WO2025042989A1 - Psma-based albumin binding agents for targeted radionuclide therapy of prostate cancer - Google Patents
Psma-based albumin binding agents for targeted radionuclide therapy of prostate cancer Download PDFInfo
- Publication number
- WO2025042989A1 WO2025042989A1 PCT/US2024/043231 US2024043231W WO2025042989A1 WO 2025042989 A1 WO2025042989 A1 WO 2025042989A1 US 2024043231 W US2024043231 W US 2024043231W WO 2025042989 A1 WO2025042989 A1 WO 2025042989A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- psma
- alb
- tumor
- cell
- compound
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K51/00—Preparations containing radioactive substances for use in therapy or testing in vivo
- A61K51/02—Preparations 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
- A61K51/04—Organic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K51/00—Preparations containing radioactive substances for use in therapy or testing in vivo
- A61K51/02—Preparations 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
- A61K51/04—Organic compounds
- A61K51/0402—Organic compounds carboxylic acid carriers, fatty acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K51/00—Preparations containing radioactive substances for use in therapy or testing in vivo
- A61K51/02—Preparations 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
- A61K51/04—Organic compounds
- A61K51/0497—Organic compounds conjugates with a carrier being an organic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/08—Drugs for disorders of the urinary system of the prostate
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07B—GENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
- C07B59/00—Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
- C07B59/008—Peptides; Proteins
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D257/00—Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
- C07D257/02—Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F5/00—Compounds containing elements of Groups 3 or 13 of the Periodic Table
- C07F5/003—Compounds containing elements of Groups 3 or 13 of the Periodic Table without C-Metal linkages
Definitions
- Prostate-specific membrane antigen is a tumor-associated antigen overexpressed in prostate adenocarcinoma cells, regardless of androgen status, in the neovasculature of solid tumors and has a low expression in benign and extra-prostatic tissues.
- PSMA Prostate-specific membrane antigen
- R 1 is selected from H, substituted aryl, substituted pyridine, and unsubstituted isoquinoline; ; arylene, and m is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; [0010] M is present or absent and when present is a metal or a radiometal; and stereoisomers and pharmaceutically acceptable salts thereof. [0011] In certain aspects, R 1 is selected from: [0012] ; [0013] thereof. In particular aspects, X is selected from 211 At, 131 I, 125 I, 124 I, 123 I, 77 Br, and 80m Br.
- M is a metal selected from Y, Lu, Tc, Zr, In, Sm, Re, Cu, Pb, Ac, Bi, Al, Ga, Re, Ho and Sc.
- M is a radiometal selected from 68 Ga, 64 Cu, 86 Y, 90 Y, 89 Zr, 111 In, 99m Tc, 177 Lu, 153 Sm, 186 Re, 188 Re, 67 Cu, 212 Pb, 225 Ac, 213 Bi, 212 Bi, 67 Ga, 203 Pb, 47 Sc, 2 42332.601_P15594-02 149 Tb, and 166 Ho.
- M is selected from 177 Lu, 225 Ac, 213 Bi, and 203 Pb. In more particular aspects, M is 177 Lu. [0015] In particular aspects, the compound of formula (I) is selected from: ; ; 42332.601_P15594-02
- nd , r treating or imaging one or more PSMA expressing tumors or cells, the method comprising contacting the one or more PSMA 4 42332.601_P15594-02 expressing tumors or cells with an effective amount of a compound of formula (I), and, when the method is an imaging method, taking an image.
- the one or more PSMA-expressing tumors or cells is selected from the group consisting of a prostate tumor or cell, a metastasized prostate tumor or cell, a lung tumor or cell, a renal tumor or cell, a glioblastoma, a pancreatic tumor or cell, a bladder tumor or cell, a sarcoma, a melanoma, a breast tumor or cell, a colon tumor or cell, a germ cell, a pheochromocytoma, an esophageal tumor or cell, a stomach tumor or cell, and combinations thereof.
- the one or more PSMA-expressing tumors or cells is a prostate tumor or cell.
- the one or more PSMA-expressing tumor or cell is in vitro, in vivo, or ex vivo. In certain aspects, the one or more PSMA-expressing tumor or cell is present in a subject. In particular aspects, the subject is human. [0025] In certain aspects, administering the compound of formula (I) to the subject results in inhibition of tumor growth. [0026]
- FIG.1 shows structures of clinically studied PSMA-targeted albumin-binding agents 177 Lu- PSMA-ALB-56, 177 Lu-EB-PSMA-617, 177 Lu-CTT1403, 177 Lu-PSMA-617, PSMA I&T, 177 Lu-L1, and 177 Lu-L14 (prior art). 5 42332.601_P15594-02 [0030] FIG. 2A and FIG. 2B show (FIG.
- FIG. 3E shows an in vitro clonogenic assay.
- FIG. 5 Clonogenic survival of PSMA+ PC3 PIP cells and PSMAPC3 flu cells treated with increasing concentrations of 177 Lu- Alb-L1 (FIG. 4A), 177 Lu-Alb-L2 (FIG. 4B), and 177 Lu-L5 (FIG. 4C) for 48 h at 37 °C. [0033] FIG. 5.
- T/B tumor-to-blood
- T/K tumor-to-kidney
- T/Sal tumor- to-salivary ratios were obtained from the biodistribution data of 177 Lu-L1 and 177 Lu-Alb- L2– 177 Lu-Alb-L6.
- FIG. 6A, FIG. 6B, and FIG. 6C show (FIG. 6A) areas under the curves (AUCs) of tumor, blood, and kidney uptakes for 177 Lu-Alb-L2– 177 Lu-Alb-L6 and 177 Lu-L1 were calculated.
- FIG. 6B AUC values of 177 Lu-Alb-L2– 177 Lu-Alb-L6 and 177 Lu-L1 are compared.
- FIG. 6C AUC0- 192h (tumor-to-blood) and AUC 0-192h (tumor-to-kidney) were analyzed.
- AUC 0-192h (tumor-to-blood) is approximately 867 and AUC0-192h(tumor-to-kidney) is approximately 8, respectively, for 177 Lu- L1, removed from the graph to provide clarity.
- the estimated maximum tumor-absorbed doses of 177 Lu-Alb-L2– 177 Lu-Alb-L6 were analyzed, assuming the maximum absorbed dose for blood 2 Gy and kidney 28 Gy, respectively.
- p-values lower than 0.05 (p ⁇ 0.05), p ⁇ 0.01, p ⁇ 0.001, and p ⁇ 0.0001 were referred to one (*), two (**), or four (****) asterisks, respectively. [0035] FIG.
- FIG. 7 shows HRMS (top) and 1 H NMR (bottom) spectra for 177 Lu-Alb-L1 in DMSO-d 6 at room temperature. 6 42332.601_P15594-02
- FIG. 8 shows HRMS (top) and 1 H NMR (bottom) spectra for 177 Lu-Alb-L2 in DMSO-d 6 at room temperature.
- FIG. 9 shows HRMS (top) and 1 H NMR (bottom) spectra for 177 Lu-Alb-L3 in DMSO-d6 at room temperature.
- FIG. 10 shows HRMS (top) and 1 H NMR (bottom) spectra for 177 Lu-Alb-L4 in DMSO-d6 at room temperature. [0039] FIG.
- FIG. 11 shows HRMS (top) and 1 H NMR (bottom) spectra for 177 Lu-Alb-L5 in DMSO-d 6 at room temperature.
- FIG. 12 shows HRMS (top) and 1 H NMR (bottom) spectra for 177 Lu-Alb-L6 in DMSO-d6 at room temperature.
- FIG.13A, FIG.13B, and FIG.13C show validation of PSMA expression in isogenic human prostate cancer PC3 sublines, PSMA+ PC3 PIP and PSMA- PC3 flu. PSMA+ PC3 PIP is designed to overexpress copious amounts of PSMA.
- FIG. 13A PSMA total protein levels in PSMA+ PC3 PIP and PSMA- PC3 flu cells by western blot analysis.
- FIG. 13B shows
- FIG.13C Flow cytometry data showing PSMA surface expression on PSMA+ PC3 PIP and PSMA- flu cells.
- R 1 is selected from H, substituted aryl, substituted pyridine, and unsubstituted isoquinoline; ; arylene, ; wherein n and m is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; [0049] M is present or absent and when present is a metal or a radiometal; and stereoisomers and pharmaceutically acceptable salts thereof.
- R 1 is selected from: [0051] ; [0052] thereof.
- X is selected from 211 At, 131 I, 125 I, 124 I, 123 I, 77 Br, and 80m Br.
- M is a metal selected from Y, Lu, Tc, Zr, In, Sm, Re, Cu, Pb, Ac, Bi, Al, Ga, Re, Ho and Sc.
- M is a radiometal selected from 68 Ga, 64 Cu, 86 Y, 90 Y, 89 Zr, 111 In, 99m Tc, 177 Lu, 153 Sm, 186 Re, 188 Re, 67 Cu, 212 Pb, 225 Ac, 213 Bi, 212 Bi, 67 Ga, 8 42332.601_P15594-02 203 Pb, 47 Sc, 149 Tb, and 166 Ho.
- M is selected from 177 Lu, 225 Ac, 213 Bi, and 203 Pb. In more particular embodiments, M is 177 Lu. [0054] In particular embodiments, the compound of formula (I) is selected from: ; ; ; 42332.601_P15594-02
- the presently disclosed subject matter provides a method for treating or imaging one or more PSMA expressing tumors or cells, the method comprising contacting the one or more PSMA expressing tumors or cells with an effective amount of a compound of formula (I), and, when the method is an imaging method, taking an image.
- the one or more PSMA-expressing tumor or cell is in vitro, in vivo, or ex vivo. In certain embodiments, the one or more PSMA-expressing tumor or cell is present in a subject. In particular embodiments, the subject is human. [0066] In certain embodiments, administering the compound of formula (I) to the subject results in inhibition of tumor growth. [0067] As used herein, the term “treating” can include reversing, alleviating, inhibiting the progression of, preventing, or reducing the likelihood of the disease, disorder, or condition to which such term applies, or one or more symptoms or manifestations of such disease, disorder, or condition.
- Preventing refers to causing a disease, disorder, condition, or symptom or manifestation of such, or worsening of the severity of such, not to occur. Accordingly, the presently disclosed compounds can be administered prophylactically to prevent or reduce the incidence or recurrence of the disease, disorder, or condition.
- the “subject” treated by the presently disclosed methods in their many embodiments is desirably a human subject, although it is to be understood that the methods described herein are effective with respect to all vertebrate species, which are intended to be included in the term “subject.” Accordingly, a “subject” can include a human subject for medical purposes, such as for the treatment of an existing condition or disease or the prophylactic treatment for preventing the onset of a condition or disease, or an animal subject for medical, veterinary purposes, or 11 42332.601_P15594-02 developmental purposes.
- an animal may be a transgenic animal.
- the subject is a human including, but not limited to, fetal, neonatal, infant, juvenile, and adult subjects.
- a “subject” can include a patient afflicted with or suspected of being afflicted with a condition or disease.
- the terms “subject” and “patient” are used interchangeably herein.
- the term “subject” also refers to an organism, tissue, cell, or collection of cells from a subject.
- a “therapeutically effective amount” of a therapeutic agent refers to the amount of the agent necessary to elicit the desired biological response.
- the effective amount of an agent may vary depending on such factors as the desired biological endpoint, the agent to be delivered, the composition of the pharmaceutical composition, the target tissue or cell, and the like.
- the term “therapeutically effective amount” refers to an amount sufficient to reduce or ameliorate the severity, duration, progression, or onset of a disease, disorder, or condition, or one or more symptoms thereof; prevent the advancement of a disease, disorder, or condition, cause the regression of a disease, disorder, or condition; prevent the recurrence, development, onset or progression of a symptom associated with a disease, disorder, or condition, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
- the term “combination” is used in its broadest sense and means that a subject is administered at least two agents, more particularly a compound disclosed herein and at least one other therapeutic agent. More particularly, the term “in combination” refers to the concomitant administration of two (or more) active agents for the treatment of a, e.g., single disease state.
- the active agents may be combined and administered in a single dosage form, may be administered as separate dosage forms at the same time, or may be administered as separate dosage forms that are administered alternately or sequentially on the same or separate days. In one embodiment of the presently disclosed subject matter, the active agents are combined and administered in a single dosage form.
- the timing of administration of a compound disclosed herein and at least one additional therapeutic agent can be varied so long as the beneficial effects of the combination of these agents are achieved.
- the phrase “in combination with” refers to the administration of a compound described herein and at least one additional therapeutic agent either simultaneously, sequentially, or a combination thereof. Therefore, a subject administered a combination of a compound described herein and at least one additional therapeutic agent can receive a compound and at least one additional therapeutic agent at the same time (i.e., simultaneously) or at different times (i.e., sequentially, in either order, on the same day or on different days), so long as the effect of the combination of both agents is achieved in the subject.
- agents administered sequentially can be administered within 1, 5, 10, 30, 60, 120, 180, 240 minutes or longer of one another. In other embodiments, agents administered sequentially, can be administered within 1, 5, 10, 15, 20 or more days of one another.
- the compound described herein and at least one additional therapeutic agent are administered simultaneously, they can be administered to the subject as separate pharmaceutical compositions, each comprising either a compound or at least one additional therapeutic agent, or they can be administered to a subject as a single pharmaceutical composition comprising both agents.
- the effective concentration of each of the agents to elicit a particular biological response may be less than the effective concentration of each agent when administered alone, thereby allowing a reduction in the dose of one or more of the agents relative to the dose that would be needed if the agent was administered as a single agent.
- the effects of multiple agents may, but need not be, additive or synergistic.
- the agents may be administered multiple times. 13 42332.601_P15594-02 [0075]
- the two or more agents can have a synergistic effect.
- the terms “synergy,” “synergistic,” “synergistically” and derivations thereof, such as in a “synergistic effect” or a “synergistic combination” or a “synergistic composition” refer to circumstances under which the biological activity of a combination of a compound described herein and at least one additional therapeutic agent is greater than the sum of the biological activities of the respective agents when administered individually.
- Synergy can be expressed in terms of a “Synergy Index (SI),” which generally can be determined by the method described by F. C.
- a “synergistic combination” has an activity higher that what can be expected based on the observed activities of the individual components when used alone.
- a “synergistically effective amount” of a component refers to the amount of the component necessary to elicit a synergistic effect in, for example, another therapeutic agent present in the composition.
- the “agent(s)” may be formulated into liquid or solid dosage forms and administered systemically or locally.
- the agents may be delivered, for example, in a timed- or sustained-slow release form as is known to those skilled in the art. Techniques for formulation and administration may be found in Remington: The Science and Practice of Pharmacy (20th ed.) Lippincott, Williams & Wilkins (2000).
- Suitable routes may include oral, buccal, by inhalation spray, sublingual, rectal, transdermal, vaginal, transmucosal, nasal or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, 14 42332.601_P15594-02 intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intra-articular, intra-sternal, intra-synovial, intra-hepatic, intralesional, intracranial, intraperitoneal, intranasal, or intraocular injections or other modes of delivery.
- the compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration.
- Such carriers enable the compounds of the disclosure to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a subject (e.g., patient) to be treated.
- the agents of the disclosure also may be formulated by methods known to those of skill in the art, and may include, for example, but not limited to, examples of solubilizing, diluting, or dispersing substances, such as saline; preservatives, such as benzyl alcohol; absorption promoters; and fluorocarbons.
- the compound disclosed herein is administered intranasally in a form selected from the group consisting of a nasal spray, a nasal drop, a powder, a granule, a cachet, a tablet, an aerosol, a paste, a cream, a gel, an ointment, a salve, a foam, a paste, a lotion, a cream, an oil suspension, an emulsion, a solution, a patch, and a stick.
- the term administrating via an "intranasal route” refers to administering by way of the nasal structures.
- compositions suitable for use in the present disclosure include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Generally, the compounds according to the disclosure are effective over a wide dosage range. For example, in the 15 42332.601_P15594-02 treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used. A non-limiting dosage is 10 to 30 mg per day.
- these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
- suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
- the preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
- compositions for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
- suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethyl-cellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone).
- CMC carboxymethyl-cellulose
- PVP polyvinylpyrrolidone
- disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
- Dragee cores are provided with suitable coatings.
- suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol (PEG), and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
- Dye-stuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
- compositions that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol.
- the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
- the active compounds may be dissolved or suspended in 16 42332.601_P15594-02 suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols (PEGs).
- PEGs liquid polyethylene glycols
- stabilizers may be added.
- compositions thereof include pharmaceutically acceptable salts.
- Pharmaceutically acceptable salts are generally well known to those of ordinary skill in the art, and include salts of active compounds that can be prepared with relatively nontoxic acids or bases, depending on the particular substituent moieties found on the compounds described herein.
- the parent form of the compound can differ from the various salt forms in certain physical properties, such as solubility, and the like.
- base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent or by ion exchange, whereby one basic counterion (base) in an ionic complex is substituted for another.
- bases include sodium, potassium, calcium, ammonium, organic amino, magnesium, and the like.
- acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent or by ion exchange, whereby one acidic counterion (acid) in an ionic complex is substituted for another.
- acid addition salts include those derived from inorganic acids, organic acids, and amino acids. See, for example, Berge et al, “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19). Compounds containing both basic and acidic functionalities allow such compounds to be converted into either base or acid addition salts.
- salts suitable for use with the presently disclosed subject matter include, by way of example but not limitation, acetate, arginate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, monohydrogencarbonate, citrate, edetate, edisylate, estolate, esylate, fumarate, galactonate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydriodic, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, isobutyrate, lactate, lactobionate, malate, maleate, malonate, mandelate, mesylate, methanesulfonate, mucate, napsylate, nitrate, pam
- hydrocarbon refers to any chemical group comprising hydrogen and carbon. A hydrocarbon group may be substituted or unsubstituted. As would be known to one of ordinary skill in the art, all valencies must be satisfied in making any substitutions.
- the hydrocarbon may be unsaturated, saturated, branched, unbranched, cyclic, polycyclic, or heterocyclic.
- aliphatic or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocyclyl”, “cycloaliphatic”, or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
- aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-4 alipatic carbon atoms. In some embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
- cycloaliphatic refers to a monocyclic C3-C7 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
- Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl, or (cycloalkyl)alkenyl.
- alkane refers to acyclic branched or unbranched hydrocarbons having the general formula CnH2n+2, and therefore consisting entirely of hydrogen atoms and saturated carbon atoms.
- alkyl refers to a univalent group derived from an alkane by removal of a hydrogen atom from any carbon atom and having the chemical formula of -CnH2n+1. The groups derived by removal of a hydrogen atom from a terminal carbon atom of unbranched alkanes form a subclass of normal alkyl (n-alkyl) groups H(CH 2 ) n .
- RCH 2 , R 2 CH (R ⁇ H), and R 3 C (R ⁇ H) are primary, secondary and tertiary alkyl groups, respectively.
- An alkyl can be a straightchain (i.e., unbranched) or branched acyclic hydrocarbon having the number of carbon atoms designated (i.e., C 1-10 means one to ten carbons, including 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10 carbons).
- the term “alkyl” refers to C 1-20 inclusive, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20 carbons.
- the alkyl can be a C1-C4 alkyl, including 1, 2, 3, and 4 carbons. In yet other embodiments, the alkyl can be a C 1 -C 6 alkyl, including 1, 2, 3, 4, 5, and 6 carbons. In even yet other embodiments, the alkyl can be a C1-C8 alkyl, including 1, 2, 3, 4, 5, 6, 7, and 8 carbons. [00104] “Lower alkyl” refers to an alkyl group having 1 to about 8 carbon atoms (i.e., a C1-8 alkyl), e.g., 1, 2, 3, 4, 5, 6, 7, or 8 carbon atoms.
- “Higher alkyl” refers to an alkyl group having about 10 to about 20 carbon atoms, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
- “alkyl” refers to straight-chain alkyls.
- “alkyl” refers to branched alkyls.
- “alkyl” refers to straight-chain and/or branched alkyls.
- “Branched” refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain.
- Representative alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, sec-pentyl, isopentyl, neopentyl, n- hexyl, sec-hexyl, n-heptyl, n-octyl, n-decyl, n-undecyl, and dodecyl.
- Alkyl groups can optionally be substituted (a “substituted alkyl”) with one or more substituents, which can be the same or different.
- substituent groups include, but are not limited to, alkyl, substituted alkyl, cycloalkyl, halogen, acyl, carboxyl, oxo, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, cyano, and mercapto.
- heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain having from 1 to 20 carbon atoms or 19 42332.601_P15594-02 heteroatoms consisting of at least one carbon atom and at least one heteroatom selected from the group consisting of O, N, P, Si and S, and wherein the nitrogen, phosphorus, and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
- the heteroatom(s) O, N, P and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which alkyl group is attached to the remainder of the molecule.
- cycloalkane refers to saturated monocyclic hydrocarbons (with or without side chains), e.g., cyclobutane. Unsaturated monocyclic hydrocarbons having one endocyclic double or one triple bond are called cycloalkenes and cycloalkynes, respectively. Those having more than one such multiple bond are cycloalkadienes, cycloalkatrienes, and the like.
- cyclic hydrocarbons having any number of such multiple bonds are cyclic olefins or cyclic acetylenes.
- cycloalkyl refer to a univalent group derived from a cycloalkane by removal of a hydrogen atom from a ring carbon atom. Cycloalkyls can be a mono- or multicyclic ring system of about 3 to about 10 carbon atoms, e.g., 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms. The cycloalkyl group also can be optionally substituted with a substituent group provided hereinabove for alkyl groups.
- Representative monocyclic cycloalkyl rings include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
- Multicyclic cycloalkyl rings include adamantyl, octahydronaphthyl, decalin, camphor, camphane, and noradamantyl, and fused ring systems, such as dihydro- and tetrahydronaphthalene, and the like.
- cycloalkylalkyl refers to a cycloalkyl group, which is attached to the parent molecular moiety through an alkylene moiety, also as defined above, e.g., a C1-20 alkylene moiety.
- alkylene moiety also as defined above, e.g., a C1-20 alkylene moiety.
- Examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.
- cycloheteroalkyl and “heterocycloalkyl” (or more generally “heterocyclic”) are used interchangeably and refer to an unsaturated ring system, such as a 3- to 10-member substituted or unsubstituted cycloalkyl ring system, including one or more heteroatoms, which can be the same or different, and are selected from the group consisting of nitrogen (N), oxygen (O), 20 42332.601_P15594-02 sulfur (S), phosphorus (P), and silicon (Si), in which the nitrogen, sulfur, and phosphorus heteroatoms may be oxidized and the nitrogen heteroatom may be quaternized.
- an unsaturated ring system such as a 3- to 10-member substituted or unsubstituted cycloalkyl ring system, including one or more heteroatoms, which can be the same or different, and are selected from the group consisting of nitrogen (N), oxygen (O), 20 42332.601_P15594-02 sulfur (S), phosphorus (P),
- the cycloheteroalkyl ring can be optionally fused to or otherwise attached to other cycloheteroalkyl rings and/or non-aromatic hydrocarbon rings.
- Representative cycloheteroalkyl ring systems include, but are not limited to pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidinyl, piperazinyl, indolinyl, quinuclidinyl, morpholinyl, thiomorpholinyl, thiadiazinanyl, tetrahydrofuranyl, and the like.
- cycloalkylene and “heterocycloalkylene” refer to the divalent derivatives of cycloalkyl and heterocycloalkyl, respectively.
- the terms “bicycloalkyl” and “bicycloheteroalkyl” refer to two cycloalkyl or cycloheteroalkyl groups that are bound to one another. Non-limiting examples include bicyclohexane and bipiperidine.
- An “unsaturated hydrocarbon” has one or more double bonds or triple bonds.
- alkene refers to an acyclic branched or unbranched hydrocarbons having one carbon–carbon double bond and the general formula CnH2n.
- Acyclic branched or unbranched hydrocarbons having more than one double bond are alkadienes, alkatrienes, and the like.
- alkenyl refers to a monovalent group derived from a C2-20 inclusive straight or branched hydrocarbon moiety having at least one carbon-carbon double bond by the removal of a single hydrogen molecule.
- Alkenyl groups include, but are not limited to, ethenyl (i.e., vinyl), 2-propenyl, butenyl, 1-methyl-2-buten-1-yl, pentenyl, 2- isopentenyl, hexenyl, octenyl, allenyl, butadienyl, crotyl (but-2-en-1-yl), 2-(butadienyl), 2,4- pentadienyl, 3-(l,4-pentadienyl), and the like, including higher homologs and isomers.
- cycloalkenyl refers to a cyclic hydrocarbon containing at least one carbon-carbon double bond.
- cycloalkenyl groups include cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadiene, cyclohexenyl, 1,3-cyclohexadiene, cycloheptenyl, cycloheptatrienyl, and cyclooctenyl.
- alkyne refers to an acyclic branched or unbranched hydrocarbons having a carbon-carbon triple bond and the general formula CnH2n-2, RC ⁇ CR.
- Acyclic branched or unbranched hydrocarbons having more than one triple bond are known as alkadiynes, alkatriynes, and the like.
- alkynyl refers to a monovalent group derived from a straight or branched C2-20 hydrocarbon of a designed number of carbon atoms containing at least one carbon- carbon triple bond.
- alkynyl groups include, but are not limited to, ethynyl, 2-propynyl (propargyl), 1-propynyl, pentynyl, hexynyl, and heptynyl groups, and the like.
- alkylene refers to an alkanediyl group having the free valencies on adjacent carbon atoms, e.g.
- alkylene by itself or a part of another substituent refers to a straight or branched bivalent aliphatic hydrocarbon group derived from an alkyl group having from 1 to about 20 carbon atoms, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
- the alkylene group can be straight, branched or cyclic.
- the alkylene group also can be optionally unsaturated and/or substituted with one or more “alkyl group substituents.” There can be optionally inserted along the alkylene group one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms (also referred to herein as “alkylaminoalkyl”), wherein the nitrogen substituent is alkyl as previously described.
- An alkylene group can have about 2 to about 3 carbon atoms and can further have 6-20 carbons. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being some embodiments of the present disclosure.
- a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
- heteroalkylene by itself or as part of another substituent means a divalent group derived from heteroalkyl, as exemplified, but not limited by, -CH 2 -CH 2 -S-CH 2 -CH 2 - and -CH 2 -S-CH 2 -CH 2 -NH-CH 2 -.
- heteroatoms also can occupy either or both of the chain termini (e.g., alkyleneoxo, alkylenedioxo, alkyleneamino, alkylenediamino, and the like).
- alkylene and heteroalkylene linking groups no orientation of the linking group is implied by the direction in which the formula 22 42332.601_P15594-02 of the linking group is written.
- the formula -C(O)OR’- represents both -C(O)OR’- and –R’OC(O)-.
- arene refers to a monocyclic and polycyclic aromatic hydrocarbon.
- aryl refers to a group derived from arenes by removal of a hydrogen atom from a ring carbon atom. Groups similarly derived from heteroarenes are sometimes subsumed in this definition.
- An aryl group can include, for example, a single ring or multiple rings (such as from 2 to 3 rings), which are fused together or linked covalently.
- heteroaryl refers to the class of heterocyclyl groups derived from heteroarenes by removal of a hydrogen atom from any ring atom.
- a “heteroaryl” group can include from one to four heteroatoms (in each separate ring in the case of multiple rings) selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
- a heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom.
- Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2- imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3- isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2- thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquino
- arylene and “heteroarylene” refer to the divalent forms of aryl and heteroaryl, respectively.
- aryl when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
- arylalkyl and heteroarylalkyl are meant to include those groups in which an aryl or heteroaryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl, furylmethyl, and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(l- 23 42332.601_P15594-02 naphthyloxy)propyl, and the like).
- an alkyl group e.g., benzyl, phenethyl, pyridylmethyl, furylmethyl, and the like
- an oxygen atom e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(l- 23 42332.601_P15594-02 naphthyloxy)propyl, and the
- haloaryl as used herein is meant to cover only aryls substituted with one or more halogens.
- a heteroalkyl, heterocycloalkyl, or heteroaryl includes a specific number of members (e.g., “3 to 7 membered”), the term “member” refers to a carbon or heteroatom.
- acyl In organic chemistry an unspecified acyl group is commonly a carboxylic acyl group.
- acyl specifically includes arylacyl groups, such as a 2-(furan-2-yl)acetyl)- and a 2-phenylacetyl group. Specific examples of acyl groups include acetyl and benzoyl.
- alkoxyl or “alkoxy” are used interchangeably herein and refer to a saturated (i.e., alkyl–O–) or unsaturated (i.e., alkenyl–O– and alkynyl–O–) group attached to the parent molecular moiety through an oxygen atom, wherein the terms “alkyl,” “alkenyl,” and “alkynyl” are as previously described and can include C1-20 inclusive, linear, branched, or cyclic, saturated or unsaturated oxo-hydrocarbon chains, including, for example, methoxyl, ethoxyl, propoxyl, isopropoxyl, n-butoxyl, sec-butoxyl, tert-butoxyl, and n-pentoxyl, neopentoxyl, n-hexoxyl, and the like.
- alkoxyalkyl refers to an alkyl-O-alkyl ether, for example, a methoxyethyl or an ethoxymethyl group.
- Aryloxyl refers to an aryl-O- group wherein the aryl group is as previously described, including a substituted aryl.
- aryloxyl as used herein can refer to phenyloxyl or hexyloxyl, and alkyl, substituted alkyl, halo, or alkoxyl substituted phenyloxyl or hexyloxyl.
- Aralkyl refers to an aryl-alkyl-group wherein aryl and alkyl are as previously described, and included substituted aryl and substituted alkyl. Exemplary aralkyl groups include benzyl, phenylethyl, and naphthylmethyl.
- Aralkyloxyl refers to an aralkyl-O– group wherein the aralkyl group is as previously described. An exemplary aralkyloxyl group is benzyloxyl, i.e., C6H5-CH2-O-. An aralkyloxyl group can optionally be substituted.
- exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, butyloxycarbonyl, and tert-butyloxycarbonyl.
- Exemplary aryloxycarbonyl groups include phenoxy- and naphthoxy-carbonyl.
- An exemplary aralkoxycarbonyl group is benzyloxycarbonyl.
- acyloxyl refers to an oxygen-centered radicals consisting of an acyl radical bonded to an oxygen atom, e.g., an acyl-O- group wherein acyl is as previously described.
- amine refers to a compound formally derived from ammonia by replacing one, two or three hydrogen atoms by hydrocarbyl groups, and having the general structures RNH2 (primary amines), R 2 NH (secondary amines), R 3 N (tertiary amines). In some embodiments, the term amino refers to the –NH2 group.
- the amino group is -NR'R”, wherein R' and R” are typically selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
- R' and R are typically selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
- acylamino and “alkylamino” refer to specific N-substituted organic radicals with acyl and alkyl substituent groups, respectively.
- aminoalkyl refers to an amino group covalently bound to an alkylene linker. More particularly, the terms alkylamino, dialkylamino, and trialkylamino as used herein refer to one, two, or three, respectively, alkyl groups, as previously defined, attached to the parent molecular moiety through a nitrogen atom.
- alkylamino refers to a group having the structure –NHR’ wherein R’ is an alkyl group, as previously defined; whereas the term dialkylamino refers to a group having the structure –NR’R”, wherein R’ and R” are each independently selected from the group consisting of alkyl groups.
- trialkylamino refers to a group having the structure –NR’R”R”’, wherein R’, R”, and R’” are each independently selected 25 42332.601_P15594-02 from the group consisting of alkyl groups. Additionally, R’, R”, and/or R’” taken together may optionally be –(CH2)k– where k is an integer from 2 to 6. Examples include, but are not limited to, methylamino, dimethylamino, ethylamino, diethylamino, diethylaminocarbonyl, methylethylamino, isopropylamino, piperidino, trimethylamino, and propylamino.
- alkylthioether and thioalkoxyl refer to a saturated (i.e., alkyl–S–) or unsaturated (i.e., alkenyl–S– and alkynyl–S–) group attached to the parent molecular moiety through a sulfur atom.
- thioalkoxyl moieties include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like.
- “Acylamino” refers to an acyl-NH– group wherein acyl is as previously described.
- “Aroylamino” refers to an aroyl-NH– group wherein aroyl is as previously described.
- the term “carboxyl” refers to the –COOH group. Such groups also are referred to herein as a “carboxylic acid” moiety.
- cyano refers to the -C ⁇ N group.
- halo halide
- halogen as used herein refer to fluoro, chloro, bromo, and iodo groups.
- haloalkyl are meant to include monohaloalkyl and polyhaloalkyl.
- halo(C1-4)alkyl is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
- hydroxyl refers to the —OH group.
- hydroxyalkyl refers to an alkyl group substituted with an –OH group. 26 42332.601_P15594-02
- mercapto refers to the –SH group.
- the term thus embraces aldehydes, carboxylic acids, ketones, sulfonic acids, amides and esters.
- keto is sometimes used as a prefix, but such use has been abandoned by IUPAC for naming specific compounds.
- oxo as used herein means an oxygen atom that is double bonded to a carbon atom or to another element.
- nitro refers to the –NO2 group.
- thiol refers to a compounds having the structure RSH (R ⁇ H), e.g., MeCH2SH ethanethiol.
- a thiol also is known by the term “mercaptan.”
- thiohydroxyl or thiol refers to a group of the formula –SH.
- thioether refers to the —SO4 group.
- sulfide refers to a compound having the structure RSR (R ⁇ H) and also are referred to as “thioethers.”
- ureido refers to a urea group of the formula –NH—CO—NH2.
- a structure represented generally by, for example, the formula: 42332.601_P15594-02 refers to a ring structure, for example, but not limited to a 3-carbon, a 4- carbon, a 5-carbon, a 6-carbon, a 7-carbon, and the like, aliphatic and/or aromatic cyclic compound, including a saturated ring structure, a partially saturated ring structure, and an unsaturated ring structure, comprising a substituent R group, wherein the R group can be present or absent, and when present, one or more R groups can each be substituted on one or more available carbon atoms of the ring structure.
- n is an integer generally having a value ranging from 0 to the number of carbon atoms on the ring available for substitution.
- the structure above where n is 0 to 2 would comprise compound groups including, but not limited to: ring structure indicates that the bond can be either present or absent in the ring. That is, a dashed line representing a bond in a cyclic ring structure indicates that the ring structure is selected from the group consisting of a saturated ring structure, a partially saturated ring structure, and an unsaturated ring structure.
- Certain compounds of the present disclosure may possess asymmetric carbon atoms (optical or chiral centers) or double bonds; the enantiomers, racemates, diastereomers, tautomers, 28 42332.601_P15594-02 geometric isomers, stereoisometric forms that may be defined, in terms of absolute stereochemistry, as (R)-or (S)- or, as D- or L- for amino acids, and individual isomers are encompassed within the scope of the present disclosure.
- the compounds of the present disclosure do not include those which are known in art to be too unstable to synthesize and/or isolate.
- the present disclosure is meant to include compounds in racemic, scalemic, and optically pure forms.
- Optically active (R)- and (S)-, or D- and L-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
- the compounds described herein contain olefenic bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers.
- structures depicted herein are also meant to include all stereochemical forms of the structure; i.e., the R and S configurations for each asymmetric center. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric mixtures of the present compounds are within the scope of the disclosure.
- the term “congener” refers to one of two or more substances related to each other by origin, structure, or function.
- enantiomer refers to one of a pair of molecular entities which are mirror images of each other and non-superposable.
- stereoisomer refers to an isomer that possess identical constitution, but which differ in the arrangement of their atoms in space.
- racemate refers to an equimolar mixture of a pair of enantiomers. It does not exhibit optical activity.
- the chemical name or formula of a racemate is distinguished from those of the enantiomers by the prefix ( ⁇ )- or rac- (or racem-) or by the symbols RS and SR.
- the term “diastereoisomerism” refers to stereoisomerism other than enantiomerism.
- Diastereoisomers (or diastereomers) are stereoisomers not related as mirror images. Diastereoisomers are characterized by differences in physical properties, and by some differences in chemical behavior towards achiral as well as chiral reagents.
- tautomer refers to one of two or more structural isomers which exist in equilibrium and which are readily converted from one isomeric form to another. 29 42332.601_P15594-02
- structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
- compounds having the present structures with the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C-enriched carbon are within the scope of this disclosure.
- the compounds of the present disclosure may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds.
- the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure.
- radioactive isotopes such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure.
- Albumin- binding moieties have been used strategically to enhance the tumor uptake and retention of existing PSMA-based investigational agents.
- a series of PSMA-based, ⁇ -particle-emitting, low-molecular- weight compounds was previously developed. From this series, 177 Lu-L1 was selected as the lead agent because of its reduced off-target radiotoxicity in preclinical studies.
- the ligand L1 of the compound 177 Lu-L1 contains a PSMA-targeting Lys-Glu urea moiety with an N-bromobenzyl substituent in the ⁇ -amino group of Lys.
- 177 Lu-L1 was structurally modified to improve tumor targeting using two albumin-binding moieties, 4-(p-iodophenyl)butyric acid moiety (IPBA) and ibuprofen (IBU), and the effects of linker length and composition was evaluated.
- IPBA 4-(p-iodophenyl)butyric acid moiety
- IBU ibuprofen
- Six structurally related PSMA- targeting ligands (Alb-L1, Alb-L2, Alb-L3, Alb-L4, Alb-L5, and Alb-L6) were synthesized based on the structure of 177 Lu-L1.
- the ligands were assessed for in vitro binding affinity and were 31 42332.601_P15594-02 radiolabeled with 177 Lu following standard protocols.
- 177 Lu-labeled analogs were studied in cell uptake and selected cell efficacy studies.
- In vivo pharmacokinetics were investigated by conducting tissue biodistribution studies for 177 Lu-Alb-L2, 177 Lu-Alb-L3, 177 Lu-Alb-L4, 177 Lu- Alb-L5, and 177 Lu-Alb-L6 (2 h, 24 h, 72 h, and 192 h) in male NSG mice bearing human PSMA+ PC3 PIP and PSMA ⁇ PC3 flu xenografts.
- Preliminary therapeutic ratios of the agents were estimated from the area under the curve (AUC 0-192h ) of the tumors, blood, and kidney uptake values.
- PSMA inhibition constants (Kis) of the ligands were in the ⁇ 10 nM range.
- the long-linker-based agents, 177 Lu-Alb-L4 and 177 Lu-Alb- L5 displayed significantly higher tumor uptake and retention (p ⁇ 0.001) than the short-linker- bearing 177 Lu-Alb-L2 and 177 Lu-Alb-L3 and a long polyethylene glycol (PEG) linker-bearing agent, 177 Lu-Alb-L6.
- the area under the curve (AUC0-192h) of the PSMA+ PC3 PIP tumor uptake of 177 Lu-Alb-L4 and 177 Lu-Alb-L5 were greater than 4-fold higher than 177 Lu-Alb-L2, 177 Lu-Alb- L3, and 177 Lu-Alb-L6, respectively.
- the PSMA+ PIP tumor uptake (AUC 0-192h ) of 177 Lu-Alb- L2 and 177 Lu-Alb-L3 was approximately 1.5-fold higher than 177 Lu-Alb-L6.
- 177 Lu-Alb-L6 displayed the highest ratios of AUC(tumor)-to-AUC(blood) and AUC(tumor)-to-AUC(kidney) values from the series.
- 177 Lu-Alb-L4 demonstrated a nearly similar ratio of AUC(tumor)-to-AUC(blood) as 177 Lu-Alb-L6.
- the tumor- to-blood ratio was the dose-limiting therapeutic ratio for all of the compounds.
- 177 Lu-Alb-L4 and 177 Lu-Alb-L6 showed high tumor uptake in PSMA+ tumors and tumor-to-blood ratios.
- the albumin- binding properties of the agents revealed a substantially higher affinity for the long-linker-based agents, 177 Lu-Alb-L4 and 177 Lu-Alb-L5, compared to 177 Lu-Alb-L6 and the short-linker-based 177 Lu-Alb-L2, as well as 177 Lu-Alb-L3 (Table 3).
- the cell uptake and internalized fraction of the 177 Lu-labeled agents are shown in FIG. 3.
- Clonogenic Survival Assay [00206] Clonogenic assays were performed to assess the cellular efficacy of 177 Lu-Alb-L2 and 177 Lu-Alb-L5 and were compared with 177 Lu-L1, as shown in FIG.4. The data revealed a decrease in the survival of PSMA+ PC3 PIP cells with increasing concentrations of radioactivity, with complete cell killing reached at a concentration of 370 kBq/mL for 177 Lu-L1, 185 kBq/mL for 177 Lu-Alb-L2, and 37 kBq/mL for 177 Lu-Alb-L5, respectively.
- the activity to reduce cell survival to 37% (A0) was 18.5 kBq/mL for 177 Lu-L1, approximately 20 kBq/mL for 177 Lu-Alb-L2, and approximately 10 kBq/mL for 177 Lu-Alb-L5, respectively.
- Biodistribution [00208] Tissue biodistribution studies of the series of compounds form 177 Lu-Alb-L2 to 177 Lu-Alb- L6 were performed at 2 h, 24 h, 48 h, and 192 h after injection to evaluate the tumor targeting and clearance of the agents from blood and normal tissues. The biodistribution of 177 Lu-L1 was conducted at 24 h post-injection for the agents for direct comparison.
- Biodistribution data of 177 Lu-L1 (3–192 h) were acquired from our previously reported studies. Banerjee et al., 2019. A fast tumor accumulation was observed for 177 Lu-Alb-L2, which reached 17.45 ⁇ 6.51%ID/g at 2 h post-injection and displayed higher uptake, 26.41 ⁇ 6.73%ID/g at 24 h and 22.00 ⁇ 4.71%ID/g at 48 h, respectively. At 192 h post-injection, tumor uptake was low, 3.39 ⁇ 1.03%ID/g.
- 177 Lu-Alb-L3 displayed similar pharmacokinetics as 177 Lu- Alb-L2, 34.05 ⁇ 1.98%ID/g at 2 h, 30.55 ⁇ 7.44%ID/g at 24 h, 19.61 ⁇ 4.46%ID/g at 48 h, and 5.94 ⁇ 1.38%ID/g at 192 h.
- the tumor uptake of 177 Lu-Alb-L4 was 40.89 ⁇ 4.73%ID/g at 2 h, 88.04 ⁇ 16.51%ID/g at 24 h, 87.74 ⁇ 35 42332.601_P15594-02 14.09%ID/g at 48 h, and 42.22 ⁇ 14.05%ID/g at 192 h, respectively.
- 177 Lu-Alb-L5 it was 28.78 ⁇ 7.25%ID/g at 2 h, 111.47 ⁇ 13.85%ID/g at 24 h, 127.44 ⁇ 22.85%ID/g at 48 h, and 70.96 ⁇ 2.34%ID/g at 192 h, respectively.
- 177 Lu-Alb-L6 displayed moderate uptake and fast clearance: 38.73 ⁇ 1.26%ID/g at 2 h, 13.72 ⁇ 3.55%ID/g at 24 h, 9.62 ⁇ 0.96%ID/g at 48 h, and 2.22 ⁇ 0.37%ID/g at 192 h.
- the tumor uptake values of 177 Lu-L1 at 24 h were 12.86 ⁇ 0.98–14.47 ⁇ 1.32%ID/g.
- the tumor uptake and clearance of 177 Lu-L1 were in the same range as 177 Lu-Alb-L6 (30.81 ⁇ 2.86%ID/g, 15.67 ⁇ 6.25%ID/g, 11.94 ⁇ 3.83%ID/g, 9.33 ⁇ 3.18%ID/g, and 4.21 ⁇ 1.45%ID/g at 192 h).
- the uptake in the PSMA ⁇ PC3 flu tumors was low for the agents.
- the highest uptake in the PSMA ⁇ PC3 flu tumor was associated with 177 Lu-Alb-L5, 3.87 ⁇ 0.70%ID/g at 2 h, 4.55 ⁇ 0.76%ID/g at 24 h, 3.95 ⁇ 0.50%ID/g at 48 h, and 1.87 ⁇ 0.23%ID/g at 192 h post-injection, respectively.
- the blood uptake levels were high at 2 h for 177 Lu-Alb-L2, with 19.59 ⁇ 6.06%ID/g; 177 Lu- Alb-L3, with 10.29 ⁇ 0.41%ID/g; 177 Lu-Alb-L4, with 15.48 ⁇ 7.12%ID/g; 177 Lu-Alb-L5, with 22.36 ⁇ 4.72%ID/g; and 177 Lu-Alb-L6, with 2.97 ⁇ 0.45%ID/g, respectively.
- 177 Lu-Alb-L6 displayed the fastest clearance from the circulation owing to the long PEG linker, and the uptake was ⁇ 0.03 ⁇ 0.01%ID/g at 24 h.
- 177 Lu-Alb-L5 displayed the slowest clearance from the blood at 192 h post-injection, 2.77 ⁇ 0.19%ID/g, followed by 177 Lu-Alb-L4 (2.19 ⁇ 0.46%ID/g), 177 Lu-Alb-L3 (0.24 ⁇ 0.12%ID/g), and 177 Lu-Alb-L2 ( ⁇ 0.01%ID/g).
- Kidney uptake was relatively high for 177 Lu-Alb-L5 (33.45 ⁇ 6.41%ID/g at 2 h, 70.23 ⁇ 16.64 at 24%ID/g, 48.17 ⁇ 15.62%ID/g at 48 h, and 5.06 ⁇ 0.99%ID/g at 192 h) and 177 Lu-PSMA- Alb-L4 (64.44 ⁇ 8.75%ID/g at 2 h, 48.81 ⁇ 19.86%ID/g at 24 h, 23.46 ⁇ 7.75%ID/g at 48 h, and 1.34 ⁇ 0.82 at 192 h).
- 177 Lu-Alb-L3 displayed the highest kidney uptake at 2 h, 73.28 ⁇ 22.85%ID/g, and the fastest clearance, 2.77 ⁇ 1.56%ID/g at 24 h.
- 177 Lu-Alb-L2 and 177 Lu-Alb-L6 displayed low kidney uptake of ⁇ 17.86%ID/g at 2 h.
- 177 Lu-Alb-L6 displayed fast renal clearance resulting in ⁇ 1%ID/g after 24 h; in contrast, 177 Lu-Alb-L2 showed a slow and steady wash-out from the kidneys from ⁇ 7.44 ⁇ 2.64% IA/g at 24 h post-injection to 3.74 ⁇ 1.05%ID/g at 48 h post- injection.
- kidney uptake and clearance values of 177 Lu-L1 were in the range of 177 Lu-Alb-L6 36 42332.601_P15594-02 (5.16 ⁇ 2.38%ID/g at 3 h, 0.28 ⁇ 0.18%ID/g at 24 h, 0.22 ⁇ 0.10%ID/g at 72 h, and 0.01 ⁇ 0.00%ID/g at 192 h).
- the activity levels in the salivary and lacrimal glands of the agents were comparable to the blood levels and were relatively high for 177 Lu-Alb-L5 (salivary glands, 5.83 ⁇ 1.06%ID/g at 2 h, 4.35 ⁇ 0.66%ID/g at 24 h, 3.20 ⁇ 0.24 h %ID/g at 48 h, and 1.18 ⁇ 0.09%ID/g at 192 h) and 177 Lu- Alb-L4 (salivary glands, 5.40 ⁇ 0.33%ID/g at 2 h, 0.86 ⁇ 0.21%ID/g at 24 h, 0.62 ⁇ 0.07%D/g at 48 h, and 0.18 ⁇ 0.06%ID/g at 192 h).
- the tumor-to-kidney ratio was less than one for the series 177 Lu-Alb-L2 to 177 Lu-Alb-L5 at 2 h and remained at ⁇ 10 during 24–48 h and increased significantly, approximately 20–50 at 192 h after injection.
- the lowest tumor-to-kidney ratio was associated with 177 Lu-Alb-L5.
- the tumor-to-kidney values were greater than 4 for 177 Lu-L1 at 24 h and remained greater than 50 at all-time points.
- a similar time course was observed for the tumor-to-liver, tumor-to- salivary, and tumor-to-lacrimal glands, as well as the tumor-to-bone ratios.
- FIG.6A The area under the curve (AUC 0-192h ) values and AUC ratios of tumor-to-blood and tumor- to-kidney over 192 h of the new agents and 177 Lu-L1 are listed in FIG.6A, FIG.6B.
- Tumor AUC0- 192h of 177 Lu-Alb-L5 was significantly higher than 177 Lu-Alb-L4 (p ⁇ 0.002).
- tumor AUC of 177 Lu-Alb-L5 and Alb-L4 were approximately 4-fold higher compared to 177 Lu-Alb-L2 and 177 Lu-Alb-L3 and approximately 6-fold higher than 177 Lu-Alb-L6 and 177 Lu-L1.
- the tumor- 37 42332.601_P15594-02 to-blood AUC 0-192h ratio of 177 Lu-Alb-L4 was approximately 2-fold higher than 177 Lu-Alb-L5, while the tumor-to-kidney AUC0-192h ratios were in the same range.
- 177 Lu-Alb-L6 and 177 Lu-L1 displayed nearly similar tumor AUC0-192h and kidney AUC0-192h, and consequently both agents had a similar tumor-to-kidney AUC 0-192h (approximately 8).
- the tumor-to-blood AUC 0-192h ratios of 177 Lu-L1 were significantly higher (approximately 867) than the tumor-to-blood AUC0- 192h ratios of 177 Lu-Alb-L6 (approximately 48).
- the tumor-to-kidney AUC0-192h ratios of 177 Lu- Alb-L6 were approximately 2-fold higher than the other albumin-binding agents from the series.
- the tumor-to-blood AUC0-192h of 177 Lu-Alb-L6 was comparable to 177 Lu-Alb-L4; it was approximately 1.14-fold higher.
- 177 Lu-Alb-L6 displayed low binding affinity, likely due to the long PEG linker, as we noted earlier with a similar construct. Chen et al., 2022. Proof-of-concept cell efficacy data further confirmed the effect of enhanced cellular uptake and internalization of 177 Lu-Alb-L5 (Structure B) compared to 177 Lu-L1 or 177 Lu-Alb-L2 (Structure A). Enhanced cellular internalization is critical to radiation-induced DNA damage and PSMA- expressing cancer cell death. [00220] The tumor uptake of 177 Lu-Alb-L2 and 177 Lu-Alb-L3 was significantly lower than our previously reported long-linker-based albumin-binding agent, 177 Lu-L14 (FIG.
- 177 Lu-Alb- L4 tumor AUC0-192h of 12,857 ⁇ 1469%ID/g.h
- 177 Lu-Alb-L5 tumor AUC0-192h of 18,842 ⁇ 1693%ID/g.h
- the blood uptake values of 177 Lu-Alb-L4 were in the range of 177 Lu-L14 (blood AUC0-192h of 314 ⁇ 37), indicating a 2-fold improvement in the tumor- to-blood ratios of 177 Lu-Alb-L4 relative to 177 Lu-L14.
- the kidney uptake of 177 Lu-Alb-L4 (AUC0-192h of 3895 ⁇ 631) was 1.5-fold higher than 177 Lu-L14 (AUC0-192h of 2550 ⁇ 347).
- the tumor and PSMA-expressing typical healthy tissue uptake values were significantly lower for 177 Lu-Alb-L6, most likely owing to low PSMA-binding and albumin-binding properties.
- the tumor and kidney AUCs of 177 Lu-Alb-L6 were in the range of 177 Lu-L1; however, the blood AUC was approximately 30-fold higher for 177 Lu-Alb-L6 than 177 Lu-L1.
- 177 Lu-Alb-L6 For all of the compounds, the dose-limiting organ was found to be the blood. 177 Lu-Alb-L6, and to a lesser extent, 177 Lu-Alb-L4, appear to be the more viable compounds with potential tumoricidal absorbed doses able to be delivered to the tumor.
- Renal radiotoxicity has not proved to be a significant issue for 177 Lu-PSMA-targeted therapy, possibly due to optimal low linear energy transfer radiations of 177 Lu ( ⁇ max 0.5 MeV, 1.7 mm).
- a minimal expression of PSMA has been found in human kidneys. It has been reported that murine PSMA, with 91% similarity to the human PSMA sequence, is overexpressed in the proximal microtubules of the murine renal cortex.
- the tumor-to-red-marrow values of 177 Lu-EB- PSMA-617 and 177 Lu-PSMA-ALB-56 were >10-fold and >5-fold higher than 177 Lu-PSMA-617, respectively.
- Kramer et al. 2021.
- the tumor dose at maximum injectable activity was 177 Lu-EB-PSMA-617 (60.1 Gy), and 177 Lu-PSMA-ALB-56 (96 Gy) was significantly lower than 177 Lu-PSMA-671 (131 Gy).
- the estimated values are tumor AUC 0-192h is 4484 ⁇ 790%ID/g.h, blood AUC0-192h is 2015 ⁇ 251%ID/g.h, and kidney AUC0-192h is 880 ⁇ 38%ID/g.h.
- the blood AUC of 177 Lu-Alb-L4 is significantly lower (greater than 6-fold) than 111 In-DOTA-5D3, albeit with increased kidney uptake (greater than 4- fold).
- albumin-binding agent might be a superior option for PSMA-based radiopharmaceutical therapy compared to antibody-based agents.
- these studies mainly used athymic nude mice for tumor implantation.
- our studies were performed using 40 42332.601_P15594-02 NSG mice because of our institutional availability of this strain.
- the other notable variables could be related to the high specific activity of the agents used in our studies obtained through HPLC purification and the relatively large tumor used for the biodistribution studies (Table 9).
- PSMA+ PC3 PIP and PSMA ⁇ PC3 flu cells have the advantage of being isogenic and androgen- independent cell lines and are anticipated to display similar biological factors for evaluating tumor pharmacokinetics, except for the PSMA expression levels. Many reported studies currently use the same tumor models for developing similar structure–activity relationship data because of the fast and predictable growth rate. There might be some variability associated with tumor sizes, although the data related to the pharmacokinetic performance are expected to be similar. [00228] Materials and Methods [00229] General Methods. All reagents and solvents were purchased from Sigma-Aldrich or Fisher Scientific unless specified and are listed in Table 1. These reagents and solvents were directly used without further purification.
- Radiochemistry was performed under standard labeling conditions in ammonium acetate buffer (0.2 M) at pH approximately 4.5 following our reported method. Banerjee et al., 2019. The radiolabeling was performed in a radiochemistry microwave chamber at 90 °C for 5 min at 40 watts (Resonance Instruments Inc., Skokie, IL, USA), and the reaction solution was purified using reverse-phase HPLC. An isocratic HPLC method was developed in each case, as listed in Table 2, to remove the unreacted ligand from the radiolabeled material to ensure high specific activity. L- Ascorbic acid was added to the isolated radiolabeled compounds in the final formulation to maintain stability and was used for in vitro and in vivo experiments.
- test tube was vortexed for 2 min and then centrifuged (4000 ⁇ g, 5 min). Aliquots (0.1 mL) from the 1-octanol and PBS phases were transferred into two test tubes for counting. The amount of radioactivity in each test tube was 43 42332.601_P15594-02 measured using the automated ⁇ -counter.
- the amount of released glutamate from NAAG was measured by incubating it with a working solution of the Amplex Red glutamic acid kit (Molecular Probes Inc., Eugene, OR, USA) at 37 °C for 60 min. Fluorescence was measured with excitation at 535 nm and emission at 590 nm using a microplate reader. Inhibition curves were determined using semi-log plots. Data were analyzed using a one-site total binding regression using GraphPad Prism version 9 for Windows (GraphPad Software, San Diego, CA, USA). The IC 50 values were determined as the concentration at which enzymatic activity was inhibited by 50%. Assays were performed in triplicate, with the entire inhibition study repeated at least once.
- Enzyme inhibitory 44 42332.601_P15594-02 constants (K i values) were generated using the Cheng-Prusoff conversion. Cheng and Prusoff, 1973.
- Protein-Binding Assay [00239] Albumin-binding properties of the compounds were analyzed following a reported method. Tsuchihashi et al., 2023. A solution of 177 Lu-Alb-L2– 177 LuAlb-L5 (74 kBq in 10 ⁇ L PBS) was added to 190 ⁇ L human serum albumin solution (45 mg HSA in 1 mL of PBS). After mixing, the solution was incubated at 37 °C for 60 min.
- PC3 PIP cells were initially generated via the transfection of PC3 cells, employing VSV-G pseudo-typed lentiviral- vector-expressing human PSMA. Leek et al., 1995; Chang et al., 1999; Liu et al., 2009. [00242] As reported previously, flow cytometry and Western blot assays are routinely used to evaluate PSMA expressions of PSMA+ PC3 PIP cells and PSMA ⁇ PC3 flu cells. Banerjee et al., 2019. Selected data generated for the studies of this report are provided in FIG. 13.
- PSMA+ PC3 PIP cells were pre-incubated with ZJ43 (10 ⁇ M final concentration) for 30 min and then washed 3 times with binding buffer followed by incubation of the radioactive dose (100 ⁇ L of 370 kBq/mL in binding buffer) for 2 h. The cell uptake studies were then conducted using the method mentioned in the previous section.
- the collected pooled washes (containing cell-surface-bound 177 Lu-Alb-L1– 177 Lu-Alb-L6) and cell pellets (containing internalized 177 Lu-Alb-L1– 177 Lu-Alb-L6) were counted in an automated ⁇ - spectrometer along with the standards. All radioactivity values were converted into a percentage of incubated dose (%ID) per million cells. Experiments were performed in triplicate and repeated 2 times. Data were fitted according to linear regression analysis. [00246] Clonogenic Survival Assay Cells (200–1000) were seeded in 60-mm culture dishes.
- Each radioligand ( 177 Lu-L1, 177 Lu-Alb- L2, 177 Lu-Alb-L5) was diluted in a prewarmed medium at different concentrations (0, 0.37, 1.85, 3.7, 18.5, 37, 185, and 370 kBq/mL) and incubated with the cells for 48 h, as we previously reported. Banerjee et al., 2019. The radiolabeled compound was replaced with fresh medium, and cells were incubated for 2 weeks or until colonies had at least 50 cells. The colonies were stained with crystal violet and counted, and the surviving fraction was normalized to the control plating efficiency, as previously described. Franken et al., 2006.
- mice bearing PSMA+ PC3 PIP and PSMA ⁇ PC3 flu xenografts were injected intravenously with the respective 177 Lu- labeled agent, 177 Lu-Alb-L2– 177 Lu-Alb-L6 (1.85 MBq) diluted in 150 ⁇ L saline.
- Mice were sacrificed at 2 h, 24 h, 48 h, and 192 h post-injection, and selected tissues were harvested, weighed, and measured radioactivity using an automated ⁇ -counter. A group of 4 mice was used for each time point; the results were listed as the percentage of the injected dose per gram of tissue mass (%ID/g). The data are presented as the average ⁇ standard deviation (SD).
- the biodistribution study of 177 Lu-L1 was performed at only 24 h during the biodistribution study of 177 Lu-Alb-L2 and 177 Lu-Alb-L5 in a single experiment.
- the biodistributions of 177 Lu-Alb-L3, 177 Lu-Alb-L4, and 177 Lu-L1 (24 h) were acquired in a separate experiment.
- the data are listed in Tables 4-10.
- Biodistribution data of 177 Lu-L1 at 3 h, 24 h, 48 h, and 72 h were obtained from our previous report. Banerjee et al., 2019.
- the 192 h post-injection data of 177 Lu-L1 are unpublished and were acquired during the same study. Table 4.
- Tissue biodistribution data of 177 Lu-Alb-L4 in male NSG mice bearing PSMA+ PC3 PIP and PSMA- PC3 flu xenografts in either flank (Data presented in 5 Table 7.
- Tissue biodistribution data of 177 Lu-Alb-L6 in male NSG mice bearing PSMA+ PC3 PIP xenografts (Data presented in %ID/g, expressed as 1 Table 9.
- PSMA+ PC PIP tumor weight (g) and expressed as mean ⁇ SD) and tumor volume 42332.601_P15594-02 Tumor NA NA NA NA size 52 42332.601_P15594-02 Table 10.
- Tissue biodistribution data of 177Lu-Alb-L1 in male NSG mice bearing PSMA+ PC3 PIP and PSMA- PC3 flu xenografts in either flank (Data presented in % ID/g, expressed as mean - e [00250]
- Statistical Analysis [00251] All graphs, AUC calculations, and statistical analyses were created and performed using the GraphPad Prism software (version 9.0). Significant differences were evaluated using a one- way ANOVA or unpaired t-test; p-values ⁇ 0.05 were considered to be significant.
- KEU was treated with 2,5-dioxopyrrolidin-1-yl 5-((tert- butoxycarbonyl)amino)pentanoate in the presence of DIPEA, followed by deprotection using trifluoroacetic acid (TFA) and methylene dichloride (DCM), which provides the corresponding intermediate compound 3 or 4.
- TFA trifluoroacetic acid
- DCM methylene dichloride
- Fmoc-L-Lys(Boc)-OSu was added to the intermediate 5 or 6 to create compound 7 or 8.
- Boc group of lysine was removed from compounds 7 or 8.
- the crude product was conjugated with ibuprofen (IBU) or 4-(p-iodophenyl) butyric acid (IPBA) to create Alb-L1, Alb-L1, and Alb- L3. 54 42332.601_P15594-02
- IBU ibuprofen
- IPBA 4-(p-iodophenyl) butyric acid
- reaction mixture was stirred overnight and concentrated to isolate the crude product.
- To the above crude was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature, and the mixture was stirred for 2 h.
- the reaction mixture was then concentrated and purified using acetonitrile (ACN)/water (H2O) on the C18 Sep-Pak column to provide compound 3 (146 mg, 85%) as a white solid.
- reaction mixture was stirred for 24 h and concentrated to get crude product.
- To the above crude was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature, and the reaction mixture was stirred for 2 h.
- the reaction mixture was then concentrated and purified using ACN/H 2 O on the C18 Sep-Pak column to provide compound 4 (143 mg, 75%) as a white solid.
- reaction mixture was stirred for 2 h and concentrated to get the crude product.
- To the isolated crude was added 2 mL of 20% piperidine in DMF (3 mL) at room temperature, and the mixture was stirred for 1 h. Concentrated and purified using ACN/H 2 O on the C18 Sep-Pak column to provide compound 6 (125 mg, 65%) as a white solid.
- reaction mixture was stirred for 2 h and purified using acetonitrile/H 2 O on the C18 Sep-Pak column to provide the desired product as a white solid (41 mg, 85% yield).
- crude product (20 mg, 0.016 mmol) was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature; the reaction mixture was stirred for 2 h and concentrated to afford a crude compound 8.
- reaction mixture was stirred for 2 h, and the resultant reaction mixture was purified by preparative RP-HPLC chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization afforded 177 Lu-Alb-L2 (19.3 mg, 90%) as a white solid.
- reaction mixture was stirred for 2 h and purified using ACN/H2O on the C18 Sep-Pak column to provide the desired product as a white solid (41 mg, 80% yield).
- To the above product (29 mg, 0.016 mmol) was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature; the reaction mixture was stirred for 2 h and concentrated to afford a crude amine 8.
- reaction mixture was stirred overnight and concentrated to obtain the crude product.
- To the above crude was added 2 mL of TFA/CH 2 Cl 2 (1:1) at room temperature, and the mixture was stirred for 2 h. Concentrated and purified using ACN/H 2 O on C18 Sep-Pak column to provide compound 10 (36 mg, 70%) as a white solid.
- Membranes were incubated in blocking buffer (2.5% BSA, 20% tween 20 in PBS) for 2 h, then washed with PBST and further incubated for 6 h with PSMA (cat#D718E, Cell Signaling Technology) and GAPDH (cat#D16H1, Cell Signaling Technology) antibodies at room temperature. Finally, membranes were incubated with HRP-coupled anti-rabbit IgG secondary antibodies, and blots were developed by ECL reagent. Digital quantification of chemiluminescence was performed using Image J software (NIH). [00274] Real-Time PCR (RT-qPCR) [00275] Cells were cultured to 80% confluence for mRNA isolation in 6 well-cell culture plates.
- RT-qPCR Real-Time PCR
- cDNA was synthesized from 1 ⁇ g of total RNA of each experimental replicate using a cDNA synthesis kit (Applied Biosystems, USA) as per the manufacturer's protocol.
- the mRNAs were amplified on Applied Biosystems 7500 Fast Detection system with SYBR green qPCR master mix per manufacturer's instructions (Applied Biosystems, USA). All reactions were performed in triplicate, and negative controls were included in each experiment.
- GAPDH was taken as internal control, and all the data sets were normalized to the level of GAPDH. Fold change in gene expression was calculated by the ⁇ 2CT method, and results were reported as arbitrary units or fold changes.
- PSMA+ PC3 PIP and PSMA- PC3 flu cells (1 ⁇ 10 6 cells) were harvested using cell dissociation buffer (Gibco) and converted into a single-cell suspension. The harvested cells were washed twice with flow cytometry buffer ( ⁇ 1 phosphate-buffered saline with 2 mM ethylenediaminetetraacetic acid and 0.5% fetal bovine serum) and passed by pipetting through a 70 ⁇ m strainer. Next, the cells were stained with PSMA-PE antibody (cat#342504BioLegend,) according to the manufactured protocol. Cells were incubated at 4°C for 1 h in the dark.
- flow cytometry buffer ⁇ 1 phosphate-buffered saline with 2 mM ethylenediaminetetraacetic acid and 0.5% fetal bovine serum
- Glu-Ureido–Based Inhibitors of Prostate-Specific Membrane Antigen Lessons Learned during the Development of a Novel Class of Low-Molecular-Weight Theranostic Radiotracers. J. Nucl. Med.2017, 58, 17S–26S. [00289] Kelly, J.; Amor-Coarasa, A.; Ponnala, S.; Nikolopoulou, A.; Williams, C.; Schlyer, D.; Zhao, Y.; Kim, D.; Babich, J.W. Trifunctional PSMA-targeting constructs for prostate cancer with unprecedented localization to LNCaP tumors. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1841–1851.
- PSMA prostate-specific membrane antigen
- PSMA Anti-Prostate-specific Membrane Antigen
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Physics & Mathematics (AREA)
- Optics & Photonics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Urology & Nephrology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
A series of albumin-binding radiolabeled PSMA-based low-molecular-weight radiotherapeutics comprising a DOTA chelator used as a complexing agent for a radiometal, such as 177Lu, albumin-binding moieties, such as 4-(p-iodophenyl)butyric acid moiety (I PBA) and ibuprofen (IBU), fatty acids, an amino acid urea moiety, and a linker, and their use for treating one or more PSMA expressing tumors or cells, including prostate cancer, are disclosed.
Description
PSMA-BASED ALBUMIN BINDING AGENTS FOR TARGETED RADIONUCLIDE THERAPY OF PROSTATE CANCER CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims benefit of U.S. Provisional Application No. 63/520,883 filed August 21, 2023, which is incorporated herein by reference in its entirety. BACKGROUND [0002] Prostate cancer is the most commonly diagnosed non-cutaneous cancer among men in the United States and globally. Siegel et al., 2023. Death from prostate cancer occurs mainly in patients with aggressive, androgen-insensitive, metastatic disease. Rebello et al., 2021; Sandhu et al., 2021. Prostate-specific membrane antigen (PSMA) is a tumor-associated antigen overexpressed in prostate adenocarcinoma cells, regardless of androgen status, in the neovasculature of solid tumors and has a low expression in benign and extra-prostatic tissues. Silver et al., 1997; Wright et al., 1995. Studies have shown that radiopharmaceutical therapy targeting PSMA with β-particle- emitting 177Lu (half-life 6.7 d) is a life-prolonging treatment option for patients with metastatic castration-resistant prostate cancer. Sartor et al., 2021. Recently, a low-molecular-weight agent, 177Lu-PSMA-617, demonstrated a low toxicity profile compared to the standard of care and received regulatory approval. Sartor et al., 2021; Hofman et al., 2021. Although highly promising, the median overall survival among randomized patients was 15.3 months in the treatment vs. 11.3 months in the control group, suggesting that further improvements in radiopharmaceutical therapies targeting PSMA are needed. Sartor et al., 2021; Sandhu et al., 2021. SUMMARY [0003] In some aspects, the presently disclosed subject matter provides a compound of formula (I): 1 42332.601_P15594-02
I); [0005] R1 is selected from H, substituted aryl, substituted pyridine, and unsubstituted isoquinoline; ;
arylene,
and m is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; [0010] M is present or absent and when present is a metal or a radiometal; and stereoisomers and pharmaceutically acceptable salts thereof. [0011] In certain aspects, R1 is selected from: [0012] ; [0013]
thereof. In particular aspects, X is selected from 211At, 131I, 125I, 124I, 123I, 77Br, and 80mBr. [0014] In certain aspects, M is a metal selected from Y, Lu, Tc, Zr, In, Sm, Re, Cu, Pb, Ac, Bi, Al, Ga, Re, Ho and Sc. In more certain aspects, M is a radiometal selected from 68Ga, 64Cu, 86Y, 90Y, 89Zr, 111In, 99mTc, 177Lu, 153Sm, 186Re, 188Re, 67Cu, 212Pb, 225Ac, 213Bi, 212Bi, 67Ga, 203Pb, 47Sc, 2 42332.601_P15594-02
149Tb, and 166Ho. In particular aspects, M is selected from 177Lu, 225Ac, 213Bi, and 203Pb. In more particular aspects, M is 177Lu. [0015] In particular aspects, the compound of formula (I) is selected from: ; ;
42332.601_P15594-02
nd , r treating or imaging one or more PSMA expressing tumors or cells, the method comprising contacting the one or more PSMA 4 42332.601_P15594-02
expressing tumors or cells with an effective amount of a compound of formula (I), and, when the method is an imaging method, taking an image. [0023] In certain aspects, the one or more PSMA-expressing tumors or cells is selected from the group consisting of a prostate tumor or cell, a metastasized prostate tumor or cell, a lung tumor or cell, a renal tumor or cell, a glioblastoma, a pancreatic tumor or cell, a bladder tumor or cell, a sarcoma, a melanoma, a breast tumor or cell, a colon tumor or cell, a germ cell, a pheochromocytoma, an esophageal tumor or cell, a stomach tumor or cell, and combinations thereof. In particular aspects, the one or more PSMA-expressing tumors or cells is a prostate tumor or cell. [0024] In certain aspects, the one or more PSMA-expressing tumor or cell is in vitro, in vivo, or ex vivo. In certain aspects, the one or more PSMA-expressing tumor or cell is present in a subject. In particular aspects, the subject is human. [0025] In certain aspects, administering the compound of formula (I) to the subject results in inhibition of tumor growth. [0026] Certain aspects of the presently disclosed subject matter having been stated hereinabove, which are addressed in whole or in part by the presently disclosed subject matter, other aspects will become evident as the description proceeds when taken in connection with the accompanying Examples and Drawings as best described herein below. BRIEF DESCRIPTION OF THE FIGURES [0027] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee. [0028] Having thus described the presently disclosed subject matter in general terms, reference will now be made to the accompanying Figures, which are not necessarily drawn to scale, and wherein: [0029] FIG.1 shows structures of clinically studied PSMA-targeted albumin-binding agents 177Lu- PSMA-ALB-56, 177Lu-EB-PSMA-617, 177Lu-CTT1403, 177Lu-PSMA-617, PSMA I&T, 177Lu-L1, and 177Lu-L14 (prior art). 5 42332.601_P15594-02
[0030] FIG. 2A and FIG. 2B show (FIG. 2A) structures of representative compounds disclosed herein derived from Structure A (Alb-L1, Alb-L2, and Alb-L3) and Structure B (Alb-L4, Alb-L5, and Alb-L6); and (FIG. 2B) binding affinities to PSMA. [0031] FIG. 3A, FIG. 3B, FIG. 3C, FIG. 3D, FIG. 3E, and FIG. 3F show cell uptake and internalization (mean ± SD, n = 3) of 177Lu-Alb-L1 (FIG.3A), 177Lu-Alb-L2 (FIG.3B), 177Lu-Alb- L3 (FIG. 3C), 177Lu-Alb-L4 (FIG. 3D), 177Lu-Alb-L5 (FIG. 3E), and 177Lu-Alb-L6 (FIG. 3F) in PSMA+ PC3 PIP cells and PSMA− PC3 flu cells (approximately 1 million) at 37 °C. PSMA expression blockade studies in PSMA+ PC3 PIP cells of the series of compounds 177Lu-Alb-L1 to 177Lu-Alb-L6 were performed at 2 h post-incubation. The cells (approximately 1 million) were pre-incubated with a known PSMA inhibitor, ZJ43 (10 µM final concentration), for 30 min before the corresponding radioactive dose was added to the medium. [0032] FIG. 4A, FIG. 4B, and FIG. 4C show an in vitro clonogenic assay. Clonogenic survival of PSMA+ PC3 PIP cells and PSMAPC3 flu cells treated with increasing concentrations of 177Lu- Alb-L1 (FIG. 4A), 177Lu-Alb-L2 (FIG. 4B), and 177Lu-L5 (FIG. 4C) for 48 h at 37 °C. [0033] FIG. 5. (A) Tissue biodistribution data of 177Lu-Alb-L2–177Lu-Alb-L6 and 177Lu-L1 are shown as the percentage of injected dose per gram of tissue, mean ± SD, n = 3–4 mice, dose: 1.87 MBq (intravenous injection via tail-vein), tumor model: PSMA+ PC3 PIP, and PSMA− PC3 flu tumor-bearing male NSG mice. (B) The tumor-to-blood (T/B), tumor-to-kidney (T/K), and tumor- to-salivary (T/Sal) ratios were obtained from the biodistribution data of 177Lu-L1 and 177Lu-Alb- L2–177Lu-Alb-L6. Biodistribution data of 177Lu-L1 were obtained from Banerjee et al., 2019. [0034] FIG. 6A, FIG. 6B, and FIG. 6C show (FIG. 6A) areas under the curves (AUCs) of tumor, blood, and kidney uptakes for 177Lu-Alb-L2–177Lu-Alb-L6 and 177Lu-L1 were calculated. (FIG. 6B) AUC values of 177Lu-Alb-L2–177Lu-Alb-L6 and 177Lu-L1 are compared. (FIG. 6C) AUC0- 192h(tumor-to-blood) and AUC0-192h(tumor-to-kidney) were analyzed. AUC0-192h(tumor-to-blood) is approximately 867 and AUC0-192h(tumor-to-kidney) is approximately 8, respectively, for 177Lu- L1, removed from the graph to provide clarity. The estimated maximum tumor-absorbed doses of 177Lu-Alb-L2–177Lu-Alb-L6 were analyzed, assuming the maximum absorbed dose for blood 2 Gy and kidney 28 Gy, respectively. p-values lower than 0.05 (p < 0.05), p < 0.01, p < 0.001, and p < 0.0001 were referred to one (*), two (**), or four (****) asterisks, respectively. [0035] FIG. 7 shows HRMS (top) and 1H NMR (bottom) spectra for 177Lu-Alb-L1 in DMSO-d6 at room temperature. 6 42332.601_P15594-02
[0036] FIG. 8 shows HRMS (top) and 1H NMR (bottom) spectra for 177Lu-Alb-L2 in DMSO-d6 at room temperature. [0037] FIG. 9 shows HRMS (top) and 1H NMR (bottom) spectra for 177Lu-Alb-L3 in DMSO-d6 at room temperature. [0038] FIG. 10 shows HRMS (top) and 1H NMR (bottom) spectra for 177Lu-Alb-L4 in DMSO-d6 at room temperature. [0039] FIG. 11 shows HRMS (top) and 1H NMR (bottom) spectra for 177Lu-Alb-L5 in DMSO-d6 at room temperature. [0040] FIG. 12 shows HRMS (top) and 1H NMR (bottom) spectra for 177Lu-Alb-L6 in DMSO-d6 at room temperature. [0041] FIG.13A, FIG.13B, and FIG.13C show validation of PSMA expression in isogenic human prostate cancer PC3 sublines, PSMA+ PC3 PIP and PSMA- PC3 flu. PSMA+ PC3 PIP is designed to overexpress copious amounts of PSMA. FIG. 13A. PSMA total protein levels in PSMA+ PC3 PIP and PSMA- PC3 flu cells by western blot analysis. FIG. 13B. Real time PCR PSMA gene expression in PSMA+ PC3 PIP and PSMA- PC3 flu cells. FIG.13C. Flow cytometry data showing PSMA surface expression on PSMA+ PC3 PIP and PSMA- flu cells. DETAILED DESCRIPTION [0042] The presently disclosed subject matter now will be described more fully hereinafter with reference to the accompanying Figures, in which some, but not all embodiments of the inventions are shown. Like numbers refer to like elements throughout. The presently disclosed subject matter may be embodied in many different forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will satisfy applicable legal requirements. Therefore, it is to be understood that the presently disclosed subject matter is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. [0043] In some embodiments, the presently disclosed subject matter provides a compound of formula (I): 7 42332.601_P15594-02
I); [0045] R1 is selected from H, substituted aryl, substituted pyridine, and unsubstituted isoquinoline; ;
arylene, ; wherein n and
m is an integer selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12; [0049] M is present or absent and when present is a metal or a radiometal; and stereoisomers and pharmaceutically acceptable salts thereof. [0050] In certain embodiments, R1 is selected from: [0051] ; [0052]
thereof. In particular embodiments, X is selected from 211At, 131I, 125I, 124I, 123I, 77Br, and 80mBr. [0053] In certain embodiments, M is a metal selected from Y, Lu, Tc, Zr, In, Sm, Re, Cu, Pb, Ac, Bi, Al, Ga, Re, Ho and Sc. In more certain embodiments, M is a radiometal selected from 68Ga, 64Cu, 86Y, 90Y, 89Zr, 111In, 99mTc, 177Lu, 153Sm, 186Re, 188Re, 67Cu, 212Pb, 225Ac, 213Bi, 212Bi, 67Ga, 8 42332.601_P15594-02
203Pb, 47Sc, 149Tb, and 166Ho. In particular embodiments, M is selected from 177Lu, 225Ac, 213Bi, and 203Pb. In more particular embodiments, M is 177Lu. [0054] In particular embodiments, the compound of formula (I) is selected from: ; ; ;
42332.601_P15594-02
nd p g g g p y g p n the epsilon amine position of the lysine linker are disclosed in U.S. Patent No.11,458,213 for Prostate-specific 10 42332.601_P15594-02
membrane antigen targeted high-affinity agents for endoradiotherapy of prostate cancer to Ray et al., issued Oct. 4, 2022, which is incorporated by reference in its entirety. [0063] In other embodiments, the presently disclosed subject matter provides a method for treating or imaging one or more PSMA expressing tumors or cells, the method comprising contacting the one or more PSMA expressing tumors or cells with an effective amount of a compound of formula (I), and, when the method is an imaging method, taking an image. [0064] In certain embodiments, the one or more PSMA-expressing tumors or cells is selected from the group consisting of a prostate tumor or cell, a metastasized prostate tumor or cell, a lung tumor or cell, a renal tumor or cell, a glioblastoma, a pancreatic tumor or cell, a bladder tumor or cell, a sarcoma, a melanoma, a breast tumor or cell, a colon tumor or cell, a germ cell, a pheochromocytoma, an esophageal tumor or cell, a stomach tumor or cell, and combinations thereof. In particular embodiments, the one or more PSMA-expressing tumors or cells is a prostate tumor or cell. [0065] In certain embodiments, the one or more PSMA-expressing tumor or cell is in vitro, in vivo, or ex vivo. In certain embodiments, the one or more PSMA-expressing tumor or cell is present in a subject. In particular embodiments, the subject is human. [0066] In certain embodiments, administering the compound of formula (I) to the subject results in inhibition of tumor growth. [0067] As used herein, the term “treating” can include reversing, alleviating, inhibiting the progression of, preventing, or reducing the likelihood of the disease, disorder, or condition to which such term applies, or one or more symptoms or manifestations of such disease, disorder, or condition. Preventing refers to causing a disease, disorder, condition, or symptom or manifestation of such, or worsening of the severity of such, not to occur. Accordingly, the presently disclosed compounds can be administered prophylactically to prevent or reduce the incidence or recurrence of the disease, disorder, or condition. [0068] The “subject” treated by the presently disclosed methods in their many embodiments is desirably a human subject, although it is to be understood that the methods described herein are effective with respect to all vertebrate species, which are intended to be included in the term “subject.” Accordingly, a “subject” can include a human subject for medical purposes, such as for the treatment of an existing condition or disease or the prophylactic treatment for preventing the onset of a condition or disease, or an animal subject for medical, veterinary purposes, or 11 42332.601_P15594-02
developmental purposes. Suitable animal subjects include mammals including, but not limited to, primates, e.g., humans, monkeys, apes, and the like; bovines, e.g., cattle, oxen, and the like; ovines, e.g., sheep and the like; caprines, e.g., goats and the like; porcines, e.g., pigs, hogs, and the like; equines, e.g., horses, donkeys, zebras, and the like; felines, including wild and domestic cats; canines, including dogs; lagomorphs, including rabbits, hares, and the like; and rodents, including mice, rats, and the like. An animal may be a transgenic animal. In some embodiments, the subject is a human including, but not limited to, fetal, neonatal, infant, juvenile, and adult subjects. Further, a “subject” can include a patient afflicted with or suspected of being afflicted with a condition or disease. Thus, the terms “subject” and “patient” are used interchangeably herein. The term “subject” also refers to an organism, tissue, cell, or collection of cells from a subject. [0069] In general, a “therapeutically effective amount” of a therapeutic agent refers to the amount of the agent necessary to elicit the desired biological response. As will be appreciated by those of ordinary skill in the art, the effective amount of an agent may vary depending on such factors as the desired biological endpoint, the agent to be delivered, the composition of the pharmaceutical composition, the target tissue or cell, and the like. In some embodiments, the term “therapeutically effective amount” refers to an amount sufficient to reduce or ameliorate the severity, duration, progression, or onset of a disease, disorder, or condition, or one or more symptoms thereof; prevent the advancement of a disease, disorder, or condition, cause the regression of a disease, disorder, or condition; prevent the recurrence, development, onset or progression of a symptom associated with a disease, disorder, or condition, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy. [0070] The term “combination” is used in its broadest sense and means that a subject is administered at least two agents, more particularly a compound disclosed herein and at least one other therapeutic agent. More particularly, the term “in combination” refers to the concomitant administration of two (or more) active agents for the treatment of a, e.g., single disease state. As used herein, the active agents may be combined and administered in a single dosage form, may be administered as separate dosage forms at the same time, or may be administered as separate dosage forms that are administered alternately or sequentially on the same or separate days. In one embodiment of the presently disclosed subject matter, the active agents are combined and administered in a single dosage form. In another embodiment, the active agents are administered 12 42332.601_P15594-02
in separate dosage forms (e.g., wherein it is desirable to vary the amount of one but not the other). The single dosage form may include additional active agents for the treatment of the disease state. [0071] Further, the compounds disclosed herein can be administered alone or in combination with adjuvants that enhance stability of the compounds, alone or in combination with one or more therapeutic agents, facilitate administration of pharmaceutical compositions containing them in certain embodiments, provide increased dissolution or dispersion, increase inhibitory activity, provide adjunct therapy, and the like, including other active ingredients. Advantageously, such combination therapies utilize lower dosages of the conventional therapeutics, thus avoiding possible toxicity and adverse side effects incurred when those agents are used as monotherapies. [0072] The timing of administration of a compound disclosed herein and at least one additional therapeutic agent can be varied so long as the beneficial effects of the combination of these agents are achieved. Accordingly, the phrase “in combination with” refers to the administration of a compound described herein and at least one additional therapeutic agent either simultaneously, sequentially, or a combination thereof. Therefore, a subject administered a combination of a compound described herein and at least one additional therapeutic agent can receive a compound and at least one additional therapeutic agent at the same time (i.e., simultaneously) or at different times (i.e., sequentially, in either order, on the same day or on different days), so long as the effect of the combination of both agents is achieved in the subject. [0073] When administered sequentially, the agents can be administered within 1, 5, 10, 30, 60, 120, 180, 240 minutes or longer of one another. In other embodiments, agents administered sequentially, can be administered within 1, 5, 10, 15, 20 or more days of one another. Where the compound described herein and at least one additional therapeutic agent are administered simultaneously, they can be administered to the subject as separate pharmaceutical compositions, each comprising either a compound or at least one additional therapeutic agent, or they can be administered to a subject as a single pharmaceutical composition comprising both agents. [0074] When administered in combination, the effective concentration of each of the agents to elicit a particular biological response may be less than the effective concentration of each agent when administered alone, thereby allowing a reduction in the dose of one or more of the agents relative to the dose that would be needed if the agent was administered as a single agent. The effects of multiple agents may, but need not be, additive or synergistic. The agents may be administered multiple times. 13 42332.601_P15594-02
[0075] In some embodiments, when administered in combination, the two or more agents can have a synergistic effect. As used herein, the terms “synergy,” “synergistic,” “synergistically” and derivations thereof, such as in a “synergistic effect” or a “synergistic combination” or a “synergistic composition” refer to circumstances under which the biological activity of a combination of a compound described herein and at least one additional therapeutic agent is greater than the sum of the biological activities of the respective agents when administered individually. [0076] Synergy can be expressed in terms of a “Synergy Index (SI),” which generally can be determined by the method described by F. C. Kull et al., Applied Microbiology 9, 538 (1961), from the ratio determined by: [0077] Qa/QA + Qb/QB = Synergy Index (SI) [0078] wherein: [0079] QA is the concentration of a component A, acting alone, which produced an end point in relation to component A; [0080] Qa is the concentration of component A, in a mixture, which produced an end point; [0081] QB is the concentration of a component B, acting alone, which produced an end point in relation to component B; and [0082] Qb is the concentration of component B, in a mixture, which produced an end point. [0083] Generally, when the sum of Qa/QA and Qb/QB is greater than one, antagonism is indicated. When the sum is equal to one, additivity is indicated. When the sum is less than one, synergism is demonstrated. The lower the SI, the greater the synergy shown by that particular mixture. Thus, a “synergistic combination” has an activity higher that what can be expected based on the observed activities of the individual components when used alone. Further, a “synergistically effective amount” of a component refers to the amount of the component necessary to elicit a synergistic effect in, for example, another therapeutic agent present in the composition. [0084] Depending on the specific conditions being treated, the “agent(s)” may be formulated into liquid or solid dosage forms and administered systemically or locally. The agents may be delivered, for example, in a timed- or sustained-slow release form as is known to those skilled in the art. Techniques for formulation and administration may be found in Remington: The Science and Practice of Pharmacy (20th ed.) Lippincott, Williams & Wilkins (2000). Suitable routes may include oral, buccal, by inhalation spray, sublingual, rectal, transdermal, vaginal, transmucosal, nasal or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, 14 42332.601_P15594-02
intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intra-articular, intra-sternal, intra-synovial, intra-hepatic, intralesional, intracranial, intraperitoneal, intranasal, or intraocular injections or other modes of delivery. [0085] For injection, the agents of the disclosure may be formulated and diluted in aqueous solutions, such as in physiologically compatible buffers such as Hank’s solution, Ringer’s solution, or physiological saline buffer. For such transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0086] Use of pharmaceutically acceptable inert carriers to formulate the compounds herein disclosed for the practice of the disclosure into dosages suitable for systemic administration is within the scope of the disclosure. With proper choice of carrier and suitable manufacturing practice, the compositions of the present disclosure, in particular, those formulated as solutions, may be administered parenterally, such as by intravenous injection. The compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration. Such carriers enable the compounds of the disclosure to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a subject (e.g., patient) to be treated. [0087] For nasal or inhalation delivery, the agents of the disclosure also may be formulated by methods known to those of skill in the art, and may include, for example, but not limited to, examples of solubilizing, diluting, or dispersing substances, such as saline; preservatives, such as benzyl alcohol; absorption promoters; and fluorocarbons. [0088] In particular embodiments, the compound disclosed herein is administered intranasally in a form selected from the group consisting of a nasal spray, a nasal drop, a powder, a granule, a cachet, a tablet, an aerosol, a paste, a cream, a gel, an ointment, a salve, a foam, a paste, a lotion, a cream, an oil suspension, an emulsion, a solution, a patch, and a stick. As used herein, the term administrating via an "intranasal route" refers to administering by way of the nasal structures. [0089] Pharmaceutical compositions suitable for use in the present disclosure include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Generally, the compounds according to the disclosure are effective over a wide dosage range. For example, in the 15 42332.601_P15594-02
treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used. A non-limiting dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, the bioavailability of the compound(s), the adsorption, distribution, metabolism, and excretion (ADME) toxicity of the compound(s), and the preference and experience of the attending physician. [0090] In addition to the active ingredients, these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. The preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions. [0091] Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethyl-cellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone). If desired, disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. [0092] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol (PEG), and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dye-stuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. [0093] Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in 16 42332.601_P15594-02
suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols (PEGs). In addition, stabilizers may be added. [0094] Further, one of ordinary skill in the art will recognize that the presently disclosed compounds, and pharmaceutical compositions thereof, include pharmaceutically acceptable salts. Pharmaceutically acceptable salts are generally well known to those of ordinary skill in the art, and include salts of active compounds that can be prepared with relatively nontoxic acids or bases, depending on the particular substituent moieties found on the compounds described herein. The parent form of the compound can differ from the various salt forms in certain physical properties, such as solubility, and the like. [0095] When compounds of the present disclosure contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent or by ion exchange, whereby one basic counterion (base) in an ionic complex is substituted for another. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, magnesium, and the like. [0096] When compounds of the present disclosure contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent or by ion exchange, whereby one acidic counterion (acid) in an ionic complex is substituted for another. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids, organic acids, and amino acids. See, for example, Berge et al, “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19). Compounds containing both basic and acidic functionalities allow such compounds to be converted into either base or acid addition salts. [0097] Accordingly, pharmaceutically acceptable salts suitable for use with the presently disclosed subject matter include, by way of example but not limitation, acetate, arginate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, monohydrogencarbonate, citrate, edetate, edisylate, estolate, esylate, fumarate, galactonate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydriodic, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, isobutyrate, lactate, lactobionate, malate, maleate, malonate, mandelate, mesylate, methanesulfonate, mucate, napsylate, nitrate, pamoate (embonate), pantothenate, phosphate, phthalate, diphosphate, 17 42332.601_P15594-02
monohydrogen phosphate, dihydrogen phosphate, polygalacturonate, propionate, salicylate, stearate, subacetate, suberate, succinate, sulfate, monohydrogensulfate, tannate, tartrate, including (+)-tartrates, (-)-tartrates, and mixtures thereof including racemic mixtures, teoclate, p- toluenesulfonate and trifluoroacetate. Other pharmaceutically acceptable salts may be found in, for example, Remington: The Science and Practice of Pharmacy (20th ed.) Lippincott, Williams & Wilkins (2000). [0098] Unless otherwise noted, the chemical definitions provided immediately herein below are intended to comply with IUPAC. Compendium of Chemical Terminology, 2nd ed. (the "Gold Book"). Compiled by A. D. McNaught and A. Wilkinson. Blackwell Scientific Publications, Oxford (1997). [0099] The term “hydrocarbon” as used herein, refers to any chemical group comprising hydrogen and carbon. A hydrocarbon group may be substituted or unsubstituted. As would be known to one of ordinary skill in the art, all valencies must be satisfied in making any substitutions. The hydrocarbon may be unsaturated, saturated, branched, unbranched, cyclic, polycyclic, or heterocyclic. [00100] The term “aliphatic” or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocyclyl”, “cycloaliphatic”, or “cycloalkyl”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-4 alipatic carbon atoms. In some embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” (or “carbocyclyl” or “cycloalkyl”) refers to a monocyclic C3-C7 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl, or (cycloalkyl)alkenyl. 18 42332.601_P15594-02
[00101] The term “alkane” refers to acyclic branched or unbranched hydrocarbons having the general formula CnH2n+2, and therefore consisting entirely of hydrogen atoms and saturated carbon atoms. [00102] The term “alkyl” refers to a univalent group derived from an alkane by removal of a hydrogen atom from any carbon atom and having the chemical formula of -CnH2n+1. The groups derived by removal of a hydrogen atom from a terminal carbon atom of unbranched alkanes form a subclass of normal alkyl (n-alkyl) groups H(CH2)n. The groups RCH2, R2CH (R ≠ H), and R3C (R ≠ H) are primary, secondary and tertiary alkyl groups, respectively. [00103] An alkyl can be a straightchain (i.e., unbranched) or branched acyclic hydrocarbon having the number of carbon atoms designated (i.e., C1-10 means one to ten carbons, including 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10 carbons). In particular embodiments, the term “alkyl” refers to C1-20 inclusive, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20 carbons. In other embodiments, the alkyl can be a C1-C4 alkyl, including 1, 2, 3, and 4 carbons. In yet other embodiments, the alkyl can be a C1-C6 alkyl, including 1, 2, 3, 4, 5, and 6 carbons. In even yet other embodiments, the alkyl can be a C1-C8 alkyl, including 1, 2, 3, 4, 5, 6, 7, and 8 carbons. [00104] “Lower alkyl” refers to an alkyl group having 1 to about 8 carbon atoms (i.e., a C1-8 alkyl), e.g., 1, 2, 3, 4, 5, 6, 7, or 8 carbon atoms. “Higher alkyl” refers to an alkyl group having about 10 to about 20 carbon atoms, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms. In certain embodiments, “alkyl” refers to straight-chain alkyls. In other embodiments, “alkyl” refers to branched alkyls. In certain other embodiments, “alkyl” refers to straight-chain and/or branched alkyls. “Branched” refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain. [00105] Representative alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, sec-pentyl, isopentyl, neopentyl, n- hexyl, sec-hexyl, n-heptyl, n-octyl, n-decyl, n-undecyl, and dodecyl. [00106] Alkyl groups can optionally be substituted (a “substituted alkyl”) with one or more substituents, which can be the same or different. Such substituent groups include, but are not limited to, alkyl, substituted alkyl, cycloalkyl, halogen, acyl, carboxyl, oxo, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, cyano, and mercapto. [00107] The term “heteroalkyl,” by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain having from 1 to 20 carbon atoms or 19 42332.601_P15594-02
heteroatoms consisting of at least one carbon atom and at least one heteroatom selected from the group consisting of O, N, P, Si and S, and wherein the nitrogen, phosphorus, and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N, P and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which alkyl group is attached to the remainder of the molecule. Examples include, but are not limited to, -CH2-CH2-O-CH3, -CH2-CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, - CH2-S-CH2-CH3, -CH2-CH2-S(O)-CH3, -CH2-CH2-S(O)2-CH3, -CH=CH-O-CH3, -Si(CH3)3, - CH2-CH=N-OCH3, -CH=CH-N(CH3)- CH3, O-CH3, -O-CH2-CH3, and -CN. Up to two or three heteroatoms may be consecutive, such as, for example, -CH2-NH-OCH3 and -CH2-O-Si(CH3)3. [00108] The term “cycloalkane” refers to saturated monocyclic hydrocarbons (with or without side chains), e.g., cyclobutane. Unsaturated monocyclic hydrocarbons having one endocyclic double or one triple bond are called cycloalkenes and cycloalkynes, respectively. Those having more than one such multiple bond are cycloalkadienes, cycloalkatrienes, and the like. The inclusive terms for any cyclic hydrocarbons having any number of such multiple bonds are cyclic olefins or cyclic acetylenes. [00109] The term “cycloalkyl” refer to a univalent group derived from a cycloalkane by removal of a hydrogen atom from a ring carbon atom. Cycloalkyls can be a mono- or multicyclic ring system of about 3 to about 10 carbon atoms, e.g., 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms. The cycloalkyl group also can be optionally substituted with a substituent group provided hereinabove for alkyl groups. Representative monocyclic cycloalkyl rings include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. Multicyclic cycloalkyl rings include adamantyl, octahydronaphthyl, decalin, camphor, camphane, and noradamantyl, and fused ring systems, such as dihydro- and tetrahydronaphthalene, and the like. [00110] The term “cycloalkylalkyl” as used herein, refers to a cycloalkyl group, which is attached to the parent molecular moiety through an alkylene moiety, also as defined above, e.g., a C1-20 alkylene moiety. Examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl. [00111] The terms “cycloheteroalkyl” and “heterocycloalkyl” (or more generally “heterocyclic”) are used interchangeably and refer to an unsaturated ring system, such as a 3- to 10-member substituted or unsubstituted cycloalkyl ring system, including one or more heteroatoms, which can be the same or different, and are selected from the group consisting of nitrogen (N), oxygen (O), 20 42332.601_P15594-02
sulfur (S), phosphorus (P), and silicon (Si), in which the nitrogen, sulfur, and phosphorus heteroatoms may be oxidized and the nitrogen heteroatom may be quaternized. The cycloheteroalkyl ring can be optionally fused to or otherwise attached to other cycloheteroalkyl rings and/or non-aromatic hydrocarbon rings. Representative cycloheteroalkyl ring systems include, but are not limited to pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidinyl, piperazinyl, indolinyl, quinuclidinyl, morpholinyl, thiomorpholinyl, thiadiazinanyl, tetrahydrofuranyl, and the like. [00112] The terms “cycloalkylene” and “heterocycloalkylene” refer to the divalent derivatives of cycloalkyl and heterocycloalkyl, respectively. [00113] As used herein the terms “bicycloalkyl” and “bicycloheteroalkyl” refer to two cycloalkyl or cycloheteroalkyl groups that are bound to one another. Non-limiting examples include bicyclohexane and bipiperidine. [00114] An “unsaturated hydrocarbon” has one or more double bonds or triple bonds. As used herein, the term “alkene” refers to an acyclic branched or unbranched hydrocarbons having one carbon–carbon double bond and the general formula CnH2n. Acyclic branched or unbranched hydrocarbons having more than one double bond are alkadienes, alkatrienes, and the like. [00115] More particularly, the term “alkenyl” as used herein refers to a monovalent group derived from a C2-20 inclusive straight or branched hydrocarbon moiety having at least one carbon-carbon double bond by the removal of a single hydrogen molecule. Alkenyl groups include, but are not limited to, ethenyl (i.e., vinyl), 2-propenyl, butenyl, 1-methyl-2-buten-1-yl, pentenyl, 2- isopentenyl, hexenyl, octenyl, allenyl, butadienyl, crotyl (but-2-en-1-yl), 2-(butadienyl), 2,4- pentadienyl, 3-(l,4-pentadienyl), and the like, including higher homologs and isomers. [00116] The term “cycloalkenyl” as used herein refers to a cyclic hydrocarbon containing at least one carbon-carbon double bond. Examples of cycloalkenyl groups include cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadiene, cyclohexenyl, 1,3-cyclohexadiene, cycloheptenyl, cycloheptatrienyl, and cyclooctenyl. [00117] The term “alkyne” as used herein refers to an acyclic branched or unbranched hydrocarbons having a carbon-carbon triple bond and the general formula CnH2n-2, RC≡CR. Acyclic branched or unbranched hydrocarbons having more than one triple bond are known as alkadiynes, alkatriynes, and the like. 21 42332.601_P15594-02
[00118] The term “alkynyl” as used herein refers to a monovalent group derived from a straight or branched C2-20 hydrocarbon of a designed number of carbon atoms containing at least one carbon- carbon triple bond. Examples of alkynyl groups include, but are not limited to, ethynyl, 2-propynyl (propargyl), 1-propynyl, pentynyl, hexynyl, and heptynyl groups, and the like. [00119] As used herein, the term “alkylene” refers to an alkanediyl group having the free valencies on adjacent carbon atoms, e.g. –CH(CH3)CH2– propylene (systematically called propane-1,2- diyl). More particularly, the term “alkylene” by itself or a part of another substituent refers to a straight or branched bivalent aliphatic hydrocarbon group derived from an alkyl group having from 1 to about 20 carbon atoms, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms. The alkylene group can be straight, branched or cyclic. The alkylene group also can be optionally unsaturated and/or substituted with one or more “alkyl group substituents.” There can be optionally inserted along the alkylene group one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms (also referred to herein as “alkylaminoalkyl”), wherein the nitrogen substituent is alkyl as previously described. Exemplary alkylene groups include methylene (–CH2– ); ethylene (–CH2–CH2–); propylene (–(CH2)3–); cyclohexylene (–C6H10–); –CH=CH–CH=CH–; –CH=CH–CH2–; -CH2CH2CH2CH2-, -CH2CH=CHCH2-, -CH2CsCCH2-, - CH2CH2CH(CH2CH2CH3)CH2-, -(CH2)q-N(R)-(CH2)r–, wherein each of q and r is independently an integer from 0 to about 20, e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and R is hydrogen or lower alkyl; methylenedioxyl (–O–CH2–O–); and ethylenedioxyl (-O-(CH2)2–O–). An alkylene group can have about 2 to about 3 carbon atoms and can further have 6-20 carbons. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being some embodiments of the present disclosure. A “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms. [00120] The term “heteroalkylene” by itself or as part of another substituent means a divalent group derived from heteroalkyl, as exemplified, but not limited by, -CH2-CH2-S-CH2-CH2- and -CH2-S-CH2-CH2-NH-CH2-. For heteroalkylene groups, heteroatoms also can occupy either or both of the chain termini (e.g., alkyleneoxo, alkylenedioxo, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula 22 42332.601_P15594-02
of the linking group is written. For example, the formula -C(O)OR’- represents both -C(O)OR’- and –R’OC(O)-. [00121] The term “arene” refers to a monocyclic and polycyclic aromatic hydrocarbon. [00122] The term “aryl” refers to a group derived from arenes by removal of a hydrogen atom from a ring carbon atom. Groups similarly derived from heteroarenes are sometimes subsumed in this definition. An aryl group can include, for example, a single ring or multiple rings (such as from 2 to 3 rings), which are fused together or linked covalently. [00123] The term “heteroaryl” refers to a group formed by removing one or more hydroxy groups from oxoacids that have the general structure RkE(=O)l(OH)m (l ≠ 0), and replacement analogues of such acyl groups. In organic chemistry an unspecified acyl group is commonly a carboxylic acyl group. [00124] The term “heteroaryl” refers to the class of heterocyclyl groups derived from heteroarenes by removal of a hydrogen atom from any ring atom. A “heteroaryl” group can include from one to four heteroatoms (in each separate ring in the case of multiple rings) selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom. Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2- imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3- isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2- thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl, and 6-quinolyl. Substituents for each of above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below. The terms “arylene” and “heteroarylene” refer to the divalent forms of aryl and heteroaryl, respectively. [00125] For brevity, the term “aryl” when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, the terms “arylalkyl” and “heteroarylalkyl” are meant to include those groups in which an aryl or heteroaryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl, furylmethyl, and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(l- 23 42332.601_P15594-02
naphthyloxy)propyl, and the like). However, the term “haloaryl,” as used herein is meant to cover only aryls substituted with one or more halogens. [00126] Where a heteroalkyl, heterocycloalkyl, or heteroaryl includes a specific number of members (e.g., “3 to 7 membered”), the term “member” refers to a carbon or heteroatom. [00127] Each of above terms defined hereinabove (e.g. , “alkyl,” “heteroalkyl,” “cycloalkyl, and “heterocycloalkyl”, “alkenyl”, “alkynyl,” “aryl,” “heteroaryl,” as well as their divalent derivatives) are meant to include both substituted and unsubstituted forms of the indicated group. Optional substituents for each type of group are provided below. [00128] As used herein, the term “acyl” refers to a group formed by removing one or more hydroxy groups from oxoacids that have the general structure RkE(=O)l(OH)m (l ≠ 0), and replacement analogues of such acyl groups. In organic chemistry an unspecified acyl group is commonly a carboxylic acyl group. For example, in some embodiments, the term acyl includes an organic acid group wherein the -OH of the carboxyl group has been replaced with another substituent and has the general formula RC(=O)-, wherein R is an alkyl, alkenyl, alkynyl, aryl, carbocylic, heterocyclic, or aromatic heterocyclic group as defined herein). As such, the term “acyl” specifically includes arylacyl groups, such as a 2-(furan-2-yl)acetyl)- and a 2-phenylacetyl group. Specific examples of acyl groups include acetyl and benzoyl. Acyl groups also are intended to include amides, -RC(=O)NR’, esters, -RC(=O)OR’, ketones, -RC(=O)R’, and aldehydes, -RC(=O)H. [00129] The terms “alkoxyl” or “alkoxy” are used interchangeably herein and refer to a saturated (i.e., alkyl–O–) or unsaturated (i.e., alkenyl–O– and alkynyl–O–) group attached to the parent molecular moiety through an oxygen atom, wherein the terms “alkyl,” “alkenyl,” and “alkynyl” are as previously described and can include C1-20 inclusive, linear, branched, or cyclic, saturated or unsaturated oxo-hydrocarbon chains, including, for example, methoxyl, ethoxyl, propoxyl, isopropoxyl, n-butoxyl, sec-butoxyl, tert-butoxyl, and n-pentoxyl, neopentoxyl, n-hexoxyl, and the like. [00130] The term “alkoxyalkyl” as used herein refers to an alkyl-O-alkyl ether, for example, a methoxyethyl or an ethoxymethyl group. [00131] “Aryloxyl” refers to an aryl-O- group wherein the aryl group is as previously described, including a substituted aryl. The term “aryloxyl” as used herein can refer to phenyloxyl or hexyloxyl, and alkyl, substituted alkyl, halo, or alkoxyl substituted phenyloxyl or hexyloxyl. 24 42332.601_P15594-02
[00132] “Aralkyl” refers to an aryl-alkyl-group wherein aryl and alkyl are as previously described, and included substituted aryl and substituted alkyl. Exemplary aralkyl groups include benzyl, phenylethyl, and naphthylmethyl. [00133] “Aralkyloxyl” refers to an aralkyl-O– group wherein the aralkyl group is as previously described. An exemplary aralkyloxyl group is benzyloxyl, i.e., C6H5-CH2-O-. An aralkyloxyl group can optionally be substituted. [00134] “Alkoxycarbonyl” refers to an alkyl-O-C(=O)– group. Exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, butyloxycarbonyl, and tert-butyloxycarbonyl. [00135] “Aryloxycarbonyl” refers to an aryl-O-C(=O)– group. Exemplary aryloxycarbonyl groups include phenoxy- and naphthoxy-carbonyl. [00136] “Aralkoxycarbonyl” refers to an aralkyl-O-C(=O)– group. An exemplary aralkoxycarbonyl group is benzyloxycarbonyl. [00137] The term “acyloxyl” refers to an oxygen-centered radicals consisting of an acyl radical bonded to an oxygen atom, e.g., an acyl-O- group wherein acyl is as previously described. [00138] The term “amine” refers to a compound formally derived from ammonia by replacing one, two or three hydrogen atoms by hydrocarbyl groups, and having the general structures RNH2 (primary amines), R2NH (secondary amines), R3N (tertiary amines). In some embodiments, the term amino refers to the –NH2 group. More generally, the amino group is -NR'R”, wherein R' and R” are typically selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. [00139] The terms “acylamino” and “alkylamino” refer to specific N-substituted organic radicals with acyl and alkyl substituent groups, respectively. [00140] An “aminoalkyl” as used herein refers to an amino group covalently bound to an alkylene linker. More particularly, the terms alkylamino, dialkylamino, and trialkylamino as used herein refer to one, two, or three, respectively, alkyl groups, as previously defined, attached to the parent molecular moiety through a nitrogen atom. The term alkylamino refers to a group having the structure –NHR’ wherein R’ is an alkyl group, as previously defined; whereas the term dialkylamino refers to a group having the structure –NR’R”, wherein R’ and R” are each independently selected from the group consisting of alkyl groups. The term trialkylamino refers to a group having the structure –NR’R”R”’, wherein R’, R”, and R’” are each independently selected 25 42332.601_P15594-02
from the group consisting of alkyl groups. Additionally, R’, R”, and/or R’” taken together may optionally be –(CH2)k– where k is an integer from 2 to 6. Examples include, but are not limited to, methylamino, dimethylamino, ethylamino, diethylamino, diethylaminocarbonyl, methylethylamino, isopropylamino, piperidino, trimethylamino, and propylamino. [00141] The terms alkylthioether and thioalkoxyl refer to a saturated (i.e., alkyl–S–) or unsaturated (i.e., alkenyl–S– and alkynyl–S–) group attached to the parent molecular moiety through a sulfur atom. Examples of thioalkoxyl moieties include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like. [00142] “Acylamino” refers to an acyl-NH– group wherein acyl is as previously described. “Aroylamino” refers to an aroyl-NH– group wherein aroyl is as previously described. [00143] The term “carbonyl” refers to a compound containing the carbonyl group, -C(=O)-. The term is commonly used in the restricted sense of aldehydes (R-C(=O)H) and ketones, although it actually includes carboxylic acids and derivatives. [00144] The term “carboxylic acid” refers to an oxoacids having the structure RC(=O)OH. The term is used as a suffix in systematic name formation to denote the –C(=O)OH group including its carbon atom. In some embodiments, the term “carboxyl” refers to the –COOH group. Such groups also are referred to herein as a “carboxylic acid” moiety. [00145] “Carbamoyl” refers to an amide group of the formula –C(=O)NH2. [00146] “Alkylcarbamoyl” refers to a R’RN–C(=O)– group wherein one of R and R’ is hydrogen and the other of R and R’ is alkyl and/or substituted alkyl as previously described. [00147] “Dialkylcarbamoyl” refers to a R’RN–C(=O)– group wherein each of R and R’ is independently alkyl and/or substituted alkyl as previously described. [00148] The term carbonyldioxyl, as used herein, refers to a carbonate group of the formula -O- C(=O)-OR. [00149] The term “cyano” refers to the -C≡N group. [00150] The terms “halo,” “halide,” or “halogen” as used herein refer to fluoro, chloro, bromo, and iodo groups. Additionally, terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl. For example, the term “halo(C1-4)alkyl” is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. [00151] The term “hydroxyl” refers to the –OH group. [00152] The term “hydroxyalkyl” refers to an alkyl group substituted with an –OH group. 26 42332.601_P15594-02
[00153] The term “mercapto” refers to the –SH group. [00154] The term “oxo compound” refers to a compounds containing an oxygen atom, =O, doubly bonded to carbon or another element. The term thus embraces aldehydes, carboxylic acids, ketones, sulfonic acids, amides and esters. Oxo used as an adjective (and thus separated by a space) modifying another class of compound, as in oxo carboxylic acids, indicates the presence of an oxo substituent at any position. To indicate a double-bonded oxygen that is part of a ketonic structure, the term keto is sometimes used as a prefix, but such use has been abandoned by IUPAC for naming specific compounds. A traditional use of keto is for indicating oxidation of CHOH to C=O in a parent compound that contains OH groups, such as carbohydrates, e.g., 3-ketoglucose. In some embodiments, the term “oxo” as used herein means an oxygen atom that is double bonded to a carbon atom or to another element. [00155] The term “nitro” refers to the –NO2 group. [00156] The term “thio” refers to replacement of an oxygen by a sulfur, e.g., PhC(=S)NH2, thiobenzamide. [00157] The term “thiol” refers to a compounds having the structure RSH (R ≠ H), e.g., MeCH2SH ethanethiol. A thiol also is known by the term “mercaptan.” [00158] The term “thiohydroxyl” or “thiol,” as used herein, refers to a group of the formula –SH. [00159] The term “sulfate” refers to the –SO4 group. [00160] The term “sulfide” refers to a compound having the structure RSR (R ≠ H) and also are referred to as “thioethers.” [00161] The term “sulfone” refers to a compound having the structure, RS(=O)2R (R ≠ H), e.g., C2H5S(=O)2CH3 ethyl methyl sulfone. [00162] The term “sulfoxide” refers to a compound having the structure R2S=O (R ≠ H), e.g., Ph2S=O diphenyl sulfoxide. [00163] The term “ureido” refers to a urea group of the formula –NH—CO—NH2. [00164] One of ordinary skill in the art would recognize that a structure represented generally by, for example, the formula:
42332.601_P15594-02
[00166] as used herein refers to a ring structure, for example, but not limited to a 3-carbon, a 4- carbon, a 5-carbon, a 6-carbon, a 7-carbon, and the like, aliphatic and/or aromatic cyclic compound, including a saturated ring structure, a partially saturated ring structure, and an unsaturated ring structure, comprising a substituent R group, wherein the R group can be present or absent, and when present, one or more R groups can each be substituted on one or more available carbon atoms of the ring structure. The presence or absence of the R group and number of R groups is determined by the value of the variable “n,” which is an integer generally having a value ranging from 0 to the number of carbon atoms on the ring available for substitution. Each R group, if more than one, is substituted on an available carbon of the ring structure rather than on another R group. For example, the structure above where n is 0 to 2 would comprise compound groups including, but not limited to:
ring structure indicates that the bond can be either present or absent in the ring. That is, a dashed line representing a bond in a cyclic ring structure indicates that the ring structure is selected from the group consisting of a saturated ring structure, a partially saturated ring structure, and an unsaturated ring structure. [00170] The symbol ( ) denotes the point of attachment of a moiety to the remainder of the molecule.
[00171] When a named atom of an aromatic ring or a heterocyclic aromatic ring is defined as being “absent,” the named atom is replaced by a direct bond. [00172] Throughout the specification and claims, a given chemical formula or name shall encompass all tautomers, congeners, and optical- and stereoisomers, as well as racemic mixtures where such isomers and mixtures exist. [00173] Certain compounds of the present disclosure may possess asymmetric carbon atoms (optical or chiral centers) or double bonds; the enantiomers, racemates, diastereomers, tautomers, 28 42332.601_P15594-02
geometric isomers, stereoisometric forms that may be defined, in terms of absolute stereochemistry, as (R)-or (S)- or, as D- or L- for amino acids, and individual isomers are encompassed within the scope of the present disclosure. The compounds of the present disclosure do not include those which are known in art to be too unstable to synthesize and/or isolate. The present disclosure is meant to include compounds in racemic, scalemic, and optically pure forms. Optically active (R)- and (S)-, or D- and L-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefenic bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. [00174] Unless otherwise stated, structures depicted herein are also meant to include all stereochemical forms of the structure; i.e., the R and S configurations for each asymmetric center. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric mixtures of the present compounds are within the scope of the disclosure. [00175] As used herein, the term “congener” refers to one of two or more substances related to each other by origin, structure, or function. [00176] The term “enantiomer” refers to one of a pair of molecular entities which are mirror images of each other and non-superposable. [00177] The term “stereoisomer” refers to an isomer that possess identical constitution, but which differ in the arrangement of their atoms in space. [00178] The term “racemate” refers to an equimolar mixture of a pair of enantiomers. It does not exhibit optical activity. The chemical name or formula of a racemate is distinguished from those of the enantiomers by the prefix (±)- or rac- (or racem-) or by the symbols RS and SR. [00179] The term “diastereoisomerism” refers to stereoisomerism other than enantiomerism. Diastereoisomers (or diastereomers) are stereoisomers not related as mirror images. Diastereoisomers are characterized by differences in physical properties, and by some differences in chemical behavior towards achiral as well as chiral reagents. [00180] It will be apparent to one skilled in the art that certain compounds of this disclosure may exist in tautomeric forms, all such tautomeric forms of the compounds being within the scope of the disclosure. The term “tautomer,” as used herein, refers to one of two or more structural isomers which exist in equilibrium and which are readily converted from one isomeric form to another. 29 42332.601_P15594-02
[00181] Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures with the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C- or 14C-enriched carbon are within the scope of this disclosure. [00182] The compounds of the present disclosure may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) or carbon-14 (14C). All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure. [00183] The term “about,” when used in connection with one or more numbers or numerical ranges, should be understood to refer to all such numbers, including all numbers in a range and modifies that range by extending the boundaries slightly above and slightly below the numerical values set forth by, for example, in some embodiments, +/-20%, +/-15%, +/-10%, +/-5%, +/-4%, +/-3%, +/- 2%, and +/-1%. The recitation of numerical ranges by endpoints includes all numbers, e.g., whole integers, including fractions thereof, subsumed within that range (for example, the recitation of 1 to 5 includes 1, 2, 3, 4, and 5, as well as fractions thereof, e.g., 1.5, 2.25, 3.75, 4.1, and the like) and any range within that range. [00184] The phrase “in one embodiment” or “in some embodiments” as used herein does not necessarily refer to the same embodiment, though it may. Furthermore, the phrase “in another embodiment” as used herein does not necessarily refer to a different embodiment, although it may. Thus, as described below, various embodiments of the invention may be readily combined, without departing from the scope or spirit of the invention. [00185] The terms “comprise(s),” “include(s),” “having,” “has,” “can,” “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that do not preclude the possibility of additional acts or structures. The singular forms “a,” “and” and “the” include plural references, i.e., “one or more,” unless the context clearly dictates otherwise. The present disclosure also contemplates other embodiments “comprising,” “consisting of” and “consisting essentially of,” the embodiments or elements presented herein, whether explicitly set forth or not. Likewise, the term “include” and its grammatical variants are intended to be non- 30 42332.601_P15594-02
limiting, such that recitation of items in a list is not to the exclusion of other like items that can be substituted or added to the listed items. EXAMPLES [00186] The following Examples have been included to provide guidance to one of ordinary skill in the art for practicing representative embodiments of the presently disclosed subject matter. In light of the present disclosure and the general level of skill in the art, those of skill can appreciate that the following Examples are intended to be exemplary only and that numerous changes, modifications, and alterations can be employed without departing from the scope of the presently disclosed subject matter. The synthetic descriptions and specific examples that follow are only intended for the purposes of illustration and are not to be construed as limiting in any manner to make compounds of the disclosure by other methods. EXAMPLE 1 [00187] Preclinical Evaluation of a Series of Albumin-Binding 177Lu-Labeled PSMA-Based Low- Molecular-Weight Radiotherapeutics [00188] Overview [00189] Prostate-specific membrane antigen (PSMA)-based low-molecular-weight agents using beta(β)-particle-emitting radiopharmaceuticals is a new treatment paradigm for patients with metastatic castration-resistant prostate cancer. Although results have been encouraging, there is a need to improve the tumor residence time of current PSMA-based radiotherapeutics. Albumin- binding moieties have been used strategically to enhance the tumor uptake and retention of existing PSMA-based investigational agents. A series of PSMA-based, β-particle-emitting, low-molecular- weight compounds was previously developed. From this series, 177Lu-L1 was selected as the lead agent because of its reduced off-target radiotoxicity in preclinical studies. The ligand L1 of the compound 177Lu-L1 contains a PSMA-targeting Lys-Glu urea moiety with an N-bromobenzyl substituent in the ε-amino group of Lys. [00190] In this Example, 177Lu-L1 was structurally modified to improve tumor targeting using two albumin-binding moieties, 4-(p-iodophenyl)butyric acid moiety (IPBA) and ibuprofen (IBU), and the effects of linker length and composition was evaluated. Six structurally related PSMA- targeting ligands (Alb-L1, Alb-L2, Alb-L3, Alb-L4, Alb-L5, and Alb-L6) were synthesized based on the structure of 177Lu-L1. The ligands were assessed for in vitro binding affinity and were 31 42332.601_P15594-02
radiolabeled with 177Lu following standard protocols. All 177Lu-labeled analogs were studied in cell uptake and selected cell efficacy studies. In vivo pharmacokinetics were investigated by conducting tissue biodistribution studies for 177Lu-Alb-L2, 177Lu-Alb-L3, 177Lu-Alb-L4, 177Lu- Alb-L5, and 177Lu-Alb-L6 (2 h, 24 h, 72 h, and 192 h) in male NSG mice bearing human PSMA+ PC3 PIP and PSMA− PC3 flu xenografts. [00191] Preliminary therapeutic ratios of the agents were estimated from the area under the curve (AUC0-192h) of the tumors, blood, and kidney uptake values. Compounds were obtained in greater than 98% radiochemical yields and greater than 99% purity. PSMA inhibition constants (Kis) of the ligands were in the ≤10 nM range. The long-linker-based agents, 177Lu-Alb-L4 and 177Lu-Alb- L5, displayed significantly higher tumor uptake and retention (p < 0.001) than the short-linker- bearing 177Lu-Alb-L2 and 177Lu-Alb-L3 and a long polyethylene glycol (PEG) linker-bearing agent, 177Lu-Alb-L6. The area under the curve (AUC0-192h) of the PSMA+ PC3 PIP tumor uptake of 177Lu-Alb-L4 and 177Lu-Alb-L5 were greater than 4-fold higher than 177Lu-Alb-L2, 177Lu-Alb- L3, and 177Lu-Alb-L6, respectively. Also, the PSMA+ PIP tumor uptake (AUC0-192h) of 177Lu-Alb- L2 and 177Lu-Alb-L3 was approximately 1.5-fold higher than 177Lu-Alb-L6. The lowest blood AUC0-192h and kidney AUC0-192h, however, were associated with 177Lu-Alb-L6 from the series. Consequently, 177Lu-Alb-L6 displayed the highest ratios of AUC(tumor)-to-AUC(blood) and AUC(tumor)-to-AUC(kidney) values from the series. Among the other agents, 177Lu-Alb-L4 demonstrated a nearly similar ratio of AUC(tumor)-to-AUC(blood) as 177Lu-Alb-L6. The tumor- to-blood ratio was the dose-limiting therapeutic ratio for all of the compounds. [00192] In summary, 177Lu-Alb-L4 and 177Lu-Alb-L6 showed high tumor uptake in PSMA+ tumors and tumor-to-blood ratios. These data suggest that linker length and composition can be modulated to generate an optimized therapeutic agent. [00193] Background [00194] One way to potentially develop improved radiopharmaceutical therapies targeting PSMA is through the pharmacokinetic optimization of agents similar to 177Lu-PSMA-617. Several approaches involving the structural manipulation of PSMA-targeting ureas are currently under intense investigation to improve the efficacy of 177Lu-PSMA-617. Sandhu et al., 2021. Examples of such optimization include the development of bivalent or high-affinity agents and the incorporation of pharmacophores to enhance their blood circulation. Banerjee et al., 2011; Kopka et al., 2017; Kelly et al., 2018; Kelly et al., 2019; Wang et al., 2022; Murce et al., 2023. 32 42332.601_P15594-02
[00195] Along this direction, incorporating an albumin-binding moiety into PSMA-based, low- molecular-weight agents has gained significant attention. Kelly et al., 2018; Wang et al., 2022; Choy et al., 2017; Benešová et al., 2018; Lau et al., 2019; Zang et al., 2020; Ling et al., 2020; Kuo et al., 2021; Reissig et al., 2022. Well-characterized, small-molecule moieties (e.g., 4-(para- iodophenyl) butyric acid (IPBA), truncated Evans blue, ibuprofen (IBU), and fatty acids), referred to as “blood half-life extenders”, non-covalently bind to serum albumin and have been utilized extensively. Zorzi et al., 2019. Several agents of this class have entered clinical trials, as shown in FIG. 1 (prior art). [00196] The majority of these agents are derived from the structure of PSMA-617. Kuo et al., 2021; Kramer et al., 2021; Tschan et al., 2022. Preclinical and clinical data have revealed increased tumor targeting of such agents. Increased radiation dose delivery to the salivary glands and kidneys, however, also was observed partly due to PSMA expression in these tissues. Wang et al., 2022; Zang et al., 2020; Kramer et al., 2021. In addition, increased off-target radiotoxicity, specifically hematologic toxicity, has been demonstrated in clinical studies. Wang et al., 2022; Kramer et al., 2021. Accordingly, current preclinical efforts are focused on optimizing the PSMA-targeting moiety, Kuo et al., 2021; Kuo et al., 2022, linker composition, Kelly et al., 2019; Deberle et al., 2020, and albumin-binding motif, Kuo et al., 2021; Deberle et al., 2020, to maximize the tumor- to-kidney radiation dose ratios. Improving tumor targeting and weakening albumin binding to reduce blood, kidney, or salivary gland uptake is the primary rationale for newly developed agents. Zang et al., 2020; . Kuo et al., 2022; Deberle et al., 2020. [00197] We recently developed a series of PSMA-based agents with reduced off-target toxicity, one of which was translated into the clinic. Banerjee et al., 2019. The lead agent, L1, also was investigated using alpha (α)-particle-emitting radionuclides, including 225Ac, 213Bi, 212Pb, and 211At. Banerjee et al., 2020; Banerjee et al., 2021; Mease et al., 2022. [00198] In this Example, we evaluated six albumin-binding agents by conjugating IBU and IPBA using two bifunctional linkers. One goal of this Example was to generate an agent with enhanced tumor uptake and retention compared to 177Lu-L1. We investigated two albumin-binding moieties and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid mono-amide (DOTA-monoamide) as a chelating unit to generate six ligands bearing either a short (A) (177Lu-Alb-L1–177Lu-Alb-L3) or long linker (B) (177Lu-Alb-L4–Alb-L6) (FIG. 2). Without wishing to be bound to any one 33 42332.601_P15594-02
particular theory, these data indicate that the linker length and composition might be critical in optimizing PSMA- and albumin-based agents for clinical use. [00199] Results [00200] Synthesis and Binding Affinities of Ligands Alb-L1 to Alb-L6 [00201] We designed and synthesized six ligands using (i) PSMA-targeting Glu-Lys (N-p-bromo- benzyl) urea moiety, (ii) short linker (Structure A) or long linker (Structure B), and (iii) albumin- binding moieties IPBA and IBU, as shown in FIG. 2. Standard solution-phase peptide conjugation chemistry was employed to synthesize these new compounds, as provided in more detail herein below. After evaluating compounds in the series 177Lu-Alb-L1 to 177Lu-Alb-L5, we studied 177Lu- Alb-L6, containing a long polyethylene glycol (PEG) linker, to reduce the blood uptake of these agents. [00202] In Vitro Characterization [00203] Binding Affinity, Cell Uptake, and Internalization [00204] The new ligands displayed high binding affinity to PSMA with Ki values ranging from 0.12 nM to 11.24 nM, as listed in FIG. 2. Ligands Alb-L4 and Alb-L5 from Structure B construct and Alb-L3 displayed significantly higher affinity (p < 0.05) than the other ligands. The albumin- binding properties of the agents revealed a substantially higher affinity for the long-linker-based agents, 177Lu-Alb-L4 and 177Lu-Alb-L5, compared to 177Lu-Alb-L6 and the short-linker-based 177Lu-Alb-L2, as well as 177Lu-Alb-L3 (Table 3). The cell uptake and internalized fraction of the 177Lu-labeled agents are shown in FIG. 3. Except for 177Lu-Alb-L1 (without a p-bromo-benzyl group on the PSMA-targeting unit), all of the agents demonstrated high cell uptake in the PSMA+ PC3 PIP cells, i.e., in the range of 28% to 75% of the incubated dose at 2 h, and this was slightly increased up to 30% to 77% at 24 h incubation. From the series, 177Lu-Alb-L4 and 177Lu-Alb-L5 displayed high uptake, 60% and 75%, respectively, at 2 h, and 63% and 77% at 24 h of incubation in PSMA+ PC3 PIP cells. The percentage of internalization for the agents in PSMA+ PC3 PIP cells was in the range of 32–47% at 2 h. The uptake of the radioligands in the PSMA− PC3 flu cells was significantly lower (approximately 100-fold) than that of the PSMA+ PC3 PIP cells, revealing the PSMA-specific binding of the agents. A PSMA expression blocking study was performed using an excess of ZJ43, Olszewski et al., 2004, a known PSMA inhibitor, which caused a significantly low uptake of the radiolabeled agents in PSMA+ PC3 PIP cells (FIG.3). These data 34 42332.601_P15594-02
further confirmed the PSMA-mediated uptake of 177Lu-Alb-L1–177Lu-Alb-L6 in PSMA+ PC3 PIP cells. [00205] Clonogenic Survival Assay [00206] Clonogenic assays were performed to assess the cellular efficacy of 177Lu-Alb-L2 and 177Lu-Alb-L5 and were compared with 177Lu-L1, as shown in FIG.4. The data revealed a decrease in the survival of PSMA+ PC3 PIP cells with increasing concentrations of radioactivity, with complete cell killing reached at a concentration of 370 kBq/mL for 177Lu-L1, 185 kBq/mL for 177Lu-Alb-L2, and 37 kBq/mL for 177Lu-Alb-L5, respectively. The activity to reduce cell survival to 37% (A0) was 18.5 kBq/mL for 177Lu-L1, approximately 20 kBq/mL for 177Lu-Alb-L2, and approximately 10 kBq/mL for 177Lu-Alb-L5, respectively. [00207] Biodistribution [00208] Tissue biodistribution studies of the series of compounds form 177Lu-Alb-L2 to 177Lu-Alb- L6 were performed at 2 h, 24 h, 48 h, and 192 h after injection to evaluate the tumor targeting and clearance of the agents from blood and normal tissues. The biodistribution of 177Lu-L1 was conducted at 24 h post-injection for the agents for direct comparison. The biodistribution studies of 177Lu-Alb-L2 and 177Lu-Alb-L5, as well as 177Lu-L1 (24 h), were performed in one experiment to keep the experimental variations minimal (for example, tumor size, animals’ age, and specific activity of 177Lu). Similarly, biodistribution studies of 177Lu-Alb-L3 and 177Lu-Alb-L4, as well as 177Lu-L1 (24 h), were conducted head-to-head in one experiment. The biodistribution study of 177Lu-Alb-L6 was performed separately using tumors of a similar size. [00209] The results of the presently disclosed agents and 177Lu-L1 are presented in FIG. 5 and Tables 4-10. Biodistribution data of 177Lu-L1 (3–192 h) were acquired from our previously reported studies. Banerjee et al., 2019. A fast tumor accumulation was observed for 177Lu-Alb-L2, which reached 17.45 ± 6.51%ID/g at 2 h post-injection and displayed higher uptake, 26.41 ± 6.73%ID/g at 24 h and 22.00 ± 4.71%ID/g at 48 h, respectively. At 192 h post-injection, tumor uptake was low, 3.39 ± 1.03%ID/g. 177Lu-Alb-L3 displayed similar pharmacokinetics as 177Lu- Alb-L2, 34.05 ± 1.98%ID/g at 2 h, 30.55 ± 7.44%ID/g at 24 h, 19.61 ± 4.46%ID/g at 48 h, and 5.94 ± 1.38%ID/g at 192 h. [00210] The long-linker-based agents (Structure B), 177Lu-Alb-L4 and 177Lu-Alb-L5, displayed significantly higher tumor uptake and retention than 177Lu-Alb-L2 and 177Lu-Alb-L3. The tumor uptake of 177Lu-Alb-L4 was 40.89 ± 4.73%ID/g at 2 h, 88.04 ± 16.51%ID/g at 24 h, 87.74 ± 35 42332.601_P15594-02
14.09%ID/g at 48 h, and 42.22 ± 14.05%ID/g at 192 h, respectively. For 177Lu-Alb-L5, it was 28.78 ± 7.25%ID/g at 2 h, 111.47 ± 13.85%ID/g at 24 h, 127.44 ± 22.85%ID/g at 48 h, and 70.96 ± 2.34%ID/g at 192 h, respectively. 177Lu-Alb-L6 displayed moderate uptake and fast clearance: 38.73 ± 1.26%ID/g at 2 h, 13.72 ± 3.55%ID/g at 24 h, 9.62 ± 0.96%ID/g at 48 h, and 2.22 ± 0.37%ID/g at 192 h. The tumor uptake values of 177Lu-L1 at 24 h were 12.86 ± 0.98–14.47 ± 1.32%ID/g. The tumor uptake and clearance of 177Lu-L1 were in the same range as 177Lu-Alb-L6 (30.81 ± 2.86%ID/g, 15.67 ± 6.25%ID/g, 11.94 ± 3.83%ID/g, 9.33 ± 3.18%ID/g, and 4.21 ± 1.45%ID/g at 192 h). The uptake in the PSMA− PC3 flu tumors was low for the agents. The highest uptake in the PSMA− PC3 flu tumor was associated with 177Lu-Alb-L5, 3.87 ± 0.70%ID/g at 2 h, 4.55 ± 0.76%ID/g at 24 h, 3.95 ± 0.50%ID/g at 48 h, and 1.87 ± 0.23%ID/g at 192 h post-injection, respectively. [00211] The blood uptake levels were high at 2 h for 177Lu-Alb-L2, with 19.59 ± 6.06%ID/g; 177Lu- Alb-L3, with 10.29 ± 0.41%ID/g; 177Lu-Alb-L4, with 15.48 ± 7.12%ID/g; 177Lu-Alb-L5, with 22.36 ± 4.72%ID/g; and 177Lu-Alb-L6, with 2.97 ± 0.45%ID/g, respectively. 177Lu-Alb-L6 displayed the fastest clearance from the circulation owing to the long PEG linker, and the uptake was <0.03 ± 0.01%ID/g at 24 h. The blood uptake values of 177Lu-L1 (0.18 ± 0.16 at 3 h, 0.01 ± 0.01 at 24 h, 0.03 ± 0.03 at 48 h, 0.02 ±0.02 at 72 h, and 0.00 ±0.00 at 192 h), however, were significantly lower than those of 177Lu-Alb-L6 and the new albumin-binding agents at all time points. 177Lu-Alb-L5 displayed the slowest clearance from the blood at 192 h post-injection, 2.77 ± 0.19%ID/g, followed by 177Lu-Alb-L4 (2.19 ± 0.46%ID/g), 177Lu-Alb-L3 (0.24 ± 0.12%ID/g), and 177Lu-Alb-L2 (≤0.01%ID/g). [00212] Kidney uptake was relatively high for 177Lu-Alb-L5 (33.45 ± 6.41%ID/g at 2 h, 70.23 ± 16.64 at 24%ID/g, 48.17 ± 15.62%ID/g at 48 h, and 5.06 ± 0.99%ID/g at 192 h) and 177Lu-PSMA- Alb-L4 (64.44 ± 8.75%ID/g at 2 h, 48.81 ± 19.86%ID/g at 24 h, 23.46 ± 7.75%ID/g at 48 h, and 1.34 ± 0.82 at 192 h). 177Lu-Alb-L3 displayed the highest kidney uptake at 2 h, 73.28 ± 22.85%ID/g, and the fastest clearance, 2.77 ± 1.56%ID/g at 24 h. 177Lu-Alb-L2 and 177Lu-Alb-L6 displayed low kidney uptake of ~17.86%ID/g at 2 h. 177Lu-Alb-L6 displayed fast renal clearance resulting in <1%ID/g after 24 h; in contrast, 177Lu-Alb-L2 showed a slow and steady wash-out from the kidneys from ~7.44 ± 2.64% IA/g at 24 h post-injection to 3.74 ± 1.05%ID/g at 48 h post- injection. The kidney uptake and clearance values of 177Lu-L1 were in the range of 177Lu-Alb-L6 36 42332.601_P15594-02
(5.16 ± 2.38%ID/g at 3 h, 0.28 ± 0.18%ID/g at 24 h, 0.22 ± 0.10%ID/g at 72 h, and 0.01 ± 0.00%ID/g at 192 h). [00213] The activity levels in the salivary and lacrimal glands of the agents were comparable to the blood levels and were relatively high for 177Lu-Alb-L5 (salivary glands, 5.83 ± 1.06%ID/g at 2 h, 4.35 ± 0.66%ID/g at 24 h, 3.20 ± 0.24 h %ID/g at 48 h, and 1.18 ± 0.09%ID/g at 192 h) and 177Lu- Alb-L4 (salivary glands, 5.40 ± 0.33%ID/g at 2 h, 0.86 ± 0.21%ID/g at 24 h, 0.62 ± 0.07%D/g at 48 h, and 0.18 ± 0.06%ID/g at 192 h). Similarly, the uptake of the other normal tissues, including the lung and liver, were high for 177Lu-Alb-L5 and decreased continuously over time. For the agents 177Lu-Alb-L2, 177Lu-Alb-L3, 177Lu-Alb-L4, and 177Lu-Alb-L6, normal tissue clearance was below the blood levels and mostly ≤2%ID/g after 24 h. [00214] The selected tumor-to-normal tissue ratios of the agents are shown in FIG. 5B. A high blood pool was observed for all of the agents at 2 h after injection, resulting in a low tumor-to- blood ratio (1.0 ± 0.2), and it increased over time. The tumor-to-blood ratios were approximately 4–8 during 24 h and 48 h post-injection for 177Lu-Alb-L2 and approximately 200–220 after 192 h injection. In contrast, 177Lu-Alb-L3 displayed fast blood clearance, approximately 200–250 at 24– 48 h, and remained in that range until 192 h. For 177Lu-Alb-L4, the ratios were in the range of approximately 50 during 24–48 h and ~100–150 at 192 h. The slowest clearance was observed for 177Lu-Alb-L5; the tumor-to-blood ratios were approximately 10–20 during 24–48 h and approximately 30 during 192 h. The tumor-to-blood values were greater than 300 for 177Lu-L1 at all time points. [00215] The tumor-to-kidney ratio was less than one for the series 177Lu-Alb-L2 to 177Lu-Alb-L5 at 2 h and remained at ≤10 during 24–48 h and increased significantly, approximately 20–50 at 192 h after injection. The lowest tumor-to-kidney ratio was associated with 177Lu-Alb-L5. In contrast, the tumor-to-kidney values were greater than 4 for 177Lu-L1 at 24 h and remained greater than 50 at all-time points. A similar time course was observed for the tumor-to-liver, tumor-to- salivary, and tumor-to-lacrimal glands, as well as the tumor-to-bone ratios. [00216] The area under the curve (AUC0-192h) values and AUC ratios of tumor-to-blood and tumor- to-kidney over 192 h of the new agents and 177Lu-L1 are listed in FIG.6A, FIG.6B. Tumor AUC0- 192h of 177Lu-Alb-L5 was significantly higher than 177Lu-Alb-L4 (p < 0.002). In addition, tumor AUC of 177Lu-Alb-L5 and Alb-L4 were approximately 4-fold higher compared to 177Lu-Alb-L2 and 177Lu-Alb-L3 and approximately 6-fold higher than 177Lu-Alb-L6 and 177Lu-L1. The tumor- 37 42332.601_P15594-02
to-blood AUC0-192h ratio of 177Lu-Alb-L4 was approximately 2-fold higher than 177Lu-Alb-L5, while the tumor-to-kidney AUC0-192h ratios were in the same range. 177Lu-Alb-L6 and 177Lu-L1 displayed nearly similar tumor AUC0-192h and kidney AUC0-192h, and consequently both agents had a similar tumor-to-kidney AUC0-192h (approximately 8). The tumor-to-blood AUC0-192h ratios of 177Lu-L1, however, were significantly higher (approximately 867) than the tumor-to-blood AUC0- 192h ratios of 177Lu-Alb-L6 (approximately 48). The tumor-to-kidney AUC0-192h ratios of 177Lu- Alb-L6 were approximately 2-fold higher than the other albumin-binding agents from the series. The tumor-to-blood AUC0-192h of 177Lu-Alb-L6 was comparable to 177Lu-Alb-L4; it was approximately 1.14-fold higher. [00217] Discussion [00218] In this Example, we investigated a new series of albumin-binding 177Lu-labeled agents based on the structure of 177Lu-L1, our previously reported lead agent. Banerjee et al., 2019. As reported by others and us, small-molecule organic moieties with low and reversible binding to serum albumin (Mol wt.67 kDa) were utilized as a possibility to extend the circulating time of the PSMA-based agents, providing prolonged exposure to the tumors. Such modification significantly increased tumor uptake for the agents, relative to our original compound, 177Lu-L1, through an increased blood half-life. These agents were designed and derived from our previously linker- based targeting platform. Banerjee et al., 2008; Banerjee et al., 2010. Only two structural features were investigated, linker length and the attachment of two albumin-binding moieties, as shown in FIG. 2. Nevertheless, several structure–activity relationships were derived from this small series of compounds, as described below. [00219] We observed significantly higher (p < 0.05) binding affinity and PSMA+ cell binding both in vitro and in vivo for new agents, specifically 177Lu-Alb-L4 and 177Lu-Alb-L5 (Structure B), compared to 177Lu-Alb-L2 and 177Lu-Alb-L3 (Structure A). 177Lu-Alb-L6 displayed low binding affinity, likely due to the long PEG linker, as we noted earlier with a similar construct. Chen et al., 2022. Proof-of-concept cell efficacy data further confirmed the effect of enhanced cellular uptake and internalization of 177Lu-Alb-L5 (Structure B) compared to 177Lu-L1 or 177Lu-Alb-L2 (Structure A). Enhanced cellular internalization is critical to radiation-induced DNA damage and PSMA- expressing cancer cell death. [00220] The tumor uptake of 177Lu-Alb-L2 and 177Lu-Alb-L3 was significantly lower than our previously reported long-linker-based albumin-binding agent, 177Lu-L14 (FIG. 2) (tumor AUC0- 38 42332.601_P15594-02
192h of 5740 ± 520%ID/g.h), although ≥1.5-fold higher than 177Lu-L1 (tumor AUC0-192h of 1734 ± 130%ID/g.h) (FIG. 6A, FIG. 6B). Banerjee et al., 2019. The long-linker-based agents, 177Lu-Alb- L4 (tumor AUC0-192h of 12,857 ± 1469%ID/g.h) and 177Lu-Alb-L5 (tumor AUC0-192h of 18,842 ± 1693%ID/g.h) demonstrated a greater than 2-fold improvement in tumor uptake compared to 177Lu-L14. The blood uptake values of 177Lu-Alb-L4 (blood AUC0-192h of 307 ± 80) were in the range of 177Lu-L14 (blood AUC0-192h of 314 ± 37), indicating a 2-fold improvement in the tumor- to-blood ratios of 177Lu-Alb-L4 relative to 177Lu-L14. Notably, the kidney uptake of 177Lu-Alb-L4 (AUC0-192h of 3895 ± 631) was 1.5-fold higher than 177Lu-L14 (AUC0-192h of 2550 ± 347). In contrast, the tumor and PSMA-expressing typical healthy tissue uptake values were significantly lower for 177Lu-Alb-L6, most likely owing to low PSMA-binding and albumin-binding properties. Notably, the tumor and kidney AUCs of 177Lu-Alb-L6 were in the range of 177Lu-L1; however, the blood AUC was approximately 30-fold higher for 177Lu-Alb-L6 than177Lu-L1. The in vivo performance of 177Lu-Alb-L6 was consistent with the earlier reports, notably, a fast kidney clearance of long PEG-linker-based 177Lu-Alb analogs, published in Kelly et al., 2018, and Kelly et al., 2019. [00221] By comparing the efficacy (in terms of therapeutic index) of the different agents to the absorbed dose thresholds of radiotoxicity for kidneys (approximately 28 Gy) and blood (approximately 2 Gy), the results for the maximum absorbed dose to the tumors of the agents were determined. FIG.6C shows these results. The maximum tumor absorbed dose was estimated to be the lowest of the two values, obtained from the kidney maximum absorbed dose and the blood maximum absorbed dose. For all of the compounds, the dose-limiting organ was found to be the blood.177Lu-Alb-L6, and to a lesser extent, 177Lu-Alb-L4, appear to be the more viable compounds with potential tumoricidal absorbed doses able to be delivered to the tumor. [00222] Renal radiotoxicity has not proved to be a significant issue for 177Lu-PSMA-targeted therapy, possibly due to optimal low linear energy transfer radiations of 177Lu (βmax 0.5 MeV, 1.7 mm). Furthermore, a minimal expression of PSMA has been found in human kidneys. It has been reported that murine PSMA, with 91% similarity to the human PSMA sequence, is overexpressed in the proximal microtubules of the murine renal cortex. Bacich et al., 2001. [00223] The high kidney uptake observed in mice upon the administration of urea-based PSMA therapeutic agents is likely due to the binding to the PSMA mouse isoform. However, most 39 42332.601_P15594-02
preclinical development of PSMA-based albumin-binding radiotherapeutics has been focused on reducing tumor-to-kidney AUC values. Kelly et al., 2019; Kuo et al., 2021; Kramer et al., 2021. [00224] Furthermore, recent in vitro studies revealed that these albumin-binding compounds are associated with significantly higher binding to human blood plasma than mouse plasma. Benešová et al., 2018; Borgna et al., 2020; Busslinger et al., 2022. Accordingly, tumor-to-blood ratios appeared to be critical in optimizing the albumin-binding agents in patient studies, as reported by Kramer et al., 2021. Their analysis from clinical studies revealed that the kidney-absorbed dose of 177Lu-PSMA-617 was approximately 4-fold lower than the albumin-based agents, 177Lu-EB- PSMA-617 and 177Lu-PSMA-ALB-56. In addition, the tumor-to-red-marrow values of 177Lu-EB- PSMA-617 and 177Lu-PSMA-ALB-56 were >10-fold and >5-fold higher than 177Lu-PSMA-617, respectively. Kramer et al., 2021. As a result, the tumor dose at maximum injectable activity was 177Lu-EB-PSMA-617 (60.1 Gy), and 177Lu-PSMA-ALB-56 (96 Gy) was significantly lower than 177Lu-PSMA-671 (131 Gy). Although speculative, projecting mouse data to human data, the estimated tumor radiation doses of 177Lu-Alb-L4 (84 Gy) and 177Lu-Alb-L6 (94 Gy), respectively, were in the range of the reported agents, considering red marrow as the dose-limiting organ. Following similar dosimetry logistics, the tumor radiation dose of 177Lu-L1 (224 Gy) is estimated to be significantly higher than the developed albumin-based agents, considering kidney as the dose-limiting organ. [00225] Furthermore, we compared these AUC values with our previously reported PSMA-based antibody 111In-DOTA-5D3, which was carried out using the same tumor models and same time points. Banerjee et al., 2019. The estimated values are tumor AUC0-192h is 4484 ± 790%ID/g.h, blood AUC0-192h is 2015 ± 251%ID/g.h, and kidney AUC0-192h is 880 ± 38%ID/g.h. The data revealed that the tumor uptake values of the low-molecular-weight albumin-binding agents, such as 177Lu-Alb-L4, are significantly (approximately 3-fold) higher than those of the large antibody- based agent, as anticipated. In comparison, the blood AUC of 177Lu-Alb-L4 is significantly lower (greater than 6-fold) than 111In-DOTA-5D3, albeit with increased kidney uptake (greater than 4- fold). The data suggest that an optimized albumin-binding agent might be a superior option for PSMA-based radiopharmaceutical therapy compared to antibody-based agents. [00226] Although many reported preclinical studies using albumin-binding PSMA-based 177Lu- labeled therapeutics were conducted using the PSMA+ PC3 PIP tumor model, these studies mainly used athymic nude mice for tumor implantation. In contrast, our studies were performed using 40 42332.601_P15594-02
NSG mice because of our institutional availability of this strain. The other notable variables could be related to the high specific activity of the agents used in our studies obtained through HPLC purification and the relatively large tumor used for the biodistribution studies (Table 9). We anticipate that blood and normal tissue uptake data could be a rational indicator to compare the performance of the radiotherapeutics, as revealed in a recent report by Tschan et al., 2021. IBU- based 177Lu-Alb-L4 displayed a low binding affinity to blood. These data suggest that an optimized PEG linker, in combination with an IBU-based moiety, may improve the pharmacokinetics of this class of agents. [00227] There are a few limitations, however, to this study. We used a transfected cell line (PSMA+ PC3 PIP) which may have displayed an unrealistically high PSMA expression and may not have reflected the natural abundance and heterogeneity of PSMA in human cancer. The PSMA+ PC3 PIP and PSMA− PC3 flu cells, however, have the advantage of being isogenic and androgen- independent cell lines and are anticipated to display similar biological factors for evaluating tumor pharmacokinetics, except for the PSMA expression levels. Many reported studies currently use the same tumor models for developing similar structure–activity relationship data because of the fast and predictable growth rate. There might be some variability associated with tumor sizes, although the data related to the pharmacokinetic performance are expected to be similar. [00228] Materials and Methods [00229] General Methods. All reagents and solvents were purchased from Sigma-Aldrich or Fisher Scientific unless specified and are listed in Table 1. These reagents and solvents were directly used without further purification. Amino acid derivatives were received from Chem-Impex International. 1,4,7,10-Tetraazacyclododecane-1,4,7,10-tetraacetic acid mono-N- hydroxysuccinimide ester (DOTA-NHS-ester) was purchased from Macrocyclics Inc, Dallas. (2S)-2-[[(1S)-1-carboxy-3-methylbutyl]carbamoylamino]pentanedioic acid (ZJ43) was synthesized in-house following that reported in Olszewski et al., 2004. All new compounds were synthesized using standard solution-phase chemistry based on our well-established methods. Banerjee et al., 2019. Table 1. List of chemicals and solvents.
41 42332.601_P15594-02
Chem-Impex L-Glutamic acid di-tert-butyl ester hydrochloride N-ε-Z-L-lysine tert-butyl ester hydrochloride te
o umn c romatograp y o nterme ates was per orme us ng otage so era ash Chromatography with SNAP Ultra C18 Sep-Pak columns. Purification of final compounds was performed using an Agilent (Santa Clara, CA, USA) high-performance liquid chromatography (HPLC) system equipped with a model 1200 quaternary pump and a model 1200 UV absorbance detector using a 250 mm × 10 mm Phenomenex Luna C18 column. Spectral characterization data of the new agents are provided herein below and in FIGS 7-12. 177LuCl3 was supplied by the US Department of Energy Isotope Program. All 177Lu-labeled radioligands were purified via HPLC to remove unreacted ligand from the radiolabeled material to ensure high specific radioactivity. 42 42332.601_P15594-02
Animal studies were undertaken according to the guidelines set forth by Johns Hopkins Animal Care and Use Committee. [00231] Radiochemistry [00232] Radiolabeling was performed under standard labeling conditions in ammonium acetate buffer (0.2 M) at pH approximately 4.5 following our reported method. Banerjee et al., 2019. The radiolabeling was performed in a radiochemistry microwave chamber at 90 °C for 5 min at 40 watts (Resonance Instruments Inc., Skokie, IL, USA), and the reaction solution was purified using reverse-phase HPLC. An isocratic HPLC method was developed in each case, as listed in Table 2, to remove the unreacted ligand from the radiolabeled material to ensure high specific activity. L- Ascorbic acid was added to the isolated radiolabeled compounds in the final formulation to maintain stability and was used for in vitro and in vivo experiments. All 177Lu-labeled compounds were stable for up to 4 h at room temperature without significant radiolysis and were stable at 4 °C for 7 data concentration of 37 MBq/mL. Table 2. HPLC Methods for 177Lu-Alb-L1-177Lu-Alb-L6. N)
[00233] Measurement of Partition Coefficients [00234] The partition coefficient of the 177Lu-labeled agents was determined in 1-octanol and phosphate-buffered saline (PBS) (pH 7.4).1-Octanol (3 mL) and PBS (3 mL) were pipetted into a 15 mL test tube containing 370 kBq of the test compound. The test tube was vortexed for 2 min and then centrifuged (4000× g, 5 min). Aliquots (0.1 mL) from the 1-octanol and PBS phases were transferred into two test tubes for counting. The amount of radioactivity in each test tube was 43 42332.601_P15594-02
measured using the automated γ-counter. The partition coefficient was calculated using the following equation: log Pow = log[counts1-octanol/countsPBS], and the data are listed in Table 3. Table 3. Partition coefficient (POctanol/water) and albumin binding properties of 177Lu-Alb-L2-177Lu-Alb-L6 [00235] In Vit
[00236] Competitive Inhibition Assays [00237] NAALADase Assay. Binding affinities of all new ligands were measured according to a previously described competitive fluorescence-based assay. Banerjee et al., 2011. In brief, cell lysates of LNCaP cell extracts were incubated with PSMA-targeted agents (0.01 nm–100 μM) in the presence of 4 μM NAAG at 37 °C for 2 h and the reference PSMA inhibitor, ZJ43 (0.01 nm– 100 μM). Olszewski et al., 2004. The amount of released glutamate from NAAG was measured by incubating it with a working solution of the Amplex Red glutamic acid kit (Molecular Probes Inc., Eugene, OR, USA) at 37 °C for 60 min. Fluorescence was measured with excitation at 535 nm and emission at 590 nm using a microplate reader. Inhibition curves were determined using semi-log plots. Data were analyzed using a one-site total binding regression using GraphPad Prism version 9 for Windows (GraphPad Software, San Diego, CA, USA). The IC50 values were determined as the concentration at which enzymatic activity was inhibited by 50%. Assays were performed in triplicate, with the entire inhibition study repeated at least once. Enzyme inhibitory 44 42332.601_P15594-02
constants (Ki values) were generated using the Cheng-Prusoff conversion. Cheng and Prusoff, 1973. [00238] Protein-Binding Assay [00239] Albumin-binding properties of the compounds were analyzed following a reported method. Tsuchihashi et al., 2023. A solution of 177Lu-Alb-L2–177LuAlb-L5 (74 kBq in 10 μL PBS) was added to 190 μL human serum albumin solution (45 mg HSA in 1 mL of PBS). After mixing, the solution was incubated at 37 °C for 60 min. Then, a 100 μL of the reaction solution was loaded onto a gel filtration column (Thermo Scientific™ Zeba™ Spin 7K MWCO size exclusion spin columns, Waltham, MA, USA), previously equilibrated with 0.1 M acetate buffer (pH 6.0), followed by centrifugation (1500× g, 2 min). The radioactivity of the column and eluate was then measured using an automated γ-counter. The data are listed in Table 3. [00240] Cell Culture [00241] Androgen-independent PSMA-high (PSMA+) PC3 PIP cells and PSMA-low (PSMA−) PC3 flu cell lines are an isogenic subline pair of human PC3 cell lines (androgen-independent PSMA-negative bone metastatic prostate carcinoma). These cell lines were generously provided by Warren Heston (Cleveland Clinic). According to the literature reports, PC3 PIP cells were initially generated via the transfection of PC3 cells, employing VSV-G pseudo-typed lentiviral- vector-expressing human PSMA. Leek et al., 1995; Chang et al., 1999; Liu et al., 2009. [00242] As reported previously, flow cytometry and Western blot assays are routinely used to evaluate PSMA expressions of PSMA+ PC3 PIP cells and PSMA− PC3 flu cells. Banerjee et al., 2019. Selected data generated for the studies of this report are provided in FIG. 13. These cells were cultured in RPMI-1640 cell culture medium supplemented with 10% fetal calf serum, L- glutamine, antibiotics, and puromycin (2 µg/mL) to maintain expression, and were used for in vitro cell uptake studies and in vivo tumor generation. All cell cultures were maintained at 5% carbon dioxide at 37 °C in a humidified incubator. Authentication of the cell lines were performed routine by JHU GRCF (grcf.jhmi.edu/biorepository-cell-center/bioprocessing/cell-line-authentication). We tested mycoplasma contaminations of the cell line cultures every two weeks using the MycoAlert PLUS mycoplasma detection kit (Lonza). [00243] Cell Uptake and Internalization Study Cell uptake studies were performed following our previously reported protocol. Banerjee et al., 2019. In brief, adherent PSMA+ PC3 PIP cells and PSMA− PC3 flu cells detached using 45 42332.601_P15594-02
nonenzymatic buffer (Gibco) and approximately 1 million cells per tube were incubated in 100 µL of each radiolabeled agent (370 kBq/mL) for 2 h and 24 h at 37 °C in the 100 µL growth medium (binding buffer (1× PBS + 2mM EDTA + 0.5% FBS)). After incubation, the medium was removed at the indicated time points, and the cells were washed three times with ice-cold PBS. The collected pooled washes and the cell pellets were counted using an automated γ- counter. The radioactivity values were converted into a percentage of incubated dose (%ID) per million cells. Experiments were performed in triplicate and repeated two times. [00244] For the PSMA blocking studies, PSMA+ PC3 PIP cells were pre-incubated with ZJ43 (10 µM final concentration) for 30 min and then washed 3 times with binding buffer followed by incubation of the radioactive dose (100 µL of 370 kBq/mL in binding buffer) for 2 h. The cell uptake studies were then conducted using the method mentioned in the previous section. [00245] For the internalization assays, cells were detached using nonenzymatic buffer, and aliquots of 1 million cells per tube were incubated with 370 kBq/mL of each radiolabeled agent for 2 h and 24 h at 37 °C along with the 100 µL of the binding buffer, as mentioned in the cell uptake study. At the indicated time points, the medium was removed, and cells were washed with binding buffer followed by a mildly acidic buffer (50 mM glycine, 150 mM NaCl (pH 3.0)) at 4 °C for 5 min. The acidic buffer was then collected, and cells were washed twice with binding buffer. The collected pooled washes (containing cell-surface-bound 177Lu-Alb-L1–177Lu-Alb-L6) and cell pellets (containing internalized 177Lu-Alb-L1–177Lu-Alb-L6) were counted in an automated γ- spectrometer along with the standards. All radioactivity values were converted into a percentage of incubated dose (%ID) per million cells. Experiments were performed in triplicate and repeated 2 times. Data were fitted according to linear regression analysis. [00246] Clonogenic Survival Assay Cells (200–1000) were seeded in 60-mm culture dishes. Each radioligand (177Lu-L1, 177Lu-Alb- L2, 177Lu-Alb-L5) was diluted in a prewarmed medium at different concentrations (0, 0.37, 1.85, 3.7, 18.5, 37, 185, and 370 kBq/mL) and incubated with the cells for 48 h, as we previously reported. Banerjee et al., 2019. The radiolabeled compound was replaced with fresh medium, and cells were incubated for 2 weeks or until colonies had at least 50 cells. The colonies were stained with crystal violet and counted, and the surviving fraction was normalized to the control plating efficiency, as previously described. Franken et al., 2006. [00247] In Vivo Experiments 46 42332.601_P15594-02
[00248] Biodistribution [00249] Five-to-six-week-old male NSG mice were purchased from Johns Hopkins University research animal resources. Briefly, approximately 14–20 days after subcutaneous inoculation of PSMA+ PC3 PIP (3 × 106 cells) or PSMA− flu cells (1 × 106 cells) in 100 µL HBSS solution on the upper flanks, tissue biodistribution studies were performed. Male NSG mice bearing PSMA+ PC3 PIP and PSMA− PC3 flu xenografts were injected intravenously with the respective 177Lu- labeled agent, 177Lu-Alb-L2–177Lu-Alb-L6 (1.85 MBq) diluted in 150 µL saline. Mice were sacrificed at 2 h, 24 h, 48 h, and 192 h post-injection, and selected tissues were harvested, weighed, and measured radioactivity using an automated γ-counter. A group of 4 mice was used for each time point; the results were listed as the percentage of the injected dose per gram of tissue mass (%ID/g). The data are presented as the average ± standard deviation (SD). The biodistribution study of 177Lu-L1 was performed at only 24 h during the biodistribution study of 177Lu-Alb-L2 and 177Lu-Alb-L5 in a single experiment. The biodistributions of 177Lu-Alb-L3, 177Lu-Alb-L4, and 177Lu-L1 (24 h) were acquired in a separate experiment. The data are listed in Tables 4-10. Biodistribution data of 177Lu-L1 at 3 h, 24 h, 48 h, and 72 h were obtained from our previous report. Banerjee et al., 2019. The 192 h post-injection data of 177Lu-L1 are unpublished and were acquired during the same study. Table 4. Tissue biodistribution data of 177Lu-Alb-L2 in male NSG mice
42332.601_P15594-02
Fat 1.71 ± 0.40 0.64 ± 0.31 0.44 ± 0.23 0.07 17.86 ± 0.05 ±
Table 5. Tissue biodistribution data of 177Lu-Alb-L3 in male NSG mice bearing d 0 3 1 0 0 1 1 8
48 42332.601_P15594-02
Table 6. Tissue biodistribution data of 177Lu-Alb-L4 in male NSG mice bearing PSMA+ PC3 PIP and PSMA- PC3 flu xenografts in either flank (Data presented in 5
Table 7. Tissue biodistribution data of 177Lu-Alb-L5 in male NSG mice bearing in 9 2 8 1 7 5 5 1 9 3 3
42332.601_P15594-02
Salivary 5.83 ± 1.06 4.35 ± 0.66 3.20 ± 0.24 1.18 ± 0.09 Lacrimal glands 7.33 ± 1.03 5.97 ± 0.51 4.44 ± 0.60 1.27 ± 0.14 1 9 4 3
50 42332.601_P15594-02
Table 8. Tissue biodistribution data of 177Lu-Alb-L6 in male NSG mice bearing PSMA+ PC3 PIP xenografts (Data presented in %ID/g, expressed as 1
Table 9. PSMA+ PC PIP tumor weight (g) and expressed as mean±SD) and tumor volume
42332.601_P15594-02
Tumor NA NA NA NA size
52 42332.601_P15594-02
Table 10. Tissue biodistribution data of 177Lu-Alb-L1 in male NSG mice bearing PSMA+ PC3 PIP and PSMA- PC3 flu xenografts in either flank (Data presented in % ID/g, expressed as mean - e
[00250] Statistical Analysis [00251] All graphs, AUC calculations, and statistical analyses were created and performed using the GraphPad Prism software (version 9.0). Significant differences were evaluated using a one- way ANOVA or unpaired t-test; p-values < 0.05 were considered to be significant. p-values lower than 0.05 (p < 0.05), p < 0.01, p < 0.001, and p < 0.0001 were referred to with one (*), two (**), three (***), or four (****) asterisks, respectively. [00252] Summary [00253] The data suggest that 177Lu-Alb-L4 could be an improved option for 177Lu-L1, and further modification in the linker construct using a PEG linker may be possible to reduce blood and kidney uptake. [00254] Synthesis and characterization of new ligands 53 42332.601_P15594-02
[00255] New agents were prepared following several solution phase reaction schemes described below.
Scheme 1. a) (I) 2,5-dioxopyrrolidin-1-yl 5-((tert-butoxycarbonyl)amino)pentanoate, DIPEA, DMF, RT, overnight. (ii) TFA:CH2Cl2 (1:1), RT, 2 h; b) (i) Fmoc-L-Lys(Boc)-OSu, DIPEA, DMF, RT, 2 h (ii) 20% piperidine, DMF, RT, 1 h; c) (i) DOTA-NHS-ester, DIPEA, DMSO, RT, 2 h. (ii) TFA:CH2Cl2 (1:1), RT, 2 h; d) IBU-NHS or IPBA-NHS, DIPEA, DMSO, RT, 2 h. [00256] Scheme 1. Protected Glu-Lys-urea (KEU) 1 or 2 was prepared from commercially available starting materials and following our previously reported methods. Banerjee et al., 2019. Initially, KEU was treated with 2,5-dioxopyrrolidin-1-yl 5-((tert- butoxycarbonyl)amino)pentanoate in the presence of DIPEA, followed by deprotection using trifluoroacetic acid (TFA) and methylene dichloride (DCM), which provides the corresponding intermediate compound 3 or 4. Then adding Fmoc-L-Lys(Boc)-OSu to the intermediate compound 3 or 4; removing Boc using TFA provided the intermediate compound 5 or 6. Next, chelating agent DOTA-NHS-ester was added to the intermediate 5 or 6 to create compound 7 or 8. Subsequently, the Boc group of lysine was removed from compounds 7 or 8. The crude product was conjugated with ibuprofen (IBU) or 4-(p-iodophenyl) butyric acid (IPBA) to create Alb-L1, Alb-L1, and Alb- L3. 54 42332.601_P15594-02
Scheme 2. a) DSS, DIPEA, DMF, RT, overnight; b) (i) DOTA-L-Lys(Dde)-L-Lys-NH 2, DIPEA, DMSO, RT, 2 h, 65% (ii) TFA:CH2Cl2 (1:1), RT, 2 h, 70%; c). 2% hydrazine-hydrate in DMF, RT, 30 min, 66%; d) IBU-NHS or IPBA-NHS, DIPEA, DMSO, RT, 2 h, 88%. [00257] In Scheme 2, the protected Glu-Lys-urea (KEU), 2, was treated with disuccinimidyl suberate (DSS) following our reported method to create the intermediate compound 9 in good yield. Banerjee et al., 2019. Then, compound 9 was treated with DOTA-L-Lys(Dde)-L-Lys-NH2 in the presence of DIPEA, followed by deprotection, which produced the intermediate 10 in good yield (65%). Subsequently, compound 10 was converted into the corresponding amine derivatives 11 with 2% hydrazine hydrate for 2 min. Next, the resultant crude compound was treated with IBU-NHS or IPBA -But-NHS, which delivered 177Lu-Alb-L4 or 177Lu-Alb-L5 in good yield.
02
Scheme 3. a) (i) 2,5-dioxopyrrolidin-1-yl 6-((tert-butoxycarbonyl)amino)hexanoate, DIPEA, DMF, RT, overnight. (ii) TFA:CH2Cl2 (1:1), RT, 2 h; b) (i) t-Boc-N-amido-PEG10-NHS ester, DMSO, TEA, RT, 2 h. (ii) TFA:CH2Cl2 (1:1), RT, 2 h; c) (i) Fmoc-L-Lys(Boc)-OSu, TEA, DMSO, RT, 2 h. (ii) 20% piperidine, DMF, RT, 1 h. iii) DOTA-NHS-ester, DIPEA, DMSO, RT, 2 h. (iv) TFA:CH2Cl2 (1:1), RT, 2 h; d) IPBA-NHS, DIPEA, DMSO, RT, 2 h. [00258] In Scheme 3, for creating 177Lu-Alb-L6, compound 2 was reacted with 2,5- dioxopyrrolidin-1-yl 6-((tert-butoxycarbonyl)amino)hexanoate to generate the corresponding intermediate 12, which was then treated with t-Boc-N-amido-PEG10-NHS ester, to deliver compound 13. Next, Fmoc-L-Lys(Boc)-OSu was added to compound 13, followed by deprotection and addition of DOTA-NHS-ester generated compound 14. Finally, 14 was conjugated with IPBA- NHS to provide 177Lu-Alb-L6 in greater than 50% yield. [00259] ((S)-5-(5-Aminopentanamido)-1-carboxypentyl)carbamoyl)-L-glutamic acid (3): To a stirred solution of 1 (200 mg, 0.410 mmol, 1.0 eq) and 2,5-dioxopyrrolidin-1-yl 5-((tert- butoxycarbonyl)amino)pentanoate (129 mg, 0.410 mmol, 1.0 eq) in DMF (2 mL) was added diisopropyl ethyl amine (DIPEA) (214 µL, 1.23 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred overnight and concentrated to isolate the crude product. To the above crude was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature, and the mixture was stirred for 2 h. The reaction mixture was then concentrated and purified using acetonitrile (ACN)/water (H2O) on the C18 Sep-Pak column to provide compound 3 (146 mg, 85%) as a white solid. 1H NMR (500 MHz, D2O) δ 4.25 (dd, J = 5.5, 9.5 Hz, 1H), 4.17 (dd, J = 5.0, 9.0 Hz, 1H), 3.17 (t, J = 6.5 Hz, 2H), 3.02- 2.92 (m, 2H), 2.50 (t, J = 7.0 Hz, 2H), 2.30-2.22 (m, 2H), 2.21-2.11 (m, 1H), 2.01-1.90 (m, 1H), 1.88-1.76 (m, 1H), 1.74-1.58 (m, 5H), 1.55-1.45 (m, 2H), 1.44-1.30 (m, 2H). [00260] (10S,23S,27S)-10-Amino-2,2-dimethyl-4,11,17,25-tetraoxo-3-oxa-5,12,18,24,26- pentaaza nonacosane-23,27,29-tricarboxylic acid (5): To a stirred solution of 3 (60 mg, 0.143 mmol, 1.0 eq) and Fmoc-L-Lys(Boc)-OSu (81 mg, 0.143 mmol, 1.0 eq) in DMF (1 mL) was added DIPEA (124 µL, 0.717 mmol, 5.0 eq)) at room temperature. The reaction mixture was stirred for 2 h and concentrated to get crude product. To the above crude was added 2 mL of 20% piperidine in DMF (2 mL) at room temperature, and the mixture was stirred for 1 h. Concentrated and purified using ACN/H2O on the C18 Sep-Pak column to provide compound 5 (65 mg, 70%) as a white solid. HRMS (ESI) m/z: [M + H]+ calcd for C28H51N6O11, 647.3611; found, 647.3610. 56 42332.601_P15594-02
[00261] (3S,7S,20S)-27-(4-Isobutylphenyl)-5,13,19,26-tetraoxo-20-(2-(4,7,10- tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido)-4,6,12,18,25- pentaazaoctacosane-1,3,7-tricarboxylic acid (177Lu-Alb-L1): To a stirred solution of 5 (22.5 mg, 0.034 mmol, 1.0 eq) and DOTA-NHS-ester (29.13 mg, 0.038 mmol, 1.1 eq) in DMSO (500 µL) was added DIPEA (72 µL, 0.417 mmol, 12.0 eq) at room temperature. The reaction mixture was then stirred for 2 h at room temperature and purified using ACN/H2O on the C18 Sep-Pak column to provide the desired compound as a white solid. To the isolated compound (26.8 mg, 0.026 mmol), was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature. The reaction mixture was then stirred for 2 h and concentrated to produce compound 7. Compound 7 was used for the next step without further purification. To the stirred solution of 7 and 2,5-dioxopyrrolidin-1-yl-2-(4-isobutyl phenyl)propanoate (8.09 mg, 0.026 mmol) in DMSO (500 µL) was added DIPEA (46 µL, 0.266 mmol, 12.0 eq) at room temperature. The reaction mixture was stirred for 2 h, and the crude product obtained after solvent evaporation was purified by preparative RP-HPLC chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization afforded compound 177Lu-Alb-L1 (23.4 mg, 60%) as a white solid. [RP-HPLC purification was achieved using Agilent System, λ 220 nm, 250 mm × 10 mm Phenomenex Luna C18 column, solvent gradient: 90% H2O (0.1% TFA) and 10% ACN (0.1% TFA), reaching 60% of ACN in 20 min at a flow rate of 10 mL/min, product eluted at 11.3 min].1H NMR (500 MHz, DMSO-d6) δ 8.64 (s, 1H), 8.03 (s, 1H), 7.93 (s, 1H), 7.79 (s, 1H), 7.18 (d, J = 7.5 Hz, 2H), 7.06 (d, J = 7.5 Hz, 2H), 6.386.25 (m, 2H), 4.22-4.14 (m, 2H), 4.12-4.05 (m, 2H), 3.07 (s, 8H), 3.01-2.86 (m, 6H), 2.50 (s, 16H), 2.38 (d, J = 7.0 Hz, 2H), 2.36-2.16 (m, 4H), 2.03 (t, J = 6.5 Hz, 2H), 1.97-1.86 (m, 1H), 1.82-1.10 (m, 16H), 1.28 (d, J = 7.0 Hz, 3H), 0.84 (d, J = 6.5 Hz, 6H); HRMS (ESI) m/z: [M + H]+ calcd for C52H85N10O17, 1121.6100; found, 1121.6088. [00262] (((S)-5-(5-Amino-N-(4-bromobenzyl)pentanamido)-1-carboxypentyl)carbamoyl)-L- glutamic acid (4): To a stirred solution of 2 (214 mg, 0.326 mmol, 1.0 eq) and 2,5-dioxopyrrolidin- 1-yl 5-((tert-butoxycarbonyl)amino)pentanoate (102.5 mg, 0.326 mmol, 1.0 eq) in DMF (2 mL) was added DIPEA (170 µL, 0.978 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 24 h and concentrated to get crude product. To the above crude was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature, and the reaction mixture was stirred for 2 h. The reaction mixture was then concentrated and purified using ACN/H2O on the C18 Sep-Pak column to provide compound 4 (143 mg, 75%) as a white solid.1H NMR (500 MHz, D2O) δ 7.55 (d, J = 8.5 Hz, 1H), 57 42332.601_P15594-02
7.51 (d, J = 8.5 Hz, 1H), 7.12 (dd, J = 2.5, 8.0 Hz, 2H), 4.59 (s, 1H), 4.51 (s, 1H), 4.294.22 (m, 1H), 4.19-4.11 (m, 1H), 3.39-3.29 (m, 2H), 3.05-2.88 (m, 2H), 2.59-2.38 (m, 4H), 2.222.10 (m, 1H), 2.02-1.90 (m, 1H), 1.82-1.46 (m, 8H), 1.39-1.24 (m, 2H). [00263] (10S,23S,27S)-10-Amino-18-(4-bromobenzyl)-2,2-dimethyl-4,11,17,25-tetraoxo-3- oxa-5,12,18,24,26-pentaazanonacosane-23,27,29-tricarboxylic acid (6): To a stirred solution of 4 (139 mg, 0.236 mmol, 1.0 eq) and Fmoc-L-Lys(Boc)-OSu (133.9 mg, 0.236 mmol, 1.0 eq) in DMF (2 mL) was added DIPEA (250 µL, 1.42 mmol, 6.0 eq) at room temperature. The reaction mixture was stirred for 2 h and concentrated to get the crude product. To the isolated crude was added 2 mL of 20% piperidine in DMF (3 mL) at room temperature, and the mixture was stirred for 1 h. Concentrated and purified using ACN/H2O on the C18 Sep-Pak column to provide compound 6 (125 mg, 65%) as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.43-8.32 (m, 1H), 8.08 (s, 2H), 7.93 (d, J = 7.5 Hz, 1H), 7.74 (d, J = 7.5 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.41 (d, J = 8.0 Hz, 1H), 6.74 (t, J = 5.0 Hz, 1H), 6.40-6.26 (m, 2H), 4.52 (s, 1H), 4.44 (s, 1H), 4.14-3.98 (m, 2H), 3.23-2.97 (m, 5H), 2.93-2.82 (m, 2H), 2.37 (t, J = 7.0 Hz, 1H), 2.31-2.15 (m, 3H), 1.96-1.43 (m, 14H), 1.35 (s, 9H), 1.29-1.15 (m, 4H); [M + H]+ calcd for C35H56BrN6O11, 815.3192; found, 815.3184. [00264] (3S,7S,20S)-12-(4-Bromobenzyl)-27-(4-isobutylphenyl)-5,13,19,26-tetraoxo-20-(2- (4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido)-4,6,12,18,25- pentaazaoctacosane-1,3,7-tricarboxylic acid (177Lu-Alb-L2): To a stirred solution of 6 (33.3 mg, 0.04 mmol, 1.0 eq) and DOTA-NHS-ester (34.2 mg, 0.045 mmol, 1.1 eq) in DMSO (500 µL) was added DIPEA (85 µL, 0.490 mmol, 12.0 eq) at room temperature. The reaction mixture was stirred for 2 h and purified using acetonitrile/H2O on the C18 Sep-Pak column to provide the desired product as a white solid (41 mg, 85% yield). To the crude product (20 mg, 0.016 mmol) was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature; the reaction mixture was stirred for 2 h and concentrated to afford a crude compound 8. To the stirred solution of crude 8 (18.3 mg, 0.0166 mmol, 1.0 eq) and 2,5-dioxopyrrolidin-1-yl 2-(4-isobutylphenyl)propanoate (6.0 mg, 0.0199 mmol, 1.2 eq) in DMSO (500 µL) was added DIPEA (35 µL, 0.199 mmol, 12.0 eq)) at room temperature. The reaction mixture was stirred for 2 h, and the resultant reaction mixture was purified by preparative RP-HPLC chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization afforded 177Lu-Alb-L2 (19.3 mg, 90%) as a white solid. [RP-HPLC purification was performed using Agilent System, λ 220 nm, 250 mm × 10 mm Phenomenex Luna C18 column, solvent gradient: 90% H2O (0.1% TFA) and 10% ACN (0.1% 58 42332.601_P15594-02
TFA), reaching 60% of ACN in 20 min at a flow rate of 10 mL/min, product eluted at 14.6 min]. 1H NMR (500 MHz, DMSO-d6) δ 8.63 (s, 1H), 8.03 (d, J = 8.5 Hz, 1H), 7.91 (s, 1H), 7.54 (d, J = 7.5 Hz, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.23-7.10 (m, 4H), 7.05 (d, J = 7.0 Hz, 2H), 6.39-6.25 (m, 2H), 4.50 (s, 1H), 4.43 (s, 1H), 4.26-4.00 (m, 4H), 3.07 (s, 8H), 2.96-2.73 (m, 6H), 2.50 (s, 16H), 2.37 (d, J = 7.5 Hz, 2H), 2.30-2.15 (m, 4H), 1.96-1.85 (m, 1H), 1.82-1.56 (m, 4H), 1.55-1.11 (m, 14H), 1.27 (d, J = 7.0 Hz, 3H), 0.83 (d, J = 6.0 Hz, 6H); HRMS (ESI) m/z: [M + H]+ calcd for C59H90BrN10O17, 1289.5642; found, 1289.5663. [00265] (3S,7S,20S)-12-(4-bromobenzyl)-29-(4-iodophenyl)-5,13,19,26-tetraoxo-20-(2-(4,7,10- tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido)-4,6,12,18,25- pentaazanonacosane-1,3,7-tricarboxylic acid (177Lu-Alb-L3): To a stirred solution of 6 (35.2 mg, 0.043 mmol, 1.0 eq) and DOTA-NHS-ester (34.5 mg, 0.045 mmol, 1.05 eq) in DMSO (300 µL) was added DIPEA (90 µL, 0.518 mmol, 12.0 eq) at room temperature. The reaction mixture was stirred for 2 h and purified using ACN/H2O on the C18 Sep-Pak column to provide the desired product as a white solid (41 mg, 80% yield). To the above product (29 mg, 0.016 mmol) was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature; the reaction mixture was stirred for 2 h and concentrated to afford a crude amine 8. To the stirred solution of crude 8 (26.5 mg, 0.0240 mmol, 1.0 eq) and 2,5-dioxopyrrolidin-1-yl 4-(4-iodophenyl)butanoate (9.77 mg, 0.0252 mmol, 1.05 eq) in DMSO (300 µL) was added DIPEA (50 µL, 0.288 mmol, 12.0 eq) at room temperature. The reaction mixture was stirred for 2 h, and crude was purified by reverse phase flash chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization afforded compound 177Lu-Alb-L3 (19.8 mg, 60%) as a white solid. [Flash chromatography purification was achieved using Biotage Isolera One system, λ 220 nm, Biotage SNAP Ultra C18 column (12 g), solvent gradient: 90% H2O (0.1% TFA) and 10% ACN (0.1% TFA), reaching 90% of ACN, product eluted at 50% to 55% of (B) in (A) fraction].1H NMR (500 MHz, DMSO-d6) δ 8.89-8.58 (m, 1H), 8.41 (s, 1H), 7.96-7.89 (m, 1H), 7.60 (d, J = 8.0 Hz, 2H), 7.53 (d, J = 8.0 Hz, 1H), 7.48 (d, J = 8.5 Hz, 1H), 7.14 (d, J = 7.0 Hz, 2H), 7.00 (d, J = 8.0 Hz, 2H), 6.71-6.35 (m, 2H), 4.43 (s, 2H), 4.20-4.09 (m, 1H), 4.04-3.95 (m, 2H), 3.51-2.86 (m, 14H), 2.50 (s, 16H), 2.39- 2.31 (m, 2H), 2.27-2.18 (m, 3H), 2.04 (t, J = 7.5 Hz, 2H), 1.80-1.69 (m, 4H), 1.68-1.30 (m, 12H), 1.29-1.13 (m, 5H); HRMS (ESI) m/z: [M + K]+ calcd for C56H82BrIKN10O17, 1411.3688; found, 1411.3719. 59 42332.601_P15594-02
[00266] di-tert-Butyl(((S)-6-(N-(4-bromobenzyl)-8-((2,5-dioxopyrrolidin-1-yl)oxy)-8- oxooctanamido) -1-(tert-butoxy)-1-oxohexan-2-yl)carbamoyl)-L-glutamate (9): To a solution of DSS (308 mg, 0.838 mmol, 2.2 eq) in 6 mL DMF was added a solution of di-tert-butyl (((S)-6- ((4-bromobenzyl)amino)-1-(tert-butoxy)-1-oxohexan-2-yl)carbamoyl dropwise)-L-glutamate 2 (250 mg, 0.381 mmol, 1.0 eq) in 2 mL DMF and TEA (58 µL, 0.419 mmol, 1.1 eq) at rt for 20 min. The reaction mixture was left stirring overnight at room temperature. The solvent was removed under vacuum, and the solid residue was purified using a silica gel column using CH3CN/CH2Cl2 as a solvent system to obtain compound 9 (225 mg, 65%). 1H NMR (500 MHz, CDCl3) δ 7.46 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 7.02 (d, J = 8.0 Hz, 1H), 5.72-5.05 (m, 2H), 4.50 (s, 1H), 4.46 (s, 1H), 4.35-4.20 (m, 2H), 3.39-3.22 (m, 1H), 3.13 (t, J = 7.5 Hz, 1H), 2.80 (s, 4H), 2.58 (dt, J = 7.5, 15.0 Hz, 2H), 2.42-2.19 (m, 4H), 2.10-1.98 (m, 1H), 1.89-1.62 (m, 6H), 1.60-1.48 (m,
, 1.49-1.34 (m, 30H), 1.33-1.18 (m, 4H); HRMS (ESI) m/z: [M + H]+ calcd for C43H66BrN4O12, 909.3846; found, 909.3855. [00267] (8S,11S,30S,34S)-25-(4-Bromobenzyl)-2-(4,4-dimethyl-3,5-dioxocyclohexylidene)- 9,17,24,32-tetraoxo-8-(2-(4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1- yl)acetamido)-3,10,16,25,31,33-hexaazahexatriacontane-11,30,34,36-tetracarboxylic acid (10): To a stirred solution of 9 (32.5 mg, 0.035 mmol, 1.0 eq) and DOTA-L-Lys(Dde)-L-Lys-NH2 (29.5 mg, 0.035 mmol, 1.0 eq) in DMSO (300 µL) was added DIPEA (38 µL, 0.214 mmol, 6.0 eq)) at room temperature. The reaction mixture was stirred overnight and concentrated to obtain the crude product. To the above crude was added 2 mL of TFA/CH2Cl2 (1:1) at room temperature, and the mixture was stirred for 2 h. Concentrated and purified using ACN/H2O on C18 Sep-Pak column to provide compound 10 (36 mg, 70%) as a white solid.1H NMR (500 MHz, DMSO-d6) δ 13.22 (t, J = 5.0 Hz, 1H), 8.70 (s, 1H), 8.35 (d, J = 5.0 Hz, 1H), 7.83-7.73 (m, 1H), 7.55 (d, J = 8.5 Hz, 1H), 7.49 (d, J = 8.5 Hz, 1H), 7.17-7.10 (m, 2H), 6.37-6.26 (m, 2H), 4.51 (s, 1H), 4.43 (s, 1H), 4.41-4.33 (m, 1H), 4.18-4.39 (m, 3H), 3.08 (s, 8H), 3.02-2.92 (m, 4H), 2.50 (s, 16H), 2.46 (s, 4H), 2.37-2.30 (m, 2H), 2.26 (s, 3H), 2.27-2.17 (m, 3H), 2.05-1.85 (m, 3H), 1.79-1.08 (m, 28H), 0.93 (s, 6H); HRMS (ESI) m/z: [M + H]+ calcd for C65H101BrN11O21, 1450.6362; found, 1450.6351. [00268] (4S,7S,26S,30S)-4-(4-Aminobutyl)-21-(4-bromobenzyl)-2,5,13,20,28-pentaoxo-1- (4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)-3,6,12,21,27,29- hexaazadotriacontane-7,26,30,32-tetracarboxylic acid (11): Compound 10 (20 mg, 0.013 mmol) was dissolved 2 mL 2% hydrazine-hydrate in DMF at room temperature followed by 200 60 42332.601_P15594-02
µL water. The solution was left stirring at room temperature for 30 min, then evaporated to dryness. The colorless residue was purified using ACN/H2O on the C18 Sep-Pak column to provide compound 11 (11.6 mg, 66%) as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.61 (s, 1H), 8.28 (s, 1H), 7.81-7.62 (m, 3H), 7.55 (d, J = 8.0 Hz, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.17-7.09 (m, 2H), 6.37-6.25 (m, 2H), 4.51 (s, 1H), 4.43 (s, 1H), 4.36-4.28 (m, 1H), 4.17-4.04 (m, 3H), 3.07 (s, 8H), 2.99-2.92 (m, 2H), 2.80-2.67 (m, 4H), 2.50 (s, 16H), 2.38-2.15 (m, 5H), 2.07-1.85 (m, 4H), 1.75-1.10 (m, 25H); HRMS (ESI) m/z: [M + H]+ calcd for C55H89BrN11O19, 1286.5504; found, 1286.5514. [00269] (3S,7S,26S,29S,36S)-12-(4-Bromobenzyl)-36-(4-isobutylphenyl)-5,13,20,28,35- pentaoxo-29-(2-(4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido)- 4,6,12,21,27,34-hexaazaheptatriacontane-1,3,7,26-tetracarboxylic acid (177Lu-Alb-L4): To the stirred solution of amine 11 (7.35 mg, 0.0057 mmol, 1.0 eq) and 2,5-dioxopyrrolidin-1-yl 2-(4- isobutylphenyl)propanoate (2.08 mg, 0.0068 mmol, 1.2 eq) in DMSO (100 µL) was added DIPEA (9.75 µL, 0.057 mmol, 10.0 eq) at room temperature. The reaction mixture was stirred for 2 h, concentrated, and the solid crude was purified by preparative RP-HPLC chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization afforded compound 177Lu-Alb-L4 (7.4 mg, 88%) as a white solid. [RP-HPLC purification was achieved using Agilent System, λ 220 nm, 250 mm × 10 mm Phenomenex Luna C18 column, solvent gradient: 95% H2O (0.1% TFA) and 5% ACN (0.1% TFA), reaching 60% of ACN in 20 min at a flow rate of 5 mL/min, product eluted at 18.3 min].1H NMR (500 MHz, DMSO-d6) δ 12.62 (broad singlet, 5H), 8.66 (s, 1H), 8.28 (d, J = 7.0 Hz,
7.92-7.85 (m, 1H), 7.80-7.71 (m, 1H), 7.55 (d, J = 7.5 Hz, 1H), 7.49 (d, J = 7.5 Hz, 1H), 7.22-7.11 (m, 4H), 7.06 (d, J = 8.0 Hz, 2H), 6.38-6.25 (m, 2H), 4.51 (s, 1H), 4.44 (s, 1H), 4.36-4.29 (m, 4H), 3.51 (q, J = 7.0 Hz, 1H), 3.24-2.84 (m, 14H), 2.50 (s, 16H), 2.38 (d, J = 7.0 Hz, 2H), 2.35-2.17 (m, 4H), 2.07-1.85 (m, 3H), 1.83-1.57 (m, 6H), 1.55-1.32 (m, 12H), 1.29-1.11 (m, 13H), 0.84 (d, J = 6.5 Hz, 6H); HRMS (ESI) m/z: [M + H]+ calcd for C68H105BrN11O20, 1474.6715; found, 1474.6715. [00270] (3S,7S,26S,29S)-12-(4-Bromobenzyl)-38-(4-iodophenyl)-5,13,20,28,35-pentaoxo-29- (2-(4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido)- 4,6,12,21,27,34-hexaazaoctatriacontane-1,3,7,26-tetracarboxylic acid (177Lu-Alb-L5): To the stirred solution of amine 11 (8.5 mg, 0.0066 mmol, 1.0 eq) and 2,5-dioxopyrrolidin-1-yl 4-(4- iodophenyl)butanoate (3.0 mg, 0.0079 mmol, 1.2 eq) in DMSO (100 µL) was added DIPEA (11.4 61 42332.601_P15594-02
µL, 0.066 mmol, 10.0 eq) at room temperature. The reaction mixture was stirred for 2 h, and the crude was purified by preparative RP-HPLC chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization afforded compound 177Lu-Alb-L5 (9.0 mg, 88%) as a white solid. RP-HPLC purification was achieved using Agilent System, λ 220 nm, 250 mm × 10 mm Phenomenex Luna C18 column, solvent gradient: 90% H2O (0.1% TFA) and 10% ACN (0.1% TFA), reaching 60% of ACN in 20 min at a flow rate of 10 mL/min, product eluted at 14.2 min].1H NMR (500 MHz, DMSO-d6) δ 8.37 (s, 1H), 8.25 (s, 1H), 7.85-7.72 (m, 2H), 7.647.59 (m, 2H), 7.55 (d, J = 8.0 Hz, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.18-7.11 (m, 2H), 7.00 (d, J = 8.0 Hz, 2H), 6.38-6.26 (m, 2H), 4.50 (s, 1H), 4.43 (s, 1H), 4.35-4.26 (m, 1H), 4.13-4.04 (m, 2H), 2.99 (s, 8H), 3.05-2.85 (m, 6H), 2.50 (s, 16H), 2.38-2.16 (m, 4H), 2.08-1.88 (m, 3H), 1.78-1.11 (m, 34H); HRMS (ESI) m/z: [M + H]+ calcd for C65H98BrIN11O20, 1558.5187; found, 1558.5212. [00271] (((1S)-5-(N-(4-bromobenzyl)-2-((S)-45-(4-iodophenyl)-35,42-dioxo-36-(2-(4,7,10 tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido) 4,7,10,13,16,19,22,25,28,31-decaoxa-34,41-diazapentatetracontanamido)hexanamido)-1- carboxypentyl)carbamoyl)-L-glutamic acid (177Lu-Alb-L6): To the stirred solution of amine 14 10 mg, 0.0062 mmol, 1.0 eq) and IPBA-NHS (2.9 mg, 0.0074 mmol, 1.2 eq) in DMSO (100 µL) was added DIPEA (10.8 µL, 0.062 mmol, 10.0 eq) at room temperature. The reaction mixture was stirred for 2 h, and crude was diluted with water and purified by preparative RP-HPLC chromatography using 0.1% TFA in H2O and 0.1% TFA in acetonitrile as eluents followed by lyophilization produced 177Lu-Alb-L6 (8 mg, 80%) as a white solid. [RP-HPLC purification was achieved using Agilent System, λ 220 nm, 250 mm × 10 mm Phenomenex Luna C18 column, solvent gradient: 90% H2O (0.1% TFA) and 10% ACN (0.1% TFA), reaching 60% of ACN in 20 min at a flow rate of 10 mL/min. 1H NMR (500 MHz, DMSO-d6): δ 8.39 (s, 1H), 8.07 (s, 1H), 7.82 (t, J = 5.0 Hz, 1H), 7.67-7.73 (m, 2H), 7.42-7.50 (m, 2H), 7.07-7.13 (m, 4H), 6.99 (d, J = 10 Hz,2H), 6.47 (d, 2H), 6.21-6.28 (m, 2H), 4.45 (s, 1H), 4.38 (s, 1H), 4.14-4.18 (m, 1H), 4.94-4.06 (m, 2H), 3.45- 3.64 (m, 8H), 3.28-3.41 (m, 30H), 3.08-3.20 (m, 9H), 2.83-2.96 (m, 13H), 2.11-2.23 (m, 8H), 1.82- 1.89 (m, 1H), 1.69-1.76 (m, 1H), 1.53-1.66 (m, 3H), 0.77-1.48 (m, 24H); HRMS (ESI) m/z: [M + H]+ calcd for C80H130BrIN11O28, 1898.7305; found, 1898.7309. [00272] Western Blot [00273] PSMA+ PC3 PIP and PSMA- PC3 flu were homogenized on ice using radioimmunoprecipitation assay buffer (Sigma-Aldrich) containing protease inhibitor cocktail and 62 42332.601_P15594-02
subsequently sonicated to obtain a clear lysate. After centrifugation (10,000g for 15 min at 4°C) to remove cell debris, the supernatant was quantified using the protein assay kit bicinchoninic acid (Thermo Fisher Scientific). About 20 µg of each sample was separated and transferred onto a nitrocellulose membrane. Membranes were incubated in blocking buffer (2.5% BSA, 20% tween 20 in PBS) for 2 h, then washed with PBST and further incubated for 6 h with PSMA (cat#D718E, Cell Signaling Technology) and GAPDH (cat#D16H1, Cell Signaling Technology) antibodies at room temperature. Finally, membranes were incubated with HRP-coupled anti-rabbit IgG secondary antibodies, and blots were developed by ECL reagent. Digital quantification of chemiluminescence was performed using Image J software (NIH). [00274] Real-Time PCR (RT-qPCR) [00275] Cells were cultured to 80% confluence for mRNA isolation in 6 well-cell culture plates. cDNA was synthesized from 1 μg of total RNA of each experimental replicate using a cDNA synthesis kit (Applied Biosystems, USA) as per the manufacturer's protocol. The mRNAs were amplified on Applied Biosystems 7500 Fast Detection system with SYBR green qPCR master mix per manufacturer's instructions (Applied Biosystems, USA). All reactions were performed in triplicate, and negative controls were included in each experiment. GAPDH was taken as internal control, and all the data sets were normalized to the level of GAPDH. Fold change in gene expression was calculated by the Δ2CT method, and results were reported as arbitrary units or fold changes. [00276] Flow Cytometry [00277] PSMA+ PC3 PIP and PSMA- PC3 flu cells (1 × 106 cells) were harvested using cell dissociation buffer (Gibco) and converted into a single-cell suspension. The harvested cells were washed twice with flow cytometry buffer (×1 phosphate-buffered saline with 2 mM ethylenediaminetetraacetic acid and 0.5% fetal bovine serum) and passed by pipetting through a 70 µm strainer. Next, the cells were stained with PSMA-PE antibody (cat#342504BioLegend,) according to the manufactured protocol. Cells were incubated at 4°C for 1 h in the dark. After 1 h, cells were washed with cold phosphate-buffered saline. The unstained and stained cells' fluorescence intensities were analyzed using flow cytometry (Attune NXT). Data were analyzed quantitatively by FlowJo software. 63 42332.601_P15594-02
REFERENCES [00278] All publications, patent applications, patents, and other references mentioned in the specification are indicative of the level of those skilled in the art to which the presently disclosed subject matter pertains. All publications, patent applications, patents, and other references are herein incorporated by reference to the same extent as if each individual publication, patent application, patent, and other reference was specifically and individually indicated to be incorporated by reference. It will be understood that, although a number of patent applications, patents, and other references are referred to herein, such reference does not constitute an admission that any of these documents form part of the common general knowledge in the art. [00279] Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin.2023, 73, 17–48. [00280] Rebello, R.J.; Oing, C.; Knudsen, K.E.; Loeb, S.; Johnson, D.C.; Reiter, R.E.; Gillessen, S.; Van der Kwast, T.; Bristow, R.G. Prostate cancer. Nat. Rev. Dis. Primers 2021, 7, 9. [00281] Sandhu, S.; Moore, C.M.; Chiong, E.; Beltran, H.; Bristow, R.G.; Williams, S.G. Prostate cancer. Lancet 2021, 398, 1075–1090. [00282] Silver, D.A.; Pellicer, I.; Fair, W.R.; Heston, W.D.; Cordon-Cardo, C. Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin. Cancer Res.1997, 3, 81–85. [00283] Wright, G.L.; Haley, C.; Beckett, M.L.; Schellhammer, P.F. Expression of prostate- specific membrane antigen in normal, benign, and malignant prostate tissues. Urol. Oncol. Semin. Orig. Investig.1995, 1, 18–28. [00284] Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177–PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Eng. J. Med.2021, 385, 1091–1103. [00285] Hofman, M.S.; Emmett, L.; Sandhu, S.; Iravani, A.; Joshua, A.M.; Goh, J.C.; Pattison, D.A.; Tan, T.H.; Kirkwood, I.D.; Ng, S.; et al.177Lu-PSMA-617 versus cabazitaxel in patients with metastatic castration-resistant prostate cancer (TheraP): A randomised, open-label, phase 2 trial. Lancet 2021, 397, 797–804. [00286] Sandhu, S.; Guo, C.; Hofman, M.S. Radionuclide Therapy in Prostate Cancer: From Standalone to Combination PSMA Theranostics. J. Nucl. Med.2021, 62, 1660–1668. 64 42332.601_P15594-02
[00287] Banerjee, S.R.; Pullambhatla, M.; Shallal, H.; Lisok, A.; Mease, R.C.; Pomper, M.G. A Modular Strategy to Prepare Multivalent Inhibitors of Prostate-Specific Membrane Antigen (PSMA). Oncotarget 2011, 2, 1244. [00288] Kopka, K.; Benešová, M.; Bařinka, C.; Haberkorn, U.; Babich, J. Glu-Ureido–Based Inhibitors of Prostate-Specific Membrane Antigen: Lessons Learned during the Development of a Novel Class of Low-Molecular-Weight Theranostic Radiotracers. J. Nucl. Med.2017, 58, 17S–26S. [00289] Kelly, J.; Amor-Coarasa, A.; Ponnala, S.; Nikolopoulou, A.; Williams, C.; Schlyer, D.; Zhao, Y.; Kim, D.; Babich, J.W. Trifunctional PSMA-targeting constructs for prostate cancer with unprecedented localization to LNCaP tumors. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1841–1851. [00290] Kelly, J.M.; Amor-Coarasa, A.; Ponnala, S.; Nikolopoulou, A.; Williams, C.; DiMagno, S.G.; Babich, J. Albumin-Binding PSMA Ligands: Implications for Expanding the Therapeutic Window. J. Nucl. Med.2019, 60, 656–663. [00291] Wang, G.; Zang, J.; Jiang, Y.; Liu, Q.; Sui, H.; Wang, R.; Fan, X.; Zhang, J.; Zhu, Z.; Chen, X. A single-arm, low-dose, prospective study of 177Lu-EB-PSMA radioligand therapy in patients with metastatic castration-resistant prostate cancer. J. Nucl. Med.2022, 64, 611–617. [00292] Murce, E.; Beekman, S.; Spaan, E.; Handula, M.; Stuurman, D.; de Ridder, C.; Seimbille, Y. Preclinical Evaluation of a PSMA-Targeting Homodimer with an Optimized Linker for Imaging of Prostate Cancer. Molecules 2023, 28, 4022. [00293] Choy, C.J.; Ling, X.; Geruntho, J.J.; Beyer, S.K.; Latoche, J.D.; Langton-Webster, B.; Anderson, C.J.; Berkman, C.E.177Lu-Labeled Phosphoramidate-Based PSMA Inhibitors: The Effect of an Albumin Binder on Biodistribution and Therapeutic Efficacy in Prostate Tumor- Bearing Mice. Theranostics 2017, 7, 1928–1939. [00294] Benešová, M.; Umbricht, C.A.; Schibli, R.; Müller, C. Albumin-Binding PSMA Ligands: Optimization of the Tissue Distribution Profile. Mol. Pharm.2018, 15, 934–946. [00295] Lau, J.; Jacobson, O.; Niu, G.; Lin, K.-S.; Bénard, F.; Chen, X. Bench to Bedside: Albumin Binders for Improved Cancer Radioligand Therapies. Bioconjug Chem. 2019, 30, 487–502. [00296] Zang, J.; Liu, Q.; Sui, H.; Wang, R.; Jacobson, O.; Fan, X.; Zhu, Z.; Chen, X. 177Lu- EB-PSMA Radioligand Therapy with Escalating Doses in Patients with Metastatic Castration- Resistant Prostate Cancer. J. Nucl. Med.2020, 61, 1772–1778. [00297] Ling, X.; Latoche, J.D.; Choy, C.J.; Kurland, B.F.; Laymon, C.M.; Wu, Y.; Salamacha, N.; Shen, D.; Geruntho, J.J.; Rigatti, L.H.; et al. Preclinical Dosimetry, Imaging, and Targeted 65 42332.601_P15594-02
Radionuclide Therapy Studies of Lu-177-Labeled Albumin-Binding, PSMA-Targeted CTT1403. Mol. Imaging Biol.2020, 22, 274–284. [00298] Kuo, H.-T.; Lin, K.-S.; Zhang, Z.; Uribe, C.F.; Merkens, H.; Zhang, C.; Bénard, F. 177Lu-Labeled Albumin-Binder–Conjugated PSMA-Targeting Agents with Extremely High Tumor Uptake and Enhanced Tumor-to-Kidney Absorbed Dose Ratio. J. Nucl. Med.2021, 62, 521–527. [00299] Reissig, F.; Zarschler, K.; Novy, Z.; Petrik, M.; Bendova, K.; Kurfurstova, D.; Bouchal, J.; Ludik, M.-C.; Brandt, F.; Kopka, K.; et al. Modulating the pharmacokinetic profile of Actinium-225-labeled macropa-derived radioconjugates by dual targeting of PSMA and albumin. Theranostics 2022, 12, 7203–7215. [00300] Zorzi, A.; Linciano, S.; Angelini, A. Non-covalent albumin-binding ligands for extending the circulating half-life of small biotherapeutics. MedChemComm 2019, 10, 1068– 1081. [00301] [00302] Kramer, V.; Fernández, R.; Lehnert, W.; Jiménez-Franco, L.D.; Soza-Ried, C.; Eppard, E.; Ceballos, M.; Meckel, M.; Benešová, M.; Umbricht, C.A.; et al. Biodistribution and dosimetry of a single dose of albumin-binding ligand [177Lu]Lu-PSMA-ALB-56 in patients with mCRPC. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 893–903. [00303] Tschan, V.J.; Borgna, F.; Busslinger, S.D.; Stirn, M.; Rodriguez, J.M.M.; Bernhardt, P.; Schibli, R.; Müller, C. Preclinical investigations using [177Lu]Lu-Ibu-DAB-PSMA toward its clinical translation for radioligand therapy of prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 3639–3650. [00304] Kuo, H.-T.; Lin, K.-S.; Zhang, Z.; Zhang, C.; Merkens, H.; Tan, R.; Roxin, A.; Uribe, C.F.; Bénard, F. What a difference a methylene makes: Replacing Glu with Asp or Aad in the Lys-urea-Glu pharmacophore of PSMA-targeting radioligands to reduce kidney and salivary gland uptake. Theranostics 2022, 12, 6179–6188. [00305] Deberle, L.M.; Benešová, M.; Umbricht, C.A.; Borgna, F.; Büchler, M.; Zhernosekov, K.; Schibli, R.; Müller, C. Development of a new class of PSMA radioligands comprising ibuprofen as an albumin-binding entity. Theranostics 2020, 10, 1678–1693. [00306] Banerjee, S.R.; Kumar, V.; Lisok, A.; Chen, J.; Minn, I.; Brummet, M.; Boinapally, S.; Cole, M.; Ngen, E.; Wharram, B.; et al.177Lu-labeled low-molecular-weight agents for PSMA- targeted radiopharmaceutical therapy. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2545–2557. [00307] Banerjee, S.R.; Minn, I.; Kumar, V.; Josefsson, A.; Lisok, A.; Brummet, M.; Chen, J.; Kiess, A.P.; Baidoo, K.; Brayton, C.; et al. Preclinical Evaluation of 203/212Pb-Labeled Low- 66 42332.601_P15594-02
Molecular-Weight Compounds for Targeted Radiopharmaceutical Therapy of Prostate Cancer. J. Nucl. Med.2020, 61, 80–88. [00308] Banerjee, S.R.; Lisok, A.; Minn, I.; Josefsson, A.; Kumar, V.; Brummet, M.; Boinapally, S.; Brayton, C.; Mease, R.C.; Sgouros, G.; et al. Preclinical evaluation of 213Bi- and 225Ac-Labeled Low-Molecular-Weight Compounds for Radiopharmaceutical Therapy of Prostate Cancer. J. Nucl. Med.2021, 62, 980–988. [00309] Mease, R.C.; Kang, C.M.; Kumar, V.; Banerjee, S.R.; Minn, I.; Brummet, M.; Gabrielson, K.L.; Feng, Y.; Park, A.; Kiess, A.P.; et al. An Improved 211At-Labeled Agent for PSMA-Targeted alpha-Therapy. J. Nucl. Med.2022, 63, 259–267. [00310] Olszewski, R.T.; Bukhari, N.; Zhou, J.; Kozikowski, A.P.; Wroblewski, J.T.; Shamimi- Noori, S.; Wroblewska, B.; Bzdega, T.; Vicini, S.; Barton, F.B.; et al. NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR. J. Neurochem.2004, 89, 876–885. [00311] Banerjee, S.R.; Foss, C.A.; Castanares, M.; Mease, R.C.; Byun, Y.; Fox, J.J.; Hilton, J.; Lupold, S.E.; Kozikowski, A.P.; Pomper, M.G. Synthesis and evaluation of technetium- 99m- and rhenium-labeled inhibitors of the prostate-specific membrane antigen (PSMA). J. Med. Chem.2008, 51, 4504–4517. [00312] Banerjee, S.R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Green, G.; Fox, J.J.; Horti, A.; Mease, R.C.; Pomper, M.G. 68Ga-labeled inhibitors of prostate-specific membrane antigen (PSMA) for imaging prostate cancer. J. Med. Chem.2010, 53, 5333–5341. [00313] Chen, Y.; Minn, I.; Rowe, S.P.; Lisok, A.; Chatterjee, S.; Brummet, M.; Banerjee, S.R.; Mease, R.C.; Pomper, M.G. A Series of PSMA-Targeted Near-Infrared Fluorescent Imaging Agents. Biomolecules 2022, 12, 405. [00314] Bacich, D.J.; Pinto, J.T.; Tong, W.P.; Heston, W.D.W. Cloning, expression, genomic localization, and enzymatic activities of the mouse homolog of prostate-specific membrane antigen/NAALADase/folate hydrolase. Mamm. Genome 2001, 12, 117–123. [00315] Borgna, F.; Deberle, L.M.; Cohrs, S.; Schibli, R.; Müller, C. Combined Application of Albumin-Binding [177Lu]Lu-PSMA-ALB-56 and Fast-Cleared PSMA Inhibitors: Optimization of the Pharmacokinetics. Mol. Pharm.2020, 17, 2044–2053. [00316] Busslinger, S.D.; Tschan, V.J.; Richard, O.K.; Talip, Z.; Schibli, R.; Müller, C. [225Ac]Ac-SibuDAB for Targeted Alpha Therapy of Prostate Cancer: Preclinical Evaluation and Comparison with [225Ac]Ac-PSMA-617. Cancers 2022, 14, 5651. [00317] Banerjee, S.R.; Kumar, V.; Lisok, A.; Plyku, D.; Novakova, Z.; Brummet, M.; Wharram, B.; Barinka, C.; Hobbs, R.; Pomper, M.G. Evaluation of 111In-DOTA-5D3, a 67 42332.601_P15594-02
Surrogate SPECT Imaging Agent for Radioimmunotherapy of Prostate-Specific Membrane Antigen. J. Nucl. Med.2019, 60, 400–406. [00318] Tschan, V.J.; Borgna, F.; Schibli, R.; Müller, C. Impact of the mouse model and molar amount of injected ligand on the tissue distribution profile of PSMA radioligands. Eur. J. Nucl. Med. Mol. Imaging 2021, 49, 470–480. [00319] Banerjee, S.R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Foss, C.A.; Green, G.; Fox, J.J.; Lupold, S.E.; Mease, R.C.; Pomper, M.G. Sequential SPECT and optical imaging of experimental models of prostate cancer with a dual modality inhibitor of the prostate-specific membrane antigen. Angew. Chem. Int. Ed.2011, 50, 9167–9170. [00320] Cheng, Y.; Prusoff, W.H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol.1973, 22, 3099–3108. [00321] Tsuchihashi, S.; Nakashima, K.; Tarumizu, Y.; Ichikawa, H.; Jinda, H.; Watanabe, H.; Ono, M. Development of Novel 111In/225Ac-Labeled Agent Targeting PSMA for Highly Efficient Cancer Radiotheranostics. J. Med. Chem.2023, 66, 8043–8053. [00322] Leek, J.; Lench, N.; Maraj, B.; Bailey, A.; Carr, I.M.; Andersen, S.; Cross, J.; Whelan, P.; MacLennan, K.A.; Meredith, D.M.; et al. Prostate-specific membrane antigen: Evidence for the existence of a second related human gene. Br. J. Cancer 1995, 72, 583–588. [00323] Chang, S.S.; Reuter, V.E.; Heston, W.D.W.; Bander, N.H.; Grauer, L.S.; Gaudin, P.B. Five Different Anti-Prostate-specific Membrane Antigen (PSMA) Antibodies Confirm PSMA Expression in Tumor-associated Neovasculature1. Cancer Res.1999, 59, 3192–3198. [00324] Liu, C.; Hasegawa, K.; Russell, S.J.; Sadelain, M.; Peng, K.-W. Prostate-specific membrane antigen retargeted measles virotherapy for the treatment of prostate cancer. Prostate 2009, 69, 1128–1141. [00325] Franken, N.A.; Rodermond, H.M.; Stap, J.; Haveman, J.; van Bree, C. Clonogenic assay of cells in vitro. Nat. Protoc.2006, 1, 2315–2319. [00326] Although the foregoing subject matter has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be understood by those skilled in the art that certain changes and modifications can be practiced within the scope of the appended claims. 68 42332.601_P15594-02
Claims
2,
3,
4,
5,
6,
7,
8,
9, 10, 11, and 12; M is present or absent and when present is a metal or a radiometal; and stereoisomers and pharmaceutically acceptable salts thereof. 2. The compound of claim 1, wherein R1 is selected from ;
42332.601_P15594-02
wherein X is independently Br, I, or At and radioisotopes thereof. 3. The compound of claim 2, wherein X is selected from 211At, 131I, 125I, 124I, 123I, 77Br, and 80mBr. 4. The compound of claim 1, wherein M is a metal selected from Y, Lu, Tc, Zr, In, Sm, Re, Cu, Pb, Ac, Bi, Al, Ga, Re, Ho and Sc. 5. The compound of claim 1, wherein M is a radiometal selected from 68Ga, 64Cu, 86Y, 90Y, 89Zr, 111In, 99mTc, 177Lu, 153Sm, 186Re, 188Re, 67Cu, 212Pb, 225Ac, 213Bi, 212Bi, 67Ga, 203Pb, 47Sc, 149Tb, and 166Ho. 6. The compound of claim 5, wherein M is selected from 177Lu, 225Ac, 213Bi, and 203Pb. 7. The compound of claim 6, wherein M is 177Lu. 8. The compound of any one of claims 1-7, wherein the compound of formula (I) is selected from: ;
42332.601_P15594-02
; 42332.601_P15594-02
I O O HN
9. A method for treating or imaging one or more PSMA expressing tumors or cells, the method comprising contacting the one or more PSMA expressing tumors or cells with an effective amount of a compound of any one of claims 1-8, and, when the method is an imaging method, taking an image.
10. The method of claim 9, wherein the one or more PSMA-expressing tumors or cells is selected from the group consisting of a prostate tumor or cell, a metastasized prostate tumor or cell, a lung tumor or cell, a renal tumor or cell, a glioblastoma, a pancreatic tumor or cell, a bladder tumor or cell, a sarcoma, a melanoma, a breast tumor or cell, a colon tumor or cell, a germ cell, a pheochromocytoma, an esophageal tumor or cell, a stomach tumor or cell, and combinations thereof.
11. The method of claim 10, wherein the one or more PSMA-expressing tumors or cells is a prostate tumor or cell. 72 42332.601_P15594-02
12. The method of claim 9, wherein the one or more PSMA-expressing tumor or cell is in vitro, in vivo, or ex vivo.
13. The method of claim 9, wherein the one or more PSMA-expressing tumor or cell is present in a subject.
14. The method of claim 13, wherein the subject is human.
15. The method of claim 9, wherein the method results in inhibition of the tumor growth. 73 42332.601_P15594-02
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363520883P | 2023-08-21 | 2023-08-21 | |
| US63/520,883 | 2023-08-21 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025042989A1 true WO2025042989A1 (en) | 2025-02-27 |
Family
ID=94732777
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/043231 Pending WO2025042989A1 (en) | 2023-08-21 | 2024-08-21 | Psma-based albumin binding agents for targeted radionuclide therapy of prostate cancer |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025042989A1 (en) |
Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20210220493A1 (en) * | 2018-02-06 | 2021-07-22 | The Johns Hopkins University | Psma targeted radiohalogenated urea-polyaminocarboxylates for cancer radiotherapy |
| US20220024882A1 (en) * | 2018-11-28 | 2022-01-27 | ITM Isotopen Technologien München AG | Novel tumor antigen binding agents and uses thereof |
-
2024
- 2024-08-21 WO PCT/US2024/043231 patent/WO2025042989A1/en active Pending
Patent Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20210220493A1 (en) * | 2018-02-06 | 2021-07-22 | The Johns Hopkins University | Psma targeted radiohalogenated urea-polyaminocarboxylates for cancer radiotherapy |
| US20220024882A1 (en) * | 2018-11-28 | 2022-01-27 | ITM Isotopen Technologien München AG | Novel tumor antigen binding agents and uses thereof |
Non-Patent Citations (1)
| Title |
|---|
| MEASE RONNIE C., KANG CHOONG MO, KUMAR VIVEK, BANERJEE SANGEETA RAY, MINN IL, BRUMMET MARY, GABRIELSON KATHLEEN L., FENG YUTIAN, P: "An Improved 211 At-Labeled Agent for PSMA-Targeted α-Therapy", THE JOURNAL OF NUCLEAR MEDICINE, SOCIETY OF NUCLEAR MEDICINE, US, vol. 63, no. 2, 1 February 2022 (2022-02-01), US , pages 259 - 267, XP093283928, ISSN: 0161-5505, DOI: 10.2967/jnumed.121.262098 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20230147035A1 (en) | Prostate-specific membrane antigen targeted high-affinity agents for endoradiotherapy of prostate cancer | |
| US20250230135A1 (en) | Metal/radiometal-labeled psma inhibitors for psma-targeted imaging and radiotherapy | |
| US20250195700A1 (en) | Prostate-specific membrane antigen targeted high-affinity agents for endoradiotherapy of prostate cancer | |
| US11911488B2 (en) | Scaffolds and multifunctional intermediates for imaging PSMA and cancer therapy | |
| Duan et al. | (S)-3-(Carboxyformamido)-2-(3-(carboxymethyl) ureido) propanoic acid as a novel PSMA targeting scaffold for prostate cancer imaging | |
| US20210040126A1 (en) | 68Ga-LABELED NOTA-CHELATED PSMA-TARGETED IMAGING AND THERAPEUTIC AGENTS | |
| JP2020522506A5 (en) | ||
| US11078166B2 (en) | Triazole conjugated ureas, thioureas, carbamates, and reversed carbamates for PSMA-targeted imaging agents and uses thereof | |
| WO2016149188A1 (en) | 68Ga-LABELED NOTA-CHELATED PSMA-TARGETED IMAGING AND THERAPEUTIC AGENTS | |
| JP2024505374A (en) | Dual mode radiotracer and its therapy | |
| WO2025042989A1 (en) | Psma-based albumin binding agents for targeted radionuclide therapy of prostate cancer | |
| AU2017260080B2 (en) | 18F-FNDP for pet imaging of soluble epoxide hydrolase (sEH) | |
| HK1259764B (en) | Prostate-specific membrane antigen targeted high-affinity agents for endoradiotherapy of prostate cancer |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24857238 Country of ref document: EP Kind code of ref document: A1 |