AU2007237157A1 - Peptides that ameliorate atherosclerosis - Google Patents

Peptides that ameliorate atherosclerosis Download PDF

Info

Publication number
AU2007237157A1
AU2007237157A1 AU2007237157A AU2007237157A AU2007237157A1 AU 2007237157 A1 AU2007237157 A1 AU 2007237157A1 AU 2007237157 A AU2007237157 A AU 2007237157A AU 2007237157 A AU2007237157 A AU 2007237157A AU 2007237157 A1 AU2007237157 A1 AU 2007237157A1
Authority
AU
Australia
Prior art keywords
seq
peptide
peptides
group
ldl
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.)
Granted
Application number
AU2007237157A
Other versions
AU2007237157B2 (en
Inventor
Gattadahalli M. Anantharamaiah
Alan M Fogelman
Mohamad Navab
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.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/645,454 external-priority patent/US6664230B1/en
Application filed by University of California filed Critical University of California
Priority to AU2007237157A priority Critical patent/AU2007237157B2/en
Publication of AU2007237157A1 publication Critical patent/AU2007237157A1/en
Application granted granted Critical
Publication of AU2007237157B2 publication Critical patent/AU2007237157B2/en
Priority to AU2009202705A priority patent/AU2009202705C1/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

PEPTIDES THAT AMELIORATE ATHEROSCLEROSIS 4 FIELD OF THE INVENTION c- [0003] This invention relates to the field of atherosclerosis. In particular, this invention pertains to the identification of a class of peptides that t are non-orally or orally administrable and that ameliorate one or more Ssymptoms of atherosclerosis.
DEFINITION
c 10 [0003a] In the specification the term "comprising" shall be understood to have a broad meaning similar to the term "including" and will be understood to imply the inclusion of a stated integer or step or group of integers-or steps-~dt not the exclusion of any other integer or step or group of integers or steps.
This definition also applies to variations on the term "comprising" such as "comprise" and "comprises".
BACKGROUND OF THE INVENTION [0004] Cardiovascular disease is a leading cause of morbidity and mortality, particularly in the United States and in Western European countries.
Several causative factors are implicated in the development of cardiovascular disease including heritary predisposition to the disease, gender; lifestyle factors such as-smoking and diet, age, hypertension, and hyperlipidemia, including hypercholesterolemia. Several of these factors, particularly.
hyperlipidemia and hypercholesteremia (high blood cholesterol concentrations) provide a significant risk factor associated with atherosclerosis.
[0005] Cholesterol is present in the blood as free and esterified cholesterol within lipoprotein particles, commonly known as chylomicrons, very low density lipoproteins (VLDLs), low density lipoproteins (LDLs), and high density lipoproteins (HDLs). Concentration of total cholesterol in the blood is influenced by absorption of cholesterol from the digestive tract, (2) synthesis of cholesterol from dietary constituents 0 such as carbohydrates, proteins, fats and ethanol, and removal of cholesterol from CN blood by tissues, especially the liver, and subsequent conversion of the cholesterol to bile acids, steroid hormones, and biliary cholesterol.
,D [0006] Maintenance of blood cholesterol concentrations is influenced by both genetic and environmental factors. Genetic factors include concentration of rate-limiting enzymes in cholesterol biosynthesis, concentration of receptors for low density lipoproteins in the liver, concentration of rate-limiting enzymes for conversion of cholesterols bile acids, rates of synthesis and secretion of lipoproteins and gender of person. Environmental factors influencing the hemostasis of blood cholesterol concentration in humans include dietary composition, incidence of smoking, physical activity, and use of a variety of pharmaceutical agents. Dietary variables include amount and type of fat (saturated and polyunsaturated fatty acids), amount of cholesterol, amount and type of fiber, and perhaps amounts of vitamins such as vitamin C and D and minerals such as calcium.
[0007] Epidemiological studies show an inverse correlation of high density lipoprotein (HDL) and apolipoprotein (apo) A-I levels with the occurrence of atherosclerotic events (Wilson et al. (1988) Arteriosclerosis 8: 737-741). Injection of HDL into rabbits fed an atherogenic diet has been shown to inhibit atherosclerotic lesion formation (Badimon et al. (1990) J. 'Clin. Invest. 85: 1234-1241).
[0008] Human apo A-I has been a subject of intense study because of its antiatherogenic properties. Exchangeable apolipoproteins, including apo A-I, possess lipidassociating domains (Brouillette and Anantharamaiah (1995) Biochim. Biophys. Acta 1256:103-129; Segrest et al. (1974) FEBSLett. 38: :247-253). Apo A-I has been postulated to possess eight tandem repeating 22mer sequences, most of which have the potential to form class A amphipathic helical structures (Segrest et al. (1974) FEBSLett.
38: :247-253). Characteristics of the class A amphipathic helix include the presence of positively charged residues at the polar-nonpolar interface and negatively charged residues at the center of the polar face (Segrest et al. (1974) FEBS Lett. 38: 247-253; Segrest et al. (1990) Proteins: Structure, Function, and Genetics 8: 103-117). Apo A-I has been shown to strongly associate with phospholipids to form complexes and to promote cholesterol efflux from cholesterol-enriched cells. The delivery and maintenance of serum 3 O levels of apo A-I to effectively mitigate one or more symptoms of atherosclerosis has heretofore proven elusive.
0 IN SUMMARY OF THE INVENTION [0009] This invention provides novel peptides, the administration of which mitigate one or more symptoms of atherosclerosis. In particular, it was the discovery of one part of this invention that peptides comprising a class A mC amphipathic helix when formulated with amino acid residue(s) and/or r- having protected amino and carboxyl termini can be orally administered to an O 10 organism, are readily taken up and delivered to the serum, and are effective to mitigate one or more symptoms of atherosclerosis. It was a discovery of another part of the invention, that forms of these peptides may be administered by a non-oral route such as by injection may also mitigate one or more symptoms of atherosclerosis.
[0010] Thus, in one embodiment, this invention provides a peptide that ameliorates a symptom of atherosclerosis, where the peptide ranges in length from about 10 to about 30 amino acids, comprises at least one class A amphipathic helix, comprises at least one amino acid residue or at least one amino acid residue, protects a phospholipid against oxidation by an oxidizing agent, and is not the D-18A peptide D-W-L-K-A-F-Y-D-K-V-A-E- K-L-K-E-A-F(SEQ ID No: 1) having all form amino acid residues).
[0010a] According to the first broad form of the invention there is provided a peptide that ameliorates a symptom of atherosclerosis, wherein said peptide has the amino acid sequence selected from the group consisting of: D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO D-W-F-K-A-F-Y- D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO D-W-L-K-A-F-Y-D-K-V A-E-K-F-K- E-A-F- (SEQ ID NO D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO D-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO D-W-L-K- A-F-Y-D-K-F-F E-K-F-K-E-F-F- (SEQ ID NO: D-W-F-K-A-F-Y-D-K-F-F-E- K-F-K-E-F-F- (SEQ ID NO (SEQ ID NO:9),D-W-L-K-A-F-Y-D-K-V-F-E-K- F-K-E-A-F- (SEQ ID NO 10), D-W-L- 3B K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ I D NO: 11), D-W-L-K-A-F-Y-D-K-V-A- E-K-F-K-E-F-F- (SEQ ID NO: 12), D-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- Z (SEQ ID NO: 13),E-W-L-K-L-F-Y-E-K-V-L-E-K-F-K-E-A-F- (SEQ ID NO: 14), IND E-W L-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO: 15), E-.W-L-K-A-F-Y-
(N
D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO: 16),E-W-L-K-A-F-Y-D-K-V-F-E-K-F-K- E-A-F- (SEQ ID NO: 17), E-W-L-K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ ID NO: 18), E-W-L-K-A-F-Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID NO E-W-L- K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO 20), A-F-Y-D-K-V-A-E-K-L-K- E-A-F- (SEQ ID NO 21),A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO 22), A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO 23),A-F-Y-D-K-F-F-E-K-F-K-E- F-F- (SEQ ID NO 24), A-F-Y-D-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO 25), A- F-Y-D-K-V-A-E-K-F-K-E-A-F (SEQ ID NO A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQID NO 27), A-F-Y-D-K-V-F-E-K-F-K-E-A-F- (SEQ ID NO 28), A-F-Y-D- K-V-F-E-K-L-K-E-F-F- (SEQ ID NO 29),A-F-Y-D-K-V- A-E-K-F-K-E-F-F- (SEQ ID NO: 30), K-A-F-Y-D-K-V-F-E-K-F-K-E-F- (SEQ ID NO L-F-Y-E- K-V-L-E-K-F-K-E-A-F- (SEQ ID NO 32), A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO 33),A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO: 34),A-F-Y-D- K-V-F-E-K-F-K-E-A-F- (SEQ ID NO 35),A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ ID NO: 36), A-F-Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID NO 37),A-F-Y-D-K-V-F- E-K-F-K-E-F-F- (SEQ ID NO 38), D-W-L-K-A-L-Y-D-K-V-A-E-K-L-K-E-A-L- (SEQ ID NO 39), D-W-F-K-A-F-Y-E-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO D-W-F-K-A-F-Y-E-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO 41), E-W-L-K-A-L-Y-E- K-V-A-E-K-L-K-E-A-L- (SEQ ID NO 42), E-W-L-K-A-F-Y-E-K-V-A-E-K-L-K-E- A-F (SEQ ID NO 43), E-W-F-K-A-F-Y-E-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO: 44), E-W-L-K-A-F-Y-E-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO Y-E-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO 46), E-W-F-K-A-F-Y-E-K-F-F-E-K-F- K-E-F-F- (SEQ ID NO 47), D-F-L-K-A-W-Y-D-K-V-A-E-K-L-K-E-A-W- (SEQ ID NO E-F-L-K-A-W-Y-E-K-V-A-E-K-L-K-E-A-W- (SEQ ID NO 49), D-F- W-K-A-W-Y-D-K-V-A-E-K-L-K-E-W-W- (SEQ ID NO E-F-W-K-A-W-Y-E- K-V-A-E-K-L-K-E-W-W- (SEQ ID NO D-K-L-K-A-F-Y-D-K-V-F-E-W-A-K- E-A-F (SEQ ID NO 52), D-K-W-K-A-V-Y-D-K-F-A-E-A-F-K-E-F-L- (SEQ ID NO: 53), E-K-L-K-A-F-Y-E-K-V-F-E-W-A-K-E-A-F- (SEQ ID NO 54),E-K-W- K-A-V-Y-E-K-F-A-E-A-F-K-E-F-L- (SEQ ID NO: 3C E-K-F-K-E-A-Y- (SEQ ID NO: 56), E-K-W-K-A-V-Y-E-K-F-A-E-A-F-K-E-F-L- (SEQ ID NO 57), D-W-L-K-A-F-V-Y-D-K-V-F-K-L-K-E-F-F (SEQ ID NO 58), Z E-W-L-K-A-F-V-Y-E-K-V-F-K-L-K-E-F-F- (SEQ ID NO: 59), D-W-L-R-A-F-Y-D- K-V-A-E-K-L-K-E-A-F- (SEQ ID NO: 60), E-W-L-R-A-F-Y-E-K-V-A-E-K-L-K-E- A-F (SEQ ID NO 61),D-W-L-K-A-F-Y-DR-V-A-E-K-L-K-E-A-F- (SEQ ID NO: 62), E-W-L-K-A-F-Y-E-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO 63),D-W-L-K-A-F- Y-D-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO 64),E-W-L-K-A-F-Y-E-K-V-A-E-R-L- K-E-A-F- (SEQ ID NO: 65), D-W-L-K-A-F-Y-D-K-V-A-E-K-L-R-E-A-F- (SEQ ID NO 66), E-W-L-K-A-F-Y-E-K-V-A-E-K-L-R-E-A-F (SEQ ID NO 67), D-W-L-K- A-F-Y-D-R-V-A-E-R-L-K-E-A-F- (SEQ ID NO 68), E-W-L-K-A-F-Y-E-R-V-A-E- R-L-K-E-A-F- (SEQ ID NO: 69), D-W-L-R-A-F-Y-D-K-V-A-E-K-L-R-E-A-F (SEQ ID NO 70),E-W-L-R-A-F-Y-E-K-V-A-E-K-L-R-E-A-F- (SEQ ID NO: 71), D-W-L-R-A-F-Y-D-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO 72),E-W-L-R-A-F-Y-E- R-V-A-E-K-L-K-E-A-F- (SEQ ID NO 73),D-W-L-K-A-F-Y-D-K-V-A-E-R-L-R-E- A-F- (SEQ ID NO 74), E-W-L-K-A-F-Y-E-K-V-A-E-R-L-R-E-A-F- (SEQ ID NO D-W-L-R-A-F-Y-D-K-V-A-E-R-L-K-E-A-F (SEQ ID NO 76), E-W-L-R-A- F-Y-E-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO 77), D-W-L-K-A-F-Y-D-K-V-A-E-K- L-K-E-A-F-P-D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F (SEQ ID NO 78), D-W-
L-K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F-P-D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F
(SEQ ID NO 79),D-W-F-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-P-D-W-F-K-A-F-Y- D-K-V-A-E-K-L- K-E-A-F (SEQ ID NO 80), D-K-L-K-A-F-Y-D-K-V-F-E-W-A-K- E-A-F-P-D-K7L-K-A-F-Y-D-K-V-F-E-W-L-K-E-A-F (SEQ ID NO D-K-W-K-
A-V-Y-D-K-F-A-E-A-F-K-E-F-L-P-D-K-W-K-A-V-Y-D-K-F-A-E-A-F-K-E-F-L
(SEQ ID NO 82), D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-P-D-W-F-K-A-F-Y- D-K-V-A-E-K-F-K-E-A-F (SEQ ID NO: 83), D-W-L-K-A-F-V-Y-D-K-V-F-K-L-K- E-F-F-P-D-W-L-K-A-F-V-Y-D-K-V-F-K-L-K-E-F-F (SEQ ID NO 84), D-W-L-K-
A-F-Y-D-K-F-A-E-K-F-K-E-F-F-P-D-W-L-K-A-F-Y-D-K-F-A-E-K-F-K-E-F-F
(SEQ ID NO 85), wherein said peptide comprises at least one amino acid residue and protects a phospholipid afgainst oxidation by an oxidizing agent.
[00Q1Ob] According to a second broad form of the invention there is provided a peptide that ameliorates a symptom of atherosclerosis, wherein 3D O said peptide has the amino acid sequence D-W-F-K-A-F-Y-D-K-V-A-E- K-F-K-E-A-F (SEQ ID said peptide comprises all amino acid residues; said peptide comprises at least one protecting group; and said peptide protects a phospholipid against oxidation by an oxidizing agent.
O [0010c] In particularly preferred embodiments, the peptide further N 10 comprises a protecting group coupled to the amino and/or carboxyl terminus.
Preferred protecting groups include, but are not limited to acetyl, amide, and 3 to 20 carbon alkyls groups, Fmoc, t-boc, 9-fluoreneacetyl group, 1fluorenecarboxylic group, 9-fluorenecarboxylic group, 9-fluorenone-1carboxylic group, benzyloxycarbonyl, Xanthyl (Xan), Trityl (Trt), 4-methyltrityl (Mtt), 4-methoxytrityl (Mmt), 4-methoxy-2,3,6-trimethyl-benzenesulphonyl (Mtr), Mesitylene-2-sulphonyl (Mts), 4,4-dimethoxybenzhydryl (Mbh), Tosyl (Tos), 2,2,5,7,8-pentamethyl chroman-6-sulphonyl (Pmc), 4-methylbenzyl (MeBzl), 4-methoxybenzyl (MeOBzl), Benzyloxy (BzlO), Benzyl (Bzl), Benzoyl 3-nitro-2-pyridinesulphenyl (Npys), 1-(4,4-dimentyl-2,6diaxocyclohexylidene)ethyl (Dde), 2,6-dichlorobenzyl (2,6-DiCI-Bzl), 2chlorobenzyloxycarbonyl 2-bromobanzyloxycarbonyl (2-Br-Z), Benzyloxymethyl (Bom), t-butyoxycarbonyl (Boc), cyclohexyloxy (cHxO), tbutoxymethyl (Bum), t-butoxy (tBuO), t-Butyl (tBu), Acetyl and Trifluoroacetyl (TFA). In certain particularly preferred embodiments the peptide further comprises a first protecting group coupled to the amino terminus and a second protecting group coupled to the carboxyl terminus.
Particularly preferred peptides closely mimic the class A amphipathic helix of human or mouse apo A-1. In certain embodiments, preferred peptides comprise greater NI than about 50% amino acid sequence identity with the polypeptide encoded by the exon o) encoding a class A amphipathic helix of human or mouse apo A-i1. In certain preferred Z emb~diments, at least about 10%, preferably at least 20%, more preferably at least about N' 5 30%, sill more preferably at least about 50%, even more preferably at least about and most preferably at least 90% and even 100% of the enantiomeric amino acids are"D tn amino acids. The peptide may be combined with a pharmacologically acceptable excipient an excipient suitable for oral administration to a mammal).
[0011] In certain particularly preferred embodiments, the peptide comprises one or More of the following amino acid sequences: D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO:2), D-W-F-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO:3), D-W-L-K-A-F- Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO:4), D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO:5), D-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO:6), D-W-L-K-A-F-Y- D-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO:7), D-W-F-K-A-F-Y-D-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO: D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO: D-W-L-K-A-F-Y-D-K-V- F-E-K-F-K-E-A-F- (SEQ ID NO: 10), D-W-L-K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ ID NO: 11), D-W-L-K-A-F-Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID NO: 12), D-W-L-K-A-F-Y-D-K- V-F-E-K-F-K-E-F-F- (SEQ DD NO: 13), E-W-L-K-L-F-Y-E-K-V-L-E-K-F-K-E-A-F- (SEQ ID NO: 14), E-W-L-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO: 15), E-W-L-K-A-F-Y-D-K- V-A-E-K-L-K-E-F-F- (SEQ ID NO: 16), E-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-A-F- (SEQ ID NO: 17), (SEQ ID NO: 18), E-W-L-K-A-F-Y-D-K- V-A-E-K-F-K-E-F-F- (SEQ ID NO: 19), E-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ ID A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO:2 A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ED NO:22), A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO:23), A-F-Y-D-K-F-F-E-K-F- K-E-F-F- (SEQ ED NO:24), A-F-Y-D-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO:2 A-F-Y-D-K-V- A-B-K-F-K-B-A-F- (SEQ ID NO:26), A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO:27), A-F- Y-D-K-V-F-E-K-F-K-E-A-F- (SEQ ID NO:2 A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ IID NO:29), A-F-Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID NO:30), K-A-F-Y-D-K-V-F-E-K-F-K-E-F- (SEQ DD NO:3 L-F-Y-E-K-V-L-E-K-F-K-E-A-F.7 (SEQ ID NO:32), A-F-Y-D-K-V-A-E-K-F- K-E-A-F- (SEQ ID NO:33), A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO:34), A-F-Y-D-K-V- F-B-K-F-K-E-A-F- (SEQ ID NO:35), A-F-Y-D-K-V-F-E-KL-KEFF- (SEQ ID NO:36), A-F- Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ED NO:37), A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ MD NO:3 D-W-L-K-A-L-Y-D-K-V-A-E-K-L-K-E-A-L- (SEQ ID~ NO:39), D-W-F-K-A-F-Y-E-K- N- V-A-E-K-L-K-E-F-F- (SEQ ID NO:40), D-W-F-K-A-F-Y-E-K-F-F-E-K-F-K-E-F-F- (SEQ ID o NO:41), E-W-L-K-A-L-Y-E-K-V-A-E-K-L-K-E-A-L- (SEQ ID NO:42), E-W-L-K-A-F-Y-E-K- Z V-A-E-K-L-K-E-A-F- (SEQ ID NO:43), E-W-F-K-A-F-Y-E-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO:44), E-W-L-K-A-F-Y-E-K-V-F-E-K-F-K-E-F-F- (SEQ ED NO:45), E-W-L-K-A-F-Y1-E-K-F- F-E-K-F-K-E-F-F- (SEQ ID NO:46), E-W-F-K-A-F-Y-E-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO:47), D-F-L-K-A-W-Y-D-K-V-A-E-K-L-K-E-A-W- (SEQ ID NO:48), E-F-L-K-A-W-Y-E-K- V-A-E-K-L-K-E-A-W- (SEQ ID NO:4 (SEQ c-I ID NO:50), E-F-W-K-A-W-Y-E-K-V-A-E-K-L-K-E-W-W- (SEQ ID NO:5 D-K-L-K-A-F-Y- C) 10 D-K-V-F-E-W-A-K-E-A-F- (SEQ ID NO:5 D-K-W-K-A-V-Y-D-K-F-A-E-A-F-K-E-F-Lc-I (SEQ ID NO:53), E-K-L-K-A-F-Y-E-K-V-F-E-W-A-K-E-A-F- (SEQ ID NO:54), E-K-W-K-A- V-Y-E-K-F-A-E-A-F-K-E-F-L- (SEQ ID NO: 55), D-W-L-K-A-F-V-D-K-F-A-E-K-F-K-E-A-Y- (SEQ ID NO:56), E-K-W-K-A-V-Y-E-K-F-A-E-A-F-K-E-F-L- (SEQ ID NO:5 D-W-L-K-A-F- V-Y-D-K-V-F-K-L-K-E-F-F- (SEQ ED NO:58), E-W-L-K..A-F-V-Y-E-K-V-F-K-L-K-E-F-F- (SEQ ID NO:59), D-W-L-R-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO:60), E-W-L-R-A- F-Y-E-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO:6 D-W-L-K-A-F-Y-D-R-V-A-E-K-L-K-E-A-F- (SEQ DD NO: 62), E-W-L-K-A-F-Y-E-R-V-A-E-K-L-K-E-A-F- (SEQ ED NO:63), D-W-L-K-A-F- Y-D-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO:64), E-W-L-K-A-F-Y-E-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO:65), D-W-L-K-A-F-Y-D-K-V-A-E-K-L-R-E-A-F- (SEQ lID NO:66), E-W-L-K-A- F-Y-E-K-V-A-E-K-L-R-E-A-F- (SEQ ID NO:67), D-W-L-K-A-F-Y-D-R-V-A-E-R-L-K-E-A-F- (SEQ ID NO:68), E-W-L-K-A-F-Y-E-R-V-A-E-R-L-K-E-A-F- (SEQ ID NO:69), D-W-L-R-A-F- Y-D-K-V-A-E-K-L-R-E-A-F- (SEQ ID NO:7 E-W-L-R-A-F-.Y-E-K-V-K.E-K-L-R-E-A-F- (SEQ ID NO:7 D-W-L-R-A-F-Y-D-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO:72), E-W-L-R-A-F- Y-E-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO D-W-L-K-A-F-Y-D-K-V-A-E-R-L-R-E-A-F- (SEQ ID NO:74), E-W-L-K-A-F-Y-E-K-V-A-E-R-L-R-E-A-F- (SEQ ID NO:75), D-W-L-R-A-F- Y-D-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO:76), E-W-L-R-A-F-Y-E-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO:7 D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-P-D-W-L-K-A-F-Y-D-K-V-A-E- K-L-K-E-A-F (SEQ ID NO: 7 D-W-L.-K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F-P-D-W-L-K-A-F- Y-D-K-V-A-E-K-L-K-E-F-F (SEQ ID NO:79), D-W-F-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-P- D-W-F-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F (SEQ ED NO: 80), D-K-L-K-A-F-Y-D-K-V-F-E-W- A-K-E-A-F-P-D-K-L-K-A-F-Y-D-K-V-F-E-W-L-K-E-A-F (SEQ ID NO:S8 D-K-W-K-A-V-Y- D-K-F-A-E-A-F-K-E-F-L-P-D-K-W-K-A-V-Y-D-K-F-A-E-A-F-K-E-F-L (SEQ ID NO: 82), D- W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-P-D-'W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F
(SEQ
S6
O
SID NO:83), D-W-L-K-A-F-V-Y-D-K-V-F-K-L-K-E-F-F-P-D-W-L-K-A-F-V-Y-D-K- O V-F-K-L-K-E-F-F (SEQ ID NO:84), D-W-L-K-A-F-Y-D-K-F-A-E-K-F-K-E-F-F-P- Z D-W-L-K-A-F-Y-D-K-F-A-E-K-F-K-E-F-F (SEQ ID NO:85), truncations of the
ID
Cabove sequences, multimeric combinations preferably ranging from dimers to trimers, tetramers, 5 mers, 8 mers, or 10 mers) of the above sequences, conservative substitutions of the above sequences, and/or the above sequences comprising amino acid analogs. The enantiomeric amino C- acids of such sequences preferably comprise at least one amino acid or Salternatively at least one amino acid. In certain preferred embodiments, at C 10 least 50%, more preferably at least 75%, and most preferably at least and even 100% of the enantiomeric amino acids are amino acids as described herein. Such peptides can also include a protecting group amide, acetyl, propionyl, and a 3 to 20 carbon alkyl, etc.) coupled to the amino or carboxyl terminus. In certain embodiments, the protecting group coupled to the carboxyl terminus is an amide. In certain embodiments, the protecting group coupled to the amino terminus is an acetyl, a propionyl, or a 3 to carbon alkyl. Certain peptides comprise both a carboxyl- and an aminoterminus protecting group. In one such embodiment, the amino terminus protecting group is a protecting group selected from the group consisting of acetyl, propionyl, and a 3 to 20 carbon alkyl; and the carboxyl terminal protecting group is an amide.
[0012] In certain embodiments, the peptide is one that protects a phospholipid against oxidation by an oxidizing agent selected from the group consisting of lipids such as hydrogen peroxide, 13(S)-HPODE, HPODE, HPETE, HODE, and HETE. The phospholipid can be a phospholipid selected from the group consisting of 1-palmitoyl-2-arachidonoyl-sn-glycero-3phosphorylcholine (PAPC), 1-stearoyl-2-arachidonoyl-sn-glycero-3phosphorylcholine (SAPC), 1 -stearoyl-2-arachidonoyl-sn-glycero-3phosphorylethanolamine (SAPE). Thus the peptide prevents the formation of lipids such as oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3phosphorylcholine (Ox-PAPC), 1-palmitoyl-2-oxovaleroyl-sn-glycero-3phosphorylcholine (POVPC), 1-palmitoyl-2-glutaroyl-sn-glycero-3- 7 C phosphorylcholine (PGPC), 1 -palmitoyl-2-epoxyisoprostane-sn-glycero-3- O phosphoryholine (PEIPC), oxidized 1-stearoyl-2-arachidonoyl-sn-glycero-3phosphorylcholine (Ox-SAPC), 1-stearoyl-2-oxovaleroyl-sn-glycero-3- Iphosphorycholine (SOVPC), 1-stearoyl-2-glutaroyl-sn-glycero-3phosphorylcholine (SGOPC), 1-stearoyl-2-glutaepoxyisoprostane-sn-glycero-3p hosp horylcho line (SG PC) 1 -stea royl-2-epoxyisoprosta ne-sn-g lycero-3phosphorylcholine (SIPC), oxidized 1-stearoyl-2-arachidonly-sn-glycero-3phosphorylethanolamine (OxSAPE), 1-stearoyl-2-oxovaleroyl-sn-glycero-3- C- phosphorylethanolamine (SOVPE), 1-stearoyl-2-glutaroyl-sn-glycero-3phosphorylethanolamine (SGPE), and 1-stearoyl-2-epoxyisoprostane-sn- CN 10 glycero-3-phosphorylethanolamine (SEI PE).
[0013] In another embodiment, this invention provides a composition, suitable for oral administration, that ameliorates a symptom of atherosclerosis.
The composition comprises a peptide that is a human apo A-1 peptide or fragment thereof comprising a class A amphipathic helix, or an analogue of a human apo A-1 peptide wherein said peptide has a first protecting group attached to an amino terminal and a second protecting group attached to a carboxyl terminal. The peptide may comprise a plurality of amino acid residues or a plurality of residues. The protecting groups include, but are not limited to the protecting groups described herein. In certain embodiments, more than half, more preferably more than 80%, and most preferably more than 90% or even all of the enantiomeric amino acids comprising the peptide are amino acids. The composition can further comprise a pharmaceutically acceptable excipient an excipient suitable for oral administration or an excipient suitable for injection). Preferred peptides are capable of protecting a phospholipid 1-palmitoyl-2-arachidonoyl-snglycero-3-phosphorylcholine (PAPC), 1-stearoyl-2-arachindonoyl-sn-glycero- 3-phosphorylcholine (SAPC), 1-stearoyl-2-arachidonoyl-sn-glycero-3phosphorylethanolamine (SAPE)] from oxidization by an oxidizing agent (e.g.
hydrogen peroxide, 13(S)-HPODE, 15(S)-HPETE, HPODE, HPETE, HODE, and HETE). Thus the peptide prevents the formation of oxidized 1-palmitoyl- 2-arachidonoyl-sn-glycero-3-phosphorylcholine (Ox-PAPC), 1-palmitoyl-2oxovaleroyl-sn-glycero-3-phosphorylcholine (POVPC), 1 -palmitoyl-2-glutaroyl- 8 Ssn-glycero-3-phosphorylcholine (PGPC), 1-palmitoyl-2-epxoyisoprostane-sn- O glycero-3-phosphorylcholine (PEIPC), oxidized 1-stearoyl-2-arachidonoyl-snglycero-3-phosphorylcholine (Ox-SAPC), 1-stearoyl-2-oxovaleroyl-sn-glycero- 3-phosphorycholine (SOVPC), -stearoyl-2-glutaroyl-sn-glycero-3cN| 3-phosphorylcholine (SOVPC), 1-stearoyl-2-glutaroyl-sn-glycero-3phosphorylcholine (SGPC), 1-stearoyl-2-epoxyisoprostane-sn-glycero-3uI phosphorylcholine (SEIPC), oxidized 1-stearoyl-2-arachidonoyl-sn-glycero-3phosphorylethanolamine (Ox-SAPE), 1-stearoyl-2-oxovaleroyl-sn-glycero-3- C phosphorylethanolamine (SOVPE), 1-stearoyl-2-gluteroyl-sn-glycero-3- 0 phosphorylethanolamine (SGPE), and 1-stearoyl-2-epoxyisoprostane-sn- C 10 glycero-3-phosphorylethanolamine (SEI PE).
[0014] This invention also provides methods of ameliorating a symptom of atherosclerosis. The methods comprise administering to an organism (e.g.
human or non-human mammal) one or more of the peptides described herein.
In particularly preferred embodiments for oral administration, such peptides comprise a plurality of amino acids and/or are protected as described herein. For administration by a non-oral route, the peptides preferably comprise a plurality of amino acids. The peptide is preferably orally administered to the organism and the organism is preferably an organism diagnosed as having or as at risk for one or more symptoms of atherosclerosis. In certain embodiments, the peptide can be provided as an isolated peptide or combined with a pharmacological excipient as described herein. The administration is preferably at a dosage sufficient to ameliorate one or more symptoms of atherosclerosis and/or to significantly reduce the likelihood of occurrence of one or more symptoms of atherosclerosis.
[0015] In still another embodiment, this invention provides a kit for ameliorating a symptom of atherosclerosis. Preferred kits include a container containing one or more of the peptides described herein. The peptides preferably comprise a plurality of amino acids or alternatively a plurality of amino acids and/or are protected as described herein. In certain embodiments, the kit can optionally further include a pharmaceutically 8A
O
ci acceptable excipient and/or the peptide is provided combined with a O pharmaceutically acceptable excipient in a unit dosage formulation).
Z Preferred kits provided the peptide(s) as a unit dosage formulation is for oral c administration. The kits also, optionally, include instructional materials teaching the use of said peptide for ameliorating one or more symptoms of I atherosclerosis and/or for reducing the likelihood of occurrence of one or more symptoms of atherosclerosis.
C( [0016] In certain embodiments, this invention excludes any one or more Speptides disclosed in U.S. Patent 4,643,988 and/or in Garber et al. (1992) N 10 Arteriosclerosis and Thrombosis, 12: 886-894. In certain embodiments this invention excludes any one or more peptides disclosed in U.S. Patent 4,643,988 and/or in Garber et al (1992) that were synthesized with all enantiomeric amino acids being amino acids or synthesized with amino acids where the peptides are blocking groups. In certain embodiments, this invention excludes peptides having the formula A 1
-B
1
-B
2
-C
1
-D-B
3
-B
4
-A
2
-C
2
-B
5 Be-A 3
-C
3
-B
7
-C
4
-A
4
-B
8
-B
9 (SEQ ID NO:87) wherein A 1
A
2
A
3 and A 4 are independently aspartic acid or glutamic acid, or homologues or analogues thereof; B 1
B
2
B
3
B
4
B
5
B
6 B, B and B 9 are independently tryptophan, phenylalanine, alanine, leucine, tyrosine, isoleucine, valine or anaphthylalanine, or homologues or analogues thereof; C1, C 2
C
3 0 and C 4 are independently lysine or arginine, and D is serine, threonine, alanine, glycine, CN histidine, or homologues or analogues thereof; provided that, when A 1 and A 2 are aspartic acid, A 3 and A 4 are glutamic acid, B2 and B 9 are leucine, B 3 and B 7 are phenylalanine, B 4 Z is tyrosine, B 5 is valine, B 6
B
8 and D are alanine, and C 1
C
2
C
3 and C 4 are lysine, B1 is N 5 not tryptophan.
Definitions.
[0017] The terms "polypeptide", "peptide" and "protein" are used interchangeably C1 herein to refer to a polymer of amino acid residues. The terms apply to amino acid Spolymers in which one or more amino acid residues is an artificial chemical analogue of a CN 10 corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers.
[0018] The term" class A amphipathic helix" refers to a protein structure that forms an a-helix producing a segregation of a polar and nonpolar faces with the positively charged residues residing at the polar-nonpolar interface and the negatively charged residues residing at the center of the polar face (see, Segrest et al. (1990) Proteins: Structure, Function, and Genetics 8: 103-117).
[0019] The term "ameliorating" when used with respect to "ameliorating one or more symptoms of atherosclerosis" refers to a reduction, prevention, or elimination of one or more symptoms characteristic of atherosclerosis and/or associated pathologies. Such a reduction includes, but is not limited to a reduction or elimination of oxidized phospholipids, a reduction in atherosclerotic plaque formation and rupture, a reduction in clinical events such as heart attack, angina, or stroke, a decrease in hypertension, a decrease in inflammatory protein biosynthesis, reduction in plasma cholesterol, and the like.
[0020] The term "enantiomeric amino acids" refers to amino acids that can exist in at least two forms that are nonsuperimposable mirror images of each other. Most amino acids (except glycine) are enantiomeric and exist in a so-called L-form (L amino acid) or D-form (D amino acid). Most naturally occurring amino acids are amino acids. The terms "D amino acid" and "L amino acid" are used to refer to absolute configuration of the amino acid, rather than a particular direction of rotation of plane-polarized light. The usage herein is consistent with standard usage by those of skill in the art.
-9r- 0 [0021] The term "protecting group" refers to a chemical group that, when attached C1 to a functional group in an amino acid a side chain, an alpha amino group, an alpha O carboxyl group, etc.) blocks or masks the properties of that functional group. Preferred Z amino-terminal protecting groups include, but are not limited to acetyl, or amino groups.
C1 5 Other amino-terminal protecting groups include, but are not limited to alkyl chains as in fatty acids, propeonyl, formyl and others. Preferred carboxyl terminal protecting groups include, but are not limited to groups that form amides or esters.
r [0022] The phrase "protect a phospholipid from oxidation by an oxidizing agent" refers to the ability of a compound to reduce the rate of oxidation of a phospholipid (or the amount of oxidized phospholipid produced) when that phospholipid is contacted with an oxidizing agent hydrogen peroxide, 13-(S)-HPODE, 15-(S)-HPETE, HPODE, HPETE, HODE, HETE, etc.).
[0023] The terms "low density lipoprotein" or "LDL" is defined in accordance with common usage of those of skill in the art. Generally, LDL refers to the lipid-protein complex which when isolated by ultracentrifugation is found in the density range d 1.019 to d 1.063.
[0024] The terms "high density lipoprotein" or "HDL" is defined in accordance with common usage of those of skill in the art. Generally "HDL" refers to a lipid-protein complex which when isolated by ultracentrifugation is found in the density range of d= 1.063 tod =1.21.
[0025] The term "Group I HDL" refers to a high density lipoprotein or components thereof apo A-I, paraoxonase, platelet activating factor acetylhydrolase, etc.) that reduce oxidized lipids in low density lipoproteins) or that protect oxidized lipids from oxidation by oxidizing agents.
[0026] The term "Group II HDL" refers to an HDL that offers reduced activity or no activity in protecting lipids from oxidation or in repairing reducing) oxidized lipids.
[0027] The term "HDL component" refers to a component molecules) that comprises a high density lipoprotein (HDL). Assays for HDL that protect lipids from oxidation or that repair reduce oxidized lipids) also include assays for components of ITDL apo A-I, paraoxonase, platelet activating factor acetylhydrolase, etc.) that C display such activity.
O [0028s The term "human apo A-I peptide" refers to a full-length human apo A-I peptide or to a fragment or domain thereof comprising a class A amphipathic helix.
[0029] A "monocytic reaction" as used herein refers to monocyte activity characteristic of the "inflammatory response" associated with atherosclerotic plaque formation. The monocytic reaction is characterized by monocyte adhesion to cells of the vascular wall cells of the vascular endothelium), andlor chemotaxis into the subendothelial space, and/or differentiation of monocytes into macrophages.
[0030] The term "absence of change" when referring to the amount of oxidized phospholipid refers to the lack of a detectable change, more preferably the lack of a statistically significant change at least at the 85%, preferably at least at the more preferably at least at the 95%, and most preferably at least at the 98% or 99% confidence level). The absence of a detectable change can also refer to assays in which oxidized phospholipid level changes, but not as much as in the absence of the protein(s) described herein or with reference to other positive or negative controls.
[0031] The following abbreviations are used herein: PAPC: L-ax-l-palmitoyl-2arachidonoyl-sn-glycero-3-phosphocholine POVPC: 1-palmitoyl-2-(5-oxovaleryl)-snglycero-3-phosphocholine; PGPC: 1-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine; PEIPC: 1-palmitoyl-2-(5,6-epoxyisoprostane
E
2 )-sn-glycero-3-phsophocholine; ChC18:2: cholesteryl linoleate; ChC18:2-OOH: cholesteryl linoleate hydroperoxide; DMPC: 1,2ditetradecanoyl-rac-glycerol-3-phosphocholine; PON: paraoxonase; HPF: Standardized high power field; PAPC: L-a-l1-pahnlmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine; POVPC: 1 -palmitoyl-2-(5-oxovaleryl)-sn-glycero-3-phosphocholine; PGPC: 1-palmitoyl- 2-glutaryl-sn-glycero-3-phosphocholine; PEIPC: 1-palmitoyl-2-(5,6-epoxyisoprostane
E
2 sn-glycero-3-phosphocholine; PON: paraoxonase; BL/6: C57BL/6J; C3H:C3H/HeJ.
[00321 The term "conservative substitution" is used in reference to proteins or peptides to reflect amino acid substitutions thatdo not substantially alter the activity (specificity for lipoproteins))or binding affinity for lipids or lipoproteins)) of the molecule. Typically conservative amino acid substitutions involve substitution one amino acid for another amino acid with similar chemical properties charge or O hydrophobicity). The following six. groups each contain amino acids that are typical N1 conservative substitutions for one another: 1) Alanine Serine Threonine 2) O Aspartic acid Glutamic acid 3) Asparagine Glutamine 4) Arginine Lysine 5) Isoleucine Leucine Methionine Valine and 6) C1 5 Phenylalanine Tyrosine Tryptophan [0033] The terms "identical" or percent "identity," in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using one of the following sequence comparison algorithms or by visual inspection. With respect to the peptides of this invention sequence identity is determined over the full length of the peptide.
[0034] For sequence comparison, typically one'sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
[0035] Optimal alignment of sequences for comparison can be conducted, by the local homology algorithm of Smith Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm ofNeedleman Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson Lipman (1988) Proc. Natl. Acad. Sci.
USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally Ausubel et al., supra).
[0036] One example of a useful algorithm is PILEUP. PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments to show relationship and percent sequence identity. It also plots a tree or dendogram showing the clustering relationships used to create the alignment. PILEUP -12ruses a simplification of the progressive alignment method of Feng Doolittle (1987) J C Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins O> Sharp (1989) CABIOS 5: 151-153. The program can align up to 300 sequences, each of a 0 Z maximum length of 5,000 nucleotides or amino acids. The multiple alignment procedure C 5 begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences. This cluster is then aligned to the next most related sequence or r cluster of aligned sequences. Two clusters of sequences are aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by C a series of progressive, pairwise alignments. The program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence C comparison and by designating the program parameters. For example, a reference sequence can be compared to other test sequences to determine the percent sequence identity relationship using the following parameters: default gap weight default gap length weight and weighted end gaps.
[0037] Another example of algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in Altschul et al. (1990) J. Mol. Biol. 215: 403-410. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al, supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always 0) and N (penalty score for mismatching residues; always For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either -13- 0 sequence is reached. The BLAST algorithm parameters W, T, and X determine the C" sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) O uses as defaults a wordlength of 11, an expectation of 10, M=5, and a Z comparison of both strands. For amino acid sequences, the BLASTP program uses as \0 C 5 defaults a wordlength of 3, an expectation of 10, and the BLOSUM62 scoring matrix (see Henikoff& Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915).
[0038] In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., SKarlin Altschul (1993) Proc. Natl. Acad. Sci. USA ,90: 5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
[0039] The term "D-18A peptide" refers to a peptide having the sequence: D-W- L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F (SEQ ID NO: 1) where all of the enantiomeric amino acids are D form amino acids.
BRIEF DESCRIPTION OF THE DRAWINGS [0040] Figure 1, panels A, B, C, and D show the association of 14 C-D-5F with blood components in an ApoE null mouse. ApoA-I mimetic peptide D-5F labeled with 1 4 C amino acids was administered by oral gavage to apo E deficient mice or incubated with their plasma in vitro. Fasting blood was collected 6 hrs after gavage and 14C association with blood, plasma, and lipoproteins determined.
[0041] Figures 2A and 2B illustrate that orally administered D peptide is active.
ApoA-I mimetic peptides D-5F and L-5F (100 jtg per animal) were administered to LDL receptor null mice by oral gavage. Blood was collected after 6 hrs, LDL and HDL were isolated by gel filtration (FPLC) and examined in the artery wall model system for HDL protective capacity (Figure 2A) and LDL resistance (Figure 2B) to oxidation by determining monocyte chemotactic activity generated. As seen, D-5F but not -14rendered the HDL markedly more protective and LDL after D-5F became highly resistant Sto oxidation.
O [0042] Figures 3A and 3B show the plasma concentration ofD vs L peptide after gavage. ApoA-I mimetic peptides D-4F (Figure 3B) and L-4F (Figure 3B) were labeled C 5 with 125I and administered by oral gavage to LDL receptor null mice Blood was collected after 3 hrs, plasma fractionated by FPLC and radioactivity determined in the t) eluted fractions. Less than 15% of the L peptide eluted as intact 18 mer whereas more b than 70% of the D-4F was intact. These studies demonstrate that the D peptide is C, dramatically more resistant to degradation in vivo compared with the L peptide.
0 10 [0043] Figure 4 illustrates the absence of antibody to D-4F in treated mice. No antibody (white precipitation line) to D-4F was detected in LDL receptor null mouse plasma following 6 weeks of treatment with peptide at 5 mg per day (lower panel). The positive control (upper panel) shows the presence of a precipitation line for apoA-I in mouse plasma. Upper panel: Center: rabbit anti ApoA-I and periphery: plasma from D- 4F mice. Lower panel: Center: Plasma from LDL mice treated with D 4F, and Periphery: Pure D-4F peptide at 0 to 80 ig.
[00441 Figure 5 shows the incidence of fatty streak lesions in the aortic root of LDL receptor null mice on a Western Diet. Groups of LDL receptor null mice were placed on a Western type diet and were given orally, vehicle (Control) 9) or peptide D-4F twice daily for 6 weeks. The mice were subsequently sacrificed, aortic arch fixed and sectioned and fatty streak lesions quantified. The mice receiving D-4F had an 81% reduction in lesion area (p<0.01).
[0045] Figure 6, panels A, B, and C illustrate the plasma distributions of peptide or apo A-I following intraperitoneal injection. Human apo A-I, mouse apo A-I, and peptide 5F were labeled with 1251 and injected intraperitoneally into C57BL/6 mice that had been fed the atherogenic diet for at least three weeks. Samples were taken during the kinetic studies described in Table 3. Representative samples were analyzed by the CLiP method, and fractions were collected for determination of radioactivity. The elution volume was based on the column pump rate only; the volume contributed by the enzymatic reagent pump was neglected. Data shown are cholesterol (as absorbance at 500 nm in arbitrary units; solid lines) and radioactivity (in counts per minute; dashed lines).
L Panels are A: human apo A-I (one hour following injection); B: mouse apo A-I (one hour), N C: 5F (1.5 hours).
0 [0046] Figure 7, panels A and B illustrate the interaction of mouse lipoproteins with human artery wall cells. LDL and HDL were isolated by FPLC from the plasma of 1 5 mice fed the atherogenic diet and injected with vehicle (PBS), or with peptide 5F at gg/mouse/day. The cocultures were treated without (No Addition) or with human LDL in (hLDL) at 200 pg/ml LDL protein, or mouse LDL (MoLDL) at 200 pg/ml or with 200 j tg/ml human LDL human HDL (hHDL) at 350 [tg/ml of HDL protein or mouse HDL C (MoHDL) at 300 g/ml. The cocultures were incubated with the above additions for 8 hrs O 10 at 370 C in the presence of 10% lipoprotein deficient serum (LPDS). The supematants C were collected and analyzed for Auerbach lipid hydroperoxide equivalents (panel The cocultures were then washed and incubated with fresh culture medium without serum or LPDS for an additional 8 hrs. The conditioned medium was collected and analyzed for monocyte chemotactic activity (panel A no cell blank (No Cell Blank) is included in both panels for comparison.
[0047] Figure 8 shows mean lesion cross-sectional areas. Data shown represent the mean lesion cross-sectional area for each animal (O and the mean I SEM of all animals in each group with error bars. Abbreviations: PBS, mice fed the atherogenic diet and injected daily with 200 pl phosphate-buffered saline; 5F, mice fed the atherogenic diet and injected daily with 20 jig of 5F in 200 pl PBS; MoAI, mice fed the atherogenic diet and injected daily with 50 pg of mouse apo A-I in 200 [il PBS. p<0.002 as determined by two-tailed t-test. A significant difference was also shown using one way analysis of variance on ranks (p<0.001).
[0048] Figure 9 shows that both the D and L isomers of apo A-I peptide mimetics prevent monocyte chemotactic activity induced by mildly oxidized LDL in vitro. Medium alone (LDL, NO CELLS or CELLS, NO LDL), control LDL from normal subjects at 250 tJg/ml (LDL), and LDL plus control HDL from normal subjects at 350 g/ml (+HDL).
Other cocultures were incubated with the control LDL together with varying amounts (micrograms shown on the abscissa) of either D-2F, or L-2F (third panel from the left, 2F) or D-37-pA or L-37pA (last panel on the right, 37pA). The data represent mean SD of 0 values obtained from quadruplicate cocultures. Values for HDL or added peptides were N all significantly different from LDL alone (first panel on the left) at the level ofp 0.01.
O [0049] Figures 10A and 10B illustrate that feeding mice ApoA-1 peptide mimetics IN of this invention renders red cells resistant to n vitro lysis. Figures 10A and 10B show 1 5 the results of in vitro red cell lysis assay at 18 hours (Figure 10A) and at 48 hours (Figure The asterisks reflect the presence of a significant difference (p<0001) between the red cell lysis for animals that received the vehicle versus those that received the peptides.
C [0050] Figure 11 shows that feeding mice ApoA-l mimetic D peptides of this r- invention renders circulating LDL resistant to oxidation. Groups of LDL receptor- 0 10 deficient mice were administered the D-peptides or the saline vehicle by gavage.
Each animal was given 100pl of saline, 100 pg/100l l of peptide D-2F or peptide D-37pA.
Blood was collected from retroorbital sinus under mild anesthesia 17 hrs later. LDL was isolated from plasma by FPLC. Cocultures of artery Wall cells were incubated with medium alone (NO ADDITION), control LDL from normal subjects (LDL), LDL plus control HDL from normal subjects Other cocultures were incubated with murine LDL following gavage with saline (SALINE LDL), with D-2F (D-2F LDL) or with D- 37pA peptide (D-37pA LDL). The cocultures were incubated for 4 hrs at 37 0 C in the presence of 10% LPDS. The supernatants were then discarded, the cocultures were washed and incubated with culture medium without serum or LPDS for an additional 4 hrs. This conditioned medium was collected and analyzed for monocyte chemotactic activity. The values are mean+-SD of quadruplicate cocultures. The asterisks indicate p<0.001.
[0051] Figure 12 illustrates the results of a chemotaxis assay comparing lipoproteins from mice given the D-form and or L-form peptides by gavage.
[0052] Figure 13A illustrates the results of a chemotaxis assay comparing control HDL and HDL from mice given the D-peptide by gavage. Figure 13B illustrates the results of a chemotaxis assay comparing LDL and VLDL/IDL from mice given the Dpeptide by gavage.
[0053] Figures 14A and 14B show electrophoresis of 2F indicating its selfassociation. Figure 14A: SDS PAGE of 2F. Lane 1 shows the molecular weight standard and lane 2 shows the band corresponding to 2F (molecular weight is 2242) S moving slightly lower than the lowest molecular weight standard 5-2. 5 kDa). Figure C 14B: Non-denaturing PAGE showing the mobilities of 100 g/ml (lane 2) and 0 250g/ml (lane 1) of2F indicating self-association in solution. Lane 3 shows the mobility of the high molecular weight standard.
[0054] Figure 15 shows that the homologous series ofpeptides stabilize the hexr phase transition of DiPoPE bilayers. Shift in TH of DiPoPE as a function of the mole fraction of added peptide. Measured by DSC at a heating scan rate of 37 0 2F; O 3F3; E 4F; D 5F;; V 6F; A 7F; S apo A-I.
[0055] Figure 16: Relative right angle light scattering monitoring of the dissolution of EPC MLVs by homologous series of peptides as a function of time. A representative EPC MLV clarification curve is shown for each of the homologous peptides. An equimolar concentration of peptide and EPC was used (105pM). Both excitation and emission wavelengths were 400nm. Triton X-100 achieved complete dissolution at a final concentration of ImM. 2F; 3F 3 ;-L 3F 14 4F;-A- 5F;-V- 6F; V- 7F; human apo A-I; -*-Triton X-100.
[0056] Figure 17 illustrates LCAT activating ability of homologous peptides.
Histograms representing activation of LCAT by the F-peptides. LCAT activity was measured using small unilamellar vesicles of EPC-cholesterol and the activity is represented as a percentage compared to that of apo A-I activity, where apo A-I activity is taken to be 100%. Each value represents an average value from triplicates. The peptide concentration used was [0057] Figure 18 shows that LDL-induced monocyte chemotaxis was inhibited by the homologous series ofpeptides. LDL alone or LDL incubated with either human HDL or the homologous series of peptides was added to the human artery wall cell cocultures for 8h in the presence of 10% LPDS. The supernatants were removed and the cocultures were washed with culture medium without serum or LPDS. The conditioned medium was then collected and analyzed for monocyte chemotactic activity. The data represent mean +SEM values (n=9 in each case). By pair-wise-comparisons with LDL all peptides except the 3F peptides were significantly more effective (at least p<0. 001, signified by and Comparisons between all peptides were analyzed by one-way ANOVA. The asterisk indicates that peptides 4F, 5F and 6F were significantly more effective than the -18- 0 homologues 2F and 7F 05 by Duckett comparison). The bracket indicates no
O
l significant difference in the ability to inhibit LDL-induced chemotaxis among these three O peptides.
1O 0058] Figure 19 shows that Influenza A infection causes an increase in hepatic oxidized phospholipids two days after infection. C57BL/6 mice on a chow diet were infected with a dose of influenza A virus intranasally such that no viremia resulted as t described by Van Lenten et al. (2001) Circulation, 103: 2283-2288. Zero, 2, 3, 5, 7, and 9 Sdays after infection the livers were removed and oxidized phospholipid content determined by ESI-MS.
O
[0059] Figure 20 shows that D-4F prevents the decrease in paraoxonase activity after Influenza A infection. Some of the mice described in Figure 19 were injected intraperitoneally with 20utg daily of D-4F and the others were injected with phosphate buffered saline (PBS). Paraoxonase activity (PON) was measured in the plasma at zero, 2, 7, and 9 days after infection.
[0060] Figure 21 shows that D-4F prevents the induction of oxidized phospholipids in aortas of mice infected with Influenza A virus. Some of the mice described in Figure 19 were injected intraperitoneally with 20pg daily of D-4F and the others were injected with phosphate buffered saline (PBS). The aortas of the mice were harvested at days zero, 2, 7 and 9 days after infection and oxidized phospholipid content was determined by ESI-MS.
[0001] Figures 22A through 22C show that, after oral administration, D-4F, but not L-4F remains intact in the circulation of LDL receptor null mice and enhances HDL's ability to protect LDL against oxidation by human artery wall cells and reduces LDLinduced monocyte chemotactic activity. Figure 22A: Peptides L-4F and D-4F were radiolabeled using the lodo-bead reagent and were administered by oral gavage to LDL receptor null mice (100 il of saline containing 100 gg of unlabeled peptide plus 125Ilabeled peptide with specific activity of 11x10 6 cpm per ulg peptide per animal, n=3).
Blood was collected after 4 hrs, plasma was separated, delipidated and analyzed by reverse phase HPLC as described in reference 12. Panels B and C: Peptides L-4F and D-4F (100 pg in 100 pl of saline per animal) were administered to LDL receptor deficient mice by oral gavage. Blood was collected after 6 hrs, plasma HDL and LDL were isolated by 0 gel filtration (FPLC) and examined in the cocultures. Figure 22B: The ability of mouse Cl and human HDL to protect a control human LDL (hLDL) against oxidation by human 0 artery wall cells and to inhibit LDL-induced monocyte chemotactic activity is shown.
Z Human LDL at 200 jpg protein/mi was added to human artery wall cell cocultures together C 5 with human HDL (hHDL) at 350 jig protein/ml or mouse HDL (mHDL) at 100 jig cholesterol/ml taken from mice that received saline (Saline Rx) or L-4F (L-4F Rx) or Dt 4F (D-4F Rx) and monocyte chemotactic activity was determined as described herein.
Figure 22C: The ability of mouse LDL to induce monocyte chemotactic activity is shown.
CI The assay controls are shown on the left as in Panel B. On the right, mouse LDL (mLDL) O 10 was isolated from mice receiving saline (Saline Rx) or L-4F (L-4F Rx) or D-4F (D-4F Rx) C and was added at 100 jig cholesterol/ml to the artery wall cell cocultures without HDL and monocyte chemotactic activity determined. Values are mean SD for four wells in two independent experiments.
[0002] Figures 23 shows that oral administration of D-4F dramatically reduces lesions in LDL receptor null mice on a Western diet. Groups of LDL receptor null mice were placed on a Western diet and were given by oral gavage, 100 jl of saline alone (Saline), n 4 animals, or 100 pl of liposomes without D-4F (Liposomes), n 5 animals, or 2.5 mg D-4F peptide in 100 pl liposomes (D-4F in liposomes), n 6 animals, twice daily for 6 weeks. The mice were bled, and subsequently sacrificed, aortic root fixed, sectioned and the extent of Oil Red O staining in fatty streak lesions quantified.
[0003] Figure 24 shows that oral administration of D-4F dramatically reduces lesions in apo E null mice on a chow diet. At 4 weeks of age, D-4F was added to the drinking water of some of the apo E null mice to give a concentration of 1 mg/ml of D-4F 4 mice). D-4F at a concentration of 2 mg/ml was added to the drinking water of another group of mice (n 4 mice) and no peptide was added to the drinking water of a third group of mice (n 5 mice). The mice all consumed approximately 2.5 ml of water per day so that one group received no peptide (Water), a second group received 2.5 mg D- 4F/mouse/day (2.5 mg D-4F) and the third group received 5.0 mg D-4F/mouse/day mg D-4F). All were continued on the chow diet for 5 weeks at which time the mice were bled and subsequently sacrificed, aortic root fixed, sectioned and the extent of Oil Red O staining in fatty streak lesions quantified.
DETAILED DESCRIPTION I. Mitigation of a symptom of atherosclerosis.
0 Z [0060] This invention pertains to the discovery that synthetic peptides designed to I, mimic the class A amphipathic helical motif (Segrest et al. (1990) Proteins: Structure, Function, and Genetics 8: 103-117) are able to associate with phospholipids and exhibit many biological properties similar to human apo-A-I. In particular, it was a discovery of this invention that when such peptides are formulated using D amino acids, the peptides n show dramatically elevated serum half-lives and, particularly when the amino and/or carboxy termini are blocked, can even be orally administered.
C1 10 [0061] Moreover, it was a surprising discovery of this invention that such D-form peptides retain the biological activity of the corresponding L-form peptide. In vivo animal studies using such D-form peptides showed effective oral delivery, elevated serum halflife, and the ability to mitigate or prevent/inhibit one or more symptoms of atherosclerosis.
[0062] We discovered that normal HDL inhibits three steps in the formation of mildly oxidized LDL. In those studies (see, copending application USSN 09/541,468, filed on March 31, 2000) we demonstrated that treating human LDL in vitro with apo A-I or an apo A-I mimetic peptide (37pA) removed seeding molecules from the LDL that included HPODE and HPETE. These seeding molecules were required for cocultures of human artery wall cells to be able to oxidize LDL and for the LDL to induce the artery wall cells to produce monocyte chemotactic activity. We also demonstrated that after injection of apo A-I into mice or infusion into humans, the LDL isolated from the mice or human volunteers after injection/infusion of apo A-I was resistant to oxidation by human artery wall cells and did not induce monocyte chemotactic activity in the artery wall cell cocultures.
[0063] The protective function of the D peptides of this invention is illustrated in Figure 1 through 5. Figure 1, panels A, B, C, and D show the association of 14C-D-5F with blood components in an ApoE null mouse. It is also demonstrated herein, that HDL from mice that were fed an atherogenic diet and injected with PBS failed to inhibit the oxidation of human LDL and failed to inhibit LDL-induced monocyte chemotactic activity in human artery wall coculures. In contrast, HDL from mice fed an atherogenic diet and injected daily with peptides described herein was as effective in inhibiting human LDL -21- -w r- 0 oxidation and preventing LDL-induced monocyte chemotactic activity in the cocultures as CI was normal human HDL (Figures 2A and 2B). In addition, LDL taken from mice fed the O atherogenic diet and injected daily with PBS was more readily oxidized and more readily Z induced monocyte chemotactic activity than LDL taken from mice fed the same diet but
\D
C 5 injected with 20 pg daily of peptide 5F. The D peptide did not appear to be immunogenic (Figure 4).
[0064] The in vitro responses of human artery wall cells to HDL and LDL from c mice fed the atherogenic diet and injected with a peptide according to this invention are consistent with the protective action of shown by such peptides in vivo. Despite, similar 0 10 levels of total cholesterol, LDL-cholesterol, IDL+VLDL-cholesterol, and lower HDLcholesterol as a percent of total cholesterol, the animals fed the atherogenic diet and injected with the peptide had significantly lower lesion scores (Figure The peptides of this invention thus prevented progression of atherosclerotic lesions in mice fed an atherogenic diet.
[0065] Thus, in one embodiment, this invention provides methods for ameliorating and/or preventing one or more symptoms of atherosclerosis. The methods preferably involve administering to an organism, preferably a mammal, more preferably a human one or more of the peptides of this invention (or mimetics of such peptides). The peptide(s) can be administered, as described herein, according to any of a number of standard methods including, but not limited to injection, suppository, nasal spray, time-release implant, transdermal patch, and the like. In one particularly preferred embodiment, the peptide(s) are administered orally as a syrup, capsule, or tablet).
[0066] The methods involve the administration of a single polypeptide of this invention or the administration of two or more different polypeptides. The polypeptides can be provided as monomers or in dimeric, oligomeric or polymeric forms. In certain embodiments, the multimeric forms may comprise associated monomers ionically or hydrophobically linked) while certain other multimeric forms comprise covalently linked monomers (directly linked or through a linker).
[0067] While the invention is described with respect to use in humans, it is also suitable for animal, e.g. veterinary use. Thus preferred organisms include, but are not limited to humans, non-human primates, canines, equines, felines, porcines, ungulates, CI largomorphs, and the like.
0 [0068] The methods of this invention are not limited to humans or non-human animals showing one or more symptom(s) of atherosclerosis hypertension,, plaque c' 5 formation and rupture, reduction in clinical events such as heart attack, angina, or stroke, high levels of plasma cholesterol, high levels of low density lipoprotein, high levels of l very low density lipoprotein, or inflammatory proteins, etc.), but are useful in a prophylactic context. Thus, the peptides of this invention (or mimetics thereof) may be C administered to organisms to prevent the onset/development of one or more symptoms of 10 atherosclerosis. Particularly preferred subjects in this context are subjects showing one or C more risk factors for atherosclerosis family history, hypertension, obesity, high alcohol consumption, smoking, high blood cholesterol, high blood triglycerides, elevated blood LDL, VLDL, IDL, or low HDL, diabetes, or a family history of diabetes, high blood lipids, heart attack, angina or stroke, etc.).
[0069] In addition to methods of use of the atherosclerosis-inhibiting peptides of this invention, this invention also provides the peptides themselves, the peptides formulated as pharmaceuticals, particularly for oral delivery, and kits for the treatment and/or prevention of one or more symptoms of atherosclerosis.
H. Mitigation of a symptom of atheroscloerosis associated with an acute inflammatory response.
[0070] The atherosclerosis-inhibiting peptides of this invention are also useful in a number of other contexts. In particular, we have observed that cardiovascular complications atherosclerosis, stroke, etc.) frequently accompany or follow the onset of an acute phase inflammatory response. Such an acute state inflammatory response is often associated with a recurrent inflammatory disease leprosy, tuberculosis, systemic lupus erythematosus, and rheumatoid arthritis), a viral infection influenza), a bacterial infection, a fungal infection, an organ transplant, a wound or other trauma, an implanted prosthesis, a biofilm, and thelike. [0071] It was a surprising discovery of this invention that administration of one or more of the peptide described herein, can reduce or prevent the formation of oxidized 0 phospholipids during or following an acute phase response and thereby mitigate or 0 CN eliminate cardiovascular complications associated with such a condition.
0 [0072] Thus, for example, we have demonstrated that a consequence of influenza IND infection is the dimunition in paraoxonase and platelet activating acetylhydrolase activity in the HDL. Without being bound by a particular theory, we believe that, as a result of the loss of these HDL enzymatic activities and also as a result of the association of prot n oxidant proteins with HDL during the acute phase response, HDL is no longer able to r prevent LDL oxidation and was no longer able to prevent the LDL-induced production of monocyte chemotactic activity by endothelial cells.
[0073] We observed that in a subject injected with very low dosages of the polypeptides of this invention 20 micrograms for mice) daily after infection with the influenza A virus paraoxonase levels did not fall and the biologically active oxidized phospholipids were not generated beyond background. This indicates that D-4F (and/or other peptides of this invention) can be administered orally or by injection) to patients with known coronary artery disease during influenza infection or other events that can generate an acute phase inflammatory response due to viral infection, bacterial infection, trauma, transplant, various autoimmune conditions, etc.) and thus we can prevent by this short term treatment the increased incidence of heart attack and stroke associated with pathologies that generate such inflammatory states.
[0074] Thus, in certain embodiments, this invention contemplates administering one or more of the peptides of this invention to a subject at risk for, or incurring, an acute inflammatory response and/or at risk for or incurring a symptom of atherosclerosis.
[0075] Thus, for example, a person having or at risk for coronary disease may prophylactically be administered a polypeptide of this invention during flu season. A person (or animal) subject to a recurrent inflammatory condition, e.g. rheumatoid arthritis, various autoimmune diseases, etc., can be treated with a polypeptide of this invention to mitigate or prevent the development of atherosclerosis or stroke. A person (or animal) subject to trauma, e.g. acute injury, tissue transplant, etc. can be treated with a polypeptide of this invention to mitigate the development of atherosclerosis or stroke.
[0076] In certain instances such methods will entail a diagnosis of the occurrence or risk of an acute inflammatory response. The acute inflammatory response typically -24- 0 involves alterations in metabolism and gene regulation in the liver. It is a dynamic N homeostatic process that involves all of the major systems of the body, in addition to the O immune, cardiovascular and central nervoqs system. Normally, the acute phase response Z lasts only a few days; however, in cases of chronic or recurring inflammation, an aberrant C 5 continuation of some aspects of the acute phase response may contribute to the underlying tissue damage that accompanies the disease, and may also lead to further complications, In for example cardiovascular diseases or protein deposition diseases such as amyloidosis.
[0077] An important aspect of the acute phase response is the radically altered biosynthetic profile of the liver. Under normal circumstances, the liver synthesizes a characteristic range of plasma proteins at steady state concentrations. Many of these proteins have important functions and higher plasma levels of these acute phase reactants (APRs) or acute phase proteins (APPs) are required during the acute phase response following an inflammatory stimulus. Although most APRs are synthesized by hepatocytes, some are produced by other cell types, including monocytes, endothelial cells, fibroblasts and adipocytes. Most APRs are induced between 50% and several-fold over normal levels. In contrast, the major APRs can increase to 1000-fold over normal levels. This group includes serum amyloid A (SAA) and either C-reactive protein (CRP) in humans or its homologue in mice, serum amyloid P component (SAP). So-called negative APRs are decreased in plasma concentration during the acute phase response to allow an increase in the capacity of the liver to synthesize the induced APRs.
[0078] In certain embodiments, the acute phase response, or risk therefore is evaluated by measuring one or more APPs. Measuring such markers is well known to those of skill in the art, and commercial companies exist that provide such measurement AGP measured by Cardiotech Services, Louisville, KY).
II. Mitigation of a symptom or condition associated with coronary calcification and osteoporosis.
[0079] We have also identified oxidized lipids as a cause of coronary calcification and osteoporosis. Moreover, without being bound to a particularly theory, we believe the same mechanisms are involved in the pathogenesis of calcific aortic stenosis.
[0080] Thus, in certain embodiments, this invention contemplates the use of the peptides described herein to inhibit or prevent a symptom of a disease such as polymyalgia 0 rheumaticapolyarteritis nodosa, scleroderma, idiopathic pulmonary fibrosis, chronic Cl obstructive pulmonary disease, Alzheimers Disease, AIDS, coronary calcification, calcific O aortic stenosis, osteoporosis, and the like.
C IV. Preferred peptides and their preparation.
5 Preferred peptides.
[0081] It was a discovery of this invention that class A peptides, are capable of mitigating one or more symptoms of atherosclerosis. Class A peptides are characterized by formation of an a-helix that produces a segregation of polar and non-polar residues N. thereby forming a polar and a nonpolar face with the positively charged residues residing at the polar-nonpolar interface and the negatively charged residues residing at the center of the polar face (see, Anantharamaiah (1986) Meth. Enzymol, 128: 626-668). It is noted that the fourth exon of apo A-I, when folded into 3.667 residues/turn produces a class A amphipathic helical structure.
[0082] One particularly preferred class A peptide, designated 18A (see, Table 1, and also Anantharamaiah (1986) Meti. Enzymol, 128: 626-668) was modified as described herein to produce peptides orally administratable and highly effective at inhibiting or preventing one or more symptoms of atherosclerosis. Without being bound by a particular theory, it is believed that the peptides of this invention act in vivo may by picking up seeding molecule(s) that mitigate oxidation of LDL.
[0083] We determined that increasing the number of Phe residues on the hydrophobic face of 18A would theoretically increase lipid affinity as determined by the computation described by Palgunachari et al. (1996) Arteriosclerosis, Thrombosis, Vascular Biology 16: 328-338. Theoretically, a systematic substitution of residues in the nonpolar face of 18A with Phe could yield six peptides. Peptides with an additional 2, 3 and 4 Phe would have theoretical lipid affinity values of 13, 14 and 15 units, respectively. However, the 1 values jumped four units if the additional Phe were increased from 4 to 5 (to 19 X units). Increasing to 6 or 7-Phe would produce a less dramatic increase (to 20 and 21 X units, respectively). Therefore, we chose 5 additional Phe (and hence the peptides designation as 5F). In one particularly preferred embodiment, the -26peptide was blocked in that the amino terminal residue was acetylated and the carboxyl NI terminal residue was ainidated.
0 [0084] The new class A peptide analog, 5F inhibited lesion development in atherosclerosis-susceptible mice. The new peptide analog, 5F, was compared with mouse apo A-I (MoA-I) for efficacy in inhibiting diet-induced atherosclerosis in these mice using peptide dosages based on the study by Levine et al. (Levine et al. (1993) Proc. Natl.
Acad Sci. USA 90:12040-12044).
[0085] A number of other class A peptides were also produced and showed varying, but significant degrees of efficacy in mitigating one or more symptoms of atherosclerosis. A number of such peptides are illustrated in Table 1.
Table 1. Preferred peptides for use in this invention.
Peptide Amino Acid Sequaence SEQ ID Name
NO.
18A D-W-L-K--A-F-Y-D-K-V-A-E-K-L-K-E-A-F 1 2F Ac-D-W-L-K-A-F-Y-D-K-V-A-E-K-LK-E-A-F-NH 2 2 3F Ac-D-W-F-K-A-F--Y-D-K-V-A-E-K-L-K-E-A-F-Ni 2 3 3F14 Ac-D-W-L-K-A--F-Y-D-K-V-A-E-K-F-K-E-A-F-NH 2 4 4F Ac-D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-NH 2 Ac-D-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F-NH 2 6 6F Ac-D-W-L--K-A-F-Y-D-K-F-F-E-K-F-K-E-F-F-NH 2 7 7F AC-D-W-F-K-A-F-Y--D-K-F-F-E-K-F-K-E-F-F-NHi 2 8 Ac-D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F-NH 2 9 Ac-D-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-A-F-NH 2 Ac-D-W-L-K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F-N1 2 11 Ac-D-W-L-K-A-F-Y-D-K-V-A--E-K-F-K-E-F-F-NH 2 12 Ac-D-W-Ir-K-A-F--Y-D-K-V-F-E-K-F-K-E-F--NH 2 13 Ac-E-W-L-K-L-F-Y-E-K-V-L-E-K-F-K-E-A-F-NH 2 14 Ac-E-W-L-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-NH 2 Ac-E-W-L-K-A--F-Y-D-K-V-A-E-K-L-K-E-F-F-NH 2 16 Ac-E-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-A-F-NH 2 17 Ac-E-W-L-K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F-NH 2 18 Ac-E-W-L-K-A-F-Y-D-K-V-A-E-K-F-K-E-F-F-NH 2 19 Ac-E-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F-NH,
AC-A-F-Y-D-K--V-A-E-K-L-K-E-A-F-NH
2 21 Ac-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-NH 2 22 Ac-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-N{ 2 23 Ac-A-F-Y-D-K-F-F-E-K-F-K-E-F-F-NH 2 24 -27oAc-A-F-Y-D-K-F-F-E-K-F-K-E-F-F-NH 2 0cAFYDKVAEKFKEAFN22 c-iAc-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-N1 2 26 Ac-A-F-Y-D-K-V-A-E-K-L-K-E--F-N1 2 27 IN Ac-A-F-Y-D-K-V-F-E-K-F-K-E-A-F-NH 2 28 zcAFYDKVAEKFKEFFN23 Ac--A-F-Y-D-K-V-F-E-K--K-E-F-NH 2 29 (NAc-A-F-Y-D-K-V-A-E-K-F-K-E-F-F-NH 2 Ac--A-F-Y-D-K-V--E-K-F-K-E-F--NH 2 31 Ac-L-F-Y-E-K-V-L-E-K--K-E---F-NH 2 32 Ac-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-NH 2 33 Ac-A-F-Y-D-K-V-A-E-K-L-K-E-F-F-N1 2 34 AcAFYDK---n---EFFN23 Ci Ac-A-F-Y-D-K-V-F-E-K-F-K-E-A-F-NH 2 S AcWL--A-F-Y-D)-K--V-F-E-K-L-K-E-F-F-NH 2 36 Ac-DW-Ac-A-F-Y-D-K-V-A-E-K-F-K-E-F-F-N1 2 37 AcDW---A-F-Y-D-K-V-F-E-K-F-K-E-F-F-NH 2 38
L-NR
2 39 Ac-D-W-F-K--A-F-Y--E-K-V-A-E-K-L-K-E-F-F-NH 2 Ac-D-W-F-K-A-F-Y-E-K-F-F--E-K-F-K-E-F-F-NH 2 41 Ac-E-W-L-K--A-L-Y-E-K-V-A-E-K-L-K-E----NH 2 42 Ac-E-W-L-K-A-F-Y-E-K-V-A--E-K-L-K-E---F-NH 2 43 Ac-E-W-F-K-A-F-Y-E-K-V-A-E-K-L-K-E-F-F-NH 2 44 Ac-E-W-L-K-A-F-Y-E-K-V-F-E-K--K-E--F-NH 2 Ac-E-W-L-K--A-F-Y-E-K-F-F--E-K-F-K-E---N1 2 46 Ac-E--WF-K-A-F--Y-E-K-F-F-E-K-F-K-E--F-NH 2 47 Ac -D-F-L-K-A-W-Y-D-K-V-A-E-K-L-K-E--W-NH 2 48 Ac- E-F-L-K-A-W-Y- E-A-W-NH 2 49 Ac 2 2 51 Ac -D-K-L-K-A-F-Y-D-K-VF-AE----K-E-AF-LNH 2 52 Ac-D-K-W-K-A--V-D--K-F-A-E-A-F-K-E-F-L-NH 2 53 Ac-E-K-L-K-A-F-Y-E-K-VF-AE-A-FK-E-A-F-NH 2 54 2 2 56 Ac-D-W-L-K-A-F-Y-D-K-V-F--K-L-K-E--F-N1 2 58 Ac-E-W-L-K-A-F-Y-E-K-V-F--K-L-K-E-F-F-NH 2 59 Ac-D-W-L-R-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-NH 2 Ac-E-W-L-R-A-F-Y-E-K-V-A-E-K-L-K-E-A-F-NH 2 61 Ac-D-W-L-K-A-F-Y-D-R-V-A-E-K-L-K-E-A-F-NH 2 62 -28o Ac-E-W-L-K-A-F-Y-E-K-V-A-E-R-L-K-E-A-F-NH 2 0 Ac-D-W-L-K-A-F-Y-D-K-V-A-E-K-L-R-E-A-F-NH2I 66 AcEWLKAFY--------REAFN26 Ac-E-W-L--K-A-F-Y-E-K-V-A-E-K-L-R-E-A-F-NHz 67 IN Ac-D-W-L-K-A-F-Y-D-R-V-A-E-R-L-K-E-A-F-N~H 69 zcDWLRAFYDKVAEKLREAFN27 Ac-E-W-L-K-A-F-Y-E-R-V-A-E-R-L-K-E-A-F-NH 2 69 CiAc-D-W-L-R-A-F-Y-D-K-V-A-E-K-L-R-E-A-F-NH 2 Ac-E-W-L-R-A-F-Y-E-K-V-A-E-K-L-R-E-A-F-NH 2 71 Ac-D-W-L-R-A-F-Y-D-R-V-A-E-'Z-L-K-E-A-F-NH2 72 __Ac-E-W-L-R-A-F-Y-E-R-V-A-E-K-L-K-E-A-F-NH 2 73 Ac-D-W-L-K-A-F-Y-D-K-V-A-E-R-L-R-E-A-F-N~H 74 CiAc-E-W-L-K-A-F-Y-E-K-V-A-E-R-L-R--E-A-F-NH2 o D-W-L--A-F-Y-D-K-V-A-E--L-K-E-A-F-NHW--K 76 O Ac-E-L-RA-F-Y- R-L-K-E-A-F -N27 D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-P-D-W-L-K- 78
A-F-Y-D-K-V-A-E-K--L-K-E-A-F'
79
A-F-Y-D-K-V-A-E-K-L--K-E-F-F
A-F-Y-D-K-V--A-E-K-L-K-E-A-F
81
A-F--Y-D-K-VF--E-W-L-K-E-A-F
82
A-V-Y-D-K-F-A-E-A-F-K-E-AFL
83
A-F-VY-D-K-V---K-F-K-E-A-F
D-W-L-K-A-F-Y-D-K V-FE-K--K-E-F-F-P-D-W-L-K- 84
F-F
'Linkers are underlined.
[0086] While various peptides of Table 1, are ilustrated with an acetyl group protecting the amino terminus and an amnide group protecting the carboxyl terminus, either or both of these protecting groups may be eliminated and/or substituted with another protecting group as described herein. In particularly preferred embodiments, the peptides comprise one or more D-form amino acids as described herein. In certain embodiments, every amino acid every enantiomeric amino acid) of the peptides of Table 1 is a D form amino acid.
0 [0087] It is also noted that Table 1 is not fully inclusive. Using the teaching provided herein, other suitable peptides can routinely be produced by conservative or O semi-conservative substitutions D replaced by extensions, deletions, and the like).
Z Thus, for example, one embodiment utilizes truncations of any one or more of peptides
IND
N 5 identified by SEQ ID Nos:2-20 and 39-85. Thus, for example, SEQ ID NO: 21 illustrates a peptide comprising 14 amino acids from the C-terminus of 18A comprising one or more I D amino acids, while SEQ ID NOS:22-38 illustrate other truncations. Longer peptides are also suitable. Such longer peptides may entirely form a class A amphipathic helix, or the class A amphipathic helix (helices) may form one or more domains of the peptide. In C 10 addition, this invention contemplates multimeric versions of the peptides. Thus, for example, the peptides illustrated in Table 1 can be coupled together (directly or through a linker a carbon linker, or one or more amino acids) with one or more intervening amino acids). Illustrative polymeric peptides include 18A-Pro-18A and the peptides of SEQ ID NOs:79-85 preferably comprising one or more D amino acids, more preferably with every amino acid a D amino acid as described herein and/or having one or both termini protected.
[0088] It was a surprising discovery of this invention that, when the class A peptides as illustrated in Table 1) incorporated D amino acids they retained their activity and, but could be administered orally. Moreover this oral administration resulted in relatively efficient uptake and significant serum half-life thereby providing an efficacious method of mitigating one or more symptoms of atherosclerosis.
[0089] Using the teaching provided herein, one of skill can routinely modify the illustrated class A peptides to produce other suitable class A peptides of this invention.
For example, routine conservative or semi-conservative substitutions E for D) can be made of the existing amino acids. The effect of various substitutions on lipid affinity of the resulting peptide can be predicted using the computational method described by Palgunachari et al. (1996) Arteriosclerosis, Thrombosis, Vascular Biology 16: 328-338.
The peptides can be lengthened or shortened as long as the class A a-helix structure is preserved. In addition, substitutions can be made to render the resulting peptide more similar to peptide(s) endogenously produced by the subject species.
[0090] In certain embodiments, the peptides of this invention comprise forms of the peptides described in U.S. Patent 4,643,988, more preferably forms having one r- 0 or both termini coupled to protecting groups. Such peptides include peptides having the CN formula AI-BI-B 2 -Ci-D-B 3
-B
4
-A
2
-C
2
-B
5
-B
6
-A
3
-C
3
-B
7
-C
4
-A
4 -B8-B 9 (SEQ ID NO:86) O wherein Ai, Az, A 3 and A 4 are independently aspartic acid or glutamic acid, or Z homologues or analogues thereof; BI, B 2
B
3
B
4
B
5
B
6
B
7 B8 and B 9 are independently
INO
CN 5 tryptophan, phenylalanine, alanine, leucine, tyrosine, isoleucine, valine or anaphthylalanine, or homologues or analogues thereof; C 1
C
2
C
3 and C 4 are independently tt lysine or arginine, and D is serine, threonine, alanine, glycine, histidine, or homologues or analogues thereof; provided that, when A 1 and Az are aspartic acid, A 3 and A 4 are CI glutamic acid, B 2 and B 9 are leucine, B 3 and B 7 are phenylalanine, B 4 is tyrosine, Bs is 0 10 valine, B 6 Bg, and D are alanine, and C 1
C
2
C
3 and C 4 are lysine, B 1 is not Tryptophan, CI where at one enantiomeric amino acid is a form amino acids. Preferably at least of the enantiomeric amino acids are form, more preferably at least 80% of the enantiomeric amino acids are form, and most preferably at least 90% or even all of the enantiomeric amino acids are form amino acids.
[0091] While, in preferred embodiments, the peptides of this invention utilize naturally-occurring amino acids or D forms of naturally occurring amino acids, substitutions with non-naturally occurring amino acids methionine sulfoxide, methionine methylsulfonium, norleucine, episilon-aminocaproic acid, 4-aminobutanoic acid, tetrahydroisoquinoline-3-carboxylic acid, 8-aminocaprylic acid, 4-aminobutyric acid, Lys(N(epsilon)-trifluoroacetyl), a-aminoisobutyric acid, and the like) are also contemplated.
[0092] In addition to the class A peptides described herein, peptidomimetics are also contemplated herein. Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide.
These types of non-peptide compound are termed "peptide mimetics" or "peptidomimetics" (Fauchere (1986)Adv. Drug Res. 15: 29; Veber and Freidinger (1985) TINS p.392; and Evans et al. (1987) J. Med. Chem. 30: 1229) and are usually developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent therapeutic or prophylactic effect.
[0093] Generally, peptidomimetics are structurally similar to a paradigm polypeptide 5F described herein), but have one or more peptide linkages optionally -31- 0 replaced by a linkage selected from the group consisting of: -CH 2 NH-, -CH 2
-CH
2
-CH
2 C, -CH=CH- (cis and trans), -COCH 2
-CH(OH)CH
2
-CH
2 SO-, etc. by methods known in O the art and further described in the following references: Spatola (1983) p. 267 in Z Chemistry and Biochemisty ofA nino Acids, Peptides, and Proteins, B. Weinstein, eds.,
\D
CN 5 Marcel Dekker, New York,; Spatola (1983) Vega Data 1(3) Peptide Backbone Modifications. (general review); Morley (1980) Trends Pharnn Sci pp. 463-468 (general tt review); Hudson et al. (1979) Int J Pept Prot Res 14:177-185 (-CH 2 NH-, CH 2
CH
2 Spatola et al. (1986) Life Sci 38:1243-1249 (-CH 2 Hann, (1982) J Chem Soc Perkin CN Trans 1307-314 cis and trans); Almquist et al. (1980) JMed Chem. 23:1392- 0 10 1398 (-COCH 2 Jennings-White et al.(1982) Tetrahedron Lett. 23:2533 (-COCH 2 CN Szelke, M. et al., European Appln. EP 45665 (1982) CA: 97:39405 (1982) CH(OH)CH2-); Holladay et al. (1983) Tetrahedron Lett 24:4401-4404 (-C(OH)CH 2 and Hruby (1982) Life Sci., 31:189-199 (-CH 2 [0094] A particularly preferred non-peptide linkage is -CH 2 NH-. Such peptide mimetics may have significant advantages over polypeptide embodiments, including, for example: more economical production, greater chemical stability, enhanced pharmacological properties (half-life, absorption, potency, efficacy, etc.), reduced antigenicity, and others.
[0095] In addition, circularly permutations of the peptides described herein or constrained peptides (including cyclized peptides) comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods known in the art (Rizo and Gierasch (1992) Ann. Rev. Biochem. 61: 387); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the peptide.
Peptide preparation.
[0096] The peptides used in this invention are chemically synthesized using standard chemical peptide synthesis techniques or, particularly where the peptide does not comprise amino acid residues, are recombihantly expressed. Where the polypeptides are recombinantly expressed, a host organism bacteria, plant, fungal cells, etc.) in cultured in an environment where one or more of the amino acids is provided to the -32- Sorganism exclusively in a D form. Recombinantly expressed peptides in such a system 0 C1 then incorporate those D amino acids.
0 [0004] In preferred embodiments the peptides are chemically synthesized by any of a number of fluid or solid phase peptide synthesis techniques known to those of skill in the art. Solid phase synthesis in which the C-terminal amino acid of the sequence is attached to an insoluble support followed by sequential addition of the remaining amino acids in the sequence is a preferred method for the chemical synthesis of the polypeptides of this invention. Techniques for solid phase synthesis are well known to those of skill in the art and are described, for example, by Barany and Merrifield (1963) Solid-Phase Peptide Synthesis; pp. 3-284 in The Peptides: Analysis, Synthesis, Biology. Vol. 2: Special Methods in PeptideSynthesis, Part Merrifield et al. (1963) J. Am. Chem. Soc., 2149-2156, and Stewart et al. (1984) Solid Phase Peptide Synthesis, 2nd ed. Pierce Chem.
Co., Rockford, I11.
[0097] In a most preferred embodiment, the peptides are synthesized by the solid phase peptide synthesis procedure using a benzhyderylamine resin (Beckman Bioproducts, 0.59 mmol of NH 2 /g of resin) as the solid support. The COOH terminal amino acid t-butylcarbonyl-Phe) is attached to the solid support through a 4-(oxymethyl)phenacetyl group. This is a more stable linkage than the conventional benzyl ester linkage, yet the finished peptide can still be cleaved by hydrogenation. Transfer hydrogenation using formic acid as the hydrogen donor is used for this purpose. Detailed protocols used for peptide synthesis and analysis of synthesized peptides are describe in a miniprint supplement accompanying Anantharamaiah et al. (1985) J. Biol. Chem., 260(16): 10248- 10255.
[0098] It is noted that in the chemical synthesis of peptides, particularly peptides comprising D amino acids, the synthesis usually produces a number of truncated peptides in addition to the desired full-length product. The purification process HPLC) typically results in the loss of a significant amount of the full-length product.
[0099] It was a discovery of this invention that, in the synthesis of a D peptide in order to prevent loss in purifying the longest form one can dialyze and use the mixture and thereby eliminate the last HPLC purification. Such a mixture loses about of the potency of the highly purified product per wt of protein product), but the mixture contains about 6 times more peptide and thus greater total activity.
D-form amino acids.
[0100] D-amino acids are incorporated at one or more positions in the peptide simply by using a D-form derivatized amino acid residue in the chemical synthesis. Dform residues for solid phase peptide synthesis are commercially available from a number of suppliers (see, Advanced Chem Tech, Louisville; Nova Biochem, San Diego; Sigma, St Louis; Bachem California Inc., Torrance, etc.). The D-form amino acids can be incorporated at any position in the peptide as desired. Thus, for example, in one embodiment, the peptide can comprise a single D-amino acid, while in other embodiments, the peptide comprises at least two, generally at least three, more generally at least four, most generally at least five, preferably at least six, more preferably at least seven and most preferably at least eight D amino acids. In particularly preferred embodiments, essentially every other (enantiomeric) amino acid is a D-form amino acid.
In certain embodiments at least 90%, preferably at least 90%, more preferably at least of the enantiomeric amino acids are D-form amino acids. In one particularly preferred embodiment, essentially every enantiomeric amino acid is a D-form amino acid.
Protecting groups.
[0101] In certain embodiments, the one or more R-groups on the constituent amino acids and/or the terminal amino acids are blocked with a protecting group. Without being bound by a particular theory, it was a discovery of this invention that blockage, particularly of the amino and/or carboxyl termini of the subject peptides of this invention greatly improves oral delivery and significantly increases serum half-life.
[0102] A wide number of protecting groups are suitable for this purpose. Such groups include, but are not limited to acetyl, amide, and alkyl groups with acetyl and alkyl groups being particularly preferred for N-terminal protection and amide groups being preferred for carboxyl terminal protection. In certain particularly preferred embodiments, the protecting groups include, but are not limited to alkyl chains as in fatty acids, propeonyl, formyl, and others. Particularly preferred carboxyl protecting groups include amides, esters, and ether-forming protecting groups. In one preferred embodiment, an -34r- 0 acetyl group is used to protect the amino terminus and an amide group is used to protect the carboxyl terminus. These blocking groups enhance the helix-forming tendencies of the S peptides. Certain particularly preferred blocking groups include alkyl groups of various Z lengths, e.g. groups having the formula: CH 3
-(CH
2 )n-CO- where n ranges from about 1 to 5 about 20, preferably from about 1 to about 16 or 18, more preferably from about 3 to about 13, and most preferably from about 3 to about tIt [0103] In certain particularly preferred embodiments, the protecting groups include, but are not limited to alkyl chains as in fatty acids, propeonyl, formyl, and others.
Particularly preferred carboxyl protecting groups include amides, esters, and ether-forming protecting groups. In one preferred embodiment, an acetyl group is used to protect the amino terminus and an amide group is used to protect the carboxyl terminus. These blocking groups enhance the helix-forming tendencies of the peptides. Certain particularly preferred blocking groups include alkyl groups of various lengths, e.g. groups having the formula: CH 3
-(CH
2 where n ranges from about 3 to about 20, preferably from about'3 to about 16, more preferably from about 3 to about 13, and most preferably from about 3 to about [0104] Other protecting groups include, but are not limited to Fmoc, tbutoxycarbonyl (t-BOC), 9-fluoreneacetyl group, 1-fluorenecarboxylic group, 9florenecarboxylic group, 9-fluorenone-l-carboxylic group, benzyloxycarbonyl, Xanthyl (Xan), Trityl (Trt), 4-methyltrityl (Mtt), 4-methoxytrityl (Mmt), 4-methoxy-2,3,6trimethyl-benzenesulphonyl (Mtr), Mesitylene-2-sulphonyl (Mts), 4,4dimethoxybenzhydryl (Mbh),Tosyl (Tos), 2,2,5,7,8-pentamethyl chroman-6-sulphonyl (Pmc), 4-methylbenzyl (MeBzl), 4-methoxybenzyl (MeOBzl), Benzyloxy (BzlO), Benzyl (Bzl), Benzoyl 3-nitro-2-pyridinesulphenyl (Npys), 1-(4,4-dimentyl-2,6diaxocyclohexylidene)ethyl (Dde), 2,6-dichlorobenzyl (2,6-DiCl-Bzl), 2chlorobenzyloxycarbonyl 2-bromobenzyloxycarbonyl (2-Br-Z), Benzyloxymethyl (Bom), cyclohexyloxy (cHxO),t-butoxymethyl (Bum), t-butoxy (tBuO), t-Butyl (tBu), Acetyl and Trifluoroacetyl (TFA).
[0105] Protecting/blocking groups are well known to those of skill as are methods of coupling such groups to the appropriate residue(s) comprising the peptides of this invention (see, Greene et al., (1991) Protective Groups in.Organic Synthesis, 2nd ed., John Wiley Sons, Inc. Somerset, In one preferred embodiment, for example, ro acetylation is accomplished during the synthesis when the peptide is on the resin using
O
CN acetic anhydride. Amide protection can be achieved by the selection of a proper resin for O the synthesis. During the synthesis of the peptides described herein in the examples, rink Z amide resin was used. After the completion of the synthesis, the semipermanent
INO
Cl 5 protecting groups on acidic bifunctional amino acids such as Asp and Glu and basic amino acid Lys, hydroxyl of Tyr are all simultaneously removed. The peptides released from I such a resin using acidic treatment comes out with the n-terminal protected as acetyl and the carboxyl protected as NH 2 and with the simultaneous removal of all of the other CI protecting groups.
cI 10 V. Enhancing peptide uptake.
[0106] It was also a surprising discovery of this invention that when an all L amino acid peptide otherwise having the sequence of the, peptides of this invention) is administered in conjunction with the D-form a peptide of this invention) the uptake of the D-form peptide is increased. Thus, in certain embodiments, this invention contemplates the use of combinations of D-form and L-form peptides in the methods of this invention. The D-form peptide and the L-form peptide can have different amino acid sequences, however, in preferred embodiments, they both have amino acid sequences of peptides described herein, and in still more preferred embodiments, they have the same amino acid sequence.
[0107] It was also a discovery of this invention that concatamers of the class A amphipathic helix peptides of this invention are also effective in mitigating one or more symptoms of atherosclerosis. The monomers comprising the concatamers can be coupled directly together or joined by a linker. In certain embodiments, the linker is an amino acid linker a proline), or a peptide linker Gly 4 Ser 3 In certain embodiments, the concatamer is a 2 mer, more preferably a 3 mer, still more preferably a 4 mer, and most preferably 5 mer, 8 mer or 10 mer.
VI. Pharmaceutical formulations.
[0108] In order to carry out the methods of the invention, one or more peptides or peptide mimetics of this invention are administered, e.g. to an individual diagnosed as having one or more symptoms of atherosclerosis, or as being at risk for atherosclerosis.
The peptides or peptide mimetics can be administered in the "native" form or, if desired, in C the form of salts, esters, amides, prodrugs, derivatives, and the like, provided the salt, S ester, amide, prodrug or derivative is suitable pharmacologically, effective in the Z present method. Salts, esters, amides, prodrugs and other derivatives of the active agents C 5 may be prepared using standard procedures known to those skilled in the art of synthetic organic chemistry and described, for example, by March (1992) Advanced Organic 'Chemistry; Reactions, Mechanisms and Structure, 4th Ed. N.Y. Wiley-Interscience.
[0109] For example, acid addition salts are prepared from the free base using conventional methodology, that typically involves reaction with a suitable acid.
10 Generally, the base form of the drug is dissolved in a polar organic solvent such as methanol or ethanol and the acid is added thereto. The resulting salt either precipitates or may be brought out of solution by addition of a less polar solvent. Suitable acids for preparing acid addition salts include both organic acids, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. An acid addition salt may be reconverted to the free base by treatment with a suitable base. Particularly preferred acid addition salts of the active agents herein are halide salts, such as may be prepared using hydrochloric or hydrobromic acids. Conversely, preparation of basic salts of the peptides or mimetics are prepared in a similar manner using a pharmaceutically acceptable base such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine, or the like. Particularly preferred basic salts include alkali metal salts, the sodium salt, and copper salts.
[0110] Preparation of esters typically involves functionalization of hydroxyl and/or carboxyl groups which may be present within the molecular structure of the drug. The esters are typically acyl-substituted derivatives of free alcohol groups, moieties that are derived from carboxylic acids of the formula RCOOH where R is alky, and preferably is lower alkyl. Esters can be reconverted to the free acids, if desired, by using conventional hydrogenolysis or hydrolysis procedures.
-37r- [0111] Amides and prodrugs may also be prepared using techniques known to those skilled in the art or described in the pertinent literature. For example, amides may O be prepared from esters, using suitable amine reactants, or they may be prepared from an Z anhydride or an acid chloride by reaction with ammonia or a lower alkyl amine. Prodrugs C1 5 are typically prepared by covalent attachment of a moiety that results in a compound that is therapeutically inactive until modified by an individual's metabolic system.
r tr [0112] The peptides or mimetics identified herein are useful for parenteral, topical, oral, nasal (or otherwise inhaled), rectal, or local administration, such as by aerosol or transdermally, for prophylactic and/or therapeutic treatment of atherosclerosis and/or symptoms thereof. The pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration. Suitable unit dosage forms, include, but are not limited to powders, tablets, pills, capsules, lozenges, suppositories, patches, nasal sprays, injectibles, implantable sustained-release formulations, lipid complexes, etc.
[0113] The peptides and/or peptide mimetics of this invention are typically combined with a pharmaceutically acceptable carrier (excipient) to form a pharmacological composition. Pharmaceutically acceptable carriers can contain one or more physiologically acceptable compound(s) that act, for example, to stabilize the composition or to increase or decrease the absorption of the active agent(s).
Physiologically acceptable compounds can include, for example, carbohydrates, such as glucose, sucrose, or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins, protection and uptake enhancers such as lipids, compositions that reduce the clearance or hydrolysis of the active agents, or excipients or other stabilizers and/or buffers.
[0114] Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid. One skilled in the art would appreciate that the choice of pharmaceutically acceptable carrier(s), including a physiologically acceptable compound depends, for example, on the route of administration of the active agent(s) and on the particular physio-chemical characteristics of the active agent(s).
-38- [0115] The excipients are preferably sterile and generally free of undesirable
O
C-i matter. These compositions may be sterilized by conventional, well-known sterilization Stechniques.
[0116] In therapeutic applications, the compositions of this invention are c, 5 administered to a patient suffering from one or more symptoms of atherosclerosis or at risk for atherosclerosis in an amount sufficient to cure or at least partially prevent or arrest VI the disease and/or its complications. An amount adequate to accomplish this is defined as a "therapeutically effective dose." Amounts effective for this use will depend upon the c severity of the disease and the general state of the patient's health. Single or multiple administrations of the compositions may be administered depending on the dosage and C frequency as required and tolerated by the patient. In any event, the composition should provide a sufficient quantity of the active agents of the formulations of this invention to effectively treat (ameliorate one or more symptoms) the patient.
[0117] The concentration of peptide or mimetic can vary widely, and will be selected primarily based on fluid volumes, viscosities, body weight and the like in accordance with the particular mode of administration selected and the patient's needs.
Concentrations, however, will typically be selected to provide dosages ranging from about 0.1 or 1 mg/kg/day to about 50 mg/kg/day and sometimes higher. Typical dosages range from about 3 mg/kg/day to about 3.5 mg/kg/day, preferably from about 3.5 mg/kg/day to about 7.2 mg/kg/day, more preferably from about 7.2 mg/kg/day to about 11.0 mg/kg/day, and most preferably from about 11.0 mg/kg/day to about 15.0 mg/kg/day. In certain preferred embodiments, dosages range from about 10 mg/kg/day to about 50 mg/kg/day.
It will be appreciated that such dosages may be varied to optimize a therapeutic regimen in a particular subject or group of subjects.
[0118] In certain preferred embodiments, the peptides or peptide mimetics of this invention are administered orally via a tablet) or as an injectable in accordance with standard methods well known to those of skill in the art. In other preferred embodiments, the peptides, may also be delivered through the skin using conventional transdermal drug delivery systems, transdermal "patches" wherein the active agent(s) are typically contained within a laminated structure that serves as a drug delivery device to be affixed to the skin. In such a structure, the drug composition is typically contained in a layer, or "reservoir," underlying an upper backing layer. It will be appreciated that the term -39- "reservoir" in this context refers to a quantity of "active ingredient(s)" that is ultimately C available for delivery to the surface of the skin. Thus, for example, the "reservoir" may O include the active ingredient(s) in an adhesive on a backing layer of the patch, or in any of Z a variety of different matrix formulations known to those of skill in the art. The patch may C1 5 contain a single reservoir, or it may contain multiple reservoirs.
[0119] In one embodiment, the reservoir comprises a polymeric matrix of a pharmaceutically acceptable contact adhesive material that serves to affix the system to the skin during drug delivery. Examples of suitable skin contact adhesive materials include, but are not limited to, polyethylenes, polysiloxanes, polyisobutylenes, polyacrylates, polyurethanes, and the like. Alternatively, the drug-containing reservoir and skin contact adhesive are present as separate and distinct layers, with the adhesive underlying the reservoir which, in this case, may be either a polymeric matrix as described above, or it may be a liquid or hydrogel reservoir, or may take some other form. The backing layer in these laminates, which serves as the upper surface of the device, preferably functions as a primary structural element of the "patch" and provides the device with much of its flexibility. The material selected for the backing layer is preferably substantially impermeable to the active agent(s) and any other materials that are present.
[0120] Other preferred formulations for topical drug delivery include, but are not limited to, ointments and creams. Ointments are semisolid preparations which are typically based on petrolatum or other petroleum derivatives. Creams containing the selected active agent, are typically viscous liquid or semisolid emulsions, often either oilin-water or water-in-oil. Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase, also sometimes called the "internal" phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant. The specific ointment or cream base to be used, as will be appreciated by those skilled in the art, is one that will provide for optimum drug delivery. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and nonsensitizing.
[0121] Unlike typical peptide formulations, the peptides of this invention comprising D-form amino acids can be administered, even orally, without protection against proteolysis by stomach acid, etc. Nevertheless, in certain embodiments, peptide delivery can be enhanced by the use of protective excipients. This is typically accomplished either by complexing the polypeptide with a composition to render it Z resistant to acidic and enzymatic hydrolysis or by packaging the polypeptide in an C 5 appropriately resistant carrier such as a liposome. Means of protecting polypeptides for oral delivery are well known in the art (see, U.S. Patent 5,391,377 describing lipid In compositions for oral delivery of therapeutic agents).
[0122] Elevated serum half-life can be maintained by the use of sustained-release c protein "packaging" systems. Such sustained release systems are well known to those of 10 skill in the art. In one preferred embodiment, the ProLease biodegradable microsphere
C
,1 delivery system for proteins and peptides (Tracy (1998) Biotechnol. Prog. 14: 108; Johnson et al. (1996), Nature Med. 2: 795; Herbert et al. (1998), Phannaceut. Res. 357) a dry powder composed of biodegradable polymeric microspheres containing the protein in a polymer matrix that can be compounded as a dry formulation with or without other agents.
[0123] The ProLease microsphere fabrication process was specifically designed to achieve a high protein encapsulation efficiency while maintaining protein integrity. The process consists of preparation of freeze-dried protein particles from bulk protein by spray freeze-drying the drug solution with stabilizing excipients, (ii) preparation of a drugpolymer suspension followed by sonication or homogenization to reduce the drug particle size, (iii) production of frozen drug-polymer microspheres by atomization into liquid nitrogen, (iv) extraction of the polymer solvent with ethanol, and filtration and vacuum drying to produce the final dry-powder product. The resulting powder contains the solid form of the protein, which is homogeneously and rigidly dispersed within porous polymer particles. The polymer most commonly used in the process, poly(lactide-co-glycolide) (PLG), is both biocompatible and biodegradable.
[0124] Encapsulation can be achieved at low temperatures -40 0 During encapsulation, the protein is maintained in the solid state in the absence of water, thus minimizing water-induced conformational mobility of the protein, preventing protein degradation reactions that include water as a reactant, and avoiding organic-aqueous interfaces where proteins may undergo denaturation. A preferred process uses solvents in 0 which most proteins are insoluble, thus yielding high encapsulation efficiencies 0 C greater than 0 [0125] In another embodiment, one or more components of the solution can be D provided as a "concentrate", in a storage container in a premeasured volume) ready for dilution, or in a soluble capsule ready for addition to a volume of water.
[0126] The foregoing formulations and administration methods are intended to be Villustrative and not limiting. It will be appreciated that, using the teaching provided C herein, other suitable formulations and modes of administration can be readily devised.
O VII. Lipid-based formulations.
[0005] In certain embodiments, the peptides of this invention are administered in conjunction with one or more lipids. The lipids can be formulated as an excipient to protect and/or enhance transport/uptake of the peptides or they can be administered separately.
[0006] Without being bound by a particular theory, it was discovered of this invention that administration oral administration) of certain phospholipids can significantly increase HDILDL ratios. In addition, it is believed that certain mediumlength phospholipids are transported by a process different than that involved in general lipid transport. Thus, co-administration of certain medium-length phospholipids with the peptides of this invention confer a number of advantages: They protect the phospholipids from digestion or hydrolysis, they improve peptide uptake, and they improve HDIJLDL ratios.
[0007] The lipids can be formed into liposomes that encapsulate the polypeptides of this invention and/or they can be simply complexed/admixed with the polypeptides.
Methods of making liposomes and encapsulating reagents are well known to those of skill in the art (see, Martin and Papahadjopoulos (1982) J. Biol. Chem., 257: 286-288; Papahadjopoulos et al. (1991) Proc. Natl. Acad. Sci. USA, 88: 11460-11464; Huang et al.
(1992) Ccmcer Res., 52:6774-6781; Lasic et al.(1992) FEBS Lett., 312: 255-258., and the like).
[0008] Preferred phospholipids for use in these methods have fatty acids ranging from about 4 carbons to about 24 carbons in the sn-1 and sn-2 positions. In certain -42preferred embodiments, the fatty acids are saturated. JIn other preferred embodiments, the fatty acids can be unsaturated. Various preferred fatty acids are illustrated in Table 2.
Table 2 Preferred fatty acids in the sn-i and/or sn-2 position of the preferred phospholipids for administration of D polypeptides.
Carbon No.
10:0 11:0 12:0 13:0 14:0 15:0 16:0 17:0 18:0 19:0 20:0 21:0 22:0 23:0 24:0 14:1 14:1 16:1 16:1 Common Name Propionoyl Butanoyl Pentanoyl Caproyl Heptanoyl Capryloyl Nonanoyl Capryl Undcanoyl Lauroyl Tridecanoyl Myristoyl Pentadecanoyl Palmitoyl Heptadecanoyl Stearoyl Nonadecanoyl Arachidoyl Heniecosanoyl Behenoyl Trucisanoyl Lignoceroyl Myristoleoyl (9-cis) Myristelaidoyl (9-trans) Palrnitoleoyl (9-cis) Palmitelaidoyl (9-trans) IUPAC Name Trianoic Tetranoic Pentanoic Hexanoic Heptanoic Octanoic Nonanoic Decanoic Undecanoic Dodecanoic Tridecanoic Tetradecanoic Pentadecanoic Hexadecanoic Heptadecanoic Octadecanoic Nonadecanoic Eicosanoic Heniecosanoic Docosanoic Trocosanoic Tetracosanoic The fatty acids in these positions can be the same or different. Particularly preferred phospholipids have phosphoryicholine at the sn-3 position.
VIII. Additional pharmacologically active agents.
S[0127] Additional pharmacologically active agents may be delivered along with O the primary active agents, the peptides of this invention. In one embodiment, such agents include, but are not limited to agents that reduce the risk of atherosclerotic events and/or complications thereof. Such agents include, but are not limited to beta blockers, beta blockers and thiazide diuretic combinations, statins, aspirin, ace inhibitors, ace f receptor inhibitors (ARBs), and the like.
[0128] Suitable beta blockers include, but are not limited to cardioselective (selective beta 1 blockers), acebutolol (Sectral
TM
atenolol (TenorminTM), betaxolol 0 10 (KerloneTM), bisoprolol (Zebeta T M metoprolol (Lopressor T M and the like. Suitable nonselective blockers (block beta 1 and beta 2 equally) include, but are not limited to carteolol (Cartrolm), nadolol (Corgard
TM
penbutolol (LevatolTM), pindolol (Visken'), propranolol (Inderal
M
timolol (Blockadrenl m labetilol (NormodyneT, TrandateM), and the like.
[0129] Suitable beta blocker thiazide diuretic combinations include, but are not limited to Lopressor HCT, ZIAC, Tenoretic, Corzide, Timolide, Inderal LA 40/25, Inderide, Normozide, and the like.
[0130] Suitable statins include, but are not limited to pravastatin (Pravachol/Bristol-Myers Squibb), simvastatin (Zocor/Merck), lovastatin (Mevacor/Merck), and the like.
[0131] Suitable ace inhibitors include, but are not limited to captopril (e.g.
Capoten M by Squibb), benazepril Lotensinm by Novartis), enalapril VasotecM by Merck), fosinopril Monopril T M by Bristol-Myers), lisinopril (e.g.
PrinivilTM by Merck or ZestrilTM by Astra-Zeneca), quinapril Accupril T m by Parke- Davis), ramipril AltaceTm by Hoechst Marion Roussel, King Pharmnaceuticals), imidapril, perindopril erbumine AceonTM by Rhone-Polenc Rorer), trandolapril MavikTM by Knoll Pharmaceutical), and the like. Suitable ARBS (Ace Receptor Blockers) include but are not limited to losartan CozaarTM by Merck), irbesartan Avaprom by Sanofi), candesartan Atacandm by Astra Merck), valsartan Diovanm by Novartis), and the like.
-44- IX. Kits for the amelioration of one or more symptoms of atherosclerosis.
C [0132] In another embodiment this invention provides kits for amelioration of one 0 or more symptoms of atherosclerosis or for the prophylactic treatment of a subject (human or animal) at risk for atherosclerosis. The kits preferably comprise a container containing
O
C 5 one or more of the peptides or peptide mimetics of this invention. The peptide or peptide mimetic may be provided in a unit dosage formulation suppository, tablet, caplet, V" patch, etc.) and/or may be optionally combined with one or more pharmaceutically acceptable excipients.
[0133] The kit can, optionally, further comprise one or more other agents used in C 10 the treatment of heart disease and/or atherosclerosis. Such agents include, but are not limited to, beta blockers, vasodilators, aspirin, statins, ace inhibitors or ace receptor inhibitors (ARBs) and the like, e.g. as described above.
[0134] In addition, the kits optionally include labeling and/or instructional materials providing directions protocols) for the practice of the methods or use of the "therapeutics" or "prophylactics" of this invention. Preferred instructional materials describe the use of one or more polypeptides of this invention to mitigate one or more symptoms of atherosclerosis and/or to prevent the onset or increase of one or more of such symptoms in an individual at risk for atherosclerosis. The instructional materials may also, optionally, teach preferred dosages/therapeutic regiment, counter indications and the like.
[0135] While the instructional materials typically comprise written or printed materials they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this invention. Such media include, but are not limited to electronic storage media magnetic discs, tapes, cartridges, chips), optical media CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials.
EXAMPLES
[0136] The following examples are offered to illustrate, but not to limit the claimed invention.
Example 1
O
C1 [0137] Several synthetic class A peptide analogs have been shown to mimic many O of the properties of human apo A-I in vitro. In this example, a new peptide (5F) with Z increased amphipathicity, was given by intraperitoneal injection, 20 gg/daily, for 16 weeks 1 5 to C57BL/6J mice fed an atherogenic diet. Mouse apo A-I (MoAI) (50 gg/daily) or phosphate buffer saline (PBS) injections were given to other mice as controls. Total Splasma cholesterol levels and lipoprotein profiles were not significantly different among the treated group and the control groups except that the mice receiving 5F or MoAI had C1 lower high density lipoprotein (HDL)-cholesterol when calculated as a percent of total cholesterol. No toxicity or production of antibodies to the injected materials was c, observed. When LDL was taken from animals injected with 5F and presented to human artery wall cells in vitro it produced less lipid hydrodroperoxides and less LDL-induced chemotactic activity than LDL taken from controls. Additionally, when HDL was taken from mice injected with 5F and presented to human artery wall cells in vitro together with human LDL, there were substantially less lipid hydroperoxides formed and substantially less LDL-induced monocyte chemotactic activity. Mice receiving peptide 5F had significantly less aortic atherosclerotic lesion area compared to mice receiving PBS.
Lesion area in mice receiving MoAI was similar to that of the PBS-injected animals. We conclude that 5F may have potential in the prevention and treatment of atherosclerosis.
Materials and Methods Peptides [0138] Peptide 5F (Ac-18A[Asp Trp Leu Lys Ala Phe Tyr Asp Lys Val Phe Glu Lys Phe Lys Glu Phe Phe]-NH 2 was synthesized by solid-phase peptide synthesis (see, Anantharamaiah and Garber (1996) Meth. Enzymol. 263: 267-282; Palgunachari et al. (1996) Arteriosclerosis, Thrombosis, Vascular Biology 16: 328-338). The purity of the synthetic peptide was established by analytical HPLC and ion-spray mass spectrometry. The peptide was dialyzed against distilled water and lyophilized before using.
[0139] MoAI was isolated from the plasma of C57BL/6J mice (EDTA plasma was purchased from Harlan Bioproducts for Science, Indianapolis, IN). MoAI was isolated -46- 0 using a combination of size-exclusion and reversed-phase column chromatography.
O
CI Briefly, plasma density was adjusted to 1.21 g/ml by addition of KBr, and centrifuged at O 50,000 rpm for 24 hours at 4 0 C (Ti70 rotor, Beckman, Fullerton, CA). The top fraction Z was collected, dialyzed against water to remove KBr, lyophilized, and delipidated. The C 5 pellet was dissolved in Gn:DTT:Tris solution (3 M guanidine HC1, 1 mM dithiothreitol, and 10 mM Tris; pH=8.0), then dialyzed against the same solution using 12,000 MW- Scutoff dialysis tubing in order to remove much of the apo A-Il and C apolipoproteins from the sample. The sample was then dialyzed against water and lyophilized. The pellet was CI dissolved in fresh Gn:DTT:Tris solution, and proteins were separated by size-exclusion 1-- 0 10 column chromatography, using an XK26/100 column (2.6 X 100 cm) packed with bulk-
O
CI phase Superose 12 (Pharmacia Biotech, Piscataway, NJ) equilibrated with Gn:DTT:Tris solution. The flow rate was 0.5 ml/min, and 2.5 ml fractions were collected. Fractions corresponding to the apo A-I peak were analyzed by SDS-PAGE, and further purified by preparative C-18 reverse-phase HPLC (Anantharamaiah and Garber (1996) Meth.
Enzymol. 263: 267-282).
Mice [0140] All experiments were performed using female C57B16J mice (Jackson Laboratory, Bar Harbor, ME). Mice were purchased at six weeks of age, and the diet studies were begun with mice at eight weeks of age. Mice weighing 20 to 22 grams were used in the turnover studies. All animal studies were prospectively reviewed and approved by the Institutional Animal Care and Use Committee of the University of Alabama at Birmingham.
Kinetic studies- [0141] The 5F peptide, MoAI, and human apo A-I were labeled with 12'I by the method of Bilheimer et al. (1972) Biochim. Biophys. Acta 260: 212-221. Mice were placed on a modified Thomas-Hartroft atherogenic diet (#TD88051; Teklad, Madison, WI) for four weeks at which time daily intraperitoneal injections of peptide or protein dissolved in 200 pl phosphate-buffered saline (PBS) were begun. Animals injected with MoAI or human apo A-I received 50 A g per animal; those injected with 5F received 20 /g Animals were not fasted for the kinetic studies and blood samples were taken under xylazine:ketamine anesthesia from the retro-orbital sinus at 15, 30, and 45 minutes, and 1, -47- 2, 3, 4, 6, 8, 12, and 24 hours following injection. Each animal provided three blood samples at different time points (all retro-orbital and alternating eyes), and at least three samples were collected (from different animals) at each time point. Samples were Z collected into heparinized capillary tubes, then placed in microcentrifuge tubes; the plasma c 5 was separated by centrifugation. Duplicate 10 pl aliquots of each sample were taken for radioactivity determination, using gamma counting (Cobra; Packard Instruments, Downers SGrove, L) for 10 minutes per sample. Total plasma volume was calculated as 4.2% body Sweight. Each sample was expressed as percent of injected CPM in total plasma. Free 125 was determined by trichloroacetic acid (TCA) precipitation (1 ml of 10% TCA per 10 p/l C 10 plasma sample). Fitting to the kinetic model was done using all data points, rather than N averages at each time point (PKAnalyst, MicroMath Scientific Software, Salt Lake City,
UT).
Injection protocol and sample collection for lesion studies- [0142] Mice were acquired at six weeks of age, and randomized into groups of except that a negative control group of 10 received no treatments and was given standard rodent chow. At eight weeks of age, the treatment groups were placed on a modified Thomas-Hartroft atherogenic diet (#TD88051; Teklad, Madison, WI), and injections were begun. The diet was stored at 40 C and was used for no longer than three months after the manufacture date in order to minimize lipid oxidation. Animals were injected intraperitoneally daily for 16 weeks, including weekends and holidays. Twenty mice in each group received daily injections of 200 pl PBS (as positive controls), or 20 pg 5F in 200 gl PBS, or 50 gg MoAI in 200 gl PBS.
[0143] Lyophilized 5F peptide was prepared in vials, with each bottle containing sufficient peptide for one day's injection. The 5F peptide was lyophilized in PBS, and was dissolved in autoclaved Milli-Q water (Millipore Corp., Bedford, MA) on the day of injection. The injection volume for all groups was maintained at 200 pl/mouse per day.
[0144] Blood samples were taken under anesthesia by retro-orbital bleeding at study entry (pre-diet) and at the time of organ harvesting. At the end of the study (week 16), at the last bleeding, the heart and the liver were excised. The hearts were kept in 0.9% saline solution for about 1 hour to eliminate blood and to permit the heart muscle to relax. They were then fixed in phosphate-buffered 4% formaldehyde for at least one week C until sectioned. The livers were removed and weighed.
O
Z Histological evaluation- [0145] Histological evaluations were performed according to the method of Paigen et al. (Paigen et al. (1990) Arteriosclerosis 10: 316-323) with some modifications.
r Briefly, hearts were fixed for at least one week in the phosphate-buffered formaldehyde solution. After removing the lower 2/3 of the hearts, the remaining tissue was frozen in I OCT medium (Tissue-Tek, Miles Inc., Elkhart, IN) and sectioned in a cryostat at -20 0
C.
Alternate 20 /m sections were saved on slides, and observed for the beginning of the C 10 aortic root. Sections were then collected for an additional 600 Am, or until the aortic cross-section was rounded and the valve cusps were no longer evident. Slides were stained with Oil Red O, and counterstained with hematoxylin. Stained lesion crosssectional areas were measured in consecutive slides 80 /m apart by image analysis (SigmaScan Pro, SPSS Scientific, Chicago, IL), and the average lesion area was determined for each aortic sinus over the 400 Aim length (five slides) providing the greatest mean lesion area.
Cocultures, Monocyte Isolation, Isolation of Lipoproteins, Determination of Lipid Hydroperoxides, and Monocyte Chemotactic Activity- [0146] Cocultures of human artery wall cells, monocyte isolation, isolation of lipoproteins by ultracentrifugation from the plasma of normal human donors or from mouse plasma by FPLC, and determination of lipid hydroperoxides and monocyte chemotactic activity were performed according to standard methods. All human subject participation was with informed consent approved by the UCLA Human Subjects Protection Committee. The protocol for testing mouse lipoproteins in the coculture was also performed as follows: Briefly, LDL and HDL were isolated by FPLC from mouse plasma from mice fed the atherogenic diet and injected with vehicle (PBS), or with peptide at 20 g/mouse/day. The cocultures were treated with human LDL at 200 g/ml LDL protein, or mouse LDL at 200 ptg/ml or with 200 g/ml human LDL human HDL at 350 ,/g/ml of HDL protein or mouse HDL at 300 /g/ml or with mouse HDL alone at 300 gig/ml. The cocultures were incubated with or without the above additions for 8 hrs at 370 -49- C in the presence of 10% lipoprotein deficient serum (LPDS). The supernatants were C- collected and analyzed for Auerbach lipid hydroperoxide equivalents. The cocultures were then washed and incubated with fresh culture medium without serum or LPDS for an Z additional 8 hrs. The conditioned medium was collected and analyzed for monocyte C, 5 chemotactic activity.
Chemical and analytical methods-Column cholesterol lipoprotein profiles (CLiP)- [0147] Plasma cholesterol lipoprotein profiles were measured using our recently- Sdeveloped CLiP method (Garber et al. (2000) J. Lipid Res. 41:1020-1026). Briefly, 5 to C1 10 10 Al of plasma were analyzed using a single Superose 6 (Pharmacia, Piscataway NJ) column. Immediately following the column, cholesterol reagent was introduced through a mixing tee, and the eluent:reagent mixture entered a post-column reaction coil.
Cholesterol content of the eluent mixture was spectrophotometrically detected at 500 nm, and data points were collected into a computer. The resulting profiles were decomposed into component peaks and analyzed for relative area using PeakFit (SPSS Science, Chicago, IL); absolute cholesterol values for total cholesterol and each component peak were determined by comparison with a control sample of known values. In some cases fractions were collected to determine distribution of radioactivity. The CLiP method allowed analysis of individual mouse samples, avoiding the use of pooled samples.
Antibody detection- [0148] To determine whether daily injections of peptides elicited any immune response in mice, indirect ELISA titration (Engvall (1980) Meth. Enzymol. 70:419-439) was carried out with plasma taken from mice at the time of organ collection (following sixteen weeks of daily injection). Plates were coated with the injected peptides or MoAI (10gg/ml). Plates were incubated overnight. After thorough washing with borate buffered saline (pH 8.2) containing 0.05% Tween 20, and blocking with buffer gelatin and 0.1% BSA in borate buffer) for lh, 200 pl of the diluted mouse plasma (1:100 dilution) samples were serially diluted 1:1 with borate-buffered saline. Biotinylated goat antibody to mouse IgG (0.l1pg/ml) was then added to the wells and the plates were treated with SA- HRP (Streptavidin-horse radish peroxidase) for an hour and developed with ABTS and 0 peroxide as substrate. The plates were incubated overnight at room temperature after C1 every addition of antigen/antibody and washed thoroughly with borate buffered saline (pH O 8.2) containing 0.05% Tween 20, and blocked with buffer gelatin and 0.1% BSA in Z borate buffer) for lh before the next addition.
cIN Statistical methods- [0149] Treatment groups were compared by two-tailed t-tests or one way analysis of variance (where the data were normally distributed), or by one way analysis of variance c% C.1 on ranks (SigmaStat; SPSS Science, Chicago, IL). Kinetics of peptide or protein turnover Swere analyzed by fitting to a first order one-compartment kinetic model assuming non- CN 10 equal input and output rates (PKAnalyst; MicroMath Scientific Software, Salt Lake City,
UT).
Results Kinetic studies- [0150] The kinetics of the clearance of peptide 5F and human and mouse apo A-I from mouse plasma following intraperitoneal injection are summarized in Table 3.
Table 3. Summary of fitted data from kinetic experiments Injected Material TV2 Time to Max. in max. CPM plasma r Human apo A-I 15.6 3.61 23.7 0.947 /g/mouse) Mouse apo A-I 15.7 1.74 13.5 0.928 ig) (20 /Lg) 6.22 2.36 14.29 0.895 Data shown represent results of fitting data to a first order one-compartment kinetic model assuming unequal input and output rates (PKAnalyst; MicroMath Scientific Software, Salt Lake City, UT). Abbreviations: TV/2: half time of clearance from plasma; Max. in plasma: percent of injected dose found in total plasma at peak levels; r 2 goodness of fit statistic of the kinetic model.
[0151] Human and mouse apo A-I had greatly prolonged clearance compared with the 5F peptide. Human apo A-I and 5F had longer times to peak plasma levels than did mouse apo A-I, although peak levels achieved were generally similar (human apo A-I reached higher peak levels than did the other materials). Analysis of plasma samples by -51r- 0 column chromatography demonstrated that peptide 5F and apo A-I (both human and C mouse) associated with plasma lipoproteins, especially with particles in the HDL-sized O region (Figure The HDL:VLDL ratio of peptide radioactivity 1.5 h following injection Z of 5F was 4.19±0.58 p<0.05). Similar results were found 5 h following injection of CN 5 5F (6.44±1.10, p<0.02). The injected peptide initially had less than 3% free 12I by TCA precipitation. However, 1.5 hours after injection, free 12 5 I radioactivity in the plasma as a
U--
tI percent of total eluted radioactivity was substantially greater for 5F being 26.9±9.4% and at 5 hours 34.4±4.8%, reflecting the expected clearance of lipoproteins and lipoprotein- C associated peptides. The rate of increase in the radioactivity due to free iodine from 1.5 to 0 10 5 hours was less than that from injection to 1.5 hours, possibly suggesting considerable C initial degradation of the peptide in the peritoneal cavity.
Survival and gross morphology on the chow or atherogenic diets- [0152] Only three mice died from unexplained causes during the course of the prolonged diet studies. Two of the animals had been receiving MoAI, and one was receiving 5F peptide. At the time of organ collection, no gross morphological differences were observed between the groups. Livers were enlarged in all animals fed the atherogenic diet, but neither liver weights nor liver weight as a percent of body weight were different between groups (Table All animals on the atherogenic diet (including PBS-injected animals) had lower body weights than the chow-fed controls (Table 4).
Table 4. Body and liver weights following treatment.
Diet Subgroup Body Weight Liver Weight Liver:Body (percentage) Chow 23.38±0.52 0.99±0.02 4.24-0.04% Atherogenic PBS (n=14) 20.55±0.32* 1.60±0.04 7.84±0.26% (n=15) 21.60±0.28 1.61±0.04 7.46±0.23% MoAI (n=14) 21.16-0.34 1.72±0.04 8.15±0.23%* Data shown are mean SEM of weights taken at the time of organ harvesting (after 16 weeks of treatment). The chow-fed animals received no injections. The other mice were maintained on the atherogenic diet as described in Methods. The PBS group received intraperitoneal injections of 200 gl phosphate-buffered saline daily. The 5F group received intraperitioneal injections of 20 Rg 5F in 200 01PBS daily and the MoAI group received 50 gg MoAI in 200 gl PBS daily.
*p<0.05 vs 5F; two-tailed t-test -52- Antigenicitv- [0153] Blood samples taken at the conclusion of the 16-week injection period were tested for the presence of antibodies against the peptides. No antibodies were detected against peptide 5F or against MoAI (data not shown). Cross experiments, where the ELISA plates were coated with peptides or protein which was not injected into the series of animals, produced results essentially identical to those in the direct determination of the presence of antibodies (data not shown).
Lipoprotein and apolipoprotein characterization- [0154] Total and lipoprotein cholesterol values as determined by the CLiP method are presented in Table 3. Accuracy of total cholesterol values was confirmed by a manual cholesterol assay (Cholesterol 1000; Sigma, St. Louis, MO) (data not shown). No significant differences in total or lipoprotein-fraction cholesterol levels were seen between the treatment groups. However, when lipoprotein fractions were expressed as a percent of total cholesterol (Table HDL-cholesterol comprised a significantly lower percentage in the 5F and MoAI groups compared with the PBS group.
Table 5: Total and lipoprotein cholesterol levels (mg/dl and percent of total cholesterol) after 16 weeks of chow or atherogenic diet.
VLDL IDL+LDL HDL TC Chow Diet 11.66±2.34 23.68±3.51 37.30±2.52 72.64+5.58 (16.61±3.55%) (31.66±3.61%) (51.73±1.75%) Atherogenic Diet PBS 88.36±5.48 75.82±7.64 24.36±2.19 188.54±14.2 (47.26±1.37%) (39.83±1.34%) (12.91-0.68%) 100.34±15.72 83.37±8.15 17.92±2.91 201.63±25.2 (47.96+3.26%) (42.80±2.51%) (9.24±1.18%*) MoAI 100.08±9.73 87.86±8.34 19.50±3.07 207.45±16.9 (48.23±2.75%) (42.44±2.46%) (9.34±1.19%*) Data are expressed as mean mg/dl SEM and, in parentheses, as percent of total cholesterol. Abbreviations: VLDL, very low density lipoprotein; IDL, intermediate density lipoprotein; LDL, low density lipoprotein; HDL, high density lipoprotein; TC, total cholesterol; MoAI, mouse apo A-I; PBS, Phosphate buffered saline. The chow-fed animals received no injections. The other mice-were maintained on the atherogenic diet as described in Methods. The PBS group received intraperitoneal injections of 200 p.1 PBS daily. The 5F group received intraperitioneal injections of 20 tg 5F in 200 pl PBS daily and the MoAI group received 50 gg MoA-I in 200 gl PBS daily. Numbers of animals are as shown in Table 4.
*p<0.05 or less compared with PBS by two-tailed t-test.
-53- 2 '1 4 Interaction of Mouse Lipoproteins with Human Artery Wall Cells- 0 Z [0155] We recently discovered that normal HDL inhibits three steps in the ND formation of mildly oxidized LDL. In those studies (see, copending application USSN 09/541,468, filed on March 31, 2000) we demonstrated that treating human LDL in vitro with apo A-I or an apo A-I mimetic peptide (37pA) removed seeding molecules from the LDL that included HPODE and HPETE. These seeding molecules were required for c cocultures of human artery wall cells to be able to oxidize LDL and for the LDL to induce the artery wall cells to produce monocyte chemotactic activity. We also demonstrated that after injection of apo A-I into mice or infusion into humans, the LDL isolated from the mice or human volunteers after injection/infusion of apo A-I was resistant to oxidation by human artery wall cells and did not induce monocyte chemotactic activity in the artery wall cell cocultures. Figure 7 demonstrates that HDL from the mice in the present study that were fed the atherogenic diet and injected with PBS failed to inhibit the oxidation of human LDL (Figure 7A) and failed to inhibit LDL-induced monocyte chemotactic activity (Figure 7B) in human artery wall coculures. In contrast, HDL from mice fed the atherogenic diet and injected daily with peptide 5F was as effective in inhibiting human LDL oxidation and preventing LDL-induced monocyte chemotactic activity in the cocultures as was normal human HDL. Figure 7 also shows that LDL taken from mice fed the atherogenic diet and injected daily with PBS was more readily oxidized and more readily induced monocyte chemotactic activity than LDL taken from mice fed the same diet but injected with 20Lg daily of peptide 5F. No cytotoxicity was noted in the artery wall cells treated with any of the lipoproteins (data not shown). Similar results were obtained in three of three separate experiments (data not shown).
Lesion formation- [0156] Mean lesion cross-sectional areas are presented in Figure 8. As expected, no lesions were observed in the group given normal mouse chow (data not shown). As previously reported (Paigen et al. (1990) Arteriosclerosis 10: 316-323), considerable variations in lesion area were observed in all groups receiving the atherogenic diet.
However, the 5F-injected animals had significantly lower mean lesion area than PBSinjected animals, whether analyzed by two-tailed t-test (p<0.002) or by one-way analysis -54r- 0 of variance on ranks (p<0.001; determined due to the non-normal distribution of mean CN lesion areas). MoAI injection produced no difference in lesion area compared with PBS O injection, and lesion area was significantly greater than in 5F-injected animals, both by t- Z test (p<0.002) and by one way analysis of variance on ranks (p<0.001).
(N
Discussion t [0157] We previously demonstrated that synthetic peptides that were designed to mimic the class A amphipathic helical motif were able to associate with phospholipids, C1 and exhibited many biological properties similar to human apo A-I (3,8,10,14,15, 20). We Salso have shown that when these peptides are administered intravenously in animals, they C1 10 are found to be associated with plasma lipoproteins This study was designed to address the hypothesis that a new peptide, 5F, with increased theoretical lipid affinity, would possess anti-atherogenic properties.
[0158] The studies presented here demonstrated that this peptide 5F entered the plasma after interperitoneal injection and achieved plasma levels that were roughly comparable to MoAI, but less than human apo A-I (Table 3 and Figure The plasma clearance half-time of 5F was shorter than either mouse or human apo A-I after peritoneal injection. After injection the majority of 5F was found in the region of HDL (Figure 6), despite the fact that the preponderance of circulating cholesterol was in the VLDL-, IDL-, and LDL-sized regions on the atherogenic diet.
[0159] Plasma cholesterol levels and distributions were not significantly different among the injected groups on the atherogenic diet (Table However, when the lipoprotein fractions were expressed as a percent of total cholesterol (Table HDLcholesterol comprised a significantly lower percentage in the 5F and MoAI groups compared with the PBS group.
[0160] Normal HDL inhibits three steps in the formation of mildly oxidized LDL.
We demonstrated that treating human LDL in vitro with apo A-I or an apo A-I mimetic peptide removed seeding molecules from the LDL that included HPODE and HPETE.
These seeding molecules were required for cocultures of human artery wall cells to be able to oxidize LDL and for the LDL to induce the artery wall cells to produce monocyte chemotactic activity (see copending copending application USSN 09/541,468, filed on March 31, 2000). We also demonstrated that after injection of apo A-I into mice or 0 infusion into humans, the LDL isolated from the mice or human volunteers after Cl injection/infusion of apo A-I was resistant to oxidation by human artery wall cells and did O not induce monocyte chemotactic activity in the artery wall cell cocultures. In the present Z studies, HDL from mice that were fed the atherogenic diet and injected with PBS failed to \0 CN 5 inhibit the oxidation of human LDL (Figure 7A) and failed to inhibit LDL-induced monocyte chemotactic activity (Figure 7B) in the human artery wall coculures. In stark I contrast, HDL from mice fed the same atherogenic diet but injected with peptide 5F was found to be as effective in inhibiting human LDL oxidation and preventing LDL-induced CI monocyte chemotactic activity in the cocultures as was normal human HDL (Figure 7).
S 10 LDL taken from mice fed the atherogenic diet and injected with 5F was less readily CI oxidized and induced less monocyte chemotactic activity than LDL taken from mice fed the same diet but injected with PBS (Figure It is possible that 5F interacted with LDL in the circulation (either before or after associating with HDL) and removed seeding molecules necessary for LDL oxidation and LDL-induced monocyte chemotactic activity in a manner similar to that described in vitro for a related peptide, 37pA (copending copending application USSN 09/541,468, filed on March 31, 2000).
[0161] The in vitro responses of human artery wall cells to HDL and LDL from mice fed the atherogenic diet and injected with peptide 5F are consistent with the protective action of 5F in vivo. Despite, similar levels of total cholesterol, LDLcholesterol, IDL+VLDL-cholesterol, and lower HDL-cholesterol as a percent of total cholesterol, the animals fed the atherogenic diet and injected with 5F had significantly lower lesion scores (Figure These results are somewhat analogous to those of Shah et al. (Shah et al. (1998) Circulation 97:780-785) who found that, despite persistence of hypercholesterolemia, apo A-IMinano prevented progression of atherosclerotic lesions in apo E-deficient mice.
[0162] The reason that human apo A-I has been used successfully to prevent/reduce atherosclerosis in animals (Wilson et al. (1988) Arteriosclerosis 8: 737- 741; Rubin et al. (1991) Nature 353:265-267; Paszty et al. (1994) J. Clin. bnvest.
94:899-903; Plump et al. (1994) Proc. Natl. Acad. Sci. USA 91:9607-9611; Shah et al.
(1998) Circulation 97:780-785) but injection of MoAI at a dose of 50 Rg daily in these studies did not is not clear. It has been shown that MoAI does not form protein:lipid complexes as stable as does human apo A-I (Gong et al. (1994) Biochim. Biophys. Acta 0 1213:335-342). Mouse HDL has also been shown to be more easily denatured by C guanidine hydrochloride than human HDL (Gong et al. (1994) Biochim. Biophys. Acta >1213:335-342) suggesting that amphipathic helical peptides might displace MoAI more Z easily from mouse HDL than human apo A-I from human HDL. These differences may or C, 5 may not explain why MoAI did not significantly reduce lesions in this study. It may also be that a higher dose of MoAI is required under the conditions that we employed. In any event, the 5F peptide was highly effective under these conditions and MoAI was not.
[0163] The ELISA analysis of plasma at the conclusion of the injection protocol
C
s I indicated that antibodies were not formed against the 5F peptide. This was not surprising in that lipid-associating peptides have been shown not to produce antibodies, presumably C, because these peptides bind lipids in such a way as to prevent the exposure of epitopes necessary to elicit an immune response (Muranishi (1997) J. Pharm. Soc. Japan 117:394- 404; Fricker and Drewer (1996) J Peptide Sci. 2:195-211).
[0164] A preliminary study by us suggested that transgenic mice expressing a class A amphipathic helical peptide (37pA) with theoretically less lipid affinity than the peptide used in this study may have been resistant to atherosclerosis (Garber et al. (1997) Circulation 96:1-490). The current study suggests that peptide 5F likely has great potential for elucidating the mechanisms involved in atherogenesis and also has therapeutic potential.
Example 2 Efficacy of D Peptides [0165] This example demonstrates the efficacy of D peptides of this invention.
Human aortic wall cocultures were incubated with medium alone (LDL, NO CELLS or CELLS, NO LDL), control LDL from normal subjects at 250 ig/ml (LDL) and LDL plus control HDL from normal subjects at 350 gg/ml Other cocultures were incubated with the control LDL together with varying amounts (micrograms shown on the abscissa) of either D-2F, or L-2F (third panel from the left, 2F) or D-37-pA or L-37pA (last panel on the right, 37pA). The data represent mean SD of values obtained from quadruplicate cocultures. Values for HDL or added peptides were all significantly different from LDL alone (first panel on the left) at the level of p 0.01.
r-- S [0166] The cocultures were incubated for 4 hrs at 37 0 C in the presence of
O
C1 LPDS to produce mildly oxidized LDL. The supernatants were then discarded, the O cocultures were washed and incubated with culture medium without serum or LPDS for an Z additional 4 hrs. This conditioned medium was collected and analyzed for monocyte N 5 chemotactic activity. As shown in Figure 9, treating LDL with the D peptides in vitro prevents their oxidation by artery wall cells.
[0167] Figure 10 demonstrates that giving the D peptides to mice renders their red blood cells resistant to hemolysis a phenomenon due to oxidation as it can be prevented with Vitamin E, data not shown). Groups of LDL receptor deficient mice (n=3) commonly used as an animal model of atherosclerotic lesion formation were administered the D-peptides or the saline vehicle by gavage. Each animal was administered 100/tl of saline, 100 j/g/100gl of peptide D-2F or peptide D-37pA. Blood was collected from retroorbital sinus under mild anesthesia 17n and 48 hrslater. Red cells were separated by centrifugation, were diluted to 10 hematocrit with PBS and incubated at 37 0 C with gentle mixing. Aliquots were removed at time points t=0, 2, 6 and 18 hrs, cell pellets spun down and the optical density due to the released hemoglobin determined.
[0168] Figure 11 demonstrates that administering the D peptides to mice by gavage and then isolating their LDL renders the LDL resistant to artery wall cell oxidation as measured by the monocyte chemotaxis bioassay.
[0169] Another experiment demonstrated that the D-peptide was absorbed from the stomach and rendered LDL unable to induce monocyte chemotactic activity in our human artery wall cell coculture model while the L-peptide of 2F did not have this property. Either saline or 2F synthesized from D amino acids or from L amino acids was instilled in the stomachs of mice by gavage (instillation in the stomach by tube). After gavage the mice were bled and their LDL isolated and added to the human artery wall cell cocultures. The D-peptide when given by gavage protected the LDL as evidenced by the reduced monocyte chemotaxis induced by the LDL taken from the mice that received the D-2F peptide (D2FLDL) (synthesized from D amino acids), while the LDL taken from mice that received the L-2F (synthesized from the natural L amino acids) (L2FLDL) readily induced monocyte chemotaxis (see Figure 12).
-58-
U--
O [0170] 2F synthesized from L amino acids when presented to LDL in vitro was as
O
C1 effective as the 2F synthesized from the D amino acids (see Figure Thus, the O difference in the results with this experiment where the peptides were given in vivo by gavage indicate that the 2F synthesized from D amino acids must have been absorbed
ID
C1 5 intact from the stomach while the 2F peptide synthesized from the natural L amino acids must have been degraded in the stomach in the process of digestion and/or in the plasma t as we hypothesized would be the case. In other studies we have not seen evidence of antibody formation against the D-2F peptide.
[0171] Figure 13A and Figure 13B are two graphs from experiments in which 10 LDL receptor knockout mice were given 50 micrograms of D-5F by gavage. The animals were bled 1.5, 3 or 6 hours later and their HDL, LDL, and VLDL/IDL isolated. As indicated in the graph, HDL taken 1.5 hours after gavage did not protect control (cont.) LDL from modification but the HDL taken after 3 hours and slightly less after 6 hours following gavage were as protective against LDL-induced monocyte chemotactic activity production'by human artery wall cells as a control HDL (Figure 13A). In the other graph (Figure 13B), 1.5, 3, or 6 h after administration of 50 micrograms of D-5F by gavage mouse LDL and VLDIJIDL were isolated. In the left panel a control LDL was added to the human artery wall cells without or with a control HDL and monocyte chemotactic activity produced by the artery wall cells was measured. In the middle panel the mouse LDL taken after 1.5, 3, or 6 hours after gavage of 50 micrograms of D-5F were added to the artery wall cells. The results indicate that after 3 h and 6 h the LDL induced significantly less monocyte chemotactic activity. On the right side of the graph the VLDIJIDL fraction of lipoproteins (V/I LDL) were added and as shown the 3 hour time point induced significantly less monocyte chemotactic activity.
Example 3 Effects of Increasing Hydrophobicity on the Physical-Chemical and Biological Properties of a Class A Amphipathic Helical Peptide List of abbreviations [0172] Ac 2 0, acetic anhydride; apo A-I, apolipoprotein A-I; BSA, Bovine serum albumin; CAD, coronary artery disease; CD, circular dichroism; DMPC, dimyristoyl phosphatidylcholine; DiPoPE, Di (16:1) palmitoleoyl phosphatidylethanolamine; DSC, -59- 0 Differential Scanning Calorimetry; EDTA, ethylene diamine tetraacetic acid; EPC, Egg CN phosphatidylcholine; FMOC, Fluorinylmethyloxycarbonyl; Gdn HC1, Guanidine O Hydrochloride; HAEC, human aortic endothelial cells; HASMC, human aortic smooth Z muscle cells; HBTU, 2-(H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium ,N 5 hexafluorophosphate; HDL, high density lipoprotein; HPLC, High Performance Liquid Chromatography; LCAT, lecithin cholesterol acyl transferase; MCP-1, monocyte chemotactic protein-1; M-CSF, macrophage colony-stimulating factor; MLV multilamellar vesicles; NMM, N-methylmorpholine; PBS, phosphate buffered saline; SPIPES, piperazine-N,N'-bis[2-ethanesulfonic acid]; RP-HPLC, reverse phase high O 10 performance liquid chromatography; TFA, trifloroacetic acid.
Abstract [0173] We have recently shown that a class A amphipathic peptide 5F with increased amphipathicity protected mice from diet-induced atherosclerosis. We have now examined the effects of increasing the hydrophobicity of a series of homologous class A amphipathic peptides, including 5F, on physical and functional properties related to atherosclerosis inhibition by systematically replacing existing nonpolar amino acids with phenylalanine. The peptides, based on the sequence Ac-D-W-L-K-A-F-Y-D-K-V-A-E-K-
L-K-E-A-F-NH
2 (SEQ ID NO:1, Ac-18A-NH 2 or 2F) were: 3F 3 (Ac-F 3 18A-NH2), 3F"(Ac-
F'
1 18A-NH 2 4F(Ac-F 3 14 18A-NH 2 5F(Ac-F 14 17 1 8A-NH 2 6F(Ac-F 1 0o 11 1 4 17 18A-NH 2 and 7F(Ac-F 3 10 14 1 7 18A-NH 2 Measurements of aqueous solubility, HPLC retention time, exclusion pressure for penetration into an egg PC monolayer, and rates of egg PC solubilization revealed an abrupt increase in the hydrophobicity between peptides 4F and this was accompanied by increased ability to associate with phospholipids. The peptides 6F and 7F were less effective, indicating a limit to increased hydrophobicity for promoting lipid interaction in these peptides. Despite this marked increase in lipid affinity, these peptides were less effective than apoA-I in activating the plasma enzyme, lecithin: cholesterol acyl transferase (LCAT), with 5F activating LCAT the best (80% of apoA-I). Peptides 4F, 5F and 6F were equally potent in inhibiting LDL-induced monocyte chemotactic activity. These studies suggest that an appropriate balance between peptidepeptide and peptide-lipid interactions is required for optimal biological activity of amphipathic peptides. These studies provide a rationale for the design of small apoA-Imimetics with increased potency for atherosclerosis inhibition.
Introduction.
O
C [0174] Plasma levels of high density lipoproteins (HDL) and apolipoprotein A-I O (apo the major protein constituent of HDL, are inversely correlated to coronary Z artery disease (CAD) (Sprecher et al. (1993) Arterioscler. Thromb. 13: 495-504; Philips N 5 et al. (1993) Circulation 88: 2762-2770). Human apo A-I is a 243 residue protein, containing eight 22-mer amphipathic helical repeats, the majority of which have been tI shown to possess the Class A motif (Segrest et al. (1990) Proteins 8: 103-117; Anantharamaiah et al. (1993) pp. 109-142 In: The Amphipathic Helix (Epand, R. M., Cc€ C ed), CRC Press, Boca Raton, FL). Class A amphipathic helices have a characteristic 0 10 charge distribution; they have a cluster of positively charged amino acids at the C polar/nonpolar boundary of the a helix and negatively charged residues at the center of the polar face (Segrest et al. (1990) Proteins 8: 103-117; Anantharamaiah et al. (1993) pp.
'109-142 In: The Amphipathic Helix (Epand, R. ed), CRC Press, Boca Raton, FL; Segrest et al. (1992) J. Lipid Res. 33: 141-166). This unique secondary structural motif has been postulated to be responsible for the lipid-associating property of apo A-I (Segrest et al. (1990) Proteins 8: 103-117). Many studies with synthetic analogues of Class A amphipathic helices have supported this concept (Segrest et al. (1994) Adv. Prot. Chem.
303-369; Brouillette and Anantharamaiah (1995) Biochim. Biophys. Acta 1256: 103- 129). Recently, we have synthesized each of the putative 22 mer helices present in human apo A-I as monomers and tandem dimers and shown that the N- and C-terminal amphipathic helices possess the maximum lipid-associating ability (Mishra et al. (1998) Biochemistry 37: 10313-10324). X-ray crystal structure and molecular modeling studies of the exon 4 (44-243 residues) of apo A-I suggests that a self-associated state of the entire apo A-Iis necessary for lipid association (Borhani et al. (1999) Proc. Natl. Acad. Sci.
USA. 94:12291-12296; Segrest et al. (2000) Current Opin. Lipidol. 11:105-115). In this model, two molecules of apo A-I are arranged in the form of a head-to-tail dimer with the monomers interacting with each other to stabilize the lipid-associated structure of apo A-I.
[0175] Experimental evidence suggests that the protective effect of apo A-I and HDL against coronary artery disease could be due to their role in "reverse cholesterol transport" (Fielding and Fielding (1995) J. Lipid Res. 36: 211-228; Glomset (1968) J.
Lipid Res. 9:155-167). Reverse cholesterol transport is the sum of three steps involving HDIJapo A-I, a) efflux of cholesterol from xx cells (Johnson et al. (1991) Biochim.
-61- Biophys. Acta. 1085: 273-298; Oram and Yokoyama (1996) J. Lipid Res. 37: 2473- C- 2491), b) esterification by LCAT of HDL-associated cholesterol (Fielding et al. (1972) Biochem. Biophys. Res. Comm. 46: 1493-1498; Jonas (1991) Biochim. Biophys. Acta Z 1084: 205-220) and c) receptor-mediated delivery of cholesterol ester to the liver (Kreiger C- 5 (1999) Ann Rev. Biochem. 68: 523-558). In vivo studies have shown that both human apo A-I and a class A synthetic amphipathic helical peptide inhibit atherosclerosis without altering plasma cholesterol levels by a mechanism that is independent of reverse cholesterol transport (Shah et al. (1998) Circulation 97: 780-785). Recently, we have C, suggested that inhibition of LDL-induced monocyte chemotaxis into artery wall cells has 0 10 been suggested to be another major role played by apo A-I and HDL in preventing atherosclerosis (Navab et al. (2000) J. LipidRes. 41: 1481-1494; Navab et al. (2000) J.
Lipid Res. 41: 1495-1508).
[0176] A peptide that has been shown to mimic the properties of human apo A-I, 18A, has also been shown to possess LCAT activating (Anantharamaiah et al. (1990) Arteriosclerosis 10: 95-105; Epand et al. (1987) J. Biol. Chem. 262: 9389-9396) and cholesterol effluxing abilities (Davidson et al. (1994) J. Biol. Chem. 269: 22975-22982; Yancey et al. (1995) Biochemistry, 34: 7955-7965). Neutralizing the terminal charges of 18A to form Ac-18A-NH 2 was shown to increase its lipid affinity, and biological activities (Yancey et al. (1995) Biochemistry, 34: 7955-7965; Venkatachalapathi et al. (1993) Proteins: Structure, Function and Genetics. 15: 349-359). Several modifications of the amino acid sequence of this 'parent' molecule, 18A, have been made in an attempt to improve its apo A-I mimicking properties (Brouillette and Anantharamaiah (1995) Biochim. Biophys. Acta 1256: 103-1291; Mishra et al. (1994) J. Biol. Chem. 269: 7185- 7191; Mishra et al. (1995) J. Biol. Chem. 270: 1602-1611 Our earlier studies (Brouillette and Anantharamaiah (1995) Biochim. Biophys. Acta 1256: 103-1291; Epand et al. (1987) J. Biol. Chem. 262: 9389-9396) have shown that an increase in the hydrophobicity of this peptide increases its lipid affinity and apo A-I-mimicking properties. A synthetic peptide 5F, an analog of Ac-18A-NH 2 with increased amphipathicity has been shown to inhibit diet-induced atherosclerosis in mice (see, e.g., Examples 1 and However, the peptide 2F did not significahtly inhibit diet-induced lesion formation in C57 BL6 mice (Garber et al. (1999) Circulation 100: 1538). A study of 18A dimer peptides indicated that increased peptide-peptide association decreased -62peptide:lipid association (Mishra et al. (1995) J. Biol. Chem. 270: 1602-1611). To c determine the maximum extent to which the lipid affinity of the 18A peptide can be increased with a positive effect on lipid-associating and apo A-I-mimicking properties, we Z designed a homologous series of peptides in which Phe residues were systematically N 5 increased by substituting hydrophobic amino acids such as, Leu and Ala on the nonpolar face with Phe. According to the experimentally determined hydrophobicity scale of Wimley and White (Wimley and White (1996) Nature Struc. Biol. 3: 842-848), Trp and Phe are the most hydrophobic amino acids in the sense that they exhibit the greatest C, partitioning into the membrane from the aqueous phase. We elected to use Phe to increase C 10 the hydrophobicity of the peptide because it is the most acid-resistant hydrophobic amino acid in membrane active peptides and Phe-containing peptides can be synthesized more easily than Trp-containing peptides. The effects of this increase in hydrophobicity on the physical and lipid associating properties, and apo A-I-mimicking biological properties such as LCAT activation and inhibition of LDL-induced chemotactic activities, were studied.
Experimental procedures.
Peptide synthesis.
[0177] The peptides were synthesized by the solid phase method using an automated solid phase synthesizer (PS3 Protein Technologies, Woburn, MA). FMOCamino acids were coupled to a rink amide resin 536 mEq/g], (Peninsula Laboratories, Inc. Belmont, CA) in the presence of HBTU and NMM, and acetylated with acetic anhydride at the N-terminus. The peptides were cleaved from the solid support using TFA in dichloromethane in presence of anisole mercaptoeathanol and tryptophan (20% by weight of the peptide resin) and purified on a VYDAC C-4 (22mm x 25cm, particle size 10 im) reversed phase HPLC (RP-HPLC) column using a gradient of to 58% acetonitrile in water containing 0. 1% TFA in 66 min. with a flow rate of 4. 8 ml/min. The purity of the peptides was verified by analytical RP-HPLC using a C 1 8 column (VYDAC, 4. 6mm x 25 cm, 5pm) and-a linear acetonitrile-water (in presence of 0.
1% TFA) gradient of 25% to 58% in 33 min., and by the mass spectral analysis.
Circular Dichroism.
C- [0178] CD spectra were recorded on an AVIV 62DS spectropolarimeter as O described by Mishra et al. (1994) J. Biol. Chen. 269: 7185-7191. Briefly, spectra were Z obtained using a cell with a 0. 1cm path length and measurements were taken every nm
\D
CN 5 from 260nm to 190nm at 25 0 C. All the CD spectra were signal averaged by adding four scans, base line corrected and smoothed. Peptide solutions in PBS, pH 7. 4, were used at a tI concentration of 11p M. Peptide-DMPC complexes (1:20 mol:mol) were used to determine the effect of lipid binding on the helicity of these peptides. These complexes Ci were prepared by adding the appropriate volume of peptide solution to DMPC O 10 multilamellar vesicles. DMPC multilamellar vesicles were prepared as follows: A known CI amount of lipid was dissolved in ethanol and the solvent was removed by evaporating slowly under a thin stream of nitrogen. Residual solvent was removed by storing the lipid film under vacuum overnight. An appropriate volume,of PBS, pH 7. 4 was added to the thin lipid film to give the required final concentration of DMPC. The lipid-peptide complexes were prepared by adding the required volume of peptide solutions to give a lipid to peptide molar ratio of 20:1. Due to the poor solubility of these peptides, a peptide concentration of 11iM was used. The mean residue ellipticity, [O]MRE (deg. cm 2 dmol-) at 222nm was calculated using the following equation: [e]MR MRW[0]/10cl where, MRW is mean residue weight of the peptide, 0 is the observed ellipticity in degrees, c is the concentration of the peptide in g/ml, and 1 is the path length of the cell in centimeters. The percent helicity of the peptide was estimated from the following equation as described by Morrisett et al. (1973) Biochemistry, 12: 1290-1299: a helicity ([]z222+3,000)/(36,000+3,000) where, [0]222 is the mean residue ellipticity at 222nm.
Differential Scanning Calorimetry.
[0179] DSC studies were carried out using a Microcal MC-2 scanning calorimeter (MicroCal, Inc. Amherst, MA) at a scan rate of 200 h' 1 for DMPC, and 37 0 C h- 1 for DiPoPE, using the procedure described by Mishra et al. (1994) J. Biol. Chem. 269: 7185- 7191. A known amount of phospholipid was dissolved in chloroform. For one set of -64samples, peptide was dissolved in methanol and added to a solution of DiPoPE in ,I chloroform/methanol For both, pure lipid samples and the organic solutions of lipid and peptide, solvent was removed under a slow stream of nitrogen. Residual solvent 0 Z was removed under vacuum. Buffer (PBS, pH 7. 4, for DMPC or 20 mM PIPES, 1 mM I 5 EDTA, 150 mM NaCI and 0. 002% NaN 3 pH 7. 4, for DiPoPE) alone or a known concentration of peptide solution in buffer to give a specific lipid/peptide molar ratio was added to the dried film and hydrated by vortexing at room temperature for 30 min. For DMPC, four consecutive scans with a 60 min. equilibration time between scans were r n taken. DSC thermograms were analyzed using the software provided by MicroCal Inc., Amherst, MA, and Origin, version 5. 0.
Surface Pressure Measurements.
[0180] Monolayer exclusion pressure measurements give the affinity of the peptides for a lipid-water interface; the procedure of Phillips and Krebs (Phillips and Krebs (1986) Methods Enzymnol. 128: 387-403; Ibdah et al. (1989) Biochim. Biophys.
Acta 1004: 300-308) was followed. An insoluble monolayer of egg phosphatidylcholine (EPC) was spread at the air-water interface in a Teflon dish at room temperature to give an initial surface pressure (7t) in the range of 5-45 dyn/cm. A solution of peptides in PBS containing 1. 5M Gdn. HC1 was carefully injected in to the subphase to give a final concentration of 50p.g/dL. The Gdn. HCI was diluted in the subphase to a final concentration of <lmM to allow the peptides to renature. The subphase was stirred continuously and the increase in EPC monolayer surface pressure (Ant) was recorded until a steady state value was obtained. The value of the initial surface pressure (ri) at which the peptides no longer penetrate the EYPC monolayer i.e. the exclusion pressure was calculated by extrapolating the ji vs At linear regression fit to An 0 dyn/cm.
Right Angle Light Scattering Measurements.
[0181] Association of these peptides with egg phosphatidylcholine was determined by following the dissolution of EPC multilamellar vesicles (MLV) by right angle light scattering using a SLM 8000C photon counting spectrofluorometer as described in (Mishra et al. (1994) J. Biol. Chem. 269: 7185-7191). EPC MLVs were prepared by evaporating a solution of EPC (Avanti Polar, AL) under nitrogen and hydrating the lipid film with phosphate-buffered saline (pH 7. The sample containing 105pM EPC and an 0 CI equimolar amount of peptide was maintained at 25 0 C and continuously stirred. Turbidity Sclarification was monitored for 30 min. Complete dissolution of EPC vesicles was Z achieved by addition of Triton X-100 to a final concentration of 1mM.
Lecithin:Cholesterol Acvltransferase (LCAT) Purification.
I[0182] LCAT was isolated from fresh normolipidemic plasma by the method of Albers et al. (1986) Metlwds Enzymol. 129: 763-783, with some modifications. The C density of the plasma was adjusted to 1. 21 g/ml and it was centrifuged at 175,000 g for 24 Sh. The LCAT containing fraction was subjected to Affi-Gel Blue chromatography CI 10 followed by DE-52 chromatography. LCAT was eluted from the DE-52 column using a to 200mM NaC1 gradient in Tris buffer (10mM, pH 7. SDS-PAGE showed greater than 90% purity of the enzyme with no human apo A-I, contamination.
Assay of LCAT activity: [0183] The substrate was prepared by sonicating egg PC/cholesterol (90:20 mol/mol) containing trace amounts of 7a- 3 H cholesterol in a Branson 250 sonifier for 12 mins to obtain small unilamellar vesicles. The substrate (50gl) was incubated with 5pg of peptide or human apo A-I and 50l of BSA (40pg/ml) for 1 h at 37 0 C. The total volume was brought up to 150l1. After incubating for lh, 100pl of LCAT was added and incubated for lh at 37 0 C and the reaction was quenched by spotting 10l on a silica strip.
Cholesterol and cholesteryl ester were separated by thin layer chromatography of the silica strip in hexane:chloroform 1v/v) mixture. Cholesterol and cholesteryl oleate standards were visualized by immersing the TLC plate in a 3% cupric acetate, 8% phosphoric acid buffer and heating it. The positions of the standards were used to cut the strip into two and the two parts were counted in scintillation fluid in a Packard Tri Carb 4530. All reactions were done in triplicate. The activation of LCAT by the peptides is expressed as a percentage of the total activation by apo A-I.
-66- 0 Electrophoresis: C [0184] Non-denaturing and SDS-PAGE and was carried out using the method of 0 Laemmli (1970) Nature 227: 680-685. Premade Novex gels were used and the gel was Z stained with Coomassie blue to identify the protein bands.
LDL-induced Monocvte Chemotactic Activity LDL-induced Monocvte l Chemotactic Activity: [0185] Cocultures of human artery wall cells, monocyte isolation, isolation of CI lipoproteins by ultracentrifugation from the plasma of normal human donors or from O mouse plasma by FPLC, and determination of lipid hydroperoxides and monocyte 1 10 chemotactic activity were performed as as described by Navab et al. (Navab et al. (1991) J. Clin. Invest. 88: 2039-2046; Navab et al. (1977) J. Clin. Invest. 99: 2005-2019).
Briefly, LDL and HDL were isolated from human plasma by the method of Havel et al.
(Havel et al. (1955) J. Clin. Invest. 43:1345-1353). Human aortic endothelial cells (HAEC) and smooth muscle cells (HASMC) were isolated as described by Navab et al.
(1991) J. Clin. Invest. 88: 2039-2046. Microtitre plates were treated with 0. 1% gelatin at 37°C overnight. HASMC were added at a confluent density of 1 x 10 5 cells/cm 2 Cells were cultured for two days, at which time they had covered the entire surface of the well and had produced a substantial amount of extracellular matrix. HAEC were subsequently added at 2 x 105 cells /cm 2 and were allowed to grow, forming a complete monolayer of confluent HAEC in two days. In all experiments, HAEC and autologous HASMC (from the same donor) were used at passage levels of four to six. Monocytes were isolated blood from normal donors as described by Fogelman et al. (1988) J. Lipid Res. 29: 1243-1247.
The cocultures were treated with native LDL(250 pg protein /ml) or presence of HDL (350 p.g protein/ml) or peptides for 8h. The cocultures were then washed and incubated with medium 199 for an additional 8 h. The resulting coculture supernatants were assayed for monocyte chemotactic activity as described by Navab et al. (1997) J Clin Invest, 99: 2005-2019.
-67- 0Results.
Analysis of the peptides.
Z [0186] Table 6 shows the sequences of the various 18A analogues that were C synthesized. The peptide Ac-18A-NH 2 which has two Phe residues at positions 6 and 18 (close to the interfacial Lys residues) is referred to as 2F. Two 3F peptides were Sy synthesized, 3F 3 or 3F 1 where Leu in position 3 and 14 (both present at the center of the Snonpolar face) is replaced by Phe, respectively. Peptide 4F has two Phe residues at the C, center of the nonpolar face that is a result of substitution of two central Leu residues. The substitutions in the peptides (3F to 7F) are shown in Table 6. With an increase in the C 10 number of Phe residues the theoretical hydrophobicity per residue on the nonpolar face increases from 2. 05 for the peptide, 2F, to 3. 15 for 7F.
Table 6: Modifications of Ac-18A-NH2 to increase hydrophobicity Peptide Sequence 1 Hydrophobicity 2 Theoretical lipid affinity
(A)
3 2F Ac-18A-NH2 2. 05 13.03 3F 3 Ac-[F18A]-NH 2 2. 20 13.84 3F 1 4 Ac-[F' 1 18A]-NH 2 2.20 13.79 4F Ac-[FF 3 1418A]-NH2 2. 35 14. 59 Ac-[F" 11 1 4 17 18A]-NH 2 2. 81 19. 07 6F Ac-[Flo' 0 14 1 718A]-NH2 2. 96 19.87 7F Ac-[ F3' 10 ,l14' 1 7 18A]-NH 2 3.15 20. 78 1Baseline sequence 18A DWLKAFYDKVAEKLKEAF (SEQ ID NO:2) SHydrophobicity is expressed as the hydrophobicity per residue on the nonpolar face.
3 Theoretical lipid affinity has been calculated as shown in (Palgunachari et al. (1996) Arterioscler. Thromb. Vasc. Biol. 16: 328-338).
[0187] The peptides were purified on a preparative Vydac C 4 column by reversedphase (RP)-HPLC using water (with 0. 1% trifluoroacetic acid) and acetonitrile 1% trifluoroacetic acid). The purity and the retention times of the peptides were determined on an analytical Vydac Cis column using a gradient of 25%-58% acetonitrile in water containing 0. 1% TFA. The purity of these peptides was also confirmed by mass spectrometry. The mass was in agreement with the calculated molecular weight. The -68- O retention times of the peptides are listed in Table 7. Although both the 3F peptides and 4F 1 have additional Phe residues compared to 2F, the retention times of these peptides on the O C 18 column are not very different (~22min). A sudden increase in the retention time is apparent with 5F, 6F and 7F (~26min). With increasing number of Phe residues, the C1 5 solubility of these peptides in PBS decreases. As can be seen from Table 7, the solubility of 2F, 3F 3 3F 1 4 and 4F 25 to 1. 4 mg/ml) are significantly higher than those of 5F, 6F In and 7F 03 to 0. 1 mg/ml).
Table 7. Physical Properties of the F-peptides.
Peptide Molecular Retention Solubility Monolayer SWeight' Time (mins) 2 (mg/ml) 3 Exclusion Pressure(t) 4 apo A-I 28000 28.0 >2.0 34 18A 2200 19.8 >2.0 37pA 4580 26.0 >2.0 41 2F 2242 22.5 >2.0 38 3F' 2276 21.0 1.25 38 3F 1 4 2276 21.2 1.45 39 4F 2310 22.0 1.30 2429 26. 5 0. 10 6F 2462 27.0 0.03 46 7F 2510 26. 0 0. 10 'The mass as determined by mass spectroscopy was very close to the theoretically calculated molecular weight.
2 The retention time is the time taken for the peptide to elute from a Vydac Cig column using the gradient 25%-58% of acetonitrile in water containing 0. 1% TFA in 33 mins.
Solubility was determined in PBS.
4Reproducibility of these measurements is ±1 dyn/cm [0188] The self-association of these amphipathic peptides was examined by nondenaturing polyacrylamide gel electrophoresis (PAGE). Figure 14 shows the mobility of 2F on both denaturing SDS (Figure 14A) and on non-denaturing (Figure 14B) gels. The molecular weight of 2F is 2242 and it can be seen as a single band on the SDS gel (Figure 14A) moving slightly lower than the lowest molecular weight standard 5-2. 5 kDa).
However, under non-denaturing conditions it forms aggregates in a concentration dependent manner as seen in Figure 14B. At lower concentrations (100p.g/ml) it forms 0 aggregates of two sizes while at a higher concentration (250Lg/ml) only the bigger C' aggregates are observed (Figure 14B). All the other peptides studied also exhibited O aggregation under non-denaturing conditions suggesting that the peptides possess a strong Z tendency to self-associate.
(N
Circular Dichroism.
[0189] The secondary structure of the peptides was determined by circular dichroism spectroscopy. Table 8 shows the percent helicity of the peptides in PBS and in C the presence of DMPC. In PBS, homologues 2F, 4F, 5F, 6F and 7F have a higher 0percentage helicity than 3F 3 and 3F 1 4 (Table Since 5F, 6F and 7F were sparingly Cl 10 soluble in PBS, the CD studies were carried out using 1 1AM of the peptides (a concentration at which they were all soluble). Peptide 2F showed 55% helicity, comparable to 5F in solution. Both 6F and 7F were slightly more helical (67% and 58% respectively) while 4F was slightly less Both the 3F peptides were much less helical. However, binding to DMPC considerably increased the helicity of all the peptides except for 6F (Table In a lipid environment, 2F, 5F and 7F showed a high helical content (68% to Although, the peptides 3F 3 and 3F 1 4 had a very small helical content in PBS, there was a significant increase in helicity in a lipid environment, from about 22% to 42% for 3F 3 and from 19% to 55% for 3F 14 The helicity of the peptides 6F and 4F did not change appreciably in the presence of lipid. However, these peptides were still less helical than peptides 2F and 5F. The CD results suggest that there is no systematic change in the helicities of the peptides with increasing substitution by Phe; peptides 2F and 5F exhibited maximum helicity in solution and in the presence of phospholipid.
Table 8. Helicities of the F-peptides in aqueous and lipid environments Peptides Percent Helicity PBS' DMPC' 2F 55 72 3F 3 22 42 3F' 4 19 4F 45 44 55 76 6F 67 7F 58 68 O1 1pM solutions of peptide was used. Peptide:DMPC ratio used was 1:20 (mol/mol). Three measurements were made and an error of was obtained.
0 Z DSC studies with DMPC and DiPoPE.
[0190] The effect of these 18A analogues on the chain melting transition of multilamellar vesicles of DIPC was studied by DSC using peptide-lipid mixtures at 100:1 S lipid/peptide molar ratio. Table 9 shows the transition temperatures and enthalpies of the j- chain melting transition of DMPC in the presence and absence of peptides. The pure lipid undergoes a pretransition at 13 0 C and a main chain melting transition at 23 0 C. The 0 10 addition of the peptides to DMPC resulted in a broadening of the gel to liquid-crystalline C transition and a lowering of the transition enthalpy (Table The pretransition was not seen in the presence of any of the peptides. Among the peptides studied, 2F, 3F 3 5F, and 6F reduced the transition enthalpy to the maximum extent (Table None of the peptides changed the transition temperature by more than 0. 2 0
C.
Table 9. Effect of the F-peptides on the chain melting transition parameters of DMPC Peptide TCM(°C) AH CM ATIn (C) (kcals/mol) DMPC 23.1 6.4 0.2 2F 23.2 4.5 3F 3 23.2 4.9 0.4 3F 1 4 23.2 5.5 0.3 4F 23.2 5.3 0.4 23.2 4.9 6F 23.1 4.0 7F 23.2 4.5 The DMPC/peptide ratio used was 100:1 (mol/mol). The concentration of the DMPC used was 1. 5 mM. TCM is the temperature at which the chain melting transition takes place, AHcM is the enthalpy of the transition and ATrn is the width at half maximum of the transition.
[0191] The shift in the bilayer to hexagonal phase transition temperature (TH) has been used to evaluate the effects of peptides oi-the intrinsic curvature properties of phospholipids (Epand (1998) Biochim. Biophys. Acta, 1376: 353-368). It was previously shown that 2F raises TH of DiPoPE (Tytler et al. (1993) J. Biol. Chem. 268: 22112- 22118). In the current study we prepared the peptide-lipid mixtures in two ways. One -71- 0 was by adding the peptide in organic solvent to the lipid in organic solvent followed by IN depositing the material as a film and subsequently hydrating with buffer. In the other 0 method, the peptide and lipid were mixed after each was hydrated separately. If the Z mixture comes to equilibrium prior to the DSC analysis, it should not matter how the NC 5 peptide and lipid are originally mixed. However, membrane systems can equilibrate slowly, in which case there may be more peptide in the lipid when it was incorporated at high concentrations into the lipid film. In general the results from both methods of sample preparation are similar (not shown) but the shift in TH tends to be larger for samples in CN1 which peptide was incorporated into a film composed of lipid and peptide. The variation 0 10 of the TH with mol fraction of peptide is shown for the various peptides and apo A-I C,1 (Figure 15). A linear increase in TH is observed for 2F and 5F while 4F behaves more like apo A-I in that a more rapid increase is observed at lower peptide concentrations. On the other hand, the two 3F analogues as well as 6F and 7F do not significantly affect TH.
Interaction of peptides with phospholipid monolavers.
[0192] The monolayer exclusion pressure, 7t, is the surface pressure at which peptides are no longer able to penetrate a monolayer of EPC. The value of 7T, reflects the theoretical lipid affinity of the peptide. The exclusion pressure of the F peptides increased with increasing number of Phe residues (Table All the peptides studied here had higher exclusion pressures than apo A-I and the parent peptide 18A. The value of 7r increased gradually from 2F to 4F (38 to 40 dyn/cm). This is in the range seen for 37pA, a tandem repeat of 18A punctuated by a proline. The exclusion pressure value increases significantly for 5F, 6F and 7F (40 to 45 dyn/cm). It is apparent that the 5F, 6F and 7F homologues possess a similar ability to interact with EPC monolayers, as determined by the exclusion pressure. It is interesting that the HPLC retention times and monolayer exclusion pressures for the F-peptides listed in Table 7 show parallel trends, with an abrupt increase between 4F and Right Angle Light Scattering.
[0193] As can be seen in Figure 16, all the peptides were able to clarify EPC MLVs, unlike apo A-I, which does not clarify EPC MLVs. The two homologous 3F peptides were the least effective in clarifying the EPC IlLVs. The homologous peptides -72- 0 2F, 5F, 6F and 7F, all clarified the EPC MLVs to a similar extents. Peptide 4F was the C, most effective in clarifying EPC MLVs with activity similar to that of Triton X-100. The S time for 50% clearance of the turbidity of EPC MLVs was also the shortest for the Z homologue 4F. Peptide 7F took the longest time to achieve 50%clearance; this was due to N 5 an initial lag period of -300 sees (Figure 16). This is probably due to the requirement for self-associated 7F molecules to dissociate before they can interact with EPC MLVs and solubilize them. The slower rates of clearance exhibited by the homologues 2F, 5F and 6F may also be due to a higher self-association of these peptides.
SActivation of the plasma enzyme LCAT.
1 10 [0194] The ability of these peptides to activate the plasma enzyme LCAT was determined by measuring the initial velocity of the LCAT reaction with egg PCcholesterol vesicles as substrate (Figure 17). LCAT activation is expressed relative to that by apo A-I, which was considered to be 100%. Activation of LCAT by 20pg/ml of peptides and apo A-I is shown in Figure 4. At this concentration, apo A-I activates LCAT better than any of the peptides. Among the peptides studied here, however, 5F is the best activator (80% of apo As far as LCAT activation is concerned, both 3F 3 and 3F 1 4 have similar activating abilities. Therefore, they have been represented as one bar (Figure 17).
LDL-induced monocyte chemotactic activity [0195] When LDL is incubated with the human artery wall coculture system, it is trapped in the subendothelial space and gets oxidized to produce biologically active lipids.
These lipids induce monocyte chemotaxis. Thus, coculture monocyte chemotaxis is a well-established assay for the formation of biologically active lipids. It has been shown that inhibition of chemotaxis is directly correlated with the removal of "seeding molecules" that are responsible for the secretion of monocyte chemotactic protein-1 (MCP-1) (Navab et al. (2000) J. LipidRes. 41: 1481-1494; Navab et al. (2000) J. Lipid Res. 41: 1495-1508) and differentiation factor macrophage colony-stimulating factor (M- CSF). Figure 18 shows that LDL after incubation with peptides exhibited varied effects with homologues 4F, 5F and 6F reducing the chemotactic properties of LDL the most.
O Peptides 3F were not at all effective compared to 2F and 7F, which were less effective Sthan the peptides 4F, 5F and 6F.
0 Z Discussion.
IND
Effect of increasing hydrophobicity of a class A amphipathic helical peptide analogue on its physical-chemical and lipid binding properties: [0196] The peptides studied in this paper are homologues of the parent peptide,
F'-
M 18A The calculated hydrophobicity per residue (according to modified GES scale (Palgunachari et al. (1996) Arterioscler. Thromb. Vasc. Biol. 16: 328-338)) on the O nonpolar face increased as the number of Phe residues increased. This increase in hydrophobicity (Table 6) is reflected in the theoretical lipid affinity, A (Ibid.). However, the A value increases gradually from 2F to 4F (from 13. 03 to 14. 59) with a sudden increase in the value from 14. 59 (for 4F) to 19. 07 for 5F. A gradual increase in A was again observed after 5F in the values for 6F and 7F (Table This is due to the substitution of Leu at positions 3 and 14 in Ac-18A-NH 2 with Phe which results in a slight increase in the hydrophobicity of the nonpolar face and thus, a slight increase in A values for the two 3F analogues and 4F. In homologues 5F, 6F and 7F however, besides the Leu to Phe substitutions, Ala in positions 11 and 17 are also substituted by Phe, resulting in a significant increase in the A values (Table Since Ala is less hydrophobic than Leu and Leu is less hydrophobic than Phe, the substitution of Ala to Phe causes a greater change in hydrophobicity and theoretical lipid affinity of the resulting peptide than a Leu to Phe substitution.
[0197] The retention time on a C 1 8 reversed phase HPLC column, solubility of these peptides and their ability to penetrate an EPC monolayer, all exhibit a trend similar to that seen in the theoretical lipid affinity values (Table The retention times of peptides 2F, 3F 3 3F 1 4 and 4F are about the same (21-22 min. and significantly less than those of 5F, 6F and 7F, which comprise a second group (26-27 min.). The peptides 2F to 4F have considerably higher aqueous solubility than homologues 5F to 7F, which are sparingly soluble (Table A gradual increase in exclusion pressure was observed from 2F to 4F after which there is an abrupt increase from 40 dyn/cm to 45 dyn/cm. The exclusion pressures for the peptides 5F, 6F and 7F are not very different from each other r.- 0 and are significantly higher than that of apo A-I (Table The parent peptide 18A CN dyn/cm) and even the dimer of 18A, 37pA (40 dyn/cm) were also significantly less O effective in penetrating into an egg PC monolayer spread at the air-water interface. Based Z on the above physical properties, the F peptides can be separated into two groups; group I C 5 with 2F, 3F 3F 4 4F and group II with peptides 5F, 6F and 7F.
[0198] The CD data (Table 8) indicate that the percent helicity value of all the peptides increases in the presence of DMPC suggesting that all of the peptides associate with lipids. The binding of these peptides to DMPC appears to be similar as suggested by DSC (Table However, the effect of these peptides on the stabilization of the bilayer structure of DiPoPE is different. 4F and 5F seem to interact better with DiPoPE because they appear to be better stabilizers than the other peptides.
[0199] While apo A-I is not able to clarify EPC MLVs, all of the peptide analogs are able to do so, but to different extents. Among the group I peptides that are easily soluble in aqueous buffer and exhibit a monolayer exclusion pressure value in the range 38-40 dyn/cm (2F, 3F analogs and 4F), 4F appears to be the most efficient and at the peptide:lipid ratio under investigation, exhibits similar kinetics to that of Triton X-100 (Figure 16). While the monolayer exclusion pressures of the peptides 2F and 3F are similar, the 3F homologues are the slowest in clarifying EPC MLVs. The reason for reduced EPC clarifying ability of the 3F homologues is not clear at this time. The group II peptides (5F, 6F and 7F) that are not easily soluble in aqueous buffer and possess surface pressure values 45dyn/cm solubilize EPC MLVs relatively slowly. These results are consistent with peptide aggregates having to disassociate and then interact with EPC. The superior reactivity of 4F can be explained by the fact that its hydrophobicity is optimal so that hydrophobic peptide:peptide interactions favoring self-association do not prevent peptide:lipid interactions.
Effect of increased hydrophobicity on LCAT activation: [0200] Activation of LCAT is a complex process and is not only dependent on lipid affinity but also on the interaction of the amphipathic helical protein with the enzyme LCAT (Jonas (2000) Biochim. Biophys. Acta 1529: 245-256). In agreement with this, the ability to activate LCAT was found to be different for the homologous peptides. The peptide 5F showed the maximum LCAT-activating ability, in agreement with the physical properties studied in Table 7 wherein an abrupt increase was seen from 4F to 5F, including exclusion pressure values at the egg PC-water interface. The fact that the peptides 6F and 7F are not as effective as 5F could be explained by the increased peptide:peptide
O
Z interaction (as reflected in the low aqueous solubility of these peptides) which does not allow for peptide:lipid or peptide:LCAT interaction. These results are in agreement with our earlier observations with the 18A dimer peptides in which the enhanced selfassociation of the dimer 18A-18A (36A) peptide reduced its ability to interact with lipids Scompared to 18A-Pro-18A peptide (Jonas (2000) Biochim. Biophys. Acta 1529: 245-256).
c Although LCAT activation by the peptides has been compared with that of apo A-I, it 10 should be noted that apo A-I and the peptides interact differently with the substrate since they all have different reactivities to EPC (Figure 16). Similar observations were made by Chung et al who showed that a synthetic peptide 18A-Pro-18A and apo A-I interact differently with EPC (Chung et al. (1985) J. Biol. Chem. 260: 10256-10262).
Effect of increased hydrophobicity of the nonpolar face on LDL-induced monocyte chemotaxis: [0201] Since removal of "seeding molecules" depends on the amphipathicity of the peptide as reported by us (Navab et al. (2000) J. Lipid Res. 41: 1481-1494; Navab et al.
(2000) J. Lipid Res. 41: 1495-1508), we examined the ability of these peptides to inhibit LDL-induced monocyte chemotaxis. In this assay, peptides 4F, 5F and 6F at 100 gg/ml level, showed significant and similar inhibition of LDL-induced chemotaxis based on one way analysis of variance. Although the homologue 2F showed some inhibitory activity, for reasons that are not clear, peptide analogs 3F showed no inhibition compared to LDL alone. These results were in agreement with the fact that the peptide 3F was not able to remove the lipid hydroperoxides (results not shown) and the reduced ability to clarify EPC MLVs. Peptide 7F was significantly less effective than peptides 4F, 5F and 6F (P<0.
001). The reduced ability of 7F can again be explained by increased self-association of the peptide that decreased its ability to interact with the lipid as seen in EPC MLV clarification studies. These results again demonstrate that the delicate balance existing between the contributions of the hydrophobicity of the peptide to self association can critically affect apo A-I-mimicking properties.
-76- [0202] In vivo administration of peptide 5F, which possesses increased LCAT- Sactivating ability and increased ability to remove "seeding molecules" protected mice from diet-induced atherosclerosis. In contrast, administration of 2F, that is similar in LCAT- 0 activating ability to 4F, but less effective than 4F and 5F in removing "seeding molecules" I 5 from LDL, did not significantly inhibit diet-induced lesion formation in C57 BL6 mice (mean lesion area for control mice administered with PBS 14. 7+ 1. 8 PLm 2 X 10 3 compared to 2F-administered mice 13. 2+ 1. 7 m 2 X10O 3 n= 15). It follows that in this mouse model, inhibition of LDL-induced monocyte chemotaxis is more anti-atherogenic than LCAT activation. Since the peptides 2F and 4F are similar in activating LCAT, and a 10 4F and 5F are similar in removing "seeding molecules" from LDL, the peptide 4F may CI serve as a reagent to distinguish between the importance of LCAT activation and the inhibition of LDL-induced monocyte chemotaxis in different atherosclerosis-sensitive mouse models. If the inhibition of LDL-induced chemotaxis is more important than the LCAT-activating ability, then 4F should be better peptide to use as an inhibitor of atherosclerosis since this peptide is more soluble than the peptides 5F, 6F and 7F.
Example 4 Peptides D-4F Maintains Paroxynase Levels and Blocks Oxidized Phospholipid Production During an Acute Inflammatory Response [0203] We have observed that intranasal instillation of the influenza A virus in mice caused a time dependent loss in the anti-inflammatory properties of HDL reaches a maximum 7 to 9 days after inoculation. The dose chosen was one that did not cause viremia and so the changes were not due directly to the virus but were due to the inflammatory state induced by the host's systemic response to the viral infection. This response is part of the innate immune system and is known as the acute phase reaction or acute phase response.
[0204] One of the consequences was dimunition in paraoxonase and platelet activating acetylhydrolase activity in the HDL of the mice after the influenza infection.
As a result of the loss of these HDL enzymatic activities and also as a result of the association of pro-oxidant proteins with HDL during the acute phase response, HDL was no longer able to prevent LDL oxidation and was no longer able to prevent the LDLinduced production of monocyte chemotactic activity by endothelial cells. Normal HDL is S able to prevent the LDL-induced production of monocyte chemotactic activity by Sendothelial cells because normal HDL contains sufficient paraoxonase and platelet activating acetylhydrolase activities to destroy the biologically active oxidized Z phospholipids.
CN 5 [0205] In this example, we demonstrate that early (two days) after influenza A infection the livers of infected mice generated these oxidized phospholipids (Figure 19) In and later (7 to 9 days after infection) these biologically active oxidized phospholipids appeared in the aorta of the mice. However, if the mice were injected with 20 micrograms CI of D-4F daily after infection with the influenza A virus paraoxonase levels did not fall C 10 (Figure 20) and the biologically active oxidized phospholipids were not generated beyond C background (Figure 21).
[0206] These data indicate that D-4F (and/or other peptides of this invention) can be given either orally or by injection to patients with khown coronary artery disease during influenza infection or other events that can generate an acute phase inflammatory response due to viral infection, bacterial infection, trauma, transplant, various autoimmune conditions, etc.) and thus we can prevent by this short term treatment the increased incidence of heart attack and stroke associated with pathologies that generate such inflammatory states.
Example Oral Administration of an Apo A-I Mimetic Peptide Synthesized from D-Amino Acids Dramatically Reduces Atherosclerosis in Mice [0009] Apo A-I mimetic peptides synthesized from either D or L amino acids were effective in vitro in protecting low density lipoprotein (LDL) against oxidation by artery wall cells. However, when the peptides were given orally to LDL receptor null mice and their HDL was isolated and tested for its ability to protect LDL against oxidation in vitro, only the peptides synthesized from D-amino acids were effective. The peptide synthesized from D-amino acids was stable in the circulation and was found in fractions associated with high density lipoproteins (HDL). The peptide synthesized from L amino acids was rapidly degraded and excreted in the urine. When the peptide synthesized from D-amino acids known as D-4F was administered orally, twice daily, to LDL receptor null mice on a Western diet, lesions decreased by 79%. When added to the drinking water of apo E null -78mice, D-4F decreased lesions by more than 84%. We conclude that orally administered Sapo A-I mimetic peptides synthesized from D-amino acids are useful for the prevention and treatment of atherosclerosis and other chronic inflammatory illnesses that are caused 0 Z by oxidized lipids.
ID
Background.
[0010] HDL-cholesterol concentrations are inversely related to the risk for atherosclerotic coronary artery disease (Miller and Miller (1975) Lancet, 1: 16-19).
r n Infusion (Badimon et al. (1990) J Clin Invest, 85: 1234-1241) or transgenic expression (Plump et al. (1994) Proc. Natl. Acad. Sci., USA, 91: 9607-9611) of apo A-I, the major apolipoprotein of HDL, has been shown to protect against atherosclerosis in animal models. The mechanisms by which apo A-I protects against the development of atherosclerosis have been postulated to include reverse cholesterol transport (Shah et al.
(2001) Circulation, 103: 3047-3050) and removal of low levels of oxidized lipids, "seeding molecules", that are required to oxidize LDL (Navab et al. (2000) JLipid Res.
41: 1481-1494; Navab et al. (2000) JLipid Res, 41: 1495-1508; Navab et al. (2001) Arterioslcer Thromb Vasc Biol. 21: 481-488). Class A amphipathic helical peptide analogs have been shown to mimic several in vitro properties of apoA-I including the removal of the "seeding molecules" that are required for LDL oxidation (Navab et al.
(2000) J Lipid Res. 41: 1481-1494; Navab et al. (2000) J Lipid Res, 41: 1495-1508).
Intraperitoneal administration of a class A amphipathic helical peptide has recently been shown to protect mice from diet induced atherosclerosis without changing their plasma cholesterol levels (Garber et al. (2001) J Lipid Res, 42: 545-552). The reduction in lesions was associated with a significant improvement in the ability of HDL to inhibit LDL oxidation in vitro Up to now, the major limitation for the use of apo A-I or apo A-I mimetic peptides as a pharmacological agent has been the need for a parenteral route of administration.
[0011] Mammalian enzymes such as proteases recognize peptides and proteins synthesized from L-amino acids but rarely recognize those synthesized from D-amino acids. We demonstrate here that specific formulations of apo A-I mimetic peptides synthesized from D-amino acids can be administered orally and dramatically inhibit atherosclerosis in mice.
Methods
O
Mice.
0 [0012] Female LDL receptor null or apo E null mice on a C57BL/6J background ,0 were purchased from the Jackson Laboratory, Bar Harbor, ME. The LDL receptor null mice were maintained on Purina chow diet (Ralston Purina Co.) until they were 4 weeks old when they were switched to a Western diet (Teklad, Madison, WI, diet 88137) for 6 In weeks. The apo E null mice were maintained on Purina chow diet throughout the study.
l"- C LDL receptor null mice received the test peptide or a vehicle control by gastric gavage twice daily for the periods indicated in the figure legends. At four weeks of age the test 0 10 peptide was added to the drinking water of some of the apo E null mice at the concentrations indicated in the figure legends and the apo E null mice were continued on the chow diet.
[0013] Mice were bled under anesthesia from the retro-orbital venous plexus, in accordance with protocols approved by the UCLA Animal Research Committee.
Atherosclerotic lesions were measured as previously described 9 Lipoproteins [0014] LDL and HDL were isolated as described (Navab et al. (2000) J Lipid Res.
41: 1481-1494, and examples herein) after obtaining informed consent from human volunteers and from the blood of mice as noted above.
Cocultures, Cellular Oxidation of LDL, Monocyte isolation, and Monocyte Chemotaxis Assay [0015] Human aortic endothelial and smooth muscle cells were isolated and cultured as previously described Cellular oxidation of LDL in the presence and absence of HDL was determined as described Human blood monocytes were isolated after obtaining informed consent and monocyte chemotaxis was determined as described previously 5 Synthesis and preparation of apo A-I mimetic peptides C [0016] Apo A-I mimetic peptides were synthesized as previously described" except that in some instances each amino acid in the peptide was the D-stereoisomer of the Z amino acid. The peptides are based on the sequence Ac-D-W-L-K-A-F-Y-D-K-V-A-E-K- C 5 L-K-E-A-F-NH 2 (SEQ ID NO: 1) (Ac-18A-NH2 or 2F). 2F or an analog of 2F with the primary amino acid sequence Ac-D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F-NH2
(SEQ
l ID NO:5, also designated 4F) was used in the studies reported here. Peptides synthesized S from the L-amino acids are designated with an L L-4F) and peptides synthesized C, from D-amino acids are designated with a D D-4F). In some cases the peptides were iodinated using IODO-BEAD reagent (Pierce, Rockford, IL) according to the C-i recommendations by the manufacturer. Liposomes made of L-a-l-palmitoyl-2-oleyl-snglycero-3-phosphocholine (Avanti Polar Lipids, Alabaster, AL) with and without D-4F were made according to the manufacturer's recommendations. Extraction and detection of intact peptides from mouse plasma was performed as described by Garber et al. (1992) Arterioscler Thromb. 12: 886-894, using reverse phase HPLC.
Other Methods [0017] Protein (Navab et al. (2000) J Lipid Res. 41: 1481-1494) and cholesterol (Van Lenten et al. (2001) Circulation, 103: 2283-2288) content of lipoproteins and statistical analyses were performed as described by Van Lenten et al. (2001) Circulation, 103: 2283-2288, with significance defined as P<0.05.
Results [0018] In vitro, both L-2F and D-2F were equally able to block LDL oxidation and LDL-induced monocyte chemotactic activity in human artery wall cell cocultures (data not shown). In vivo however, as shown in Figure 22 A after oral administration, only D-4F remained intact in the circulation and was able to enhance HDL protective capacity (Figure22 B) and decrease LDL-induced monocyte chemotactic activity (Figure 22C).
Two hours after oral administration of 13I -L-4F or, 125 I -D-4F the urine from mice given L-4F had approximately 15 times more radioactivity than was the case for mice given D- 4F (data not shown).
[0019] Figure 23, demonstrates that twice-daily administration of D-4F by gavage O reduced atherosclerotic lesions in LDL receptor null mice on a Western diet by 79%. Total plasma cholesterol levels did not significantly differ in the LDL receptor null mice given
O
z D-4F and those given liposomes alone or saline alone. Total cholesterol was 761± 69 IN 5 mg/dl for the D-4F group, 677±52 mg/dl for the liposome group and 699± 31 for the saline group. The mean HDL-cholesterol level was slightly higher in the D-4F group, 73±8.7 mg/dl, compared to 65.9±9.2 for the liposome group and 67.1±6.3 for the saline group, but these differences did not reach statistical significance.
C [0020] Figure 24 demonstrates that the apo E null mice given D-4F in their 0 10 drinking water had more than an 84% reduction in lesions and was not significantly CI different whether the mice were given 2.5 mg/day/mouse or 5.0 mg/day/mouse. There was no significant difference in the amount of water consumed (2.5 ml/day/mouse) between the apo E null mice receiving no peptide, or those receiving 2.5 mg D- 4F/mouse/day or 5.0 mg D-4F/mouse/day and there was no significant difference in body, heart, or liver weights of the apo E null mice in the three groups (data not shown).
Furthermore, the plasma total cholesterol concentration was not significantly different in the apo E null mice that received D-4F (478±149 mg/dl for mice receiving water without peptide, 534±12.3 mg/dl for mice receiving D-4F at 2.5 mg/mouse/day, and 579±4.6 mg/dl for mice receiving D-4F at 5.0 mg/mouse/day). Mean HDL-cholesterol was mildly increased in the mice receiving D-4F but the differences did not reach statistical significance (32.2±7 mg/dl for mice receiving water without peptide, 38.7±5 for mice I receiving D-4F at 2.5 mg/mouse/day, and 43.4±6 mg/dl for mice receiving D-4F at mg/mouse/day).
Discussion [0021] Up to this point in time, the use of apo A-I and apo A-I mimetic peptides as pharmacological agents has been limited by the need for parenteral administration. The marked reduction in atherosclerotic lesions in the present study occurred despite the absence of significant changes in total plasma cholesterol. Although, there was a trend toward slightly higher HDL-cholesterol levels in the mice receiving D-4F, this did not reach statistical significance. The studies presented here suggest that orally administered apo A-I mimetic peptides synthesized from D-amino acids may be useful for the -82prevention and treatment of atherosclerosis and other chronic inflammatory illnesses that C1 are caused by oxidized lipids.
0 [0207] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof C 5 will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, I) and patent applications cited herein are hereby incorporated by reference in their entirety r- for all purposes.
-83-

Claims (15)

1. A peptide that ameliorates a symptom of atherosclerosis, z wherein said peptide has the amino acid sequence selected from the group INO 5consisting of: D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO D-W- F-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO: D-W-L-K-A-F-Y-D-K-V A-E-K-F-K-E-A-F- (SEQ ID NO D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO D-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO 6), D-W-L-K-A-F-Y-D-K-F-F E-K-F-K-E-F-F- (SEQ ID NO D-W-F-K-A-F-Y-D- K-F-F-E-K-F-K-E-F-F- (SEQ ID NO D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E- F-F- (SEQ ID NO D-W-L-K-A-F-Y-D-K-V-F-E-K- F-K-E-A-F- (SEQ ID NO: D-W-L-K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ I D NO: 11), D-W-L-K-A-F- Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID D-W-L-K-A-F-Y-D-K-V-F-E-K-F- K-E-F-F- (SEQ ID NO: 13), E-W-L-K-L-F-Y-E-K-V-L-E-K-F-K-E-A-F- (SEQ ID NO: 14), E-W L-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO: 15), E-W-L- K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO: 16), E-W-L-K-A-F-Y-D-K-V-F- E-K-F-K-E-A-F- (SEQ ID NO: 17), E-W-L-K-A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ ID NO: 18), E-W-L-K-A-F-Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID NO: 19), E-W-L-K-A-F-Y-D-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO 20), A-F-Y-D-K-V-A-E- K-L-K-E-A-F- (SEQ ID NO 21), A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO: 22), A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO 23), A-F-Y-D-K-F-F-E-K-F- K-E-F-F- (SEQ ID NO 24), A-F-Y-D-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO A-F-Y-D-K-V-A-E-K-F-K-E-A-F (SEQ ID NO 26), A-F-Y-D-K-V-A-E-K-L-K-E- F-F- (SEQID NO 27), A-F-Y-D-K-V-F-E-K-F-K-E-A-F- (SEQ ID NO 28), A-F- Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ I D NO 29), A-F-Y-D-K-V- A-E-K-F-K-E-F-F- (SEQ ID NO 30), K-A-F-Y-D-K-V-F-E-K-F-K-E-F- (SEQ ID NO 31), L-F-Y-E- K-V-L-E-K-F-K-E-A-F- (SEQ ID NO 32), A-F-Y-D-K-V-A-E-K-F-K-E-A-F- (SEQ ID NO 33), A-F-Y-D-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO: 34), A-F-Y- D-K-V-F-E-K-F-K-E-A-F- (SEQ ID NO 35), A-F-Y-D-K-V-F-E-K-L-K-E-F-F- (SEQ ID NO 36), A-F-Y-D-K-V-A-E-K-F-K-E-F-F- (SEQ ID NO 37), A-F-Y- D-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO 38), D-W-L-K-A-L-Y-D-K-V-A-E-K-L-K- E-A-L- (SEQ ID NO: 39), D-W-F-K-A-F-Y-E-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO 40), D-W-F-K-A-F-Y-E-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO 41), E-W-L- K-A-L-Y-E-K-V-A-E-K-L-K-E-A-L- (SEQ ID NO 42), E-W-L-K-A-F-Y-E-K-V-A- 84 E-K-L-K-E-A-F (SEQ ID NO 43), E-W-F-K-A-F-Y-E-K-V-A-E-K-L-K-E-F-F- (SEQ ID NO 44), E-W-L-K-A-F-Y-E-K-V-F-E-K-F-K-E-F-F- (SEQ ID NO: E-W-L-K-A-F-Y-E-K-F-F-E-K-F-K-E-F-F- (SEQ ID NO: 46), E-W-F-K-A-F-Y-E- z K-F-F-E-K-F-K-E-F-F- (SEQ ID NO: 47), D-.F-L-K-A-W-Y-D-K-V-A-E-K-L-K-E- IND 5 A-W- (SEQ ID NO: 48), E-F-L-K-A-W-Y-E-K-V-A-E-K-L-K-E-A-W- (SEQ ID NO 49), D-F-W-K-A-W-Y-D-K-V-A-E-K-L-K-E-W-W- (SEQ ID NO: 50), E-F- W-K-A-W-Y-E-K-V-A-E-K-L-K-E-W-W- (SEQ ID NO: 51), D-K-L-K-A-F-Y-D-K- V-F-E-W-A-K-E-A-F (SEQ ID NO:. 52), D-K-W-K-A-V-Y-D-K-F-A-E-A-F-K-E-F- L- (SEQ ID NO 53), E-K-L-K-A-F-Y-E-K-V-F-E-W-A-K-E-A-F- (SEQ ID NO: 54), E-K-W-K-A-V-Y-E-K-F-A-E-A-F-K-E-F-L- (SEQ ID NO: 55), D-W-L-K-A- F-V-D-K-F-A-E-K-F-K-E-A-Y- (SEQ ID NO 56), E-K-W-K-A-V-Y-E-K-F-A-E-A- F-K-E-F-L- (SEQ ID NO: 57), D-W-L-K-A-F-V-Y-D-K-V-F-K-L-K-E-F-F (SEQ ID NO: 58), E-W-L-K-A-F-V-Y-E-K-V-F-K-L-K-E-F-F- (SEQ ID NO 59), D-W- L-R-A-F-Y-D-K-V-A-E-K-L-K-E-A-F- (SEQ ID NO 60), E-W-L-R-A-F-Y-E-K-V- A-E-K-L-K-E-A-F- (SEQ ID NO 61), D-W-L-K-A-F-Y-DR-V-A-E-K-L-K-E-A-F- (SEQ ID NO 62), E-W-L-K-A-F-Y-E-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO 63), D-W-L-K-A-F-Y-D-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO 64), E-W-L-K-A-F-Y- E-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO 65), D-W-L-K-A-F-Y-D-K-V-A-E-K-L-R- E-A-F- (SEQ ID NO 66), E-W-L-K-A-F-Y-E-K-V-A-E-K-L-R-E-A-F (SEQ ID NO 67), D-W-L-K-A-F-Y-D-R-V-A-E-R-L-K-E-A-F- (SEQ ID NO 68), E-W-L- K-A-F-Y-E-R-V-A-E-R-L-K-E-A-F- (SEQ ID NO 69), D-W-L-R-A-F-Y-D-K-V- A-E-K-L-R-E-A-F (SEQ ID NO: 70), E-W-L-R-A-F-Y-E-K-V-A-E-K-L-R-E-A-F- (SEQ ID NO 71), D-W-L-R-A-F-Y-D-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO: 72), E-W-L-R-A-F-Y-E-R-V-A-E-K-L-K-E-A-F- (SEQ ID NO 73), D-W-L-K-A- F-Y-D-K-V-A-E-R-L-R-E-A-F- (SEQ ID NO 74), E-W-L-K-A-F-Y-E-K-V-A-E-R- L-R-E-A-F- (SEQ ID NO 75), D-W-L-R-A-F-Y-D-K-V-A-E-R-L-K-E-A-F (SEQ ID NO 76), E-W-L-R-A-F-Y-E-K-V-A-E-R-L-K-E-A-F- (SEQ ID NO 77), D-W- L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-P-D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F (SEQ ID NO 78), D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-F-F-P-D-W-L-K-A-F-Y- D-K-V-A-E-K-L-K-E-F-F (SEQ ID NO 79), D-W-F-K-A-F-Y-D-K-V-A-E-K-L-K- E-A-F-P-D-W-F-K-A-F-Y-D-K-V-A-E-K-L- K-E-A-F (SEQ ID NO 80), D-K-L-K- A-F-Y-D-K-V-F-E-W-A-K-E-A-F-P-D-K-L-K-A-F-Y-D-K-V-F-E-W-L-K-E-A-F (SEQ ID NO.- 81), D-K-W-K-A-V-Y-D-K-F-A-E-A-F-K-E-F-L-P-D-K-W-K-A-V- Y-D-K-F-A-E-A-F-K-E-F-L (SEQ ID NO 82), D-W-F-K-A-F-Y-D-K-V-A-E-K-F- 85 r- K-E-A-F-P-D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E-A-F (SEQ ID NO: 83), D-W-L- 0 K-A-F-V-Y-D-K-V-F-K-L-K-E-F-F-P-D-W-L-K-A-F-V-Y-D-K-V-F-K-L-K-E-F-F (SEQ ID NO 84), D-W-L-K-A-F-Y-D-K-F-A-E-K-F-K-E-F-F-P-D-W-L-K-A-F-Y- D-K-F-A-E-K-F-K-E-F-F (SEQ ID NO 85), wherein said peptide comprises at least one amino acid residue and protects a phospholipid against oxidation by an oxidizing agent. The peptide of claims 1 which comprises all residues.
3. The peptide of claim 1 or claim 2, wherein said peptide further comprises at least one protecting group.
4. The peptide of claim 3, wherein said at least one protecting group is coupled to the amino and/or carboxyl terminus. The peptide of claim 4, wherein a first protecting group is coupled to the amino terminus and a second protecting group is coupled to the carboxyl terminus.
6. The peptide of claim 5, wherein said first protecting group and said second protecting group are independently selected from the group consisting of acetyl, amide, 3 to 20 carbon alkyl groups, Emoc, t-boc, 9- fluoreneacetyl group, 1-fluorenecarboxylic group, 9-florenecarboxylic group, 9- fluorenone-I-carboxylic group, benzyloxycarbonyl, Xanthyl (Xan), Trityl (Trt), 4-methyltrityl 4-methoxytrityl (Mint), 4-methoxy-2, 3,6-triinethyl- benzenesulphonyl (Mtr), Mesitylene-2-sulphonyl (Mts), 4,4- dimethoxybenzhydryl (Mbh),Tosyl (Tos), 2,2,5,7,8-pentamethyl chroman-6- sulphonyl (Pmc), 4-methylbenzyl (MeBzl), 4-methoxybenzyl (MeOBzl), Benzyloxy (BzlO), Benzyl (Bzl), Benzoyl (13z), 3-nitro-2-pyridinesulphenyl (Npys), 1 -(4,4-dimentyl-2 ,6-diaxocyclohexylidene)ethyI (Dde), 2,6- dichlorobenzyl (2,6-DiCI-Bzl), 2-chlorobenzyloxycarbonyl 2- bromobenzyloxycarbonyl Benzyloxymethyl (Born), t-butoxycarbonyl (Boc), cyclohexyloxy (cHxO), t-butoxymethyl (Bum), t-butoxy (tBuO), t-Butyl 86 (tBu), Acetyl a benzoyl group, a carbobenzoxy group, a propyl group, a 0butyl group, a pentyl group, a hexyl group, and Trifluoroacetyl (TFA). S7. The peptide of claim 4, wherein a said protecting group is Z coupled to the amino terminal and said amino terminal protecting group is a S 5 protecting group selected from the group consisting of a benzoyl group, an acetyl, a propionyl, a carbobenzoxy, a propyl, a butyl, a pentyl, a hexyl, and a 3 to 20 carbon alkyl.
8. The peptide of claim 4 or claim 7 wherein a peptide is coupled to the carboxyl terminal and said carboxyl terminal protecting group is an amide. c 10 9. The peptide of claim 3, wherein said amino terminal protecting group is an acetyl. The peptide of any one of claims 1 to 9, wherein said oxidizing agent is selected from the group consisting of hydrogen peroxide, 13(S)- HPODE, 15(S)-HPETE, HPODE, HPETE, HODE, and HETE.
11. The peptide of any one of claims 1 to 10, wherein said phospholipid is selected from the group consisting of 1-palmitoyl-2- arachidonoyl-sn-glycero-3-phosphorylcholine (PAPC), 1-stearoyl-2- arachidonoyl-sn-glycero-3-phosphorylcholine (SAPC), 1-stearoyl-2- arachidonyl-sn-glycero-3-phosphorylethanolamine (SAPE).
12. A pharmaceutical formulation comprising the peptide of any one of claims 1 to 11, wherein said peptide is mixed with a pharmacologically acceptable excipient.
13. The pharmaceutical formulation of claim 12, wherein said pharmacologically acceptable excipient is suitable for oral administration to a mammal.
14. The pharmaceutical formulation of claim 12, wherein said excipient is suitable for non-oral administration to a mammal. A peptide according to any one of claims 1 to 11 for use in the prophylaxis or treatment of atherosclerosis. -87-
16. A peptide according to any one of claims 1 to 11 for use in the prophylaxis or treatment of a coronary complication associated with an acute phase response to an inflammation. O S17. The use of a peptide according to any one of claims 1 to 11 for the manufacture of a medicament for the prophylaxis or treatment of atherosclerosis.
18. The use of claim 17 wherein said peptide is formulated as a unit t' dosage formulation in a pharmaceutically acceptable excipient.
19. The use of a peptide according to any one of claims 1 to 11 for c 10 the manufacture of a medicament for the prophylaxis or treatment of a coronary complication associated with an acute phase response to an inflammation. The use of claim 19, wherein said peptide is formulated as a unit dosage formulation in a pharmaceutically acceptable excipient.
21. A peptide that ameliorates a symptom of atherosclerosis, wherein said peptide is obtained by deletion, elongation, or conservative substitution of the amino acid sequence D-W-F-K-A-F-Y-D-K-V-A-E-K-F-K-E- A-F- (SEQ ID NO said peptide comprises all amino acid residues, and said peptide protects a phospholipid against oxidation by an oxidizing agent.
22. A method for the prophylaxis or treatment of atherosclerosis in a mammal, the method comprising administering to the mammal a therapeutically effective amount of the peptide of any one of claims 1 to 11 or the pharmaceutical formulation of any one of claims 12 to 14.
23. A method for the prophylaxis or treatment of a coronary complication in a mammal that is associated with an acute phase response to an inflammation comprising administering to said mammal a therapeutically effective amount of a peptide of any one of claims 1 to 11 or the pharmaceutical formulation of any one of claims 12 to 14. -88-
AU2007237157A 2000-08-24 2007-11-26 Peptides that ameliorate atherosclerosis Expired AU2007237157B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2007237157A AU2007237157B2 (en) 2000-08-24 2007-11-26 Peptides that ameliorate atherosclerosis
AU2009202705A AU2009202705C1 (en) 2000-08-24 2009-07-03 Peptides that ameliorate atherosclerosis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US09/645,454 2000-08-24
US09/645,454 US6664230B1 (en) 2000-08-24 2000-08-24 Orally administered peptides to ameliorate atherosclerosis
AU2006200035A AU2006200035B2 (en) 2000-08-24 2006-01-06 Peptides that ameliorate atherosclerosis
AU2007237157A AU2007237157B2 (en) 2000-08-24 2007-11-26 Peptides that ameliorate atherosclerosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2006200035A Division AU2006200035B2 (en) 2000-08-24 2006-01-06 Peptides that ameliorate atherosclerosis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2009202705A Division AU2009202705C1 (en) 2000-08-24 2009-07-03 Peptides that ameliorate atherosclerosis

Publications (2)

Publication Number Publication Date
AU2007237157A1 true AU2007237157A1 (en) 2007-12-13
AU2007237157B2 AU2007237157B2 (en) 2009-04-09

Family

ID=38846692

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007237157A Expired AU2007237157B2 (en) 2000-08-24 2007-11-26 Peptides that ameliorate atherosclerosis

Country Status (1)

Country Link
AU (1) AU2007237157B2 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4643988A (en) * 1984-05-15 1987-02-17 Research Corporation Amphipathic peptides
CN1109047C (en) * 1996-03-29 2003-05-21 D·博菲利 Amphipathic molecules as cholesterol and other lipid uptake inhibitors
US6037323A (en) * 1997-09-29 2000-03-14 Jean-Louis Dasseux Apolipoprotein A-I agonists and their use to treat dyslipidemic disorders

Also Published As

Publication number Publication date
AU2007237157B2 (en) 2009-04-09

Similar Documents

Publication Publication Date Title
AU2001286732B2 (en) Orally administered peptides to ameliorate atherosclerosis
US7144862B2 (en) Orally administered peptides to ameliorate atherosclerosis
CA2480217C (en) G-type peptides to ameliorate atherosclerosis
AU2001286732A1 (en) Orally administered peptides to ameliorate atherosclerosis
AU2005287004B2 (en) G-type peptides and other agents to ameliorate atherosclerosis and other pathologies
AU2007237157B2 (en) Peptides that ameliorate atherosclerosis

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired