WO2018175589A1 - Modified peptides and uses thereof for treating cancer - Google Patents

Modified peptides and uses thereof for treating cancer Download PDF

Info

Publication number
WO2018175589A1
WO2018175589A1 PCT/US2018/023573 US2018023573W WO2018175589A1 WO 2018175589 A1 WO2018175589 A1 WO 2018175589A1 US 2018023573 W US2018023573 W US 2018023573W WO 2018175589 A1 WO2018175589 A1 WO 2018175589A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
protein
pharmaceutical composition
weight
group
Prior art date
Application number
PCT/US2018/023573
Other languages
French (fr)
Inventor
Matthew P. THOMPSON
Nathan C. Gianneschi
Paul A. Bertin
Cassandra E. CALLMANN
Original Assignee
The Regents Of The University Of California
Vybyl Holdings, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Regents Of The University Of California, Vybyl Holdings, Inc. filed Critical The Regents Of The University Of California
Publication of WO2018175589A1 publication Critical patent/WO2018175589A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/38Albumins
    • A61K38/385Serum albumin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons

Definitions

  • the present disclosure generally provides compounds useful for treating cancer.
  • the disclosure provides containing compounds that are chemically modified to have one or more moieties that include hydrophobic portions.
  • the disclosure provides compositions that include such modified peptides and another protein, such as albumin or albumin mimetics. Further, the disclosure provides various uses of these compounds and compositions.
  • Cancer refers to a group of diseases characterized by the formation of malignant tumors or neoplasms, which involve abnormal cell growth and have the potential to invade adjacent tissue and spread to other parts of the body. There are more than 14 million new diagnoses of cancer annually. Moreover, cancer accounts for more than 8 million deaths each year, which is about 15% of all deaths worldwide. In developed countries, cancer accounts for an even higher percentage of deaths.
  • cytotoxic agents have been used to treat cancer. And while these agents have generally afforded effective means of treating cancer, they often exhibit off-target activity towards other cells in the body. Therefore, patients undergoing such treatment often suffer from a number of side-effects.
  • Peptide-based therapies for cancer offer the possibility of creating molecules that are specifically designed to act selectively against certain cancerous cells without having a significant effect on other non-cancerous cells. This is especially true as researchers develop an increasingly detailed understanding of the specific genetic makeup of certain cancers. Such compounds come in a wide variety of different classes. Some peptide- based drugs act in ways that are similar to traditional cytotoxins.
  • ADCs Antibody-drug conjugates
  • the present disclosure provides compounds and compositions that can deliver increasingly high quantities of peptide-based cancer therapies to cancer cells (e.g., in a solid tumor) with reduced off-target side effects and improved pharmacokinetic lifetime.
  • the compounds are prodrugs of peptide compounds, such that the prodrug permits improved delivery of the peptide compound to a solid tumor in a mammal.
  • the disclosure also provides methods and uses of those compounds and compositions for the treatment of cancer.
  • a 1 is an organic group, a hydrophilic group, or a hydrogen atom
  • a 2 is a peptide moiety
  • X 1 is a hydrophobic group
  • a 1 is a hydrophilic group, such as a carboxylic acid group (-COOH) or a pharmaceutically acceptable salt thereof.
  • the peptide moiety is an anticancer peptide moiety.
  • the hydrophobic group is a C 12-22 hydrocarbylene group, which is optionally substituted.
  • compositions e.g., pharmaceutical compositions
  • a compound of any embodiments of the first aspect e.g., and a protein.
  • the protein is an albumin or an albumin mimetic.
  • compositions e.g., pharmaceutical compositions
  • a compound of any embodiments of the first aspect a protein, wherein the protein is an albumin or an albumin mimetic; and a carrier, which includes water; wherein the compound and the protein are non-covalently associated with each other; and wherein the compound and the protein are solvated by the carrier.
  • the disclosure provides methods of treating cancer, which include administering to a subject a compound or composition of any embodiments of any of the foregoing aspects. In some further embodiments thereof, the disclosure provides methods of treating cancer that include administering to a subject one or more immunotherapy agents.
  • the disclosure provides methods of inducing apoptosis in a cancer cell, which include contacting the cancer cell with a compound or composition of any embodiments of any of the first through the third aspects. In some further embodiments thereof, the disclosure provides methods of inducing apoptosis in a cancer cell that include contacting the cancer cell with one or more immunotherapy agents.
  • the disclosure provides methods for inhibiting growth of a cancerous tumor, which includes contacting the cancerous tumor with a compound of any embodiments of the first aspect. In some further embodiments thereof, the disclosure provides methods of inhibiting growth of a cancerous tumor that include contacting the cancerous tumor with one or more immunotherapy agents.
  • the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects as a medicament.
  • the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects for treating cancer. In some further embodiments thereof, the disclosure provides uses that include use in combination with one or more immunotherapy agents.
  • the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects in the manufacture of a medicament. In a tenth aspect, the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects in the manufacture of a medicament for treating cancer.
  • the disclosure provides methods of making compounds of the first and second aspects and compositions of the third and fourth aspects.
  • FIG. 1 shows a non-limiting example of a compound of formula (I), where the compound includes a peptide moiety, which is modified to include a long-chain dibasic acid moiety.
  • hydrocarbon refers to an organic group composed of carbon and hydrogen, which can be saturated or unsaturated, and can include aromatic groups.
  • hydrocarbyl refers to a monovalent or polyvalent (e.g., divalent or higher) hydrocarbon moiety. In some cases, a divalent hydrocarbyl group is referred to as a "hydrocarbylene” group.
  • alkyl refers to a straight or branched chain saturated hydrocarbon having 1 to 30 carbon atoms, which may be optionally substituted, as herein further described, with multiple degrees of substitution being allowed.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, isobutyl, n-butyl, sec-butyl, tert-butyl, isopentyl, n-pentyl, neopentyl, n-hexyl, and 2-ethylhexyl.
  • the "alkyl” group can be divalent, in which case, the group can alternatively be referred to as an "alkylene” group.
  • one or more of the carbon atoms in the alkyl or alkylene group can be replaced by a heteroatom (e.g., selected from nitrogen, oxygen, or sulfur, including N-oxides, sulfur oxides, sulfur dioxides, and carbonyl groups, where feasible), and is referred to as a "heteroalkyl” or “heteroalkylene” group, respectively.
  • Non-limiting examples include “oxyalkyl” or “oxyalkylene” groups, which refer to groups where a carbon atom in the alkyl or alkylene group is replaced by oxygen.
  • Non-limiting examples of oxyalkyl or oxyalkylene groups include alkyl or alkylene chains that contain a carbonyl group, and also alkoxylates, polyalkylene oxides, and the like.
  • C z refers to a group of compound having z carbon atoms
  • C x-y refers to a group or compound containing from x to y, inclusive, carbon atoms.
  • Ci-6 alkyl represents an alkyl group having from 1 to 6 carbon atoms and, for example, includes, but is not limited to, methyl, ethyl, n-propyl, isopropyl, isobutyl, n-butyl, sec-butyl, tert-butyl, isopentyl, n-pentyl, neopentyl, and n-hexyl.
  • alkenyl refers to a straight or branched chain non-aromatic hydrocarbon having 2 to 30 carbon atoms and having one or more carbon-carbon double bonds, which may be optionally substituted, as herein further described, with multiple degrees of substitution being allowed.
  • alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-butenyl, and 3-butenyl. In some instances, the
  • alkenyl group can be divalent, in which case the group can altematively be referred to as an "alkenylene” group. Also, in some instances, one or more of the carbon atoms in the alkenyl or alkenylene group can be replaced by a heteroatom (e.g., selected from nitrogen, oxygen, or sulfur, including N-oxides, sulfur oxides, sulfur dioxides, and carbonyl groups, where feasible), and is referred to as a “heteroalkenyl” or “heteroalkenylene” group, respectively.
  • a heteroatom e.g., selected from nitrogen, oxygen, or sulfur, including N-oxides, sulfur oxides, sulfur dioxides, and carbonyl groups, where feasible
  • cycloalkyl refers to an aliphatic saturated or unsaturated hydrocarbon ring system having 3 to 20 carbon atoms, which may be optionally substituted, as herein further described, with multiple degrees of substitution being allowed. In some embodiments, the term refers only to saturated hydrocarbon ring systems, substituted as herein further described. Examples of “cycloalkyl,” as used herein, include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, adamantyl, and the like.
  • the "cycloalkyl” group can be divalent, in which case the group can alternatively be referred to as a "cycloalkylene” group.
  • Cycloalkyl and cycloalkylene groups can also be referred to herein as "carbocyclic rings.”
  • one or more of the carbon atoms in the cycloalkyl or cycloalkylene group can be replaced by a heteroatom (e.g., selected independently from nitrogen, oxygen, silicon, or sulfur, including N-oxides, sulfur oxides, and sulfur dioxides, where feasible), and is referred to as a "heterocyclyl” or “heterocyclylene” group, respectively.
  • a heteroatom e.g., selected independently from nitrogen, oxygen, silicon, or sulfur, including N-oxides, sulfur oxides, and sulfur dioxides, where feasible
  • heterocyclic ring can also be used interchangeably with either of these terms.
  • the cycloalkyl and heterocyclyl groups are fully saturated.
  • the cycloalkyl and heterocyclyl groups can contain one or more carbon-carbon double bonds.
  • halogen refers to a fluorine, chlorine, bromine, or iodine atom. In some embodiments, the terms refer to a fluorine or chlorine atom.
  • organic group refers to a monovalent or polyvalent functional group having at least one carbon atom, which optionally contains one or more additional atoms selected from the group consisting of hydrogen atoms, halogen atoms, nitrogen atoms, oxygen atoms, phosphorus atoms, and sulfur atoms, and which does not include covalently bound metal or semi-metal atoms.
  • these terms can include metal salts of organic groups, such as alkali metal or alkaline earth metal salts of organic anions.
  • pharmacophore refers to a type of organic functional group.
  • Standard pharmacophores are hydrophobic pharmacophores, hydrogen-bond donating pharmacophores, hydrogen-bond accepting pharmacophores, positive ionizable
  • hydrophobic group As used herein, the terms “hydrophobic group,” “hydrophobic moiety,” or
  • hydrophobic residue refer to an organic group that consists essentially of hydrophobic pharmacophores. In some embodiments, the terms refer to an organic group that consists of hydrophobic pharmacophores.
  • hydrophilic group refers to an organic group that comprises one pharmacophores selected from the group consisting of hydrogen bond donors, hydrogen bond acceptors, negative ionizable groups, or positive ionizable groups.
  • the terms refer to an organic group that consist essentially of pharmacophores selected from the group consisting of hydrogen bond donors, hydrogen bond acceptors, negative ionizable groups, or positive ionizable groups.
  • peptide moiety refers to a peptide compound, or a pharmaceutically acceptable salt thereof, where an atom or a group of atoms is absent, thereby creating a monovalent or polyvalent moiety.
  • peptide compound refers to a compound formed from the condensation of two or more amino acid compounds. Any suitable amino acid compounds can be used, including L-amino acids or D-amino acids.
  • the amino acids can be alpha-amino acids, beta-amino acids, gamma-amino acids, and delta-amino acids. In some embodiments, the amino acids are L-alpha-amino acids.
  • At least 80% by number, or at least 85% by number, or at least 90% by number, or at least 95% by number of the amino acids forming the peptide moiety are selected from the 22 proteinogenic amino acids.
  • Conjugates can be formed in any suitable way.
  • a hydrogen atom is absent from the N-terminal end of the peptide compound, thereby creating a monovalent moiety.
  • a non-limiting example of such a "peptide moiety,” is the moiety of the following formula:
  • the bond line-structure method is used to depict chemical compounds or moieties.
  • the lines represent chemical bonds, and the carbon atoms are not explicitly shown (but are implied by the intersection of the lines).
  • the hydrogen atoms are also not explicitly shown, except in instances where they are attached to heteroatoms. Heteroatoms, however, are explicitly shown.
  • the structures shown below are for 2-methylpropane, 1 -methoxypropane, and 1 -propanol:
  • aromatic rings are typically represented merely by one of the contributing resonance structures.
  • the following structures are for benzene, pyridine,
  • a "protein binding moiety” is a moiety that binds non-covalently to one or more sites on a protein with a binding constant (Kb) of at least 100 M "1 in water at 25 °C.
  • amino acid refers to a compound having the structure
  • H2N-R x -COOH where R x is an organic group, and where the NH2 may optionally combine with Rx (e.g., as in the case of proline).
  • R x is an organic group
  • NH2 may optionally combine with Rx (e.g., as in the case of proline).
  • the term includes any known amino acids, including, but not limited to, alpha amino acids, beta amino acids, gamma amino acids, delta amino acids, and the like. In some embodiments, the term can refer to alpha amino acids.
  • hydroxy acid refers to a compound having the structure
  • R y is an organic group.
  • Non-limiting examples include gly colic acid, lactic acid, and caprolactone.
  • alkanol amine refers to a compound having the structure
  • R z is an optionally substituted alkylene group.
  • Non-limiting examples include ethanol amine.
  • administer means to introduce, such as to introduce to a subject a compound or composition.
  • the term is not limited to any specific mode of delivery, and can include, for example, subcutaneous delivery, intravenous delivery, intramuscular delivery, intracisternal delivery, delivery by infusion techniques, transdermal delivery, oral delivery, nasal delivery, and rectal delivery.
  • the administering can be carried out by various individuals, including, for example, a health-care professional (e.g., physician, nurse, etc.), a pharmacist, or the subj ect (i.e., self-administration).
  • treat or “treating” or “treatment” can refer to one or more of:
  • delaying the progress of a disease, disorder, or condition controlling a disease, disorder, or condition; ameliorating one or more symptoms characteristic of a disease, disorder, or condition; or delaying the recurrence of a disease, disorder, or condition, or characteristic symptoms thereof, depending on the nature of the disease, disorder, or condition and its characteristic symptoms.
  • subject refers to any mammal such as, but not limited to, humans, horses, cows, sheep, pigs, mice, rats, dogs, cats, and primates such as chimpanzees, gorillas, and rhesus monkeys.
  • the "subject” is a human.
  • the "subject” is a human who exhibits one or more symptoms characteristic of a disease, disorder, or condition.
  • the term “subject” does not require one to have any particular status with respect to a hospital, clinic, or research facility (e.g., as an admitted patient, a study participant, or the like).
  • compound includes free acids, free bases, and salts thereof.
  • composition is used to denote a composition that may be administered to a mammalian host, e.g., orally, topically, parenterally, by inhalation spray, or rectally, in unit dosage formulations containing conventional non-toxic carriers, diluents, adjuvants, vehicles and the like.
  • a mammalian host e.g., orally, topically, parenterally, by inhalation spray, or rectally
  • unit dosage formulations containing conventional non-toxic carriers, diluents, adjuvants, vehicles and the like.
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or by infusion techniques. Also included within the scope of the disclosure are the individual enantiomers of the compounds represented by Formula (I) or pharmaceutically acceptable salts thereof, as well as any wholly or partially racemic mixtures thereof. The disclosure also covers the individual enantiomers of the compounds represented by Formula (I) or pharmaceutically acceptable salts thereof, as well as mixtures with diastereoisomers thereof in which one or more stereocenters are inverted. 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 structure, except for the replacement of a hydrogen atom by a deuterium or tritium, or the replacement of a carbon atom by a 1 C- or 14 C-enriched carbon are within the scope of the disclosure.
  • mixture refers broadly to any combining of two or more compositions.
  • the two or more compositions need not have the same physical state; thus, solids can be “mixed” with liquids, e.g., to form a slurry, suspension, or solution. Further, these terms do not require any degree of homogeneity or uniformity of composition. This, such “mixtures” can be homogeneous or heterogeneous, or can be uniform or nonuniform. Further, the terms do not require the use of any particular equipment to carry out the mixing, such as an industrial mixer.
  • optional event means that the subsequently described event(s) may or may not occur. In some embodiments, the optional event does not occur. In some other embodiments, the optional event does occur one or more times.
  • substituted refers to substitution of one or more hydrogen atoms of the designated moiety with the named substituent or substituents, multiple degrees of substitution being allowed unless otherwise stated, provided that the substitution results in a stable or chemically feasible compound.
  • a stable compound or chemically feasible compound is one in which the chemical structure is not substantially altered when kept at a temperature from about -80 °C to about +40 °C, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • the phrases “substituted with one or more... " or “substituted one or more times ... " refer to a number of substituents that equals from one to the maximum number of substituents possible based on the number of available bonding sites, provided that the above conditions of stability and chemical feasibility are met.
  • multi-atom bivalent species are to be read from left to right.
  • D is defined as -OC(O)-
  • the resulting group with D replaced is: A-OC(0)-E and not A-C(0)0-E.
  • the disclosure provides compounds of formula (I):
  • a 1 is a hydrophilic group a hydrogen atom, or an organic group
  • a 2 is a peptide moiety
  • X 1 is a hydrophobic group
  • a 1 is an organic group.
  • a 1 can contain any suitable number of carbon atoms. In some embodiments, for example, A 1 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms, or from 1 to 10 carbon atoms, or from 1 to 6 carbon atoms.
  • a 1 can also contain one or more heteroatoms, such as nitrogen, oxygen, sulfur, or phosphorus.
  • a 1 is a hydrophilic group or moiety.
  • a hydrophilic group include, but are not limited to, a carboxylic acid moiety, an ester moiety, an amide moiety, a urea moiety, an amine moiety, an ether moiety, an alcohol moiety, a thioether moiety, a thiol moiety, a ketone moiety, an aldehyde moiety, a sulfate moiety, a thiosulfate moiety, a sulfite moiety, a thiosulfite moiety, a phosphate moiety, a phosphonate moiety, a phosphinate moiety, a phosphite moiety, a borate moiety, or a boronate moiety.
  • a 1 is selected from the group consisting of a carboxylic acid group (-COOH), a carboxylate anion (-COO " ), or a carboxylate ester (-COOR a , where R a is an organic group such as an alkyl or alkoxylate group). In some such embodiments, A 1 is a carboxylic acid group. In some such embodiments,
  • a 1 is a carboxylate ester group.
  • a 1 is a hydrogen atom. In some other embodiments of any of the aforementioned embodiments, A 1 is a hydroxyl (-OH) group.
  • X 1 can be a hydrophobic group having any suitable number of carbon atoms. In some embodiments, for example, X 1 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms.
  • X 1 is Cs-3o hydrocarbylene, which is optionally substituted. In some further embodiments, X 1 is C 12-22 hydrocarbylene, which is optionally substituted. In some further embodiments, X 1 is C 12-22 alkylene. In some further embodiments, X 1 is -(CH2)i2-, -(CH2)i4-, -(CH2)i6-, -(CH2)i8-, -(CH2)2o-, or -(CH2)22-. In some other embodiments, X 1 is -(CH2)i6-. In some further embodiments, X 1 is C 12-22 alkenylene. In some further such embodiments, X 1 is
  • X 1 is C 12-22 hydrocarbylene, which is optionally substituted. In some such embodiments, X 1 is C 12-22 hydrocarbylene. In some further such embodiments, X 1 is C 14-22 hydrocarbylene. In some further such embodiments, X 1 is C16-22 hydrocarbylene. In some embodiments of any of the aforementioned embodiments, X 1 is C12-22 hydrocarbylene, wherein A 1 and X 2 (or, if X 2 is a direct bond, A 2 ) are separated from each other by at least 6, or by at least 8, or by at least 10, or by at least 12, or by at least 14, carbon atoms.
  • X 1 is Ci4-22 hydrocarbylene, wherein A 1 and X 2 (or, if X 2 is a direct bond, A 2 ) are separated from each other by at least 6, or by at least 8, or by at least 10, or by at least 12, or by at least 14, carbon atoms.
  • X 1 is Ci6-22 hydrocarbylene, wherein A 1 and X 2 (or, if X 2 is a direct bond, A 2 ) are separated from each other by at least 6, or by at least 8, or by at least 10, or by at least 12, or by at least 14, carbon atoms.
  • X 1 is C12-22 straight-chain alkylene, or C 14-22 straight-chain alkylene, or C16-22 straight-chain alkylene. In some further embodiments of any of the aforementioned embodiments, X 1 is C12-22 straight-chain alkenylene, or C 14-22 straight-chain alkenylene, or C16-22 straight-chain alkenylene.
  • X 2 is a direct bond.
  • X 2 is an organic group. In some embodiments, X 2 is a hydrophilic group. In some embodiments, X 2 is a heteroalkylene group.
  • X 2 can contain any suitable number of carbon atoms. In some embodiments, for example, X 2 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms, or from 1 to 10 carbon atoms, or from 1 to 6 carbon atoms.
  • X 2 can contain any suitable number of carbon atoms. In some embodiments, for example, X 2 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms, or from 1 to 10 carbon atoms, or from 1 to 6 carbon atoms.
  • X 2 can contain certain groups.
  • groups that X 2 can contain are polyalkylene oxide groups, such as polyethylene glycol (PEG) and various polypeptide chains.
  • X 2 is an organic group selected from the group consisting of
  • R c , R d , and R e are, independently at each occurrence, a hydrogen atom or Ci-io alkyl.
  • X 2 comprises one or more moieties formed from alkylene glycols, such as a short poly(ethylene glycol) chain having 1 to 25 ethylene glycol units.
  • X 2 comprises one or more moieties formed from amino acids, such as an oligopeptide chain having 1 to 25 amino acid units.
  • X 2 comprises one or more moieties formed from hydroxy acids, such as moieties formed from gly colic acid, lactic acid, or caprolactone.
  • X 2 comprises a combination of a poly(ethylene glycol) chain having 1 to 25 ethylene glycol units and an oligopeptide having 1 to 25 amino acid units, and optionally one or more units formed from hydroxy acids.
  • X 2 will depend on the type of functional group through which it is linked to the peptide moiety, so as to avoid making compounds that are chemically unstable or impossible.
  • the skilled artisan will be able to select combinations of X 2 and A 2 that result in chemically stable compounds, which are compounds in which the chemical structure is not substantially altered when kept at a temperature from about -80 °C to about +40 °C, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • a 2 can be any suitable peptide moiety.
  • the peptide moiety can contain any suitable number of amino acid units (i.e., units in the peptide chain formed from an amino acid).
  • the peptide moiety comprises from 2 to 100 amino acid units, or from 3 to 50 amino acid units, or from 4 to 35 amino acid units, or from 5 to 30 amino acid units, or from 5 to 25 amino acid units, or from 5 to 20 amino acid units.
  • the peptide moiety is an anticancer peptide moiety.
  • anticancer peptide moiety refers to a moiety of a peptide compound that has anticancer activity, either directly as a cytotoxin, or indirectly, for example, as an immunotherapy agent.
  • the peptide moiety is an anticancer peptide moiety, which is a moiety of a peptide compound selected from the group consisting of: leuteinizing hormone-releasing hormone (LHRH) agonists, LHRH antagonists, tumor-associated antigens, angiogenesis inhibitors (such as VEGF modulators), gastric-releasing peptide receptor (GPRP) antagonists, monoclonal antibodies, and immunotherapy agents, such as checkpoint inhibitors.
  • LHRH leuteinizing hormone-releasing hormone
  • LHRH antagonists LHRH antagonists
  • tumor-associated antigens such as VEGF modulators
  • angiogenesis inhibitors such as VEGF modulators
  • GPRP gastric-releasing peptide receptor
  • monoclonal antibodies such as checkpoint inhibitors.
  • nl is an integer
  • n2 is an integer from 13 to 25
  • n3 is an integer from 1 to 25.
  • -X 2 -X 1 -A 1 is selected from the group consisting of:
  • -X 2 -X 1 -A 1 is selected from the group consisting of:
  • nl is an integer from 12 to 24.
  • nl is an integer from 14 to
  • n2 is an integer from 15 to 23, or from 17 to 21. In some embodiments of any of the
  • n3 is an integer from 1 to 15, or from 1 to 10, or from 1 to 6.
  • nl is an integer from 14 to 22, or from 16 to 20.
  • n2 is an integer from 15 to 23, or from 17 to 21.
  • n3 is an integer from 1 to 15, or from 1 to 10, or from 1 to 6. . In some such embodiments,
  • -X ⁇ X ⁇ A 1 is -0-(CH 2 ) n3 -OH, where n3 is an integer from 14 to 26, or an integer from 16 to 24, or an integer from 18 to 22.
  • pharmaceutically acceptable salts refers to salts of the compounds which are not biologically or otherwise undesirable and are generally prepared by reacting the free base with a suitable organic or inorganic acid or by reacting the acid with a suitable organic or inorganic base.
  • Representative salts include the following salts: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate,
  • an acidic substituent such as -COOH
  • an acidic substituent such as -COOH
  • ammonium, morpholinium, sodium, potassium, barium, calcium salt, and the like for use as the dosage form.
  • a basic group such as amino or a basic heteroaryl radical, such as pyridyl
  • an acidic salt such as hydrochloride, hydrobromide, phosphate, sulfate, trifluoroacetate, trichloroacetate, acetate, oxalate, maleate, pyruvate, malonate, succinate, citrate, tartarate, fumarate, mandelate, benzoate, cinnamate, methanesulfonate, ethanesulfonate, picrate, and the like.
  • the compounds of the foregoing embodiments are useful as anticancer agents or prodrugs thereof, and are therefore useful as compounds for the treatment of cancer.
  • Table 3 shows various examples of compounds that are contemplated by the present disclosure. Table 3 refers to various combinations of an A 2 - moiety with a
  • Table 1 shows illustrative example moieties for the A 2 - moiety, wherein A 2 can be the moiety shown or can also be a pharmaceutically acceptable salt thereof.
  • Table 2 shows illustrative example moieties for -X 2 -X 1 -A 1 .
  • Table 3 shows non-limiting illustrative combinations of the moieties from Tables 1 and 2, which can come together to form compounds of the present disclosure.
  • the compounds disclosed in Table 3 can be made by methods analogous to those illustrated in the Examples, and by common synthetic methods known to those of ordinary skill in the art.
  • the compounds of any of the preceding embodiments may be formulated into pharmaceutical compositions in any suitable manner.
  • such pharmaceutical formulations are aqueous formulations suitable for parenteral administration, such as intravenous or intra-arterial administration.
  • compositions that include one or more compounds of formula (I) (according to any of the foregoing
  • the protein is an albumin or an albumin mimetic.
  • the protein is human serum albumin (HSA) or a mimetic thereof, i.e., a protein whose sequence is at least 50% equivalent to that of HSA, or at least 60% equivalent to that of HSA, or at least 70% equivalent to that of HSA, or at least 80% equivalent to that of HSA, or at least 90% equivalent to that of HSA, or at least 95% equivalent to that of HSA, at least 97% equivalent to that of HSA, at least 99% equivalent to that of HSA.
  • the protein is human serum albumin.
  • the pharmaceutical composition also includes a carrier, such as a liquid carrier.
  • the carrier includes water.
  • water makes up at least 50% by volume, or at least 60% by volume, or at least 70% by volume, or at least 80% by volume, or at least 90% by volume, based on the total volume of liquid materials in the pharmaceutical composition.
  • the carrier can also include other liquid ingredients, such as liquid ingredients commonly included in aqueous pharmaceutical formulations for parenteral administration.
  • the compounds of formula (I) bind non-covalently to the protein in the pharmaceutical formulation.
  • the compound of formula (I) and the protein e.g., human serum albumin
  • Kb binding constant
  • the compound of formula (I) and the protein are solvated by the carrier.
  • at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 98% by weight, or at least 99% by weight of the compounds of formula (I) in the composition are bound non-covalently to the protein with a binding constant (Kb) of at least 10 2 M "1 , or at least 10 3 M "1 , or at least 10 4 M “1 , or at least 10 5 M "1 at 25 °C in the aqueous composition.
  • the composition is substantially free of agglomerates or nanoparticles.
  • no more than 5% by weight, or no more than 4% by weight, or no more than 3% by weight, or no more than 2% by weight, or no more than 1% by weight of the protein-compound (i.e., non-covalently bound conjugates between the protein and one or more compounds of formula (I)) in the aqueous composition have a radius greater than 7 nm, or a radius greater than 5 nm, or a radius greater than 4 nm, as measured by dynamic light scattering.
  • the compound of formula (I) can have any suitable molar ratio to the protein in the formulation.
  • the molar ratio of the compound of formula (I) to the protein ranges from 1 : 10 to 20: 1, or from 1 :5 to 15: 1, or from 1 :2 to 10: 1.
  • the molar ratio of the compound of formula (I) to the protein is about 1 : 1, or is about 2: 1, or is about 3: 1, or is about 4: 1, or is about 5: 1, or is about 6: 1, or is about 7: 1, wherein the term "about,” in this instance means ⁇ 0.5: 1, such that "about 5: 1" refers to a range from 4.5: 1 to 5.5: 1.
  • compositions that include: a compound, which comprises a peptide moiety and a protein binding moiety; a protein, wherein the protein is an albumin or an albumin mimetic; and a carrier, which comprises water.
  • the protein is human serum albumin (HSA) or a mimetic thereof, i.e., a protein whose sequence is at least 50% equivalent to that of HSA, or at least 60% equivalent to that of HSA, or at least 70% equivalent to that of HSA, or at least 80% equivalent to that of HSA, or at least 90% equivalent to that of HSA, or at least 95% equivalent to that of HSA, at least 97% equivalent to that of HSA, at least 99% equivalent to that of HSA.
  • HSA human serum albumin
  • the protein is human serum albumin.
  • the carrier includes water.
  • water makes up at least 50% by volume, or at least 60% by volume, or at least 70% by volume, or at least 80% by volume, or at least 90% by volume, based on the total volume of liquid materials in the pharmaceutical composition.
  • the carrier can also include other liquid ingredients, such as liquid ingredients commonly included in aqueous pharmaceutical formulations for parenteral administration.
  • the compounds bind non-covalently to the protein in the pharmaceutical formulation.
  • the compound and the protein e.g., human serum albumin
  • Kb binding constant
  • the compound and the protein are solvated by the carrier.
  • at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 98% by weight, or at least 99% by weight of the compounds of formula (I) in the composition are bound non-covalently to the protein with a binding constant (Kb) of at least 10 2 M "1 , or at least 10 3 M "1 , or at least 10 4 M “1 , or at least 10 5 M "1 at 25 °C in the aqueous composition.
  • the composition is substantially free of agglomerates or nanoparticles.
  • no more than 5% by weight, or no more than 4% by weight, or no more than 3% by weight, or no more than 2% by weight, or no more than 1% by weight of the protein-compound (i.e., non-covalently bound conjugates between the protein and one or more compounds of formula (I)) in the aqueous composition have a radius greater than 7 nm, or a radius greater than 5 nm, or a radius greater than 4 nm, as measured by dynamic light scattering.
  • the compound of formula (I) can have any suitable molar ratio to the protein in the formulation.
  • the molar ratio of the compound of formula (I) to the protein ranges from 1 : 10 to 20: 1, or from 1 :5 to 15: 1, or from 1 :2 to 10: 1.
  • the molar ratio of the compound of formula (I) to the protein is about 1 : 1, or is about 2: 1, or is about 3: 1, or is about 4: 1, or is about 5: 1, or is about 6: 1, or is about 7: 1, wherein the term "about,” in this instance means ⁇ 0.5: 1, such that "about 5: 1" refers to a range from 4.5: 1 to 5.5: 1.
  • compositions of any of the foregoing aspects and embodiments can also include certain additional ingredients, such as those commonly employed in pharmaceutical compositions for parenteral administration.
  • the compounds or compositions of any of the foregoing embodiments are useful in the treatment of cancer and related disorders. Therefore, these compounds and compositions can be used for administration to a subject who has or has had a cancerous tumor.
  • the disclosure provides methods of treating cancer, including administering to a subject a compound or composition of any of the foregoing aspects and embodiments.
  • the subject is a human.
  • the subject is a subject in need of such treatment, e.g., a human in need of such treatment.
  • the disclosure provides methods of inducing apoptosis in a cancer cell, including contacting the cancer cell with a compound or composition of any of the foregoing aspects and embodiments.
  • the disclosure provides methods of inhibiting proliferation of a cancerous tumor, including contacting the cancerous tumor with a compound or composition of any of the foregoing aspects and embodiments.
  • the disclosure provides uses of a compound or composition of any of the foregoing aspects and embodiments as a medicament.
  • the disclosure provides uses of a compound or composition of any of the foregoing aspects and embodiments for treating cancer.
  • the disclosure provides uses of a compound of any of the foregoing aspects and embodiments in the manufacture of a medicament.
  • the disclosure provides uses of a compound of any of the foregoing aspects and embodiments in the manufacture of a medicament for treating cancer.
  • the compounds or compositions of any of the foregoing embodiments are useful when used in conjunction with immunotherapy agents, such as checkpoint inhibitors, toll like receptor modulators, and various antibodies, including, but not limited to, alemtuzumab, atezolizumab, ipilimumab, ofatumumab, nivolumab, pembrolizumab, and rituximab.
  • immunotherapy agents such as checkpoint inhibitors, toll like receptor modulators, and various antibodies, including, but not limited to, alemtuzumab, atezolizumab, ipilimumab, ofatumumab, nivolumab, pembrolizumab, and rituximab.
  • LRMS Liquid chromatography / low-resolution mass spectrometry
  • EDC l-Ethyl-3-(3-dimethylaminopropyl)carbodiirnide
  • DIPEA N,N-diisopropylethylamine
  • HBTU 2-(lH-benzotriazol-l-yl)-l,l,3,3-tetramethyluronium
  • HSA Human serum albumin
  • Peptides were synthesized using standard solid phase synthesis procedures on an automatic peptide synthesizer using 1 g Rink resin with a loading of 0.68 mmol/g.
  • the terminal Fmoc group was removed by treating 500 mg resin with 20% 4-methyl piperidine in DMF (10 mL) for 10 min, the solvent was removed and the resin treated with another 10 mL of 20% 4-methyl piperidine in DMF for 5 min. The solvent was removed and the resin washed with DMF (25 mL x5).

Abstract

The present disclosure generally provides compounds useful for treating cancer. In some aspects, the disclosure provides containing compounds that are chemically modified to have one or more moieties that include hydrophobic portions. In some aspects, the disclosure provides compositions that include such modified peptides and another protein, such as albumin or albumin mimetics. Further, the disclosure provides various uses of these compounds and compositions.

Description

MODIFIED PEPTIDES AND USES THEREOF FOR TREATING CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
The present application claims the benefit of priority to United States Provisional Application No. 62/475, 175, filed March 22, 2017, which is hereby incorporated by reference as though set forth herein in its entirety.
TECHNICAL FIELD
The present disclosure generally provides compounds useful for treating cancer. In some aspects, the disclosure provides containing compounds that are chemically modified to have one or more moieties that include hydrophobic portions. In some aspects, the disclosure provides compositions that include such modified peptides and another protein, such as albumin or albumin mimetics. Further, the disclosure provides various uses of these compounds and compositions.
DESCRIPTION OF RELATED ART
Cancer refers to a group of diseases characterized by the formation of malignant tumors or neoplasms, which involve abnormal cell growth and have the potential to invade adjacent tissue and spread to other parts of the body. There are more than 14 million new diagnoses of cancer annually. Moreover, cancer accounts for more than 8 million deaths each year, which is about 15% of all deaths worldwide. In developed countries, cancer accounts for an even higher percentage of deaths.
Therapies for cancer have improved significantly over the years. Traditionally, cytotoxic agents have been used to treat cancer. And while these agents have generally afforded effective means of treating cancer, they often exhibit off-target activity towards other cells in the body. Therefore, patients undergoing such treatment often suffer from a number of side-effects. Peptide-based therapies for cancer offer the possibility of creating molecules that are specifically designed to act selectively against certain cancerous cells without having a significant effect on other non-cancerous cells. This is especially true as researchers develop an increasingly detailed understanding of the specific genetic makeup of certain cancers. Such compounds come in a wide variety of different classes. Some peptide- based drugs act in ways that are similar to traditional cytotoxins. Others have an indirect effect, and operate by stimulating the immune system to develop its own defenses against the cancerous cells. Even so, developing effective peptide-based therapies presents a number of difficulties. One of the chief difficulties relates to improving the half-life of the peptide compound long enough to function effectively to treat the cancer, either directly or indirectly. Because the human body contains a number of enzymes designed to break down peptides into their constituent amino acids, such peptides are often "chewed up" by the body before they have an opportunity to be effective. Such compounds may also have difficulty finding their way to the cancer cell. This is especially true if the cancer cells in question do not overexpress a surface protein that has an affinity for the peptide. Antibody-drug conjugates (ADCs) have provided a partial solution to this problem, wherein the toxic pay load is conjugated to an antibody that selectively binds to antigens that are overexpressed on the surface of certain cancer cells. But such a strategy is often not easily generalized to from one anti-cancer agent to the next. Thus, despite the immense promise of peptide-based therapies for treating cancer, there ae still very few such drugs.
Therefore, there is a continuing need to discover new ways of selectively directing peptide-based cancer drugs to cancer cells and tumors, so as to reduce off-target effects and to improve their pharmacokinetic profile.
SUMMARY
The present disclosure provides compounds and compositions that can deliver increasingly high quantities of peptide-based cancer therapies to cancer cells (e.g., in a solid tumor) with reduced off-target side effects and improved pharmacokinetic lifetime. In some embodiments, the compounds are prodrugs of peptide compounds, such that the prodrug permits improved delivery of the peptide compound to a solid tumor in a mammal. The disclosure also provides methods and uses of those compounds and compositions for the treatment of cancer.
In a first aspect, the disclosure provides compounds of formula (I):
A1 X1 X2 A2 (I) wherein: A1 is an organic group, a hydrophilic group, or a hydrogen atom; A2 is a peptide moiety; X1 is a hydrophobic group; and X2 is a direct bond, an organic group, or a heteroatom group selected from the group consisting of -0-, -S-, -S(=0)-, -S(=0)2-, -S-S-, -N=, =N-, -N(H)-, -N=N-N(H)-, -N(H)-N=N-, -N(OH)-, or -N(=0)-. In some embodiments, A1 is a hydrophilic group, such as a carboxylic acid group (-COOH) or a pharmaceutically acceptable salt thereof. In some embodiments, the peptide moiety is an anticancer peptide moiety. In some embodiments, the hydrophobic group is a C 12-22 hydrocarbylene group, which is optionally substituted. In some embodiments, X2 is an organic group, such as a carbonyl group, i.e., -C(=0)-.
In a second aspect, the disclosure provides compositions (e.g., pharmaceutical compositions) that include: a compound of any embodiments of the first aspect; and a protein. In some embodiments, the protein is an albumin or an albumin mimetic.
In a third aspect, the disclosure provides compositions (e.g., pharmaceutical compositions) that include: a compound of any embodiments of the first aspect; a protein, wherein the protein is an albumin or an albumin mimetic; and a carrier, which includes water; wherein the compound and the protein are non-covalently associated with each other; and wherein the compound and the protein are solvated by the carrier.
In a fourth aspect, the disclosure provides methods of treating cancer, which include administering to a subject a compound or composition of any embodiments of any of the foregoing aspects. In some further embodiments thereof, the disclosure provides methods of treating cancer that include administering to a subject one or more immunotherapy agents.
In a fifth aspect, the disclosure provides methods of inducing apoptosis in a cancer cell, which include contacting the cancer cell with a compound or composition of any embodiments of any of the first through the third aspects. In some further embodiments thereof, the disclosure provides methods of inducing apoptosis in a cancer cell that include contacting the cancer cell with one or more immunotherapy agents.
In a sixth aspect, the disclosure provides methods for inhibiting growth of a cancerous tumor, which includes contacting the cancerous tumor with a compound of any embodiments of the first aspect. In some further embodiments thereof, the disclosure provides methods of inhibiting growth of a cancerous tumor that include contacting the cancerous tumor with one or more immunotherapy agents.
In a seventh aspect, the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects as a medicament.
In a eighth aspect, the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects for treating cancer. In some further embodiments thereof, the disclosure provides uses that include use in combination with one or more immunotherapy agents.
In a ninth aspect, the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects in the manufacture of a medicament. In a tenth aspect, the disclosure provides uses of a compound or composition of any embodiments of any of the first through the third aspects in the manufacture of a medicament for treating cancer.
In a eleventh aspect, the disclosure provides methods of making compounds of the first and second aspects and compositions of the third and fourth aspects.
Further aspects and embodiments are provided in the drawings, the detailed description, the claims, and the abstract.
BRIEF DESCRIPTION OF DRAWINGS
The following drawings are provided for purposes of illustrating various embodiments of the compounds, compositions, methods, and uses disclosed herein. The drawings are provided for illustrative purposes only, and are not intended to describe any preferred compounds or compositions or any preferred methods or uses, or to serve as a source of any limitations on the scope of the claimed inventions.
FIG. 1 shows a non-limiting example of a compound of formula (I), where the compound includes a peptide moiety, which is modified to include a long-chain dibasic acid moiety.
DETAILED DESCRIPTION
The following description recites various aspects and embodiments of the inventions disclosed herein. No particular embodiment is intended to define the scope of the invention. Rather, the embodiments provide non-limiting examples of various compositions, and methods that are included within the scope of the claimed inventions. The description is to be read from the perspective of one of ordinary skill in the art. Therefore, information that is well known to the ordinarily skilled artisan is not necessarily included.
Definitions
The following terms and phrases have the meanings indicated below, unless otherwise provided herein. This disclosure may employ other terms and phrases not expressly defined herein. Such other terms and phrases shall have the meanings that they would possess within the context of this disclosure to those of ordinary skill in the art. In some instances, a term or phrase may be defined in the singular or plural. In such instances, it is understood that any term in the singular may include its plural counterpart and vice versa, unless expressly indicated to the contrary. As used herein, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. For example, reference to "a substituent" encompasses a single substituent as well as two or more substituents, and the like.
As used herein, "for example," "for instance," "such as," or "including" are meant to introduce examples that further clarify more general subject matter. Unless otherwise expressly indicated, such examples are provided only as an aid for understanding
embodiments illustrated in the present disclosure, and are not meant to be limiting in any fashion. Nor do these phrases indicate any kind of preference for the disclosed embodiment.
As used herein, "hydrocarbon" refers to an organic group composed of carbon and hydrogen, which can be saturated or unsaturated, and can include aromatic groups. The term "hydrocarbyl" refers to a monovalent or polyvalent (e.g., divalent or higher) hydrocarbon moiety. In some cases, a divalent hydrocarbyl group is referred to as a "hydrocarbylene" group.
As used herein, "alkyl" refers to a straight or branched chain saturated hydrocarbon having 1 to 30 carbon atoms, which may be optionally substituted, as herein further described, with multiple degrees of substitution being allowed. Examples of "alkyl," as used herein, include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, isobutyl, n-butyl, sec-butyl, tert-butyl, isopentyl, n-pentyl, neopentyl, n-hexyl, and 2-ethylhexyl. In some instances, the "alkyl" group can be divalent, in which case, the group can alternatively be referred to as an "alkylene" group. Also, in some instances, one or more of the carbon atoms in the alkyl or alkylene group can be replaced by a heteroatom (e.g., selected from nitrogen, oxygen, or sulfur, including N-oxides, sulfur oxides, sulfur dioxides, and carbonyl groups, where feasible), and is referred to as a "heteroalkyl" or "heteroalkylene" group, respectively. Non-limiting examples include "oxyalkyl" or "oxyalkylene" groups, which refer to groups where a carbon atom in the alkyl or alkylene group is replaced by oxygen. Non-limiting examples of oxyalkyl or oxyalkylene groups include alkyl or alkylene chains that contain a carbonyl group, and also alkoxylates, polyalkylene oxides, and the like.
The number of carbon atoms in any group or compound can be represented by the terms. Thus, "Cz" refers to a group of compound having z carbon atoms, and "Cx-y", refers to a group or compound containing from x to y, inclusive, carbon atoms. For example, "Ci-6 alkyl" represents an alkyl group having from 1 to 6 carbon atoms and, for example, includes, but is not limited to, methyl, ethyl, n-propyl, isopropyl, isobutyl, n-butyl, sec-butyl, tert-butyl, isopentyl, n-pentyl, neopentyl, and n-hexyl. The same logic applies to other types of functional groups, defined below. As used herein, "alkenyl" refers to a straight or branched chain non-aromatic hydrocarbon having 2 to 30 carbon atoms and having one or more carbon-carbon double bonds, which may be optionally substituted, as herein further described, with multiple degrees of substitution being allowed. Examples of "alkenyl," as used herein, include, but are not limited to, ethenyl, 2-propenyl, 2-butenyl, and 3-butenyl. In some instances, the
"alkenyl" group can be divalent, in which case the group can altematively be referred to as an "alkenylene" group. Also, in some instances, one or more of the carbon atoms in the alkenyl or alkenylene group can be replaced by a heteroatom (e.g., selected from nitrogen, oxygen, or sulfur, including N-oxides, sulfur oxides, sulfur dioxides, and carbonyl groups, where feasible), and is referred to as a "heteroalkenyl" or "heteroalkenylene" group, respectively.
As used herein, "cycloalkyl" refers to an aliphatic saturated or unsaturated hydrocarbon ring system having 3 to 20 carbon atoms, which may be optionally substituted, as herein further described, with multiple degrees of substitution being allowed. In some embodiments, the term refers only to saturated hydrocarbon ring systems, substituted as herein further described. Examples of "cycloalkyl," as used herein, include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, adamantyl, and the like. In some instances, the "cycloalkyl" group can be divalent, in which case the group can alternatively be referred to as a "cycloalkylene" group. Cycloalkyl and cycloalkylene groups can also be referred to herein as "carbocyclic rings." Also, in some instances, one or more of the carbon atoms in the cycloalkyl or cycloalkylene group can be replaced by a heteroatom (e.g., selected independently from nitrogen, oxygen, silicon, or sulfur, including N-oxides, sulfur oxides, and sulfur dioxides, where feasible), and is referred to as a "heterocyclyl" or "heterocyclylene" group, respectively. The term
"heterocyclic ring" can also be used interchangeably with either of these terms. In some embodiments, the cycloalkyl and heterocyclyl groups are fully saturated. In some other embodiments, the cycloalkyl and heterocyclyl groups can contain one or more carbon-carbon double bonds.
As used herein, "halogen," "halogen atom," or "halo" refer to a fluorine, chlorine, bromine, or iodine atom. In some embodiments, the terms refer to a fluorine or chlorine atom.
As used herein, the terms "organic group," "organic moiety," or "organic residue" refer to a monovalent or polyvalent functional group having at least one carbon atom, which optionally contains one or more additional atoms selected from the group consisting of hydrogen atoms, halogen atoms, nitrogen atoms, oxygen atoms, phosphorus atoms, and sulfur atoms, and which does not include covalently bound metal or semi-metal atoms. In some embodiments, these terms can include metal salts of organic groups, such as alkali metal or alkaline earth metal salts of organic anions.
As used herein, the term "pharmacophore" refers to a type of organic functional group. Standard pharmacophores are hydrophobic pharmacophores, hydrogen-bond donating pharmacophores, hydrogen-bond accepting pharmacophores, positive ionizable
pharmacophores, and negative ionizable pharmacophores. The classification of organic functional groups within a compound is carried out according to standard classification systems known in the art.
As used herein, the terms "hydrophobic group," "hydrophobic moiety," or
"hydrophobic residue" refer to an organic group that consists essentially of hydrophobic pharmacophores. In some embodiments, the terms refer to an organic group that consists of hydrophobic pharmacophores.
As used herein, the terms "hydrophilic group," "hydrophilic moiety," or "hydrophilic residue" refer to an organic group that comprises one pharmacophores selected from the group consisting of hydrogen bond donors, hydrogen bond acceptors, negative ionizable groups, or positive ionizable groups. In some embodiments, the terms refer to an organic group that consist essentially of pharmacophores selected from the group consisting of hydrogen bond donors, hydrogen bond acceptors, negative ionizable groups, or positive ionizable groups.
As used herein, the term "peptide moiety" refers to a peptide compound, or a pharmaceutically acceptable salt thereof, where an atom or a group of atoms is absent, thereby creating a monovalent or polyvalent moiety. As used herein, the term "peptide compound" refers to a compound formed from the condensation of two or more amino acid compounds. Any suitable amino acid compounds can be used, including L-amino acids or D-amino acids. In addition, the amino acids can be alpha-amino acids, beta-amino acids, gamma-amino acids, and delta-amino acids. In some embodiments, the amino acids are L-alpha-amino acids. In some embodiments, at least 80% by number, or at least 85% by number, or at least 90% by number, or at least 95% by number of the amino acids forming the peptide moiety are selected from the 22 proteinogenic amino acids. Conjugates can be formed in any suitable way. In some embodiments, for example, a hydrogen atom is absent from the N-terminal end of the peptide compound, thereby creating a monovalent moiety. A non-limiting example of such a "peptide moiety," is the moiety of the following formula:
Figure imgf000010_0001
where a hydrogen atom is absent to create a monovalent moiety that, within a compound, bonds to the rest of the molecule through the remaining nitrogen atom, and wherein Rk, Rm, and Rn are amino acid substituents. Note that the term "peptide moiety" is not limited to any particular procedure for making such compounds or moieties.
Various methods of drawing chemical structures are used herein. In some instances, the bond line-structure method is used to depict chemical compounds or moieties. In the line- structure method, the lines represent chemical bonds, and the carbon atoms are not explicitly shown (but are implied by the intersection of the lines). The hydrogen atoms are also not explicitly shown, except in instances where they are attached to heteroatoms. Heteroatoms, however, are explicitly shown. Thus, using that methodology, the structures shown below are for 2-methylpropane, 1 -methoxypropane, and 1 -propanol:
Figure imgf000010_0002
In that methodology, aromatic rings are typically represented merely by one of the contributing resonance structures. Thus, the following structures are for benzene, pyridine,
Figure imgf000010_0003
As used herein, a "protein binding moiety" is a moiety that binds non-covalently to one or more sites on a protein with a binding constant (Kb) of at least 100 M"1 in water at 25 °C.
As used herein, "amino acid" refers to a compound having the structure
H2N-Rx-COOH, where Rx is an organic group, and where the NH2 may optionally combine with Rx (e.g., as in the case of proline). The term includes any known amino acids, including, but not limited to, alpha amino acids, beta amino acids, gamma amino acids, delta amino acids, and the like. In some embodiments, the term can refer to alpha amino acids. As used herein, "hydroxy acid" refers to a compound having the structure
HO-Ry-COOH, where Ry is an organic group. Non-limiting examples include gly colic acid, lactic acid, and caprolactone.
As used herein, "alkanol amine" refers to a compound having the structure
HO-Rz-NH2, where Rz is an optionally substituted alkylene group. Non-limiting examples include ethanol amine.
As used herein, "administer" or "administering" means to introduce, such as to introduce to a subject a compound or composition. The term is not limited to any specific mode of delivery, and can include, for example, subcutaneous delivery, intravenous delivery, intramuscular delivery, intracisternal delivery, delivery by infusion techniques, transdermal delivery, oral delivery, nasal delivery, and rectal delivery. Furthermore, depending on the mode of delivery, the administering can be carried out by various individuals, including, for example, a health-care professional (e.g., physician, nurse, etc.), a pharmacist, or the subj ect (i.e., self-administration).
As used herein, "treat" or "treating" or "treatment" can refer to one or more of:
delaying the progress of a disease, disorder, or condition; controlling a disease, disorder, or condition; ameliorating one or more symptoms characteristic of a disease, disorder, or condition; or delaying the recurrence of a disease, disorder, or condition, or characteristic symptoms thereof, depending on the nature of the disease, disorder, or condition and its characteristic symptoms.
As used herein, "subject" refers to any mammal such as, but not limited to, humans, horses, cows, sheep, pigs, mice, rats, dogs, cats, and primates such as chimpanzees, gorillas, and rhesus monkeys. In some embodiments, the "subject" is a human. In some such embodiments, the "subject" is a human who exhibits one or more symptoms characteristic of a disease, disorder, or condition. The term "subject" does not require one to have any particular status with respect to a hospital, clinic, or research facility (e.g., as an admitted patient, a study participant, or the like).
As used herein, the term "compound" includes free acids, free bases, and salts thereof.
As used herein, the term "pharmaceutical composition" is used to denote a composition that may be administered to a mammalian host, e.g., orally, topically, parenterally, by inhalation spray, or rectally, in unit dosage formulations containing conventional non-toxic carriers, diluents, adjuvants, vehicles and the like. The term
"parenteral" as used herein, includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or by infusion techniques. Also included within the scope of the disclosure are the individual enantiomers of the compounds represented by Formula (I) or pharmaceutically acceptable salts thereof, as well as any wholly or partially racemic mixtures thereof. The disclosure also covers the individual enantiomers of the compounds represented by Formula (I) or pharmaceutically acceptable salts thereof, as well as mixtures with diastereoisomers thereof in which one or more stereocenters are inverted. 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 structure, except for the replacement of a hydrogen atom by a deuterium or tritium, or the replacement of a carbon atom by a 1 C- or 14C-enriched carbon are within the scope of the disclosure.
As used herein, "mix" or "mixed" or "mixture" refers broadly to any combining of two or more compositions. The two or more compositions need not have the same physical state; thus, solids can be "mixed" with liquids, e.g., to form a slurry, suspension, or solution. Further, these terms do not require any degree of homogeneity or uniformity of composition. This, such "mixtures" can be homogeneous or heterogeneous, or can be uniform or nonuniform. Further, the terms do not require the use of any particular equipment to carry out the mixing, such as an industrial mixer.
As used herein, "optionally" means that the subsequently described event(s) may or may not occur. In some embodiments, the optional event does not occur. In some other embodiments, the optional event does occur one or more times.
As used herein, "substituted" refers to substitution of one or more hydrogen atoms of the designated moiety with the named substituent or substituents, multiple degrees of substitution being allowed unless otherwise stated, provided that the substitution results in a stable or chemically feasible compound. A stable compound or chemically feasible compound is one in which the chemical structure is not substantially altered when kept at a temperature from about -80 °C to about +40 °C, in the absence of moisture or other chemically reactive conditions, for at least a week. As used herein, the phrases "substituted with one or more... " or "substituted one or more times ... " refer to a number of substituents that equals from one to the maximum number of substituents possible based on the number of available bonding sites, provided that the above conditions of stability and chemical feasibility are met.
As used herein, "comprise" or "comprises" or "comprising" or "comprised of refer to groups that are open, meaning that the group can include additional members in addition to those expressly recited. For example, the phrase, "comprises A" means that A must be present, but that other members can be present too. The terms "include," "have," and "composed of and their grammatical variants have the same meaning. In contrast, "consist of or "consists of or "consisting of refer to groups that are closed. For example, the phrase "consists of A" means that A and only A is present. As used herein, the phrases "consist essentially of," "consists essentially of," and "consisting essentially of refer to groups that are open, but which only includes additional unnamed members that would not materially affect the basic characteristics of the claimed subject matter.
As used herein, "or" is to be given its broadest reasonable interpretation, and is not to be limited to an either/or construction. Thus, the phrase "comprising A or B" means that A can be present and not B, or that B is present and not A, or that A and B are both present. Further, if A, for example, defines a class that can have multiple members, e.g., Ai and A2, then one or more members of the class can be present concurrently.
As used herein, the various functional groups represented will be understood to have a point of attachment at the functional group having the hyphen or dash (-) or a dash used in combination with an asterisk (*). In other words, in the case of -CH2CH2CH3 or
♦-CFhCFhCFb, it will be understood that the point of attachment is the CH2 group at the far left. If a group is recited without an asterisk or a dash, then the attachment point is indicated by the plain and ordinary meaning of the recited group.
As used herein, multi-atom bivalent species are to be read from left to right. For example, if the specification or claims recite A-D-E and D is defined as -OC(O)-, the resulting group with D replaced is: A-OC(0)-E and not A-C(0)0-E.
Other terms are defined in other portions of this description, even though not included in this subsection.
Modified Peptide Compounds
In at least one aspect, the disclosure provides compounds of formula (I):
A1 X1 X2 A2 (I) wherein: A1 is a hydrophilic group a hydrogen atom, or an organic group; A2 is a peptide moiety; X1 is a hydrophobic group; and X2 is a direct bond, an organic group, or a group selected from the group consisting of -0-, -S-, -S(=0)-, -S(=0)2-, -S-S-, -N=, =N-, -N(H)-, -N=N-N(H)-, -N(H)-N=N-, -N(OH)-, or -N(=0)-.
In some embodiments, A1 is an organic group. A1 can contain any suitable number of carbon atoms. In some embodiments, for example, A1 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms, or from 1 to 10 carbon atoms, or from 1 to 6 carbon atoms. A1 can also contain one or more heteroatoms, such as nitrogen, oxygen, sulfur, or phosphorus.
In some embodiments according to any of the foregoing embodiments, A1 is a hydrophilic group or moiety. Non-limiting examples of a hydrophilic group include, but are not limited to, a carboxylic acid moiety, an ester moiety, an amide moiety, a urea moiety, an amine moiety, an ether moiety, an alcohol moiety, a thioether moiety, a thiol moiety, a ketone moiety, an aldehyde moiety, a sulfate moiety, a thiosulfate moiety, a sulfite moiety, a thiosulfite moiety, a phosphate moiety, a phosphonate moiety, a phosphinate moiety, a phosphite moiety, a borate moiety, or a boronate moiety.
In some embodiments of any of the aforementioned embodiments, A1 is selected from the group consisting of a carboxylic acid group (-COOH), a carboxylate anion (-COO"), or a carboxylate ester (-COORa, where Ra is an organic group such as an alkyl or alkoxylate group). In some such embodiments, A1 is a carboxylic acid group. In some such
embodiments, A1 is a carboxylate ester group.
In some other embodiments of any of the aforementioned embodiments, A1 is a hydrogen atom. In some other embodiments of any of the aforementioned embodiments, A1 is a hydroxyl (-OH) group.
In any of the aforementioned embodiments, X1 can be a hydrophobic group having any suitable number of carbon atoms. In some embodiments, for example, X1 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms.
In some embodiments of any of the aforementioned embodiments, X1 is Cs-3o hydrocarbylene, which is optionally substituted. In some further embodiments, X1 is C 12-22 hydrocarbylene, which is optionally substituted. In some further embodiments, X1 is C 12-22 alkylene. In some further embodiments, X1 is -(CH2)i2-, -(CH2)i4-, -(CH2)i6-, -(CH2)i8-, -(CH2)2o-, or -(CH2)22-. In some other embodiments, X1 is -(CH2)i6-. In some further embodiments, X1 is C 12-22 alkenylene. In some further such embodiments, X1 is
-(CH2)7-CH=CH-(CH2)7-.
In some further embodiments of any of the aforementioned embodiments, X1 is C 12-22 hydrocarbylene, which is optionally substituted. In some such embodiments, X1 is C 12-22 hydrocarbylene. In some further such embodiments, X1 is C 14-22 hydrocarbylene. In some further such embodiments, X1 is C16-22 hydrocarbylene. In some embodiments of any of the aforementioned embodiments, X1 is C12-22 hydrocarbylene, wherein A1 and X2 (or, if X2 is a direct bond, A2) are separated from each other by at least 6, or by at least 8, or by at least 10, or by at least 12, or by at least 14, carbon atoms. In some further such embodiments, X1 is Ci4-22 hydrocarbylene, wherein A1 and X2 (or, if X2 is a direct bond, A2) are separated from each other by at least 6, or by at least 8, or by at least 10, or by at least 12, or by at least 14, carbon atoms. In some further such embodiments, X1 is Ci6-22 hydrocarbylene, wherein A1 and X2 (or, if X2 is a direct bond, A2) are separated from each other by at least 6, or by at least 8, or by at least 10, or by at least 12, or by at least 14, carbon atoms. In some further embodiments of any of the aforementioned embodiments, X1 is C12-22 straight-chain alkylene, or C 14-22 straight-chain alkylene, or C16-22 straight-chain alkylene. In some further embodiments of any of the aforementioned embodiments, X1 is C12-22 straight-chain alkenylene, or C 14-22 straight-chain alkenylene, or C16-22 straight-chain alkenylene.
In some embodiments of any of the aforementioned embodiments, X2 is a direct bond.
In some other embodiments of any of the aforementioned embodiments, X2 is an organic group. In some embodiments, X2 is a hydrophilic group. In some embodiments, X2 is a heteroalkylene group.
In any of the aforementioned embodiments where X2 is an organic group, X2 can contain any suitable number of carbon atoms. In some embodiments, for example, X2 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms, or from 1 to 10 carbon atoms, or from 1 to 6 carbon atoms.
In any of the aforementioned embodiments where X2 is a heteroalkylene group, X2 can contain any suitable number of carbon atoms. In some embodiments, for example, X2 contains from 1 to 100 carbon atoms, or from 1 to 50 carbon atoms, or from 1 to 25 carbon atoms, or from 1 to 10 carbon atoms, or from 1 to 6 carbon atoms.
In some of the aforementioned embodiments, X2 can contain certain groups. Some non-limiting examples of such groups that X2 can contain are polyalkylene oxide groups, such as polyethylene glycol (PEG) and various polypeptide chains.
In some embodiments, X2 is an organic group selected from the group consisting of
-C(=0)-, -C≡C-, -C(H)=C(H)-, -C(=0)-0-, -0-C(=0)-, -C(=0)-NH-, -NH-C(=0)-,
-NH-C(=0)-0-, -0-(C=0)-NH-, -0-C(=0)-0-, -C(=N-NH2)-, -C(=N-Rb)- (where Rb is a hydrogen atom or an alkyl group), -C(=N-OH)-, -NH-C(=0)-NH-, -NH-C(=S)-NH-, -NH-C(=S)-0-, -0-C(=S)-NH-, -NH-C(=0)-S-, -S-C(=0)-NH-,-NH-C(=S)-S-,
-S-C(=S)-NH-, and the cyclic structures shown below:
Figure imgf000016_0001
where Rc, Rd, and Re are, independently at each occurrence, a hydrogen atom or Ci-io alkyl. In some further embodiments, X2 is -C(=0)-.
In some embodiments, X2 is a group selected from the group consisting of -0-, -S-, -S(=0)-, -S(=0)2-, -S-S-, -N= =N-, -N(H)-, -N=N-N(H)-, -N(H)-N=N-, -N(OH)-, and -N(0)-.
In some embodiments, X2 comprises one or more moieties selected from the group consisting of: -C(=0)-, -0-C(=0)-, -NH-C(=0)-, one or more moieties formed from a alkylene glycols, one or more units formed from alkanol amines, one or more units formed from amino acids, and one or more units formed from hydroxyl acids. Thus, in some embodiments, X2 comprises one or more moieties formed from alkylene glycols, such as a short poly(ethylene glycol) chain having 1 to 25 ethylene glycol units. In some
embodiments, X2 comprises one or more moieties formed from amino acids, such as an oligopeptide chain having 1 to 25 amino acid units. In some embodiments, X2 comprises one or more moieties formed from hydroxy acids, such as moieties formed from gly colic acid, lactic acid, or caprolactone. In some embodiments, X2 comprises a combination of a poly(ethylene glycol) chain having 1 to 25 ethylene glycol units and an oligopeptide having 1 to 25 amino acid units, and optionally one or more units formed from hydroxy acids..
In any of the above embodiments, the selection of X2 will depend on the type of functional group through which it is linked to the peptide moiety, so as to avoid making compounds that are chemically unstable or impossible. The skilled artisan will be able to select combinations of X2 and A2 that result in chemically stable compounds, which are compounds in which the chemical structure is not substantially altered when kept at a temperature from about -80 °C to about +40 °C, in the absence of moisture or other chemically reactive conditions, for at least a week. In the above embodiments, A2 can be any suitable peptide moiety. The peptide moiety can contain any suitable number of amino acid units (i.e., units in the peptide chain formed from an amino acid). In some embodiments, the peptide moiety comprises from 2 to 100 amino acid units, or from 3 to 50 amino acid units, or from 4 to 35 amino acid units, or from 5 to 30 amino acid units, or from 5 to 25 amino acid units, or from 5 to 20 amino acid units. In some embodiments of any of the aforementioned embodiments, the peptide moiety is an anticancer peptide moiety. As used herein, the term "anticancer peptide moiety" refers to a moiety of a peptide compound that has anticancer activity, either directly as a cytotoxin, or indirectly, for example, as an immunotherapy agent.
In some embodiments of any of the aforementioned embodiments, the peptide moiety is an anticancer peptide moiety, which is a moiety of a peptide compound selected from the group consisting of: leuteinizing hormone-releasing hormone (LHRH) agonists, LHRH antagonists, tumor-associated antigens, angiogenesis inhibitors (such as VEGF modulators), gastric-releasing peptide receptor (GPRP) antagonists, monoclonal antibodies, and immunotherapy agents, such as checkpoint inhibitors.
The selection of -X^X^A1 can depend on the nature of the connection to the peptide moiety.
For example, in embodiments where the -X2-X1-A1 connects to an oxygen atom or an
NH group on the drug moiety, as is the case of conjugation to the N-terminus of the protein in Table 1 , then -X^-A1 is selected from the group consisting of: -C(=0)-(CH2)m-C(=0)-OH;
-C(=0)-(CH2)ni-C(=0)-OCH3;
Figure imgf000017_0001
alkylene)-C(=0)-0-(CH2)n2-C(=0)-OH;
alkylene)-NH-C(=0)-(CH2)ni-C(=0)-OH;
Figure imgf000017_0002
alkylene)-C(=0)-0-[(CH2)2-0-]n3(CH2)n2-C(=0)-OH;
-C(=0)-0-(CH2)n2-C(=0)-OH; and -C(=0)-NH-(CH2)n2-C(=0)-OH; wherein nl is an integer
12 to 24, n2 is an integer from 13 to 25, and n3 is an integer from 1 to 25. In some further such embodiments, -X2-X1-A1 is selected from the group consisting of:
-C(=0)-(CH2)ni-C(=0)-OH; -C(=0)-(CH2)ni-C(=0)-OCH3;
alkylene)-C(=0)-0-(CH2)n2-C(=0)-OH;
alkylene)-NH-C(=0)-(CH2)ni-C(=0)-OH;
Figure imgf000017_0003
alkylene)-C(=0)-0-[(CH2)2-0-]n3(CH2)n2-C(=0)-OH;
-C(=0)-0-(CH2)n2-C(=0)-OH; and -C(=0)-NH-(CH2)n2-C(=0)-OH. In some further such embodiments, -X2-X1-A1 is selected from the group consisting of:
-C(=0)-(CH2)ni-C(=0)-OH; -C(=0)-0-(CH2)n2-C(=0)-OH; and -C(=0)-NH-(CH2)n2-C(=0)-OH. In some other embodiments, -X^-A1 is
-C(=0)-(Ci-6 alkylene)-0-C(=0)-(CH2)m-C(=0)-OH, where nl is an integer from 12 to 24.
In some embodiments of any of the aforementioned embodiments, nl is an integer from 14 to
22, or from 16 to 20. In some embodiments of any of the aforementioned embodiments, n2 is an integer from 15 to 23, or from 17 to 21. In some embodiments of any of the
aforementioned embodiments, n3 is an integer from 1 to 15, or from 1 to 10, or from 1 to 6.
In some such embodiments,
Figure imgf000018_0001
alkylene)-C(=0)-0-(CH2)n3-OH, where n3 is an integer from 14 to 26, or an integer from 16 to 24, or an integer from 18 to 22.
In embodiments where the -X^X^A1 connects to a -C(=0) group on the drug moiety, such as on the C-terminus of the peptide moieties recited in Table 1, then -X^X^A1 is selected from the group consisting of: -0-(CH2)n2-C(=0)-OH; -NH-(CH2)n2-C(=0)-OH;
-NH-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OH;
-0-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OH;
-NH-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OCH3;
-0-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OCH3;
-NH-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-CH3; -0-(Ci-e alkylene)-0-C(=0)-(CH2)ni-CH3;
-NH-(Ci-6 alkylene)-C(=0)-0-[(CH2)2-0-]n3(CH2)n2-C(=0)-OH; and
-0-(Ci-6 alkylene)-C(=0)-0-[(CH2)2-0-]n3(CH2)n2-C(=0)-OH; wherein nl is an integer 12 to 24, n2 is an integer from 13 to 25, and n3 is an integer from 1 to 25. In some further such embodiments, -X^X^A1 is selected from the group consisting of: -0-(CH2)n2-C(=0)-OH; -NH-(CH2)n2-C(=0)-OH; -NH-(Ci-e alkylene)-0-C(=0)-(CH2)ni-C(=0)-OH;
-0-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OH;
-NH-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OCH3; and
-0-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OCH3. In some further such embodiments, -X^X!-A1 is selected from the group consisting of: -0-(CH2)n2-C(=0)-OH;
-NH-(CH2)n2-C(=0)-OH; -NH-(Ci-e alkylene)-0-C(=0)-(CH2)ni-C(=0)-OH; and
-0-(Ci-6 alkylene)-0-C(=0)-(CH2)ni-C(=0)-OH. In some embodiments of any of the aforementioned embodiments, nl is an integer from 14 to 22, or from 16 to 20. In some embodiments of any of the aforementioned embodiments, n2 is an integer from 15 to 23, or from 17 to 21. In some embodiments of any of the aforementioned embodiments, n3 is an integer from 1 to 15, or from 1 to 10, or from 1 to 6. . In some such embodiments,
-X^X^A1 is -0-(CH2)n3-OH, where n3 is an integer from 14 to 26, or an integer from 16 to 24, or an integer from 18 to 22. The compounds described in any of the above embodiments can also exist as pharmaceutically acceptable salts. The term "pharmaceutically acceptable salts" refers to salts of the compounds which are not biologically or otherwise undesirable and are generally prepared by reacting the free base with a suitable organic or inorganic acid or by reacting the acid with a suitable organic or inorganic base. Representative salts include the following salts: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate,
methylbromide, methylnitrate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, N-methylglucamine, oxalate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, potassium, salicylate, sodium, stearate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, trimethylammonium, and valerate. When an acidic substituent is present, such as -COOH, there can be formed the ammonium, morpholinium, sodium, potassium, barium, calcium salt, and the like, for use as the dosage form. When a basic group is present, such as amino or a basic heteroaryl radical, such as pyridyl, there can be formed an acidic salt, such as hydrochloride, hydrobromide, phosphate, sulfate, trifluoroacetate, trichloroacetate, acetate, oxalate, maleate, pyruvate, malonate, succinate, citrate, tartarate, fumarate, mandelate, benzoate, cinnamate, methanesulfonate, ethanesulfonate, picrate, and the like.
The compounds above can be made by standard organic synthetic methods, such as those illustrated in: Wuts et al., Greene 's Protective Groups in Organic Synthesis (4th ed.,
2006) ; Larock, Comprehensive Organic Transformations (2nd ed., 1999); and Smith et al, March 's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure (6th ed.,
2007) ; Methods of Molecular Biology, 35, Peptide Synthesis Protocols, (M. W. Pennington and B. M. Dunn Eds), Springer, 1994; Methods of Enzymology, 289, Solid Phase Peptide Synthesis, (G. B. Fields Ed.), Academic Press, 1997; Chemical Approaches to the Synthesis of Peptides and Proteins, (P. Lloyd-Williams, F. Albericio, and E. Giralt Eds), CRC Press, 1997; Fmoc Solid Phase Peptide Synthesis, A Practical Approach, (W. C. Chan, P. D. White Eds), Oxford University Press, 2000; Solid Phase Synthesis, A Practical Guide, (S. F. Kates, F Albericio Eds), Marcel Dekker, 2000; P. Seneci, Solid-Phase Synthesis and Combinatorial Technologies, John Wiley & Sons, 2000; Synthesis of Peptides and Peptidomimetics (M. Goodman, Editor-in-chief, A. Felix, L. Moroder, C. Tmiolo Eds), Thieme, 2002; N. L. Benoiton, Chemistry of Peptide Synthesis, CRC Press, 2005. Specific non-limiting examples are shown below in the Examples.
The compounds of the foregoing embodiments, including their pharmaceutically acceptable salts, are useful as anticancer agents or prodrugs thereof, and are therefore useful as compounds for the treatment of cancer.
Table 3 (below) shows various examples of compounds that are contemplated by the present disclosure. Table 3 refers to various combinations of an A2- moiety with a
-X^X^A1, which together form compounds of the present disclosure. Table 1 shows illustrative example moieties for the A2- moiety, wherein A2 can be the moiety shown or can also be a pharmaceutically acceptable salt thereof. Table 2 shows illustrative example moieties for -X2-X1-A1. Table 3 shows non-limiting illustrative combinations of the moieties from Tables 1 and 2, which can come together to form compounds of the present disclosure. The compounds disclosed in Table 3 can be made by methods analogous to those illustrated in the Examples, and by common synthetic methods known to those of ordinary skill in the art. Suitable methods of making such compounds are illustrated in: Wuts et al., Greene 's Protective Groups in Organic Synthesis (4th ed., 2006); Larock, Comprehensive Organic Transformations (2nd ed., 1999); and Smith et al, March 's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure (6th ed., 2007); Methods of Molecular Biology, 35, Peptide Synthesis Protocols, (M. W. Pennington and B. M. Dunn Eds), Springer, 1994; Methods of Enzymology, 289, Solid Phase Peptide Synthesis, (G. B. Fields Ed.), Academic Press, 1997; Chemical Approaches to the Synthesis of Peptides and Proteins, (P. Lloyd- Williams, F. Albericio, and E. Giralt Eds), CRC Press, 1997; Fmoc Solid Phase Peptide Synthesis, A Practical Approach, (W. C. Chan, P. D. White Eds), Oxford University Press, 2000; Solid Phase Synthesis, A Practical Guide, (S. F. Kates, F Albericio Eds), Marcel Dekker, 2000; P. Seneci, Solid-Phase Synthesis and Combinatorial Technologies, John Wiley & Sons, 2000; Synthesis of Peptides and Peptidomimetics (M. Goodman, Editor-in-chief, A. Felix, L. Moroder, C. Tmiolo Eds), Thieme, 2002; N. L. Benoiton, Chemistry of Peptide Synthesis, CRC Press, 2005.
Table 1
Figure imgf000020_0001
Table 2
Figure imgf000021_0001
-X^X!-A1 Moieties
HB25 -C(=0)-0-(CH2)i5-C(=0)-OH
HB26 -C(=0)-0-(CH2)i7-C(=0)-OH
HB27 -C(=0)-0-(CH2)i9-C(=0)-OH
HB28 -C(=0)-0-(CH2)8-CH=CH-(CH2)7-C(=0)-OH
HB29 -C(=0)-NH-(CH2)i5-C(=0)-OH
HB30 -C(=0)-NH-(CH2)i7-C(=0)-OH
HB31 -C(=0)-NH-(CH2)i9-C(=0)-OH
HB32 -C(=0)-NH-(CH2)8-CH=CH-(CH2)7-C(=0)-OH
HB33 -0-(CH2)i5-C(=0)-OH
HB34 -0-(CH2)i7-C(=0)-OH
HB35 -0-(CH2)i9-C(=0)-OH
HB36 -0-(CH2)8-CH=CH-(CH2)7-C(=0)-OH
HB37 -NH-(CH2)2-0-C(=0)-(CH2)i4-C(=0)-OH
HB38 -NH-(CH2)2-0-C(=0)-(CH2)i6-C(=0)-OH
HB39 -NH-(CH2)2-0-C(=0)-(CH2)i8-C(=0)-OH
HB40 -NH-(CH2)2-0 -C(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-OH
HB41 -0-(CH2)2-0-C(=0)-(CH2)i4-C(=0)-OH
HB42 -0-(CH2)2-0-C(=0)-(CH2)i6-C(=0)-OH
HB43 -0-(CH2)2-0-C(=0)-(CH2)i8-C(=0)-OH
HB44 -0-(CH2)2-0 -C(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-OH
HB45 -NH-CH2-C(=0)-0-[(CH2)2-0-]eC(=0)-(CH2)i4-C(=0)-OH
HB46 -NH-CH2-C(=0)-0-[(CH2)2-0-]eC(=0)-(CH2)i6-C(=0)-OH
HB47 -NH-CH2-C(=0)-0-[(CH2)2-0-]eC(=0)-(CH2)i8-C(=0)-OH
HB48 -NH-CH2-C(=0)-0-[(CH2)2-0-]eC(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-OH
HB49 -NH-(CH2)2-0 -C(=0)-(CH2)i4-C(=0)-0-CH3 -X^X!-A1 Moieties
HB50 -NH-(CH2)2-0 -C(=0)-(CH2)i6-C(=0)-0-CH3
HB51 -NH-(CH2)2-0 -C(=0)-(CH2)i8-C(=0)-0-CH3
HB52 -NH-(CH2)2-0-C(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-0-CH3
HB53 -C(=0)-CH2-0-C(=0)-(CH2)i4-C(=0)-OH
HB54 -C(=0)-CH2-0-C(=0)-(CH2)i6-C(=0)-OH
HB55 -C(=0)-CH2-0-C(=0)-(CH2)i8-C(=0)-OH
HB56 -C(=0)-CH2-0-C(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-OH
HB57 -C(=0)-CH(CH3)-0-C(=0)-(CH2)i4-C(=0)-OH
HB58 -C(=0)-CH(CH3)-0-C(=0)-(CH2)i6-C(=0)-OH
HB59 -C(=0)-CH(CH3)-0-C(=0)-(CH2)i8-C(=0)-OH
HB60 -C(=0)-CH(CH3)-0-C(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-OH
HB61 -C(=0)-(CH2)5-0-C(=0)-(CH2)i4-C(=0)-OH
HB62 -C(=0)-(CH2)5-0-C(=0)-(CH2)i6-C(=0)-OH
HB63 -C(=0)-(CH2)5-0-C(=0)-(CH2)i8-C(=0)-OH
HB64 -C(=0)-(CH2)5-0-C(=0)-(CH2)7-CH=CH-(CH2)7-C(=0)-OH
Table 3
Figure imgf000023_0001
Pharmaceutical Compositions
In certain aspects, the compounds of any of the preceding embodiments may be formulated into pharmaceutical compositions in any suitable manner. In general, as compounds for the treatment of cancer, such pharmaceutical formulations are aqueous formulations suitable for parenteral administration, such as intravenous or intra-arterial administration.
In at least one aspect, the disclosure provides pharmaceutical compositions that include one or more compounds of formula (I) (according to any of the foregoing
embodiments) and a protein. In some embodiments, the protein is an albumin or an albumin mimetic. In some such embodiments, the protein is human serum albumin (HSA) or a mimetic thereof, i.e., a protein whose sequence is at least 50% equivalent to that of HSA, or at least 60% equivalent to that of HSA, or at least 70% equivalent to that of HSA, or at least 80% equivalent to that of HSA, or at least 90% equivalent to that of HSA, or at least 95% equivalent to that of HSA, at least 97% equivalent to that of HSA, at least 99% equivalent to that of HSA. In some embodiments, the protein is human serum albumin.
In certain embodiments of any of the foregoing embodiments, the pharmaceutical composition also includes a carrier, such as a liquid carrier. In some embodiments, the carrier includes water. For example, in some such embodiments, water makes up at least 50% by volume, or at least 60% by volume, or at least 70% by volume, or at least 80% by volume, or at least 90% by volume, based on the total volume of liquid materials in the pharmaceutical composition. The carrier can also include other liquid ingredients, such as liquid ingredients commonly included in aqueous pharmaceutical formulations for parenteral administration.
In certain embodiments having an aqueous carrier, the compounds of formula (I) bind non-covalently to the protein in the pharmaceutical formulation. In some embodiments, the compound of formula (I) and the protein (e.g., human serum albumin) are non-covalently associated with each other with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1 at 25 °C in the aqueous composition.
In some embodiments having an aqueous carrier, the compound of formula (I) and the protein are solvated by the carrier. In some such embodiments, at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 98% by weight, or at least 99% by weight of the compounds of formula (I) in the composition are bound non-covalently to the protein with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1 at 25 °C in the aqueous composition. In some further such embodiments, the composition is substantially free of agglomerates or nanoparticles. For example, in some embodiments of any of the aforementioned embodiments, no more than 5% by weight, or no more than 4% by weight, or no more than 3% by weight, or no more than 2% by weight, or no more than 1% by weight of the protein-compound (i.e., non-covalently bound conjugates between the protein and one or more compounds of formula (I)) in the aqueous composition have a radius greater than 7 nm, or a radius greater than 5 nm, or a radius greater than 4 nm, as measured by dynamic light scattering.
The compound of formula (I) can have any suitable molar ratio to the protein in the formulation. For example, in some embodiments of any of the foregoing embodiments, the molar ratio of the compound of formula (I) to the protein ranges from 1 : 10 to 20: 1, or from 1 :5 to 15: 1, or from 1 :2 to 10: 1. In some embodiments of any of the foregoing embodiments, the molar ratio of the compound of formula (I) to the protein is about 1 : 1, or is about 2: 1, or is about 3: 1, or is about 4: 1, or is about 5: 1, or is about 6: 1, or is about 7: 1, wherein the term "about," in this instance means ±0.5: 1, such that "about 5: 1" refers to a range from 4.5: 1 to 5.5: 1.
In at least one aspect, the disclosure provides pharmaceutical compositions that include: a compound, which comprises a peptide moiety and a protein binding moiety; a protein, wherein the protein is an albumin or an albumin mimetic; and a carrier, which comprises water.
In some embodiments, the protein is human serum albumin (HSA) or a mimetic thereof, i.e., a protein whose sequence is at least 50% equivalent to that of HSA, or at least 60% equivalent to that of HSA, or at least 70% equivalent to that of HSA, or at least 80% equivalent to that of HSA, or at least 90% equivalent to that of HSA, or at least 95% equivalent to that of HSA, at least 97% equivalent to that of HSA, at least 99% equivalent to that of HSA. In some embodiments, the protein is human serum albumin.
As noted above, in some embodiments, the carrier includes water. For example, in some such embodiments, water makes up at least 50% by volume, or at least 60% by volume, or at least 70% by volume, or at least 80% by volume, or at least 90% by volume, based on the total volume of liquid materials in the pharmaceutical composition. The carrier can also include other liquid ingredients, such as liquid ingredients commonly included in aqueous pharmaceutical formulations for parenteral administration.
In certain embodiments, the compounds bind non-covalently to the protein in the pharmaceutical formulation. In some embodiments, the compound and the protein (e.g., human serum albumin) are non-covalently associated with each other with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1 at 25 °C in the aqueous composition.
In some embodiments having an aqueous carrier, the compound and the protein are solvated by the carrier. In some such embodiments, at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 98% by weight, or at least 99% by weight of the compounds of formula (I) in the composition are bound non-covalently to the protein with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1 at 25 °C in the aqueous composition. In some further such embodiments, the composition is substantially free of agglomerates or nanoparticles. For example, in some embodiments of any of the aforementioned embodiments, no more than 5% by weight, or no more than 4% by weight, or no more than 3% by weight, or no more than 2% by weight, or no more than 1% by weight of the protein-compound (i.e., non-covalently bound conjugates between the protein and one or more compounds of formula (I)) in the aqueous composition have a radius greater than 7 nm, or a radius greater than 5 nm, or a radius greater than 4 nm, as measured by dynamic light scattering.
The compound of formula (I) can have any suitable molar ratio to the protein in the formulation. For example, in some embodiments of any of the foregoing embodiments, the molar ratio of the compound of formula (I) to the protein ranges from 1 : 10 to 20: 1, or from 1 :5 to 15: 1, or from 1 :2 to 10: 1. In some embodiments of any of the foregoing embodiments, the molar ratio of the compound of formula (I) to the protein is about 1 : 1, or is about 2: 1, or is about 3: 1, or is about 4: 1, or is about 5: 1, or is about 6: 1, or is about 7: 1, wherein the term "about," in this instance means ±0.5: 1, such that "about 5: 1" refers to a range from 4.5: 1 to 5.5: 1.
The pharmaceutical compositions of any of the foregoing aspects and embodiments can also include certain additional ingredients, such as those commonly employed in pharmaceutical compositions for parenteral administration.
Methods and Uses
The compounds or compositions of any of the foregoing embodiments are useful in the treatment of cancer and related disorders. Therefore, these compounds and compositions can be used for administration to a subject who has or has had a cancerous tumor.
Thus, in certain aspects, the disclosure provides methods of treating cancer, including administering to a subject a compound or composition of any of the foregoing aspects and embodiments. In some embodiments, the subject is a human. In some embodiments, the subject is a subject in need of such treatment, e.g., a human in need of such treatment.
In some aspects, the disclosure provides methods of inducing apoptosis in a cancer cell, including contacting the cancer cell with a compound or composition of any of the foregoing aspects and embodiments.
In some aspects, the disclosure provides methods of inhibiting proliferation of a cancerous tumor, including contacting the cancerous tumor with a compound or composition of any of the foregoing aspects and embodiments.
In some aspects, the disclosure provides uses of a compound or composition of any of the foregoing aspects and embodiments as a medicament.
In some aspects, the disclosure provides uses of a compound or composition of any of the foregoing aspects and embodiments for treating cancer.
In some aspects, the disclosure provides uses of a compound of any of the foregoing aspects and embodiments in the manufacture of a medicament.
In some aspects, the disclosure provides uses of a compound of any of the foregoing aspects and embodiments in the manufacture of a medicament for treating cancer.
Combination Therapies
The compounds or compositions of any of the foregoing embodiments are useful when used in conjunction with immunotherapy agents, such as checkpoint inhibitors, toll like receptor modulators, and various antibodies, including, but not limited to, alemtuzumab, atezolizumab, ipilimumab, ofatumumab, nivolumab, pembrolizumab, and rituximab.
EXAMPLES
The following examples show certain illustrative embodiments of the compounds, compositions, and methods disclosed herein. These examples are not to be taken as limiting in any way. Nor should the examples be taken as expressing any preferred embodiments, or as indicating any direction for further research.
The examples may use abbreviations for certain common chemicals. The following abbreviations refer to the compounds indicated.
DMF Dimethylformamide
DCM Dichloromethane
NMR Nuclear magnetic resonance HPLC = High-performance liquid chromatography
RP-HLPC = Reverse-phase high-performance liquid chromatography
LRMS = Liquid chromatography / low-resolution mass spectrometry
HRMS = Liquid chromatography / high-resolution mass spectrometry TIPS = Triisopropylsilyl
DMAP = 4-(Dimethylamino)pyridine
EDC = l-Ethyl-3-(3-dimethylaminopropyl)carbodiirnide
THF = Tetrahydrofuran
DIPEA = N,N-diisopropylethylamine
HBTU = 2-(lH-benzotriazol-l-yl)-l,l,3,3-tetramethyluronium
hexafluorophosphate)
DCC = N,N'-dicyclohexylcarbodiimide
HSA = Human serum albumin
Coupling
Figure imgf000028_0001
Scheme 1. Synthesis of KLA peptide-C18 prodrug.
Example 1 - General procedure for the synthesis of KLA peptide-C18 prodrug.
Peptides were synthesized using standard solid phase synthesis procedures on an automatic peptide synthesizer using 1 g Rink resin with a loading of 0.68 mmol/g. The terminal Fmoc group was removed by treating 500 mg resin with 20% 4-methyl piperidine in DMF (10 mL) for 10 min, the solvent was removed and the resin treated with another 10 mL of 20% 4-methyl piperidine in DMF for 5 min. The solvent was removed and the resin washed with DMF (25 mL x5). To a solution of the Mono-TIPS -protected octadecanedioic acid (316.9 mg, 0.673 mmol) in DMF was added DIPEA (234 uL, 1.34 mmol) followed by HBTU (255 mg, 0.673 mmol), this solution was mixed for 2 min then added to the resin. The mixture was left to mix ovemight then the solvent was removed and the resin was washed with DMF (25 mL x5) and DCM (25 mL x6) then dried under vacuum. The peptide was cleaved from the resin using a mixture of 95:2.5 :2.5 (TFA:TIPS:H20) for 2 h. The peptide was precipitated by addition to cold ether. Centrifuge and dry under vacuum. Purify by preparative HPLC. ESI, 910.77 [M+2H]2+.

Claims

1. A compound of formula (I)
A1 X1 X2 A2 (I) wherein:
A1 is an organic group; or A1 is a hydrophilic group or a hydrogen atom;
A2 is a peptide moiety;
X1 is a hydrophobic group; and
X2 is a direct bond, an organic group, -0-, -S-, -S(=0)-, -S(=0)2-, -S-S-, -N=, =N-, -N(H)-, -N=N-N(H)-, -N(H)-N=N-, -N(OH)-, or -N(=0)-.
2. The compound of claim 1, wherein A1 is a carboxylic acid group, a carboxylate anion, or a carboxylate ester.
3. The compound of claim 2, wherein A1 is a carboxylic acid group.
4. The compound of any one of claims 1 to 3, wherein the peptide moiety comprises from 2 to 100 amino acid units, or from 3 to 50 amino acid units, or from 4 to 35 amino acid units, or from 5 to 30 amino acid units, or from 5 to 25 amino acid units, or from 5 to 20 amino acid units.
5. The compound of any one of claims 1 to 4, wherein the peptide moiety is an anticancer peptide moiety.
6. The compound of claim 5, wherein the anticancer peptide moiety is a moiety of an anticancer peptide selected from the group consisting of: leuteinizing hormone-releasing hormone (LHRH) agonists, LHRH antagonists, tumor-associated antigens, angiogenesis inhibitors (such as VEGF modulators), gastric-releasing peptide receptor (GPRP) antagonists, monoclonal antibodies, and immunotherapy agents, such as checkpoint inhibitors.
7. The compound of claim 6, wherein the anticancer peptide is an angiogenesis inhibitor.
8. The compound of claim 7, wherein the angiogenesis inhibitor is a vascular endothelial growth factor (VEGF) protein.
9. The compound of any one of claims 1 to 8, wherein X1 is C 12-22 hydrocarbylene, which is optionally substituted.
10. The compound of claim 9, wherein X1 is C12-22 alkylene group.
11. The compound of claim 10, wherein X1 is -(CH2)i2-, -(CH2)i4-, -(CH2)i6-, -(CH2)i8-, -(CH2)2o-, or -(CH2)22-.
12. The compound of claim 1 1, wherein X1 is -(CH2)i6-.
13. The compound of claim 12, wherein X2 is -C(=0)-.
14. The compound of claim 1 , wherein -X^-A1 is -C(=0)-(CH2)i6-C(=0)OH and -A2 is an anticancer peptide moiety, and wherein the anticancer peptide is a vascular endothelial growth factor (VEGF) protein and binds to -X^X^A1 via the N-terminus of the protein.
15. A pharmaceutical composition comprising:
a compound of any one of claims 1 to 14; and
a protein, wherein the protein is human serum albumin or a protein whose sequence is at least 50% equivalent to that of human serum albumin.
16. The pharmaceutical composition of claim 15, wherein the protein is human serum albumin.
17. The pharmaceutical composition of claim 15 or 16, further comprising a carrier.
18. The pharmaceutical composition of claim 17, wherein the carrier comprises water.
19. The pharmaceutical composition of claim 18, wherein the compound and the protein are non-covalently associated with each other with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1.
20. The pharmaceutical composition of any one of claims 17 to 19, wherein the compound and the protein are solvated by the carrier.
21. The pharmaceutical composition of any one of claims 17 to 20, which contains one or more compounds of any one of claims 1 to 16 and one or more proteins, wherein at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 99% by weight, of the compounds in the composition are bound to proteins with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1.
22. The pharmaceutical composition of claim 21, wherein at least at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 99% by weight, of the protein- bound particles in the composition have a radius no greater than 5 nm, or no greater than 4 nm, as measured by dynamic light scattering.
23. The pharmaceutical composition of any one of claims 17 to 22, wherein the
pharmaceutical composition is suitable for parenteral administration to a mammal, e.g., a human.
24. The pharmaceutical composition of any one of claims 17 to 22, wherein the
pharmaceutical composition is suitable for intravenous administration to a mammal, e.g., a human.
25. A pharmaceutical composition comprising:
a compound, which comprises a peptide moiety and a protein binding moiety;
a protein, wherein the protein is human serum albumin or a protein whose sequence is at least 50% equivalent to that of human serum albumin; and
a carrier, which comprises water;
wherein the compound and the protein are non-covalently associated with each other with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1; and
wherein the compound and the protein are solvated by the carrier.
26. The pharmaceutical composition of claim 25, wherein the compound is a compound of any one of claims 1 to 16.
27. The pharmaceutical composition of claim 25 or 26, wherein the protein is human serum albumin.
28. The pharmaceutical composition of any one of claims 25 to 27, which contains one or more compounds of any one of claims 1 to 16 and one or more proteins, wherein at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 99% by weight, of the compounds in the composition are bound to proteins with a binding constant (Kb) of at least 102 M"1, or at least 103 M"1, or at least 104 M"1, or at least 105 M"1.
29. The pharmaceutical composition of claim 28, wherein at least at least 90% by weight, or at least 95% by weight, or at least 97% by weight, or at least 99% by weight, of the protein- bound particles in the composition have a radius of no greater than 5 nm, or no greater than 4 nm, as measured by dynamic light scattering.
30. The pharmaceutical composition of any one of claims 25 to 29, wherein the
pharmaceutical composition is suitable for parenteral administration to a mammal, e.g., a human.
31. The pharmaceutical composition of any one of claims 25 to 29, wherein the
pharmaceutical composition is suitable for intravenous administration to a mammal, e.g., a human.
32. A method of treating cancer, comprising:
administering to a subject a compound of any one of claims 1 to 14 or a composition of any one of claims 15 to 31.
33. The method of claim 32, further comprising administering to a subject an
immunotherapy agent.
34. The method of claim 33, wherein administering the immunotherapeutic agent to the subject is carried out concurrently with, or within no more than three days before or after, administering to the subject the compound of any one of claims 1 to 14 or the composition of any one of claims 15 to 31.
35. A method of inducing apoptosis in a cancer cell, comprising:
contacting the cancer cell with a compound of any one of claims 1 to 14 or a composition of any one of claims 15 to 31.
36. A method of inhibiting proliferation of a cancerous tumor, comprising:
contacting the cancerous tumor with a compound of any one of claims 1 to 14 or a composition of any one of claims 15 to 31.
37. Use of a compound of any one of claims 1 to 14 or a composition of any one of claims 15 to 31 as a medicament.
38. Use of a compound of any one of claims 1 to 14 or a composition of any one of claims 15 to 31 for treating cancer.
39. Use of a compound of any one of claims 1 to 14 in the manufacture of a medicament.
40. Use of a compound of any one of claims 1 to 14 in the manufacture of a medicament for treating cancer.
PCT/US2018/023573 2017-03-22 2018-03-21 Modified peptides and uses thereof for treating cancer WO2018175589A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762475175P 2017-03-22 2017-03-22
US62/475,175 2017-03-22

Publications (1)

Publication Number Publication Date
WO2018175589A1 true WO2018175589A1 (en) 2018-09-27

Family

ID=63585737

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/023573 WO2018175589A1 (en) 2017-03-22 2018-03-21 Modified peptides and uses thereof for treating cancer

Country Status (1)

Country Link
WO (1) WO2018175589A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021207630A1 (en) * 2020-04-10 2021-10-14 Northwestern University Oxidized tumor cell lysates encapsulated in liposomal spherical nucleic acids as potent cancer immunotherapeutics
EP4076496A4 (en) * 2019-12-20 2024-01-10 Univ Northwestern Terlipressin-octadecanedioic acid conjugate for vasoconstrictive therapy

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100239659A1 (en) * 2007-07-19 2010-09-23 Allexcel., Inc. Self-assembling amphiphilic polymers as anti-cancer agents
KR20140033676A (en) * 2012-09-10 2014-03-19 코웨이 주식회사 Multi block copolymer-cell penetrating peptide conjugate, a preparation thereof, and the use of the same
US20140135254A1 (en) * 2010-08-20 2014-05-15 Cerulean Pharma Inc. Therapeutic peptide-polymer conjugates, particles, compositions, and related methods

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100239659A1 (en) * 2007-07-19 2010-09-23 Allexcel., Inc. Self-assembling amphiphilic polymers as anti-cancer agents
US20140135254A1 (en) * 2010-08-20 2014-05-15 Cerulean Pharma Inc. Therapeutic peptide-polymer conjugates, particles, compositions, and related methods
KR20140033676A (en) * 2012-09-10 2014-03-19 코웨이 주식회사 Multi block copolymer-cell penetrating peptide conjugate, a preparation thereof, and the use of the same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LEVY, 0. E. ET AL.: "Novel Exenatide Analogs with Peptidic Albumin Binding Domains: Potent Anti-Diabetic Agents with Extended Duration of Action", PLOS ONE, vol. 9, no. 2, 2014, pages e87704, XP055232184 *
WANG, Y. ET AL.: "Peptide-drug conjugates as effective prodrug strategies for targeted delivery", ADVANCED DRUG DELIVERY REVIEW, vol. 110 - 11, 29 June 2016 (2016-06-29), pages 112 - 126, XP085062902 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4076496A4 (en) * 2019-12-20 2024-01-10 Univ Northwestern Terlipressin-octadecanedioic acid conjugate for vasoconstrictive therapy
WO2021207630A1 (en) * 2020-04-10 2021-10-14 Northwestern University Oxidized tumor cell lysates encapsulated in liposomal spherical nucleic acids as potent cancer immunotherapeutics

Similar Documents

Publication Publication Date Title
ES2733355T3 (en) Prodrug comprising a self-cleavable linker
CA3007976C (en) Cnp prodrugs with large carrier moieties
CA3007979C (en) Cnp prodrugs with carrier attachment at the ring moiety
US10654864B2 (en) Modified cytotoxins and their therapeutic use
US20170267727A1 (en) Conjugates of pH Low Insertion Peptide and Monomethyl Auristatins in the Treatment of Solid Tumors
JP2009520040A5 (en)
JP2021138761A (en) Conjugates of quaternized tubulysin compounds
BR112020008974A2 (en) ligand-drug conjugates as substrates for selective cleavage by cathepsin b exopeptidase activity
WO2018175589A1 (en) Modified peptides and uses thereof for treating cancer
WO2018175622A1 (en) Modified anthracycline compounds and their therapeutic use
US20190381179A1 (en) Modified cytotoxins and their therapeutic use
JP5105166B2 (en) Method for producing polyether
WO2018175601A1 (en) Modified platinum compounds and therapeutic uses thereof
US20200048198A1 (en) Modified histone deacetylase inhibitors and uses thereof
EP3615088A2 (en) Modified mri contrast agents and uses thereof
US20220273607A1 (en) Methods of using modified cytotoxins to treat cancer
US20200046846A1 (en) Modified oligonucleotides and therapeutic uses thereof
EP3844297A2 (en) Peptides having immunomodulatory properties
US7368431B2 (en) Polypeptide, the conjugate thereof with doxorubicine and a pharmaceutical composition based thereon
Kweon et al. Coordinated ASBT and EGFR Mechanisms for Optimized Liraglutide Nanoformulation Absorption in the GI Tract

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: 18771878

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18771878

Country of ref document: EP

Kind code of ref document: A1