WO2025208006A1 - Heterobifunctional molecules for binding na v1.7 and methods of treating medical conditions using same - Google Patents
Heterobifunctional molecules for binding na v1.7 and methods of treating medical conditions using sameInfo
- Publication number
- WO2025208006A1 WO2025208006A1 PCT/US2025/021976 US2025021976W WO2025208006A1 WO 2025208006 A1 WO2025208006 A1 WO 2025208006A1 US 2025021976 W US2025021976 W US 2025021976W WO 2025208006 A1 WO2025208006 A1 WO 2025208006A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- pain
- bond
- alkyl
- certain embodiments
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
Definitions
- the invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain.
- pain persists beyond its usefulness.
- Such unnecessary suffering from pain can impair a subject’s physical mobility, mental performance, and even contribute to depression.
- Such unnecessary suffering from pain can be due to acute pain and/or chronic pain.
- Such pain can also be characterized according to whether the pain is neuropathic pain or nociceptive pain.
- Substantial resources have been devoted over the years to researching the causes of various types of pain and to the development of medicine to attenuate pain experienced by a patient.
- heterobifuctional compounds that are able to simultaneously bind a target cell-surface protein as well as an exogenous antibody protein are described in, for example, international patent application publication nos. WO 2018/134731, WO 2023/017484, and WO 2023/017483.
- Such heterobifuctional compounds are used with an anti-cotinine antibody to treat a variety of medical diseases and conditions.
- Additional heterobifunctional coti nine-containing compounds that bind voltage-gated sodium channel Na> 1.7 and an exogenous antibody protein are needed and would provide benefits to patients suffering from voltage-gated sodium channel Na v l 7-associated diseases and conditions, such as pain.
- the invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain.
- one aspect of the invention provides a collection of heterobifunctional cotinine- containing compounds represented by Formula I: or a pharmaceutically acceptable salt thereof, where the variables are as defined in the detailed description. Further description of additional collections of heterobifunctional cotinine- containing compounds are described in the detailed description.
- the compounds may be part of a pharmaceutical composition comprising a pharmaceutically acceptable carrier.
- the heterobifunctional compounds bind to voltage-gated sodium channel Na v l .7.
- the heterobifunctional compounds also bind to an anti-cotinine antibody or fragment thereof that binds cotinine. In this way, the heterobifunctional compounds may be characterized as an antibody recruiting molecule (ARM).
- ARM antibody recruiting molecule
- Another aspect of the invention provides a method of treating or preventing a Na v l .7- associated disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigenbinding fragment thereof.
- a heterobifunctional compound described herein such as a compound of Formula I
- Another aspect of the invention provides a method of depleting voltage-gated sodium channel Na v 1.7-expressing cells, wherein the method comprises contacting the cells with an effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
- a heterobifunctional compound described herein such as a compound of Formula I
- an anti-cotinine antibody or antigen-binding fragment thereof.
- Another aspect of the invention provides a combination comprising a heterobifunctional compound described herein (such as a compound of Formula I) and an anti- cotinine antibody, or antigen-binding fragment thereof.
- the invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain.
- the practice of the present invention employs, unless otherwise indicated, conventional techniques of organic chemistry, pharmacology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology. Such techniques are explained in the literature, such as in “Comprehensive Organic Synthesis” (B.M. Trost & I. Fleming, eds., 1991- 1992); “Handbook of experimental immunology” (D.M. Weir & C.C. Blackwell, eds.); “Current protocols in molecular biology” (F.M.
- aliphatic or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “cycloaliphatic”), that has a single point of attachment to the rest of the molecule.
- aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms.
- aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
- “cycloaliphatic” refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
- bicyclic ring or “bicyclic ring system” refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or having one or more units of unsaturation, having one or more atoms in common between the two rings of the ring system.
- the term includes any permissible ring fusion, such as o/VAo-fused or spirocyclic.
- heterocyclic is a subset of “bicyclic” that requires that one or more heteroatoms are present in one or both rings of the bicycle.
- a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen).
- a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
- Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom.
- a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted.
- Exemplary bicyclic rings include:
- Exemplary bridged bicyclics include: [0022]
- the term “lower alkyl” refers to a C1-4 straight or branched alkyl group.
- Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
- lower haloalkyl refers to a C1-4 straight or branched alkyl group that is substituted with one or more halogen atoms.
- heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2/f-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
- Ci-s saturated or unsaturated, straight or branched, hydrocarbon chain
- bivalent Ci-s (or Ci-e) saturated or unsaturated, straight or branched, hydrocarbon chain refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
- alkylene refers to a bivalent alkyl group.
- An “alkylene chain” is a polymethylene group, i.e., -(CH2) n - wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3.
- a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
- alkenylene refers to a bivalent alkenyl group.
- a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
- halogen means F, Cl, Br, or I.
- aryl used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
- aryl may be used interchangeably with the term “aryl ring.”
- aryl refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents.
- aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
- phenylene refers to a multivalent phenyl group having the appropriate number of open valences to account for groups attached to it.
- phenylene is a bivalent phenyl group when it has two groups attached t
- phenylene is a trivalent phenyl group when it has three groups attached
- arylene refers to a bivalent aryl group.
- heteroaryl and “heteroar-,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or
- Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4/7 quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, and tetrahydroisoquinolinyl.
- a heteroaryl group may be mono- or bicyclic.
- heteroaryl may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted.
- heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
- heteroarylene refers to a multivalent heteroaryl group having the appropriate number of open valences to account for groups attached to it. For example, “heteroarylene” is a bivalent heteroaryl group when it has two groups attached to it; “heteroarylene” is a trivalent heteroaryl group when it has three groups attached to it.
- heterocycle As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
- nitrogen includes a substituted nitrogen.
- a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
- saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, 2-oxa-6- azaspiro[3.3]heptane, and quinuclidinyl.
- oxo-heterocyclylene refers to a multivalent oxo-heterocyclyl group having the appropriate number of open valences to account for groups attached to it.
- partially unsaturated refers to a ring moiety that includes at least one double or triple bond.
- partially unsaturated is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
- R* is Ci-6 aliphatic
- R* is optionally substituted with halogen, - R*, -(haloR*), -OH, -OR*, -O(haloR*), -CN, -C(O)OH, -C(O)OR*, -NH 2 , -NHR*, -NR* 2 , or -NO 2
- each R* is independently selected from Ci-4 aliphatic, -CH 2 Ph, -0(CH 2 )o iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R* is unsubstituted or where preceded by halo is substituted only with one or more halogens.
- salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphor sulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pect
- Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N'(Ci-4alkyl)4 salts.
- Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
- Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
- structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
- the invention includes compounds that differ only in the presence of one or more isotopically enriched atoms.
- Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization.
- Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers.
- an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride
- a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis.
- diastereomeric salts are formed with an appropriate optically active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means known in the art, and subsequent recovery of the pure enantiomers.
- Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
- Chiral center(s) in a compound of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
- a compound described herein may exist as an atropisomer (e.g., substituted biaryls)
- all forms of such atropisomer are considered part of this invention.
- Chemical names, common names, and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name, and an ambiguity exists between the structure and the name, the structure predominates. It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
- alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l -propyl, 2-m ethyl -2-propyl, 2-methyl-l -butyl, 3- methyl-1 -butyl, 2-m ethyl-3 -butyl, 2,2-dimethyl-l -propyl, 2-methyl-l -pentyl, 3-methyl-l-pentyl, 4-methyl-l -pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l- butyl, 3,3-dimethyl-l-butyl, 2-ethyl-l -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl
- haloalkyl refers to an alkyl group that is substituted with at least one halogen.
- exemplary haloalkyl groups include -CH2F, -CHF2, -CF3, -CH2CF3, -CF2CF3, and the like.
- haloalkylene refers to a bivalent haloalkyl group.
- alkenyl and alkynyl are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
- a cyclopentane susbstituted with an oxo group is cyclopentanone.
- the terms “subject” and “patient” are used interchangeably and refer to organisms to be treated by the methods of the present invention.
- Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans.
- the term “effective amount” refers to the amount of a compound sufficient to effect beneficial or desired results (e.g., a therapeutic, ameliorative, inhibitory or preventative result).
- An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
- the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
- the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
- the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents.
- the compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see e.g., Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975],
- a compound of the invention contains both a basic moiety (such as, but not limited to, a pyridine or imidazole) and an acidic moiety (such as, but not limited to, a carboxylic acid) zwitterions (“inner salts”) may be formed.
- acidic and basic salts used within the scope of the invention are pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts.
- Such salts of the compounds of the invention may be formed, for example, by reacting a compound of the invention with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
- One aspect of the invention provides a compound represented by Formula I: or a pharmaceutically acceptable salt thereof, wherein:
- X 1 is a bond or a C2-8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -N(CHs)-, -O-, piperidinylene, or C3-C6 cycloalkylene;
- X 2 is $-(Ci-5 alkylene)-N(H)- or a covalent bond, wherein is a bond to L;
- R 2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl) or Ci-4 alkyl;
- R 3 and R 4 each represent independently for each occurrence hydrogen or C1-4 alkyl
- R 7 , R 8 , R 9 , and R 10 each represent independently for each occurrence hydrogen or C1-4 alkyl
- R 11 is a bond to X 1 ;
- R 12 is hydrogen, C1-3 haloalkyl, or C1-3 alkyl
- R 13 is thiazolyl or 1,2,4-thiadiazolyl, each of which is substituted with 0 or 1 occurrence of Ci -4 alkyl;
- Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene
- L lb is -CH 2 -NH-C(O)-, -NHC(O)-, or -C(O)NH-
- L 2b is Ce-i 2 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NR lb -, - wherein n is 1 , 2, 3, or 4, and represents a covalent bond to L lb
- each R lb is independently hydrogen or C1-3 alkyl
- L-d is C 12-22 linear alkylene, wherein 1, 2, 3, 4, or 5 methylene units are replaced with -NH-, -O-, -C(O)NH-, -NHC(O)-, or -NHC(O)-NH-;
- Z 4 and Z 5 is independently CH or N, provided that no more than two of Z 2 , Z 3 , Z 4 and Z ⁇ are N;
- L lj is -NH-, -C(O)NH-, -NHC(O)-, or -O-;
- L 2 ' is Ci-6 linear alkylene or 11 , , represents a covalent bond to L 1 '; and
- I' represents a single bond or a double bond;
- each of Z 1 and Z 2 is independently CH or N;
- L lk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and
- L 2k is a C3-8
- X 1 is a bond or a C 2 -8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -N(CH3)-, -O-, piperidinylene, or C3-C6 cycloalkylene.
- X 1 is a bond.
- X 1 is a C 2 -s bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -O-, piperidinylene, or C3-C6 cycloalkylene.
- X 1 is a C2-8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)- or -O-.
- X 1 is -(Ci-4 alkylcne)-pipcrazinylcnc-T, wherein T is a bond to L. In certain embodiments, X 1 is , wherein T is a bond to L.
- R 13 is fluoro, chloro, phenyl, or hydrogen; and x is 1 or 2.
- R 6 is hydrogen, fluoro, chloro, or C1.4 alkyl
- R 7 is hydrogen or Ci-4 alkyl
- Y 1 is one of the following:
- Y 1 is one of the following:
- Y ⁇ X 1 - is one of the following:
- Y ⁇ X 1 - is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
- R 7 represents independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R 7 is hydrogen or CM alkyl. In certain embodiments, R 7 is hydrogen. In certain embodiments, R 7 represents independently for each occurrence CM alkyl. In certain embodiments, R 7 is Ci-4 alkyl. In certain embodiments, R 7 is methyl. In certain embodiments, R 7 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
- R 9 represents independently for each occurrence hydrogen or CM alkyl. In certain embodiments, R 9 is hydrogen or CM alkyl. In certain embodiments, R 9 is hydrogen. In certain embodiments, R 9 represents independently for each occurrence CM alkyl. In certain embodiments, R 9 is CM alkyl. In certain embodiments, R 9 is methyl. In certain embodiments, R 9 is selected from the groups depicted in the compounds in Tables 1 , 2, and 3, below.
- R 11 is a bond to X 1 .
- R 11 replaces R 10 .
- R 11 replaces R 9 .
- R 11 replaces R 7 .
- R 11 occurs on R 2 .
- R 11 occurs on a phenyl ring of Y 1 .
- R 11 occurs on a pyridinyl ring of Y 1 .
- the position of R 11 on Y 1 is selected from the positions depicted in the compounds in Table 1, below.
- R 13 is thiazolyl or 1,2,4-thiadiazolyl, each of which is substituted with 0 or 1 occurrence of C1-4 alkyl. In certain embodiments, R 13 is thiazolyl substituted with 0 or 1 occurrence of C1-4 alkyl. In certain embodiments, R 13 is thiazolyl. In certain embodiments, R 13 is 1,2,4-thiadiazolyl substituted with 0 or 1 occurrence of C1-4 alkyl. In certain embodiments, R 13 is 1,2,4-thiadiazolyl. In certain embodiments, R 13 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
- R 14 is -N(R 10 )2 or furanyl. In certain embodiments, R 14 is -N(R 1O ) 2 . In certain embodiments, R 14 is -N(R 10 )(R 11 ). In certain embodiments, R 14 is - N(CH3)(R n ). In certain embodiments, R 14 is furanyl. In certain embodiments, R 14 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below. [0113] As generally defined above, R 13 is fluoro, chloro, phenyl, or hydrogen. In certain embodiments, R 15 is fluoro, chloro, or phenyl. In certain embodiments, R 15 is fluoro or chloro.
- R 13 is fluoro. In certain embodiments, R 15 is chloro. In certain embodiments, R 13 is phenyl. In certain embodiments, R 15 is hydrogen. In certain embodiments, R 15 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
- L is a divalent linker selected from:
- L 2b is wherein n is 1, 2, 3, or 4, and represents a covalent bond to L lb ; and each R lb is independently hydrogen or C1.3 alkyl;
- L 2 ' is Ci-6 linear alkylene or , wherein n is 1 *** or 2, and represents a covalent bond to L 1 '; and ! ⁇ represents a single bond or a double bond;
- L 2m is C3-6 linear alkylene, C3-6 cycloalkylene, or , wherein n is 1 or 2, and represents a covalent bond to
- L-n-iv 0, 1, or 2 hydrogen atoms are replaced with F
- L lp is a bond, -C(O)-, -C(O)NH- , -NHC(O)-, -S(O)2NH-, or -NHS(O)2-
- L 2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-; wherein each represents a covalent bond to X 1 , and each represents a covalent bond to X 2 .
- X 1 and represents a covalent bond to X 2 .
- variable L Further description and embodiments for variable L are provided below in, for example, Part B.
- the compound of Formula (I) is represented by Formula la or a pharmaceutically acceptable salt thereof: wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula la. [0120] In certain embodiments, the compound of Formula (I) is represented by Formula lb or a pharmaceutically acceptable salt thereof: wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula lb.
- the compound of Formula (I) is represented by Formula Ic or
- the compound is a compound of Formula Ic or Id. In certain embodiments, the compound is a compound of Formula Ic, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ic. In certain embodiments, the compound is a compound of Formula Id, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Id. [0122] In certain embodiments, the compound of Formula (I) is represented by Formula le or
- the compound of Formula (I) is represented by Formula Ig or Ih or a pharmaceutically acceptable salt thereof:
- the compound is a compound of Formula Ig or Ih. In certain embodiments, the compound is a compound of Formula Ig, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ig. In certain embodiments, the compound is a compound of Formula Ih, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ih.
- the compound of Formula (I) is represented by Formula li or a pharmaceutically acceptable salt thereof: wherein each of the variables is as defined in embodiments herein.
- the compound is a compound of Formula li.
- L is a divalent linker selected from:
- Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene
- L lb is -CH2-NH-C(0)-, -NHC(O)-, or -C(O)NH-
- L 2b is Ce-12 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NR lb -, -C(O)NR lb -, or -
- L 2b is wherein n is 1, 2, 3, or 4, and represents a covalent bond to L lb ; and each R lb is independently hydrogen or C1.3 alkyl;
- L-c O (L-c), wherein L lc is C2 -10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; and L 2c is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or - C(O)NH-;
- L 2f is a bond, -NHC(O)-, -C(O)NH-, or a Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z 1 and Z 2 is independently N or CH; (viii) (L-g), wherein Ring A is a 5 to 6 membered heteroarylene having 1 or 2 nitrogen ring atoms; L lg is a bond, -CH2-, -NH-, or -O-; and L 2g is wherein n is 1, 2, 3, 4, or 5, and represents a covalent bond to L lg ; wherein each Z 1 is independently N or CH; L lh is a bond, -C(O)-, -C(O)-NH-, or -NHC
- L 21 is a bond, C1-12 linear alkylene, or , , , , , represents a covalent bond to HN; and L 31 is a bond or -C(O)-; wherein Z 1 is C, CH, or N; each of Z 2 , Z 3 , Z 4 and Z 3 is independently CH or N, provided that no more than two of Z 2 , Z 3 , Z 4 and Z 5 are N; L lj is -NH-, -C(O)NH-, -NHC(O)-, or -O-; L 2j is C1-6 linear alkylene or , wherein n is 1 or 2, and represents a covalent bond to L lj , and I' represents a single bond or a double bond;
- Ring A is phenylene or a 5- or 6- membered heteroarylene having 1 or 2 nitrogen ring atoms; each of Z 1 and Z 2 is independently CH or N; L lk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and L 2k is a C3-8 straight chain alkylene or , wherein n is 1, 2, or 3, and represents a covalent bond to L lk ;
- L lm is a bond, -C(O)-, -
- L 2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-;
- L is a bivalent, saturated or unsaturated, straight or branched Ci-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(CI- 6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O) 2 -, -N(H)S(O) 2 -, -N(Ci- 6 alkyl)S(O) 2 -, -S(O) 2 N(H)-, -S(O) 2 N(CI- 6 alkyl)-, -N(H)C(O)-, -N(CI- 6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C i-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(H)-
- L is a divalent linker of Formula (L-a), (L-b), (L-c), (L-d), (L-e), (L-f), (L-g), (L-h), (L-i), (L-j), (L-k), (L-m), (L-n-i), (L-n-ii), (L-n-iii), or (L-n-iv).
- L is a divalent linker of Formula (L-a): wherein:
- Ring A and Ring B are each independently C4-6 cycloalkylene
- L la is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NR a -; each R a is independently hydrogen or C1-3 alkyl; and
- L 2a is -O-, -NHC(O)-, or -CH2-O-; wherein represents a represents a covalent bond to X 2 .
- Ring A and Ring B of Formula (L-a) are each independently
- L is a divalent linker of Formula (L-a-i): wherein:
- I represents a covalent bond to represents a covalent bond to X 2 .
- L is a divalent linker of Formula (L-a-ii): -ii); wherein:
- L la is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NR a -; each R a is independently hydrogen or C1-3 alkyl;
- L 2a is -O-, -NHC(O)-, or -CH 2 -O-; p is 1 or 2; and m is 1 or 2; wherein represents a covalent bond to X 1 , and represents a covalent bond to X 2 .
- L lb is -CH 2 -NH-C(O)-, -NHC(O)-, or -C(O)NH-;
- L 2b is , wherein n is 1, 2, 3, or 4, and represents a covalent bond to L lb ; and each R lb is independently hydrogen or C1-3 alkyl; i _ wherein I represents a covalent bond to X 1 , and represents a covalent bond to X 2 .
- L 2b of Formula (L-b) or (L-b-i) is selected from
- L is a divalent linker of Formula (L-b) selected from the group consisting of: .
- L is a divalent linker of Formula (L-c): wherein:
- L lc is C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, - NHC(O)-, or -C(O)NH-;
- Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene
- L 2C is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; wherein represents a represents a covalent bond to X 2 .
- Ring A of Formula (L-c) is
- L is a divalent linker of Formula (L-c-i): wherein:
- L lc is C 2 -10 linear alkylene, wherein I, 2, or 3 methylene units are replaced with -O-, -NH-, - NHC(O)-, or -C(O)NH-;
- L 2C is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; p is 1 or 2; and m is 1 or 2;
- L lc of Formula (L-c) or (L-c-i) is selected from wherein: j is 1, 2, 3, 4, 5, 6, 7, 8, or 9; k is 0, 1, 2, 3, 4, 5, 6, 7, or 8; the sum of j and k is 1, 2, 3, 4, 5, 6, 7, 8, or 9; q is 1, 2, 3, 4, 5, 6, or 7; r is 1, 2, 3, 4, 5, 6, or 7; s is 0, 1, 2, 3, 4, 5, or 6; the sum of q, r, and s is 2, 3, 4, 5, 6, 7, or 8; t is 1, 2, 3, 4, or 5; u is 1, 2, 3, 4, or 5; v is 1, 2, 3, 4, or 5; w is 0, 1, 2, 3, or 4; the sum of t, u, v, and w is 3, 4, 5, 6, or 7; and
- L 2c of Formula (L-c) or (L-c-i) is selected from
- j is O, 1, 2, 3, 4, 5, 6, 7, 8, or 9; k is 0, 1, 2, 3, 4, 5, 6, 7, 8, or 9; the sum of j and k is 1, 2, 3, 4, 5, 6, 7, 8, or 9; q is 0, 2, 3, 4, 5, 6, or 7; r is 1, 2, 3, 4, 5, 6, 7, or 8; s is 0, 1, 2, 3, 4, 5, 6, or 7; the sum of q, r, and s is 1, 2, 3, 4, 5, 6, 7, or 8; t is 0, 1, 2, 3, 4, or 5; u is 1, 2, 3, 4, 5, or 6; v is 1, 2, 3, 4, 5, or 6; w is 0, 1, 2, 3, 4, or 5; the sum of t, u, v, and w is 2, 3, 4, 5, 6, or 7; and
- X’ A , X 2A and X 3A are independently -O-, -NH-, -NHC(O)-, or -C(O)NH-;
- I represents a covalent bond to the ring of Formula (L-c) or (L-c-i), and represents a covalent bond to X 2 .
- L is a divalent linker of Formula (L-c) selected from the group consisting of:
- p he carbonyl group is the attachment point to X 1 .
- L ld of Formula (L-d) is selected from wherein: j is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; k is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; the sum of j and k is 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21; q is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19; r is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19; s is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; the sum of q, r, and s is 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; t is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; u is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; v is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 11, 12, 13, 14, 15, 16, or 17
- L is selected from the group consisting of:
- L is a divalent linker of Formula (L-e): wherein: n is an integer of 3 to 50; wherein I represents a covalent bond to X 1 , and represents a covalent bond to X 2 .
- n of Formula (L-e) is 3 to 25, 3 to 10, 3 to 8, 3 to 7, 3 to 5, or 3 to 4. In certain embodiments, n of Formula (L-e) is 3, 4, 5, 7, 8, 22, or 50.
- L is a divalent linker of Formula (L-f): wherein:
- L lf is a bond; Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-, -NH- , or -C(O)-; or -(C3-6 cycloalkylene)-NHC(O)-;
- L 2t is a bond, -NHC(O)-, -C(O)NH-, or a Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z 1 and Z 2 is independently N or CH;
- I represents a covalent bond to X 1
- ‘ represents a covalent bond to X 2 .
- L lf of Formula (L-f) is selected from ; wherein: j is 1, 2, 3, 4, or 5; k is 0, 1, 2, 3, or 4; the sum of j and k is 1, 2, 3, 4, or 5; q is 1, 2, or 3; r is 1, 2, or 3; s is 0, 1, 2; the sum of q, r, and s is 2, 3, or 4; and
- X 1A and X 2A are independently -O-, -NH-, or -C(O)-; or -(C3-6 cycloalkylene)-NHC(O)-; ####[ ##### wherein 1 represents a covalent bond to the C(O) group of Formula (L-f), represents a covalent bond to the ring of Formula (L-f).
- L 2 - 1 is Ci-6 linear alkylene or , wherein n is 1 or 2, and • represents a covalent bond to L 1 ';
- I _ _ ** wherein I represents a covalent bond to X 1 , and ’ represents a covalent bond to X 2 .
- L is a divalent linker of Formula (L-k) selected from the group consisting of: .
- L-k divalent linker of Formula (L-k) selected from the group consisting of: .
- L is a divalent linker of Formula (L-m): m is 1 or 2; p is 1 or 2;
- L lm is a bond, -C(O)-, -C(O)NH-, -NHC(O)-, -S(O) 2 NH-, or -NHS(O) 2 -;
- L 2m is C3-6 linear alkylene, C3-6 cycloalkylene, or , wherein n is 1 or 2, and represents a covalent bond to L lm ; ** wherein represents a covalent bond to X 1 , and represents a covalent bond to X 2 .
- L is selected from the group consisting of: carbonyl group is the attachment point to X 1 .
- L is a divalent linker of Formula (L-n-i):
- L is a divalent linker of Formula (L-n-ii):
- L is one of the following: wherein the point of attachment indicated on the carbon-bound carbonyl group is the attachment point to X 1 .
- the compound is a compound in Table 1 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 1. In certain embodiments, the compound is a compound in Table 2 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 2. Tn certain embodiments, the compound is any one of compounds II-l to 11-26 in Table 2 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 3 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 3. In certain embodiments, the compound is a compound in Table 1, 2, or 3, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 1, 2, or 3.
- step 1 of Scheme 1-1 cyclopentylmethoxy aniline compound 1 is cyclized with thiocyanate 2 and bromine in acetic acid to form intermediate A (using, e.g., (1) AcOH and (2) Br 2 , AcOH).
- the synthetic route illustrated in Scheme 1-2 is a general method for preparing an intermediate B, which may be used in representative syntheses of a first series of benzothiazolyl benzenesulfonamide compounds shown in Table 1.
- step 1 of Scheme 1-2 the primary amino group of cyclohexyl carbamate compound 3 is protected by reaction with 4-nitrobenzenesulfonyl chloride 4 (using, e.g., TEA, DCM) to form nosyl-protected carbamate compound 5.
- step 2 the nosyl-protected carbamate compound 5 is selectively alkylated (using, e.g., CS2CO3, CH3I, DMF), as described in WO2021156792A1, which is incorporated herein by reference in its entirety, to form methylated nosyl-protected carbamate compound 6.
- step 3 methylated nosyl-protected carbamate compound 6 is deprotected (using, e.g., thioglycolic acid, CS2CO3, MeOH, DMF, 1 h, rt), as further described in WO2021156792 Al, to form benzyl (methylamino)cyclohexyl carbamate 7.
- step 4 reductive amination of the secondary amine of benzyl (methylamino)cyclohexyl carbamate 7 (using, e.g., sodium triacetoxyborohydride) with carbamate aldehyde 8 followed by removal of the Cbz group (using, e.g., Pd/C, H2) provides intermediate B.
- the synthetic route illustrated in Scheme 1-3 is a general method for preparing an intermediate C, which may be used in representative syntheses of a first series of benzothiazolyl benzenesulfonamide compounds shown in Table 1.
- step 1 of Scheme 1-3 cyclohexyl tert-butyl carbamate compound 9 is subjected to rhodium-catalyzed ether formation with ethyl diazoacetate 10 (using, e.g., cat. Rti2(AcO)4, DCM), as described in WO2023125121A1, which is incorporated herein by reference in its entirety, to form ethyl (cyclohexylmethoxy)acetate compound 11.
- ethyl (cyclohexylmethoxy) acetate compound 11 is partially reduced (using, e.g., DIBAL-H, -78 °C) to provide aldehyde compound 12.
- step 3 reductive amination of aldehyde compound 12 with (methylamino)cyclohexyl carbamate compound 13 (using, e.g., sodium triacetoxyborohydride) followed by removal of the Cbz group (using, e g., Pd/C, H2) provides intermediate C.
- (methylamino)cyclohexyl carbamate compound 13 using, e.g., sodium triacetoxyborohydride
- removal of the Cbz group using, e g., Pd/C, H2
- step 1 of Scheme 1-4 reductive amination of the primary amine of intermediate A (using, c.g., (1) TiCl(Oz-Pr)s, DCM; (2) NaBH(OAc)s) with benzaldehyde 14, as described in W02023028077A1, forms dimethoxybenzene compound 15.
- dimethoxybenzene compound 15 is converted to sulfonamide 17 by reaction with sulfonyl chloride 16 (using, e.g., (1) LiHMDS, THF -78 °C, 30 min, 0 °C; (2) -78 °C solution addition (THF) followed by 3 h RT), as further described in W02023028077A1.
- step 3 sulfonamide 17 is subjected to nucleophilic aromatic substitution (SNAr) with intermediate B or intermediate C (using, e.g., DMSO, RT), as further described in W02023028077A1, to form cyclohexyl amino compound 18.
- SNAr nucleophilic aromatic substitution
- step 4 cyclohexyl amino compound 18 is globally deprotected (using, e.g., formic acid), as further described in W02023028077A1, to form primary amino compound 19.
- step 5 primary amino compound 19 is reacted with (17?,4r)-4-(4-(((lS,47?)-4-(2-((2S,3S)-l- methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 21.
- HATU HATU
- DIPEA DIPEA
- step 1 of Scheme 1-5 reductive amination of the primary amine of bromo benzothiazole 22 (using, e.g., (1) TiCl(Oz-Pr)3, DCM; (2) NaBH(OAc)s) with benzaldehyde 14, as described in W02023028077A1, forms dimethoxybenzene compound 23.
- step 2 dimethoxybenzene compound 23 is converted to sulfonamide 24 by reaction with sulfonyl chloride 16 (using, e.g., (1) LiHMDS, THF -78 °C, 30 min, 0 °C; (2) -78 °C solution addition (THF) followed by 3 h RT), as further described in W02023028077A1.
- step 3 sulfonamide 24 is subjected to nucleophilic aromatic substitution (SNAr) with cyclohexyl bis-amino compound 25 (using, e.g., DMSO, RT), as further described in W02023028077A1, to form cyclohexyl amino compound 26.
- SNAr nucleophilic aromatic substitution
- step 4 cyclohexyl amino compound 26 is subjected to a Cu- catalyzed coupling reaction with /c/'Z-butyl carbamate compound 27 (using, e.g., 2 mol% Cui, 2 mol% DPEO ligand, 1.2 eq. NaO/Bu, 4 A MS, dioxane, 60 °C), as described in J. Am. Chem. Soc., 2019, 141, 3541, which is incorporated herein by reference in its entirety, to form alkyl aryl ether compound 28.
- /c/'Z-butyl carbamate compound 27 using, e.g., 2 mol% Cui, 2 mol% DPEO ligand, 1.2 eq. NaO/Bu, 4 A MS, dioxane, 60 °C
- alkyl aryl ether compound 28 is globally deprotected (using, e.g., formic acid), as further described in W02023028077A1, to form primary amino compound 29.
- primary amino compound 29 is reacted with (17?,4r)-4-(4-(((15,4A)-4-(2- ((2S,35)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3- carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane-1 -carboxylic acid 20 (using, e.g, HATU, DIPEA) to form amide 30.
- step 1 of Scheme 2-1 amine intermediate 31, prepared as described in US20170275275A1, which is incorporated herein by reference in its entirety, is reacted with (17?,4r)-4-(4-(((lS,47?)-4-(2-((2£,3S)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3- carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 32.
- HATU HATU
- DIPEA DIPEA
- step 1 of Scheme 2-2 4-iodopyridine 33 is converted to bromo biaryl intermediate 35 by reaction with aryl boronic acid 34 in a Suzuki -Miyaura coupling reaction (using, e.g., Pd(PPh3)4).
- step 2 bromo biaryl intermediate 35 is converted to triaryl intermediate 37 by reaction with aryl boronic acid 36 in a Suzuki -Miyaura coupling reaction (using, e.g., Pd(PPh3)4).
- step 3 the nitrile moiety in triaryl intermediate 37 is hydrogenated (using, e.g., 0.5 mol% [Ru(cod)methylallyl2], 0.5 mol% DPPF, 10 mol% /BuOK, H2, toluene), as described in Chemistry: A European Journal, 2008, 14, 9491, which is incorporated herein by reference in its entirety, to form benzyl amine 38.
- step 4 the methyl ether in benzyl amine 38 is removed (using, e.g., 1 -decanethiol, NaOH, DMSO, heat) to form phenol intermediate 39.
- step 5 the benzyl amine moiety in phenol intermediate 39 is selectively protected with Boc via a biphasic reaction (using, e.g., BOC2O, dioxane, NaOH) to form tert-butyl carbamate compound 40.
- step 6 Zc/7-butyl carbamate compound 40 is subjected to nucleophilic aromatic substitution (SNAr) with commercially available thiadiazolyl benzenesulfonamide 41 (using, e.g., K2CO3, DMSO) to form thiadiazolyl Zc77-butyl carbamate compound 42.
- SNAr nucleophilic aromatic substitution
- step 7 the BOC protecting group is removed from thiadiazolyl Zt'rt-butyl carbamate compound 42 (using, e.g., HC1) to provide benzyl amine 43.
- step 7 the benzyl amine 43 is reacted with (U?,4r)-4-(4-(((15,4J?)- 4-(2-((25 , ,3 ⁇ S)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido)ethoxy) cyclohexyl)oxy)butanamido) cyclohexane-1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 44.
- step 1 of Scheme 2-3 reductive amination of benzyl amine 43, which may be prepared as described above in reference to Scheme 2-2, with piperidinyl aldehyde 45 (using, e.g., NaBEU) forms the thiadiazolyl piperidinyl compound 46.
- step 2 the secondary amino moiety in thiadiazolyl piperidinyl compound 46 is protected as a trifluoroacetamide (e.g., trifluoroacetic anhydride 47) to form trifluoroacetylated compound 48.
- trifluoroacetamide e.g., trifluoroacetic anhydride 47
- step 3 trifluoroacetylated compound 48 is reacted with (17?,4r)-4-(4-(((lS,47?)-4-(2-((2S',3S)-l-methyl-5-oxo-2-(pyridin-3- yl)pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cy cl ohexane-1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) followed by removal of the trifluoroacetamide protecting group (using, e.g., MeOH, K2CO3) the to form amide 49.
- HATU, DIPEA trifluoroacetamide protecting group
- step 1 of Scheme 3-1 /erZ-butyl thiazol-2-ylcarbamate 50 is deprotonated (using, e.g., LiHMDS) and reacted with sulfonyl chloride 51 to form sulfonamide intermediate D.
- deprotonated using, e.g., LiHMDS
- sulfonyl chloride 51 to form sulfonamide intermediate D.
- step 1 of Scheme 3-2 2,4-difluoro-5-nitropyridine 52 is subjected to nucleophilic aromatic substitution (SNAr) with mono-BOC protected 1,4-diamine 53 (using, c.g., K2CO3, DMF) followed by reduction of the nitro group (using, e.g, Na2S2C>4, EtOH/H2O) to form amino pyridine compound 54.
- amino pyridine compound 54 is cross-coupled with sulfonamide intermediate D (using, e.g, (1) Pd(OAc)2, rac-BINAP) followed by global acid- mediated deprotection (using, e.g., HC1) to form amino compound 55.
- step 3 amino compound 55 is reacted with (lA,4/')-4-(4-(((15',47?)-4-(2-((2S,35)-l-methyl-5-oxo-2-(pyridin-3- yl)pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 56.
- step 1 of Scheme 3-3 the benzyl amino group of commercially available 6- (aminomethyl)-4-chloropyridin-3-amine 57 is protected (using, e.g., BOC2O) to provide Boc- protected amino pyridine compound 58.
- Boc-protected amino pyridine compound 58 is reacted with 3-furanylboronic acid under Suzuki-Miyaura coupling conditions to form bi-aryl compound 59.
- bi-aryl compound 59 is cross-coupled with sulfonamide intermediate D (using, e.g, (1) Pd(OAc)2, rac-BINAP) followed by global acid-mediated deprotection (using, c.g, HC1) to form amino compound 60.
- sulfonamide intermediate D using, e.g, (1) Pd(OAc)2, rac-BINAP
- global acid-mediated deprotection using, c.g, HC1
- step 4 amino compound 60 is reacted with (lA,4r)-4-(4-(((lS,4A)-4-(2-((25,35 Y )-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido) ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, c.g., HATU, DIPEA) to form amide 61.
- an antibody, or antigen-binding fragment thereof that binds to a cotinine moiety.
- the term “anti-cotinine antibody or antigen-binding fragment thereof’ refers to an antibody, or antigen binding fragment thereof that binds to a cotinine moiety.
- Cotinine has the following structure:
- the term “cotinine moiety” refers to cotinine or an analog of cotinine.
- Compounds of Formula (I) described herein comprise a cotinine moiety linked via a linker to a Na v l .7-binding moiety.
- the cotinine moiety has the following structure: wherein R 1 is C1.4 alkyl or C3-6 cycloalkyl.
- R 1 is methyl, ethyl, 1- propyl, 2-propyl, 1-butyl, 2-butyl, or t-butyl.
- R 1 is methyl.
- R 1 is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
- antibody is used herein in the broadest sense to refer to molecules with an immunoglobulin-like domain (for example IgG, IgM, IgA, IgD or IgE) and includes monoclonal, recombinant, polyclonal, chimeric, human, humanised, multispecific antibodies, including bispecific antibodies, and heteroconjugate antibodies; a single variable domain (e.g., a domain antibody (DAB)), antigen binding antibody fragments, Fab, F(ab’)2, Fv, disulphide linked Fv, single chain Fv, disulphide-linked scFv, diabodies, TANDABS, etc. and modified versions of any of the foregoing (for a summary of alternative “antibody” formats see Holliger and Hudson, Nature Biotechnology, 2005, 23(9): 1126-1136).
- DAB domain antibody
- the term, full, whole or intact antibody refers to a heterotetrameric glycoprotein with an approximate molecular weight of 150,000 daltons.
- An intact antibody is composed of two identical heavy chains (HCs) and two identical light chains (LCs) linked by covalent disulphide bonds. This H2L2 structure folds to form three functional domains comprising two antigen-binding fragments, known as ‘Fab’ fragments, and a ‘Fc’ crystallisable fragment.
- the Fab fragment is composed of the variable domain at the aminoterminus, variable heavy (VH) or variable light (VL), and the constant domain at the carboxyl terminus, CHI (heavy) and CL (light).
- the Fc fragment is composed of two domains formed by dimerization of paired CH2 and CH3 regions.
- the Fc may elicit effector functions by binding to receptors on immune cells or by binding Clq, the first component of the classical complement pathway.
- the five classes of antibodies IgM, IgA, IgG, IgE and IgD are defined by distinct heavy chain amino acid sequences, which are called p, a, y, s and 8 respectively, each heavy chain can pair with either a K or X light chain.
- the majority of antibodies in the serum belong to the IgG class, there are four isotypes of human IgG (IgGl, IgG2, IgG3 and IgG4), the sequences of which differ mainly in their hinge region.
- CDRs are defined as the complementarity determining region amino acid sequences of an antibody or antigen binding fragment thereof. These are the hypervariable regions of immunoglobulin heavy and light chains. There are three heavy chain and three light chain CDRs (or CDR regions) in the variable portion of an immunoglobulin. Thus, “CDRs” as used herein refers to all three heavy chain CDRs, all three light chain CDRs, all heavy and light chain CDRs, or at least two CDRs.
- Table 4 represents one definition using each numbering convention for each CDR or binding unit. It should be noted that some of the CDR definitions may vary depending on the individual publication used.
- the anti-cotinine antibody is humanized.
- the Fc region of the anti-cotinine antibody is modified to increase ADCC activity, ADCP activity, and/or CDC activity, suitable modifications of which are provided below.
- the Fc region of the anti-cotinine antibody is modified to increase ADCC activity.
- Fc engineering methods can be applied to modify the functional or pharmacokinetics properties of an antibody. Effector function may be altered by making mutations in the Fc region that increase or decrease binding to Clq or Fey receptors and modify CDC or ADCC activity respectively. Modifications to the glycosylation pattern of an antibody can also be made to change the effector function. The in vivo half-life of an antibody can be altered by making mutations that affect binding of the Fc to the FcRn (neonatal Fc receptor).
- Effector function can be assessed in a number of ways including, for example, evaluating ADCC effector function of antibody coated to target cells mediated by Natural Killer (NK) cells via FcyRIII, or monocytes/macrophages via FcyRI, or evaluating CDC effector function of antibody coated to target cells mediated by complement cascade via Clq.
- NK Natural Killer
- an antibody, or antigen binding fragment thereof, of the present invention can be assessed for ADCC effector function in a Natural Killer cell assay.
- Examples of assays to determine CDC function include those described in J Imm Meth, 1995, 184: 29-38.
- amino acid residues in Fc regions, in antibody sequences or full-length antigen binding protein sequences are numbered according to the EU index numbering convention.
- Enhanced CDC Fc engineering can be used to enhance complement-based effector function.
- Fc engineering can be used to enhance complement-based effector function.
- K326W/E333S; S267E/H268F/S324T; and IgGl/IgG3 cross subclass can increase Clq binding;
- E345R Diebolder et al., Science, 2014, 343: 1260-1293
- E345R/E430G/S440Y results in preformed IgG hexamers (Wang et al., Protein Cell, 2018, 9(1): 63-73).
- Fc engineering can be used to enhance ADCC.
- Fc engineering can be used to enhance ADCC.
- F243L/R292P/Y300L/V305I/P396L; S239D/I332E; and S298A/E333A/K334A increase FcyRIIIa binding
- S239D/I332E/A330L increases FcyRIIIa binding and decreases FcyRIIb binding
- G236A/S239D/I332E improves binding to FcyRIIa, improves the FcyRIIa/FcyRIIb binding ratio (activating/inhibitory ratio), and enhances phagocytosis of antibody-coated target cells by macrophages.
- the chimeric antibody, or antigen binding fragment thereof comprises an IgGl CHI domain, an IgG3 CH2 domain, and an IgG3 CH3 domain. In a further embodiment, the chimeric antibody, or antigen binding fragment thereof, comprises an IgGl CHI domain, an IgG3 CH2 domain, and an IgG3 CH3 domain except for position 435 that is histidine.
- the chimeric antibody, or antigen binding fragment thereof comprises an IgGl CHI domain and at least one CH2 domain from IgG3.
- the chimeric antibody, or antigen binding fragment thereof comprises an IgGl CHI domain and the following residues, which correspond to lgG3 residues, in a CH2 domain: 274Q, 276K, 296F, 300F and 339T.
- the chimeric antibody, or antigen binding fragment thereof also comprises 356E, which corresponds to an IgG3 residue, within a CH3 domain.
- the antibody, or antigen binding fragment thereof also comprises one or more of the following residues, which correspond to IgG3 residues within a CH3 domain: 358M, 384S, 392N, 397M, 4221, 435R, and 436F.
- Also provided is a method of producing an antibody, or antigen binding fragment thereof, according to the invention comprising the steps of: a) culturing a recombinant host cell comprising an expression vector comprising a nucleic acid sequence encoding a chimeric Fc region having both IgGl and IgG3 Fc region amino acid residues (e.g. as described above); and b) recovering the antibody, or antigen binding fragment thereof.
- Such methods for the production of antibody, or antigen binding fragment thereof, with chimeric heavy chain constant regions can be performed, for example, using the COMPLEGENT technology system available from BioWa, Inc. (Princeton, NJ) and Kyowa Hakko Kirin Co., Ltd.
- the COMPLEGENT system comprises a recombinant host cell comprising an expression vector in which a nucleic acid sequence encoding a chimeric Fc region having both IgGl and IgG3 Fc region amino acid residues is expressed to produce an antibody, or antigen binding fragment thereof, having enhanced CDC activity, i.e.
- CDC activity is increased relative to an otherwise identical antibody, or antigen binding fragment thereof, lacking such a chimeric Fc region, as described in WO 2007/011041 and US 2007/0148165, each of which are incorporated herein by reference.
- CDC activity may be increased by introducing sequence specific mutations into the Fc region of an IgG chain.
- Also provided is a method of producing an antibody, or antigen binding fragment thereof, according to the invention comprising the steps of: a) culturing a recombinant host cell comprising an expression vector comprising a nucleic acid encoding the antibody, or antigen binding fragment thereof, optionally wherein the FUT8 gene encoding alpha- 1,6-fucosyltransf erase has been inactivated in the recombinant host cell; and b) recovering the antibody, or antigen binding fragment thereof.
- Such methods for the production of an antibody, or antigen binding fragment thereof can be performed, for example, using the POTELLIGENT technology system available from BioWa, Inc. (Princeton, NJ) in which CHOK1SV cells lacking a functional copy of the FUT8 gene produce monoclonal antibodies having enhanced ADCC activity that is increased relative to an identical monoclonal antibody produced in a cell with a functional FUT8 gene as described in US Patent No. 7,214,775, US Patent No. 6,946,292, WO 00/61739 and WO 02/31240, all of which are incorporated herein by reference. Those of ordinary skill in the art will also recognize other appropriate systems.
- the antibody, or antigen binding fragment thereof is produced in a host cell in which the FUT8 gene has been inactivated. In a further embodiment, the antibody, or antigen binding fragment thereof, is produced in a -I- FUT8 host cell. In a further embodiment, the antibody, or antigen binding fragment thereof, is afucosylated at Asn297 (IgGl).
- an antibody, or antigen binding fragment thereof comprising a heavy chain constant region that comprises a both a mutated and chimeric heavy chain constant region, individually described above.
- an antibody, or antigen binding fragment thereof comprising at least one CH2 domain from IgG3 and one CH2 domain from IgGl, and wherein the IgGl CH2 domain has one or more mutations at positions selected from 239, 332 and 330 (for example the mutations may be selected from S239D, I332E and A330L), such that the antibody, or antigen binding fragment thereof, has enhanced effector function, e.g.
- Another aspect of the invention provides a method of treating or preventing a Na v 1.7- associated disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigenbinding fragment thereof.
- a heterobifunctional compound described herein such as a compound of Formula I
- the pain is chronic pain. In certain embodiments, the pain is acute pain. In certain embodiments, the pain is neuropathic pain. In certain other embodiments, the pain is inflammatory pain. In certain embodiments, the pain is arthritis pain. In certain embodiments, the pain is arthritis pain selected from osteoarthritis pain and rheumatoid arthritis pain. [0283] In certain other embodiments, the pain is pain due to cancer. In certain embodiments, the pain is due to a cancer selected from the group consisting of a solid tumor, leukemia, and lymphoma.
- the pain is due to a cancer selected from the group consisting of a bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, endometrial cancer, esophageal cancer, leukemia, lung cancer, liver cancer, skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, renal cancer, stomach cancer, testicular cancer, and uterine cancer.
- a cancer selected from the group consisting of a bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, endometrial cancer, esophageal cancer, leukemia, lung cancer, liver cancer, skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, renal cancer, stomach cancer, testicular cancer, and uterine cancer.
- the pain is complex regional pain syndrome.
- the complex regional pain syndrome is reflex sympathetic dystrophy pain.
- the pain is trauma pain.
- the pain is due to surgery.
- the pain is a neuropathic pain selected from the group consisting of low back pain, hip pain, leg pain, non-herpetic neuralgia, post-herpetic neuralgia, diabetic neuropathy pain, lumbosacral radiculopathy pain, nerve injury-induced pain, acquired immune deficiency syndrome (AIDS) related neuropathic pain, head trauma pain, phantom limb pain, multiple sclerosis pain, root avulsion pain, painful traumatic mononeuropathy, painful polyneuropathy, thalamic pain syndrome, post-stroke pain, central nervous system injury pain, post-surgical pain, carpal tunnel syndrome pain, trigeminal neuralgia pain, post mastectomy syndrome pain, post-thoracotomy syndrome pain, stump pain, repetitive motion pain, neuropathic pain associated hyperalgesia and allodynia, drug-induced pain, toxin-caused nerve injury pain, chemotherapy-caused nerve injury pain, and combinations thereof.
- AIDS acquired immune deficiency syndrome
- the method may be further characterized according to the amount of reduction in pain intensity relative to pain observed without performing the method. Accordingly, in certain embodiments, the method is characterized by achieving at least a 20% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least a 40% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least a 60% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least an 80% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least a 90% reduction in pain intensity relative to pain observed without performing the method.
- the method may be further characterized according to the duration of reduction in pain intensity. Accordingly, in certain embodiments, the reduction in pain intensity lasts for at least 1 week. In certain embodiments, the reduction in pain intensity lasts for at least 2 weeks. In certain embodiments, the reduction in pain intensity lasts for at least 4 weeks. In certain embodiments, the reduction in pain intensity lasts for at least 2 months. In certain embodiments, the reduction in pain intensity lasts for at least 3 months. In certain embodiments, the reduction in pain intensity lasts for at least 6 months.
- the reduction in pain intensity lasts for at a duration of 2 months to six months. In certain embodiments, the reduction in pain intensity lasts for a duration of 3 months to 9 months. In certain embodiments, the reduction in pain intensity lasts for a duration of 6 months to 9 months. In certain embodiments, the reduction in pain intensity lasts for a duration of 6 months to 12 months.
- Another aspect of the invention provides a method of depleting voltage-gated sodium channel Na v 1.7-expressing cells, wherein the method comprises contacting the cells with an effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
- a heterobifunctional compound described herein such as a compound of Formula I
- the compound and the antibody, or antigen-binding fragment thereof are administered sequentially.
- the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a heavy chain variable region (VH) having SEQ ID NO: 7, and the light chain comprising a light chain variable region (VL) having SEQ ID NO: 8.
- the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase ADCC activity.
- the substitution in the Fc region is S239D/I332E, wherein residue numbering is according to the EU Index.
- Another aspect of the invention provides for the use of a heterobifunctional compound described herein (such as a compound of Formula I, or other compounds in Section I) in the manufacture of a medicament.
- the medicament is for treating a disorder described herein, such as pain.
- Another aspect of the invention provides for the use of a heterobifunctional compound described herein (such as a compound of Formula I, or other compounds in Section I) for treating a medical disorder, such as a medical disorder described herein, such as pain.
- a heterobifunctional compound described herein such as a compound of Formula I, or other compounds in Section I
- a medical disorder such as a medical disorder described herein, such as pain.
- Another aspect of the invention provides a heterobifunctional compound described herein (such as a compound of Formula I, or other compounds in Section I) for use in treating a medical disorder, such as a medical disorder described herein, such as pain.
- a heterobifunctional compound described herein such as a compound of Formula I, or other compounds in Section I
- a medical disorder such as a medical disorder described herein, such as pain.
- Another aspect of the invention provides a combination comprising a heterobifunctional compound described herein (e.g., a compound of Formula I) and an anti- cotinine antibody, or antigen-binding fragment thereof.
- a heterobifunctional compound described herein e.g., a compound of Formula I
- an anti- cotinine antibody, or antigen-binding fragment thereof e.g., an anti- cotinine antibody, or antigen-binding fragment thereof.
- the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a CDR1 having SEQ ID NO: 1, a CDR2 having SEQ ID NO: 2, and a CDR3 having SEQ ID NO: 3, and the light chain comprising a CDR1 having SEQ ID NO: 4, a CDR2 having SEQ ID NO: 5, and a CDR3 having SEQ ID NO: 6.
- the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase ADCC activity.
- the anti-cotinine antibody has a heavy chain comprising SEQ ID NO: 9 and a light chain comprising SEQ ID NO: 10.
- Heterobifunctional compounds may be evaluated for biological activity using one or more of the assays described below.
- Assay 1 Antibody Dependent Cellular Cytotoxicity Reporter Assay
- An antibody dependent cellular cytoxocity reporter assay is conducted using the following four assay components: (i) ARM compound of Formula (I) targeting Na v 1.7 (concentrations ranging from 1 pM to 10 pM) (ii) anti-cotinine antibody having a heavy chain sequence of SEQ ID NO: 11 and a light chain sequence of SEQ ID NO: 12 (rabbit variable region with human IgGl Fc domain containing a DE mutation (S239D/I332E)) (concentrations ranging from 0.01 pg / mb to 200 pg / mL); (iii) target cells: cells engineered to overexpress human Na v 1.7 (typically 1000-20,000 cells per well) and (iv) reporter cells: Reagents are combined in a final volume of 20 pL in a 384 - well tissue culture treated plate.
- Luminescence signal is measured on a microplate reader and signal background is calculated by dividing the signal of a test well by the signal obtained when no heterobivalent compound of Formula (I) was added.
- Assay 2 Binding of Anti-Cotinine Antibody to ARM Compounds of Formula I Measured by Surface Plasmon Resonance (SPR)
- Anti-cotinine antibodies having a heavy chain sequence of SEQ ID NO: 11 and a light chain sequence of SEQ ID NO: 12 can be captured on one or more flow cells of a protein A sensor chip (Cytiva) using a Biacore T200 while reserving flow cell 1 as a reference. Following capture, a 3000 second wait step is included to reduce drift during compound analysis.
- ARM compounds e.g., of Formula I are then injected at 100 pL/min with 200 and 2000 second association and dissociation times.
- the entire experiment may be run at 37°C with running buffer containing 10 mM HEPES pH 7.4, 150 mM NaCl, 0.005% P20 and 1% DMSO.
- ARM compounds of Formula (I) are titrated with a top concentration of 200 nM using a 3-fold 5-point dilution series and a corresponding 5-injection buffer cycle is run for blank subtraction. Data are double referenced by subtracting the response of the reference flow cell from that of the antibody-containing flow cell and subsequently subtracting the referenced blank sensorgrams. Following compound analysis, the surface is regenerated using a 30 second injection of pH 1 .5 glycine at a flow rate of 30 pL/min after which antibody is re-captured.
- the experiment may be run using Biacore T200 control software and evaluated using Biacore T200 Evaluation software. Curves are fit with a 1 : 1 kinetic binding model to obtain k on (1/Ms), koff (1/s), Kd (M) determined as koff/kon, and residence time determined as l/k 0 ⁇ r. (s).
- Anti-cotinine antibodies having a heavy chain sequence of SEQ ID NO: 9 and a light chain sequence of SEQ ID NO: 10 (humanized version) or a heavy chain sequence of SEQ ID NO: 11 and a light chain sequence of SEQ ID NO: 12 (rabbit version) may be captured via the Fc domain to a protein A or A/G surface on flow cell 2 of a CM5 sensor chip using the Biacore 8k. Both antibodies may have a human IgGl Fc domain containing a DE mutation (S239D/I332E).
- ARM compounds of Formula (I) are then flowed over the captured antibodies at varying top concentrations ranging from 250 nM to 4 pM. The top concentration is diluted 4-fold over 5 dilutions (with a 0 nM compound cycle included to blank subtract the data).
- Association and dissociation times of the ARM compounds of Formula (I) may vary between experiments as follows: reference 1- association for 600 seconds and dissociation for 1200 seconds at 20pl/min; reference 2- association for 240 seconds and dissociation for 300 seconds at 30pl/min; reference 3- association for 360 seconds and dissociation for 600 seconds at 30pl/min.
- Anti-cotinine antibody bound to the protein A or A/G surface is regenerated using 50mM NaOH.
- Experiments are run at 25°C at pH 7.4 using HBS-EP+ buffer using Biacore 8K control software and evaluated using Biacore Insight Evaluation software. Curves are fit with the 1 : 1 kinetic fit inherent to the software, utilising a local Rmax and local drift setting.
- Heterobifunctional compounds described herein such as a compound of Formula I, or other compounds in Section I
- additional therapeutic agents such as pain.
- the present invention provides a method of treating a disclosed disease or condition comprising administering to a patient in need thereof an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof and co-administering simultaneously or sequentially an effective amount of one or more additional therapeutic agents, such as those described herein.
- the method further comprises administering an anti-cotinine antibody, or antigen-binding fragment thereof.
- the method includes co-administering one additional therapeutic agent.
- the method includes co-administering two additional therapeutic agents.
- the combination of the disclosed compound and the additional therapeutic agent or agents acts synergistically.
- One or more other therapeutic agent may be administered separately from a compound or composition of the invention, as part of a multiple dosage regimen.
- one or more other therapeutic agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition.
- one or more other therapeutic agent and a compound or composition of the invention may be administered simultaneously, sequentially or within a period of time from one another, for example within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 18, 20, 21, 22, 23, or 24 hours from one another.
- one or more other therapeutic agent and a compound or composition of the invention are administered as a multiple dosage regimen more than 24 hours apart.
- Additional therapeutic agents for treating pain include, for example, an opioid analgesic (e.g., alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl; heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphano
- the doses and dosage regimen of the active ingredients used in the combination therapy may be determined by an attending clinician.
- the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) are administered in doses commonly employed when such agents are used as monotherapy for treating the disorder.
- the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) are administered in doses lower than the doses commonly employed when such agents are used as monotherapy for treating the disorder.
- the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) are present in the same composition, which is suitable for oral administration.
- the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) may act additively or synergistically.
- a synergistic combination may allow the use of lower dosages of one or more agents and/or less frequent administration of one or more agents of a combination therapy.
- a lower dosage or less frequent administration of one or more agents may lower toxicity of the therapy without reducing the efficacy of the therapy.
- kits comprising a therapeutically effective amount of the compound described herein (such as a compound of Formula I, or other compounds in Section I), a pharmaceutically acceptable carrier, vehicle or diluent, and optionally at least one additional therapeutic agent listed above.
- the kit further comprises an anti -cotinine antibody, or antigen-binding fragment thereof.
- the invention provides pharmaceutical compositions, which comprise a therapeutically effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
- the pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary,
- phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
- wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
- antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
- water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
- oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), le
- Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration.
- the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
- the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
- the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
- a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and poly anhydrides; and a compound of the present invention.
- an aforementioned formulation renders orally bioavailable a compound of the present invention.
- Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients.
- the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
- Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient.
- a compound of the present invention may also be administered as a bolus, electuary or paste.
- the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fdlers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants, such
- compositions may also comprise buffering agents.
- Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
- a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
- Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
- Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
- the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried.
- compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
- These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
- embedding compositions which can be used include polymeric substances and waxes.
- the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
- Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
- the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
- inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents
- the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
- adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
- Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
- suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
- Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
- suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
- Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
- Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
- the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
- the ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
- excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
- Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
- Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
- Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body.
- dosage forms can be made by dissolving or dispersing the compound in the proper medium.
- Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
- Ophthalmic formulations are also contemplated as being within the scope of this invention.
- compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
- aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
- polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
- vegetable oils such as olive oil
- injectable organic esters such as ethyl oleate.
- Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
- compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
- adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
- Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
- biodegradable polymers such as polylactide-polyglycolide.
- Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
- the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier.
- the preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred.
- parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
- systemic administration means the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient’s system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
- These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intraci sternal ly and topically, as by powders, ointments or drops, including buccally and sublingually.
- the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
- compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
- the selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
- a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
- the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
- a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
- the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg.
- the effective amount may be less than when the agent is used alone.
- the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day.
- the invention further provides a unit dosage form (such as a tablet or capsule) comprising a compound described herein in a therapeutically effective amount for the treatment of a medical disorder described herein.
- a unit dosage form such as a tablet or capsule
- Variable light chain amino acid sequence (SEQ ID NO: 8): DIQMTQSPSSLSASVGDRVTITCQSSQSVYSAKLSWYQQKPGKAPKLLIYYGSTLASGVP SRFSGSGSGTQFTLTISSLQPEDFATYYCQGTFYGPDWYFAFGGGTKVEIK
- Heavy chain amino acid sequence (SEQ ID NO: 9): EVQLVESGGGLVQPGGSLRLSCAASGFTFSNYWMSWVRQAPGKGLEWVGDIHGNRGF NYHASWAKGRFTVSRSKNTLYLQMNSLRAEDTAVYYCAKADDSGSHDIWGQGTLVTV SSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ S SGL YSL S S VVTVP S S SLGTQT YICNVNHKP SNTKVDKKVEPKSCDKTHTCPPCP APELL GGPDVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPRE EQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPEEKTISKAKGQPREPQVYTL PP SRDELTKNQ VSLTCLVKGF YP SDIAVEWE SNGQ
- OQOLVESG GRLVTPGGSLTLTCTASGFSLNNYWMSWVRQAPGKGLEWIGDIHGNRGF NYHASWAKGRFTVSRTSTTVDLRMTSLTTEDTAIYFCARADDSGSHDIWGPGTLVTVS S AKTT APS VYPLAP VC GDTTGS S VTLGCL VKGYFPEP VTLTWNSGSL S SGVHTFP AVLQ S
- Step 1 Preparation of tert-butyl 3-(2-((25',3KS)-l-methyl-5-oxo-2-(pyridin-3-yl) pyrrolidine-3-carboxamido)ethoxy)propanoate (3).
- Step 2 Preparation of 3-(2-((25,35)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-
- Step 3 Preparation of (25,3 ⁇ )-A f -(2-(3-(4-(2-(((4-(4-(4-(Af-(l,2,4-thiadiazol-5-yl) sulfamoyl)-2-cyanophenoxy)-3'-(trifluoromethyl)-[l,l'-biphenyl]-3-yl)pyridin-2-yl)niethyl) amino)ethyl)piperidin-l-yl)-3-oxopropoxy)ethyl)-l-methyl-5-oxo-2-(pyridin-3-yl) pyrrolidine-3-carboxamide (11-27).
- Blood collection and plasma isolation (simple extraction method): 30-40 pl of blood was collected in a tube containing anticoagulant (4% EDTA, w/v) at specified time points via saphenous vein. The collected blood samples were centrifuged at 13,000 rpm for 5 minutes set at 4 °C. Plasma samples were stored at dry ice/-80 °C until bioanalysis.
- Naive WB difference - pre dose WB difference was defined as the MIA window.
- Compounds were co-dosed intravenously on day 7 at 1 mg/kg with 50 mg/kg of antibody. Animals were weighed at the start of the study, pre- and post-MIA to monitor any weight loss post-MIA. Animals were then weighed daily from day 7 to 10.
- MIA-induced hyperalgesia effect was evaluated as a comparison of weight load on hind paws (Static Weight Bearing). Specifically, weight bearing (g) readings were taken for both right and left hind paws and the difference calculated. Data are expressed as % ratio ipsilateral/contralateral ((WB left/WB right)* 100) (mean ⁇ s.e.m ).
- Weight bearing readings were taken on Day 7 at 2 hours post administration, on Day 8 at 24 hours post administration, on Day 9 at 48 hours post administration and on Day 10 at 72 hours post administration. Data was analysed by comparing treatment groups to vehicle control at each time point. Statistical analysis was done by repeated measures ANOVA followed by Dunnett’s test using GraphPad Prism (p ⁇ 0.05 considered significant).
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Pain & Pain Management (AREA)
- Rheumatology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain.
Description
HETEROBIFUNCTIONAL MOLECULES FOR BINDING NAvl.7 AND METHODS OF TREATING MEDICAL CONDITIONS USING SAME
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of and priority to United States Provisional Patent Application serial number 63/711,389, filed October 24, 2024, and United States Provisional Patent Application serial number 63/571 ,523, filed March 29, 2024; the contents of each of which are hereby incorporated by reference in their entirety.
REFERENCE TO AN ELECTRONIC SEQUENCE LISTING
[0002] This application contains a sequence listing which has been submitted electronically in ST.26 format and is hereby incorporated by reference in its entirety (said ST.26 copy, created on March 24, 2025, is named “216918_seqlist.xml” and is 15,201 bytes in size.
FIELD OF THE INVENTION
[0003] The invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain.
BACKGROUND
[0004] Pain can function as a protective mechanism that allows healthy human beings and animals to avoid tissue damage and/or prevent further damage to injured tissue. However, there are many instances in which pain persists beyond its usefulness. Such unnecessary suffering from pain can impair a subject’s physical mobility, mental performance, and even contribute to depression. Such unnecessary suffering from pain can be due to acute pain and/or chronic pain. Such pain can also be characterized according to whether the pain is neuropathic pain or nociceptive pain. Substantial resources have been devoted over the years to researching the causes of various types of pain and to the development of medicine to attenuate pain experienced by a patient. Exemplary classes of common pain-relief medications include opioids, non-steroidal antiinflammatory agents, corticosteroids, and centrally acting agents such as anti -depressants and antiepileptics. However, existing therapies for treating pain are not effective for all patients and/or can have adverse side effects.
[0005] Voltage-gated sodium channel Nav1.7 is expressed at high levels in (i) nociceptive neurons at dorsal root ganglion and trigeminal ganglion and (ii) sympathetic ganglion neurons, and is involved in nociception. Molecules that inhibit Navl .7 have been reported for treatment of pain. See, for example, P.T. Nguyen et al. In Front. Pharmacol. (2022) vol. 13, article 842032; L.A. McDermott et al. InNeuron (2019) vol. 101(5), pages 905-919; and U.S. Patent 10,179,781.
[0006] The use of heterobifuctional compounds that are able to simultaneously bind a target cell-surface protein as well as an exogenous antibody protein are described in, for example, international patent application publication nos. WO 2018/134731, WO 2023/017484, and WO 2023/017483. Such heterobifuctional compounds are used with an anti-cotinine antibody to treat a variety of medical diseases and conditions. Additional heterobifunctional coti nine-containing compounds that bind voltage-gated sodium channel Na> 1.7 and an exogenous antibody protein are needed and would provide benefits to patients suffering from voltage-gated sodium channel Navl 7-associated diseases and conditions, such as pain.
[0007] The present invention addresses the foregoing need and provides other related advantages.
SUMMARY
[0008] The invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain. In particular, one aspect of the invention provides a collection of heterobifunctional cotinine- containing compounds represented by Formula I:
or a pharmaceutically acceptable salt thereof, where the variables are as defined in the detailed description. Further description of additional collections of heterobifunctional cotinine- containing compounds are described in the detailed description. The compounds may be part of a pharmaceutical composition comprising a pharmaceutically acceptable carrier.
[0009] The heterobifunctional compounds bind to voltage-gated sodium channel Navl .7. The heterobifunctional compounds also bind to an anti-cotinine antibody or fragment thereof that binds cotinine. In this way, the heterobifunctional compounds may be characterized as an antibody recruiting molecule (ARM).
[0010] Another aspect of the invention provides a method of treating or preventing a Navl .7- associated disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigenbinding fragment thereof.
[0011] Another aspect of the invention provides a method of treating or preventing a disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof, wherein the disease or condition is selected from pain, cough, acute itch, or chronic itch.
[0012] Another aspect of the invention provides a method of treating or preventing pain in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
[0013] Another aspect of the invention provides a method of increasing antibody-dependent cell cytotoxicity (ADCC) of voltage-gated sodium channel Nav1.7-expressing cells, wherein the method comprises contacting the cells with an effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
[0014] Another aspect of the invention provides a method of depleting voltage-gated sodium channel Nav1.7-expressing cells, wherein the method comprises contacting the cells with an effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
[0015] Another aspect of the invention provides a combination comprising a heterobifunctional compound described herein (such as a compound of Formula I) and an anti- cotinine antibody, or antigen-binding fragment thereof.
DETAILED DESCRIPTION
[0016] The invention provides heterobifunctional cotinine-containing compounds, pharmaceutical compositions, and methods of using same to treat medical conditions, such as pain. The practice of the present invention employs, unless otherwise indicated, conventional techniques of organic chemistry, pharmacology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology. Such techniques are explained in the literature, such as in “Comprehensive Organic Synthesis” (B.M. Trost & I. Fleming, eds., 1991- 1992); “Handbook of experimental immunology” (D.M. Weir & C.C. Blackwell, eds.); “Current protocols in molecular biology” (F.M. Ausubel etal., eds., 1987, and periodic updates); and “Current protocols in immunology” (J.E. Coligan et al., eds., 1991), each of which is herein incorporated by reference in its entirety.
[0017] Various aspects of the invention are set forth below in sections; however, aspects of the invention described in one particular section are not to be limited to any particular section. Further, when a variable is not accompanied by a definition, the previous definition of the variable controls.
Definitions
[0018] Compounds of the present invention include those described generally herein, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. These definitions apply regardless of whether a term is used by itself or in combination with other terms, unless otherwise indicated. Hence, the definition of “alkyl” applies to “alkyl” as well as the “alkyl” portions of “-O-alkyl” etc. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in “Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and “March’s Advanced Organic Chemistry”, 5th Ed.,
Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0019] The term “aliphatic” or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “cycloaliphatic”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
[0020] As used herein, the term “bicyclic ring” or “bicyclic ring system” refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or having one or more units of unsaturation, having one or more atoms in common between the two rings of the ring system. Thus, the term includes any permissible ring fusion, such as o/VAo-fused or spirocyclic. As used herein, the term “heterobicyclic” is a subset of “bicyclic” that requires that one or more heteroatoms are present in one or both rings of the bicycle. Such heteroatoms may be present at ring junctions and are optionally substituted, and may be selected from nitrogen (including N-oxides), oxygen, sulfur (including oxidized forms such as sulfones and sulfonates), phosphorus (including oxidized forms such as phosphates), boron, etc. In some embodiments, a bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. As used herein, the term “bridged bicyclic” refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge. As defined by IUPAC, a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal
atoms (excluding hydrogen). In some embodiments, a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom.
Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted. Exemplary bicyclic rings include:
[0021] Exemplary bridged bicyclics include:
[0022] The term “lower alkyl” refers to a C1-4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
[0023] The term “lower haloalkyl” refers to a C1-4 straight or branched alkyl group that is substituted with one or more halogen atoms.
[0024] The term “heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2/f-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)).
[0025] The term “unsaturated,” as used herein, means that a moiety has one or more units of unsaturation.
[0026] As used herein, the term “bivalent Ci-s (or Ci-e) saturated or unsaturated, straight or branched, hydrocarbon chain”, refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
[0027] The term “alkylene” refers to a bivalent alkyl group. An “alkylene chain” is a polymethylene group, i.e., -(CH2)n- wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0028] The term “-(Co alkylene)-“ refers to a bond. Accordingly, the term “-(C0-3 alkylene)-” encompasses a bond (i.e., Co) and a -(C1.3 alkylene)- group.
[0029] The term “alkenylene” refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0030] The term “halogen” means F, Cl, Br, or I.
[0031] The term “aryl” used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term “aryl” may be used interchangeably with the term “aryl ring.” In certain embodiments of the present invention, “aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may
bear one or more substituents. Also included within the scope of the term “aryl,” as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like. The term “phenylene” refers to a multivalent phenyl group having the appropriate number of open valences to account for groups attached to it. For example, “phenylene” is a bivalent phenyl group when it has two groups attached t “phenylene” is a trivalent phenyl group when it has three groups attached
The term “arylene” refers to a bivalent aryl group.
[0032] The terms “heteroaryl” and “heteroar-,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or
9 ring atoms; having 6, 10, or 14 z electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term “heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms “heteroaryl” and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where unless otherwise specified, the radical or point of attachment is on the heteroaromatic ring or on one of the rings to which the heteroaromatic ring is fused. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4/7 quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, and tetrahydroisoquinolinyl. A heteroaryl group may be mono- or bicyclic. The term “heteroaryl” may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted. The term “heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
[0033] The term “heteroarylene” refers to a multivalent heteroaryl group having the appropriate number of open valences to account for groups attached to it. For example, “heteroarylene” is a bivalent heteroaryl group when it has two groups attached to it; “heteroarylene” is a trivalent heteroaryl group when it has three groups attached to it.
[0034] As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro- 2/7 pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in N- -substituted pyrrolidinyl).
[0035] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, 2-oxa-6- azaspiro[3.3]heptane, and quinuclidinyl. The terms “heterocycle,” “heterocyclyl,” “heterocyclyl ring,” “heterocyclic group,” “heterocyclic moiety,” and “heterocyclic radical,” are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 37/ indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl. A heterocyclyl group may be mono- or bicyclic. The term “heterocyclylalkyl” refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted. The term “oxo-heterocyclyl” refers to a heterocyclyl substituted by one or more oxo group. The term “heterocyclylene” refers to a multivalent heterocyclyl group having the appropriate number of open valences to account for groups attached to it. For example, “heterocyclylene” is a bivalent heterocyclyl group when it has two groups attached to it; “heterocyclylene” is a trivalent heterocyclyl group when it has three groups attached to it. The term “oxo-heterocyclylene” refers to a multivalent oxo-heterocyclyl group having the appropriate number of open valences to account for groups attached to it.
[0036] As used herein, the term “partially unsaturated” refers to a ring moiety that includes at least one double or triple bond. The term “partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[0037] As described herein, compounds of the invention may contain “optionally substituted” moieties. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[0038] Each optional substituent on a substitutable carbon is a monovalent substituent independently selected from halogen; -(CH2)o 4R0; -(CH2)o 4OR0; -0(CH2)o-4R°, -0-(CH2)o- 4C(O)OR°; -(CH2)O-4CH(OR°)2; -(CH2)O 4SRO; -(CH2)O 4Ph, which may be substituted with R°; ~(CH2)o-40(CH2)o-iPh which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -(CH2)o-40(CH2)o-i-pyridyl which may be substituted with R°; -NO2; -CN; - N3; -(CH2)O-4N(R°)2; -(CH2)O 4N(RO)C(O)R°; -N(R°)C(S)R°; -(CH2)O- ^N(RO)C(O)NR°2; -N(RO)C(S)NR°2; -(CH2)O^N(R°)C(0)OR°; -N(R°)N(R°)C(O)R°; -N(R°)N(R°)C(O)NRO2; -N(R°)N(R°)C(O)OR°; -(CH2)o 4C(O)R°; -C(S)R°; -(CH2)O-IC(0)OR°; -(CH2)o 4C(O)SR°; -(CH2)o 4C(O)OSiR°3; -(CH2)o 4OC(O)R°; -OC(0)(CH2)o 4SR- SC(S)SR°; -(CH2)o 4SC(O)R°; -(CH2)O 4C(O)NRO 2; -C(S)NRO 2; -C(S)SR°; -SC(S)SR°, -(CH2)O^OC(0)NR0 2;
-C(O)N(OR°)R°; -C(O)C(O)R°; -C(O)CH2C(O)R°; -C(NOR°)R°; -(CH2)o 4SSR°; -(CH2)o 4S(O)2R°; -(CH2)O 4S(O)2ORO; -(CH2)O- ^OS(O)2RO; -S(O)2NRO 2; -S(O)(NR°)R°; - S(O)2N=C(NR°2)2; -(CH2)O 4S(O)R°; -N(R°)S(0)2NRO 2; -N(R°)S(O)2R°; -N(OR°)R°; -
C(NH)NR°2; -P(O)2RO; -P(O)RO 2; -OP(O)RO 2; -OP(O)(ORO)2; SiR°3; -(Ci^ straight or branched alkylene)O-N(R°)2; or -(Ci-4 straight or branched alkylene)C(O)O-N(R°)2.
[0039] Each R° is independently hydrogen, Ci-6 aliphatic, -CH2Ph, -0(CH2)o-iPh, -CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted by a divalent substituent on a saturated carbon atom of R° selected from =0 and =S; or each R° is optionally substituted with a monovalent substituent independently selected from halogen, (CH2)o 2R*, -(haloR*), (CH2)o 2OH, (CH2)o 2OR*, - (CH2)0 2CH(OR*)2; -O(haloR’), -CN, -N3, -(CH2)0 2C(O)R*, -(CH2)o 2C(O)OH, -(CH2)o- 2C(O)OR*, -(CH2)O-2SR*, -(CH2)O 2SH, -(CH2)O 2NH2, -(CH2)O 2NHR*, -(CH2)O 2NR’2, -NO2, -SiR*3, -OSiR*3, -C(O)SR* -(Ci-4 straight or branched alkylene)C(O)OR*, or -SSR*.
[0040] Each R* is independently selected from Ci^ aliphatic, -CH2Ph, -0(CH2)o iPh, or a 5- 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R* is unsubstituted or where preceded by halo is substituted only with one or more halogens; or wherein an optional substituent on a saturated carbon is a divalent substituent independently selected from =0, =S, =NNR*2, =NNHC(O)R*, =NNHC(O)OR*, =NNHS(O)2R\ =NR*, =N0R*, -O(C(R*2))2 3O-, or- S(C(R*2))2-3S-, or a divalent substituent bound to vicinal substitutable carbons of an “optionally substituted” group is -O(CR*2)2-3O-, wherein each independent occurrence of R* is selected from hydrogen, Ci-6 aliphatic or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0041] When R* is Ci-6 aliphatic, R* is optionally substituted with halogen, - R*, -(haloR*), -OH, -OR*, -O(haloR*), -CN, -C(O)OH, -C(O)OR*, -NH2, -NHR*, -NR*2, or -NO2, wherein each R* is independently selected from Ci-4 aliphatic, -CH2Ph, -0(CH2)o iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R* is unsubstituted or where preceded by halo is substituted only with one or more halogens.
[0042] An optional substituent on a substitutable nitrogen is independently -R1, -NR1’ 2, -
C(NH)NR1 2, or -N(R1)S(O)2R1; wherein each R1 is independently hydrogen, C1-6 aliphatic, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, two independent occurrences of R1, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; wherein when R1 is C1-6 aliphatic, R1 is optionally substituted with halogen, -R*, -(haloR*), -OH, -OR*, - O(haloR*), -CN, C(O)OH, C(O)OR*, NH2, NHR*, NR*2, or NO2, wherein each R* is independently selected from C1-4 aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R* is unsubstituted or where preceded by halo is substituted only with one or more halogens.
[0043] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphor sulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate,
malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
[0044] Further, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al., Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al., Journal of Pharmaceutical Sciences (1977) 66(1) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33 201-217; Anderson et al., The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference.
[0045] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N'(Ci-4alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
[0046] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. The invention includes compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
[0047] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Alternatively, a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis. Still further, where the molecule contains a basic functional group (such as amino) or an acidic functional group (such as carboxylic acid) diastereomeric salts are formed with an appropriate optically active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means known in the art, and subsequent recovery of the pure enantiomers.
[0048] Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. Chiral center(s) in a compound of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. Further, to the extent a compound described herein may exist as an atropisomer (e.g., substituted biaryls), all forms of such atropisomer are considered part of this invention.
[0049] Chemical names, common names, and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name, and an ambiguity exists between the structure and the name, the structure predominates. It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
[0050] The terms “a” and “an” as used herein mean “one or more” and include the plural unless the context is inappropriate.
[0051] The term “alkyl” refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-12, 1-10, or 1-6 carbon atoms, referred to herein as C1-C12 alkyl, C1-C10 alkyl, and Ci-Ce alkyl, respectively. Exemplary alkyl groups include, but are not limited
to, methyl, ethyl, propyl, isopropyl, 2-methyl-l -propyl, 2-m ethyl -2-propyl, 2-methyl-l -butyl, 3- methyl-1 -butyl, 2-m ethyl-3 -butyl, 2,2-dimethyl-l -propyl, 2-methyl-l -pentyl, 3-methyl-l-pentyl, 4-methyl-l -pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l- butyl, 3,3-dimethyl-l-butyl, 2-ethyl-l -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc.
[0052] The term “cycloalkyl” refers to a monovalent saturated cyclic, bicyclic, or bridged cyclic (e.g., adamantyl) hydrocarbon group of 3-12, 3-8, 4-8, or 4-6 carbons, referred to herein, e.g., as “C3-C6 cycloalkyl,” derived from a cycloalkane. Exemplary cycloalkyl groups include cyclohexyl, cyclopentyl, cyclobutyl, and cyclopropyl. The term “cycloalkylene” refers to a bivalent cycloalkyl group.
[0053] The term “haloalkyl” refers to an alkyl group that is substituted with at least one halogen. Exemplary haloalkyl groups include -CH2F, -CHF2, -CF3, -CH2CF3, -CF2CF3, and the like. The term “haloalkylene” refers to a bivalent haloalkyl group.
[0054] The term “hydroxyalkyl” refers to an alkyl group that is substituted with at least one hydroxyl. Exemplary hydroxyalkyl groups include -CH2CH2OH, -C(H)(OH)CH3, -CH2C(H)(OH)CH2CH2OH, and the like.
[0055] The terms “alkenyl” and “alkynyl” are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
[0056] The terms “alkoxyl” or “alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, terZ-butoxy and the like. The term “haloalkoxyl” refers to an alkoxyl group that is substituted with at least one halogen. Exemplary haloalkoxyl groups include -OCH2F, -OCEIF2, -OCF3, -OCH2CF3, -OCF2CF3, and the like.
[0057] The term “oxo” is art-recognized and refers to a “=O” substituent. For example, a cyclopentane susbstituted with an oxo group is cyclopentanone.
[0058] The symbol “ */wv ” indicates a point of attachment.
[0059] When any substituent or variable occurs more than one time in any constituent or the compound of the invention, its definition on each occurrence is independent of its definition at every other occurrence, unless otherwise indicated.
[0060] One or more compounds of the invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. “Solvate” means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. “Solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. “Hydrate” is a solvate wherein the solvent molecule is H2O.
[0061] As used herein, the terms “subject” and “patient” are used interchangeably and refer to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans.
[0062] The term “IC50” is art-recognized and refers to the concentration of a compound that is required to achieve 50% inhibition of the target.
[0063] As used herein, the term “effective amount” refers to the amount of a compound sufficient to effect beneficial or desired results (e.g., a therapeutic, ameliorative, inhibitory or preventative result). An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. As used herein, the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
[0064] As used herein, the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
[0065] As used herein, the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see e.g., Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975],
[0066] For therapeutic use, salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non- pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
[0067] In addition, when a compound of the invention contains both a basic moiety (such as, but not limited to, a pyridine or imidazole) and an acidic moiety (such as, but not limited to, a carboxylic acid) zwitterions (“inner salts”) may be formed. Such acidic and basic salts used within the scope of the invention are pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts. Such salts of the compounds of the invention may be formed, for example, by reacting a compound of the invention with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
[0068] Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.
[0069] As a general matter, compositions specifying a percentage are by weight unless otherwise specified.
I. Heterobifunctional Cotinine-Containing Compounds
[0070] One aspect of the invention provides heterobifunctional cotinine-containing compounds. The compounds may be used in the pharmaceutical compositions and therapeutic
methods described herein. Exemplary compounds are described in the following sections, along with exemplary procedures for making the compounds.
Part A: Compounds of Formula I
[0071] One aspect of the invention provides a compound represented by Formula I:
or a pharmaceutically acceptable salt thereof, wherein:
R1 is Ci-4 alkyl or C3-6 cycloalkyl;
X1 is a bond or a C2-8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -N(CHs)-, -O-, piperidinylene, or C3-C6 cycloalkylene;
X2 is $-(Ci-5 alkylene)-N(H)- or a covalent bond, wherein is a bond to L;
Y1 is defined by Formula 1-1 that is substituted by one occurrence of R11, wherein Formula 1-1 is one of the following:
wherein:
R2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl) or Ci-4 alkyl;
R3 and R4 each represent independently for each occurrence hydrogen or C1-4 alkyl;
R? represents independently for each occurrence fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or Ci-4 alkyl;
R7, R8, R9, and R10 each represent independently for each occurrence hydrogen or C1-4 alkyl;
R11 is a bond to X1;
R12 is hydrogen, C1-3 haloalkyl, or C1-3 alkyl;
R13 is thiazolyl or 1,2,4-thiadiazolyl, each of which is substituted with 0 or 1 occurrence of Ci -4 alkyl;
R14 is -N(R10)2 or furanyl;
R1? is fluoro, chloro, phenyl, or hydrogen; x is 1 or 2;
L is a divalent linker selected from:
(i) a bivalent, saturated or unsaturated, straight or branched Ci-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, - S-, -N(H)-, -N(CI-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, -N(CI-6 alkyl)S(O)2-, -S(O)2N(H)-, -S(O)2N(CI-6 alkyl)-, -N(H)C(O)-, -N(CI-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C 1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(CI-6 alkyl)-, -N(H)C(O)O- , -N(CI-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or optionally substituted 5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
wherein Ring A and Ring B are each independently C4-6 cycloalkylene; Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and L2a is -O-, -NHC(O)-, or -CH2-O-;
(iii)
wherein Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; Llb is -CH2-NH-C(O)-, -NHC(O)-, or -C(O)NH-; L2b is Ce-i2 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NRlb-, -
wherein n is 1 , 2, 3, or 4, and represents a covalent bond to Llb; and each Rlb is independently hydrogen or C1-3 alkyl;
(iv) O (L-c), wherein Llc is C2 -10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; and L2c is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, - NHC(O)-, or -C(O)NH-;
(v) O (L-d), wherein Lld is C 12-22 linear alkylene, wherein 1, 2, 3, 4, or 5 methylene units are replaced with -NH-, -O-, -C(O)NH-, -NHC(O)-, or -NHC(O)-NH-;
(vii) O (L-f), wherein Llf is a bond; C1-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-, -NH-, or -C(O)-; or -(C3-6 cycloalkylene)-NHC(O)-; L2f is a bond, -NHC(O)-, -C(O)NH-, or a C1-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z1 and Z2 is independently N or CH;
O
(viii)
** (L-g), wherein Ring A is a 5 to 6 membered heteroarylene having 1 or 2 nitrogen ring atoms; Llg is a bond, -CH2-, -NH-, or -O-; and
L g is
wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to Llg;
, , , , , represents a covalent bond to Llh
represents a covalent bond to L3h; L3h is a bond, -C(0)CH2-, -O-(C3-6 cycloalkylene)-O-, or -C(O)NH(CH2)3OCH2-; L4h is a bond, -C(O)-, -CH2C(O)-, or -
C(O)CH2-; and m is 1, 2, or 3;
, , , , , , bond to L31 and
represents a covalent bond to NH; L21 is a bond, C1-12 linear
***** $ _ alkylene, or
, wherein n is 1, 2, 3, 4, or 5, and I represents a covalent bond to HN; and L31 is a bond or -C(O)-;
wherein Z1 is C, CH, or N; each of Z2, Z3,
Z4 and Z5 is independently CH or N, provided that no more than two of Z2, Z3, Z4 and Z~ are N; Llj is -NH-, -C(O)NH-, -NHC(O)-, or -O-; L2' is Ci-6 linear alkylene or 11
, , represents a covalent bond to L1'; and I' represents a single bond or a double bond;
(xii)
wherein Ring A is phenylene or a 5- or
6-membered heteroarylene having 1 or 2 nitrogen ring atoms; each of Z1 and Z2 is
independently CH or N; Llk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and L2k is a C3-8
*** I _ straight chain alkylene or
, wherein n is 1, 2, or 3, and ’ represents a covalent bond to Llk;
3-6 cycoa yene, or , w eren n s or , an represents a covalent bond to Llm;
1 or 2; 0, 1, or 2 hydrogen atoms of Y ' NA are replaced with F; L p is a bond, -
C(O)-, -C(O)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-; and wherein each
represents a covalent bond to X1, and each
represents a covalent bond to X2.
[0072] The definitions of variables in Formula I above encompass multiple chemical groups. The application contemplates embodiments where, for example, i) the definition of a variable is a single chemical group selected from those chemical groups set forth above, ii) the definition of a variable is a collection of two or more of the chemical groups selected from those set forth above, and iii) the compound is defined by a combination of variables in which the variables are defined by (i) or (ii).
[0073] In certain embodiments, the compound is a compound of Formula I.
[0074] As generally defined above, R1 is Ci-4 alkyl or C3-6 cycloalkyl. In certain embodiments, R1 is C1-4 alkyl. In certain embodiments, R1 is C3-6 cycloalkyl. In certain embodiments, R1 is -CH3. In certain embodiments, R1 is cyclopropyl. In certain embodiments, R1 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0075] As generally defined above, X1 is a bond or a C2-8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -N(CH3)-, -O-, piperidinylene, or C3-C6 cycloalkylene. In certain embodiments, X1 is a bond. In certain embodiments, X1 is a C2-s bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -O-, piperidinylene, or C3-C6 cycloalkylene. In certain
embodiments, X1 is a C2-8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)- or -O-.
[0076] In certain embodiments, X1 is -(C1-6 alkylene)-N(H)-'P, wherein T is a bond to L. In certain embodiments, X1 is -(C 1-4 alkylene)-N(H)-T, wherein T is a bond to L. In certain embodiments, X1 is -(CH2)-N(H)-'P, wherein T is a bond to L. In certain embodiments, X1 is -(CH2)2-N(H)-'P, wherein T is a bond to L. In certain embodiments, X1 is -(CH2)3-N(H)-'P, wherein T is a bond to L.
[0077] In certain embodiments, X1 is -(C1-4 alkylene)-O-(Ci-4 alkylene)-(C3-6 cycloalkylene)- (Ci-4 alkylene)-N(H)-T, wherein T is a bond to L. In certain embodiments, X1 is -(C1.3 alkylene)-O-(CH2)-(C3-6 cycloalkylene)-(CH2)-N(H)-'P, wherein T is a bond to L. In certain embodiments, X1 is
, wherein T is a bond to L. In certain embodiments, X1 is -(Ci-4 alkylene)-N(H)-(Ci-4 alkylene)-piperidinylene-T/, wherein T is a bond to L. In certain embodiments, X1 is -(C1-3 alkylene)-N(H)-(Ci-3 alkylene)-piperidinylene-'P,
wherein T is a bond to L. In certain embodiments, X is H , wherein T is a bond to L. In certain embodiments, X1 is -(C1.4 alkylene)-N(H)-(Ci-4 alkylene)-piperazinylene- T, wherein T is a bond to L. In certain embodiments, X1 is -(Ci-4 alkylcne)-pipcrazinylcnc-T, wherein T is a bond to L. In certain embodiments, X1 is
, wherein T is a bond to L.
[0078] In certain embodiments, X1 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0079] As generally defined above, X2 is <t>-(C 1-5 alkylene)-N(H)- or a covalent bond, wherein <[> is a bond to L. In certain embodiments, X2 is $-(Ci-s alkylene)-N(H)-. In certain embodiments, X2 is c[)-CH2CH2-N(H)-. In certain embodiments, X2 is $-C(H)(CH3)-N(H)-. In certain embodiments, X2 is a covalent bond. In certain embodiments, X2 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0080] As generally defined above, Y1 is defined by Formula 1-1 that is substituted by one occurrence of R11, wherein Formula 1-1 is one of the following:
wherein:
R2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl) or Ci-4 alkyl;
R3 and R4 each represent independently for each occurrence hydrogen or C1-4 alkyl;
R5 represents independently for each occurrence fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or C1.4 alkyl;
R7, R8, R9, and R10 each represent independently for each occurrence hydrogen or C1-4 alkyl;
R11 is a bond to X1;
R12 is hydrogen, C1-3 haloalkyl, or C1-3 alkyl;
R13 is thiazolyl or 1,2,4-thiadiazolyl, each of which is substituted with 0 or 1 occurrence of C1-4 alkyl;
R14 is -N(R10)2 or furanyl;
R13 is fluoro, chloro, phenyl, or hydrogen; and x is 1 or 2.
[0081] In certain embodiments, Y1 is defined by the following formula that is substituted by one occurrence of R11 :
[0082] In certain embodiments,
; wherein:
R2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl);
R3 and R4 are independently hydrogen or Ci-4 alkyl;
R? is fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or C1.4 alkyl;
R7 is hydrogen or Ci-4 alkyl;
R8 is Ci-4 alkyl or hydrogen; and
R9 is hydrogen or Ci-4 alkyl.
[0083] In certain embodiments, Y1 is one of the following:
wherein:
R3 and R4 are independently hydrogen or C1-4 alkyl;
R3 is fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or Ci-4 alkyl;
R7 is hydrogen or Ci-4 alkyl;
R8 is hydrogen or Ci-4 alkyl;
R9 is Ci-4 alkyl; and
R10 is hydrogen or Ci-4 alkyl.
[0085] In certain embodiments, Y1 is one of the following:
[0086] In certain embodiments, Y1 is one of the following:
[0087] In certain embodiments, Y1 is defined by one of the following formulae which are substituted by one occurrence of R11:
[0088] In certain embodiment
, certain
[0089] In certain embodiments, Y1 is one of the following:
[0090] In certain embodiments, Y1 is defined by the following formula that is substituted by one occurrence
[0091] In certain embodiments,
certain embodiments,
certain embodiments, Y1 is
o o . In certain embodiments, Y1 is O O
[0092] In certain embodiments, Y1 is one of the following:
[0093] In certain embodiments, Y1 is selected from the groups depicted in the compounds in Tables 1, 2, and 3.
[0094] In certain embodiments, Y^X1- is one of the following:
[0095] In certain embodiments, Y’-X1- is one of the following:
[0096] In certain embodiments, Y^X1- is one of the following:
[0097] In certain embodiments, Y^X1- is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0098] As generally defined above, R2 is -(Ci-4 alkylene)-(Cs-6 cycloalkyl) or Ci-4 alkyl. In certain embodiments, R2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl). In certain embodiments, R2 is -(Ci- 2 alkylene)-(Cs-6 cycloalkyl). In certain embodiments, R2 is -(CH2)-cyclohexyl. In certain embodiments, R2 is -(CH2)-cyclopentyl. In certain embodiments, R2 is Ci-4 alkyl. In certain embodiments, R2 is methyl. In certain embodiments, R2 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0099] As generally defined above, R3 and R4 each represent independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R3 and R4 are independently hydrogen or Ci-4 alkyl. In certain embodiments, R3 and R4 are hydrogen.
[0100] In certain embodiments, R3 represents independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R3 is hydrogen. In certain embodiments, R3 represents independently for each occurrence Ci-4 alkyl. In certain embodiments, R3 is methyl. In certain embodiments, R3 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0101] In certain embodiments, R4 represents independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R4 is hydrogen. In certain embodiments, R4 represents independently for each occurrence Ci-4 alkyl. In certain embodiments, R4 is methyl. In certain embodiments, R4 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0102] As generally defined above, R5 represents independently for each occurrence fluoro, chloro, or cyano. In certain embodiments, R5 is fluoro, chloro, or cyano. In certain embodiments, R5 is fluoro. In certain embodiments, R5 is chloro. In certain embodiments, R3 is
cyano. In certain embodiments, R5 represents independently for each occurrence fluoro or chloro. In certain embodiments, R5 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0103] As generally defined above, R6 is hydrogen, fluoro, chloro, or CM alkyl. In certain embodiments, R6 is hydrogen, fluoro, or chloro. In certain embodiments, R6 is fluoro or chloro. In certain embodiments, R6 is hydrogen. In certain embodiments, R6 is fluoro. In certain embodiments, R6 is chloro. In certain embodiments, R6 is Ci-4 alkyl. In certain embodiments, R6 is methyl. In certain embodiments, R6 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0104] As generally defined above, R7, R8, R9, and R10 each represent independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R7 is hydrogen, and R8 is hydrogen. In certain embodiments, R7, R8, R9, and R10 are selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0105] In certain embodiments, R7 represents independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R7 is hydrogen or CM alkyl. In certain embodiments, R7 is hydrogen. In certain embodiments, R7 represents independently for each occurrence CM alkyl. In certain embodiments, R7 is Ci-4 alkyl. In certain embodiments, R7 is methyl. In certain embodiments, R7 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0106] In certain embodiments, R8 represents independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R8 is hydrogen or Ci-4 alkyl. In certain embodiments, R8 is hydrogen. In certain embodiments, R8 represents independently for each occurrence CM alkyl. In certain embodiments, R8 is CM alkyl. In certain embodiments, R8 is methyl. In certain embodiments, R8 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0107] In certain embodiments, R9 represents independently for each occurrence hydrogen or CM alkyl. In certain embodiments, R9 is hydrogen or CM alkyl. In certain embodiments, R9 is hydrogen. In certain embodiments, R9 represents independently for each occurrence CM alkyl. In certain embodiments, R9 is CM alkyl. In certain embodiments, R9 is methyl. In certain
embodiments, R9 is selected from the groups depicted in the compounds in Tables 1 , 2, and 3, below.
[0108] In certain embodiments, R10 represents independently for each occurrence hydrogen or Ci-4 alkyl. In certain embodiments, R10 is hydrogen or Ci-4 alkyl. In certain embodiments, R10 is hydrogen. In certain embodiments, R10 represents independently for each occurrence Ci-4 alkyl. In certain embodiments, R10 is Ci-4 alkyl. In certain embodiments, R10 is methyl. In certain embodiments, R10 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0109] As generally defined above, R11 is a bond to X1. In certain embodiments, R11 replaces R10. In certain embodiments, R11 replaces R9. In certain embodiments, R11 replaces R7. In certain embodiments, R11 occurs on R2. In certain embodiments, R11 occurs on a phenyl ring of Y1. In certain embodiments, R11 occurs on a pyridinyl ring of Y1. In certain embodiments, the position of R11 on Y1 is selected from the positions depicted in the compounds in Table 1, below.
[0110] As generally defined above, R12 is hydrogen, C1.3 haloalkyl, or C1-3 alkyl. In certain embodiments, R12 is hydrogen or C1-3 haloalkyl. In certain embodiments, R12 is C1-3 haloalkyl or C1-3 alkyl. In certain embodiments, R12 is hydrogen. In certain embodiments, R12 is C1-3 haloalkyl. In certain embodiments, R12 is trifluorom ethyl. In certain embodiments, R12 is C1.3 alkyl. In certain embodiments, R12 is methyl. In certain embodiments, R12 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0111] As generally defined above, R13 is thiazolyl or 1,2,4-thiadiazolyl, each of which is substituted with 0 or 1 occurrence of C1-4 alkyl. In certain embodiments, R13 is thiazolyl substituted with 0 or 1 occurrence of C1-4 alkyl. In certain embodiments, R13 is thiazolyl. In certain embodiments, R13 is 1,2,4-thiadiazolyl substituted with 0 or 1 occurrence of C1-4 alkyl. In certain embodiments, R13 is 1,2,4-thiadiazolyl. In certain embodiments, R13 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0112] As generally defined above, R14 is -N(R10)2 or furanyl. In certain embodiments, R14 is -N(R1O)2. In certain embodiments, R14 is -N(R10)(R11). In certain embodiments, R14 is - N(CH3)(Rn). In certain embodiments, R14 is furanyl. In certain embodiments, R14 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0113] As generally defined above, R13 is fluoro, chloro, phenyl, or hydrogen. In certain embodiments, R15 is fluoro, chloro, or phenyl. In certain embodiments, R15 is fluoro or chloro. In certain embodiments, R13 is fluoro. In certain embodiments, R15 is chloro. In certain embodiments, R13 is phenyl. In certain embodiments, R15 is hydrogen. In certain embodiments, R15 is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0114] As generally defined above, x is 1 or 2. In certain embodiments, x is 1. In certain embodiments, x is 2. In certain embodiments, x is selected from the values depicted in the compounds in Tables 1, 2, and 3, below.
[0115] As generally defined above, L is a divalent linker selected from:
(i) a bivalent, saturated or unsaturated, straight or branched Ci-60 hydrocarbon chain, wherein 0- 20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(CI-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, -N(CI-6 alkyl)S(O)2-, -S(O)2N(H)-, - S(O)2N(CI-6 alkyl)-, -N(H)C(O)-, -N(CI-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(CI-6 alkyl)-, - OC(O)N(H)-, -OC(O)N(CI-6 alkyl)-, -N(H)C(O)O-, -N(CI-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or optionally substituted 5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
wherein Ring A and Ring B are each independently C4-6 cycloalkylene; Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and L2a is -O-, - NHC
(iii)
wherein Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; Llb is -CH2-NH-C(O)-, -NHC(O)-, or -C(O)NH-; L2b is C6-12 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NRlb-, -C(O)NRlb-, or -
***
NRlbC(O)-; or L2b is
wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llb; and each Rlb is independently hydrogen or C1.3 alkyl;
(iv) O (L-c), wherein Lle is C2 -10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; and L2c is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -
wherein Lld is C 12-22 linear alkylene, wherein 1, 2, 3, 4, or 5 methylene units are replaced with -NH-, -O-, -C(O)NH-, -NHC(O)-, or -NHC(O)-NH-;
(vii) O (L-f), wherein Llf is a bond; C1-6 linear alkylene, wherein
0, 1, or 2 methylene units are replaced with -O-, -NH-, or -C(O)-; or -(C3-6 cycloalkylene)- NHC(O)-; L2f is a bond, -NHC(O)-, -C(O)NH-, or a Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z1 and Z2 is independently N or CH;
(L-g), wherein Ring A is a 5 to 6 membered heteroarylene having 1 or 2 nitrogen ring atoms; Llg is a bond, -CH2-, -NH-, or -O-; and L2g is
wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to Llg;
is a bond, -C(O)-, -C(O)-NH-, or -NHC(O)-; L2h is C2-10 linear alkylene
wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llh and
represents a covalent bond to L3h; L3h is a bond, -C(0)CH2-, -O-(C3-6 cycloalkylene)-O-, or - C(O)NH(CH2)3OCH2-; L4h is a bond, -C(O)-, -CH2C(O)-, or -C(O)CH2-; and m is 1, 2, or 3;
, w ere n n s , , , , or , an represents a cova ent ond to L31
represents a covalent bond to NH; L21 is a bond, C1-12 linear alkylene, or
, , , , , , represents a covalent bond to HN; and L31 is a bond or -C(O)-;
wherein Z1 is C, CH, or N; each of Z2, Z3, Z4 and
Z5 is independently CH or N, provided that no more than two of Z2, Z3, Z4 and Z5 are N; Llj is -
NH-, -C(O)NH-, -NHC(O)-, or -O-; L2' is Ci-6 linear alkylene or
, wherein n is 1 *** or 2, and represents a covalent bond to L1'; and !■ represents a single bond or a double bond;
(xii)
wherein Ring A is phenylene or a 5- or 6- membered heteroarylene having 1 or 2 nitrogen ring atoms; each of Z1 and Z2 is independently
CH or N; Llk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and L2k is a C3-8 straight chain alkylene
, , , represents a covalent bond to Llk;
, , , ,
C(0)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2m is C3-6 linear alkylene, C3-6 cycloalkylene, or
, wherein n is 1 or 2, and
represents a covalent bond to
Llm;
(L-n-iv); or
0, 1, or 2 hydrogen atoms
are replaced with F; Llp is a bond, -C(O)-, -C(O)NH- , -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-; wherein each
represents a covalent bond to X1, and each
represents a covalent bond to X2.
[0116] In certain embodiments,
wherein
Ring A and Ring B are each independently C4-6 cycloalkylene; Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and L2a is -O- or -CH2-O-; wherein
represents a covalent bond to
X1, and represents a covalent bond to X2.
[0117] In certain embodiments, L is selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0118] Further description and embodiments for variable L are provided below in, for example, Part B.
[0119] In certain embodiments, the compound of Formula (I) is represented by Formula la or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula la.
[0120] In certain embodiments, the compound of Formula (I) is represented by Formula lb or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula lb.
[0121] In certain embodiments, the compound of Formula (I) is represented by Formula Ic or
Id or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula Ic or Id. In certain embodiments, the compound is a compound of Formula Ic, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ic. In certain embodiments, the compound is a compound of Formula Id, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Id.
[0122] In certain embodiments, the compound of Formula (I) is represented by Formula le or
If or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula le or If. In certain embodiments, the compound is a compound of Formula le, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula le. In certain embodiments, the compound is a compound of Formula If, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula If.
[0123] In certain embodiments, the compound of Formula (I) is represented by Formula Ig or Ih or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula Ig or Ih. In certain embodiments, the compound is a compound of Formula Ig, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ig. In certain embodiments, the compound is a compound of Formula Ih, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ih.
[0124] In certain embodiments, the compound of Formula (I) is represented by Formula li or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula li.
[0125] In certain embodiments, the compound of Formula (I) is represented by Formula Ij or Ik or a pharmaceutically acceptable salt thereof:
wherein each of the variables is as defined in embodiments herein. In certain embodiments, the compound is a compound of Formula Ij or Ik. In certain embodiments, the compound is a compound of Formula Ij, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ij. In certain embodiments, the compound is a compound of Formula Ik, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ik.
[0126] The description above describes multiple embodiments relating to compounds of Formula I. The patent application specifically contemplates all combinations of the embodiments.
Part B: Additional Description of the Linker
[0127] As generally defined above, L is a divalent linker selected from:
(i) a bivalent, saturated or unsaturated, straight or branched Ci-60 hydrocarbon chain, wherein 0- 20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(CI-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, -N(CI-6 alkyl)S(O)2-, -S(O)2N(H)-, - S(O)2N(C i-6 alkyl)-, -N(H)C(O)-, -N(CI-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(CI-6 alkyl)-, - OC(O)N(H)-, -OC(O)N(C i-6 alkyl)-, -N(H)C(O)O-, -N(CI-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or optionally substituted 5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
independently C4-6 cycloalkylene; Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and L2a is -O-, - NHC(O)-, or -CH2-O-;
(iii)
wherein Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; Llb is -CH2-NH-C(0)-, -NHC(O)-, or -C(O)NH-; L2b is Ce-12 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NRlb-, -C(O)NRlb-, or -
NRlbC(O)-; or L2b is
wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llb; and each Rlb is independently hydrogen or C1.3 alkyl;
(iv) O (L-c), wherein Llc is C2 -10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; and L2c is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or - C(O)NH-;
(v) O (L-d), wherein Lld is C 12-22 linear alkylene, wherein 1, 2, 3, 4, or 5 methylene units are replaced with -NH-, -O-, -C(O)NH-, -NHC(O)-, or -NHC(O)-NH-;
(vii) O (L-f), wherein Llf is a bond; C1-6 linear alkylene, wherein
0, 1 , or 2 methylene units are replaced with -O-, -NH-, or -C(O)-; or -(C3-6 cycloalkylene)- NHC(O)-; L2f is a bond, -NHC(O)-, -C(O)NH-, or a Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z1 and Z2 is independently N or CH;
(viii)
(L-g), wherein Ring A is a 5 to 6 membered heteroarylene having 1 or 2 nitrogen ring atoms; Llg is a bond, -CH2-, -NH-, or -O-; and L2g is
wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to Llg;
wherein each Z1 is independently N or CH; Llh is a bond, -C(O)-, -C(O)-NH-, or -NHC(O)-; L2h is C2-10 linear alkylene
_ I **** wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llh and « represents a covalent bond to L31’; L3h is a bond, -C(0)CH2-, -O-(Cs-6 cycloalkylene)-O-, or -
C(O)NH(CH2)3OCH2-; L4h is a bond, -C(O)-, -CH2C(O)-, or -C(O)CH2-; and m is 1, 2, or 3;
, , , , , , d to L31
****
represents a covalent bond to NH; L21 is a bond, C1-12 linear alkylene, or
, , , , , , represents a covalent bond to HN; and L31 is a bond or -C(O)-;
wherein Z1 is C, CH, or N; each of Z2, Z3, Z4 and Z3 is independently CH or N, provided that no more than two of Z2, Z3, Z4 and Z5 are N; Llj is -NH-,
-C(O)NH-, -NHC(O)-, or -O-; L2j is C1-6 linear alkylene or
, wherein n is 1 or 2, and represents a covalent bond to Llj, and I' represents a single bond or a double bond;
(xii)
wherein Ring A is phenylene or a 5- or 6- membered heteroarylene having 1 or 2 nitrogen ring atoms; each of Z1 and Z2 is independently CH or N; Llk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and L2k is a C3-8 straight chain alkylene or
, wherein n is 1, 2, or 3, and
represents a covalent bond to Llk;
(xiii)
wherein Z1 is CH or N; m is 1 or 2; p is 1 or
2; 0, 1, or 2 hydrogen atoms
are replaced with F; Llm is a bond, -C(O)-, -
C(O)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2m is C3-6 linear alkylene, C3-6 cycloalkylene, or
, wherein n is 1 or 2, and
represents a covalent bond to
Llm;
(L-n-iv); or
, , , , -
, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-;
* I _ _ I ** wherein each 1 represents a covalent bond to X1, and each 1 represents a covalent bond to X2.
[0128] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched Ci-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(CI-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, -N(Ci-6 alkyl)S(O)2-, -S(O)2N(H)-, -S(O)2N(CI-6 alkyl)-, -N(H)C(O)-, -N(CI-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C i-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(CI-6 alkyl)-, -N(H)C(O)O-, -N(CI-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or optionally substituted 5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[0129] In certain embodiments, L is a divalent linker of Formula (L-a), (L-b), (L-c), (L-d), (L-e), (L-f), (L-g), (L-h), (L-i), (L-j), (L-k), (L-m), (L-n-i), (L-n-ii), (L-n-iii), or (L-n-iv).
[0130] In certain embodiments, L is a divalent linker of Formula (L-a):
wherein:
Ring A and Ring B are each independently C4-6 cycloalkylene;
Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and
L2a is -O-, -NHC(O)-, or -CH2-O-; wherein represents a
represents a covalent bond to X2.
[0131] In certain embodiments, Ring A and Ring B of Formula (L-a) are each independently
[0132] In certain embodiments, L is a divalent linker of Formula (L-a-i):
wherein:
Ring A is C4-6 cycloalkylene;
Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and L2a is -O-, -NHC(O)-, or -CH2-O-;
* ] — wherein I represents a covalent bond to
represents a covalent bond to X2.
[0133] In certain embodiments, Ring A of Formula (L-a-i) is
[0134] In certain embodiments, L is a divalent linker of Formula (L-a-ii):
-ii); wherein:
Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl;
L2a is -O-, -NHC(O)-, or -CH2-O-; p is 1 or 2; and m is 1 or 2; wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0135] In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from
wherein: j is 1, 2, 3, or 4; k is 0, 1, 2, or 3; the sum of j and k is 2, 3, or 4; q is 1 or 2; r is 1 or 2; s is 0 or 1; the sum of q, r, and s is 2 or 3;
X1A and X2A are independently -O- or NRa; and each Ra is independently hydrogen or C1-3 alkyl; wherein represents a covalent bond to the C(O) group of Formula (L-a), (L-a-i), or (L-a-ii), and represents a covalent bond to Ring B of Formula (L-a) or to the cyclohexylene group of Formula (L-a-i) or (L-a-ii).
[0136] In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from - (CH2)2O-, -(CH2)3O-, -(CH2)4O-, -(CH2)2OCH2-, -(CH2)3OCH2-, -(CH2)2O(CH2)2-, -CH2OCH2-, -CH2O(CH2)2-, -CH2O(CH2)3-, -CH2OCH2O-, or -CH2OCH2OCH2-. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -(CH2)2O-, -(CH2)3O-, -(CH2)2OCH2-, or -(CH2)3OCH2-. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -(CH2)2NRa-, -(CH2)3NRa-, -(CH2)4NRa-, -(CH2)2NRaCH2-, -(CH2)3NRaCH2-, - (CH2)2NRa(CH2)2-, -CH2NRaCH2-, -CH2NRa(CH2)2-, -CH2NRa(CH2)3-, -CH2NRaCH2NRa-, or - CH2NRaCH2NRaCH2-, wherein each Ra is independently hydrogen or C1-3 alkyl. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -(CH2)2NRa-, - (CH2)3NRa-, -(CH2)2NRaCH2-, or -(CH2)3NRaCH2-, wherein Ra is hydrogen or C1-3 alkyl. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -(CH2)2NH-, - (CH2)3NH-, -(CH2)4NH-, -(CH2)2NHCH2-, -(CH2)3NHCH2-, -(CH2)2NH(CH2)2-, -CH2NHCH2-, - CH2NH(CH2)2-, -CH2NH(CH2)3-, -CH2NHCH2NH-, or -CH2NHCH2NHCH2-. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -(CH2)2NH-, -(CH2)3NH- , -(CH2)2NHCH2-, or -(CH2)3NHCH2-. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -CH2OCH2NRa-, -CH2NRaCH2O-, -CH2OCH2NRaCH2-, - CH2NRaCH2OCH2-, wherein Ra is independently hydrogen or C1-3 alkyl. In certain embodiments, Lla of Formula (L-a), (L-a-i), or (L-a-ii) is selected from -CH2OCH2NH-, - CH2NHCH2O-, -CH2OCH2NHCH2-, -CH2NHCH2OCH2-.
[0137] In certain embodiments, L is a divalent linker of Formula (L-a-iii):
p is 1 or 2; m is 1 or 2; and n is 1, 2, or 3; i _ wherein I represents a covalent bond to X1, and
represents a covalent bond to X2.
[0138] In certain embodiments, L is a divalent linker of Formula (L-a) selected from the group consisting of:
embodiments, for linkers of Formula (L-a), the point of attachment indicated on the cycloalkyl- bound carbonyl group is the attachment point to X1.
[0139] In certain embodiments, L is selected from the group consisting of:
indicated on the cycloalkyl-bound carbonyl group is the attachment point to X1.
[0140] In certain embodiments, L is a divalent linker of Formula (L-b):
wherein:
Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene;
Llb is -CH2-NH-C(O)-, -NHC(O)-, or -C(O)NH-;
L2b is C6 -12 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NRlb-, - C(O)NRlb-, or -NRlbC(O)-; or
L2b is
, wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llb; and each Rlb is independently hydrogen or C1-3 alkyl; i _ wherein I represents a covalent bond to X1, and
represents a covalent bond to X2.
[0142] In certain embodiments, L is a divalent linker of Formula (L-b-i):
)NH-;
L2b is Ce-12 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NRlb-, - C(O)NRlb-, or -NRlbC(O)-; or
L2b is
, wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llb; each Rlb is independently hydrogen or C1-3 alkyl; p is 1 or 2; and m is 1 or 2;
* wherein represents a covalent bond to X , and represents a covalent bond to X .
[0143] In certain embodiments, L2b of Formula (L-b) or (L-b-i) is selected from
; wherein: j is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; k is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; the sum ofj and k is 5, 6, 7, 8, 9, 10, or 11; q is 1 , 2, 3, 4, 5, 6, 7, 8, or 9; r is 1, 2, 3, 4, 5, 6, 7, 8, or 9; s is O, 1, 2, 3, 4, 5, 6, 7, or 8; the sum of q, r, and s is 4, 5, 6, 7, 8, 9, or 10; t is 1, 2, 3, 4, 5, 6, or 7; u is 1, 2, 3, 4, 5, 6, or 7; v is 1, 2, 3, 4, 5, 6, or 7; w is 0, 1, 2, 3, 4, 5, or 6; the sum of t, u, v, and w is 3, 4, 5, 6, 7, 8, or 9; a is 1, 2, 3, 4, or 5; b is 1, 2, 3, 4, or 5; c is 1, 2, 3, 4, or 5; d is 1, 2, 3, 4, or 5; e is 0, 1, 2, 3, or 4; the sum of a, b, c, d, and e is 4, 5, 6, 7, or 8;
X1A, X2A, X3A, and X4A are independently -O-, -NRlb-, -C(O)NRlb-, or -NRlbC(O)-; and each Rlb is independently hydrogen or C 1-3 alkyl;
*** I _ _ I ** wherein I represents a covalent bond to Llb of Formula (L-b) or (L-b-i), and « represents a covalent bond to X2.
[0144] In certain embodiments, L is a divalent linker of Formula (L-b) selected from the group consisting of:
. In certain embodiments, for linkers of Formula
(L-b), the point of attachment indicated on the cycloalkyl-bound carbonyl group is the attachment point to X1.
[0145] In certain embodiments, L is selected from the group consisting of
indicated on the cycloalkyl-bound carbonyl group is the attachment point to X1.
[0146] In certain embodiments, L is a divalent linker of Formula (L-c):
wherein:
Llc is C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, - NHC(O)-, or -C(O)NH-;
Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; and
L2C is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; wherein represents a
represents a covalent bond to X2.
[0147] In certain embodiments, Ring A of Formula (L-c) is
[0148] In certain embodiments, L is a divalent linker of Formula (L-c-i):
wherein:
Llc is C2 -10 linear alkylene, wherein I, 2, or 3 methylene units are replaced with -O-, -NH-, - NHC(O)-, or -C(O)NH-;
L2C is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; p is 1 or 2; and m is 1 or 2;
* I _ _ | ** wherein I represents a covalent bond to X1, and » represents a covalent bond to X2.
[0149] In certain embodiments, Llc of Formula (L-c) or (L-c-i) is selected from
wherein: j is 1, 2, 3, 4, 5, 6, 7, 8, or 9; k is 0, 1, 2, 3, 4, 5, 6, 7, or 8; the sum of j and k is 1, 2, 3, 4, 5, 6, 7, 8, or 9; q is 1, 2, 3, 4, 5, 6, or 7; r is 1, 2, 3, 4, 5, 6, or 7; s is 0, 1, 2, 3, 4, 5, or 6; the sum of q, r, and s is 2, 3, 4, 5, 6, 7, or 8; t is 1, 2, 3, 4, or 5; u is 1, 2, 3, 4, or 5; v is 1, 2, 3, 4, or 5;
w is 0, 1, 2, 3, or 4; the sum of t, u, v, and w is 3, 4, 5, 6, or 7; and
X1A, X2A and X3A are independently -O-, -NH-, -NHC(O)-, or -C(O)NH-;
####! _ wherein 1 represents a covalent bond to the C(O) group of Formula (L-c) or (L-c-i), and
represents a covalent bond to the ring of Formula (L-c) or (L-c-i).
[0150] In certain embodiments, L2c of Formula (L-c) or (L-c-i) is selected from
, , or wherein: j is O, 1, 2, 3, 4, 5, 6, 7, 8, or 9; k is 0, 1, 2, 3, 4, 5, 6, 7, 8, or 9; the sum of j and k is 1, 2, 3, 4, 5, 6, 7, 8, or 9; q is 0, 2, 3, 4, 5, 6, or 7; r is 1, 2, 3, 4, 5, 6, 7, or 8; s is 0, 1, 2, 3, 4, 5, 6, or 7; the sum of q, r, and s is 1, 2, 3, 4, 5, 6, 7, or 8; t is 0, 1, 2, 3, 4, or 5; u is 1, 2, 3, 4, 5, or 6; v is 1, 2, 3, 4, 5, or 6; w is 0, 1, 2, 3, 4, or 5; the sum of t, u, v, and w is 2, 3, 4, 5, 6, or 7; and
X’A, X2A and X3A are independently -O-, -NH-, -NHC(O)-, or -C(O)NH-;
### | _ wherein I represents a covalent bond to the ring of Formula (L-c) or (L-c-i), and
represents a covalent bond to X2.
[0151] In certain embodiments, L is a divalent linker of Formula (L-c) selected from the group consisting of:
In certain embodiments, for linkers of Formula (L-c), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0152] In certain embodiments, L is selected from the group consisting of:
p he carbonyl group is the attachment point to X1.
[0153] In certain embodiments, L is a divalent linker of Formula (L-d):
wherein:
Lld is C 12-22 linear alkylene, wherein 1, 2, 3, 4, or 5 methylene units are replaced with -NH-, -O-, -C(O)NH-, -NHC(O)-, or -NHC(0)-NH-; wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0154] In certain embodiments, Lld of Formula (L-d) is selected from
wherein: j is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; k is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; the sum of j and k is 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21; q is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19; r is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19; s is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; the sum of q, r, and s is 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; t is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; u is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; v is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; w is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16; the sum of t, u, v, and w is 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19; a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; b is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; c is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; d is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; e is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14; the sum of a, b, c, d, and e is 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; f is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, or 13; g is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13; h is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13; i is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13; y is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13; z is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12; the sum of f, g, h, i, y, and z is 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; and
X1A, X2A, X3A, X4A, and X5A are independently -NH-, -0-, -C(O)NH-, -NHC(O)-, or -NHC(O)- NH-;
### | ** wherein represents a covalent bond to the C(O) group of Formula (L-d), and » represents a covalent bond to X2.
[0155] In certain embodiments, L is a divalent linker of Formula (L-d) selected from the group consisting of:
In certain embodiments, for linkers of Formula (L-d), the point of attachment indicated on the carbon-bound carbonyl group is the attachment point to X1.
[0156] In certain embodiments, L is selected from the group consisting of:
wherein the point of attachment indicated on the carbon-bound carbonyl group is the attachment point to X1.
[0157] In certain embodiments, L is a divalent linker of Formula (L-e):
wherein: n is an integer of 3 to 50; wherein I represents a covalent bond to X1, and
represents a covalent bond to X2.
[0158] In certain embodiments, n of Formula (L-e) is 3 to 25, 3 to 10, 3 to 8, 3 to 7, 3 to 5, or 3 to 4. In certain embodiments, n of Formula (L-e) is 3, 4, 5, 7, 8, 22, or 50.
[0159] In certain embodiments, L is a divalent linker of Formula (L-f):
wherein:
Llf is a bond; Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-, -NH- , or -C(O)-; or -(C3-6 cycloalkylene)-NHC(O)-;
L2t is a bond, -NHC(O)-, -C(O)NH-, or a Ci-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z1 and Z2 is independently N or CH;
♦ I _ _ I ** wherein I represents a covalent bond to X1, and ‘ represents a covalent bond to X2.
[0160] In certain embodiments, Llf of Formula (L-f) is selected from
; wherein: j is 1, 2, 3, 4, or 5; k is 0, 1, 2, 3, or 4; the sum of j and k is 1, 2, 3, 4, or 5; q is 1, 2, or 3; r is 1, 2, or 3; s is 0, 1, 2; the sum of q, r, and s is 2, 3, or 4; and
X1A and X2A are independently -O-, -NH-, or -C(O)-; or -(C3-6 cycloalkylene)-NHC(O)-;
####[ ##### wherein 1 represents a covalent bond to the C(O) group of Formula (L-f),
represents a covalent bond to the ring of Formula (L-f).
[0161] In certain embodiments, L2f of Formula (L-f) is selected from
wherein: j is 1, 2, 3, 4, or 5; k is 0, 1, 2, 3, or 4; the sum of j and k is 1, 2, 3, 4, or 5; q is 1, 2, or 3; r is 1, 2, or 3; s is 0, 1, 2; and the sum of q, r, and s is 2, 3, or 4;
####! _ _ I ** wherein ? represents a covalent bond to the ring of Formula (L-f), and ’ represents a covalent bond to X2.
[0162] In certain embodiments, L is a divalent linker of Formula (L-f) selected from the group consisting of:
. In certain embodiments, for linkers of Formula (L-f), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0163] In certain embodiments, L is selected from the group consisting of
attachment indicated on the carbonyl group is the attachment point to X1.
[0164] In certain embodiments, L is a divalent linker of Formula (L-g):
wherein:
Ring A is a 5 to 6 membered heteroarylene having 1 or 2 nitrogen ring atoms;
Llg is a bond, -CH2-, -NH-, or -O-; and
L2g is
wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to Llg;
H ** wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0165] In certain embodiments, L is a divalent linker of Formula (L-g-i):
wherein:
Llg is a bond, -CH2-, -NH-, or -O-;
L2g is
wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to Llg;
Z1, Z2, and Z3 are each independently selected from N or CH, provided that one or two of Z1, Z2, and Z3 is N; ** wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0166] In certain embodiments, L is a divalent linker of Formula (L-g) selected from the group consisting of:
. In certain embodiments, for linkers of
Formula (L-g), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0167] In certain embodiments, L is selected from the group consisting of:
[0168] In certain embodiments, L is a divalent linker of Formula (L-h):
each Z1 is independently N or CH;
Llh is a bond, -C(O)-, -C(O)-NH-, or -NHC(O)-;
L211 is C2-10 linear alkylene or
, wherein n is 1, 2, 3, or 4, and
represents a covalent bond to Llh and
represents a covalent bond to L3h;
L3h is a bond, -C(0)CH2-, -O-(C3-6 cycloalkylene)-O-, or -C(O)NH(CH2)3OCH2-;
L4h is a bond, -C(O)-, -CH2C(O)-, or -C(O)CH2-; and m is 1, 2, or 3;
* I _ _ I ** wherein 1 represents a covalent bond to X1, and « represents a covalent bond to X2.
[0169] In certain embodiments, L is a divalent linker of Formula (L-h) selected from the group consisting of:
In certain embodiments, for linkers of Formula (L-h), the point of attachment indicated on the carbonyl group or cyclic group is the attachment point to X1.
[0170] In certain embodiments, L is a divalent linker of Formula (L-h) selected from the group consisting of:
wherein the point of attachment indicated on the carbonyl group or cyclic group is the attachment point to X1.
[0171] In certain embodiments, L is a divalent linker of Formula (L-i):
wherein:
L11 is a bond, C1-12 linear alkylene, or
, wherein n is 1, 2, 3, 4, or 5, and
★ ★★ represents a covalent bond to L31 and
represents a covalent bond to NH;
L21 is a bond, C1-12 linear alkylene, or
, wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to HN; and
L31 is a bond or -C(O)-; ** wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0172] In certain embodiments, L is a divalent linker of Formula (L-i) selected from the group consisting of:
certain embodiments, for linkers of
Formula (L-i), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0173] In certain embodiments, L is a divalent linker of Formula (L-j):
each of Z2, Z3, Z4 and Z? is independently CH or N, provided that no more than two of Z2, Z3, Z4 and Z5 are N;
L1J is -NH-, -C(O)NH-, -NHC(O)-, or -O-;
*** I _
L2-1 is Ci-6 linear alkylene or
, wherein n is 1 or 2, and • represents a covalent bond to L1'; and
' represents a single bond or a double bond;
I _ _ | ** wherein I represents a covalent bond to X1, and ’ represents a covalent bond to X2.
[0174] In certain embodiments, L is a divalent linker of Formula (L-j) selected from the group consisting of:
. In certain embodiments, for linkers of
Formula (L-j ), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0175] In certain embodiments, L is selected from the group consisting of
; wherein the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0176] In certain embodiments, L is a divalent linker of Formula (L-k):
wherein:
Ring A is phenylene or a 5- or 6-membered heteroarylene having 1 or 2 nitrogen ring atoms; each of Z1 and Z2 is independently CH or N;
Llk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and
L2k is a C3-8 straight chain alkylene or
, wherein n is 1, 2, or 3, and
represents a covalent bond to Llk;
* | _ _ I ** wherein I represents a covalent bond to X1, and « represents a covalent bond to X2.
[0177] In certain embodiments, L is a divalent linker of Formula (L-k) selected from the group consisting of:
. In certain embodiments, for linkers of
Formula (L-k), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0178] In certain embodiments, L is a selected from the group consisting of:
on the carbonyl group is the attachment point to X1.
[0179] In certain embodiments, L is a divalent linker of Formula (L-m):
m is 1 or 2; p is 1 or 2;
0, 1, or 2 hydrogen atoms
are replaced with F;
Llm is a bond, -C(O)-, -C(O)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and
L2m is C3-6 linear alkylene, C3-6 cycloalkylene, or
, wherein n is 1 or 2, and represents a covalent bond to Llm; ** wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0180] In certain embodiments, L is a divalent linker of Formula (L-m) selected from the group consisting of:
m), the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0181] In certain embodiments, L is selected from the group consisting of:
carbonyl group is the attachment point to X1.
[0182] In certain embodiments, L is a divalent linker of Formula (L-n-i):
H ** wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0183] In certain embodiments, L is a divalent linker of Formula (L-n-ii):
! _ _ | ** wherein I represents a covalent bond to X , and « represents a covalent bond to X .
[0184] In certain embodiments, L is a divalent linker of Formula (L-n-iii):
-iii) ** wherein represents a covalent bond to X1, and
represents a covalent bond to X2.
[0185] In certain embodiments, L is a divalent linker of Formula (L-n-iv):
iv)
« I _ wherein I represents a covalent bond to X1, and
represents a covalent bond to X2.
[0186] In certain embodiments,
wherein Z1 is CH or N; m is 1 or 2; p is 1 or 2; 0, 1, or 2 hydrogen atoms
are replaced with
F; Llp is a bond, -C(O)-, -C(0)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-.
[0187] In certain embodiments, L is
certain embodiments, for linkers of Formula (L-p), the point of attachment indicated on the cycloalkyl- bound carbonyl group is the attachment point to X1. In certain embodiments, L is
wherein the point of attachment indicated on the cycloalkyl-bound carbonyl group is the attachment point to X1.
on the carbonyl group is the attachment point to X1.
[0189] In certain embodiments, L is one of the following:
wherein the point of attachment indicated on the carbon-bound carbonyl group is the attachment point to X1.
[0190] In certain embodiments, L is one of the following:
carbonyl group is the attachment point to X1.
[0191] In certain embodiments,
; wherein the point of attachment indicated on the carbonyl group is the attachment point to X1.
[0192] In certain embodiments, Lis selected from the groups depicted in the compounds in Table 1, below. In certain embodiments, L is selected from the groups depicted in the compounds in Table 2, below. In certain embodiments, L is selected from the groups depicted in the compounds in Table 3, below. In certain embodiments, Lis selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
Part C: Additional Embodiments
[0193] In certain embodiments,
taken together are one of the following:
[0194] In certain embodiments,
taken together are one of the following:
[0195] In certain embodiments,
taken together are one of the following:
[0196] In certain embodiments,
selected from the groups depicted in the compounds in Tables 1, 2, and 3, below.
[0197] The description above describes multiple embodiments relating to compounds of Formula I. The patent application specifically contemplates all combinations of the embodiments.
Exemplary Specific Compounds
[0198] In certain embodiments, the compound is a compound in Table 1 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 1. In certain embodiments, the compound is a compound in Table 2 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound
in Table 2. Tn certain embodiments, the compound is any one of compounds II-l to 11-26 in Table 2 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 3 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 3. In certain embodiments, the compound is a compound in Table 1, 2, or 3, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 1, 2, or 3.
TABLE 1. Exemplary Compounds
TABLE 2. Exemplary Compounds
ʼnll
TABLE 3. Exemplary Compounds
Synthetic Methods
[0199] Methods for preparing compounds described herein are illustrated in the following synthetic schemes. The schemes are provided for the purpose of illustrating the invention and are not intended to limit the scope or spirit of the invention. Starting materials shown in the schemes can be obtained from commercial sources or can be prepared based on procedures described in the literature.
[0200] In the schemes, it is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated (for example, use of protecting groups or alternative reactions). Protecting group chemistry and strategy is well known in the art, for example, as described in detail in Protecting Groups in Organic Synthesis, 3rd Edition, T. W. Greene and P. G. M. Wuts, John Wiley & Sons, 1999 and Greene's Protective Groups in Organic Synthesis, 5th Ed., (Peter G. M. Wuts, John Wiley & Sons: 2014), the entire contents of both of which are hereby incorporated by reference.
[0201] The synthetic route illustrated in Scheme 1-1 is a general method for preparing an intermediate A, which may be used in representative syntheses of a first series of N- benzothiazolyl benzenesulfonamide compounds shown in Table 1.
Scheme 1-1.
[0202] In step 1 of Scheme 1-1, cyclopentylmethoxy aniline compound 1 is cyclized with thiocyanate 2 and bromine in acetic acid to form intermediate A (using, e.g., (1) AcOH and (2) Br2, AcOH).
[0203] The synthetic route illustrated in Scheme 1-2 is a general method for preparing an intermediate B, which may be used in representative syntheses of a first series of benzothiazolyl benzenesulfonamide compounds shown in Table 1.
Scheme 1-2.
[0204] In step 1 of Scheme 1-2, the primary amino group of cyclohexyl carbamate compound 3 is protected by reaction with 4-nitrobenzenesulfonyl chloride 4 (using, e.g., TEA, DCM) to form nosyl-protected carbamate compound 5. In step 2, the nosyl-protected carbamate compound 5 is selectively alkylated (using, e.g., CS2CO3, CH3I, DMF), as described in WO2021156792A1, which is incorporated herein by reference in its entirety, to form methylated nosyl-protected carbamate compound 6. In step 3, methylated nosyl-protected carbamate compound 6 is deprotected (using, e.g., thioglycolic acid, CS2CO3, MeOH, DMF, 1 h, rt), as further described in WO2021156792 Al, to form benzyl (methylamino)cyclohexyl carbamate 7. Finally, in step 4, reductive amination of the secondary amine of benzyl (methylamino)cyclohexyl carbamate 7 (using, e.g., sodium triacetoxyborohydride) with carbamate aldehyde 8 followed by removal of the Cbz group (using, e.g., Pd/C, H2) provides intermediate B.
[0205] The synthetic route illustrated in Scheme 1-3 is a general method for preparing an intermediate C, which may be used in representative syntheses of a first series of benzothiazolyl benzenesulfonamide compounds shown in Table 1.
Scheme 1-3.
[0206] In step 1 of Scheme 1-3, cyclohexyl tert-butyl carbamate compound 9 is subjected to rhodium-catalyzed ether formation with ethyl diazoacetate 10 (using, e.g., cat. Rti2(AcO)4, DCM), as described in WO2023125121A1, which is incorporated herein by reference in its
entirety, to form ethyl (cyclohexylmethoxy)acetate compound 11. In step 2, ethyl (cyclohexylmethoxy) acetate compound 11 is partially reduced (using, e.g., DIBAL-H, -78 °C) to provide aldehyde compound 12. Finally, in step 3, reductive amination of aldehyde compound 12 with (methylamino)cyclohexyl carbamate compound 13 (using, e.g., sodium triacetoxyborohydride) followed by removal of the Cbz group (using, e g., Pd/C, H2) provides intermediate C.
[0207] The synthetic route illustrated in Scheme 1-4 is a general method for preparing a cyclopentylmethoxy benzenesulfonamide compound 21.
Scheme 1-4.
[0208] In step 1 of Scheme 1-4, reductive amination of the primary amine of intermediate A (using, c.g., (1) TiCl(Oz-Pr)s, DCM; (2) NaBH(OAc)s) with benzaldehyde 14, as described in W02023028077A1, forms dimethoxybenzene compound 15. In step 2, dimethoxybenzene compound 15 is converted to sulfonamide 17 by reaction with sulfonyl chloride 16 (using, e.g., (1) LiHMDS, THF -78 °C, 30 min, 0 °C; (2) -78 °C solution addition (THF) followed by 3 h RT), as further described in W02023028077A1. In step 3, sulfonamide 17 is subjected to nucleophilic aromatic substitution (SNAr) with intermediate B or intermediate C (using, e.g., DMSO, RT), as further described in W02023028077A1, to form cyclohexyl amino compound 18. In step 4, cyclohexyl amino compound 18 is globally deprotected (using, e.g., formic acid), as further described in W02023028077A1, to form primary amino compound 19. Finally, in step 5, primary amino compound 19 is reacted with (17?,4r)-4-(4-(((lS,47?)-4-(2-((2S,3S)-l- methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 21.
[0209] The synthetic route illustrated in Scheme 1-5 is a general method for preparing a benzenesulfonamide compound 30, which may be applied with appropriate adjustments to make additional compounds shown in Table 1.
Scheme 1-5.
[0210] In step 1 of Scheme 1-5, reductive amination of the primary amine of bromo benzothiazole 22 (using, e.g., (1) TiCl(Oz-Pr)3, DCM; (2) NaBH(OAc)s) with benzaldehyde 14, as described in W02023028077A1, forms dimethoxybenzene compound 23. In step 2, dimethoxybenzene compound 23 is converted to sulfonamide 24 by reaction with sulfonyl chloride 16 (using, e.g., (1) LiHMDS, THF -78 °C, 30 min, 0 °C; (2) -78 °C solution addition (THF) followed by 3 h RT), as further described in W02023028077A1. In step 3, sulfonamide 24 is subjected to nucleophilic aromatic substitution (SNAr) with cyclohexyl bis-amino compound 25 (using, e.g., DMSO, RT), as further described in W02023028077A1, to form cyclohexyl amino compound 26. In step 4, cyclohexyl amino compound 26 is subjected to a Cu- catalyzed coupling reaction with /c/'Z-butyl carbamate compound 27 (using, e.g., 2 mol% Cui, 2 mol% DPEO ligand, 1.2 eq. NaO/Bu, 4 A MS, dioxane, 60 °C), as described in J. Am. Chem. Soc., 2019, 141, 3541, which is incorporated herein by reference in its entirety, to form alkyl aryl ether compound 28. In step 5, alkyl aryl ether compound 28 is globally deprotected (using, e.g., formic acid), as further described in W02023028077A1, to form primary amino compound 29. Finally, in step 6, primary amino compound 29 is reacted with (17?,4r)-4-(4-(((15,4A)-4-(2- ((2S,35)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-
carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane-1 -carboxylic acid 20 (using, e.g, HATU, DIPEA) to form amide 30.
[0211] The synthetic route illustrated in Scheme 2-1 is a general method for preparing a thiadiazolyl benzenesulfonamide compound 32, which may be applied with appropriate adjustments to make additional compounds shown in Table 2.
Scheme 2-1.
[0212] In step 1 of Scheme 2-1, amine intermediate 31, prepared as described in US20170275275A1, which is incorporated herein by reference in its entirety, is reacted with (17?,4r)-4-(4-(((lS,47?)-4-(2-((2£,3S)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3- carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 32.
[0213] The synthetic route illustrated in Scheme 2-2 is a general method for preparing a thiadiazolyl benzenesulfonamide compound 44, which may be applied with appropriate adjustments to make additional compounds shown in Table 2.
Scheme 2-2.
[0214] In step 1 of Scheme 2-2, 4-iodopyridine 33 is converted to bromo biaryl intermediate 35 by reaction with aryl boronic acid 34 in a Suzuki -Miyaura coupling reaction (using, e.g., Pd(PPh3)4). In step 2, bromo biaryl intermediate 35 is converted to triaryl intermediate 37 by reaction with aryl boronic acid 36 in a Suzuki -Miyaura coupling reaction (using, e.g., Pd(PPh3)4). In step 3, the nitrile moiety in triaryl intermediate 37 is hydrogenated (using, e.g., 0.5 mol% [Ru(cod)methylallyl2], 0.5 mol% DPPF, 10 mol% /BuOK, H2, toluene), as described in Chemistry: A European Journal, 2008, 14, 9491, which is incorporated herein by reference in its entirety, to form benzyl amine 38. In step 4, the methyl ether in benzyl amine 38 is removed (using, e.g., 1 -decanethiol, NaOH, DMSO, heat) to form phenol intermediate 39. In step 5, the benzyl amine moiety in phenol intermediate 39 is selectively protected with Boc via a biphasic reaction (using, e.g., BOC2O, dioxane, NaOH) to form tert-butyl carbamate compound 40. In step 6, Zc/7-butyl carbamate compound 40 is subjected to nucleophilic aromatic substitution (SNAr) with commercially available thiadiazolyl benzenesulfonamide 41 (using, e.g., K2CO3, DMSO) to form thiadiazolyl Zc77-butyl carbamate compound 42. In step 7, the BOC protecting group is removed from thiadiazolyl Zt'rt-butyl carbamate compound 42 (using, e.g., HC1) to provide benzyl amine 43. Finally, in step 7, the benzyl amine 43 is reacted with (U?,4r)-4-(4-(((15,4J?)- 4-(2-((25,,3<S)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido)ethoxy)
cyclohexyl)oxy)butanamido) cyclohexane-1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 44.
[0215] The synthetic route illustrated in Scheme 2-3 is a general method for preparing a thiadiazolyl benzenesulfonamide compound 49, which may be applied with appropriate adjustments to make additional compounds shown in Table 2.
[0216] In step 1 of Scheme 2-3, reductive amination of benzyl amine 43, which may be prepared as described above in reference to Scheme 2-2, with piperidinyl aldehyde 45 (using, e.g., NaBEU) forms the thiadiazolyl piperidinyl compound 46. In step 2, the secondary amino moiety in thiadiazolyl piperidinyl compound 46 is protected as a trifluoroacetamide (e.g., trifluoroacetic anhydride 47) to form trifluoroacetylated compound 48. Finally, in step 3, trifluoroacetylated compound 48 is reacted with (17?,4r)-4-(4-(((lS,47?)-4-(2-((2S',3S)-l-methyl-5-oxo-2-(pyridin-3- yl)pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cy cl ohexane-1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) followed by removal of the trifluoroacetamide protecting group (using, e.g., MeOH, K2CO3) the to form amide 49.
[0217] The synthetic route illustrated in Scheme 3-1 is a general method for preparing an intermediate D, which may be used in representative syntheses of a first and second series of thiazolyl benzenesulfonamide compounds shown in Table 3.
Scheme 3-1.
[0218] In step 1 of Scheme 3-1, /erZ-butyl thiazol-2-ylcarbamate 50 is deprotonated (using, e.g., LiHMDS) and reacted with sulfonyl chloride 51 to form sulfonamide intermediate D.
[0219] The synthetic route illustrated in Scheme 3-2 is a general method for preparing a thiazolyl benzenesulfonamide compound 56, which may be applied with appropriate adjustments to make additional compounds shown in Table 3.
Scheme 3-2.
[0220] In step 1 of Scheme 3-2, 2,4-difluoro-5-nitropyridine 52 is subjected to nucleophilic aromatic substitution (SNAr) with mono-BOC protected 1,4-diamine 53 (using, c.g., K2CO3, DMF) followed by reduction of the nitro group (using, e.g, Na2S2C>4, EtOH/H2O) to form amino pyridine compound 54. In step 2, amino pyridine compound 54 is cross-coupled with sulfonamide intermediate D (using, e.g, (1) Pd(OAc)2, rac-BINAP) followed by global acid- mediated deprotection (using, e.g., HC1) to form amino compound 55. Finally, in step 3, amino compound 55 is reacted with (lA,4/')-4-(4-(((15',47?)-4-(2-((2S,35)-l-methyl-5-oxo-2-(pyridin-3- yl)pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, e.g., HATU, DIPEA) to form amide 56.
[0221] The synthetic route illustrated in Scheme 3-3 is a general method for preparing a thiazolyl benzenesulfonamide compound 61, which may be applied with appropriate adjustments to make additional compounds shown in Table 3.
Scheme 3-3.
[0222] In step 1 of Scheme 3-3, the benzyl amino group of commercially available 6- (aminomethyl)-4-chloropyridin-3-amine 57 is protected (using, e.g., BOC2O) to provide Boc- protected amino pyridine compound 58. In step 2, Boc-protected amino pyridine compound 58 is reacted with 3-furanylboronic acid under Suzuki-Miyaura coupling conditions to form bi-aryl compound 59. In step 3, bi-aryl compound 59 is cross-coupled with sulfonamide intermediate D (using, e.g, (1) Pd(OAc)2, rac-BINAP) followed by global acid-mediated deprotection (using, c.g, HC1) to form amino compound 60. Finally, in step 4, amino compound 60 is reacted with (lA,4r)-4-(4-(((lS,4A)-4-(2-((25,35Y)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido) ethoxy)cyclohexyl)oxy)butanamido) cyclohexane- 1 -carboxylic acid 20 (using, c.g., HATU, DIPEA) to form amide 61.
[0223] The modular synthetic route illustrated in Schemes 1-1, 1-2, 1-3, 1-4, 1-5, 2-1, 2-2, 2-3, 3-1, 3-2, and 3-3 can be adjusted to provide additional benzothiazolyl benzenesulfonamide,
thiadi azolyl benzenesulfonamide, thiazolyl benzenesulfonamide, and related compounds by conducting functional group transformations on the intermediate and final compounds. Such functional group transformations are well known in the art, as described in, for example, Comprehensive Organic Synthesis (B.M. Trost & I. Fleming, eds., 1991-1992); Organic Synthesis, 3rd Ed. (Michael B. Smith, Wavefunction, Inc., Irvine: 2010); Modern Methods of Organic Synthesis, 4th Ed. (William Carruthers and Iain Coldham, Cambridge University Press, Cambridge: 2004); March ’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 8th Ed, (Michael B. Smith, John Wiley & Sons, New York: 2020); and Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 3rd Ed. (Richard C. Larock, ed., John Wiley & Sons, New York: 2018).
II. Anti-cotinine Antibody
[0224] Also provided is an antibody, or antigen-binding fragment thereof, that binds to a cotinine moiety. As used herein, the term “anti-cotinine antibody or antigen-binding fragment thereof’ refers to an antibody, or antigen binding fragment thereof that binds to a cotinine moiety. Cotinine has the following structure:
[0225] As used herein, the term “cotinine moiety” refers to cotinine or an analog of cotinine. Compounds of Formula (I) described herein comprise a cotinine moiety linked via a linker to a Navl .7-binding moiety. In certain embodiments, the cotinine moiety has the following structure:
wherein R1 is C1.4 alkyl or C3-6 cycloalkyl. In certain embodiments, R1 is methyl, ethyl, 1- propyl, 2-propyl, 1-butyl, 2-butyl, or t-butyl. In certain embodiments, R1 is methyl. In certain embodiments, R1 is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
[0226] The term “antibody” is used herein in the broadest sense to refer to molecules with an immunoglobulin-like domain (for example IgG, IgM, IgA, IgD or IgE) and includes monoclonal, recombinant, polyclonal, chimeric, human, humanised, multispecific antibodies, including bispecific antibodies, and heteroconjugate antibodies; a single variable domain (e.g., a domain antibody (DAB)), antigen binding antibody fragments, Fab, F(ab’)2, Fv, disulphide linked Fv, single chain Fv, disulphide-linked scFv, diabodies, TANDABS, etc. and modified versions of any of the foregoing (for a summary of alternative “antibody” formats see Holliger and Hudson, Nature Biotechnology, 2005, 23(9): 1126-1136).
[0227] The term, full, whole or intact antibody, used interchangeably herein, refers to a heterotetrameric glycoprotein with an approximate molecular weight of 150,000 daltons. An intact antibody is composed of two identical heavy chains (HCs) and two identical light chains (LCs) linked by covalent disulphide bonds. This H2L2 structure folds to form three functional domains comprising two antigen-binding fragments, known as ‘Fab’ fragments, and a ‘Fc’ crystallisable fragment. The Fab fragment is composed of the variable domain at the aminoterminus, variable heavy (VH) or variable light (VL), and the constant domain at the carboxyl terminus, CHI (heavy) and CL (light). The Fc fragment is composed of two domains formed by dimerization of paired CH2 and CH3 regions. The Fc may elicit effector functions by binding to receptors on immune cells or by binding Clq, the first component of the classical complement pathway. The five classes of antibodies IgM, IgA, IgG, IgE and IgD are defined by distinct heavy chain amino acid sequences, which are called p, a, y, s and 8 respectively, each heavy chain can pair with either a K or X light chain. The majority of antibodies in the serum belong to the IgG class, there are four isotypes of human IgG (IgGl, IgG2, IgG3 and IgG4), the sequences of which differ mainly in their hinge region.
[0228] CDRs” are defined as the complementarity determining region amino acid sequences of an antibody or antigen binding fragment thereof. These are the hypervariable regions of immunoglobulin heavy and light chains. There are three heavy chain and three light chain CDRs (or CDR regions) in the variable portion of an immunoglobulin. Thus, “CDRs” as used herein
refers to all three heavy chain CDRs, all three light chain CDRs, all heavy and light chain CDRs, or at least two CDRs.
[0229] Throughout this specification, amino acid residues in variable domain sequences and variable domain regions within full-length antigen binding sequences, e.g. within an antibody heavy chain sequence or antibody light chain sequence, are numbered according to the Kabat numbering convention. Similarly, the terms “CDR”, “CDRL1”, “CDRL2”, “CDRL3”, “CDRH1”, “CDRH2”, “CDRH3” used in the Examples follow the Kabat numbering convention. For further information, see Kabat et al., Sequences of Proteins of Immunological Interest, 4th Ed., U.S. Department of Health and Human Services, National Institutes of Health (1987).
[0230] It will be apparent to those skilled in the art that there are alternative numbering conventions for amino acid residues in variable domain sequences and full-length antibody sequences. There are also alternative numbering conventions for CDR sequences, for example those set out in Chothia et al., Nature, 1989, 342: 877-883. The structure and protein folding of the antigen binding protein may mean that other residues are considered part of the CDR sequence and would be understood to be so by a skilled person.
[0231] Other numbering conventions for CDR sequences available to a skilled person include “AbM” (University of Bath) and “contact” (University College London) methods.
[0232] Table 4 below represents one definition using each numbering convention for each CDR or binding unit. It should be noted that some of the CDR definitions may vary depending on the individual publication used.
Table 4.
[0233] In a further embodiment, the anti-cotinine antibody is humanized. In a further embodiment, the Fc region of the anti-cotinine antibody is modified to increase ADCC activity,
ADCP activity, and/or CDC activity, suitable modifications of which are provided below. In a further embodiment, the Fc region of the anti-cotinine antibody is modified to increase ADCC activity.
[0234] Fc engineering methods can be applied to modify the functional or pharmacokinetics properties of an antibody. Effector function may be altered by making mutations in the Fc region that increase or decrease binding to Clq or Fey receptors and modify CDC or ADCC activity respectively. Modifications to the glycosylation pattern of an antibody can also be made to change the effector function. The in vivo half-life of an antibody can be altered by making mutations that affect binding of the Fc to the FcRn (neonatal Fc receptor).
[0235] The term “effector function” as used herein refers to one or more of antibody- mediated effects including antibody-dependent cell-mediated cytotoxicity (ADCC), antibody- mediated complement activation including complement-dependent cytotoxicity (CDC), complement-dependent cell-mediated phagocytosis (CDCP), antibody dependent complement- mediated cell lysis (ADCML), and Fc-mediated phagocytosis or antibody-dependent cellular phagocytosis (ADCP).
[0236] The interaction between the Fc region of an antigen binding protein or antibody and various Fc receptors (FcR), including FcyRI (CD64), FcyRII (CD32), FcyRIII (CD16), FcRn, Clq, and type II Fc receptors is believed to mediate the effector functions of the antigen binding protein or antibody. Significant biological effects can be a consequence of effector functionality. Usually, the ability to mediate effector function requires binding of the antigen binding protein or antibody to an antigen and not all antigen binding proteins or antibodies will mediate every effector function.
[0237] Effector function can be assessed in a number of ways including, for example, evaluating ADCC effector function of antibody coated to target cells mediated by Natural Killer (NK) cells via FcyRIII, or monocytes/macrophages via FcyRI, or evaluating CDC effector function of antibody coated to target cells mediated by complement cascade via Clq. For example, an antibody, or antigen binding fragment thereof, of the present invention can be assessed for ADCC effector function in a Natural Killer cell assay. Examples of such assays can be found in Shields et al., The Journal of Biological Chemistry, 2001, 276: 6591-6604; Chappel
et al., The Journal of Biological Chemistry, 1993, 268: 25124-25131 ; Lazar et al., PNAS, 2006, 103: 4005-4010.
[0238] Examples of assays to determine CDC function include those described in J Imm Meth, 1995, 184: 29-38.
[0239] The effects of mutations on effector functions (e.g., FcRn binding, FcyRs and Clq binding, CDC, ADCML, ADCC, ADCP) can be assessed, e.g., as described in Grevys et al., J Immunol., 2015,194(11): 5497-5508; Tam et al., Antibodies, 2017, 6(3): 12; or Monnet et al., mAbs, 2014, 6(2): 422-436.
[0240] Throughout this specification, amino acid residues in Fc regions, in antibody sequences or full-length antigen binding protein sequences, are numbered according to the EU index numbering convention.
[0241] Human IgGl constant regions containing specific mutations have been shown to enhance binding to Fc receptors. In some cases these mutations have also been shown to enhance effector functions, such as ADCC and CDC, as described below. Antibodies, or antigen binding fragments thereof, of the present invention may include any of the following mutations.
[0242] Enhanced CDC: Fc engineering can be used to enhance complement-based effector function. For example (with reference to IgGl), K326W/E333S; S267E/H268F/S324T; and IgGl/IgG3 cross subclass can increase Clq binding; E345R (Diebolder et al., Science, 2014, 343: 1260-1293) and E345R/E430G/S440Y results in preformed IgG hexamers (Wang et al., Protein Cell, 2018, 9(1): 63-73).
[0243] Enhanced ADCC: Fc engineering can be used to enhance ADCC. For example (with reference to IgGl), F243L/R292P/Y300L/V305I/P396L; S239D/I332E; and S298A/E333A/K334A increase FcyRIIIa binding; S239D/I332E/A330L increases FcyRIIIa binding and decreases FcyRIIb binding; G236A/S239D/I332E improves binding to FcyRIIa, improves the FcyRIIa/FcyRIIb binding ratio (activating/inhibitory ratio), and enhances phagocytosis of antibody-coated target cells by macrophages. An asymmetric Fc in which one heavy chain contains L234Y/L235Q/G236W/S239M/H268D/D270E/S298A mutations and D270E/K326D/A330M/K334E in the opposing heavy chain, increases affinity for FcyRIIIa FlSS
(a lower-affinity allele) and FcyRIIIa VI 58 (a higher-affinity allele) with no increased binding affinity to inhibitory FcyRIIb (Mimoto et al., mAbs, 2013, 5(2): 229-236).
[0244] Enhanced ADCP: Fc engineering can be used to enhance ADCP. For example (with reference to IgGl), G236A/S239D/I332E increases FcyRIIa binding and increases FcyRIIIa binding (Richards, J. et al., Mol. Cancer Ther., 2008, 7: 2517-2527).
[0245] Increased co-engagement: Fc engineering can be used to increase co-engagement with FcRs. For example (with reference to IgGl), S267E/L328F increases FcyRIIb binding; N325S/L328F increases FcyRIIa binding and decreases FcyRIIIa binding Wang et al., Protein Cell, 2018, 9(1): 63-73).
[0246] In a further embodiment, an antibody, or antigen binding fragment thereof, of the present invention may comprise a heavy chain constant region with an altered glycosylation profile, such that the antibody, or antigen binding fragment thereof, has an enhanced effector function, e.g., enhanced ADCC, enhanced CDC, or both enhanced ADCC and CDC. Examples of suitable methodologies to produce an antibody, or antigen binding fragment thereof, with an altered glycosylation profile are described in WO 2003/011878, WO 2006/014679 and EP1229125.
[0247] The absence of the al, 6 innermost fucose residues on the Fc glycan moiety on N297 of IgGl antibodies enhances affinity for FcyRIIIA As such, afucosylated or low fucosylated monoclonal antibodies may have increased therapeutic efficacy (Shields et al., J Biol Chem., 2002, 277(30): 26733-40 and Monnet et al., mAbs, 2014, 6(2): 422-436).
[0248] In certain embodiments there is provided an antibody, or antigen binding fragment thereof, comprising a chimeric heavy chain constant region. In an embodiment, the antibody, or antigen binding fragment thereof, comprises an IgGl/IgG3 chimeric heavy chain constant region, such that the antibody, or antigen binding fragment thereof, has an enhanced effector function, for example enhanced ADCC or enhanced CDC, or enhanced ADCC and CDC functions. For example, a chimeric antibody, or antigen binding fragment thereof, of the invention may comprise at least one CH2 domain from IgG3. In one such embodiment, the antibody, or antigen binding fragment thereof, comprises one CH2 domain from IgG3 or both CH2 domains may be from IgG3. In a further embodiment, the chimeric antibody, or antigen binding fragment thereof, comprises an IgGl CHI domain, an IgG3 CH2 domain, and an IgG3
CH3 domain. In a further embodiment, the chimeric antibody, or antigen binding fragment thereof, comprises an IgGl CHI domain, an IgG3 CH2 domain, and an IgG3 CH3 domain except for position 435 that is histidine.
[0249] In a further embodiment, the chimeric antibody, or antigen binding fragment thereof, comprises an IgGl CHI domain and at least one CH2 domain from IgG3. In an embodiment, the chimeric antibody, or antigen binding fragment thereof, comprises an IgGl CHI domain and the following residues, which correspond to lgG3 residues, in a CH2 domain: 274Q, 276K, 296F, 300F and 339T. In an embodiment, the chimeric antibody, or antigen binding fragment thereof, also comprises 356E, which corresponds to an IgG3 residue, within a CH3 domain. In an embodiment, the antibody, or antigen binding fragment thereof, also comprises one or more of the following residues, which correspond to IgG3 residues within a CH3 domain: 358M, 384S, 392N, 397M, 4221, 435R, and 436F.
[0250] Also provided is a method of producing an antibody, or antigen binding fragment thereof, according to the invention comprising the steps of: a) culturing a recombinant host cell comprising an expression vector comprising a nucleic acid sequence encoding a chimeric Fc region having both IgGl and IgG3 Fc region amino acid residues (e.g. as described above); and b) recovering the antibody, or antigen binding fragment thereof.
[0251] Such methods for the production of antibody, or antigen binding fragment thereof, with chimeric heavy chain constant regions can be performed, for example, using the COMPLEGENT technology system available from BioWa, Inc. (Princeton, NJ) and Kyowa Hakko Kirin Co., Ltd. The COMPLEGENT system comprises a recombinant host cell comprising an expression vector in which a nucleic acid sequence encoding a chimeric Fc region having both IgGl and IgG3 Fc region amino acid residues is expressed to produce an antibody, or antigen binding fragment thereof, having enhanced CDC activity, i.e. CDC activity is increased relative to an otherwise identical antibody, or antigen binding fragment thereof, lacking such a chimeric Fc region, as described in WO 2007/011041 and US 2007/0148165, each of which are incorporated herein by reference. In an alternative embodiment, CDC activity may be increased by introducing sequence specific mutations into the Fc region of an IgG chain. Those of ordinary skill in the art will also recognize other appropriate systems.
[0252] Also provided is a method of producing an antibody, or antigen binding fragment thereof, according to the invention comprising the steps of: a) culturing a recombinant host cell comprising an expression vector comprising a nucleic acid encoding the antibody, or antigen binding fragment thereof, optionally wherein the FUT8 gene encoding alpha- 1,6-fucosyltransf erase has been inactivated in the recombinant host cell; and b) recovering the antibody, or antigen binding fragment thereof.
[0253] Such methods for the production of an antibody, or antigen binding fragment thereof, can be performed, for example, using the POTELLIGENT technology system available from BioWa, Inc. (Princeton, NJ) in which CHOK1SV cells lacking a functional copy of the FUT8 gene produce monoclonal antibodies having enhanced ADCC activity that is increased relative to an identical monoclonal antibody produced in a cell with a functional FUT8 gene as described in US Patent No. 7,214,775, US Patent No. 6,946,292, WO 00/61739 and WO 02/31240, all of which are incorporated herein by reference. Those of ordinary skill in the art will also recognize other appropriate systems.
[0254] In certain embodiments, the antibody, or antigen binding fragment thereof, is produced in a host cell in which the FUT8 gene has been inactivated. In a further embodiment, the antibody, or antigen binding fragment thereof, is produced in a -I- FUT8 host cell. In a further embodiment, the antibody, or antigen binding fragment thereof, is afucosylated at Asn297 (IgGl).
[0255] It will be apparent to those skilled in the art that such modifications may not only be used alone but may be used in combination with each other in order to further enhance effector function.
[0256] In one such embodiment, there is provided an antibody, or antigen binding fragment thereof, comprising a heavy chain constant region that comprises a both a mutated and chimeric heavy chain constant region, individually described above. For example, an antibody, or antigen binding fragment thereof, comprising at least one CH2 domain from IgG3 and one CH2 domain from IgGl, and wherein the IgGl CH2 domain has one or more mutations at positions selected from 239, 332 and 330 (for example the mutations may be selected from S239D, I332E and
A330L), such that the antibody, or antigen binding fragment thereof, has enhanced effector function, e.g. enhanced ADCC or enhanced CDC, or enhanced ADCC and enhanced CDC in comparison to an equivalent antibody, or antigen binding fragment thereof, with an IgGl heavy chain constant region lacking said mutations. In certain embodiments, the IgGl CH2 domain has the mutations S239D and I332E. In certain embodiments, the IgGl CH2 domain has the mutations S239D, A330L, and I332E.
[0257] In an alternative embodiment, there is provided an antibody, or antigen binding fragment thereof, comprising both a chimeric heavy chain constant region and an altered glycosylation profile, as individually described above. In an embodiment, the antibody, or antigen binding fragment thereof, comprises an altered glycosylation profile such that the ratio of fucose to mannose is 0.8:3 or less. In one such embodiment, the heavy chain constant region comprises at least one CH2 domain from IgG3 and one CH2 domain from IgGl and has an altered glycosylation profile such that the ratio of fucose to mannose is 0.8:3 or less, for example wherein the antibody, or antigen binding fragment thereof, is defucosylated. Said antibody, or antigen binding fragment thereof, has an enhanced effector function, e.g. enhanced ADCC or enhanced CDC, or enhanced ADCC and enhanced CDC, in comparison to an equivalent antibody, or antigen binding fragment thereof, with an IgGl heavy chain constant region lacking said glycosylation profile.
[0258] In an alternative embodiment, the antibody, or antigen binding fragment thereof, has at least one IgG3 heavy chain CH2 domain and at least one heavy chain constant domain from IgGl wherein both IgG CH2 domains are mutated in accordance with the limitations described herein.
[0259] In one aspect, there is provided a method of producing an antibody, or antigen binding fragment thereof, according to the invention described herein comprising the steps of: a) culturing a recombinant host cell containing an expression vector comprising a nucleic acid sequence encoding a chimeric Fc domain having both IgGl and IgG3 Fc domain amino acid residues (e.g. as described above); and wherein the FUT8 gene encoding alpha- 1,6-fucosyltransferase has been inactivated in the recombinant host cell; and b) recovering the antibody, or antigen binding fragment thereof.
[0260] Such methods for the production of an antibody, or antigen binding fragment thereof, can be performed, for example, using the ACCRETAMAB technology system available from BioWa, Inc. (Princeton, NJ) that combines the POTELLIGENT and COMPLEGENT technology systems to produce an antibody, or antigen binding fragment thereof, having both enhanced ADCC and CDC activity relative to an otherwise identical monoclonal antibody that lacks a chimeric Fc domain and that is fucosylated.
[0261] In certain embodiments, there is provided an antibody, or antigen binding fragment thereof, comprising a mutated and chimeric heavy chain constant region wherein said antibody, or antigen binding fragment thereof, has an altered glycosylation profde such that the antibody, or antigen binding fragment thereof, has enhanced effector function, e.g. enhanced ADCC or enhanced CDC, or both enhanced ADCC and CDC. In certain embodiments the mutations are selected from positions 239, 332 and 330, e.g. S239D, I332E and A33OL. In a further embodiment the heavy chain constant region comprises at least one CH2 domain from IgG3 and one CHI domain from IgGl. In certain embodiments the heavy chain constant region has an altered glycosylation profile such that the ratio of fucose to mannose is 0.8:3 or less, e.g. the antibody, or antigen binding fragment thereof, is defucosylated, such that said antibody, or antigen binding fragment thereof, has an enhanced effector function in comparison with an equivalent non-chimeric antibody, or antigen binding fragment thereof, lacking said mutations and lacking said altered glycosylation profile.
[0262] In a further embodiment, the anti-cotinine antibody, or antigen binding fragment thereof, comprises a heavy chain CDR1 having SEQ ID NO: 1, a heavy chain CDR2 having SEQ ID NO: 2, a heavy chain CDR3 having SEQ ID NO: 3, a light chain CDR1 having SEQ ID NO: 4, a light chain CDR2 having SEQ ID NO: 5, and a light chain CDR3 having SEQ ID NO: 6. In a further embodiment, the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a CDR1 having SEQ ID NO: 1, a CDR2 having SEQ ID NO: 2, and a CDR3 having SEQ ID NO: 3, and the light chain comprising a CDR1 having SEQ ID NO: 4, a CDR2 having SEQ ID NO: 5, and a CDR3 having SEQ ID NO: 6. In a further embodiment, the anti- cotinine antibody is of IgGl isotype. In a further embodiment, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase or enhance ADCC activity. In a further embodiment, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase or enhance ADCC activity, wherein the substitution is S239D/I332E or
S239D/I332E/A330L, wherein residue numbering is according to the EU Index. In a further embodiment, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase or enhance ADCC activity, wherein the substitution is S239D/I332E, wherein residue numbering is according to the EU Index.
[0263] In a further embodiment, the anti-cotinine antibody, or antigen binding fragment thereof, comprises a heavy chain variable region (VH) having SEQ ID NO: 7, a light chain variable region (VL) having SEQ ID NO: 8. In a further embodiment, the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a heavy chain variable region (VH) having SEQ ID NO: 7, and the light chain comprising a light chain variable region (VL) having SEQ ID NO: 8. In a further embodiment, the anti-cotinine antibody is of IgGl isotype. In a further embodiment, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase or enhance ADCC activity. In a further embodiment, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase or enhance ADCC activity, wherein the substitution is S239D/I332E or S239D/I332E/A330L, wherein residue numbering is according to the EU Index. In a further embodiment, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase or enhance ADCC activity, wherein the substitution is S239D/I332E, wherein residue numbering is according to the EU Index.
[0264] In a further embodiment, the anti-cotinine antibody has a heavy chain comprising SEQ ID NO: 9 and a light chain comprising SEQ ID NO: 10.
[0265] Also provided is a pharmaceutical composition comprising an anti-cotinine antibody, or antigen binding fragment thereof as disclosed herein, and a pharmaceutically acceptable excipient, carrier, or diluent.
[0266] Also provided is a combination comprising a heterobifunctional compound described herein (e.g., a compound of Formula (I)), and an anti-cotinine antibody, or antigen-binding fragment thereof as disclosed herein. The heterobifunctional compound and anti-cotinine antibody, or antigen binding fragment thereof can be present in the same composition or in separate compositions. In certain embodiments, a combination comprises a pharmaceutical composition comprising a heterobifunctional compound (e.g., a compound of Formula (I)) and an anti-cotinine antibody, or antigen binding fragment thereof as disclosed herein, and a
pharmaceutically acceptable carrier, diluent, or excipient. In certain embodiments, a combination comprises a first pharmaceutical composition comprising a heterobifunctional compound (e.g., a compound of Formula (I)) and a pharmaceutically acceptable carrier, diluent, or excipient; and a second pharmaceutical composition comprising an anti-cotinine antibody or antigen binding fragment thereof as disclosed herein, and a pharmaceutically acceptable carrier, excipient, or diluent.
[0267] The heterobifunctional compounds (e.g., a compound of Formula (1)) and pharmaceutically acceptable salts thereof are capable of simultaneously binding a cell surface- expressed Nav1.7 and an anti-cotinine antibody, or antigen binding fragment thereof to form a ternary complex for the treatment and/or prevention of diseases or disorders associated with Nav1.7-expressing cells.
[0268] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered simultaneously. In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered simultaneously from a single composition, including as a fixed-dose composition or by pre-mixing the compound and the antibody, or antigen-binding fragment thereof, prior to administration. For example, the compound and the antibody, or antigen-binding fragment thereof, can be pre-mixed about 2 seconds to about 30 seconds, about 30 seconds to about 2 minutes, about 2 minutes to about 10 minutes, about 10 minutes to about 30 minutes, or about 30 minutes to about 2 hours prior to administration. In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered simultaneously from two separate compositions.
[0269] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered sequentially.
[0270] In certain embodiments, the compound and the antibody, or antigen-binding fragment thereof, whether administered simultaneously or sequentially, may be administered by the same route or may be administered by different routes. In certain embodiments, the compound and the antibody, or antigen-binding fragment thereof, are both administered intraveneously or subcutaneously, in the same composition or in separate compositions. In certain embodiments, the compound is administered orally and the antibody or antigen-binding fragment thereof is administered intravenously or subcutaneously.
[0271] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered in a molar ratio of compound to antibody, or antigen-binding fragment thereof, of about 2:1, about 1.8:1, about 1.6:1, about 1.5:1, about 1.4:1, about 1.3:1, about 1.2:1, about 1:1, about 1:1.2, about 1:1.3, about 1:1.4, about 1:1.5, about 1:1.6, about 1:1.8, about 1:2, about2:l to about 1.5:1, about 1.5:1 to about 1.2:1, about 1.2:1 to about 1:1, about 1:1 to about 1:1.2, about 1 : 1.2 to about 1 : 1.5, or about 1 : 1.5 to about 1 :2.
[0272] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are present as a combination in a molar ratio of compound to antibody, or antigenbinding fragment thereof, of about 2:1, about 1.8:1, about 1.6:1, about 1.5:1, about 1.4:1, about 1.3:1, about 1.2:1, about 1:1, about 1:1.2, about 1:1.3, about 1:1.4, about 1:1.5, about 1:1.6, about 1:1.8, about 1:2, about 2:1 to about 1.5:1, about 1.5:1 to about 1.2:1, about 1.2:1 to about 1:1, about 1:1 to about 1:1.2, about 1:1.2 to about 1:1.5, or about 1:1.5 to about 1:2.
[0273] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered at a dosage of compound of 0.0001 mg/kg to 1 mg/kg and antibody of 0.01 mg/kg to 100 mg/kg. For example, in a further embodiment, the compound is administered at a dosage of about 0.0001 mg/kg to about 0.0002 mg/kg, about 0.0002 mg/kg to about 0.0003 mg/kg, about 0.0003 mg/kg to about 0.0004 mg/kg, about 0.0004 mg/kg to about 0.0005 mg/kg, about 0.0005 mg/kg to about 0.001 mg/kg, about 0.001 mg/kg to about 0.002 mg/kg, about 0.002 mg/kg to about 0.003 mg/kg, about 0.003 mg/kg to about 0.004 mg/kg, about 0.004 mg/kg to about 0.005 mg/kg, about 0.005 mg/kg to about 0.01 mg/kg, about 0.01 mg/kg to about 0.02 mg/kg, about 0.02 mg/kg to about 0.03 mg/kg, about 0.03 mg/kg to about 0.04 mg/kg, about 0.04 mg/kg to about 0.05 mg/kg, about 0.05 mg/kg to about 0.1 mg/kg, about 0.1 mg/kg to about 0.2 mg/kg, about 0.2 mg/kg to about 0.3 mg/kg, about 0.3 mg/kg to about 0.4 mg/kg, about 0.4 mg/kg to about 0.5 mg/kg, and/or about 0.5 mg/kg to about 1 mg/kg, and the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 0.01 mg/kg to about 0.02 mg/kg, about 0.02 mg/kg to about 0.03 mg/kg, about 0.03 mg/kg to about 0.04 mg/kg, about 0.04 mg/kg to about 0.05 mg/kg, about 0.05 mg/kg to about 0.1 mg/kg, about 0.1 mg/kg to about 0.2 mg/kg, about 0.2 mg/kg to about 0.3 mg/kg, about 0.3 mg/kg to about 0.4 mg/kg, about 0.4 mg/kg to about 0.5 mg/kg, about 0.5 mg/kg to about 1 mg/kg, about 1 mg/kg to about 2 mg/kg, about 2 mg/kg to about 3 mg/kg, about 3 mg/kg to about 4 mg/kg, about 4 mg/kg to about 5 mg/kg, about 5 mg/kg to about 10 mg/kg, about 10 mg/kg to about 15 mg/kg, about 15 mg/kg to
about 20 mg/kg, about 20 mg/kg to about 25 mg/kg, about 25 mg/kg to about 30 mg/kg, about 30 mg/kg to about 35 mg/kg, about 35 mg/kg to about 40 mg/kg, about 40 mg/kg to about 45 mg/kg, about 45 mg/kg to about 50 mg/kg, about 50 mg/kg to about 60 mg/kg, about 60 mg/kg to about 70 mg/kg, about 70 mg/kg to about 80 mg/kg, about 80 mg/kg to about 90 mg/kg, and/or about 90 mg/kg to about 100 mg/kg.
[0274] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered at a dosage of compound of 0.007 mg to 70 mg and antibody of 0.7 mg to 7000 mg. For example, in a further embodiment, the compound is administered at a dosage of about 0.007 mg to about 0.01 mg, about 0.01 mg to about 0.02 mg, about 0.02 mg to about 0.03 mg, about 0.03 mg to about 0.04 mg, about 0.04 mg to about 0.05 mg, about 0.05 mg to about 0.1 mg, about 0.1 mg to about 0.2 mg, about 0.2 mg to about 0.3 mg, about 0.3 mg to about 0.4 mg, about 0.4 mg to about 0.5 mg, about 0.5 mg to about 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, about 5 mg to about 10 mg, about 10 mg to about 20 mg, about 20 mg to about 30 mg, about 30 mg to about 40 mg, about 40 mg to about 50 mg, about 50 mg to about 60 mg, and/or about 60 mg to about 70 mg, and the antibody, or antigen-binding fragment thereof, is administered at a dosage of about 0.7 mg to about 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, about 5 mg to about 10 mg, about 10 mg to about 20 mg, about 20 mg to about 30 mg, about 30 mg to about 40 mg, about 40 mg to about 50 mg, about 50 mg to about 100 mg, about 100 mg to about 500 mg, about 500 mg to about 1000 mg, about 1000 mg to about 1500 mg, about 1500 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2500 mg to about 3000 mg, about 3000 mg to about 3500 mg, about 3500 mg to about 4000 mg, about 4000 mg to about 4500 mg, about 4500 mg to about 5000 mg, about 5000 mg to about 5500 mg, about 5500 mg to about 6000 mg, about 6000 mg to about 6500 mg, and/or about 6500 mg to about 7000 mg.
[0275] In a further embodiment, the compound and the antibody, or antigen-binding fragment thereof, are administered in a molar ratio and/or dosage as described herein once every week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, or once every six weeks for a period of one week to one year, such as a period of one week, one month, two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, or twelve months.
III. Therapeutic Applications
[0276] One aspect of the invention provides a method of treating or preventing a disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (e.g., a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
[0277] Another aspect of the invention provides a method of treating or preventing a Nav1.7- associated disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigenbinding fragment thereof.
[0278] Another aspect of the invention provides a method of treating or preventing a disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof, wherein the disease or condition is selected from pain, cough, acute itch, or chronic itch.
[0279] In certain embodiments, the method is to treating the disease or condition. In certain embodiments, the method is to preventing the disease or condition.
[0280] Another aspect of the invention provides a method of treating or preventing pain in a patient in need thereof, wherein the method comprises administering to the patient a therapeutically effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
[0281] In certain embodiments, the method is to treating pain. In certain embodiments, the method is to preventing pain.
[0282] In certain embodiments, the pain is chronic pain. In certain embodiments, the pain is acute pain. In certain embodiments, the pain is neuropathic pain. In certain other embodiments, the pain is inflammatory pain. In certain embodiments, the pain is arthritis pain. In certain embodiments, the pain is arthritis pain selected from osteoarthritis pain and rheumatoid arthritis pain.
[0283] In certain other embodiments, the pain is pain due to cancer. In certain embodiments, the pain is due to a cancer selected from the group consisting of a solid tumor, leukemia, and lymphoma. In certain embodiments, the pain is due to a cancer selected from the group consisting of a bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, endometrial cancer, esophageal cancer, leukemia, lung cancer, liver cancer, skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, renal cancer, stomach cancer, testicular cancer, and uterine cancer.
[0284] In certain other embodiments, the pain is complex regional pain syndrome. In certain embodiments, the complex regional pain syndrome is reflex sympathetic dystrophy pain. In certain other embodiments, the pain is trauma pain. In certain embodiments, the pain is due to surgery.
[0285] In certain other embodiments, the pain is located in the patient’s hand, wrist, arm, shoulder, back, leg, knee, ankle, foot, toe, neck, or head. In certain embodiments, the pain is low back pain. In certain embodiments, the pain is chronic low back pain.
[0286] In certain other embodiments, the pain is a neuropathic pain selected from the group consisting of low back pain, hip pain, leg pain, non-herpetic neuralgia, post-herpetic neuralgia, diabetic neuropathy pain, lumbosacral radiculopathy pain, nerve injury-induced pain, acquired immune deficiency syndrome (AIDS) related neuropathic pain, head trauma pain, phantom limb pain, multiple sclerosis pain, root avulsion pain, painful traumatic mononeuropathy, painful polyneuropathy, thalamic pain syndrome, post-stroke pain, central nervous system injury pain, post-surgical pain, carpal tunnel syndrome pain, trigeminal neuralgia pain, post mastectomy syndrome pain, post-thoracotomy syndrome pain, stump pain, repetitive motion pain, neuropathic pain associated hyperalgesia and allodynia, drug-induced pain, toxin-caused nerve injury pain, chemotherapy-caused nerve injury pain, and combinations thereof.
Amount of Reduction in Pain Intensity
[0287] The method may be further characterized according to the amount of reduction in pain intensity relative to pain observed without performing the method. Accordingly, in certain embodiments, the method is characterized by achieving at least a 20% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least a 40% reduction in pain intensity relative to pain observed
without performing the method. In certain embodiments, the method is characterized by achieving at least a 60% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least an 80% reduction in pain intensity relative to pain observed without performing the method. In certain embodiments, the method is characterized by achieving at least a 90% reduction in pain intensity relative to pain observed without performing the method.
Duration of Reduction in Pain Intensity
[0288] The method may be further characterized according to the duration of reduction in pain intensity. Accordingly, in certain embodiments, the reduction in pain intensity lasts for at least 1 week. In certain embodiments, the reduction in pain intensity lasts for at least 2 weeks. In certain embodiments, the reduction in pain intensity lasts for at least 4 weeks. In certain embodiments, the reduction in pain intensity lasts for at least 2 months. In certain embodiments, the reduction in pain intensity lasts for at least 3 months. In certain embodiments, the reduction in pain intensity lasts for at least 6 months.
[0289] In certain embodiments, the reduction in pain intensity lasts for at a duration of 2 months to six months. In certain embodiments, the reduction in pain intensity lasts for a duration of 3 months to 9 months. In certain embodiments, the reduction in pain intensity lasts for a duration of 6 months to 9 months. In certain embodiments, the reduction in pain intensity lasts for a duration of 6 months to 12 months.
Increasing Antibody-dependent Cell Cytotoxicity
[0290] Another aspect of the invention provides a method of increasing antibody-dependent cell cytotoxicity (ADCC) of voltage-gated sodium channel Nav1 /-expressing cells, wherein the method comprises contacting the cells with an effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti -cotinine antibody, or antigen-binding fragment thereof.
Depleting
[0291] Another aspect of the invention provides a method of depleting voltage-gated sodium channel Nav1.7-expressing cells, wherein the method comprises contacting the cells with an
effective amount of a heterobifunctional compound described herein (such as a compound of Formula I) and an anti-cotinine antibody, or antigen-binding fragment thereof.
Administration Aspects
[0292] In certain embodiments, the compound and the antibody, or antigen-binding fragment thereof, are administered simultaneously.
[0293] In certain embodiments, the compound and the antibody, or antigen-binding fragment thereof, are administered sequentially.
Aspects of the Anti-cotinine Antibody
[0294] In certain embodiments, the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a CDR1 having SEQ ID NO: 1, a CDR2 having SEQ ID NO: 2, and a CDR3 having SEQ ID NO: 3, and the light chain comprising a CDR1 having SEQ ID NO: 4, a CDR2 having SEQ ID NO: 5, and a CDR3 having SEQ ID NO: 6.
[0295] In certain embodiments, the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a heavy chain variable region (VH) having SEQ ID NO: 7, and the light chain comprising a light chain variable region (VL) having SEQ ID NO: 8.
[0296] In certain embodiments, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase ADCC activity.
[0297] In certain embodiments, the substitution in the Fc region is S239D/I332E, wherein residue numbering is according to the EU Index.
[0298] In certain embodiments, the anti-cotinine antibody has a heavy chain comprising SEQ ID NO: 9 and a light chain comprising SEQ ID NO: 10.
Subjects
[0299] In certain embodiments, the subject is a human. In certain embodiments, the subject is an adult human. In certain embodiments, the subject is a pediatric human.
Medical Uses
[0300] Another aspect of the invention provides for the use of a heterobifunctional compound described herein (such as a compound of Formula I, or other compounds in Section I) in the
manufacture of a medicament. In certain embodiments, the medicament is for treating a disorder described herein, such as pain.
[0301] Another aspect of the invention provides for the use of a heterobifunctional compound described herein (such as a compound of Formula I, or other compounds in Section I) for treating a medical disorder, such as a medical disorder described herein, such as pain.
[0302] Another aspect of the invention provides a heterobifunctional compound described herein (such as a compound of Formula I, or other compounds in Section I) for use in treating a medical disorder, such as a medical disorder described herein, such as pain.
Combination
[0303] Another aspect of the invention provides a combination comprising a heterobifunctional compound described herein (e.g., a compound of Formula I) and an anti- cotinine antibody, or antigen-binding fragment thereof.
[0304] In certain embodiments, the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a CDR1 having SEQ ID NO: 1, a CDR2 having SEQ ID NO: 2, and a CDR3 having SEQ ID NO: 3, and the light chain comprising a CDR1 having SEQ ID NO: 4, a CDR2 having SEQ ID NO: 5, and a CDR3 having SEQ ID NO: 6.
[0305] In certain embodiments, the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a heavy chain variable region (VH) having SEQ ID NO: 7, and the light chain comprising a light chain variable region (VL) having SEQ ID NO: 8.
[0306] In certain embodiments, the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase ADCC activity.
[0307] In certain embodiments, the substitution in the Fc region is S239D/I332E, wherein residue numbering is according to the EU Index.
[0308] In certain embodiments, the anti-cotinine antibody has a heavy chain comprising SEQ ID NO: 9 and a light chain comprising SEQ ID NO: 10.
IV. Biological Assays for Evaluating Compound Activity
[0309] Heterobifunctional compounds may be evaluated for biological activity using one or more of the assays described below.
Assay 1; Antibody Dependent Cellular Cytotoxicity Reporter Assay
[0310] An antibody dependent cellular cytoxocity reporter assay is conducted using the following four assay components: (i) ARM compound of Formula (I) targeting Nav1.7 (concentrations ranging from 1 pM to 10 pM) (ii) anti-cotinine antibody having a heavy chain sequence of SEQ ID NO: 11 and a light chain sequence of SEQ ID NO: 12 (rabbit variable region with human IgGl Fc domain containing a DE mutation (S239D/I332E)) (concentrations ranging from 0.01 pg / mb to 200 pg / mL); (iii) target cells: cells engineered to overexpress human Nav1.7 (typically 1000-20,000 cells per well) and (iv) reporter cells: Reagents are combined in a final volume of 20 pL in a 384 - well tissue culture treated plate. All four assay components are incubated together for about 12-18 hours. Thereafter, BioGio Detection reagent (Promega) is added to the wells to lyse the cells and provide a substrate for the luciferase reporter protein. Luminescence signal is measured on a microplate reader and signal background is calculated by dividing the signal of a test well by the signal obtained when no heterobivalent compound of Formula (I) was added. EC50 calculations are done using Graphpad Prism Software, specifically a nonlinear regression curve fit ( Y = Bottom + ( Top - Bottom ) / ( 1 + 10 A ( ( Log EC50 - X ) * HillSlope ) ) ).
Assay 2: Binding of Anti-Cotinine Antibody to ARM Compounds of Formula I Measured by Surface Plasmon Resonance (SPR)
[0311] Anti-cotinine antibodies having a heavy chain sequence of SEQ ID NO: 11 and a light chain sequence of SEQ ID NO: 12 (rabbit variable region with human IgGl Fc domain containing a DE mutation (S239D/I332E)) can be captured on one or more flow cells of a protein A sensor chip (Cytiva) using a Biacore T200 while reserving flow cell 1 as a reference. Following capture, a 3000 second wait step is included to reduce drift during compound analysis. ARM compounds (e.g., of Formula I) are then injected at 100 pL/min with 200 and 2000 second association and dissociation times. The entire experiment may be run at 37°C with running buffer containing 10 mM HEPES pH 7.4, 150 mM NaCl, 0.005% P20 and 1% DMSO. ARM compounds of Formula (I) are titrated with a top concentration of 200 nM using a 3-fold 5-point dilution series and a corresponding 5-injection buffer cycle is run for blank subtraction. Data are double referenced by subtracting the response of the reference flow cell from that of the antibody-containing flow cell and subsequently subtracting the referenced blank sensorgrams.
Following compound analysis, the surface is regenerated using a 30 second injection of pH 1 .5 glycine at a flow rate of 30 pL/min after which antibody is re-captured. The experiment may be run using Biacore T200 control software and evaluated using Biacore T200 Evaluation software. Curves are fit with a 1 : 1 kinetic binding model to obtain kon (1/Ms), koff (1/s), Kd (M) determined as koff/kon, and residence time determined as l/k0±r. (s).
[0312] Anti-cotinine antibodies having a heavy chain sequence of SEQ ID NO: 9 and a light chain sequence of SEQ ID NO: 10 (humanized version) or a heavy chain sequence of SEQ ID NO: 11 and a light chain sequence of SEQ ID NO: 12 (rabbit version) may be captured via the Fc domain to a protein A or A/G surface on flow cell 2 of a CM5 sensor chip using the Biacore 8k. Both antibodies may have a human IgGl Fc domain containing a DE mutation (S239D/I332E).
[0313] After antibody capture, a 1500 second wait step is included to reduce drift in the binding step. ARM compounds of Formula (I) are then flowed over the captured antibodies at varying top concentrations ranging from 250 nM to 4 pM. The top concentration is diluted 4-fold over 5 dilutions (with a 0 nM compound cycle included to blank subtract the data). Association and dissociation times of the ARM compounds of Formula (I) may vary between experiments as follows: reference 1- association for 600 seconds and dissociation for 1200 seconds at 20pl/min; reference 2- association for 240 seconds and dissociation for 300 seconds at 30pl/min; reference 3- association for 360 seconds and dissociation for 600 seconds at 30pl/min. Anti-cotinine antibody bound to the protein A or A/G surface is regenerated using 50mM NaOH. Experiments are run at 25°C at pH 7.4 using HBS-EP+ buffer using Biacore 8K control software and evaluated using Biacore Insight Evaluation software. Curves are fit with the 1 : 1 kinetic fit inherent to the software, utilising a local Rmax and local drift setting.
V. Combination Therapy
[0314] Another aspect of the invention provides for combination therapy. Heterobifunctional compounds described herein (such as a compound of Formula I, or other compounds in Section I) or their pharmaceutically acceptable salts may be used in combination with additional therapeutic agents to treat medical disorders, such as pain.
[0315] In some embodiments, the present invention provides a method of treating a disclosed disease or condition comprising administering to a patient in need thereof an effective amount of
a compound disclosed herein or a pharmaceutically acceptable salt thereof and co-administering simultaneously or sequentially an effective amount of one or more additional therapeutic agents, such as those described herein. In some embodiments, the method further comprises administering an anti-cotinine antibody, or antigen-binding fragment thereof. In some embodiments, the method includes co-administering one additional therapeutic agent. In some embodiments, the method includes co-administering two additional therapeutic agents. In some embodiments, the combination of the disclosed compound and the additional therapeutic agent or agents acts synergistically.
[0316] One or more other therapeutic agent may be administered separately from a compound or composition of the invention, as part of a multiple dosage regimen. Alternatively, one or more other therapeutic agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as a multiple dosage regime, one or more other therapeutic agent and a compound or composition of the invention may be administered simultaneously, sequentially or within a period of time from one another, for example within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 18, 20, 21, 22, 23, or 24 hours from one another. In some embodiments, one or more other therapeutic agent and a compound or composition of the invention are administered as a multiple dosage regimen more than 24 hours apart.
[0317] Additional therapeutic agents for treating pain include, for example, an opioid analgesic (e.g., alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl; heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacyl morphan, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbuphine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, normorphine, norpipanone, opium, oxycodone, oxymorphone, papaveretum, pentazocine, phenadoxone, phenomorphan, phenazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propiram, propoxyphene, sufentanil, tramadol, tilidine, salts thereof and mixtures thereof), a non-steroidal anti-inflammatory drug
(e.g., aspirin, ibuprofen, diclofenac, diflunisal, ibuprofen, naproxen, fenoprofen, piroxicam, flurbiprofen, mefenamic acid, sulindac, salts thereof and mixtures thereof), and acetaminophen.
Additional Considerations
[0318] The doses and dosage regimen of the active ingredients used in the combination therapy may be determined by an attending clinician. In certain embodiments, the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) are administered in doses commonly employed when such agents are used as monotherapy for treating the disorder. In other embodiments, the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) are administered in doses lower than the doses commonly employed when such agents are used as monotherapy for treating the disorder. In certain embodiments, the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) are present in the same composition, which is suitable for oral administration.
[0319] In certain embodiments, the compound described herein (such as a compound of Formula I, or other compounds in Section I) and the additional therapeutic agent(s) may act additively or synergistically. A synergistic combination may allow the use of lower dosages of one or more agents and/or less frequent administration of one or more agents of a combination therapy. A lower dosage or less frequent administration of one or more agents may lower toxicity of the therapy without reducing the efficacy of the therapy.
[0320] Another aspect of this invention is a kit comprising a therapeutically effective amount of the compound described herein (such as a compound of Formula I, or other compounds in Section I), a pharmaceutically acceptable carrier, vehicle or diluent, and optionally at least one additional therapeutic agent listed above. In some embodiments, the kit further comprises an anti -cotinine antibody, or antigen-binding fragment thereof.
VI. Pharmaceutical Compositions and Dosing Considerations
[0321] As indicated above, the invention provides pharmaceutical compositions, which comprise a therapeutically effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. The pharmaceutical compositions may be specially formulated for administration in
solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally. In certain embodiments, the invention provides a pharmaceutical composition comprising a compound described herein (such as a compound of Formula I, or other compounds in Section I) and a pharmaceutically acceptable carrier.
[0322] The phrase “therapeutically effective amount” as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.
[0323] The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0324] Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
[0325] Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
[0326] Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
[0327] In certain embodiments, a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and poly anhydrides; and a compound of the present invention. In certain embodiments, an aforementioned formulation renders orally bioavailable a compound of the present invention.
[0328] Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
[0329] Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.
[0330] In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules, trouches and the like), the active ingredient is mixed with one or
more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fdlers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, zinc stearate, sodium stearate, stearic acid, and mixtures thereof; (10) coloring agents; and (11) controlled release agents such as crospovidone or ethyl cellulose. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
[0331] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
[0332] The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved
in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
[0333] Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
[0334] Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
[0335] Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
[0336] Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
[0337] Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
[0338] Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
[0339] The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
[0340] Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
[0341] Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
[0342] Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
[0343] Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
[0344] Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol,
propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
[0345] These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
[0346] In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
[0347] Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
[0348] When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier.
[0349] The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For
example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred.
[0350] The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
[0351] The phrases “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient’s system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
[0352] These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intraci sternal ly and topically, as by powders, ointments or drops, including buccally and sublingually.
[0353] Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
[0354] Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
[0355] The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular
compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
[0356] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
[0357] In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Preferably, the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. When the compounds described herein are co-administered with another agent (e.g., as sensitizing agents), the effective amount may be less than when the agent is used alone.
[0358] If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day.
[0359] The invention further provides a unit dosage form (such as a tablet or capsule) comprising a compound described herein in a therapeutically effective amount for the treatment of a medical disorder described herein.
VII. SEQUENCE LISTINGS
[0360] Heavy chain CDR1 amino acid sequence (SEQ ID NO: 1): NYWMS
[0361] Heavy chain CDR2 amino acid sequence (SEQ ID NO: 2):
DIHGNRGFNYHASWAKG
[0362] Heavy chain CDR3 amino acid sequence (SEQ ID NO: 3): ADDSGSHDI
[0363] Light chain CDR1 amino acid sequence (SEQ ID NO: 4): QSSQSVYSAKLS
[0364] Light chain CDR2 amino acid sequence (SEQ ID NO: 5): YGSTLAS
[0365] Light chain CDR3 amino acid sequence (SEQ ID NO: 6): QGTFYGPDWYFA
[0366] Variable heavy chain amino acid sequence (SEQ ID NO: 7):
EVQLVESGGGLVQPGGSLRLSCAASGFTFSNYWMSWVRQAPGKGLEWVGDIHG
NRGFNYHASWAKGRFTVSRSKNTLYLQMNSLRAEDTAVYYCAKADDSGSHDIW GQGTLVTVSS
[0367] Variable light chain amino acid sequence (SEQ ID NO: 8): DIQMTQSPSSLSASVGDRVTITCQSSQSVYSAKLSWYQQKPGKAPKLLIYYGSTLASGVP SRFSGSGSGTQFTLTISSLQPEDFATYYCQGTFYGPDWYFAFGGGTKVEIK
[0368] Heavy chain amino acid sequence (SEQ ID NO: 9): EVQLVESGGGLVQPGGSLRLSCAASGFTFSNYWMSWVRQAPGKGLEWVGDIHGNRGF NYHASWAKGRFTVSRSKNTLYLQMNSLRAEDTAVYYCAKADDSGSHDIWGQGTLVTV SSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ S SGL YSL S S VVTVP S S SLGTQT YICNVNHKP SNTKVDKKVEPKSCDKTHTCPPCP APELL GGPDVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPRE EQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPEEKTISKAKGQPREPQVYTL PP SRDELTKNQ VSLTCLVKGF YP SDIAVEWE SNGQPENNYKTTPP VLD SDGSFFL YSKLT VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0369] Light chain amino acid sequence (SEQ ID NO: 10):
DIQMTQ SP S SL S AS VGDRVTITCQ S SQ S VYS AKLS WYQQKPGKAPKLLIYYGST LASGVPSRFSGSGSGTQFTLTISSLQPEDFATYYCQGTFYGPDWYFAFGGGTKV EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGN SQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC [0370] Heavy chain amino acid sequence (SEQ ID NO: 11): QQQLVESGGR LVTPGGSLTL TCTASGFSLN NYWMSWVRQA PGKGLEWIGD IHGNRGFNYH ASWAKGRFTY SRTSTTVDLR MTSLTTEDTA IYFCARADDS GSHDIWGPGT LVTVSSASTK GPSVFPLAPS SKSTSGGTAA LGCLVKDYFP EPVTVSWNSG ALTSGVHTFP AVLQSSGLYS LSSVVTVPSS SLGTQTYICN VNHKPSNTKV DKKVEPKSCD KTHTCPPCPA PELLGGPDVF LFPPKPKDTL MISRTPEVTC VVVDVSHEDP EVKFNWYVDG VEVHN AKTKP REEQYNSTYR VVS VLTVLHQ DWLNGKEYKC KVSNKALPAP EEKTISKAKG QPREPQVYTL PPSRDELTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGK [0371] Light chain amino acid sequence (SEQ ID NO: 12):
ELDLTOTPSPVSAAVGDTVTINCOSSOSVYSAKLSWYOQKPGOPPKLLIYYGSTLASGV
PSRFKGSGSGTQFSLTISDVOCADAATYYCQGTYYGPDWYFAFGGGTEVVVKRTVAA PSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDS TYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
[0372] Heavy chain amino acid sequence (SEQ ID NO: 13):
OQOLVESGGRLVTPGGSLTLTCTASGFSLNNYWMSWVRQAPGKGLEWIGDIHGNRGF NYHASWAKGRFTVSRTSTTVDLRMTSLTTEDTAIYFCARADDSGSHDIWGPGTLVTVS S AKTT APS VYPLAP VC GDTTGS S VTLGCL VKGYFPEP VTLTWNSGSL S SGVHTFP AVLQ S
DLYTLSSSVTVTSSTWPSQSITCNVAHPASSTKVDKKIEPRGPTIKPCPPCKCPAPNLLGG PSVFIFPPKIKDVLMISLSPMVTCVVVDVSEDDPDVQISWFVNNVEVLTAQTQTHREDYN STLRVVSALPIQHQDWMSGKEFKCKVNNKALPAPIERTISKPKGSVRAPQVYVLPPPEEE MTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSKLRVEK KNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGK
[0373] Light chain amino acid sequence (SEQ ID NO: 14):
ELDLTQTPSPVSAAVGDTVTINCOSSOSVYSAKLSWYOQKPGOPPKLLIYYGSTLASGV PSRFKGSGSGTQFSLTISDVOCADAATYYCQGTYYGPDWYFAFGGGTEVVVKRADAA PTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQNGVLNSWTDQDSKDST YSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC
EXAMPLES
[0374] The invention now being generally described, will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
EXAMPLE 1 - Synthesis of (2X,3^-Af-(2-(3-(4-(2-(((4-(4-(4-(N-(l,2,4-thiadiazol-5-yl) sulfamoyI)-2-cyanophenoxy)-3'-(trifluoromethyl)-[l,l'-biphenyl]-3-yl)pyridin-2-yI) methyl)amino)ethyl)piperidin-l-yl)-3-oxopropoxy)ethyl)-l-methyl-5-oxo-2-(pyridin-3- yl)pyrrolidine-3-carboxamide, compound (11-27).
[0375] Step 1: Preparation of tert-butyl 3-(2-((25',3KS)-l-methyl-5-oxo-2-(pyridin-3-yl) pyrrolidine-3-carboxamido)ethoxy)propanoate (3). To a stirred solution of (25,3S)-l-methyl- 5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxylic acid (1) (0.5 g, 2.270 mmol) in dichloromethane (DCM) (8 mL), hexafluorophosphate azabenzotriazole tetramethyl uronium (HATU) (1.036 g, 2.72 mmol) and triethylamine (0.949 ml, 6.81 mmol) were added at room temperature. The mixture was stirred for 10 minutes. A solution of tert-butyl 3-(2-aminoethoxy)propanoate (2) (0.430 g, 2.270 mmol) in DCM (2 mL) was then added at room temperature. The reaction mixture was stirred at room temperature for 16 hours. The reaction was monitored by liquid chromatography-mass spectrometry (LCMS) / thin-layer chromatography (TLC). When deemed complete, the reaction mixture was quenched with water and extracted with DCM. The organic
layer was washed with brine and then evaporated under reduced pressure to yield the crude product. Crude product was purified via silica gel chromatography (0-9% methanol (MeOH):DCM) to afford the title compound 3 (0.7 g, 1.35 mmol, 59% yield) as a pale-yellow liquid. LC-MS m/z 392.2 (M+H)+.
[0376] Step 2: Preparation of 3-(2-((25,35)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-
3-carboxamido)ethoxy)propanoic acid (4). To a stirred solution of /e/7-butyl 3-(2-((25,35)-l- methyl-5-oxo-2-(pyridin-3-yl)pyrrolidine-3-carboxamido)ethoxy)propanoate (3) (0.7 g, 1.788 mmol) in DCM (0.8 ml), HC1 (4 M solution in dioxane, 2.68 mL, 10.73 mmol) was added at 0 °C. The reaction was allowed to come to room temperature. After 3 hours, the reaction was deemed complete by TLC, and the reaction mixture was concentrated. The crude product was triturated with diethyl ether. The diethyl ether was removed, and the obtained residue was dried under high vacuum to afford the title compound 4 (0.45 g, 1.03 mmol, 58%). LC-MS m/z 336.2 (M+H)+.
[0377] Step 3: Preparation of (25,3 ^)-Af-(2-(3-(4-(2-(((4-(4-(4-(Af-(l,2,4-thiadiazol-5-yl) sulfamoyl)-2-cyanophenoxy)-3'-(trifluoromethyl)-[l,l'-biphenyl]-3-yl)pyridin-2-yl)niethyl) amino)ethyl)piperidin-l-yl)-3-oxopropoxy)ethyl)-l-methyl-5-oxo-2-(pyridin-3-yl) pyrrolidine-3-carboxamide (11-27). To a stirred solution of 3-(2-((25,35)-l-methyl-5-oxo-2- (pyridin-3-yl)pyrrolidine-3-carboxamido)ethoxy)propanoic acid (4) (0.05 g, 0.149 mmol) in /V,.V- dimethylformamide (DMF) (0.5 mL), HATU (0.062 g, 0.164 mmol) was added, and the mixture was stirred at 10 °C. The resulting solution was added slowly to a stirred solution of 3-cyano-4- ((3-(2-(((2-(piperidin-4-yl)ethyl)amino)methyl)pyridin-4-yl)-3'-(trifluoromethyl)-[l,r-biphenyl]-
4-yl)oxy)-A-(l,2,4-thiadiazol-5-yl)benzenesulfonamide (5) (0.111 g, 0.134 mmol) and A,A- di isopropyl ethyl amine (DIPEA) (0.156 ml, 0.895 mmol) in DMF (0.5 mL) at 0 °C. The reaction was stirred at 0 °C for 2 hours, quenched with cold water, and then extracted with DCM. The collected extracts were washed with brine and then evaporated under reduced pressure. The crude product was purified by reverse phase preparative chromatography to afford the title compound 11-27 (54 mg, 0.051 mmol, 34%) as an off-white solid. LC-MS m/z 1037.3 (M+H)+.
EXAMPLE 2 - Synthesis of (21S.3iS)- V-(2-(((l/?,4.S)-4-(4-(((l/’.4/?)-4-(4-(2-(((4-(4-(4-( V-(1.2,4- thiadiazol-5-yl)sulfamoyl)-2-cyanophenoxy)-3'-(trifluoromethyl)-[l,l'-biphenyl]-3-yl) pyridin-2-yl)methyl)amino)ethyl)piperidine-l-carbonyl)cyclohexyl)amino)-4-oxobutoxy) cyclohexyl)oxy)ethyI)-l-methyl-5-oxo-2-(pyridin-3-yl)pyrroIidine-3-carboxamide (II-2)
[0378] To a solution of ( l/?,4/')-4-(4-((( LS',4/?)-4-(2-((2,S',3,S')- l -methyl-5-oxo-2-(pyridin-3-yl) pyrrolidine-3-carboxamido)ethoxy)cyclohexyl)oxy)butanamido)cyclohexane-l-carboxylic acid (8) (27.6 mg, 0.048 mmol) and HATU (18.34 mg, 0.048 mmol) in dimethyl sulfoxide (DMSO) (2 ml), DIPEA (31.2 mg, 0.241 mmol) was added. The resulting mixture was stirred at room temperature for 10 minutes. 3-Cyano-4-((3-(2-(((2-(piperidin-4-yl)ethyl)amino)methyl)pyridin- 4-yl)-3'-(trifluoromethyl)-[l,T-biphenyl]-4-yl)oxy)-A-(l,2,4-thiadiazol-5-yl)benzenesulfonamide (7) (40 mg, 0.048 mmol) was then added, and the reaction mixture was stirred for 1.5 hours, after which the reaction was deemed complete based on LC-MS analysis. The reaction mixture was poured over a saturated NH4CI solution (15 mL), and the resulting solid precipitate was
collected, washed with water, and dried. The resulting pale-yellow solid was purified by reverse phase preparative chromatography to afford the title compound II-2 (30 mg, 0.023 mmol, 48%) as a fluffy, white solid. LC-MS m/z \T1^A (M+H)+.
EXAMPLE 3 - In vitro antagonism assay
[0379] The Qube is an automated whole-cell patch-clamp instrument used to record the inward sodium currents in a multi-hole mode.
[0380] Only wells with current amplitudes > 2000 pA at pulse 1 control stage were analyzed. The amplitude of the sodium current was calculated by measuring the difference between the peak inward current on stepping to -lOmV (z.e., peak of the current) and the leak current. The sodium current was assessed in vehicle control conditions and then at the end of each five (5) minute compound application. Individual well result was normalized to the vehicle control amplitude and the mean was calculated for each compound concentration. These values were then plotted, and estimated EC50 was calculated from the curve fit.
[0381] Materials and Methods: All compounds were tested in the presence of 0.1% Pol oxamer 188 Non-Ionic Surfactant and at approximately room temperature. Navi.7 Human Sodium Ion Channel Cell Based Two Pulse Protocol Assay Cells were held at -130 mV for 100 ms before stepping to -10 mV for 500 ms to completely inactivate the sodium channels (pulse 1), and stepped back to -120 mV for 10 ms (to completely recover from inactivation; however, channels that have inhibitors bound to them do not recover from inactivation) before stepping to -10 mV for 50 ms (pulse 2). The sweep interval was 20 s (0.05 Hz). Each concentration of compound was applied for 5 minutes. The assay was performed at approximately room temperature.
EXAMPLE 4 - Pharmacokinetic (PK) Analysis
[0382] Stock solution preparation: Stock solutions of compounds of Formula (I) were prepared at 20 mg/mL and 100 mg/mL in DMSO for pharmacokinetic/pharmacodynamic (PKZPD) and dose tolerability studies, respectively. Stock solutions were stored at -20 °C until further usage.
[0383] Formulations preparation: On the day of experiment, the stock solution of the compound of Formula (I) was removed from storage at -20 °C and thawed at room temperature. Anti-cotinine antibody, if required was removed from storage at -80 °C and thawed at room
temperature. Antibody vials were immediately transferred into wet ice after thawing. Compound s of Formula (I) were further diluted in DMSO as per experimental requirements. [0384] Formulation composition: The formulation composition was Saline: DMSO: PBS. Saline was added based on the quantity required followed by stock solution of the compound of Formula (I) prepared in DMSO followed by addition of antibody in PBS. Formulations were incubated at room temperature for 30 minutes before administration to the mouse. DMSO was used at 1 to 2 % (v/v) in the final formulation.
[0385] Administration to Animal: Solution formulation of antibody and compound of Formula (I) was injected (bolus injection) to the restrained mouse in the right/left lateral tail vein. Animals were dosed with 0.2 or 1.0 milligram per kilogram (mpk) of the compound of Formula (I) and 10 or 50 mpk of the anti-cotinine antibody.
[0386] Blood collection and plasma isolation (simple extraction method): 30-40 pl of blood was collected in a tube containing anticoagulant (4% EDTA, w/v) at specified time points via saphenous vein. The collected blood samples were centrifuged at 13,000 rpm for 5 minutes set at 4 °C. Plasma samples were stored at dry ice/-80 °C until bioanalysis.
[0387] Blood drug concentration analysis: Drug concentration in plasma samples was determined by an LC-MS/MS-based bioanalytical method. Samples were analyzed on Q-Trap, API-5500 LC-MS/MS system coupled with Exion UHPLC system from SCIEX, USA operated in multiple reaction monitoring mode employing electrospray ionization technique in positive polarity. Analyte and internal standard peaks were resolved on Synergi Polar, 75 x 2 0 mm, 4- um column using mobile phase 5 mM ammonium acetate in Milli-Q water as phase A and acetonitrile as Phase B. Gradient elution was performed with initial composition 85 % Phase A at 0 0 min, holding it for 0 5 minutes, ramping to 40 % by 0.8 minute, keeping the same for next 1.0 minutes, ramping to 25% by 1.9 minute and keeping the same level for 1.3 minutes before coming back to 85 % by 3.5 minutes. The total run time was 2 minutes. Area of desired peak was quantitated using standard curve.
EXAMPLE 5 - In vivo efficacy in a rat monosodium iodoacetate (MIA) induced acute joint pain model
[0388] Naive Sprague Dawley rats (source: Charles River UK; weight range: 180-200g) were acclimatized to the procedure room for at least 1 hour in their home cages, with food and water available ad libitum. Habituation to the apparatus and baseline weight bearing readings were
taken over several days (day -3 to -1), with baseline weight bearing readings were taken on the final day (day-1). Osteoarthritis (OA) was induced in rats via intra-articular injection of 2mg MIA (25 pl of 80 mg/mL) into the left knee on day 0. Weight bearing measurements were taken on day 3, animals were ranked and randomised to treatment groups (n=10 per group) according to their MIA window using a Latin square design. Naive WB difference - pre dose WB difference was defined as the MIA window. Compounds were co-dosed intravenously on day 7 at 1 mg/kg with 50 mg/kg of antibody. Animals were weighed at the start of the study, pre- and post-MIA to monitor any weight loss post-MIA. Animals were then weighed daily from day 7 to 10. MIA-induced hyperalgesia effect was evaluated as a comparison of weight load on hind paws (Static Weight Bearing). Specifically, weight bearing (g) readings were taken for both right and left hind paws and the difference calculated. Data are expressed as % ratio ipsilateral/contralateral ((WB left/WB right)* 100) (mean ± s.e.m ). Weight bearing readings were taken on Day 7 at 2 hours post administration, on Day 8 at 24 hours post administration, on Day 9 at 48 hours post administration and on Day 10 at 72 hours post administration. Data was analysed by comparing treatment groups to vehicle control at each time point. Statistical analysis was done by repeated measures ANOVA followed by Dunnett’s test using GraphPad Prism (p<0.05 considered significant).
[0389] A summary of data for compounds tested in the assays of Examples 3-5 is provided in Table 5.
Table 5
INCORPORATION BY REFERENCE
[0390] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.
EQUIVALENTS
[0391] The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.
Claims
Claims:
1. A compound represented by Formula (I):
or a pharmaceutically acceptable salt thereof, wherein:
R1 is Ci-4 alkyl or C3-6 cycloalkyl;
X1 is a bond or a C2-8 bivalent saturated straight or branched hydrocarbon chain wherein one, two, or three methylene units of the chain are independently replaced by -N(H)-, -N(CH3)-, -O-, piperidinylene, or C3-C6 cycloalkylene;
X2 is $-(Ci-5 alkylene)-N(H)- or a covalent bond, wherein is a bond to L;
Y1 is defined by Formula 1-1 that is substituted by one occurrence of R11, wherein Formula 1-1 is one of the following:
wherein:
R2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl) or Ci-4 alkyl;
R3 and R4 each represent independently for each occurrence hydrogen or C1-4 alkyl;
R? represents independently for each occurrence fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or Ci-4 alkyl;
R7, R8, R9, and R10 each represent independently for each occurrence hydrogen or C1-4 alkyl;
R11 is a bond to X1;
R12 is hydrogen, C1-3 haloalkyl, or C1-3 alkyl;
R13 is thiazolyl or 1,2,4-thiadiazolyl, each of which is substituted with 0 or 1 occurrence of Ci -4 alkyl;
R14 is -N(R10)2 or furanyl;
R1? is fluoro, chloro, phenyl, or hydrogen; x is 1 or 2;
L is a divalent linker selected from:
(i) a bivalent, saturated or unsaturated, straight or branched Ci-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(CI-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, -N(CI-6 alkyl)S(O)2-, -S(O)2N(H)-, -S(O)2N(Ci-6 alkyl)-, -N(H)C(O)-, -N(CI-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C 1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(CI-6 alkyl)-, -N(H)C(O)O- , -N(CI-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or optionally substituted 5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
wherein Ring A and Ring B are each independently C4-6 cycloalkylene; Lla is C3-5 linear alkylene, wherein 1 or 2 methylene units are replaced with -O- or -NRa-; each Ra is independently hydrogen or C1-3 alkyl; and
wherein Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; Llb is -CH2-NH-C(0)-, -NHC(O)-, or -C(O)NH-; L2b is C6-12 linear alkylene, wherein 1, 2, 3, or 4 methylene units are replaced with -O-, -NRlb-,
wherein n is 1, 2, 3, or 4, and
*** represents a covalent bond to Llb; and each Rlb is independently hydrogen or C1-3 alkyl;
(iv) O (L-c), wherein Llc is C2 -10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -NHC(O)-, or -C(O)NH-; Ring A is C4-6 cycloalkylene or C7-9 bridged bicyclic cycloalkylene; and L2c is -O- or a saturated C2-10 linear alkylene, wherein 1, 2, or 3 methylene units are replaced with -O-, -NH-, -;
wherein Lld is C 12-22 linear alkylene, wherein 1, 2, 3, 4, or 5 methylene units are replaced with -NH-, -O-, -C(O)NH-, -NHC(O)-, or -NHC(O)-NH-;
(vii) O (L-f), wherein Llf is a bond; C1-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-, -NH-, or -C(O)-; or -(C3-6 cycloalkylene)-NHC(O)-; L2f is a bond, -NHC(O)-, -C(O)NH-, or a C1-6 linear alkylene, wherein 0, 1, or 2 methylene units are replaced with -O-; and each of Z1 and Z2 is independently N or CH;
wherein Ring A is a 5 to 6 membered heteroarylene having 1 or 2 nitrogen ring atoms; L1§ is a bond, -CH2-, -NH-, or -O-; and
represents a covalent bond to L3h; L3h is a bond, -C(0)CH2-, -O-(Cs-6 cycloalkylene)-O-, or -C(O)NH(CH2)3OCH2-; L4h is a bond, -C(O)-, -CH2C(O)-, or
, , , , , , p bond to L31 and
represents a covalent bond to NH; L21 is a bond, C1-12 linear alkylene, or
, wherein n is 1, 2, 3, 4, or 5, and
represents a covalent bond to HN; and L31 is a bond or -C(O)-;
wherein Z1 is C, CH, or N; each of Z2, Z3,
Z4 and Z5 is independently CH or N, provided that no more than two of Z2, Z3, Z4 and Z~ are N; Llj is -NH-, -C(O)NH-, -NHC(O)-, or -O-; L2' is Ci-6 linear alkylene or 11
, , represents a covalent bond to L1'; and represents a single bond or a double bond;
(xii)
wherein Ring A is phenylene or a 5- or
6-membered heteroarylene having 1 or 2 nitrogen ring atoms; each of Z1 and Z2 is independently CH or N; Llk is a bond, -C(O)-, -C(O)NH-, or -NHC(O)-; and L2k is a C3-8
*** straight chain alkylene or
, wherein n is 1, 2, or 3, and
represents a covalent bond to Llk;
3-6 cycoa yene, or , w eren n s or , an represents a covalent bond to Llm;
1 or 2; 0, 1, or 2 hydrogen atoms of Y ' NA are replaced with F; L p is a bond,
-C(O)-, -C(O)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-; and d _ _ I** wherein each I represents a covalent bond to X1, and each ‘ represents a covalent bond to X2.
2. The compound of claim 1, wherein the compound is a compound of Formula I.
3. The compound of claim 1 or 2, wherein R1 is -CH3.
4. The compound of any one of claims 1-3, wherein X2 is <f)-(Ci-5 alkylene)-N(H)-.
5. The compound of any one of claims 1-3, wherein X2 is <J)-CH2CH2-N(H)-.
6. The compound of any one of claims 1-3, wherein X2 is $-C(H)(CH3)-N(H)-.
7. The compound of any one of claims 1-6, wherein Y1 is defined by the following formula that is substituted by one occurrence of R11:
8. The compound of any one of claims 1-6, wherein Y1 is
; wherein:
R2 is -(Ci-4 alkylene)-(C3-6 cycloalkyl);
R3 and R4 are independently hydrogen or C1-4 alkyl;
R5 is fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or C1-4 alkyl;
R7 is hydrogen or C1-4 alkyl;
R8 is C1-4 alkyl or hydrogen; and
R9 is hydrogen or C1-4 alkyl.
9. The compound of any one of claims 1-6, wherein Y1 is one of the following:
10. The compound of any one of claims 1-6, wherein Y1 is
; wherein:
R3 and R4 are independently hydrogen or C1-4 alkyl;
R? is fluoro, chloro, or cyano;
R6 is hydrogen, fluoro, chloro, or Ci-4 alkyl;
R7 is hydrogen or C1-4 alkyl;
R8 is hydrogen or Ci-4 alkyl;
R9 is Ci-4 alkyl; and
R10 is hydrogen or C1-4 alkyl.
11. The compound of any one of claims 1-8 or 10, wherein R3 is hydrogen.
12. The compound of any one of claims 1-8, 10, or 11, wherein R4 is hydrogen.
13. The compound of any one of claims 1-8 or 10-12, wherein R7 is hydrogen, and R8 is hydrogen.
14. The compound of any one of claims 1-6, wherein Y1 is one of the following:
15. The compound of claim 1, wherein the compound is represented by Formula la or a pharmaceutically acceptable salt thereof:
16. The compound of any one of claims 1-8 or 15, wherein R2 is -(C1-2 alkylene)-(C5-6 cycloalkyl).
17. The compound of any one of claims 1-8 or 15, wherein R2 is -(CH2)-cyclopentyl.
18. The compound of claim 1, wherein the compound is represented by Formula lb or a pharmaceutically acceptable salt thereof:
19. The compound of any one of claims 1-7, 10-13, or 18, wherein R10 is C1-4 alkyl.
20. The compound of any one of claims 1-7, 10-13, or 18, wherein R10 is methyl.
21. The compound of any one of claims 1-8, 10-13, or 18-20, wherein R9 is methyl.
22. The compound of any one of claims 1-8, 10-13, or 15-21, wherein R5 is cyano.
23. The compound of any one of claims 1-8, 10-13, or 15-21, wherein R5 is fluoro.
24. The compound of any one of claims 1-8, 10-13, or 15-23, wherein R6 is hydrogen.
25. The compound of any one of claims 1-8, 10-13, or 15-23, wherein R6 is chloro.
26. The compound of any one of claims 1-25, wherein X1 is -(C1-6 alkylene)-N(H)-'P, wherein T is a bond to L.
27. The compound of any one of claims 1-25, wherein X1 is -(CH2)s-N(H)-'P, wherein T is a bond to L.
28. The compound of any one of claims 1-25, wherein X1 is -(CH2)2-N(H)-'IJ, wherein T is a bond to L.
29. The compound of any one of claims 1-25, wherein X1 is -(C1-3 alkylene)-O-(CH2)-(Cs-6 cycloalkylene)-(CH2)-N(H)-T, wherein T is a bond to L.
30. The compound of any one of claims 1-25, wherein X1 is
wherein T is a bond to L.
31. The compound of any one of claims 1-6, wherein Y'-X1- is one of the following:
32. The compound of any one of claims 1-6, wherein Y1 is defined by one of the following formulae which are substituted by one occurrence of R11:
33. The compound of any one of claims 1-6, wherein Y1 is
34. The compound of any one of claims 1-6, wherein Y1 is
35. The compound of any one of claims 1-6, wherein Y1 is
36. The compound of any one of claims 1-6, wherein Y1 is
37. The compound of any one of claims 1-6, wherein Y1 is
38. The compound of any one of claims 1-6, wherein Y1 is
0. The compound of claim 1, wherein the compound is represented by Formula Ic or Id or a pharmaceutically acceptable salt thereof:
41. The compound of claim 1, wherein the compound is represented by Formula le or If or a pharmaceutically acceptable salt thereof:
If.
42. The compound of claim 1, wherein the compound is represented by Formula Ig or Ih or a pharmaceutically acceptable salt thereof:
43. The compound of any one of claims 32-38 or 40-42, wherein R3 is hydrogen.
44. The compound of any one of claims 32-38 or 40-43, wherein R4 is hydrogen.
45. The compound of any one of claims 32-38 or 40-44, wherein R12 is hydrogen.
46. The compound of any one of claims 32-38 or 40-44, wherein R12 is C1-3 haloalkyl.
47. The compound of any one of claims 32-38 or 40-44, wherein R12 is trifluoromethyl.
48. The compound of any one of claims 32-47, wherein X1 is -(C1-6 alkylene)-N(H)-'P, wherein T is a bond to L.
49. The compound of any one of claims 32-47, wherein X1 is -(CH2)-N(H)-'P, wherein T is a bond to L.
50. The compound of any one of claims 32-47, wherein X1 is -(C1-3 alkylene)-N(H)-(Ci-3 alkylene)-piperidinylene-T', wherein T is a bond to L.
51. The compound of any one of claims 32-47, wherein X1 is
wherein T is a bond to L.
52. The compound of any one of claims 1-6, wherein Y^X1- is one of the following:
53. The compound of any one of claims 1-6, wherein Y1 is defined by the following formula that is substituted by one occurrence of R11:
54. The compound of any one of claims 1-6, wherein
55. The compound of any one of claims 1-6, wherein
56. The compound of any one of claims 1-6, wherein
57. The compound of any one of claims 1-6, wherein Y1 is
58. The compound of any one of claims 1-6, wherein Y1 is one of the following:
59. The compound of claim 1, wherein the compound is represented by Formula Ti or a pharmaceutically acceptable salt thereof:
li.
60. The compound of any one of claims 53-55 or 59, wherein R15 is fluoro.
61. The compound of any one of claims 53-55 or 59, wherein R15 is phenyl.
62. The compound of claim 1, wherein the compound is represented by Formula Ij or Ik or a pharmaceutically acceptable salt thereof:
63. The compound of any one of claims 53, 56, 57, or 62, wherein R14 is -N(R10)2.
64. The compound of any one of claims 53, 56, 57, or 62, wherein R14 is furanyl.
65. The compound of any one of claims 53-57 or 59-64, wherein R13 is thiazolyl.
66. The compound of any one of claims 53-57 or 59-64, wherein R13 is 1 ,2,4-thiadiazolyl.
67. The compound of any one of claims 53-57 or 59-66, wherein R3 is hydrogen.
68. The compound of any one of claims 53-57 or 59-67, wherein R4 is hydrogen.
69. The compound of any one of claims 53-57 or 59-68, wherein R5 represents independently for each occurrence fluoro or chloro.
70. The compound of any one of claims 53-57 or 59-68, wherein R5 is cyano.
71. The compound of any one of claims 53-57 or 59-70, wherein x is 2.
72. The compound of any one of claims 53-57 or 59-70, wherein x is 1.
73. The compound of any one of claims 53-72, wherein X1 is -(Ci-4 alkyl ene)-N(H)-T, wherein T is a bond to L.
74. The compound of any one of claims 53-72, wherein X1 is -(CH2)2-N(H)-'P, wherein T is a bond to L.
75. The compound of any one of claims 53-72, wherein X1 is -(CH2)-N(H)-'P, wherein T is a bond to L.
76. The compound of any one of claims 53-72, wherein X1 is -(Ci-4alkylene)-piperazinylene- T, wherein T is a bond to L.
77. The compound of any one of claims 53-72, wherein X1 is
, wherein
T is a bond to L.
78. The compound of any one of claims 1-6, wherein Y^X1- is one of the following:
9. The compound of any one of claims 1-78, wherein L is a divalent linker of Formula (L-a- i):
wherein Ring
are as defined for Formula (L-a).
80. The compound of any one of claims 1-78, wherein L is a divalent linker of Formula (L-a- ii):
-ii), wherein
are as defined for Formula (L-a); p is 1 or 2; and m is 1 or 2.
81. The compound of any one of claims 1-78, wherein L is a divalent linker of Formula (L-a-
82. The compound of any one of claims 1-78, wherein L is selected from the group
; wherein the point of attachment indicated on the cycloalkyl-bound carbonyl group is the attachment point to X1.
83. The compound of any one of claims 1-78, wherein L is a divalent linker of Formula (L-b-
are as defined for Formula (L-b); p is 1 or 2; and m is 1 or 2.
84. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
indicated on the cycloalkyl -bound carbonyl group is the attachment point to X1.
85. The compound of any one of claims 1-78, wherein L is a divalent linker of Formula (L-
are as defined for
Formula (L-c); p is 1 or 2; and m is 1 or 2.
86. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
indicated on the carbonyl group is the attachment point to X1.
87. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
wherein the point of attachment indicated on the carbon-bound carbonyl group is the attachment point to X1.
88. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
point of attachment indicated on the carbonyl group is the attachment point to X1.
89. The compound of any one of claims 1-78, wherein L is a divalent linker of Formula (L-
are as defined for Formula (L-g); Z1, Z2, and Z3 are each independently selected from N or CH, provided that one or two of Z1, Z2, and Z3 is N.
90. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
indicated on the carbonyl group is the attachment point to X1.
91. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
indicated on the carbonyl group is the attachment point to X1.
92. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
indicated on the carbonyl group is the attachment point to X1.
93. The compound of any one of claims 1-78, wherein L is a selected from the group consisting of:
indicated on the carbonyl group is the attachment point to X1.
94. The compound of any one of claims 1-78, wherein L is selected from the group consisting of:
the carbonyl group is the attachment point to X1.
95. The compound of any one of claims 1-78, wherein L is
; , , y g p , ,
-C(O)NH-, -NHC(O)-, -S(O)2NH-, or -NHS(O)2-; and L2p is -(4-6 membered saturated heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur)-C(O))-.
96. The compound of any one of claims 1-78, wherein L is
the cycloalkyl-bound carbonyl group is the attachment point to X1.
97. The compound of any one of claims 1-78, wherein L is one of the following:
wherein the point of attachment indicated on the carbon-bound carbonyl group is the attachment point to X1.
98. The compound of any one of claims 1-78, wherein L is one of the following:
wherein the point of attachment indicated on the carbonyl group is the attachment point to X1.
99. The compound of any one of claims 1-78, wherein
taken together are one of the following:
00. The compound of any one of claims 1-78, wherein
taken together are one of the following:
101. A compound in Table 1, 2, or 3, or a pharmaceutically acceptable salt thereof.
102. A pharmaceutical composition, comprising a compound of any one of claims 1-101 and pharmaceutically acceptable carrier.
103. A method of treating or preventing a Nav1.7-associated disease or condition in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1-101 and an anti-cotinine antibody, or antigen-binding fragment thereof.
104. The method of claim 103, wherein the Nav1.7-associated disease or condition is pain, cough, acute itch, or chronic itch.
105. A method of treating or preventing pain in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of the compound of any one of claims 1-101 and an anti-cotinine antibody, or antigen-binding fragment thereof.
106. The method of claim 105, wherein the method is to treating pain.
107. The method of claim 105 or 106, wherein the pain is chronic pain.
108. The method of claim 105 or 106, wherein the pain is acute pain.
109. The method of any one of claims 105-108, wherein the pain is neuropathic pain.
110. The method of any one of claims 105-108, wherein the pain is inflammatory pain.
111. The method of any one of claims 105-108, wherein the pain is arthritis pain.
112. The method of any one of claims 105-108, wherein the pain is arthritis pain selected from osteoarthritis pain and rheumatoid arthritis pain.
113. The method of any one of claims 105-108, wherein the pain is due to cancer.
114. The method of any one of claims 105-108, wherein the pain is due to a cancer selected from the group consisting of a solid tumor, leukemia, and lymphoma.
115. The method of any one of claims 105-108, wherein the pain is complex regional pain syndrome.
116. The method of claim 115, wherein the complex regional pain syndrome is reflex sympathetic dystrophy pain.
117. The method of any one of claims 105-108, wherein the pain is trauma pain.
118. The method of any one of claims 105-108, wherein the pain is due to surgery.
119. The method of any one of claims 105-118, wherein the pain is located in the patient’s hand, wrist, arm, shoulder, back, leg, knee, ankle, foot, toe, neck, or head.
120. The method of any one of claims 105-108, wherein the pain is low back pain.
121. The method of claim 105 or 106, wherein the pain is chronic low back pain.
122. The method of any one of claims 105-108, wherein the pain is a neuropathic pain selected from the group consisting of low back pain, hip pain, leg pain, non-herpetic neuralgia, post-herpetic neuralgia, diabetic neuropathy pain, lumbosacral radiculopathy pain, nerve injury-induced pain, acquired immune deficiency syndrome (AIDS) related neuropathic pain, head trauma pain, phantom limb pain, multiple sclerosis pain, root avulsion pain, painful traumatic mononeuropathy, painful polyneuropathy, thalamic pain syndrome, post-stroke pain, central nervous system injury pain, post-surgical pain, carpal tunnel syndrome pain, trigeminal neuralgia pain, post mastectomy syndrome pain, postthoracotomy syndrome pain, stump pain, repetitive motion pain, neuropathic pain associated hyperalgesia and allodynia, drug-induced pain, toxin-caused nerve injury pain, chemotherapy-caused nerve injury pain, and combinations thereof.
123. The method of any one of claims 105-122, wherein the compound and the antibody, or antigen-binding fragment thereof, are administered simultaneously.
124. The method of any one of claims 105-122, wherein the compound and the antibody, or antigen-binding fragment thereof, are administered sequentially.
125. A method of increasing antibody-dependent cell cytotoxicity (ADCC) of voltage-gated sodium channel Nav1.7-expressing cells, comprising contacting the cells with an effective amount of the compound of any one of claims 1-101 and an anti-cotinine antibody, or antigen-binding fragment thereof.
126. A method of depleting voltage-gated sodium channel Nav l 7-expressing cells, comprising contacting the cells with an effective amount of the compound of any one of claims 1-101 and an anti-cotinine antibody, or antigen-binding fragment thereof.
127. The method of any one of claims 103-126, wherein the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a CDR1 having SEQ ID NO: 1, a CDR2 having SEQ ID NO: 2, and a CDR3 having SEQ ID NO: 3, and the light chain comprising a CDR1 having SEQ ID NO: 4, a CDR2 having SEQ ID NO: 5, and a CDR3 having SEQ ID NO: 6.
128. The method of any one of claims 103-126, wherein the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a heavy chain variable region (VH) having SEQ ID NO: 7, and the light chain comprising a light chain variable region (VL) having SEQ ID NO: 8.
129. The method of any one of claims 103-128, wherein the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase ADCC activity.
130. The method of claim 129, wherein the substitution in the Fc region is S239D/1332E, wherein residue numbering is according to the EU Index.
131. The method of any one of claims 103-126, wherein the anti-cotinine antibody has a heavy chain comprising SEQ ID NO: 9 and a light chain comprising SEQ ID NO: 10.
132. A combination comprising the compound of any one of claims 1-101 and an anti-cotinine antibody, or antigen-binding fragment thereof.
133. The combination of claim 132, wherein the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a CDR1 having SEQ ID NO: 1, a CDR2 having SEQ ID NO: 2, and a CDR3 having SEQ ID NO: 3, and the light chain comprising a CDR1 having SEQ ID NO: 4, a CDR2 having SEQ ID NO: 5, and a CDR3 having SEQ ID NO: 6.
134. The combination of claim 132, wherein the anti-cotinine antibody has a heavy chain and a light chain, the heavy chain comprising a heavy chain variable region (VH) having SEQ ID NO: 7, and the light chain comprising a light chain variable region (VL) having SEQ ID NO: 8.
135. The combination of any one of claims 132-134, wherein the anti-cotinine antibody is of IgGl isotype comprising a substitution in an Fc region to increase ADCC activity.
136. The combination of claim 135, wherein the substitution in the Fc region is S239D/I332E, wherein residue numbering is according to the EU Index.
137. The combination of claim 132, wherein the anti-cotinine antibody has a heavy chain comprising SEQ ID NO: 9 and a light chain comprising SEQ ID NO: 10.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202463571523P | 2024-03-29 | 2024-03-29 | |
| US63/571,523 | 2024-03-29 | ||
| US202463711389P | 2024-10-24 | 2024-10-24 | |
| US63/711,389 | 2024-10-24 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025208006A1 true WO2025208006A1 (en) | 2025-10-02 |
Family
ID=97219470
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2025/021976 Pending WO2025208006A1 (en) | 2024-03-29 | 2025-03-28 | Heterobifunctional molecules for binding na v1.7 and methods of treating medical conditions using same |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025208006A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2015181797A1 (en) * | 2014-05-30 | 2015-12-03 | Pfizer Inc. | Benzenesulfonamides useful as sodium channel inhibitors |
| WO2023017484A1 (en) * | 2021-08-13 | 2023-02-16 | Glaxosmithkline Intellectual Property Development Limited | Cytotoxicity targeting chimeras |
| WO2023028077A1 (en) * | 2021-08-24 | 2023-03-02 | Genentech, Inc. | Sodium channel inhibitors and methods of designing same |
-
2025
- 2025-03-28 WO PCT/US2025/021976 patent/WO2025208006A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2015181797A1 (en) * | 2014-05-30 | 2015-12-03 | Pfizer Inc. | Benzenesulfonamides useful as sodium channel inhibitors |
| WO2023017484A1 (en) * | 2021-08-13 | 2023-02-16 | Glaxosmithkline Intellectual Property Development Limited | Cytotoxicity targeting chimeras |
| WO2023028077A1 (en) * | 2021-08-24 | 2023-03-02 | Genentech, Inc. | Sodium channel inhibitors and methods of designing same |
Non-Patent Citations (1)
| Title |
|---|
| STORER, R. I. ET AL.: "Highly potent and selective Nav1.7 inhibitors for use as intravenous agents and chemical probes", BIOORG. MED. CHEM. LETT., vol. 27, 2017, pages 4805 - 4811, XP085238129, DOI: 10.1016/j.bmcl.2017.09.056 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| AU2017274442B2 (en) | Glucocorticoid receptor agonist and immunoconjugates thereof | |
| US11234977B2 (en) | Fused tricyclic pyrazolo-dihydropyrazinyl-pyridone compounds as antivirals | |
| CN115279766B (en) | Combination therapy containing A2A/A2B and PD-1/PD-L1 inhibitors | |
| WO2019108795A1 (en) | Treatment of indolent or aggressive b-cell lymphomas using a combination comprising btk inhibitors | |
| US20250136722A1 (en) | Cytotoxicity targeting chimeras | |
| US20240325392A1 (en) | Cytotoxicity targeting chimeras for ccr2-expressing cells | |
| WO2019050780A1 (en) | Compounds for reducing the viscosity of biological formulations | |
| TW202327620A (en) | An anti-inflammatory compound and use thereof | |
| KR20250039375A (en) | Antibody-drug conjugate and method for producing and using the same | |
| JP2025513163A (en) | Antibody-drug conjugates, pharmaceutical compositions thereof and uses thereof | |
| US20250057827A1 (en) | Cytotoxicity targeting chimeras for prostate specific membrane antigen-expressing cells | |
| US20250154288A1 (en) | Cytotoxicity targeting chimeras for c-c chemokine receptor 2-expressing cells | |
| CN118678972A (en) | Pharmaceutical conjugates of glucocorticoids | |
| US20250049932A1 (en) | Cytotoxicity targeting chimeras for folate receptor-expressing cells | |
| WO2025208006A1 (en) | Heterobifunctional molecules for binding na v1.7 and methods of treating medical conditions using same | |
| US20250064947A1 (en) | Cytotoxicity targeting chimeras for fibroblast activation protein-expressing cells | |
| US20250057958A1 (en) | Cytotoxicity targeting chimeras for ccr2-expressing cells | |
| US20250051479A1 (en) | Cytotoxicity targeting chimeras for cxcr3-expressing cells | |
| WO2023161877A1 (en) | Cytotoxicity targeting chimeras for integrin avb6-expressing cells | |
| WO2025208012A1 (en) | HETEROBIFUNCTIONAL MOLECULES FOR BINDING NA v1.8 AND METHODS OF TREATING MEDICAL CONDITIONS USING SAME | |
| TW202527994A (en) | An antiviral conjugate | |
| TW202430226A (en) | Anti-cd40 antibody drug conjugates, preparation methods and pharmaceutical uses thereof | |
| TW202539743A (en) | Antibody-drug conjugate (adc) comprising a linker system | |
| WO2024255740A1 (en) | Camptothecin derivative for treating or preventing cancer, and antibody-drug conjugate thereof | |
| CN119954820A (en) | A compound and its application |
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: 25778469 Country of ref document: EP Kind code of ref document: A1 |