US20180297929A1 - Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor - Google Patents

Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor Download PDF

Info

Publication number
US20180297929A1
US20180297929A1 US15/956,172 US201815956172A US2018297929A1 US 20180297929 A1 US20180297929 A1 US 20180297929A1 US 201815956172 A US201815956172 A US 201815956172A US 2018297929 A1 US2018297929 A1 US 2018297929A1
Authority
US
United States
Prior art keywords
gemcabene
compound
less
pharmaceutically acceptable
subject
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
US15/956,172
Other languages
English (en)
Inventor
Daniela Carmen Oniciu
Charles Larry Bisgaier
José Rui GOMES
Stefan Heckhoff
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.)
Corden Pharma Switzerland LLC
Neurobo Pharmaceuticals Inc
Original Assignee
Gemphire Therapeutics LLC
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
Priority to US15/956,172 priority Critical patent/US20180297929A1/en
Application filed by Gemphire Therapeutics LLC filed Critical Gemphire Therapeutics LLC
Assigned to GEMPHIRE THERAPEUTICS INC. reassignment GEMPHIRE THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BISGAIER, CHARLES LARRY, ONICIU, DANIELA CARMEN
Assigned to CORDEN PHARMA SWITZERLAND LLC reassignment CORDEN PHARMA SWITZERLAND LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GOMES, JOSÉ RUI
Assigned to GEMPHIRE THERAPEUTICS INC. reassignment GEMPHIRE THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CORDEN PHARMA SWITZERLAND LLC
Assigned to CORDEN PHARMA SWITZERLAND LLC reassignment CORDEN PHARMA SWITZERLAND LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HECKHOFF, Stefan
Assigned to GEMPHIRE THERAPEUTICS INC. reassignment GEMPHIRE THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CORDEN PHARMA SWITZERLAND LLC
Publication of US20180297929A1 publication Critical patent/US20180297929A1/en
Priority to US16/402,853 priority patent/US20200148617A1/en
Assigned to GEMPHIRE THERAPEUTICS INC. reassignment GEMPHIRE THERAPEUTICS INC. CHANGE OF ADDRESS FOR ASSIGNEE Assignors: GEMPHIRE THERAPEUTICS INC.
Assigned to NEUROBO PHARMACEUTICALS, INC. reassignment NEUROBO PHARMACEUTICALS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: GEMPHIRE THERAPEUTICS INC.
Priority to US17/583,081 priority patent/US20220332672A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C62/00Compounds having carboxyl groups bound to carbon atoms of rings other than six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C62/08Saturated compounds containing ether groups, groups, groups, or groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C51/00Preparation of carboxylic acids or their salts, halides or anhydrides
    • C07C51/42Separation; Purification; Stabilisation; Use of additives
    • C07C51/43Separation; Purification; Stabilisation; Use of additives by change of the physical state, e.g. crystallisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C51/00Preparation of carboxylic acids or their salts, halides or anhydrides
    • C07C51/42Separation; Purification; Stabilisation; Use of additives
    • C07C51/47Separation; Purification; Stabilisation; Use of additives by solid-liquid treatment; by chemisorption
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/235Saturated compounds containing more than one carboxyl group
    • C07C59/305Saturated compounds containing more than one carboxyl group containing ether groups, groups, groups, or groups

Definitions

  • This invention provides pharmaceutically acceptable salts of 6-(5-carboxy-5-methyl-hexyloxy)-2,2-dimethyl-hexanoic acid (“gemcabene”), wherein the pharmaceutically acceptable salts have a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m as measured by laser light diffraction, and compositions comprising (i) an effective amount of a pharmaceutically acceptable salt of gemcabene, wherein the pharmaceutically acceptable salt has a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m as measured by laser light diffraction, and (ii) a pharmaceutically acceptable carrier or vehicle.
  • This invention further provides methods for purifying crude gemcabene, comprising dissolving the crude gemcabene in in heptane to provide a heptane solution of the crude gemcabene and cooling the heptane solution to a temperature ranging from 10° C. to 15° C. to precipitate gemcabene.
  • the invention further provides pharmaceutically acceptable salts of gemcabene as synthesized or purified by the methods of the invention.
  • the pharmaceutically acceptable salts of gemcabene and compositions thereof are useful for treating or preventing liver disease or an abnormal liver condition, a disorder of lipoprotein or glucose metabolism, a cardiovascular or related vascular disorder, a disease caused by fibrosis (such as liver fibrosis), or a disease associated with inflammation (such as liver inflammation).
  • LDL-C low-density lipoprotein cholesterol
  • triglycerides are associated with mixed dyslipidemia including type IIb hyperlipidemia.
  • Type IIb is characterized by elevation of apolipoprotein B, very low-density lipoprotein cholesterol (VLDL-C), intermediate density lipoprotein cholesterol (IDL), and small dense low-density lipoprotein (LDL) levels, in addition to elevation in LDL-C and triglyceride levels.
  • VLDL-C very low-density lipoprotein cholesterol
  • IDL intermediate density lipoprotein cholesterol
  • LDL small dense low-density lipoprotein
  • FVF familial combined hyperlipidemia
  • Familial hyperlipidemias can be classified according to the Fredrickson classification, which is based on the pattern of lipoprotein migration in electrophoresis or ultracentrifugation.
  • type IIb patients have a high risk of developing non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatosis hepatitis (NASH), which are forms of fatty liver that can develop due to hepatic triglyceride overproduction and accumulation.
  • NAFLD non-alcoholic fatty liver disease
  • NASH non-alcoholic steatosis hepatitis
  • NAFLD is strongly associated with features of metabolic syndrome, including obesity, insulin resistance, type-2 diabetes mellitus, and dyslipidemia. NASH can cause the liver to swell and become damaged. NASH tends to develop in people who are overweight or obese, or have diabetes, or mixed dyslipidemia, or high cholesterol or high triglycerides or an inflammatory condition. NASH is marked by hepatocyte ballooning and liver inflammation, which can lead to liver damage and progress to scarring and irreversible changes, similar to the damage caused by heavy alcohol use.
  • NAFLD NAFLD
  • NASH or fatty liver can lead to metabolic complications including elevation of liver enzymes, fibrosis, cirrhosis, hepatocellular carcinoma, and liver failure.
  • Liver failure is life-threatening and therefore there is a need to develop therapies to delay development, prevent formation or reverse the condition of a fatty liver, such as in type IIb patients and other patients at risk for, or present with fatty liver disease.
  • statins are very effective at lowering LDL-C, in general they are not very effective at also lowering triglyceride concentrations. Further, high dose statin therapy is often not well tolerated because it can cause muscle pain (myalgia) and increase patient's risk for serious muscle toxicity, such as rhabdomyolysis. Also, commonly used triglyceride lowering agents that are given in combination with statins are not well-tolerated. Fibrates when given with statins are known to have drug-drug interactions resulting in increased statin blood drug levels and present an increased safety risk.
  • a safe and efficacious treatment for type IIb hyperlipidemia which can lower one or both LDL-C concentrations and triglyceride concentrations, treatment or prevention of liver disease or an abnormal liver condition, a disorder of lipoprotein or glucose metabolism, a cardiovascular or related vascular disorder, a disease caused by increased levels of fibrosis, or a disease associated with increased inflammation, with minimal risks or side effects.
  • a pharmaceutically acceptable salt of gemcabene having a PSD90 of less than 30 ⁇ m can be difficult to handle due to its low density and/or increased electrostatic properties.
  • particles having low density and/or high electrostatic properties render tableting these particles difficult, particularly in manufacturing processes.
  • the present invention provides pharmaceutically acceptable salts of gemcabene, the pharmaceutically acceptable salts having a particle size distribution characterized by a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m as measured by laser light diffraction and providing a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m as measured by laser light diffraction
  • a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention still further provides pharmaceutically acceptable salts of gemcabene, the pharmaceutically acceptable salts having a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m as measured by laser light diffraction and providing a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • the present invention still further provides methods for purifying crude gemcabene, wherein the crude gemcabene comprises no more than 1% w/w of 2,2,7,7-tetramethyl-octane-1,8-dioic acid as determined by high-performance liquid chromatography, comprising: dissolving the crude gemcabene in heptane to provide a heptane solution of the crude gemcabene; and cooling the heptane solution to a temperature ranging from 10° C. to 15° C. to precipitate gemcabene, wherein the gemcabene comprises 0.5% w/w or less of 2,2,7,7-tetramethyl-octane-1,8-dioic acid by area as determined by high-performance liquid chromatography.
  • the present invention still further provides gemcabene purified by the methods of the present invention.
  • the present invention still further provides pharmaceutically acceptable salts of gemcabene prepared from the gemcabene purified by the methods of the present invention.
  • a gemcabene pharmaceutically acceptable salt disclosed herein is a “compound of the invention”.
  • compositions comprising an effective amount of a compound of the invention, and a pharmaceutically acceptable carrier or vehicle (each composition being a “composition of the invention”).
  • the present invention still further provides methods for treating or preventing a liver disease or an abnormal liver condition, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing a disorder of lipoprotein metabolism, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for reducing in a subject's blood plasma or blood serum the subject's total cholesterol concentration, low-density lipoprotein cholesterol concentration, low-density lipoprotein concentration, very low-density lipoprotein cholesterol concentration, very low-density lipoprotein concentration, non-HDL cholesterol concentration, non-HDL concentration, apolipoprotein B concentration, triglyceride concentration, apolipoprotein C-III concentration, C-reactive protein concentration, fibrinogen concentration, lipoprotein(a) concentration, interleukin-6 concentration, angiopoietin-like protein 3 concentration, angiopoietin-like protein 4 concentration, PCSK9 concentration, or serum amyloid A concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for elevating in the subject's blood plasma or blood serum the subject's high-density lipoprotein cholesterol concentration, high-density lipoprotein concentration, high-density cholesterol triglyceride concentration, adiponectin concentration or apolipoprotein A-I concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing thrombosis, a blood clot, a primary cardiovascular event, a secondary cardiovascular event, progression to nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, liver cirrhosis hepatocellular carcinoma, liver failure, pancreatitis, pulmonary fibrosis or hyperlipoproteinemia type IIB, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for reducing a subject's risk of developing thrombosis, a blood clot, a primary cardiovascular event, a secondary cardiovascular event, progression to nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, liver cirrhosis, hepatocellular carcinoma, liver failure, pancreatitis, pulmonary fibrosis or hyperlipoproteinemia type IIB, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods of reducing or inhibiting progression of fibrosis, steatosis, ballooning or inflammation in the liver of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for reducing post-prandial lipemia or preventing prolonged post-prandial lipemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for reducing a fibrosis score or a nonalcoholic fatty liver disease activity score in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for stabilizing, regressing, or maintaining a fibrosis score or a nonalcoholic fatty liver disease activity score in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for slowing the progression of a fibrosis score or a nonalcoholic fatty liver disease activity score in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for reducing a fat content in a liver of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing a disorder of glucose metabolism, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing a cardiovascular disorder or a related vascular disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing inflammation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for preventing or reducing the risk of developing pancreatitis, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing a pulmonary disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for treating or preventing musculoskeletal discomfort, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention still further provides methods for lowering a subject's LDL-C concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • FIG. 1A is a line graph showing a dissolution profile of gemcabene from a composition of the invention in the form of a film-coated tablet.
  • FIG. 1B is a line graph showing a dissolution profile of gemcabene from a composition of the invention in the form of a film-coated tablet.
  • FIG. 2 is a scanning electron micrograph of gemcabene calcium salt hydrate Crystal Form 1 having a particle size distribution characterized by a PSD90 of about 58 ⁇ m as measured by laser light diffraction.
  • FIG. 3 is a line graph showing LDL-C concentrations of three familial hypercholesterolemia patients (1F, 2M and 3M) as measured during the course of their treatment with gemcabene calcium salt hydrate Crystal Form 1 having a particle size distribution characterized by a PSD90 of 52 ⁇ m as measured by laser light diffraction (gemcabene calcium salt hydrate Crystal Form 1, 300-mg strength film-coated tablet, Tablet D).
  • FIG. 4 is a line graph showing values for percent change from baseline of LDL-C concentrations of the three familial hypercholesterolemia patients (1F, 2M and 3M) shown in FIG. 3 as measured during the course of their treatment with gemcabene calcium salt hydrate Crystal Form 1 having a particle size distribution characterized by a PSD90 of 52 ⁇ m as measured by laser light diffraction (gemcabene calcium salt hydrate Crystal Form 1 300-mg strength film-coated tablet, Tablet D).
  • FIG. 5A shows photomicrographs of hematoxylin and eosin-stained liver sections of STAMTM model mice treated with gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (ID: 306) or with vehicle (ID: 208) and photomicrographs of hematoxylin and eosin-stained liver sections of normal mice treated with vehicle (ID: 103).
  • FIG. 5B shows photomicrographs of hematoxylin and eosin-stained liver sections of STAMTM model mice treated with gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (ID: 402 and 508) and photomicrographs of hematoxylin and eosin-stained liver sections of STAMTM model mice treated with reference compound telmisartan.
  • FIG. 6 shows photomicrographs of Sirius red-stained liver sections of STAMTM model mice treated with vehicle (ID: 208), treated with gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (ID: 303, 403, 501), or treated with reference compound telmisartan (ID: 606) and photomicrographs of Sirius red-stained liver sections of normal mice treated with vehicle (ID: 102).
  • FIG. 7 shows graphs with components of the NAFLD Activity Score (NAS) of STAMTM model mice treated with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction or reference compound telmisartan and normal mice treated with vehicle.
  • NAS NAFLD Activity Score
  • FIG. 8A shows a graph of the NAS in STAMTM model mice treated with (a) vehicle, gemcabene calcium salt hydrate Crystal Form 1 with a PSD90 of 52 ⁇ m as measured by laser light diffraction or reference compound telmisartan.
  • FIG. 8B shows a graph of the liver Sirius-red positive area (the fibrosis area) in STAMTM model mice treated with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction or reference compound telmisartan.
  • FIG. 9 is a graph showing non-fasting plasma triglyceride concentrations in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 10 is a graph showing gene expression levels of hepatic sulfatase 2 (Sulf-2) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • Sulf-2 hepatic sulfatase 2
  • gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 11 is a graph showing gene expression levels for hepatic apolipoprotein C-III (ApoC-III) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 12 is a graph showing gene expression levels for hepatic sterol regulatory element binding transcription factor 1 (SREBP-1) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • SREBP-1 hepatic sterol regulatory element binding transcription factor 1
  • FIG. 13 is a graph showing gene expression levels for hepatic chemokine (C-C motif) ligand 4 (MIP-1 ⁇ ) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • C-C motif hepatic chemokine
  • FIG. 14 is a graph showing gene expression levels for hepatic chemokine (C-C motif) receptor 5 (CCR5) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • C-C motif C-C motif receptor 5
  • FIG. 15 is a graph showing gene expression levels for chemokine (C-C motif) receptor 2 (CCR2) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • CCR2 chemokine receptor 2
  • FIG. 16 is a graph showing gene expression levels for hepatic nuclear factor of kappa light polypeptide gene enhancer in B cells 1 (NF- ⁇ B) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 17 is a graph showing gene expression levels for hepatic C-reactive protein, pentraxin-related (CRP) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • CRP pentraxin-related
  • FIG. 18 is a graph showing gene expression levels for hepatic low-density lipoprotein receptor (LDL-receptor) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • LDL-receptor hepatic low-density lipoprotein receptor
  • FIG. 19 is a graph showing gene expression levels for hepatic acetyl-coenzyme A carboxylase alpha (ACC1) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • ACC1 hepatic acetyl-coenzyme A carboxylase alpha
  • FIG. 20 is a graph showing gene expression levels for hepatic acetyl-coenzyme A carboxylase beta (ACC2) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • ACC2 hepatic acetyl-coenzyme A carboxylase beta
  • FIG. 21 is a graph showing gene expression levels for hepatic patatin-like phospholipase domain containing 3 (PNPLA3) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • PNPLA3 hepatic patatin-like phospholipase domain containing 3
  • FIG. 22 is a graph showing gene expression levels for hepatic matrix metalloproteinase 2 (MMP-2) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • MMP-2 hepatic matrix metalloproteinase 2
  • gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 23 is a graph showing gene expression levels for hepatic alcohol dehydrogenase 4 (class II), pi polypeptide (ADH4) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 24 is a graph showing hepatic gene expression levels for tumor necrosis factor alpha (TNF- ⁇ ) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • TNF- ⁇ tumor necrosis factor alpha
  • FIG. 25 is a graph showing gene expression levels for hepatic chemokine (C-C motif) ligand 2 (MCP-1) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • C-C motif hepatic chemokine
  • FIG. 26 is a graph showing hepatic gene expression levels for actin, alpha smooth muscle actin ( ⁇ -SMA) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • ⁇ -SMA alpha smooth muscle actin
  • FIG. 27 is a graph showing gene expression levels for hepatic tissue inhibitor of metalloproteinase 1 (TIMP-1) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • TMP-1 hepatic tissue inhibitor of metalloproteinase 1
  • FIG. 28 is a powder X-ray diffractogram of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (Sample 4 in Table 2).
  • FIG. 29 is a powder X-ray diffractogram of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 62 ⁇ m as measured by laser light diffraction (Sample 7 in Table 2).
  • FIG. 30 shows measurements of amorphous gemcabene calcium particle size distribution.
  • FIG. 31 shows the effect of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction on the correlation between hepatic ApoC-III or hepatic Sulf-2 and plasma triglycerides in a diabetic mouse model.
  • FIG. 32 is a graph showing hepatic gene expression levels for interleukin 6 (IL-6) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • IL-6 interleukin 6
  • FIG. 32 is a graph showing hepatic gene expression levels for interleukin 6 (IL-6) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 33 is a graph showing hepatic gene expression levels for interleukin 1 ⁇ (IL-1 ⁇ ) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • IL-1 ⁇ interleukin 1 ⁇
  • FIG. 34 is a graph showing hepatic gene expression levels for chemokine (C-X-C motif) ligand 1 (CXCL1/KC) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 35 is a graph showing hepatic gene expression levels for stearoyl-coenzyme A desaturase (SCD) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • SCD stearoyl-coenzyme A desaturase
  • FIG. 36 is a graph showing hepatic gene expression levels for lipoprotein lipase (LPL) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • LPL lipoprotein lipase
  • FIG. 37 is a graph showing hepatic gene expression levels for angiopoietin-like protein 3 (ANGPTL3) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • ANGPTL3 angiopoietin-like protein 3
  • FIG. 38 is a graph showing hepatic gene expression levels for angiopoietin-like protein 4 (ANGPTL4) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • ANGPTL4 angiopoietin-like protein 4
  • FIG. 39 is a graph showing hepatic gene expression levels for angiopoietin-like protein 8 (ANGPTL8) in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • ANGPTL8 angiopoietin-like protein 8
  • FIG. 40 is a graph showing hepatic gene expression levels for fetuin-A in normal mice treated with vehicle and NASH-induced mice treated for three weeks with vehicle, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 52 ⁇ m as measured by laser light diffraction (30, 100 or 300 mg/kg) or reference compound telmisartan (10 mg/kg).
  • FIG. 41A shows arithmetic-mean concentration of gemcabene ( ⁇ SD) versus time, overlaid by dose for the time points collected 0-24 h post-dose displayed on linear axes.
  • FIG. 41B shows arithmetic-mean concentration of gemcabene ( ⁇ SD) versus time, overlaid by dose for the time points collected 0-24 h post-dose displayed on semi-log axes.
  • FIG. 42A shows arithmetic-mean predose (C trough ) concentration of gemcabene ( ⁇ SD) versus time, overlaid by dose.
  • FIG. 42B shows arithmetic-mean predose (C trough ) concentration of gemcabene ( ⁇ SD) versus time, overlaid by dose with the 900 mg Day 28 trough concentration from patient 006-003 excluded.
  • FIG. 43 is a line graph showing values for percent change from baseline of LDL-C concentrations of the eight familial hypercholesterolemia patients in Example 19 as measured during the course of their treatment with gemcabene calcium salt hydrate Crystal Form 1 having a particle size distribution characterized by a PSD90 of 52 ⁇ m as measured by laser light diffraction (gemcabene calcium salt hydrate Crystal Form 1 300-mg strength film-coated tablet, Tablet D).
  • FIG. 44 is a line graph showing values for percent change from baseline of LDL-C concentrations of the three familial hypercholesterolemia patients, who were determined to have homozygous familial hypercholesterolemia (HoFH) genotype based on post-trial genetic assessment, as measured during the course of their treatment with gemcabene calcium salt hydrate Crystal Form 1 having a particle size distribution characterized by a PSD90 of 52 ⁇ m as measured by laser light diffraction (gemcabene calcium salt hydrate Crystal Form 1 300-mg strength film-coated tablet, Tablet D).
  • HoFH homozygous familial hypercholesterolemia
  • FIG. 45 is a line graph showing values for percent change from baseline of LDL-C concentrations of the three familial hypercholesterolemia patients, who were determined to have heterozygous familial hypercholesterolemia (HeFH) genotype based on post-trial genetic assessment, as measured during the course of their treatment with gemcabene calcium salt hydrate Crystal Form 1 having a particle size distribution characterized by a PSD90 of 52 ⁇ m as measured by laser light diffraction (gemcabene calcium salt hydrate Crystal Form 1 300-mg strength film-coated tablet, Tablet D).
  • HeFH heterozygous familial hypercholesterolemia
  • LS least square
  • LS least square
  • FIG. 52A is a X-ray powder diffractogram of amorphous gemcabene calcium salt.
  • FIG. 52B is an overlay of a thermogravimetric analysis (TGA) thermogram and differential thermal analysis (DTA) thermogram of amorphous gemcabene calcium salt.
  • TGA thermogravimetric analysis
  • DTA differential thermal analysis
  • FIG. 52C is a differential scanning calorimetry (DSC) thermogram of amorphous gemcabene calcium salt.
  • FIG. 53A is a X-ray powder diffractogram of gemcabene calcium salt Crystal Form 2.
  • FIG. 53B is an overlay of a thermogravimetric analysis (TGA) thermogram and differential thermal analysis (DTA) thermogram of gemcabene calcium salt Crystal Form 2.
  • TGA thermogravimetric analysis
  • DTA differential thermal analysis
  • FIG. 54A is a X-ray powder diffractogram of gemcabene calcium salt Crystal Form C3.
  • FIG. 54B is an overlay of a thermogravimetric analysis (TGA) thermogram and differential thermal analysis (DTA) thermogram of gemcabene calcium salt Crystal Form C3.
  • TGA thermogravimetric analysis
  • DTA differential thermal analysis
  • FIG. 54C is a differential scanning calorimetry (DSC) thermogram of gemcabene calcium salt Crystal Form C3.
  • FIG. 55A is a X-ray powder diffractogram of crystalline gemcabene calcium salt ethanol solvate.
  • FIG. 55B is an overlay of a thermogravimetric analysis (TGA) thermogram and differential thermal analysis (DTA) thermogram of crystalline gemcabene calcium salt ethanol solvate.
  • TGA thermogravimetric analysis
  • DTA differential thermal analysis
  • the present invention provides compounds of the invention.
  • the compound of the invention is gemcabene calcium salt.
  • the compound of the invention is gemcabene calcium salt hydrate.
  • the compound of the invention is amorphous or crystalline pharmaceutically acceptable salt of gemcabene.
  • Gemcabene has been previously described, e.g., in U.S. Pat. No. 5,648,387, which is hereby incorporated by reference in its entirety.
  • Various gemcabene calcium salt hydrates have been previously described, e.g., in U.S. Pat. No. 6,861,555, which is hereby incorporated by reference in its entirety.
  • compositions of the invention further provide compositions of the invention.
  • the compositions of the invention further comprise an additional pharmaceutically active agent.
  • the compositions of the invention further comprise two or more additional pharmaceutically active agents.
  • the compositions of the invention are useful for treating or preventing various diseases including liver disease or an abnormal liver condition, a disorder of lipoprotein or glucose metabolism, a cardiovascular or related vascular disorder, a disease caused by increased levels of fibrosis, or a disease associated with increased inflammation.
  • the invention further provides methods for treating or preventing liver disease or an abnormal liver condition, a disorder of lipoprotein or glucose metabolism, a cardiovascular or related vascular disorder, a disease caused by increased levels of fibrosis, or a disease associated with increased inflammation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • Each of the therapeutic or prophylactic methods disclosed herein is a “therapeutic or prophylactic method of the invention”.
  • a compound of the invention has a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 35 ⁇ m to about 85 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 35 ⁇ m to about 80 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 35 ⁇ m to about 75 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m.
  • the compound of the invention has a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 45 ⁇ m to 75 ⁇ m. In some embodiments, the compound of the invention has a PSD90 ranging from 50 ⁇ m to 75 ⁇ m.
  • the compounds of the invention have a dissolution profile having a value of at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm. In some embodiments, the compound of the invention has a dissolution profile having a value of at least 85% in no more than 45 minutes. In some embodiments, the compound of the invention has a dissolution profile having a value of at least 90% in no more than 45 minutes.
  • the compounds of the invention have a dissolution profile having a n value of at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 30 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • the compound of the invention is a gemcabene calcium salt. In other embodiments, the compound of the invention is a gemcabene calcium salt hydrate. In some embodiments, the compound of the invention is an amorphous solid. In some embodiments, the compound of the invention is a crystalline polymorph. In some embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form 1. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form 2. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C1. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C2. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C3. In some embodiments, the compound of the invention is an amorphous gemcabene calcium salt. In some embodiments, the compound of the invention is an amorphous gemcabene calcium salt hydrate.
  • the compound of the invention has a water content of about 2% w/w to about 5% w/w of the compound of the invention. In other embodiments, the compound of the invention has the water content of about 2% w/w to about 4% w/w. In some embodiments, the water content is about 3% w/w to about 5% w/w. In other embodiments, the water content is about 3% w/w to about 4% w/w.
  • the compound of the invention is a gemcabene calcium salt solvate. In some embodiments, the compound of the invention is a gemcabene calcium salt alcohol solvate. In some embodiments, the compound of the invention is a gemcabene calcium salt ethanol solvate. In some embodiments, the compound of the invention is a gemcabene calcium salt n-propyl solvate. In some embodiments, the compound of the invention is a gemcabene calcium salt isopropyl solvate. In some embodiments, the compound of the invention is a gemcabene calcium salt methanol solvate. In some embodiments, the compound of the invention is a gemcabene calcium salt n-butyl solvate.
  • the compound of the invention has an ethanol content of about 0% w/w to about 0.5% w/w of the compound of the invention. In some embodiments, the compound of the invention has an ethanol content of about 0.5% w/w to about 8% w/w of the compound of the invention.
  • the composition of the invention is in a form of a tablet or a capsule. In some embodiments, the composition of the invention further comprises an effective amount of an additional pharmaceutically active agent. In other embodiments, the composition of the invention further comprises an effective amount of two or more additional pharmaceutically active agents.
  • the additional pharmaceutically active agent is a statin.
  • the statin is atorvastatin, simvastatin, pravastatin, rosuvastatin, fluvastatin, lovastatin, pitavastatin, mevastatin, dalvastatin, dihydrocompactin, or cerivastatin, or a pharmaceutically acceptable salt thereof.
  • the pharmaceutically acceptable salt of the statin is a calcium salt. In some embodiments, the statin is atorvastatin calcium.
  • additional pharmaceutically active agents include, but are not limited to, a lipid lowering agent, a PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor, a cholesterol absorption inhibitor, an ACC (acetyl-CoA carboxylase) inhibitor, an ApoC-III (apolipoprotein C-III) inhibitor, an ApoB (apolipoprotein B) synthesis inhibitor, an ANGPTL 3 (angiopoietin-like protein 3) inhibitor, an ANGPTL 4 (angiopoietin-like protein 4) inhibitor, an ANGPTL 8 (angiopoietin-like protein 8) inhibitor, an ACL (adenosine triphosphate citrate lyase) inhibitor, a microsomal transfer protein inhibitor, a fenofibric acid, a fish oil, a fibrate, a thyroid hormone beta receptor agonist, a farnesoid X receptor (FXR), a CCR
  • PCSK9
  • the additional pharmaceutically active agent is a contraceptive agent.
  • a “contraceptive agent” refers to any pharmaceutically active agent that promotes the prevention of conception, impregnation, or implantation or prevents or reduces the likelihood of pregnancy.
  • the contraceptive agent is one or both of ethinyl estradiol and norethindrone.
  • the contraceptive agent is a combination of ethinyl estradiol and norethindrone.
  • the contraceptive agent is estrogen, an estrogen derivative, progestin or a progestin derivative.
  • the present invention provides methods for treating or preventing a liver disease or an abnormal liver condition, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • Illustrative liver diseases or abnormal liver conditions include, but are not limited to, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, alcoholic steatohepatitis, cirrhosis, inflammation, fibrosis, partial fibrosis, primary biliary cirrhosis, primary sclerosing cholangitis, liver failure, hepatocellular carcinoma, liver cancer, hepatic steatosis, hepatocyte ballooning, hepatic lobular inflammation, and hepatic triglyceride accumulation.
  • the liver disease or liver condition is nonalcoholic fatty liver disease or nonalcoholic steatohepatitis.
  • the present invention provides methods for treating or preventing an abnormal fibrosis of an internal organ of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the abnormal fibrosis of an internal organ is in a human subject.
  • the present invention provides methods for treating or preventing a disease or an abnormal condition generated by an inflammatory response of an organ in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the inflammatory response is in an internal organ.
  • the subject is a human.
  • the present invention provides methods for treating or preventing a disorder of lipoprotein metabolism, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • disorders of lipoprotein metabolism include, but are not limited to, dyslipidemia, dyslipoproteinemia, mixed dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), type IIb hyperlipidemia, familial combined hyperlipidemia, familial hypercholesterolemia, familial chylomicronemia syndrome, hypertriglyceridemia, dysbetalipoproteinemia, lipoprotein overproduction, lipoprotein deficiency, elevation of total cholesterol, elevation of low-density lipoprotein cholesterol concentration, elevation of very low-density lipoprotein cholesterol concentration, elevation of non-HDL cholesterol concentration, elevation of apolipoprotein B concentration, elevation of apolipoprotein C-III concentration, elevation of C-reactive protein concentration, elevation of fibrinogen concentration, elevation of lipoprotein(a) concentration, elevation of interleukin-6 concentration, elevation of angiopo
  • the disorder of lipoprotein metabolism is mixed dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), type IIb hyperlipidemia, or familial combined hyperlipidemia. In some embodiments, the disorder of lipoprotein metabolism is familial hypercholesterolemia.
  • the present invention provides methods for reducing a subject's total cholesterol, low-density lipoprotein cholesterol concentration, very low-density lipoprotein cholesterol concentration, non-HDL cholesterol concentration, apolipoprotein B concentration, apolipoprotein C-III concentration, C-reactive protein concentration, fibrinogen concentration, lipoprotein(a) concentration, interleukin-6 concentration, angiopoietin-like protein 3 concentration, angiopoietin-like protein 4 concentration, serum amyloid A concentration, PCSK9 concentration, low-density lipoprotein concentration, very low-density lipoprotein concentration, or triglyceride concentration, comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a subject's triglyceride concentration or LDL-cholesterol, comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a subject's cholesterol-rich remnant ApoB-lipoprotein or triglyceride-rich remnant ApoB-lipoprotein concentration in the subject's blood serum or plasma, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a subject's cholesterol- and triglyceride-rich remnant ApoB-lipoproteins (C-TRLs) in the subject's plasma, comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • the present invention provides methods for increasing hepatic clearance of cholesterol-rich remnant ApoB-lipoprotein or triglyceride-rich remnant ApoB-lipoprotein in a subject, comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • the present invention provides methods for enhancing or increasing hepatic clearance of C-TRLs in a subject, comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • fast hepatic clearance of C-TRLs lead to less cholesterol deposition (less plaque buildup) in arteries.
  • increasing hepatic clearance of cholesterol-rich remnant ApoB-lipoprotein, triglyceride-rich remnant ApoB-lipoprotein, or C-TRLs can be useful in treating or preventing cardiovascular diseases including atherosclerosis.
  • the present invention provides methods for reducing a subject's risk of thrombosis or blood clot, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for treating or preventing a disorder of glucose metabolism, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • disorders of glucose metabolism include, but are not limited to, insulin resistance, impaired glucose tolerance, impaired fasting glucose (concentration in blood), diabetes mellitus, familial partial lipodystrophy, lipodystrophy, obesity, peripheral lipoatrophy, diabetic nephropathy, diabetic retinopathy, renal disease, and septicemia.
  • obesity is central obesity.
  • the present invention provides methods for treating or preventing an atherometabolic syndrome, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a subject's risk of developing an atherometabolic syndrome, comprising administering to a subject in need thereof, an effective amount of a compound of the invention.
  • Atherometabolic syndrome like type 2 diabetes, increases plasma levels of cholesterol- and triglyceride-rich remnant ApoB-lipoproteins (C-TRLs).
  • atherometabolic syndrome includes metabolic syndrome, which can be defined by a cluster of symptoms that include abdominal obesity, impaired glucose tolerance, dyslipidemia, and raised blood pressure.
  • atherometabolic syndrome includes one or more conditions associated with increased risk of cardiovascular disease or one or more conditions associated with increased blood pressure, increased LDL-C, lowered HDL-C, and/or increased blood sugar level.
  • the present invention provides methods for treating or preventing a cardiovascular disorder or a related vascular disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • cardiovascular disorders or related vascular disorders include, but are not limited to, arteriosclerosis, atherosclerosis, hypertension, coronary artery disease, myocardial infarction, arrhythmia, atrial fibrillation, heart valve disease, heart failure, cardiomyopathy, myopathy, pericarditis, impotence, and a thrombotic disorder.
  • the present invention provides methods for treating or preventing a C-reactive protein-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the C-reactive protein related disorder is inflammation, ischemic necrosis, or a thrombotic disorder.
  • the present invention provides methods for treating or preventing disorders related to modulating inflammation markers or C-reactive proteins, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the disorder related to modulating inflammation markers or C-reactive proteins is inflammation, ischemic necrosis, or a thrombotic disorder.
  • the present invention provides methods for treating or preventing Alzheimer's disease, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for treating or preventing Parkinson's disease, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for treating or preventing pancreatitis, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for preventing or reducing the risk of developing pancreatitis, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for treating or preventing pulmonary disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the pulmonary disorder is chronic obstructive pulmonary disease or an idiopathic pulmonary fibrosis.
  • the present invention provides methods for treating or preventing musculoskeletal discomfort, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the musculoskeletal discomfort is myalgia.
  • the musculoskeletal discomfort is myositis.
  • the present invention provides methods for treating or preventing a sulfatase-2-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the sulfatase-2-related disorder is a hepatic sulfatase-2-related disorder.
  • the sulfatase-2-related disorder is a disorder of lipogenesis or lipid modulation.
  • disorders of lipogenesis include, but are limited to, diabetes and related conditions, obesity, hepatic steatosis, non-alcoholic steatohepatitis, cancer, cardiovascular disease (hypertriglyceridemia), and skin disorders.
  • disorders of lipid modulation include, but are not limited to, elevated total cholesterol, elevated low-density lipoprotein cholesterol (LDL-C), elevated apolipoprotein B (Apo B), elevated triglyceride and elevated non-high-density lipoprotein cholesterol.
  • LDL-C low-density lipoprotein cholesterol
  • Apo B elevated apolipoprotein B
  • triglyceride elevated non-high-density lipoprotein cholesterol
  • the present invention provides methods for downregulating hepatic sulfatase-2 expression in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for treating or preventing an ApoC-III related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the ApoC-Ili related disorder is a disorder of lipogenesis or lipid modulation, described herein.
  • the present invention provides methods for treating or preventing an ACC1-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the ACC1-related disorder is a disorder of lipogenesis or lipid modulation, described herein.
  • the present invention provides methods for treating or preventing an ADH-4-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the ADH-4-related disorder is a disorder of lipogenesis or lipid modulation, described herein.
  • the present invention provides methods for treating or preventing a TNF- ⁇ -related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the TNF- ⁇ -related disorder is inflammation.
  • the present invention provides methods for treating or preventing a MCP-1-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the MCP-1-related disorder is inflammation.
  • the present invention provides methods for treating or preventing a MIP-1 ⁇ -related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the MIP-1 ⁇ -related disorder is inflammation.
  • the present invention provides methods for treating or preventing a CCR5-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the CCR5-related disorder is inflammation.
  • the present invention provides methods for treating or preventing a CCR2-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the CCR2-related disorder is inflammation.
  • the present invention provides methods for treating or preventing a NF- ⁇ B-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the NF- ⁇ B-related disorder is inflammation.
  • the present invention provides methods for treating or preventing a TIMP-1-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the TIMP-1-related disorder is fibrosis.
  • the fibrosis is hepatic fibrosis.
  • the present invention provides methods for treating or preventing a MMP-2-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the MMP-2-related disorder is hepatic carcinogenesis or cancer.
  • the therapeutic or prophylactic methods of the invention further comprise administering an effective amount of an additional pharmaceutically active agent. In some embodiments, the therapeutic or prophylactic methods of the invention further comprise administering an effective amount of two or more additional pharmaceutically active agent.
  • the additional pharmaceutically active agent is a statin. In some embodiments, the statin is atorvastatin, simvastatin, pravastatin, rosuvastatin, fluvastatin, lovastatin, pitavastatin, mevastatin, dalvastatin, dihydrocompactin, or cerivastatin or a pharmaceutically acceptable salt thereof. In some embodiments, the statin is atorvastatin calcium.
  • additional pharmaceutically active agents are as disclosed herein.
  • the additional pharmaceutically active agent is a human hormone FGF19.
  • a numerical value means ⁇ up to 20% of the numerical value, in some embodiments, ⁇ up to 19%, ⁇ up to 18%, ⁇ up to 17%, ⁇ up to 16%, ⁇ up to 15%, ⁇ up to 14%, ⁇ up to 13%, ⁇ up to 12%, ⁇ up to 11%, ⁇ up to 10%, ⁇ up to 9%, ⁇ up to 8%, ⁇ up to 7%, ⁇ up to 6%, ⁇ up to 5%, ⁇ up to 4%, ⁇ up to 3%, ⁇ up to 2%, ⁇ up to 1%, ⁇ up to less than 1%, or any other value or range of values therein.
  • a “subject” is a human or non-human mammal, e.g., a bovine, horse, feline, canine, rodent, or non-human primate.
  • the human can be a male or female, child, adolescent or adult.
  • the female can be premenarcheal or postmenarcheal.
  • the “gemcabene” (United States Adopted Name) has the chemical name 6-(5-carboxy-5-methyl-hexyloxy)-2,2-dimethyl-hexanoic acid, which is also known as 6-(5-carboxy-5-methyl-hexyloxy)-2,2-dimethylhexanoic acid or 6,6′-oxybis(2,2-dimethylhexanoic acid), and has the structure:
  • Illustrative pharmaceutically acceptable salts of a basic compound include those of an inorganic or organic acid, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, camphorsulfonic acid, oxalic acid, maleic acid, succinic acid, citric acid, formic acid, hydrobromic acid, benzoic acid, tartaric acid, fumaric acid, salicylic acid, mandelic acid, or carbonic acid.
  • an inorganic or organic acid for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, camphorsulfonic acid, oxalic acid, maleic acid, succinic acid, citric acid, formic acid, hydrobromic acid, benzoic acid, tartaric acid, fumaric acid, salicylic acid, mandelic acid, or carbonic acid.
  • examples of inorganic or organic acids suitable to form an acid addition salt include but are not limited to, hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, camphorsulfonic acid, oxalic acid, maleic acid, succinic acid, citric acid, formic acid, hydrobromic acid, benzoic acid, tartaric acid, fumaric acid, salicylic acid, mandelic acid, carbonic acid, etc.
  • Illustrative pharmaceutically acceptable salts of an acidic compound, e.g., gemcabene include alkali metal salts, (e.g., lithium, sodium and potassium salts), alkaline earth metal salts (e.g., calcium and magnesium salts), aluminum salts, ammonium salts, and salts with organic amines such as benzathine (N,N′-dibenzylethylenediamine), choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), benethamine (N-benzylphenethylamine), diethylamine, piperazine, tromethamine (2-amino-2-hydroxymethyl-1,3-propanediol) and procaine.
  • alkali metal salts e.g., lithium, sodium and potassium salts
  • alkaline earth metal salts e.g., calcium and magnesium salts
  • aluminum salts e.g., ammonium salts
  • a pharmaceutically acceptable salt derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Pharmaceutically acceptable salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine,
  • an “effective amount” when used in connection with a compound of the invention means an amount of the compound of the invention that, when administered to a subject for treating or preventing a disorder or abnormal condition, is effective to treat or prevent the disorder or abnormal condition, alone or in combination with an additional pharmaceutically active agent.
  • an “effective amount” when used in connection with an additional pharmaceutically active agent means an amount of the additional pharmaceutically active agent that, when administered to a subject for treating or preventing a disorder or abnormal condition, is effective to treat or prevent the disorder or abnormal condition, alone or in combination with a compound of the invention.
  • D 90 or “PSD90”, means that 90% of the particles of a compound of the invention have a diameter that is less than the indicated diameter.
  • a D 90 or a PSD90 of 75 ⁇ m means that 90% of the cumulative volume of the particles of the indicated compound of the invention have a diameter that is less than 75 ⁇ m.
  • D 90 ” or “PSD50” means that 50% of the cumulative volume of the particles of a compound of the invention have a diameter that is less than the indicated diameter.
  • D 10 or “PSD10” means that 10% of the cumulative volume of the particles of a compound of the invention have a diameter that is less than the indicated diameter.
  • an “immediate-release” composition refers to a composition of the invention that releases at least 75% (by weight) of a compound of the invention within one hour of administration to a subject. In some embodiments, an immediate-release composition of the invention releases at least 75% by weight, at least 80% by weight, at least 85% by weight, or at least 90% by weight of a compound of the invention within 45 minutes of administration to a subject.
  • AUC (0-24) refers to area under the plasma concentration-time curve from time 0 to 24 hours following a compound's administration.
  • AUC last which is synonymous with “AUC (0-tldc) ”, “AUC (0-tlqc) ”, “AUC (0-tc) ”, and “AUC (0-t) ”, refers to area under the plasma concentration-time curve from time 0 to the last detectable concentration of a compound following its administration.
  • baseline plasma or blood serum LDL-C refers to plasma or blood serum LDL-C of a subject as measured prior to administration of the compound of the invention.
  • a subject “on a stable dose” of a lipid-lowering medication, drug or agent, such as a statin refers to a subject that has been taking the same dose of lipid-lowering medication (e.g., statins) for a period of time in which the subject's blood serum or plasma concentration of LDL-C has stabilized.
  • lipid-lowering medication e.g., statins
  • stabilized means that a new steady state level of LDL-C in the subject's blood serum or plasma concentration has been achieved at a time after beginning the lipid-lowering medication and remains relatively constant from day today within reasonable margins ( ⁇ 15%) of the new steady state level.
  • a “statin therapy” refers to a treatment where a subject is administered a statin.
  • the subject is “undergoing statin therapy”, i.e., being administered with a statin.
  • the stain therapy is maximally tolerated statin therapy.
  • the statin therapy is ineffective to treat or prevent a disease or condition as disclosed herein.
  • the statin therapy is ineffective to lower the subject's LDL-C concentration, lower the subject's triglyceride concentration, or raise the subject's HDL-C concentration to a normal value or to the subject's goal value.
  • maximally tolerated statin therapy refers to therapeutic regimen comprising the administration of daily dose of a statin that is the maximally tolerated dose for a particular subject. “Maximally tolerated dose” means the highest dose of statin that can be administered to a subject without causing unacceptable adverse side effects in the subject.
  • a subject with homozygous familial hypercholesterolemia or “an HoFH subject” is a subject determined to have HoFH by genetic confirmation or clinical diagnosis.
  • a subject with HoFH (1) has a genetic confirmation of two mutant alleles at the LDL-receptor, apolipoprotein B, PCSK9 or the LDL-RAP1 (LDL-receptor adaptor protein 1) gene locus.
  • the subject may have paired or same (homozygous) or two unpaired or dissimilar (compound homozygous or compound heterozygous) mutations at alleles on the LDL-receptor, apolipoprotein B, PCSK9, or the LDL-RAP1 gene locus; or (2) is clinically determined to have (a) untreated LDL-C>500 mg/dL (12.92 mmol/L) or treated LDL-C ⁇ 300 mg/dL (7.76 mmol/L) together with either appearance of cutaneous or tendinous xanthoma before 10 years of age, or evidence of heterozygous familial hypercholesterolemia in both parents, or (b) LDL-C>300 mg/dL (7.76 mmol/L) on maximally tolerated lipid-lowering drug therapy.
  • the clinically diagnosis is only indicative of HoFH, but there are some subjects that does not meet the clinical LDL-C limitations (e.g., subjects have LDL-C ⁇ 500 mg/dL or LDL-C ⁇ 300 mg/dL) yet have HoFH by genetic confirmation. Similarly, subjects can be clinically diagnosed as having HoFH but not by genetic confirmation.
  • a subject with heterozygous familial hypercholesterolemia or “an HeFH subject” is a subject determined to have HeFH by genetic confirmation or clinical diagnosis.
  • a subject with HeFH is clinically determined to have LDL-C ⁇ 190 mg/dL.
  • Genotype analysis for each of four genes is not commonly conducted as the analysis is lengthy, expensive and interpretations of results are controversial.
  • polymorphic changes in DNA that result in a single amino acid or small changes may result in little or no functional change in the protein, but this genetic variation is considered a “mutation” or “varian” of the predominant gene in the population.
  • the loose interpretation of functional activity does not allow precision in genetic classification.
  • other genetic and environmental factors result in phenotypic variation.
  • familial hypercholesterolemia and more specifically homozygous familial hypercholesterolemia, is generally based on clinical interpretation. The clinical interpretation is sometimes supported by follow-up gene sequence analysis for both alleles of the LDL-receptor, apolipoprotein B, PCSK9 and LDL-RAP1 for the subject and if feasible the parents, siblings, and other relatives.
  • the PSD90 of the compounds of the invention is achieved by reducing the particles' size, e.g., by micronizing or milling.
  • the micronizing or milling is achieved using a pinmill.
  • the micronizing or milling is achieved using a Fitzmill.
  • the compounds of the invention have a PSD90 ranging from 35 ⁇ m to about 90 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 36 ⁇ m to about 90 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 37 ⁇ m to about 90 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 38 ⁇ m to about 90 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 39 ⁇ m to about 90 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 40 ⁇ m to about 90 ⁇ m.
  • the compounds of the invention have a PSD90 ranging from 35 ⁇ m to about 85 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 36 ⁇ m to about 85 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 37 ⁇ m to about 85 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 38 ⁇ m to about 85 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 39 ⁇ m to about 85 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 40 ⁇ m to about 85 ⁇ m.
  • the compounds of the invention have a PSD90 ranging from 35 ⁇ m to about 80 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 36 ⁇ m to about 80 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 37 ⁇ m to about 80 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 38 ⁇ m to about 80 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 39 ⁇ m to about 80 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 40 ⁇ m to about 80 ⁇ m.
  • the compounds of the invention have a PSD90 ranging from 35 ⁇ m to about 75 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 36 ⁇ m to about 75 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 37 ⁇ m to about 75 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 38 ⁇ m to about 75 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 39 ⁇ m to about 75 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m.
  • the compounds of the invention have a PSD90 ranging from 45 ⁇ m to about 90 ⁇ m. In other embodiments, the compounds of the invention have a PSD90 ranging from 45 ⁇ m to about 85 ⁇ m. In other embodiments, the compounds of the invention have a PSD90 ranging from 45 ⁇ m to about 80 ⁇ m. In other embodiments, the compounds of the invention have a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m.
  • the compounds of the invention have a PSD90 ranging from 50 ⁇ m to about 90 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 50 ⁇ m to about 85 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 50 ⁇ m to about 80 ⁇ m. In some embodiments, the compounds of the invention have a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the compounds of the invention have a PSD90 of 35 ⁇ m, 36 ⁇ m, 37 ⁇ m, 38 ⁇ m, 39 ⁇ m, 40 ⁇ m, 41 ⁇ m, 42 ⁇ m, 43 ⁇ m, 44 ⁇ m, 45 ⁇ m, 46 ⁇ m, 47 ⁇ m, 48 ⁇ m, 49 ⁇ m, 50 ⁇ m, 51 ⁇ m, 52 ⁇ m, 53 ⁇ m, 54 ⁇ m, 55 ⁇ m, 56 ⁇ m, 57 ⁇ m, 58 ⁇ m, 59 ⁇ m, 60 ⁇ m, 61 ⁇ m, 62 ⁇ m, 63 ⁇ m, 64 ⁇ m, 65 ⁇ m, 66 ⁇ m, 67 ⁇ m, 68 ⁇ m, 69 ⁇ m, 70 ⁇ m, 71 ⁇ m, 72 ⁇ m, 73 ⁇ m, 74 ⁇ m, 75 ⁇ m, 76 ⁇ m, 77 ⁇ m, 78 ⁇ m, 79
  • the compounds of the invention have a PSD90 of about 44 ⁇ m, about 45 ⁇ m, about 46 ⁇ m, about 47 ⁇ m, about 48 ⁇ m, about 49 ⁇ m, about 50 ⁇ m, about 51 ⁇ m, about 52 ⁇ m, about 53 ⁇ m, about 54 ⁇ m, about 55 ⁇ m, about 56 ⁇ m, about 57 ⁇ m, about 58 ⁇ m, about 59 ⁇ m, about 60 ⁇ m, about 61 ⁇ m, about 62 ⁇ m, about 63 ⁇ m, about 64 ⁇ m, about 65 ⁇ m, about 66 ⁇ m, about 67 ⁇ m, about 68 ⁇ m, about 69 ⁇ m, about 70 ⁇ m, about 71 ⁇ m, about 72 ⁇ m, about 73 ⁇ m, about 74 ⁇ m, about 75 ⁇ m, about 76 ⁇ m, about 77 ⁇ m, about 78 ⁇ m, about 79 ⁇ m, about 80 ⁇ m,
  • the compounds of the invention having a PSD90 of about 50 ⁇ m to about 62 ⁇ m particularly enable compressed tablet formulation with desired properties such as high drug loading, good compressibility, fast dissolution profile, and minimal to no cracking.
  • the particle size distribution and the PSD90 of a compound of the invention is determined by the laser light diffraction particle size distribution analysis.
  • the particle size distribution is determined in accordance with the Fraunhofer light diffraction method. In this method, a coherent laser beam passes through the sample and the resulting diffraction pattern is focused on a multi-element detector. Since the diffraction pattern depends, among other parameters, on particle size, the particle size distribution can be calculated based on the measured diffraction pattern of the sample. The method is described in more detail in USP38-NF33, ⁇ 429> Light Diffraction Measurement of Particle Size.
  • the compound of the invention has a dissolution profile characterized by its (% dissolution) over time.
  • the dissolution profile can have a (% dissolution) value of at least 80% in 45 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • the compound of the invention is a calcium salt.
  • the calcium salt is a calcium salt hydrate.
  • the compound of the invention is an amorphous solid.
  • the compound of the invention is a crystalline polymorph.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form 1.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form 2.
  • the compound of the invention is gemcabene calcium salt hydrate Crystal Form C3.
  • the compound of the invention is gemcabene calcium salt hydrate Crystal Form C2.
  • the compound of the invention is gemcabene calcium salt hydrate Crystal Form C1.
  • the compound of the invention is a calcium salt solvate.
  • the calcium salt solvate is a calcium salt ethanol solvate.
  • a compound of the invention has a dissolution profile characterized by % dissolution value of at least 85% gemcabene in 45 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. and as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • a compound of the invention has a dissolution profile characterized by % dissolution value of at least 90% gemcabene in 45 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. and as measured by high-performance liquid chromatography using a detection wavelength of 210 nm. See Example 13 for detailed method of determining % dissolution values.
  • a compound of the invention has a dissolution profile characterized by % dissolution value of at least 80%, at least 81%, at least 82%, at least 83%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95%, or any value ranging from these percentages (e.g., 85%-90% dissolution), in 45 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. and as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • the compound of the invention has a dissolution profile characterized by % dissolution value of at least 70% in 30 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • the pharmaceutically acceptable salt is a calcium salt.
  • the calcium salt is a calcium salt hydrate.
  • the compound of the invention is an amorphous solid.
  • the compound of the invention is a crystalline polymorph.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form 1.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form 2.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form C3.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form C2.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form C1.
  • TGA thermogravimetric analysis
  • DTA differential thermal analysis
  • DSC differential scanning calorimetry
  • GC gas chromatography
  • KF Karl-Fisher
  • HPLC/CAD high-performance liquid chromatography with charged aerosol detector
  • PSD particle size distribution
  • a compound of the invention has a dissolution profile characterized by % dissolution value of at least 85% in 45 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. as measured by high-performance liquid chromatography using a detection wavelength of 210 nm. In some embodiments, a compound of the invention has a dissolution profile characterized by % dissolution value of at least 90% gemcabene in 45 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • a compound of the invention has a dissolution profile characterized by % dissolution value of, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, or at least 75%, or a value ranging from and to any of these percentages, in 30 minutes or less in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • a compound of the invention comprises an amorphous form or a crystalline form of gemcabene or a pharmaceutically acceptable salt thereof having a dissolution profile comprising a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm and (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 30 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • the present invention further provides a pharmaceutically acceptable salt of gemcabene, the pharmaceutically acceptable salt having (a) a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m as measured by laser light diffraction and (b) a dissolution profile characterized by a % dissolution value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 30 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm.
  • the dissolution profile is measured using the composition of the invention. In some embodiments, the dissolution profile of a compound of the invention is measured using a composition of the invention that is in the form of a tablet. In some embodiments, the tablet is a compressed tablet. In some embodiment, the compressed tablet is a film-coated compressed tablet.
  • the dissolution profile of a compound of the invention is measured using a composition of the invention that is in the form of a capsule.
  • the compound of the invention has a water content of about 1% w/w to about 6% w/w of the compound of the invention. In some embodiments, the compound of the invention has a water content of about 2% w/w to about 5% w/w of the compound of the invention. In some embodiments, the water content of the compound of the invention is about 2% w/w to about 5%, about 2% w/w to about 4% w/w, about 3% w/w to about 5% w/w, or about 3% w/w to about 4% w/w of the compound of the invention, or a value ranging from and to any of these percent by weight values.
  • the water content of the compound of the invention is about 2.0%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, about 3.0%, about 3.1%, about 3.2%, about 3.3%, about 3.4%, about 3.5%, about 3.6%, about 3.7%, about 3.8%, about 3.9%, about 4.0%, about 4.1%, about 4.2%, about 4.3%, about 4.4%, about 4.5%, about 4.6%, about 4.7%, about 4.8%, about 4.9%, or about 5.0% by weight of the compound of the invention. In other embodiments, the water content of the compound of the invention is about 3.4%, about 3.5%, about 3.6%, or about 3.7% by weight of the compound of the invention.
  • the compound of the invention has an ethanol content of about 0% w/w to about 0.5% w/w of the compound of the invention. In some embodiments, the ethanol content of the compound of the invention is about 0.0%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, or about 0.5% by weight of the compound of the invention.
  • a compound of the invention has an ethanol content that is less than about 5000 ppm of the compound of the invention. In some embodiments, a compound of the invention has an ethanol content that is less than about 4000 ppm of the compound of the invention. In some embodiments, a compound of the invention has an ethanol content that is less than about 3000 ppm of the compound of the invention. In some embodiments, a compound of the invention has an ethanol content that is less than about 2000 ppm of the compound of the invention.
  • the ethanol content is less than about 500 ppm, less than about 600 ppm, less than about 700 ppm, less than about 800 ppm, less than about 900 ppm, less than about 1000 ppm, less than about 1100 ppm, less than about 1200 ppm, less than about 1300 ppm, less than about 1400 ppm, less than about 1500 ppm, less than about 1600 ppm, less than about 1700 ppm, less than about 1800 ppm, less than about 1900 ppm, or less than about 2000 ppm, of the compound of the invention.
  • the compound of the invention has an ethanol content of about 0.5% w/w to about 8% w/w of the compound of the invention. In some embodiment, the compound of the invention is an ethanol solvate having an ethanol content of about 0.5% w/w to about 8% w/w of the compound of the invention.
  • the ethanol content of the compound of the invention is about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0% by, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, 2.0%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, about 3.0%, about 3.1%, about 3.2%, about 3.3%, about 3.4%, about 3.5%, about 3.6%, about 3.7%, about 3.8%, about 3.9%, about 4.0%, about 4.1%, about 4.2%, about 4.3%, about 4.4%, about 4.5%, about 4.6%, about 4.7%, about 4.8%, about 4.9%, about 5.0%, about 5.1%, about 5.2%, about 5.3%, about 5.4%, about 5.5%, about 5.6%, about 5.7%, about 5.8%, about 5.9%, about 6.0%, about 6.1%, about 6.2%, about 6.3%, about 6.4%, about 5.5%
  • a compound of the invention has an ethanol content is about 20,000 ppm to about 40,000 ppm of the compound of the invention. In some embodiments, a compound of the invention is an ethanol solvate having an ethanol content is about 20,000 ppm to about 40,000 ppm of the compound of the invention.
  • a compound of the invention has an ethanol content that is about 20,000 ppm, about 21,000 ppm, about 22,000 ppm, about 23,000 ppm, about 24,000 ppm, about 25,000 ppm, about 26,000 ppm, about 27,000 ppm, about 28,000 ppm, about 29,000 ppm, about 30,000 ppm, about 31,000 ppm, about 32,000 ppm, about 33,000 ppm, about 34,000 ppm, about 35,000 ppm, about 36,000 ppm, about 37,000 ppm, about 38,000 ppm, about 39,000 ppm, about 40,000 ppm of the compound of the invention.
  • a compound of the invention has an ethanol content that is about 28,000 ppm, about 28,100 ppm, about 28,200 ppm, about 28,300 ppm, about 28,400 ppm, about 28,500 ppm, about 28,600 ppm, about 28,700 ppm, about 28,800 ppm, or about 28,900 ppm of the compound of the invention.
  • a steady state plasma concentration of gemcabene in a subject is achieved within about 5-20 days following the start of repeated dose administration of the compound of the invention or following increase in daily dosing of the compound of the invention. In some embodiments, a steady state plasma concentration of gemcabene in a subject is achieved within about 14 days following the start of repeated dose administration of the compound of the invention or following increase in daily dosing of the compound of the invention. In some embodiments, the steady state is achieved within 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 days following the start of daily administration of the compound of the invention at a dose of about 50 mg/day to about 900 mg/day or following the increase in daily dose of the compound of the invention to a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. 5° C. in no more than 30 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm, and providing a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides pharmaceutically acceptable salts of gemcabene, the pharmaceutically acceptable salts having (a) a particle size distribution characterized by a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m as measured by laser light diffraction (b) a dissolution profile characterized by a % dissolution value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 250 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 250 ⁇ g ⁇ hr/mL at steady state to about 5750 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 300 ⁇ g ⁇ hr/mL at steady state to about 5500 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 200 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject in an amount that is molar equivalent to about 50 mg of gemcabene per day to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 250 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject in an amount that is molar equivalent to about 50 mg of gemcabene per day to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 250 ⁇ g ⁇ hr/mL at steady state to 5750 ⁇ g ⁇ hr/mL at steady state when administered to a human subject in an amount that is molar equivalent to about 50 mg of gemcabene per day to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 300 ⁇ g ⁇ hr/mL at steady state to 5500 ⁇ g ⁇ hr/mL at steady state when administered to a human subject in an amount that is molar equivalent to about 50 mg of gemcabene per day to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) of about 200 ⁇ g ⁇ hr/mL, about 250 ⁇ g ⁇ hr/mL, about 300 ⁇ g ⁇ hr/mL, about 350 ⁇ g ⁇ hr/mL, about 400 ⁇ g ⁇ hr/mL, about 450 ⁇ g ⁇ hr/mL, about 500 ⁇ g ⁇ hr/mL, about 550 ⁇ g ⁇ hr/mL, about 600 ⁇ g ⁇ hr/mL, about 650 ⁇ g ⁇ hr/mL, about 700 ⁇ g ⁇ hr/mL, about 750 ⁇ g ⁇ hr/mL, about 800 ⁇ g ⁇ hr/mL, about 850 ⁇ g ⁇ hr/mL, about 900 ⁇ g ⁇ hr/mL, about 950 ⁇ g ⁇ hr/mL, about 1000 ⁇ g ⁇ hr/mL, about 1100 ⁇ g ⁇ hr/mL, about 1200 ⁇ g ⁇ h
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state or from about 250 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day, about 60 mg/day, about 70 mg/day, about 80 mg/day, about 90 mg/day, about 100 mg/day, about 110 mg/day, about 120 mg/day, about 130 mg/day, about 140 mg/day, about 150 mg/day, about 160 mg/day, about 170 mg/day, about 180 mg/day, about 190 mg/day, about 200 mg/day, about 210 mg/day, about 220 mg/day, about 230 mg/day, about 240 mg/day, about 250 mg/day, about 260 mg/day, about 270 mg/day, about 280 mg/day, about 290
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 200 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state or from 250 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject in an amount that is molar equivalent to about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 1000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day or in an amount that is molar equivalent to about 50 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 500 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day or in an amount that is molar equivalent to about 50 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 300 ⁇ g ⁇ hr/mL at steady state to about 1500 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 150 mg/day or in an amount that is molar equivalent to about 150 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 500 ⁇ g ⁇ hr/mL at steady state to about 1200 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 150 mg/day or in an amount that is molar equivalent to about 150 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 500 ⁇ g ⁇ hr/mL at steady state to about 2500 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 300 mg/day or in an amount that is molar equivalent to about 300 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 1000 ⁇ g ⁇ hr/mL at steady state to about 2000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 300 mg/day or in an amount that is molar equivalent to about 300 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 750 ⁇ g ⁇ hr/mL at steady state to about 3250 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 450 mg/day or in an amount that is molar equivalent to about 450 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 1250 ⁇ g ⁇ hr/mL at steady state to about 3000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 450 mg/day or in an amount that is molar equivalent to about 450 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 1500 ⁇ g ⁇ hr/mL at steady state to about 5000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 600 mg/day or in an amount that is molar equivalent to about 600 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 1500 ⁇ g ⁇ hr/mL at steady state to about 4500 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 600 mg/day or in an amount that is molar equivalent to about 600 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 2000 ⁇ g ⁇ hr/mL at steady state to 4000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 600 mg/day or in an amount that is molar equivalent to about 600 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 3000 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 900 mg/day or in an amount that is molar equivalent to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from 3250 ⁇ g ⁇ hr/mL at steady state to about 5750 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 900 mg/day or in an amount that is molar equivalent to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 500 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose ranging from about 300 mg/day to about 900 mg/day or in an amount that is molar equivalent in a range from about 300 mg to about 900 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 1500 ⁇ g ⁇ hr/mL at steady state to about 5250 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose ranging from about 450 mg/day to about 750 mg/day or in an amount that is molar equivalent in a range from about 450 mg to about 750 mg of gemcabene per day.
  • the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 1500 ⁇ g ⁇ hr/mL at steady state to about 5250 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose ranging from about 500 mg/day to about 700 mg/day or in an amount that is molar equivalent in a range from about 500 mg to about 700 mg of gemcabene per day.
  • the present invention provides compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. 5° C. in no more than 30 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm, and providing a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • the present invention provides a pharmaceutically acceptable salt of gemcabene, the pharmaceutically acceptable salt having (a) a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m as measured by laser light diffraction and (b) a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 150 ⁇ g ⁇ hr/mL to about 5750 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 400 ⁇ g ⁇ hr/mL to about 5500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 500 ⁇ g ⁇ hr/mL to about 5250 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of the compound of the invention in an amount that is molar equivalent to about 50 mg of gemcabene to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 150 ⁇ g ⁇ hr/mL to about 5750 ⁇ g ⁇ hr/mL after a single dose administration of the compound of the invention in an amount that is molar equivalent to about 50 mg of gemcabene to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 400 ⁇ g ⁇ hr/mL to about 5500 ⁇ g ⁇ hr/mL after a single dose administration of the compound of the invention in an amount that is molar equivalent to about 50 mg of gemcabene to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 500 ⁇ g ⁇ hr/mL to about 5250 ⁇ g ⁇ hr/mL after a single dose administration of the compound of the invention in an amount that is molar equivalent to about 50 mg of gemcabene to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 500 ⁇ g ⁇ hr/mL to about 5500 ⁇ g ⁇ hr/mL after a single dose administration of the compound of the invention in an amount that is molar equivalent to about 50 mg of gemcabene to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last of about 50 ⁇ g ⁇ hr/mL, about 100 ⁇ g ⁇ hr/mL, about 150 ⁇ g ⁇ hr/mL, about 200 ⁇ g ⁇ hr/mL, about 250 ⁇ g ⁇ hr/mL, about 300 ⁇ g ⁇ hr/mL, about 350 ⁇ g ⁇ hr/mL, about 400 ⁇ g ⁇ hr/mL, about 450 ⁇ g ⁇ hr/mL, about 500 ⁇ g ⁇ hr/mL, about 550 ⁇ g ⁇ hr/mL, about 600 ⁇ g ⁇ hr/mL, about 650 ⁇ g ⁇ hr/mL, about 700 ⁇ g ⁇ hr/mL, about 750 ⁇ g ⁇ hr/mL, about 800 ⁇ g ⁇ hr/mL, about 850 ⁇ g ⁇ hr/mL, about 900 ⁇ g ⁇ hr/mL, about 950 ⁇ g ⁇ hr/mL
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single administration of about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, 400 mg, about 410 mg, about 420 mg, about 430 mg, about 440 mg, about 450 mg, about 460 mg, about 470 mg, about 480 mg, about 490
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single administration of the compound of the invention in an amount that is molar equivalent to about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, 400 mg, about 410 mg, about 420 mg, about 430 mg, about 440 mg, about 450 mg, about 460
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 750 ⁇ g ⁇ hr/mL after single administration to a human subject at a dose of about 50 mg or in an amount that is molar equivalent to about 50 mg of gemcabene. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 100 ⁇ g ⁇ hr/mL to about 500 ⁇ g ⁇ hr/mL after single administration to a human subject at a dose of about 50 mg or in an amount that is molar equivalent to about 50 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 100 ⁇ g ⁇ hr/mL to about 1250 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 150 mg or in an amount that is molar equivalent to about 150 mg of gemcabene. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 200 ⁇ g ⁇ hr/mL to about 1000 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 150 mg or in an amount that is molar equivalent to about 150 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 500 ⁇ g ⁇ hr/mL to about 2250 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 300 mg or in an amount that is molar equivalent to about 300 mg of gemcabene. In some embodiments, the compound of the invention provides a plasma gemcabene AUC (0-24) ranging from about 750 ⁇ g ⁇ hr/mL to about 2000 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 300 mg or in an amount that is molar equivalent to about 300 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 1000 ⁇ g ⁇ hr/mL to about 4000 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 600 mg or in an amount that is molar equivalent to about 600 mg of gemcabene. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 1500 ⁇ g ⁇ hr/mL to about 3500 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 600 mg or in an amount that is molar equivalent to about 600 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 1750 ⁇ g ⁇ hr/mL to about 3750 ⁇ g ⁇ hr/mL after single administration to a human subject at a dose of about 600 mg or in an amount that is molar equivalent to about 600 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 2500 ⁇ g ⁇ hr/mL to about 6000 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 900 mg or in an amount that is molar equivalent to about 900 mg of gemcabene. In some embodiments, the compound of the invention provides a plasma gemcabene AUC last ranging from about 2750 ⁇ g ⁇ hr/mL to about 5500 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 900 mg or in an amount that is molar equivalent to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 500 ⁇ g ⁇ hr/mL to about 5500 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 300 mg to about 900 mg or in an amount that is molar equivalent to about 300 mg to about 900 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 750 ⁇ g ⁇ hr/mL to about 5000 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 450 mg to about 750 mg or in an amount that is molar equivalent to about 450 mg to about 750 mg of gemcabene.
  • the compound of the invention provides a plasma gemcabene AUC last ranging from about 1000 ⁇ g ⁇ hr/mL to about 4500 ⁇ g ⁇ hr/mL after single dose administration to a human subject at a dose of about 500 mg to about 700 mg or in an amount that is molar equivalent to about 500 mg to about 700 mg of gemcabene.
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum low-density lipoprotein cholesterol (LDL-C) by about 1% to about 80% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 5% to about 75% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • LDL-C blood serum low-density lipoprotein cholesterol
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 10% to about 75% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 15% to about 70% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 11%, at least about 12%, at least about 13%, at least about 14%, at least about 15%, at least about 16%, at least about 17%, at least about 18%, at least about 19%, at least about 20%, at least about 21%, at least about 22%, at least about 23%, at least about 24%, at least about 25%, at least about 26%, at least about 27%, at least about 28%, at least about 29%, at least about 30%, at least about 31%, at least about 32%, at least about 33%, at least about 34%, at least about 35%, at least about 36%, at least about 37%, at least about 38%, at least about
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum total cholesterol by about 1% to about 80%, including all subranges therein, when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 1% to about 80% when administered to a human subject in an amount that is molar equivalent to about 50 mg to about 900 mg gemcabene per day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 5% to about 75%, about 10% to about 75%, or about 15% to about 70%, when administered to a human subject in an amount that is molar equivalent to about 50 mg to about 900 mg gemcabene per day.
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 11%, at least about 12%, at least about 13%, at least about 14%, at least about 15%, at least about 16%, at least about 17%, at least about 18%, at least about 19%, at least about 20%, at least about 21%, at least about 22%, at least about 23%, at least about 24%, at least about 25%, at least about 26%, at least about 27%, at least about 28%, at least about 29%, at least about 30%, at least about 31%, at least about 32%, at least about 33%, at least about 34%, at least about 35%, at least about 36%, at least about 37%, at least about 38%, at least about
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum total cholesterol by about 1% to about 80%, all subranges therein, when administered to a human subject in an amount that is molar equivalent to about 50 mg to about 900 mg gemcabene per day.
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum LDL-C by about 1% to about 80% or by about 1% to about 75% when administered to a human subject in an amount that is molar equivalent to about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, 400 mg, about 410 mg, about 420 mg, about 430 mg, about 440 mg, about
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum apolipoprotein B (Apo B) by about 1% to about 50% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1% to about 40% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1% to about 30% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • Apo B blood serum apolipoprotein B
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 5% to about 30% when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 4
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 11%, at least about 12%, at least about 13%, at least about 14%, at least about 15%, at least about 16%, at least about 17%, at least about 18%, at least about 19%, at least about 20%, at least about 21%, at least about 22%, at least about 23%, at least about 24%, at least about 25%, at least about 26%, at least about 27%, at least about 28%, at least about 29%, at least about 30%, at least about 31%, at least about 32%, at least about 33%, at least about 34%, at least about 35%, at least about 36%, at least about 37%, at least about 38%, at least about 3
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1% to about 50% when administered to a human subject in an amount that is molar equivalent to about 50 mg to about 900 mg gemcabene per day. In some embodiments, the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1% to about 40%, about 1% to about 30%, or about 5% to about 30%, when administered to a human subject in an amount that is molar equivalent to about 50 mg to about 900 mg gemcabene per day.
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 5
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 11%, at least about 12%, at least about 13%, at least about 14%, at least about 15%, at least about 16%, at least about 17%, at least about 18%, at least about 19%, at least about 20%, at least about 21%, at least about 22%, at least about 23%, at least about 24%, at least about 25%, at least about 26%, at least about 27%, at least about 28%, at least about 29%, at least about 30%, at least about 31%, at least about 32%, at least about 33%, at least about 34%, at least about 35%, at least about 36%, at least about 37%, at least about 38%, at least about 3
  • the compound of the invention provides reduction in a human subject's baseline plasma or blood serum Apo B by about 1% to about 50% when administered to a human subject in an amount that is molar equivalent to about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, 400 mg, about 410 mg, about 420 mg, about 430 mg, about 440 mg, about 450 mg, about 460 mg, about 470 mg, about 480 mg, about 490 mg,
  • the present invention provides compounds of the invention having (a) an amorphous form or a crystalline form and (b) a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. 5° C.
  • the present invention provides compounds of the invention having (a) an amorphous form or a crystalline form and (b) a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. 5° C.
  • the present invention provides compounds of the invention having an amorphous form or a crystalline form and providing a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides compounds of the invention having an amorphous form or a crystalline form and providing a plasma gemcabene AUC (0-24) ranging from 200 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention providing a plasma gemcabene AUC (0-24) ranging from about 200 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention providing a plasma gemcabene AUC (0-24) ranging from 200 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides compounds of the invention having an amorphous form or a crystalline form and providing a plasma gemcabene AUC (0-24) ranging from 250 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention providing a plasma gemcabene AUC (0-24) ranging from 250 ⁇ g ⁇ hr/mL at steady state to 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides compounds of the invention having an amorphous form or a crystalline form and providing a plasma gemcabene AUC (0-24) ranging from about 250 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention providing a plasma gemcabene AUC (0-24) ranging from about 250 ⁇ g ⁇ hr/mL at steady state to about 6000 ⁇ g ⁇ hr/mL at steady state when administered to a human subject at a dose of about 50 mg/day to about 900 mg/day.
  • the present invention provides amorphous or crystalline compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides amorphous or crystalline compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides amorphous or crystalline compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides amorphous or crystalline compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • the present invention provides compounds of the invention having (a) an amorphous form or a crystalline form and (b) a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention having a dissolution profile having a value of (1) at least 80% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C. in no more than 45 minutes as measured by high-performance liquid chromatography using a detection wavelength of 210 nm or (2) at least 70% in pH 5.0 potassium acetate buffer at 37° C. ⁇ 5° C.
  • a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • the present invention provides compounds of the invention having an amorphous form or a crystalline form and providing a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • the present invention provides pharmaceutical compositions comprising an amorphous form or a crystalline form of the compounds of the invention providing a plasma gemcabene AUC last ranging from about 50 ⁇ g ⁇ hr/mL to about 7500 ⁇ g ⁇ hr/mL after a single dose administration of about 50 mg to about 900 mg to a human subject.
  • an effective dose of the compound of the invention can be a dose that achieves ⁇ 10% mean reduction in low-density lipoprotein cholesterol (LDL-C) after 4 weeks of treatment. In some embodiments, an effective dose of the compound of the invention can be a dose that achieves ⁇ 15% mean reduction in LDL-C after 4 weeks of treatment. In some embodiments, an effective dose of the compound of the invention can be a dose that achieves ⁇ 5%, ⁇ 6%, ⁇ 7%, ⁇ 8%, ⁇ 9%, ⁇ 10%, ⁇ 11%, ⁇ 12%, ⁇ 13%, ⁇ 14%, or 15% mean reduction in LDL-C after 4 weeks of treatment.
  • LDL-C low-density lipoprotein cholesterol
  • an effective dose of the compound of the invention can be a dose that achieves ⁇ 5%, ⁇ 6%, ⁇ 7%, ⁇ 8%, ⁇ 9%, ⁇ 10%, ⁇ 11%, ⁇ 12%, ⁇ 13%, ⁇ 14%, or 15% mean reduction in LDL-C after 4 weeks of daily administration of the compound of the invention in about 50 mg to about 900 mg per day.
  • the pharmacokinetic values and properties of a compound of the invention is measured with a composition of the invention that is in the form of a tablet.
  • the tablet is a compressed tablet.
  • the compressed tablet is a film-coated compressed tablet.
  • the pharmacokinetic values and properties of a compound of the invention is measured using a composition of the invention that is in the form of a capsule.
  • AUC (0-24) or AUC last of a compound of the invention is measured with a composition of the invention that is in the form of a tablet.
  • the tablet is a compressed tablet.
  • the compressed tablet is a film-coated compressed tablet.
  • AUC (0-24) or AUC last of a compound of the invention is measured using a composition of the invention that is in the form of a capsule.
  • the pharmacokinetic values and properties disclosed herein are in connection with a human subject.
  • the present invention further provides methods for making gemcabene.
  • Gemcabene is useful for making the compounds of the invention.
  • Gemcabene or gemcabene calcium can be prepared by a synthetic process as shown in Scheme 1.
  • Isobutyric acid is converted to an alkali metal salt.
  • isobutyric acid is converted to an alkali metal salt using an alkali metal hydroxide.
  • the alkali metal hydroxide is lithium hydroxide, sodium hydroxide or potassium hydroxide.
  • the alkali metal hydroxide is sodium hydroxide.
  • the alkali metal hydroxide is lithium hydroxide, which converts isobutyric acid to lithium isobutyrate. In some embodiments, the alkali metal hydroxide is sodium hydroxide, which converts isobutyric acid to sodium isobutyrate. In some embodiments, the alkali metal hydroxide is potassium hydroxide, which converts isobutyric acid to potassium isobutyrate.
  • the alkali metal hydroxide is present in an aqueous solution or suspension. In some embodiments, the alkali metal hydroxide is present in an about 30% (w/w) in aqueous solution.
  • the alkali metal salt is sodium hydroxide.
  • the sodium hydroxide is present in an aqueous solution. In some embodiments, the aqueous solution of sodium hydroxide is 30% (w/w).
  • isobutyric acid is converted to an alkali metal salt in the presence of an organic solvent.
  • the organic solvent is a hydrocarbon solvent.
  • the hydrocarbon solvent is benzene, toluene, xylene or an alkane.
  • the alkane is a C5-C12 alkane.
  • the alkane is pentane, hexane or heptane.
  • the alkane is n-pentane, n-hexane or n-heptane.
  • the alkane is n-heptane.
  • heterogeneous azeotropic distillation composition in azeotrope: 12.9% water and 87.1% heptane; b.p. 79.2° C.
  • heterogeneous azeotropic distillation of water is performed at about 100 to about 110° C.
  • heterogeneous azeotropic distillation of water is performed at about 105° C.
  • heterogeneous azeotropic distillation of water is performed at about 900 mbar to about 1100 mbar.
  • heterogeneous azeotropic distillation of water is performed at about 1000 mbar.
  • the removal of water for example, by heterogeneous azeotropic distillation, can be measured by volume.
  • Karl-Fisher analysis can be performed.
  • water, if any, present in the reaction mixture prior to the addition of the enolate-forming base is ⁇ 0.05% w/w of the reaction mixture as determined by Karl-Fisher analysis.
  • water, if any, present in the reaction mixture prior to the addition of the enolate-forming base is 0.05% w/w or less, 0.04% w/w or less, 0.03% w/w or less, 0.02% w/w or less, 0.015% w/w or less, 0.0125% w/w or less, or 0.01% w/w or less of the reaction mixture as determined by Karl-Fisher analysis.
  • water, if any, present in the reaction mixture prior to the addition of the enolate-forming base is less than 0.05% w/w, less than 0.04% w/w, less than 0.03% w/w, less than 0.02% w/w, less than 0.015% w/w, less than 0.0125% w/w, or less than 0.01% w/w of the reaction mixture as determined by Karl-Fisher analysis.
  • the alkali metal salt of isobutyric acid is converted to an enolate using an enolate-forming base.
  • the enolate-forming base is lithium hexamethyldisilazide, lithium diisopropylamide (LDA), lithium tetramethylpiperidide (LiTMP), or lithium diethylamide (LiNEt 2 ).
  • the enolate-forming base is LDA and is prepared in situ using diisopropylamine and an organolithium reagent, such as n-butyllithium, n-hexyllithium or n-heptyllithium.
  • the enolate-forming base is generated in an aprotic solvent. In some embodiments, the enolate-forming base is obtained commercially and is present in an aprotic solvent. In some embodiments, the enolate-forming base is generated in THF or solvent mixture comprising THF. In some embodiments, the enolate-forming base is in THF or solvent mixture comprising THF.
  • the LDA is pre-made and obtained commercially, particularly in view of organolithium reagents' highly pyrogenic properties.
  • the LDA is pre-made.
  • the pre-made LDA is present in solution.
  • the pre-made LDA solution is about 25% w/w to about 30% w/w LDA.
  • the LDA is 28% w/w in heptane/THF/ethylbenzene.
  • the pre-made LDA is present in solution.
  • the pre-made LDA solution is about 1.5M to about 2.5M.
  • the LDA is 2.0M to 2.2M in heptane/THF/ethylbenzene.
  • the addition of the enolate-forming base is performed under anhydrous conditions. In some embodiments, the addition of the enolate-forming base is performed under substantially anhydrous conditions. In some embodiments, the addition of the enolate-forming base is performed under conditions where the water content is ⁇ 0.05% w/w of the reaction mixture as determined by Karl-Fisher analysis.
  • the enolate-forming base is admixed with the alkali metal salt of isobutyric acid to provide an enolate of the alkali metal salt of isobutyric acid.
  • the enolate-forming base can be added to the alkali metal salt of isobutyric acid, or vice versa.
  • the enolate-forming base is LDA
  • the alkali metal salt of isobutyric acid is sodium isobutyrate
  • the LDA is added to the sodium isobutyrate.
  • the enolate-forming base and the alkali metal salt of isobutyric acid are admixed at a temperature ranging from about 10° C. to about 15° C.
  • the reaction mixture is heated at 42° C. ⁇ 2° C. In some embodiments, the reaction mixture is heated at 42° C. ⁇ 2° C. for about 30 minutes to 2 hours. In some embodiments, the reaction mixture is heated at 42° C. ⁇ 2° C. for about 1 hour.
  • the enolate-forming base and the alkali metal salt of isobutyric acid are admixed in the presence of heptane, tetrahydrofuran (THF), or combination thereof. In some embodiments, the enolate-forming base and the alkali metal salt of isobutyric acid are admixed in the presence of n-heptane, tetrahydrofuran (THF), or combination thereof.
  • the enolate of the alkali metal salt of isobutyric acid is admixed with a bis-(4-halobutyl)ether.
  • the enolate can be added to the bis-(4-halobutyl)ether, or vice versa.
  • the bis-(4-halobutyl)ether is bis-(4-chlorobutyl)ether; in some embodiments, the bis-(4-halobutyl)ether is bis-(4-bromobutyl)ether; and in some embodiments, the bis-(4-halobutyl)ether is bis-(4-iodobutyl)ether.
  • about two equivalents of the enolate of the alkali metal salt of isobutyric acid are admixed with a bis-(4-halobutyl)ether. In some embodiments, about two to about three equivalents of the enolate of the alkali metal salt of isobutyric acid are admixed with a bis-(4-halobutyl)ether. In some embodiments, 2.2 to 2.5 equivalents of the enolate of the alkali metal salt of isobutyric acid are admixed with a bis-(4-halobutyl)ether.
  • the bis-(4-halobutyl)ether is added to the enolate dropwise. In some embodiments, the bis-(4-halobutyl)ether is added to the enolate dropwise over about 1 hour to about 5 hours. In some embodiments, the bis-(4-halobutyl)ether is added to the enolate dropwise over about 1 hour to about 4 hours. In some embodiments, the bis-(4-halobutyl)ether is added to the enolate at a temperature ranging from about 40° C. to about 45° C. In some embodiments, the bis-(4-halobutyl)ether is added to the enolate at a temperature ranging from 40° C. to 44° C.
  • the bis-(4-halobutyl)ether is added to the enolate as a solution in THF.
  • the bis-(4-halobutyl)ether is bis-(4-chlorobutyl)ether
  • the enolate is a lithium enolate of sodium isobutyrate
  • the bis-(4-chlorobutyl)ether is added as a solution in THF to the lithium enolate of sodium isobutyrate at a temperature ranging from 40° C. to 44° C.
  • the reaction mixture is allowed to stir a temperature ranging from about 40° C. to about 45° C. In some embodiments, after the addition of the bis-(4-halobutyl)ether, the reaction mixture is allowed to stir at a temperature ranging from 40° C. to 44° C. In some embodiments, after the addition of the bis-(4-halobutyl)ether, the reaction mixture is allowed to stir for about 8 hours to about 30 hours. In some embodiments, after the addition of the bis-(4-halobutyl)ether, the reaction mixture is allowed to stir for at least 10 hours.
  • the reaction mixture is allowed to stir for about 10 hours to about 24 hours. In some embodiments, after the addition of the bis-(4-halobutyl)ether, the reaction mixture is allowed to stir for about 14 hours to about 24 hours.
  • the reaction mixture is allowed to stir at a temperature ranging from 40° C. to 44° C. and until quantitative 1 H NMR analysis indicates ⁇ 5% bis-(4-halobutyl)ether in the reaction mixture (e.g., ⁇ 95% conversion of bis-(4-halobutyl)ether). In some embodiments, after the addition of bis-(4-halobutyl)ether, the reaction mixture is allowed to stir at a temperature ranging from 40° C. to 44° C.
  • the reaction mixture is allowed to stir at a temperature ranging from 40° C. to 44° C. and until 1 H NMR analysis indicates less than 5%, less than 4%, less than 3%, less than 2%, or less than 1.5% bis-(4-halobutyl)ether in the reaction mixture.
  • an aqueous work-up can be performed to extract the gemcabene salt product into an aqueous phase.
  • the aqueous phase can be acidified, for example, with a mineral acid, such as hydrochloric acid.
  • the gemcabene salt converted to gemcabene the gemcabene can be extracted with an organic solvent.
  • Useful organic solvents include heptane, hexane, methyl tetrahydrofuran, toluene, ethyl acetate, butyl acetate, cyclohexane, 2-butanone, and diisopropyl ether.
  • the organic solvent is heptane.
  • the organic solvent is n-heptane.
  • the aqueous phase is extracted multiple times with the organic solvent.
  • the organic solvent used in the extractions after the bis-(4-halobutyl)ether reaction is complete or substantially complete has a temperature ranging from about 40° C. to about 60° C.
  • the organic solvent used in the extractions after the bis-(4-halobutyl)ether reaction is complete or substantially complete has a temperature ranging from about 48° C. to about 54° C. In some embodiments, the extractions are performed at a temperature ranging from about 40° C. to about 60° C. (temperature indicates the temperature of the solvents used in extractions).
  • the organic layer containing gemcabene can be evaporated to substantial dryness.
  • the resultant crude gemcabene can be admixed with water, which can be subsequently evaporated.
  • the water is evaporated at ⁇ 60° C.
  • the further resultant crude gemcabene can be dissolved in an organic solvent, such as heptane, and the organic solution can be washed with water and evaporated to substantial dryness.
  • This process can be repeated one or more times. In some embodiments, the process is performed twice. In some embodiments, the process is performed at least twice.
  • isobutyric acid impurity resulting from, for example, use of more than two equivalents of the enolate of the alkali metal salt of isobutyric acid per equivalent of bis-(4-halobutyl)ether, can be removed by co-distillation with water. Without being bound by theory, it is believed that the isobutyric acid is removed as an azeotrope with water. The presence of isobutyric acid impurity in the crude gemcabene can adversely affect its crystallization and the purity of crystallized gemcabene.
  • co-distillation of water is performed at a temperature ranging from about 100° C. to about 110° C. In some embodiments, co-distillation of water is performed at a temperature ranging from about 100° C. to about 105° C. In some embodiments, co-distillation of water is performed at ambient pressure. In some embodiments, co-distillation of water is performed at reduced pressure. In some embodiments, co-distillation of water is performed at reduced pressure such that co-distillation of water is performed at a temperature in ranging from about 35° C. to about 70° C.
  • co-distillation of water is performed at reduced pressure such that co-distillation of water is performed at a temperature ranging from about 40° C. to about 60° C. In some embodiments, co-distillation of water is performed at about 10 mbar to about 100 mbar.
  • a first co-distillation with water provides crude gemcabene comprising isobutyric acid impurity in 5% w/w or less of the crude gemcabene as determined by ion chromatography. In some embodiments, a first co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 5% w/w or less, 4% w/w or less, 3% w/w or less, 2% w/w or less, or 1% w/w or less of the crude gemcabene as determined by ion chromatography.
  • a first co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in less than 5% w/w, less than 4% w/w, less than 3% w/w, less than 2% w/w, or less than 1% w/w of the crude gemcabene as determined by ion chromatography.
  • a first co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 0.9% w/w or less, 0.8% w/w or less, 0.7% w/w or less, 0.6% w/w or less, or 0.5% w/w or less of the crude gemcabene as determined by ion chromatography.
  • a first co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in less than 0.9% w/w, less than 0.8% w/w, less than 0.7% w/w, less than 0.6% w/w, or less than 0.5% w/w of the crude gemcabene as determined by ion chromatography. In some embodiments, a first co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 0.8% w/w or less of the crude gemcabene as determined by ion chromatography.
  • a second co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 1% w/w or less of the crude gemcabene as determined by ion chromatography. In some embodiments, a second co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 1.0% w/w or less, 0.9% w/w or less, 0.8% w/w or less, 0.7% w/w or less, 0.6% w/w or less, 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, or 0.2% w/w or less of the crude gemcabene as determined by ion chromatography.
  • a second co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in less than 1.0% w/w, less than 0.9% w/w, less than 0.8% w/w, less than 0.7% w/w, less than 0.6% w/w, less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, or less than 0.2% w/w of the crude gemcabene as determined by ion chromatography.
  • a second co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, or 0.2% w/w or less of the crude gemcabene as determined by ion chromatography. In some embodiments, a second co-distillation with water provides the crude gemcabene comprising isobutyric acid impurity in 0.3% w/w or less of the crude gemcabene as determined by ion chromatography.
  • a water/heptane heterogeneous azeotropic distillation can be performed in order to remove substantially all water content as determined by Karl-Fisher analysis.
  • the water content, if any, is ⁇ 0.05% w/w of the reaction mixture as determined by Karl-Fisher analysis.
  • the water content, if any, is 0.05% w/w or less, or 0.04% w/w or less of the reaction mixture as determined by Karl-Fisher analysis.
  • the water content, if any is less than 0.05% w/w, or less than 0.04% w/w of the reaction mixture as determined by Karl-Fisher analysis.
  • the crude gemcabene is passed through silica gel to remove impurities, such as any colored or polar impurities.
  • silica gel filtration is performed using 5% (v/v) THF in heptane as an eluent.
  • the silica gel is washed with only heptane.
  • heptane is n-heptane.
  • the gemcabene-containing fractions from silica gel filtration can be evaporated to substantial dryness and the resultant residue can be crystallized from an organic solvent or mixture of organic solvents.
  • the organic solvent is heptane or a mixture of heptane and THF.
  • the organic solvent is heptane in the absence of THF.
  • heptane is n-heptane.
  • crude gemcabene is dissolved in the organic solvent at a temperature ranging from about 20° C. to about 50° C. In some embodiments, the crude gemcabene is dissolved in the organic solvent at a temperature ranging from 35° C. to 50° C.
  • the organic solution is cooled to 15° C. ⁇ 2° C. In some embodiments, the organic solution is cooled to 15° C. ⁇ 2° C. and subsequently seeded with one or more gemcabene crystals.
  • the organic solvent is heptane. In some embodiments, the organic solvent is n-heptane.
  • the gemcabene is allowed to crystallize at a temperature ranging from 9° C. to 16° C. In some embodiments, the gemcabene is allowed to crystallize at a temperature ranging from 10° C. to 15° C. In some embodiments, the gemcabene is allowed to crystallize at a temperature ranging from 10° C. to 14° C. In some embodiments, the gemcabene is allowed to crystallize at a temperature of 10° C., 11° C., 12° C., 13° C., 14° C., or 15° C. In some embodiments, the gemcabene is allowed to crystallize at a temperature of 12° C.
  • the crude gemcabene before recrystallization comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity. Allowing gemcabene to crystallize from heptane at a temperature ranging from 10° C. to 15° C. yields gemcabene containing substantially less 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity than gemcabene that is allowed to crystallize from heptane at a temperature below 10° C. Moreover, as shown in Table C, the gemcabene of Entry 4, which was allowed to crystallize from heptane maintained at 12-14° C.
  • heptane is n-heptane.
  • a first gemcabene crystallization from heptane at a temperature ranging from 9° C. to 16° C. yields gemcabene comprising 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in ⁇ 0.5% w/w of the crystallized gemcabene as determined by high-performance liquid chromatography (HPLC).
  • gemcabene comprising 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity, if any, in 0.5% w/wor less, 0.4% w/wor less, 0.3% w/wor less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the crystallized gemcabene as determined by HPLC.
  • gemcabene comprising 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the crystallized gemcabene as determined by HPLC.
  • HPLC is equipped with charged aerosol detector (CAD).
  • HPLC is equipped with ultraviolet detector (UV).
  • heptane is n-heptane.
  • a first gemcabene crystallization from heptane at a temperature ranging between 10° C. to 14° C. yields gemcabene containing 2,2,7,7-tetramethyl-octane-1,8-dioic acid in a range of 0.5% w/w to 0.1% w/w, 0.4% w/w to 0.1% w/w, 0.3% w/w to 0.1% w/w, or 0.2% w/w to 0.1% w/w of the crystallized gemcabene as determined by HPLC.
  • gemcabene comprising 2,2,7,7-tetramethyl-octane-1,8-dioic acid in a range of 0.5% w/w to 0.01% w/w, 0.4% w/w to 0.01% w/w, 0.3% w/w to 0.01% w/w, or 0.2% w/w to 0.01% w/w of the crystallized gemcabene as determined by HPLC.
  • gemcabene comprising 2,2,7,7-tetramethyl-octane-1,8-dioic acid in a range of 0.5% w/w to 0.001% w/w, 0.4% w/w to 0.001% w/w, 0.3% w/w to 0.001% w/w, or 0.2% w/w to 0.001% w/w of the crystallized gemcabene as determined by HPLC.
  • heptane is n-heptane.
  • the concentration of the crystallization solution affects the recovery of gemcabene.
  • the crystallization solution has a concentration greater than 0.3 g/mL crude gemcabene in the organic solvent or mixtures of organic solvent.
  • the crystallization solution has a concentration of ⁇ 0.4 g/mL, ⁇ 0.5 g/mL, or ⁇ 0.6 g/mL crude gemcabene in the organic solvent or mixtures of organic solvent.
  • the crystallization solution has a concentration ranging from 0.3 g of crude gemcabene/mL of heptane to 0.9 g of crude gemcabene/mL of heptane.
  • crystallization solution has a concentration ranging from 0.5 g of crude gemcabene/mL of heptane to 0.8 g of crude gemcabene/mL of heptane. In some embodiments, the crystallization solution has a concentration ranging from 0.5 g of crude gemcabene/mL of heptane to 0.7 g of crude gemcabene/mL of heptane. In some embodiments, crystallization solution has a concentration of 0.6 g crude gemcabene/mL of heptane. In some embodiments, heptane is n-heptane.
  • the yield of gemcabene can be affected by the number of equivalents of isobutyric acid, alkali metal hydroxide or enolate-forming base in relation to bis-(4-halobutyl)ether.
  • molar equivalents ranging from 2.05 to 3.00 of each of isobutyric acid, alkali metal hydroxide, and enolate-forming base are used compared to 1.00 molar equivalent of bis-(4-halobutyl)ether.
  • molar equivalents ranging from 2.15 to 2.50 of each of isobutyric acid, alkali metal hydroxide, and enolate-forming base are used compared to 1.0 molar equivalent of bis-(4-halobutyl)ether.
  • molar equivalents ranging from 2.20 to 2.40 of each of isobutyric acid, alkali metal hydroxide, and enolate-forming base are used compared to 1.0 molar equivalent of bis-(4-halobutyl)ether.
  • 2.20 equivalents of each of isobutyric acid, alkali metal hydroxide, and enolate-forming are used compared to 1.0 molar equivalent of bis-(4-chlorobutyl)ether.
  • the alkali metal hydroxide is sodium hydroxide and the enolate-forming base is LDA.
  • the alkali metal hydroxide is sodium hydroxide
  • the enolate-forming base is LDA
  • the bis-(4-halobutyl)ether is bis-(4-iodobutyl)ether.
  • gemcabene made according to any one of the methods disclosed herein has a purity ranging from about 85% w/w to 100% w/w as determined by high-performance liquid chromatography (HPLC). In some embodiments, gemcabene has a purity ranging from about 90% w/w to100% w/w as determined by HPLC. In some embodiments, gemcabene has a purity ranging from about 95% w/w to 100% w/w as determined by HPLC. In some embodiments, gemcabene has a purity ranging from about 98% w/w to 100% w/w as determined by HPLC. In some embodiments, gemcabene has a purity ranging from about 99% w/w to 100% w/w as determined by HPLC.
  • gemcabene has a purity ranging from 99.0% to 100% as determined by HPLC. In some embodiments, gemcabene has a purity ranging from about 99.5% w/w to 100% w/w as determined by HPLC. In some embodiments, HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • CAD charged aerosol detector
  • UV ultraviolet detector
  • gemcabene made according to any one of the methods disclosed herein comprises isobutyric acid impurity in ⁇ 0.5% w/w of the gemcabene as determined by ion chromatography (IC).
  • gemcabene comprises isobutyric acid impurity, if any, in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene as determined by IC.
  • gemcabene comprises isobutyric acid impurity in less than 0.5%, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene as determined by IC.
  • gemcabene comprises isobutyric acid impurity in 0.05% w/w or less of the gemcabene as determined by IC.
  • gemcabene is substantially free of isobutyric acid impurity.
  • isobutyric acid impurity in gemcabene is below the quantification limit of the IC.
  • the quantification limit of isobutyric acid using an IC is 0.05% w/w.
  • gemcabene made according to any one of the methods disclosed herein comprises 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid impurity in ⁇ 0.5% w/w of the gemcabene as determined by high-performance liquid chromatography (HPLC).
  • gemcabene comprises 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid impurity in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene as determined by HPLC.
  • gemcabene comprises 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid impurity, if any, in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • gemcabene made according to any one of the methods disclosed herein comprises (Z)-2,2-dimethyl-hex-4-enoic acid impurity in ⁇ 0.5% w/w of the gemcabene as determined by high-performance liquid chromatography (HPLC).
  • gemcabene comprises (Z)-2,2-dimethyl-hex-4-enoic acid impurity in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene as determined by HPLC.
  • gemcabene comprises (Z)-2,2-dimethyl-hex-4-enoic acid impurity, if any, in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • gemcabene made according to any one of the methods disclosed herein comprises (E)-2,2-dimethyl-hex-4-enoic acid impurity in ⁇ 1.0% w/w of the gemcabene as determined by high-performance liquid chromatography (HPLC).
  • gemcabene comprises (E)-2,2-dimethyl-hex-4-enoic acid impurity in ⁇ 0.5% of the gemcabene as determined by HPLC.
  • gemcabene comprises (E)-2,2-dimethyl-hex-4-enoic acid impurity in less than 1.0% w/w, less than 0.9% w/w, less than 0.8% w/w, less than 0.7% w/w, less than 0.6% w/w, less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene as determined by HPLC.
  • gemcabene comprises (E)-2,2-dimethyl-hex-4-enoic acid impurity, if any, in 1.0% w/w or less, 0.9% w/w or less, 0.8% w/w or less, 0.7% w/w or less, 0.6% w/w or less, 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • the present invention further provides gemcabene made according to any one of the methods disclosed herein.
  • the present invention further provides gemcabene purified according to any one of the methods disclosed herein.
  • the present invention further provides gemcabene purified by dissolving the crude gemcabene in heptane and cooling the heptane solution to a temperature ranging from 10° C. to 15° C. to precipitate gemcabene.
  • heptane is n-heptane.
  • the present invention further provides a pharmaceutically acceptable salt of gemcabene, wherein gemcabene is synthesized according to any one of the methods disclosed herein.
  • the present invention further provides a pharmaceutically acceptable salt of gemcabene, wherein gemcabene is purified according to any one of the methods disclosed herein.
  • the present invention further provides a pharmaceutically acceptable salt of gemcabene, wherein gemcabene is purified by dissolving the crude gemcabene in heptane and cooling the heptane solution to a temperature ranging from 10° C. to 15° C. to precipitate gemcabene.
  • heptane is n-heptane.
  • gemcabene synthesized according to any one of the methods disclosed herein can be converted into gemcabene calcium.
  • gemcabene is allowed to react with calcium oxide.
  • gemcabene is allowed to react with calcium oxide in ethanol.
  • gemcabene is allowed to react with calcium oxide in ethanol under refluxing conditions.
  • the reaction mixture can be stirred at 22° C. ⁇ 2° C. for about one hour and then can be filtered.
  • the filtered product can then be dried under vacuum. In some embodiments, the drying is performed under stream of nitrogen under vacuum.
  • purified water is added to the dried gemcabene calcium and heated. In some embodiments, purified water is added to the dried gemcabene calcium at atmospheric pressure and heated to a temperature range of about 80 to about 110° C. In some embodiments, purified water is added to the dried gemcabene calcium at atmospheric pressure and heated to a temperature range of about 85° C. to about 95° C. for about 5 hours to about 10 hours. In some embodiments, purified water is added to the dried gemcabene calcium at atmospheric pressure and heated to 90° C. for about 6 hours. Heating gemcabene calcium with purified water provides gemcabene calcium salt hydrate.
  • gemcabene calcium salt hydrate is dried under vacuum. In some embodiments, gemcabene calcium salt hydrate is dried under vacuum at a temperature range of about 80° C. to about 110° C. In some embodiments, gemcabene calcium salt hydrate is dried under vacuum at a temperature range of about 85° C. to about 95° C. for at least 5 hours, at least 10 hours, or at least 15 hours. In some embodiments, gemcabene calcium salt hydrate is dried under vacuum at a temperature of 90° C. for at least 16 hours to yield gemcabene calcium salt hydrate Crystal Form 1. Similarly, gemcabene calcium salt solvate can be obtained with alcohol solvents, such as ethanol.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein has a purity ranging from about 85% w/w to 100% w/w as determined by high-performance liquid chromatography (HPLC). In some embodiments, gemcabene calcium salt hydrate or solvate has a purity ranging from about 90% w/w to 100% w/w as determined by HPLC. In some embodiments, gemcabene calcium salt hydrate or solvate has a purity ranging from about 95% w/w to 100% w/w as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate has a purity ranging from about 98% w/w to 100% w/w as determined by HPLC. In some embodiments, gemcabene calcium salt hydrate or solvate has a purity ranging from about 99% w/w to 100% w/w as determined by HPLC. In some embodiments, gemcabene calcium salt hydrate or solvate has a purity ranging from about 99.5% w/w to 100% w/w as determined by HPLC. In some embodiments, gemcabene calcium salt hydrate or solvate has a purity ranging from 99.5% w/w to 100% w/w as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate has a purity ranging from 99.7% w/w to 100% w/w as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • CAD charged aerosol detector
  • UV ultraviolet detector
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid impurity in ⁇ 0.5% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • HPLC high-performance liquid chromatography
  • gemcabene calcium salt hydrate or solvate comprises 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid impurity, if any, in less than 0.5%, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate comprises 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid impurity in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in ⁇ 0.5% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • HPLC high-performance liquid chromatography
  • gemcabene calcium salt hydrate or solvate comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity, if any, in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises isobutyric acid impurity in ⁇ 0.5% w/w of the gemcabene calcium salt hydrate or solvate as determined by ion chromatography (IC).
  • gemcabene calcium salt hydrate or solvate comprises isobutyric acid impurity in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene calcium salt hydrate or solvate as determined by IC.
  • gemcabene calcium salt hydrate or solvate comprises isobutyric acid impurity, if any, in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by IC.
  • gemcabene calcium salt hydrate or solvate comprises isobutyric acid impurity in 0.07% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by IC.
  • gemcabene calcium salt hydrate or solvate comprises isobutyric acid impurity in 0.05% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by IC. In some embodiments, gemcabene calcium salt hydrate or solvate is substantially free of isobutyric acid impurity. In some embodiments, isobutyric acid impurity in gemcabene calcium salt hydrate or solvate is below the quantification limit of the IC. In one embodiment, the quantification limit of isobutyric acid using an IC is 0.05% w/w.
  • gemcabene calcium salt hydrate or solvate made from gemcabene synthesized according to any one of the methods disclosed herein comprises (Z)-2,2-dimethyl-hex-4-enoic acid impurity in ⁇ 0.5% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • gemcabene calcium salt hydrate or solvate comprises (Z)-2,2-dimethyl-hex-4-enoic acid impurity in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate comprises (Z)-2,2-dimethyl-hex-4-enoic acid impurity, if any, in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • gemcabene calcium salt hydrate or solvate made from gemcabene synthesized according to any one of the method disclosed herein comprises E)-2,2-dimethyl-hex-4-enoic acid impurity in ⁇ 0.5% w/w (of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate comprises (E)-2,2-dimethyl-hex-4-enoic acid impurity in less than 0.5% w/w, less than 0.4% w/w, less than 0.3% w/w, less than 0.2% w/w, less than 0.15% w/w, less than 0.1% w/w, or less than 0.05% w/w of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • gemcabene calcium salt hydrate or solvate comprises (E)-2,2-dimethyl-hex-4-enoic acid impurity, if any, in 0.5% w/w or less, 0.4% w/w or less, 0.3% w/w or less, 0.2% w/w or less, 0.15% w/w or less, 0.1% w/w or less, or 0.05% w/w or less of the gemcabene calcium salt hydrate or solvate as determined by HPLC.
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV).
  • gemcabene calcium salt hydrate or solvate made from gemcabene synthesized according to any one of the methods disclosed herein comprises ⁇ 2.5 ppm bis-(4-chlorobutyl)ether impurity as determined by gas chromatography (GC).
  • gemcabene calcium salt hydrate or solvate comprises less than 2.5 ppm, less than 2.0 ppm, less than 1.5 ppm or less than 1.0 ppm bis-(4-chlorobutyl)ether impurity as determined by GC.
  • gemcabene calcium salt hydrate or solvate comprises 2.5 ppm or less, 2.0 ppm or less, 1.5 ppm or less, or 1.0 ppm or less bis-(4-chlorobutyl)ether impurity as determined by GC.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the method disclosed herein contains ⁇ 2.5 ppm 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid impurity as determined by gas chromatography (GC).
  • gemcabene calcium salt hydrate or solvate contains less than 2.5 ppm, less than 2.0 ppm, less than 1.5 ppm or less than 1.0 ppm 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid impurity as determined by GC.
  • gemcabene calcium salt hydrate or solvate contains 2.5 ppm or less, 2.0 ppm or less, 1.5 ppm or less, or 1.0 ppm or less 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid impurity as determined by GC.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the method disclosed herein contains ⁇ 2.5 ppm 1-chloro-4-hydroxybutane impurity as determined by gas chromatography (GC). In some embodiments, gemcabene calcium salt hydrate or solvate contains less than 2.5 ppm, less than 2.0 ppm, less than 1.5 ppm or less than 1.0 ppm 1-chloro-4-hydroxybutane impurity as determined by GC.
  • gemcabene calcium salt hydrate or solvate contains 2.5 ppm or less, 2.0 ppm or less, 1.5 ppm or less, or 1.0 ppm or less 1-chloro-4-hydroxybutane impurity as determined by GC.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the method disclosed herein contains ⁇ 8 ppm collectively the sum of 1-chloro-4-hydroxybutane, 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid and (bis-(4-chlorobutyl)ether impurities as determined by gas chromatography (GC).
  • GC gas chromatography
  • gemcabene calcium salt hydrate or solvate contains less than 8 ppm, less than 7.0 ppm, less than 6 ppm or less than 5.0 ppm collectively the sum of 1-chloro-4-hydroxybutane, 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid and (bis-(4-chlorobutyl)ether impurities as determined by GC.
  • gemcabene calcium salt hydrate or solvate contains 8 ppm or less, 7.5 ppm or less, 7.0 ppm or less, or 6.5 ppm or less 1-chloro-4-hydroxybutane impurity as determined by GC.
  • gemcabene calcium salt hydrate made from gemcabene synthesized according to any one of the methods disclosed herein comprises water in the range of about 2.0% w/w to about 5.0% w/w of the gemcabene calcium salt hydrate as determined by Karl-Fisher analysis.
  • gemcabene calcium salt hydrate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises water in the range of 2.0% w/w to 5.0% w/w of the gemcabene calcium salt hydrate as determined by Karl-Fisher analysis.
  • gemcabene calcium salt hydrate or solvate made from gemcabene synthesized according to any one of the methods disclosed herein comprises calcium in a range from about 10% m/m to about 15 m/m of the gemcabene calcium salt hydrate or solvate as determined by inductively coupled plasma optical emission spectrometry (ICP-OES).
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises calcium in a range from about 10% m/m to about 14% m/m of the gemcabene calcium salt hydrate or solvate as determined by ICP-OES.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises calcium in a range from 9.8% m/m to 13.8% m/m of the gemcabene calcium salt hydrate or solvate as determined by ICP-OES. In some embodiments, gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises calcium in a range from 11.5% m/m to 12.5% m/m of the gemcabene calcium salt hydrate or solvate as determined by ICP-OES.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises calcium in about 11.77% m/m of the gemcabene calcium salt hydrate or solvate as determined by ICP-OES.
  • gemcabene calcium salt hydrate or solvate made from gemcabene synthesized according to any one of the methods disclosed herein comprises a gemcabene conjugate base component ranging from about 82% w/w to about 92% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC), wherein the gemcabene conjugate base has the structure:
  • gemcabene calcium salt hydrate or solvate made from gemcabene made according to any one of the methods disclosed herein comprises a gemcabene conjugate base component ranging from 82% w/w to 92% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • the gemcabene conjugate base component is percentage of the gemcabene calcium salt hydrate or solvate without accounting for water, solvent, and calcium content.
  • HPLC is equipped with an ultraviolet detector (UV).
  • gemcabene calcium salt hydrate or solvate made from gemcabene made according to any one of the methods disclosed herein has an anhydrous gemcabene calcium content from about 98% w/w to about 105% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • gemcabene calcium salt hydrate or solvate made from gemcabene made according to any one of the methods disclosed herein has an anhydrous gemcabene calcium content from 98% w/w to 105% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • gemcabene calcium salt hydrate or solvate made from gemcabene made according to any one of the methods disclosed herein comprises 2.0% or less of total impurities as determined by high-performance liquid chromatography.
  • gemcabene calcium salt hydrate or solvate prepared from gemcabene synthesized according to any one of the methods disclosed herein comprises total impurities in less than 2.0% w/w of the gemcabene calcium salt hydrate or solvate as determined by high-performance liquid chromatography (HPLC).
  • HPLC is equipped with a charged aerosol detector (CAD) or with an ultraviolet detector (UV). Different HPLC instrument's impurity analyses can be added to provide the sum of impurities.
  • CAD charged aerosol detector
  • UV ultraviolet detector
  • an “impurities” refers to any organic compounds that are not gemcabene or a pharmaceutically acceptable salt of gemcabene that is detectable by HPLC.
  • impurities for example, isobutyric acid and bis-(4-halobutyl)ether are examples of impurities.
  • Other examples of related substances are presented in Table D.
  • the present invention further provides methods for purifying crude gemcabene, wherein the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 5% w/w of the crude gemcabene as determined by high-performance liquid chromatography (HPLC), comprising: dissolving the crude gemcabene in heptane to provide a heptane solution of the crude gemcabene; and cooling the heptane solution to a temperature ranging from 10° C. to 15° C.
  • HPLC high-performance liquid chromatography
  • the gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in 0.5% w/w or less of the gemcabene of as determined by high-performance liquid chromatography.
  • the present invention further provides methods for purifying crude gemcabene, wherein the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 3% w/w of the crude gemcabene as determined by high-performance liquid chromatography (HPLC), comprising: dissolving the crude gemcabene in heptane to provide a heptane solution of the crude gemcabene; and cooling the heptane solution to a temperature ranging from 10° C. to 15° C.
  • HPLC high-performance liquid chromatography
  • the gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in 0.5% w/w or less of the gemcabene of as determined by high-performance liquid chromatography.
  • the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 2.5% w/w of the crude gemcabene as determined by HPLC.
  • the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 2% w/w of the crude gemcabene as determined by HPLC.
  • the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 1.5% w/w of the crude gemcabene as determined by HPLC. In some embodiments, the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 1% w/w of the crude gemcabene as determined by HPLC.
  • the present invention further provides methods for purifying crude gemcabene, wherein the crude gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in no more than 1% w/w of the crude gemcabene as determined by high-performance liquid chromatography, comprising: dissolving the crude gemcabene in heptane to provide a heptane solution of the crude gemcabene; and cooling the heptane solution to a temperature ranging from 10° C. to 15° C.
  • the gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in 0.5% w/w or less of the gemcabene of as determined by high-performance liquid chromatography.
  • the crude gemcabene prior to purification comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in greater than 0.7% w/w and no more than 1% w/w of the crude gemcabene as determined by high-performance liquid chromatography (HPLC).
  • the crude gemcabene prior to purification comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in greater than 0.5% w/w and no more than 1% w/w of the crude gemcabene as determined by HPLC.
  • the crude gemcabene prior to purification comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid impurity in a range of 1.0% w/w to 0.5% w/w of the crude gemcabene as determined by HPLC.
  • the gemcabene after purification comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid from 0.01% w/w to 0.5% w/w of the gemcabene as determined by high-performance liquid chromatography.
  • the temperature of the heptane solution for purification ranges from 10° C. to 14° C. In some embodiments, the temperature of the heptane solution for purification is 12° C. In some embodiments, the temperature of the heptane solution during crystallization ranges from 10° C. to 14° C. In some embodiments, the temperature of the heptane solution during crystallization is 12° C.
  • the crude gemcabene further comprises isobutyric acid in 0.5% w/w or less of the crude gemcabene as determined by ion chromatography. In some embodiments, the crude gemcabene comprises isobutyric acid in 0.3% or less of the crude gemcabene as determined by ion chromatography.
  • the concentration of crude gemcabene in the heptane solution ranges from 0.3 g of crude gemcabene/mL of heptane to 0.8 g of crude gemcabene/mL of heptane. In some embodiments, the concentration of crude gemcabene in the heptane solution ranges from 0.5 g of crude gemcabene/mL of heptane to 0.7 g of crude gemcabene/mL of heptane. In some embodiments, the concentration of crude gemcabene in the heptane solution is 0.6 g of crude gemcabene/mL of heptane.
  • the method of purifying crude gemcabene further comprises: dissolving the gemcabene in heptane to provide a heptane solution of the gemcabene; and cooling the heptane solution to a temperature ranging from 10° C. to 15° C. to precipitate recrystallized gemcabene.
  • heptane is n-heptane.
  • the method of purifying crude gemcabene further comprises: allowing an enolate of an alkali metal salt of isobutyric acid to react with a bis-(4-halobutyl)ether to provide crude gemcabene salt and acidifying the crude gemcabene salt to provide the crude gemcabene.
  • the enolate of an alkali metal salt of isobutyric acid to react is allowed to react with the bis-(4-halobutyl)ether under conditions essentially free of water.
  • the method further comprising allowing sodium isobutyrate to react with an enolate-forming base to provide the enolate of sodium isobutyrate.
  • the method further comprising allowing isobutyric acid to react with sodium hydroxide to provide the sodium isobutyrate.
  • the bis-(4-halobutyl)ether is bis-(4-chlorobutyl)ether.
  • the enolate of the alkali metal salt of isobutyric acid is an enolate of sodium isobutyrate.
  • the enolate-forming base is lithium hexamethyldisilazide, lithium diisopropylamide, lithium tetramethylpiperidide, or lithium diethylamide.
  • the sodium hydroxide is in a water solution, and further comprising removing the water via evaporation after allowing the isobutyric acid to react with sodium hydroxide and before allowing the sodium isobutyrate to react with the enolate-forming base.
  • the sodium isobutyrate has a water content of 0.05% w/w or less of the reaction mixture comprising sodium isobutyrate as determined by Karl-Fisher analysis.
  • the sodium isobutyrate has a water content of about 0.05% w/w or less of the reaction mixture comprising sodium isobutyrate as determined by Karl-Fisher analysis.
  • the enolate of the alkali metal salt of isobutyric acid is present in an amount of two or more molar equivalents and the bis-(4-halobutyl)ether present in an amount of one molar equivalent. In some embodiments, the enolate of an alkali metal salt of isobutyric acid is present in an amount of 2.1 to 2.4 molar equivalents and the bis-(4-halobutyl)ether present in an amount of one molar equivalent.
  • the crude gemcabene further comprises isobutyric acid.
  • the isobutyric acid is removed from the crude gemcabene via distillation after acidifying the crude gemcabene salt and before precipitating gemcabene from the heptane solution at a temperature ranging from 10° C. to 15° C.
  • the removal of isobutyric acid further comprising admixing the crude gemcabene and water prior to removing at least some of the isobutyric acid.
  • the distillation removes water and isobutyric acid.
  • the admixing the crude gemcabene and water and removing the water and at least some of the isobutyric acid is performed at least two times.
  • the crude gemcabene after distillation comprises isobutyric acid in 0.5% w/w or less of the distilled crude gemcabene as determined by ion chromatography. In some embodiments, the crude gemcabene after distillation comprises isobutyric acid in 0.3% or less of the distilled crude gemcabene as determined by ion chromatography.
  • the present invention further provides gemcabene made by or purified by any one of the methods disclosed herein.
  • gemcabene comprises isobutyric acid in 0.10% w/w or less of the gemcabene as determined by ion chromatography.
  • gemcabene comprises isobutyric acid in 0.05% w/w or less of the gemcabene as determined by ion chromatography.
  • the present invention further provides a pharmaceutically acceptable salt of gemcabene made by or purified by any one of the methods disclosed herein.
  • the pharmaceutically acceptable salt is a calcium salt.
  • the calcium salt is a hydrate.
  • the calcium salt hydrate is Crystal Form 1.
  • the calcium salt hydrate is Crystal Form 2.
  • the calcium salt hydrate is Crystal Form C3.
  • the calcium salt is an ethanol solvate.
  • the pharmaceutically acceptable salt of gemcabene comprises 2,2,7,7-tetramethyl-octane-1,8-dioic acid in 0.5% w/w or less of the pharmaceutically acceptable salt of gemcabene as determined by high-performance liquid chromatography.
  • the pharmaceutically acceptable salt gemcabene comprises water in 2% w/w to 5% w/w of the pharmaceutically acceptable salt of gemcabene as determined by Karl-Fisher analysis.
  • the pharmaceutically acceptable salt of gemcabene comprises isobutyric acid in 0.5% w/w or less of the pharmaceutically acceptable salt of gemcabene as determined by ion chromatography.
  • the pharmaceutically acceptable salt gemcabene comprises isobutyric acid in 0.10% w/w or less of the pharmaceutically acceptable salt of gemcabene as determined by ion chromatography. In some embodiments, the pharmaceutically acceptable salt gemcabene comprises isobutyric acid in 0.05% w/w or less of the pharmaceutically acceptable salt of gemcabene as determined by ion chromatography.
  • the pharmaceutically acceptable salt gemcabene comprises 2.5 ppm or less of bis-(4-chlorobutyl)ether as determined by gas chromatography. In some embodiments, the pharmaceutically acceptable salt gemcabene comprises 2.5 ppm or less of 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid as determined by gas chromatography. In some embodiments, the pharmaceutically acceptable salt gemcabene comprises 2.5 ppm or less of 1-chloro-4-hydroxybutane as determined by gas chromatography.
  • the pharmaceutically acceptable salt gemcabene comprises 8 ppm or less of sum of all genotoxic impurities, including but not limited to, bis-(4-chlorobutyl)ether, 1-chloro-4-hydroxybutane and 6-(4-chlorobutoxy)-2,2-dimethyl-hexanoic acid as determined by gas chromatography.
  • the pharmaceutically acceptable salt gemcabene comprises total impurities in 2.0% w/w or less of the pharmaceutically acceptable salt of gemcabene as determined by high-performance liquid chromatography.
  • the pharmaceutically acceptable salt gemcabene comprises a gemcabene conjugate base component in a range of 82% w/w to 92% w/w of the pharmaceutically acceptable salt of gemcabene as determined by high-performance liquid chromatography, wherein the gemcabene conjugate base component has the structure:
  • the pharmaceutically acceptable salt gemcabene comprises calcium in about 10% m/m to about 14% m/m of the pharmaceutically acceptable salt of gemcabene as determined by inductively coupled plasma optical emission spectrometry. In some embodiments, the pharmaceutically acceptable salt gemcabene comprises calcium in about 9.8% m/m to 13.8% m/m of the pharmaceutically acceptable salt of gemcabene as determined by inductively coupled plasma optical emission spectrometry.
  • the present invention further provides pharmaceutical compositions comprising a pharmaceutically acceptable salt of gemcabene and a pharmaceutically acceptable carrier or vehicle, wherein gemcabene is synthesized according to any one of the methods disclosed herein.
  • the present invention further provides pharmaceutical compositions comprising a pharmaceutically acceptable salt of gemcabene and a pharmaceutically acceptable carrier or vehicle, wherein gemcabene is purified according to any one of the methods disclosed herein.
  • the present invention further provides pharmaceutical compositions comprising a pharmaceutically acceptable salt of gemcabene and a pharmaceutically acceptable carrier or vehicle, wherein gemcabene is purified according to any one of the methods disclosed by dissolving the crude gemcabene in heptane and cooling the heptane solution to a temperature ranging from 10° C. to 15° C. to precipitate gemcabene.
  • heptane is n-heptane.
  • the present invention provides methods for treating or preventing various diseases and conditions as disclosed herein, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is human.
  • the present invention provides methods for treating or preventing liver disease or an abnormal liver condition, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • liver disease or liver conditions include, but are not limited to, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), alcoholic steatohepatitis, cirrhosis, inflammation, liver fibrosis, partial fibrosis, primary biliary cirrhosis, primary sclerosing cholangitis, liver failure, hepatocellular carcinoma (HCC), liver cancer, hepatic steatosis, hepatocyte ballooning (also known as hepatocellular ballooning), hepatic lobular inflammation, and hepatic triglyceride accumulation.
  • the liver disease or the liver condition is NAFLD or NASH.
  • the liver disease or the liver condition is NAFLD. In other embodiments, the liver disease or the liver condition is NASH. In some embodiments, the liver disease or the liver condition is hepatic steatosis. In some embodiments, the liver disease or the liver condition is liver fibrosis.
  • treating or preventing liver fibrosis, NAFLD, or NASH includes regressing, stabilizing, or inhibiting progression of liver fibrosis, NAFLD, or NASH.
  • the present invention further provides methods for reducing liver fat (fat content of the liver), stabilizing the amount of liver fat, or reducing the accumulation of liver fat, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for reducing liver steatosis (fat content of the liver), stabilizing the amount of liver triglycerides, or reducing the accumulation of liver triglycerides, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for treating or preventing lobular inflammation or hepatocyte ballooning, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • treating or preventing lobular inflammation or hepatocyte ballooning is slowing the progression of, stabilizing, or reducing the lobular inflammation or hepatocyte ballooning.
  • the present invention further provides methods for treating or preventing a disorder of lipoprotein metabolism, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • disorders of lipoprotein metabolism include, but are not limited to, dyslipidemia, dyslipoproteinemia, mixed dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), type IIb hyperlipidemia or familial combined hyperlipidemia, familial hypercholesterolemia, familial chylomicronemia syndrome, hypertriglyceridemia, dysbetalipoproteinemia, lipoprotein overproduction or deficiency, elevation of total cholesterol, elevation of low-density lipoprotein cholesterol concentration, elevation of very low-density lipoprotein cholesterol concentration, elevation of non-high-density lipoprotein (non-HDL) cholesterol concentration, elevation of apolipoprotein B concentration, elevation of apolipoprotein C-III concentration, elevation of C-reactive protein concentration, elevation of fibrinogen concentration, elevation of lipoprotein(a) concentration, elevation of interleukin-6 concentration, elevation of angiopoietin-like protein 3 concentration, elevation of angiopoietin-like protein 4 concentration, elevation of serum amyloid A
  • the disorder of lipoprotein metabolism is dyslipidemia, dyslipoproteinemia, mixed dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), type IIb hyperlipidemia, familial combined hyperlipidemia, familial hypercholesterolemia, familial chylomicronemia syndrome, hypertriglyceridemia, dysbetalipoproteinemia, metabolic syndrome, lipoprotein overproduction, lipoprotein deficiency, non-insulin dependent diabetes, abnormal lipid elimination in bile, a metabolic disorder, abnormal phospholipid elimination in bile, an abnormal oxysterol elimination in bile, an abnormal bile production, hypercholesterolemia, hyperlipidemia or visceral obesity.
  • ASCVD atherosclerotic cardiovascular disease
  • the disorder of lipoprotein metabolism is mixed dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), type IIb hyperlipidemia, familial combined hyperlipidemia, or familial hypercholesterolemia.
  • ASCVD atherosclerotic cardiovascular disease
  • type IIb hyperlipidemia familial combined hyperlipidemia
  • familial hypercholesterolemia is hypertriglyceridemia.
  • the disorder of lipoprotein metabolism is hypercholesterolemia.
  • the hypertriglyceridemia is a severe hypertriglyceridemia. “Severe hypertriglyceridemia” is where a subject has a baseline plasma triglyceride concentration of greater than or equal to 500 mg/dl.
  • familial hypercholesterolemia (FH) is homozygous FH (HoFH) or heterozygous FH (HeFH).
  • the present invention further provides methods for treating or preventing a peroxisome proliferator activated receptor-associated disorder.
  • the present invention further provides methods for reducing a subject's plasma or blood serum triglyceride concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum, the subject's total cholesterol concentration, low-density lipoprotein cholesterol concentration, low-density lipoprotein concentration, very low-density lipoprotein cholesterol concentration, very low-density lipoprotein concentration, non-HDL cholesterol concentration, non-HDL concentration, apolipoprotein B concentration, triglyceride concentration, apolipoprotein C-III concentration, C-reactive protein concentration, fibrinogen concentration, lipoprotein(a) concentration, interleukin-6 concentration, angiopoietin-like protein 3 concentration, angiopoietin-like protein 4 concentration, PCSK9 concentration, or serum amyloid A concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing in the subject's blood plasma or blood serum, the subject's triglyceride concentration or low-density lipoprotein cholesterol concentrations, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for lowering in a subject's blood plasma or blood serum, the subject's low-density lipoprotein cholesterol (LDL-C) concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention, wherein the subject is on a stable dose of a statin.
  • LDL-C low-density lipoprotein cholesterol
  • the present invention provides methods for elevating in a subject's blood plasma or blood serum, the subject's high-density lipoprotein cholesterol concentration, high-density lipoprotein concentration, high-density cholesterol triglyceride concentration, adiponectin concentration or apolipoprotein A-I concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for cholesterol or triglyceride mobilization from a subject's endothelial and epithelial cells to the subject's blood plasma or blood serum and transport for clearance and excretion, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a subject's risk of developing a thrombosis, a blood clot, a primary cardiovascular event, a secondary cardiovascular event, progression to nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, liver cirrhosis, hepatocellular carcinoma, liver failure, pancreatitis, pulmonary fibrosis, or hyperlipoproteinemia type IIB, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a subject's risk of developing pancreatitis.
  • the present invention provides methods for reducing a subject's risk of developing an ApoC-II deficiency.
  • the present invention provides methods for treating or preventing fibrosis, steatosis, ballooning or inflammation in the liver of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • treating or preventing ballooning or inflammation in the liver of a subject is reducing ballooning or inflammation in the liver of a subject.
  • the present invention further provides reducing or inhibiting progression of fibrosis, steatosis, ballooning or inflammation in the liver of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing post-prandial lipemia or preventing prolonged post-prandial lipemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for decreasing the extent and duration of post-prandial lipemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for decreasing the extent and duration of post-prandial lipemia, comprising administering to a subject in need thereof a composition of the invention.
  • the present invention provides methods for treating or preventing hypoalphalipoproteinemia.
  • the present invention provides methods for reducing a magnitude or duration of post-prandial lipemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a fat content of the liver of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention provides methods for reducing a steatosis of the liver of a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for reducing a subject's risk of thrombosis or blood clot, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the therapeutic or prophylactic methods of the invention are effective to reduce a subject's plasma or blood serum triglyceride concentration to below about 200 mg/dl or to below about 150 mg/dl. In some embodiments, the therapeutic or prophylactic methods of the invention are effective to reduce a subject's plasma or blood serum triglyceride concentration to below about 200 mg/dl or to below about 150 mg/dl within about 8 to about 12 weeks after administering a compound of the invention.
  • the therapeutic or prophylactic methods of the invention are effective to reduce the subject's plasma or blood serum triglyceride concentration by at least 10% in a subject whose baseline plasma or blood serum triglyceride concentration is 500 mg/dl or higher, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the therapeutic or prophylactic methods of the invention are effective to reduce the subject's plasma or blood serum triglyceride concentration by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, or any range between any of these values, of the baseline plasma or blood serum triglyceride concentration where the subject has a baseline plasma or blood serum triglyceride concentration of 500 mg/dl or higher.
  • the therapeutic or prophylactic methods of the invention are effective to reduce the subject's plasma or blood serum triglyceride concentration by up to about 60% of the baseline plasma or blood serum triglyceride concentration in a subject whose baseline plasma or blood serum triglyceride concentration is 500 mg/dl or higher, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the therapeutic or prophylactic methods of the invention are effective to reduce the subject's plasma or blood serum triglyceride concentration by at least 10% in a subject whose baseline plasma or blood serum triglyceride concentration is 200 mg/dl or higher, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the therapeutic or prophylactic methods of the invention are effective to reduce the subject's plasma or blood serum triglyceride concentration by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, or any range between any of these values, of the baseline plasma or blood serum triglyceride concentration where the subject has a baseline plasma or blood serum triglyceride concentration is 200 mg/dl or higher.
  • the therapeutic or prophylactic methods of the invention are effective to reduce the subject's plasma or blood serum triglyceride concentration by up to about 35%, by up to about 36%, by up to about 37%, by up to about 38%, by up to about 39%, or by up to about 40% of the baseline plasma or blood serum triglyceride concentration in a subject whose baseline plasma or blood serum triglyceride concentration is 200 mg/dl or higher, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for reducing a subject's plasma or blood serum LDL cholesterol concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present methods are effective to reduce the subject's plasma or blood serum LDL cholesterol concentration to below about 130 mg/dl. In some embodiments, the present methods are effective to reduce the subject's plasma or blood serum LDL cholesterol concentration to below about 130 mg/dl within about 8 to about 12 weeks of administering a compound of the invention.
  • the present invention further provides methods for reducing a subject's ApoB concentration, comprising administering to a subject in need thereof an effective amount a compound of the invention.
  • the methods are effective to reduce the subject's ApoB concentration to below about 120 mg/dl.
  • the methods are effective to reduce the subject's ApoB concentration to below about 120 mg/dl within about 8 to about 12 weeks following administering a compound of the invention.
  • the subject has atherometabolic syndrome, metabolic syndrome, type-2 diabetes, impaired glucose tolerance, obesity, dyslipidemia, hepatitis B, hepatitis C, a human immunodeficiency virus (HIV) infection, or a metabolic disorder such as Wilson's disease, a glycogen storage disorder, galactosemia, an inflammatory condition or an elevated body mass index above what is normal for the subject's gender, age or height.
  • a metabolic disorder such as Wilson's disease, a glycogen storage disorder, galactosemia, an inflammatory condition or an elevated body mass index above what is normal for the subject's gender, age or height.
  • metabolic syndrome type-2 diabetes, impaired glucose tolerance, obesity, dyslipidemia, hepatitis B, hepatitis C, an HIV infection, or a metabolic disorder such as Wilson's disease, a glycogen storage disorder or galactosemia is believed to be a risk factor for developing fatty liver (steatosis).
  • the subject has an HIV infection.
  • the subject has an HIV infection and the subject is being administered with a highly active antiretroviral therapy (HAART) agent such as an antiretroviral inhibitor.
  • HAART highly active antiretroviral therapy
  • a compound of the invention is believed to be catabolized to a much lesser extent by the same P450 enzymes that metabolize antiretroviral inhibitors when treating an HIV subject undergoing an antiretroviral inhibitor treatment.
  • the present invention further provides methods for treating or preventing an HIV-associated the liver disease or the liver condition. In some embodiments, the present invention further provides methods for treating or preventing an HIV-associated NAFLD. In some embodiments, the present invention further provides methods for treating or preventing an HIV-associated lipodystrophy. In some embodiments, the present invention further provides methods for treating or preventing a liver disease or the liver condition, comprising administering an effective amount of a compound of the invention to a subject who has an HIV infection. In some embodiments, the present invention further provides methods for treating or preventing NAFLD, comprising administering an effective amount of a compound of the invention to a subject who has an HIV infection.
  • the present invention further provides methods for treating or preventing a disorder of glucose metabolism, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • disorders of glucose metabolism include, but are not limited to, is insulin resistance, impaired glucose tolerance, impaired fasting glucose (concentrations in blood), diabetes mellitus, lipodystrophy, familial partial lipodystrophy, obesity, peripheral lipoatrophy, diabetic nephropathy, diabetic retinopathy, renal disease, and septicemia.
  • obesity is central obesity.
  • the present invention further provides methods for treating or preventing a disorder of glucose metabolism, comprising administering an effective amount of a compound of the invention to a subject who has an HIV infection, In some embodiments, the present invention further provides methods for treating or preventing lipodystrophy, comprising administering an effective amount of a compound of the invention to a subject who has an HIV infection.
  • the present invention further provides methods for treating or preventing a cardiovascular disorder or a related vascular disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • cardiovascular disorders and related vascular disorders include, but are not limited to, arteriosclerosis, atherosclerosis, hypertension, coronary artery disease, myocardial infarction, arrhythmia, atrial fibrillation, heart valve disease, heart failure, cardiomyopathy, myopathy, pericarditis, impotence, and a thrombotic disorder.
  • the present invention further provides methods for reducing a subject's risk of having a cardiovascular or vascular event, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the cardiovascular or vascular event is primary cardiovascular event.
  • the cardiovascular event is secondary cardiovascular event.
  • cardiovascular events include, but are not limited to, myocardial infarction, stroke, angina, acute coronary syndrome, coronary artery bypass graft surgery and cardiovascular death.
  • a primary cardiovascular event is the first cardiovascular event that a subject experiences. If the same subject experiences a second cardiovascular event, then the second cardiovascular event is a secondary cardiovascular event.
  • the present invention further provides methods for treating or preventing a disease caused by an increased level of fibrosis, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the disease caused by an increased level of fibrosis is a lung disease.
  • the disease caused by an increased level of fibrosis is a heart disease.
  • the disease caused by an increased level of fibrosis is a skin disease.
  • diseases caused by an increased level of fibrosis include, but are not limited to, chronic obstructive pulmonary disease, cystic fibrosis, idiopathic pulmonary fibrosis, emphysema, nephrogenic fibrosis, endometrial fibrosis, perineural fibrosis, hepatic fibrosis, myocardial fibrosis, acute lung injury, radiation-induced lung injury following treatment for cancer, progressive massive fibrosis, a complication of coal workers' pneumoconiosis (lungs), cirrhosis (liver), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, arterial stiffness (heart), glial scar (brain), arthrofibrosis (knee, shoulder, other joints), Crohn's Disease (intestine), Dupuytren's contracture (hands, fingers), keloid (skin), mediastinal fibrosis (soft tissue of
  • the present invention further provides methods for treating or preventing a disease associated with increased inflammation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the disease associated with increased inflammation is an autoimmune disease.
  • diseases associated with increased inflammation include, but are not limited to, multiple sclerosis, inflammatory bowel disease, celiac disease, Crohn's disease, antiphospholipid syndrome, atherosclerosis, autoimmune encephalomyelitis, autoimmune hepatitis, Graves' disease, ulcerative colitis, multiple sclerosis, myasthenia gravis, myositis, polymyositis, Raynaud's phenomenon, rheumatoid arthritis, scleroderma, Sjogren's syndrome, systemic lupus, type 1 diabetes and uveitis.
  • the disease associated with increased inflammation is multiple sclerosis, inflammatory bowel disease, celiac disease, or Crohn's disease.
  • the present invention further provides methods for preventing death from or increasing survival from a disease associated with increased inflammation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the disease associated with increased inflammation is influenza, sepsis, or a viral disease.
  • viral diseases include, but are not limited to, influenza, human immunodeficiency virus infection, hepatitis B, and hepatitis C.
  • the present invention further provides methods for treating or preventing an inflammation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the inflammation is indicated by an increased concentration of C-reactive protein in a patient's plasma or serum.
  • C-reactive protein related disorders include, but are not limited to, inflammation, ischemic necrosis, and a thrombotic disorder.
  • the present invention further provides methods for treating or preventing a sulfatase-2-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • sulfatase-2-related disorders include, but are not limited to, disorders of lipogenesis or lipid modulation, elevated plasma or blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention further provides methods for treating or preventing an apolipoprotein C-III-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • apolipoprotein C-III-related disorders include, but are not limited to, disorders of lipogenesis or lipid modulation, elevated plasma or blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention further provides methods for treating or preventing Alzheimer's disease, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for treating or preventing Parkinson's disease, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for treating or preventing pancreatitis, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for treating or preventing the risk of developing pancreatitis, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for treating or preventing a pulmonary disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the pulmonary disorder is a chronic obstructive pulmonary disease or an idiopathic pulmonary fibrosis.
  • the present invention further provides methods for treating or preventing musculoskeletal discomfort, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for reducing a subject's plasma or blood serum fibrinogen concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject's plasma or blood serum fibrinogen concentration is greater than 300 mg/dl. In some embodiments, the subject's plasma or blood serum fibrinogen concentration is greater than 400 mg/dl.
  • the present invention further provides methods for reducing a fibrosis score or a nonalcoholic fatty liver disease activity score in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the nonalcoholic fatty liver disease activity score (NAS or NAFLD score) is a composite score that measures changes in NAFLD during therapeutic trials.
  • NAS is a composite score comprised of three components that includes scores for steatosis, lobular inflammation and hepatocyte ballooning (Table 15).
  • NAS is the unweighted sum of the scores for steatosis, lobular inflammation and hepatocyte ballooning.
  • Steatosis grade is quantified as the percentage of hepatocytes that contain fat droplets.
  • the fibrosis stage of the liver is evaluated separately from NAS by histological evaluation of the intensity of Sirius red staining of collagen in the pericentral region of liver lobules.
  • the present invention provides methods for slowing the progression of a component of NAS, comprising administering to a subject in need thereof a compound of the invention.
  • the present invention provides methods for slowing the progression of a component of NAS, comprising administering to a subject in need thereof a composition of the invention.
  • the present invention provides methods for slowing the progression of steatosis, lobular inflammation, or hepatocyte ballooning, comprising administering to a subject in need thereof a compound of the invention.
  • the present invention provides methods for slowing the progression of steatosis, lobular inflammation, or hepatocyte ballooning, comprising administering to a subject in need thereof a composition of the invention.
  • the present invention provides methods for slowing the progression of steatosis, comprising administering to a subject in need thereof a compound of the invention or a composition of the invention.
  • the present invention provides methods for slowing the progression of lobular inflammation, comprising administering to a subject in need thereof a compound of the invention or a composition of the invention.
  • the present invention provides methods for slowing the progression of hepatocyte ballooning, comprising administering to a subject in need thereof a compound of the invention or a composition of the invention.
  • the present invention further provides methods for reducing elevated total cholesterol, low-density lipoprotein cholesterol (LDL-C), apolipoprotein B (Apo B), triglyceride or non-high-density lipoprotein cholesterol in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for increasing high-density lipoprotein cholesterol in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has primary hyperlipidemia.
  • the primary hyperlipidemia is heterozygous familial.
  • the primary hyperlipidemia is homozygous familial.
  • the primary hyperlipidemia isnon-familial.
  • the subject has mixed hyperlipidemia.
  • the present invention further provides methods for treating or preventing a condition or disease associated with hepatic overexpression of sulfatase-2 (Sulf-2) mRNA, comprising administering to a subject in need thereof an effect amount of a compound of the invention.
  • Sulf-2 inhibits hepatic disposal of C-TRLs, thereby increasing plasma or blood serum triglyceride concentration in a subject.
  • Conditions or diseases associated with hepatic overexpression of Sulf-2 include but are not limited to, elevated plasma or blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention further provides methods for treating or preventing a condition or disease associated with hepatic overexpression of ApoC-III mRNA, comprising administering to a subject in need thereof an effect amount of a compound of the invention.
  • overexpression of ApoC-III mRNA leads to increased plasma or blood serum triglyceride concentration in a subject.
  • Conditions or diseases associated with hepatic overexpression of ApoC-III include, but are not limited to, elevated blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention further provides methods for treating or preventing a condition or disease associated with hepatic overexpression of ANGPTL3 mRNA, comprising administering to a subject in need thereof an effect amount of a compound of the invention.
  • overexpression of ANGPTL3 mRNA leads to blockage of lipoprotein lipase activity and elevated plasma or blood serum triglyceride concentration in a subject.
  • Conditions or diseases associated with hepatic overexpression of ANGPTL3 include, but are not limited to, elevated blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention further provides methods for treating or preventing a condition or disease associated with hepatic overexpression of ANGPTL4 mRNA, comprising administering to a subject in need thereof an effect amount of a compound of the invention.
  • overexpression of ANGPTL4 mRNA leads to blockage of lipoprotein lipase activity and elevated plasma or blood serum triglyceride concentration in a subject.
  • Conditions or diseases associated with hepatic overexpression of ANGPTL4 include, but are not limited to, elevated blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention further provides methods for treating or preventing a condition or disease associated with hepatic overexpression of ANGPTL8 mRNA, comprising administering to a subject in need thereof an effect amount of a compound of the invention.
  • overexpression of ANGPTL8 mRNA leads to blockage of lipoprotein lipase activity and elevated plasma or blood serum triglyceride concentration in a subject.
  • Conditions or diseases associated with hepatic overexpression of ANGPTL8 include, but are not limited to, elevated blood serum triglycerides or hyperlipidemia, hypercholesterolemia, diabetes, fatty liver disease, obesity, atherosclerosis, and/or cardiovascular diseases.
  • the present invention provides methods for lowering a subject's blood plasma or blood serum LDL-C concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention or a composition of the invention.
  • the present invention further provides methods for reducing a subject's blood plasma or blood serum elevated total cholesterol or elevated LDL-C, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has homozygous familial hypercholesterolemia (HoFH).
  • the subject is known to have HoFH.
  • the subject has heterozygous familial hypercholesterolemia (HeFH).
  • the subject is known to have HeFH.
  • the therapeutic or prophylactic methods of the invention can further comprise administering an additional pharmaceutically active agent to a subject.
  • the therapeutic or prophylactic methods of the invention can further comprise administering two or more additional pharmaceutically active agents to a subject.
  • the subject is on a stable dose of statin.
  • the present invention provides methods for lowering a subject's LDL-C concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention, wherein the subject is on a stable dose of a statin.
  • the additional pharmaceutically active agent is a statin, lipid lowering agent, a PCSK9 inhibitor, Vitamin E, an ANGPTL3 inhibitor, an ANGPTL4 inhibitor, an ANGPTL8 inhibitor, a cholesterol absorption inhibitor, a ACC inhibitor, an ApoC-III inhibitor, an ACL inhibitor, a fish oil, a fibrate, a thyroid hormone beta receptor agonist, a farnesoid X receptor (FXR), a CCR2/CCR5 (C-C chemokine receptor types 2 (CCR2) and 5 (CCR5)) inhibitor or antagonist, a caspase protease inhibitor, an ASK-1 (Apoptosis signal-regulating kinase 1) inhibitor, a galectin-3 protein, a NOX (Nicotinamide adenine dinucleotide phosphate oxidase) inhibitor, an ileal bile acid transporter, a PPAR (peroxisome proliferator-activated receptor) agonist,
  • the therapeutic or prophylactic methods of the invention can further comprise administering a statin and ezetimibe.
  • the subject is undergoing gastric bypass surgery.
  • the present invention further provides methods for treating or preventing heterozygous familial hypercholesterolemia (HeFH), comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for treating or preventing atherosclerotic cardiovascular disease (ASCVD), comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the atherosclerotic cardiovascular disease is a clinical atherosclerotic cardiovascular disease.
  • the subject is an adult.
  • the subject is on statin therapy.
  • the statin therapy is maximally tolerated statin therapy.
  • the methods further comprise administering a statin to the subject.
  • the subject has abnormally high plasma or blood serum LDL-C.
  • the maximally tolerated statin therapy is insufficient to lower the subject's plasma or blood serum LDL-C. In some embodiment, the maximally tolerated statin therapy is insufficient to lower the subject's plasma or blood serum LDL-C to the subject's goal plasma or blood serum LDL-C concentration.
  • a subject's goal plasma or blood serum LDL-C concentration varies with the subject's risk factor or factors, pre-existing conditions, and/or health status.
  • LDL-C goal concentration for all human subjects, including human subjects with CHD (coronary heart disease) and other clinical forms of atherosclerotic disease should be less than 100 mg/dL.
  • a reasonable or a desirable LDL-C goal concentration for all human subject with CHD and other clinical forms of atherosclerotic disease can be less than 70 mg/dL (Smith et al. Circulation. 2006; 113:2363-2372).
  • the present invention further provides methods for treating or preventing HoFH, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is on one or more other low-density lipoprotein (LDL) lowering therapies.
  • the methods further comprise administering an LDL-lowering therapy to the subject.
  • LDL-lowering therapies include statins, ezetimibe and LDL apheresis.
  • the subject has abnormally high LDL-C.
  • the other LDL-lowering therapy is insufficient to lower the subject's LDL-C.
  • the other LDL-lowering therapy is insufficient to lower the subject's LDL-C to the subject's goal concentration.
  • the methods further comprise administering one or more additional pharmaceutically active agents, as disclosed herein.
  • the present invention further provides methods for reducing risk of a cardiovascular event, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has coronary heart disease (CHD).
  • the subject has a history of acute coronary syndrome (ACS).
  • ACS acute coronary syndrome
  • the subject has been previously treated with a statin. In other embodiments, the subject has not been previously treated with a statin.
  • the present invention further provides methods for treating or preventing primary hypercholesterolemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the primary hypercholesterolemia can be HeFH or non-familial hypercholesterolemia.
  • the present invention further provides methods for treating or preventing mixed hyperlipidemia in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject or the subject's symptoms are not effectively treated with statin therapy alone.
  • statin therapy alone means that the subject's plasma or blood serum LDL-C is not lowered to the subject's goal concentration with a given treatment.
  • the subject had been administered with a statin and/or ezetimibe prior to administration of a compound of the invention.
  • the subject was treated with a statin and/or ezetimibe previously, prior to administration of a compound of the invention.
  • the methods further comprise administering a one or both of a statin and ezetimibe to the subject.
  • the present invention further provides methods for treating or preventing HoFH, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the method further comprises administering an adjunctive treatment.
  • the adjunctive treatment can be one or more of a statin, ezetimibe and LDL apheresis.
  • the adjunctive treatment is LDL-lowering therapy.
  • the adjunctive treatment can be one or more of a statin, ezetimibe, LDL apheresis, PCSK9 inhibitor, and bile acid sequestrant.
  • the adjunctive treatment can be one or more of a statin, ezetimibe, LDL apheresis, PCSK9 inhibitor, bile acid sequestrant, lomitapide (Juxtapid®) and mipomersen (Kynamro®).
  • the adjunctive treatment can be one or more additional pharmaceutically active agents, as disclosed herein.
  • the present invention further provides methods for reducing risk of having myocardial infarction, having a stroke, needing a revascularization procedure or having angina, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject does not have coronary heart disease (CHD).
  • CHD coronary heart disease
  • the subject has one or more risk factors for CHD.
  • risk factors for CHD include, but are not limited to, high plasma or blood serum cholesterol, high plasma or blood serum triglyceride, high blood pressure, diabetes, prediabetes, overweight or obesity, smoking, lack of physical activity, unhealthy diets, stress.
  • age, gender, and family history of early CHD can be a risk factor for CHD.
  • the present invention further provides methods for reducing a subject's risk of myocardial infarction or stroke, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has type 2 diabetes.
  • the subject has type 2 diabetes and does not have CHD.
  • the subject has one or more risk factors for CHD.
  • the present invention further provides methods for reducing a subject's risk of non-fatal myocardial infarction, risk of fatal stroke or non-fatal stroke, need for a revascularization procedure, risk of congestive heart failure (CHF) or risk of angina, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has CHD.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated total cholesterol, LDL-C, Apo B or triglyceride concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for increasing high-density lipoprotein cholesterol in a subject, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is an adult.
  • the subject has primary hyperlipidemia.
  • Primary hyperlipidemia can be heterozygous familial or non-familial.
  • the subject has mixed dyslipidemia.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated triglyceride concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has hypertriglyceridemia.
  • the subject has primary dysbetalipoproteinemia.
  • the subject has hypoalphalipoproteinemia.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum total cholesterol or LDL-C concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has HoFH.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated total cholesterol, LDL-C or Apo B concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is a human male or a human female (e.g., postmenarcheal female) who is 10-17 years of age.
  • the subject has HeFH.
  • the subject's diet is insufficient to reduce the subject's elevated total cholesterol, LDL-C or Apo B.
  • the subject's life-style or diet and life-style is insufficient to reduce the subject's elevated total cholesterol, LDL-C or Apo B.
  • the present invention further provides methods for reducing a subject's risk of mortality, CHD death, non-fatal myocardial infarction, stroke or need for a revascularization procedure, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is at high risk of a coronary event.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated total cholesterol, LDL-C, Apo B or triglyceride concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for increasing in a subject's blood plasma or blood serum high-density lipoprotein cholesterol, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has primary hyperlipidemia.
  • the primary hyperlipidemia is HeFH.
  • the primary hyperlipidemia is non-familial hyperlipidemia.
  • the subject has mixed dyslipidemia.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated triglyceride concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has hypertriglyceridemia.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum triglyceride or very-low-density lipoprotein cholesterol (VLDL-C), comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has primary dysbetalipoproteinemia.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated total cholesterol or LDL-C concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is an adult.
  • the subject has HoFH.
  • the present invention further provides methods for treating or preventing hypertriglyceridemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the method further comprises adjusting the subject's diet.
  • the method further comprises placing the subject on a low-fat diet.
  • the present invention further provides methods for treating or preventing primary dysbetalipoproteinemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the primary dysbetalipoproteinemia is Type III hyperlipoproteinemia.
  • the method further comprises adjusting the subject's diet.
  • the method further comprises placing the subject on a low-fat diet.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum total cholesterol, LDL-C or Apo B concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has HoFH.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated LDL-C, total cholesterol, Apo B or triglyceride concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the present invention further provides methods for increasing in a subject's blood plasma or blood serum high-density lipoprotein cholesterol concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is an adult.
  • the subject has primary hypercholesterolemia.
  • the subject has mixed dyslipidemia.
  • the present invention further provides methods for treating or preventing severe hypertriglyceridemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is an adult.
  • the present invention further provides methods for reducing the rate or incidence of myocardial infarction or stroke, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has acute coronary syndrome (ACS).
  • the subject has non-ST-segment elevation ACS (unstable angina (UA)/non-ST-elevation myocardial infarction (NSTEMI)).
  • the subject has ST-elevation myocardial infarction (STEMI).
  • the ST segment connects the QRS complex and the T wave.
  • the subject has had a previous myocardial infarction, previous stroke or established peripheral arterial disease.
  • the subject has had a recent myocardial infarction or recent stroke. In some embodiments, recent myocardial infarction or a recent stroke took event within one year. In some embodiments, recent myocardial infarction or a recent stroke took event within three months.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum total cholesterol, LDL-C or Apo B concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has primary hypercholesterolemia.
  • Primary hypercholesterolemia can be heterozygous familial or non-familial.
  • the method further comprises administering an HMG-CoA reductase inhibitor to the subject.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum total cholesterol or LDL-C concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has HoFH.
  • the method further comprises administering an additional lipid-lowering treatment to the subject.
  • the additional lipid-lowering treatment may be a statin (e.g., atorvastatin or simvastatin) or LDL apheresis.
  • the present invention further provides methods for reducing in a subject's blood plasma or blood serum elevated sitosterol or campesterol concentration, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has homozygous familial sitosterolemia.
  • the present invention further provides methods for treating or preventing Type IV or Type V hyperlipidemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has a risk of pancreatitis.
  • a change in the subject's diet does not adequately lower the subject's plasma or blood serum triglyceride concentrations.
  • a normal blood serum triglyceride concentration is less than 150 mg/dL according to ATP III Classification of serum triglycerides (National Institute of Health Publication No. 01-3305; May 2001; Cholesterol Guidelines).
  • the subject has an abnormally high serum triglyceride concentration.
  • the subject has a blood serum triglyceride concentration of over 2000 mg/dL and optionally has an elevation of VLDL-cholesterol or has fasting chylomicronemia. In some embodiments, the subject has a triglyceride of from 1000 to 2000 mg/dL and optionally has a history of pancreatitis or of recurrent abdominal pain typical of pancreatitis.
  • the present invention further provides methods for reducing risk of developing coronary heart disease, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject has Type IIb hyperlipidemia.
  • the subject does not have history of or symptoms of existing coronary heart disease.
  • the subject has had weight loss, dietary therapy, exercise, or was administered another pharmacologic agent (e.g., a bile acid sequestrant or nicotinic acid) that was ineffective to treat the subject's hyperlipidemia.
  • the subject has in a subject's blood plasma or blood serum, one or more of an abnormally low HDL-cholesterol concentration, an abnormally high LDL-cholesterol concentration and an abnormally high triglyceride concentration.
  • the therapeutic or prophylactic methods of the invention further comprise administering an effective amount of an additional pharmaceutically active agent. In some embodiments, the therapeutic or prophylactic methods of the invention further comprise administering an effective amount of two or more additional pharmaceutically active agent.
  • the additional pharmaceutically active agent is a statin.
  • statin is atorvastatin, simvastatin, pravastatin, rosuvastatin, fluvastatin, lovastatin, pitavastatin, mevastatin, dalvastatin, dihydrocompactin, or cerivastatin, or a pharmaceutically acceptable salt thereof.
  • the statin is atorvastatin calcium.
  • the additional pharmaceutically active agent is a statin. In some embodiments, the additional pharmaceutically active agent is an HMG-CoA (3-hydroxy-3-methyl-glutaryl-coenzyme A) reductase inhibitor.
  • the additional pharmaceutically active agent is a lipid modifying agent, lipid lowering agent, anti-fibrolytic agent, or an anti-inflammatory agent.
  • the additional pharmaceutically active agent is a cholesterol lowering agent.
  • the additional pharmaceutically active agent is a cholesterol absorption inhibitor.
  • the cholesterol absorption inhibitor is ezetimibe.
  • the additional pharmaceutically active agent is a PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor, Vitamin E, an ANGPTL3 inhibitor, an ANGPTL4 inhibitor, an ANGPTL8 inhibitor, a cholesterol absorption inhibitor, an ACC (acetyl-CoA carboxylase) inhibitor, an ApoC-III (apolipoprotein C-III) inhibitor, an ApoB (apolipoprotein B) synthesis inhibitor, an ACL (adenosine triphosphate citrate lyase) inhibitor, a microsomal transfer protein inhibitor, a fenofibric acid, a fish oil, a fibrate, a thyroid hormone beta receptor agonist, a farnesoid X receptor (FXR), a CCR2/CCR5 (C-C chemokine receptor types 2 (CCR2) and 5 (CCR5)) inhibitor or antagonist, a caspase protease inhibitor, an ASK-1 (Apoptosis signal-regulating PCSK9 (
  • the additional lipid lowering agent is PCSK9 inhibitor.
  • the additional lipid lowering agent is bempedoic acid, nicotinic acid, gemfibrozil, niacin, a bile-acid resin, a fibric acid derivative, or a cholesterol absorption inhibitor.
  • the additional lipid lowering agent is bempedoic acid, nicotinic acid, or gemfibrozil.
  • the lipid-reducing agent is gemfibrozil.
  • the one or more pharmaceutically active agent is bempedoic acid.
  • fish oils examples include, but are not limited to, salmon oil, sardine oil, cod liver oil, tuna oil, herring oil, menhaden oil, mackerel oil, refined fish oils, and mixtures thereof.
  • Fish oils comprise omega-3 fatty acids: eicosapentaenoic acid and docosahexaenoic acid.
  • the fish oil is prescription fish oil.
  • the eicosapentaenoic acid is enriched or esterified, such as, but not limited to an ethyl ester.
  • the eicosapentaenoic acid is enriched and esterified.
  • the CETP inhibitor is dalcetrapib (CAS 211513-37-0), torcetrapib (CAS 262352-17-0), anacetrapib (CAS 875446-37-0), evacetrapib (CAS 1186486-62-3), BAY 60-5521 (CAS 893409-49-9), obicetrapib (866399-87-3), ATH-03 (Affris), DRL-17822 (Dr.
  • the additional pharmaceutically active agent is an antibody to CETP.
  • the antibody to CETP is a monoclonal antibody.
  • the antibody to CETP is a monoclonal antibody (Mab, TP1) to CETP.
  • the additional pharmaceutically active agent is an antibody against CETP. In some embodiments, the additional pharmaceutically active agent induces antibodies against CETP and is a vaccine. In some embodiments, the vaccine is TT/CETP (Rittershaus, C. W. et al., Arteriosclerosis, Thrombosis, and Vascular Biology. 2000; 20:2106-2112). In other embodiments, the additional pharmaceutically active agent induces antibodies against CETP and is CETi-1 (Celldex Therapeutics).
  • the additional pharmaceutically active agent immunizes a subject with CETP or CETP protein fragment.
  • the additional pharmaceutically active agent reduces CETP by inhibition with an SiRNA to CETP mRNA.
  • the additional pharmaceutically active agent targets CETP transcription by administration of DNAi to the CETP gene. In other embodiments, the additional pharmaceutically active agent targets CETP transcription by administration of DNAi in an appropriate deliver vehicle such as a SmarticleTM.
  • the additional pharmaceutically active agent is an anti-coagulation agent or a lipid regulating agent.
  • the anti-coagulation agent is aspirin, dabigatran, rivaroxaban, apixaban clopidogrel, clopNPT (conjugate of clopidogrel with 3-nitropyridine-2-thiol), prasugrel, ticagrelor, cangrelor, a platelet P2Y 12 receptor inhibitor, thienopyridine, warfarin (Coumadin) acenocoumarol, phenprocoumon, atromentin, phenindione, edoxaban betrixaban, letaxaban eribaxaban hirudin, lepirudin, bivalirudin, argatroban, dabigatran, ximelagatran, batroxobin, hementin, a heparin or vitamin E.
  • the additional pharmaceutically active agent is sizumab (CAS 1318075-13-6), selonsertib (CAS 1448428-04-3), GS-9674 (Gilead Sciences), GS-0976 (Gliead Sciences), obeticholic acid (CAS 459789-99-2; Intercept), or cenicriviroc (CAS 497223-25-3; Allergan-Takeda), or pharmaceutically acceptable salt thereof.
  • the additional pharmaceutically active agent is, but is not limited to, elafibranor (Genfit), seladelpar (Cymabay), or EDP-305 (Enanta Pharmaceuticals).
  • the additional pharmaceutically active agent is an anti-inflammatory agent, an anti-hypertensive agent, an anti-diabetic agent, an anti-obesity, an anti-fibrotic or an anti-coagulation agent.
  • the additional pharmaceutically active agent disclosed herein can be a pharmaceutically acceptable salt thereof.
  • the pharmaceutically acceptable salt can be an acid addition salt where the pharmaceutically active agent is basic, e.g., includes a basic nitrogen atom, and can be a cationic salt.
  • the pharmaceutically acceptable salt can be a base addition salt where the pharmaceutically active agent is acidic.
  • the therapeutic or prophylactic methods of the invention do not induce hepatotoxicity or a musculoskeletal disorder.
  • a subject to which a compound of the invention or composition of the invention is administered is on statin therapy.
  • the statin is atorvastatin, simvastatin, pravastatin, rosuvastatin, fluvastatin, lovastatin, pitavastatin, mevastatin, dalvastatin, dihydrocompactin, or cerivastatin, or a pharmaceutically acceptable salt thereof.
  • the statin is atorvastatin calcium.
  • the therapeutic or prophylactic methods of the invention comprises administering to a subject in need thereof an effective amount of a compound of the invention. In some embodiments, any one of the therapeutic or prophylactic methods as disclosed herein can comprise administering to a subject in need thereof an effective amount of a composition of the invention in place of an effective amount of a compound of the invention. In some embodiments, any one of the therapeutic or prophylactic methods as disclosed herein can comprise administering to a subject in need thereof an effective amount of a composition of the invention.
  • compositions of the invention comprise (i) an effective amount of a compound of the invention and (ii) a pharmaceutically acceptable carrier or vehicle.
  • compositions of the invention further comprise an effective amount of an additional pharmaceutically active agent, such as disclosed herein. In other embodiments, the compositions of the invention further comprise an effective amount of two or more additional pharmaceutically active agent as disclosed herein.
  • the pharmaceutically acceptable carrier or vehicle includes, but is not limited to, a binder, filler, diluent, disintegrant, wetting agent, lubricant, glidant, coloring agent, dye-migration inhibitor, sweetening agent or flavoring agent.
  • Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression.
  • Suitable binders or granulators include, but are not limited to, starches, such as corn starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, Panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyeth
  • Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder is hydroxypropylcellulose.
  • the binder or filler can be present from about 2% to about 49% by weight of the compositions of the invention provided herein or any range within these values. In some embodiments, the binder or filler is present in the composition of the invention from about 5% to about 15% by weight. In some embodiments, the binder or filler is present in the composition of the invention at about 5%, 6%, 7%, 8%, 9%, 8%, 10%, 11%, 12%, 13%, 14%, or 15% by weight or any range within any of these values.
  • Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar.
  • Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.
  • the diluent is lactose monohydrate.
  • the diluent is lactose monohydrate Fast-Flo 316 NF.
  • compositions of the invention can comprise from about 5% to about 49% of a diluent by weight of composition or any range between any of these values.
  • the diluent is present in the compositions of the invention from about 15% to about 30% by weight.
  • the diluent is present in the composition of the invention at about 15%, 16%, 17%, 18%, 19%, 18%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, or 30% by weight or any range within any of these values.
  • Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof.
  • the amount of disintegrant in the compositions of the invention can vary.
  • the disintegrant is croscarmellose sodium.
  • the disintegrant is croscarmellose sodium NF (Ac-Di-Sol).
  • compositions of the invention can comprise from about 0.5% to about 15% or from about 1% to about 10% by weight of a disintegrant.
  • the compositions of the invention comprise a disintegrant in an amount of about 5%, 6%, 7%, 8%, 9%, 8%, 10%, 11%, 12%, 13%, 14%, or 15% by weight of the composition or in any range within any of these values.
  • Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL® R 200 (W.R. Grace Co., Baltimore, Md.) and CAB-O-SIL® (Cabot Co. of Boston, Mass.); and mixtures thereof.
  • the lubricant is magnesium stearate.
  • compositions can of the invention can comprise about 0.1 to about 5% by weight of a lubricant.
  • the compositions of the invention comprise a lubricant in an amount of about 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 0.8%, 1.0%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2.0%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, or 3.0%, by weight of the composition or in any range within any of these values.
  • Suitable glidants include colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston, Mass.), and talc, including asbestos-free talc.
  • Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof.
  • Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds that provide a pleasant taste sensation, such as peppermint and methyl salicylate.
  • Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, sucralose, and artificial sweeteners, such as saccharin and aspartame.
  • Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate.
  • Suspending and dispersing agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrolidone.
  • Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether.
  • Solvents include glycerin, sorbitol, ethyl alcohol, and syrup.
  • non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil.
  • Organic acids include citric and tartaric acid.
  • Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
  • the compounds of the invention and the compositions of the invention can be formulated for administration by a variety of means including orally, parenterally, by inhalation spray, topically, or rectally in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used here includes subcutaneous, intravenous, intramuscular, and intraarterial injections with a variety of infusion techniques. Intraarterial and intravenous injection as used herein includes administration through catheters.
  • compositions of the invention can be formulated in accordance with the routine procedures adapted for desired administration route. Accordingly, the compositions of the invention can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the compounds of the invention and the compositions of the invention can be formulated as a preparation suitable for implantation or injection.
  • pharmaceutically acceptable salt of gemcabene and the compositions of the invention can be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt).
  • suitable polymeric or hydrophobic materials e.g., as an emulsion in an acceptable oil
  • ion exchange resins e.g., as a sparingly soluble salt
  • sparingly soluble derivatives e.g., as a sparingly soluble salt
  • suitable vehicle e.g., sterile pyrogen-free water
  • compositions of the invention are suitable for oral administration. These compositions can comprise solid, semisolid, gelmatrix or liquid dosage forms suitable for oral administration. As used herein, oral administration includes buccal, lingual, and sublingual administration. Suitable oral dosage forms include, without limitation, tablets, capsules, pills, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, granules, bulk powders, effervescent or non-effervescent powders or granules, solutions, emulsions, suspensions, solutions, wafers, sprinkles, elixirs, syrups or any combination thereof. In some embodiments, compositions of the invention suitable for oral administration are in the form of a tablet or a capsule. In some embodiments, the composition of the invention is in a form of a tablet. In some embodiments, the composition of the invention is in a form of a capsule. In some embodiments, the compound of the invention is contained in a capsule.
  • capsules are immediate release capsules.
  • Non-limiting example of a capsule is a Coni-snap® hard gelatin capsule.
  • compositions of the invention can be in the form of compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets.
  • Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach.
  • Enteric-coatings include, but are not limited to, fatty acids, fats, phenylsalicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates.
  • Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
  • Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
  • Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate.
  • a film coating can impart the same general characteristics as a sugar coating.
  • Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
  • the coating is a film coating.
  • the film coating comprises Opadry White and simethicone emulsion 30% USP.
  • the compound of the invention is contained in a tablet. In some embodiments, the compound of the invention is contained in a compressed tablet. In some embodiments, the compound of the invention is contained in a film-coated compressed tablet. In some embodiments, the compositions of the invention are in the form of film-coated compressed tablets.
  • the compositions of the invention is prepared by fluid bed granulation of the compound of the invention with one or more pharmaceutically acceptable carrier, vehicle, or excipients.
  • the compositions of the invention prepared by fluid bed granulation process can provide tablet formulation with good flowability, good compressibility, fast dissolution, good stability, and/or minimal to no cracking.
  • the fluid bed granulation process allows preparation of formulations having high drug loading, such as over 70% or over 75% of a compound of the invention.
  • compositions of the invention can be in the form of soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate.
  • the hard gelatin capsule also known as the dry-filled capsule (DFC)
  • DFC dry-filled capsule
  • the soft elastic capsule is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol.
  • the soft gelatin shells can contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid.
  • liquid, semisolid, and solid dosage forms can be encapsulated in a capsule.
  • suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides.
  • Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545.
  • the capsules can also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • compositions of the invention can be in liquid or semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups.
  • An emulsion can be a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil.
  • Emulsions can include a pharmaceutically acceptable non-aqueous liquids or solvent, emulsifying agent, and preservative.
  • Suspensions can include a pharmaceutically acceptable suspending agent and preservative.
  • Aqueous alcoholic solutions can include a pharmaceutically acceptable acetal, such as a di-(lower alkyl)acetal of a lower alkyl aldehyde (the term “lower” means an alkyl having between 1 and 6 carbon atoms), e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol.
  • Elixirs can be clear, sweetened, and hydroalcoholic solutions.
  • Syrups can be concentrated aqueous solutions of a sugar, for example, sucrose, and can comprise a preservative.
  • a solution in a polyethylene glycol can be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
  • compositions of the invention for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems.
  • Miccellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.
  • compositions of the invention can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form.
  • Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders can include diluents, sweeteners, and wetting agents.
  • Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders can include organic acids and a source of carbon dioxide.
  • Coloring and flavoring agents can be used in all of the above dosage forms. And, flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.
  • compositions of the invention can be formulated as immediate or modified release dosage forms, including delayed-, extended, pulsed-, controlled, targeted-, and programmed-release forms.
  • compositions of the invention comprise a film-coating.
  • compositions of the invention can comprise another active ingredient that does not impair the composition's therapeutic or prophylactic efficacy or can comprise a substance that augments or supplements the composition's efficacy.
  • the tablet dosage forms can comprise a pharmaceutically acceptable salt of gemcabene in powdered, crystalline, or granular form, and can further comprise a carrier or vehicle described herein, including binder, disintegrant, controlled-release polymer, lubricant, diluent, or colorant.
  • the compositions of the invention comprise from about 50 mg to about 900 mg, about 150 mg to about 600 mg, or about 150 mg to about 300 mg of a compound of the invention.
  • the compositions of the invention comprise a compound of the invention in an amount of about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, about 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, about 400 mg, about 410 mg, about 420 mg, about 430 mg, about 440 mg, about 450 mg, about
  • compositions of the invention comprise about 50 mg of a compound of the invention. In some embodiments, the compositions of the invention comprise about 150 mg of a compound of the invention. In some embodiments, the compositions of the invention comprise about 300 mg of a compound of the invention. In some embodiments, the compositions of the invention comprise about 600 mg of a compound of the invention.
  • compositions of the invention comprise a compound of the invention in an amount that is a molar equivalent to 50 mg to about 900 mg, about 150 mg to about 600 mg, or about 150 mg to about 300 mg of gemcabene. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is a molar equivalent to about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, about 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, about 400 mg, about 410 mg,
  • compositions of the invention comprise a pharmaceutically acceptable salt of gemcabene in an amount that is a molar equivalent to about 50 mg. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is a molar equivalent to about 150 mg of gemcabene. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is a molar equivalent to about 300 mg. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is a molar equivalent to about 600 mg.
  • compositions of the invention comprise a compound of the invention in an amount of about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325 mg, about 350 mg, about 375 mg, about 400 mg, about 425 mg, about 450 mg, about 475 mg, about 500 mg, about 525 mg, about 550 mg, about 575 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, or any amount ranging from and to these values.
  • the compound of the invention is gemcabene calcium salt hydrate Crystal Form 1. In some embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form 2. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C1. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C2. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C3. In some embodiments, the compound of the invention is an amorphous gemcabene calcium salt hydrate.
  • compositions of the invention comprise a compound of the invention in an amount that is a molar equivalent to about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325 mg, about 350 mg, about 375 mg, about 400 mg, about 425 mg, about 450 mg, about 475 mg, about 500 mg, about 525 mg, about 550 mg, about 575 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, or about 900 mg gemcabene, or any amount ranging from and to these values.
  • the compound of the invention is gemcabene calcium salt hydrate Crystal Form 1. In some embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form 2. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C1. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C2. In other embodiments, the compound of the invention is gemcabene calcium salt hydrate Crystal Form C3. In some embodiments, the compound of the invention is an amorphous gemcabene calcium salt hydrate.
  • compositions of the invention are in the form of a tablet or a capsule.
  • the compositions of the invention comprise a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule.
  • the compositions of the invention comprise a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule.
  • the tablet or the capsule comprises about 50 mg of a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In one aspect, the tablet or the capsule comprises about 50 mg of a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In one aspect, the tablet or the capsule comprises about 50 mg of a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 50 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 50 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 50 mg of gemcabene.
  • the tablet or the capsule comprises about 150 mg of a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In one aspect, the tablet or the capsule comprises about 150 mg of a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In one aspect, the tablet or the capsule comprises about 150 mg of a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 150 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 150 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 150 mg of gemcabene.
  • the tablet or the capsule comprises about 300 mg of a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m In some embodiments, the tablet or the capsule comprises about 300 mg of a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m In some embodiments, the tablet or the capsule comprises about 300 mg of a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 300 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 300 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 300 mg of gemcabene.
  • the tablet or the capsule comprises about 600 mg of a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 600 mg of a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 600 mg of a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 600 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 600 mg of gemcabene. In some embodiments, the tablet or the capsule comprises a compound of the invention having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m in an amount that is a molar equivalent to about 600 mg of gemcabene.
  • the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule.
  • the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 150 mg of gemcabene.
  • the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 300 mg of gemcabene.
  • the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 600 mg of gemcabene.
  • the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In other embodiments, the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m. In other embodiments, the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 900 mg of gemcabene.
  • compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule.
  • the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 150 mg of gemcabene.
  • the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 300 mg of gemcabene.
  • the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 600 mg of gemcabene.
  • the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m.
  • the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form 2 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 900 mg of gemcabene.
  • compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 50 ⁇ m to about 75 ⁇ m and are in the form of a tablet or a capsule.
  • the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 150 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 150 mg of gemcabene.
  • the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 300 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 300 mg of gemcabene.
  • the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 600 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 600 mg of gemcabene.
  • the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m. In some embodiments, the tablet or the capsule comprises about 900 mg of gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 45 ⁇ m to about 75 ⁇ m or from 50 ⁇ m to about 75 ⁇ m In some embodiments, the tablet or the capsule comprises gemcabene calcium salt hydrate Crystal Form C3 having a PSD90 ranging from 40 ⁇ m to about 75 ⁇ m, from 45 ⁇ m to about 75 ⁇ m, or from 50 ⁇ m to about 75 ⁇ m, in an amount that is molar equivalent to about 900 mg of gemcabene.
  • compositions of the invention comprise a compound of the invention in an amount of about 38.5 wt % to about 99.9 wt %, about 79 wt % to about 98 wt %, about 65% to about 98 wt %, or about 50 wt % to about 70 wt % of the total weight of the pharmaceutical composition.
  • compositions of the invention comprise a compound of the invention in an amount of about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about 70%, about 71%, about 72%, about 73%, about 74%, about 75%, about 76%, about 77%, about 78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 96%, about 98%
  • the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 in an amount of about 38.5 wt % to about 99.9 wt %, about 79 wt % to about 98 wt %, about 65% to about 98 wt %, or about 50 wt % to about 70 wt % of the total weight of the pharmaceutical composition.
  • the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 2 in an amount of about 38.5 wt % to about 99.9 wt %, about 79 wt % to about 98 wt %, about 65% to about 98 wt %, or about 50 wt % to about 70 wt % of the total weight of the pharmaceutical composition.
  • the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form C3 in an amount of about 38.5 wt % to about 99.9 wt %, about 79 wt % to about 98 wt %, about 65% to about 98 wt %, or about 50 wt % to about 70 wt % of the total weight of the pharmaceutical composition.
  • compositions of the invention comprise amorphous gemcabene calcium salt hydrate in an amount of about 38.5 wt % to about 99.9 wt %, about 79 wt % to about 98 wt %, about 65% to about 98 wt %, or about 50 wt % to about 70 wt % of the total weight of the pharmaceutical composition.
  • compositions of the invention further comprise another pharmaceutically active agent. In some embodiments, the compositions of the invention further comprise about 0.1 mg to about 100 mg, about 5 mg to about 80 mg, about 10 mg to about 60 mg or about 10 mg to about 40 mg of a statin or a pharmaceutically acceptable salt thereof.
  • compositions of the invention comprise a statin or a pharmaceutically acceptable salt thereof in an amount of about 0.1 mg, about 0.2 mg, about 0.3 mg, about 0.4 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg, about 30 mg, 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, 41 mg, about 42 mg, about 43 mg, about 44 mg, about 45 mg, about 46 mg, about 47
  • a composition of the invention comprising a compound of the invention further comprises a statin or a pharmaceutically acceptable salt thereof in an amount of about 0.001 wt % to about 75 wt %, about 0.005 wt % to about 61.5 wt %, about 2 wt % to about 35 wt %, or about 2 wt % to about 21 wt % of the composition.
  • compositions of the invention comprise a statin or a pharmaceutically acceptable salt thereof in an amount of about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%,
  • compositions of the invention comprise a statin or a pharmaceutically acceptable salt thereof in an amount of about 61%, about 61.1%, about 61.2%, about 61.3%, about 61.4%, about 61.5%, about 61.6%, about 61.7%, about 61.8%, about 61.9%, or about 62.0%, by weight of the composition, or an amount ranging from and to these values.
  • compositions of the invention further comprise about 0.1 mg to about 50 mg, about 1 mg to about 30 mg, about 5 mg to about 20 mg or about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof.
  • the compositions of the invention comprise ezetimibe or a pharmaceutically acceptable salt thereof in an amount of about 0.1 mg, about 0.2 mg, about 0.3 mg, about 0.4 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg, about 30 mg, 31 mg, about 32 mg,
  • compositions of the invention further comprise two pharmaceutically active agents.
  • the compositions of the invention further comprise a) about 0.1 mg to about 50 mg, about 1 mg to about 30 mg, about 5 mg to about 20 mg or about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof and b) about 0.1 mg to about 100 mg, about 5 mg to about 80 mg, about 10 mg to about 60 mg or about 10 mg to about 40 mg of a statin or a pharmaceutically acceptable salt thereof.
  • compositions of the invention comprise a) ezetimibe or a pharmaceutically acceptable salt thereof in an amount of about 0.1 mg, about 0.2 mg, about 0.3 mg, about 0.4 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg, about 30 mg, 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, 41 mg, about 42 mg, about 43 mg, about 44 mg, about 45 mg, about 46
  • a composition of the invention comprising gemcabene calcium salt hydrate Crystal Form 1, gemcabene calcium salt hydrate Crystal Form 2 or gemcabene calcium salt hydrate Crystal Form C3 further comprises a statin or a pharmaceutically acceptable salt thereof in an amount of about 0.001 wt % to about 75 wt %, about 0.005 wt % to about 61.5 wt %, about 2 wt % to about 35 wt %, or about 2 wt % to about 21 wt % of the composition.
  • the compositions of the invention comprise a compound of the invention in an amount of about 50 mg to about 900 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 1 mg to about 80 mg. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount of about 150 mg to about 600 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 10 mg to about 40 mg. In some embodiments, the compositions of the invention a compound of the invention in an amount of about 150 mg to about 300 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 10 mg to about 40 mg. In some embodiments, the compositions of the invention a compound of the invention in an amount of about 150 mg to about 900 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 10 mg to about 60 mg.
  • the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 in an amount of about 50 mg to about 900 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 1 mg to about 80 mg. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 in an amount of about 150 mg to about 600 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 10 mg to about 40 mg. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 in an amount of about 150 mg to about 300 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 10 mg to about 40 mg. In some embodiments, the compositions of the invention comprise gemcabene calcium salt hydrate Crystal Form 1 in an amount of about 150 mg to about 900 mg and statin or a pharmaceutically acceptable salt thereof in an amount of about 10 mg to about 60 mg.
  • compositions of the invention comprise a compound of the invention in an amount of about 38.5 wt % to about 99.9 wt % and a statin or a pharmaceutically acceptable salt thereof in an amount of about 0.1 wt % to about 61.5 wt % of the composition.
  • the compositions of the invention comprise a compound of the invention in an amount of about 65 wt % to about 98 wt % and a statin or a pharmaceutically acceptable salt thereof in an amount of about 2 wt % to about 35 wt % of the composition.
  • compositions of the invention comprise a compound of the invention in an amount of about 79 wt % to about 98 wt % and a statin or a pharmaceutically acceptable salt thereof in an amount of about 2 wt % to about 21 wt % of the composition.
  • the pharmaceutically acceptable salt is a calcium salt.
  • the calcium salt is a calcium salt hydrate.
  • the calcium salt hydrate is calcium salt hydrate Crystal Form 1.
  • the additional pharmaceutically active agent is present in an amount of about 10 mg to 100 mg or about 5 mg to 50 mg in the compositions of the invention. In some embodiments, the additional pharmaceutically active agent is present in an amount of about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, or any range between any of these values in the compositions of the invention.
  • compositions of the invention can further comprise an excipient such as a diluent, a disintegrant, a wetting agent, a binder, a glidant, a lubricant, or any combination thereof.
  • a tablet comprises a binder.
  • the binder comprises microcrystalline cellulose, dibasic calcium phosphate, sucrose, corn starch, polyvinylpyrridone, hydroxypropyl cellulose, hydroxymethyl cellulose, or any combination thereof.
  • the tablet comprises a disintegrant.
  • the disintegrant comprises sodium croscarmellose, sodium starch glycolate, or any combination thereof.
  • the tablet comprises a lubricant.
  • the lubricant comprises magnesium stearate stearic acid, hydrogenated oil, sodium stearyl fumarate, or any combination thereof.
  • compositions of the invention are in the form of a tablet that comprises a binder such as any of the binders described herein.
  • compositions of the invention are in the form of a tablet that comprises a disintegrant such as any of the disintegrants described herein.
  • compositions of the invention are in the form of a tablet that comprises a lubricant such as any of the lubricants described herein.
  • the compositions of the invention can be in a modified release or a controlled release dosage form.
  • the compositions of the invention can comprise particles exhibiting a particular release profile.
  • the composition of the invention can comprise a compound of the invention in an immediate release form while also comprising a statin or a pharmaceutically acceptable salt thereof in a modified release form, both compressed into a single tablet.
  • Other combination and modification of release profile can be achieved as understood by one skilled in the art. Examples of modified release dosage forms suited for pharmaceutical compositions of the instant invention are described, without limitation, in U.S. Pat. Nos.
  • the compositions of the invention are a matrix-controlled release dosage form.
  • the compositions of the invention can comprise about 300 mg to about 600 mg of a compound of the invention provided as a matrix-controlled release form.
  • a matrix-controlled release form can further comprise an additional pharmaceutically active agent.
  • the release profile of the compound of the invention and of the additional pharmaceutically active agent is the same or different. Suitable matrix-controlled release dosage forms are described, for example, in Takada et al in “Encyclopedia of Controlled Drug Delivery,” Vol. 2, Mathiowitz ed., Wiley, 1999.
  • compositions of the invention comprise from about 10 mg to about 40 mg of the statin and from about 300 mg to about 600 mg of a compound of the invention, wherein the composition is in a matrix-controlled modified release dosage form.
  • the matrix-controlled release form comprises an erodible matrix comprising water-swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • the erodible matrix of the matrix-controlled release form comprises chitin, chitosan, dextran, or pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, or scleroglucan; starches, such as dextrin or maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), carrrboxymethyl ethyl cellulose (CMEC) hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose
  • compositions of the invention are in a matrix-controlled modified release form comprising a non-erodible matrix.
  • the statin, the compound of the invention is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered.
  • the non-erodible matrix of the matrix-controlled release form comprises an insoluble polymer, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, a methyl acrylate-methyl methacrylate copolymer, an ethylene-vinylacetate copolymer, an ethylene/propylene copolymer, an ethylene/ethyl acrylate copolymer, a vinylchloride copolymer with vinyl acetate, a vinylidene chloride, an ethylene or a propylene, an ionomer polyethylene terephthalate, a butyl rubber epichlorohydrin rubber, an ethylene/vinyl alcohol copolymer, an ethylene/vinyl acetate/vinyl alcohol terpolymer, an ethylene/vinyloxyethanol copolymer
  • compositions of the invention that are in a modified release dosage form can be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, melt-granulation followed by compression.
  • the compositions of the invention comprise a tablets-in-capsule system, which can be a multifunctional and multiple unit system comprising versatile mini-tablets in a hard gelatin capsule.
  • the mini-tablets can be rapid-release, extended-release, pulsatile, delayed-onset extended-release minitablets, or any combination thereof.
  • combinations of mini-tablets or combinations of mini-tablets and minibeads comprising multiple active pharmaceutical agents can each have specific lag times, of release multiplied pulsatile drug delivery system (DDS), site-specific DDS, slow-quick DDS, quick/slow DDS and zero-order DDS.
  • DDS release multiplied pulsatile drug delivery system
  • compositions of the invention are in an osmotic-controlled release dosage form.
  • the osmotic-controlled release device comprises a one-chamber system, a two-chamber system, asymmetric membrane technology (AMT), an extruding core system (ECS), or any combination thereof.
  • such devices comprise at least two components: (a) the core which contains the active pharmaceutical agent(s); and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core.
  • the semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).
  • the core of the osmotic device optionally comprises an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device.
  • PEO
  • osmogens which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating.
  • Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic salts, such as sodium s
  • Osmotic agents of different dissolution rates can be employed to influence how rapidly the compound of the invention dissolves following administration.
  • an amorphous sugar such as Mannogeme EZ (SPI Pharma, Lewes, Del.) can be included to provide faster delivery during the first couple of hours (e.g., about 1 to about 5 hrs) to promptly produce prophylactic or therapeutic efficacy, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time.
  • the gemcabene or pharmaceutically acceptable salt thereof is released from the compositions of the invention at such a rate to replace the amount of the compound of the invention metabolized or excreted by the subject.
  • the core can also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.
  • Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking.
  • Suitable polymers useful in forming the coating include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CA ethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxlated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copo
  • the semipermeable membranes can also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water vapor, as disclosed in U.S. Pat. No. 5,798,119.
  • Such hydrophobic but water-vapor permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes.
  • the delivery port(s) on the semipermeable membrane can be formed post-coating by mechanical or laser drilling. Delivery port(s) can also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports can be formed during coating process, as in the case of asymmetric membrane coatings of the type disclosed in U.S. Pat. Nos. 5,612,059 and 5,698,220.
  • the total amount of the compound of the invention released and the release rate can substantially be modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.
  • the pharmaceutical composition in an osmotic controlled-release dosage form can further comprise additional conventional excipients as described herein to promote performance or processing of the formulation.
  • the osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy , supra; Santus and Baker, J. Controlled Release 1995, 35, 1-21; Verma et al., Drug Development and Industrial Pharmacy 2000, 26, 695-708; Verma et al., J. Controlled Release 2002, 79, 7-27).
  • the pharmaceutical composition provided herein is formulated as asymmetric membrane technology (AMT) controlled-release dosage form that comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients.
  • AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.
  • the pharmaceutical composition provided herein is formulated as ESC controlled-release dosage form that comprises an osmotic membrane that coats a core comprising the compound of the invention, hydroxylethyl cellulose, and other pharmaceutically acceptable excipients.
  • compositions of the invention are a modified release dosage form that is fabricated as a multiparticulate-controlled release dosage form that comprises a plurality of particles, granules, or pellets, microparticulates, beads, microcapsules and microtablets, ranging from about 10 ⁇ m to about 3 mm, about 50 ⁇ m to about 2.5 mm, or from about 100 ⁇ m to 1 mm in diameter.
  • the multiparticulate-controlled release dosage forms can provide a prolonged release dosage form with an improved bioavailability.
  • Suitable carriers to sustain the release rate of the compound of the invention include, without limitation, ethyl cellulose, HPMC, HPMC-phtalate, colloidal silicondioxide and Eudragit-RSPM.
  • Pellets suitable to be used in the compositions and therapeutic or prophylactic methods of the invention comprise 50-80% (w/w) of a drug and 20-50% (w/w) of microcrystalline cellulose or other polymers.
  • Suitable polymers include, but are not limited to, microcrystalline wax, pregelatinized starch and maltose dextrin.
  • Beads can be prepared in capsule and tablet dosage forms. Beads in tablet dosage form can demonstrate a slower dissolution profile than microparticles in capsule form.
  • Microparticle fillers suitable for compositions and therapeutic or prophylactic methods of the invention include, without limitation, sorbitan monooleate (Span 80), HPMC, or any combination thereof.
  • Suitable dispersions for controlled release latex include, for example, ethyl-acrylate and methyl-acrylate.
  • compositions of the invention are in the form or microcapsules and/or microtablets.
  • microcapsules comprise extended release polymer microcapsules containing a statin and a compound of the invention with various solubility characteristics. Extended release polymer microcapsules can be prepared with colloidal polymer dispersion in an aqueous environment.
  • microcapsules suitable for the compositions and methods provided herein can be prepared using conventional microencapsulating techniques (Bodmeier & Wang, 1993).
  • Such multiparticulates can be made by the processes known to those skilled in the art, including wet- and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores. See, for example, Multiparticulate Oral Drug Delivery ; Marcel Dekker: 1994; and Pharmaceutical Pelletization Technology ; Marcel Dekker: 1989. Excipients for such technologies are commercially available and described in US Pharmacopeia, and gemcabene salts are prepared as described in U.S. Pat. No. 6,861,555 or in International Application Publication WO 2016/077832 as, for example, gemcabene calcium salt single polymorph.
  • compositions of the invention can be blended with the compositions of the invention to aid in processing and forming the multiparticulates.
  • the resulting particles can themselves constitute the multiparticulate dosage form or can be coated by various film-forming materials, such as enteric polymers, water-swellable, or water-soluble polymers.
  • the multiparticulates can be further processed as a capsule or a tablet.
  • compositions of the invention are in a dosage form that has an instant releasing component and at least one delayed releasing component, and is capable of giving a discontinuous release of the compound in the form of at least two consecutive pulses separated in time from 0.1 hrs to 24 hrs.
  • the invention further provides a kit comprising a composition of the invention and instructions for its use.
  • the kit can further comprise a composition comprising an additional pharmaceutically active agent.
  • the kit comprises a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention and another composition comprising from about 0.1 mg to about 80 mg of a statin; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention and another composition comprising from about 10 mg to about 80 mg of a statin; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 150 mg to about 600 mg of a compound of the invention and from about 10 mg to about 40 mg of a statin; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention and another composition comprising from about 5 mg to about 80 mg of an atorvastatin or a pharmaceutically acceptable salt thereof; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention and another composition comprising from about 10 mg to about 80 mg of an atorvastatin or a pharmaceutically acceptable salt thereof; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 150 mg to about 600 mg of a compound of the invention and from about 10 mg to about 40 mg of an atorvastatin or a pharmaceutically acceptable salt thereof; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention and another composition comprising from about 5 mg to about 20 mg of ezetimibe or a pharmaceutically acceptable salt thereof; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention and another composition comprising from about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof; and instructions for the use thereof.
  • the kit comprises a composition of the invention comprising from about 150 mg to about 600 mg of a compound of the invention and another composition comprising from about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof; and instructions for the use thereof.
  • the kit comprises a) a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention, b) a composition comprising from about 5 mg to about 80 mg of a statin or a pharmaceutically acceptable salt thereof, c) a composition comprising from about 5 mg to about 20 mg of ezetimibe or a pharmaceutically acceptable salt thereof, and d) instructions for the use thereof.
  • the kit comprises a) a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention, b) a composition comprising from about 10 mg to about 80 mg of a statin or a pharmaceutically acceptable salt thereof, c) a composition comprising about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof, and d) instructions for the use thereof.
  • the kit comprises a) a composition of the invention comprising from about 150 mg to about 600 mg of a compound of the invention, b) a composition comprising from about 10 mg to about 40 mg of a statin or a pharmaceutically acceptable salt thereof, c) a composition comprising about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof, and d) instructions for the use thereof.
  • the kit comprises a) a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention, b) a composition comprising from about 5 mg to about 80 mg of an atorvastatin or a pharmaceutically acceptable salt thereof, c) a composition comprising from about 5 mg to about 20 mg of ezetimibe or a pharmaceutically acceptable salt thereof, and d) instructions for the use thereof.
  • the kit comprises a) a composition of the invention comprising from about 50 mg to about 900 mg of a compound of the invention, b) a composition comprising from about 10 mg to about 80 mg of an atorvastatin or a pharmaceutically acceptable salt thereof, c) a composition comprising about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof, and d) instructions for the use thereof.
  • the kit comprises a) a composition of the invention comprising from about 150 mg to about 600 mg of a compound of the invention, b) a composition comprising from about 10 mg to about 40 mg of an atorvastatin or a pharmaceutically acceptable salt thereof, c) a composition comprising about 10 mg of ezetimibe or a pharmaceutically acceptable salt thereof, and d) instructions for the use thereof.
  • composition of the invention and the other composition are contained in separate containers. In some embodiments, the composition of the invention and the other composition are contained in the same container.
  • the container is a bottle, vial, blister pack, or any combination thereof. In some embodiments, the container is a bottle, vial, blister pack, or any combination thereof with a closure (e.g., a cap, a top, or a sealed package to provide the composition of the invention in a closed system).
  • a closure e.g., a cap, a top, or a sealed package to provide the composition of the invention in a closed system.
  • the statin is atorvastatin, simvastatin, pravastatin, rosuvastatin, fluvastatin, lovastatin, pitavastatin, mevastatin, dalvastatin, dihydrocompactin, or cerivastatin, or any pharmaceutically acceptable salt thereof. In some embodiments, the statin is atorvastatin or a pharmaceutically acceptable salt thereof.
  • the composition of the invention or the other composition is in the form of a tablet.
  • the tablet comprises one or more excipients selected from a diluent, a disintegrant, a wetting agent, a binder, a glidant, a lubricant, or any combination thereof.
  • compositions of the invention are administered to a subject in need thereof.
  • the composition of the invention is in a unit dose form.
  • unit dose or “unit-dose” refers to a specific formulation containing a specific amount of a compound of the invention.
  • a unit dose of can be a tablet comprising about 300 mg of a compound of the invention.
  • a unit dose comprises about 50 mg, about 150 mg, about 300 mg, or about 600 mg of a compound of the invention.
  • a unit dose comprises a compound of the invention in an amount that is molar equivalent to about 150 mg, about 300 mg, or about 600 mg gemcabene.
  • the compositions of the invention are administered to a subject in need thereof, once, twice, three times, or four times a day. In some embodiments, the compositions of the invention are administered to a subject in need thereof in ways that allows a daily dose of about 600 mg to about 900 mg of a compound of the invention. In some embodiments, the compositions of the invention are administered to a subject in need thereof in ways that allows a daily dose in an amount that is molar equivalent to about 600 mg to about 900 mg of gemcabene. In some embodiments, the daily dose is about 600 mg of a compound of the invention. In another embodiment, the daily dose is an amount that is molar equivalent to 600 mg of gemcabene.
  • compositions of the invention comprise about 300 mg of a compound of the invention and are administered to a subject in need thereof once a day. In some embodiments, the compositions of the invention comprise about 300 mg of a compound of the invention and are administered to a subject in need thereof twice a day. In some embodiments, the compositions of the invention comprise about 300 mg of a compound of the invention and are administered to a subject in need thereof three times a day.
  • compositions of the invention comprise a compound of the invention in an amount that is molar equivalent to about 300 mg of gemcabene and are administered to a subject in need thereof once a day. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is molar equivalent to about 300 mg of gemcabene and are administered to a subject in need thereof twice a day. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is molar equivalent to about 300 mg of gemcabene and are administered to a subject in need thereof three times a day.
  • compositions of the invention comprise about 600 mg of a compound of the invention and are administered to a subject in need thereof once a day. In some embodiments, the compositions of the invention comprise a compound of the invention in an amount that is molar equivalent to about 600 mg of gemcabene and are administered to a subject in need thereof once a day.
  • compositions of the invention comprise about 150 mg of a compound of the invention and are administered to a subject in need thereof once a day. In some embodiments, the compositions of the invention comprise about 150 mg of a compound of the invention and are administered to a subject in need thereof twice a day. In some embodiments, the compositions of the invention comprise about 150 mg of a compound of the invention and are administered to a subject in need thereof three times a day. In some embodiments, the compositions of the invention comprise about 150 mg of a compound of the invention and are administered to a subject in need thereof four times a day.
  • two separate unit doses each comprising about 150 mg of a compound of the invention, are administered to a subject in need thereof once a day. In some embodiments, two separate unit doses, each comprising about 150 mg of a compound of the invention, are administered to a subject in need thereof twice a day (total 600 mg/day). In some embodiments, two separate unit doses, each comprising about 150 mg of a compound of the invention, are administered to a subject in need thereof three times a day (total 900 mg/day).
  • compositions of the invention comprising a compound of the invention in an amount that is molar equivalent to about 150 mg of gemcabene is administered to a subject in need thereof once a day. In some embodiments, the compositions of the invention comprising a compound of the invention in an amount that is molar equivalent to about 150 mg of gemcabene is administered to a subject in need thereof twice a day. In some embodiments, the compositions of the invention comprising a compound of the invention in an amount that is molar equivalent to about 150 mg of gemcabene is administered to a subject in need thereof three times a day. In some embodiments, the compositions of the invention comprising a compound of the invention in an amount that is molar equivalent to about 150 mg of gemcabene is administered to a subject in need thereof four times a day.
  • two separate unit doses each comprising a compound of the invention in an amount that is molar equivalent to about 150 mg of gemcabene, are administered to a subject in need thereof once a day.
  • two separate unit doses, each comprising a compound of the invention in an amount that is molar equivalent to about 150 mg of gemcabene are administered to a subject in need thereof three times a day (total 900 mg/day).
  • Tetrahydrofuran (THF) (279 kg) was added followed by a lithium diisopropylamide solution (lithium diisopropylamide 28% w/w in heptane/THF/ethylbenzene, 174.6 kg, 2.2 equiv) at 10° C.-15° C.
  • THF Tetrahydrofuran
  • a lithium diisopropylamide solution lithium diisopropylamide 28% w/w in heptane/THF/ethylbenzene, 174.6 kg, 2.2 equiv
  • the product-containing organic layer was separated, and the aqueous layer was extracted with heptane (106 kg) at 50° C. ⁇ 2° C. The aqueous layer was then discarded.
  • the combined product-containing heptane layer was washed twice with water (64 kg) at 50° C. ⁇ 2° C. and the aqueous layers were discarded.
  • the heptane layer was evaporated to dryness at ⁇ 60° C.
  • the resultant residue was mixed twice with water (320 kg each wash) and evaporated to dryness at ⁇ 60° C.
  • the remaining material was dissolved in heptane (286 kg) at 22° C. ⁇ 2° C. and washed with water (193 kg) and the aqueous layer was discarded.
  • the heptane layer was evaporated to dryness at ⁇ 60° C. and co-evaporated three-times with heptane (each 109 kg). Karl-Fisher analysis indicated water content was 0.04%.
  • the resultant residue was dissolved in heptane (130 kg) and THF (1.4 kg) at 22° C. 2° C., filtered through silica gel (64.0 kg) and the silica gel was washed first with heptane (246 kg)/THF (16.0 kg) mixture and then only with heptane (492 kg).
  • the collected filtrate was concentrated to a volume of about 150 L at ⁇ 60° C.
  • the solution was transferred to a smaller vessel (ST-164, glass-lined, 160 l) with heptane (44 kg), followed by evaporation to dryness at ⁇ 60° C.
  • 1 H NMR analysis of the crude indicated 96.7% purity.
  • the crude gemcabene was dissolved in heptane (55.0 kg) at 40° C. ⁇ 5° C. and the heptane solution was cooled to 15° C. ⁇ 2° C. After seeding with gemcabene crystals (30 g), the solution was cooled to 12° C.
  • the purified gemcabene contained 0.4% 2,2,7,7-Tetramethyl-octane-1,8-dioic acid.
  • Gemcabene (50.5 kg; 167 mol, 1.00 equiv, from Step 1) was dissolved in ethanol (347 kg, denatured with 1% cyclohexane) and filtered through a 1.2 ⁇ m filter in the reaction vessel (ST-1005, glass-lined, 1600 l). The equipment was flushed with additional ethanol (38 kg). Calcium oxide (9.35 kg, 167 mol, 1.00 equiv) was added at 22° C. under stirring, and the mixture is heated at reflux for 20-25 hours. The resulting mixture was cooled to 52° C. ⁇ 2° C. and tert-butyl methyl ether (125 kg, filtered through a 1.2 ⁇ m filter) was charged.
  • the crystalline ethyl alcohol solvate was isolated by filtration in an agitated filter dryer (FT-1001, stainless steel, 1000 l) and washed with tert-butyl methyl ether in three portions (3 ⁇ 37 kg, filtered through a 1.2 ⁇ m filter).
  • the crystalline ethyl alcohol solvate was dried with interval agitation (3 minutes stirring, 15 minutes not stirring) at a jacket temperature of 30° C. for 66 minutes, 50° C. for 30 minutes, 70° C. for 30 minutes, and 90° C. for at least 12 hours in vacuum with a stream of 20 L/h nitrogen.
  • the gemcabene calcium salt hydrate Crystal Form 1 obtained in Step 2 (53.2 kg, 157 mol) was milled using a pinmill (MP160) with a dedicated rotor and stator equipped with 4 pin rows (n. 699), under nitrogen flow. An amount of 49.3 kg of gemcabene calcium Crystal Form 1 with a PSD90 ranging from 40 ⁇ m to 75 ⁇ m was obtained in 93% yield.
  • Instrument Type Agilent 1200 Series or ThermoScientific Ultimate 3000 UHPLC (QC-HPLC-26), or equivalent
  • Mobile Phase A Water + 0.1% v/v formic acid
  • Mobile Phase B Acetonitrile + 0.1% v/v formic acid Column Temperature: 40° C.
  • Sample Solution (10 mg/mL): 100 mg ( ⁇ 5 mg) sample was added to a 10 mL-flask and Sample Solvent was added to the mark.
  • Reference Mix Stock Solutions (0.5 mg/mL for gemcabene and 0.25 mg/L for other substances): 10 mg ( ⁇ 1 mg) gemcabene+5 mg ( ⁇ 1 mg) 2,2,7,7-tetramethyl-octane-1,8-dioic acid+5 mg ( ⁇ 1 mg) 6-(4-hydroxybutoxy)-2,2-dimethylhexanoic acid+5 mg ( ⁇ 1 mg) 2,2-dimethyl-hex-4-enoic acid (E/Z ratio approximately 5:1) were added in a 20 mL-flask and the Sample Solvent was added to the mark (Reference Mix Stock). 2.0 mL of Reference Mix Stock was added to a 20 mL-flask and the Sample Solvent was added to the mark (Diluted Reference Stock).
  • UV Reporting threshold: 0.05% w/w
  • HPLC High-Performance Liquid Chromatography
  • Instrument Type Agilent 1200 Series or ThermoScientific Ultimate 3000 UHPLC (QC-HPLC-26), or equivalent
  • Mobile Phase A Water + 0.1% v/v formic acid
  • Mobile Phase B Acetonitrile + 0.1% v/v formic acid Column Temperature: 40° C.
  • Instrument Type Dionex ICS-5000 + SP, ICS-5000 + EG, Dionex ICS-5000 + DC, Autosampler AS, or equivalent IC
  • Eluent Generator KOH e.g., Dionex EGC III KOH Eluent Generator Cartridge - Product no. 074532
  • Suppressor set 112 mA Cell Temperature: 35° C.
  • Injection Volume 10 ⁇ L Oven temperature 30° C.
  • sample 25 mg ( ⁇ 1.0 mg) sample was added to a 5 mL-flask and dissolved with water/acetonitrile 1:1+0.05% trifluoroacetic acid and filled to the mark.
  • the flask was placed in an ultrasound bath for 10 min and then left to cool for approx. 1 hr. Then the solution was observed to make sure it was clear and had no particles (no deposit). If there were particles, sample was filtered into a vial via the syringe filter (e.g. filter 0.45 um—for organic PTFE solutions) (discarded 2-3 ml filtrate in advance to saturate the filter). If no particles were present, the sample can be analyzed without filtration.
  • the syringe filter e.g. filter 0.45 um—for organic PTFE solutions
  • Limit of quantification was 2.50 ⁇ g/mL which corresponds to 0.05% w/w.
  • Exactly 125 mg (5 ppm*) of bis-(4-chlorobutyl)ether was added to a 20 mL volumetric flask containing 10 ml of N-methyl-2-pyrrolidone (NMP). The volumetric flask was filled to the mark with NMP. *The value in ppm refers to 100 ⁇ I of stock uplift and 125 mg nominal weight.
  • 125 mg (1000 ppm*) n-hexane, 250 mg (2000 ppm*) THF, 125 mg (1000 ppm*) diisopropylamine, 250 mg (2000 ppm*) ethylbenzene and 125 mg (1000 ppm*) of cyclohexane were accurately measured into a 20 mL volumetric flask containing approximately 10 mL of NMP. The volumetric flask was filled to the mark with NMP and solution was mixed until it became homogeneous. *The value in ppm refers to 100 ⁇ I of stock uplift and 125 mg nominal weight.
  • n-heptane, t-butyl methyl ether, and ethanol were weighed into a 20 mL volumetric flask containing approximately 10 mL NMP.
  • 20 ⁇ L of Stock bis-(4-chlorobutyl)ether and 4 mL of Stock Solution was added to the volumetric flask. Then, the volumetric flask was filled to the mark with NMP and solution was mixed until it became homogeneous.
  • the value in ppm refers to 100 ⁇ I of stock uplift and 125 mg nominal weight.
  • ICP-OES Inductively Coupled Plasma Optical Emission Spectrometry
  • ICP - Optical Emission spectrometer Vista- PRO, Agilent or suitable equivalent
  • Emission wavelength 317.93 nm
  • Plasma observation Axial Plasma power 1200 W
  • Plasma gas flow 16.5 L/min
  • Ar Auxiliary gas flow 1.5 L/min
  • Nebulizer Concentric Spray chamber Cyclon spray chamber Nebulizer gas flow 0.75 L/min Pump rate 20 rpm
  • Reagents Water Deionized or higher purity Nitric Acid Conc.
  • Quality Control Analyzed the quality control sample at least once. The relative deviation from the expected analyte concentration must not exceed ⁇ 10% for the analyte Linearity Check: Evaluated data from linear regression calculation. Correlation coefficients must be ⁇ 0.999 Blank Solutions: The blank solutions were analyzed. For Bach determination, the Ca concentration of the blank solution must not be more than 1% of the lowest concentration of the test solutions
  • Calibration solutions Laboratory prepared mixtures with known and suitable element concentrations. Prepared at least 4 calibration solutions (including zero solution) from the stock standard solution by appropriate dilution with water and addition of 2 mL of conc. HNO 3 per 100 mL; selected the calibration range according to the expected analyte concentration. E.g. for an element content of 11.8% m/m (8.0, 12.0 and 16.0) mg/L would be suitable concentrations. Zero solution Prepared as described for the calibration solutions, but without addition of any standard solution. Test solutions Test solutions were prepared in duplicate. Weighed approx. 25 mg of the test substance accurately (to within 0.01 mg) into a digestion vessel.
  • the emission of the zero solution and the calibration solutions were measured using suitable Instrument parameters (see above). The emission of blank solution, quality control solution and test solutions were measured. If necessary, the test solutions were diluted with zero solution (dilution factor 0 to obtain a reading within the calibration range. Alternatively, new calibration solutions were prepared in order to adapt the calibration range.
  • the calibration function was determined using the corresponding readings.
  • the analyte element concentration was calculated in the test solutions from the measured emission with this calibration function subtracting the reading of the zero solution.
  • the analyte element concentration was calculated in the test substance using the equation below. These calculations were done by the Instrument software.
  • Gemcabene calcium salt Crystal Form 1 was prepared as described in Example 1. Approximately 40 g of gemcabene calcium salt Crystal Form 1 was weighed. To this, approximately 800 mL of water was added and mixed at ambient temperature for dissolution. After approximately 4 hours, the solid was found to have dissolved and the solution was transferred to a 2 L round bottom flask. The solution was then frozen before being placed on a freeze dryer for approximately 72 hours. X-ray powder diffraction (XRPD) analysis of a combined lot of material showed that the diffractogram is consistent with reference amorphous data ( FIG. 52A ). Polarized light microscope (PLM) images showed glass-like particles with limited birefringence.
  • XRPD X-ray powder diffraction
  • Thermogravimetric analysis showed a weight loss of 3.1% up to 150° C. ( FIG. 52B ). No thermal events were noted in the differential thermal analysis (DTA) or in the differential scanning calorimetry (DSC) ( FIGS. 52B and 52C ). The moisture content of the material was determined to be 2.62% by Karl-Fisher titration.
  • the amorphous gemcabene calcium salt was determined to have a gemcabene content (% gemcabene) of 88.85% on a % w/w basis by high-performance liquid chromatography equipped with charged aerosol detector (HPLC-CAD).
  • Particle size distribution (PSD) analysis returned a D10 value of 5.2 ⁇ m, a D50 value of 26.4 ⁇ m and a D90 value of 60.3 ⁇ m.
  • the amorphous form was obtained by drying gemcabene calcium ethanol solvate.
  • the amorphous solid was difficult to handle due to electrostatic properties and a relatively low bulk density of ⁇ 0.3 g/mL (tapped).
  • Gemcabene calcium salt Crystal Form 1 was prepared as described in Example 1. To a 5 L glass reactor held at 70° C., approximately 160 g of gemcabene calcium salt Crystal Form 1 was added, along with approximately 2.4 L of an ethanol:water (90:10 v/v %) solution. The slurry was then mixed at 120 RPM using a 4 pitch-blade PTFE impeller for approximately 2 hours. After 2 hours, a further 824 mL water was added to the slurry (new solvent ratio of ethanol:water (67:33 v/v %)) and the material was then left to slurry for approximately 18 hours. The crystallization was then cooled to 40° C. and the stirring rate decreased to 100 RPM. The crystallization was held for 2 hours then separated by filtration.
  • the mother liquor was determined to have a concentration of 18.47 mg/mL by high-performance liquid chromatography (HPLC).
  • the gemcabene calcium salt Crystal Form 2 was determined to have a gemcabene content (% gemcabene) of 86.91% w/w.
  • Gas chromatography analysis of the material showed a residual ethanol content of 61 ppm.
  • Particle size distribution (PSD) analysis was carried out and gave a D10 value of 5.0 ⁇ m, a D50 of 14.4 ⁇ m and a D90 of 38.2 ⁇ m.
  • Amorphous form of gemcabene calcium salt was prepared as described in Example 3. To a large crystallization dish, approximately 50 g of amorphous gemcabene calcium salt was added. To the crystallization dish, 250 mL ethanol was added in 50 mL aliquots, with the material mixed between additions to ensure even solvent distribution. The mixture was mixed several times during drying to minimize large aggregate formation. The material was then dried at ambient under vacuum for approximately 72 hours. X-ray powder diffraction (XRPD) spectroscopy analysis showed that the dried material was consistent with gemcabene calcium salt Crystal Form C3. Polarized light microscope (PLM) images showed agglomerated particles with limited birefringence. Thermogravimetric analysis showed a weight loss of 5.5% up to 160° C.
  • XRPD X-ray powder diffraction
  • FIG. 54B A single endothermic event was noted in the differential thermal analysis (DTA) at onset 121° C., with a peak at 129° C. ( FIG. 54B ).
  • DTA differential thermal analysis
  • DSC Differential scanning calorimetry
  • the moisture content of the material was determined to be 2.1% by Karl-Fisher titration.
  • the gemcabene calcium salt Crystal Form C3 was determined to have a gemcabene content (% gemcabene) of 83.98% on a % w/w basis was determined by high-performance liquid chromatography equipped with charged aerosol detector (HPLC-CAD). Gas chromatography analysis showed a residual ethanol content of 76070 ppm. Particle size distribution (PSD) analysis returned a D10 value of 8.8 ⁇ m, a D50 value of 20.4 ⁇ m and a D90 value of 44.3 ⁇ m.
  • PSD particle size distribution
  • the mother liquor was determined to have a concentration of 21.59 mg/mL by high-performance liquid chromatography (HPLC).
  • HPLC high-performance liquid chromatography
  • the crystalline gemcabene calcium salt ethanol solvate was determined to have a gemcabene content (% gemcabene) of 90.51% w/w.
  • Gas chromatography analysis of the material showed a residual ethanol content of 28628 ppm and a residual t-BME content of 511 ppm.
  • Particle size distribution (PSD) analysis was performed and gave a D10 value of 3.3 ⁇ m, a D50 of 31.8 ⁇ m and a D90 of 85 ⁇ m.
  • Example 7 Gemcabene Calcium Salt Hydrate Crystal Forms C1, C2 and C3 (Collectively, Gemcabene Calcium Salt Hydrate Crystal Form C)
  • Gemcabene calcium salt hydrate Crystal Forms C1-C3 were obtained by way of prolonged drying by charging the wet amorphous form of gemcabene calcium salt hydrate product to an agitated pan dryer at temperatures of 80° C. for at least 24 hours then at higher temperature up to 100° C. for 24 hours or more. Depending on the temperature of drying and the duration of drying, various forms of Crystal Form C were obtained, including Crystal Form C1, Crystal Form C2, and Crystal Form C3.
  • the particle size distribution was determined in accordance with the Fraunhofer light diffraction method. A coherent laser beam was passed through the sample and the resulting diffraction pattern was focused on a multi-element detector. Because the diffraction pattern depends, among other parameters, on particle size, the particle size distribution (PSD) was calculated based on the measured diffraction patter of the sample.
  • PSD particle size distribution
  • Stock dispersion solution was prepared by adding a few drops of the dispersing aid (e.g., 1% w/w solution of a detergent in white spirit such as Span 80, Fluka (85548-250 ml)) to an appropriate amount of substance and mixed carefully. The dispersion was then slowly diluted to a final volume of about 10 ml while vortexing. The suspension cell of the instrument (Malvern Mastersizer 2000 equipped with Hydro 2000S sample dispersion unit) was filled with dispersion medium and a background measurement was taken. The stock dispersion was added to the suspension cell until an optical concentration of 5% to 15% was reached. Once the measurement was initialized, the final optical concentration increased following the internal ultrasonication step and did not exceed 25%. The cumulative volume distribution was determined in accordance with the instrument's instruction manual.
  • the dispersing aid e.g., 1% w/w solution of a detergent in white spirit such as Span 80, Fluka (85548-250 ml)
  • the dispersion was then slowly diluted to
  • PSD10, PSD50, and PSD90 values were determined from the cumulative volume distribution of each measurement. Values smaller than 10 ⁇ m were reported to one decimal place. Results larger than 10 ⁇ m were reported as single digit values. Sample parameters used in analysis are shown below:
  • Scanning electron micrographs were obtained using a FEI Phenom SEM using 5 kV of accelerating voltage.
  • the samples were prepared for imaging by mounting a small quantity (about 1 mg-10 mg) of sample to an aluminum sample stub using a piece of two-sided carbon tape. A conductive gold/palladium coating was applied to the sample to prevent charging effects from interfering with the imaging process. Electron micrographs were then collected. Magnification, image height, and a graduated micron bar can be found at the bottom of each micrograph.
  • Gemcabene calcium salt hydrate Crystal Form 1 having various particle sizes were prepared by use of different milling techniques. Total of nine Samples (Samples 1-9, Table 2) of gemcabene calcium salt hydrate Crystal Form 1 were subjected to the laser light diffraction particle size analysis. The PSD90 of each Sample determined by laser light diffraction is shown in Table 2.
  • Sample 5 (Table 2) under different conditions.
  • Sample 1 was prepared by milling Sample 5 using Fitzpatrick Comminuting Machine model L1A at high speed (8946 RPM) through an 80-mesh screen.
  • the resulting particle size after milling had PSD90 of about 150 ⁇ m.
  • Sample 2 was prepared by further milling Sample 1 using a pinmill.
  • the PSD90 of Sample 2 was about 75 ⁇ m.
  • Sample 3 was prepared by further milling Sample 1 using a Fitzpatrick Comminuting Machine model L1A at high speed.
  • the PSD90 of Sample 3 was about 110 ⁇ m.
  • Gemcabene calcium salt hydrate Crystal Form 1 of Samples 1-4 and 6-8 (Table 2) was used to produce drug product tablets by wet granulation in fluidized bed. Tablets could not be manufactured from Samples 5 and 9 (Table 2) because the particle size distribution was too large and the particles did not fluidize in the fluidized bed granulation. Further, gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 less than about 30 ⁇ m showed difficulties in the formulation process due to electrostatic properties and low loose density.
  • Each PXRD pattern was analyzed using the GSAS II crystallography data analysis software program, utilizing the peak fit function. Peaks were selected and peak position, intensity, and full width at half maximum (“FWHM”) were allowed to refine freely. The small residual background was fit using a 5-term polynomial function which was allowed to refine freely.
  • the water contents of two other gemcabene calcium salt hydrate Crystal Form 1 samples having PSD90 of 55 ⁇ m (Sample 10) and PSD90 of 47 ⁇ m (Sample 11) were determined to be about 3.7% w/w in each sample, which corresponds to about 0.82 equivalents of water per a mole of gemcabene calcium salt.
  • the water content of gemcabene calcium salt hydrate Crystal Form 1 with PDS90 ranging from 47 ⁇ m to 62 ⁇ m had a water content of about 3.5% w/w to about 3.7% w/w, corresponding to about 0.78 to about 0.82 equivalents of water per a mole of gemcabene calcium salt.
  • the ethanol content specification is less than 5000 ppm.
  • the ethanol content in Samples 4 and 6-10 was determined to range from 710 ppm to 1840 ppm.
  • the loose bulk densities of Samples 4 and 6-10 ranged from 0.25 g/mL to 0.30 g/mL and the tapped bulk densities of Samples 4 and 6-10 ranged from 0.33 g/mL to 0.49 g/mL (Table 3).
  • Gemcabene calcium salt hydrate Crystal Form 1 from each of Samples 1-4 and 6-8 of Table 2 was granulated with excipients using a fluid bed granulation process.
  • a sample batch formula for gemcabene calcium salt hydrate Crystal Form 1 granulation is shown in Table 4.
  • the binding solution was prepared by weighing 41.06 kg of purified water and adding to a stainless-steel mixer and mixed. The mixing took about 1.5-2.5 hours. Hydroxylpropyl cellulose was slowly added to the water while mixing. The mixer speed was maintained to sufficiently mix the hydroxypropyl cellulose without generating foam. The mixing was continued until the hydroxypropyl cellulose was completely dissolved and a clear homogeneous solution was obtained.
  • the spray pump was verified to deliver a rate of 100 to 350 g/minute of the hydroxypropyl cellulose solution.
  • the Glatt 30 fluid bed granulator was set with a process air volume of 500 m 3 per hour, an inlet air temperature of 70° C., and exhaust temperature of 45° C. ⁇ 10° C.
  • Gemcabene calcium salt hydrate Crystal Form 1 and lactose monohydrate were milled with a 45R mesh screen, for example with Quadro Comil 197 Ultra equipped with a Round Impeller (45R screen; 0.045′′ opening size; round), to de-lump and the material was captured in a container double lined with polyethylene bags.
  • the fluid bed granulator was charged with de-lumped gemcabene calcium salt hydrate Crystal Form land lactose monohydrate. Once the powder fluidization began, the binding solution was sprayed over the powder. After the powder was wet, the spray rate was reduced and the air volume was adjusted until all binder solution was sprayed. The inlet air volume was adjusted to ensure fluidization of granules and the target temperature was kept at about 28° C. After all binder solution was applied, granulation was continued with water to achieve acceptable visual granulation endpoint. The granulation was dried to a loss on drying (LOD) value of not more than 2.0%.
  • LOD loss on drying
  • the rate of spraying the binding solution can vary depending on the scale of the granulation, etc. For example, for 22 L granulator/drying bowl size scale, the spray rate of the binder can be at 75-90 g/min for first 30-45 minutes then 50-65 g/min for the remaining time until theoretical amount is sprayed on. Further, if required, purified water can be added to continue granulation until visually acceptable granulation is achieved before drying.
  • Samples 1G, 2G, 3G, 4G, 6G, 7G, and 8G are referred to as Samples 1G, 2G, 3G, 4G, 6G, 7G, and 8G, respectively.
  • Samples 1G, 2G, 3G, 4G, 6G, 7G and 8G were each milled through a 39R mesh screen and collected in a container double-lined with polyethylene bags (e.g., Quadro Comil 197 Ultra equipped with a round bar impeller) to provide Samples 1M, 2M, 3M, 4M, 6M, 7M, and 8M, respectively.
  • polyethylene bags e.g., Quadro Comil 197 Ultra equipped with a round bar impeller
  • a V-blender was charged with the milled Samples 1M-4M and 6M-8M. Croscarmellose Sodium was passed through a 20 mesh hand screen and charged into the V-blender with the granulation and blended for 10 minutes. A bag containing the magnesium stearate component was rinsed with the granulation blend. The mixture was filtered through a 20 mesh screen, added to the V-blender and blended for about 3 minutes. The final granulation blend was discharged into drums which are double lined with polyethylene bags and sealed.
  • Samples 1M-4M and 6M-8M are referred to as Samples 1FB, 2FB, 3FB, 4FB, 6FB, 7FB, and 8FB, respectively.
  • Samples 1FB-4FB and 6FB-8FB were compressed 300-mg film-coated tablets.
  • a sample tablet formula is shown in Table 5.
  • Samples 1FB-4FB were added, separately, to a tablet press equipped with a force feeder. Samples 1FB-4FB were respectively compressed per specified parameters in Table 6. The tablet weight and hardness were adjusted to target tablet weight and hardness, and were passed through a metal detector and tablet de-duster and collected into double lined polyethylene bags.
  • Samples 1FB, 2FB, 3FB, 4FB, 6FB, 7FB, and 8FB were compressed on a rotary tablet press using 0.2759′′ ⁇ 0.6285′′ oval tooling to a theoretical fill weight of 470 mg. See Table 6 below for compression parameters, batch weight variation and tablet properties. All tablets compressed well and had a low relative standard deviation (RSD) for tablet weight variation. Tablets prepared from Samples 1FB, 2FB, 3FB, 4FB, 6FB, 7FB, and 8FB are referred to as Tablets A, B, C, D, F, G, and H, respectively.
  • Each batch was film-coated in either the experimental Vector Coater LDCS instrument (Tablets A-C, Table 6) or the GMP Compu-Lab 24 (Tablet D, Table 6).
  • the film-coating suspension consisted of Opadry White YS 1-7040 and Simethicone Emulsion 30% USP.
  • Purified water was weighed into a stainless-steel container and mixed to create a vortex. Simethicone emulsion and Opadry White YS 1-7040 were added to the purified water and mixed for a minimum of 50 minutes or until the suspension was visually uniform. Tablets A-D were, separately, divided into two batches and are weighed out for coating. The Tablets were charged into a coating pan heated to an outlet temperature of 42° C. ( ⁇ 2° C.). The Tablets were film-coated to a 3.0% weight gain ( ⁇ 1.0%). After 90% of theoretical amount of film-coating suspension for each batch were sprayed, the average weight was checked and spraying continued to achieve a weight gain of 2.0% to 4.0%. Tablets were allowed to dry and cool down. The Tablets were packaged in tared containers double lined with polyethylene bags.
  • Film-coated Tablets F-H were prepared by the same process used for making Tablet D.
  • the laboratory scale fluid-bed granulation equipment was a Freund-Vector MFL-01 laboratory fluid-bed processor configured for a top-spray process, which is a scaled-down Glatt equipment used for the granulation of the clinical batches.
  • Table 7A gives the quantitative theoretical composition of the tablet formulation and the laboratory scale batch size.
  • High-performance liquid chromatography indicated that the amorphous gemcabene calcium contained 80.9% (w/w) molar equivalent of gemcabene.
  • the amount of the amorphous gemcabene calcium charged to the batch was adjusted by this factor, resulting in 92.71 g of amorphous gemcabene calcium being dispensed with a commensurate decrease in the lactose monohydrate quantity to 9.75 g.
  • the amorphous gemcabene calcium was screened to form a uniform powder for use in the granulation process using a #40 mesh (425 ⁇ m) sieve and 92.72 g of the screened material was dispensed into the granulator.
  • Amorphous gemcabene calcium and Lactose Monohydrate were charged into the fluid-bed's expansion chamber and were allowed to mix for 2 minutes using a process air flow of 50 L per minute (LPM).
  • LPM liquid hourly fluid
  • the fluid bed charge was then granulated by the addition of a granulation solution, consisting of water and Hydroxypropyl Cellulose (Klucel® EF). This solution was dispensed into the granulator as an atomized spray from the fluid-bed's air atomized spray nozzle.
  • Target granulation process parameters were scaled for the MFL-01 fluid-bed from the large-scale granulation process. Table 7D reports the target processing parameters.
  • Example 13 Dissolution Profiles of Gemcabene Film-Coated Tablets (300 mg) Prepared from Gemcabene Calcium Salt Crystal Form 1 having various PSD90 values
  • the average % release values at 20, 30 and 45 minutes are lower with Tablets A and C when compared with % dissolution of Tablets B and D.
  • the amount of gemcabene detected at 45 minutes was about 8% to 15% lower for Tablets A and C than that of the Tablets made from gemcabene calcium salt hydrate Crystal Form 1 having a smaller particle size (Tablets B and D).
  • Tablets G and H made from gemcabene calcium salt hydrate Crystal Form 1 having a PSD90 of 62 ⁇ m and 48 ⁇ m, respectively, show a more favorable dissolution profile with almost 40% average release at 10 min and essentially 100% average release at 30 min. Meanwhile, when drug substance gemcabene Calcium Salt Hydrate Crystal Form 1 is used neat (recrystallized only), the dissolution profiles show lower release profiles.
  • gemcabene was accurately weighed and transferred to a 100 mL volumetric flask, then dissolved in about 10 mL of acetonitrile (ACN). If necessary, sonication can be used to dissolve gemcabene.
  • ACN acetonitrile
  • the gemcabene solution was diluted to volume with the dissolution medium.
  • Dissolution Medium 50 mM Potassium Acetate in water, adjusted to pH 5.0 with glacial acetic acid Dissolution Apparatus: USP Type 2, Paddles Rotation Speed: 50 rpm
  • Sampling times 10, 20, 30, 45, 60, and/or 75 minutes
  • Sampling technique Withdraw 2 mL sample with a syringe and stainless-steel cannula with a 45 ⁇ m filter tip into an HPLC vial
  • the concentration (mg/mL) of the sample solution was calculated for each time point as follows or by using validated software such as OpenLAB or equivalent.
  • Sample ⁇ ⁇ concentration ⁇ ⁇ ( mg ⁇ / ⁇ mL ) Sample ⁇ ⁇ area ⁇ Sample ⁇ ⁇ area Mean ⁇ ⁇ standard ⁇ ⁇ area
  • Gemcabene detected (mg) in dissolution medium pH 5.0 potassium acetate
  • “gemcabene released” which is calculated for each vessel as follows or using a validated software such as DataCal, OpenLAB, or equivalent.
  • % ⁇ ⁇ release mg ⁇ ⁇ gemcabene ⁇ ⁇ released ⁇ 100 _ 300 ⁇ ⁇ mg * ⁇ ⁇ ( theoretical )
  • each gemcabene calcium salt hydrate Crystal Form 1 300-mg film-coated tablet comprises gemcabene calcium salt hydrate Crystal Form 1 in an amount that is a molar equivalent to 300 mg gemcabene.
  • Dissolution data for Tablets A-D and F—H are shown in the following Tables, respectively: Tables 8a and 8b; Table 9; Table 10; Table 11; Table 12; Table 13; and Table 14, where the release of gemcabene is determined by the amount of gemcabene measured by the above described HPLC method.
  • Dissolution profiles of Tablets A-D and F—H are shown in FIG. 1A and dissolution profiles of Tablets B-D are separately shown in FIG. 1B .
  • the dissolution profile of Tablets B, D, and F-H was more favorable than the dissolution profile of Tablets A and C which comprise gemcabene calcium salt hydrate Crystal Form 1 having a higher PSD90, 151 ⁇ m and 110 ⁇ m, respectively. Without bound to any theory, it is believed that the more favorable (fast) dissolution profile is a useful indicator of the tablet having good bioavailability. Further, it was unexpected that Tablet C, comprising gemcabene calcium salt hydrate Crystal Form 1 having PSD90 of 110 ⁇ m, had a significantly slower dissolution profile.
  • Tablets were tested for content uniformity using HPLC in accordance with USP ⁇ 905>.
  • sample solutions were evaluated via HPLC against the sensitivity solution, working standard, check standards, marker solutions, and a mobile phase A blank. Data were collected using validated HPLC system software. Content uniformity results were consistent among all batches and did not appear to be affected by the particle size distribution of the gemcabene calcium salt hydrate Crystal Form 1.
  • gemcabene reference standard was weighed into a 25 mL volumetric flask and diluted to volume with mobile phase A to yield a concentration of 2.4 mg/mL (expressed as free diacid).
  • 1.0 mL of the gemcabene working or check standard was transferred into a 100 mL volumetric flask, diluted to volume with mobile phase A and mixed well. 1.0 mL of this solution was transferred into a 20 mL volumetric flask. Diluted to volume with mobile phase A and mixed well for a nominal concentration of 1.2 ⁇ g/mL of gemcabene.
  • the content uniformity was calculated based on the following formula.
  • PAsmp Peak area of gemcabene
  • PAstd Average peak area of gemcabene in all working standard injections
  • each gemcabene calcium salt hydrate Crystal Form 1 300-mg film-coated tablet comprises gemcabene calcium salt hydrate Crystal Form 1 in an amount that is a molar equivalent to 300 mg gemcabene theoretical gemcabene molar equivalent of gemcabene calcium salt in each tested 300 mg tablets.
  • Example 10 Each granulation of Example 10 did not over wet or require additional water to complete. Each granulation produced a blend with exceptional flow properties and tablets of adequate hardness with low friability. Thus, further optimization can be necessary to perform at larger batch sizes.
  • NASH-HCC non-alcoholic steatosis hepatitis
  • the murine STAMTM model of NASH-HCC is a high-fat caloric (HFC)-fed mouse model, in which pathological progression is very similar to that in humans as they develop liver steatosis, inflammation, and partial fibrosis (Kohli and Feldstein, J Hepatol, 155, 941-943, doi:10.1016/j.jhep.2011.04.010 (2011)).
  • HFC high-fat caloric
  • mice Two-day old neonatal C57BL/6 male mice were administered low-dose streptozotocin (STZ), and were subsequently fed a HFC diet from 4 weeks of age. In this model, the mice typically develop liver steatosis and diabetes, reaching steatohepatitis within 3 weeks, followed by cirrhosis within 8 weeks, and carcinoma within 16 weeks.
  • mice were administered daily oral gemcabene calcium salt hydrate Crystal Form 1 starting at week 6 of age and were sacrificed at week 9. Telmisartan (with antisteatotic, anti-inflammatory and antifibrotic effects in STAMTM mice) was used as a positive comparator.
  • a baseline reference group was administered vehicle at day 2 of age, and from week 6 of age was vehicle-treated and chow-fed.
  • Five STAMTM groups were streptozocin-treated at day 2 of age and fed a HFC-diet beginning with week 4 of age.
  • These STAMTM groups were orally administered from week 6 one of the following: water-vehicle, gemcabene calcium salt hydrate Crystal Form 1 at 30, 100 and 300 mg/kg daily, or telmisartan (MICARDIS®) 10 mg/kg daily. Telmisartan (MICARDIS®) was purchased from Boehringer Ingelheim GmbH (Germany) and dissolved in pure water. All groups were sacrificed at week 9. The treatment schedule is summarized in Table 17. Vehicle, gemcabene calcium salt hydrate Crystal Form 1, or telmisartan were administered by oral gavage once daily.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
US15/956,172 2017-04-18 2018-04-18 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor Abandoned US20180297929A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/956,172 US20180297929A1 (en) 2017-04-18 2018-04-18 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
US16/402,853 US20200148617A1 (en) 2017-04-18 2019-05-03 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
US17/583,081 US20220332672A1 (en) 2017-04-18 2022-01-24 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201762486728P 2017-04-18 2017-04-18
US201762486822P 2017-04-18 2017-04-18
US201762569358P 2017-10-06 2017-10-06
US201762584576P 2017-11-10 2017-11-10
US15/956,172 US20180297929A1 (en) 2017-04-18 2018-04-18 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/402,853 Continuation US20200148617A1 (en) 2017-04-18 2019-05-03 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor

Publications (1)

Publication Number Publication Date
US20180297929A1 true US20180297929A1 (en) 2018-10-18

Family

ID=63791570

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/956,172 Abandoned US20180297929A1 (en) 2017-04-18 2018-04-18 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
US16/402,853 Abandoned US20200148617A1 (en) 2017-04-18 2019-05-03 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
US17/583,081 Pending US20220332672A1 (en) 2017-04-18 2022-01-24 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/402,853 Abandoned US20200148617A1 (en) 2017-04-18 2019-05-03 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
US17/583,081 Pending US20220332672A1 (en) 2017-04-18 2022-01-24 Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor

Country Status (9)

Country Link
US (3) US20180297929A1 (fr)
EP (1) EP3612171A4 (fr)
JP (2) JP2020516622A (fr)
KR (1) KR20200054910A (fr)
CN (1) CN110996928A (fr)
CA (1) CA3059073A1 (fr)
IL (1) IL269884B2 (fr)
TW (1) TW201902471A (fr)
WO (1) WO2018195163A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10227285B2 (en) 2014-11-14 2019-03-12 Gemphire Therapeutics Inc. Processes and intermediates for preparing alpha,omega-dicarboxylic acid-terminated dialkane ethers
WO2020081837A1 (fr) * 2018-10-18 2020-04-23 Gemphire Therapeutics Inc. Gemcabène, sels pharmaceutiquement acceptables de celui-ci, compositions correspondantes et procédés d'utilisation associés

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1197242A (fr) * 1983-03-02 1985-11-26 Jean A. Gauthier Derives de l'acide pyrimido¬1,2-a|pyrrolo ¬2,1-c|¬1,4|benzodiazepine-3-carboxylique
US5648387A (en) * 1995-03-24 1997-07-15 Warner-Lambert Company Carboxyalkylethers, formulations, and treatment of vascular diseases
US6861555B2 (en) * 2000-01-25 2005-03-01 Warner-Lambert Company Calcium dicarboxylate ethers, methods of making same, and treatment of vascular disease and diabetes therewith
HUP0204422A3 (en) * 2000-01-25 2003-08-28 Warner Lambert Co Calcium dicarboxylate ethers, methods of making the compounds, and pharmaceutical compositions containing them
KR20050058454A (ko) * 2002-08-22 2005-06-16 워너-램버트 캄파니 엘엘씨 골관절염의 치료 방법
US20050026979A1 (en) * 2003-07-31 2005-02-03 Maha Ghazzi Methods for treating inflammation and inflammation-associated diseases with a statin and ether
WO2012170676A1 (fr) * 2011-06-08 2012-12-13 Sti Pharma, Llc Préparation organique pharmaceutique hydrosoluble à absorption contrôlée administrée une fois par jour
CA2861643C (fr) * 2012-01-06 2020-10-06 Charles L. Bisgaier Methodes de reduction du risque de maladie cardiovasculaire
KR20170083535A (ko) * 2014-11-14 2017-07-18 젬파이어 세러퓨틱스 인코포레이티드 α,ω-다이카복실산-말단 다이알케인 에터를 제조하기 위한 방법 및 중간체
BR112018007857A2 (pt) * 2015-11-06 2018-10-30 Gemphire Therapeutics Inc combinações de gemcabene para o tratamento de doença cardiovascular

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10227285B2 (en) 2014-11-14 2019-03-12 Gemphire Therapeutics Inc. Processes and intermediates for preparing alpha,omega-dicarboxylic acid-terminated dialkane ethers
WO2020081837A1 (fr) * 2018-10-18 2020-04-23 Gemphire Therapeutics Inc. Gemcabène, sels pharmaceutiquement acceptables de celui-ci, compositions correspondantes et procédés d'utilisation associés

Also Published As

Publication number Publication date
IL269884B1 (en) 2023-05-01
KR20200054910A (ko) 2020-05-20
CA3059073A1 (fr) 2018-10-25
US20200148617A1 (en) 2020-05-14
TW201902471A (zh) 2019-01-16
WO2018195163A1 (fr) 2018-10-25
EP3612171A1 (fr) 2020-02-26
IL269884B2 (en) 2023-09-01
JP2020516622A (ja) 2020-06-11
IL269884A (fr) 2019-11-28
US20220332672A1 (en) 2022-10-20
CN110996928A (zh) 2020-04-10
JP2023057082A (ja) 2023-04-20
EP3612171A4 (fr) 2020-10-21

Similar Documents

Publication Publication Date Title
US20220332672A1 (en) Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
JP2020514352A (ja) 組合せ療法のための医薬組成物
JP2023096079A (ja) 化合物の製剤およびそれらの使用
US8834858B2 (en) Treatment for dyslipidemia
JP4901218B2 (ja) 併用医薬
US20200253877A1 (en) Gemcabene, pharmaceutically acceptable salts thereof, compositions thereof and methods of use therefor
JP2021522247A (ja) 肝疾患における好中球エラスターゼ阻害薬の使用
US20120141586A1 (en) Thrombin receptor antagonist and clopidogrel fixed dose tablet
Markel The resurgence of niacin: from nicotinic acid to niaspan/laropiprant
JP2011525194A (ja) 線維性疾患または病態を治療するための組成物
TWI651086B (zh) 用以治療或預防心血管疾病之膽固醇酯轉蛋白質(cetp)抑制劑及包含該抑制劑之醫藥組成物
US20190008779A1 (en) Gemcabene compositions and methods of use thereof
EP2734198A1 (fr) Méthode et composition pharmaceutique améliorée destinées à améliorer l'absorption d'un promédicament ester
CA2749534A1 (fr) Inhibiteur de phospholipase a2 (spla2) secretoire, compositions de medicament a base de niacine et methodes de traitement des maladies cardiovasculaires et de la dyslipidemie
JP2021533190A (ja) LDLコレステロールの細胞内クリアランスを増強しかつスタチンとの併用療法で効力を高めかつ副作用を減らすための、カルバメートβフェニルエタノールアミン類似体の新規使用法
BR102013028912A2 (pt) composição farmacêutica, forma farmacêutica oral, cápsula, comprimido de dupla camada, usos, método de tratamento de hipercolesterolemia, hipertrigliceridemia e/ou dislipidemia mista e método de prevenção de aterosclerose, diabetes ou prevenção secundária de outras doenças cardiovasculares
TWI684465B (zh) 含雙醋瑞因之製劑及使用其於降低尿酸血中濃度的方法
WO2024054996A1 (fr) Méthodes pour le traitement de la dépression
CA3117958A1 (fr) Traitement et inhibition de la fibrose chez des patients atteints d'une maladie hepatique non alcoolique
WO2011052499A1 (fr) Composition pharmaceutique présentant une stabilité au stockage améliorée
WO2022051319A1 (fr) Combinaisons de doses fixes de chs-131 et d'un inhibiteur de dpp-4
Radiosensitizer Azelnidipine Antihypertensive
JP2018513203A (ja) ジアセレインを含有する製剤及びそれを用いて尿酸の血中濃度を低下させる方法
KR20100109840A (ko) 분비성 포스포리파제 a2 (spla2) 억제제를 사용한 심혈관 질환 및 이상지혈증의 치료 및 spla2 억제제 조합 요법

Legal Events

Date Code Title Description
AS Assignment

Owner name: GEMPHIRE THERAPEUTICS INC., MICHIGAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ONICIU, DANIELA CARMEN;BISGAIER, CHARLES LARRY;REEL/FRAME:045677/0829

Effective date: 20180430

AS Assignment

Owner name: GEMPHIRE THERAPEUTICS INC., MICHIGAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CORDEN PHARMA SWITZERLAND LLC;REEL/FRAME:045732/0804

Effective date: 20180504

Owner name: CORDEN PHARMA SWITZERLAND LLC, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GOMES, JOSE RUI;REEL/FRAME:045732/0799

Effective date: 20180504

AS Assignment

Owner name: GEMPHIRE THERAPEUTICS INC., MICHIGAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CORDEN PHARMA SWITZERLAND LLC;REEL/FRAME:046551/0585

Effective date: 20180730

Owner name: CORDEN PHARMA SWITZERLAND LLC, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HECKHOFF, STEFAN;REEL/FRAME:046551/0573

Effective date: 20180730

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: GEMPHIRE THERAPEUTICS INC., MICHIGAN

Free format text: CHANGE OF ADDRESS FOR ASSIGNEE;ASSIGNOR:GEMPHIRE THERAPEUTICS INC.;REEL/FRAME:051136/0270

Effective date: 20191126

AS Assignment

Owner name: NEUROBO PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:GEMPHIRE THERAPEUTICS INC.;REEL/FRAME:052724/0193

Effective date: 20191230