (10 mL) and water (10 mL) several times. The ether layer was dried (MgSO4), filtered, and concentrated under vacuum to provide QC04001 (2.53 g, quantitative), which was used without furher purification. 1H NMR (400 MHz, CDC13) 5 = 3.47—3.50 (m, 2 H), 3.42—3.45 (m, 2 H), 3.38 (br, s, 1 H), 3.28— 3.34 (m, 2 H), 3.16—3.28 (m, 2 H), 2.86—2.99 (m, 4 H), 1.48 (s, 18 H); 13C NMR (101 MHz, CDCl3) 8 = , 155.85 (C = O), 79.80, 79.70 ("Bu), 53.20, 52.62, 52.52, 51.78, 50.50, 49.91, 49.63, 48.39, 48.23, 47.83, 47.46 (TACN ring from 5320—4746), 28.60 (t'Bu). >_(O OZN4©—>_\_> 02N4©—>_\ HZN OMe "‘2N OH BocHN OH 0004010 Qco4o11 EXAMPLE. tert-Butyl [2-Hydroxy—1-(4-nitrobenzyl)ethyl]carbamate (QC04011)[19]. With minor revision to the reported procedure,[19] where the HCl salt of methyl 2-amino-3—(4-nitrophenyl)propanoate was used directly without neutralization with Et3N, to a solution of the methyl 2-amino—3 -(4-nitrophenyl)propanoate hydrochloride salt (6.22 g, 23.9 mmol) in MeOH (70 mL) at 23 0C was added NaBH4 (2.86 g, 71.4 mmol) in multiple ns. The reaction was monitored by TLC and LC-MS. The mixture was heated to reflux (with water bath at ~ 70 0 C), and NaBH4 was added portion-wise as needed until most of the starting al eared, requiring about 6 grams of NaBH4 in total. After evaporation of the solvent, the residue was treated with H20 (70 mL) and extracted with DCM/IPA (3/ 1). The combined organic layer was dried, filtered, and concentrated under vacuum to provide white solid QC04010 (4.4 g, 94), which was used t r cation.
EXAMPLE. QC04010 (4.4 g, 22.7 mmol) was dissolved in CH3CN (30 mL) at ambient temperature, to which was added BOC-ON (11.2 g, 27.2 mmol, 12 eq.) portionwise. To the above mixture was added DIPEA (5.24 mL, 3.76 g, 29.2 mmol, M.W.: 129.24, d: 0.742), the resulting e was stirred for 4 h and evaporated. The residue was partitioned between ether (50 mL) and 10% NaOH solution (20 mL). The ether layer was separated and washed with 10% NaOH solution (10 mL) and water (10 mL) sequentially. The ether layer was dried, ed, and concentrated under vacuum. The residue was washed with ether (20 mL) to e QC04011 (5.31 g, 75%), which was used without further purification. To prepare an analytical sample, the residue is purified via column chromatography on $102 g with Hexane/Ethyl e (3/1 to 1/1 with 1% of MeOH) to afford pure QC04011 as a white solid. 1H NMR (400 MHZ, CDCl3) 6 = 8.15 (d, J = 8.8 MHZ, 2 H), 7.40 (d, J=8.8 MHZ, 2 H), 4.84 (d, J: 6.8 MHZ, 1 H), 3.90 (s, 1 H), 3.68 (dd, J: 3.1 MHZ, 1 H), 3.57 (dd, J: 3.1 MHZ, 1 H), 2.98 (d, J = 6.0 MHZ, 2 H), 1.39 (s, 9 H); 13C NMR (101 MHZ, CDCl3) 5 = 156.0, 146.4, 146.2, 130.1, 123.5, 79.8, 63.3, 53.1, 37.3, 28.0.
OZN —> 02N BocHN 0H BocHN \o Qco4o11 QC04012 EXAMPLE. tert-Butyl (1-(4-nitropheny1)oxopropanyl)carbamate.
QC04011 (1.27 g, 4.3 mmol) was dissolved in CH2Clz (40 mL), and cooled to 0 °C, to which Dess—Martin periodinane (1.70 g, 5.16 mmol, 1.2 equiv) was added in one portion. After stirring for 15 min at 0 0C, the reaction was warmed to 23 0C and stirred for 45 min. The reaction was quenched by addition of a basic aq Na2S203 solution (50/50, v/v of aq NazszO3 and aq NazHCO3), and the ing mixture was vigorously stirred for 15 min. After extraction with CH2C12 (3 x), the organic phases were washed successively with water and brine, dried overNast4, filtered and concentrated in vacuo to provide QC04012, which was used without further purification.
BOON mm aCJioBocl—\N N02 </Boc BocHN QCMM2 QC04001 0604013 EXAMPLE. Reductive amination of QC04012 and QC04001 to prepare QC04013 :4. 1 ,4-Di-tert-buty1 7-(2- { [(tert-Butoxy)carbonyl]amino} 3-(4-nitrophenyl) propyl)-1,4,7-triaZ0nane-1,4-dicarboxylate (QC04013): nd QC04012 (4.3 mmol in theory) was added to a solution of QC04001 (1.40 g, 4.3 mmol) in DCE (100 mL) at 0 °C .
The ing solution was stirred for 10 min and sodium toxyborohydride (1.28 g, 6.02 mmol, 1.4 eq.) was added portionwise to the solution over 30 min. The mixture was stirred at ambient temperature ght. The reaction mixture was concentrated, treated with a WO 73678 saturated aqueous solution of NaHCO3 (50 mL), and extracted with ethyl acetate (3 x 50 mL).
The combined organic layers were dried with NazSO4, ed, and trated in vacuo.
The residue was purified via flash chromatography (SiOz, Hex/EA = 3/1) to provide QC04013 (2.31 g, 88.5 % for 2 steps, based on 2.61 g in theory) as a pale yellow semi-solid. 1H NMR (400 MHz, CDC13) 5 = 8.11 (2 H, d, J = 7.6 Hz), 7.35 (2 H, d, J = 7.6 Hz), 5.28 (1 H, s, br), 3.54—3.88 (2 H, m), 3.39—3.54 (2 H, m), 3.32—3.40 (1 H, m), 3.15-3.32 (2 H, m), 2.79—3.15 (4 H, m), 2.37—2.73 (6 H, m), 1.43 (9 H, s), 1.42 (9 H, s), 1.38 (9 H, s); 13C NMR (101 MHz, CDC13) 5 = 156.15, , 155.70, 155.56, 147.00, 146.95, 146.81, 146.76, 130.36, 123.73, 123.65, 123.60, 80.07, 79.99, 79.92, 79.81, 79.57, 79.46, 60.79, 60.47, 55.52, 54.33, 54.06, 53.64, 53.15, 53.28, 51.54, 50.80, 50.71, 50.42, 49.87, 49.07, 48.12, 39.67, 39.45, 28.74, 28.61. MS m/z: MS—API: Calcd. for C30H50N508 ([M+H]+): 608.4, Found: 608.3; NHBoc Bee 1 \ HCI/Dioxane N02—>NHH </BOC QC04013 4HC| QC04014 EXAMPLE. 1-(4—Nitrophenyl)-3 -(1,4,7-triazonanyl)propanamine. 3 (2.31 g, 3.8 mmol) was dispersed in 30 mL of 4 M HCl/Dioxane, the resulting mixture was stirred at room temperature for 20 hours. The reaction e was rapidly added to cold EtzO to precipitate a white solid. The solid was collected and dried in air to afford the pure product QC04014 (1.71 g, in quantitative yield) as a pale-white solid. MS m/z: MS-API: Calcd. for C15H26N502 ([M+H]+): 3082, Found: 308.2; o O tB 1:BUO )LOtBu .,01BBr/Nr u “2H ‘Wo W J4HCI <N/NJ >—<\/[—\ KPo QCO4015 Qco4o14 </KFO ‘BuO ‘BuO QCO4015 EXAMPLE. Introduction of the tri-tert-butyl ethylacetatelb. To a solution of QC04014 (78 mg, 0.19 mmol) and DIPEA (0.272 mL, 202 mg, 1.56 mmol, 8.2 eq. M.W.: 129.24, d: 0.742) in DMF (2 mL) was added NaI (233.8 mg, 1.56 mmol, 8.2 eq. M.W.: 149.89) and tert-Butyl bromoacetate (0.126 mL, 168 mg, 0.86 mmol, 4.5 eq. M.W.: 195.05, d: 1.321) slowly at room temperature. The resulting mixture was warmed to 60-70 °C and stirred for 20 hs. After completion, monitored by TLC and LC-MS, the reaction was ed by water and extracted with EtzO. The combined organic solvent was washed successively with water and brine, and dried over Nast4. After filtration, the solvent was evaporated under , and ing deep-colored oil residue was purified by flash chromatography on SiOz (DCM/MeOH = 100/1-100/4) to provide QC04015 (14 mg, 10 %) as a yellow oil and QC04015’ (61 mg, 49.4 %). MS m/z: MS—API: Calcd. for C39H66N5010 ([M+H]+): 764.5, Found: 764.4; O‘Bu otBu OX O>‘OtBu Ojg jOtBu0 ‘Buoh&WN%1atmOCNmWWZpd/C H2 ‘Bu0>_\ N L L tBUO QC04016 EXAMPLE. To a solution of 5 (20 mg, 0.039 mmol) in MeOH (2 mL) was added 10% Pd/C catalyst (5 mg). The resulting mixture was subjected to hydrogenolysis by agitation with H2 (g) at 1 atm (~ 15 psi) at ambient temperature for 14 h.
The reaction e was diluted with excess DCM and filtered through celite, and the filtrate was concentrated in vacuo to provide QC04016 (13 mg, 67.5 %). MS m/z: MS-API: Calcd. for C39H63N508 ([M+H]+): 734.5, Found: 734.4; FOLATE TARGETED EXAMPLES o o 002Me PyBOP DIPEA 0'3“ c W013“ DMSO 24°C_:2NHNJ/VL:f” OCF3 (:OCF3 QC02023 N‘o-TFA-Pteroic Acid EXAMPLE. (S)tert-butyl l-methyl 2-(4-(N-((2-aminooxo-3,4- dihydropteridinyl)methyl)-2,2,2-trifluoroacetamido)benzamido)pentanedioate 23).
HCl-HzN-Glu(OtBu)-0Me (350 mg, 1.38 mmol) was added to a solution of A-Pteroic Acid (560 mg, 1.37 mmol) and DIPEA (1.2 mL, 6.85 mmol) in DMSO (6.0 mL) at 23 0C under N2. After stirring for 15 min at 23 °C, PyBOP (720 mg, 1.0 mmol) was added, and the reaction mixture was stirred for 24 h at 23 OC. Volatile material was removed under reduced vacuum to afford the crude product as a semi-solid, which was further purified via solid extraction with Hex/EA (l/1) 3 times to provide QC02023 as a pale—yellow solid in quantitative yield, which was used without further purification. Xmax = 280 nm; LC—MS (Agilent G6130B Quadrupole : Mobile phase: Buffer (pH 7)-CH3CN; Column: Analytic C18 column; Method: 0—100 CH3CN—15 min, U; = 5.62 min. MS m/z: : Calcd. for C26H29F3N7O7 ([M+H]+): 608.2, Found: 608.1; o 902Me 902Me Poll—iN’VYOtB”O TFA/DCMCOCF3 —>HZNJ\HNJE ijiNWOHfCOCF3 Qcozozs 1/3, RT QC02024 EXAMPLE. (S)-4—(4-(N-((2-amino—4-oxo—3,4-dihydropteridiny1)methy1)— 2,2,2-trifluoroacetamido)benzamido)methoxy-5—oxopentanoic acid (QC02024). 224 mg of QC02023 was treated with TFA/DCM (15 mL, 1/3) at 23 OC. The reaction was stirred at 23 OC and monitored by TLC. After 1.5 hours, starting material was not observed by TLC.
The volatile material was removed under reduced pressure resulting in a semi-solid residue, which was d with cold EtzO, to provide a pale white solid precipitate, which was collected by filtration and dried in air to provide (S)(4-(N-((2-aminooxo-3,4- dihydropteridinyl)methyl)-2,2,2-tn'fluoroacetamido)benzamido)-5 -methoxy oxopentanoic acid QC02024 (169 mg, 83 % for 2 steps). 7mm = 280 nm; LC—MS (Agilent G6130B Quadrupole : Mobile phase: Buffer (pH 7)-CH3CN; Column: Analytic C18 column; Method: 0—100 15 min, U; = 3.40 min. MS m/z: MS-API: Calcd. for F3N7O7 ([M+H]+): 552.1, Found: 552.1; 1H NMR (400 MHz, DMSO) 5 = 12.16 (s, br, 1 H), 8.88 (d, J: 7.2 Hz, 1 H), 8.65 (s, 1 H), 7.92 (d, J: 8.0 Hz, 2 H), 7.64 (d, J: 8.0 Hz, 2 H), 7.16 (s, br, 1 H), 5.14 (s, 2 H), 4.38—4.55 (m, 1 H), 3.64 (s, 3 H), 2.28—2.40 (m, 2 H), 2.00- 2.12 (m, 1 H), 1.87—2.00 (m, 1 H); 13C NMR (101 MHz, DMSO) 5 = 173.91, 172.36, 165.93, 161.03, 156.11, 155.76 (d, J: 35.8 Hz), 154.19, 149.40, , 141.80, 134.30, 128.89, 128.62, 128.29, 117.91 (d, J = 48.5 Hz), 5390, 52.23, 52.06, 30.26, 25.81; 19F NMR (377 MHz, CDCl3) 5 = -62.87.
H2N N N EC 1777 C25H31N907 Exact Mass: 569.23 MOI. Wt.: 569.57 EXAMPLE. lu—Lys—OH (EC1777). EC1777 was prepared using solid phase peptide synthesis as follows. nd mmol Equivalent Molecular ty Weight (grams) Fmoc-Lys—Resin 0.5 1 1.00 (Loading ~0.5mmol/g) FmOC-Glu-O‘Bu 425-5 0426 NIO-TFA-Pteroic 0.65 1.3 408 0.265 PyBOP 520.31 DIPEA 1.5 3 129.24 0.168 (d=0.742) In a peptide synthesis vessel, Fmoc-Lys-resin (1.0g, 0.5mmol) was placed and washed with DMF (3 X 10 ml). Initial Fmoc deprotection was performed using 20% piperidine in DMF (3 x 10 ml) solution for 10 mins per cycle. Subsequent washes of DMF (3 X 10 ml) and i-PrOH (3 X 10 ml), a Kaiser test was done to determine reaction completion.
Following another DMF wash (3 x 10 ml); an amino acid solution (2.0 eq.) in DMF, PyBOP (2.0 eq.) and DIPEA (3.0 eq.) were added to the vessel and the solution bubbled with Argon for 1 hour. The coupling solution was filtered, the resin was washed with DMF (3 x 10 ml) and i-PrOH (3 X 10 ml) and a Kaiser test was done to assess reaction completion. The above process was performed successively for the additional coupling. Resin cleavage was performed with a cocktail consisting of 95% CF3CO2H, 2.5% H20 and 2.5% triisopropylsilane. The cleavage il (10 ml) was poured onto the resin and bubbled with Argon for 30 mins, followed by filtration into a clean flask. Further ge was performed twice successively with fresh cleavage cocktail for 10 mins of bubbling. The combined filtrate was poured onto cold diethyl ether, the precipitate formed was collected by centrifugation at 4000 rpm for 5 mins (3X). The itate was obtained following decanting and drying of the solid under vacuum. Deprotection of the trifluoro-acetyl group was ed by dissolving the crude itate in H20 (15 ml), which was basified with Na2CO3 to pH 9 with Argon ng. Upon completion of the reaction, confirmed by LCMS, the solution was acidified to pH 3 using 2 M HCl and the desired linker was purified by preparative HPLC (mobile phase A = 10mM Ammonium acetate, pH = 5; Organic phase B = Acetonitrile; Method; 10% B to 100%B in 30 mins) to yield EC 1777 (112 mg, 39%); 1H NMR (500 MHz DMSO—d6) Pivotal s: 5 8.60 (s, 1H), 7.58 (d, 2H), 6.60 (d, 2H), 4.45 (s, 2H). [M+H]+ = Calculated 570.23, found 570.582 0 QOZH H H H ' N N N HNJK/[NjAN o cogH 8 NJ H2NJ\\N H N/ H020J E01778 C45H57N13O138 Exact Mass: 9 Mol. Wl.: 1020.08 EXAMPLE. Pte-yGlu-Lys—NOTA. In a dry flask, EC 1777 (30.5 mg, 0.054 mmol, 1.0 eq.), 1,1,3,3-tetramethy1guanidine (13.45 ul, 0.107 mmol, 2.0 eq.) and DMSO (2.5 ml) under Argon were sonicated for 1 hour. DIPEA (0.19 ml, 1.07 mmol, 20 eq.) was added to the on, followed by sonication for an addition hour. To the transparent solution was added p—SCN—Bn—NOTA.3HCl (33 mg, 0.059 mmol, 1.1 eq.) and the on was moitored until completion by LCMS and purified using ative HPLC (mobile phase A = 10mM Ammonium acetate, pH = 5; Organic phase B = Acetonitrile; Method; 10% B to 100%B in mins) to yield EC 1778 (16 mg, 29%). 1H NMR (500 MHz DMSO-d6) Pivotal s: 8 8.60 (s, 1H), 7.58 (d, 2H), 7.29 (d, 2H), 7.07 (d, 2H), 6.61 (d,2H), 4.45 (s, 2H), 4.20 (t, 1H).
[M+H]+ = Calculated 1020.39, found 1020.63.
EXAMPLE. Pte-yGlu-Lys-NOTA -Al-18F is prepared by reaction of Pte- yGlu-Lys—NOTA with A118F3-3H20 (1 step method) or with 3H20 followed by reaction with NalSF (2 step method) using published processes. 0 _ TENT” H O 1 O H2N \N N/ OJNCF3 be\ & N NH EXAMPLE. N10-TFA-Pte—yGlu-OtBu-Arg(be)-Arg(be)-Lys(Mtt)—resin 3.
The general procedure described for the synthesis of resin bound folate-peptide resin 1 was followed for the coupling of 2X Fmoc-L—Arg(be)—OH, Fmoc-Glu-OtBu, and N10-TFA-Pte— OH to —Lys(Mtt)—Wang resin.
HZN NH o J::j)Lm/\¢Ajy ; H N O ;If\/A\/ jS],N\[::L\/<é{_\NACOZF HQN N N NH H020 HZN/L3NH E02217 C571-181N21O15S Exact Mass: 1331 .59 Mol. Wt: 1332.45 EXAMPLE. Pte-yGlu-Arg—Arg-Lys-Bn-NOTA 4 (EC2217). In a peptide synthesis vessel, A—Pte-yGlu-OtBu—Arg(be)-Arg(be)-Lys(Mtt)-resin (0.28g, 0.07mmol) was placed and washed with DCM (3 x 10 m1). Selective Mtt deprotection was performed by adding a 2% CF3C02H/ DCM solution to the vessel and bubbling with Argon for 10 min. After filtering, the resin was washed with dichloromethane followed by a fresh solution of 2% H/ DCM. This process was repeated until there was no more yellow solution being yielded and a Kaiser test was done. Following a DMF wash (3 X 10 ml); p- SCN-Bn-NOTA.3HC1 (50 mg, 0.09 mmol, 1.2 eq.) in DMF, and DIPEA (80 ul, 0.45 mmol, 6.0 eq.) were added to the vessel and the solution bubbled with Argon for 2 hour. The coupling solution was filtered, the resin was washed with DMF (3 x 10 m1) and i-PrOH (3 x ml) and a Kaiser test was done to assess reaction completion. Resin cleavage/global tert- butyl ester ection was performed with a cocktail consisting of 95% CF3COzH, 2.5% H20 and 2.5% triisopropylsilane. The cleavage cocktail (10 ml) was poured onto the resin and bubbled with Argon for 60 mins, followed by filtration into a clean flask. Further cleavage was performed twice successively with fresh ge cocktail for 20 mins of bubbling. The combined filtrate was poured onto cold diethyl ether, the precipitate formed was ted by centrifugation at 4000 rpm for 5 mins (3x). The precipitate was obtained following decanting and drying of the solid under vacuum. Deprotection of the ro- acetyl group was achieved by dissolving the crude precipitate in H20 (15 ml), which was basified with NazCO3 to pH 9 with Argon bubbling. Upon completion of the reaction, confirmed by LCMS, the solution was acidified to pH 5 using 2 M HCl and the desired linker was purified by preparative HPLC (mobile phase A = 10mM Ammonium acetate, pH = 5; Organic phase B = itrile; Method; 10% B to 100%B in 30 mins) to yield EC2217 (35mg, 35%). 1H NMR (500 MHz DMSO—d6) Pivotal signals: 5 8.61 (s, 1H), 7.54 (d, J = 8.4 Hz, 2H), 7.17 - 7.03 (m, 2H), 6.99 (d, J = 8.0 Hz, 2H), 6.66 (d, J = 8.5 Hz, 2H), 4.52 - 4.45 (m, 1H), 4.17 (dt, J = 8.9, 4.6 Hz, 2H), 4.12 (s, 1H), 4.07 — 3.97 (m, 1H). [M+H]+ = Calculated 1332.59, found 1332.87 be’NYNH + NH 0V0 o o : NJ H o IW NHMtt N H’Vfig i g: H HM ‘W Y o 1 HZN \N N/ OJ\CF3 >ro NH Mann/km EXAMPLE. N10-TFA-Pte-yGlu-OtBu-Asp(OtBu)-Arg(be)-Arg(be)- Lys(Mtt)-resin 5. The general ure bed for the synthesis of resin bound folate- e resin 1 was followed for the coupling of 2X Fmoc—L-Arg(be)-OH, Fmoc-L— Bu)-OH, Fmoc-Glu—OtBu, and N10—TFA-Pte-OH to —Lys(Mtt)-Wang resin.
HZNYNH HO cxl2 \ Oo§_/0H OVOH N o NAcogH : N N N WJL 0 NHW N h] N N : B _ H H HOZCJ )\\ If” OH 1 H2N N N NH HgN/kNH EC2218 CG1H86N2201BS Exact Mass: 1446.62 Mol. Wt: 1447.54 EXAMPLE. Pte-yGlu-Asp-Arg-Arg-Lys-Bn-NOTA 6 (EC2218). Pte-yGlu- Asp-Arg-Arg-Lys-Bn-NOTA, EC2218 was prepared in 18% yield according to the process described for folate-peptide-NOTA, 4. 1H NMR (500 MHz DMSO-d6) Pivotal signals: 5 8.58 (s, 1H), 7.52 (d, J = 9.0 Hz, 2H), 7.14 — 7.08 (m, 4H), 6.61 (d, J = 9.0 Hz, 2H), 4.16 — 4.09 (m, 2H), 4.06 (dd, J = 10.0, 4.3 Hz, 1H), 3.90 (dd, J = 7.8, 4.7 Hz, 1H). [M+H]+ = Calculated 1449.64, found 1449.76 HNjfiNfN O H2N)\\N N/ 0%CF3 \L Pka/KNH H EXAMPLE. N10—TFA-Pte—yGlu-OtBu-Arg(be)-Lys(Mtt)—resin 7. The general procedure described for the synthesis of resin bound folate-peptide resin 1 was followed for the coupling of -Arg(be)-OH, Fmoc—Glu-OtBu, and N10-TFA-Pte-OH to Fmoc-L—Lys(Mtt)-Wang resin.
Ho 02 I—\ o OVOH o OVOH NAcogH UW imm o u H N H028 N 1% HgN/KN”“51 3”” N/ 1 HZN’l§NH E02219 CS1H69N17O14S Exact Mass: 1175.49 Mel. W1.: 1176.27 WO 73678 EXAMPLE. Pte-yGlu-Arg—Lys-Bn—NOTA 8 (EC2219). Pte-yGlu-Arg-Lys— Bn-NOTA, EC2219 was preapred in 20% yield ing to the process described for folate— peptide-NOTA, 4. 1H NMR (500 MHz DMSO-d6) l signals: 5 8.68 (s, 1H), 7.60 (d, J = 8.4 Hz, 3H), 7.27 — 6.97 (m, 4H), 6.77 - 6.69 (m, 2H), 4.28 —f 4.19 (m, 2H), 4.08 (dd, J = 9.0, 5.4 Hz, 1H), 4.01 (dd, J = 8.5, 5.4 Hz, 1H). [M+H]+ = Calculated 1, found 1178.7 H/£:::I/H\H NQKH H N /—\ 0 NW N IW \n/\N N’\ lN o <\1\H; o 0 0 OH <tjf‘i> COZH EC2222 C49H74N20014 Exact Mass: 1166.57 Mol. Wt.: 1167.24 EXAMPLE. Pte-yGlu—Arg—Arg—Lys—NOTA 9 (EC2222). In a peptide synthesis vessel, N10-TFA-Pte-yGlu-OtBu-Arg(be)-Arg(be)-Lys(Mtt)-resin (0.5g, 0.12mmol) was placed and washed with DCM (3 x 10 m1). ive Mtt deprotection was performed by adding a 2% CF3C02H / DCM solution to the vessel and bubbling with Argon for 10 min. After filtering, the resin was washed with dichloromethane ed by a fresh solution of 2% CF3COzH/ DCM. This process was repeated until there was no more yellow solution being d and a Kaiser test was done. Following a DMF wash (3 X 10 ml); NOTA-Bis(tBu)ester (0.10 g, 0.24 mmol, 20 eq.) in DMF, PyBOP (0.14 g, 0.26 mmol, 2.2 eq) and DIPEA (64 ul, 0.36 mmol, 3.0 eq.) were added to the vessel and the solution bubbled with Argon for 2 hour. The coupling solution was filtered, the resin was washed with DMF (3 x 10 ml) and i-PrOH (3 x 10 ml) and a Kaiser test was done to assess reaction completion.
Resin cleavage/global tert—butyl ester deprotection was performed with a cocktail consisting of 95% CF3COzH, 2.5% H20 and 2.5% triisopropylsilane. The cleavage cocktail (10 ml) was poured onto the resin and bubbled with Argon for 1hr, followed by filtration into a clean flask. Further cleavage was performed twice successively with fresh cleavage cocktail for 10 mins of bubbling. The combined filtrate was poured onto cold diethyl ether, the itate formed was collected by centrifugation at 4000 rpm for 5 mins (3X). The precipitate was obtained following decanting and drying of the solid under vacuum. Deprotection of the trifluoro-acetyl group was achieved by dissolving the crude precipitate in H20 (15 ml), which was basified with Na2CO3 to pH 9 with Argon bubbling. Upon completion of the reaction, confirmed by LCMS, the solution was acidified to pH 5 using 2 M HCl and the desired linker was purified by preparative HPLC (mobile phase A = 10mM Ammonium acetate, pH = 5; Organic phase B = itrile; Method; 10% B to 100%B in 30 mins) to yield EC2222 (28mg, 20%). 1H NMR (500 MHz DMSO—d6) Pivotal s: 8 8.60 (s, 1H), 7.51 (d, J = 8.1 Hz, 2H), 6.64 (d, J = 8.4 Hz, 2H), 4.21 — 4.09 (m, 2H), 4.09 — 4.03 (m, 1H), 3.98 — 3.88 (m, 1H), 3.50 (s, 1H). [M+H]+ = Calculated 1167.57, found 1167.8 0 902Me O NHW PyBOP, DIPEA WailNfem,N O H2N\/\O/\/o\/\NHBOC —> HN \ N + o 0002024 DMSO,23 C 902Me ngkNNWNwO/VowNHBOCQC07010rooms EXAMPLE. thyl 18-(4-(N-((2-aminooxo—3,4-dihydropteridin y1)methyl)—2,2,2-trifluoroacetamido)benzamido)-2,2—dimethyl-4,15—dioxo-3 ,8,1 1-trioxa-5,14— diazanonadecanoate (QC07010). QC02024 (100 mg, 0.181 mmol) is added to a solution of Mono-Boc-PEG-NHZ (45 mg, 0.181 mmol) and DIPEA (0.158 mL, 0.905 mmol) in DMSO (2 mL) at 23 0C under N2. After being d for 15 min at 23 OC, PyBOP (94.2 mg, 0.181 mmol) was added, and the reaction mixture was stirred for 24 h at 23 OC. Volatile material was removed under reduced vacuum, the crude material was further ed by SPE cation: extract successively with ACN (2 X), EA (1 X) and EtzO (1 x) to afford pure product QC07010 (127mg, 90%). Xmax = 280 nm; LC-MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH CN; : Analytic C18 column; Method: 0— 100 CH3CN—15 min, tR = 5.06 min. MS m/z: MS-API: Calcd. for C33H43F3N9010 ([M+H]+): 782.3, Found: 782.2; 1H NMR (400 MHz, DMSO) 5 = 11.59 (s, br, 1 H), 8.92 (d, J = 7.2 Hz, 1 H), 8.64 (s, 1 H), 7.85—8.02 (m, 3 H), 7.64 (d, J = 8.0 Hz, 2 H), 6.75 (t, J = 5.2 Hz, 1 H), 5.13 (s, 2 H), 4.33—4.48 (m, 1 H), 3.64 (s, 3 H), 3.46 (s, 4 H), 3.30—3.41 (s, 4 H), 3.14—3.23 (m, 2 H), 3.01-3.08 (m, 2 H), 2.19—2.30 (m, 2 H), 2.02-2.12 (m, 1 H), 1.89—2.00 (m, 1 H), 1.35 (s, 9 H); 13C NMR (101 MHz, DMSO) 8 = 172.43, 171.46, 165.73, 160.87, 156.80, 15570 (d, J = 35.5 Hz), 155.67, 154.17, 149.49, 144.20, 141.73, 134.30, , 128.55, 128.23, 116.20 (d, J = 290.0 Hz), 77.65, 69.58, 69.50, 69.193, 69.192, 53.88, 52.52, 51.96, 38.89, 38.62, 31.65, 28.23, 26.32; 19F NMR (377 MHZ, CDC13) 6 = -62.87.
O EOZMe H o TFA/DCM 0 0*N/V\n/ \/\O/\/ \/\NHBOC—> HNJENjA'll o 1l3 QC0701O N)\\N N/ COCF3 O gone H NfifiNfEOCFg,N duwwvmQCO7011 E. (S)-methyl 2-(4-(N-((2-aminooxo-3,4-dihydropteridin y1)methyl)-2,2,2—trifluoroacetamido)benzamido)-5—((2-(2—(2— aminoethoxy)ethoxy)ethy1)amino)—5-oxopentanoate (QC07011). QC07010 (274 mg, 0.35 mmol) was treated with TFA/DCM (4 mL, 1/3) at 23 oC. The reaction was stirred at 23 OC and monitored by LC-MS. After 1.5 h, TLC showed that all starting material eared.
The mixture was diluted with CH3CN and evaporated to dry via ap. Residue TFA (b.p. 72.4 0C) was d through azeotropic distillation with ACN to afford the product QC07011 in quantitative yield, which was used without further purification. kmax = 280 nm; LC—MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH 7)—ACN; : Analytic C18 column; : 0-100 ACN 15 min, tR = 3.84 min. MS m/z: MS-API: Calcd. for C28H35F3N903 ([M+H]+): 682.2, Found: 682.2. 0 902Me I«(j/k” = “WNV\O/\/o\/\NH2—>NOTA-NHS o HNJj:N\ o DIPEA,DMSO NkN | rCOCFs QC07011 COZH :NOiNW N002Me N if!“/>N\_/¥002H EXAMPLE. (S)-2,2'—(7-(4-(4—(N-((2—amino—4—oxo-3,4—dihydropteridin—6- hyl)-2,2,2-tn'fluoroacetamido)benzamido)-3 ,7,18-trioxo-2,11,14-trioxa-8,17- diazanonadecany1)-1,4,7-t1iazonane-1,4-diy1)diacetic acid (QC07013). QC07011 (15.7 mg, 0.023 mmol) in DMSO (0.5 ml) was added NOTA-NHS (18.2 mg, 0.028 mmol) followed by DIPEA (15 uL, 0.084 mmol). The reaction was stirred at 23 °C, monitored by LC—MS, and most of the starting material was converted to QC07013 in 5 hours. The product was purified by RP-Clg HPLC to afford the pure product QC07013 (13.0 mg, 58.5 %). Xmax = 280 nm; LC—MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH 7)- CH3CN; Method: 0—100 CH3CN—15 min, tR = 3.74 min. MS m/z: MS—API: Calcd. for WO 73678 C40H54F3N12013 ([M+H]+): 967.4, Found: 967.2; HPLC (Agilent ative C18 Column): Mobile phase: Buffer (pH 7)-CH3CN; Method: 0-100 30 min, tR = 10.75 min c0211 O COZMQ O<N\N/N> O /\/\n/N\/\O/\/O\/\NJJ\/N\—/LCO2H HN N\ NQXH J\\ I COCF3 / QC07013 N N N 1 M NaOH (aq) C02H 23 °C 15 min 60 % afterOHPLC ON/V\n/ \/\O/\/0\/\NCOZH OFN/> N /U\/N 1¥C02H QC07017 EXAMPLE. (S)—2,2'—(7—(1-(4—(((2-amino—4—oxo—3,4—dihydropteridin—6— yl)methyl)amino)phenyl)-3 -carboxy-1,6,17-trioxo-10, 13 -dioxa-2,7,16-triazaoctadecan y1)—1,4,7-triazonane—1,4-diyl)diacetic acid (FA—PEGl—NOTA, QC07017). QC07013 (20.8 mg, 0.022 mmol ) was stirred in 1.2 mL of 1 M NaOH (aq.) at 23 OC and the reaction was monitored by LC-MS. After 15 min, all ng material was transformed to product, the crude material was purified by RP-C18 HPLC to afford QC07017 (11.3 mg, 60%). kmax = 280 nm; HPLC (Agilent Preparative C18 Column): Mobile phase: Buffer (pH 7)—CH3CN; Method: 0-30 CH3CN-30 min, tR = 11.49 min. LC-MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH CN; Method: 0—100 CH3CN 15 min, tR = 2.72 min. MS m/z: MS-API: Calcd. for C37H53N120l2 ([M+H]+): 857.4, Found: 857.2. 1H NMR (400 MHz, DMSO) 5 = 8.62 (s, 1 H), 8.28 (t, J = 5.6 Hz, 1 H), 7.99 (t, J = 5.6 Hz, 1 H), 7.85 (d, J = 7.2 Hz, 1 H), 776—780 (S, br, 2 H), 7.58 (d, J = 8.8 Hz, 2 H), 7.00 (t, J = 6.0 Hz, 1 H), 6.62 (d, J = 8.8 Hz, 2 H), 4.47 (d, J = 5.2 Hz, 2 H), 4.13—4.18 (m, 1 H), 3.43 (s, 4 H), 3.31—3.41 (m, 4 H), 3.29—3.32 (m, 2 H), 3.10—3.24 (m, 4 H), 3.03—3.10(s,br, 2 H), 2.90—3.03 (3, br, 2 H), 2.10—2.14 (m, 2 H), .05 (m, 1 H), 1.84—1.91 (m, 1 H); 13C NMR (101 MHz, DMSO) 6 = 174.33, 172.21, 171.17, 170.35, 165.70, 161.85, , 154.95, 150.56, 148.45, 148.32, 128.62, 127.87, 121.84, 111.38, 69.44, 69.30, 69.08, 68.70, 60.95, 57.48, 53.11, 50.85, 49.41, 48.91, 45.88, 38.60, 38.18, 32.04, 27.52.
Q 00N’\/ 2 1)Swe||DCM2h DMF2h ® 000W0WOVEONNHFWC 2) Fmoc—NH—(PEG)5——COOH HATU/DIPEA/DMF Trt-EDA Resin 1) 20% Piperidine/DMF, 1) 20% PiperidinelDMF, H 00213” H 30 mln; min; N Q 00”N WCk/igoA/NWNHFmoc—> 0 0 2) Fmoc—GIu-O‘Bu 2) N‘O-(TFA)pteroic acid HATU/DIPEA/DMF, 2 h; 0 HATU/DIPEA/DMF, 2h 1) TFA/HQO/TIPS (95:25:25); tBUOZC o H H @ O ”wNwoflo/VNWN 2) Sat Na2C03, HPLC purification O o o ACFgN/\::fLNHNHZ o 902H H H o N\/\or»:OWN\/\ N o o \ Nofiw HZNJNjK/[NfH Qc03019 EXAMPLE. Solid Phase Synthesis (SPS) of FA-PEGa-EDA-NHZ Precursor (QC03019). 1,2-Diaminoethane trityl resin (1.2 mmol/g, 100 mg, 0.12 mmol) was swollen with dichloromethane (DCM, 3mL) followed by dimethyl formamide (DMF, 3 mL). After swelling the resin in DMF, a solution of fluorenylmethoxycarbonyl -PEG6-OH (1.5 equiv), HATU (1.5 , and DIPEA (2.0 equiv) in DMF was added. Argon was bubbled for 2 h, and resin was washed with DMF (3 x 3 mL) and i-PrOH (3 x 3 mL). The above sequence was repeated for two more coupling steps for conjugation of Fmoc-Glu-(OtBu)-OH and Nlo-TFA-Ptc-OH. The final product was cleaved from the resin using a trifluoroacetic acid HzOztriisopropylsilane cocktail (95:2.5:2.5) and concentrated under vacuum. The concentrated product was precipitated in l ether and dried under vacuum, which was then incubated in Sat. Na2C03 and monitored by LC-MS. 1 hour later, the mixture was neutralized to pH = 7 with 2 M HCl (aq.) which was purified by preparative with preparative RP-CIS HPLC [solvent nt: 0% B to 50% B in 30 min; A = 10 mM NH4OAC, pH = 7; B = CH3CN]. Acetonitrile was removed under vacuum, and the residue was freeze-dried to yield QC03019 as a yellow solid (59 mg, 60%). Analytical RP—C18 HPLC: tR = 4.22 min (A = 10 mM , pH = 7.0; B = CH3CN, solvent gradient: 0% B to 50% B in 15 min); ative RP-C18 HPLC: tR = 11.7 min (A = 10 mM NH4OAC, pH = 7.0; B = CH3CN, t gradient: 0% B to 50% B in 30 min); kmx = 280 nm; HPLC (Agilent Preparative C18 ): Mobile phase: Buffer (pH 7)-CH3CN; Method: 0-30 CH3CN-30 min, tR = 11.7 min.
LC-MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH 7)-CH3CN; Method: 0—50 CH3CN-15 min, tR = 4.22 min. MS m/z: : Calcd. for C36H55N10012 ([M+H]+): 819.4, found, 819.2. 1H NMR (DMSO—ds/DzO) 5 = 8.63 (s, 1H), 7.64 (d, J = 8.8 Hz, 2H), 6.64 (d, J = 8.8 Hz, 2H), 4.48 (s, 2H), 4.12—4.21 (m, 1 H), 3.58 (t, J: 6.4 Hz, 2 H), 3.41—3.53 (m, 24 H), 3.18—3.25 (m, 2 H), 3.11—3.18 (m, 2 H), 2.28 (t, J: 6.4, 2H), 2.15 (t, J: 7.4, 2H), 2.03 (m, 1H), 1.88 (m, 1H) ppm.
NWNwO’(\/OWN\/\NH2 —>NOTA—NHS 2)qu o DIPEA, DMSO 0003019 NWNwoNOWNWNkflNFCO2H FwtfiEL/[NNjANN/(j)kN 0 ocovozs {:ch EXAMPLE. FA—PEGs—NOTA. To QC03019 (9.5 mg, 0.011 mmol) in DMSO (0.40 ml, with a tration at 0.029 M) was added NOTA-NHS (8.6 mg, 0.013 mmol) ed by DIPEA (7.0 uL, 0.039 mmol). The reaction was stirred at 23 OC, monitored by LC-MS, and most of the starting material was ormed to the corresponding product in 5 hours. The crude material was ed by RP-C1g HPLC to afford the pure product QC07029 (5.5 mg, 45 %). Analytical RP-Cls HPLC: tR = 3.91 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 50% B in 15 min); Preparative RP-C18 HPLC: tR = 10.51 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 50% B in min); max = 280 nm; HPLC (Agilent Preparative C18 Column): Mobile phase: Buffer (pH 7)—CH3CN; Method: 0—30 CH3CN—30 min, 61 = 10.51 min. LC-MS (Agilent G6130B pole LC/MS): Mobile phase: Buffer (pH 7)—ACN; Method: 0-50 ACN-15 min, tR = 3.91 min MS m/z: MS-API: Calcd. for C48H74N13017 ([M+H]+): 1104.5, Found: 1104.4 1'1sz wmuoNWNHMVOPNFCZJla)AIFa; pH4. orpH 4.5-5 b) AICI3; then NaF, n=1-20 H N NACOZH N o m31a HN n N J\\ H N N < n: 1-20 E. FA-NOTA-Al—lSF Radiotracer[2]. Two s for the formation of FANOTA-Al-lSF are described herein. Conditions including the pH value, concentration of the ates and temperature for the chelating reaction with 18F-Al can be varied. The general methods for FA-NOTA-Al-lSF are described as followed: Method a). FA-NOTA Precursor was dissolved in 2 mM NaOAc (pH 4.5) and 0.5 mL of ethanol, which was treated with A118F3-3H20 (1.5 eq.) which was freshly prepared before ation. The pH was adjusted to 4.5-5.0, and the reaction mixture was refluxed for -30 min with pH kept at 45-50. After being cooled down to room temperature, the crude material was loaded to a cartridge, and the radiotracer was eluted into vial. After sterile filtration and being diluted to appropriate radioactivity (5-10 mCi) and specific activity (> 1 Ci/umol), the radiotracer was ready for in vivo PET imaging study.
Method b). A Precursor was dissolved in 2 mM NaOAc (pH 4.5), and treated with 3HzO (1.5 eq.). The pH was adjusted to 4.5—5.0, and the reaction mixture was refluxed for 15-30 min with pH kept at 45-50. The crude material was purified by RP-HPLC to afford the A-Al-OH intermediate ready for 18F— labeling.
Appropriate amount of FA—NOTA—Al-OH was treated with NalSF saline solution and ethanol (1/ 1, v/v), and the whole mixture was heated at 100-110OC for 15 min. After being cooled down to room temperature, the crude material was loaded to a cartridge, and the radiotracer was eluted into vial. After e filtration and being diluted to appropriate radioactivity (5-10 mCi) and specific activity (> 1 l), the radiotracer was ready for in viva PET imaging study.
O COZH O<N\N/> O 'LCOZH HN N\ NdHWN\/\O/\/O\/\Nj)J\/N folate-NOTA (1) Ax I H N N N iAI‘BFa rcozH o OCH2 0 {N3 : H 0 o mmN \/\ /\/ \/\. k/NAFBFN u uL HzNifiNjflNN -NOTA-AI13F (2) EXAMPLE. Standard Protocol for the Formulation of Folate-NOTA-AllSF Radiotracer. The resin containing 18F was first washed with 1.5 mL of ultrapure water, and then 18F was eluted out from resin by using 1.0 mL of 0.4 M KHC03 solution. 100 uL of the eluting solution containing 18F was added to a stem vial charged with 10 uL acetic acid, 25 uL AlC13 (2 mM in 0.1 M NaOAc pH 4 ) and 125 ”L 0.1 M NaOAc pH 4 buffer. The whole mixture was incubated for 2 min before 0.25 mg folate-NOTA precursor (1) in 125 uL of 0.1 M NaOAc pH 4 buffer was transferred to the same stem via]. The reaction was immediately heated to 100 0C for 15 min.
After cooling to room temperature, the crude material was mixed with 0.7 mL 0.1% formic acid and purified by radioactive HPLC on a Xselect CSH C18 (250 x 10 mm) column using MeCN and 0.1% formic acid as the mobile phase. The fraction at 11.5 min was collected to afford pure radiotracer in ~ 40-50% hemical yield (RCY) with ~ 98% radiochemical purity (RCP). The total radiochemical synthesis of folate-NOTA-APSF (2, AllSF-QC07017) was accomplished in ~37 min with a specific activity (SA) of 70 i 18.4 ol. After sterile filtration and appropriate dilution in isotonic saline to the desired radioactivity, the folate-NOTA-AllSF (2) racer was ready for PET imaging study.
Using same strategy, radiochemcial synthesis of FA—PEG1z—NOTA—Al-18F radiotracer (QC07043) was accomplished in ~35 min with a specific activity (SA) of 49 i 17.1 GBq/umol. Although the radiochemical purity is excellent, 100% after radioactive HPLC purification, the total hemical yield (RCY) is relatively low, ~ 25-30%. After sterile filtration and appropriate dilution in isotonic saline to the desired ctivity, the FA- PEGiz—NOTA-Al—lsF radiotracer was ready for PET imaging study.
NH «s 00NM1) Swell DCM 2 h DMF 2 h Q5 00 ~“WV)?W0 ~NHFmoc 2) FmocNH(PEG)5COOH HATU/DIPEA/DMF 0” Trt-EDA Resin 1) 20% Piperidine/DMF, 1) 20% PiperidinelDMF, H COZ‘Bu min; min; N Q 00”N WOWONNWNHFFHOC—p 0 0 2) Fmoc—GIu-O‘Bu 2) N‘O-(TFA)pteroic acid HATU/DIPEA/DMF, 2 h; 0 HATU/DIPEA/DMF, 2h 1) TFA/HQO/TIPS (95:25:25); H tBUOZC @ O 2) Sat Na2C03 HPLC purification HNNNWOwO/VNWNN/‘KQjL/T 0 ° %CF3\N wrjfiwiwwwwQC07041 EXAMPLE. Solid Phase Synthesis (SPS) of FA-PEGiz-EDA-NHZ (QC07042)[11]. 1,2-Diamin0ethane trityl resin (1.2 mmol/g, 50 mg, 0.06 mmol) was swollen with dichloromethane (DCM, 3mL) followed by dimethyl formamide (DMF, 3 mL). After swelling the resin in DMF, a solution of fluorenylmethoxycarbonyl (Fmoc)-PEG12-OH (1.5 equiv), HATU (1.5 equiv), and DIPEA (2.0 equiv) in DMF was added. Argon was d for 2 h, and resin was washed with DMF (3 x 3 mL) and i-PrOH (3 x 3 mL). The above sequence was repeated for two more coupling steps for conjugation of Fmoc-Glu-(OtBu)-OH and N10-TFA-Ptc-OH. The final product was cleaved from the resin using a roacetic acid (TFA):HzOztriisopropylsilane cocktail :25) and concentrated under vacuum. The concentrated product was precipitated in diethyl ether and dried under vacuum, which was then ted in Sat. Na2C03 and monitored by LC-MS. 1 hour later, the mixture was neutralized to pH = 7 with 2 M HCl (aq.) which was ed by preparative with preparative RP-CIS HPLC [solvent gradient: 0% B to 50% B in 30 min; A = 10 mM NH4OAC, pH = 7; B = CH3CN]. Acetonitrile was removed under vacuum, and the residue was freeze-dried to yield pure QC07042 as a yellow solid (32.5 mg, 50 %). Analytical RP—C18 HPLC: tR = 4.76 min (A = 10 mM NH4OAC, pH = 7.0; B = CH3CN, t gradient: 0% B to 50% B in 15 min); ative RP-Cig HPLC: tR = 13.75 min (A = 10 mM NH4OAC, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 50% B in 30 min); UV-Vis: max = 280 nm; Preparative RP—C18 HPLC: HPLC (Agilent Preparative C18 Column): Mobile phase: Buffer (pH 7)- CH3CN; Method: 0—50 CH3CN, 30 min, tR = 13.75 min. LC-MS: LC—MS (Agilent G6130B Quadrupole LC/MS) of t Mobile phase: Buffer (pH CN; Method: 0-50 CH3CN, min, tR = 4.76 min. MS m/z: MS-API: Calcd. for C48H79N10018 ([M+H]+): 1083.6, Found: 1083.4; o 902H _ H H _ HS HNWNth/OWNwNHZ —>11 N o o DIPEA,DMSO HN \ N A ' JAH QC7042 \ / N N O ”WHwOA/OWHWNJLPSQOZH 0 co H2 O O Qco7o43 N HzNXNHNfiNjflNH K N/ COzH EXAMPLE. FA—PEG12—EDA—NH2—NOTA (QCO7043). To FA—PEG12— EDA—NHz (QCO7042, 4.78 mg, 0.004 mmol, M.W.:1082.5) in DMSO (0.25 ml, with a concentration at 0.025 M) was added NOTA-NHS (3.5 mg, 0.005 mmol, 1.2 eq.) ed by DIPEA (2.7 ”L, 0.039 mmol). The whole mixture was stirred at 23 OC and red by LC- MS. 4 hours later, LC—MS showed that almost all of the starting material was transformed to the product. The crude material was then purified by preparative RP-HPLC to afford the pure FA—PEG12—EDA-NH2—NOTA (QCO7043, 4.09 mg, 68 %). Analytical RP-C18 HPLC: tR = 6.21 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 30% B in min); Preparative RP-Clg HPLC: tR = 15.60 min (A = 10 mM NH4OAC, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 30% B in 30 min); UV-Vis: kmax = 280 nm; LC—MS: LC— MS (Agilent G6130B Quadrupole LC/MS) of t Mobile phase: Buffer (pH 7)—CH3CN; Method: 0——8.00 (1n, 1 H), 7.55 (d, J: 6.4 Hz, 1 H), 7.54 (3, br, 2 H), 6.81—6.93 (m, 1 H), 6.62 (d, J = 8.0 Hz, 2 H), 4.45 (d, J = 4.4 Hz, 2 H), 3.95—4.03 (m, 1 H), 3.64-3.70 (m, 2 H), 3.56—3.63 (m, 6 H), 3.38—3.50 (m, 28 H), 3.33—3.36 (m, 6 H), 3.20—3.24 (m, 4 H), 3.09—3.18 (m, 10 H), 3.04-3.09 (m, 4 H), 2.50 (s, 12 H, overlapping with the residue peak of DMSO), 2.27—2.34 (m, 2 H), 2.02—2.12 (m, 2 H), 1.99—2.01 (m, 2 H).
N o O gozH H \/\O/\/ \/\O O N NM“_< >‘ N/—l\\l/—<OH N @HW O </N\7 +5: folate--C-NETA I jflu H2N \N N/ Oi) E. C-NETA and folate-C—NETA. A PyBOP promoted coupling between QC04018 and compound 6, followed by deprotection of tert-butyl ester with TFA, provided folate-C-NETA. The folate-C-NETA is used to evaluate the labeling efficiency with A118F and 68Ga and evaluate the in vivo PET imaging. 0 O OMe OMe F MeZN CM CM QC07002 EXAMPLE. Methyl 3-cyano(dimethylamino)benzoate 02)[1]. To a stirred solution of methyl 3-cyanofluorobenzoate (5 g, 27.9 mmol) in DMSO (6 ml) was added dimethylamine hydrochloride (2.75 g, 33.7 mmol) ed by potassium carbonate (8.1 g, 58.6 mmol). The reaction mixture was stirred at room temperature ght and trated. The residue was dissolved in dichloromethane (50 ml) and washed with water (2 X 25 m1), brine, dried over NazSO4 and concentrated in vacuo to give the methyl 3—cyano— 4—(dimethylamino)benzoate (QC07002) in quantitative yield and was used without further purification. 0 o ’ (B MeZN Me3N CN CF 860 ON QC07002 3 3 0607003 EXAMPLE. 2-Cyano(methoxycarbonyl)—N,N,N—trimethylbenzenaminium trifluoromethanesulfonate (QC07003). To a stirred solution of methyl 3-cyano (dimethylamino)benzoate (3.4 g, 16.7 mmol) in anhydrous dichloromethane (17 ml) was added methyl trifluoromethanesulfonate (10 g, 60.9 mmol, M.W. 164.1) dropwise. The reaction was stirred at RT for 16 h and another portion of methyl trifluoromethanesulfonate (10 g, 60.9 mmol, M.W.: 164.1) was added. The reaction was stirred for another 16 hours and tert-butylmethylether (20 ml) was added slowly. The suspension was filtered and the collected solid was washed with tert-butylmethylether. The crude product was purified by RP—C18 HPLC: (acetonitrile/water-gradient 1:99 to 80:20) to afford product QC07003 (3.69 g) in 60 % yield. Analytical RP-Clg HPLC: tR = 0.49 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B in 15 min); kmax = 275 nm; LC-MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH 7)-CH3CN; Column: ic C13 column;Method: 0-100 CH3CN-15 min, tR = 0.49 min. MS m/z: MS-API: Calcd. for C12H15N202 ([M]+): 219.1, Found: 219.0; 1H NMR (400 MHz, D20) 5 = 8.67 (d, J=2.1 Hz, 1 H), 8.44 (dd, J=9.1, 2.1 Hz, 1 H), 8.15 (d, J=9.1 Hz, 1 H), 3.93 (s, 3 H), 3.87 (s, 9 H) ppm. 0 0 OMG OH (+) —> 9 MegN MegN e 9 C" CN CF3SO3 CF3803 QC07003 QC07004 EXAMPLE. 4-Carboxycyano-N,N,N-trimethylbenzenaminium oromethanesulfonate (QC07004). A solution of QC07003 (3.6 g, 9.8 mmol) in water (83 ml) and TFA (83 ml) was heated at 120°C for 48 h. The reaction e was concentrated in vacuo, the light green oil was treated with diethylether to result a suspension.
This solid was collected by filtration, washed with diethylether and dried in vacuo to give 4— carboxycyano-N,N,Ntrimethy1benzenaminium oromethanesulfonate 4 (2.8 g, 82%). Analytical RP-CIS HPLC: U; = 0.61 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B1n 15 min); Mm: 240 nm. LC-MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH 7)-CH3CN; : Analytic C18 column; Method: 0-100 CH3CN 15 min, tR = 0.61 min. MS m/z: MS-API: Calcd. for C11H13N202 ([M]+): 205.1, Found: 205.1; 1H NMR (400 MHZ, DMSO) 5 = 8.58 (d, J = 2.07 Hz, 1 H), 8.39 — 8.49 (m, 1H), 8.23 - 8.35 (m, 1 H) 3.85 (s, 9 H).
COZMG @010 0.00wN\/\ /\/O\/\o e Me3N BACOCFS Qco7o11 CF3803 4 o gone o H e :qu N/\/\n/N\/\o/\/O\/\N CF3803 o @ COCF3 CN Qco7oos EXAMPLE. FA—PEGi-TMA Precursor (QC07005). QC07004 (62 mg, 0.17 mmol) was added to the solution of QC07011 (0.14 mmol) and DIPEA (87 “L, 1.75 mmol) in DMSO (2.0 mL) at 23 0C under N2. After being stirred for 15 min at 23 OC, PyBOP (91 mg, 0.17 mmol) was added, and the reaction mixture was stirred for 24 h at 23 0C. Volatile material was removed under reduced vacuum to afford the crude product which was r purified by RP-HPLC (C13) to afford the pure compound QC07005 as pale yellow colored solid (125.1 mg, 72 %). Analytical RP-Clg HPLC: tR = 4.17 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B in 15 min); Max: 280 nm; LC-MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: Buffer (pH CN; Column: ic C18 column; Method: 0—100 CH3CN—15 min, ti: = 4.17 min. MS m/z: MS—API: Calcd. for C28H35F3N908 ([M]+): 868.3, Found: 868.2. oN/"\/\n/N\/\o/\/O\/\No902Me 6 CF3303 ”N 0|:j/\N QC07005 NMes COCF3 CN )iNN/\/”1(N\/\o/\/O\/\HJkI;:LF0007006 CN LE. General procedure for the one—pot 19F— introduction and deprotection. 8.3 pL of freshly prepared KF—Kryptofix (1/ 1.5) (0.0012 mmol, 0.144 M) solution was azeotropically dried with CH3CN at 90-100 0C, to which 1.2 mg (0.0012 mmol, 1.0 equiv.) QC07005 in 50 ul of anhydrous DMSO was added with a concentration of precursor at 0.024 M. The resulting mixture was immediately immersed into an oil bath ted to 70-75 0C and kept at 70-75 0C for 10 min. After being cooled down to room temperature, 200 111 of 1M NaOH (aq.) was added with a concentration of NaOH (aq.) at 0.8 M. The on was monitored by LC—MS and found complete after 5 min, which was neutralized with 1 M HCl (aq.) and ed by LC—MS (QC07006). And the total labeling efficiency was about 30 % based on the analysis of LC—MS. Analytical RP-C18 HPLC: A = mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B in 15 min; km“: 280 nm; LC—MS: Method: 0—100 CH3CN—15 min, tR = 5.13 min. MS m/z: MS-API: Calcd. for C36H37F4N1009 ([M+H]+): 829.3, Found: 829.1. 0 002m COZH Wow:@WFA-Tris 0002024 3 :flOiN/\/\n/N“1460‘0/80/ HZNA HFolate—Tris-Borate KF/ Kryptofix 222 oim{..002H —> Hle-fiLJENNj/\NFolate-Tris— Borate-18F PET Agent EXAMPLE. Folate—lSF-Boronate PET Imaging Agent PSMA TARGETED EXAMPLES HCI o H H NH2 #0 YNJOJ<i 1) rophenyl chloroformate, o 2) H-Lys(Z)-OtBu, DIPEA, O O I 1)Pd/CH2 MeOH 2) 4--Nitropheny| chloroformate, DIPEA, DCM C31H48N4011EC1380HN\©\ NO2 Exact Mass: 652.33 Mol. Wt.: 652.73 EXAMPLE. EC1380, 10. In a dry flask, H—Glu(OtBu)—OtBu.HCl (2.48g , 8.41 mmol) and 4—nitrophenyl formate (1.86 g, 9.25 mmol, 1.1 eq) were added, dissolved in CH2Clz (30ml) under Argon atmosphere. The stirring solution was chilled to 0 °C, followed by the dropwise addition of DIPEA (4.50 ml, 25.2 mmol, 3 eq). The reaction mixture was allowed to warm to room temperature and stirred for 1 hr. To the stirring solution was added H-Lys—(Z)-O‘Bu (4.39 g, 11.8 mmol, 1.4 eq), DIPEA (4.50 ml, 25.2 mmol, 3 eq) and stirred for 1 hr. Upon completion, the reaction was quenched with saturated NaHCOs and extracted with CH2C12 three times. The organic extracts were combined, dried over NazSO4, filtered and the solvent was removed via reduced pressure. The product was purified using silica gel tography with petroleum ether and ethyl acetate. The Cbz protected amine was erred to a round bottom flask with 10% Pd/C (10% wt eq), dissolved in MeOH (30 ml) under Hydrogen atmosphere (latm) and stirred for 3 hr. Upon completion, the reaction mixture was filtered through celite and the t was removed Via reduced pressure to yield the crude amine. The amine was taken up in CH2C12 (30ml) under Argon atmosphere and chilled to 0 °C. To the chilled on was added 4-nitropheny1 chloroformate (2.2 g, 10.9 mmol, 1.3 eq) and DIPEA (6.0 ml, 33.6 mmol, 4 eq) subsequently and stirred for 2 hr at room temperature. The reaction mixture was quenched with saturated NH4Cl and extract three times with ethyl acetate. The c extracts were combined, dried over Na2804, filtered, and solvent was removed under vacuum and purified using silica gel chromatography to yield the desired activated amine, EC1380 (2.54 g, 46%). 1‘0 Oyoj< 0k NHMtt 'JL JOL Worm NW N 0 (:3 O EXAMPLE. Glu(O‘Bu)—OtBu—Lys—O‘Bu—AMPAA—Asp(O‘Bu)—Asp(O’Bu)— Lys(Mtt)-resin 11. The general procedure bed for the synthesis of resin bound folate- peptide resin 1 was followed for the ng of 2 X Fmoc—L-Asp(O‘Bu)-OH, Fmoc- AMPAA-OH, Fmoc-L-Lys(Z)-O‘Bu, and Fmoc-(L)—Glu(O‘Bu) to Fmoc-L—Lys(Mtt)—Wang resin. The resin bound penta-peptide was subjected to rd Fmoc deprotection, washings and Kaiser test. Following another DMF wash (3 x 10 m1); an EC1380 on (2.0 eq.) in DMF, and DIPEA (3.0 eq.) were added to the vessel and the solution bubbled with Argon for 2 hour. The coupling solution was filtered, the resin was washed with DMF (3 x 10 m1) and i- PrOH (3 x 10 ml) and a Kaiser test was done to assess reaction completion.
O 902H 0 H020“) \ /\:JLW JL COZH COZH H020 N N N N O 902H H H H H NEJiNHWHJL H00)2 0 '\ E02209 CS6H78N1ZOZSS Exact Mass: 1318.50 MOI. Wt.: 1319.35 EXAMPLE. Glu-Lys-AMPAA-Asp-Asp-Lys-Bn-NOTA 12. Glu-Lys- AMPAA—Asp-Asp-Lys-Bn-NOTA, EC2209 was prepared in 47% yield according to the process bed for folate-peptide-NOTA, 4. 1H NMR (500 MHZ DMSO—dg) l signals: 5 7.25 — 7.18 (m, 2H), 7.14 (d, J: 8.1 Hz, 1H), 7.12 — 7.06 (m, 5H), 4.47 (ddd, J: 17.8, 7.5, 5.6 Hz, 2H), 4.11 — 4.08 (m, 3H), 4.08 — 4.02 (m, 2H), 3.98 (dd, J: 8.2, 5.1 Hz, 1H). [M+H]+ = Calculated 1319.50, found 1319.70 beHNYNH beHNY NWNJQENHE):\Og/éNH—{ENHfifg:NHMtt COQtBUOE \ngtBL?OIW ButOZC\ N N COgtBu EXAMPLE. Bu)—O‘Bu—Lys—OtBu—Aoc—Phe—Phe—Arg(be)—Asp(OtBu)— Arg(be)—Lys(Mtt)—resin 13. The general procedure described for the sis of resin bound folate-peptide resin 1 was followed for the coupling of —Arg(be)-OH, Fmoc- L—Asp(O’Bu)-OH, Fmoc—L—Arg(be)-OH, 2 X Fmoc-Phe—OH, Fmoc-Aoc—OH, Fmoc-L- Lys(Z)-O’Bu, Fmoc-(L)-Glu(OtBu) and ECl380 to Fmoc-L—Lys(Mtt)-Wang resin.
H2NYNH HZNVNH NH NH O O O 001.111;an “VLuJi'VLuH H H H l—\ “MNrN NAC02H H0201. O \O O \COZHO 002” O <jNJ HNYN H020 0 COZH EC2390 C73H114N20023 Exact Mass: 4 Mol. Wt.: 1639.81 EXAMPLE. Glu—Lys-Aoc—Phe-Phe-Arg-Asp-Arg—Lys-NOTA 14. Glu-Lys— Aoc-Phe-Phe-Arg—Asp-Arg-Lys-NOTA, EC2390 was prepared in 37% yield according to the process described for folate-peptide-NOTA, 4. 1H NMR (500 MHZ DMSO-ds) Pivotal signals: 6 7.25 — 7.14 (m, 6H), 7.16 — 7.08 (m, 3H), 4.47 (dd, J = 9.0, 4.7 Hz, 1H), 4.42 (t, J: .9 Hz, 1H), 4.36 (dd, J: 10.4, 4.4 Hz, 1H), 4.27 (t, J: 6.9 Hz, 1H), 4.16 (t, J: 5.6 Hz, 1H), 3.97 — 3.88 (m, 2H). [M+H]+ = Calculated 1639.84, found 1640.22 000:3” COOBn COO’Bu COOH COOtBu a i 6" i HOOC NH2.HC| {BuOOC N N i COO’Bu tBuOOC i ’ H H H H H H H H H COO Bu 1 2 DUPA_1 Reagents and conditions: (a) triphosgene, TEA/ DCM, -78 °C; (b) H-L-GIu(OBn)-OtBuHC|; (0) H2; Pd-C/DCM.
EXAMPLE. DUPA-EAOA—Phe-Arg-Lys-NHz, 2-[3—(3-Benzyloxycarbonyl- 1—tert—butoxycarbonyl-propyl)-ureido]pentanedioic acid di—tert-butyl ester (2). [1, 2] To a solution of L-glutamate t-butyl ester hydrochloride 1 (1.0 g, 3.39 mmol) and triphosgene (329.8 mg, 1.12 mmol) in DCM (25.0 mL) at —78 OC, tn'ethylamine (TEA, 1.0 mL, 8.19 mmol) was added. After stirring for 2 h at —78 0C under argon, a solution of L- Glu(OBn)—OtBu (1.2 g, 3.72 mmol) and TEA (600 ,uL, 4.91 mmol) in DCM (5.0 mL) was added. The reaction mixture was allowed to come to room temperature (rt) over a period of 1 h and stirred at ambient temperature overnight. The reaction was quenched with 1 M HCl, and the organic layer was washed with brine and dried over NazSO4. The crude product was purified using flash chromatography (hexane:EtOAc ) 1:1) to yield the ediate 2 (1.76 g, 90.2%) as a colorless oil and crystallized using hexane:DCM. Rf) 0.67 (hexane:EtOAc ) 1:1). 1H NMR (CDClg): 6 1.43 (s, 9H, CH3—tBu); 1.44 (s, 9H, CH3—tBu); 1.46 (s, 9H, CH3—tBu); 1.85 (m, 1H, Glu—H); 1.87 (m, 1H, Glu—H); 2.06 (m, 1H, Glu-H); 2.07 (m, 1H, Glu—H); 2.30 (m, 2H, ; 2.44 (m, 2H, Glu—H); 4.34 [s (broad), 1H, RH]; 4.38 [s (broad), 1H, R-H]; .10 (s, 2H, CH2—Ar); 5.22 [s (broad), 2H, Urea-H); 7.34 (m, 5H, Ar-H). EI-HRMS (m/z): (M + H)+ calcd for C30H47N209, 82; found, 89.
EXAMPLE. 2-[3-(1,3-Bis—tert-butoxycarbonyl-propyl)-ureido]pentanedioic Acid 1-tert-Butyl Ester, DUPA_1. To a on of 2 (250 mg, 432 mmol) in DCM, 10% Pd/C was added. The reaction mixture was enated at 1 atm for 24 h at rt. Pd/C was filtered through a Celite pad and washed with DCM. The crude product was purified using flash chromatography (hexane: EtOAc ) 40:60) to yield DUPA_1 (169 mg, 80.2%) as a colorless oil, and crystallized using hexanezDCM. Rf: 0.58 (hexane: EtOAc = 40:60). 1H NMR (CDCl3): 6 1.46 (m, 27H, CH3— tBu); 1.91 (m, 2H,Glu—H); 2.07 (m, 1H, Glu—H); 2.18 (m,1H, Glu-H); 2.33 (m, 2H, Glu-H); 2.46 (m, 2H, ; 4.31(s (broad), 1H, RH); 4.35 (s (broad), 1H, R-H); 5.05 (t, 2H,Urea-H); EI-HRMS (m/z): (M + H)+ calcd for C23H41N209,489.2812; found, 489.2808.
H2N NH DUPA-EAOA-Phe-Arg-Lys-NH2 Reagents and conditions: (a) (i) 20 idine/DMF,room temperature,10min;(ii)Fmoc— Arg(Boc)2-OH, HBTU, HOBt, DMF-DIPEA, 2h. (b) (i) 20%piperidine/DMF,room temperature,10min; (ii) Fmoc-Phe—OH, HBTU, HOBt, DMF-DIPEA, 2h. (c) (i) 20% piperidine/DMF, room temperature,10min; (ii) Fmocamino- octanoic(EAO)acid,HBTU,HOBt,DMF/DIPEA,2h. (d) (i) 20% dine/DMF, room temperature,10min; (ii) (tBuO)3-DUPA-OH, HBTU, HOBt, DIPEA, 2h. (e) TFA/H20/TIPS (95:2.5:2.5),1h EXAMPLE. DUPA—EAOA—Phe-Arg-Lys-NHz. Fmoc-Lys(Boc)-Wang resin (0.43 mM) was swollen with DCM (3 mL) followed by yl formamide (DMF, 3 mL). A solution of 20% piperidine in DMF (3 x 3 mL) was added to the resin, and argon was bubbled for 5 min. The resin was washed with DMF (3 x 3 mL) and isopropyl alcohol (1'- PrOH, 3 x 3 mL). Formation of free amine was ed by the Kaiser test. After swelling the resin in DMF, a solution of Fmoc-Arg(Boc)2-OH (2.5 equiv), HBTU (2.5 equiv), HOBt (2.5 , and DIPEA (4.0 equiv) in DMF was added. Argon was bubbled for 2 h, and resin was washed with DMF (3 x 3 mL) and i-PrOH (3 x 3 mL). The coupling efficiency was assessed by the Kaiser Test. The above sequence was repeated for 3 more coupling steps to introduce the phenylanaline (Phe), 8—amino-octanoic acid (EAO), and DUPA successively. Final compound was cleaved from the resin using a trifluoroacetic acid (TFA):HzO:triisopropylsilane cocktail (95:25:25) and trated under . The concentrated product was precipitated in cold diethyl ether and dried under vacuum. The crude product was purified using preparative RP-HPLC [(11) 210 nm; solvent gradient: 0% B to 50% B in 30 min run; mobile phase: A) 0.1 % TFA, pH = 2; B) acetonitrile (ACN)]. ACN was removed under vacuum, and pure fractions were —dried to yield DUPA-EAOA- Phe-Arg-Lys—NHz as a white solid. UV/vis: lmax = 205 nm. Analytical C: tR = 6.2 min (A = 0.1 % TFA; B = CH3CN, solvent gradient: 0% B to 50% B in 15 min); ESI-MS (m/z): (M + H)+ calcd for C40H65N10013, 893.5; found, 893.4. co H2 n O JL O—N HOZC N N COZH N N H H A + J o H2N NH N O DUPA-EAOA-Phe-Arg-Lys-NH2 (ocosoo1) HO 0)2 Chemical Formula: C40H64N1oo13 Exact Mass: 892.5 Molecular Weight: 893.0 H2N NH QC08002 Chemical Formula: N13013 Exact Mass: 1177.6 Molecular Weight: 1178.3 EXAMPLE. DUPA-EAOA-Phe—Arg—Lys—NHz—NOTA. To DUPA—EAOA— Phe—Arg-Lys—NHz (QC08001, 5.0 mg, 0.0056 mmol, M.W.:893.0) in DMSO (0.20 ml, with a concentration at 0.028 M) was added NOTA-NHS (5.5 mg, 0.0084 mmol, 1.5 eq.) followed by DIPEA (2.9 uL, 0.017 mmol). The on was stirred at 23 OC, monitored by LC—MS, and most of the starting material was transformed to the corresponding product in 5 hours.
The crude material was purified by RP—C13 HPLC. ACN was removed under vacuum, and pure fractions were freeze-dried to yield the pure AOA-Phe-Arg-Lys-NH2—NOTA (QC08002, 3.3 mg, 50 %). ical RP-CIS HPLC: tR = 5.98 min (A = 0.1 % TFA; B = CH3CN, solvent gradient: 0% B to 50% B in 15 min); Preparative RP—C18 HPLC: tR = 16.16 min (A = 0.1 % TFA; B = CH3CN, solvent gradient: 0% B to 50% B in 30 min); UV-Viszkmax = 201 nm; HPLC (Agilent Preparative C18 Column): Mobile phase: A = 0.1 % TFA; B = CH3CN; Method: 0-50 CH3CN-30 min, tR = 16.16 min LC—MS (Agilent G6130B Quadrupole LC/MS): Mobile phase: A = 0.1 % TFA; B = CH3CN; Method: 0-50 30 min, tR = 5.98 min; MS m/z: MS-API: Calcd. for C52H84N13018 ([M+H]+): 1178.6, Found: 1178.4.
O s COZH E EL Jk0 f KCOZH H020 N M COZH HN HZN/kNH EXAMPLE. DUPA-EAOA-Phe—Arg—Lys—NH2—NOTA—A118F. Method a):.
DUPA-EAOA-Phe-Arg-Lys-NH2—NOTA is dissolved in 2 mM NaOAc (pH 4.5) and 0.5 mL of ethanol, and treated with A118F3-3H20 (1.5 eq.) which is y ed before application. The pH is ed to 4.5-5.0, and the reaction mixture is refluxed for 15-30 min with pH kept at 45-50. After cooling to room temperature, the crude material is loaded to a cartridge, and the radiotracer eluted into vial. After sterile filtration and being diluted to appropriate radioactivity (5-10 mCi) and specific activity (> 1 Ci/umol), the racer is used in in vivo PET imaging.
Method b). DUPA—EAOA-Phe-Arg-Lys—NHz—NOTA is dissolved in 2 mM NaOAc (pH 4.5), and treated with A1Cl3-3H20 (1.5 eq.). The pH is ed to 4.5-5.0, and the reaction mixture is refluxed for 15-30 min with the pH kept at 45-50. The crude material is purified by RP-HPLC to afford the DUPA-EAOA-Phe-Arg-Lys-NHz-NOTA-A1—OH intermediate ready for 18F- labeling. riate amount of DUPA-EAOA-Phe-Arg-Lys- NHz—NOTA—Al—OH is d with Na18F saline solution and ethanol (1/ 1, v/v), and the whole mixture is heated at 100-110 0C for 15 min. After cooling to room temperature, the crude material is loaded to a cartridge, and the racer eluted into vial. After e filtration and being diluted to appropriate radioactivity (5-10 mCi) and specific activity (> 1 Ci/umol), the radiotracer is ready for use in in vivo PET imaging. 0 (e)H02C c02H DUPAEAOAPhePheEDA.NH2 Trt-EDA Resin Reagents and conditions: (a) Fmoc-Phe-OH, HBTU, HOBt, DMF/DIPEA, 2h. (b) (i) 20 %piperidine/DMF,room temperature,10min; (ii) Fmoc-Phe—OH, HBTU, HOBt, DMF/DIPEA, 2h. (c) (i) 20%piperidine/DMF,room temperature,10min; (ii) Fmoc-S—amino- octanoic (EAO) acid, HBTU, HOBt, DMF/DIPEA, 2h. (d) (i) 20% piperidine/DMF, room temperature,10min; (ii) (tBuO)3-DUPA-OH, HBTU, HOBt, DIPEA, 2h. (e) TFA/HzO/TIPS (95:2.5:2.5),1h.
EXAMPLE. Solid Phase Peptide Synthesis (SPPS) of AOA—Phe- A—NH2.[2, 3]. As described herein for AOA-Phe-Arg—Lys—NH2 01), DUPA-EAOA-Phe—Phe-EDA-NH2 is preapred. The commercially available Trt-EDA resin was swollen with DCM (3 mL) followed by dimethyl formamide (DMF, 3 mL), to which a solution of Fmoc-Phe-OH (2.5 equiv), HBTU (2.5 equiv), HOBt (2.5 equiv), and DIPEA (4.0 equiv) in DMF was added. Argon was bubbled for 2 h, and resin was washed with DMF (3 x 3 mL) and i-PrOH (3 x 3 mL). The ng efficiency was assessed by the Kaiser Test. A solution of 20% piperidine in DMF (3 x 3 mL) was added to the resin, and argon was bubbled for 5 min. The resin was washed with DMF (3 x 3 mL) and isopropyl alcohol (1'- PrOH, 3 x 3 mL). Formation of free amine was assessed by the Kaiser test. The above sequence was repeated for 3 more coupling steps to introduce the second phenylanaline (Phe), 8-amino-octanoic acid (EAO), and DUPA successively. Final compound was cleaved from the resin using a trifluoroacetic acid (TFA):H20:triisopropylsilane cocktail (95:2.5:2.5) and concentrated under vacuum. The concentrated product was precipitated in cold diethyl ether and dried under vacuum. The crude product was ed using preparative RP-HPLC [/1] 210 nm; solvent gradient: 0% B to 100% B in 30 min run; mobile phase: A) 10 mM NH4OAc (pH = 7, buffer); B) acetonitrile (ACN)]. ACN was removed under vacuum, and pure fractions were freeze—dried to yield AOA-Phe—Phe-EDA-NH2 as a white solid.
Analytical RP—C13 HPLC: tR = 3.99 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B in 15 min); Preparative RP-C18 HPLC: tR = 16.05 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, t gradient: 0% B to 100% B in 30 min); UV-Vis: kmax = 209 nm; LC-MS: LC-MS (Agilent G6130B pole LC/MS) of Product Mobile phase: Buffer (pH 7)-CH3CN; Method: 0-100 ACN-15 min, tR = 3.99 min. MS m/z: MS-API: Calcd. for C39H56N7011([M+H]+): 7984, Found: 798.3; Calcd. for C39H55N7O11K ([M+K]+): 836.4, Found: 836.3. HPLC (Agilent Preparative C18 Column): Mobile phase: Buffer (pH 7)-CH3CN; Method: 0—100 ACN-30 min, tR = 16.05 min. o o Hozcfil \ o—N O “W H2 N N/\g/ H I H + <1JO 0 J HPFe-TFA JL U H020MoecuaI I rW'eig ht" 660 38. .
H020 m M COzH DUPA-EAOA-Phe-Phe-EDA-NHZ QC08008 Chemical Formula: C39H55N7O11 Exact Mass: 797.4 Molecular Weight: 797.9 O O H H F”\/\/\/\)L i/\NHKCOZ DIPEA u 0 “MNHV” NNJ_ ’ 0 DMSO JL \\cozH HOZC COZH ocosooa U n u Chemical Formula: C51H74N1OO16 Exact Mass: 1082.5 Molecular Weight: 1083.2 EXAMPLE. To DUPA—EAOA—Phe-Phe—EDA—NH2 (QC08008, 5.9 mg, 0.0074 mmol, M.W.:797.4) in DMSO (0.25 ml, with a concentration at 0.025 M) was added NOTA—NHS (7.3 mg, 0.011 mmol, 1.5 eq.) followed by 4 drops of DIPEA. The mixture was stirred at 23 OC and monitored by LC-MS. 4 hours later, LC—MS showed that almost all of the starting material was transformed to the product. The crude al was then purified by preparative RP-HPLC to afford the pure DUPA-EAOA-Phe—Phe-NOTA (QC08009, 4.50 mg, 56 %, based on 8.02 mg in theory, 97 % purity by HPLC at 210 nm). Analytical RP-C18 HPLC: tR = 3.45 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B in 15 min); Preparative RP-CIS HPLC: tR = 10.09 min (A = 10 mM NH4OAc, pH = 7.0; B = CH3CN, solvent gradient: 0% B to 100% B in 30 min); UV-Vis: max = 211 nm; LC-MS: LC-MS (Agilent G6130B Quadrupole LC/MS) of Product Mobile phase: Buffer (pH CN; Method: 0-100 ACN—15 min, tR = 3.45 min. MS m/z: MS-API: Calcd. for C51H75N10016 ([M+H]+): 1083.5, Found: 1083.3; HPLC (Agilent Preparative C18 ): Mobile phase: Buffer (pH 7)-CH3CN; Method: 0—100 ACN-30 min, tR = 10.09 min. 1H NMR (400 MHz, DMSO—d6) 5 = 10.13 (br, 1 H), 8.98 (br, 1 H), 8.43 (br, 1 H), 7.90 (br, 3 H), 7.30— 7.10 (m, 10 H), 6.37 (br, 1 H), 6.28 (br, 1 H), 4.60—4.52 (m, 1 H), 4.32—4.44 (m, 1 H), 4.24— 4.31 (m, 2 H), .03 (m, 2 H), 3.85—3.92 (m, 2 H), 3.28 (s, 4 H), 3.25 (s, 2 H), 3.09 (m, 1 H), 3.05 (m, 1 H), 2.92—3.02 (m, 4 H), 2.54-2.67 (m, 12 H), 2.31—2.38 (m, 2 H), 2.19-2.31 (m, 3 H), 2.11—2.18 (m, 2 H),2.02—2.10 (m, 3 H), 1.52-1.72 (m, 4 H), 1.25—1.37 (m, 4 H), 1.05— 1.13 (m, 2 H), o o COZH 0 fl co H F”H\/\/\/\)L ? r 2 COZH N N$NWNJLN H ; H H Of NNJ \\COZH A DUPA-EAOA-Phe-Arg-LyS-NOTA N N H QC08004 64CuCIZ-NH4OAc pH 5.5, 95 00 o o COZH 0 m 00 H HM H ? r 2 COzH FN NJNWNJLN s HOZC N N COZH HN H H A DUPA-EAOA-Phe-Arg-Lys-NOTA-B‘Cu H2N NH EXAMPLE. Radiochemical Synthesis of DUPA-EAOA-Phe—Arg-Lys- NOTA-“Cu Radiotracer. NOTA based chelators have also been ed and ed in the formulation of NOTA—64/67Cu for nuclear ne/radiotherapy.[14-16] The corresponding DUPA-NOTA-64Cu was prepare for the dual purpose of imaging and therapy, also referred to as ostics. DUPA-EAOA-Phe—Arg-Lys—NOTA—64Cu was prepared according a standard protocol with minor modifications. [4, 14-16] The 64Cu(OAc)2, in situ ed from z with 0.1 M ammonium acetate (pH 5.5),was added to the reaction tube containing the DUPA-NOTA precursor. The resulting mixture was then heated to 95 0C for 15 min. After cooling to room temperature, the crude material was purified by radioactive HPLC on a C18 column using MeCN and 0.1% TFA as the mobile phase to afford the target radiotracer with ~ 90% radiochemical purity (RCP). Sterile filtration and dilution in isotonic saline to the desired radioactivity provided the radiotracer ready for PET imaging.
COZH i H020 NJLN COzH DUPA-EAOA-Phe-Phe-NOTA H H (Qcosoos) H COZH E H020EarN COZH H M = 68Ga, 64Cu or Al-13F DUPA-EAOA-Phe-Phe-NOTA-“CulAl-“F EXAMPLE. Radiochemical sis of DUPA-EAOA-Phe-Phe-NOTA- 64Cu/Al—13F. o N COZH ; o o o \\ JP COZH DU PA-EAOA-Phe-Phe-NOTA H (0008009) U COZH F HOZC N N COZH H H DUPA-EAOA-Phe-Phe-NOTA-“Ga EXAMPLE. Radiochemical synthesis of DUPA-EAOA-Phe-Phe-NOTA- 68Ga.
EXAMPLE. l procedure for 68Ga labeling: 68Ga was eluted from the 68Ge/68Ga generator with 0.1N HCl. A predetermined amount of 68'Ga in 0.1N HCl was added to a DUPA-NOTA on in acetate buffer (pH 4.8). The labeling mixture was incubated at room temperature, and labeling efficiencies were d by radioactive HPLC. The radiolabeled product was purified by radioactive HPLC and the DUPA-NOTA-68Ga peak sample was collected. After sterile filtration and being diluted to appropriate radioactivity (5- 10 mCi) and specific activity (> 1 l), the radiotracer was ready for in vivo PET imaging study.
COZH E H02C N N COzH H H DUPA-C-NETA DUPA-C-NETA-M OH M = AI-13F, 68Ga, 1”Lu or say EXAMPLE. Radiochemical synthesis of DUPA-C-NETA based theranostics. 51) Qcosoos.
, DIPEA; b) TFA HgLfiNHNHQfdl—Lfio”o COZH FHW ° “ H020 ”k” COZH DUPA-C-NETA 0?: EXAMPLE. ation of the NOTA Derivatives. Bifunctional conjugates, also referred to as theranotics, are described herein. Compounds described herein can tightly chelate both radionuclides such as 18F and 68Ga for PET imaging, and radionuclides 177Lu and 90Y for radiotherapy. C—NETA, a NOTA derivative, has been reported to e AllSF with about twice the efficiency (87%) of 17] Moreover, C-NETA also reportedly chelates the commonly used radiotherapeutic es, such as 177Lu and 90Y, with high labeling ency.[18] Thus, it is appreciated , that C—NETA is useful as a bifunctional chelator that can be used for both PET imaging and radiotherapy, where the radionuclide is a metal or metal halide, such as A118F, 68Ga, 177Lu or 90Y.
EXAMPLE. A PyBOP promoted coupling between QC04018 and QC08008, followed by deprotection of tert-butyl ester with TFA provides DUPA-C-NETA. DUPA-C- NETA is used to evaluate the labeling efficiency to AllSF, 68Ga, 177Lu and 90Y, and evaluate the in viva PET imaging and radiotherapy.
METHOD EXAMPLES EXAMPLE. The specificity of the radionuclide containing conjugates binding to FR is evaluated against KB xenografts homogenates and Cal5l xenografts homogenates.
Concentration dependent binding was evaluated for F-QCO7017 and 18F—AIF- QC07043, and separated into specific and non-specific binding. Significant non-specific binding was not observed in KB homogenates. Minor non—specific binding was observed in Cal5l homogenates, with a specific/non-specific g ratio of >3:l at all trations up to about 30 nM for 18F—AIF—QCO7017, and a specific/non—specific binding ratio of >221 at all concentrations up to about 20 nM for 18F—AIF-QC07043. Minor non-specific binding was observed in A549 homogenates, with a specific/non-specific binding ratio of >2:l at all trations up to about 10 nM for 18F—AIF-QC07043. Scatchard analyses were also performed. Displaceable and saturable binding of 18F—AIF—QC07017 in human tumor xenografts (KB and Ca151) by self competition was observed. Both 18F-AIF—QCO7017 and F-QC07043 bound one site with high affinity in all cell xenografts. The high ratio of Bmax/Kd indicated a high specific g affinity to KB xenografts. Moderate binding affinity was observed for Ca151 afts, and the lowest binding affinity was observed for A549 xenografts. Without being bound by , it is believed herein that the moderate expression of FR in Ca151 xenografts accounts for the lower g affinity. g affinities of 18F—AIF-QCO7017 (2) to FR in KB and Ca151 tumor crude homogenate.
Folate-NOTA-A118F (2) Bmax / Kd Binding ties of 18F—AIF-QC07043 to FR in KB and Ca151 tumor crude homogenate.
FA-PEG12-NOTA-A118F Bmax/Kd EXAMPLE. uPET imaging was performed on nude mice bearing KB tumor xenografts under baseline and competed conditions to evaluate the in vivo binding specificity of 18F-AIF—QCO7017 (2) to FR. Nude mice bearing KB tumor xenografts on their left shoulder were injected with 0.30-0.40 mCi (2). The ed group received 100 pg of folic acid 10 min before the iv. injection of (2), and the ent group was injected with a corresponding volume of phosphate buffer. Time course inspection of PET images obtained at various time points revealed that the data acquired in 60—90 min post tracer injection gave the best visual PET imaging. The KB tumors were clearly visualized in the treated group, whereus the uptake of (2) was completely inhibited by competing with folic acid, ting a high specificity of (2) binding to FR in vivo. Without being bound by theory, it is believed herein that the high ctivity found in kidneys was due to the uptake mediated by FR that is expressed in the proximal tubule cells in kidneys and the potential accumulation of radiotracer via renal excretion, which was further supported by the biodistribution studies described herein. With the exception of the liver, significant uptake in other organs was not observed. A significant blocking effect in liver uptake was observed in under competed conditions.
E. Ex vivo tribution study of compounds described herein under both baseline and competed conditions in nude mice g KB tumor xenografts on their left shoulder demonstrates a high and specific uptake in FR(+) tumors. Radiotracer levels of 18F—AIF-QC07017 and 18'F—AIF-QC07043 were determined in whole blood, plasma, heart, kidney, liver, lung, muscle, spleen, KB xenograft tumor tissues and A549 xenograft tumor s (, , and ). The highest signal was observed in the kidneys. Accumulation was observed to a substantially less extent in the liver. Without being bound by theory, it is believed herein that the highest accumulation of radioactivity in kidneys, along with the relative low uptake of radiotracer in the hepatobiliary system i.e. liver, bile and ine/feces supports that renal elimination is the predominant ion pathway. Except for the kidneys, accumulation in KB xenograft tumor tissues was greatest, and significantly greater than in the liver. lation in A549 xenograft tumor tissues was comparable to the liver. Accumulation in both KB xenograft tumor tissues and A549 xenograft tumor tissues was blocked under competition conditions with folic acid ( and ). The FR specificity of 18F—AIF-QC07‘017 and 18F—AIF—QC07043 was comparable to latide (EC20), a compound in clinical trials, in both KB xenograft tumor tissues and A549 xenograft tumor tissues.
Uptake in KB xenografts Example Uptake Uptake under competed (SUV) conditions (i SEM) (SUV) (i SEM) 18F-AIF—QC07017 2.84 i 0.76 0.34 i 0.02 F—QC07043 2.33 i 0.13 0.41 i 0.06 99mTc—EC20 2.75 i 0.14 0.43 i 0.05 P values: 99mTc—EC20 vs 18F—AIF—QC07043, p=0.15; 18F—AIF-QC07017 vs 18F-AIF— QC07043, p=0.48; EC20 VS 18F—AIF—QC07017, p=0.85.
Uptake in A549 xenografts Example Uptake Uptake under competed (SUV) ions (i SEM) (SUV) (i SEM) 18F-AIF-QC07017 0.64 i 0.16 0.03 i 0.01 18F—AIF—QC07043 0.53 i 0.06 0.04 i 0.01 99mTc-EC20 0.71 i 0.09 0.08 i 0.02 P : 99mTc-EC20 vs l8F—AIF—QC07043, p=0.l3; 18F-AIF-QC07017 vs F— QC07043, p=0.50; 99mTc—EC20 vs 18F—AIF—QC07017, p=0.74 E. In vitro evaluation of DUPA-EAOA-Phe-Phe-NOTA-68Ga radiotracer (68Ga-QC08009). 67Ga has a longer half life than 68Ga (about 3.3 days versus about 68 minutes, respectively). Thus, 67Ga is used as a surrogate of 68Ga for in vitro evaluation of Kd values and tissue imaging. It is to be understood that the in vitro evaluation of Kd values and tissue imaging observed for 67Ga is predictive of 68Ga. DUPA-EAOA-Phe- Phe-NOTA-67Ga (67Ga—NOTA—LC-PSMA2) was prepared in nearly tative hemical yield. In vitro study in both the PSMA(-) cell line (PC3) and the PSMA(+) cell lines (LnCaP and PIP—PC3) ed a PSMA mediated high and specific uptake with a Kd = 8.45i2.l6 nM. PC3 is a PSMA (-) cell line; LnCap is a PSMA (+) cell line; and PIP- PC3 is a transfect cell line with higher PSMA expression. Uptake of 68Ga—QC08009 by PC3 cells was minimal, and did not change when competed. Uptake of C08009 by LnCaP and PIP-PC3 was substantial, with PIP-PC3 cells showing the highest uptake. In both cases, Uptake of 68Ga-QC08009 by LnCaP and PIP-PC3 is blocked by competing ligand.
Compared to 67Ga-DKFZ—PSMAll, an imaging agent in clinical trials, 67Ga-NOTA-LC— PSMA2 demonstrated superior binding to PSMA(+) prostate cancer tissues.
EXAMPLE. In Vivo PET imaging and BioD study of DUPA-EAOA-Phe-Phe- NOTA-63Ga radiotracer (63Ga-QC08009). In vivo micro-PET/CT scan with 68Ga-NOTA-LC— PSMA2 racer in mice carrying PSMA (+) LnCaP xenografts showed 4.29 %ID uptake in PSMA(+) tumor. At 1 hour post-injection, most of the racer was found in bladder.
Without being bound by theory, it is ed herein that the data support that the y elimination pathway is in urine. In addition, compared to other tissues, minor accumulation of radiotracer was observed in the kidneys. Without being bound by theory, it is believed herein that the relatively high PSMA expression in mouse kidneys, compared to other tissues, accounts at least in part for the minor accumulation of 68Ga—NOTA—LC—PSMA2 radiotracer in kidneys.

Claims (13)

WHAT IS CLAIMED IS:
1. A conjugate of the formula B-L-P or a pharmaceutically acceptable salt thereof, wherein B is a folic acid moiety, L is a divalent linker comprising a polyether, and P comprises a on-emitting isotope having a half-life of more than 80 minutes.
2. The conjugate of claim 1, comprising folate-Asp.
3. The conjugate of claim 1, comprising folate-Arg.
4. The conjugate of claim 1 wherein the linker ses a polypeptide comprising lysine, ne, or aspartic acid, or a combination thereof.
5. The conjugate of any one of claims 1 to 4, wherein the polyether is a polyethylene glycol (PEG).
6. The conjugate of any one of claims 1 to 5, wherein the positron-emitting e has a half-life of more than 90 minutes.
7. The conjugate of any one of claims 1 to 5, wherein the positron-emitting isotope has a half-life of more than 100 minutes.
8. The ate of any one of claims 1 to 5, wherein the positron-emitting isotope has a half-life of 80 minutes to 8 hours.
9. The conjugate of any one of claims 1 to 4, wherein the positron-emitting isotope is selected from the group consisting of 45Ti, 61Cu, 66Ga, and 18F.
10. The conjugate of claim 1 having the formula wherein n is from 1 to 20.
11. The conjugate of claim 1, comprising the formula
12. A composition comprising a ate according to any one of claims 1 to 11, and an acceptable carrier.
13. A dosage form comprising a conjugate according to any one of claims 1 to 11, in a solution. WO 73678 5E3.» :mmfim .63..
NZ719939A 2013-11-14 2014-11-13 Compounds for positron emission tomography NZ719939B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ758917A NZ758917A (en) 2013-11-14 2014-11-13 Compounds for positron emission tomography

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201361904387P 2013-11-14 2013-11-14
US201361904400P 2013-11-14 2013-11-14
US61/904,400 2013-11-14
US61/904,387 2013-11-14
US201361909822P 2013-11-27 2013-11-27
US61/909,822 2013-11-27
PCT/US2014/065467 WO2015073678A1 (en) 2013-11-14 2014-11-13 Compounds for positron emission tomography

Publications (2)

Publication Number Publication Date
NZ719939A NZ719939A (en) 2022-03-25
NZ719939B2 true NZ719939B2 (en) 2022-06-28

Family

ID=

Similar Documents

Publication Publication Date Title
US20220125958A1 (en) Compounds for positron emission tomography
US11344623B2 (en) Cholecystokinin B receptor targeting for imaging and therapy
ES2914593T3 (en) PSMA binding agents and uses thereof
ES2472423T3 (en) Imaging agents targeting the folate receptor
ES2972577T3 (en) Imaging and therapy targeting fibroblast activation protein (FAP)
EP3656403B1 (en) Preparation process of psma binding ligand-linker conjugates
AU2024203858A1 (en) Conjugates for treating diseases caused by PSMA expressing cells
AU2019225154A1 (en) Chemical conjugates of evans blue derivatives and their use as radiotherapy and imaging agents for targeting prostate cancer
NZ719939B2 (en) Compounds for positron emission tomography