US20050075337A1 - Pharmaceutical compositions including carotenoid analogs or derivatives for the inhabition and amelioration of disease - Google Patents

Pharmaceutical compositions including carotenoid analogs or derivatives for the inhabition and amelioration of disease Download PDF

Info

Publication number
US20050075337A1
US20050075337A1 US10/793,702 US79370204A US2005075337A1 US 20050075337 A1 US20050075337 A1 US 20050075337A1 US 79370204 A US79370204 A US 79370204A US 2005075337 A1 US2005075337 A1 US 2005075337A1
Authority
US
United States
Prior art keywords
pharmaceutical composition
derivative
carotenoid
carotene
carotenoid analog
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
US10/793,702
Other languages
English (en)
Inventor
Samuel Lockwood
Sean O'Malley
David Watumull
Laura Hix
Henry Jackson
Geoff Nadolski
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.)
Cardax Pharmaceuticals Inc
Original Assignee
Hawaii Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hawaii Biotech Inc filed Critical Hawaii Biotech Inc
Priority to US10/793,702 priority Critical patent/US20050075337A1/en
Assigned to HAWAII BIOTECH, INC. reassignment HAWAII BIOTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HIX, LAURA M., JACKSON, HENRY, LOCKWOOD, SAMUEL FOURNIER, NADOLSKI, GEOFF, O'MALLEY, SEAN, WATUMULL, DAVID G.
Publication of US20050075337A1 publication Critical patent/US20050075337A1/en
Assigned to CARDAX PHARMACEUTICALS, INC. reassignment CARDAX PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAWAII BIOTECH, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C403/00Derivatives of cyclohexane or of a cyclohexene or of cyclohexadiene, having a side-chain containing an acyclic unsaturated part of at least four carbon atoms, this part being directly attached to the cyclohexane or cyclohexene or cyclohexadiene rings, e.g. vitamin A, beta-carotene, beta-ionone
    • C07C403/24Derivatives of cyclohexane or of a cyclohexene or of cyclohexadiene, having a side-chain containing an acyclic unsaturated part of at least four carbon atoms, this part being directly attached to the cyclohexane or cyclohexene or cyclohexadiene rings, e.g. vitamin A, beta-carotene, beta-ionone having side-chains substituted by six-membered non-aromatic rings, e.g. beta-carotene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/401Proline; Derivatives thereof, e.g. captopril
    • 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
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • 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
    • A61P1/10Laxatives
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/16Emollients or protectives, e.g. against radiation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • 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
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/301,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/58One oxygen atom, e.g. butenolide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/117Esters of phosphoric acids with cycloaliphatic alcohols
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H13/00Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids
    • C07H13/02Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by carboxylic acids
    • C07H13/04Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by carboxylic acids having the esterifying carboxyl radicals attached to acyclic carbon atoms

Definitions

  • 60/472,831 entitled “Structural Carotenoid Analogs for the Inhibition and Amelioration of Disease” filed on May 22, 2003; Provisional Patent Application No. 60/473,741 entitled “Structural Carotenoid Analogs for the Inhibition and Amelioration of Disease” filed on May 28, 2003; and Provisional Patent Application No. 60/485,304 entitled “Structural Carotenoid Analogs for the Inhibition and Amelioration of Disease” filed on Jul. 3, 2003.
  • the invention generally relates to the fields of medicinal and synthetic chemistry. More specifically, the invention relates to the synthesis and use of carotenoid analogs or derivatives.
  • CVD cardiovascular disease
  • CAD coronary artery disease
  • CVD cardiovascular disease
  • Ischemia is the lack of an adequate oxygenated blood supply to a particular tissue. Ischemia underlies many acute and chronic disease states including, but not limited to:
  • systemic inflammation e.g., serum levels of C-reactive protein or CRP
  • CRP C-reactive protein
  • Myocardial salvage appears to be maximal in a 2 to 6 hour “therapeutic window” subsequent to acute plaque rupture and thrombosis.
  • this therapeutic window is even narrower, generally less than 3 hours post-thrombosis.
  • Recombinant tissue-type plasminogen activator administered within 3 hours of ischemic stroke significantly improves clinical outcome, but increases the risk of hemorrhage.
  • Ischemia creates changes in the affected tissue, with the potential fmal result of contraction band and/or coagulation necrosis of at-risk myocardium.
  • Pathologic changes in ischemic myocardium include, but are not limited to:
  • Gap junctions are a unique type of intercellular junction found in most animal cell types. They form aqueous channels that interconnect the cytoplasms of adjacent cells and enable the direct intercellular exchange of small (less than approximately 1 kiloDalton) cytoplasmic components. Gap junctions are created across the intervening extracellular space by the docking of two hemichannels (“connexons”) contributed by each adjacent cell. Each hemichannel of is an oligomer of six connexin molecules.
  • Connexin 43 was the second connexin gene discovered and it encodes one of the most widely expressed connexins in established cell lines and tissues. Gap junctions formed by connexin 43 have been implicated in development, cardiac function, and growth control.
  • Cardiac arrhythmia is generally considered a disturbance of the electrical activity of the heart that manifests as an abnormality in heart rate or heart rhythm. Patients with a cardiac arrhythmia may experience a wide variety of symptoms ranging from palpitations, to fainting (“syncope”), and sudden cardiac death.
  • connexin 43 The major connexin in the cardiovascular system is connexin 43. Gap junctional coordination of cellular responses among cells of the vascular wall, in particular the endothelial cells, is thought to be critical for the local modulation of vasomotor tone and for the maintenance of circulatory homeostasis. Controlling the upregulation of connexin 43 may also assist in the maintenance of electrical stability in cardiac tissue. Maintaining electrical stability in cardiac tissue may benefit the health of hundreds of thousands of people a year with some types of cardiovascular disease [e.g., ischemic heart disease (IHD) and arrhythmia], and may prevent the occurrence of sudden cardiac death in patients at high risk for arrhythmia.
  • IHD ischemic heart disease
  • Cancer is generally considered to be characterized by the uncontrolled, abnormal growth of cells.
  • Connexin 43 is also associated with cellular growth control. Growth control by connexin 43 is likely due to connexin 43's association with gap junctional communication. Maintenance, restoration, or increases of functional gap junctional communication inhibits the proliferation of transformed cells. Therefore, upregulation and/or control of the availability of connexin 43 may potentially inhibit and/or ameliorate the spread of cancerous cells.
  • Chronic liver injury regardless of etiology, may lead to a progressive spectrum of pathology from acute and chronic inflammation, to early stage fibrosis, and finally to cirrhosis, end-stage liver disease (ESRD), and hepatocellular carcinoma (HCC).
  • ESRD end-stage liver disease
  • HCC hepatocellular carcinoma
  • a cascade of inflammatory events secondary to the initiating injury including the release of cytokines and the formation of reactive oxygen species (ROS), activates hepatic stellate cells (HSC).
  • HSC produce extracellular matrix components (ECM), including collagen, and are critical in the process which generates hepatic fibrosis.
  • End-stage liver disease [manifested as either cirrhosis or hepatocellular carcinoma (HCC)] is the eighth leading cause of disease-related death in the United States.
  • Chronic inflammation in the liver resulting from viral infection, alcohol abuse, drug-induced toxicity, iron and copper overload, and many other factors can initiate hepatic fibrosis.
  • By-products of hepatocellular damage activate Kupffer cells, which then release a number of cytokines, ROS (including in particular superoxide anion), and other paracrine and autocrine factors which in turn act upon hepatic stellate cells (HSC).
  • ROS including in particular superoxide anion
  • HSC hepatic stellate cells
  • ROS can induce HSC cells.
  • Elevated levels of indirect markers of oxidative stress e.g., thiobarbituric acid reactive species or TBARS
  • TBARS thiobarbituric acid reactive species
  • levels of gluthathione, glutathione peroxidase, superoxide dismutase, carotenoids, and a-tocopherol (vitamin E) are significantly lower in patients with chronic liver disease.
  • Supplying these endogenous and/or exogenous antioxidants reverses many of the signs of chronic liver disease, including both surrogate markers for the disease process, as well as direct measurements of hepatic fibrosis. Therefore, they are likely potent agents for therapeutic intervention in liver disease.
  • the administration of structural analogs or derivatives of carotenoids may inhibit and/or ameliorate the occurrence of diseases in subjects.
  • Maladies which may be treated with structural analogs or derivatives of carotenoids may include any disease that involves production of reactive oxygen species and/or other radical and non-radical species (for example singlet oxygen, a reactive oxygen species but not a radical).
  • water-soluble analogs of carotenoids may be used to treat a disease that involves production of reactive oxygen species. Oxidation of DNA, proteins, and lipids by reactive oxygen species and other radical and non-radical species has been implicated in a host of human diseases.
  • Radicals may be the primary cause for the following conditions, may make the body more susceptible to other disease-initiating factors, may inhibit endogenous defenses and repair processes, and/or may enhance the progression of incipient disease(s).
  • In the first category are those disease conditions in which a single organ is primarily affected, and for which evidence exists that radicals and/or non-radicals are involved in the pathology of the disease. These examples are not to be seen as limiting, and additional disease conditions will be obvious to those skilled in the art.
  • RAnget al. Throat age-related macular degeneration
  • AMD Throat age-related macular degeneration
  • hypertensive retinal disease uveitis, choroiditis, vitreitis, ocular hemorrhage, degenerative retinal damage, cataractogenesis and cataracts, retinopathy of prematurity, Meuniere's disease, drug-induced ototoxicity (including is aminoglycoside and furosemide toxicity), infectious and idiopathic otitis, otitis media, infectious and allergic sinusitis, head and neck cancer;
  • senile dementia including Alzheimer's dementia
  • Neuman-Pick's disease including Alzheimer's dementia
  • neurotoxin reactions including hyperbaric oxygen effects, Parkinson's disease, cerebral and spinal cord trauma, hypertensive cerebrovascular injury, stroke (thromboembolic, thrombotic, and hemorrhagic), infectious encephalitis and meningitis, allergic encephalomyelitis and other demyelinating diseases, amyotrophic lateral sclerosis (ALS), multiple sclerosis, neuronal ceroid lipofuscinoses, ataxia-telangiectasia syndrome, aluminum, iron, and other heavy metal(s) overload, primary brain carcinoma/malignancy and brain metastases;
  • ALS amyotrophic lateral sclerosis
  • Cardiovascular arteriosclerosis, atherosclerosis, peripheral vascular disease, myocardial infarction, chronic stable angina, unstable angina, idiopathic surgical injury (during CABG, PTCA), inflammatory heart disease [as measured and influenced by C-reactive protein (CRP) and myeloperoxidase (MPO)], vascular restenosis, low-density lipoprotein oxidation (ox-LDL), cardiomyopathies, cardiac arrhythmia (ischemic and post-myocardial infarction induced), congestive heart failure (CHF), drug toxicity (including adriamycin and doxorubicin), Keshan disease (selenium deficiency), trypanosomiasis, alcohol cardiomyopathy, venous stasis and injury (including deep venous thrombosis or DVT), thrombophlebitis;
  • CRP C-reactive protein
  • MPO myeloperoxidase
  • vascular restenosis vascular
  • Pulmonary asthma, reactive airways disease, chronic obstructive pulmonary disease (COPD or emphysema), hyperoxia, hyperbaric oxygen effects, cigarette smoke inhalation effects, environmental oxidant pollutant effects, acute respiratory distress syndrome (ARDS), bronchopulmonary dysplasia, mineral dust pneumoconiosis, adriamycin toxicity, bleomycin toxicity, paraquat and other pesticide toxicities, chemical pneumonitis, idiopathic pulmonary interstitial fibrosis, infectious pneumonia (including fungal), sarcoidosis, asbestosis, lung cancer (small- and large-cell), anthrax infection, anthrax toxin exposure;
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • bronchopulmonary dysplasia mineral dust pneumoconiosis
  • adriamycin toxicity bleomycin toxicity
  • paraquat and other pesticide toxicities chemical pneumonitis, id
  • Renal hypertensive renal disease, end-stage renal disease, diabetic renal disease, infectious glomerulonephritis, nephrotic syndrome, allergic glomerulonephritis, type I-IV hypersensitivity reactions, renal allograft rejection, nephritic antiglomerular basement membrane disease, heavy metal nephrotoxicity, drug-induced (including aminoglycoside, furosemide, and non-steroidal anti-inflammatory) nephrotoxicity, rhabdomyolisis, renal carcinoma;
  • Hepatic carbon tetrachloride liver injury, endotoxin and lipopolysaccharide liver injury, chronic viral infection (including Hepatitis infection), infectious hepatitis (non-viral etiology), hemachromatosis, Wilson's disease, acetaminophen overdose, congestive heart failure with hepatic congestion, cirrhosis (including alcoholic, viral, and idiopathic etiologies), hepatocellular carcinoma, hepatic metastases;
  • Gastrointestinal inflammatory bowel disease (including Crohn's disease, ulcerative colitis, and irritable bowel syndrome), colon carcinoma, polyposis, infectious diverticulitis, toxic megacolon, gastritis (including Helicobacter pylori infection), gastric carcinoma, esophagitis (including Barrett's esophagus), gastro-esophageal reflux disease (GERD), Whipple's disease, gallstone disease, pancreatitis, abetalipoproteinemia, infectious gastroenteritis, dysentery, nonsteroidal anti-inflammatory drug-induced toxicity;
  • Hematopoietic/Hematologic Pb (lead) poisoning, drug-induced bone marrow suppression, protoporphyrin photo-oxidation, lymphoma, leukemia, porphyria(s), parasitic infection (including malaria), sickle cell anemia, thallasemia, favism, pernicious anemia, Fanconi's anemia, post-infectious anemia, idiopathic thrombocytopenic purpura (ITP), autoimmune deficiency syndrome (AIDS);
  • Genitourinary infectious prostatitis, prostate carcinoma, benign prostatic hypertrophy (BPH), urethritis, orchitis, testicular torsion, cervicitis, cervical carcinoma, ovarian carcinoma, uterine carcinoma, vaginitis, vaginismus;
  • Musculoskeletal osteoarthritis, rheumatoid arthritis, tendonitis, muscular dystrophy, degenerative disc disease, degenerative joint disease, exercise-induced skeletal muscle injury, carpal tunnel syndrome, Guillan-Barre syndrome, Paget's disease of bone, ankylosing spondilitis, heterotopic bone formation; and
  • Integumentary solar radiation injury (including sunburn), thermal injury, chemical and contact dermatitis (including Rhus dermatitis), psoriasis, Bloom syndrome, leukoplakia (particularly oral), infectious dermatitis, Kaposi's sarcoma.
  • aging including age-related immune deficiency and premature aging disorders, cancer, cardiovascular disease, cerebrovascular disease, radiation injury, alcohol-mediated damage (including Wernicke-Korsakoff's syndrome), ischemia-reperfusion damage, inflammatory and auto-immune disease, drug toxicity, amyloid disease, overload syndromes (iron, copper, etc.), multi-system organ failure, and endotoxemia/sepsis.
  • Maladies which may be treated with structural carotenoid analogs or derivatives, may include, but are not limited to, cardiovascular inflammation, hepatitis C infection, cancer (hepatocellular carcinoma and prostate), macular degeneration, rheumatoid arthritis, stroke, Alzheimer's disease, and/or osteoarthritis.
  • the administration of water soluble analogs or derivatives of carotenoids to a subject may inhibit and/or ameliorate the occurrence of ischemia-reperfusion injury in subjects.
  • water soluble and other structural carotenoid analogs or derivatives may be administered to a subject alone or in combination with other structural carotenoid analogs or derivatives.
  • ischemia-reperfusion injury in a human subject that is experiencing, or has experienced, or is predisposed to experience myocardial infarction, stroke, peripheral vascular disease, venous or arterial occlusion and/or restenosis, organ transplantation, coronary artery bypass graft surgery, percutaneous transluminal coronary angioplasty, and cardiovascular arrest and/or death may be inhibited or ameliorated by the administration of therapeutic amounts of water soluble and/or other structural carotenoid analogs or derivatives to the subject.
  • Water soluble structural carotenoid analogs or derivatives are those analogs or derivatives which may be formulated in aqueous solution, either alone or with excipients.
  • Water soluble carotenoid analogs or derivatives may include those compounds and synthetic derivatives which form molecular self-assemblies, and may be more properly termed “water dispersible” carotenoid analogs or derivatives.
  • Water soluble and/or “water-dispersible” carotenoid analogs or derivatives may be preferred in some embodiments of the current invention.
  • Water soluble carotenoid analogs or derivatives may have a water solubility of greater than about 1 mg/mL in some embodiments. In certain embodiments, water soluble carotenoid analogs or derivatives may have a water solubility of greater than about 10 mg/mL. In some embodiments, water soluble carotenoid analogs or derivatives may have a water solubility of greater than about 50 mg/mL.
  • the administration of water soluble analogs or derivatives of carotenoids to a subject may inhibit and/or ameliorate some types of cardiovascular disease associated with cardiac arrhythmia.
  • water soluble analogs or derivatives of carotenoids may be administered to a subject alone or in combination with other carotenoid analogs or derivatives.
  • Carotenoid analogs or derivatives may assist in the maintenance of electrical stability in cardiac tissue. Assistance in the maintenance of electrical stability in cardiac tissue may inhibit and/or ameliorate some types of cardiovascular disease, including in particular sudden cardiac death attributable to lethal cardiac arrhythmia.
  • the administration of water soluble analogs or derivatives of carotenoids to a subject may inhibit and/or ameliorate the occurrence of liver disease in the subject.
  • water soluble analogs or derivatives of carotenoids may be administered to a subject alone or in combination with other carotenoid analogs or derivatives.
  • the liver disease may be a chronic liver disease such as, for example, Hepatitis C infection.
  • water soluble analogs or derivatives of carotenoids may inhibit and/or ameliorate the proliferation and propagation of initiated, transformed and/or cancerous or pre-cancerous cell(s).
  • water soluble analogs or derivatives of carotenoids may be administered to a subject alone or in combination with other carotenoid analogs or derivatives.
  • Carotenoid analogs or derivatives may inhibit the proliferation rate of carcinogen-initiated cells.
  • Carotenoid analogs or derivatives may increase connexin 43 expression. Increase of connexin 43 expression may increase, maintain, or restore gap junctional intercellular communication and thus inhibit the growth of carcinogen-initiated cells.
  • Embodiments may be further directed to pharmaceutical compositions comprising combinations of structural carotenoid analogs or derivatives to said subjects.
  • the composition of an injectable structural carotenoid analog or derivative of astaxanthin may be particularly useful in the therapeutic methods described herein.
  • an injectable astaxanthin structural analog or derivative is administered with another astaxanthin structural analog or derivative and/or other carotenoid structural analogs or derivatives, or in formulation with other antioxidants and/or excipients that further the intended purpose.
  • one or more of the astaxanthin structural analogs or derivatives are water soluble.
  • carotenoid analog and carotenoid derivative may generally refer to in some embodiments chemical compounds or compositions derived from a naturally occurring carotenoid.
  • terms such as carotenoid analog and carotenoid derivative may generally refer to chemical compounds or compositions which are synthetically derived from non-carotenoid based parent compounds; however, which ultimately substantially resemble a carotenoid derived analog.
  • terms such as carotenoid analog and carotenoid derivative may generally refer to a synthetic derivative of a naturally occurring carotenoid.
  • a chemical compound including a carotenoid derivative may have the general structure (I): Each R 3 may be independently hydrogen or methyl. R 1 and R 2 may be independently H, an acyclic alkene with one or more substituents, or a cyclic ring including one or more substituents. y may be 5 to 12. In some embodiments, y may be about 3 to about 15. In certain embodiments, the maximum value of y may only be limited by the ultimate size of the chemical compound, particularly as it relates to the size of the chemical compound and the potential interference with the chemical compound's biological availability as discussed herein. In some embodiments, substituents may be at least partially hydrophilic. In some embodiment, substituents may be each independently coupled to a carotenoid analog or derivative via an ether and/or an ester functionality. These carotenoid derivatives may be used in a pharmaceutical composition.
  • a chemical compound including a carotenoid derivative may have the general structure (Ia): Each R 3 may be independently hydrogen or methyl. R 1 and R 2 may be independently H, an acyclic alkene with one or more substituents, or a cyclic ring including one or more substituents. In some embodiments, substituents may be at least partially hydrophilic. These carotenoid derivatives may be used in a pharmaceutical composition. In one embodiment, a pharmaceutical composition that includes carotenoid structural analogs or derivatives having general structure (Ia) may be used for treating ischemia-reperfusion injury.
  • the terms “disodium salt disuccinate astaxanthin derivative”, “dAST”, “Cardax”, “Cardax”, “rac”, and “astaxanthin disuccinate derivative (ADD)” represent varying nomenclature for the use of the disodium salt disuccinate astaxanthin derivative in various stereoisomer and aqueous formulations, and represent illustrative embodiments for the intended use of this structural carotenoid analog.
  • the diacid disuccinate astaxanthin derivative (astaCOOH) is the protonated form of the derivative utilized for flash photolysis studies for direct comparison with non-esterified, “racemic” (i.e., mixture of stereoisomers) astaxanthin.
  • Cardax-C is the disodium salt disuccinate di-vitamin C derivative (derivative XXIII) utilized in superoxide anion scavenging experiments assayed by electron paramagnetic resonance (EPR) spectroscopy.
  • FIG. 1 depicts a graphic representation of several examples of “parent” carotenoid structures as found in nature.
  • FIG. 2 depicts an effect of disodium salt disuccinate astaxanthin derivative on the reactive oxygen species superoxide anion as monitored using electron paramagnetic resonance (EPR) spectroscopy.
  • EPR electron paramagnetic resonance
  • FIG. 3 depicts an effect of a disodium salt disuccinate astaxanthin derivative/free vitamin C solution on the reactive oxygen species superoxide anion as monitored using electron paramagnetic resonance (EPR) spectroscopy.
  • EPR electron paramagnetic resonance
  • FIG. 4 depicts a graphical representation of a relative reduction of infarct size in male Sprague-Dawley rats with pre-treatment using a disodium salt disuccinate astaxanthin derivative intravenous formulation (CardaxTM).
  • CardaxTM disodium salt disuccinate astaxanthin derivative intravenous formulation
  • FIG. 5 depicts the chemical structure of the all-trans (all-E) disodium salt disuccinate ester derivative of meso-astaxanthin (3R,3′S- or 3S,3!R-dihydroxy- ⁇ , ⁇ -carotene-4,4′-dione; DAST) synthesized for the current study (shown as the all-E dianionic bolamphiphile).
  • FIG. 8 depicts the induced CD and UV/Vis spectra obtained by titration of human serum albumin (HSA) with dAST in Ringer buffer solution (pH 7.4) at low LP ratios.
  • HSA human serum albumin
  • dAST dAST in Ringer buffer solution
  • FIG. 8 depicts the induced CD and UV/Vis spectra obtained by titration of human serum albumin (HSA) with dAST in Ringer buffer solution (pH 7.4) at low LP ratios.
  • Insets molar circular dichroic absorption coefficients ( ⁇ in M ⁇ 1 cm ⁇ 1 ) and molar absorption coefficients ( ⁇ in M ⁇ 1 cm ⁇ 1 ) of the induced CD and absorption bands calculated on the basis of total
  • FIG. 11 depicts an illustration of right-handed chiral arrangements of two meso-carotenoid molecules for which excitonic interactions produce long-wavelength positive and short-wavelength negative Cotton effects in the CD spectrum. Gray-colored molecules lie behind the plane of the paper.
  • FIG. 12 depicts (upper figure): fluorescence quenching of HSA by dAST measured in 0.1 M pH 7.4 phosphate buffer solution at 37° C. Initial and final concentrations of HSA and the ligand were varied between 4.2 ⁇ 10 ⁇ 6 M ⁇ 4.0 ⁇ 10 ⁇ 6 M and 1.3 ⁇ 10 ⁇ 6 M ⁇ 1.4 ⁇ 10 ⁇ 5 M, respectively. LIP ratios are noted on curves. The lower figure depicts an effect of DMSO alone on the intrinsic fluorescence of HSA.
  • FIG. 13 depicts the X-ray crystallographic structure of fatty acid-free HSA. Subdomains and the two primary drug-binding sites of HSA are indicated. Dotted bar represents spatial dimension of the interdomain cleft, and asterisk indicates the position of Trp214. The inter-atomic distance between the 3 and 3′ chiral carbon atoms of the DAST molecule is 28 ⁇ .
  • FIG. 14 depicts that the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivative (“rac” in Figure Legends) induces functional gap junctional communication in murine embryonic fibroblast (10T1 ⁇ 2) cells. Confluent cultures were treated for 4 days as described in text, then assayed for the ability to transfer the fluorescent dye Lucifer Yellow. Arrows indicate the cell injected with Lucifer Yellow.
  • FIG. 15 A depicts connexin 43 protein expression in cells treated with the mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivatives as assessed by quantitative Western blot analysis.
  • the upper bands are believed to represent the phosphorylated forms of the protein assembled into gap junctions; lower bands unassembled proteins (Saez, 1998).
  • Lane 1 1:2 ethanol (EtOH)/ H 2 O (solvent only negative control); Lane 2: TTNPB, a synthetic retinoid, in acetone at 10 ⁇ 8 M (positive control); Lane 3: Retinyl acetate in acetone at 10 ⁇ 5 M (positive control); Lane 4: Statistical mixture (“rac”) of stereoisomers of the disodium salt disuccinate astaxanthin derivative at 10 ⁇ 5 M delivered in a 1:2 formulation of EtOH/ H 2 O; Lane 5: 3R,3′R-disodium salt disuccinate astaxanthin derivative at 10 ⁇ 5 M delivered in a 1:2 formulation of EtOH/ H 2 O; Lane 6: 3S,3′S disodium salt disuccinate astaxanthin derivative at 10 ⁇ 5 M delivered in a 1:2 formulation of EtOH/ H 2 O; Lane 7: Meso (3R,3′S) disodium salt disuccinate astaxanthin derivative at 10 ⁇ 5 M delivered in a 1:2 formulation of EtOH/ H 2 O.
  • FIG. 15B depicts an immunoblot stained with Coomassie blue to demonstrate equal protein loading of all the bands. This confirms that differences in immunolabeling are not an artifact due to variability in total protein loaded and/or transferred to the membrane.
  • FIG. 15D depicts the dose-response curve of Cx43 protein expression in murine embryonic fibroblast cells (10T1 ⁇ 2) treated with the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivatives as assessed by quantitative Western blot analysis.
  • the upper bands are believed to represent the phosphorylated forms of the protein assembled into gap junctions; lower bands unassembled proteins.
  • Lane 1 1:2 EtOH/ H 2 O (solvent only negative control).
  • Lane 2 T[NPB in acetone at 10 ⁇ 8 M (positive control).
  • Lane 3 disodium salt disuccinate astaxanthin derivative (“rac”) at 10 ⁇ 5 M delivered in a 1:2 formulation of EtOH/ H 2 O.
  • Lane 4 disodium salt disuccinate astaxanthin derivative (“rac”) at 5 ⁇ 10 ⁇ 6 M delivered in a 1:2 formulation of EtOH/ H 2 O.
  • Lane 5 disodium salt disuccinate astaxanthin derivative (“rac”) at 10 ⁇ 6 M delivered in a 1:2 formulation of EtOH/ H 2 O.
  • FIG. 16 depicts that the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivative increases the assembly of Cx43 immunoreactive junctional plaques.
  • Confluent cultures of 10T1 ⁇ 2 cells were treated for 4 days as described above with the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivative: (1) at 10 ⁇ 5 M in 1:2 EtOH/ H 2 O; (2) with 1:2 EtOH/H 2 O as solvent only negative control; or (3) TTNPB at 10 ⁇ 8 M in tetrahydrofuran (THF) solvent as positive control.
  • TTNPB tetrahydrofuran
  • Panel A the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivative at 10 ⁇ 5 M in 1:2 EtOHI H 2 O; Panel C: 1:2 EtOH/ H 2 O as solvent control; Panel E: TTNPB at 10 ⁇ 8 M in tetrahydrofuran (THF) solvent as positive control.
  • Panels B, D, and F digital analysis of panels A, C, and E, respectively, demonstrating pixels above a fixed set threshold positive for fluorescent intensity.
  • Light gray arrows immunoreactive junctional plaques
  • dark gray arrows position of cell nuclei.
  • junctional immunoreactive plaques in the cultures treated with the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivative in comparison with solvent-only treated controls.
  • the junctional plaques shown in Panels C and D represent infrequent plaques seen in controls; most cells in these cultures were negative for Cx43 staining.
  • FIG. 17 depicts the 3 stereoisomers of the disodium disuccinate diester of astaxanthin synthesized for the current studies (shown as the all-E geometric isomers); the mixture of stereoisomers, or individual stereoisomers, were used in separate applications (see Figure legends). Note that the meso forms (3R,3′S and 3S,3′R) are identical.
  • FIG. 18 depicts the mean percent inhibition of superoxide anion signal as detected by DEPMPO spin trap by the disodium disuccinate derivatives of astaxanthin in pure aqueous formulation.
  • Mixture statistical mixture of stereoisomers [3S,3′S, meso (3R,3′S and 3′R,3S), 3R,3′R in a 1:2:1 ratio].
  • Each derivative in aqueous formulation was standardized to control EPR signal detected without addition of compound (set at 0% inhibition by convention). Note the absence of superoxide inhibition by 3S,3′S formulation in water. In each case, the aqueous formulation is less potent than the corresponding formulation in EtOH ( FIG. 19 ).
  • FIG. 19 depicts the mean percent inhibition of superoxide anion signal as detected by DEPMPO spin trap by the disodium disuccinate derivatives of astaxanthin in ethanolic formulation.
  • Mixture statistical mixture of stereoisomers [3S,3′S, meso (3R,3′S and 3′R,3S), 3R,3′R in a 1:2:1 ratio].
  • the mixture, meso, and 3R,3′R stock solutions were 1:2 ethanol/water (331 ⁇ 3% EtOH); the 3S,3′S stock solution was 1:1 ethanol/water (50% EtOH).
  • Final concentration of EtOH in the isolated neutrophil test assay was 0.3% and 0.5%, respectively.
  • Each derivative in ethanolic formulation was standardized to control EPR signal detected without addition of compound (set at 0% inhibition by convention).
  • FIG. 20 depicts the mean percent inhibition of superoxide anion signal as detected by DEPMPO spin trap by the mixture of stereoisomers of the disodium disuccinate derivative of astaxanthin (tested in 1:2 EtOH/water formulation; final EtOH concentration in isolated neutrophil assay 0.3%). As the concentration of the derivative increases, inhibition increases in a non-linear, dose-dependent manner. At 3 mM, near-complete inhibition of superoxide anion signal is seen (95.0% inhibition).
  • FIG. 21 depicts the mean percent inhibition of superoxide anion signal as detected by DEPMPO spin trap by the hydrochloride salt dilysine astaxanthin derivative.
  • This derivative was highly water soluble (>50 mg/mL), and did not require a co-solvent for excellent radical-quenching ability in this assay. Compare the superoxide anion inhibition of this derivative with that depicted in FIG. 20 , for a derivative that forms supramolecular assemblies in pure aqueous formulation.
  • FIG. 22 depicts a standard plot of concentration of non-esterified, free astaxanthin versus time for plasma after single dose oral gavage in black mice. Only non-esterified, free astaxanthin is detected in plasma, corroborating the complete de-esterification of the carotenoid analog or derivative in the mammalian gut.
  • FIG. 23 depicts a standard plot of concentration of non-esterified, free astaxanthin verses time for liver after single dose oral gavage in black mice. Only non-esterified, free astaxanthin is detected in liver, also corroborating (see FIG. 22 for plasma) the complete de-esterification of the carotenoid analog or derivative in the mammalian gut, as has been described previously. At every time point, liver levels of non-esterified, free astaxanthin are greater than that observed in plasma, a finding suggesting vastly improved solid-organ delivery of free carotenoid in the novel emulsion vehicle used in this study.
  • FIG. 24 depicts the effect of the disodium disuccinate astaxanthin derivative at 500 mg/kg by oral gavage on lipopolysaccharide (LPS)-induced liver injury in mice (as measured by elevation in serum alanine aminotransferase, or ALT).
  • LPS lipopolysaccharide
  • ALT serum alanine aminotransferase
  • FIG. 25 depicts a graphical representation of a relative reduction of infarct size in male Sprague-Dawley rats with pre-treatment using a disodium salt disuccinate astaxanthin derivative intravenous formulation (CardaxTM).
  • CardaxTM disodium salt disuccinate astaxanthin derivative intravenous formulation
  • FIG. 26 depicts a graphical representation of a relative reduction of infarct size in male Sprague-Dawley rats with pre-treatment using a disodium salt disuccinate astaxanthin derivative intravenous formulation (CardaxTM).
  • CardaxTM disodium salt disuccinate astaxanthin derivative intravenous formulation
  • FIG. 27 depicts transient absorption versus delay for the diacid disuccinate astaxanthin derivative (astaCOOH) using flash photolysis.
  • the experiment was performed in acetonitrile (MeCN) using nitronaftalin (NN) as photosensitizer.
  • the spectra obtained demonstrate that the diacid disuccinate astaxanthin derivative behaves identically to non-esterified, free racemic astaxanthin as a radical quencher (formation of the carotenoid radical cation), identifying the derivative as an active “soft-drug” which generates non-esterified, free astaxanthin in vivo after both oral and intravenous delivery.
  • FIG. 28 depicts transient absorption versus delay for the reference compound non-esterified, free racemic astaxanthin (asta)] using flash photolysis.
  • the experiment was performed in acetonitrile (MeCN) using nitronaftalin (NN) as photosensitizer.
  • the spectra obtained are nearly superimposable on those obtained for the diacid disuccinate astaxanthin derivative (astaCOOH), suggesting identical radical-cation forming properties for both compounds.
  • FIG. 29 depicts a pictorial representation of a Western blot of a polyacrylamide gel with anti-connexin 43 antibody.
  • FIG. 30 depicts a pictorial representation of quantitative densitometric images of Western blots with anti-connexin 43 antibodies followed by HRP chemiluminescence on a Biorad imager.
  • FIG. 31 depicts a graph of relative fold-induction of connexin 43 expression by positive control (TTNPB, potent synthetic retinoid) and test compounds (disodium salt disuccinate astaxanthin derivative in four water and/or ethanol (EtOH)/water formulations: H 2 0-10 ⁇ 5 , H 2 0-10 ⁇ 6 , H 2 0-10 ⁇ 7 , and EtOH/H 2 0-10 ⁇ 5 ) versus sterile water control (H 2 0) at 96 hours post-dosing.
  • TTNPB potent synthetic retinoid
  • test compounds sodium salt disuccinate astaxanthin derivative in four water and/or ethanol (EtOH)/water formulations: H 2 0-10 ⁇ 5 , H 2 0-10 ⁇ 6 , H 2 0-10 ⁇ 7 , and EtOH/H 2 0-10 ⁇ 5
  • H 2 0 sterile water control
  • FIG. 32 depicts a graph of mean levels of non-esterified, free astaxanthin in plasma and liver after eleven (11) days of oral gavage of 500 mg/kg disodium disuccinate astaxanthin derivative (ADD) in emulsion vehicle to black mice. Both peak and trough levels in plasma and liver achieved were >200 nM, considered to be protective against oxidative stress and hepatic injury in vivo. The peak levels obtained in liver at 6 hours post-11 th dose were nearly 9 times the protective levels necessary (1760 nM).
  • ADD disodium disuccinate astaxanthin derivative
  • FIG. 33 depicts the mean percent inhibition of superoxide anion signal as detected by DEPMPO spin trap by the disodium salt disuccinate di-vitamin C derivative [derivative (XXIII)]. As the concentration of the derivative increases, inhibition increases in a dose-dependent manner. At 60 ⁇ M, nearly complete inhibition of superoxide anion signal is seen. This derivative was also highly water soluble, and was introduced into the test assay without a co-solvent (see FIG. 21 ). The novel derivative was comparable in radical-quenching efficacy to the formulation of the disodium salt disuccinate astaxanthin derivative in a 1:2 formulation with vitamin C (see FIG. 3 ), suggesting active, “soft-drug” properties for this derivative. This co-antioxidant derivative strategy increased the relative radical scavenging potency (when compared with the disodium salt disuccinate astaxanthin derivative) by 50-fold.
  • FIG. 34 depicts effects of non-esterified, free astaxanthin (as the all-trans mixture of stereoisomers) on MCA-induced neoplastic transformation in mouse embryonic fibroblast (10T1 ⁇ 2) cells.
  • Non-esterified, free astaxanthin is produced rapidly in vivo after oral and intravenous administration of novel carotenoid derivatives, and is detected in high concentration in both plasma and solid organs (see FIG. 22 and FIG. 23 ).
  • Non-esterified, free astaxanthin demonstrated levels of reduction of neoplastic transformation (100%) above any other carotenoid tested in this assay at similar concentrations, demonstrating the increased utility of this compound for cancer chemoprevention applications.
  • FIG. 35 depicts a comparison of an astaxanthin-treated dish to control dishes (see description for FIG. 34 ).
  • FIG. 36 depicts a comparison of astaxanthin (as the mixture of stereoisomers) to previously tested carotenoids in this laboratory using this assay (see description for FIG. 34 ).
  • FIG. 37 depicts a graphical representation of a relative reduction of infarct size in male New Zealand rabbits with pre-treatment using a disodium salt disuccinate astaxanthin derivative intravenous formulation (CardaxTM).
  • CardaxTM disodium salt disuccinate astaxanthin derivative intravenous formulation
  • FIG. 38 depicts a graphical representation of a relative reduction of circulating levels of plasma C-reactive protein (CRP) in male New Zealand rabbits with pre-treatment using a disodium disuccinate astaxanthin derivative intravenous formulation (CardaxTM).
  • CRP plasma C-reactive protein
  • CardaxTM disodium disuccinate astaxanthin derivative intravenous formulation
  • Parenter carotenoids may generally refer to those natural compounds utilized as starting scaffold for structural carotenoid analog or derivative synthesis.
  • Carotenoid derivatives may be derived from a naturally occurring carotenoid.
  • Naturally occurring carotenoids may include lycopene, lycophyll, lycoxanthin, astaxanthin, betacarotene, lutein, zeaxanthin, and/or canthaxanthin to name a few.
  • Carotenoids are a group of natural pigments produced principally by plants, yeast, and microalgae. The family of related compounds now numbers greater than 700 described members, exclusive of Z and E isomers. Fifty (50) have been found in human sera or tissues. Humans and other animals cannot synthesize carotenoids de novo and must obtain them from their diet. All carotenoids share common chemical features, such as a polyisoprenoid structure, a long polyene chain forming the chromophore, and near symmetry around the central double bond. Tail-to-tail linkage of two C 20 geranylgeranyl diphosphate molecules produces the parent C 40 carbon skeleton.
  • Carotenoids without oxygenated functional groups are called “carotenes”, reflecting their hydrocarbon nature; oxygenated carotenes are known as “xanthophylls.” Cyclization at one or both ends of the molecule yields 7 identified end groups (illustrative structures shown in FIG. 1 ).
  • Carotenoids with chiral centers may exist either as the R (rectus) or S (sinister) configurations.
  • astaxanthin (with 2 chiral centers at the 3 and 3′ carbons) may exist as 3 possible stereoisomers: 3S, 3′S; 3R, 3′S and 3S, 3′R (identical meso forms); or 3R,3′R.
  • the relative proportions of each of the stereoisomers may vary by natural source.
  • Haematococcus pluvialis microalgal meal is 99% 3S,3′S astaxanthin, and is likely the predominant human evolutionary source of astaxanthin.
  • Krill (3R,3′R) and yeast sources yield different stereoisomer compositions than the microalgal source.
  • Synthetic astaxanthin produced by large manufacturers such as Hoffmnann-LaRoche AG, Buckton Scott (USA), or BASF AG, are provided as defined geometric isomer mixtures of a 1:2:1 stereoisomer mixture [3S, 3′S; 3R, 3′S, (meso); 3R, 3′R] of non-esterified, free astaxanthin.
  • Natural source astaxanthin from salmonid fish is predominantly a single stereoisomer (3S,3′S), but does contain a mixture of geometric isomers. Astaxanthin from the natural source Haematococcus pluvialis may contain nearly 50% Z isomers.
  • the Z conformational change may lead to a higher steric interference between the two parts of the carotenoid molecule, rendering it less stable, more reactive, and more susceptible to reactivity at low oxygen tensions.
  • the Z forms in relation to the all-E form, the Z forms: (1) may be degraded first; (2) may better suppress the attack of cells by reactive oxygen species such as superoxide anion; and (3) may preferentially slow the formation of radicals. Overall, the Z forms may initially be thermodynamically favored to protect the lipophilic portions of the cell and the cell membrane from destruction.
  • the all-E form of astaxanthin unlike ⁇ -carotene, retains significant oral bioavailability as well as antioxidant capacity in the form of its dihydroxy- and diketo-substitutions on the ⁇ -ionone rings, and has been demonstrated to have increased efficacy over ⁇ -carotene in most studies.
  • the all-E form of astaxanthin has also been postulated to have the most membrane-stabilizing effect on cells in vivo. Therefore, it is likely that the all-E form of astaxanthin in natural and synthetic mixtures of stereoisomers is also extremely important in antioxidant mechanisms, and may be the form most suitable for particular pharmaceutical preparations.
  • the antioxidant mechanism(s) of carotenoids includes singlet oxygen quenching, direct radical scavenging, and lipid peroxidation chain-breaking.
  • the polyene chain of the carotenoid absorbs the excited energy of singlet oxygen, effectively stabilizing the energy transfer by delocalization along the chain, and dissipates the energy to the local environment as heat. Transfer of energy from triplet-state chlorophyll (in plants) or other porphyrins and proto-porphyrins (in mammals) to carotenoids occurs much more readily than the alternative energy transfer to oxygen to form the highly reactive and destructive singlet oxygen ( 1 O 2 ).
  • Carotenoids may also accept the excitation energy from singlet oxygen if any should be formed in situ, and again dissipate the energy as heat to the local environment. This singlet oxygen quenching ability has significant implications in cardiac ischemia, macular degeneration, porphyria, and other disease states in which production of singlet oxygen has damaging effects. In the physical quenching mechanism, the carotenoid molecule may be regenerated (most frequently), or be lost. Carotenoids are also excellent chain-breaking antioxidants, a mechanism important in inhibiting the peroxidation of lipids. Astaxanthin can donate a hydrogen (H) to the unstable polyunsaturated fatty acid (PUFA) radical, stopping the chain reaction.
  • H hydrogen
  • PUFA unstable polyunsaturated fatty acid
  • Peroxyl radicals may also, by addition to the polyene chain of carotenoids, be the proximate cause for lipid peroxide chain termination.
  • the appropriate dose of astaxanthin has been shown to completely suppress the peroxyl radical chain reaction in liposome systems. Astaxanthin shares with vitamin E this dual antioxidant defense system of singlet oxygen quenching and direct radical scavenging, and in most instances (and particularly at low oxygen tension in vivo) is superior to vitamin E as a radical scavenger and physical quencher of singlet oxygen.
  • Carotenoids and in particular astaxanthin, are potent direct radical scavengers and singlet oxygen quenchers and possess all the desirable qualities of such therapeutic agents for inhibition or amelioration of ischemia-reperfusion injury.
  • Synthesis of novel carotenoid derivatives with “soft-drug” properties i.e. active as antioxidants in the derivatized form), with physiologically relevant, cleavable linkages to pro-moieties, can generate significant levels of free carotenoids in both plasma and solid organs.
  • this is a particularly useful embodiment (characteristics specific to non-esterified, free astaxanthin below):
  • antioxidants which are potent singlet oxygen quenchers and direct radical scavengers, particularly of superoxide anion, should limit hepatic fibrosis and the progression to cirrhosis by affecting the activation of hepatic stellate cells early in the fibrogenetic pathway. Reduction in the level of ROS by the administration of a potent antioxidant can therefore be crucial in the prevention of the activation of both HSC and Kupffer cells.
  • This protective antioxidant effect appears to be spread across the range of potential therapeutic antioxidants, including water-soluble (e.g., vitamin C, glutathione, resveratrol) and lipophilic (e.g., vitamin E, ⁇ -carotene, astaxanthin) agents. Therefore, a co-antioxidant derivative strategy in which water-soluble and lipophilic agents are combined synthetically is a particularly useful embodiment.
  • Vitamin E is generally considered the reference antioxidant.
  • carotenoids are more efficient in quenching singlet oxygen in homogenenous organic solvents and in liposome systems. They are better chain-breaking antioxidants as well in liposomal systems. They have demonstrated increased efficacy and potency in vivo. They are particularly effective at low oxygen tension, and in low concentration, making them extremely effective agents in disease conditions in which ischemia is an important part of the tissue injury and pathology.
  • These carotenoids also have a natural tropism for the heart and liver after oral administration. Therefore, therapeutic administration of carotenoids should provide a greater benefit in limiting fibrosis than vitamin E.
  • the parent carotenoid may have a structure of any naturally occurring carotenoid.
  • Some examples of naturally occurring carotenoids that may be used as parent compounds are shown in FIG. 1 .
  • Aaptopurpurin Actinioerythrin; Actinioerythrol; Adonirubin; Adonixanthin; A.g.470; A.g.471; Agelaxanthin C; Aleuriaxanthin; Alloxanthin; Amarouciaxanthin A; Amarouciaxanthin B; Anchovyxanthin; 3′,4′-Anhydrodiatoxanthin; Anhydrodeoxyflexixanthin; Anhydroeschscholtzxanthin; Anhydrolutein; Anhydroperidinin; Anhydrorhodovibrin; Anhydrosaproxanthin; Anhydrowarmingol; Anhydrowarmingone; Antheraxanthin; Aphanicin; Aphanicol; Aphanin; Aphanol; Aphanizophyll; 8′-Apo- ⁇ -caroten-8′-al; 10′-Apo- ⁇ -caroten-10′-al; 12′-Apo-p-caroten
  • Decahydro- ⁇ -carotene 1,2,7,8,11,12,7′,8′,11′,12′-Decahydro- ⁇ , ⁇ -carotene; 7,8,11,12,15,7′,8′,11′,12′,15′Decahydro- ⁇ , ⁇ -carotene; 1,2,7,8,11,12,7′,8′,11′,12′-Decahydro- ⁇ , ⁇ -caroten-1-ol; Decahydrolycopene; Decaprenoxanthin; Decaprenoxanthin diglucoside; Decaprenoxanthin monoglucoside; Deepoxyneoxanthin; Dehydro—see also Bisdehydro-, Didehydro-, MonodehydroDehydroadonirubin; Dehydroadonixanthin; Dehydrocarotene II; Dehydrocarotene III; Dehydro- ⁇ -carotene; 3,4-Dehydro- ⁇ -carotene; 3′,4
  • Flavacin Flavochrome; Flavorhodin; Flavoxanthin; Flexixanthin; Foliachrome; Foliaxanthin; Fritschiellaxanthin; Fucochrome; Fucoxanthin; Fucoxanthinol; Fucoxanthol;
  • Gazaniaxanthin ⁇ ,D-Gentiobiosyl ⁇ ,D-glucosyl 8,8′-diapocarotene-8,8′-dioate; Gentiobiosyl hydrogen-8,8′-dioate; Gentiobiosyl neapolitanosyl 8,8′-diapocarotene-8,8′-dioate; ⁇ ,D-Glucosyl hydrogen-4,4′-diapocarotene-4,4′-dioate; 4′- ⁇ ,D-Glucosyl 4-hydrogen-7′,8′-dihydro-4,4′-diapocarotene-4,4′-dioate; ⁇ ,D-Glucosyl hydrogen-8,8′-diapocarotene-8,8′-dioate; ⁇ ,D-Glucosyl methyl-8,8′-diapocarotene-8,8′-dioate; Glucopyranosyloxy (see Glucosy
  • Idoxanthin Isoagelaxanthin A; Isobixin; Isocarotene; Iso- ⁇ -carotene; Iso- ⁇ -carotene; Isocrocetin; Isocryptoxanthin; Isofucoxanthin; Isofucoxanthinol; Isolutein; Isomethylbixin; Isomytiloxanthin; 2-Isopentenyl-3,4-dehydrorhodopin; Isorenieratene; ⁇ -Isorenieratene; 3,3′-Isorenieratenediol; 3-Isorenieratenol; Isotedaniaxanthin; Isotedanin; Isozeaxanthin;
  • Keto- see also oxo or -one Ketocapsanthin; 4-Ketocapsanthin; 4-Keto- ⁇ -carotene; 4-Keto- ⁇ -carotene; 4-Keto- ⁇ -carotene; 4-Ketocynthiaxanthin; 4-Keto-3,4′-dehydro- ⁇ -carotene; 4-Keto-1′,2′-dihydro-1′-hydroxytorulene; 2-Keto-7′,8′-dihydrorhodovibrin; 4-Keto-3,3′-dihydroxy- ⁇ -carotene; 4′-Keto-3-hydroxy- ⁇ -carotene; 4-Keto-3′-hydroxylycopene; 4-Ketolutein 332 4-Ketomyxol 2′-(methylpentoside); 4-Ketomyxoxanthophyll; 2-Keto-OH-spirilloxanthin; 4-K
  • Neocarotene Neochrome; Neo- ⁇ -carotene B; Neo- ⁇ -cryptoxanthin A; Neoxanthin; Neoxanthin 3-acetate; Neurosporaxanthin; Neurosporaxanthin methyl ester; Neurosporene; Nonaprenoxanthin; 2′-Nor-astaxanthin diester; Norbixin; Nostoxanthin;
  • Vaucheriaxanthin Violaxanthin; Violeoxanthin; Violerythrin;
  • ⁇ -Zeacarotene ⁇ -Zeacarotene; ⁇ -Zeacarotene; ⁇ 1 -Zeacarotene; ⁇ -Zeacarotene-3,17′-diol; ⁇ -Zeacarotene-3,17′-diol; ⁇ -Zeacaroten-3-ol; Zeaxanthene; Zeaxanthin; Zeaxanthin diepoxide; Zeaxanthin dimethyl ether; Zeaxanthin dirhamnoside; Zeaxanthin dipalmitate; Zeaxanthin 5,6-epoxide; Zeaxanthin 5,8-epoxide; Zeaxanthin furanoxide; Zeaxanthin monomethyl ether; Zeaxanthin monorhamnoside; Zeaxanthol; and Zeinoxanthin.
  • the above list of naturally occurring carotenoids is meant to a be a non-limiting example of naturally occurring carotenoids.
  • the total synthesis of naturally occurring as well as synthetic carotenoids as starting scaffolds for carotenoid analogs or derivatives may be a method of generation of said carotenoid analogs or derivatives.
  • the carotenoid derivatives may include compounds having a structure including a polyene chain (i.e., backbone of the molecule).
  • the polyene chain may include between about 5 and about 15 unsaturated bonds.
  • the polyene chain may include between about 7 and about 12 unsaturated bonds.
  • a carotenoid derivative may include 7 or more conjugated double bonds to achieve acceptable antioxidant properties.
  • decreased antioxidant properties associated with shorter polyene chains may be overcome by increasing the dosage administered to a subject or patient.
  • a chemical compound including a carotenoid derivative may have the general structure (I): Each R 3 may be independently hydrogen or methyl. R 1 and R 2 may be independently H, an acyclic alkene with one or more substituents, or a cyclic ring including one or more substituents. y may be 5 to 12. In some embodiments, y may be about 3 to about 15. In certain embodiments, the maximum value of y may only be limited by the ultimate size of the chemical compound, particularly as it relates to the size of the chemical compound and the potential interference with the chemical compound′s biological availability as discussed herein. In some embodiments, substituents may be at least partially hydrophilic. These carotenoid derivatives may be used in a pharmaceutical composition.
  • the carotenoid derivatives may include compounds having the structure (Ia): Each R 3 may be independently hydrogen, methyl, alkyl, alkenyl, or aromatic substituents. R 1 and R 2 may be independently H, an acyclic alkene with at least one substituent, or a cyclic ring with at least one substituent having general structure (II): where n may be between 4 to 10 carbon atoms. W is the substituent.
  • each cyclic ring may be independently two or more rings fused together to form a fused ring system (e.g., a bycyclic system).
  • a fused ring system e.g., a bycyclic system
  • Each ring of the fused ring system may independently contain one or more degrees of unsaturation.
  • Each ring of the fused ring system may be independently aromatic. Two or more of the rings forming the fused ring system may form an aromatic system.
  • a chemical compound including a carotenoid derivative may have the general structure (Ib): Each R 3 may be independently hydrogen or methyl. Each Y may be independently O or H 2 . Each R may be independently OR 1 or R 1 Each R 1 ay be independently -alkyl-NR 2 3 + , -aromatic-NR 2 3 + , -alkyl-CO 2 ⁇ , -aromatic-CO 2 , -amino acid-NH 3 + , -phosphorylated amino acid-NH 3 + , polyethylene glycol, dextran, H, alkyl, or aryl. Each R 2 may be independently H, alkyl, or aryl. z may be 5 to 12.
  • z may be about 3 to about 15. In certain embodiments, the maximum value of z may only be limited by the ultimate size of the chemical compound, particularly as it relates to the size of the chemical compound and the potential interference with the chemical compound′s biological availability as discussed herein.
  • substituents may be at least partially hydrophilic. These carotenoid derivatives may be used in a pharmaceutical composition.
  • a chemical compound including a carotenoid derivative may have the general structure (Ic): Each R 3 may be independently hydrogen or methyl. Each Y may be independently O or H 2 . Each X is independently -alkyl-NR 1 3 + , -aromatic-NR 1 3 + , -alkyl-CO 2 ⁇ , -aromatic-CO 2 ⁇ , -amino acid-NH 3 + , -phosphorylated amino acid-NH 3 + , polyethylene glycol, dextran, alkyl, or aryl.
  • Each R 1 is independently -alkyl-NR 2 3 + , -aromatic-NR 2 3 + , -alkyl-CO 2 ⁇ , -aromatic-CO 2 ⁇ , -amino acid-NH 3 + , -phosphorylated amino acid-NH 3 + , polyethylene glycol, dextran, H, alkyl, aryl, or alkali salt.
  • Each R 2 may be independently H, alkyl, or aryl.
  • z may be 5 to 12. In some embodiments, z may be about 3 to about 15.
  • the maximum value of z may only be limited by the ultimate size of the chemical compound, particularly as it relates to the size of the chemical compound and the potential interference with the chemical compound's biological availability as discussed herein.
  • substituents may be at least partially hydrophilic. These carotenoid derivatives may be used in a pharmaceutical composition.
  • five- and/or six-membered ring carotenoid derivatives may be more easily synthesized. Synthesis may come more easily due to, for example, the natural stability of five- and six-membered rings. Synthesis of carotenoid derivatives including five- and/or six-membered rings may be more easily synthesized due to, for example, the availability of naturally occurring carotenoids including five- and/or six-membered rings. In some embodiments, five-membered rings may decrease steric hindrance associated with rotation of the cyclic around the molecular bond connecting the cyclic ring to the polyene chain.
  • Reducing steric hindrance may allow greater overlap of any ⁇ oribitals within a cyclic with the polyene chain, thereby increasing the degree of conjugation and effective chromophore length of the molecule. This may have the salutatory effect of increasing antioxidant capacity of the carotenoid derivatives.
  • a substituent (W) may be at least partially hydrophilic.
  • a hydrophilic substituent may assist in increasing the water solubility of a carotenoid derivative.
  • a carotenoid derivative may be at least partially water soluble.
  • the cyclic ring may include at least one chiral center.
  • the acyclic alkene may include at least one chiral center.
  • the cyclic ring may include at least one degree of unsaturation.
  • the cyclic ring may be aromatic. One or more degrees of unsaturation within the ring may assist in extending the conjugation of the carotenoid derivative.
  • the cyclic ring may include a substituent.
  • the substituent may be hydrophilic.
  • the cyclic ring may include, for example (a), (b), or (c):
  • the substituent may include, for example, a carboxylic acid, an amino acid, an ester, an alkanol, an amine, a phosphate, a succinate, a glycinate, an ether, a glucoside, a sugar, or a carboxylate salt.
  • each substituent —W may independently include -XR.
  • Each X may independently include O, N, or S.
  • each substituent -W may independently comprises amino acids, esters, carbamates, amides, carbonates, alcohol, phosphates, or sulfonates.
  • the substituent may include, for example (d) through (rr): where each R is, for example, independently -alkyl-NR 1 3 + , -aromatic-NR 1 3 + , -alkyl-CO 2 ⁇ , -aromatic-CO 2 ⁇ , -amino acid-NH 3 + , -phosphorylated amino acid-NH 3 + , polyethylene glycol, dextran, H, alkyl, or aryl.
  • substituents may include any combination of (d) through (rr).
  • negatively charged substituents may include alkali metals, one metal or a combination of different alkali metals in an embodiment with more than one negatively charged substituent, as counter ions.
  • Alkali metals may include, but are not limited to, sodium, potassium, and/or lithium.
  • Water soluble carotenoid analogs or derivatives may have a water solubility of greater than about 1 mg/mL in some embodiments. In certain embodiments, water soluble carotenoid analogs or derivatives may have a water solubility of greater than about 10 mg/mL. In some embodiments, water soluble carotenoid analogs or derivatives may have a water solubility of greater than about 50 mg/mL.
  • carotenoid derivative in 3 dimensions is important when considering its use in biological and/or medicinal applications. Some of the largest naturally occurring carotenoids are no greater than about C 50 . This is probably due to size limits imposed on molecules requiring incorporation into and/or interaction with cellular membranes. Cellular membranes may be particularly co-evolved with molecules of a length of approximately 30 nm. In some embodiments, carotenoid derivatives may be greater than or less than about 30 nm in size. In certain embodiments, carotenoid derivatives may be able to change conformation and/or otherwise assume an appropriate shape which effectively enables the carotenoid derivative to efficiently interact with a cellular membrane.
  • alkenes in the E configuration this should not be seen as limiting.
  • Compounds discussed herein may include embodiments where alkenes are in the Z configuration or include alkenes in a combination of Z and E configurations within the same molecule.
  • the compounds depicted herein may naturally convert between the Z and E configuration and/or exist in equilibrium between the two configurations.
  • a chemical compound may include a carotenoid derivative having the structure (III)
  • Each Y may be independently O or H 2 .
  • Each R may be independently OR 1 or R 1 .
  • Each R 1 may be independently -alkyl-NR 2 3 + , -aromatic-NR 2 3 + , -alkyl-CO 2 ⁇ , -aromatic-CO 2 ⁇ , -amino acid-NH 3 + , -phosphorylated amino acid-NH 3 + , polyethylene glycol, dextran, H, alkyl, peptides, poly-lysine or aryl.
  • each R 2 may be independently H, alkyl, or aryl.
  • the carotenoid derivative may include at least one chiral center.
  • the carotenoid derivative has the structure (IV)
  • the carotenoid derivative has the structure (V)
  • a chemical compound may include a carotenoid derivative having the structure (VI)
  • Each Y may be independently O or H 2 .
  • Each R may be independently H, alkyl, or aryl.
  • the carotenoid derivative may include at least one chiral center.
  • Y may be H 2 , the carotenoid derivative having the structure (VII)
  • the carotenoid derivative has the structure (VIII)
  • a chemical compound may include a carotenoid derivative having the structure (IX)
  • Each Y may be independently O or H 2 .
  • Each R 1 may be CH 2 .
  • n may be 1 to 9.
  • Each X may be independently
  • Each R may be independently -alkyl-NR 1 3 ⁇ , -aromatic-NR 1 3 + , -alkyl-CO 2 ⁇ , -aromatic-CO 2 ⁇ , -amino acid-NH 3 + , -phosphorylated amino acid-NH 3 + , polyethylene glycol, dextran, H, alkyl, or aryl.
  • Each R 1 may be independently H, alkyl, or aryl.
  • the carotenoid derivative may include at least one chiral center.
  • the carotenoid derivative has the structure (X) In a specific embodiment where Y is H 2 , the carotenoid derivative has the structure (X) In a specific embodiment where Y is O, the carotenoid derivative has the structure (XI)
  • a chemical compound may include a carotenoid derivative having the structure (XII) Each Y may be independently O or H 2 .
  • the carotenoid derivative may include at least one chiral center.
  • Y may be H 2 , the carotenoid derivative having the structure (XIII)
  • the carotenoid derivative has the structure (XIV)
  • a chemical compound may include a disuccinic acid ester carotenoid derivative having the structure (XV)
  • a chemical compound may include a disodium salt disuccinic acid ester carotenoid derivative having the structure (XVI)
  • a chemical compound may include a carotenoid derivative with a co-antioxidant, in particular one or more analogs or derivatives of vitamin C (i.e., L ascorbic acid) coupled to a carotenoid.
  • vitamin C i.e., L ascorbic acid
  • Some embodiments may include carboxylic acid and/or carboxylate derivatives of vitamin C coupled to a carotenoid (e.g., structure (XVII)) Carbohydr.
  • Res. 1978, 60, 251-258 herein incorporated by reference, discloses oxidation at C-6 of ascorbic acid as depicted in EQN. 5.
  • Some embodiments may include vitamin C and/or vitamin C analogs or derivatives coupled to a carotenoid.
  • Vitamin C may be coupled to the carotenoid via an ether linkage (e.g., structure (XVIII))
  • Some embodiments may include vitamin C disuccinate analogs or derivatives coupled to a carotenoid (e.g., structure (XIX))
  • Some embodiments may include solutions or pharmaceutical preparations of carotenoids and/or carotenoid derivatives combined with co-antioxidants, in particular vitamin C and/or vitamin C analogs or derivatives.
  • Pharmaceutical preparations may include about a 2:1 ratio of vitamin C to carotenoid respectively.
  • co-antioxidants may increase solubility of the chemical compound.
  • co-antioxidants e.g., vitamin C
  • co-antioxidants may decrease toxicity associated with at least some carotenoid analogs or derivatives.
  • co-antioxidants e.g., vitamin C
  • co-antioxidants may increase the potency of the chemical compound synergistically.
  • Co-antioxidants may be coupled to a carotenoid derivative.
  • Co-antioxidants may coupled (e.g., a covalent bond) to the carotenoid derivative.
  • Co-antioxidants may be included as a part of a pharmaceutically acceptable formulation.
  • a carotenoid e.g., astaxanthin
  • vitamin C may be coupled to vitamin C forming an ether linkage.
  • the ether linkage may be formed using the Mitsunobu reaction as in EQN. 1.
  • vitamin C may be selectively esterified.
  • Vitamin C may be selectively esterified at the C-3 position (e.g., EQN. 2).
  • a carotenoid may be coupled to vitamin C.
  • Vitamin C may be coupled to the carotenoid at the C-6, C-5 diol position as depicted in EQNS. 3 and 4 forming an acetal.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a glyoxylate linker as depicted in EQN. 6.
  • a water soluble moiety e.g., vitamin C
  • a glyoxylate linker as depicted in EQN. 6.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a glyoxylate linker as depicted in EQN. 7.
  • a water soluble moiety e.g., vitamin C
  • a glyoxylate linker as depicted in EQN. 7.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a phosphate linker as depicted in EQN. 8.
  • a water soluble moiety e.g., vitamin C
  • a phosphate linker as depicted in EQN. 8.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a phosphate linker as depicted in EQN. 9.
  • a water soluble moiety e.g., vitamin C
  • a phosphate linker as depicted in EQN. 9.
  • Carbohydr. Res. 1988, 176, 73-78, herein incorporated by reference discloses the 6-bromo derivative of vitamin C's reaction with phosphates.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a phosphate linker as depicted in EQN. 10.
  • a water soluble moiety e.g., vitamin C
  • a phosphate linker as depicted in EQN. 10.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a phosphate linker as depicted in EQN. 11.
  • Vitamin C may be coupled to the carotenoid using selective esterification at C-3 of unprotected ascorbic acid with primary alcohols.
  • a carotenoid may be coupled to a water soluble moiety (e.g., vitamin C) with a phosphate linker as in LXVII.
  • Structure LXVII may include one or more counterions (e.g., alkali metals).
  • EQN. 12 depicts an example of a synthesis of a protected form of LXVII.
  • a chemical compound may include a carotenoid derivative including one or more amino acids (e.g., lysine) and/or amino acid analogs or derivatives (e.g., lysine hydrochloric acid salt) coupled to a carotenoid [e.g., structure (XX)].
  • a carotenoid derivative including one or more amino acids (e.g., lysine) and/or amino acid analogs or derivatives (e.g., lysine hydrochloric acid salt) coupled to a carotenoid [e.g., structure (XX)].
  • a carotenoid analog or derivative may include:
  • the carotenoid derivatives may be synthesized from naturally occurring carotenoids.
  • the carotenoids may include structures 2A-2E depicted in FIG. 1 .
  • the carotenoid derivatives may be synthesized from a naturally occurring carotenoid including one or more alcohol substituents.
  • the carotenoid derivatives may be synthesized from a derivative of a naturally occurring carotenoid including one or more alcohol substituents.
  • the synthesis may result in a single stereoisomer.
  • the synthesis may result in a single geometric isomer of the carotenoid derivative.
  • the synthesis/synthetic sequence may include any prior purification or isolation steps carried out on the parent carotenoid.
  • the synthesis may be a total synthesis.
  • An example may include, but is not limited to, a 3S,3′S all-E carotenoid derivative, where the parent carotenoid is astaxanthin.
  • the synthetic sequence may include protecting and subsequently deprotecting various functionalities of the carotenoid and/or substituent precursor.
  • the alcohols may be deprotonated with a base.
  • the deprotonated alcohol may be reacted with a substituent precursor with a good leaving group.
  • the base may include any non-nucleophilic base known to one skilled in the art such as, for example, dimethylaminopyridine (DMAP).
  • DMAP dimethylaminopyridine
  • the deprotonated alcohol may act as a nucleophile reacting with the substituent precursor, displacing the leaving group.
  • Leaving goups may include, but are not limited to, Cl, Br, tosyl, brosyl, mesyl, or trifyl. These are only a few examples of leaving groups that may be used, many more are known and would be apparent to one skilled in the art. In some embodiments, it may not even be necessary to deprotonate the alcohol, depending on the leaving group employed. In other examples the leaving group may be internal and may subsequently be included in the final structure of the carotenoid derivative, a non-limiting example may include anhydrides or strained cyclic ethers. For example, the deprotonated alcohol may be reacted with succinic anhydride.
  • the disuccinic acid ester of astaxanthin may be further converted to the disodium salt.
  • Examples of synthetic sequences for the preparation of some of the specific embodiments depicted are described in the Examples section.
  • the example depicted below is a generic non-limiting example of a synthetic sequence for the preparation of carotenoid derivatives.
  • Reperfusion of ischemic myocardium results in significant cellular and local alterations in at-risk tissue which exacerbate damage created by the ischemic insult. Specifically, vascular and microvascular injury, endothelial dysfunction, accelerated cellular necrosis, and granulocyte activation occur subsequent to ischemia-reperfusion.
  • vascular and microvascular injury results from complement activation, the interaction of circulating and localized C-reactive protein with C1q and phosphocholine on exposed cells forming the membrane attack complex (MAC) with ensuing cell death and increased endothelial permeability, superoxide anion (O 2 —) generation by affected endothelium and activated leukocytes, microemboli, cytokine release (in particular IL-6), and activation of platelets with IIbIIIa receptor activation, and subsequent release of ADP and serotonin.
  • Endothelial dysfunction follows, with subsequent generation of superoxide anion by the dysfunctional endothelium, further damaging the affected endothelium in a positive feedback cycle.
  • ischemia-reperfusion results in early and severe injury to the vasculature, which further compromises myocyte survival.
  • Granulocyte activation also occurs during ischemia-reperfusion.
  • MPO myeloperoxidase
  • elastases elastases
  • proteases elastases
  • oxygen-derived radical and non-radical species most importantly superoxide anion, hypochlorite, singlet oxygen, and hydrogen peroxide after the “respiratory burst”.
  • Oxygen-derived radical and non-radical e.g.
  • TBARS thiobarbituric acid reactive substances
  • MDA malondialdehyde
  • ROS reactive oxygen species
  • the (O)-form using molecular oxygen as the electron acceptor, produces the superoxide anion O 2 — in the coronary endothelium.
  • Superoxide anion is then available to create additional tissue damage in the local environment.
  • the superoxide anion is not the most reactive or destructive radical species in biological systems on its own. However, it is the source of some shorter- and longer-lived, more damaging radicals and/or ROS such as the hydroxyl radical, hydrogen peroxide, singlet oxygen, and peroxyl radicals (e.g. peroxynitrite). As such, it can be considered the “lynchpin” radical in I/R injury.
  • the biological reactions of the superoxide radical to form these important oxidants are shown below:
  • (1) superoxide anion may accept a single electron (“monovalent reduction”), producing peroxide (O 2 ⁇ 2 ). Coupled with 2 protons, peroxide then forms hydrogen peroxide (H 2 O 2 ). H 2 O 2 diffuses easily through cell membranes and cannot readily be excluded from the cytoplasm, where it may react with cellular components or activate central inflammatory cascades such as nuclear factor kappa-B (NF-kappa-B), which are also implicated in the additional inflammatory damage in I/R injury.
  • NF-kappa-B nuclear factor kappa-B
  • superoxide anion typically reacts with itself to produce hydrogen peroxide and oxygen (“dismutation”).
  • superoxide dismutation may be spontaneous, or catalyzed by the enzyme superoxide dismutase (SOD), a reaction which results in the formation of oxidized SOD: 2O 2 ⁇ +2H + ⁇ H 2 O 2 + 3 O 2
  • (3) superoxide anion may serve as a reducing agent and donate a single electron (“monovalent reduction”) to a metal cation.
  • ferric iron (Fe 3+ ) is reduced and subsequently acts as a catalyst to convert hydrogen peroxide (H 2 O 2 ) into the hydroxyl radical (HO.).
  • Ferrous iron (Fe 2+ ) the reduced metal cation, subsequently catalyzes the breaking of the oxygen-oxygen bond of hydrogen peroxide. This produces one hydroxyl radical (HO′) and one hydroxide ion (HO ⁇ ).
  • the reaction is known as the Fenton reaction, particularly important in ischemia-reperfusion injury where iron and/or copper compartmentalization has been lost (typically through hemolysis of red blood cells, RBCs): Fe 2+ +H 2 O 2 ⁇ Fe 3+ +HO.+HO ⁇ (step 2)
  • Hydroxyl radicals readily cross cellular membranes. Hydroxyl radical damage is “diffusion rate-limited”, that is, the 3-dimensional distance in which damage may be inflicted is related to the radical's rate of diffusion.
  • the hydroxyl radical is a particularly toxic ROS. Hydroxyl radicals may add to organic substrates (represented by R in the reaction below) and form a hydroxylated adduct which is itself a radical.
  • PUFAs polyunsaturated fatty acids
  • endothelial and myocyte membranes are particularly susceptible to hydroxyl radical damage: HO.+R ⁇ HOR. (hydroxylated adduct)
  • the adduct formed above may further oxidize in the presence of metal cations or molecular oxygen. This results in oxidized, stable product(s). In the first case, the extra electron is transferred to the metal ion, and in the second case, to oxygen (forming superoxide). Two adduct radicals may also react with each other forming oxidized, stable, and crosslinked products plus water. This is an important process in the oxidation of membrane proteins: HOR.+HOR. ⁇ R—R+2H 2 O
  • hydroxyl radicals may oxidize organic substrates by abstracting electrons from such molecules: HO.+R ⁇ R.+OH ⁇
  • the oxidized substrate (R.) is a radical. Such radicals may react with other molecules in a chain reaction. Carotenoids are particularly efficient lipid-peroxidation chain breakers. In one instance, the reaction with ground-state oxygen produces peroxyl radicals (ROO.): R.+ 3 O 2 ⁇ ROO. Peroxyl radicals are very reactive. They may react with other organic substrates in a chain reaction: ROO.+RH ⁇ ROOH+R.
  • Chain reactions are common in the oxidative damage of PUFAs and other susceptible membrane lipids. Measurement of the rate of oxygen consumption is one indication of the initiation and progress of the chain reaction. It is important to note that, in liposomal model systems, non-esterified, free astaxanthin at the appropriate dose is capable of complete suppression of the chain reaction and accompanying oxygen consumption.
  • superoxide anion may react with the hydroxyl radical (HO.) to form singlet oxygen ( 1 O 2 *).
  • Singlet oxygen is not a radical, but is highly reactive and damaging in cardiac biological systems. Singlet oxygen has been implicated in the destruction of membrane-bound proteins such as 5′-nucleotidase, important in the maintenance or restoration of local concentrations of vasodilatory compounds such as adenosine (shown to be effective in humans for reduction of infarct size): O 2 ⁇ +HO. ⁇ 1 O 2 *+HO ⁇
  • superoxide anion may also react with the radical nitric oxide (NO.), producing peroxynitrite (OONO ⁇ ).
  • NO. radical nitric oxide
  • OONO ⁇ peroxynitrite
  • Peroxynitrite is a highly reactive and damaging molecule in biological systems. O 2 ⁇ +NO. ⁇ OONO ⁇
  • PMNs Polymorphonuclear leukocytes
  • neutrophils neutrophils
  • activated macrophages are a rich source of oxygen-derived radical and non-radical species.
  • the NADPH-oxidase system located in phagocyte cell membranes is an important source of radicals following stimulation.
  • the PMNs and activated macrophages rapidly consume oxygen in the “respiratory burst” and convert it to superoxide anion and subsequently hydrogen peroxide (H 2 O 2 ), as well as significant amounts of singlet oxygen.
  • PMNs are additionally a source of hypochlorite, another damaging reactive oxygen species. While important in phagocytic cell activity in infection, in the local environment during ischemia and reperfusion, further cellular injury occurs as these ROS attack normal and damaged host cells in the local area.
  • Neutrophils are a primary source of oxygen radicals during ischemia-reperfusion after prolonged myocardial ischemia, particularly in animal models of experimental infarction. Many prior studies have documented oxygen radical formation during ischemia-reperfusion, but few addressed the source(s) of such radicals in vivo, or had examined radical generation in the context of prolonged myocardial ischemia. Neutrophils are recruited in large amounts within the previously ischemic tissue and are thought to induce injury by local release of various mediators, chiefly oxygen radicals. Previously, the contribution of activated neutrophils to ischemia-reperfusion injury and potential myocardial salvage remained unclear. A methodology was developed to detect radicals, in particular superoxide anion, without interfering with the blood-borne mechanisms of radical generation.
  • Ischemia causes depletion of ATP in cells in the affected area.
  • At the level of the mitochondrial electron transport chain which normally “leaks” approximately 5% of the processed electrons in healthy tissue, further leakage of partially-reduced oxygen species (in particular O 2 —) is favored when the respiratory chain becomes largely reduced. This happens primarily during ischemia.
  • the net effect in the local cellular environment is a tip in the balance of the redox status from anti-oxidant to pro-oxidant, which is at the same time less capable of absorbing additional radical insult(s) without further cellular damage.
  • the following compounds have been evaluated, either in animal models or in limited human trials, as therapeutic agents for the reduction of ischemia-reperfusion injury and/or myocardial salvage during acute myocardial infarction (AMI). Most are biological antioxidants.
  • the plasma half-lives of carotenoids administered orally range from approximately 21 hours for the xanthophylls (“oxygenated” carotenoids including astaxanthin, capsanthin, lutein, and zeaxanthin) to 222 hours for carotenes (“hydrocarbon” carotenoids such as lycopene).
  • oxygenated carotenoids including astaxanthin, capsanthin, lutein, and zeaxanthin
  • carotenes hydrocarbon carotenoids such as lycopene
  • Astaxanthin as a xanthophyll carotenoid, is highly lipid soluble in natural form. It is also small in size (597 Da). Therefore, an injectable astaxanthin structural analog or derivative has a low likelihood of ilnmunogenicity in the right formulation, and is a particularly desirable compound for the current therapeutic indication.
  • Carotenoids have been evaluated, mostly in animal models, for their possible therapeutic value in the prevention and treatment of cancer.
  • the antioxidant properties of carotenoids were the focus of studies directed towards carotenoids and their use in cancer prevention.
  • Studies conducted by Bertram et al. (1991) pointed towards the fact that although carotenoids were antioxidants, this particular property did not appear to be the major factor responsible for their activity as cancer chemopreventive agents. It was, however, discovered that the activity of carotenoids was strongly correlated with their ability to upregulate gap junctional communication. It has been postulated that gap junctions serve as conduits for antiproliferative signals generated by growth-inhibited normal cels.
  • Connexin 43 which is capable of being induced by carotenoids, is the most widely expressed connexin in human tissues. Upregulation of connexin 43, therefore, may be the mechanism by which carotenoids are useful in the chemoprevention of cancer in humans and other animals. And recently, a human study by Nishino et al. (2003) demonstrated that a cocktail of carotenoids (10 mg lycopene, 5 mg each of ⁇ - and ⁇ -carotene) given by chronic oral administration was efficacious in the chemoprevention of hepatocellular carcinoma in high-risk cirrhotic patients in Japan. It is likely, then, that more potent cancer-chemopreventive carotenoids (such as astaxanthin), which accumulate more dramatically in liver, will be particularly useful embodiments.
  • cancer-chemopreventive carotenoids such as astaxanthin
  • the terms “inhibiting” and “ameliorating” are generally defined as the prevention and/or reduction of the negative consequences of a disease state.
  • the methods and compositions described herein may have value as both an acute and a chronic (prophylactic) modality.
  • ischemia-reperfusion injury is generally defined as the pathology attributed to reoxygenation of previously ischemic tissue (either chronically or acutely ischemic), which includes atherosclerotic and thromboembolic vascular disease and its related illnesses.
  • ischemic tissue either chronically or acutely ischemic
  • major diseases or processes including myocardial infarction, stroke, peripheral vascular disease, venous or arterial occlusion and/or restenosis, organ transplantation, coronary artery bypass graft surgery, percutaneous transluminal coronary angioplasty, and cardiovascular arrest and/or death are included, but are not seen as limiting for other pathological processes which involve reperfusion of ischemic tissue in their individual pathologies.
  • arrhythmia is generally defined as any variation from the normal rhythm of the heart beat, including sinus arrhythmia, premature beat, heart block, atrial fibrillation, atrial flutter, ventricular tachycardia, ventricular fibrillation, torsades de pointes, pulsus alternans and paroxysmal tachycardia.
  • cardiac arrhythmia is generally defmed as a disturbance of the electrical activity of the heart that manifests as an abnormality in heart rate or heart rhythm. Arrhythmia is most commonly related to cardiovascular disease, and in particular, ischemic heart disease.
  • cancer is generally considered to be characterized by the uncontrolled, abnormal growth of cells.
  • cancer may refer to tissue in a diseased state including pre-cancerous, carcinogen-initiated and carcinogen-transformed cells.
  • structural carotenoid analogs or derivatives may be generally defmed as carotenoids and the biologically active structural analogs or derivatives thereof.
  • “Derivative” in the context of this application is generally defined as a chemical substance derived from another substance either directly or by modification or partial substitution.
  • “Analog” in the context of this application is generally defined as a compound that resembles another in structure but is not necessarily an isomer. Typical analogs or derivatives include molecules which demonstrate equivalent or improved biologically useful and relevant function, but which differ structurally from the parent compounds.
  • Parent carotenoids are selected from the more than 700 naturally-occurring carotenoids described in the literature, and their stereo- and geometric isomers.
  • Such analogs or derivatives may include, but are not limited to, esters, ethers, carbonates, amides, carbamates, phosphate esters and ethers, sulfates, glycoside ethers, with or without spacers (linkers).
  • the synergistic combination of more than one structural analog or derivative of carotenoids may be generally defined as any composition including one structural carotenoid analog or derivative combined with one or more other structural carotenoid analogs or derivatives or co-antioxidants, either as derivatives or in solutions and/or formulations.
  • subject may be generally defined as all mammals, in particular humans.
  • administration may be generally defined as the administration of the pharmaceutical or over-the-counter (OTC) or nutraceutical compositions by any means that achieves their intended purpose.
  • administration may include parenteral, subcutaneous, intravenous, intracoronary, rectal, intramuscular, intra-peritoneal, transdermal, or buccal routes.
  • administration may be by the oral route.
  • the dosage administered will be dependent upon the age, health, weight, and/or disease state of the recipient, kind of concurrent treatment, if any, frequency of treatment, and/or the nature of the effect desired.
  • techniques described herein may be applied to the inhibition and/or amelioration of any disease or disease state related to reactive oxygen species. Any techniques described herein directed towards the inhibition of ischemia-reperfusion injury may also be applied to the inhibition or amelioration of a liver disease, a non-limiting example being Hepatitis C infection. Techniques described herein directed towards the inhibition and/or amelioration of ischemia-reperfusion injury may also be applied to the inhibition and/or amelioration of arrhythmia. Techniques described herein directed towards the inhibition and/or amelioration of ischemia-reperfusion injury may also be applied to the inhibition and/or amelioration of cancer. In some embodiments, techniques described herein may be used for controlling connexin 43 expression. Techniques described herein may be used to control gap junctional communication. In some embodiments, techniques described herein may be used for controlling C-reactive protein levels.
  • An embodiment may include the administration of structural carotenoid analogs or derivatives alone or in combination to a subject such that the occurrence of ischemia-reperfusion injury is thereby inhibited and/or ameliorated.
  • the structural carotenoid analogs or derivatives may be water soluble and/or water dispersible derivatives.
  • the carotenoid derivatives may include any substituent that substantially increases the water solubility of the naturally occurring carotenoid.
  • the carotenoid derivatives may retain and/or improve the antioxidant properties of the parent carotenoid.
  • the carotenoid derivatives may retain the non-toxic properties of the parent carotenoid.
  • the carotenoid derivatives may have increased bioavailability, relative to the parent carotenoid, upon administration to a subject.
  • the parent carotenoid may be naturally occurring.
  • compositions comprised of the synergistic combination of more than one structural analog or derivative of carotenoids to a subject such that the occurrence of ischemia-reperfusion injury is thereby reduced.
  • the composition may be a “racemic” (i.e. mixture of the potential stereoisomeric forms) mixture of carotenoid derivatives.
  • pharmaceutical compositions comprised of structural analogs or derivatives of carotenoids in combination with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier may be serum albumin.
  • structural analogs or derivatives of carotenoids may be complexed with human serum albumin (i.e., HSA) in a solvent. HSA may act as a pharmaceutically acceptable carrier.
  • compositions may include all compositions of 1.0 gram or less of a particular structural carotenoid analog, in combination with 1.0 gram or less of one or more other structural carotenoid analogs or derivatives and/or co-antioxidants, in an amount which is effective to achieve its intended purpose. While individual subject needs vary, determination of optimal ranges of effective amounts of eachcomponent is with the skill of the art.
  • a structural carotenoid analog or derivative may be administered to mammals, in particular humans, orally at a dose of 5 to 100 mg per day referenced to the body weight of the mammal or human being treated for ischemia-reperfusion injury.
  • a structural carotenoid analog or derivative may be administered to mammals, in particular humans, parenterally at a dose of between 5 to 1000 mg per day referenced to the body weight of the mammal or human being treated for ischemia-reperfusion injury. In other embodiments, about 100 mg of a structural carotenoid analog or derivative is either orally or parenterally administered to treat or prevent ischemia-reperfusion injury.
  • the unit oral dose may comprise from about 0.25 mg to about 1.0 gram, or about 5 to 25 mg, of a structural carotenoid analog.
  • the unit parenteral dose may include from about 25 mg to 1.0 gram, or between 25 mg and 500 mg, of a structural carotenoid analog.
  • the unit intracoronary dose may include from about 25 mg to 1.0 gram, or between 25 mg and 100 mg, of a structural carotenoid analog.
  • the unit doses may be administered one or more times daily, on alternate days, in loading dose or bolus form, or titrated in a parenteral solution to commonly accepted or novel biochemical surrogate marker(s) or clinical endpoints as is with the skill of the art.
  • the compounds may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers, preservatives, excipients and auxiliaries which facilitate processing of the structural carotenoid analog or derivative which may be used pharmaceutically.
  • suitable pharmaceutically acceptable carriers such as tablets, softgels, lozenges, dragees, and capsules
  • the pharmaceutical preparations may be manufactured in a manner which is itself known to one skilled in the art, for example, by means of conventional mixing, granulating, dragee-making, softgel encapsulation, dissolving, extracting, or lyophilizing processes.
  • pharmaceutical preparations for oral use may be obtained by combining the active compounds with solid and semi-solid excipients and suitable preservatives, and/or co-antioxidants.
  • the resulting mixture may be ground and processed.
  • the resulting mixture of granules may be used, after adding suitable auxiliaries, if desired or necessary, to obtain tablets, softgels, lozenges, capsules, or dragee cores.
  • Suitable excipients may be fillers such as saccharides (e.g., lactose, sucrose, or mannose), sugar alcohols (e.g., marmitol or sorbitol), cellulose preparations and/or calcium phosphates (e.g., tricalcium phosphate or calcium hydrogen phosphate).
  • binders may be used such as starch paste (e.g., maize or corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone).
  • Disintegrating agents may be added (e.g., the above-mentioned starches) as well as carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof (e.g., sodium alginate).
  • Auxiliaries are, above all, flow-regulating agents and lubricants (e.g., silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol, or PEG).
  • Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices.
  • Softgelatin capsules are providedwith suitable coatings, which, typically, contain gelatin and/or suitable edible dye(s).
  • animal component-free and kosher gelatin capsules may be particularly suitable for the embodiments described herein for wide availability of usage and consumption.
  • concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol (PEG) and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures, including dimethylsulfoxide (DMSO), tetrahydrofuran (THF), acetone, ethanol, or other suitable solvents and co-solvents.
  • DMSO dimethylsulfoxide
  • THF tetrahydrofuran
  • cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate
  • Dye stuffs or pigments may be added to the tablets or dragee coatings or softgelatin capsules, for example, for identification or in order to characterize combinations of active compound doses, or to disguise the capsule contents for usage in clinical or other studies.
  • Other pharmaceutical preparations which may be used orally include push-fit capsules made of gelatin, as well as soft, thermally-sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules may contain the active compounds in the form of granules which may be mixed with fillers such as, for example, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers and/or preservatives.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils such as rice bran oil or peanut oil or palm oil, or liquid paraffin.
  • stabilizers and preservatives may be added.
  • pulmonary administration of a pharmaceutical preparation may be desirable.
  • Pulmonary administration may include, for example, inhalation of aerosolized or nebulized liquid or solid particles of the pharmaceutically active component dispersed in and surrounded by a gas.
  • Possible pharmaceutical preparations which may be used rectally include, for example, suppositories, which consist of a combination of the active compounds with a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, or parrafin hydrocarbons.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base.
  • Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffm hydrocarbons.
  • Suitable formulations for parenteral administration include, but are not limited to, aqueous solutions of the active compounds in water-soluble and/or water dispersible form, for example, water-soluble salts, esters, carbonates, phosphate esters or ethers, sulfates, glycoside ethers, together with spacers and/or linkers.
  • Suspensions of the active compounds as appropriate oily injection suspensions may be administered, particularly suitable for intramuscular injection.
  • Suitable lipophilic solvents, co-solvents (such as DMSO or ethanol), and/or vehicles including fatty oils, for example, rice bran oil or peanut oil and/or palm oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides, may be used.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, dextran, and/or cyclodextrins. Cyclodextrins (e.g., ⁇ -cyclodextrin) may be used specifically to increase the water solubility for parenteral injection of the structural carotenoid analog.
  • Liposomal formulations in which mixtures of the structural carotenoid analog or derivative with, for example, egg yolk phosphotidylcholine (E-PC), may be made for injection.
  • the suspension may contain stabilizers, for example, antioxidants such as BHT, and/or preservatives, such as benzyl alcohol.
  • reagents were purchased from commercial sources and used as received unless otherwise indicated. Solvents for reactions and isolations were reagent grade and used without purification unless otherwise indicated. All of the following reactions were performed under nitrogen (N 2 ) atmosphere and were protected from direct light.
  • N 2 nitrogen
  • “Racemic” astaxanthin (as the mixture of stereoisomers 3S,3′S, meso, and 3R,3′R in a 1:2:1 ratio) was purchased from Divi's Laboratories, Ltd (Buckton Scott, India). “Racemic” lutein and zeaxanthin were purchased from Indofme Chemical Co., Inc.
  • TLC Thin-layer chromatography
  • IPC in-process control
  • Disuccinic acid ester of astaxanthin XV (2 g, 2.509 mmol) and 200 mL ethanol were stirred at room temperature under nitrogen in a 500 mL round-bottom flask.
  • Sodium ethoxide (340 mg, 5.019 mmol, Acros #A012556101) was added as a solid in a single portion and the solution was allowed to stir overnight. The following day, the precipitate was filtered off and washed with ethanol followed by methylene chloride to afford a purple solid, the disodium salt of the disuccinic acid ester of astaxanthin, XVI [1.41 g, 67%] and was placed on a high vacuum line to dry.
  • the reaction was quenched by pouring the cold reaction mixture into a separatory funnel containing 1.00 L of IPAC and 500 mL of a saturated solution of ammonium chloride and 500 mL of water.
  • the organic layer was concentrated to a white solid.
  • the solid was reslurried in dichloromethane (250 mL) for 2 h and heptane (1.00 L) was added and stirred for 1 h.
  • the mixture was concentrated under vacuum to a volume of 500 mL.
  • Racemic lutein 2B (“xanthophyll”) was purchased from ChemPacific. Flash chromatography was performed on Natland International Corporation 230400 mesh silica gel using the indicated solvents.
  • sample sodium disuccinate astaxanthin derivative, as the all-trans mixture of stereoisomers 3S,3′S, meso, and 3R,3′R in a 1:2:1 ratio
  • sample sodium disuccinate astaxanthin derivative, as the all-trans mixture of stereoisomers 3S,3′S, meso, and 3R,3′R in a 1:2:1 ratio
  • sterile-filtered (0.2 ⁇ M Millipore®) deionized (DI) water in a 15 mL glass centrifuge tube.
  • DI deionized
  • a 1 mL volume of filtrate was then diluted appropriately with DI water, and the concentration of the solution was measured at 480 run using a four point calibration curve prepared from fresh sample. After taking the dilutions into account, the concentration of the saturated solution of the disodium disuccinate astaxanthin derivative was 8.64 mg/mL.
  • FIG. 27 and FIG. 28 depict the results of spectral analysis after flash photolysis of the formation of triplet and carotenoid cation radical states for non-esterified, free astaxanthin 2E and the diacid disuccinate astaxanthin derivative XV were obtained.
  • Formation of the carotenoid cation radical is a measure of the potential biophysical behavior of the novel derivative as an antioxidant. If a derivative retains the antioxidant behavior of non-esterified, free astaxanthin, then all previously documented (i.e. literature precedent) therapeutic applications for astaxanthin can be reasonably assumed for the novel derivative, including at least singlet oxygen quenching, lipid peroxidation chain-breaking, and/or direct radical scavenging.
  • Negative peaks in the spectra demonstrate ground state depletion of NN and astaCOOH XV.
  • the positive peak at 550 nm shows the formation of the astaCOOH XV triplet; the positive peak at 850 nm shows the formation of the astaCOOH XV cation radical.
  • the 3 car decays rather quickly. After 15 is, half of the 3 car has disappeared, and after 50 ⁇ s, no 3 car is left. The car + is stable within this time frame.
  • mouse embryonic fibroblast CH3/10T 1/2 cells were treated with the following formulations in a 4 mL cell culture system with media containing 2% calf serum:
  • TTNPB is a highly potent retinoid that is effective at inducing connexin 43 expression at the 96-hour time point at 10 ⁇ 8 M.
  • GJC Intercellular Gap Junctional Communication
  • GJC gap junctional communication
  • junctional permeability was assayed by microinjection of the fluorescent dye Lucifer Yellow CH (Sigma, St. Louis, Mo.) into individual confluent cells essentially as described previously (Zhang, 1994). Briefly, confluent cultures of C3H/10T1 ⁇ 2 cells were treated for 4 days with: (1) the disodium salt disuccinate astaxanthin derivative XVI (1 ⁇ 10 ⁇ 5 M) dissolved in a 1:2 ethanol/water (EtOH/ H 2 O) formulation; (2) a synthetic retinoid, TTNPB (1 ⁇ 10 ⁇ 8 M) dissolved in tetrahydrofuran as a positive control; or (3) 1:2 EtOH/H 2 O treated cells as a negative control.
  • the number of fluorescent cells adjacent to the injected cell was later determined by digital image analysis using an unbiased density threshold method and the SigmaScan software program (Jandel Scientific). This number of communicating cells was used as an index of junctional communication, as described previously (Hossain, 1993).
  • Panel A treatment with the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin at 1 ⁇ 10 ⁇ 5 M in 1:2 EtOH/H 2 O;
  • Panel C 1:2 EtOH/ H 2 O solvent negative control
  • Panel E TTNPB at 1 ⁇ 10 ⁇ 8 M in tetrahydrofuran as solvent, positive control;
  • Panels B, D, F digital analysis of micrographs A, C, E respectively, demonstrating pixels above a set threshold positive for Lucifer Yellow fluorescence. Because cell nuclei have the most volume, they accumulate the most Lucifer Yellow and exhibit the most fluorescence.
  • mouse embryonic fibroblast C3H/10T 1 ⁇ 2 cells were cultured in Eagle's basal medium with Earle's salts (Atlanta Biologicals, Atlanta, Ga.), supplemented with 5% fetal calf serum (Atlanta Biologicals, Atlanta, Ga.) and 25 ⁇ g/mL gentamicin sulfate (Sigma, St. Louis, Mo.), and incubated at 37° C. in 5% CO 2 .
  • the confluent cells were treated for four days with the disodium salt disuccinate astaxanthin derivatives XVI and then harvested and analyzed for Cx43 protein induction as described.
  • Protein content was measured using the Protein Assay Reagent kit (Pierce Chemical Co., Rockford, Ill.) according to manufacturer's instructions.
  • Cell lysates containing 100 ⁇ g of protein were analyzed by Western blotting using the NuPage western blotting kit and apparatus (Invitrogen, Carlsbad, Calif.) and Cx43 protein detected using a rabbit polyclonal antibody (Zymed, San Francisco, Calif.) raised against a synthetic polypeptide corresponding to the C-terminal domain of mouse, human and rat Cx43.
  • Cx43 immunoreactive bands were visualized by chemiluminescence using an anti-rabbit HRP-conjugated secondary antibody (Pierce Chemical Co., Rockford, Ill.).
  • Digital images were obtained with a cooled CCD camera, and quantitative densitometry was then performed (Bio-Rad, Richmond, Calif.). Equal protein loading of the lanes was confirmed by staining with Coomassie blue protein stain and digital image analysis.
  • disodium salt disuccinate astaxanthin derivatives XVI were added to cell cultures in a formulation of 1:2 ethanol/ H 2 O at 1 ⁇ 10 ⁇ 5 M.
  • the statistical mixture of stereoisomers and purified enantiomeric forms demonstrated increased expression of Cx43 in comparison to cell cultures treated with 1:2 ethanol/H 2 O alone ( FIG. 15A and FIG. 15B ).
  • Treatment with the statistical mixture of stereoisomers of the disodium salt disuccinate astaxanthin derivative XVI elicited the highest induction level of Cx43 of all derivatives tested.
  • induction levels were several-fold less than induction levels seen with the retinoids tetrahydrotetramethylnapthyl propenylbenzoic acid (TTNPB) (Hofftnan-LaRoche, Nutley, N.J.) and retinyl acetate (Sigma, St. Louis, Mo.) included as positive controls; this relative potency difference is consistent with previous studies.
  • TTNPB tetrahydrotetramethylnapthyl propenylbenzoic acid
  • retinyl acetate Sigma, St. Louis, Mo.
  • Non-esterified, free astaxanthin 2E is generated in the mammalian gut after oral administration of esterified astaxanthin. Only free astaxanthin is found in mammalian plasma and solid organs. This was again demonstrated in single- and multiple dose oral pharmacokinetic studies; the results are described herein. Inherent esterase activity of serum albumin, and the action of promiscuous esterases in serum and solid organs rapidly generates non-esterified, free astaxanthin after parenteral administration of the disodium disuccinate astaxanthin derivative (XVl).
  • Flash photolysis experiments also demonstrated that the disodium disuccinate astaxanthin derivative XVI and non-esterified, free astaxanthin have identical antioxidant behavior in terms of formation of the carotenoid cation radical.
  • An experiment was performed to assess the ability of non-esterified, free astaxanthin (the in vivo final cleavage product of the disodium salt disuccinate astaxanthin derivative (XVI), tested as the all-trans mixture of stereoisomers 3S,3′S, meso, and 3R,3′R in a 1:2:1 ratio) to inhibit neoplastic transformation in the C3H10T1 ⁇ 2 cell culture model developed in the lab of the late Charles Heidelberger (Reznikoff, 1973).
  • FIG. 34 depicts effects of non-esterified, free astaxanthin (as the all-trans mixture of stereoisomers) on MCA-induced neoplastic transformation.
  • Graph represents a total of 68 cultures treated with astaxanthin 2E at 3 ⁇ 10 ⁇ 6 M, 1 ⁇ 10 ⁇ 6 M and 3 ⁇ 10 ⁇ 7 M, delivered in a THF vehicle of 0.3%, 0.1% and 0.03%, respectively.
  • Controls were as follows: a total of 16 dishes did not receive carcinogen and were treated with 0.05% ethanol solvent; controls did not exhibit any transformation events. A total of 20 dishes were treated with MCA and 1% THF solvent, yielding a transformation rate of 0.92 foci/dish. Percent reduction (% reduction) of transformation in astaxanthin-treated dishes was calculated by a comparison of the mean foci/dish of each treatment with the MCA-treated controls. Inferential statistics were performed using the paired Student's t-test; calculated P values of 0.00004, 0.00001, and 0.00006, respectively, were obtained. P ⁇ 0.05 was considered significant. Treatment with 3 ⁇ 10 ⁇ 6 M astaxanthin 2E resulted in complete suppression of the transformed phenotype ( FIG. 35 ).
  • FIG. 35 depicts a comparison of astaxanthin-treated dish to control dishes. Representative dishes treated with: A, no MCA with solvent control; B, MCA 5.0 ⁇ g/ml with 1% THF as solvent control; C, MCA with 3 ⁇ 10 ⁇ 6 M astaxanthin (as the all-trans mixture of stereoisomers) in THF. It is notable that this level of inhibition far exceeded that reported previously for all other carotenoids tested using identical protocols (Bertram, 1991). A comparison of the current data to data previously reported for percent reduction in neoplastic transformation at the concentrations tested revealed astaxanthin 2E to be a far more potent inhibitor of transformation than either ⁇ -carotene or canthaxanthin ( FIG. 36 ). FIG.
  • neutrophils were isolated on a Percoll gradient from whole blood from a human volunteer. The isolated neutrophils were then re-suspended in phosphate-buffered saline, and maximally stimulated with phorbol ester to induce the respiratory burst and production of superoxide anion.
  • the disodium salt disuccinate astaxanthin derivative XVI was added at various concentrations, and the superoxide signal [as measured with electron paramagnetic resonance (EPR) spectroscopy] was subsequently measured.
  • the disodium salt disuccinate astaxanthin derivative XVI (as the mixture of stereoisomers) reduced the measured superoxide anion signal in a dose-dependent manner ( FIG.
  • FIG. 2 demonstrates the strong superoxide signal after activation in controls, then the results of titration with the disodium salt disuccinate astaxanthin derivative XVI from 100 ⁇ M to 3 mM.
  • FIG. 3 depicts an effect of a disodium salt disuccinate astaxanthin derivative XVI/Vitamin C solution on reactive oxygen species (superoxide anion) as monitored using EPR spectroscopy.
  • the solution included a mixture of about 2 to about 1 of vitamin C to disodium salt disuccinate astaxanthin derivative XVI respectively.
  • the disodium salt disuccinate astaxanthin derivative XVI/Vitamin C solution reduced the measured superoxide anion signal in a dose-dependent manner ( FIG. 3 ); complete suppression of the superoxide anion signal was achieved at 0.02 ⁇ M concentration.
  • FIG. 3 demonstrates the strong superoxide signal after activation in controls, then the results of titration with the disodium salt disuccinate astaxanthin derivative XVI[Vitamin C solution from 0.01 ⁇ M to 0.02 ⁇ M.
  • neutrophils were again isolated on a Percoll gradient from whole blood from a second human volunteer.
  • the isolated neutrophils were then re-suspended in phosphate-buffered saline, and maximally stimulated with phorbol ester to induce the respiratory burst and production of superoxide anion.
  • the hydrochloride salt dilysinate astaxanthin derivative (XX) was added at four (4) concentrations, and the superoxide signal (as measured with EPR spectroscopy) was subsequently measured.
  • the hydrochloride salt dilysinate astaxanthin derivative XX also reduced the measured superoxide anion signal in a dose-dependent manner ( FIG.
  • Non-esterified, all-E astaxanthin 2E [1:2:1 statistical mixture of stereoisomers 3S,3′S, meso (identical 3S,3′R and 3′S,3R), and 3R,3′R] was purchased from Buckton Scott (India) and used as supplied (>95% purity by HPLC). Astaxanthin 2E was dissolved in HPLC grade dimethylsulfoxide (DMSO; Sigma-Aldrich, St. Louis, Mo.).
  • DMSO dimethylsulfoxide
  • the disodium disuccinate derivatives XVI of astaxanthin 2E were tested separately in nine formulations: statistical mixture of stereoisomers (as for astaxanthin, above, a 1:2:1 mixture of all-E; labeled as “mixture” in all tables and figures); 3S,3′S, and 3R,3′R (optical isomers or enantiomers); and meso (mixture of identical 3S,3′R and 3′S,3R; diastereomers of the enantiomeric pair). All disuccinate derivatives were synthesized at >90% purity by HPLC. The disuccinate derivatives were first tested at the appropriate final concentrations in pure aqueous solution (deionized water) from stock solutions of 10 mM.
  • each of the four disuccinate derivatives were then tested from stock solutions prepared in a 1:2 mixture of ethanol (final concentration of EtOH in stock solution 33 1 ⁇ 3%; final concentration in isolated neutrophil assay 0.3%; HPLC grade ethanol, Sigma-Aldrich, St. Louis, Mo.) at 10 mM.
  • the 3S,3′S derivative was also tested from a 50% EtOH concentration stock solution (final concentration in isolated neutrophil assay 0.5%).
  • Ethanolic formulation of the disuccinate derivatives has been shown to completely disaggregate the supramolecular assemblies which form in pure aqueous solution, providing monomeric solutions of the derivatives immediately before introduction into the test assay.
  • a carotenoid derivative [Succinic acid mono-(4- ⁇ 18-[4-(3-carboxy-propionyloxy)-2,6,6-trimethyl-3-oxo-cyclohex-1-enyl]-3,7,12,16-tetramethyl-octadeca-1,3,5,7,9,11,13,15,17-nonaenyl ⁇ -3,5,5-trimethyl-2-oxo-cyclohex-3-enyl) ester; FIG. 17 ] and its stereoisomeric forms were synthesized, disodium disuccinate derivatives XVI of astaxanthin 2E, in all-trans (all-E) form.
  • the derivatives are symmetric chiral molecules with 2 chiral centers at the 3 and 3′ carbon positions, comprising 4 stereoisomers: 3R,3′R and 3S,3′S (optical isomers, or enantiomers), as well as the diastereomeric meso forms (identical 3R,3′S and 3′R,3S).
  • the statistical mixture of stereoisomers synthesized from the commercial source of astaxanthin contains 3R,3′R, meso (identical 3R,3′S and 3′R,3S), and 3S,3′S stereoisomeric forms in a 1:2:1 ratio.
  • All individual stereoisomers and the statistical mixture were synthesized at >90% purity by HPLC, allowing direct comparison of the individual efficacy of these forms as direct radical scavengers.
  • the all-E forms of the stereoisomers used in this study were linear, rigid molecules (bolaamphiphiles) owing to the lack of cis (or Z) configuration(s) in the polyene chain of the spacer material.
  • the disodium disuccinate diesters XVI of astaxanthin 2E demonstrate increased water “dispersibility” over the parent compound astaxanthin 2E.
  • the water dispersibilities of the individual stereoisomers and the statistical mixture were all greater than 8 mg/mL (approximately 10 mM), allowing them to be introduced into the buffered aqueous test system without a co-solvent.
  • the tendency for the parent carotenoids such as astaxanthin 2E (Salares, 1977), as well as carotenoid derivatives (e.g. capsanthin derivatives) (Zsila, 2001 and Bikadi, 2002) to form supramolecular assemblies in aqueous solution was also observed for the derivatives tested in the current study.
  • PMNs Human polymorphonuclear leukocytes
  • Erythrocytes were lysed by addition of 18 mL of ice-cold water for 30 s, followed by 2 mL of 10 ⁇ PIPES buffer (25 mM PIPES, 110 mM NaCl, and 5 mM KCl, titrated to pH 7.4 with NaOH). Cells were pelleted at 4° C., the supernatant was decanted, and the procedure was repeated. After the second hypotonic lysis, cells were washed twice with PAG buffer (PIPES buffer containing 0.003% human serum albumin and 0.1% glucose). Afterward, PMNs were counted by light microscopy on a hemocytometer. The fmal pellet was then suspended in PAG-CM buffer (PAG buffer with 1 mM CaCl 2 and 1 mM MgCl 2 ).
  • PAG buffer PAG buffer with 1 mM CaCl 2 and 1 mM MgCl 2 .
  • the potent SOD mimetic produced by Metaphore, Inc. served as a positive control at study outset. As has been observed repeatedly in the Zweier laboratory, the 10 ⁇ M dose in water-only vehicle nearly completely eliminated the superoxide anion signal as detected with DEPMPO (97% inhibition; Table 1). An ethanol-alone negative control (final concentration 0.3%) was also evaluated, as ethanol shows minor scavenging activity in these systems; 5.7% inhibition was seen at this concentration. This amount of inhibition was not subtracted from formulations containing ethanol in the descriptive data in Table 1, as the utility of the dosing vehicle itself (disodium disuccinate derivative XVI in EtOH) in direct scavenging was being evaluated in this study. Non-esterified, free astaxanthin in DMSO (100 ⁇ M) was evaluated as a reference standard for direct comparison to the novel derivatives synthesized for this study; mean inhibition of the astaxanthin/DMSO vehicle was 28% (Table 1).
  • FIG. 18 shows the relative scavenging ability of each of the 3 stereoisomers (mixture and 3 individual stereoisomers) in water, at a final concentration of 100 ⁇ M. Except for the 3R,3′R enantiomer (28.7% inhibition), all other derivative formulations showed decreased scavenging ability relative to the astaxanthin/DMSO formulation (range ⁇ 2.0% to 19.3% inhibition; Table 1). As can be seen, the 3S,3′S formulation did not exhibit any mean scavenging activity. When introduced into the isolated neutrophil test system in ethanolic formulation, however, in each case the scavenging ability increased over that of the same derivative formulated in water ( FIG. 19 ; range 38.0% to 42.5%).
  • FIG. 20 shows the results of titration of superoxide signal inhibition by increasing concentrations of the mixture of stereoisomers of disodium disuccinate astaxanthin XVI in ethanolic formulation.
  • concentration was increased from 100 ⁇ M to 3 mM, near complete inhibition of superoxide signal was noted (95.0% inhibition at the 3 mM dose; Table 1 and FIG. 18 ).
  • the dose-response curve was non-linear. Adjusting for percent inhibition and tested dose, the disodium disuccinate derivative was between one and two orders of magnitude less potent than the SOD mimetic used as a positive control in the current study (Table 1).
  • Table 1 depicts descriptive statistics for various formulations of disodium disuccinate derivatives of astaxanthin tested in the current study.
  • Astaxanthin 2E is a potent lipophilic antioxidant that normally exerts its 5 antioxidant properties in lipid-rich cellular membranes, lipoproteins, and other tissues (Britton, 1995).
  • the pure aqueous formulations of the novel derivatives were less potent than the ethanolic formulations in terms of direct scavenging ability.
  • Supramolecular assembly of the water soluble carotenoid derivatives in some solvents may explain their lack of potency in those solvents.
  • the aggregation is of the helical, “card-pack” type, with aggregates greater than 240 nmn in size forming in pure aqueous solution. Increasing ionic strength of buffer solutions may increase both the size and stabilility of these aggregates.
  • the disodium disuccinate astaxanthin derivative XVI is one to two orders of magnitude less potent than the SOD mimetic.
  • these derivatives decay to free astaxanthin, which becomes active in the lipid-rich membranes of cells [including the mitochondrial and nuclear membranes (Goto, 2001)], therefore providing dual protection (aqueous and lipid-phase radical scavenging), not achievable with water-soluble proteins and enzyme mimetics.
  • Non-esterified, free astaxanthin (when provided as a dietary supplement at 0.02% of feed wt/wt) is cardioprotective against the ROS-mediated strenuous exercise insult to both skeletal and cardiac muscle (Aoi et al. 2003). Therefore, this characteristic (i.e. dual-phase radical scavenging) should provide additional utility for this class of compounds as clinical therapeutic agents in those indications for which radical and reactive oxygen species prevention is important (Cross, 1987).
  • the study demonstrates for the first time direct scavenging of superoxide anion detected by EPR spectroscopy by a group of carotenoid derivatives.
  • the compounds were found to form supramolecular assemblies in pure aqueous solution. Formation of supramolecular assemblies may limit their scavenging potency relative to monomeric solutions of the same compounds. No significant differences in scavenging ability were seen among the 3 stereoisomers of the carotenoid derivatives.
  • Dose-ranging studies revealed that the concentration of derivative could be increased to near-complete suppression of the induced superoxide anion signal.
  • this class of compounds may be used as both an aqueous phase and lipid phase scavenger, which should find wide application in those acute and chronic disease conditions for which potent radical scavengers have demonstrated efficacy.
  • neutrophils were isolated on a Percoll gradient from whole blood from a human volunteer. The isolated neutrophils were then re-suspended in phosphate-buffered saline, and maximally stimulated with phorbol ester to induce the respiratory burst and production of superoxide anion.
  • EPR electron paramagnetic resonance
  • the disodium disuccinate di-vitamin C astaxanthin derivative (XXIII) (semi-systematic name Succinic acid 4-[18-(4- ⁇ 3-[2-(3,4-dihydroxy-5-oxo-2,5-dihydrofuran-2-yl)-2-hydroxy-ethoxycarbonyl]-propionyloxyl-2,6,6-trimethyl-2-oxo-cyclohex-1-enyl)-3,7,12,16-tetramethyl-octadeca-1,3,5,7,9,11,13,15,17-nonaenyl]-3,5,5-trimethyl-2-oxo-cyclohex-3-enyl ester 2-(3,4-dihydroxy-5-oxo-2,5-dihydrofuran-2-yl)-2-hydroxy-ethyl ester) was added at various concentrations, and the superoxide signal (as measured with EPR spectroscopy
  • the disodium disuccinate di-vitamin C astaxanthin derivative (XXII) reduced the measured superoxide anion signal in a dose-dependent manner ( FIG. 33 ); complete suppression of the superoxide anion signal was achieved at 60 ⁇ M concentration. This represents a 50-fold increase in potency over the disodium disuccinate astaxanthin derivative (XVI) also synthesized for the current series of experiments.
  • the purity of the derivative as tested was 88% (by HPLC area under the curve, or AUC).
  • the potency of the derivative (XXIII) approached that of the formulation of disodium disuccinate astaxanthin (XVI) with free vitamin C in a 1:2 molar ratio (which completely suppressed the superoxide anion signal in a 20 ⁇ M/40 ⁇ M disodium disuccinate astaxanthin derivative (XVI)/free vitamin C formulation).
  • Derivative (XXIII) which generates 2 moles of free vitamin C and 1 mole of non-esterified, free astaxanthin for every mole of derivative in vivo may be particularly preferred for certain clinical indications.
  • Derivative (XXIII) will also likely show increased efficacy in those clinical situations in which aqueous-phase scavenging (by the intact parent derivative, as well as free vitamin C) as well as lipid-phase scavenging (by non-esterified, free astaxanthin) are important for reduction in the pathology attributable to ROS and other radical species injury.
  • FIG. 4 , FIG. 25 , and FIG. 26 depict graphical representations of the reduction of infarct size in male Sprague-Dawley rats.
  • Male Sprague-Dawley rats were pre-treated with the disodium salt disuccinate astaxanthin derivative XVI (as the mixture of stereoisomers) in aqueous solution before performing an occlusion and inducing a myocardial infarction.
  • Male Sprague-Dawley rats (175-200 grams) were anaesthetized with 100 mg/kg of Inactin, instrumented, and the heart exposed.
  • the left coronary artery had a suture placed around it and was subjected to 30 minutes of total coronary artery occlusion followed by 2 hours of reperfusion, at which time infarct size was measured in hearts excised from the animal.
  • the hearts were washed in buffer and incubated in triphenyltetrazolium chloride (TTC) staining solution kept at 37° C. in phosphate buffer at pH of 7.40.
  • TTC triphenyltetrazolium chloride
  • I infarct size
  • IS/AAR % of the area at risk
  • mice Male C57BIJ6 mice, approximately 25 g, were housed in cages (three mice/cage) and fed standard mouse chow (Purina Mouse Chow, Ralston Purina, St. Louis) and water ad libitum for at least five days prior to the start of the experiment.
  • standard mouse chow Purina Mouse Chow, Ralston Purina, St. Louis
  • water ad libitum for at least five days prior to the start of the experiment.
  • the disodium disuccinate astaxanthin derivative XVI was mixed with the following components to make an emulsion suitable for oral gavage:
  • the disodium disuccinate astaxanthin derivative XVI demonstrates water-solubility of approximately 8.64 mg/mL in pure aqueous formulation. In the emulsion described above, solubility was increased to approximately 50 mg/mL, allowing for dosing up to 500 mg/kg by gavage in these animals. This significant 6-fold increase in solubility in the dosing vehicle greatly facilitated gavage studies in these small mice.
  • the emulsion was given by oral gavage at 500 mg/kg body weight in a single dose. Food was withdrawn from all cages the evening prior to the experiment. One hour after administration of the emulsion, food and water were restored to all animals.
  • Free astaxanthin concentration was also determined, at the same time points as for plasma samples, in liver. Livers were removed from each animal in the pharmacokinetic study after sacrifice, and snap frozen in liquid nitrogen. Liver tissue was prepared for HPLC analysis as described (Jewell, 1999). Therefore, simultaneous examination of liver accumulation of free astaxanthin was performed at the same time points as the plasma analyses.
  • liver Up to 300 mg of liver from each animal was snap frozen in liquid nitrogen. Tissue homogenization and extraction were performed with a mixture of chloroform/methanol/water, according to the methods of Jewell (1999). Non-esterified, free astaxanthin accumulation in liver was then evaluated by HPLC as described above for plasma samples.
  • disodium disuccinate astaxanthin derivative XVI in the emulsion described above is a superior vehicle for delivery of therapeutic concentrations of free carotenoid to tissues of interest after oral dosing.
  • the C max (Table 4) of 0.9 mg/L is also unprecedented in rodents, animals which absorb only a small percentage of the oral dose of carotenoids. It is significant that these plasma and liver levels of free carotenoid were obtained after just a single dose of compound in the emulsion vehicle.
  • Osterlie et al. (2000) have described C max plasma levels of 1.3 mg/L after a single dose of 100 mg (approximately 1.1 mg/kg oral dose) of non-esterified, free astaxanthin in olive oil vehicle. Humans typically absorb 40 to 50% of the oral dose of carotenoid when provided in fatty vehicle, as opposed to a few percentage points for rodents.
  • the influence of parenteral administration of the disodium disuccinate astaxanthin derivative (XVI) on induced infarct size and induced levels of circulating C-reactive protein (CRP) in rabbits was investigated using the methods of Barrett et al. (2002) with slight modifications.
  • the purpose of the current study was to investigate the ability of the disodium disuccinate astaxanthin derivative (XVI) to reduce inflammation as measured by CRP in the setting of experimental myocardial ischemia-reperfusion injury in the rabbit heart. It has been suggested that CRP, commonly used as a marker for the acute inflammatory (“acute-phase”) response, may actually have a pro-inflammatory effect mediated through the activation of the complement cascade.
  • Myocardial ischemia-reperfusion injury which is accompanied by an increase in the formation of oxygen radicals (ROS) has also been shown to activate the complement system. It has been demonstrated that (1) the endogenous increase in plasma CRP secondary to a remote inflammatory lesion was associated with an increase in myocardial tissue injury secondary to regional ischemia and reperfusion; (2) this increase in injury (manifested as increased infarct size) was mediated by complement activity; and (3) CRP was an “effector”, and not merely an indirect measure of systemic inflammation, in this system. Therefore, reduction of circulating CRP levels, together with the reduction(s) in infarct size previously noted with CardaxTM in rodents, would form a powerful anti-inflammatory therapeutic modality in the acute coronary syndrome setting.
  • ROS oxygen radicals
  • the rabbits were anesthetized with a mixture of xylazine (3 mg/kg) and ketamine (35 mg/kg) followed by pentobarbital (90 mg/kg) intramuscularly. Additional pentobarbital was administered as necessary to maintain anesthesia.
  • the rabbits were ventilated with room air, and the heart was exposed via a left thoracotomy. The heart was then supported in a pericardial cradle and a 3-0 silk ligature was placed around the left anterior descending coronary artery. The artery was occluded for 30 minutes by exerting traction on the ligature and subsequently reperfused for 180 minutes. Shortly before completing the protocol, a venous blood sample was obtained for determination of plasma CRP.
  • the hearts were removed and cannulated by the aorta on the Langendorff perfusion apparatus.
  • the hearts were then perfused with a modified Krebs-Henseleit buffer for 10 to 15 minutes (20-25 muminute).
  • the hearts were perfused with 80 mL of 0.4% 2,3,5-triphenyltetrazolium chloride (TTC) at 37° C. for determination of the area-at-risk (AAR).
  • TTC 2,3,5-triphenyltetrazolium chloride
  • AAR area-at-risk
  • the left circumflex coronary artery was then ligated in the same area as it was during the surgical preparation/experimental infarction.
  • FIG. 37 Mean infarct size in control animals and CardaxTM—treated animals is shown in FIG. 37 .
  • Levels of circulating CRP in control animals and CardaxTM—treated animals (shown as the mean difference between baseline levels and induced levels at the time of reperfusion) is shown in FIG. 38 .
  • Reductions in infarct size of approximately 55.4% percent were seen in CardaxTM—treated rabbits; ischemic area-at-risk was similar in both groups.
  • the mean increase in circulating CRP levels in controls ( ⁇ 23.5%) over baseline was completely abrogated in the CardaxTM—treated animals, to mean levels below those observed at baseline ( ⁇ 15.7%).
  • CRP is both an effector in the acute coronary syndrome—resulting in an increased infarct size in the presence of elevated levels of this acute phase reactant—and a strong independent predictor of cardiovascular risk in primary and secondary prevention cardiac patients—reductions in the levels of this circulating protein forms a strong therapeutic modality.
  • mice Three-month old male ICR mice were treated with LPS and galactosamine in order to induce liver injury (Leist, 1995). Mice were first orally gavaged with either an olive oil/water/ lecithin emulsion (10 ml/kg, or 0.3 mL for a 30 gram mouse), or the same emulsion containing the disodium disuccinate astaxanthin derivative XVI (50 mg/mL) for a final disodium disuccinate astaxanthin dose of 500 mg/kg. Two hours later mice were injected intraperitoneally (IP) with either saline (10 ml/kg) or a solution of E.
  • IP intraperitoneally
  • coli LPS (3 mg/kg, Sigma catalog number L-3755) and D-galactosamine (700 mg/kg). Animals were sacrificed by carbon dioxide (CO 2 ) asphyxiation 5 hours after the EP injection, and plasma was then collected for ALT determination.
  • CO 2 carbon dioxide
  • ROS including the radical nitric oxide NO,
  • substantial systemic inflammation occurs after LPS insult, for which non-esterified, free astaxanthin is protective (Ohgami et al. 2003)
  • the utility of the novel derivative for clinical indications in which such inflammation is promoted represents a particularly useful embodiment.
  • both peak and trough levels were taken (peak levels taken 6 hours after dosing at the probable C max ; trough levels obtained 6 hours after C max , or 12 hours post-dose).
  • Mean peak levels in plasma at peak and trough, respectively were 485 nM and 231 nM; mean peak levels in liver at peak and trough, respectively, were 1760 nM and 519 nM.
  • protective levels were achieved and maintained to 11 days post-multiple dosing; in the case of liver, levels almost 9 times the protective level were achieved.
  • the accumulation in liver was greater than that observed in plasma, demonstrating the increased utility of this dosing vehicle for targeting to this solid organ ( FIG. 32 ). It is also apparent from this data set that chronic administration of the disodium disuccinate astaxanthin derivative XVI will be efficacious in hepatoprotection.
  • a single maximum dose of the disodium disuccinate astaxanthin derivative XVI (500 mglkg) was given by oral gavage in the emulsion vehicle to black mice, and the accumulation of non-esterified, free astaxanthin was measured in four (4) animals at the probable C max and T max (6 hours), as deduced from plasma and liver samples in the prior study.
  • FIG. 5 depicts a carotenoid derivative, the disodium salt disuccinate derivative XVI (dAST) of synthetic meso-astaxanthin (3R,3′S-dihydroxy- ⁇ , ⁇ -carotene-4,4′-dione), in all-trans (all-E) form.
  • the symmetric C 40 -xanthophyll used to generate the new derivative has two chiral centers at the 3 and 3′ positions.
  • C 40 -xanthophyll exhibits no optical activity, as these stereocenters have opposite absolute configurations and internally compensate each other.
  • Natural carotenoid molecules possessing carboxylic functionality bind preferentially to human serum albumin (HSA), the most abundant protein in the blood.
  • HSA human serum albumin
  • the novel derivative dAST XVI was synthesized from crystalline astaxanthin 2E [3R,3′R, 3R,3′S, 3S,3′S (25:50:25)], a statistical mixture of stereioisomers obtained commercially (Buckton Scott, India).
  • the astaxanthin stereoisomers were separated by high-pressure liquid chromatography (HPLC), allowing for the synthesis of the purified meso-disodium salt disuccinate derivative XVI for testing in the current study.
  • the all-trans (all-E) form of the meso stereoisomer used was a linear, rigid molecule owing to the lack of cis (or Z) configuration(s) in the polyene chain of the spacer material ( FIG. 5 ).
  • the disodium salt disuccinate derivative XVI of synthetic meso-astaxanthin was successfully synthesized at >99% purity by HPLC.
  • Essentially fatty acid-free human serum albumin (catalog No. A-1887, lot No. 14H9319) were obtained from Sigma and used as supplied. Double-distilled water and spectroscopy grade dimethyl sulfoxide (DMSO, Scharlau Chemie S.A., Barcelona, Spain) and ethanol (Chemolab, Budapest, Hungary) were used. All other chemicals were of analytical grade.
  • HSA was dissolved in pH 7.4 Ringer or 0.1 M pH 7.4 phosphate buffer solutions.
  • the molecular weight of HSA was defined as 66500 Da.
  • Circular Dichroism and UV/Vis Absorption Spectroscopy were recorded on a Jasco J-715 spectropolarimeter at 25 ⁇ 0.2 and 37 ⁇ 0.2° C. in a rectangular cuvette with 1 cm pathlength. Temperature control was provided by a Peltier thermostat equipped with magnetic stirring. All spectra were accumulated three times with a bandwidth of 1.0 mn and a resolution of 0.5 nm at a scan speed of 100 nm/min. Induced CD was defined as the CD of the dAST XVI-HSA mixture minus the CD of HSA alone at the same wavelengths, and is expressed as ellipticity in millidegrees (mdeg).
  • Phosphate buffer, LP values from 0.82 to 13.10: 2 mL of 2.2 ⁇ 10 ⁇ 6 M HSA solution was placed in the cuvette with 1 cm optical pathlength and ⁇ L volumes of the ligand stock solution (c 3.6 ⁇ 10 ⁇ 4 ) were added with an automatic pipette.
  • initial and final concentrations of HSA and DAST were 4.2 ⁇ 10 ⁇ 6 M ⁇ 4.0 ⁇ 10 ⁇ 6 M and 1.3 ⁇ 10 ⁇ 7 M ⁇ 1.4 ⁇ 10 ⁇ 5 M, respectively.
  • the meso-carotenoid/HSA molar ratio was varied between 0.03 and 3.53.
  • final DMSO concentration did not exceed 5 v/v%.
  • a control experiment was also performed, in which the fluorescence of HSA during addition of 20, 50 and 100 ⁇ L DMSO to the solution was measured.
  • dAST XVI exhibited intense light absorption in the visible spectrum ( FIG. 6 ).
  • the main bell-shaped absorption band centered at 481.5 nm was due to the lowest energy electronic dipole allowed, a ⁇ * transition polarized along the long axis of the polyene chain.
  • the vibrational sub-bands were indeed present beneath this curve, as revealed by the second derivative of the spectrum ( FIG. 6 ). Additionally, in the near-UV region, further transitions were present.
  • the electronic transition moment ( ⁇ ) of the moderately intense band around 300 nm is polarized parallel to the long axis of the DAST XVI molecule.
  • the band at 371 nm ⁇ is oriented along the twofold, C 2 symmetry axis of the conjugated system.
  • the weak n ⁇ * transitions of the carbonyl groups were obscured by the other bands.
  • the meso-carotenoid compound did not show any CD bands in ethanol since the effects of the two opposite chiral centers (3R,3′S) canceled each other (data not shown).
  • carotenoid aggregates built up by chiral monomers also exhibit induced Cotton effects (CE) due to the chiral intermolecular arrangement determined by asymmetric centers.
  • CE Cotton effects
  • FIG. 8 Upon addition of DAST to the HSA solution prepared in pH 7.4 Ringer buffer, two definite, oppositely-signed induced CD bands appeared between 300 and 450 nm with a zero cross-over point at 367 nm ( FIG. 8 ).
  • the figure inserts show the intensities of the induced Cotton effects and the main absorption band at different L/P ratios ( ⁇ and ⁇ values are calculated with respect to the total meso-carotenoid concentration). Magnitudes of the CEs increased with increasing concentration of the ligand, however, their shape and wavelength positions remain unchanged. As mentioned above, there are two transitions below 450 nm which might be responsible for the observed optical activity.
  • the absorption band around 300 nm has transition symmetry B, and the corresponding electric and magnetic transition moments are perpendicular to the twofold symmetry axis along the polyene chain.
  • the electric and magnetic transition moments of the band at 372.5 nm are polarized parallel to the C 2 axis, its transition symmetry is A. It is reasonable to assume that upon protein binding, these bands shift to longer wavelengths due to the changing microenvironment surrounding the polyene chain. It has been well established that CD spectra of carotenoids in which the chromophoric portions belong to the C 2 point group conform to the C 2 -rule: if the overall conjugated system acquires right-handed chirality (i.e.
  • the meso-carotenoid binds to HSA in such a manner that the protein environment fixes the terminal rings in a well-defined chiral conformation that results in the observed negative- and positive-induced CD bands.
  • the absolute configurations of the chiral 3 and 3′centers do not determine the chiroptical property of the molecule; rather, the asymmetric protein environment of the albumin molecule (via non-covalent chemical interactions) determines the observed activity.
  • the direction of the transition dipole moment is known; it is polarized along the long axis of the polyene chain.
  • the neighboring meso-carotenoid molecules are arranged in such a manner that their long axes form a positive (clockwise) intermolecular overlay angle.
  • Chiral arrangements of two conjugated chains shown in FIG. 11 satisfy the former condition; in these cases, a long-wavelength positive and a short wavelength negative band would appear in the CD spectrum.
  • the spectroscopic behavior of the absorption band helps to differentiate between these spatial arrangements. Due to unfavourable Coulombic interactions between the transition dipole moments of neighbouring meso-carotenoid molecules in the case of a and b ( FIG.
  • dAST XVI molecules form a right-handed chiral array in which the long axes of meso-carotenoid monomers form an acute, positive angle ( FIG. 11 , a and b ).
  • the first few meso-carotenoid molecules bind to HSA in right-handed arrangement, and subsequent meso-carotenoid monomers build upon this chiral architecture.
  • HSA provides the first essential step, the chiral initiation (“chiral seeding”); after this the self-assembly continues automatically.
  • chiral seeding chiral seeding
  • the 3 and 3′ chiral centers play a decisive role in allowing the aggregates to form the chiral self-assembly on the HSA molecules.
  • the meso-carotenoid molecules form right- and left-handed assemblies to an equal extent, due to the lack of chiral discrimination.
  • the single tryptophan residue (Trp214) located in the depth of subdomain IIA is largely responsible for the intrinsic fluorescence of HSA.
  • the fluorescence emission spectrum of HSA overlaps with the absorption spectrum of the meso-carotenoid. Therefore, fluorescence spectroscopic measurements were obtained after incremental addition of dAST XVI in DMSO to a solution of HSA. The results clearly demonstrated that the meso-carotenoid molecules were able to effectively quench the intrinsic fluorescence of HSA ( FIG. 12 ).
  • the DMSO used to prepare the stock solution of dAST XVI exhibited a negligible effect on the intrinsic HSA fluorescence ( FIG. 12 ). At an L/P ratio of 0.7, the baseline fluorescence intensity decreased by 50%.
  • dAST XVI binds to other long-chain (C18, C20) fatty acid binding sites of HSA, which have been well-characterized by high resolution X-ray crystallography.
  • the disodium salt disuccinate derivative XVI of synthetic, achiral meso-astaxanthin formed optically inactive, card-pack type aggregates in aqueous buffer solutions, as indicated by the large blue-shift of the main visible absorption band versus the band observed in ethanolic solution.
  • the meso-carotenoid appears to be preferentially associated with HSA in monomeric fashion.
  • the concentration of albumin in human blood in vivo is approximately 0.6 mM, suggesting that at doses of up to 500 mg, the meso-carotenoid (molecular weight 841 Da) will associate with the albumin in monomeric fashion (excluding additional potential non-specific binding to circulating blood cells and lipoproteins, which would increase the potential non-aggregating dose).
  • the meso-carotenoid molecular weight 841 Da
  • Bound meso-carotenoid molecules exhibited induced CD bands which were adequately explained by a right-handed helical conformation of the conjugated system.
  • Fibrinolytic therapy and n-acetylcysteine in the treatment of patients with acute myocardial infarction its influence on authentic plasma hydroperoxide levels and polymorphonuclear neutrophil oxygen metabolism. Cardiology 91: 60-65.
US10/793,702 2002-07-29 2004-03-04 Pharmaceutical compositions including carotenoid analogs or derivatives for the inhabition and amelioration of disease Abandoned US20050075337A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/793,702 US20050075337A1 (en) 2002-07-29 2004-03-04 Pharmaceutical compositions including carotenoid analogs or derivatives for the inhabition and amelioration of disease

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US39919402P 2002-07-29 2002-07-29
US46797303P 2003-05-05 2003-05-05
US47283103P 2003-05-22 2003-05-22
US47374103P 2003-05-28 2003-05-28
US48530403P 2003-07-03 2003-07-03
US10/629,538 US7145025B2 (en) 2002-07-29 2003-07-29 Structural carotenoid analogs for the inhibition and amelioration of disease
US10/793,702 US20050075337A1 (en) 2002-07-29 2004-03-04 Pharmaceutical compositions including carotenoid analogs or derivatives for the inhabition and amelioration of disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/629,538 Continuation US7145025B2 (en) 2002-07-29 2003-07-29 Structural carotenoid analogs for the inhibition and amelioration of disease

Publications (1)

Publication Number Publication Date
US20050075337A1 true US20050075337A1 (en) 2005-04-07

Family

ID=31192475

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/629,538 Expired - Lifetime US7145025B2 (en) 2002-07-29 2003-07-29 Structural carotenoid analogs for the inhibition and amelioration of disease
US10/793,696 Abandoned US20050065097A1 (en) 2002-07-29 2004-03-04 Carotenoid ether analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US10/793,702 Abandoned US20050075337A1 (en) 2002-07-29 2004-03-04 Pharmaceutical compositions including carotenoid analogs or derivatives for the inhabition and amelioration of disease
US10/793,703 Expired - Lifetime US7317008B2 (en) 2002-07-29 2004-03-04 Carotenoid ester analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US11/357,897 Expired - Lifetime US7592449B2 (en) 2002-07-29 2006-02-17 Structural carotenoid analogs for the inhibition and amelioration of disease

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/629,538 Expired - Lifetime US7145025B2 (en) 2002-07-29 2003-07-29 Structural carotenoid analogs for the inhibition and amelioration of disease
US10/793,696 Abandoned US20050065097A1 (en) 2002-07-29 2004-03-04 Carotenoid ether analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/793,703 Expired - Lifetime US7317008B2 (en) 2002-07-29 2004-03-04 Carotenoid ester analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US11/357,897 Expired - Lifetime US7592449B2 (en) 2002-07-29 2006-02-17 Structural carotenoid analogs for the inhibition and amelioration of disease

Country Status (12)

Country Link
US (5) US7145025B2 (US20050075337A1-20050407-C00072.png)
EP (2) EP1532108B1 (US20050075337A1-20050407-C00072.png)
JP (2) JP4601549B2 (US20050075337A1-20050407-C00072.png)
KR (1) KR20050069975A (US20050075337A1-20050407-C00072.png)
CN (2) CN101845009B (US20050075337A1-20050407-C00072.png)
AU (1) AU2003256982A1 (US20050075337A1-20050407-C00072.png)
BR (1) BRPI0313155B8 (US20050075337A1-20050407-C00072.png)
CA (1) CA2495167C (US20050075337A1-20050407-C00072.png)
HK (2) HK1084380A1 (US20050075337A1-20050407-C00072.png)
MX (1) MXPA05001202A (US20050075337A1-20050407-C00072.png)
NO (1) NO20050619L (US20050075337A1-20050407-C00072.png)
WO (1) WO2004011423A2 (US20050075337A1-20050407-C00072.png)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050004235A1 (en) * 2002-07-29 2005-01-06 Lockwood Samuel Fournier Carotenoid analogs or derivatives for the inhibition and amelioration of liver disease
US20050009788A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling connexin 43 expression
US20050009758A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050009930A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling connexin 43 expression
US20050026874A1 (en) * 2002-07-29 2005-02-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of liver disease
US20050037995A1 (en) * 2002-07-29 2005-02-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050049248A1 (en) * 2002-07-29 2005-03-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for controlling C-reactive protein levels
US20050059659A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling C-reactive protein levels
US20050059635A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling C-reactive protein levels
US20050065096A1 (en) * 2002-07-29 2005-03-24 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050075316A1 (en) * 2002-07-29 2005-04-07 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ester analogs or derivatives for the inhibition and amelioration of disease
US20050090469A1 (en) * 2002-07-29 2005-04-28 Lockwood Samuel F. Carotenoid ester analogs or derivatives for the inhibition and amelioration of liver disease
US20060088904A1 (en) * 2004-10-01 2006-04-27 Lockwood Samuel F Methods for the synthesis of astaxanthin
US10071141B2 (en) 2015-05-08 2018-09-11 Spectral Platforms, Inc. Albumin-based non-covalent complexes and methods of use thereof
US11105747B2 (en) 2017-03-20 2021-08-31 Spectral Platforms, Inc. Spectroscopic methods to detect and characterize microorganisms

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070160645A1 (en) * 2001-10-25 2007-07-12 Jakob Vinten-Johansen PostConditioning System And Method For The Reduction Of Ischemic-Reperfusion Injury In The Heart And Other Organs
CA2463415C (en) 2001-10-25 2012-02-07 Emory University Catheter for modified perfusion
EA200401111A1 (ru) * 2002-02-25 2005-02-24 Диффьюжн Фармасьютикалз Ллс Биполярные соли транс-каротиноидов и их применение
US7759506B2 (en) * 2002-02-25 2010-07-20 Diffusion Pharmaceuticals Llc Bipolar trans carotenoid salts and their uses
EP1753708B1 (en) * 2004-01-20 2018-02-21 Brigham Young University Novel sirtuin activating compounds and methods for making the same
CA2564066A1 (en) * 2004-04-14 2005-11-03 Hawaii Biotech, Inc. Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation
US20060058269A1 (en) * 2004-04-14 2006-03-16 Lockwood Samuel F Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation
JP2007238441A (ja) * 2004-12-03 2007-09-20 Fuji Chem Ind Co Ltd アスタキサンチンを有効成分とする体脂肪減少用組成物
AU2005319144A1 (en) * 2004-12-22 2006-06-29 Emory University Therapeutic adjuncts to enhance the organ protective effects of postconditioning
US20060270589A1 (en) * 2005-02-22 2006-11-30 Lockwood Samuel F Carotenoids, carotenoid analogs, or carotenoid derivatives for the treatment of visual disabilities
EP1853544A4 (en) 2005-02-24 2010-06-16 Diffusion Pharmaceuticals Llc TRANSCAROTENOIDS AND THEIR SYNTHESIS, THEIR PREPARATION AND USES THEREOF
CA2606329A1 (en) * 2005-03-09 2006-09-21 Cardax Pharmaceuticals, Inc. Carotenoids, carotenoid analogs, or carotenoid derivatives for the treatment of proliferative disorders
UA94038C2 (ru) 2005-03-18 2011-04-11 Майкробиа, Инк. Продуцирование каротиноидов в маслянистых дрожжах и грибах
US20070015735A1 (en) * 2005-03-23 2007-01-18 Lockwood Samuel F Water-dispersible carotenoids, including analogs and derivatives
US20060270590A1 (en) * 2005-03-29 2006-11-30 Lockwood Samuel F Reduction in complement activation and inflammation during tissue injury by carotenoids, carotenoid analogs, or derivatives thereof
JP2006016407A (ja) * 2005-06-15 2006-01-19 Yamaha Motor Co Ltd ホスホジエステラーゼ阻害剤
JP2006016408A (ja) * 2005-06-23 2006-01-19 Yamaha Motor Co Ltd 血中中性脂肪抑制剤
US20070238793A1 (en) * 2005-12-07 2007-10-11 Lockwood Samuel F Structural carotenoid analogs or derivatives for the modulation of systemic and/or target organ redox status
JP2007153845A (ja) * 2005-12-07 2007-06-21 Yamaha Motor Co Ltd 脂肪蓄積抑制剤
US20070135521A1 (en) * 2005-12-14 2007-06-14 Yamaha Hatsudoki Kabushiki Kaisha Agent for Preventing Metabolic Syndrome
WO2007090095A2 (en) * 2006-01-27 2007-08-09 Cardax Pharmaceuticals, Inc. Synthesis of carotenoid analogs or derivatives with improved antioxidant characteristics
WO2007147163A2 (en) * 2006-06-16 2007-12-21 Cardax Pharmaceuticals, Inc. Compositions comprising carotenoid analogs or derivatives and methods for synthesis
WO2008017455A1 (en) * 2006-08-08 2008-02-14 Nestec S.A. Stable and bioavailable compositions of isomers of carotenoids for skin and hair
US8691555B2 (en) 2006-09-28 2014-04-08 Dsm Ip Assests B.V. Production of carotenoids in oleaginous yeast and fungi
ITMI20061874A1 (it) * 2006-09-29 2006-12-29 Rosario Ammirante Composizione per il trattamento degli stati patologici neurodegenerativi del sistema nervoso centrale ed in particolare nel morbo di parkinson
WO2008118862A1 (en) * 2007-03-23 2008-10-02 Cardax Pharmaceuticals, Inc. Carotenoid analogs and derivatives for the prevention of platelet aggregation
US8293804B2 (en) 2007-04-13 2012-10-23 Diffusion Pharmaceuticals Llc Use of bipolar trans carotenoids as a pretreatment and in the treatment of peripheral vascular disease
JP2011502125A (ja) * 2007-10-31 2011-01-20 ディフュージョン・ファーマシューティカルズ・エルエルシー 小分子拡散を促進する新しい種類の治療法
US20090297492A1 (en) * 2008-05-30 2009-12-03 Yamaha Hatsudoki Kabushiki Kaisha Method for Improving Cognitive Performance
WO2010120686A2 (en) * 2009-04-13 2010-10-21 Parsons J Kellogg Methods for the diagnosis and treatment of benign prostatic hyperplasia
CA2765697C (en) 2009-06-22 2019-11-12 Diffusion Pharmaceuticals Llc Diffusion enhancing compounds and their use alone or with thrombolytics
ES2593880T3 (es) * 2009-09-11 2016-12-13 Nestec, S.A. Composiciones y métodos para mejorar las funciones cognitivas y relacionadas en animales
US8541645B2 (en) * 2009-10-22 2013-09-24 University Of Calcutta Animal model for cigarette-smoke-induced atherosclerosis and related methods
CN103124498A (zh) 2010-06-02 2013-05-29 扩散药品有限公司 双极性反式类胡萝卜素的口服制剂
EP2600890A1 (en) * 2010-08-04 2013-06-12 Instituto Europeo di Oncologia S.r.l. Method of antigen loading for immunotherapy
CN102389407A (zh) * 2011-12-01 2012-03-28 滨州医学院 虾青素在制备治疗和预防肺纤维化的药品中的应用
GB201120772D0 (en) * 2011-12-02 2012-01-11 Ip Science Ltd Cocoa-based food products
WO2013082458A1 (en) 2011-12-02 2013-06-06 The Regents Of The University Of California Reperfusion protection solution and uses thereof
CN104780782A (zh) * 2012-07-09 2015-07-15 诺格罗有限责任公司 使用膳食补充剂预防酒精反应
CN103073471B (zh) * 2013-01-30 2014-11-05 江苏省农业科学院 一种叶黄素二琥珀酸酯的超声辅助合成方法
US9482675B1 (en) * 2013-07-31 2016-11-01 University Of Kentucky Research Foundation Methods and systems for prognosis and diagnosis of brain damage
JP6183746B2 (ja) * 2013-11-13 2017-08-23 国立大学法人高知大学 遅延性アレルギー抑制剤
AU2015262405B2 (en) 2014-05-20 2019-01-03 Asta Pharmaceuticals Co., Ltd. Carotenoid derivative, pharmaceutically acceptable salt thereof, and pharmaceutically acceptable ester or amide thereof
EP3207028B1 (en) * 2014-10-19 2020-05-27 Shenkar College of Engineering and Design Astaxanthin based polymer and uses thereof
US20160367620A1 (en) 2015-06-19 2016-12-22 Harry B. Demopoulos Glutathione
US9572783B1 (en) * 2015-10-08 2017-02-21 Chuen Wei Lu Use of xanthophylls for the treatment of cancers
CN105646869B (zh) * 2016-01-04 2018-01-16 中国海洋大学 一种水溶性虾青素衍生物及其制备方法
KR20230014850A (ko) * 2016-03-24 2023-01-30 디퓨젼 파마슈티컬즈 엘엘씨 암을 치료하기 위한, 화학 요법 및 방사선 요법과의 양극성 트랜스 카로티노이드의 용도
CN106748946A (zh) * 2017-02-14 2017-05-31 烟台固特丽生物科技股份有限公司 一种含水溶性虾青素作物营养液的制备方法
WO2019213538A1 (en) * 2018-05-03 2019-11-07 L.E.A.F. Holdings Group Llc Carotenoid compositions and uses thereof
JP2021176821A (ja) * 2018-08-01 2021-11-11 アスタファーマシューティカルズ株式会社 新規カロテノイド関連誘導体、その塩又はエステル類若しくはアミド類
CN109678771A (zh) * 2018-12-28 2019-04-26 广州立达尔生物科技股份有限公司 一种叶黄素甘氨酸酯及其盐酸盐的制备方法
JP2022549503A (ja) * 2019-09-27 2022-11-25 カーダックス、インコーポレイティッド アスタキサンチンエステルおよびその使用方法
FR3105790B1 (fr) * 2019-12-26 2022-01-14 Biophytis Composés chimiques ciblant l’œil et leur utilisation dans le traitement de maladies oculaires
CN111257466B (zh) * 2020-02-27 2022-04-01 南宁海关技术中心 一种红心鸭蛋中类胡萝卜素含量的测定方法
CN113788867B (zh) * 2021-06-30 2023-12-26 东北林业大学 一种叶黄素水溶性衍生物及其制备工艺

Citations (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3755422A (en) * 1970-01-15 1973-08-28 Rhone Poulenc Sa Preparation of carotenoid compounds
US3965261A (en) * 1975-04-29 1976-06-22 University Of Virginia Method for treating papillomas
US3975519A (en) * 1975-06-09 1976-08-17 University Of Virginia Method for increasing the oxygen partial pressure in the bloodstream of mammals
US4009270A (en) * 1975-11-21 1977-02-22 The University Of Virginia Method for treating spinal cord injury
US4038144A (en) * 1976-04-19 1977-07-26 The University Of Virginia Method of increasing fermentation yields
US4046880A (en) * 1976-04-20 1977-09-06 The University Of Virginia Method of treating hypertension
US4070460A (en) * 1975-11-10 1978-01-24 University Of Virginia Patents Foundation Method for treating cerebral edema
US4176179A (en) * 1978-04-17 1979-11-27 The University Of Virginia Alumni Patents Foundation Method for treating arthritis
US4245109A (en) * 1978-06-02 1981-01-13 Hoffmann-La Roche Inc. Process for producing astaxanthin
US4283559A (en) * 1978-06-02 1981-08-11 Hoffmann-La Roche Inc. Process for the manufacture of cyclohexene derivatives
US4304784A (en) * 1978-03-31 1981-12-08 Takeda Chemical Industries, Ltd. Composition and method for inhibiting platelet aggregation
US4435427A (en) * 1980-12-19 1984-03-06 Basf Aktiengesellschaft Stable injectable β-carotene micellar solutions and their preparation
US4491574A (en) * 1983-03-02 1985-01-01 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Reduction of high dose aspirin toxicity by dietary vitamin A
US4585885A (en) * 1982-08-20 1986-04-29 Hoffmann-La Roche Inc. Cyclohexenone derivatives and process for making same
US4952716A (en) * 1987-03-27 1990-08-28 Hoffmann-La Roche Inc. Ethynylcyclohexene compounds
US5057494A (en) * 1988-08-03 1991-10-15 Ethicon, Inc. Method for preventing tissue damage after an ischemic episode
US5153001A (en) * 1984-01-28 1992-10-06 Roshdy Ismail Agent for treating heart disease
US5221668A (en) * 1992-02-26 1993-06-22 Abbott Laboratories Nutritional product for trauma and surgery patients
US5278189A (en) * 1990-06-04 1994-01-11 Rath Matthias W Prevention and treatment of occlusive cardiovascular disease with ascorbate and substances that inhibit the binding of lipoprotein (A)
US5310554A (en) * 1992-10-27 1994-05-10 Natural Carotene Corporation High purity beta-carotene
US5326757A (en) * 1992-01-06 1994-07-05 Health Maintenance Programs, Inc. Pharmaceutically active antioxidant containing composition and the method of its use to prevent and treat restenosis following angioplasty
US5328845A (en) * 1992-03-27 1994-07-12 Universal Foods Corporation Fungal negative microorganism capable of producing high levels of beta-carotene
US5346488A (en) * 1985-04-08 1994-09-13 The General Hospital Corporation Laser-induced ablation of atherosclerotic plaque
US5364563A (en) * 1989-07-25 1994-11-15 Hoffmann-La Roche Inc. Powdered aqueous carotenoid dispersions
US5422247A (en) * 1992-03-27 1995-06-06 Universal Foods Corporation Blakeslea trispora mated culture capable of increased beta-carotene production
US5455362A (en) * 1993-07-05 1995-10-03 Basf Aktiengesellschaft Preparation of astaxanthin novel intermediates therefor and the preparation thereof
US5457135A (en) * 1992-05-08 1995-10-10 Baranowitz; Steven Treatment of age related macular degeneration with beta-carotene
US5492701A (en) * 1993-07-08 1996-02-20 Rhone-Poulenc Nutrition Animale Process for the preparation of spherules
US5536504A (en) * 1993-11-19 1996-07-16 Marigen S.A. Ultramicroemulsions from spontaneously dispersible concentrates containing xanthophyll esters and having antitumor activity
US5589468A (en) * 1995-01-13 1996-12-31 Clintec Nutrition Co. Method for providing nutrition to elderly patients
US5607839A (en) * 1993-07-22 1997-03-04 Nippon Oil Company, Ltd. Bacteria belonging to new genus process for production of carotenoids using same
US5607707A (en) * 1992-09-15 1997-03-04 Smithkline Beecham P.L.C. Compositions
US5612485A (en) * 1992-06-04 1997-03-18 Betatene Ltd Of Cheltenham High cis beta-carotene composition
US5643943A (en) * 1994-12-23 1997-07-01 Alcon Laboratories, Inc. Systemic administration of esters and amides of antioxidants which may be used as antioxidant prodrug therapy for oxidative and inflammatory pathogenesis
US5668183A (en) * 1992-04-14 1997-09-16 Roche Vitamins Inc. Water dispersible compositions
US5705180A (en) * 1993-03-22 1998-01-06 Betatene Limited Therapeutic agent for the treatment of melanomas
US5744502A (en) * 1994-09-19 1998-04-28 Astacarotene Ab Method for increasing the production of/in breeding and production animals in the poultry industry
US5773026A (en) * 1993-03-22 1998-06-30 Betatene Limited Aqueous formulations of water-insoluble therapeutic agent comprising carotenoids and/or tocopherols
US5801159A (en) * 1996-02-23 1998-09-01 Galileo Laboratories, Inc. Method and composition for inhibiting cellular irreversible changes due to stress
US5811446A (en) * 1997-04-18 1998-09-22 Cytos Pharmaceuticals Llc Prophylactic and therapeutic methods for ocular degenerative diseases and inflammations and histidine compositions therefor
US5837224A (en) * 1996-01-19 1998-11-17 The Regents Of The University Of Michigan Method of inhibiting photoaging of skin
US5849345A (en) * 1994-12-21 1998-12-15 Roche Vitamins Inc. Carotenoid ketones and esters
US5854015A (en) * 1995-10-31 1998-12-29 Applied Food Biotechnology, Inc. Method of making pure 3R-3'R stereoisomer of zeaxanthin for human ingestion
US5858700A (en) * 1997-04-03 1999-01-12 Kemin Foods, Lc Process for the isolation and purification of lycopene crystals
US5863953A (en) * 1996-11-27 1999-01-26 Basf Aktiengesellschaft Liquid, oil-miscible carotenoid preparations
US5871766A (en) * 1990-10-01 1999-02-16 Brigham And Women's Hospital Beta-carotene vitamin E therapy for inhibition of major vascular events
US5876782A (en) * 1997-05-14 1999-03-02 Kemin Industries, Inc. Method for the conversion of xanthophylls in plant material
US5886053A (en) * 1995-02-03 1999-03-23 Basf Aktiengesellschaft Use of carotenoids for producing drugs for the treatment of dermatoses
US5891907A (en) * 1996-03-11 1999-04-06 Basf Aktiengesellschaft Stable aqueous solubilizates of carotenoids and vitamins
US5895659A (en) * 1993-03-11 1999-04-20 Basf Akteingesellschaft Finely dispersed carotenoid and retinoid suspension and their preparation
US5925684A (en) * 1996-03-11 1999-07-20 Basf Aktiengesellschaft Stable carotenoid emulsions suitable for parenteral administration
US5959138A (en) * 1997-11-25 1999-09-28 Industrial Organica S.A. De C.V. Short chain diesters and process for making the same
US5968251A (en) * 1996-09-09 1999-10-19 Basf Aktiengesellschaft Production of carotenoid preparations in the form of coldwater-dispersible powders, and the use of the novel carotenoid preparations
US5976575A (en) * 1995-12-28 1999-11-02 Amway Corporation Dry carotenoid-oil powder and process for making same
US6020003A (en) * 1998-02-23 2000-02-01 Basf Corporation Method of making spray-dried powders with high edible-oil loadings based on non-hydrolyzed gelatin
US6040147A (en) * 1997-04-02 2000-03-21 The Brigham And Women's Hospital, Inc. Systemic inflammatory markers as diagnostic tools in the prevention of atherosclerotic diseases and as tools to aid in the selection of agents to be used for the prevention and treatment of atherosclerotic disease
US6043259A (en) * 1998-07-09 2000-03-28 Medicure Inc. Treatment of cardiovascular and related pathologies
US6046181A (en) * 1995-10-17 2000-04-04 Showa Denko K.K. Highly purified tocopheryl phosphate, process for producing the same, analytical method therefor and cosmetic
US6051587A (en) * 1998-04-16 2000-04-18 Medicure, Inc. Treatment of iatrogenic and age-related hypertension and pharmaceutical compositions useful therein
US6054491A (en) * 1996-03-27 2000-04-25 Astacarotene Ab Agent for increasing the production of/in breeding and production mammals
US6060511A (en) * 1995-10-05 2000-05-09 Gainer; John L. Trans-sodium crocetinate, methods of making and methods of use thereof
US6075058A (en) * 1998-12-12 2000-06-13 Tufts University Compositions for increased bioavailability of carotenoids
US6083520A (en) * 1994-05-19 2000-07-04 Ewos Aktiebolag Bioactive feed
US6093348A (en) * 1996-05-14 2000-07-25 Roche Vitamins Inc. Process for manufacture of carotenoid compositions
US6132790A (en) * 1991-09-06 2000-10-17 Betatene Limited Carotenoid composition
US6218436B1 (en) * 1993-06-28 2001-04-17 The Howard Foundation Pharmaceutically active carotenoids
US6232060B1 (en) * 1996-01-19 2001-05-15 Galileo Laboratories, Inc. Assay system for anti-stress agents
US6245818B1 (en) * 1997-09-04 2001-06-12 Astacarotene Ab Medicament for improvement of duration of muscle function or treatment of muscle disorders or diseases
US6258855B1 (en) * 2000-02-08 2001-07-10 Cyanotech Corporation Method of retarding and ameliorating carpal tunnel syndrome
US6265450B1 (en) * 1995-10-26 2001-07-24 Suntory Limited Anti-stress composition
US6313169B1 (en) * 1997-04-04 2001-11-06 Phyllis E. Bowen Lutein esters having high bioavailability
US6331537B1 (en) * 1998-06-03 2001-12-18 Gpi Nil Holdings, Inc. Carboxylic acids and carboxylic acid isosteres of N-heterocyclic compounds
US6335015B1 (en) * 1997-12-16 2002-01-01 Astacarotene Ab Method of the prophylactic treatment of mastitis
US6344214B1 (en) * 1999-12-13 2002-02-05 Cyanotech Corporation Method for retarding and ameliorating fever blisters and canker sores
US6399105B1 (en) * 1999-01-20 2002-06-04 Peter Donald Collin Sea cucumber carotenoid lipid fraction products and methods of use
US6428816B1 (en) * 1994-04-08 2002-08-06 Cognis Australia Pty., Ltd. Carotenoid agent for inhibiting the conversion of epithelial cells to tumors
US6540654B2 (en) * 2001-02-23 2003-04-01 Luis W. Levy Carotenoid esters
US6541519B2 (en) * 2000-04-06 2003-04-01 Coastside Bio Resources Methods and compositions for treating lipoxygenase-mediated disease states
US20040162329A1 (en) * 2002-07-29 2004-08-19 Lockwood Samuel Fournier Structural carotenoid analogs for the inhibition and amelioration of disease
US20050004235A1 (en) * 2002-07-29 2005-01-06 Lockwood Samuel Fournier Carotenoid analogs or derivatives for the inhibition and amelioration of liver disease
US20050009930A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling connexin 43 expression
US20050009758A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050009788A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling connexin 43 expression
US20050026874A1 (en) * 2002-07-29 2005-02-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of liver disease
US20050049248A1 (en) * 2002-07-29 2005-03-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for controlling C-reactive protein levels
US20050059659A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling C-reactive protein levels
US20050059635A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling C-reactive protein levels
US20050065096A1 (en) * 2002-07-29 2005-03-24 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050075316A1 (en) * 2002-07-29 2005-04-07 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ester analogs or derivatives for the inhibition and amelioration of disease
US20050090469A1 (en) * 2002-07-29 2005-04-28 Lockwood Samuel F. Carotenoid ester analogs or derivatives for the inhibition and amelioration of liver disease
US20050113372A1 (en) * 2002-07-29 2005-05-26 Lockwood Samuel F. Carotenoid analogs or derivatives for the inhibition and amelioration of disease
US20050143475A1 (en) * 2002-07-29 2005-06-30 Lockwood Samuel F. Carotenoid analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050148517A1 (en) * 2002-07-29 2005-07-07 Lockwood Samuel F. Carotenoid ether analogs or derivatives for controlling connexin 43 expression
US20050261254A1 (en) * 2004-04-14 2005-11-24 Lockwood Samuel F Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation
US20060058269A1 (en) * 2004-04-14 2006-03-16 Lockwood Samuel F Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation

Family Cites Families (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH314001A (de) * 1952-12-18 1956-05-31 Hoffmann La Roche Verfahren zur Herstellung von Vitamin-A-Estern
CH420822A (de) * 1960-10-17 1966-09-15 Hoffmann La Roche Wasserdispergierbare Carotinoidzubereitung
US3354218A (en) 1963-05-10 1967-11-21 Hoffmann La Roche Process for preparing 4-(2, 6, 6-trimethyl-4-methoxy-1-cyclohexen-1-yl)-3-buten-2-one
US3788468A (en) * 1973-05-01 1974-01-29 Univ Virginia Process for increasing oxygen diffusivity
US3853993A (en) * 1973-05-01 1974-12-10 Univ Virginia Process for increasing oxygen diffusivity and method for treating atherosclerosis
US3989757A (en) 1973-08-29 1976-11-02 Hoffmann-La Roche Inc. Isomerizing cis-carotenoids to all-trans-carotenoids
CH609320A5 (US20050075337A1-20050407-C00072.png) * 1974-06-20 1979-02-28 Hoffmann La Roche
CH623304A5 (US20050075337A1-20050407-C00072.png) * 1975-11-30 1981-05-29 Hoffmann La Roche
EP0077439B1 (de) * 1981-10-16 1986-09-24 F. HOFFMANN-LA ROCHE & CO. Aktiengesellschaft Verfahren zur Herstellung von Cyclohexenderivaten, sowie ein neues Ausgangsprodukt und neue Zwischenprodukte in diesem Verfahren
ATE13666T1 (de) * 1982-02-09 1985-06-15 Hoffmann La Roche Verfahren zur herstellung von cyclohexenderivaten.
US4851339A (en) * 1986-04-01 1989-07-25 Hills Christopher B Extraction of anti-mutagenic pigments from algae and vegetables
DE69424719T2 (de) * 1993-06-24 2000-11-02 Hoffmann La Roche Pigmentierung mit Carotinoiden
JP3249867B2 (ja) * 1993-10-21 2002-01-21 株式会社クラレ アスタキサンチンの製造方法
JPH0873312A (ja) * 1994-09-02 1996-03-19 Noevir Co Ltd 皮膚外用剤
US5527533A (en) 1994-10-27 1996-06-18 Board Of Trustees Of The University Of Illinois Method of retarding and ameliorating central nervous system and eye damage
US5532009A (en) * 1995-06-07 1996-07-02 The Procter & Gamble Company Fat substitutes containing water soluble beta-carotene
JPH08337592A (ja) * 1995-06-13 1996-12-24 Kaiyo Bio Technol Kenkyusho:Kk 新規カロテノイド
JPH0984591A (ja) * 1995-09-26 1997-03-31 Kaiyo Bio Technol Kenkyusho:Kk カロテノイド硫酸エステルおよびその製造方法
JPH09202730A (ja) * 1996-01-24 1997-08-05 Nippon Mektron Ltd 発ガン抑制作用剤
WO1998029111A1 (fr) * 1996-12-25 1998-07-09 Designer Foods Association Ltd. Inhibiteurs de la carcinogenese
JPH10327865A (ja) * 1997-05-29 1998-12-15 Kirin Brewery Co Ltd カロテノイド配糖体およびその製造法
US6008417A (en) 1997-10-20 1999-12-28 Roche Vitamins Inc. Process for making metabolites of lycopene
DE19802134A1 (de) * 1998-01-21 1999-07-22 Basf Ag Verwendung von Carotinoid-Aggregaten als Färbemittel
NL1010351C2 (nl) * 1998-10-19 2001-01-08 Werklust & Beheer B V Esters van caroteno´den voor gebruik in de preventie en behandeling van oogaandoeningen.
JP4491090B2 (ja) * 1999-10-08 2010-06-30 ヒガシマル醤油株式会社 アポトーシス誘導剤
US6426362B1 (en) 1999-10-08 2002-07-30 Galileo Laboratories, Inc. Formulations of tocopherols and methods of making and using them
EP1238068A1 (en) 1999-12-08 2002-09-11 California Institute Of Technology Directed evolution of biosynthetic and biodegration pathways
US20030104090A1 (en) 2000-05-05 2003-06-05 Levy Pedro E. Supplements containing annatto extracts and carotenoids and methods for using the same
US6579544B1 (en) 2000-05-31 2003-06-17 Nutriex, L.L.C. Method for supplementing the diet
US20020110604A1 (en) 2000-08-11 2002-08-15 Ashni Naturaceuticals, Inc. Composition exhibiting synergistic antioxidant activity
JP2004515508A (ja) 2000-12-16 2004-05-27 アベンティス・ファーマ・ドイチユラント・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング 化合物の健康促進組成物
US6984523B2 (en) 2001-08-02 2006-01-10 E.I. Du Pont De Nemours And Company Carotenoid ketolase gene
GB0119052D0 (en) 2001-08-03 2001-09-26 Mars Uk Ltd Foodstuff
MX247604B (es) * 2002-02-06 2007-07-30 Dsm Ip Assets Bv Esteres de astaxantina.
US7514107B2 (en) 2002-03-21 2009-04-07 Mars, Incorporated Treatment of diseases involving defective gap junctional communication
US20050228188A1 (en) * 2002-04-30 2005-10-13 Suntory Limited Astaxanthin medium-chain fatty acid ester, process for producing the same and composition containing the ester
WO2006039685A2 (en) * 2004-10-01 2006-04-13 Hawaii Biotech, Inc. Methods for synthesis of chiral intermediates of carotenoids, carotenoid analogs, and carotenoid derivatives

Patent Citations (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3755422A (en) * 1970-01-15 1973-08-28 Rhone Poulenc Sa Preparation of carotenoid compounds
US3965261A (en) * 1975-04-29 1976-06-22 University Of Virginia Method for treating papillomas
US3975519A (en) * 1975-06-09 1976-08-17 University Of Virginia Method for increasing the oxygen partial pressure in the bloodstream of mammals
US4070460A (en) * 1975-11-10 1978-01-24 University Of Virginia Patents Foundation Method for treating cerebral edema
US4009270A (en) * 1975-11-21 1977-02-22 The University Of Virginia Method for treating spinal cord injury
US4038144A (en) * 1976-04-19 1977-07-26 The University Of Virginia Method of increasing fermentation yields
US4046880A (en) * 1976-04-20 1977-09-06 The University Of Virginia Method of treating hypertension
US4304784A (en) * 1978-03-31 1981-12-08 Takeda Chemical Industries, Ltd. Composition and method for inhibiting platelet aggregation
US4176179A (en) * 1978-04-17 1979-11-27 The University Of Virginia Alumni Patents Foundation Method for treating arthritis
US4245109A (en) * 1978-06-02 1981-01-13 Hoffmann-La Roche Inc. Process for producing astaxanthin
US4283559A (en) * 1978-06-02 1981-08-11 Hoffmann-La Roche Inc. Process for the manufacture of cyclohexene derivatives
US4435427A (en) * 1980-12-19 1984-03-06 Basf Aktiengesellschaft Stable injectable β-carotene micellar solutions and their preparation
US4585885A (en) * 1982-08-20 1986-04-29 Hoffmann-La Roche Inc. Cyclohexenone derivatives and process for making same
US4491574A (en) * 1983-03-02 1985-01-01 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Reduction of high dose aspirin toxicity by dietary vitamin A
US5153001A (en) * 1984-01-28 1992-10-06 Roshdy Ismail Agent for treating heart disease
US5346488A (en) * 1985-04-08 1994-09-13 The General Hospital Corporation Laser-induced ablation of atherosclerotic plaque
US4952716A (en) * 1987-03-27 1990-08-28 Hoffmann-La Roche Inc. Ethynylcyclohexene compounds
US5057494A (en) * 1988-08-03 1991-10-15 Ethicon, Inc. Method for preventing tissue damage after an ischemic episode
US5364563A (en) * 1989-07-25 1994-11-15 Hoffmann-La Roche Inc. Powdered aqueous carotenoid dispersions
US5278189A (en) * 1990-06-04 1994-01-11 Rath Matthias W Prevention and treatment of occlusive cardiovascular disease with ascorbate and substances that inhibit the binding of lipoprotein (A)
US5871766A (en) * 1990-10-01 1999-02-16 Brigham And Women's Hospital Beta-carotene vitamin E therapy for inhibition of major vascular events
US6132790A (en) * 1991-09-06 2000-10-17 Betatene Limited Carotenoid composition
US5326757A (en) * 1992-01-06 1994-07-05 Health Maintenance Programs, Inc. Pharmaceutically active antioxidant containing composition and the method of its use to prevent and treat restenosis following angioplasty
US5221668A (en) * 1992-02-26 1993-06-22 Abbott Laboratories Nutritional product for trauma and surgery patients
US5328845A (en) * 1992-03-27 1994-07-12 Universal Foods Corporation Fungal negative microorganism capable of producing high levels of beta-carotene
US5422247A (en) * 1992-03-27 1995-06-06 Universal Foods Corporation Blakeslea trispora mated culture capable of increased beta-carotene production
US5668183A (en) * 1992-04-14 1997-09-16 Roche Vitamins Inc. Water dispersible compositions
US5457135A (en) * 1992-05-08 1995-10-10 Baranowitz; Steven Treatment of age related macular degeneration with beta-carotene
US5612485A (en) * 1992-06-04 1997-03-18 Betatene Ltd Of Cheltenham High cis beta-carotene composition
US5607707A (en) * 1992-09-15 1997-03-04 Smithkline Beecham P.L.C. Compositions
US5310554A (en) * 1992-10-27 1994-05-10 Natural Carotene Corporation High purity beta-carotene
US5895659A (en) * 1993-03-11 1999-04-20 Basf Akteingesellschaft Finely dispersed carotenoid and retinoid suspension and their preparation
US5773026A (en) * 1993-03-22 1998-06-30 Betatene Limited Aqueous formulations of water-insoluble therapeutic agent comprising carotenoids and/or tocopherols
US5897871A (en) * 1993-03-22 1999-04-27 Betatine Limited Therapeutic agent for the treatment of melanomas
US5705180A (en) * 1993-03-22 1998-01-06 Betatene Limited Therapeutic agent for the treatment of melanomas
US6218436B1 (en) * 1993-06-28 2001-04-17 The Howard Foundation Pharmaceutically active carotenoids
US5455362A (en) * 1993-07-05 1995-10-03 Basf Aktiengesellschaft Preparation of astaxanthin novel intermediates therefor and the preparation thereof
US5492701A (en) * 1993-07-08 1996-02-20 Rhone-Poulenc Nutrition Animale Process for the preparation of spherules
US5607839A (en) * 1993-07-22 1997-03-04 Nippon Oil Company, Ltd. Bacteria belonging to new genus process for production of carotenoids using same
US5536504A (en) * 1993-11-19 1996-07-16 Marigen S.A. Ultramicroemulsions from spontaneously dispersible concentrates containing xanthophyll esters and having antitumor activity
US6428816B1 (en) * 1994-04-08 2002-08-06 Cognis Australia Pty., Ltd. Carotenoid agent for inhibiting the conversion of epithelial cells to tumors
US6083520A (en) * 1994-05-19 2000-07-04 Ewos Aktiebolag Bioactive feed
US5744502A (en) * 1994-09-19 1998-04-28 Astacarotene Ab Method for increasing the production of/in breeding and production animals in the poultry industry
US5849345A (en) * 1994-12-21 1998-12-15 Roche Vitamins Inc. Carotenoid ketones and esters
US5643943A (en) * 1994-12-23 1997-07-01 Alcon Laboratories, Inc. Systemic administration of esters and amides of antioxidants which may be used as antioxidant prodrug therapy for oxidative and inflammatory pathogenesis
US5589468A (en) * 1995-01-13 1996-12-31 Clintec Nutrition Co. Method for providing nutrition to elderly patients
US5886053A (en) * 1995-02-03 1999-03-23 Basf Aktiengesellschaft Use of carotenoids for producing drugs for the treatment of dermatoses
US6060511A (en) * 1995-10-05 2000-05-09 Gainer; John L. Trans-sodium crocetinate, methods of making and methods of use thereof
US6046181A (en) * 1995-10-17 2000-04-04 Showa Denko K.K. Highly purified tocopheryl phosphate, process for producing the same, analytical method therefor and cosmetic
US6265450B1 (en) * 1995-10-26 2001-07-24 Suntory Limited Anti-stress composition
US5854015A (en) * 1995-10-31 1998-12-29 Applied Food Biotechnology, Inc. Method of making pure 3R-3'R stereoisomer of zeaxanthin for human ingestion
US5976575A (en) * 1995-12-28 1999-11-02 Amway Corporation Dry carotenoid-oil powder and process for making same
US5837224A (en) * 1996-01-19 1998-11-17 The Regents Of The University Of Michigan Method of inhibiting photoaging of skin
US6232060B1 (en) * 1996-01-19 2001-05-15 Galileo Laboratories, Inc. Assay system for anti-stress agents
US5801159A (en) * 1996-02-23 1998-09-01 Galileo Laboratories, Inc. Method and composition for inhibiting cellular irreversible changes due to stress
US5925684A (en) * 1996-03-11 1999-07-20 Basf Aktiengesellschaft Stable carotenoid emulsions suitable for parenteral administration
US5891907A (en) * 1996-03-11 1999-04-06 Basf Aktiengesellschaft Stable aqueous solubilizates of carotenoids and vitamins
US6054491A (en) * 1996-03-27 2000-04-25 Astacarotene Ab Agent for increasing the production of/in breeding and production mammals
US6093348A (en) * 1996-05-14 2000-07-25 Roche Vitamins Inc. Process for manufacture of carotenoid compositions
US5968251A (en) * 1996-09-09 1999-10-19 Basf Aktiengesellschaft Production of carotenoid preparations in the form of coldwater-dispersible powders, and the use of the novel carotenoid preparations
US5863953A (en) * 1996-11-27 1999-01-26 Basf Aktiengesellschaft Liquid, oil-miscible carotenoid preparations
US6040147A (en) * 1997-04-02 2000-03-21 The Brigham And Women's Hospital, Inc. Systemic inflammatory markers as diagnostic tools in the prevention of atherosclerotic diseases and as tools to aid in the selection of agents to be used for the prevention and treatment of atherosclerotic disease
US5858700A (en) * 1997-04-03 1999-01-12 Kemin Foods, Lc Process for the isolation and purification of lycopene crystals
US6313169B1 (en) * 1997-04-04 2001-11-06 Phyllis E. Bowen Lutein esters having high bioavailability
US5811446A (en) * 1997-04-18 1998-09-22 Cytos Pharmaceuticals Llc Prophylactic and therapeutic methods for ocular degenerative diseases and inflammations and histidine compositions therefor
US5876782A (en) * 1997-05-14 1999-03-02 Kemin Industries, Inc. Method for the conversion of xanthophylls in plant material
US6245818B1 (en) * 1997-09-04 2001-06-12 Astacarotene Ab Medicament for improvement of duration of muscle function or treatment of muscle disorders or diseases
US5959138A (en) * 1997-11-25 1999-09-28 Industrial Organica S.A. De C.V. Short chain diesters and process for making the same
US6335015B1 (en) * 1997-12-16 2002-01-01 Astacarotene Ab Method of the prophylactic treatment of mastitis
US6020003A (en) * 1998-02-23 2000-02-01 Basf Corporation Method of making spray-dried powders with high edible-oil loadings based on non-hydrolyzed gelatin
US6051587A (en) * 1998-04-16 2000-04-18 Medicure, Inc. Treatment of iatrogenic and age-related hypertension and pharmaceutical compositions useful therein
US6331537B1 (en) * 1998-06-03 2001-12-18 Gpi Nil Holdings, Inc. Carboxylic acids and carboxylic acid isosteres of N-heterocyclic compounds
US6043259A (en) * 1998-07-09 2000-03-28 Medicure Inc. Treatment of cardiovascular and related pathologies
US6075058A (en) * 1998-12-12 2000-06-13 Tufts University Compositions for increased bioavailability of carotenoids
US6399105B1 (en) * 1999-01-20 2002-06-04 Peter Donald Collin Sea cucumber carotenoid lipid fraction products and methods of use
US6344214B1 (en) * 1999-12-13 2002-02-05 Cyanotech Corporation Method for retarding and ameliorating fever blisters and canker sores
US6258855B1 (en) * 2000-02-08 2001-07-10 Cyanotech Corporation Method of retarding and ameliorating carpal tunnel syndrome
US6541519B2 (en) * 2000-04-06 2003-04-01 Coastside Bio Resources Methods and compositions for treating lipoxygenase-mediated disease states
US6540654B2 (en) * 2001-02-23 2003-04-01 Luis W. Levy Carotenoid esters
US20050004235A1 (en) * 2002-07-29 2005-01-06 Lockwood Samuel Fournier Carotenoid analogs or derivatives for the inhibition and amelioration of liver disease
US20050059635A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling C-reactive protein levels
US20050009930A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling connexin 43 expression
US20050009758A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050009788A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling connexin 43 expression
US20050026874A1 (en) * 2002-07-29 2005-02-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of liver disease
US20050037995A1 (en) * 2002-07-29 2005-02-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050049248A1 (en) * 2002-07-29 2005-03-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for controlling C-reactive protein levels
US20050059659A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling C-reactive protein levels
US20040162329A1 (en) * 2002-07-29 2004-08-19 Lockwood Samuel Fournier Structural carotenoid analogs for the inhibition and amelioration of disease
US20050065096A1 (en) * 2002-07-29 2005-03-24 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050065097A1 (en) * 2002-07-29 2005-03-24 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050075316A1 (en) * 2002-07-29 2005-04-07 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ester analogs or derivatives for the inhibition and amelioration of disease
US20050090469A1 (en) * 2002-07-29 2005-04-28 Lockwood Samuel F. Carotenoid ester analogs or derivatives for the inhibition and amelioration of liver disease
US20050113372A1 (en) * 2002-07-29 2005-05-26 Lockwood Samuel F. Carotenoid analogs or derivatives for the inhibition and amelioration of disease
US20050143475A1 (en) * 2002-07-29 2005-06-30 Lockwood Samuel F. Carotenoid analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050148517A1 (en) * 2002-07-29 2005-07-07 Lockwood Samuel F. Carotenoid ether analogs or derivatives for controlling connexin 43 expression
US20050261254A1 (en) * 2004-04-14 2005-11-24 Lockwood Samuel F Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation
US20060058269A1 (en) * 2004-04-14 2006-03-16 Lockwood Samuel F Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7145025B2 (en) 2002-07-29 2006-12-05 Hawaii Biotech, Inc. Structural carotenoid analogs for the inhibition and amelioration of disease
US20050009758A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US7763649B2 (en) 2002-07-29 2010-07-27 Cardax Pharmaceuticals, Inc. Carotenoid analogs or derivatives for controlling connexin 43 expression
US20050009930A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling connexin 43 expression
US20050026874A1 (en) * 2002-07-29 2005-02-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of liver disease
US20050037995A1 (en) * 2002-07-29 2005-02-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050049248A1 (en) * 2002-07-29 2005-03-03 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for controlling C-reactive protein levels
US20050059659A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid analogs or derivatives for controlling C-reactive protein levels
US20050059635A1 (en) * 2002-07-29 2005-03-17 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling C-reactive protein levels
US20050065097A1 (en) * 2002-07-29 2005-03-24 Lockwood Samuel Fournier Carotenoid ether analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050065096A1 (en) * 2002-07-29 2005-03-24 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050075316A1 (en) * 2002-07-29 2005-04-07 Lockwood Samuel Fournier Pharmaceutical compositions including carotenoid ester analogs or derivatives for the inhibition and amelioration of disease
US20050090469A1 (en) * 2002-07-29 2005-04-28 Lockwood Samuel F. Carotenoid ester analogs or derivatives for the inhibition and amelioration of liver disease
US7723327B2 (en) 2002-07-29 2010-05-25 Cardax Pharmaceuticals, Inc. Carotenoid ester analogs or derivatives for the inhibition and amelioration of liver disease
US7592449B2 (en) 2002-07-29 2009-09-22 Cardax Pharmaceuticals, Inc. Structural carotenoid analogs for the inhibition and amelioration of disease
US20050009788A1 (en) * 2002-07-29 2005-01-13 Lockwood Samuel Fournier Carotenoid ester analogs or derivatives for controlling connexin 43 expression
US20050004235A1 (en) * 2002-07-29 2005-01-06 Lockwood Samuel Fournier Carotenoid analogs or derivatives for the inhibition and amelioration of liver disease
US7247752B2 (en) 2004-10-01 2007-07-24 Cardax Pharmaceuticals, Inc. Methods for the synthesis of astaxanthin
US20060178538A1 (en) * 2004-10-01 2006-08-10 Lockwood Samuel F Methods for the synthesis of chiral dihydroxy intermediates useful for the chiral synthesis of carotenoids
US20060183947A1 (en) * 2004-10-01 2006-08-17 Lockwood Samuel F Methods for the synthesis of astaxanthin
US20060183185A1 (en) * 2004-10-01 2006-08-17 Lockwood Samuel F Method for the synthesis of astaxanthin
US20060155150A1 (en) * 2004-10-01 2006-07-13 Lockwood Samuel F Methods for the synthesis of lutein
US20060167319A1 (en) * 2004-10-01 2006-07-27 Lockwood Samuel F Methods for the synthesis of unsaturated ketone intermediates useful for the synthesis of carotenoids
US20060111580A1 (en) * 2004-10-01 2006-05-25 Lockwood Samuel F Methods for the synthesis of chiral dihydroxy ketone intermediates useful for the chiral synthesis of carotenoids
US20060088904A1 (en) * 2004-10-01 2006-04-27 Lockwood Samuel F Methods for the synthesis of astaxanthin
US20060088905A1 (en) * 2004-10-01 2006-04-27 Lockwood Samuel F Methods for the synthesis of zeazanthin
US10071141B2 (en) 2015-05-08 2018-09-11 Spectral Platforms, Inc. Albumin-based non-covalent complexes and methods of use thereof
US10342855B2 (en) 2015-05-08 2019-07-09 Spectral Platforms, Inc. Albumin-based non-covalent complexes and methods of use thereof
US10940183B2 (en) 2015-05-08 2021-03-09 Spectral Platforms, Inc. Albumin-based non-covalent complexes and methods of use thereof
US11105747B2 (en) 2017-03-20 2021-08-31 Spectral Platforms, Inc. Spectroscopic methods to detect and characterize microorganisms

Also Published As

Publication number Publication date
CN1708480B (zh) 2010-12-15
AU2003256982A1 (en) 2004-02-16
JP2010248243A (ja) 2010-11-04
HK1084380A1 (en) 2006-07-28
EP1532108A2 (en) 2005-05-25
BRPI0313155B1 (pt) 2018-11-06
JP4601549B2 (ja) 2010-12-22
US20050037995A1 (en) 2005-02-17
CA2495167A1 (en) 2004-02-05
HK1148997A1 (en) 2011-09-23
EP1532108B1 (en) 2016-06-29
JP5187700B2 (ja) 2013-04-24
WO2004011423A2 (en) 2004-02-05
WO2004011423A3 (en) 2004-05-06
US20060229446A1 (en) 2006-10-12
US7145025B2 (en) 2006-12-05
CN101845009B (zh) 2012-10-03
BRPI0313155B8 (pt) 2021-05-25
BR0313155A (pt) 2005-07-12
NO20050619L (no) 2005-04-27
US7592449B2 (en) 2009-09-22
CA2495167C (en) 2018-08-21
KR20050069975A (ko) 2005-07-05
CN1708480A (zh) 2005-12-14
CN101845009A (zh) 2010-09-29
US20050065097A1 (en) 2005-03-24
US7317008B2 (en) 2008-01-08
EP2392562B1 (en) 2018-03-07
JP2006517197A (ja) 2006-07-20
US20040162329A1 (en) 2004-08-19
EP2392562A1 (en) 2011-12-07
MXPA05001202A (es) 2005-11-23

Similar Documents

Publication Publication Date Title
US7317008B2 (en) Carotenoid ester analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US7521584B2 (en) Carotenoid analogs or derivatives for the inhibition and amelioration of disease
US7723327B2 (en) Carotenoid ester analogs or derivatives for the inhibition and amelioration of liver disease
US7345091B2 (en) Carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050009788A1 (en) Carotenoid ester analogs or derivatives for controlling connexin 43 expression
US20050143475A1 (en) Carotenoid analogs or derivatives for the inhibition and amelioration of ischemic reperfusion injury
US20050148517A1 (en) Carotenoid ether analogs or derivatives for controlling connexin 43 expression
US20050026874A1 (en) Carotenoid ether analogs or derivatives for the inhibition and amelioration of liver disease
US7763649B2 (en) Carotenoid analogs or derivatives for controlling connexin 43 expression
US7375133B2 (en) Pharmaceutical compositions including carotenoid ether analogs or derivatives for the inhibition and amelioration of disease
US20050004235A1 (en) Carotenoid analogs or derivatives for the inhibition and amelioration of liver disease
US7320997B2 (en) Pharmaceutical compositions including carotenoid ester analogs or derivatives for the inhibition and amelioration of disease
US20050059659A1 (en) Carotenoid analogs or derivatives for controlling C-reactive protein levels
US7691901B2 (en) Carotenoid analogs or derivatives for the inhibition and amelioration of inflammation
US20050059635A1 (en) Carotenoid ester analogs or derivatives for controlling C-reactive protein levels
US20050049248A1 (en) Carotenoid ether analogs or derivatives for controlling C-reactive protein levels

Legal Events

Date Code Title Description
AS Assignment

Owner name: HAWAII BIOTECH, INC., HAWAII

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LOCKWOOD, SAMUEL FOURNIER;O'MALLEY, SEAN;WATUMULL, DAVID G.;AND OTHERS;REEL/FRAME:015677/0989

Effective date: 20040802

AS Assignment

Owner name: CARDAX PHARMACEUTICALS, INC., HAWAII

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HAWAII BIOTECH, INC.;REEL/FRAME:018311/0413

Effective date: 20060808

STCB Information on status: application discontinuation

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