US20170202821A1 - Method of treating c3 glomerulopathy - Google Patents

Method of treating c3 glomerulopathy Download PDF

Info

Publication number
US20170202821A1
US20170202821A1 US15/404,610 US201715404610A US2017202821A1 US 20170202821 A1 US20170202821 A1 US 20170202821A1 US 201715404610 A US201715404610 A US 201715404610A US 2017202821 A1 US2017202821 A1 US 2017202821A1
Authority
US
United States
Prior art keywords
human
independently selected
group
compound
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/404,610
Other languages
English (en)
Inventor
Petrus BEKKER
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chemocentryx Inc
Original Assignee
Chemocentryx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chemocentryx Inc filed Critical Chemocentryx Inc
Priority to US15/404,610 priority Critical patent/US20170202821A1/en
Publication of US20170202821A1 publication Critical patent/US20170202821A1/en
Assigned to CHEMOCENTRYX, INC. reassignment CHEMOCENTRYX, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEKKER, Petrus
Priority to US16/184,535 priority patent/US11285138B2/en
Priority to US17/670,604 priority patent/US11779576B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/451Non condensed piperidines, e.g. piperocaine having a carbocyclic group directly attached to the heterocyclic ring, e.g. glutethimide, meperidine, loperamide, phencyclidine, piminodine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys

Definitions

  • C3 glomerulopathy is a rare disease of the kidney (the prevalence of C3G is estimated at 2-3 per 1,000,000 people).
  • C3G is characterized by deposition of the protein known as C3 (a component of the body's complement system) in the filtration units (the glomeruli) of the kidney, indicating complement involvement in causing kidney damage.
  • C3 glomerulopathy is characterized by evidence of alternative complement activation based on C3 deposition in the glomeruli.
  • DDD dense deposit disease
  • MPGN membranoproliferative glomerulonephritis
  • C3GN C3 glomerulonephritis
  • C3 glomerulopathy Genetic lesions leading to defective complement regulation, including mutations in complement factor H (CFH) have been described in these patients.
  • C3 glomerulopathy often have high proteinuria and progressive deterioration in renal function.
  • C3GN There is no approved treatment for patients with C3 glomerulopathy, including C3GN.
  • C3G Invariably leads to kidney failure, and kidney transplant is frequently the only option. Even after transplantation, the new kidney will frequently fail due to recurrence of the disease.
  • the present disclosure is directed to a method of treating a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a C5aR antagonist.
  • the C5aR antagonist is a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the C5aR antagonist is a compound having the formula:
  • FIG. 1 represents the patient's Estimated glomerular filtration rate (eGFR) before and after treatment with compound 1.
  • FIG. 2 represents the histopathological improvement following treatment with compound 1.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain hydrocarbon radical, having the number of carbon atoms designated (i.e. C 1-8 means one to eight carbons).
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • alkenyl refers to an unsaturated alkyl group having one or more double bonds.
  • alkynyl refers to an unsaturated alkyl group having one or more triple bonds.
  • unsaturated alkyl groups include vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • cycloalkyl refers to hydrocarbon rings having the indicated number of ring atoms (e.g., C 3-6 cycloalkyl) and being fully saturated or having no more than one double bond between ring vertices.
  • Cycloalkyl is also meant to refer to bicyclic and polycyclic hydrocarbon rings such as, for example, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, etc.
  • heterocycloalkyl refers to a cycloalkyl group that contain from one to five heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • the heterocycloalkyl may be a monocyclic, a bicyclic or a polycyclic ring system.
  • heterocycloalkyl groups include pyrrolidine, imidazolidine, pyrazolidine, butyrolactam, valerolactam, imidazolidinone, hydantoin, dioxolane, phthalimide, piperidine, 1,4-dioxane, morpholine, thiomorpholine, thiomorpholine-S-oxide, thiomorpholine-S,S-oxide, piperazine, pyran, pyridone, 3-pyrroline, thiopyran, pyrone, tetrahydrofuran, tetrahydrothiophene, quinuclidine, and the like.
  • a heterocycloalkyl group can be attached to the remainder of the molecule through a ring carbon or a heteroatom.
  • alkylene by itself or as part of another substituent means a divalent radical derived from an alkane, as exemplified by —CH 2 CH 2 CH 2 CH 2 —.
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present disclosure.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having four or fewer carbon atoms.
  • alkenylene” and alkynylene refer to the unsaturated forms of “alkylene” having double or triple bonds, respectively.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group.
  • the heteroatom Si may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule.
  • Examples include —CH 2 —CH 2 —O—CH 3 , —CH 2 —CH 2 —NH—CH 3 , —CH 2 —CH 2 —N(CH 3 )—CH 3 , —CH 2 —S—CH 2 —CH 3 , —CH 2 —CH 2 , —S(O)—CH 3 , —CH 2 —CH 2 —S(O) 2 —CH 3 , —CH ⁇ CH—O—CH 3 , —Si(CH 3 ) 3 , —CH 2 —CH ⁇ N—OCH 3 , and —CH ⁇ CH—N(CH 3 )—CH 3 .
  • heteroalkenyl and “heteroalkynyl” by itself or in combination with another term, means, unless otherwise stated, an alkenyl group or alkynyl group, respectively, that contains the stated number of carbons and having from one to three heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group.
  • heteroalkylene by itself or as part of another substituent means a divalent radical, saturated or unsaturated or polyunsaturated, derived from heteroalkyl, as exemplified by —CH 2 —CH 2 —S—CH 2 CH 2 — and —CH 2 —S—CH 2 —CH 2 —NH—CH 2 —, —O—CH 2 —CH ⁇ CH—, —CH 2 —CH ⁇ C(H)CH 2 —O—CH 2 — and —S—CH 2 —C ⁇ C—.
  • heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like).
  • alkoxy alkylamino and “alkylthio” (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively. Additionally, for dialkylamino groups, the alkyl portions can be the same or different and can also be combined to form a 3-7 membered ring with the nitrogen atom to which each is attached. Accordingly, a group represented as —NR a R b is meant to include piperidinyl, pyrrolidinyl, morpholinyl, azetidinyl and the like.
  • halo or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl.
  • C 1-4 haloalkyl is mean to include trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • aryl means, unless otherwise stated, a polyunsaturated, typically aromatic, hydrocarbon group which can be a single ring or multiple rings (up to three rings) which are fused together or linked covalently.
  • heteroaryl refers to aryl groups (or rings) that contain from one to five heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • Non-limiting examples of aryl groups include phenyl, naphthyl and biphenyl, while non-limiting examples of heteroaryl groups include pyridyl, pyridazinyl, pyrazinyl, pyrimindinyl, triazinyl, quinolinyl, quinoxalinyl, quinazolinyl, cinnolinyl, phthalaziniyl, benzotriazinyl, purinyl, benzimidazolyl, benzopyrazolyl, benzotriazolyl, benzisoxazolyl, isobenzofuryl, isoindolyl, indolizinyl, benzotriazinyl, thienopyridinyl, thienopyrimidinyl, pyrazolopyrimidinyl, imidazopyridines, benzothiaxolyl, benzofuranyl, benzothienyl, indolyl, quinoly
  • aryl when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
  • arylalkyl is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl and the like).
  • alkyl in some embodiments, will include both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below.
  • aryl and heteroaryl will refer to substituted or unsubstituted versions as provided below, while the term “alkyl” and related aliphatic radicals is meant to refer to unsubstituted version, unless indicated to be substituted.
  • Substituents for the alkyl radicals can be a variety of groups selected from: -halogen, —OR′, —NR′R′′, —SR′, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —CONR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′—C(O)NR′′R′′′, —NR′′C(O) 2 R′, —NH—C(NH 2 ) ⁇ NH, —NR′C(NH 2 ) ⁇ NH, —NH—C(NH 2 ) ⁇ NR′.
  • R′, R′′ and R′′′ each independently refer to hydrogen, unsubstituted C 1-8 alkyl, unsubstituted heteroalkyl, unsubstituted aryl, aryl substituted with 1-3 halogens, unsubstituted C 1-8 alkyl, C 1-8 alkoxy or C 1-8 thioalkoxy groups, or unsubstituted aryl-C 1-4 alkyl groups.
  • R′ and R′′ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 3-, 4-5-, 6-, or 7-membered ring.
  • —NR′R′′ is meant to include 1-pyrrolidinyl and 4-morpholinyl.
  • substituents for the aryl and heteroaryl groups are varied and are generally selected from: -halogen, —OR′, —OC(O)R′, —NR′R′′, —SR′, —R′, —CN, —NO 2 , —CO 2 R′, —CONR′R′′, —C(O)R′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′′C(O) 2 R′, —NR′—C(O)NR′′R′′′, —NH—C(NH 2 ) ⁇ NH, —NR′C(NH 2 ) ⁇ NH, —NH—C(NH 2 ) ⁇ NR′, —S(O)R′, —S(O) 2 R′, —S(O) 2 NR′R′′, —NR′S(O) 2 R′′, —N 3 , perfluoro(C 1 -C 4 )alkoxy, and perfluoro
  • Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -T-C(O)—(CH 2 ) q —U—, wherein T and U are independently —NH—, —O—, —CH 2 — or a single bond, and q is an integer of from 0 to 2.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ) r —B—, wherein A and B are independently —CH 2 —, —O—, —NH—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 NR′— or a single bond, and r is an integer of from 1 to 3.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula —(CH 2 ) s —X—(CH 2 ) t —, where s and t are independently integers of from 0 to 3, and X is —O—, —NR′—, —S—, —S(O)—, —S(O) 2 —, or —S(O) 2 NR′—.
  • the substituent R′ in —NR′— and —S(O) 2 NR′— is selected from hydrogen or unsubstituted C 1-6 alkyl.
  • heteroatom is meant to include oxygen (O), nitrogen (N), sulfur (S) and silicon (Si).
  • ionic liquid refers to any liquid that contains mostly ions.
  • “ionic liquid” refers to the salts whose melting point is relatively low (e.g., below 250° C.).
  • Examples of ionic liquids include but are not limited to 1-butyl-3-methylimidazolium tetrafluoroborate, 1-hexyl-3-methylimidazolium tetrafluoroborate, 1-octyl-3-methylimidazolium tetrafluoroborate, 1-nonyl-3-methylimidazolium tetrafluoroborate, 1-decyl-3-methylimidazolium tetrafluoroborate, 1-hexyl-3-methylimidazolium hexafluorophosphate and 1-hexyl-3-methylimidazolium bromide, and the like.
  • salts are meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • salts derived from pharmaceutically-acceptable inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like.
  • Salts derived from pharmaceutically-acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperadine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
  • arginine betaine
  • caffeine choline
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge, S. M., et al, “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • the neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present disclosure.
  • the present disclosure is directed to a method of treating a human suffering from or susceptible to complement 3 glomerulopathy comprising administering to the human an effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the compound has the formula (Ib):
  • X 1 is selected from the group consisting of CH and CR 1 ; the subscript n is an integer of from 0 to 2; X 2 is selected from the group consisting of CH and CR 2 ; and the subscript m is an integer of from 0 to 2.
  • X 1 is selected from the group consisting of CH and CR 1 ; the subscript n is an integer of from 0 to 2; X 2 is selected from the group consisting of CH and CR 2 ; and the subscript m is an integer of from 0 to 2.
  • the compound is selected from the group consisting of
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • a method of reducing glomerular inflammation in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the compound has the formula (Ie):
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • a method of reducing C3 deposits and/or C5b-9 deposits in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the compound has the formula (Ie):
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the human suffers from complement 3 glomerulonephritis. In some embodiments, the human suffers from progressive complement 3 glomerulonephritis. In some embodiments, the human suffers from recurrent complement 3 glomerulonephritis after a renal transplant. In some embodiments, the human suffers from dense deposit disease.
  • a method of clearing glomerular endocapillary proliferation in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • a method of reducing glomerular inflammatory macrophages in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the compound has the formula (Ie):
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • a method of reducing proteinuria in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof,
  • the compound has the formula (Ie):
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the human suffers from complement 3 glomerulonephritis. In some embodiments, the human suffers from progressive complement 3 glomerulonephritis. In some embodiments, the human suffers from recurrent complement 3 glomerulonephritis after a renal transplant. In some embodiments, the human suffers from dense deposit disease. In some embodiments, the human had refractory disease to immunosuppressive drugs.
  • the compound is selected from the group consisting of
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the method comprises one or more of: slowing the rate of decline in Estimated Glomerular Filtration Rate (eGFR) in the human, reducing glomerular inflammation in the human, clearing glomerular endocapillary proliferation in the human, reducing glomerular inflammatory macrophages in the human, reducing proteinuria in the human, slowing down the progression of renal disease in the human, stopping the progression of renal disease in the human, delaying end stage renal disease in the human, improving renal histology in the human, decreasing proteinuria in the human, slowing the increase in proteinuria in the human.
  • the improvements may be supported by kidney biopsy.
  • a method of slowing the rate of decline in Estimated Glomerular Filtration Rate (eGFR) in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of slowing the rate of decline in Estimated Glomerular Filtration Rate (eGFR) in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing glomerular inflammation in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing glomerular inflammation in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing C3 deposits and/or C5b-9 deposits in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of clearing glomerular endocapillary proliferation in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of clearing glomerular endocapillary proliferation in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing glomerular inflammatory macrophages in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing glomerular inflammatory macrophages in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing proteinuria in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • a method of reducing proteinuria in a human suffering from or susceptible to C3 glomerulopathy comprising administering to the human an effective amount of a compound having the following formula:
  • the human suffers from complement 3 glomerulonephritis. In some embodiments, the human suffers from progressive complement 3 glomerulonephritis. In some embodiments, the human suffers from recurrent complement 3 glomerulonephritis after a renal transplant. In some embodiments, the human suffers from dense deposit disease.
  • the complement 3 glomerulopathy is refractory to treatment. In some embodiments, the complement 3 glomerulonephritis is refractory to other treatment. In some embodiments, the human has refractory disease to immunosuppressive drugs. In some embodiments, the human has refractory disease to one or more of rituximab, cyclophosphamide, mycophenolate mofetil, tacrolimus, and steroids. In some embodiments, the human has refractory disease to one or more of rituximab, cyclophosphamide, mycophenolate mofetil, tacrolimus, and glucocorticosteroids. In some embodiments, the human shows improved health-related quality of life changes.
  • the health-related quality-of-life is based on Short Form-36 version 2 (SF-36 v2) or EuroQOL-5D-5L (EQ-5D-5L) assessment. In some embodiments, the health-related quality-of-life is based on Short Form-36 version 2 (SF-36 v2) assessment. In some embodiments, the health-related quality-of-life is based on EuroQOL-5D-5L (EQ-5D-5L) assessment.
  • the compound is administered twice daily. In some embodiments, the compound is administered once a day. In some embodiments, the compound is administered every other day. In some embodiments, the compound is administered every 3 days. In some embodiments, the compound is administered 3 times per day. In some embodiments, the compound is administered 4 times per day.
  • the human receives 30 mg of the compound daily. In some embodiments, the human receives 20 mg of the compound daily. In some embodiments, the human receives 10 mg of the compound daily. In some embodiments, the human receives 40 mg of the compound daily. In some embodiments, the human receives 60 mg of the compound daily.
  • the human receives 50 mg, 70 mg, 80 mg, 90 mg, 100 mg, 150 mg or 200 mg of the compound daily.
  • the human receives 30 mg of the compound twice daily. In some embodiments, the human receives 20 mg of the compound twice daily. In some embodiments, the human receives 10 mg of the compound twice daily.
  • the compound is administered orally.
  • the human has a Complement factor H related protein 5 (CFHR5) mutation.
  • CFHR5 Complement factor H related protein 5
  • the human receives treatment for 12 weeks. In some embodiments, the human receives treatment for 26 weeks. In some embodiments, the human receives treatment for 52 weeks. In some embodiments, the human receives chronic treatment. In some embodiments, the human receives continuous treatment.
  • the method further comprises administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
  • the one or more additional therapeutic agents is administered sequentially or concurrently in the same composition or not.
  • the one or more additional therapeutic agents is selected from immunosuppressive drugs, angiotensin-converting enzyme (ACE) inhibitors, angiotensin II type-1 receptor blockers (ARBs) and corticosteroids.
  • ACE angiotensin-converting enzyme
  • ARBs angiotensin II type-1 receptor blockers
  • the one or more additional therapeutic agents is selected from the group consisting of cyclophosphamide, mycophenolate mofetil, rituximab, eculizumab, tacrolimus, belimumab, OMS721, ACH-4471, AMY-101, Acthar Gel, SAND-5, corticotropin, CDX-1135, ramipril, perindopril, lisinopril, perindopril arginine, captopril, spirapril, quinapril, enalapril, imidapril, fosinopril, zofenopril, benazepril, trandolapril, verapamil, benazepril, amlodipine, trandolapril, P-003, cilazapril, delapril, moexipril, quinapril, fosinopril, temocapril
  • Protein kinase C stimulators Protein kinase C alpha inhibitors, CD74 antagonists, Immunoglobulin gamma Fc receptor IIB antagonists, T-cell antigen CD7 inhibitors, CD95 antagonists, N acetylmannosamine kinase stimulators, Cardiotrophin-1 ligands, Leukocyte elastase inhibitors, CD40 ligand receptor antagonists, CD40 ligand modulators, IL-17 antagonists, TLR-2 antagonists, complement factor D inhibitors, complement factor B inhibitors, complement C5 inhibitors, MASP-2 inhibitors, MASP-3 inhibitors, C3 inhibitors, pegylated APL-1, C1s inhibitors, C6 inhibitors, and T cell receptor antagonists.
  • the one or more additional therapeutic agents is selected from the group consisting of obinutuzumab, rituximab, ocrelizumab, cyclophosphamide, prednisone, hydrocortisone, hydrocortisone acetate, cortisone acetate, tixocortol pivalate, prednisolone, methylprednisolone, triamcinolone acetonide, triamcinolone alcohol, mometasone, amcinonide, budesonide, desonide, fluocinonide, fluocinolone acetonide, halcinonide, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium phosphate, fluocortolone, hydrocortisone-17-valerate, halometasone, alclometasone dipropionate, beclomethasone, betamethasone valerate,
  • Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure.
  • Certain compounds of the present disclosure possess asymmetric carbon atoms (optical centers) or double bonds, the racemates, diastereomers, geometric isomers, regioisomers and individual isomers (e.g., separate enantiomers) are all intended to be encompassed within the scope of the present disclosure.
  • the compounds of the present disclosure may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are intended to be encompassed within the scope of the present disclosure.
  • the compounds disclosed herein are also meant to encompass all pharmaceutically acceptable compounds of Formulas (I), (Ia), (Ib), (Ic), (Id), (Ie) and compound 1 being isotopically-labeled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively.
  • radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action.
  • Certain isotopically-labeled compounds of Formulas (I), (Ia), (Ib), (Ic), (Id), (Ie) and compound 1 for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability. For example, in vivo half-life may increase or dosage requirements may be reduced. Thus, heavier isotopes may be preferred in some circumstances.
  • Isotopically-labeled compounds of Formulas (I), (Ia), (Ib), (Ic), (Id), (Ie) and compound 1 can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • the methods, compositions, kits and articles of manufacture provided herein use or include compounds (e.g., (I), (Ia), (Ib), (Ic), (Id), (Ie) and compound 1) or pharmaceutically acceptable salts, prodrugs, or solvates thereof, in which from 1 to n hydrogen atoms attached to a carbon atom may be replaced by a deuterium atom or D, in which n is the number of hydrogen atoms in the molecule.
  • the deuterium atom is a non-radioactive isotope of the hydrogen atom.
  • Such compounds may increase resistance to metabolism, and thus may be useful for increasing the half-life of compounds or pharmaceutically acceptable salts, prodrugs, or solvates thereof, when administered to a mammal. See, e.g., Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism”, Trends Pharmacol. Sci., 5(12):524-527 (1984).
  • Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogen atoms have been replaced by deuterium.
  • Treatment methods provided herein include, in general, administration to a patient an effective amount of the compounds provided herein.
  • Suitable patients include those patients suffering from or susceptible to (i.e., prophylactic treatment) C3 glomerulonephritis.
  • treatment methods provided herein comprise administering to a patient an effective amount of a compound provided herein.
  • the compound(s) of the disclosure are preferably administered to a patient (e.g., a human) orally or topically.
  • the compound(s) of the disclosure are administered to a patient (e.g., a human) systemically (intravenously or subcutaneously).
  • the effective amount may be an amount sufficient to modulate C5a receptor activity and/or an amount sufficient to reduce or alleviate the symptoms presented by the patient.
  • the amount administered is sufficient to yield a plasma concentration of the compound (or its active metabolite, if the compound is a pro-drug) high enough to detectably inhibit white blood cell (e.g., neutrophil) chemotaxis in vitro.
  • Treatment regimens may vary depending on the compound used and the particular condition to be treated; for treatment of most disorders, a frequency of administration of 4 times daily or less is preferred. In general, a dosage regimen of 2 times daily is more preferred, with once a day dosing particularly preferred.
  • the specific dose level and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination (i.e., other drugs being administered to the patient) and the severity of the particular disease undergoing therapy, as well as the judgment of the prescribing medical practitioner. In general, the use of the minimum dose sufficient to provide effective therapy is preferred. Patients may generally be monitored for therapeutic effectiveness using medical or veterinary criteria suitable for the condition being treated or prevented.
  • Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment or preventions of conditions involving pathogenic C5a activity (about 0.5 mg to about 7 g per human patient per day).
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient.
  • the compound be administered orally, transdermally, intravaneously, or subcutaneously, it is preferred that sufficient amount of the compound be administered to achieve a serum concentration of 5 ng (nanograms)/mL-10 ⁇ g (micrograms)/mL serum, more preferably sufficient compound to achieve a serum concentration of 20 ng-1 ⁇ g/ml serum should be administered, most preferably sufficient compound to achieve a serum concentration of 50 ng/ml-200 ng/ml serum should be administered.
  • a serum concentration of approximately 1 micromolar for direct injection into the synovium (for the treatment of arthritis) sufficient compounds should be administered to achieve a local concentration of approximately 1 micromolar.
  • Frequency of dosage may also vary depending on the compound used and the particular disease treated. However, for treatment of most disorders, a dosage regimen of 4 times daily, three times daily, or less is preferred, with a dosage regimen of once daily or 2 times daily being particularly preferred. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination (i.e., other drugs being administered to the patient), the severity of the particular disease undergoing therapy, and other factors, including the judgment of the prescribing medical practitioner.
  • compositions which will typically contain a pharmaceutical carrier or diluent.
  • composition as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • the pharmaceutical composition further comprises one or more additional therapeutic agents.
  • compositions for the administration of the compounds of this disclosure may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy and drug delivery. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions and self-emulsifications as described in U.S. Patent Application 2002-0012680, hard or soft capsules, syrups, elixirs, solutions, buccal patch, oral gel, chewing gum, chewable tablets, effervescent powder and effervescent tablets.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents, antioxidants and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as cellulose, silicon dioxide, aluminum oxide, calcium carbonate, sodium carbonate, glucose, mannitol, sorbitol, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example PVP, cellulose, PEG, starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated, enterically or otherwise, by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the techniques described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and U.S. Pat. No. 4,265,874 to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, polyethylene glycol (PEG) of various average sizes (e.g., PEG400, PEG4000) and certain surfactants such as cremophor or solutol, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • emulsions can be prepared with a non-water miscible ingredient such as oils and stabilized with surfactants such as mono- or di-glycerides, PEG esters and the like.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy-propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxy-ethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan mono
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerin, glycerin, glycerin, glycerin, glycerin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol
  • the pharmaceutical compositions of the disclosure may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • Oral solutions can be prepared in combination with, for example, cyclodextrin, PEG and surfactants.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the compounds of the present disclosure may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials include cocoa butter and polyethylene glycols.
  • the compounds can be administered via ocular delivery by means of solutions or ointments.
  • transdermal delivery of the subject compounds can be accomplished by means of iontophoretic patches and the like.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present disclosure are employed.
  • topical application is also meant to include the use of mouth washes and gargles.
  • the compounds of this disclosure may also be coupled a carrier that is a suitable polymers as targetable drug carriers.
  • suitable polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxy-propyl-methacrylamide-phenol, polyhydroxyethyl-aspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • the compounds of the disclosure may be coupled to a carrier that is a class of biodegradable polymers useful in achieving controlled release of a drug, for example polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross linked or amphipathic block copolymers of hydrogels.
  • Polymers and semipermeable polymer matrices may be formed into shaped articles, such as valves, stents, tubing, prostheses and the like.
  • the compound of the disclosure is coupled to a polymer or semipermeable polymer matrix that is formed as a stent or stent-graft device.
  • kits comprising a compound of Formula (I), (Ia), (Ib), (Ic), (Id) or (le), or compound 1, or a pharmaceutically acceptable salt thereof, and instructions for its administration are provided.
  • kits comprising a compound of Formula (I), (Ia), (Ib), (Ic), (Id) or (Ie), or compound 1, or a pharmaceutically acceptable salt thereof, in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents and instructions for their administration are provided.
  • the compounds of this disclosure are formulated into administration units which are packaged in a single packaging.
  • the single packaging encompasses but is not limited to a bottle, a child-resistant bottle, an ampoule, and a tube.
  • the compounds of this disclosure and optionally additional therapeutic agents are formulated into administration units and every single administration unit is individually packaged in a single packaging.
  • Such individually packaged units may contain the pharmaceutical composition in any form including but not limited to liquid form, solid form, powder form, granulate form, an effervescent powder or tablet, hard or soft capsules, emulsions, suspensions, syrup, suppositories, tablet, troches, lozenges, solution, buccal patch, thin film, oral gel, chewable tablet, chewing gum, and single-use syringes.
  • Such individually packaged units may be combined in a package made of one or more of paper, cardboard, paperboard, metal foil and plastic foil, for example a blister pack.
  • One or more administration units may be administered once or several times a day.
  • One or more administration units may be administered three times a day.
  • One or more administration units may be administered twice a day.
  • One or more administration units may be administered on a first day and one or more administration units may be administered on the following days.
  • Compound 1 has the formula:
  • Example 1 Study of Compound 1 in a Patient with Progressive Complement 3 Glomerulonephritis
  • a C3 glomerulonephritis patient received treatment with the orally administered complement inhibitor compound 1, following the protocol detailed below.
  • the patient had refractory disease despite a kidney transplant and prior treatment with the broadly immunosuppressive drugs rituximab, cyclophosphamide, mycophenolate mofetil, tacrolimus, and steroids. Renal allograft biopsies were taken pre-dose, 2 and 7 months during therapy.
  • the patient's condition improved in response to compound 1 treatment.
  • the improvement seen with compound 1 treatment in this patient was based on the on-treatment kidney biopsy histologic findings that showed clearance ofglomerular endocapillary proliferation and a marked decrease in glomerular inflammatory macrophages compared to the pre-treatment biopsy. Proteinuria dropped approximately 80% with compound 1 treatment.
  • eGFR estimated glomerular filtration rate
  • FIG. 1 shows the eGFR prior and after treatment with compound 1).
  • FIG. 1 shows the eGFR prior and after treatment with compound 1).
  • Repeat biopsies showed resolution of glomerular endocapillary hypercellularity and reduction in glomerular macrophages.
  • Compound 1 stabilized eGFR and reduced glomerular inflammation.
  • Endocapillary hypercellularity/ Immunoflourescence CD68-positive total microscopy cells/glomerulus Pre- 3/11 C3, 2+; IgM, trace 11 treatment 2 months 0/36 C3, 2+; IgM, neg 2-3 7 months 0/14 C3, 2+; IgM, neg 1-2
  • FIG. 2 represents the histopathological improvement following treatment with compound 1.
  • H&E Haemotoxylin and Eosin staining before treatment with compound 1 demonstrates fibrinoid necrosis and multiple inflammatory cells.
  • C Periodic acid-Schiff (PAS) staining after treatment with compound 1 shows a reduction in endocapillary hypercellularity and glomerular inflammation.
  • B CD68 staining before treatment with compound 1.
  • D CD68 staining after treatment with compound 1 demonstrates a reduction in glomerular macrophages.
  • the aim of this study is to evaluate the efficacy, safety, and tolerability of compound 1 in a patient with progressive complement 3 (C3) glomerulonephritis.
  • the primary safety objective of this study is to evaluate the safety and tolerability of compound 1.
  • the primary efficacy objective is to evaluate the efficacy of compound 1 based on change from baseline in eGFR (MDRD, Estimated Glomerular Filtration Rate) and proteinuria.
  • the secondary objectives of this study include assessment of:
  • the patient will have biopsy proven recurrent C3GN prior to start of dosing, and be deemed eligible based on the inclusion and exclusion criteria.
  • Screening procedures will includerecording of demographics, medical history, medication history, physical examination and vital signs, serum chemistry, hematology, urinalysis (including UPCR measurement), viral screening (if not performed within prior 12 weeks), and estimated glomerular filtration rate (eGFR) assessment based on serum creatinine.
  • the baseline eGFR needs to be at least 25 mL/min/1.73 m 2 for study eligibility.
  • the patient On Day 1, the patient will start compound 1 treatment. Patients will take compound 1 30 mg orally twice daily for an initial period of 84 days. The patient will visit the study center on Days 1, 8, 15, 29, 57, and 85. The compound 1 dose will be taken in the morning optimally within one hour after breakfast and in the evening optimally within one hour after dinner. If the patient's clinical condition stabilizes or improves, and there are no adverse events preventing further treatment, the patient may be treated for another 84-day treatment cycle. The 84-day cycles may be repeated for a total of up to 4 cycles under this protocol. For the 84-day cycles after the first cycle, the patient will visit the study center every 4 weeks. There will be 4-week follow-up period after the patient stops the compound 1 treatment.
  • Duration of treatment with compound 1 84 days with up to 3 repeats of the 84-day cycle for a total period of up to 336 days.
  • the patient's condition will be evaluated by the Investigator at the end of the study and appropriate standard of care medical treatment will be provided as needed.
  • the patient will be screened within a period not to exceed 21 days prior to Day 1.
  • the compound 1 treatment period is at least 84 days and up to 336 days, and the patient will be followed for 4 weeks (28 days) after dosing is stopped.
  • any adverse events that are deemed study drug-related and are ongoing at discharge will be followed-up to resolution or until a determination is made that the unresolved event is stable.
  • the patient's condition will be evaluated by the Investigator at the end of the study and appropriate standard of care medical treatment will be provided as needed.
  • Safety assessments include adverse events, physical examination abnormalities, vital signs, and clinical laboratory tests (including blood chemistry, hematology, and urinalysis).
  • Concentrations of compound 1 and possible metabolites will be determined in plasma from 2-mL blood samples collected in EDTA tubes on Days 8, 15, 29, 57, and 85. The date and time of the last dose of compound 1 prior to sample collection for compound 1 measurement will be recorded. The samples will be kept frozen at ⁇ 70° C. or lower and shipped on dry ice for assay.
  • Plasma samples will continue to be collected every 4 weeks during any subsequent 84-day cycles.
  • Plasma samples will be collected on Day 1 (pre-dose), and Days 8, 15, 29, 57, and 85 for pharmacodynamic marker measurements, including, for example, complement fragments, and inflammatory cytokine and chemokine levels.
  • Urine samples will also be collected on Day 1 (pre-dose) and Days 8, 15, 29, 57, and 85 for biomarker assessments including, for example, MCP-1, complement fragments, and inflammatory chemokine and cytokine levels.
  • Plasma and urine samples will continue to be collected every 4 weeks during any subsequent 84-day cycles.
  • Renal biopsies will be analyzed by periodic acid-Schiff (PAS) staining, immunofluorescence staining for C3, C5b-9, and potentially other markers. Electron microscopy may also be performed.
  • PAS periodic acid-Schiff
  • the primary safety endpoint is the patient incidence of adverse events.
  • Treatment-emergent adverse events will be listed by System Organ Class, by relatedness and by maximum severity. Serious adverse events and adverse events leading to withdrawal will be listed. Vital signs and change from baseline in vital signs will be listed by study visit. Laboratory data (actual values and change from baseline) will be listed by study visit. Abnormal laboratory values will be flagged.
  • the primary efficacy endpoints are the change from baseline over the treatment period in eGFR and first morning urinary PCR.
  • Plasma samples will be collected on Days 8, 15, 29, 57, and 85 to determine the plasma concentrations of compound 1 (and metabolites). Plasma concentrations of compound 1 will be listed and plotted by study visit.
  • Example 2 A Randomized, Double-Blind, Placebo-Controlled Phase 2 Study to Evaluate the Safety and Efficacy of Compound 1 in Patients with C3 Glomerulopathy
  • the aim of this study is to evaluate the effect of compound 1 treatment on renal disease activity in patients with complement 3 glomerulopathy (C3G).
  • C3G complement 3 glomerulopathy
  • the primary objective is to evaluate the efficacy of compound 1 compared to placebo based on histologic changes in C3G pathology from kidney biopsies taken before and during treatment.
  • the secondary objectives of this study include assessment of:
  • changes from baseline in markers of alternative complement pathway involvement e.g., C3, C3d, C3c, C3adesArg, C5, C5a, C5b-9, C5adesArg, and other markers of inflammation, may be assessed in plasma/serum or urine over the course of the treatment period.
  • Patients will then be randomized, 1:1, to receive 30 mg compound 1 twice daily or matching placebo for 26 weeks in a double-blind, placebo-controlled manner.
  • the 26-week double-blind period will be followed by a 26-week period during which all patients will receive compound 1 treatment.
  • Patients will be screened for enrollment based on biopsy proven C3 glomerulopathy (i.e., ⁇ 2-levels of magnitude greater staining of C3 than any combination of IgG, IgM, IgA, and C1q) and evidence of inflammation based on leukocyte infiltration and/or endocapillary proliferation.
  • C3 glomerulopathy i.e., ⁇ 2-levels of magnitude greater staining of C3 than any combination of IgG, IgM, IgA, and C1q
  • evidence of inflammation based on leukocyte infiltration and/or endocapillary proliferation.
  • the screening period will be up to 28 days. Screening procedures will include written informed consent, demographics, medical history, medication history, physical examination and vital signs, 12-lead ECG, serum pregnancy test for women of childbearing potential, serum chemistry (including serum creatinine), hematology, urinalysis, urinary protein:creatinine ratio (PCR), viral and TB screening. If a patient did not have a renal biopsy in the past 12 weeks, a renal biopsy needs to be done prior to dosing. Prior to starting study drug treatment, blood samples will be collected for the following measurements to create a baseline profile for all patients:
  • Patients meeting inclusion criteria will start study drug treatment on Day 1. Patients will take compound 1 30 mg or matching placebo orally twice daily. The treatment period is 52 weeks (364 days). The study drug will be taken in the morning preferably with food and in the evening preferably with food, approximately 12 hours after the morning dose. Patients who receive placebo during the first 26 weeks, will receive compound 1 in a blinded cross-over. After the 364-day treatment period, all patients will be followed for 8 weeks (56 days) without study drug treatment.
  • the dose(s) of concomitant immunosuppressive treatment may not be increased during the study.
  • Treatment with these other drugs may be reduced or stopped during the study, if the patient's condition justifies it. No new treatments may be added during the study period (active treatment period or follow up), unless the patient's condition deteriorates to the extent that the investigator deems it in the best interest of the patient to do so. This will be considered a treatment failure.
  • compound 1 or placebo dosing will initially be given based on the body weight at screening and the dose will be adjusted based on compound 1 plasma levels as shown in the table below.
  • AUC 0-6 AUC 0-6 thresholds are based on the mean compound 1 plasma exposure (525 ng*hr/mL) and one standard deviation (174 ng ⁇ hr/mL) above or below the mean in adult patients from Phase 2 study CL002_168 in AAV.
  • Patients will be screened within a period not to exceed 28 days prior to Day 1.
  • the treatment period is 52 weeks (364 days) and all patients will be followed for 8 weeks (56 days) after the dosing period.
  • any adverse events that are deemed study drug-related and are ongoing at discharge will be followed-up to resolution or until a determination is made that the unresolved event is stable.
  • the patient's condition will be evaluated by the Investigator at the end of the clinical trial and appropriate standard of care medical treatment will be provided to all patients as needed.
  • Safety assessments include adverse events, physical examination abnormalities, vital signs, and clinical laboratory tests (including blood chemistry, hematology, and urinalysis).
  • Plasma/serum samples will be collected according to the Time and Events Table for pharmacodynamic marker measurements, including, for example, complement fragments, and inflammatory cytokine and chemokine levels.
  • Urine samples will also be collected according to the Time and Events Table for biomarker assessments including, for example, complement fragments, sCD 163, and inflammatory chemokine and cytokine levels.
  • renal biopsy samples will be assessed by immunofluorescence staining for C3 and immunoglobulins.
  • Patients must have biopsy-proven C3 glomerulopathy, either DDD or C3GN, with ⁇ 2-levels of magnitude greater staining of C3 than any combination of IgG, IgM, IgA, and C1q, and with evidence of inflammation, based on leukocyte infiltration or endocapillary proliferation, observed in a renal biopsy taken within 12 weeks prior to screening or during screening.
  • All renal biopsies will also be analyzed based on hematoxylin-eosin (H&E) staining, periodic acid-Schiff (PAS) staining, trichrome, and Jones methenamine silver staining. These renal biopsies will be evaluated by a central reader, blinded to treatment assignment from either slides or high-resolution electronic images.
  • H&E hematoxylin-eosin
  • PAS periodic acid-Schiff
  • trichrome trichrome
  • Jones methenamine silver staining will be evaluated by a central reader, blinded to treatment assignment from either slides or high-resolution electronic images.
  • the central reader will determine the degree of disease activity and chronicity.
  • the primary efficacy endpoint is the percent change from baseline to week 26 in the C3G Histologic Index (CHI) for disease activity.
  • the compound 1 and placebo groups will be compared by ANCOVA with treatment group and randomization strata (C3GN or DDD, and renal transplant or not) as factors, and baseline as covariate. Point estimates and corresponding 95% confidence intervals will be estimated for the difference between the compound 1 and placebo control group.
  • Continuous variables including eGFR, urinary PCR, urinary MCP-1:creatinine ratio, EQ-5D-5L, and SF-36 v2 will be analyzed using a mixed effects model for repeated measures (MMRM) with treatment group, visit, treatment-by-visit interaction, and randomization strata (C3GN or DDD, and renal transplant or not) as factors, and baseline as covariate. Patients will be considered as repeated measure units over visits. Point estimates and corresponding 95% confidence intervals will be estimated for the difference between the compound 1 group and the control group across 26 weeks using simple contrast from the model. Similar to the primary endpoint, the second 26 weeks will be compared to the first 26 weeks for the placebo group.
  • MMRM mixed effects model for repeated measures
  • Treatment-emergent serious adverse events and adverse events leading to withdrawal will be summarized by treatment group.
  • Plasma samples will be collected over the course of the study to determine the PK profile of compound 1 (and metabolites). Individual plasma concentrations of compound 1 (and metabolites) will be listed, plotted, and summarized descriptively and graphically. PK parameters will be calculated based on plasma compound 1 concentrations at the time of sample collection in relation to time of administration of the most recent dose of study medication. PK parameters of significant metabolites may also be calculated.
  • Plasma and urinary PD markers will be summarized and may be analyzed using methods analogous to the efficacy parameters. The following parameters will be determined, where possible, in 12-17 year old patients:
US15/404,610 2016-01-14 2017-01-12 Method of treating c3 glomerulopathy Abandoned US20170202821A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/404,610 US20170202821A1 (en) 2016-01-14 2017-01-12 Method of treating c3 glomerulopathy
US16/184,535 US11285138B2 (en) 2016-01-14 2018-11-08 Method of treating C3 glomerulopathy
US17/670,604 US11779576B2 (en) 2016-01-14 2022-02-14 Method of treating C3 glomerulopathy

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201662278788P 2016-01-14 2016-01-14
US201662280346P 2016-01-19 2016-01-19
US201662347450P 2016-06-08 2016-06-08
US201662397527P 2016-09-21 2016-09-21
US15/404,610 US20170202821A1 (en) 2016-01-14 2017-01-12 Method of treating c3 glomerulopathy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/184,535 Continuation US11285138B2 (en) 2016-01-14 2018-11-08 Method of treating C3 glomerulopathy

Publications (1)

Publication Number Publication Date
US20170202821A1 true US20170202821A1 (en) 2017-07-20

Family

ID=59312069

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/404,610 Abandoned US20170202821A1 (en) 2016-01-14 2017-01-12 Method of treating c3 glomerulopathy
US16/184,535 Active US11285138B2 (en) 2016-01-14 2018-11-08 Method of treating C3 glomerulopathy
US17/670,604 Active US11779576B2 (en) 2016-01-14 2022-02-14 Method of treating C3 glomerulopathy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/184,535 Active US11285138B2 (en) 2016-01-14 2018-11-08 Method of treating C3 glomerulopathy
US17/670,604 Active US11779576B2 (en) 2016-01-14 2022-02-14 Method of treating C3 glomerulopathy

Country Status (17)

Country Link
US (3) US20170202821A1 (es)
EP (1) EP3402486A4 (es)
JP (3) JP7339733B2 (es)
KR (1) KR20180102642A (es)
CN (1) CN108601790A (es)
AU (1) AU2017207359C1 (es)
BR (2) BR112018014222A2 (es)
CA (1) CA3010735C (es)
IL (1) IL260539B (es)
MA (1) MA43872A (es)
MX (2) MX2018008624A (es)
NZ (1) NZ744083A (es)
RU (1) RU2742888C2 (es)
SG (1) SG11201805828YA (es)
TW (1) TWI791423B (es)
WO (1) WO2017123716A1 (es)
ZA (1) ZA201804513B (es)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10035768B2 (en) 2010-06-24 2018-07-31 Chemocentryx, Inc. C5aR antagonists
US10266492B2 (en) 2014-09-29 2019-04-23 Chemocentryx, Inc. Processes and intermediates in the preparation of C5aR antagonists
WO2019227102A1 (en) * 2018-05-25 2019-11-28 Achillion Pharmaceuticals, Inc. Complement alternative pathway-associated nephropathy biomarkers
WO2020051538A1 (en) * 2018-09-06 2020-03-12 Achillion Pharmaceuticals, Inc. Morphic forms of complement factor d inhibitors
US10660897B2 (en) 2008-12-22 2020-05-26 Chemocentryx, Inc. C5aR antagonists
US11084800B2 (en) 2017-03-01 2021-08-10 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11285138B2 (en) 2016-01-14 2022-03-29 Chemocentryx, Inc. Method of treating C3 glomerulopathy
US11447465B2 (en) 2017-03-01 2022-09-20 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for treatment of medical disorders
US11649229B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US11649223B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US11807627B2 (en) 2018-09-25 2023-11-07 Achillon Pharmaceuticals, Inc. Morphic forms of complement factor D inhibitors
US11814391B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for the treatment of medical disorders

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020529227A (ja) * 2018-01-30 2020-10-08 プロクター アンド ギャンブル インターナショナル オペレーションズ エスエーProcter & Gamble International Operations Sa 懸濁粒子を含む相安定で噴霧可能なフレッシュニング組成物
WO2020112961A1 (en) 2018-11-30 2020-06-04 Chemocentryx, Inc. Capsule formulations
WO2022028586A1 (en) * 2020-08-07 2022-02-10 Kira Pharmaceuticals (Suzhou) Ltd. Compounds as c5ar inhibitors
CN116635075A (zh) * 2020-12-21 2023-08-22 坎莫森特里克斯公司 使用c5a抑制剂治疗c3肾小球病
CN115192563B (zh) * 2022-05-09 2023-10-13 北京大学第一医院 C3a/C3aR通路拮抗剂治疗原发性膜性肾病的用途

Family Cites Families (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1268002A (en) 1917-09-12 1918-05-28 William M Goodwin Measuring instrument.
US4166452A (en) 1976-05-03 1979-09-04 Generales Constantine D J Jr Apparatus for testing human responses to stimuli
US4256108A (en) 1977-04-07 1981-03-17 Alza Corporation Microporous-semipermeable laminated osmotic system
US4265874A (en) 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4686100A (en) 1985-04-02 1987-08-11 The Board Of Trustees Of The Leland Stanford Junior University Method for the treatment of adult respiratory distress syndrome
AU772477B2 (en) 1998-08-28 2004-04-29 Scios Inc. Inhibitors of p38-alpha kinase
RU2197288C2 (ru) * 1999-08-03 2003-01-27 Тверская медицинская академия Способ лечения больных хроническим гломерулонефритом
US6713502B2 (en) 2000-05-05 2004-03-30 Smithkline Beecham Corporation Anti-infectives
US7105567B2 (en) 2000-08-10 2006-09-12 Mitsubishi Pharma Corporation 3-substituted urea derivatives and medicinal use thereof
CA2422342C (en) 2000-09-14 2009-12-08 Mitsubishi Pharma Corporation Novel amide derivatives and medicinal use thereof
AU7622500A (en) 2000-09-29 2002-07-01 Neurogen Corp High affinity small molecule c5a receptor modulators
JP4958387B2 (ja) 2001-08-17 2012-06-20 ジェネンテック, インコーポレイテッド C5bの形成を妨害することなくc5及びc5aに結合する補体経路阻害剤
WO2003037888A1 (en) 2001-09-21 2003-05-08 Mitsubishi Pharma Corporation 3-substituted-4-pyrimidone derivatives
ITMI20012025A1 (it) 2001-09-28 2003-03-28 Dompe Spa Sali di ammonio quaternari di omega-amminoalchilammidi di acidi r 2-aril-propionici e composizioni farmaceutiche che li contengono
US20030195192A1 (en) 2002-04-05 2003-10-16 Fortuna Haviv Nicotinamides having antiangiogenic activity
EP1487796A4 (en) 2002-03-28 2005-11-16 Neurogen Corp SUBSTITUTED BIARYLAMIDES AS MODULATORS OF THE C5A RECEPTOR
AU2003218374A1 (en) 2002-03-28 2003-10-13 Neurogen Corporation Substituted tetrahydroisoquinolines as c5a receptor modulators
US20040014782A1 (en) 2002-03-29 2004-01-22 Krause James E. Combination therapy for the treatment of diseases involving inflammatory components
US20040014744A1 (en) 2002-04-05 2004-01-22 Fortuna Haviv Substituted pyridines having antiangiogenic activity
AU2003257094A1 (en) 2002-08-08 2004-02-25 Boehringer Ingelheim Pharmaceuticals, Inc. Substituted benzimidazole compounds
US7169775B2 (en) 2002-08-21 2007-01-30 Neurogen Corporation Amino methyl imidazoles as C5a receptor modulators
US7455837B2 (en) 2002-11-05 2008-11-25 The Regents Of The University Of Michigan Compositions and methods for the diagnosis and treatment of sepsis
FR2846654A1 (fr) 2002-11-05 2004-05-07 Servier Lab Nouveaux derives de la 2,3-dihydro-4(1h)-pyridinone, leur procede de preparation et les compositions pharmaceutiques qui les contiennent
WO2004043925A2 (en) 2002-11-08 2004-05-27 Neurogen Corporation 3-substituted-6-aryl pyridined as ligands of c5a receptors
AU2003902354A0 (en) 2003-05-15 2003-05-29 Harkin, Denis W. Treatment of haemorrhagic shock
MXPA05013770A (es) 2003-06-19 2006-03-08 Pfizer Prod Inc Antagonista de neuroquinina 1.
WO2005007087A2 (en) 2003-07-03 2005-01-27 Neurogen Corporation Substituted (heterocycloalkyl)methyl azole derivatives as c5a receptor modulators
GB0325956D0 (en) 2003-11-06 2003-12-10 Addex Pharmaceuticals Sa Novel compounds
BRPI0512535A (pt) 2004-06-24 2008-03-25 Incyte Corp compostos de piperidinas n-substituìdas, suas composições e métodos de modulações
WO2006071958A1 (en) 2004-12-29 2006-07-06 Millennium Pharmaceuticals, Inc. Compounds useful as chemokine receptor antagonists
US8399489B2 (en) 2005-02-18 2013-03-19 Astrazeneca Ab Antibacterial piperdine derivatives
WO2007035428A1 (en) 2005-09-15 2007-03-29 Bristol-Myers Squibb Company Met kinase inhibitors
US20070112015A1 (en) 2005-10-28 2007-05-17 Chemocentryx, Inc. Substituted dihydropyridines and methods of use
AU2006316452B2 (en) 2005-11-24 2013-01-10 Dompe' Farmaceutici S.P.A. (R)-arylalkylamino derivatives and pharmaceutical compositions containing them
DK2359834T5 (en) 2006-03-15 2017-02-06 Alexion Pharma Inc Treatment of paroxysmal nocturnal hemoglobinuria patients with a complement inhibitor
WO2008022060A2 (en) 2006-08-14 2008-02-21 Novartis Ag Imidazo-pyridine derivatives for modulating protein kinase activity
WO2008053270A2 (en) * 2006-10-31 2008-05-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of an egfr antagonist for the treatment of glomerolonephritis
RU2010102899A (ru) 2007-06-29 2011-08-10 Торрент Фармасьютикалз Лтд. (In) Новые замещенные пиперидоны в качестве индукторов hsp
CA2737219C (en) 2008-08-11 2017-02-28 Tracy Keller Halofuginone analogs for inhibition of trna synthetases and uses thereof
WO2010025510A1 (en) 2008-09-03 2010-03-11 Xenome Ltd Libraries of peptide conjugates and methods for making them
US20100074863A1 (en) 2008-09-17 2010-03-25 Yat Sun Or Anti-infective pyrrolidine derivatives and analogs
AU2009313203B2 (en) 2008-11-10 2015-08-27 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2010075257A1 (en) 2008-12-22 2010-07-01 Chemocentryx, Inc. C5ar antagonists
US20110275639A1 (en) 2008-12-22 2011-11-10 Chemocentryx, Inc. C5aR ANTAGONISTS
US20120252792A1 (en) 2009-09-17 2012-10-04 The Regents Of The University Of Michigan Methods and compositions for modulating rho-mediated gene transcription
PL2585064T3 (pl) 2010-06-24 2017-09-29 Chemocentryx, Inc. Antagoniści C5AR
US8147400B1 (en) * 2011-01-17 2012-04-03 Coloplast A/S Penile implant with dilatant liquid
AU2013277154A1 (en) * 2012-06-20 2014-12-18 Jonathan E. Grob Complement pathway modulators and uses thereof
CN105392803B (zh) 2013-05-08 2019-12-06 诺和诺德股份有限公司 C5aR拮抗剂的用途
CN113121415A (zh) 2014-09-29 2021-07-16 凯莫森特里克斯股份有限公司 制备C5aR拮抗剂的方法和中间体
BR112018014222A2 (pt) 2016-01-14 2018-12-11 Chemocentryx, Inc. método de tratamento da glomerulopatia c3
MX2018011831A (es) 2016-04-04 2019-02-13 Chemocentryx Inc Antagonistas solubles de receptor de c5aa(c5ar).
CN116635075A (zh) 2020-12-21 2023-08-22 坎莫森特里克斯公司 使用c5a抑制剂治疗c3肾小球病

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10660897B2 (en) 2008-12-22 2020-05-26 Chemocentryx, Inc. C5aR antagonists
US10035768B2 (en) 2010-06-24 2018-07-31 Chemocentryx, Inc. C5aR antagonists
US10266492B2 (en) 2014-09-29 2019-04-23 Chemocentryx, Inc. Processes and intermediates in the preparation of C5aR antagonists
US11845729B2 (en) 2014-09-29 2023-12-19 Chemocentryx, Inc. Processes and intermediates in the preparation of C5aR antagonists
US10532982B2 (en) 2014-09-29 2020-01-14 Chemocentryx, Inc. Processes and intermediates in the preparation of C5aR antagonists
US11649229B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US11649223B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US11779576B2 (en) 2016-01-14 2023-10-10 Chemocentryx, Inc. Method of treating C3 glomerulopathy
US11285138B2 (en) 2016-01-14 2022-03-29 Chemocentryx, Inc. Method of treating C3 glomerulopathy
US11708351B2 (en) 2017-03-01 2023-07-25 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11447465B2 (en) 2017-03-01 2022-09-20 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for treatment of medical disorders
US11084800B2 (en) 2017-03-01 2021-08-10 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
WO2019227102A1 (en) * 2018-05-25 2019-11-28 Achillion Pharmaceuticals, Inc. Complement alternative pathway-associated nephropathy biomarkers
CN112839945A (zh) * 2018-09-06 2021-05-25 艾其林医药公司 补体因子d抑制剂的形态形式
WO2020051538A1 (en) * 2018-09-06 2020-03-12 Achillion Pharmaceuticals, Inc. Morphic forms of complement factor d inhibitors
US11814391B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for the treatment of medical disorders
US11814363B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Morphic forms of danicopan
US11807627B2 (en) 2018-09-25 2023-11-07 Achillon Pharmaceuticals, Inc. Morphic forms of complement factor D inhibitors

Also Published As

Publication number Publication date
JP2021183625A (ja) 2021-12-02
RU2018129378A (ru) 2020-02-14
JP2023126235A (ja) 2023-09-07
KR20180102642A (ko) 2018-09-17
TWI791423B (zh) 2023-02-11
ZA201804513B (en) 2024-01-31
US20190201388A1 (en) 2019-07-04
AU2017207359B2 (en) 2022-04-14
EP3402486A4 (en) 2019-08-28
EP3402486A1 (en) 2018-11-21
US11779576B2 (en) 2023-10-10
JP2019501935A (ja) 2019-01-24
NZ744083A (en) 2022-07-01
CN108601790A (zh) 2018-09-28
CA3010735C (en) 2023-06-13
BR122024002810A2 (pt) 2024-03-12
MA43872A (fr) 2021-05-19
MX2022006069A (es) 2023-01-12
WO2017123716A1 (en) 2017-07-20
SG11201805828YA (en) 2018-08-30
CA3010735A1 (en) 2017-07-20
US11285138B2 (en) 2022-03-29
RU2742888C2 (ru) 2021-02-11
RU2018129378A3 (es) 2020-05-20
AU2017207359A1 (en) 2018-07-26
BR112018014222A2 (pt) 2018-12-11
MX2018008624A (es) 2018-12-10
IL260539B (en) 2021-05-31
JP7339733B2 (ja) 2023-09-06
US20220354837A1 (en) 2022-11-10
AU2017207359C1 (en) 2022-09-01
TW201726134A (zh) 2017-08-01

Similar Documents

Publication Publication Date Title
US11779576B2 (en) Method of treating C3 glomerulopathy
US10487098B2 (en) Soluble C5aR antagonists
US20230105869A1 (en) C5aR INHIBITOR REDUCTION OF URINARY sCD163
US20220257580A1 (en) Treatment of c3 glomerulopathy using a c5a inhibitor
CA3155596A1 (en) Free base crystalline form of a complement component c5a receptor

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHEMOCENTRYX, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BEKKER, PETRUS;REEL/FRAME:046096/0813

Effective date: 20180220

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION